Download as pdf or txt
Download as pdf or txt
You are on page 1of 469

Pathogenesis

of
Human
Pulmonary
Tuberculosis
Insights from the Rabbit Model
This page intentionally left blank
Pathogenesis
of
Human
Pulmonary
Tuberculosis
Insights from the Rabbit Model

Arthur M. Dannenberg, Jr., M.D., Ph.D.

Center for Tuberculosis Research


Departments of Environmental Health Sciences, Molecular Microbiology
and Immunology, and Epidemiology, Bloomberg School of Public Health
Department of Pathology, School of Medicine
Johns Hopkins University, Baltimore, Maryland 21205

Washington, D.C.
Max B. Lurie, M.D. (1893–1966)
Frontispiece: photo of Max B. Lurie courtesy of the late Peter Zappasodi, Henry Phipps Insti-
tute, School of Medicine, University of Pennsylvania, Philadelphia

Back cover: photo of Arthur M. Dannenberg, Jr., courtesy of Frederick W. Dubs, Photography
Laboratory of the Dept. of Pathology, Johns Hopkins Medical Institutions

Cover illustration by Robert Margulies © 1993 Hospital Practice

Copyright © 2006 ASM Press


American Society for Microbiology
1752 N Street, N.W.
Washington, DC 20036-2804

Library of Congress Cataloging-in-Publication Data

Dannenberg,Arthur M.
Pathogenesis of human pulmonary tuberculosis : insights from the rabbit model / Arthur
M. Dannenberg.
p. ; cm.
Includes bibliographical references and index.
ISBN-13: 978-1-55581-373-4
ISBN-10: 1-55581-373-9
1. Tuberculosis—Animal models 2. Tuberculosis—Pathophysiology I. American
Society for Microbiology. II.Title.
[DNLM: 1. Lurie, Max B. (Max Bernard), 1893-1966. 2. Tuberculosis, Pulmonary—
physiopathology. 3. Models,Animal. 4. Mycobacterium bovis—pathogenicity. 5.
Rabbits. 6. Tuberculosis, Pulmonary—microbiology WF 300 D188p 2006]

RC311.D362 2006
616.99507—dc22
2006021123

All Rights Reserved


Printed in the United States of America

10 9 8 7 6 5 4 3 2 1

Address editorial correspondence to:ASM Press, 1752 N St., N.W.,Washington, DC


20036-2904, U.S.A.

Send orders to:ASM Press, P.O. Box 605, Herndon,VA 20172, U.S.A.
Phone: 800-546-2416; 703-661-1593
Fax: 703-661-1501
Email: Books@asmusa.org
Online: estore.asm.org
Dedication
To Aileen Hart Dannenberg, my treasured wife of 58 years (to date), whose love
and continuous support have made my professional career and this book possible
CONTENTS

Preface xi
Introduction 1
Major contributions of Max B. Lurie and Arthur M. Dannenberg, Jr.

SECTION 1. PATHOGENESIS OF TUBERCULOSIS


1. Overview 7
Childhood and adult tuberculosis, bacillary virulence, host resistance, contagion,
and prevention
2. Stages in the Pathogenesis of Human and Rabbit
Tuberculosis 22
3. Types of Human Pulmonary Tuberculosis 34
4. Liquefaction of Caseous Foci and Cavity Formation 65

SECTION 2. IMMUNOLOGY OF TUBERCULOSIS


5. Delayed-Type Hypersensitivity, Cell-Mediated Immunity,
and Antibodies in Tuberculosis 97
Their local and systemic natures; innate immunity
6. Macrophages and Other Cells in Tuberculous Lesions 120
Including dendritic cells and lymphocytes

SECTION 3. TUBERCULOUS LESIONS


7. Structural Components of Tuberculous Lesions 155
8. Microvascular Density in Tuberculous Lesions 161
In developing and healing BCG lesions and in tuberculin reactions

vii
viii 䡵 CONTENTS

9. Early Pulmonary Lesions in Rabbits 170


Produced by an intravenous injection of tubercle bacilli
10. Macrophage Turnover, Division, and Activation in Tuberculous
Lesions 177
In developing, peak, and “healed” BCG lesions
11. Lurie’s Pulmonary Tubercle-Count Method 196
To assess bacillary virulence, genetic resistance of the host, and vaccine efficacy

SECTION 4. TUBERCULOSIS IN RABBITS AND OTHER


COMMON LABORATORY ANIMALS
12. Natural Airborne Infection 215
Resistance to the establishment of tuberculosis and to its progress
13. Response of Rabbits to Inhaled Tubercle Bacilli Including
BCG 230
Response to inhaled bovine- and human-type tubercle bacilli and to inhaled BCG
14. Characteristics of Resistance and Susceptibility to Tuberculosis
in Lurie’s Inbred Rabbits 235
15. Comparisons of Tuberculosis in Rabbits, Mice, and Guinea
Pigs 246

SECTION 5. EFFECTS OF HORMONES AND X-IRRADIATION


ON TUBERCULOSIS
16. Effects of Cortisone and Adrenocorticotropic Hormone
on Tuberculosis 273
17. Effects of Estrogen, Chorionic Gonadotropin, and Thyroid
Hormones on Tuberculosis 285
18. Effects of Whole-Body X-Irradiation on Tuberculosis 292

SECTION 6. CYTOKINES AND VASCULAR ADHESION


MOLECULES IN TUBERCULOUS LESIONS
19. Cytokine Production in Primary BCG Lesions 301
Nonspecific and antigen-specific cytokine production in developing and healing
primary BCG lesions
20. Cytokine Production in Reinfection BCG Lesions and
in Tuberculin Reactions 312
Effects of immunization on cell composition and cytokines
21. Vascular Adhesion Molecules in Tuberculous Lesions 327
ICAM-1,VCAM-1, and ELAM-1 in developing and healing rabbit
dermal BCG lesions
CONTENTS 䡵 ix

SECTION 7. TUBERCULOSIS VACCINES


22. Principles and Guidelines for Developing Better Tuberculosis
Vaccines 341
23. Characteristics of Rabbit BCG Lesions and Efficacies of BCG
and Mycobacterium microti Vaccines 354

SECTION 8. PAST, PRESENT, AND FUTURE


24. Summary and Conclusions 367
25. Suggested Future Research and Unanswered Questions 373
Research on the host; research on the bacillus; vaccines and immunotherapy;
development of new drugs

APPENDIXES
A. Award of the Trudeau Medal for 1955 385
The Trudeau Medal of the National Tuberculosis Association for 1955 awarded to
Max B. Lurie
B. Obituary of Max B. Lurie, M.D. (1893–1966) 387
by Esmond R. Long, M.D., Ph.D., Sc.D.
C. Publications of Max B. Lurie 389
D. Publications of Arthur M. Dannenberg, Jr. 399
E. Guidelines for Preventing the Transmission of Mycobacterium
tuberculosis in Health-Care Settings, 2005
(Table of Contents) 417
Department of Health and Human Services, Centers for Disease Control and
Prevention,Atlanta, Georgia
F. Collected Abstracts of Chapters in This Volume 419
G. Acknowledgments 429

Glossary 431
Index 439
This page intentionally left blank
PREFACE

This book is a review and update of my contributions to the experimental pathol-


ogy of tuberculosis, as well as those of my mentor Max B. Lurie (see Frontispiece).
The book describes 40 years of his scientific contributions, which were followed
by 40 years of mine. I was in his laboratory during the first 11 years that I worked
in this field.

BACKGROUND
Max B. Lurie, M.D.
Tuberculosis has afflicted many of the past workers in the field or their close fam-
ily members. Lurie probably caught tuberculosis from his mother, who died from
it. He became ill with the disease during his final year at Cornell Medical School,
and after graduating in 1921 he went to the National Jewish Hospital for Con-
sumptives in Denver, Colorado, for treatment.While there, he began his career in
tuberculosis research (with H. J. Corper as his mentor) and continued working on
the host-parasite relationships of this disease for the rest of his life.
When he had recovered from tuberculosis in 1926, he joined the research staff of
the University of Pennsylvania’s Henry Phipps Institute for the Cure and Preven-
tion of Tuberculosis, where he spent the rest of his career, attaining the rank of full
Professor (in Experimental Pathology). Lurie’s publications are listed in Appendix C.
Most of Lurie’s experiments are summarized in his book Resistance to Tuberculo-
sis: Experimental Studies in Native and Acquired Defensive Mechanisms (1). It almost seems
that his determination to complete that book kept him alive during the last few years
of his life. He died of an acute myocardial infarction in 1966, at the age of 73.

Arthur M. Dannenberg, Jr., M.D.


My mother’s first husband died of tuberculosis 6 months after they were married.
As a result, she became a social worker at the Henry Phipps Institute (where Lurie
worked). There she met my father, a practicing pediatrician, who devoted each

xi
xii 䡵 PREFACE

Tuesday afternoon to the care of tuberculous children in the Institute’s clinic.After


clinic, my father would go upstairs to Lurie’s laboratory, where he learned of his
latest experiments.
Lurie was an extremely enthusiastic person, and my father came home quite
inspired.Therefore, it was only natural that he would introduce me to Lurie when,
in 1948, I was deciding whether to pursue a clinical or a research career. I did not
realize at that time that Lurie was one of the world’s leading experimental pathol-
ogists of tuberculosis.
I was associated with Lurie for about 11 years—first as a postdoctoral fellow from
1948 to 1952, and then as an assistant professor from 1956 to 1964. During this time,
I had so many scientific discussions with him that his knowledge of tuberculosis has
became an integral part of my own thinking. In fact, I cite Lurie’s principles every
time I write or lecture.
Tuberculosis was (and is) one of the major diseases of humankind, especially in
developing countries. It kills about 2 million people in the world each year, more
than any other infectious disease (2). If I were going to pursue a research career
instead of a clinical one, I wanted to contribute to the elimination of a disease impor-
tant to mankind.
Fortunately, Lurie outlined what I should pursue in tuberculosis research at the
beginning of my career, specifically, the role of the macrophage, especially its
hydrolytic and metabolic enzymes. Macrophages are the host’s main defense against
the bacillus.The role of lymphocytes was just beginning to be recognized at that
time. More insight was needed from the disciplines of immunology and biochem-
istry into how these cell types function. In addition, the histopathology of the dis-
ease, as observed under the microscope, needed to be correlated with immunological
and biochemical cell functions.
Therefore, I learned the techniques of these (and other) basic sciences and have
used them in different stages of my career to understand the role of mononuclear
cells in tuberculosis lesions (see my publication list in Appendix D). Concurrently,
the whole discipline of histochemistry has evolved.This discipline enables scien-
tists to visualize structural and enzymatic cell characteristics in tissue sections.We
in my laboratory, therefore, used histochemistry to gain deeper insight into the patho-
genesis of tuberculous lesions and how the host controls them.

THE RABBIT MODEL OF TUBERCULOSIS


Lurie favored the rabbit model of tuberculosis because the disease in this labora-
tory animal most closely resembles that found in humans (1, 3–5). Caseous necro-
sis, liquefaction, and cavity formation with bronchial spread of the disease can be
readily produced in rabbits.Tuberculous lesions in mice, on the other hand, are gran-
ulomas that slowly progress with little or no caseation, and cavities never form. How-
ever, mice are an excellent model in which to study cell-mediated immunity in
tuberculosis. For such studies, many reagents are commercially available.
Tuberculosis in guinea pigs progresses rapidly with much caseation and hematoge-
nous and lymphatic spread of the disease. It resembles the susceptible form found
in infants and immunosuppressed persons, but not the slowly progressing fibrotic
cavity form found in immunocompetent humans and rabbits. Guinea pig lesions
occasionally cavitate, but significant bronchial spread is rare. Because of these
species differences, Lurie began rather early in his career to work with rabbits, after
working initially with guinea pigs. I have continued to use the rabbit model.
PREFACE 䡵 xiii

DERMAL BCG LESIONS


Many of my own studies in the rabbit model were made on developing, peak, and
healing dermal BCG lesions. In rabbits, these lesions resemble pulmonary lesions
produced by human-type tubercle bacilli in that they develop and then regress. Der-
mal BCG lesions begin with numerous bacilli injected into one site. Therefore,
caseation, liquefaction, and ulceration readily occur. However, for the study of nat-
urally occurring tuberculosis, there is no substitute for aerosol infection, where a
single unit of 1 to 3 bacilli in rabbits produces lesions that closely resemble those
occurring in human populations.
For each stage of pulmonary lesion development and healing, a rabbit must be
sacrificed (euthanized). In contrast, multiple dermal BCG lesions of various ages can
be produced in a single rabbit, or multiple lesions started at the same time can be
biopsied at various times under local anesthesia. However, the results of such stud-
ies must be confirmed in rabbits with no previous biopsy and in rabbits with all
lesions of the same age. Nevertheless, the use of multiple lesions on the same rab-
bit in part compensates for the marked difference in cost of purchase and mainte-
nance between rabbits and mice.

PURPOSE AND USE OF THIS BOOK


The purpose of this book is to present in one place our current understanding of
the pathogenesis of tuberculosis derived from Lurie’s and my own research.The book
is written for both clinicians and laboratory researchers. Clinicians will read parts
of it to gain insight into the pathogenesis of tuberculosis as a guide for the care and
treatment of their patients. Laboratory researchers will read parts of it to plan some
of their experiments. For such researchers, I have sometimes provided more details
and references.
I have used Lurie’s original publications as references for many of the statements
made herein and used his book as a reference when it contributed additional infor-
mation. Statements that I heard personally from him are also referenced to his book.

ORGANIZATION OF THIS BOOK


This book was organized for several types of readers, as follows.
(i) Readers wanting a quick understanding of the pathology and immunology
of tuberculosis can read chapter 24,“Summary and Conclusions,” and if more detail
is desired, they can read the chapter abstracts, which are assembled as Appendix F.
(ii) Readers wanting information on the individual subjects can read the perti-
nent chapters.At the head of each chapter, I have provided both an abstract and a
list of the main headings within the chapter.These should enable the reader to more
rapidly find the information he or she is looking for.The book was written so that
each chapter is complete within itself, which results in occasional duplication of text,
figures, and references but is a necessity for the reader of only individual chapters.
Finally, (iii) some readers may want to read the entire book for the various ori-
entations presented.
Many chapters contain more experimental details than others. These chapters
describe types of tuberculosis research that are usually not performed in other lab-
oratories and are therefore unfamiliar to many readers. Full details on the technol-
ogy, however, only appear in the original publications cited.
The “see” before a reference number in this book signifies that the reference con-
tains additional information.This information is pertinent but does not necessarily
xiv 䡵 PREFACE

support the statement to which the reference is attached.Without the “see,” the ref-
erence is supportive.
Note. Lurie (1) used the words “native” and “acquired” resistance.Today, because
of Janeway’s extensive studies (6), the words “innate” and “adaptive” immunity have
replaced Lurie’s terminology. In this book, we mostly used Lurie’s terminology,
because it seemed more appropriate for tuberculosis in his inbred rabbits.

OTHER BOOKS ON TUBERCULOSIS


The classic books on tuberculosis are those of Rich (Johns Hopkins University; 1951)
on the immunopathology (7), Canetti (Pasteur Institute; 1955) on the human
pathology (8), Lurie (1964) on the experimental pathology (1), and Iseman (2000)
on clinical tuberculosis (9). Multiauthored texts by authorities in each respective field
are those edited by Bloom (1994) (10), Reichman (2000) (11), Schlossberg (2006)
(12), Cole et al. (1995) (13), and Rom and Garay (2004) (14).These multiauthored
texts provide more details on the various subjects reviewed herein, but very few stud-
ies in the rabbit model of tuberculosis are included.

REFERENCES
1. Lurie, M. B. 1964. Resistance to Tuberculosis: Experimental Studies in Native and Acquired
Defensive Mechanisms. Harvard University Press, Cambridge, Mass.
2. Dye, C., S. Scheele, P. Dolin,V. Pathania, and M. Raviglione. 1999. Global burden of
tuberculosis. JAMA 282:677–686.
3. Lurie, M. B., and A. M. Dannenberg, Jr. 1965. Macrophage function in infectious dis-
ease with inbred rabbits. Bacteriol. Rev. 29:466–476.
4. Dannenberg, A. M., Jr. 1994. Rabbit model of tuberculosis, p. 149–156. In B. R. Bloom
(ed.), Tuberculosis: Pathogenesis, Protection, and Control.ASM Press,Washington, D.C.
5. Dannenberg, A. M., Jr., and E. M. Collins. 2001. Progressive pulmonary tuberculosis is
not due to increasing numbers of viable bacilli in rabbits, mice and guinea pigs, but is due
to a continuous host response to mycobacterial products. Tuberculosis 81:229–242.
6. Janeway, C. A. 2002. A trip through my life with an immunological theme. Annu. Rev.
Immunol. 20:1–28.
7. Rich, A. R. 1951. The Pathogenesis of Tuberculosis, 2nd ed. Charles C Thomas, Springfield, Ill.
8. Canetti, G. 1955. The Tubercle Bacillus in the Pulmonary Lesion of Man. Springer Publishing
Company, Inc., New York, N.Y.
9. Iseman, M. D. 2000. A Clinician’s Guide to Tuberculosis. Lippincott Williams and Wilkins,
Philadelphia, Pa.
10. Bloom, B. R. (ed.). 1994. Tuberculosis: Pathogenesis, Protection, and Control. ASM Press,
Washington, D.C.
11. Reichman, L. B., and E. S. Hershfield. 2000. Tuberculosis: a Comprehensive International
Approach, 2nd ed. Dekker, New York, N.Y.
12. Manabe,Y. C., and A. M. Dannenberg, Jr. 2006. Pathophysiology: basic aspects, p. 18-51.
In D. Schlossberg (ed.), Tuberculosis and Nontuberculous Mycobacterial Infections, 5th ed.
McGraw-Hill, New York, N.Y.
13. Cole, S.T., K. D. Eisenach, D. N. McMurray, and W. R. Jacobs, Jr. (ed.). 2005. Tuber-
culosis and the Tubercle Bacillus.ASM Press,Washington, D.C.
14. Rom,W. N., and S. M. Garay. 2004. Tuberculosis, 2nd ed. Lippincott Williams & Wilkins,
Philadelphia, Pa.
INTRODUCTION

Contributions of Max B. Lurie 1


Contributions of Arthur M. Dannenberg, Jr. 2

In the early 1900s, experimental tuberculosis was surrounding tissues) after the host becomes sen-
produced in rabbits and guinea pigs.About the sitive to their tuberculin-like products. (iv)
middle of the 20th century, René Dubos at the Caseous necrosis (and all of the tissue destruc-
Rockefeller Institute (now University) intro- tion found in tuberculosis) is due to host
duced the mouse model of this disease. Because delayed-type hypersensitivity to products of the
of the cost and size of mice and the availability bacilli, especially tuberculin-like products. (v)
of specific reagents, the mouse model has been The extracellular growth of tubercle bacilli in
used ever since by most of the scientists in the pulmonary cavities can overwhelm good native
field. However, Max B. Lurie (until his death in and acquired host resistance by the bronchial
1966) and I continued to use rabbits to study this spread of large numbers of bacilli. Although
disease. caseation, liquefaction, and cavity formation
All stages of human pulmonary tuberculosis have been well recognized ever since tubercu-
(including liquefaction and cavity formation) losis was described as a disease, the roles of these
can be readily produced in rabbits. Guinea pigs processes in its pathogenesis were significantly
develop the susceptible childhood form of the clarified by Lurie’s studies in the rabbit model.
disease that only rarely forms cavities, and mouse 2. Development of inbred rabbit strains
granulomas show little or no caseous necrosis that produced the childhood and adult
and never develop cavities. forms of this disease (chapter 13). Lurie’s
The rabbit model of tuberculosis has been susceptible rabbits developed the childhood
occasionally used by other laboratories (1-12), but form of tuberculosis with hematogenous spread
the majority of the studies with this model have of the disease and did not produce cavities. His
been made in Lurie’s and my laboratories. Our resistant rabbits developed the adult form with
principal contributions are summarized below. cavities and bronchial spread of the disease.
3. Identification of the characteristics
CONTRIBUTIONS OF MAX B. LURIE of native (genetic) and acquired (adap-
Most of Lurie’s experiments are summarized in tive) resistance that affect the pathogenesis
his book Resistance to Tuberculosis: Experimental of tuberculosis (chapter 14). When com-
Studies in Native and Acquired Defensive Mecha- pared to his inbred susceptible rabbits, Lurie’s re-
nisms (13). His most significant contributions sistant rabbits showed (i) greater maturation of
include the following. epithelioid cells (now known as “activated
1. Insights into the pathogenesis of macrophages”) that are capable of destroying
tuberculosis (chapter 2). (i) Intracellular or inhibiting tubercle bacilli, (ii) greater destruc-
tubercle bacilli are nontoxic and initially grow tion of bacilli in the primary and secondary
well within macrophages without injuring these lesions, (iii) greater interstitial inflammation
cells. (ii) A mature epithelioid cell is a (containing macrophages and lymphocytes), (iv)
macrophage that has developed the power to kill increased lymphatic drainage from pulmonary
or inhibit the tubercle bacilli that it ingests. (iii) lesions to the hilar lymph nodes (resulting from
Tubercle bacilli only kill macrophages (and the the increased interstitial inflammation), and

1
2 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

(v) liquefaction with cavity formation and Other contributions. Lurie’s other con-
bronchial spread. tributions are described in the list of his publi-
4. Demonstration of similarities in the cations presented in Appendix C.
host reaction to virulent bovine-type
bacilli, virulent human-type bacilli, and CONTRIBUTIONS OF ARTHUR M.
BCG (chapter 13). Although the host DANNENBERG, JR.
responds to all tubercle bacilli in a similar fash- The major contributions of my laboratory group
ion, its ability to arrest the disease varies inversely to experimental tuberculosis include the fol-
with the virulence of the bacilli. Both live and lowing.
dead virulent bovine-type tubercle bacilli are 1. Deeper insight into macrophage acti-
harder for rabbits to destroy and eliminate than vation and heterogeneity (chapter 6).
are virulent human-type bacilli. 2. Studies on the local nature of ac-
5. Demonstration of the efficacy of UV quired resistance to tuberculosis (chapter
light in preventing tuberculosis. 5). An expanded, recirculating, antigen-specific
6. The pulmonary tubercle-count lymphocyte population is systemic. However,
method (chapter 11). Lurie developed this these lymphocytes produce delayed-type hyper-
method to measure bacillary virulence, host re- sensitivity (DTH) and acquired cellular resistance
sistance to tuberculosis, and the efficacy of BCG reactions only at sites where tubercle bacilli
immunization. and/or their antigens are located. The local
7. Studies on the effects of hormones on nature of tuberculous lesions explains the many
the pathogenesis of tuberculosis. Lurie variations found in the clinical disease.
demonstrated that estrogens and chorionic 3. Clarification of the interplay of
gonadotropin in did not appreciably affect host delayed-type hypersensitivity (DTH) and
resistance to tuberculosis (chapter 17). How- cell-mediated immunity (CMI) (chapter
ever, glucocorticoids and adrenocorticotropic 5). DTH kills macrophages containing many
hormone (ACTH) in pharmacological doses tubercle bacilli, because these bacilli produce
decreased the host resistance, and ACTH in high local concentrations of tuberculin-like
physiological doses sometimes increased such products. Tubercle bacilli do not grow in the
resistance (chapter 16).Thyroid hormones some- resulting solid caseous tissue. CMI activates local
times increased host resistance, but thyroidectomy macrophages because of cytokines from antigen-
(or propylthiouracil administration) decreased it specific lymphocytes, so that the macrophages
(chapter 17). can inhibit the intracellular growth of ingested
8. Correlation of bacillary growth in tubercle bacilli. Both DTH and CMI are needed
the host with the histopathology of tuber- to arrest tuberculosis. In the absence of either
culous lesions (chapter 2). Lurie identified DTH or CMI, the host cannot stop the progress
(i) an initial bacillary inhibition, presumably by of the disease.These concepts explain all aspects
pulmonary alveolar macrophages; (ii) a loga- of Lurie’s bacillary growth curves in the host
rithmic (symbiotic) growth phase in mac- (outlined in item 8 in the section above).They
rophages entering the lesions from the blood; also explain why DTH developed throughout
(iii) a stationary growth phase after the host evolution: DTH is necessary to stop the intra-
becomes tuberculin-positive and during which cellular multiplication of tubercle bacilli when-
caseous necrosis is present in rabbits, in guinea ever CMI (i.e., acquired cellular resistance) can-
pigs, and undoubtedly in humans; (iv) extra- not stop it.
cellular bacillary growth associated with liq- 4. DTH is a major factor in controlling
uefaction and cavity formation; and (v) a longer the growth of tubercle bacilli (chapter 5).
symbiotic bacillary growth phase reaching Mice develop poor DTH and their tissues con-
higher titers (before the stationary phase) tain large numbers of tubercle bacilli (chapter
whenever the inhaled tubercle bacilli were 15). Guinea pigs develop poor DTH and their
more virulent. tissues contain smaller numbers of these bacilli.
INTRODUCTION 䡵 3

Yet mice are considered the more resistant 8. Causes of liquefaction and cavity for-
species, because in mice much less tissue destruc- mation (chapter 4). Liquefaction of the
tion occurs than in guinea pigs.The DTH:CMI lesion’s solid caseous center perpetuates tuber-
ratio might be altered for the benefit of the culosis in humans because it enables extracellu-
host by an appropriate vaccine (chapter 22). lar growth of tubercle bacilli and cavity forma-
5.The beneficial role of antibodies (chap- tion. We showed that proteases, DNases, and
ter 5). Hosts that have been immunized to tuber- RNases (and probably other hydrolases) are
culosis usually have circulating antibodies as well involved. Yamamura and coworkers (6, 7, 9)
as an expanded population of antigen-specific showed that DTH to the tuberculin-like prod-
Th1 lymphocytes.After virulent tubercle bacilli ucts of tubercle bacilli is involved. Much more
are inhaled by such hosts, a local antigen-antibody research is needed to understand the cause of
reaction immediately produces the C5a compo- liquefaction and cavity formation and how to
nent of complement and probably other chemo- inhibit them (see chapter 25). (The rabbit is
taxins.This results in a rapid local accumulation the only common laboratory animal in which
of macrophages and antigen-specific T lympho- the human form of chronic fibrosing tubercu-
cytes at sites of bacillary lodgement. Therefore, lous cavities can be readily produced.)
even though circulating antibodies apparently
have little or no direct effect on tubercle bacilli,
they increase the rate at which existing DTH and REFERENCES
CMI control local bacillary growth. In this man- 1. Ratcliffe, H. L., and W. F.Wells. 1948.Tubercu-
ner, antibodies play an important role in host losis of rabbits induced by droplet nuclei infec-
tion. I. Initial response to infection. J. Exp. Med.
defense against endogenous and exogenous rein- 87:575–584.
fection with tubercle bacilli. 2. Ratcliffe, H. L., and W. F.Wells. 1948.Tubercu-
6. Bacillary latency and dormancy losis of rabbits induced by droplet nuclei infec-
(chapter 10). Macrophages in tuberculous tion. II. Response to reinfection. J. Exp. Med.
lesions have a high turnover rate. Therefore, 87:585–594.
3. Weiss, C., and M. L. Boyar-Manstein. 1951. On
nonactivated macrophages from the bloodstream the mechanism of liquefaction of tubercles. I.The
continually infiltrate tuberculous lesions. Even behavior of endocellular proteinases in tubercles
in seemingly arrested tuberculous lesions, some developing in the lungs of rabbits. Am. Rev.Tuberc.
of the nonactivated macrophages probably ingest 63:694–705.
occasional tubercle bacilli that escape from the 4. Weiss, C., and F. M. Singer. 1953. Mechanism of
softening of tubercles. II. Behavior of desoxyri-
caseous center. If so, such bacilli will start to mul- bonuclease in tubercles developing in the lungs of
tiply intracellularly until they are again inhibited rabbits. Arch. Pathol. 55:516–530.
by DTH and CMI as described above. Com- 5. Weiss, C., J. Tabachnick, and H. P. Cohen.
plete bacillary dormancy within arrested tuber- 1954. Mechanism of softening of tubercles. III.
culous lesions may be rare and may occur only Hydrolysis of protein and nucleic acid during
anaerobic autolysis of normal and tuberculous lung
extracellularly in solid caseous necrotic tissues. tissue in vitro. Arch. Pathol. 57:179–193.
7. Cytokine dynamics (chapters 19 and 6. Yamamura, Y., S. Yasaka, M. Yamaguchi, K.
20). Immunized hosts rapidly produce cyto- Endo, H. Iwakura, S. Nakamura, and Y.
kines, especially chemokines, which in turn Ogawa. 1954. Studies on the experimental tuber-
cause a rapid accumulation of macrophages and culous cavity. Med. J. Osaka Univ. 5:187–197.
7. Yamamura,Y. 1958.The pathogenesis of tuber-
lymphocytes at sites of bacillary lodgement. culous cavities. Adv.Tuberc. Res. 9:13–37.
After a sufficient number of these defense cells 8. Kruml, J., L.Trnka, R. Urbancík, and J. Kuska.
have accumulated locally, the production of 1965. Histologic differences between experimen-
chemokines is rapidly downregulated. What tal cavernous tuberculosis in rabbits and human
determines the amount of local cell infiltration cavitary disease. Am. Rev. Respir. Dis. 92:299–302.
9. Yamamura, Y., Y. Ogawa, H. Yamagata, and
is worthy of further investigation. Such infiltra- Y. Yamamura. 1968. Prevention of tuberculous
tion was most pronounced in reinfected rabbits, cavity formation by immunosuppressive drugs.
as well as in Lurie’s resistant rabbits. Am. Rev. Respir. Dis. 98:720–723.
4 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

10. Yamamura,Y.,Y. Ogawa, H.Yamagata, and Y. 12. Kambara,T.,T. Hiraoka, and I. Kukita. 1985.
Yamamura. 1974. Prevention of tuberculous cav- Neutral proteases in lymph node cells of rabbits
ity formation by desensitization with tuberculin- immunized with heat-killed tubercle bacilli.
active peptide. Am. Rev. Respir. Dis. 109:594–601. Kumamoto Med. J. 38:153–163.
11. Kitano, M., S. Kume, and T. Kambara. 1982. 13. Lurie, M. B. 1964. Resistance to Tuberculosis: Exper-
Evaluation of delayed hypersensitivity skin reactions imental Studies in Native and Acquired Defensive
in rabbits immunized with live BCG. Bull. Ginkyo Mechanisms. Harvard University Press, Cambridge,
Coll. Med.Technol. 6:1–10. Mass.
Section 1.

PATHOGENESIS OF TUBERCULOSIS

5
This page intentionally left blank
1
OVERVIEW

Types of human disease 9


Contagiousness: contracting tuberculosis by the respiratory route 11
Size of infectious particles 12
Virulence of bacillary strain 13
Factors influencing the establishment of a pulmonary tuberculous lesion
(recognized by the conversion of the tuberculin skin test) 15
Factors controlling resistance to tuberculosis 16
Prevention of clinical disease 17

Abstract. Tuberculosis is still one of the major diseases of the world, especially in devel-
oping countries. It kills over 2 million people each year, more than any other infectious
disease.
Childhood-type pulmonary tuberculosis is a disease of susceptible hosts, such as infants
and immunosuppressed individuals. From the primary parenchymal lesion, the bacilli fre-
quently spread via lymphatics and cause caseous lesions of the hilar lymph nodes.The bacilli
may also spread via the bloodstream and cause lesions elsewhere in the host.The primary
lesion, as well as the metastatic lesions, often progresses until the host succumbs.
Adult-type pulmonary tuberculosis is a disease of innately resistant hosts, a category that
includes most immunocompetent persons.The active parenchymal lesion (often subapi-
cal) frequently forms a cavity, in which the bacilli may multiply extracellularly. If so, the
bacilli may spread via the bronchial tree to other parts of the lung. In such a resistant host,
many metastatic microscopic lesions do not progress, but those that do may form new cav-
ities.Also, caseous bronchopneumonia may occur when an appreciable amount of lique-
fied caseum enters the bronchial tree. Cavity formation perpetuates tuberculosis in
humankind because coughing spreads bacilli from the lungs into the environment, where
they may infect other people.
Contracting clinical tuberculosis depends on (i) the size and physiological state of the
bacillary particle, (ii) its virulence, and (iii) the native and acquired resistance of the host.
How to protect personnel against tuberculosis is also discussed in this chapter. UV lights
(shielded to protect people’s eyes) or HEPA-filtered air purifiers should be used more fre-
quently in hospital areas where tubercle bacilli are likely to be present.

Tuberculosis remains one of the major infectious disease during the 2 years after infection,
diseases in the world.Tuberculin testing indicates and about 5% develop it later due to debili-
that about one-third of the world’s population tation in old age, immunosuppression, and other
is infected with Mycobacterium tuberculosis (about causes of decreased host resistance. About
1.9 billion people), mostly in developing coun- 30% of HIV-infected patients die of tuber-
tries (1).The disease kills more people than any culosis. In fact, HIV-infected patients have a
other infectious disease, and in recent years the yearly risk of developing clinical tuberculosis
combined infection of human immunodefi- equivalent to the lifetime risk of most non-
ciency virus (HIV) and M. tuberculosis has accel- HIV-infected patients. This chapter presents
erated the death rate. Ninety percent of those an overview of the pathogenesis of the
infected with virulent M. tuberculosis have an human disease. More details are presented in
inapparent disease. About 5% develop clinical chapter 3.

7
8 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

FIGURE 1 Rapidly progressing miliary tuberculosis in an 11-month-old infant.The


rabbit counterpart of this type of tuberculosis is shown in Fig. 3. Most of the multiple
caseous tubercles are of hematogenous origin.The primary lesion is marked by an arrow.
Note the large size of the homolateral caseous hilar lymph nodes (which seeded the
blood entering the lungs; see text). From the collection of the late professors A. R. Rich
and W. G. MacCallum, Department of Pathology, Johns Hopkins School of Medicine,
Baltimore, Md.
1. OVERVIEW 䡵 9

TYPES OF HUMAN DISEASE and cell-mediated immunity (CMI) several


Humans inhaling occasional virulent tubercle weeks after a primary microscopic tubercle is
bacilli over a period of months develop only one established. DTH and CMI keep other devel-
primary pulmonary tubercle for the following oping tubercles from reaching a grossly visible
reasons. (i) Relatively few bacilli are in the air size (see chapters 5 and 11).
that people breathe, even in households with
active cases of tuberculosis. Therefore, often Childhood-Type Tuberculosis
weeks may pass before another inhaled bacillus In childhood-type tuberculosis, the bacilli in
begins to multiply in the lung. (ii) Pulmonary the primary tubercle drain via lymphatics to
alveolar macrophages are a highly activated cell the hilar nodes, where they produce caseous
population, which in humans probably destroy lesions (Fig. 1, 2, and 3). Bacilli from both the
most inhaled tubercle bacilli before they mul- primary lung lesion and the hilar nodes then
tiply (see chapters 2 and 3). (iii) The host devel- enter the bloodstream and may cause miliary
ops both delayed-type hypersensitivity (DTH) lesions throughout the body.

FIGURE 2 Miliary tuberculosis of the lungs in a 19-year-old young adult.The tubercles appear
smaller than those in Fig. 1 merely because the lungs of this individual are so much larger than
those of the infant. Caseous hilar lymph nodes were the source of most of these miliary tuber-
cles. In this patient, caseous plaques (not shown) in branches of the pulmonary veins seeded tuber-
cle bacilli into the general circulation and caused miliary tubercles in many other organs. From
the collection of the late professors A. R. Rich and W. G. MacCallum, Department of Pathology,
Johns Hopkins School of Medicine, Baltimore, Md.
FIGURE 3 Organs of Lurie’s inbred susceptible rabbit F4-33, which died of a gen-
eralized progressive tuberculosis of 3.3 months’ duration.This rabbit had a large, sin-
gle, completely caseous nonliquefied primary lesion in the middle of the left lung,
massive enlargement and caseation of the homolateral hilar lymph nodes, and
hematogenous spread of the disease to the lungs, kidney, pleura, and knee joint.The
rabbit was naturally infected in a room containing airborne tubercle bacilli from rab-
bits with tuberculous kidneys (see chapter 12). Reproduced with permission from
reference 3.
Note that the disease in this susceptible inbred rabbit was similar to the disease
in humans shown in Fig. 1 and 2.The tubercles shown in these three figures could
be almost the same size, because the photographs of the human specimens are so much
smaller than the actual specimens.

10
1. OVERVIEW 䡵 11

The secondary lesions in the lung usually native and acquired resistance of the host, bacil-
originate from bacilli in the hilar lymph nodes, lary growth is usually inhibited.The hilar nodes
because the efferent lymph from these nodes remain small with relatively little caseation, and
enters the venous blood. Bacilli in the venous few bacilli enter the efferent lymph draining into
blood enter the right side of the heart and then the great veins to return to the lungs. If they do
the lungs via the pulmonary arteries. so, they usually do not cause progressive sec-
The secondary lesions in the rest of the body ondary pulmonary lesions, because of the strong
usually originate from bacilli in primary (and immunity developed by the host during the
secondary) lung lesions. Such bacilli enter the primary infection.
pulmonary veins and then the left side of the In such immunocompetent hosts, the disease
heart to be distributed to sites throughout the may progress if the solid caseous center lique-
body via the aorta.These bacilli only go back to fies and forms a cavity (Fig. 4, 5, and 6). In the
the lungs if they pass through peripheral capil- liquefied caseum, the bacilli may grow extra-
lary beds and enter the venous blood. cellularly (for the first time during the course of
In summary, in childhood-type tuberculosis this disease) and (after the cavity forms) may
most of the secondary tubercles found in the reach such large numbers (Fig. 7) that even the
lungs usually come from caseous lesions in the high native and acquired resistance of the host
hilar nodes, and most of the secondary tubercles is overwhelmed.
in the other organs usually come from caseous Bacilli from the cavities are often
lesions in the lungs themselves. coughed into the environment where they
Infants and immunosuppressed individuals, may infect other people.Therefore, lique-
including those with HIV/AIDS infection, often faction with cavity formation is a major
develop hematogenously spread childhood-type reason why tuberculosis is perpetuated in
disease.The Ghon complex is the typical form humankind (2).
of this type of tuberculosis. It is characterized by Lurie developed inbred rabbit families that
a caseous lesion in the lung parenchyma (usu- mimicked these two types of human disease
ally not in an apical location) with enlarged (3–5). His susceptible rabbits developed the child-
caseous hilar lymph nodes. hood type of rapidly progressing disease with
hematogenous spread (Fig. 3). His resistant rab-
Adult-Type Tuberculosis bits developed the adult type of slowly progress-
Immunocompetent adults often develop adult- ing disease with cavitary formation and bronchial
type tuberculosis, whether or not they had the spread (Fig. 6).The principles established exper-
disease in childhood or even converted their imentally with these inbred rabbits have direct
tuberculin skin test from an inapparent infection. application to the human disease, which is why
Adults who develop this type of tuberculosis as I have continued to use the rabbit model of
a primary infection have rather good native re- tuberculosis throughout my research career.
sistance and can develop rather strong acquired Chapter 3 describes and illustrates the types of
resistance after infection. human tuberculosis in more detail.
Adult-type tuberculosis is characterized by
minimal involvement of the hilar lymph nodes CONTAGIOUSNESS: CONTRACTING
and few, if any, progressive secondary lesions of TUBERCULOSIS BY THE
hematogenous origin.The disease usually begins RESPIRATORY ROUTE
as a single subapical lesion that often cavitates. Several factors influence the infectiousness of
From the cavity, the bacilli spread into the air- patients with tuberculosis: (i) the frequency
ways and then into other parts of the lungs and and force of coughing and whether the patient
to the outside environment. coughs into the environment or into cupped
In this type of tuberculosis, tubercle bacilli hands, (ii) the number of viable bacilli in the
reach the hilar nodes, but because of the high patient’s sputum, (iii) the consistency of the
12 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

FIGURE 4 Bilateral tuberculous cavities in the upper lobes of a 39-year-old diabetic woman.
Below each cavity are areas of caseous lung tissue.Although infected, the hilar lymph nodes are
not markedly enlarged. In the lung on the right, an applicator stick marks the communication
between the cavity and the bronchus.The rabbit counterpart of this type of tuberculosis is shown
in Fig. 6. From the collection of the late professors A. R. Rich and W. G. MacCallum, Depart-
ment of Pathology, Johns Hopkins School of Medicine, Baltimore, Md.

sputum (it is more easily aerosolized when it is discussion of factors influencing the transmis-
more fluid), (iv) the amount of albumin-like sion of tuberculosis.
protein present (which aids bacillary survival in
air), (v) the size of the aerosolized particles SIZE OF INFECTIOUS PARTICLES
(see below), and (vi) the virulence of the bacilli When inhaled, only fine particles containing 1
(see below). When patients are receiving to 3 tubercle bacilli are capable of initiating the
antimicrobials, these drugs get concentrated infection, because they remain suspended in the
as the fluids in the aerosolized droplets evap- airstream that enters the alveolar spaces (4; see
orate. Drug-resistant bacilli survive such further discussion in references 13 and 14).The
increased concentrations better than do drug- heavier bacillary particles (containing more than
susceptible bacilli. Moisture in the air protects 3 bacilli, often with bits of caseous material)
tubercle bacilli from drying out, as do their impinge upon the mucosal surfaces of both the
wax-like coats. Finally, UV light, including that nasopharynx and the bronchial tree.These bacil-
from direct sunlight, is lethal to tubercle bacilli lary particles are moved up the bronchial tree by
(6–11). See reference 12 for a more complete cilia and are eventually swallowed.The mucosal
1. OVERVIEW 䡵 13

FIGURE 5 An apical cavity of moderate size in an adult patient. Below the cavity is an area of
caseous consolidation of pneumonic origin. Several caseous foci are also present in the other lung,
probably also pneumonic in origin.The hilar lymph nodes (not shown) contained a few caseous
foci but were only slightly enlarged. From the collection of the late professors A. R. Rich and
W. G. MacCallum, Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Md.

surfaces of the respiratory and gastrointestinal In brief, tubercle bacilli may vary in virulence,
tracts are rather resistant to tuberculosis and both genetically and phenotypically. Genetically,
develop lesions only when exposed to large Mycobacterium bovis BCG (an attenuated bovine-
numbers of tubercle bacilli. type bacillus),virulent human-type tubercle bacilli
(e.g., H37Rv), and virulent bovine-type tuber-
VIRULENCE OF BACILLARY STRAIN cle bacilli (e.g., Ravenel) are increasingly patho-
The virulence of the bacillus causing tuberculo- genic for rabbits (4, 5). Phenotypically, a viable
sis depends on the host’s ability to prevent and bacillus that has dried out or been exposed to
control the disease that the bacillus produces (see direct sunlight would often be too weak to start
reference 14). Bacillary virulence and the innate an infection. However, a viable bacillus kept moist
and acquired (adaptive) immunity of the host in a dark place would be quite infectious if
affect each other in opposite directions. This aerosolized by air currents.
book is about the host’s response to the bacillus, The pathogenicity of a specific strain of tuber-
but I discuss below some of the attributes of vir- cle bacillus is due to both its genotype and the
ulence because of their importance to the devel- response of the host.The most common measures
opment of new antimicrobials. Such attributes of virulence are (i) the number of bacilli in a par-
have recently been discussed in detail (15–19).For ticular organ, (ii) the amount of pathology pro-
a review of the older literature, see reference 20. duced, and (iii) the time of death of the host.
14 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

FIGURE 6 Organs of Lurie’s


inbred resistant rabbit A2-6, which
died of tuberculosis 9 months after its
tuberculin skin test converted.A sin-
gle primary encapsulated pulmonary
cavity was present in the right lung,
from which tubercle bacilli spread
(via the bronchial tree) to the larynx
and the intestines. No grossly visible
tuberculous lesions were found in
the hilar lymph nodes or in the kid-
neys. Nonprogressive secondary
tubercles were present in both lungs
(probably bronchogenic in origin).
Like the susceptible rabbit depicted
in Fig. 3, this resistant rabbit was nat-
urally infected in a room containing
airborne tubercle bacilli from rab-
bits with tuberculous kidneys (see
chapter 12). Reproduced with per-
mission from reference 3.
Note that the disease in this
inbred resistant rabbit was similar to
the cavitary disease in humans shown
in Fig. 4 and 5.

The virulence of H37Rv can sometimes be onymous and nonsynonymous base substitutions
reduced by repeated subculturing (2), which (25, 26). However, some deletions and insertions
demonstrates that its genotype is not completely have caused changes in the virulence of various
stable. The virulence of clinical isolates of M. members of the M. tuberculosis complex (19, 27).
tuberculosis has also been found to be variable: For example, despite the 99% genomic identity
several strains isolated from humans with active with virulent mycobacterial strains, the absence
tuberculosis in India, Southeast Asia, and Hong of certain genes in BCG and Mycobacterium microti
Kong were reduced in virulence for guinea pigs caused them to be attenuated for humans (19, 28,
(21–23). 29).The attenuation of BCG was produced by
The entire genome of M.tuberculosis (which has a serial passage of a virulent M. bovis in media
now been sequenced [24]) has changed relatively containing ox bile (30, 31; M.A. Behr and P. M.
little over the years: it has had a low rate of syn- Small, Letter, Nature 389:133–134, 1997).
1. OVERVIEW 䡵 15

FIGURE 7 Tubercle bacilli growing profusely in the liquefied caseum in a pulmonary cav-
ity of a rabbit that inhaled about 340 virulent bovine-type tubercle bacilli 33 weeks previously.
Such profuse growth occurs only in some lesions with liquefied centers, presumably where the
composition of the liquefied caseum is most favorable or the adaptation of the bacillus to extra-
cellular growth is most complete. Similar bacillary growth has been found in many human cav-
itary lesions (20). Bacilli were stained with carbol-fuchsin and counterstained with methylene
blue. Magnification, ⫻600. Photograph reproduced with permission from reference 59.

The virulence of various mycobacterial strains other adverse environments (including solid
for rabbits and mice may differ, because these caseum), which is facilitated by sigma factors (16,
strains probably contain or produce slightly dif- 40) and the synthesis of lipids (18).There are 13
ferent antigens (32–34). Virulent human-type sigma factors among the 190 transcriptional reg-
tubercle bacilli (e.g., H37Rv) are virulent in ulators that have been described in the M. tuber-
mice and humans but are partly attenuated in culosis genome (40). These factors are carefully
rabbits. Recently, the HN878 strain of human- controlled by anti-sigma factors, as well as anti-
type tubercle bacilli (which caused a miniepi- anti-sigma factors (40). See chapters 6 and 25 for
demic in Houston, Tex.) was found to have additional comments on virulence and chapters
increased virulence for both rabbits and mice 6 and 10 for additional comments on dormancy.
(33).Among virulent human-type strains of M.
tuberculosis, the Erdman strain is more virulent FACTORS INFLUENCING THE
than the H37Rv strain, which is in turn more ESTABLISHMENT OF A PULMONARY
virulent than the CDC1551 strain (34; Y. C. TUBERCULOUS LESION
Manabe et al., submitted for publication). (RECOGNIZED BY THE CONVERSION
OF THE TUBERCULIN SKIN TEST)
Several attributes of virulence have recently
been extensively investigated: (i) the entry of Number of Tubercle Bacilli Inhaled
tubercle bacilli into monocytes/macrophages (35, The inhaled population of virulent tubercle
36), (ii) their survival in these cells (37–39) (see bacilli is not uniform. It contains bacilli that
chapter 6), and (iii) their survival in a variety of are too weak to start any infection, as well as
16 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

robust bacilli that are always ready to do so. If


many bacilli are inhaled, a robust bacillus is
likely to be among them.

Microbicidal Power of the Pulmonary


Alveolar Macrophages
Alveolar macrophages vary in their capacity to
destroy the bacillus (4, 5). Some alveolar
macrophages are rich in enzymes (41) and
microbicidins, while others are poor in both.The
ratio of “rich” to “poor” alveolar macrophages
seems to be determined by the native genetic re-
FIGURE 8 An example of an effective disposable
sistance of each individual as well as by pheno- mask.This mask is made of HEPA-filtering material with
typic factors. an exhaust valve in the center. It is backed by a ring of
foam rubber that fits tightly against one’s face (but not
Establishment of the Infection quite as tightly as the less flexible mask shown in Fig.
9). Similar masks are available from Lab Safety Supply,
In humans and rabbits,pulmonary infection (with 401 S.Wright Road, Janesville,Wis. 53546.Tel: 1-800-
virulent human-type tubercle bacilli) begins only 356-0783. http://www.labsafety.com.
after a strongly endowed bacillus is ingested by a
weakly endowed alveolar macrophage. No one
knows exactly how many fine particles contain- On the host side, infants are rather suscepti-
ing human-type tubercle bacilli must be inhaled ble to tuberculosis, apparently because their
by humans before such a combination is reached. immune system is still developing. Susceptibil-
The mean number is probably between 5 and ity is also greater during adolescence in some
200, whereas the mean number for commercial human populations, but not in others. Persons
New Zealand White rabbits is between 500 and debilitated by old age and other conditions are
1,000 (see chapter 11). also more susceptible.
One report (42) indicated that the alveolar Both native and acquired resistance to tuber-
macrophages of people of African descent (i.e., culosis is lowered by immunosuppressive drugs,
blacks) were less effective in destroying inhaled glucocorticoids, and immunosuppressive viruses
tubercle bacilli than the alveolar macrophages of (e.g., HIV); silicosis, uremia, diabetes, alcoholism,
people of European descent (i.e., whites). Blacks
converted to tuberculin positivity more frequently
than whites after similar exposures to tubercle
bacilli in a nursing home.Among the converters,
however, the incidence of clinical disease was
the same (42), indicating that both groups pro-
duced a satisfactory immune response. See chap-
ter 12 for a more complete discussion of the
establishment and progress of tuberculosis.

FACTORS CONTROLLING
RESISTANCE TO TUBERCULOSIS
Some strains of tubercle bacilli may be inher-
ently more virulent than others. Such genetic
(and/or phenotypic) differences in bacillary FIGURE 9 3M 7200S Half-Facepiece Respirator
with 7255 Easi-Air High Efficiency Filter (approved by
virulence would affect the course and type of the National Institute of Occupational Safety and Health
tuberculosis that these strains produce (see ref- [NIOSH]). Similar masks are available from Lab Safety
erence 43). Supply, Janesville,Wis. (See legend to Fig. 8.)
1. OVERVIEW 䡵 17

and certain neoplastic diseases; gastrectomy; par- fibrotic encapsulation of larger foci of infec-
asitic infection (e.g., with hookworm); starvation tion. However, even after effective chemother-
and other nutritional deficiencies; and physical apy, residual tuberculous foci may reactivate if the
misery, e.g., in the concentration camps of World resistance of the person becomes depressed.
War II (44) (see chapters 3 and 16). Two or Antimicrobial therapy is discussed further in
more of these conditions are often synergistic. chapter 25.
The anti-inflammatory action of glucocorti- Immunotherapy with killed Mycobacterium
coids may mask the clinical symptoms of reac- vaccae in combination with chemotherapy shows
tivated tuberculosis. promise (47). (M. vaccae is a nonvirulent acid-fast
However, most adult human beings are rather bacillus isolated from the soil.) Immunotherapy
resistant to tuberculosis, some more so than may also prove useful in the treatment of
others (4, 45). In addition, children in develop- multidrug-resistant tuberculosis.
ing countries often develop some acquired
immunity from the environmental tubercle PREVENTION OF CLINICAL DISEASE
bacilli that they ingest. People contract pulmonary tuberculosis mainly
By killing (or inhibiting) tubercle bacilli, by inhaling small airborne particles of 1 to 3
effective antimicrobial agents, such as isoniazid, bacilli. Such particles are most often produced
rifampin, pyrazinamide, ethambutol, and strep- when a patient coughs or sneezes. Rooms where
tomycin (46), greatly aid the macrophages of the tuberculous animals are housed may contain
host in (i) arresting the progress of the disease, infectious particles if the animals cough, sneeze,
(ii) preventing its complications, (iii) healing or shed the bacilli in their feces or urine.Vacuum
small foci of infection, and (iv) promoting cleaners and air conditioners do not filter small

FIGURE 10 3M Air-Mate High Efficiency Particle (HEPA) Powered Air Purifying Respira-
tor (PAPR)—HEPA 10 Head Cover and HEPA 12 Hood—with the HEPA PAPR assembly
shown in Fig. 11 (NIOSH approved).Available from Lab Safety Supply, Janesville,Wis. (See leg-
end to Fig. 8.) Photograph is reproduced with permission from the 3M Occupational Health and
Environmental Safety Division.
18 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

FIGURE 11 Components of the 3M Air-Mate HEPA 10 Head Cover and Air-Mate HEPA
12 Hood with the HEPA PAPR assembly shown in Fig. 10 (NIOSH approved).Available from
Lab Safety Supply, Janesville,Wis. (See legend to Fig. 8.) Photograph is reproduced with permission
from the 3M Occupational Health and Environmental Safety Division.

infectious particles of 1 to 3 tubercle bacilli out Necropsies on tuberculous patients or animals


of the air.To our knowledge, only a few exist- should, if possible, be performed under laminar
ing vacuum cleaners and none of the common flow hoods, and electric bone cutters (and other
air conditioners contain HEPA filters in their devices that readily aerosolize bacilli) should
exhaust systems. Machines without HEPA filters not be used in open spaces. UV lights shielded
only filter out large noninfectious airborne par- to irradiate only the upper air of rooms (away
ticles of tubercle bacilli and may actually dis- from the eyes of personnel) are the most prac-
seminate smaller infectious particles. Large bacil- tical and inexpensive way to reduce the num-
lary particles are not infectious, because during ber of viable bacilli in local areas (8–12, 48). UV
respiration they do not reach the pulmonary lights can be used over doorways if they are
alveoli where tuberculous lesions begin, but shielded to produce a narrow beam of light
impinge on the mucosal surfaces of the airways directed only to the floor and to the ceiling.
(which are quite resistant to infection by tuber- Taking the following measures would reduce
cle bacilli).The efficacy of air ionizers in remov- the incidence of new cases of clinical tubercu-
ing infectious particles remains to be tested. losis (49–55; J. A. Schaefer, unpublished data).
1. OVERVIEW 䡵 19

(i) Tuberculous patients should cough and and treatment, room-air purification (UV irra-
sneeze into tissues or cupped hands. (ii) Attend- diation and HEPA filtration), and disinfection of
ing personnel should wear tight-fitting effective the patient’s room and utensils. The table of
masks (Fig. 8 and 9) or HEPA-filtered forced- contents of this extensive prevention guide (55)
air head covers or hoods (Fig. 10 and 11) is reproduced in appendix E.
(Schaefer, unpublished). Putting a mask on the
patient may be of some help but is not as effec- REFERENCES
tive as putting one on the attending personnel, 1. Dye, C., S. Scheele, P. Dolin,V. Pathania, and
because the patient’s coughing forces fine par- M. C. Raviglione. 1999. Consensus statement.
ticles of bacilli through the interstices of the Global burden of tuberculosis: estimated incidence,
mask. (iii) If they are likely to be exposed to prevalence, and mortality by country. JAMA 282:
tubercle bacilli, tuberculin-negative persons 677–686.
2. Grosset, J. 2003. Mycobacterium tuberculosis in the
should be vaccinated with an effective BCG extracellular compartment: an underestimated
strain, or if developed in the future, a more adversary. Antimicrob.Agents Chemother. 47:833–836.
effective vaccine. Finally, (iv) the airborne bacilli 3. Lurie, M. B. 1941. Heredity, constitution and
in patients’ rooms should be reduced by an tuberculosis, an experimental study. Am. Rev.Tuberc.
exhaust fan, by a portable HEPA-filtered air 44(Suppl. 3):1–125.
4. Lurie, M. B. 1964. Resistance to Tuberculosis: Exper-
purifier (56), or by UV lights (6, 8–12, 48) imental Studies in Native and Acquired Defensive Mech-
aimed at the ceiling and shielded to protect anisms. Harvard University Press, Cambridge, Mass.
the eyes of personnel. 5. Lurie, M. B., and A. M. Dannenberg, Jr. 1965.
Both portable HEPA-filtered air purifiers Macrophage function in infectious disease with
and shielded UV lights are effective in remov- inbred rabbits. Bacteriol. Rev. 29:466–476.
6. Lurie, M. B. 1944. Experimental epidemiology of
ing viable tubercle bacilli from air in a room. tuberculosis: the prevention of natural air-borne
HEPA-filtered air purifiers are more expensive, contagion of tuberculosis in rabbits by ultraviolet
but the filters usually do not need to be irradiation. J. Exp. Med. 79:559–572.
changed for years, depending on the accumu- 7. Riley, R. L., W. F. Wells, C. C. Mills, W. Nyka,
lation of dust in them. Shielded UV lights are and R. L. McLean. 1957. Air hygiene in tuber-
culosis: quantitative studies of infectivity and con-
less expensive, but every few months (when trol in a pilot ward. Am. Rev. Respir. Dis. 75:420–431.
cool), they need to be washed with water con- 8. Riley, R. L., C. C. Mills, F. O’Grady, L. U. Sul-
taining a detergent or wiped with alcohol to tan, F. Wittstadt, and D. N. Shivpuri. 1962.
remove oily films that accumulate on their Infectiousness of air from a tuberculosis ward. Ultra-
surface and block the transmission of the ger- violet irradiation of infected air: comparative infec-
tiousness of different patients. Am. Rev. Respir. Dis.
micidal UV wavelengths (57, 58). Nardell rec- 85:511–525.
ommends replacing the UV lamp yearly (E.A. 9. O’Grady, F., and R. L. Riley. 1963. Experi-
Nardell, unpublished data). mental airborne tuberculosis. Adv. Tuberc. Res.
These prophylactic measures are especially 12:150–190.
important for persons who may be exposed to 10. Riley, R. L., and E. A. Nardell. 1989. Clearing
the air: the theory and application of ultraviolet air
antimicrobial-resistant tubercle bacilli. Cough- disinfection. Am. Rev. Respir. Dis. 139:1286–1294.
inducing procedures (such as endotracheal suc- 11. Riley, R. L., M. Knight, and G. Middlebrook.
tioning and bronchoscopy) aerosolize numerous 1976. Ultraviolet susceptibility of BCG and viru-
tubercle bacilli.The advantages and disadvantages lent tubercle bacilli. Am. Rev. Respir. Dis. 113:413–
of the various types of protective respirators are 418.
12. Nardell, E. A. 2004. Catching droplet nuclei:
reviewed by Schaefer (unpublished). toward a better understanding of tuberculosis trans-
Complete guidelines for preventing the trans- mission. Am. J. Respir. Crit. Care Med. 169:553–554.
mission of tuberculosis in health-care settings 13. Dannenberg, A. M., Jr. 1989. Immune mecha-
were recently published by the Centers for Dis- nisms in the pathogenesis of pulmonary tubercu-
ease Control and Prevention, Atlanta, Ga. (55). losis. Rev. Infect. Dis. 11(Suppl. 2):S369–S378.
14. Manabe, Y. C., and A. M. Dannenberg, Jr.
They cover every aspect of tuberculosis control, 2006. Pathophysiology: basic aspects. Pathogenesis
including early screening of patients with pos- of tuberculosis. II. Immunology of tuberculosis,
sible tuberculosis, masks and respirators, diagnosis p. 18–51. In D. Schlossberg (ed.), Tuberculosis and
20 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

Nontuberculous Mycobacterial Infections, 5th ed. 25. Sreevatsan, S., X. Pan, K. E. Stockbauer, N. D.
McGraw-Hill Book Co., New York, N.Y. Connell, B. N. Kreiswirth,T. S.Whittam, and
15. Cole, S. T., K. D. Eisenach, D. N. McMurray, J. M. Musser. 1997. Restricted structural gene
and W. R. Jacobs, Jr. (ed.). 2005. Tuberculosis and polymorphism in the Mycobacterium tuberculosis com-
the Tubercle Bacillus.ASM Press,Washington, D.C. plex indicates evolutionarily recent global dissem-
16. Smith, I. 2003. Mycobacterium tuberculosis patho- ination. Proc. Natl.Acad. Sci. USA 94:9869–9874.
genesis and molecular determinants of virulence. 26. Fleischmann, R. D., D. Alland, J. A. Eisen, L.
Annu. Rev. Microbiol. 16:463–496. Carpenter, O.White, J. Peterson, R. DeBoy, R.
17. Gomez, J. E., and J. D. McKinney. 2004. M. Dodson, M. Gwinn, D. Haft, E. Hickey, J. F.
tuberculosis persistence, latency, and drug tolerance. Kolonay, W. C. Nelson, L. A. Umayam, M.
Tuberculosis 84:29–44. Ermolaeva, S. L. Salzberg, A. Delcher, T.
18. Daniel, J., C. Deb,V. S. Dubey,T. D. Sirakova, Utterback, J. Weidman, H. Khouri, J. Gill, A.
B. Abomoelak, H. R. Morbidoni, and P. E. Mikula, W. Bishai, W. R. Jacobs, Jr., J. C.Ven-
Kolattukudy. 2004. Induction of a novel class of ter, and C. M. Fraser. 2002. Whole-genome
diacylglycerol acyltransferase and triglycerol accu- comparison of Mycobacterium tuberculosis clinical and
mulation in Mycobacterium tuberculosis as it goes into laboratory strains. J. Bacteriol. 184:5479–5490.
a dormancy-like state in culture. J. Bacteriol. 186: 27. Brosch, R., S. V. Gordon, M. Marmiesse, P.
5017–5030. Brodin, C. Buchrieser, K. Eiglmeier, T. Gar-
19. Brodin, P., C. Demangel, and S.T. Cole. 2005. nier, C. Gutierrez, G. Hewinson, K. Kremer,
Introduction to functional genomics of the Mycobac- M. J. Parsons, A. S. Pym, S. Samper, D. van
terium tuberculosis complex. In S.T. Cole, K. D. Eise- Soolingen, and S. T. Cole. 2002. A new evolu-
nach, D. N. McMurray, and W. R. Jacobs, Jr. (ed.), tionary scenario for the Mycobacterium tuberculosis
Tuberculosis and the Tubercle Bacillus.ASM Press,Wash- complex. Proc. Natl.Acad. Sci. USA 99:3684–3689.
ington, D.C. 28. Brodin, P., K. Eiglmeier, M. Marmiesse, A.
20. Long, E. R. 1958. The Chemistry and Chemother- Billault, T. Garnier, S. Niemann, S. T. Cole,
apy of Tuberculosis, 3rd ed.The Williams & Wilkins and R. Brosch. 2002. Bacterial artificial chro-
Co., Baltimore, Md. mosome-based comparative genomic analysis
21. Dickinson, J. M, M. J. Lefford, J. Lloyd, and identifies Mycobacterium microti as a natural ESAT-
D.A. Mitchison. 1963.The virulence in the guinea- 6 deletion mutant. Infect. Immun. 70:5568–5578.
pig of tubercle bacilli from patients with pulmonary 29. Manabe, Y. C., C. P. Scott, and W. R. Bishai.
tuberculosis in Hong Kong. Tubercle 44:446–451. 2002. Naturally attenuated, orally administered
22. Comstock, G. W., and R. J. O’Brien. 1991. Mycobacterium microti as a tuberculosis vaccine is
Tuberculosis, p. 745–771. In A. S. Evans and P. S. better than subcutaneous Mycobacterium bovis BCG.
Brachman (ed.), Bacterial Infections of Humans: Epi- Infect. Immun. 70:1566–1570.
demiology and Control, 2nd ed. Plenum Medical 30. Behr, M. A., and P. M. Small. 1999. A histori-
Book Co., New York, N.Y. cal and molecular phylogeny of BCG strains. Vaccine
23. Mitchison, D. A., A. L. Bhatia, S. Radha- 17:915–922.
krishna, J. B. Selkon,T.V. Subbaiah, and J. G. 31. Behr, M. A., M. A.Wilson,W. P. Gill, H. Sala-
Wallace. 1961.The virulence in the guinea-pig of mon, G. K. Schoolnik, S. Rane, and P. M.
tubercle bacilli isolated before treatment from South Small. 1999. Comparative genomics of BCG vac-
Indian patients with pulmonary tuberculosis. I. cines by whole-genome DNA microarray. Science
Homogeneity of the investigation and a critique of 284:1520–1523.
the virulence test. Bull.W. H. O. 25:285–312. 32. Manca, C., L. Tsenova, C. E. Barry III, A.
24. Cole, S. T., R. Brosch, J. Parkhill, T. Garnier, Bergtold, S. Freeman, P. A. Haslett, J. M.
C. Churcher, D. Harris, S. V. Gordon, Musser,V. H. Freedman, and G. Kaplan. 1999.
K. Eiglmeier, S. Gas, C. E. Barry III, F.Tekaia, Mycobacterium tuberculosis CDC1551 induces a more
K. Badcook, D. Basham, D. Brown, T. Chil- vigorous host response in vivo and in vitro, but is
lingworth, R. Connor, R. Davies, K. Devlin, not more virulent than other clinical isolates. J.
T. Feltwell, S. Gentles, N. Hamlin, S. Hol- Immunol. 162:6740–6746.
royd, T. Hornsby, K. Jagels, A. Krogh, 33. Manca, C., L. Tsenova, A. Bergtold, S. Free-
J. McLean, S. Moule, L. Murphy, K. Oliver, J. man, M. Tovey, J. M. Musser, C. E. Barry III,
Osborne, M. A. Quail, M. A. Rajandream, V. H. Freedman, and G. Kaplan. 2001.Virulence
J. Rogers, S. Rutter, K. Seeger, J. Skelton, R. of a Mycobacterium tuberculosis clinical isolate in mice
Squares, S. Squares, J. E. Sulston, K. Taylor, is determined by failure to induce Th1 type immu-
S. Whitehead, and B. G. Barrell. 1998. Deci- nity and is associated with induction of IFN-alpha/
phering the biology of Mycobacterium tuberculosis beta. Proc. Natl.Acad. Sci. USA 98:5752–5757.
from the complete genome sequence. Nature 34. Manabe, Y. C., A. M. Dannenberg, Jr., S. K.
393:537–544. Tyagi, C. L. Hatem, M.Yoder, S. C.Woolwine,
1. OVERVIEW 䡵 21

B. C. Zook, M. L. Pitt, and W. R. Bishai. 2003. upper-room germicidal ultraviolet irradiation.


Different strains of Mycobacterium tuberculosis cause J. Occup. Environ. Hyg. 2:285–292.
various spectrums of disease in the rabbit model of 49. Dannenberg,A. M., Jr. 1978. Pathogenesis of pul-
tuberculosis. Infect. Immun. 71:6004–6011. monary tuberculosis in man and animals: protection
35. Schlesinger, L. S. 1996. Role of mononuclear of personnel against tuberculosis, p. 65–75. In R. J.
phagocytes in M. tuberculosis pathogenesis. J. Inves- Montali (ed.), Mycobacterial Infections of Zoo Ani-
tig. Med. 44:312–323. mals. Smithsonian Institution Press, Washington,
36. Ernst, J. D. 1998. Macrophage receptors for D.C.
Mycobacterium tuberculosis. Infect. Immun. 66:1277– 50. Maryland Department of Health and Mental
1281. Hygiene. 1991. Guidelines for preventing the
37. Russell, D. G. 2001. Mycobacterium tuberculosis: transmission of tuberculosis in health-care settings.
here today, and here tomorrow. Nat. Rev. Mol. Cell. State of Maryland Communicable Diseases Bulletin,
Biol. 2:569–577. April 1991.
38. Russell, D. G., G. E. Purdy, R. M. Owens, 51. Iseman, M. D. 2000. A Clinician’s Guide to Tuber-
K. H. Rohde, and R. M.Yates. 2005. Mycobac- culosis, p. 431–448. Lippincott Williams & Wilkins,
terium tuberculosis and the four-minute phagosome. Philadelphia, Pa.
ASM News 71:459–463. 52. Dowdle, W. R., and the Centers for Disease
39. Pieters, J. 2001. Entry and survival of pathogenic Control. 1989.A strategic plan for the elimination
mycobacteria in macrophages. Microbes Infect. 3:249– of tuberculosis in the United States. Morb. Mortal.
255. Wkly. Rep. 38(Suppl. 3):1–25.
40. Manganelli, R., R. Proveddi, S. Rodrigue, J. 53. Centers for Disease Control and Prevention.
Beaucher, L. Gaudreau, and I. Smith. 2004. 1994. Guidelines for preventing the transmission of
Sigma factors and global gene regulation in Mycobac- Mycobacterium tuberculosis in health-care facilities,
terium tuberculosis. J. Bacteriol. 186:895–902. 1994. Morb. Mortal.Wkly. Rep. 43:1–132.
41. Dannenberg, A. M., Jr., M. S. Burstone, P. C. 54. Centers for Disease Control and Prevention.
Walter, and J.W. Kinsley. 1963.A histochemical 1998. Prevention and treatment of tuberculosis
study of phagocytic and enzymatic functions of among patients infected with human immunode-
rabbit mononuclear and polymorphonuclear exu- ficiency virus: principles of therapy and revised
date cells and alveolar macrophages. I. Survey and recommendations. Morb. Mortal.Wkly. Recomm. Rep.
quantitation of enzymes, and states of cellular acti- 47(RR-20):1–58.
vation. J. Cell Biol. 17:465–486. 55. Jensen, P. A., L. A. Lambert, M. F. Iademarco,
42. Stead, W., J. W. Senner, W. T. Reddick, and R. Ridzon, and the Centers for Disease Con-
J. P. Lofgren. 1990. Racial differences in suscep- trol and Prevention. 2005. Guidelines for pre-
tibility to infection by Mycobacterium tuberculosis. venting the transmission of Mycobacterium tuberculosis
N. Engl. J. Med. 322:422–427. in health-care settings, 2005. Morb. Mortal. Wkly.
43. Dannenberg,A. M., Jr., and J. F.Tomashefski, Recomm. Rep. 54:1–141.
Jr. 1998. Pathogenesis of pulmonary tuberculosis, 56. Tepper, B. S. 1994. Portable HEPA filtration TB
p. 2447–2471. In A. P. Fishman (ed.), Fishman’s Pul- isolation in hospitals and clinics, p. 143–156. In W.
monary Diseases and Disorders, 3rd ed., vol. 2. Charney (ed.), Essentials of Modern Hospital Safety,
McGraw-Hill Book Co., New York, N.Y. vol. 3. Lewis Publishers, Chelsea, Mich.
44. Dubos, R., and J. Dubos. 1952. The White Plague: 57. California Department of Health Services.
Tuberculosis, Man and Society. Little, Brown & Co., 1990. Using Ultraviolet Radiation and Ventilation to
Boston, Mass. Control Tuberculosis. Tuberculosis Control and
45. Rich, A. 1951. The Pathogenesis of Tuberculosis, 2nd Refugee Health Programs Unit, Infectious Disease
ed. Charles C Thomas Publisher, Springfield, Ill. Branch, Sacramento, Calif.
46. Bass, J. B., Jr., L. S. Farer, P. C. Hopewell, 58. Centers for Disease Control and Prevention.
R. O’Brien, R. F. Jacobs, F. Ruben, D. E. 1994. Guidelines for preventing the transmission of
Snider, Jr., and G. Thornton. 1994.Treatment Mycobacterium tuberculosis in health-care facilities,
of tuberculosis and tuberculosis infection in adults 1994—CDC. Notice of final revisions. Fed. Regist.
and children. Official Statement of the American 59:54242–54303.
Thoracic Society, Medical Section of the American 59. Converse, P. J., A. M. Dannenberg, Jr.,T. Shi-
Lung Association. Am. J. Respir. Crit. Care Med. genaga, D. N. McMurray, S. W. Phalen, J. L.
149:1359–1374. Stanford, G. A. W. Rook, T. Koru-Sengul, H.
47. Stanford, J., C. Stanford, and J. Grange. 2004. Abbey, J. E. Estep, and M. L. M. Pitt. 1998. Pul-
Immunotherapy with Mycobacterium vaccae in the monary bovine-type tuberculosis in rabbits: bacil-
treatment of tuberculosis. Front. Biosci. 9:1701–1719. lary virulence, inhaled dose effects, tuberculin sen-
48. First, M. W., R. A. Weker, S. Yasui, and E. A. sitivity, and Mycobacterium vaccae immunotherapy.
Nardell. 2005. Monitoring human exposures to Clin. Diagn. Lab. Immunol. 5:871–881.
2
STAGES IN THE PATHOGENESIS OF
HUMAN AND RABBIT TUBERCULOSIS

First stage of tuberculosis: ingestion, and often destruction, of inhaled bacilli


by alveolar macrophages 22
Second stage of tuberculosis: logarithmic “symbiotic” growth of the bacilli
in nonactivated monocytes/macrophages 24
Third stage of tuberculosis: ending of the logarithmic stage of bacillary
multiplication by DTH (and CMI) 25
Fourth stage of tuberculosis: progression or arrest of caseous lesions 28
Fifth stage of tuberculosis: cavity formation and bronchial spread of the
disease 30
Summary 31

Abstract. After the inhalation of tubercle bacilli by rabbits and humans, the disease may
progress through the following stages. In humans, the disease begins with the establishment
of only a single primary pulmonary tubercle.
Stage 1: Ingestion and often destruction of bacilli by pulmonary alveolar macrophages.
Stage 2: Logarithmic growth of bacilli within nonactivated macrophages that entered
the developing tubercle from the bloodstream.
Stage 3:Arrest of the logarithmic bacillary growth by delayed-type hypersensitivity, which
kills the bacilli-laden macrophages and often forms a solid caseous center in the tubercle.
Stage 4a: In hosts with weakly developed cell-mediated immunity, enlargement of the
tubercle and its caseous center with hematogenous dissemination of the bacilli.
Stage 4b: In hosts with strongly developed cell-mediated immunity, stabilization or regres-
sion of the tubercle.
Stage 5: Liquefaction of the caseous center, extracellular bacillary growth, cavity for-
mation, and bronchial dissemination of the bacilli.
These stages are not distinct but blend into each other.Also, stages 3, 4, and 5 may occur
in the same lung and even in different parts of the same lesion, depending on the local con-
centration of bacilli and their tuberculin-like products. Native and acquired resistance is
never absolute, because a large number of tubercle bacilli (which have grown extracellu-
larly in a cavity) can overwhelm even the best-developed host resistance and cause sec-
ondary pulmonary lesions.

FIRST STAGE OF TUBERCULOSIS:


INGESTION, AND OFTEN are highly activated cells (5). They have been
DESTRUCTION, OF INHALED BACILLI activated nonspecifically by many stimulating
BY ALVEOLAR MACROPHAGES factors, including the ingestion and digestion of
Stage 1 (Fig. 1) begins when a bacillary particle a variety of inhaled microorganisms and organic
containing 1 to 3 bacilli is inhaled into an alve- matter (see reference 6), as well as an occasional
olus. (Larger particles impinge on the bronchial extravasated erythrocyte.Therefore, in humans
mucosa and never reach the alveoli.) (See ref- and in rabbits inhaling human-type tubercle
erences 1 through 4.) Once there, an alveolar bacilli, the alveolar macrophages may destroy the
macrophage will ingest the bacilli and often bacilli before they multiply. In such cases, the
destroy them, depending on the inherent micro- tuberculin skin test remains negative.
bicidal power of the alveolar macrophage and on Seven days after the inhalation of human
the genetic and phenotypic virulence of the tubercle bacilli, the lungs of Lurie’s inbred sus-
ingested bacillus. Most alveolar macrophages ceptible rabbits contained 20- to 30-fold more

22
2. STAGES OF HUMAN AND RABBIT TUBERCULOSIS 䡵 23

viable bacilli than did the lungs of his inbred re-


sistant rabbits (7, 8) (Fig. 2).Therefore, the alve-
olar macrophages of the resistant host must have
destroyed or inhibited more inhaled bacilli than
did those of the susceptible host. In other words,
the alveolar macrophages of the resistant rabbits
were evidently nonspecifically activated to a
greater extent than were those of the suscepti-
ble rabbits.That the alveolar macrophages from
FIGURE 1 Stage 1. An alveolar macrophage that
has ingested and killed the two tubercle bacilli in a the resistant rabbits were able to phagocytize
phagocytic vacuole.The cytoplasm of this macrophage particles more readily than those from the sus-
is darkly shaded to depict a high degree of activation, ceptible rabbits supports this conclusion (9).
i.e., high levels of lysosomal and oxidative enzymes. In vitro metabolic studies on mineral oil-
Most alveolar macrophages are nonspecifically activated induced peritoneal macrophages confirmed that
by the variety of inhaled particles that they ingest. In
humans, an alveolar macrophage is usually able to kill an macrophages from Lurie’s resistant rabbits could
inhaled tubercle bacillus, except when the bacillus is be nonspecifically activated to a greater extent
unusually virulent or the macrophage is poorly activated. (by the oil irritant) than those from his suscep-
Reproduced with permission from reference 4. tible rabbits (10).

FIGURE 2 Changes in the number of virulent human-type tubercle bacilli


(H37Rv) in the lungs of Lurie’s natively resistant and natively susceptible rabbits
at different intervals after an aerosol infection. By 7 days, the resistant rabbits had
inhibited the growth of the bacilli 20 to 30 times more effectively than did the sus-
ceptible rabbits, but, from then on, the two curves were parallel. At 4 to 5 weeks,
the susceptible rabbits had about 13 times more primary pulmonary tubercles in
their lungs than did the resistant rabbits.The means and their standard errors are
shown. Reproduced with permission from reference 8.
The number of tubercle bacilli in the lungs of the resistant rabbits failed to
decrease during the period illustrated, because liquefaction with extracellular mul-
tiplication of the bacillus readily occurred in these rabbits (7, 8). Liquefaction did
not occur in the susceptible rabbits (7, 8), because their macrophages probably devel-
oped only low levels of hydrolytic enzymes.
Inhaled virulent bovine-type tubercle bacilli (Ravenel S) grew to greater titers
than did H37Rv, but produced similar curves (28) (shown in chapter 15).
24 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

SECOND STAGE OF TUBERCULOSIS: serum (such as C5a released when complement


LOGARITHMIC “SYMBIOTIC” is activated [11]) (see chapters 19 and 20).
GROWTH OF THE BACILLI IN In time, macrophages from the bloodstream
NONACTIVATED
MONOCYTES/MACROPHAGES become completely responsible for the fate of
Stage 2 (Fig. 3) is called symbiotic (7) because the early lesion. In such a lesion, the alveolar
the number of bacilli in the lesions increases macrophages rarely participate, because they
without apparent damage to the host, and the remain peripheral, rather far from the bacilli,
number of macrophages also increases without which are almost always located more centrally
apparent damage to the bacilli. (See references (Fig. 4).
1 through 4.) If the original alveolar macrophage The macrophages from the bloodstream are
fails to destroy or inhibit the inhaled bacillary nonactivated and immature.They readily ingest
particle that it ingested, the bacilli multiply the bacilli released from alveolar macrophages.
intracellularly until the macrophage ruptures. A symbiotic relationship then develops, in which
The released bacilli are then ingested by other the macrophages of the host and the bacilli can-
alveolar macrophages and by monocytes/macro- not injure each other. These nonactivated
phages arriving from the bloodstream. Both macrophages cannot inhibit or destroy the
types of macrophages are attracted to the site by bacilli, and the bacilli cannot injure the
chemotaxins released from the bacilli (such as N- macrophages (Fig. 3 and 5) because the host is
formyl-methionyl-leucyl-phenylalanine [11]), not yet tuberculin positive. With time, many
by chemotaxins from live alveolar macrophages macrophages and many bacilli accumulate in
(such as leukotriene B4 [11] and monocyte this early lesion, which now has become a
chemoattractant protein 1 [12]), by chemotax- microscopic tubercle.
ins from dying alveolar macrophages (11) (such In this symbiotic stage (between 7 and 21 days
as uric acid [13]), and by chemotaxins from after infection), the bacilli grow logarithmically
at the same rate in both resistant and susceptible
rabbits (Fig. 2) (8). Evidently, intracellular bacilli
inhibit the microbicidal mechanisms (14–16) of
these immature macrophages equally well in both
the resistant and the susceptible hosts (see chap-
ters 6 and 15).
In rabbits, the more virulent bovine-type
bacilli (Ravenel S) and the less virulent human-
type bacilli (H37Rv) also grow at the same rate
in nonactivated macrophages. However, the
more virulent bovine-type bacilli grow for a
longer period of time (see chapter 15). Evi-
dently, acquired (adaptive) immunity develops
FIGURE 3 Stage 2. An early primary pulmonary more slowly with tubercle bacilli of greater vir-
tubercle, in which tubercle bacilli have multiplied loga- ulence. Perhaps some of the antigens that cause
rithmically within macrophages that have immigrated acquired immunity are released more slowly
into the lesion from the bloodstream.These newly arrived
macrophages are nonactivated and incompetent. Their from the more virulent strain, because the
cytoplasm is unshaded to depict the lack of activation. bovine type seems to remain intact longer.
In fact, the phagocytic vacuoles in the cytoplasm of Even though little or no difference existed in
these nonactivated macrophages seem to provide an the ability of Lurie’s resistant and susceptible rab-
ideal environment for mycobacterial multiplication. bits to control the growth of tubercle bacilli in
Stage 2 is called the stage of symbiosis, because the
bacilli are multiplying, the macrophages are accumulat- nonactivated macrophages (Fig. 2), a marked dif-
ing, and neither the host nor the parasite is injured by the ference existed in the ability of resistant and sus-
other. Reproduced with permission from reference 4. ceptible rabbits to activate macrophages. The
2. STAGES OF HUMAN AND RABBIT TUBERCULOSIS 䡵 25

FIGURE 4 An early pulmonary


tuberculous lesion with a caseous
center surrounded by blood-borne
macrophages. The alveolar macro-
phages (recognized by their dark
staining for the lysosomal enzyme -
galactosidase) have accumulated
peripherally in nearby alveoli. More
details on this lesion are given in
Fig. 5 of chapter 9. Magnification,
⫻330. Reproduced with permission
from reference 4.

resistant rabbits apparently activated their pul- THIRD STAGE OF TUBERCULOSIS:


monary alveolar macrophages nonspecifically to ENDING OF THE LOGARITHMIC
a greater degree than did the susceptible rabbits STAGE OF BACILLARY
MULTIPLICATION BY DTH
before inhalation of tubercle bacilli (see above), (AND CMI)
and the resistant rabbits immunologically activated Stage 3 (Fig. 6) is the early stage of caseous
to a greater degree the blood-borne macrophages necrosis. It occurs in rabbits 2 to 3 weeks after
that entered their tuberculous lesions than did the the inhalation of tubercle bacilli, which is the
susceptible rabbits (see below). time when the antigen-specific immune

FIGURE 5 A 2-week pulmonary


lesion produced in a rabbit by the
inhalation of virulent human-type
tubercle bacilli (H37Rv) (8). The
lesion consists of blood-borne non-
activated macrophages in which
large numbers of bacilli had multi-
plied intracellularly. Note that, in
this stage of symbiosis (i.e., before
DTH develops), even numerous
intracellular bacilli do not injure the
macrophages. In other words, the
bacillus is rather innocuous before
DTH develops, and it is the host’s
own DTH reaction that kills tissues
during this disease. Stained with car-
bol-fuchsin, counterstained with
methylene blue. Magnification,
⫻730. Reproduced with permission
from reference 8.
26 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

The concept that DTH kills the nonacti-


vated macrophages in which many tubercle
bacilli are growing, thereby eliminating the
favorable intracellular environment, was advo-
cated by several research groups (17–23) and had
been predicted many years ago (24, 25). In fact,
Robert Koch described it before DTH and
CMI were in our vocabulary (26). Tubercle
bacilli may survive in this solid caseous material,
but they cannot multiply, probably owing to
the anoxic conditions, reduced pH, and the
presence of inhibitory fatty acids (25, 27).
But why did the susceptible rabbits, which are
known to develop rather weak DTH and CMI
FIGURE 6 Stage 3. A tubercle 3 weeks of age with responses (7), stop the logarithmic growth of the
a caseous necrotic center and a peripheral accumulation bacillus just as effectively as the resistant strain
of partly activated macrophages (lightly shaded) and of rabbits? The answer is that the large number
lymphocytes (small dark cells). The first stages of
caseation occur when the tissue-damaging DTH
of bacilli in the susceptible rabbits produced
response to a high concentration of tuberculin-like high levels of tuberculin-like products that
products kills the nonactivated macrophages that have caused a strong local tissue-damaging DTH
allowed the bacilli to multiply logarithmically within response: their tuberculous lesions had larger
them.The dead and dying macrophages are depicted by caseous centers than those of the resistant rab-
fragmented cell membranes. Intact and fragmented
bacilli are present, both within macrophages and within
bits (8, 28).The large number of bacilli in the
the caseum. Tubercle bacilli do not multiply in solid susceptible rabbits also enhanced their dermal
caseum. Reproduced with permission from reference 4. tuberculin reactivity.
The greater antigenic stimulus in the sus-
ceptible rabbits did not, however, increase their
response becomes established. (See references 1 CMI to the levels found in the resistant rabbits.
through 4.) The development of tissue-damag- The lesions of the susceptible rabbits contained
ing delayed-type hypersensitivity (DTH) that many more bacilli but relatively few mature
kills macrophages harboring more than a few epithelioid cells. In other words, their macro-
tubercle bacilli eliminates the favorable intra- phages were not activated to the degree found
cellular environment for bacillary growth (Fig. in the resistant rabbits. Perhaps the lympho-
6). Blood vessels are damaged, and solid caseous cytes of the susceptible rabbits did not produce
necrosis is produced within which the bacillus the cytokines required to do so. Perhaps their
cannot grow. In other words, tissue-damaging macrophages were unable to respond appropri-
DTH to high local concentrations of tuber- ately to such cytokines. Or, perhaps, both
culin-like bacillary products is used by both the occurred, or other factors were involved.
resistant and susceptible rabbits to end the log- In brief, in stage 3 (the early stage of caseous
arithmic stage of bacillary growth. necrosis), both the resistant and susceptible rab-
Unexpectedly, Lurie’s susceptible hosts inhib- bits locally destroyed the macrophages that had
ited further increases in the number of viable allowed the bacillus to grow during the sym-
bacilli just as efficiently as did the resistant hosts biotic stage. Only after such control was estab-
(Fig. 2). Cell-mediated immunity (CMI) could lished could CMI (producing highly activated
not be responsible, because the susceptible hosts macrophages surrounding the caseous focus)
developed weaker CMI. The marked inhibi- prevent the progression of the disease. Since sus-
tion of bacillary growth found in both strains of ceptible rabbits do not develop good CMI, the
rabbits must therefore be due to the tissue- susceptible rabbits continued (in stage 4) to use
damaging DTH. tissue-damaging DTH to inhibit bacillary
2. STAGES OF HUMAN AND RABBIT TUBERCULOSIS 䡵 27

growth, but in doing so they destroyed increas- in the tuberculous lesions where the bacillary
ing amounts of their own tissues. antigens are located. Antigen-specific lympho-
Ending the logarithmic (symbiotic) stage of cytes expand and accumulate in the draining
bacillary growth may be due to several factors, but lymph nodes and lesions before such lympho-
killing of bacilli-laden macrophages by tissue- cytes are available to enter dermal tuberculin
damaging DTH is the major factor in humans, reactions (see chapter 5).
rabbits, and guinea pigs. CMI develops simulta- The role of nonspecific factors in ending the
neously with DTH and stops the growth of logarithmic stage is minimal (discussed further
tubercle bacilli within macrophages when they in the last section of chapter 5). The bacilli
contain relatively few microorganisms. There- release few, if any, endotoxin-like factors while
fore, CMI acts synergistically with DTH to con- they are growing intracellularly in nonactivated
trol bacillary growth. macrophages. If they did, the macrophages
Tuberculin-positive skin tests develop sev- would be activated during the first 24 h (see ref-
eral days after both CMI and DTH are expressed erence 29). Before DTH and CMI develop,

FIGURE 7 (A) Stage 4a. An established tubercle 4 to 5 weeks of age similar to those found in
Lurie’s susceptible rabbits. It has an enlarging caseous center.The bacilli escaping from the edge
of this center are ingested by poorly activated incompetent macrophages. In such macrophages,
the bacilli again find a favorable intracellular environment in which to multiply.They do so until
the tissue-damaging DTH (to high concentrations of tuberculin-like products) again kills these
new bacilli-laden macrophages and enlarges the caseous necrotic center.This sequence may be
repeated multiple times. Lung tissue is destroyed, and the bacilli are spread by the lymphatic and
hematogenous routes to other sites. Metastatic lesions develop in which the tissue destruction
continues. This pattern of tuberculosis is seen in immunosuppressed individuals, such as HIV/
AIDS patients. In this cartoon, several partly activated macrophages are lightly shaded to indicate
that such susceptible hosts develop only relatively weak CMI. Reproduced with permission from
reference 4. (B) Stage 4b.An established tubercle 4 or 5 weeks of age similar to those found in Lurie’s
resistant rabbits.The caseous center remains small, because the bacilli escaping from its edge are
ingested by the highly activated (competent) macrophages (darkly shaded) that surround the caseum.
In these activated macrophages, the bacilli cannot multiply and are eventually destroyed. Such effec-
tive macrophages were activated by T cells and their cytokines. If the caseous center remains solid
and does not liquefy, the disease will be arrested by this CMI response.This scenario occurs in healthy
immunocompetent humans who show positive tuberculin reactions and yet no clinical and often
no X-ray evidence of the disease. Reproduced with permission from reference 4.
28 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

intracellular tubercle bacilli inhibit macrophage phogenous and hematogenous spread of the
activation and their microbicidal abilities (see bacilli occurs, and progressive secondary lesions
chapters 1 and 6). After DTH and CMI develop, develop in the hilar nodes, lungs, and other
the bacilli are killed and many of their compo- organs (see chapter 13).
nents are released. Only then can many of these If CMI is strong (Fig. 7B), as in Lurie’s resis-
components activate macrophages nonspecifi- tant rabbits, virulent tubercle bacilli escaping
cally and act synergistically with DTH and CMI from the caseous center are ingested and
to control the progress of the disease. destroyed (or inhibited) by highly activated
FOURTH STAGE OF TUBERCULOSIS: macrophages that have accumulated perifocally.
PROGRESSION OR ARREST OF The lesions then are stabilized and even regress.
CASEOUS LESIONS The bacilli that reach the hilar lymph nodes of
Stage 4 (Fig. 7) consists of an interplay of CMI these rabbits are inhibited by the same process.
and tissue-damaging DTH. During this stage, In rabbits, virulent human-type tubercle bacilli
the disease may become clinically apparent, at are much less virulent than the bovine type. (In
least by radiograph, or it may be arrested with humans, both types are of similar virulence.) The
little visible evidence remaining except for con- disease produced by virulent human-type bacilli
version of the tuberculin reaction. usually heals in a year or so in both susceptible and
If CMI is weak (Fig. 7A), as in Lurie’s sus- resistant inbred rabbits, but the disease produced
ceptible rabbits, virulent tubercle bacilli escap- by virulent bovine-type bacilli progresses to death
ing from the edge of the caseous centers again in both rabbit strains (see chapter 13). Figure 2
multiply intracellularly in poorly activated shows the bacillary titers in the lungs of Lurie’s
macrophages (Fig. 8). Again, these infected rabbits that had inhaled human-type bacilli. Fig-
macrophages are killed by tissue-damaging ure 1 in chapter 15 shows the bacillary titers for
DTH, and the caseous center enlarges. Lym- rabbits inhaling the bovine type.

FIGURE 8 Photograph of stage


4a: a caseous tubercle in the lungs of
one of Lurie’s susceptible rabbits 5
weeks after the inhalation of human-
type tubercle bacilli. Bacilli escaping
from the caseous center (lower left)
are ingested by the surrounding
poorly activated macrophages, i.e.,
incompetent immature epithelioid
cells (upper right), where they again
find a favorable intracellular envi-
ronment in which to grow (Fig. 7A).
Magnification, ⫻550. Reproduced
with permission from reference 39.
2. STAGES OF HUMAN AND RABBIT TUBERCULOSIS 䡵 29

FIGURE 9 Stage 5.A recently formed cavity discharging liquefied caseous ma-
terial into a bronchus. In liquefied caseum, the bacilli may multiply extracellularly,
reaching large numbers. High concentrations of tuberculin-like products are pro-
duced and local tissues are destroyed, including the wall of an adjacent bronchus (illus-
trated here).The liquefied caseous material is then discharged into the airways, and
the bacilli disseminate to other parts of the lung and to the environment. Repro-
duced with permission from reference 4.
The large quantities of bacilli and their antigens in liquefied caseum may over-
whelm a formerly effective CMI, causing progression of the disease in Lurie’s re-
sistant rabbits, as well as in immunocompetent humans.Also, among such large num-
bers of bacilli, mutations causing antimicrobial resistance may occur. Lurie’s
susceptible rabbits do not liquefy caseum or form cavities.

FIGURE 10 Wall of a cavity from one


of Lurie’s genetically resistant rabbits 8
weeks after the inhalation of human-
type bacilli (H37Rv). The liquefied
caseous tissue (right) and liquefying
caseous tissue (left) contain a large num-
ber of (rod-shaped) acid-fast bacilli. Such
bacilli were formerly inhibited in solid
caseous tissue, but they grew profusely in
the liquefied caseum in the wall of this
cavity. Stained with carbol-fuchsin, coun-
terstained with methylene blue. Magni-
fication, ⫻540. Reproduced with per-
mission from reference 8.
30 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

TABLE 1 Five stages of tuberculosis


Stage 1
Pulmonary alveolar macrophages ingest the inhaled bacilli. In rabbits (and humans), most of the resident alveolar
macrophages are highly activated (5) and destroy or inhibit the bacilli before they multiply appreciably (Fig. 1).
However, if a poorly activated alveolar macrophage ingests an inhaled bacillus, it can multiply intracellularly
and eventually destroy the alveolar macrophage.
Stage 2
The bacilli are released from poorly activated alveolar macrophages and are ingested by immature (nonactivated)
monocytes/macrophages that enter the tubercle from the bloodstream.Within these immature macrophages,
the bacilli grow logarithmically (Fig. 3).This stage is called symbiotic (4, 7) because both tubercle bacilli and
macrophages accumulate in the developing lesion without harming each other.
Stage 3
Caseous necrosis ends the symbiotic stage of bacillary growth in humans, rabbits, and guinea pigs (Fig. 6).
Tubercle bacilli cannot multiply within solid caseum. Caseous necrosis is caused by a tissue-damaging delayed-
type hypersensitivity (DTH) reaction to the high local concentrations of tuberculin-like antigens that occur
whenever the bacilli are numerous within (poorly activated) macrophages (2, 3).Therefore, DTH and the
resulting caseous necrosis play major roles in stopping the progression of tuberculous lesions.
At this stage, the lesion contains a solid caseous center. Surrounding this center are nonactivated macrophages,
which permit intracellular bacillary multiplication, as well as some activated macrophages, which do not.The
activated macrophages are produced by the cell-mediated immunity (CMI) that develops along with the DTH
(see chapter 5).
Stage 4
This stage determines whether the early tubercle becomes clinically apparent. Here, CMI (causing local acquired
cellular resistance) plays a major role (see chapters 5 and 11). If only weak CMI develops (Fig. 7A), the bacilli
escaping from the edge of the caseous necrosis multiply again in nonactivated and partly activated macro-
phages.The tissue-damaging DTH immune response again kills these macrophages, causing enlargement of
the caseous center and progression of the disease.
If a strong CMI develops (Fig. 7B), a mantle of highly activated macrophages is produced that surrounds the
caseous center.These macrophages ingest and destroy (or inhibit) the bacilli escaping from the edge of the
caseous center and often arrest the lesion at a subclinical stage.
Stage 5
The solid caseum liquefies, and a cavity forms. In liquefied caseum, the bacillus may multiply extracellularly (for
the first time) and may reach tremendous numbers (Fig. 9).The high local concentration of tuberculin-like
products released by numerous bacilli causes a tissue-damaging DTH response that erodes a nearby bronchial
wall, so that a cavity forms.The liquefied caseum and tubercle bacilli are discharged into the airways, and may
spread to other parts of the lung and to the outside environment—most commonly during spells of coughing.
Arrest of the disease at this stage depends on whether the antigenic load (of both the bacilli and their products)
remains small enough for the host to control.When numerous bacilli are present, even a host with a well-
developed native and acquired resistance may not stop the progress of the disease.

FIFTH STAGE OF TUBERCULOSIS: The liquefied caseum is frequently, but not


CAVITY FORMATION AND always (33), an excellent growth medium for
BRONCHIAL SPREAD OF THE
DISEASE tubercle bacilli (7, 24, 34). In liquefied caseum,
Stage 5 (Fig. 9) is the stage of the liquefaction the bacillus may multiply extracellularly (for
of solid caseum and the formation of cavities. the first time during the course of the disease)
These events can cause the disease to progress, and may reach tremendous numbers (Fig. 10).
even in resistant hosts with good CMI, such as Bacilli in large numbers are toxic to tissues
in Lurie’s resistant rabbits and in adult humans because of the host’s DTH to their tuberculin-
with a fully competent immune system. Lique- like products. Necrosis of a nearby bronchus
faction with cavity formation perpetuates the occurs, and a cavity is formed.Then, the bacilli
disease in humankind (4, 24, 30–32, 32a). in the liquefied caseum are discharged into the
2. STAGES OF HUMAN AND RABBIT TUBERCULOSIS 䡵 31

airways and are distributed to other parts of the tuberculous lesions in human necropsy (or sur-
lung and to the outside environment, where gical) material can never duplicate primary
they may infect other persons. lesions in their early stages.The early develop-
Since liquefied caseum contains tuberculin- ment of primary lesions can only be studied in
like proteins, it causes an exudate whenever it is animal models.
aspirated into the alveolar spaces. Caseous bron-
chopneumonia results when this aspiration is REFERENCES
extensive (see chapter 3). 1. Dannenberg, A. M., Jr. 1999. Pathophysiology:
Furthermore, mutant tubercle bacilli that are basic aspects. I. Pathogenesis of tuberculosis. II.
resistant to antimicrobial agents may arise among Immunology of tuberculosis, p. 17–47. In D.
the large number of bacilli in the liquefied Schlossberg (ed.), Tuberculosis and Nontuberculous
Mycobacterial Infections, 4th ed. The W. B. Saunders
caseum. For this reason, tuberculosis is usually Co., Philadelphia, Pa.
treated simultaneously with two to four antimi- 2. Dannenberg, A. M., Jr. 1991. Delayed-type
crobials. hypersensitivity and cell-mediated immunity in the
Cavities almost never occurred in Lurie’s sus- pathogenesis of tuberculosis. Immunol.Today 12:228–
ceptible rabbits (7, 31), and cavities rarely occur 233.
3. Dannenberg, A. M., Jr. 1993. Immunopatho-
in infants or in immunosuppressed adults (30, 35). genesis of pulmonary tuberculosis. Hosp. Pract.
Macrophages do not survive in liquefied 28:51–58.
caseous material (7). High concentrations of 4. Dannenberg, A. M., Jr., and G. A. W. Rook.
tuberculin-like antigens exist in liquefied caseum, 1994. Pathogenesis of pulmonary tuberculosis: an
and macrophages may have been passively sensi- interplay of tissue-damaging and macrophage-
activating immune responses—dual mechanisms
tized to such antigens by cytophilic antibodies. that control bacillary multiplication, p. 459–483. In
Also, macrophages may be killed by the toxic fatty B. R. Bloom (ed.), Tuberculosis: Pathogenesis, Protec-
acids present (25, 27).Therefore, even activated tion, and Control.American Society for Microbiol-
macrophages (produced by well-developed CMI) ogy,Washington, D.C.
cannot control the extracellular multiplication 5. Dannenberg, A. M., Jr., M. S. Burstone, P. C.
Walter, and J. W. Kinsley. 1963.A histochemical
of tubercle bacilli within cavities. study of phagocytic and enzymatic functions of
The cause of liquefaction is largely unknown. rabbit mononuclear and polymorphonuclear exu-
Hydrolytic enzymes (32a, 33, 36) play a role, as date cells and alveolar macrophages. I. Survey and
well as DTH to the tuberculin-like products of quantitation of enzymes, and states of cellular acti-
the bacillus (37, 38) (reviewed in chapter 4).At vation. J. Cell Biol. 17:465–486.
6. Dannenberg, A. M., Jr., P. C.Walter, and F. A.
present, no therapeutic agent exists to prevent Kapral. 1963. A histochemical study of phago-
liquefaction, but, if developed, such an agent cytic and enzymatic functions of rabbit mononu-
could greatly aid antimicrobial therapy in clear and polymorphonuclear exudate cells and
reducing the number of viable tubercle bacilli alveolar macrophages. II. The effect of particle
and arresting the disease. ingestion on enzyme activity; two phases of in vi-
tro activation. J. Immunol. 90:448–465.
7. Lurie, M. B. 1964. Resistance to Tuberculosis: Exper-
SUMMARY imental Studies in Native and Acquired Defensive Mech-
Table 1 summarizes the major characteristics anisms. Harvard University Press, Cambridge, Mass.
of each of the five stages of tuberculosis.These 8. Lurie, M. B., P. Zappasodi, and C. Tickner.
stages are not distinct entities, because one stage 1955. On the nature of genetic resistance to tuber-
culosis in the light of the host-parasite relationships
blends into the next one. However, they do in natively resistant and susceptible rabbits. Am.
provide a structure for understanding the vari- Rev.Tuberc. 72:297–329.
ous types of tuberculosis that occur in humans 9. Henderson, H. J., A. M. Dannenberg, Jr., and
(see chapter 3). M. B. Lurie. 1963. Phagocytosis of tubercle bacilli
In secondary lesions during the primary infec- by rabbit pulmonary alveolar macrophages and its
relation to native resistance to tuberculosis. J.
tion and in lesions of exogenous reinfection, Immunol. 91:553–556.
each stage occurs at an accelerated rate because 10. Allison, M. J., P. Zappasodi, and M. B. Lurie.
of the presence of DTH and CMI. Therefore, 1961. Metabolic studies of mononuclear cells from
32 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

rabbits of varying genetic resistance to tuberculosis. 24. Canetti, G. 1955. The Tubercle Bacillus in the Pul-
I. Studies on cells of normal non-infected animals. monary Lesion of Man, p. 130. Springer Publishing
Am. Rev. Respir. Dis. 84:364–370. Company, Inc., New York, N.Y.
11. Majno, G., and I. Joris. 2004. Cells,Tissues, and 25. Poole, J. C. F., and H.W. Florey. 1970. Chronic
Disease: Principles of General Pathology, 2nd ed. Oxford inflammation and tuberculosis, p. 1183–1224. In H.
University Press, New York, N.Y. W. Florey (ed.), General Pathology, 4th ed. The
12. Leonard, E. J., and T.Yoshimura. 1990. Human W. B. Saunders Co., Philadelphia, Pa.
monocyte chemoattractant protein-1 (MCP-1). 26. Koch, R. 1891. Fortsetzung der Mittheilungen
Immunol.Today 11:97–101. über ein Heilmittel gegen Tuberculose [Contin-
13. Shi,Y., J. E. Evans, and K. L. Rock. 2003. Mo- uation of the communication concerning a treat-
lecular identification of a danger signal that alerts ment for tuberculosis]. Dtsch. Med.Wsch. 15:101–
the immune system to dying cells. Nature 425:516– 102.
521. 27. Hemsworth, G. R., and I. Kochan. 1978.
14. Pabst, M. J., J. M. Gross, J. P. Brozna, and Secretion of antimycobacterial fatty acids by
M. B. Goren. 1988. Inhibition of macrophage normal and activated macrophages. Infect. Immun.
priming by sulfatide from Mycobacterium tuberculo- 19: 170–177.
sis. J. Immunol. 140:634–640. 28. Allison, M. J., P. Zappasodi, and M. B. Lurie.
15. Brozna, J. P., M. Horan, J. M. Rademacher, 1962. Host-parasite relationships in natively resis-
K. M. Pabst, and M. J. Pabst. 1991. Monocyte tant and susceptible rabbits on quantitative inhala-
responses to sulfatide from Mycobacterium tuberculo- tion of tubercle bacilli: their significance for the
sis: inhibition of priming for enhanced release of nature of genetic resistance. Am. Rev. Respir. Dis.
superoxide, associated with increased secretion of 85:553–569.
interleukin-1 and tumor necrosis factor alpha, and 29. Cohn, Z. A., and B. Benson. 1965.The in vi-
altered protein phosphorylation. Infect. Immun. tro differentiation of mononuclear phagocytes. III.
59:2542–2548. The reversibility of granule and hydrolytic enzyme
16. Rook, G.A.W. 1990. Mycobacteria, cytokines and formation and the turnover of granule con-
antibiotics. Pathol. Biol. 38:276–280. stituents.
17. Kaufmann, S. H. E. 1989. In vitro analysis of the J. Exp. Med. 122:455–466.
cellular mechanisms involved in immunity to tuber- 30. Dannenberg, A. M., Jr., and J. F. Tomashef-
culosis. Rev. Infect. Dis. 11(Suppl. 2):S448–S454. ski, Jr. 1998. Pathogenesis of pulmonary tuber-
18. Kaufmann, S. H. E. 1988. CD8⫹ T lymphocytes culosis, p. 2447–2471. In A. P. Fishman (ed.), Fish-
in intracellular microbial infections. Immunol.Today man’s Pulmonary Diseases and Disorders, 3rd ed.,
9:168–174. vol. 2. McGraw-Hill Book Co., New York, N.Y.
19. Ottenhoff, T. H. M., and R. R. P. de Vries. 31. Lurie, M. B., and A. M. Dannenberg, Jr.
1990. Antigen reactivity and autoreactivity: two 1965. Macrophage function in infectious disease
sides of the cellular immune response induced with inbred rabbits. Bacteriol. Rev. 29:466–476.
by mycobacteria. Curr.Top. Microbiol. Immunol. 155: 32. Rich, A. R. 1951. The Pathogenesis of Tubercu-
111–121. losis, 2nd ed. Charles C Thomas Publisher,
20. Boom, W. H., R. S. Wallis, and K. A. Cher- Springfield, Ill.
venak. 1991. Human Mycobacterium tuberculosis- 32a. Dannenberg, A. M., Jr., and M. Sugimoto.
reactive CD4⫹ T-cell clones: heterogeneity in anti- 1976. Liquefaction of caseous foci in tubercu-
gen recognition, cytokine production, and losis. Am. Rev. Respir. Dis. 113:257–259. (Edi-
cytotoxicity for mononuclear phagocytes. Infect. torial.)
Immun. 59:2737–2743. 33. Converse, P. J., A. M. Dannenberg, Jr., J. E.
21. Kaleab, B., T. Ottenhoff, P. Converse, Estep, K. Sugisaki, Y. Abe, B. H. Schofield,
E. Halapi, G. Tadesse, M. Rottenberg, and and M. L. M. Pitt. 1996. Cavitary tuberculosis
R. Kiessling. 1990. Mycobacterial-induced cyto- produced in rabbits by aerosolized virulent tuber-
toxic T cells as well as nonspecific killer cells derived cle bacilli. Infect. Immun. 64:4776–4787.
from healthy individuals and leprosy patients. Eur. 34. Long, E. R. 1958. The Chemistry and Chemother-
J. Immunol. 20:2651–2659. apy of Tuberculosis, 3rd ed. Lippincott Williams &
22. Lowrie, D. B. 1990. Is macrophage death on the Wilkins, Baltimore, Md.
field of battle essential to victory, or a tactical weak- 35. Barnes, P. F.,A. B. Bloch, P.T. Davidson, and
ness in immunity against tuberculosis? Clin. Exp. D. E. Snider, Jr. 1991. Tuberculosis in patients
Immunol. 80:301–303. with human immunodeficiency virus infection. N.
23. Lewinsohn, D.A.,A. S. Heinzel, J. M. Gardner, Engl. J. Med. 324:1644–1650.
L. Zhu, M. R.Alderson, and D. M. Lewinsohn. 36. Tsuda, T., A. M. Dannenberg, Jr., M. Ando,
2003. Mycobacterium tuberculosis-specific CD8⫹ T O. Rojas-Espinosa, and K. Shima. 1974.
cells preferentially recognize heavily infected cells. Enzymes in tuberculous lesions hydrolyzing pro-
Am. J. Respir. Crit. Care Med. 168:1346–1352. tein, hyaluronic acid and chondroitin sulfate: a
2. STAGES OF HUMAN AND RABBIT TUBERCULOSIS 䡵 33

study of isolated macrophages and developing and cavity formation by desensitization with tuberculin-
healing rabbit BCG lesions with substrate film active peptide. Am. Rev. Respir. Dis. 109:594–
techniques; the shift of enzyme pH optima towards 601.
neutrality in “intact” cells and tissues. J. Reticuloen- 39. Lurie, M. B., S. Abramson, and A. G. Hep-
dothel. Soc. 16:220–231. pleston. 1952. On the response of genetically re-
37. Yamamura,Y. 1958.The pathogenesis of tuber- sistant and susceptible rabbits to the quantitative
culous cavities. Adv.Tuberc. Res. 9:13–37. inhalation of human-type tubercle bacilli and the
38. Yamamura, Y., Y. Ogawa, H. Maeda, and nature of resistance to tuberculosis. J. Exp. Med.
Y. Yamamura. 1974. Prevention of tuberculous 95:119–134.
3
TYPES OF HUMAN
PULMONARY TUBERCULOSIS

Types of clinical tuberculosis 34


Establishment of the infection 35
The early primary lesion 38
Encapsulated caseous or calcified nodules 38
Proliferative and exudative types of pulmonary lesions 38
Small, discrete tubercles of hematogenous origin and miliary
tuberculosis 40
Liquefied caseous lesions and cavities and bronchogenic spread 42
Progressive, locally destructive lesions 46
Advanced fibrocaseous tuberculosis 46
Tuberculous pneumonia and pleurisy 47
Childhood tuberculosis: lymphatic spread and the Ghon complex 49
Adult tuberculosis: endogenous and exogenous reinfection and subapical
localization 51
Causes of subapical localization 52
Multidrug-resistant tubercle bacilli 57
Recent analyses of human cavitary tuberculous lesions 59
Tuberculosis in the immunocompromised host 59
Inapparent arrested primary tuberculous lesions in healthy humans
compared with rabbits 60

Abstract. Human tuberculosis most frequently occurs as a tiny inapparent lesion that stays
dormant throughout the life of the host. If clinical disease is produced, it varies from the
rapidly progressing, hematogenously spread disease that occurs in infants and immuno-
suppressed individuals to a chronic, slowly progressing cavitary disease that is commonly
found in immunocompetent adults.A brief overview of both the childhood and adult types
of pulmonary tuberculosis is presented in chapter 1.
In this chapter, the gross and histopathological characteristics of each type are described
in more detail. Included are the possible causes of subapical localization in adult-type tuber-
culosis, the location of multidrug-resistant tubercle bacilli in the lungs, the characteristics
of tuberculosis in immunocompromised individuals, and a comparison of inapparent
arrested primary lesions in humans and rabbits. For further discussion of many of the top-
ics in this chapter, see reference 1.

TYPES OF CLINICAL TUBERCULOSIS Active pulmonary tuberculosis, once devel-


In most persons who are infected with the oped in humans, is a locally controlled disease
tubercle bacillus, the disease is arrested as a sin- (1–5). Each lesion is handled by the host almost
gle, inapparent primary pulmonary lesion that as if the other lesions did not exist.Thus, lesions
is not large enough to be detected in a chest in one area of the lung may liquefy and progress,
X-ray but is sufficient to produce a positive while lesions in another area of the same lung
tuberculin reaction. The progression of such may stabilize or even regress. Even parts of a sin-
inapparent primary lesions is usually stopped gle lesion may progress, while other parts remain
by the development of delayed-type hypersen- stable or regress. Also, tuberculous lesions may
sitivity (DTH) and cell-mediated immunity change over time from one type to another or
(CMI) (see chapters 2 and 5). Some primary may be a composite of several types. Finally, the
lesions, however, do not heal while they are disease as a whole may fluctuate between peri-
tiny, but progress to a larger size. ods of exacerbation and remission.

34
3. TYPES OF HUMAN PULMONARY TUBERCULOSIS 䡵 35

TABLE 1 Basic types of pulmonary tuberculosisa numbers of actively multiplying bacilli in a given
lesion produce large amounts of tuberculin-like
Types of lesions
Encapsulated caseous, liquefied, or calcified nodules products, which cause caseous necrosis, but small
Proliferative type of pulmonary lesions numbers of bacilli produce small amounts of
Exudative type of pulmonary lesions tuberculin-like products, which enhance host re-
Cavities sistance by attracting and activating both lym-
Types of disease phocytes and macrophages with little, if any,
Small discrete tubercles of hematogenous origin; necrosis.The amount of tissue damage (or ben-
focally localized or scattered diffusely throughout efit) is determined not only by the number of
both lungs (miliary tuberculosis) bacilli, but also by the amount of sensitivity that
Liquefied caseous lesions with cavity formation and the host develops to the tuberculin-like products.
bronchogenic spread Details on the clinical types of human tuber-
Progressive, locally destructive lesions culosis are published in references 6 and 7.
a
Modified from Rich (5). Details on its various pathological manifestations
are published in references 3 through 5, 8, and
Table 1 and Fig. 1 list the types of lesions and 9 and are succinctly summarized in reference 10.
types of disease found in patients with pulmonary
tuberculosis.Any combination of lesion type and ESTABLISHMENT
disease type can exist in different parts of the OF THE INFECTION
lungs of the same patient, depending on the local An inhaled particle containing more than 3
concentration of bacillary products and the host’s tubercle bacilli is too large to reach the alveo-
DTH and CMI responses to these products.Large lar spaces. It impinges on the walls of the

FIGURE 1 Development, arrest, or progression of human pulmonary tuberculosis. Once a tiny


(0.5 to 1.0 mm) pulmonary lesion is established, the person becomes tuberculin positive.The tiny
lesion is usually arrested, but in about 5% of cases, it progresses to form a larger caseous lesion.
Such a lesion may (i) stabilize with bacillary dormancy, (ii) progress with or without hematoge-
nous spread of the disease, or (iii) liquefy and produce a cavity with or without bronchial spread
of the disease.Any stabilized lesion may reactivate years later if liquefaction and cavity formation
occur with extracellular bacillary growth. Antimicrobials can arrest the disease at any stage,
unless the bacillus becomes drug resistant.
36 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

bronchial tree, moves toward the pharynx on the not, develop depending, respectively, on
mucociliary escalator, and is subsequently swal- the capacity of the bacillus for intracellu-
lowed.The bronchial tree and alimentary canal lar growth and the capacity of the alveo-
are rather resistant to low numbers of tubercle lar macrophage to inhibit such growth. A
bacilli and do not usually produce progressive weak bacillus in a strong alveolar macrophage
lesions. will never cause a primary lesion.A strong bacil-
Small inhaled particles, containing up to 3 vir- lus in a weak alveolar macrophage will always
ulent tubercle bacilli, can, however, remain sus- cause such a lesion.
pended in the airstream and reach the alveolar Within an alveolar macrophage, the bacillus
spaces (11).About one-third of such particles do may be killed immediately, it may multiply, or it
so. (The remaining two-thirds impinge on the may remain dormant for various periods of
walls of the bronchial tree.) In the alveolar time. If it multiplies, its progeny will eventually
spaces, the bacillary particle is usually ingested kill the original alveolar macrophage, which
by an alveolar macrophage (12, 13). Sometimes then releases the bacilli into the alveolar spaces.
it is exhaled from the alveolus, and sometimes Tubercle bacilli are highly chemotactic and
it drains to lymphoid tissues, where it is even- attract to the site other alveolar macrophages, as
tually destroyed. well as many monocytes/macrophages from the
The bacilli in these inhaled particles may be bloodstream. The newly arrived blood-borne
healthy and genetically and phenotypically vir- macrophages ingest the bacilli but are initially
ulent, or they may be injured, weakened, and less incapable of limiting their intracellular growth.
virulent. A tuberculous lesion will, or will (They must first be activated, whereas most

FIGURE 2 A 10-day lesion, produced in a rabbit by the intravenous injec-


tion of BCG.Alveolar macrophages, staining darkly for -galactosidase (our
marker for macrophage activation), have accumulated in the surrounding alve-
olar area (rather far from the bacilli in the small caseous center).Around the
caseous center are viable, young, -galactosidase-negative macrophages from
the bloodstream, which control the fate of the tuberculous lesion.Within the
caseous center are disintegrated -galactosidase-negative macrophages and
more than 10 faintly stained tubercle bacilli (not easily seen in this photo-
graph). Stained with 5-bromo-4-chloro-3-indolyl--D-galactoside, carbol-
fuchsin, and hematoxylin. Magnification, ⫻330. Reprinted with permission
from reference 33.
3. TYPES OF HUMAN PULMONARY TUBERCULOSIS 䡵 37

alveolar macrophages are at least partly acti- ery of the lesion, far from the more centrally
vated.) The developing lesion soon becomes located bacilli (Fig. 2). However, the alveolar
almost entirely composed of blood-borne macrophages may prevent the development of
macrophages (Fig. 2), many of which contain secondary lesions when small numbers of bacilli
multiplying bacilli. are discharged into the airways from a tubercu-
Therefore, alveolar macrophages determine lous cavity.
whether or not a tuberculous lesion becomes Genetic resistance seems to play a major role
established, but have little effect on its subse- in determining whether an inhaled bacillary
quent course.They usually remain at the periph- particle establishes a primary lesion. Lurie’s

FIGURE 3 Left lung of a 57-


year-old woman showing an arrested
Ghon complex. A calcified arrested
primary tuberculous lesion is present
subpleurally in the lower lobe, and an
inactive caseous lymph node is pre-
sent at the hilus. The pulmonary
lesion was probably acquired dur-
ing childhood. From the collection
of the late professors A. R. Rich and
W. G. MacCallum, Department of
Pathology, Johns Hopkins School of
Medicine, Baltimore, Md.
38 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

genetically resistant rabbits had alveolar capable of establishing another lesion) usually
macrophages that were more adept than those occur weeks after the initial lesion has begun. By
of his susceptible rabbits at destroying inhaled that time, the host has acquired sufficient immu-
tubercle bacilli or inhibiting their growth (11, nity to prevent the new lesion from reaching
14, 15). In other words, when compared with grossly visible size (11).
alveolar macrophages of the susceptible rabbits,
the alveolar macrophages of the resistant rabbits ENCAPSULATED CASEOUS
were activated to a greater degree. (Such OR CALCIFIED NODULES
macrophages are activated nonspecifically by If a primary lesion reaches 4 to 12 mm in diam-
the ingestion of dust, molds, bacteria, and other eter, it may be detected in a chest X-ray. In most
inhaled particles.) In established tuberculous individuals, a lesion of this size is arrested and
lesions, the macrophages of the resistant rabbits encapsulated by fibrous tissue. Its caseous center
were also activated to a greater degree (11) by may remain caseous, or it may liquefy, calcify, and
antigen-specific lymphocytes and their cytokines occasionally ossify. During calcification, fine,
(see chapters 2 and 14). dustlike particles or more solid masses of calcium
Humans resemble Lurie’s resistant rabbits carbonate and calcium phosphate are deposited
(infected with human-type tubercle bacilli) in within caseous material, making the lesion more
that their macrophages can usually be activated visible in the chest X-ray. Such an encapsulated
immunologically to arrest the development of nodule usually remains inactive for the life of the
tuberculous lesions. Therefore, human pul- patient (Fig. 3). See the last section of this chap-
monary alveolar macrophages should also be ter for the properties and fate of lesions smaller
able to be activated nonspecifically to destroy or than 4 mm in diameter.
inhibit inhaled tubercle bacilli. Roentgenographically, a diffuse distribution
of calcium deposits is characteristic of infec-
THE EARLY PRIMARY LESION tious granulomas, such as those caused by tuber-
The earliest clinical evidence of a tuberculous culosis, histoplasmosis, and coccidioidomyco-
lesion somewhere in the body is a positive sis, whereas eccentric calcification is common in
tuberculin skin test.The test becomes positive malignancies. In addition, the edges of infectious
when the tubercle bacilli in the early lesion granulomas look smooth, whereas the edges of
have multiplied sufficiently in the primary lesion malignant lesions usually look irregular.
to produce a sensitizing antigenic load; in
humans this is usually 1 to 2 months after the PROLIFERATIVE AND EXUDATIVE
bacilli are inhaled. At that time, the lesion is a TYPES OF PULMONARY LESIONS
small tubercle with a caseous center.This cen- Proliferative lesions (Fig. 4 and 5), sometimes
ter is composed of incompletely autolyzed called “hard” tubercles, are more common when
macrophages and pulmonary tissue, bacillary small quantities of bacilli and their tuberculin-
breakdown products, and dead and live (often like products are present in a host with high re-
dormant) intact bacilli. Surrounding it are the sistance to tuberculosis. The cellular infiltrate
blood-borne macrophages that control the sub- consists of macrophages, lymphocytes, plasma
sequent fate of the lesion (Fig. 2). cells, and fibroblasts. Mature epithelioid cells
Usually, only one primary lesion is found in are common, and Langhan’s giant cells are some-
humans, not multiple primary lesions. This is times present. Connective tissue fibers, inter-
because (i) only an occasional unit of 1 to 3 vir- spersed among the cells, contribute to the com-
ulent bacilli is inhaled at one time, (ii) an esti- pact nature of this type of lesion. Bacillary
mate of 20 to 200 bacillary units must be inhaled multiplication is minimal because of the good
to establish the infection (a one-time exposure defense reaction produced by the host.
of humans during a long airplane flight is In humans,the classic orientation (i.e.,a caseous
described in references 16 and 17), and (iii) center surrounded by viable tuberculous granu-
subsequent inhalations of bacillary units (that are lation tissue containing epithelioid cells) is com-
3. TYPES OF HUMAN PULMONARY TUBERCULOSIS 䡵 39

FIGURE 4 A proliferative type of miliary tubercle in the liver of an 8-month-old male


infant. Parts of four Langhans’ giant cells are seen. Stained with hematoxylin and eosin. Mag-
nification, ⫻130. From the collection of the late professors A. R. Rich and W. G. MacCallum,
Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Md.

mon in young lesions, such as those in miliary 7). Caseation frequently occurs. Exudative lesions
tuberculosis. In older lesions, this classic orienta- may resolve almost completely, leaving only
tion is frequently indiscernible;instead,epithelioid microscopic residues, but, in the absence of
and giant cells are intermixed with caseous ma- antimicrobial agents, they usually progress.
terial and fibrous tissue. Proliferative lesions may Lesions exhibiting both proliferative and
become chronic, with some areas undergoing exudative features (Fig. 8) are more common
caseous necrosis and other areas undergoing fibro- than those of one type alone. Also, as men-
sis. Such lesions may eventually progress and give tioned above, proliferative lesions may occur in
rise to satellite tubercles that coalesce, or they one part of the lung, and exudative lesions may
may stabilize with encapsulation by collagenous concurrently occur in another part, depending
connective tissue that is often hyalinized and on the concentration of bacilli and the tuber-
sometimes contains calcium deposits. culin-like components in each place.The lesions
Exudative lesions (Fig. 6 and 7) are more com- may be limited or extensive in scope and some-
mon when the host is highly sensitive to tuber- times change from one predominant form to the
culin, especially when large quantities of bacilli other in response to shifts in the delicate balance
and their tuberculin-like products are present at between host and bacillus. The most striking
the local site. Mononuclear cells and some gran- change in this balance occurs when tubercle
ulocytes accumulate in the alveolar spaces as a bacilli proliferate luxuriantly and extracellularly
rather loose exudate containing variable amounts during liquefaction and cavity formation (see
of fibrin. In this exudate, the bacilli multiply “Liquefied Caseous Lesions and Cavities and
readily, especially in a host with weak defenses. Bronchogenic Spread” below). Only then,
These lesions vary from small, loose “soft” tuber- because of such large quantities of bacilli and
cles (which are frequently confluent) to small or their products, does an exudative response occur
large areas of tuberculous pneumonia (Fig. 6 and in a relatively resistant host.
40 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

FIGURE 5 A well-encapsulated fibrocaseous pulmonary lesion formed from confluent tuber-


cles in a 5-month-old infant dying of tuberculous meningitis.The central area of necrosis is sur-
rounded by smaller satellite necrotic areas. One Langhans’ giant cell is visible.This was a small
proliferative tubercle that exacerbated as the patient became terminally ill. Stained with hema-
toxylin and eosin. Magnification, ⫻123. From the collection of the late professors A. R. Rich and
W. G. MacCallum, Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Md.

SMALL, DISCRETE TUBERCLES the lesion resolves. Occasionally, the bacilli are
OF HEMATOGENOUS ORIGIN not destroyed, and one or more grossly visible
AND MILIARY TUBERCULOSIS tubercles form in the lungs, kidney, spleen, liver,
Most tuberculous lesions, particularly those that bone, testes, ovary, or brain. In the brain, a semi-
are progressing, shed at least a few bacilli into the solid or liquefied caseous tubercle near the sub-
bloodstream from time to time. Pulmonary arachnoid or ventricular space may rupture and
lesions shed bacilli into local capillaries and discharge its contents into the cerebrospinal
venules. Lesions in the hilar lymph nodes shed fluid, causing tuberculous meningitis.
bacilli into the efferent lymphatics, which trans- Miliary tuberculosis occurs when a massive
port these bacilli into the bloodstream. Caseous dose of tubercle bacilli is discharged from a
lesions in a hilar lymph node may occasionally caseous or liquefied focus into the bloodstream
erode a blood vessel and shed bacilli directly into and the resistance of the host is inadequate, as in
the bloodstream. Such lesions may also erode an early infancy, in old age, and in immunologically
adjacent part of the bronchial tree and shed depressed persons of any age. In this type of
bacilli into the airways. tuberculosis, many small tubercles of uniform size
Wherever the bacilli are deposited, they are and hematogenous origin occur simultaneously
ingested by macrophages and often form a in the lungs (Fig. 9) and/or in the liver, spleen,
microscopic tubercle. Usually, the macrophages or kidneys.A miliary tubercle is 2 to 4 mm, the
are rapidly activated, the bacilli are destroyed, and size of a millet seed (hence the name “miliary”).
3. TYPES OF HUMAN PULMONARY TUBERCULOSIS 䡵 41

FIGURE 6 An exudative type of tuberculous lesion in the lung of a 47-year-old man.


Depicted is an area of tuberculous pneumonia.A necrotic exudate fills the alveolar spaces, and
the alveolar walls are thickened by infiltrating cells. Stained with hematoxylin and eosin. Mag-
nification, ⫻266.

Tubercle bacilli are distributed to extrapul- heart.The discharging lesions may even be in the
monary organs after entering the left side of the walls of pulmonary veins themselves. Relatively
heart. These bacilli usually originate from few bacilli pass through the peripheral capillary
caseous pulmonary lesions that discharge bacilli beds and return to the lungs.
into pulmonary veins carrying oxygenated Tubercle bacilli are usually distributed into
blood from the lungs to the left side of the both lungs after entering the right side of the

FIGURE 7 An exudative lesion


similar to that shown in Fig. 6 but
stained for tubercle bacilli, some of
which may be discerned in this pho-
tograph (arrows). Stained with Ziehl-
Neelsen, counterstained with meth-
ylene blue. Magnification, ⫻450.
From the collection of the late pro-
fessors A. R. Rich and W. G. Mac-
Callum, Department of Pathology,
Johns Hopkins School of Medicine,
Baltimore, Md.
42 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

FIGURE 8 Part of a typical pulmonary tubercle that shows both proliferative and exudative
features and a Langhans’ giant cell. It was from a 12-month-old infant with miliary tuberculo-
sis. Stained with hematoxylin and eosin. Magnification, ⫻300.
Langhans’ giant cells form when macrophages surrounding bits of caseum fuse to each
other.The nuclei of Langhans’ giant cells are in the periphery and may continue to divide. In
tuberculous lesions, Langhans’ giant cells are a sign of chronicity. From the collection of the late
professors A. R. Rich and W. G. MacCallum, Department of Pathology, Johns Hopkins School
of Medicine, Baltimore, Md.

heart. These bacilli most often originate from and occasional giant cells, i.e., the proliferative
caseous hilar lymph nodes. The hilar lymph type of lesion with or without caseous centers
nodes drain the bacilli (via efferent lymphatics) (Fig. 4 and 5), or (ii) a loosely formed exudative
into the great systemic veins, which carry them type, or “soft” tubercle (Fig. 6). (Both are
(via the right side of the heart) to pulmonary described in the preceding section.) The exuda-
arteries carrying nonoxygenated blood to the tive type contains more bacilli (Fig. 7), which
lungs. Less frequently, endogenous bacilli reach continue to multiply, at least for a while. This
the lungs from tuberculous lesions elsewhere in type usually undergoes early and complete
the body, because such lesions also release bacilli caseation and progresses rapidly. Tubercles with
into the venous side of the circulation. Miliary mixed hard and soft characteristics are com-
tubercles will occur in only one lung if a tuber- mon (Fig. 8).
culous lesion (in the pulmonary hilus) discharges
bacilli directly into a branch of the main pul- LIQUEFIED CASEOUS LESIONS AND
monary artery. CAVITIES AND BRONCHOGENIC
Miliary tubercles may be of two types, SPREAD
depending on the resistance of the host: (i) a The liquefaction of caseous foci perpetuates pul-
compact “hard” tubercle with epithelioid cells monary tuberculosis in the human population,
3. TYPES OF HUMAN PULMONARY TUBERCULOSIS 䡵 43

FIGURE 9 Miliary tuberculosis of the lung in a 19-year-old man. Caseous hilar lymph nodes
are present. From caseous plaques (not shown) in the branches of the pulmonary veins, tubercle
bacilli seeded the general circulation and were carried to other organs in the body. From the col-
lection of the late professors A. R. Rich and W. G. MacCallum, Department of Pathology, Johns
Hopkins School of Medicine, Baltimore, Md.

because it results in cavity formation. From the large numbers of bacilli and their antigenic
cavity, the bacilli can spread via the bronchial components, even persons with high degrees of
tree to other parts of the lung and to the outside acquired resistance to tuberculosis often may
environment (18). not be able to stop progression of the disease.
Before a caseous focus liquefies, tubercle (See chapter 4 for a discussion of the causes of
bacilli multiply primarily intracellularly within liquefaction and cavity formation.)
poorly activated macrophages, which readily Among the large numbers of bacilli within a
ingest every bacillus they encounter (12, 13).The liquefied or cavitary lesion, mutants may be
extracellular bacilli in solid caseous tissue rarely present that are resistant to one or more of the
multiply. However, after a caseous focus lique- antimicrobial agents. For this reason, tuberculosis
fies (Fig. 10), the extracellular bacilli may mul- is usually treated with multiple antimicrobials
tiply tremendously (Fig. 11), especially after a given together over many months.
cavity forms because of the additional oxygen A cavity is formed when a caseous focus rup-
provided by direct contact with the air in the tures through the wall of a nearby bronchus
bronchial tree (Fig. 12).When faced with such and discharges its contents into the air passages
44 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

FIGURE 10 Early liquefaction in a small tubercle from a 12-month-old infant who died of
miliary tuberculosis.The lesion is partly surrounded by fibrous tissue. Stained with hematoxylin
and eosin. Magnification, ⫻108. From the collection of the late professors A. R. Rich and W. G.
MacCallum, Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Md.

(Fig. 13 and 14).The walls of most cavities con- Erosion of incompletely thrombosed vessels
sist of an external zone of collagen (the cavity’s in the intermediate zone leads to hemorrhage
capsule, which occasionally is hyalinized) and a into the wall of the cavity (Fig. 15).There, blood
caseous (often liquefying) internal zone, where may pool and give rise to some hemoptysis.
the high oxygen content from the ambient air Massive hemoptysis, which is sometimes fatal, is
nurtures the growth of bacilli (Fig. 12). By usually due to the leakage or rupture of a fully
coughing, the patient aerosolizes this infectious patent blood vessel located in the wall of the
material, disseminating bacilli to other parts of cavity or traversing its lumen.
the lung and to the outside environment. When a liquefied caseous lesion in the lung or
Between the external and internal zones of the hilar lymph node discharges its contents into the
cavity wall is an intermediate zone of granula- air passages, the resulting lesions range from small
tion tissue rich in capillaries, granulocytes, pneumonic foci, to larger areas of pneumonia, to
macrophages, lymphocytes, and fibroblasts. At complete lobar pneumonia.The extent of the dis-
times, it contains typical tubercles. The three ease is determined by the amount of liquefied
zones are of variable thickness and not clearly caseum aspirated into the bronchial tree, as well
demarcated from one another. With newly as by the number of (live and dead) bacilli and the
formed cavities, the internal caseous zone is quantity of tuberculin-like products that this liq-
thickest.With older, still-active cavities, the exter- uefied caseum contains.
nal capsule is thickest.Around the capsule, usu- Local deposits containing small quantities of
ally between the pleura and the cavity, an area of bacilli and their components commonly cause
atelectasis is often present. This atelectatic area scattered compact proliferative-type tubercles
may prevent perforation into the pleural spaces. or (more rarely) confluent proliferative tubercles
3. TYPES OF HUMAN PULMONARY TUBERCULOSIS 䡵 45

FIGURE 11 A rabbit pulmonary tuberculous lesion showing tubercle bacilli that had grown
profusely in the liquefied caseum.This lesion was probably beginning to cavitate, because some
of the spaces in the liquefied caseum are larger than usual. Such profuse growth occurs only in
some of the lesions with liquefied centers, presumably where the composition of the liquefied
caseum is most favorable and/or the adaptation of the bacillus to extracellular growth is most
complete. Similar bacillary growth has been found in many human cavitary lesions. Stained with
carbol-fuchsin, counterstained with methylene blue. Magnification, ⫻600. Reprinted with per-
mission from reference 34.

with small foci of encapsulated caseous pneu- ity. Fibrosis is often incomplete, and variable
monia (Fig. 16). However, local deposits con- amounts of caseous and fibrocaseous material
taining large quantities of bacilli and their com- may remain within the fibrotic lesion. Calcium
ponents cause exudative-type lesions that soon deposition is common, and occasionally ossifi-
caseate, producing small or large areas of tuber- cation occurs.A “healed” cavity, like a “healed”
culous pneumonia (Fig. 6 and 7). caseous focus, is seldom completely free of
Whether there is healing or progression tubercle bacilli, which may persist for years,
depends on the extent of the disease, as well as often in a dormant state.
the number of bacilli and their rate of multipli- With effective antimicrobial therapy, “open
cation. Small scattered foci readily heal or are healing” of a cavity may occur. Inflammation
encapsulated, but areas of tuberculous pneu- decreases after the antimicrobial agents have
monia may progress into adjacent alveoli and eliminated most of the bacilli, and the necrotic
bronchioles. contents drain through a still-patent bron-
Cavitary tuberculous lesions may never heal chocavitary junction. Such drainage is facili-
during the life of the patient.They may enlarge tated if the cavity is located in an upper lobe (the
or shrink or remain stable. Spontaneous healing usual place). Metaplastic bronchial epithelium
results from a gradual collapse of the cavity, (squamous with no cilia) may eventually line the
progressive fibrosis from without, and/or cavity.The open-healed cavity may persist as one
obstruction of the bronchocavitary junction with a thick or thin fibrotic wall, or as an
followed by absorption of the air within the cav- emphysematous bleb or bulla.
46 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

FIGURE 12 The wall of a pulmonary cavity in a rabbit showing tubercle bacilli that had
grown profusely in the liquefied caseum.The bacilli are more numerous near the lumen of
the cavity (on the left), presumably because the oxygen tension there is highest. Such profuse
growth occurs only in the walls of some, not all, cavities. Stained with carbol-fuchsin, coun-
terstained with methylene blue. Magnification, ⫻250. Reprinted with permission from ref-
erence 34.

PROGRESSIVE, LOCALLY host resistance is low. Such lesions grow periph-


DESTRUCTIVE LESIONS erally as a loose exudate extending into nearby
Tuberculous lesions may progress slowly or alveolar spaces. The exudate (which soon
rapidly, depending on the number of extracel- becomes necrotic) contains young macrophages,
lular and intracellular bacteria, and how easily granulocytes, fibrin, erythrocytes, and serum
the host can destroy them. Slowly progressing protein. This type of tuberculosis commonly
caseous or cavitary foci are the most common occurs in young children. It is rare in adults,
type of progressive tuberculosis in adults (Fig. except when they are immunosuppressed, or
13).These foci have compact walls that contain when they aspirate large numbers of bacilli from
epithelioid and giant cells, and show consider- a cavity. Rapidly formed cavities may occa-
able reparative fibrosis (Fig. 17). Progression is sionally occur (Fig. 19).
intermittent.The advancing edge of the lesion
contains fresh tubercles, or fresh or organizing ADVANCED FIBROCASEOUS
pneumonia in the adjacent alveolar spaces (Fig. TUBERCULOSIS
14). Encapsulated caseous foci and tubercles, The most common form of advanced tubercu-
embedded in fibrous tissue, are prominent; hence losis seen in adults is the fibrocaseous type,
the term fibrocaseous (Fig. 5). Satellite lesions are which may progress until death. In this form of
frequent, and cavitation and extension via the the disease, different lobes of the lung may con-
bronchial passages sometimes occur. tain confluent cavities, confluent caseous foci,
Rapidly progressing caseous foci with incon- areas of caseous pneumonia, or combinations of
spicuous fibrosis (Fig. 18) are often present when these conditions (Fig. 20 and 21). Because of the
3. TYPES OF HUMAN PULMONARY TUBERCULOSIS 䡵 47

FIGURE 13 Bilateral tuberculous cavities in the upper lobes in a 39-year-old diabetic woman.
Caseous areas surround the cavities. Although infected, the hilar lymph nodes are not markedly
enlarged. In the lung on the right, an applicator stick marks the communication between the cav-
ity and the bronchus. From the collection of the late professors A. R. Rich and W. G. MacCallum,
Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Md.

large numbers of bacilli that ascend the bronchial childhood tuberculosis, in which large caseous
tree and are subsequently swallowed, lesions hilar nodes often play a major role in the pro-
may occur in the bronchi, trachea, larynx, and gression of the disease, usually by seeding bacilli
intestinal tract—all of which are rather resistant into the bloodstream via efferent lymphatics.
to small numbers of bacilli.The patient whose
lungs are shown in Fig. 20 had lesions in each TUBERCULOUS PNEUMONIA
of these locations. Figure 22 shows an example AND PLEURISY
of tuberculous laryngitis, which, however, rarely Tuberculous pneumonia is an exudative response
occurs under effective chemotherapy. Occa- in the alveolar spaces caused by the presence of
sionally, an infected bronchus may undergo cica- numerous live and dead tubercle bacilli and
tricial stenosis and become obstructed, produc- their antigenic products. Small or extensive areas
ing atelectasis of the lung distal to that site. of pneumonia commonly occur when a lique-
Despite the extensive fibrocaseous pulmonary fied caseous focus in the lung (or in a hilar
disease, the hilar nodes of adults usually show lymph node) discharges its contents into the
minimal involvement. This is in contrast to airways (Fig. 6, 19, 20, 21, and 23). Since the
48 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

FIGURE 14 An apical cavity of moderate size in the left lung of an adult. Caseous pulmonary
consolidation of pneumonic origin surrounds the cavity. Several caseous foci are also present in
the other lung.The hilar lymph nodes contain a few caseous areas but are only slightly enlarged.
From the collection of the late professors A. R. Rich and W. G. MacCallum, Department of Pathol-
ogy, Johns Hopkins School of Medicine, Baltimore, Md.

caseous material is impregnated with tubercu- especially if only small numbers of viable bacilli
loprotein and other bacillary components, it are present and the lung structure is preserved.
causes a pneumonic exudate, even if the num- Pleurisy results when subpleural caseous lesions
ber of intact bacilli is small.This exudate under- in the lung (or in the hilar lymph nodes) leak or
goes caseous necrosis if high amounts of tuber- rupture, discharging bacilli and their compo-
culin-like products are present. The initial nents into the pleural cavity (Fig. 20). (Similarly,
exudate contains mononuclear cells, granulo- tuberculous pericarditis can develop from a rup-
cytes, and a coagulum of precipitated protein and ture of such a lesion into the pericardial sac.)
fibrin. In time, epithelioid cells, lymphocytes, Clinically, the pleurisy is manifested in many
plasma cells, fibroblasts, and, occasionally, giant forms: mild, uncomplicated roughenings of the
cells appear. pleural surface, large or small numbers of miliary
The confluence of progressing exudative tubercles on the pleural surface,or serous effusions
lesions results in consolidation of small or large from large or numerous surface lesions. In more
segments of the pulmonary lobe.At one end of than half of the patients with tuberculous pleurisy,
the spectrum, the pneumonia leads to death.At the serous effusion is bacteriologically sterile,
the other end of the spectrum, the pneumonia because it is only a response to tuberculin-like
undergoes resolution with minimal scarring, products released into the pleural cavity.
3. TYPES OF HUMAN PULMONARY TUBERCULOSIS 䡵 49

FIGURE 15 A tissue section of a rabbit pulmonary cavity wall. On the left are disintegrat-
ing mature epithelioid cells (see Fig. 15 in chapter 4). On the right is a small blood vessel extend-
ing into the cavity’s lumen. Such exposed vessels are the source of blood in the sputum. In
humans, when a larger blood vessel is similarly exposed in a cavity and ruptures, massive
hemoptysis and sometimes fatal hemorrhage may occur. Glycol methacrylate-embedded
tissue section stained with Giemsa. Magnification, ⫻400. Reprinted with permission from
reference 35.

Tuberculous empyema was a serious compli- sometimes calcification. Depending on the


cation before the antimicrobial era. It consisted amount of exudate in the pleural space, a large
of a purulent exudate in the pleural cavity, usu- or small portion of the lung may collapse. Orga-
ally surrounded by a fibrocaseous capsule, and nization of the exudate and increasing fibrosis
the exudate was frequently superinfected with tend to maintain this collapse. References 6 and
pyogenic bacteria. 7 provide more details on tuberculous pleurisy.
The type of pleural disease is determined by
(i) the number of live and dead bacilli, (ii) the CHILDHOOD TUBERCULOSIS:
quantity of their tuberculin-like products in the LYMPHATIC SPREAD AND THE
caseous material, (iii) the level of tuberculin GHON COMPLEX
sensitivity of the host, (iv) the friction of the Tuberculosis in children differs from tuberculo-
roughened visceral pleura against the parietal sis in adults in several respects: (i) young children,
pleura, (v) the number and types of cells in the especially infants, usually have lower native and
exudate, (vi) the inherent ability of the host to acquired resistance to this disease; (ii) their hilar
localize and fibrose new sites of tuberculous lymph nodes more commonly become enlarged
infection, and (vii) the presence or absence of and caseous (Fig. 18, 23, and 24); (iii) blood-
superinfection with other bacteria. borne dissemination occurs more readily (Fig. 18;
Tuberculous pleurisy may resolve completely also see Fig. 9) but is still an exception rather than
or may heal with fibrosis, focal adhesions, and the rule; (iv) calcification of caseous foci is more
50 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

FIGURE 16 A cavity in the base of


the upper lobe of the left lung of a 17-
month-old infant.The apical portion was
adherent to the chest wall and solidified
with confluent proliferative tubercles and
encapsulated caseous pneumonic foci.
From the collection of the late professors
A. R. Rich and W. G. MacCallum, Depart-
ment of Pathology, Johns Hopkins School
of Medicine, Baltimore, Md.

common; and (v) the primary pulmonary lesion stitutes the Ghon complex (Fig. 24), which was
in children is usually subpleural in well-ventilated named after Anton Ghon (1866–1936), a Prague
regions of the lung (Fig. 24), whereas such lesions pathologist.
in adults are usually subapical (see below). Sometimes, a tuberculous hilar node obstructs
Tubercle bacilli, either free or within imma- a bronchus by external compression or by exten-
ture macrophages (or granulocytes), spread via sion of the disease process into the bronchial wall.
the lymphatics from the primary lesion to the The obstruction causes segmental or lobar col-
hilar lymph nodes. There, in children (and in lapse and pneumonitis, which is called the right-
adults with low resistance), the bacilli frequently middle lobe syndrome, as it frequently occurs in
cause large caseous lesions.The combination of that lobe. Sometimes, such a node ruptures into
one or more enlarged caseous hilar nodes plus a bronchus (Fig. 18 and 23), causing distally focal
a tuberculous focus in the lung periphery con- or diffuse pneumonic lesions. Such lesions can be
3. TYPES OF HUMAN PULMONARY TUBERCULOSIS 䡵 51

FIGURE 17 A portion of a large cavity in the lung of a rabbit, 18 weeks after the inhala-
tion of about 5,000 virulent bovine-type tubercle bacilli.The liquefied caseum (below) is sur-
rounded by a thick fibrous capsule. In this capsule are many fibroblasts, as well as some
macrophages, lymphocytes, and plasma cells. At the right is an oval metaplastic alveolus next
to a small blood vessel. Glycol methacrylate-embedded tissue section stained with Giemsa. Mag-
nification, ⫻250. Reprinted with permission from reference 1.

minimal or extensive, depending on the con- in the body. However, the resistance of immuno-
centration of bacilli and their antigenic products competent tuberculous adults may be over-
and on the host’s reaction to them. Both situa- whelmed when a liquefied caseous focus rup-
tions occur mainly in infants, since older children tures and seeds large numbers of bacilli directly
and adults only rarely have extensive involvement into a bronchus or into a blood or lymphatic
of the hilar nodes. vessel of appreciable size.
Reinfection-type or adult-type tuberculosis
consists of a small to large pulmonary lesion that
ADULT TUBERCULOSIS:
ENDOGENOUS AND EXOGENOUS is not accompanied by any marked enlargement
REINFECTION AND SUBAPICAL of the hilar lymph nodes.The pulmonary lesion
LOCALIZATION is most often located in the subapical region. In
As a rule, tuberculosis progresses more slowly in this type of tuberculosis, the native and acquired
adults than it does in infants, because the resis- resistance of the host is sufficient to destroy or
tance of adults is usually higher. Secondary inhibit most of the bacilli that reach the hilar
endogenous tuberculous foci of lymphatic or nodes.Adult-type tuberculosis may occur as a pri-
hematogenous origin occur less often in adults, mary infection or reinfection in either adults or
because most tubercle bacilli are soon destroyed children if their resistance is high enough to
after they are carried in the lymph to the hilar restrict bacillary multiplication in the hilar nodes.
nodes or carried in the blood to sites elsewhere It may be caused by either endogenous or
52 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

FIGURE 18 Rapidly progressing disseminated tuberculosis in an 11-month-old infant. Both


hematogenous and bronchogenic spread from caseous hilar lymph nodes occurred in the lungs.
Most of the caseous foci are hematogenous in origin.At the hilus in the lung on the right is an
area of caseous pneumonia caused by the rupture of a tuberculous lymph node into a nearby
bronchus. From the collection of the late professors A. R. Rich and W. G. MacCallum, Depart-
ment of Pathology, Johns Hopkins School of Medicine, Baltimore, Md.

exogenous tubercle bacilli (see reference 11). In CAUSES OF SUBAPICAL


other words, adult-type tuberculosis may be LOCALIZATION
caused by a reactivation of a previously arrested Source of Tubercle Bacilli
disease or by a primary infection with virulent Exogenous bacilli, giving rise to adult-type
tubercle bacilli inhaled from the environment. tuberculosis, would be inhaled directly into the
The active subapical lesion frequently consists subapical pulmonary regions, while endoge-
of a localized area of bronchopneumonia con- nous bacilli would be carried to these regions by
taining macrophages, epithelioid cells, lympho- the bloodstream (see Fig. 13, 14, 20, and 21).The
cytes, giant cells, and fibroblasts.The lesion usu- endogenous tubercle bacilli (originating from
ally heals, and the resulting fibrosis may thicken the hilar nodes) could reach the apical region at
the overlying pleura. Sometimes, a cavity forms the time of the primary infection and remain
either at the time of the initial subapical infec- dormant there until reactivation occurs (see ref-
tion or, more commonly, years later if reactiva- erence 19).Alternatively, these bacilli could have
tion of a formerly dormant subapical lesion been dormant in the hilar lymph nodes and
occurs.Then, the disease may progress into one could have seeded the subapical region at the
of the more serious types described above. time of reactivation of the disease. Scars in the
3. TYPES OF HUMAN PULMONARY TUBERCULOSIS 䡵 53

FIGURE 19 A large, rapidly formed


tuberculous cavity in the upper lobe.
Caseous pneumonia is present throughout
the rest of the lung. Fibrosis is minimal.
The hilar node is markedly enlarged and
mostly caseous. From the collection of
the late professors A. R. Rich and W. G.
MacCallum, Department of Pathology,
Johns Hopkins School of Medicine, Bal-
timore, Md.

apical region, the so-called Simon foci (20), The lower regions of the lung are ventilated
could have been caused by either exogenous or more than the upper regions mainly because of
endogenous tubercle bacilli. the descent of the diaphragm during inspiration.
Also, when the person is in the upright position,
Physiology: Apical versus Basal the lower region has a much greater blood flow
Pulmonary Regions (i.e., perfusion) within its vasculature.A greater
In humans in the upright position, the apical perfusion occurs there because the higher
region of the lung differs from the basal region hydrostatic pressure both dilates the lower vas-
in the following manner. culature and increases the flow of blood within
Apical alveoli are more distended owing to this vasculature (21).
less compression from the weight of the rest of In the lower regions of the lung, more O2 is
the lung, which is mostly due to the hydrosta- absorbed and more CO2 is released than in the
tic pressure from the blood it contains. upper regions, because both the ventilation of
54 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

FIGURE 20 Advanced fibrocaseous tuberculosis in an 11-year-old girl, who died before the
advent of the antimicrobial era.The bilateral cavities in the upper lobes have caused adjacent
areas of caseous bronchopneumonia. On the right is fibrous tissue from pleural adhesions.The
hilar lymph nodes contain several caseous foci. From the collection of the late professors
A. R. Rich and W. G. MacCallum, Department of Pathology, Johns Hopkins School of Med-
icine, Baltimore, Md.

air and the perfusion of blood are increased. In Effects of the Subapical Localization
the upper regions, therefore, less O2 is absorbed on the Establishment of Tuberculous
and less CO2 is released. Lesions
The lymph flow from the apex is also In both the upper and lower alveoli, pulmonary
reduced, because with decreased blood entering, alveolar macrophages are activated nonspecifi-
less lymph is produced. Lymph forms intersti- cally by dust and inhaled microorganisms. In
tially from fluids extravasated from the blood- humans (and rabbits), such activation enables
stream and is moved into the terminal lym- the majority of alveolar macrophages to destroy
phatics by the pumping action of the lungs as inhaled virulent human-type tubercle bacilli or
they expand and contract.This pumping action inhibit their growth (see chapter 11). More
is greater in the lower, more ventilated part of inhaled particles enter the lower alveoli, because
the lungs. they receive a greater proportion of the tidal air
3. TYPES OF HUMAN PULMONARY TUBERCULOSIS 䡵 55

FIGURE 21 Caseous broncho-


pneumonia originating from a large
subapical cavity. From the collection
of the late professors A. R. Rich and
W. G. MacCallum, Department of
Pathology, Johns Hopkins School of
Medicine, Baltimore, Md.

than the apical alveoli receive (22).Therefore, in into clinically apparent disease. In early micro-
humans, the alveolar macrophage population scopic lesions, the alveolar walls are infiltrated
in the lower alveoli might (but not necessarily) with macrophages, dendritic cells, and lympho-
be more highly activated than those in the upper cytes from the bloodstream, and an exudate con-
alveoli. If so, the alveolar macrophages in the taining these cells enters the adjacent alveoli. In
lower alveoli would prevent initial establish- a person in the upright position,the apical regions
ment of tuberculous lesions more readily than are perfused with less blood than the basal pul-
those in the apical regions.This possibility, how- monary regions (because of gravity), their supply
ever, remains to be investigated. of O2 is limited (because of less ventilation), and
CO2 (from the cells in the lesion) is less rapidly
Effects of the Subapical Localization removed (see above).The reduced availability of
on the Progress of Tuberculous O2 should decrease the ability of the infiltrating
Lesions macrophages to produce reactive oxygen and
The apical location should have marked effects on nitrogen intermediates, which kill (or inhibit)
the progress of an established microscopic lesion intracellular tubercle bacilli, and the reduced
56 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

bacilli were collected in a Rosebury impinger


and were cultured on a modified Lowenstein’s
solid egg medium (11, 23). (The aerosol samples
contained bacillary units of mostly 1 to 3 tuber-
cle bacilli.) When low levels of CO2 were added
to the air in the electric incubator, many more
mycobacterial colonies developed on the culture
medium than when no CO2 was provided. Units
of 1 to 3 tubercle bacilli suspended in air are eas-
ily desiccated and need the most favorable con-
ditions to survive and multiply.The presence of
low levels of CO2 evidently enhances their abil-
ity to survive.
In basal pulmonary regions of an individual
in the upright position, more ventilation occurs
and more blood is perfused through the alveo-
lar capillaries (21), so the early tuberculous lesion
would have more O2 available, and less CO2
would accumulate. In the basal regions, the
increased perfusion of blood would supply
defense cells more rapidly, and the increased
respiratory excursion would increase the flow of
interstitial fluid into the lymphatics.Therefore,
tubercle bacilli (usually within macrophages and
FIGURE 22 Tuberculous laryngitis in a rabbit that dendritic cells) and their (secreted) antigens
had inhaled 300 virulent bovine-type tubercle bacilli 33 would be carried more rapidly to the draining
weeks previously.The lung of this rabbit had 14 lesions, lymph nodes, where acquired (adaptive) immu-
4 of which had formed cavities.The cavity in the right nity takes place (see chapters 5 and 6).Therefore,
lower lobe of the lung had apparently discharged so
many bacilli into the bronchial tree that the larynx when a tuberculous lesion is located in the basal
became infected.This laryngeal lesion partly obstructed region, a good immune response would develop
the airway. In humans, tuberculous laryngitis has been sooner and prevent an early tuberculous lesion
rare ever since antimicrobials for this disease became from reaching clinically apparent size.
available. Magnification, ⫻5.4. Reprinted with permis- Another possible explanation of why tuber-
sion from reference 35.
culous lesions of endogenous origin develop
apically is that small particles of caseous tissue
containing tubercle bacilli are preferentially dis-
removal of CO2 might possibly enable more tributed there by the bloodstream. Both tuber-
tubercle bacilli to survive intracellularly (see cle bacilli and the caseous tissue containing
below).Also, in the apical regions, the decreased them are rich in lipids and tend to float. Caseous
flow of lymph would carry fewer bacilli to the hilar lymph nodes (via efferent lymphatics)
draining lymph nodes and leave more bacilli would drain such bacilli into the great veins.
within the developing lesion. The immune When the person is upright, the bacilli in these
response begins in draining lymph nodes.There- veins enter the heart and then would probably
fore,the immune response to inhaled bacilli in the enter the upper branches of the pulmonary
apical regions would be less rapid (and therefore arteries, where they would be distributed to
less effective) than the immune response to the upper parts of the lungs. (Human infants and
inhaled bacilli in the basal regions. rabbits, however, spend a large part of the day in
Lurie (unpublished) found that small amounts the horizontal position, and their primary lung
of CO2 enhanced the in vitro growth of tuber- lesions are usually located in the middle and
cle bacilli. Aerosol samples of virulent tubercle lower lobes, rather than subapically.)
3. TYPES OF HUMAN PULMONARY TUBERCULOSIS 䡵 57

FIGURE 23 Extensive confluent


caseous pneumonia in a 13-month-old
infant, caused by the rupture of a large
liquefied caseous hilar lymph node into
an adjacent bronchus. From the collec-
tion of the late professors A. R. Rich and
W. G. MacCallum, Department of
Pathology, Johns Hopkins School of
Medicine, Baltimore, Md.

All of the factors listed above probably play patient’s lung and also other persons.The bacil-
a role in the subapical localization of clinically lary population that is tested for drug resistance
apparent tuberculous lesions in adults, but how in sputum samples almost always comes from a
they interact and the relative importance of cavity in the patient’s lungs. In fact, tubercle
each remain to be determined. bacilli in noncavitary lesions usually do not
develop drug resistance.
MULTIDRUG-RESISTANT Analyses of bacilli from cavitary lesions by
TUBERCLE BACILLI IS6110-based RFLP (restriction fragment
Multidrug-resistant tubercle bacilli usually arise length polymorphism) technology showed that
in bacillary populations that are multiplying different types of antimicrobial resistance could
within cavities (24).The bacilli then ascend the develop independently by mutations within
bronchial tree and may infect other parts of the different cavitary lesions located in the same
58 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

FIGURE 24 A progressive Ghon complex in the mid portion of a lung from a 6-year-old boy
who died of tuberculous meningitis.At the lower edge of the specimen is the primary subpleural
caseous lesion. Near the center is a markedly enlarged caseous hilar lymph node. Many tubercles
of hematogenous origin are also present. From the collection of the late professors A. R. Rich and
W. G. MacCallum, Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Md.

lungs (24). In other words, the initial infecting were arrested before drug resistance developed
mycobacterial strain that developed (or had) in the cavities.
drug resistance in the first cavity can acquire re- Tubercle bacilli in arrested caseous foci seem
sistance to additional drugs in subsequent cav- to be quite stable.The genome of Mycobacterium
ities arising in the same patient (24). Con- tuberculosis bacilli collected from patients during
versely, at least some arrested noncavitary lesions the 1990s was found by IS6110-based RFLP
in the lungs of such patients should contain technology to be identical to the genome of
drug-susceptible tubercle bacilli if such lesions freeze-dried M. tuberculosis bacilli collected from
3. TYPES OF HUMAN PULMONARY TUBERCULOSIS 䡵 59

these patients was refractory to antimicrobial


therapy.) In three patients, both the sputum and
the cavitary lesions contained numerous tuber-
cle bacilli. However, in two patients both the
sputum and the cavitary lesions contained no
visible or culturable bacilli. In these two patients,
the surface of their cavities was epithelialized, and
no solid or liquefied caseous necrosis was present.
The cavity walls of the three sputum-positive
patients had an inner liquefied area containing
macrophages and many tubercle bacilli, sur-
rounded in turn by an area of acellular caseous
necrosis with few, if any, visible bacilli, followed
by an outer granulomatous fibrotic area con-
taining epithelioid macrophages, Langhans’ giant
cells, and lymphocytes (24).
In the area containing lymphocytes and
macrophages,the mRNAs of interleukin-2,inter-
leukin-12, gamma interferon, and inducible nitric
oxide synthase were measured by quantitative
reverse transcription-PCR. These markers of
good acquired resistance to tubercle bacilli were
found in ample quantities, which provides direct
evidence in humans for what Lurie observed in
his resistant inbred rabbits (11), namely, that hosts
with cavitary tuberculosis usually have good CMI
FIGURE 25 Confluent, multidrug-resistant, tuber-
(acquired cellular resistance) but cannot control
culous pneumonia in a patient with AIDS. The lung
parenchyma was replaced by pale nodular zones of the large numbers of bacilli that grow extracel-
caseation necrosis. Reprinted with permission from lularly in the cavity lesions.
reference 1. Many of the tubercle bacilli on the luminal
surface of the cavity walls resided in macrophages
(24). These macrophages probably were aspi-
the same patients during the 1960s (25). This rated into the cavity during spells of coughing,
finding suggests that the patients in the 1990s because many macrophages enter the bronchial
had reactivation of dormant tuberculous lesions tree from the alveolar spaces. Macrophages could
acquired 30 years earlier. The persistence and not have entered the cavity by migrating through
dormancy in tubercle bacilli have been thor- the adjacent area of caseous necrosis.
oughly reviewed (26).
TUBERCULOSIS IN THE
RECENT ANALYSES OF HUMAN IMMUNOCOMPROMISED HOST
CAVITARY TUBERCULOUS LESIONS HIV is a major cause of the overall increased inci-
When the patient’s sputum contained many dence of tuberculosis in the world today (20, 27).
tubercle bacilli, many tubercle bacilli were also In 1 year, the incidence of active disease in HIV-
present extracellularly in the liquefied caseum infected tuberculin-positive persons is the same
of the cavity wall (see references 3 and 5).This as the incidence of active disease in immuno-
correlation was recently confirmed by Kaplan et competent tuberculin-positive persons during
al. (24) with surgically removed lung specimens their entire lifespan. In the immunocompro-
from five patients with cavitary lesions. (The mised patient, tuberculosis is most often due to
surgery was performed because the disease in a reactivation of a latent focus acquired many
60 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

FIGURE 26 A tissue section from the lung depicted in Fig. 25. Caseosuppurative necrosis
can be seen on the left, and poorly organized tuberculous granulation tissue is on the right.
Stained with hematoxylin and eosin. Magnification, ⫻150. Reprinted with permission from
reference 1.

years previously, but primary tuberculosis also In patients with HIV/AIDS, the histologic
occurs frequently—possibly because some pul- pattern of tuberculosis generally correlates with
monary alveolar macrophages are infected with the degree of immunosuppression. In pre-
HIV and therefore have a decreased ability to mortem biopsies, granuloma formation is more
destroy inhaled tubercle bacilli. In such patients, proliferative (and less exudative) than in
tuberculosis is usually more severe and more necropsy-derived tissues. As the number of
rapidly progressive than in immunocompetent peripheral CD4 lymphocytes decreases, the pat-
patients. Their mortality rates are higher, and tern of necrosis changes from caseous to caseo-
extrapulmonary involvement is more common suppurative, i.e., caseous necrosis containing
(20, 27). In HIV/AIDS patients, the disease of neutrophils and more undigested nuclear debris
reactivation often resembles childhood tuber- (Fig. 26). Also, the number of mycobacteria
culosis with hilar adenopathy, frequent lower within lesions increases (28). See reference 29 for
lobe involvement, and the absence of both cav- a complete overview of the types of tuberculo-
itation and extensive fibrosis (Fig. 25 and 26). sis found in HIV/AIDS patients.
In all immunosuppressed patients, the mor-
phology of tuberculous lesions reflects poor host INAPPARENT ARRESTED PRIMARY
resistance. For example, in corticosteroid-treated TUBERCULOUS LESIONS IN
patients (Fig. 27), tuberculous lesions show abun- HEALTHY HUMANS COMPARED
WITH RABBITS
dant caseation with little or no encapsulation or
granulomatous reaction (Fig. 28A). Large num- Inapparent Lesions in Humans
bers of bacteria are usually present within the Lindgren described a series of unselected necrop-
lesions (Fig. 28B). These changes are morpho- sies (made in Finland), among which were indi-
logically similar to those found experimentally in viduals who had been vaccinated with Mycobac-
corticosteroid-treated tuberculous animals (11). terium bovis BCG and individuals who had not
3. TYPES OF HUMAN PULMONARY TUBERCULOSIS 䡵 61

FIGURE 27 Progressive miliary


tuberculosis and tuberculous pneu-
monia in the left upper lobe of an
immunocompromised 42-year-old
woman receiving corticosteroids for
dermatomyositis.An infarct is present
in the lower lobe. Reprinted with
permission from reference 1.

(30–32).The cases were evaluated at a time when of the lesions detected were inactive and had
the majority of the Finnish population had been completely arrested for years. Only one of
become tuberculin positive because of inhaling the patients in the necropsied series died of
occasional virulent tubercle bacilli over many active tuberculosis.
years. In the BCG-vaccinated group, an average BCG vaccination did not prevent the estab-
of 11.2 years elapsed between the time when lishment of detectable primary pulmonary
BCG was given and when the necropsy was lesions caused by virulent human-type tuber-
performed (31). cle bacilli. However, the lesions in the BCG
These studies included X-ray examinations of group were smaller than those in the unvacci-
air-inflated lungs to detect calcified lesions nated control group, and the involvement of
(down to 0.5 mm in diameter), many of which the regional lymph nodes was substantially
were not identified by other methods.Almost all reduced (30–32). In fact, the nodes of the
62 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

FIGURE 28 (A) Tissue section of an exudative tuberculous lesion from the lung shown in
Fig. 27, showing extensive caseous necrosis without granuloma formation or Langhans’ giant cells.
Within the adjacent alveoli (on the right) is an exudate of fibrin and caseous debris. Stained with
hematoxylin and eosin. Magnification, ⫻170. (B) Numerous intracellular and extracellular acid-
fast bacilli found at the edge of a necrotic area in the lung from the same patient. Stained with
Ziehl-Neelsen, counterstained with methylene blue. Magnification, ⫻1,700. Reprinted with per-
mission from reference 1.

vaccinated group often showed no visible evi- The difference between rabbits and humans
dence of disease. in the development of visible pulmonary lesions
is at least in part due to the tissue-damaging
Comparison of Inapparent DTH reaction that these species develop.Tuber-
Lesions in Humans and Rabbits culous humans are about 100 times more sen-
Lindgren’s studies clearly demonstrated that sitive than rabbits to tuberculin. Therefore,
humans are more susceptible than rabbits to caseous necrosis and eventual calcification more
virulent human-type tubercle bacilli. In tuber- readily occur in humans.
culin-positive humans, each primary pulmonary
lesion developed to a size that is detectable at REFERENCES
necropsy. These lesions had a caseous center 1. Dannenberg,A. M., Jr., and J. F.Tomashefski,
and were often calcified (32). In tuberculin- Jr. 1998. Pathogenesis of pulmonary tuberculosis,
p. 2447–2471. In A. P. Fishman (ed.), Pulmonary
positive rabbits infected by aerosol, many micro- Diseases and Disorders, 3rd ed., vol. 2. McGraw-Hill
scopic lesions apparently regressed before they Book Co., New York, N.Y.
became visible (see chapter 11). 2. Dannenberg, A. M., Jr. 1999. Pathophysiology:
Humans and rabbits are both rather resistant basic aspects. I. Pathogenesis of tuberculosis. II.
to tuberculosis caused by human-type tubercle Immunology of tuberculosis, p. 17–47. In D. Schloss-
berg (ed.), Tuberculosis and Nontuberculous Mycobac-
bacilli, because 95% of the lesions in humans do terial Infections, 4th ed. The W. B. Saunders Co.,
not progress to clinically active disease, but Philadelphia, Pa.
remain inactive unless host resistance is lowered. 3. Canetti, G. 1955. The Tubercle Bacillus in the Pul-
Nonetheless, humans are slightly more suscep- monary Lesion of Man, p. 130. Springer Publishing
tible to tuberculosis than rabbits, because 5% of Company, Inc., New York, N.Y.
4. Poole, J. C. F., and H.W. Florey. 1970. Chronic
humans do not arrest the infection without inflammation and tuberculosis, p. 1183–1224. In
antimicrobial therapy, but almost all rabbits do H. W. Florey (ed.), General Pathology, 4th ed. The
so (11). W. B. Saunders Co., Philadelphia, Pa.
3. TYPES OF HUMAN PULMONARY TUBERCULOSIS 䡵 63

5. Rich,A. R. 1951. The Pathogenesis of Tuberculosis, 2nd 21. West, J. B. 2000. Respiratory Physiology.The Essen-
ed. Charles C Thomas Publisher, Springfield, Ill. tials, 6th ed., p. 50–59. Lippincott Williams &
6. Iseman, M. D. 2000. A Clinician’s Guide to Tuber- Wilkins, Philadelphia, Pa.
culosis. Lippincott Williams & Wilkins, Philadel- 22. Bennett,W. D., M. S. Messina, and G. C. Smal-
phia, Pa. done. 1985. Deposition (normalized for lung vol-
7. Hopewell, P. C., and R. M. Jasmer. 2005. ume) is generally greater in basal region of lung than
Overview of clinical tuberculosis, p. 15–31. In S.T. in apical region. Effect of exercise on deposition and
Cole, K. D. Eisenach, D. N. McMurray, and W. R. subsequent retention of inhaled particles. J. Appl.
Jacobs, Jr. (ed.), Tuberculosis and the Tubercle Bacillus. Physiol. 59:1046–1054.
ASM Press,Washington, D.C. 23. Lurie, M. B., A. G. Heppleston, S. Abramson,
8. Medlar, E. M. 1955.The behavior of pulmonary and I. B. Swartz. 1950. An evaluation of the
tuberculous lesions.A pathological study. Am. Rev. method of quantitative airborne infection and its use
Tuberc. 71:1–244. in the study of the pathogenesis of tuberculosis. Am.
9. Wolinsky, E. 1989. Tuberculosis, p. 465–519. In Rev.Tuberc. 61:765–797.
G. L. Baum and E.Wolinsky (ed.), Textbook of Pul- 24. Kaplan, G., F. A. Post, A. L. Moreira, H.Wain-
monary Diseases, 4th ed., vol. I. Little, Brown & Co., wright, B. N. Kreiswirth, M. Tanverdi, B.
Boston, Mass. Mathema, S.V. Ramaswamy, G.Walther, L. M.
10. Tomashefski, J. F., Jr. 1994, Tuberculosis and Steyn, C. E. Barry III, and L.-G. Bekker.
atypical mycobacterial infections, p. 451–463. In (2003) Mycobacterium tuberculosis growth at the cav-
M. J. Saldana (ed.), Pathology of Pulmonary Disease. ity surface: a microenvironment with failed immu-
Lippincott Williams & Wilkins, Philadelphia, Pa. nity. Infect. Immun. 71:7099–7108.
11. Lurie, M. B. 1964. Resistance to Tuberculosis: Exper- 25. Lillebaek,T.,A. Dirksen, E.Vynnycky, I. Baess,
imental Studies in Native and Acquired Defense Mech- V. O.Thomsen, and A. B. Andersen. 2003. Sta-
anisms. Harvard University Press, Cambridge, Mass. bility of DNA patterns and evidence of Mycobac-
12. Schlesinger, L. S. 1993. Macrophage phagocyto- terium tuberculosis reactivation occurring decades
sis of virulent but not attenuated strains of Mycobac- after initial infection. J. Infect. Dis. 188:1032–1039.
terium tuberculosis is mediated by mannose receptors
26. Zhang,Y. 2004. Persistent and dormant tubercle
in addition to complement receptors. J. Immunol.
bacilli and latent tuberculosis. Front. Biosci. 9:1136–
150:2920–2130.
1156.
13. Schlesinger, L. S., C. G. Bellinger-Kawahara,
N. R. Payne, and M. A. Horwitz. 1990. Phago- 27. Barnes, P. F., A. B. Bloch, P.T. Davidson, and
cytosis of Mycobacterium tuberculosis is mediated by D. E. Snider, Jr. 1991.Tuberculosis in patients with
human monocyte complement receptors and com- human immunodeficiency virus infection. N. Engl.
plement component C3. J. Immunol. 144:2771– J. Med. 324:1644–1650.
2780. 28. Hill, A. R., P. Somasundaram, S. Brustein, K.
14. Dannenberg, A. M., Jr. 1991. Delayed-type Vaidya, S. Powell, P. Li, and B. Suster. 1991.
hypersensitivity and cell-mediated immunity in the Disseminated tuberculosis in the acquired immu-
pathogenesis of tuberculosis. Immunol.Today 12:228– nodeficiency syndrome era. Am. Rev. Respir. Dis.
233. 144:1164–1170.
15. Dannenberg, A. M., Jr. 1993. Immunopatho- 29. Lucas, S., and A. M. Nelson. 1994. Pathogenesis
genesis of pulmonary tuberculosis. Hosp. Pract. of tuberculosis in human immunodeficiency virus-
28:33–40 (Office edition, 51–58). infected people, p. 503–513. In B. R. Bloom (ed.),
16. Kenyon, T. A., S. E. Valway, W. W. Ihle, I. M. Tuberculosis: Pathogenesis, Protection, and Control.
Onorato, and K. G. Castro. 1996.Transmission of ASM Press,Washington, D.C.
multidrug-resistant Mycobacterium tuberculosis during 30. Lindgren, I. 1961. Anatomical and roentgeno-
a long airplane flight. N. Engl. J. Med. 334:933–938. logic studies of tuberculosis infection in BCG-
17. Wenzel, R. P. 1996. Airline travel and infection. vaccinated and non-vaccinated subjects, with bio-
N. Engl. J. Med. 334:981–982. physical investigations of calcified foci. Acta Radiol.
18. Grosset, J. 2003. Mycobacterium tuberculosis in the 209(Suppl.):1–101.
extracellular compartment: an underestimated 31. Lindgren, I. 1965.The pathology of tuberculous
adversary. Antimicrob.Agents Chemother. 47:833–836. infection in BCG-vaccinated humans. Adv.Tuberc.
19. Balasubramanian, V., E. H. Wiegeshaus, and Res. 14:202–234.
D. W. Smith. 1994. Mycobacterial infection in 32. Sutherland, I., and I. Lindgren. 1979.The pro-
guinea pigs. Immunobiology 191:395–401. tective effect of BCG vaccination as indicated by
20. Hruban, R. H., and G. M. Hutchins. 1993. autopsy studies. Tubercle 60:225–231.
Mycobacterial infections, p. 331–350. In D. H. Dial 33. Shima, K., A. M. Dannenberg, Jr., M. Ando,
and S. P. Hammar (ed.), Pulmonary Pathology. S. Chandrasekhar, J. A. Seluzicki, and J. I.
Springer-Verlag, New York, N.Y. Fabrikant. 1972. Macrophage accumulation,
64 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

division, maturation, and digestive and micro- Pulmonary bovine-type tuberculosis in rabbits: bacil-
bicidal capacities in tuberculous lesions. I. Studies lary virulence, inhaled dose effects, tuberculin sen-
involving their incorporation of tritiated thymidine sitivity, and Mycobacterium vaccae immunotherapy.
and their content of lysosomal enzymes and bacilli. Clin. Diagn. Lab. Immunol. 5:871–881.
Am. J. Pathol. 67:159–180. 35. Converse, P. J., A. M. Dannenberg, Jr., J. E.
34. Converse, P. J., A. M. Dannenberg, Jr.,T. Shi- Estep, K. Sugisaki,Y.Abe, B. H. Schofield, and
genaga, D. N. McMurray, S. W. Phalen, J. L. M. L. M. Pitt. 1996. Cavitary tuberculosis pro-
Stanford, G. A. W. Rook, T. Koru-Sengul, duced in rabbits by aerosolized virulent tubercle
H. Abbey, J. E. Estep, and M. L. M. Pitt. 1998. bacilli. Infect. Immun. 64:4776–4787.
4
LIQUEFACTION OF CASEOUS
FOCI AND CAVITY FORMATION

Overview 65
Part I. Liquefaction of caseous tissue and cavity formation: a review of the
literature 66
Role of delayed-type hypersensitivity 66
Role of hydrolytic enzymes 66
Caseation and liquefaction are two distinct processes 67
Bronchoscope production of cavities in rabbits 68
Dermal BCG lesions in pilot studies on caseation and
liquefaction 68
Effects of large numbers of tubercle bacilli on liquefaction 68
Part II. Tuberculosis produced in rabbits by aerosolized virulent
M. bovis 71
Background 71
Low-dose Experiments I and III 71
Unique events in the late disease of the low-dose
experiments 74
High-dose Experiment II 74
Tuberculin sensitivity 78
Histopathology 79
Part III. Recent experiments attempting to reduce liquefaction and cavity
formation 84
Studies with Mycobacterium vaccae 84
Preliminary studies with the proteinase inhibitor Ritonavir 86
Summary of cavity formation and suggestions for its prevention 87
A proposed method to study liquefaction and cavity formation 90

Abstract. Part I is a review of the literature on liquefaction and cavity formation. Liq-
uefaction seems to be a delayed-type hypersensitivity reaction to the tuberculin-like
products of the bacillus. It seems to be carried out by hydrolytic enzymes from the sur-
rounding host cells and possibly by enzymes within the caseum after inhibitors have dis-
sipated. Liquefied caseum and cavities occur frequently in rabbits inhaling virulent bovine-
type tubercle bacilli.They also occur occasionally in rabbits inhaling virulent human-type
tubercle bacilli. Part II describes experiments of long duration in which pulmonary cav-
ities were produced in commercial New Zealand White rabbits by aerosolized virulent
bovine-type bacilli (Mycobacterium bovis, Ravenel strain).After the inhalation of a low num-
ber of these bacilli, liquefied caseum and cavities occurred in 6 to 10 weeks.After the inhala-
tion of a high number of these bacilli, liquefied caseum and cavities occurred sooner. Details
on the gross pathology and histopathology of these pulmonary lesions are presented. Part
III describes two attempts to reduce liquefaction and cavity formation in tuberculous rab-
bits. One was immunotherapy with Mycobacterium vaccae.The other was therapy with Riton-
avir, a proteinase inhibitor. Neither treatment (as administered) had any observable effect.

OVERVIEW Liquefaction of solid caseous foci in the lung


The chronic human-type tuberculosis with liq- perpetuates tuberculosis in humans because such
uefaction and cavity formation is easily pro- liquefaction enables the extracellular growth of
duced in rabbits, but it is not as easily produced tubercle bacilli and the formation of cavities
in guinea pigs and cannot be produced in mice. that discharge such bacilli into the airways and

65
66 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

into the environment. In liquefied caseum, espe- apparently does so in the following manner. By
cially that in the cavity walls, tubercle bacilli means of cytokines (including chemokines) (14,
sometimes grow to tremendous numbers, which 15), DTH increases the number of macrophages
makes the development of antimicrobial resis- surrounding the caseous center. Then, cell-
tance more likely. mediated immunity (CMI) (and DTH) acti-
Chapter 7 describes the pathophysiology of vates these cells (16), thereby raising their level
liquefaction and cavity formation. Chapter 3 of hydrolytic enzymes.Also, DTH increases the
presents details on the human disease. Refer- blood flow through the lesions, which probably
ences 1 through 7a below review various aspects lowers the level of the enzyme inhibitors in
of cavitary tuberculosis. the solid caseum. An accompanying antigen-
antibody reaction may play a role by activating
PART I. LIQUEFACTION OF CASEOUS complement, because C3a, C4a, and C5a cause
TISSUE AND CAVITY FORMATION: vasodilation, and the chemotaxin C5a also
A REVIEW OF THE LITERATURE
attracts leukocytes into the site (15).
Role of Delayed-Type Hypersensitivity
Yamamura’s group published the most significant Role of Hydrolytic Enzymes
studies on the causes of liquefaction (8–10; see Hydrolytic enzymes play a major role in lique-
also reference 11). Pulmonary cavities were pro- faction and cavity formation (6). Products result-
duced in 2 to 4 weeks by the injection of vari- ing from the hydrolysis of proteins and other
ous materials (emulsified in paraffin oil and lano- macromolecules increase the osmolarity of the
lin) through the chest wall directly into the lungs caseum.Water is absorbed, and the fluidity of the
of rabbits. Both living and dead tubercle bacilli, caseum is increased.The most significant stud-
paraffin-oil extracts of heat-killed tubercle bacilli, ies on the hydrolytic enzymes involved in liq-
and the particulate and proteolipid fractions of uefaction were performed in the laboratories of
Mycobacterium bovis BCG were all effective if the Weiss (17–19) and Dannenberg (20–23).
rabbits were previously made tuberculin positive Weiss and associates produced tubercles in
by subcutaneous injections of heat-killed tuber- rabbits by airborne infection with virulent M.
cle bacilli in adjuvant. Rabbits not previously sen- bovis. Caseous centers were present after 7 weeks,
sitized did not develop cavities as frequently, and and liquefied centers were present after 12
those that did had become tuberculin positive weeks. By 20 weeks, the cavities were large and
from the intrapulmonary injection. Similar results coalescing. Samples of normal lung, tuberculous
were obtained in guinea pigs. Desensitization of granulation tissue from the walls of caseous and
tuberculin-positive rabbits with tuberculoproteins liquefying foci, solid caseum, and completely liq-
(9) or a tuberculin-active peptide (10) prevented uefied caseum were homogenized and assayed
cavities from forming. for proteases and nucleases by two different
Azathioprine, an immunosuppressive drug, methods.
also prevented cavities (12), probably by reduc- In the first method, substrates, e.g., benzoyl-
ing the amount of delayed-type hypersensitivity argininamide, leucinamide, DNA, or RNA,
(DTH) (6).When live tubercle bacilli (H37Rv) were added to the homogenates, and their
were injected (through the chest wall) directly hydrolytic products were measured. In the sec-
into the lungs of rabbits, no cavities formed in ond method, the homogenates were allowed to
the antimicrobial-treated rabbits simultaneously autolyze under toluene at 35°C for 1 to 6 days,
treated with azathioprine, but cavities readily after which the breakdown products of pro-
formed in the antimicrobial-treated controls that tein, DNA, and RNA were measured. Both
were not treated with azathioprine (12). methods yielded similar results. Tuberculous
References 2, 3, 4, and 13 describe additional granulation tissue had high protease and nucle-
studies supporting the premise that DTH is ase activity. Normal lung tissue and recently
required for solid caseum to liquefy. DTH caseating tissue had moderate activity. Older
4. LIQUEFACTION OF CASEOUS FOCI AND CAVITY FORMATION 䡵 67

caseous tissue and liquefied tissue had little or no BCG lesions when studied in tissue sections by
activity. gelatin-substrate film techniques (23).
Using histochemical techniques, including the The level of -galactosidase, our classic lyso-
Daoust substrate film technique (24–27), our somal marker for macrophage activation, was
laboratory (20–23) provided direct visual evi- always highest in macrophages surrounding liq-
dence for the findings of Weiss and associates. uefied caseum and was decreased in areas farther
Proteinase (cathepsin D), DNase, RNase, away (11). The high levels of -galactosidase
hyaluronidase, and other enzymes, e.g., acid phos- and other hydrolytic enzymes in macrophages
phatase, -galactosidase, -glucuronidase, esterase, immediately surrounding the caseous and liq-
succinic dehydrogenase, and cytochrome oxi- uefied centers were possibly due to their inges-
dase, were most active in live and dead macro- tion and digestion of the necrotic material.“A
phages surrounding the caseous and liquefied good meal in the belly of macrophages” increases
centers (7a).The activity of these enzymes dimin- their content of digestive enzymes (33).
ished to near zero in fully liquefied areas. Most of Once started, the liquefaction of solid caseous
these studies were evaluated in rabbit dermal material seems to be autocatalyic.The ingestion
BCG lesions with liquefied caseous centers, but of liquefied caseous material by the surround-
some were evaluated in rabbit cavitary pulmonary ing macrophages increases the level of their
lesions caused by virulent bovine-type tubercle hydrolytic enzymes, and the higher level of
bacilli (Ravenel) (7a, 20). Immunohistochemical these enzymes hydrolyzes solid caseous tissue
methods confirmed that cathepsin D was present more efficiently. Table 1 summarizes the main
in epithelioid cells at the edge of the liquefied causes and results of liquefaction.
caseum of a pulmonary cavity (20).
In pulmonary tuberculous lesions of Caseation and Liquefaction
humans, guinea pigs, mice, and rabbits, Oshima Are Two Distinct Processes
and coworkers evaluated lipases by histo- References 11 and 34 through 36 review the lit-
chemical methods (28).The Gomori calcium- erature on caseous necrosis and cavity formation.
lead method was used with polyoxyethylene However, the distinction between these two
sorbitan-monostearate and Tween 80 as sub- processes is not clearly addressed in references 34
strates. The caseous material in these lesions and 35. Solid caseum is caused by the killing of
showed no lipase activity. Other studies on viable tissue, whereas liquefied caseum is caused
lipases and esterases in tuberculosis are reviewed
in references 3 and 18.
Enzyme inhibitors from the bacilli and the TABLE 1 Causes and results of liquefactiona
necrotic host tissues probably exist in caseous Causes
material.Their presence may be the reason why Delayed-type hypersensitivity (DTH) to tuberculin-
solid caseum does not liquefy for at least a num- like bacillary products
ber of weeks (3, 18, 19).To date, such inhibitors Hydrolytic enzymes, especially proteases, DNases, and
have not been characterized. RNases, and probably lipases
Pepstatin is a nontoxic pentapeptide from Results
Actinomycetes sp. (29) discovered by Umezawa’s Extracellular bacillary multiplication (that is
sometimes tremendous), which may allow
group. Pepstatin is a highly effective inhibitor of antimicrobial drug-resistant mutants to develop
pepsin (29–31) and rabbit macrophage cathepsin High concentrations of tuberculin-like products that
D (32). However, in two pilot studies, pepstatin cause caseous necrosis of the bronchial wall and a
had no observable effect on rabbit dermal BCG cavity to form
lesions when administered either subcutaneously Spread of bacilli through the air passages to other
or directly into these lesions (32), so these stud- parts of the lungs and to the environment, where
ies were not continued. Pepstatin also did not they may infect other people
affect the proteinase activity of macrophages or a
Adapted from reference 6.
68 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

by the hydrolysis of solid caseum after it has to produce cavities more readily than did the
formed. Caseous necrosis may lead to liquefac- instillation of virulent human-type bacilli. If
tion and cavity formation, but in both rabbits the rabbits were not sensitized to tuberculin, cav-
and humans the majority of caseous lesions do ities were not produced as easily, which con-
not liquefy or form cavities.The caseum merely firmed Yamamura’s findings (described above).
inspissates, may be walled off by fibrosis, and in This bronchoscopic method has the advan-
humans often calcifies (see chapter 3). Follow- tage of not requiring an aerosol apparatus to
ing the inhalation of fully virulent bovine-type infect the rabbits.Also, it produces cavities more
bacilli, Lurie’s susceptible rabbits produced pul- quickly, i.e., in 3 to 5 weeks rather than in 10
monary tuberculous lesions with considerable weeks. However, the bronchial instillation of a
caseous necrosis, but these lesions never lique- large number of tubercle bacilli does not match
fied and cavitated (4). the normal way of developing pulmonary tuber-
Tuberculous lesions in guinea pigs show con- culous cavities, in which each pulmonary tuber-
siderable caseous necrosis, but liquefaction and culous lesion develops from a single inhaled
cavities only occur in some guinea pigs and unit containing only 1 to 3 bacilli.
then only if 1 or 2 pulmonary lesions are pre-
sent (see chapter 15). Reference 36 is a rather Dermal BCG Lesions in Pilot Studies
complete review of caseation and cavity for- on Caseation and Liquefaction
mation in rabbits, mice, guinea pigs, humans, and A simpler way to produce liquefaction with
nonhuman primates. Cavities are readily pro- large numbers of bacilli in one location would
duced in rabbits and humans, but they are not be to inject BCG intradermally (see proposed
easily produced in guinea pigs and never occur method beginning on p. 90). The resulting
in mice. lesions caseate, liquefy, and ulcerate, discharging
However, like Lurie’s resistant rabbits (4), the liquefied caseum, similar to tuberculous
commercial New Zealand White rabbits (20, 37) lesions produced bronchoscopically in the lungs.
often form cavities after inhaling virulent In rabbits already sensitive to tuberculin, BCG
bovine-type tubercle bacilli if given sufficient lesions ulcerate more quickly (called the Koch
time. After the inhalation of virulent human- phenomenon).
type tubercle bacilli, these hosts may also form Cathepsin D is a major protease of mac-
cavities (4, 36, 38, 39), but they do so less fre- rophages (21, 40–43). It exists at high levels in
quently than after the inhalation of virulent macrophages surrounding the liquefied caseous
bovine-type bacilli. Cavity formation in com- centers of BCG lesions (21).This distribution
mercially available New Zealand White rabbits of cathepsin D is similar to that found in the
with aerosolized virulent bovine-type tubercle walls of pulmonary cavities produced by
bacilli is described in part II of this chapter. aerosols of virulent tubercle bacilli (20).There-
fore, for pilot studies of liquefaction and cavity
Bronchoscope Production of Cavities formation, we highly recommend the dermal
in Rabbits BCG model. It does not require an aerosol
Rabbits were sensitized to tuberculin by inject- apparatus and can be performed in biological
ing subcutaneously 107 heat-killed virulent level 2 facilities.
bovine-type tubercle bacilli (Ravenel strain) in
a mineral oil-lanolin adjuvant on four different Effects of Large Numbers of
occasions 3 or 4 days apart (see reference 35).At Tubercle Bacilli on Liquefaction
21 days after the last injection, 2 ⫻ 104 live vir- Both bronchoscopically induced pulmonary
ulent bovine-type tubercle bacilli (Ravenel) lesions and dermal BCG lesions are produced by
were instilled with a bronchoscope into the injecting large numbers of tubercle bacilli in
right lower lung. Pulmonary cavities occurred one location.The presence of a large number of
in every rabbit within 3 to 5 weeks.The instal- bacilli apparently accelerates the liquefying
lation of bovine-type tubercle bacilli seemed process (see reference 9). Tubercle bacilli and
4. LIQUEFACTION OF CASEOUS FOCI AND CAVITY FORMATION 䡵 69

FIGURE 1 A multiloculated cavity in the lung of a


rabbit (#8, Experiment I) that had inhaled 620 virulent
bovine-type tubercle bacilli (Ravenel S) 19 weeks previ-
ously.The hole at the upper right side of the photograph
is an intact bronchus.The hole at the lower left is part of
the loculated cavity.The center of each is black because the
back was sliced from the specimen to stabilize it for pho-
tography. Note the fibrosis around the large cavity in the
center of the photograph. Magnification, ⫻3. Reprinted
with permission from reference 20.
This is an example of the slowly progressive cavitary
disease frequently found in humans (see chapter 3).
Note:Table 2 of reference 20 lists the necropsy find-
ings on each rabbit that lived 18 or more weeks in the
three experiments that we performed. I did not reproduce
the entire table in this chapter but have listed the rabbit
and experiment numbers in many of the figures, so that
interested readers can easily obtain (from reference 20)
more details on the characteristics of disease in the rab-
bits providing the specimens for these photographs.

FIGURE 2 Multiple cavities in the upper part of the


right lower lobe of the lung of a rabbit (#5, Experiment
III) euthanized 33 weeks after the inhalation of 340 vir-
ulent bovine-type tubercle bacilli.These are mostly sec-
ondary and tertiary cavities that developed locally from
the bronchogenic spread of large numbers of bacilli from
one or two primary lesions. Despite the extensive tuber-
culosis in the upper part of this lobe, no secondary lesions
were grossly visible in the lower part. In addition, the
whole left lung of this rabbit was free of grossly visible
tuberculous lesions. Magnification, ⫻1. Reprinted with
permission from reference 20.
This is an example of the local nature of tuberculosis and
the efficacy of acquired (adaptive) immunity that prevents
the progress of tuberculous lesions arising elsewhere from
relatively low numbers of inhaled bacilli.
70 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

FIGURE 3 Specimen sliced from the right side of the


right lower lobe of rabbit #5, Experiment III (Fig. 2). Note
the absence of functional lung tissue, the large size of the
cavities, the residual semisolid caseous tissue (center), and
the extensive fibrosis. Magnification, ⫻4.

FIGURE 4 Tuberculous lesions in the right upper


lobe of rabbit #5 (Experiment III). (The right lower
lobe of this rabbit is depicted in Fig. 2.) Two large cavi-
ties have been sliced open. Note that most of the lesions
are well encapsulated with fibrous tissue. Fibrous encap-
sulation of tuberculous lesions is common in chronic
human cavitary disease. Magnification, ⫻2.
4. LIQUEFACTION OF CASEOUS FOCI AND CAVITY FORMATION 䡵 71

their tuberculin-like products are highly chemo-


tactic for monocytes/macrophages and other
cells.These macrophages produce proteases and
other hydrolytic enzymes (see chapter 6), which
can liquefy necrotic tissue.
Tubercle bacilli themselves produce pro-
teinases, nucleases, and lipases (3), which may or
may not play a role in the liquefaction of solid
caseum. Granulocytes also contain such
hydrolytic enzymes, but they are not a promi-
nent feature of most liquefying caseum (9).

PART II. TUBERCULOSIS PRODUCED


IN RABBITS BY AEROSOLIZED
VIRULENT M. BOVIS
Background
Lurie produced liquefaction and cavities in his
inbred resistant rabbits by allowing them to
breathe air containing a very low number of vir-
ulent bovine-type tubercle bacilli (Ravenel
strain) in the animal room over a period of
many months—an experiment not likely to be
repeated today (see chapter 12). Lurie (4, 44), as
well as others (45, 46), also produced cavities by FIGURE 5 Tuberculous laryngitis in a rabbit (#4,
aerosols of bovine-type tubercle bacilli. In fact, Experiment III) that had inhaled 300 virulent bovine-
even aerosols of human-type bacilli, which are type tubercle bacilli 33 weeks previously.The lung of this
less virulent for rabbits, sometimes produce cav- rabbit had 14 lesions, 4 of which had formed cavities.
A cavity in the right lower lung apparently discharged
ities (4, 38, 39). so many bacilli into the bronchial tree that the larynx
Rabbits do not usually develop the extensive became infected. Eventually, the airway became partly
bronchopneumonia that some humans develop obstructed, so the animal was breathing with difficulty
(see chapter 3). Extensive bronchopneumonia in at the time it was euthanized. Magnification, ⫻5.4.
humans is apparently caused by a DTH reaction Reprinted with permission from reference 20.
Before antimicrobials were available to treat tuber-
to the tuberculin-like products in the liquefied culosis, a similar tuberculous laryngitis occasionally
caseum that enters the alveoli, and humans are occurred in humans. Such patients required a tra-
at least 100 times more sensitive than rabbits to cheotomy to stop the labored breathing.
tuberculin.
Rabbits do not cough.Therefore, the spread
of the disease to other animals housed in the Low-Dose Experiments I and III
same room is less frequent than in rooms where In our low-dose experiments, commercial New
tuberculous monkeys are housed. Rabbits Zealand White rabbits (20, 37) inhaled 220 to
sneeze, however, which may aerosolize tubercle 940 virulent bovine-type tubercle bacilli
bacilli lodged in their nasal passages. (Ravenel S from the ATCC [20]).This strain was
The following is a description of our exper- of slightly reduced virulence for rabbits, but
iments in which rabbits inhaled aerosols of vir- still sufficiently virulent to cause extensive dis-
ulent bovine-type tubercle bacilli. They show ease and even death (20, 37). In Experiment I,
that cavitary disease is easily produced in this two rabbits (each time) were euthanized at 5, 8,
animal species. Similar experiments were de- 10, and 13 weeks after the aerosol infection.
scribed by Lurie in his inbred rabbits (4). Their lungs contained 1 to 18 grossly visible or
72 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

FIGURE 7 A caseous tubercle partly obstructing the


lumen of the trachea (above) and constricting the esoph-
agus (bisected below) (rabbit #12, Experiment III).
Magnification, ⫻4.9.
The esophageal constriction apparently caused such
malnutrition that the resistance of the rabbit was
decreased and many small metastatic pulmonary lesions
developed.
FIGURE 6 A caseous tubercle at the bifurcation of
a bronchus near the hilus in a rabbit (#12, Experiment
III) that had inhaled 420 virulent bovine-type tubercle
bacilli (Ravenel S). We euthanized this rabbit at 30 considerable fibrosis. In Experiment III, one
weeks, because it had labored breathing. Magnification,
rabbit had four 2- to 3-mm pulmonary lesions
⫻4.9. Reprinted with permission from reference 20.
Before antimicrobials were available to treat tuber- that were almost healed.
culosis, similar lesions occasionally occurred in the Secondary pulmonary lesions were found in
bronchial tree in human beings. the lungs of several rabbits.They probably arose
by bronchial spread of the disease from one or
more cavities.The secondary lung lesions were
usually much smaller than the primary lesions.
palpable primary pulmonary tubercles. By 5 Therefore, most of the primary lesions could be
weeks, the tubercles had developed solid caseous identified at necropsy.The lymphoid tissue of the
centers. By 8 weeks, some of the centers had liq- ileocecal junction and appendix of these rabbits
uefied. And, by 10 and 13 weeks, some of the often contained 1- to 2-mm lesions, which
lesions contained small cavities. When the came from the numerous bacilli (growing in
remaining rabbits were euthanized (at 19 and 22 cavities) that ascended the bronchial escalator
weeks in Experiment I and at 33 weeks in and were swallowed (47). One rabbit had two
Experiment III), over half of the solid caseous small (3 to 4 mm) tubercles in one of its kidneys
foci had liquefied, and many cavities had formed (probably of hematogenous origin).
(Fig. 1 through 4). The tracheobronchial lymph nodes were
The disease varied greatly among the indi- slightly enlarged and occasionally contained
vidual rabbits. In some, hardly any disease was tuberculous granulomas. Similar to immuno-
present, whereas in others, half of a pulmonary competent adult humans, rabbits with moder-
lobe was destroyed. Most of the rabbits appeared ate to high native resistance to tuberculosis do
to have their disease under control, because not develop progressive lesions in their hilar
their pulmonary lesions were surrounded by nodes (4, 5, 47).
4. LIQUEFACTION OF CASEOUS FOCI AND CAVITY FORMATION 䡵 73

TABLE 2 Data on rabbits dying of pulmonary tuberculosis in the high-dose Experiment II


Approximate A B C
time of death No. of No. of Ratios:
Treatment (or time inhaled visible A/B No. of
(rabbit no.) euthanized virulent cavitiesb Comment
primary for each
when near bacillary lesionsa rabbit
death) (wk) units
Control #2 5 4,800 550 8.7 None Numerous 3- to 4-mm
lesions with minute
caseous centers
M. vaccae #9 6 5,800 210 27.6 Many Over half of the lesions
had cavitated
Control #11 7 4,700 260 18.1 Many Over half of the lesions
had cavitated
Control #10 8.5 3,900 250 15.6 Many Over half of the lesions
had cavitated
Control #4 9 5,300 500 10.6 Many Numerous cavities were
present
Means 4,900 354 13.8
a
Includes primary lesions with cavities. Details on the remaining six rabbits of this high-dose experiment are presented in Table
1 of reference 20. Reproduced with permission from reference 37.
b
Note that, in this high-dose experiment, cavities were present at 6 to 9 weeks after infection.

FIGURE 8 Pulmonary tuberculosis in the left lung of a rab-


bit (#8, Experiment II) that had inhaled 5,500 virulent bovine-
type tubercle bacilli 18 weeks previously.The pleural surface of
the lung is roughened by numerous coalescing tubercles. Mag-
nification, ⫻1.2.
Tuberculous pleurisy frequently occurs in humans due to
similar lesions just below the pleural surface (see chapter 3).
74 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

FIGURE 9 The same lung shown in Fig. 8, bisected horizontally. The lung contains
numerous primary and secondary lesions and many minute cavities. Note how little functional
pulmonary tissue remains in the upper lobe. Magnification, ⫻1.2.
Similar local consolidation occurs in humans (see chapter 3).

Unique Events in the Late Disease of rowed the airways (Fig. 6 and 7). One of these
the Low-Dose Experiments lesions also partly obstructed the adjacent esoph-
At 33 weeks, one rabbit showed honeycomb agus (Fig. 7).The rabbit euthanized at 30 weeks
formations, i.e., multiple coalescing cavities in was the only rabbit in any of the three experi-
both its right upper and right lower lung lobes ments that had numerous metastatic caseous
(20) (Fig. 2 through 4). Evidently, one or more lesions (0.5 to 2.0 mm in diameter) in the lungs,
primary cavitary lesions had discharged many which were probably caused by a decrease in
bacilli locally.These bacilli created new lesions host resistance from the malnutrition (1).
nearby, which in turn cavitated. However, despite
the extensive destruction in these two lung High-Dose Experiment II
lobes, the rabbit remained in good health, and In Experiment II, New Zealand White rabbits
the remainder of its lungs showed no grossly vis- (20, 37) (inbred by L. F. M. van Zutphen at the
ible evidence of tuberculosis. University of Utrecht, The Netherlands)
Two other rabbits were euthanized because inhaled 3,900 to 5,800 virulent bovine-type
they showed labored breathing and had lost tubercle bacilli (Ravenel S). Between 5 and 9
weight.The rabbit euthanized at 33 weeks had weeks, about half of the rabbits had to be euth-
extensive tuberculous laryngitis, which nar- anized because of impaired breathing. Their
rowed the airways (Fig. 5).The rabbit euthanized lungs contained between 210 and 550 pro-
at 30 weeks had two caseous lesions that nar- gressing primary lesions (Table 2). Most of
4. LIQUEFACTION OF CASEOUS FOCI AND CAVITY FORMATION 䡵 75

FIGURE 10 Pulmonary cavities from two rabbits (in high-dose Experiment II) that had inhaled
about 5,000 virulent bovine-type tubercle bacilli 18 weeks previously. Below each cavity are
encapsulated fibrotic lesions with large semisolid caseous centers that had not discharged into
bronchi.The amount of softening of the caseous material cannot be accurately assessed in for-
malin-fixed tissues, because it is hardened by the fixation process.At necropsy, the consistency
of the caseum varied from that of a “hard” cheese to that of a rather “soft” cheese. Magnifica-
tion, ⫻2.7. Reprinted with permission from reference 20.

these pulmonary lesions had liquefied centers, averaged only 6.7 and 8.8 primary tubercles,
and after 6 weeks many cavities were present respectively (Table 3).
(Table 2). When compared with the rabbits in the low-
The remaining rabbits were euthanized at dose experiments, these rabbits showed larger tra-
18 weeks. Because they survived so long, their cheobronchial lymph nodes with more caseation,
pulmonary lesions were well encapsulated (Fig. apparently because of increased numbers of
8 through 11).These “high-dose” rabbits, how- bacilli. However, none of these lymph nodes
ever, were less active, more emaciated, and often showed the extensive caseous necrosis of Lurie’s
had dyspnea upon exertion.All of these rabbits susceptible rabbits (4, 5, 7). In other words, an
showed secondary lesions in the appendix and adult type rather than a childhood type of tuber-
ileocecal junction, apparently from swallowing culosis was produced (1, 6) (see chapter 1).
bacilli that ascended the bronchial tree from These inbred van Zutphen rabbits and Lurie’s
the multiple cavities that were present. highly resistant strain III rabbits originated from
Due to the large number of primary pul- the same inbred stock at the Jackson Laborato-
monary lesions and the long duration of the ries in Bar Harbor, Me. (see chapter 14). As
experiment, we could not always differentiate expected, the variation in the number of pri-
secondary lesions from primary lesions in the mary pulmonary tubercles found among them
lung, but we could do so with about 90% seemed to be less than that found among the
accuracy.The rabbits in the high-dose exper- commercial rabbits (20, 37), but too few rabbits
iment averaged 310 primary tubercles, whereas were in these experiments to prove this obser-
the rabbits in the two low-dose experiments vation statistically.
FIGURE 11 Walled-off tuberculous lesions from the lungs of rabbits (in high-dose Exper-
iment II) 18 weeks after the inhalation of about 5,000 virulent bovine-type tubercle bacilli.
In the specimen on the right, there are several semisolid caseous lesions with thick fibrous cap-
sules and one cavity. In the specimen on the left, the amount of fibrosis is so extensive that
little functional lung tissue remains. Magnification, ⫻3.1. Fibrocaseous tuberculosis is a fre-
quent occurrence in humans.

TABLE 3 Ratios: number of inhaled bovine-type tubercle bacilli that produced one grossly visible primary
pulmonary lesion in commercial rabbitsa
A B
Inhaled dose of No. of C
Experiment
viable bovine-type visible primary Ratios: A/B
no.
tubercle bacilli (Ravenel S) pulmonary lesions for each rabbitb
(bacillary units) (including cavities)
Experiment I 523 ⫾ 137 6.7 ⫾ 2.3 107 ⫾ 29
(low dose) (523/6.7 ⫽ 78)
6 rabbits
Experiment II 5,100 ⫾ 186 310 ⫾ 36 18.2 ⫾ 1.7c
(high dose) (5,100/310 ⫽ 16.5)
11 rabbits
Experiment III 418 ⫾ 24 8.8 ⫾ 1.2 63 ⫾ 13
(low dose) (418/8.8 ⫽ 48)
12 rabbits
a
The grossly visible primary lesions of the M. vaccae and control groups were pooled to prepare this table.The means and their
standard errors are listed.The P values for the ratios in column C are as follows: for 107 and 18.2, P ⫽ 0.001; for 107 and 63, P
⫽ 0.124; and for 18.2 and 63, P ⫽ 0.003 (Student’s t test). Reproduced with permission from reference 37.
b
The ratio for each rabbit is the number of inhaled bacilli divided by the number of visible primary pulmonary lesions.The
means in the ratio column were derived from individual ratios.They are not the means in column A divided by the means in
column B, which are given in parentheses.The difference between the two methods is discussed in chapter 11.
c
The difference in ratios between Experiment II and Experiments I and III suggests that the acquired immune response has
a limited capacity to prevent developing primary pulmonary lesions from reaching grossly visible size (see text).

76
4. LIQUEFACTION OF CASEOUS FOCI AND CAVITY FORMATION 䡵 77

TABLE 4 M. vaccae immunotherapy of rabbits with active tuberculosis produced by the inhalation of virulent
bovine-type tubercle bacilli (Ravenel S)a
High-dose Experiment II Low-dose Experiment III
(survivors euthanized (survivors euthanized
at 18 weeks) at 33 weeks)
M. vaccae Controls M. vaccae Controls
Immunotherapy schedule 5, 9, and 13 weeks 7, 12, 17, 22, and 27 weeks
Rabbit no. 1, 3, 5, 7, 9 2, 4, 6, 8, 10, 11 2, 4, 6, 7, 9, 12 1, 3, 5, 8, 10, 11
No. of inhaled virulent 5,400 ⫾ 270 4,850 ⫾ 230 395 ⫾ 39 440 ⫾ 29
bacillary units P ⫽ 0.149 P ⫽ 0.417
No. of grossly visible 252 ⫾ 26.3 359 ⫾ 56.8 8.0 ⫾ 1.9 9.5 ⫾ 1.5
primary lesions in lungs P ⫽ 0.145 P ⫽ 0.545
(including those with
cavities)
No. of visible primary 3.0 ⫾ 0.9 1.8 ⫾ 0.8
pulmonary lesions P ⫽ 0.341
with cavities
Percentage of visible 35.7 ⫾ 11.9 18.7 ⫾ 9.5
primary lesions that P ⫽ 0.291
cavitated
Dermal tuberculin 2,606 ⫾ 562 mm3; 2,657 ⫾ 396 mm3
sensitivity at 4 weeks
Dermal tuberculin 1,038 ⫾ 191 mm3; 1,283 ⫾ 340 mm3
sensitivity at 33 weeks
No. of metastatic lesions 428 ⫾ 258 320 ⫾ 50 89 ⫾ 47 149 ⫾ 82
in the ileocecal and P ⫽ 0.796 P ⫽ 0.539
appendix lymphoid
tissues
Weight at time of 2.5 ⫾ 0.2 2.7 ⫾ 0.2 4.0 ⫾ 0.3 4.2 ⫾ 0.3
necropsy (in kg) P ⫽ 0.570 P ⫽ 0.578
a
This table summarizes information published in references 20 and 37.The number of primary lesions was derived from all
of the rabbits in each group, including those in Table 2 of this chapter that died early.The other data are from the survivors at the
two times of euthanasia (above). Means and their standard errors are presented. Student’s t test was used to determine significance.
Note that M. vaccae immunotherapy had no apparent effect on the number of grossly visible primary lesions, on the percentage
of those lesions that cavitated, or on the tuberculin sensitivity of the host.

In brief, the high dose of virulent bovine-type were required to produce one visible tubercle in
tubercle bacilli probably taxed host resistance to the high-dose experiment, whereas 63 and 107
its limits, so that the hosts with somewhat inhaled bacilli were required to do so in the
weaker resistance succumbed to the disease by low-dose experiments. The large number of
9 weeks. However, most of the hosts with inhaled bacilli evidently reduced the local effi-
stronger resistance lived comfortably until they cacy of the host’s innate and acquired immune
were euthanized at 18 weeks. responses, so that 3 to 6 times the number of
The high dose also had an unexpected effect microscopic tubercles reached visible size.There-
on the number of inhaled bacillary particles fore, the host immune system apparently has a
required to produce one grossly visible primary limited capacity, and this capacity was exceeded
tubercle (called “the ratio” by Lurie [4]).Table 3 in the high-dose experiment. Many factors may
shows that, when calculated by the Lurie method be involved in this dose effect, such as a limited
described in chapter 11, only 18 inhaled bacilli number of defense cells, changes in Th1/Th2
78 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

FIGURE 12 A glycol methacrylate-embedded tissue section of a tuberculous lesion, from


a rabbit (#8, Experiment I) that had inhaled 620 virulent bovine-type tubercle bacilli 19 weeks
previously.The section was stained histochemically for -galactosidase, which is our marker for
activated macrophages: the darker the blue color produced, the more activated the macrophage
(see chapter 6). This photograph shows portions of three caseous centers, surrounded by
tuberculous granulation tissue containing numerous -galactosidase-positive macrophages
(epithelioid cells). Peripheral to these macrophages are lymphocytes, plasma cells, fibroblasts,
and unactivated (-galactosidase-negative) macrophages.At the lower left are a few alveoli (with
thickened walls) containing accumulations of highly activated alveolar macrophages, which are
easily recognized by their dark -galactosidase-staining cytoplasm. Counterstained with
Giemsa. Magnification, ⫻100. Reprinted with permission from reference 20.

ratios (discussed in reference 37), and increased (Tuberculous guinea pigs are about twice as
glucocorticoid production. sensitive as tuberculous rabbits.) In the rabbits of
The dose effect did not, however, occur when low-dose Experiment III with well-controlled
virulent human-type tubercle bacilli were tuberculosis, tuberculin sensitivity decreased
inhaled, apparently because the human type is about 50% between 4 and 33 weeks (Table 4)
less virulent for rabbits than the bovine type (20, 37). Humans with well-controlled clinical
(39). In fact, the dose effect with the human type tuberculosis may not show as great a reduction
seemed to be in the opposite direction: when a in tuberculin sensitivity. These species differ-
higher number were inhaled, more (not fewer) ences may be due to partial desensitization to
human-type bacilli were apparently required to tuberculin by the nonpathogenic mycobacteria
produce each visible tubercle. See chapter 11 for in the diet of rabbits (see reference 8).
a complete discussion of tubercle counts. The studies of Yamamura (8–10, 12) indicate
that tuberculin sensitivity is necessary for cavi-
Tuberculin Sensitivity ties to form in rabbits. In these studies, tubercle
When infected with tubercle bacilli, humans bacilli (or their components) were injected
are about 100 times more sensitive to tuberculin directly into the lungs through the chest wall
than are rabbits (see PPD in the glossary). (discussed above in part I).
4. LIQUEFACTION OF CASEOUS FOCI AND CAVITY FORMATION 䡵 79

FIGURE 13 A glycol methacrylate-embedded tissue section of a tuber-


culous lesion from a rabbit (#8, Experiment I) that had inhaled 620 virulent
bovine-type tubercle bacilli 19 weeks previously.This section was stained for
acid phosphatase, a bright-red color (black in this photograph). Acid phos-
phatase is another marker for macrophage activation (see chapter 6).
The edge of a cavity is at the lower right. Next (proceeding to the upper
left) are unstained macrophages (epithelioid cells), some of which are disinte-
grating, and then an area containing many acid phosphatase-positive
macrophages.The outer layer (upper left) is the surrounding capsule contain-
ing many fibroblasts, a few of which also stain positive for acid phosphatase.
The dark circle in the capsule is a metaplastic alveolus (see Fig. 21–24). Not
all of the mature epithelioid cells (large cells with a rounded outline) stain pos-
itive for acid phosphatase, indicating heterogeneity in this cell population (see
reference 40). Counterstained with Giemsa. Magnification, ⫻330. Reprinted
with permission from reference 20.

In our inhalation experiments, we found no within cavities and provided a large antigenic
correlation between the degree of tuberculin stimulation for maintaining tuberculin sensitiv-
sensitivity at 4 weeks and the percentage of ity (20, 37).
lesions that cavitated by 33 weeks (37). Cavity
formation is a local phenomenon. Tuberculin
skin testing may not accurately reflect what is Histopathology
occurring in the lesions themselves. The local Cavity Formation. Macrophages (epithe-
nature of cavity formation and the variability lioid cells) surrounding caseous and liquefied
inherent in outbred rabbits may be why our sites are often highly activated (Fig. 12 and 13),
inhalation experiments show no correlation i.e., they contain high levels of hydrolytic and
between the initial dermal sensitivity to tuber- oxidative enzymes (discussed above and in
culin and the percentage of lesions that later chapter 6).Their hydrolytic enzymes—DNases,
formed cavities. RNases, proteinases, and probably lipases—
Rabbits with the most cavities at 33 weeks apparently play major roles in the liquefaction
tended to maintain their tuberculin sensitivity at of solid caseous material (7a, 20–23). Cathep-
higher levels than rabbits with fewer cavities sin D, which is the main proteolytic enzyme of
(20, 37).This trend was probably produced by macrophages (21), is shown histochemically in
the numerous bacilli that grew extracellularly Fig. 14.
80 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

FIGURE 14 A tissue section of the wall of a tuberculous cavity produced in a rabbit (#4, Exper-
iment I) that had inhaled 880 virulent bovine-type tubercle bacilli 22 weeks previously. Immuno-
stained for cathepsin D, a major proteinase of macrophages.
Intact bronchial epithelium is shown at the upper right. At the lower left, this epithelium is
ruptured, allowing the liquefied caseum in the cavity (below) to be discharged into the bronchus.
Cathepsin D (a brown color not easily identified in this black and white photograph) is present
in both live and dead macrophages (epithelioid cells) adjacent to the liquefied caseum.This find-
ing suggests that cathepsin D plays an important role in the liquefaction process. Immunostained
with polyclonal goat antibody to rabbit cathepsin D and the avidin-biotin peroxidase technique.
Magnification, ⫻420. Reprinted with permission from reference 20.

Hemoptysis. In clinical tuberculosis, a fre- lioid cell population.The granules were autoflu-
quent occurrence is blood-tinged sputum and orescent and birefringent and stained darkly with
occasionally frank hemoptysis. The cause of Sudan Black B,which has an affinity for both lipid
bloody sputum is the erosion of blood vessels in and acidic components. Some of these granules
the cavity wall by the necrotizing process (Fig.15). were even dark in unstained tissue sections.They
did not stain with carbol-fuchsin for acid-fast
Granulated Macrophages. Macrophages material or with Alcian blue or periodic acid-
containing granules that stained darkly with Schiff reagents for carbohydrates. These dark
Giemsa, methylene blue, or azure A were occa- granules may contain lipofuchsin, ceroid, or tis-
sionally found in the intact tuberculous granula- sue and bacillary debris, but their exact role in the
tion tissue that surrounded the caseous, liquefied, pathogenesis is unknown (20).
and cavitary centers of the pulmonary lesions
(Fig. 16).These macrophages were present both Epithelioid Cells. Large mature epithe-
as isolated cells and as small groups but composed lioid cells are known to be cells that have
no more than 5% of the macrophage/epithe- destroyed many or all of the tubercle bacilli
4. LIQUEFACTION OF CASEOUS FOCI AND CAVITY FORMATION 䡵 81

FIGURE 15 A tissue section of a rabbit pulmonary cavity wall.A small blood vessel extends
into the cavity’s lumen at the right of the photograph. Such exposed vessels are the source of
blood in the sputum. In humans, when a larger blood vessel is similarly exposed in a cavity and
ruptures, massive hemoptysis and sometimes fatal hemorrhage may occur.
Adjacent to the small blood vessel in this photograph are disintegrating mature epithelioid
cells. Lurie (4, 48) showed that such large ovoid cells have destroyed many of the bacilli that
they once contained. However, adjacent to the liquefied caseum in the wall of the cavity, the
concentration of bacillary tuberculin-like products is often too high for such epithelioid cells
to survive very long. The glycol methacrylate-embedded tissue section was stained with
Giemsa. Magnification, ⫻400. Reprinted with permission from reference 20.

they once contained (4, 48). Their histologic Number of Bacilli. Since we used thin 1-
appearance is due to bacillary components being to 2-m glycol methacrylate-embedded tissue
finely dispersed throughout their cytoplasm (4, sections (50, 53, 54) (instead of the usual 7-m
48, 49).We used the histochemical stains for - paraffin-embedded sections), the number of
galactosidase and acid phosphatase to charac- tubercle bacilli seen microscopically appeared to
terize such cells (16, 22, 40, 50, 51). be less than that described in the literature.With
Many epithelioid cells were present in the fluorescent staining, we usually found more
tuberculous granulation tissue that surrounded bacilli in liquefied caseum (Fig. 17) than in solid
the caseous, liquefied, and cavitary centers of the caseum.With acid-fast staining, we also found
lesions (Fig. 12 and 13), but not all of these tubercle bacilli within tissue spaces (which were
cells stained for -galactosidase and acid phos- possibly lymphatics) (Fig. 18), within tissue
phatase.The negative-staining epithelioid cells macrophages, and within the exudate present in
may have been activated for other products, nearby alveolar spaces.
e.g., reactive oxygen and nitrogen intermediates The number of bacilli that could be cultured
(discussed in reference 40), or they may have from liquefied caseum showed considerable
been deactivated (52) or were dying. variability (Table 5) (20). Some liquefied caseum
82 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

FIGURE 16 An area slightly deeper into the cavity wall than that depicted in Fig. 15. In the
center are macrophages containing dark-staining granules.These macrophages occur in some
places within the cavity wall, but, in most places, nongranulated macrophages predominate.The
nature of these granules is unknown (see text).
Part of the capsule (which surrounds this cavitary lesion) is shown at the left of the photo-
graph. It contains fibroblasts and some lymphocytes and plasma cells. Many of the fibroblasts con-
tain translucent (probably secretory) granules.The glycol methacrylate-embedded tissue section
was stained with Giemsa. Magnification, ⫻600. Reprinted with permission from reference 20.

had tremendous numbers, while other liquefied bacillary multiplication (4). Figure 20 clearly
caseum in the same rabbit had relatively few (see demonstrates this phenomenon: the most numer-
rabbits #4A and 4B in Table 5). In other rabbits ous tubercle bacilli are present in the liquefied
(e.g., rabbit #10), no viable bacilli were recov- caseum adjacent to the air within the cavity.
ered from the liquefied caseum. The rate of
bacillary multiplication in liquefied caseum Fibroblasts. At 19 and 22 weeks, fibroblasts
probably was influenced both by the composi- were common in the peripheral regions of the
tion of the liquefied material, including its pH tuberculous granulation tissue, and collagen fibers
(3), and by the ability of the bacilli to change formed a capsule around the lesion (Fig. 21 and
from dormancy in solid caseum to active extra- 22).Were it not for the extracellular multiplica-
cellular growth in liquefied caseum. tion of tubercle bacilli in the liquefied caseum,the
In liquefied caseous centers an appreciable disease would probably have been arrested.
number of acid-fast bacilli were often visible
microscopically (4, 20, 37), but in solid caseous Lymphocytes and Plasma Cells. From
centers only a few bacilli were visible (4).The 5 weeks on, the disease appeared to be slowly
number of bacilli usually increased substantially progressing. Many lymphocytes and plasma cells
after a cavity formed (Fig. 19). were present in the lesions (Fig. 22 and 23). Dur-
The increased amount of oxygen provided by ing the later stages of the disease (at 19 and 22
connection to the bronchial tree is a stimulus for weeks), occasional lymphoid nodules were
4. LIQUEFACTION OF CASEOUS FOCI AND CAVITY FORMATION 䡵 83

FIGURE 17 A tissue section of the liquefied caseum in a pulmonary lesion of a rabbit (Exper-
iment II) that had inhaled about 5,000 virulent bovine-type tubercle bacilli 7 weeks previously.
The bacilli were stained with TB Fluorostain (rhodamine B/auramine 0) (Polysciences, Inc.,
Warrington, Pa.) and visualized with an epifluorescent microscope at wavelengths 495 nm (exci-
tation) and 535 nm (emission). Liquefied caseum contains variable numbers of tubercle bacilli,
both microscopically in tissue sections and by culture (see Table 5). Magnification, ⫻400.

found in the granulation tissue surrounding type 1 cells (48) or type 2 cells (which can dif-
some of the cavities. Since cavities are con- ferentiate into type 1 cells) (55). Many of the
nected to the bronchial tree, some of these nod- epithelial cells shown in Fig. 23 were vacuolated,
ules may have been bronchus-associated lym- suggesting that they were type 2 cells. Type 2
phoid tissue rather than nodules arising de novo cells secrete surfactant (mostly phospholipids),
from the infiltrating lymphocytes. and the secretory granules containing these
lipids produce the vacuolated appearance shown
Metaplastic Alveolar Epithelium and in Fig. 23.
Chemotaxis of Alveolar Macrophages. In Alveolar macrophages were present within
the outer layers of the tuberculous granulation the metaplastic alveoli (Fig. 23 and 24), as well
tissue of lesions 10 or more weeks old, the as in adjacent alveoli (see Fig. 5 in chapter 9).
epithelium of some of the alveoli and respiratory The alveolar macrophages frequently had a vac-
bronchioles was cuboidal and formed air-filled uolated appearance (Fig. 23), probably from the
saccules (Fig. 21 through 24). These saccules ingestion of surfactant. Chemotactic factors
probably arose when the alveolar connection to from both the infiltrating host cells and the
the airways was at least partly obstructed. tubercle bacilli themselves had evidently caused
The cuboidal epithelial cells that lined these alveolar macrophages to migrate into the sur-
saccules could be either metaplastic alveolar rounding alveolar spaces.The perifocal alveolar
84 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

FIGURE 18 A tissue section of a portion of the cavity wall. Bacilli can be seen in the clear
spaces (possibly lymphatics) found among the intact and disintegrating mature epithelioid cells.
Liquefied caseum is shown in the lower left corner of the photograph.The glycol methacry-
late-embedded tissue section (1 to 2 m thick) was stained with carbol-fuchsin and Giemsa.
Magnification, ⫻1,200.
In rabbits (and humans) with cavitary tuberculosis, tubercle bacilli enter the lymphatics and
the microvasculature. However, the native and acquired resistance of such hosts is usually so
high that metastatic lesions arising by the lymphogenous and hematogenous routes usually
fail to progress. Progressive metastatic pulmonary lesions arise in such resistant hosts when large
numbers of tubercle bacilli enter the airways from the cavity. In this case, many bacilli may
land in one site and may be difficult for the host to control.

macrophages occasionally contained ingested (reviewed in references 37 and 63). Injections of


tubercle bacilli that had entered the bronchial heat-killed M. vaccae did not convert the tuber-
tree from a cavity. culin skin test of healthy tuberculin-negative
humans (64).These properties enabled M. vac-
PART III. RECENT EXPERIMENTS cae to be used as an immunotherapeutic agent
ATTEMPTING TO REDUCE in clinical trials with favorable results (57–59,
LIQUEFACTION AND CAVITY 63, 65–70), although no effect was found in
FORMATION
some trials (63, 71, 72). To date, M. vaccae
Studies with Mycobacterium vaccae immunotherapy of tuberculosis has shown no
M. vaccae is a rapidly growing avirulent acid-fast adverse effects. However, further studies are
bacillus that was originally found free in the pas- indicated to determine its best role in the treat-
tures of cattle and in their milk (56). When ment of this disease (63, 72).
heat-killed and injected intradermally into In two experiments, we administered M. vac-
humans, M. vaccae apparently enhanced the CMI cae to rabbits with tuberculosis caused by the
response to virulent tubercle bacilli without inhalation of virulent bovine-type tubercle bacilli
appreciably increasing the DTH response (57– (37) in order to determine whether M. vaccae
60). M. vaccae increases the Th1 cytokine could reduce liquefaction and cavity formation.
response more than the Th2 response (61, 62) Autoclaved M. vaccae (strain NCTC 11659) was
4. LIQUEFACTION OF CASEOUS FOCI AND CAVITY FORMATION 䡵 85

TABLE 5 Number of tubercle bacilli cultured from the liquefied caseum in selected lesions from rabbits in
low-dose Experiment III at 33 weeksa
No. of viable
No. of primary lesions bacilli in approximately
Rabbit no. (caseous or liquefied) No. of cavities 10 l of liquefied caseum
in entire lung in entire lung from individual noncavitary
pulmonary lesionsb
#1 12 3 85,000
#2A, #B 8 17 A: 450
B: 300
#3 4 0 ⬍10
#4A, #B 10 4 A: 160
B: 600,000
#8 8 1 550
#10 10 0 ⬍1
#11 9 2 300
a
Adapted from reference 20.
b
Note that the number of viable bacilli in 10 l of liquefied caseum varies from ⬍1 to 600,000. Evidently, the bacilli must
change from a dormant state to one of extracellular growth, and the liquefied caseum must be of a composition to favor such growth.

FIGURE 19 A tissue section of a rabbit pulmonary tuberculous lesion (rabbit #4, Exper-
iment III) showing virulent bovine-type tubercle bacilli that had grown profusely in lique-
fied caseum. On the left, the lesion was probably beginning to cavitate, because some of the
spaces in the liquefied caseum seem larger than usual. Such profuse growth occurs only in some
lesions with liquefied centers (see Table 5), presumably where the composition of the lique-
fied caseum is most favorable and/or the adaptation of the bacillus to extracellular growth is
most complete. Stained with carbol-fuchsin, counterstained with methylene blue. Magnifi-
cation, ⫻600. Reprinted with permission from reference 37.
86 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

FIGURE 20 A tissue section of the wall of a rabbit pulmonary cavity (rabbit #5, Experi-
ment III) showing virulent bovine-type tubercle bacilli growing in the liquefied caseum.The
bacilli are growing more profusely near the lumen of the cavity (on the left) where the oxy-
gen tension is highest. Stained with carbol-fuchsin, counterstained with methylene blue. Mag-
nification, ⫻250. Reprinted with permission from reference 37.

injected intradermally into rabbits 3 or 5 times, the treatment of HIV infection that causes
4 or 5 weeks apart, and beginning 5 or 7 weeks AIDS. Among the HIV proteinase inhibitors,
after infection, respectively. Ritonavir is thought to be the most active
We found that M. vaccae had no significant against host cathepsin D. Cathepsin D is the
effect on cavity formation or on any other major glutamyl/aspartyl proteinase of rabbit
observable aspect of tuberculosis, including macrophages (41, 42).
tuberculin sensitivity (Table 4) (37). In these Ritonavir (50 mg/kg) was orally adminis-
rabbit M. vaccae experiments, no antimicrobial tered twice daily to five New Zealand White
drugs were given, whereas in the human trials, rabbits starting 5 weeks after the rabbits had
antimicrobials were simultaneously given.This inhaled sufficient virulent M. bovis (Ravenel) to
difference and others (such as the number of produce 50 to 100 primary pulmonary tuber-
individuals evaluated) may have contributed to cles. Diluent was administered to five control
our inability to confirm in rabbits the favorable rabbits that were similarly infected. One Riton-
results of M. vaccae immunotherapy in humans. avir rabbit and one control rabbit were eutha-
Reference 63 is a complete review of studies nized at 1 month after the beginning of the
employing M. vaccae, including descriptions of therapy, one treated and one control rabbit were
the various clinical trials. euthanized at 2 months, and the remaining three
treated rabbits and three controls were eutha-
Preliminary Studies with the nized at 3 months. At necropsy, rabbits treated
Proteinase Inhibitor Ritonavir with Ritonavir showed no apparent decrease in
Ritonavir (Abbott Laboratories, Inc., Chicago, the number of solid caseous tuberculous lesions
Ill.) is an aspartyl proteinase inhibitor used in that had liquefied. If there was such a decrease,
4. LIQUEFACTION OF CASEOUS FOCI AND CAVITY FORMATION 䡵 87

FIGURE 21 A tissue section of the wall of a rabbit pulmonary cavity (Experiment II) 18
weeks after the inhalation of about 5,000 virulent bovine-type tubercle bacilli.To the right is
the cavity’s air space. Next to it is the adjacent liquefied caseum. In the middle of the photo-
graph is the fibrous capsule.The area at the left contains alveoli (saccules) with metaplastic epithe-
lium and a few large macrophages in the air space. Between these metaplastic alveoli are
macrophages, plasma cells, lymphocytes, fibroblasts, and dilated microvessels (probably venules).
The glycol methacrylate-embedded tissue section was stained with Giemsa. Magnification, ⫻125.
Reprinted with permission from reference 20.

it was obscured by variations in the disease in the lungs of commercial New Zealand White
found among the rabbits. rabbits (20, 37) that was quite similar to the
This preliminary experiment (unpublished) form frequently found in adult human popula-
suggests that a combination of inhibitors of sev- tions (1, 2, 73–75).
eral hydrolytic enzymes will be necessary to Tubercle bacilli are inhibited in solid caseous
inhibit the liquefaction of solid caseous lesions, material before liquefaction occurs, but may
e.g., those that inhibit DNases, RNases, and/or grow to tremendous numbers after it occurs,
lipases in addition to various proteinases.Whether especially in cavities.The exact cause of lique-
or not such therapy would have adverse effects faction is not known. It seems to be a DTH
on host resistance remains to be determined. reaction to products of the bacilli.Also, the pro-
An alternative or additional approach would be teinases, nucleases, and lipases from live (76)
to increase the CMI/DTH ratio by an appro- and dead macrophages seem to be involved.
priate vaccine, such as M. vaccae (described in the These hydrolases apparently diffuse into solid
preceding section). caseum from the periphery, and inhibitors of
these hydrolases that are present in the solid
SUMMARY OF CAVITY FORMATION caseum apparently disappear (see Fig. 25 and 26)
AND SUGGESTIONS FOR ITS (1, 3, 7a, 18).
PREVENTION To prevent liquefaction and cavity forma-
An aerosol of bovine-type tubercle bacilli (of tion, we highly recommend that multiple hydro-
slightly reduced virulence) readily produces a lase inhibitors be developed and evaluated for
slowly progressing cavitary form of the disease effectiveness and toxicity in the rabbit cavitary
88 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

FIGURE 22 Another portion of the same cavity as in Fig. 21, shown at higher magnifica-
tion to identify the types of cells present.A thick fibrous capsule (upper half) surrounds the liq-
uefied caseum (below).The capsule contains many fibroblasts, as well as some macrophages, lym-
phocytes, and plasma cells.An oval metaplastic alveolus and a small dilated blood vessel are present
on the right. Upon fixation, liquefied caseum often contains spaces between the coagulated pro-
teins, whereas solid caseum usually does not.
On gross examination, all gradations of caseum fluidity were found among the various lesions.
The gross examination is a more accurate test of fluidity, because fixation solidifies the protein-
rich material. The glycol methacrylate-embedded tissue section was stained with Giemsa.
Magnification, ⫻250.

FIGURE 23 An alveolus (saccule) with metaplastic


epithelium containing alveolar macrophages in the wall of
the same cavity shown in Fig. 21 and 22. Many of the alve-
olar epithelial cells are vacuolated, and the macrophages
within this alveolus are also vacuolated (see text). Nearby
are areas rich in lymphocytes and plasma cells, containing
dilated capillaries or venules. Accumulations of plasma
cells and dilated microvessels are frequent components of
tuberculous granulation tissue.The glycol methacrylate-
embedded tissue section (cut 1 to 2 m) was stained with
Giemsa. Magnification, ⫻350.
Metaplastic alveoli probably arise from partial obstruc-
tion of the connecting bronchus.They no longer func-
tion in the oxygenation of blood.
FIGURE 24 A tissue section
of a rabbit pulmonary cavity wall
(Experiment II) 18 weeks after the
inhalation of about 5,000 virulent
bovine-type tubercle bacilli. The
lumen of the cavity is at the right.
Less fibrosis is present than that in
Fig. 21 and 22, and the metaplastic
alveoli are closer to the cavity
lumen. Several dark acid-phos-
phatase-positive macrophages are
present in the cavity wall, as well as
in the lumen of the metaplastic
alveoli. The glycol methacrylate-
embedded tissue section (cut 1 to 2
m) was stained for acid phos-
phatase and counterstained with
Giemsa. Magnification, ⫻210.

FIGURE 25 A tissue section of a rabbit dermal BCG lesion, 28 days of age, immunostained for cathepsin D, a
major macrophage proteinase.The caseous center has liquefied and ulcerated.The viable and dead macrophages sur-
rounding the liquefied caseum contain high levels of cathepsin D.The dead macrophages within the liquefied caseum
and the viable macrophages in the more peripheral tuberculous granulation tissue contain much-reduced levels of
this proteinase. Stained with goat antiserum to cathepsin D, rabbit anti-goat immunoglobulin (in excess), horseradish
peroxidase–goat antiperoxidase complex, hydrogen peroxide, and 3-amino-9-ethylcarbazole. No counterstain. Mag-
nification, ⫻100. Reprinted with permission from reference 21.
Note how closely this ulcerating dermal BCG lesion resembles the pulmonary cavitary lesion that was produced
by an aerosol of virulent bovine-type tubercle bacilli and shown in Fig. 14. Cathepsin D has a similar distribution
in both lesions. Experiments to study factors involved in liquefaction and cavity formation, as well as drugs to inhibit
such factors, could be performed with dermal BCG lesions and then confirmed with virulent tubercle bacilli in
the lungs.

89
90 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

FIGURE 26 A tissue section of a rabbit dermal BCG lesion, 28 days of age, stained for the
lysosomal enzyme -galactosidase, our marker for activated macrophages. As in Fig. 25, the
caseous center has liquefied and ulcerated.The viable and dead macrophages surrounding the liq-
uefied caseum (in the top third of the photograph) contain the highest levels of -galactosidase.
The dead macrophages within the liquefied caseum contain little -galactosidase, and the viable
macrophages in the peripheral tuberculous granulation tissue (in the lower half of the photograph)
show reduced levels of this hydrolytic enzyme. Stained with 5-bromo-4-chloro-3-indolyl--D-
galactoside, lightly counterstained with hematoxylin. Magnification, ⫻100. Reprinted with per-
mission from reference 21.
This figure demonstrates that macrophages at the edge of liquefying caseum become highly
activated for lysosomal enzymes. Lysosomal proteinases, nucleases, and lipases seem to be major
causes of liquefaction.

model of tuberculosis. Inhibitors of cavity for- growth would be a welcome addition to our
mation would be useful in the treatment of control of this disease.
both drug-susceptible and drug-resistant tuber- Another approach to the control of tubercu-
culosis and would reduce the spread of this dis- losis would be to identify the genes that enable
ease in human populations. tubercle bacilli to change from a dormant state
Also, vaccines that increase the CMI/DTH to an actively multiplying state. Then, drugs
ratio, i.e., cause a reduction in DTH with respect could be developed to prevent this occurrence.
to CMI, may not only prevent clinical tuber-
culosis, but may also reduce the amount of cav- A PROPOSED METHOD
itation should such clinical disease occur. See M. TO STUDY LIQUEFACTION
vaccae studies (described above) and chapter 22. AND CAVITY FORMATION
The composition of liquefied caseum that Research on liquefaction and cavity formation
enables tubercle bacilli to multiply extracellularly in tuberculosis has been impeded by the lack
is not fully known. Drugs that prevent a com- of a convenient method to produce these
position favorable to extracellular bacillary phenomena. Here is one suggestion that may
4. LIQUEFACTION OF CASEOUS FOCI AND CAVITY FORMATION 䡵 91

circumvent this difficulty. It may be a break- variations among rabbits in skin composition
through or may be perfectly worthless, but and in their inflammatory and immune responses
because it is simple, it should be evaluated. may prevent the development of a workable assay
system. Nevertheless, the quantitative dermal
Premise. (i) Liquefaction and ulceration in response to tubercle bacilli should be evaluated
the skin are surrogates for liquefaction and cav- because it may greatly hasten the ability to under-
ity formation in the lungs. (ii) These phenom- stand and inhibit liquefaction and cavity forma-
ena mainly occur after a threshold number of tion,which are the major processes responsible for
tubercle bacilli is reached. the perpetuation of tuberculosis in humans.
Liquefaction and ulceration in the skin could
be used to evaluate drugs that inhibit the host REFERENCES
enzymes and/or the bacillary components that 1. Rich, A. R. 1951. The Pathogenesis of Tuberculo-
may cause liquefaction and cavities.An increase sis, 2nd ed. Charles C Thomas Publisher, Spring-
in the number of bacilli required to produce liq- field, Ill.
uefaction in the skin would be a measure of the 2. Canetti, G. 1955. The Tubercle Bacillus in the Pul-
potential efficacy of the procedure. If estab- monary Lesion of Man: Histobacteriology and Its Bear-
ing on the Therapy of Pulmonary Tuberculosis. Springer
lished, this intradermal method would be a con- Publishing Co., New York, N.Y.
venient, sensitive, and quantitative measure of the 3. Long, E. R. 1958. The Chemistry and Chemo-
host’s ability to liquefy caseum and form pul- therapy of Tuberculosis: a Compilation and Critical
monary cavities. Review of Existing Knowledge on the Chemistry of
Similarities of ulceration in dermal BCG Tubercle Bacilli and Their Products, Chemical Changes
and Processes in the Host, and Chemical Aspects of the
lesions and cavity formation in pulmonary lesions Treatment of Tuberculosis, 3rd ed., p. 52–61, 163–
produced by virulent tubercle bacilli are dis- 182. Lippincott Williams & Wilkins, Philadel-
cussed above (p. 68). Figures 25 and 26 show that phia, Pa.
hydrolase distribution is apparently the same in 4. Lurie, M. B. 1964. Resistance to Tuberculosis: Exper-
both pulmonary and dermal cavitary lesions. imental Studies in Native and Acquired Defensive
Mechanisms. Harvard University Press, Cambridge,
Mass.
Procedure. Live and dead BCG, dead vir- 5. Lurie, M. B., and A. M. Dannenberg, Jr.
ulent human-type tubercle bacilli (Erdman or 1965. Macrophage function in infectious disease
H37Rv), and dead bovine-type tubercle bacilli with inbred rabbits. Bacteriol. Rev. 29:466–476.
(Ravenel) should be injected intradermally into 6. Dannenberg, A. M., Jr., and J. F. Tomashef-
ski, Jr. 1998. Pathogenesis of pulmonary tuber-
rabbits in a range of doses. The lowest dose culosis, p. 2447–2471. In A. P. Fishman (ed.), Fish-
required to produce liquefaction and ulcera- man’s Pulmonary Diseases and Disorders, 3rd ed.,
tion at the site of injection could then be deter- vol. 2. McGraw-Hill Book Co., New York, N.Y.
mined. Initially, each bacillary strain could be 7. Dannenberg, A. M., Jr. 1994. Rabbit model of
used on a single rabbit in a wide range of doses, tuberculosis, p. 149–156. In B. R. Bloom (ed.),
Tuberculosis: Pathogenesis, Protection and Control.
and the results could be confirmed in several ASM Press,Washington, D.C.
rabbits with fewer doses near the threshold con- 7a. Dannenberg, A. M., Jr., and M. Sugimoto.
centration. The experiment could then be 1997. Liquefaction of caseous foci in tuberculo-
repeated in a biological level 3 facility with sis. Am. Rev. Respir. Dis. 113:257–259. (Editorial.)
intradermal live virulent tubercle bacilli, and 8. Yamamura,Y., S.Yasaka, M.Yamaguchi, K.
Endo, H. Iwakura, S. Nakamura, and Y.
finally confirmed in a more analogous system, Ogawa. 1954. Studies on the experimental tuber-
namely, in rabbits inhaling aerosolized live vir- culous cavity. Med. J. Osaka Univ. 5:187–197.
ulent tubercle bacilli. 9. Yamamura,Y. 1958.The pathogenesis of tuber-
culous cavities. Adv.Tuberc. Res. 9:13–37.
Caveats. Liquefaction and ulceration of 10. Yamamura,Y.,Y. Ogawa, H.Yamagata, and
Y.Yamamura. 1974. Prevention of tuberculous
tuberculous lesions in the skin should reflect liq- cavity formation by desensitization with tuber-
uefaction and cavitation in the lungs because the culin-active peptide. Am. Rev. Respir. Dis. 109:
same host response occurs in each. However, 594–601.
92 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

11. Kruml, J., L.Trnka, R. Urbancík, and J. Kuska. ically in dermal BCG lesions. J. Immunol. 100:931–
1965. Histologic differences between experimen- 941.
tal cavernous tuberculosis in rabbits and human 23. Tsuda,T.,A. M. Dannenberg, Jr., M.Ando, O.
cavitary disease. Am. Rev. Respir. Dis. 92:299–302. Rojas-Espinosa, and K. Shima. 1974. Enzymes
12. Yamamura,Y.,Y. Ogawa, H.Yamagata, and Y. in tuberculous lesions hydrolyzing protein,
Yamamura. 1968. Prevention of tuberculous cav- hyaluronic acid and chondroitin sulfate: a study of
ity formation by immunosuppressive drugs. Am. isolated macrophages and developing and healing
Rev. Respir. Dis. 98:720–723. rabbit BCG lesions with substrate film techniques;
13. Pagel,W. 1936.The role of the bacillus and of “het- the shift of enzyme pH optima towards neutrality
eroallergy” in tuberculous liquefaction. J. Pathol. in “intact” cells and tissues. J. Reticuloendothel. Soc.
Bacteriol. 42:417–424. 16:220–231.
14. Sugisaki, K., A. M. Dannenberg, Jr., Y. Abe, 24. Daoust, R., and H. Amano. 1961. Improved
J. Tsuruta, W.-J. Su, W. Said, L. Feng, DNA film method for localizing DNAase activity
T.Yoshimura, P. J. Converse, and P. Mounts. in tissue sections. Exp. Cell. Res. 24:559–564.
1998. Non-specific and immune-specific up- 25. Daoust, R. 1966. Modified procedure for the his-
regulation of cytokines in rabbit dermal tubercu- tochemical localization of ribonuclease activity by
lous (BCG) lesions. J. Leukoc. Biol. 63:440–450. the substrate film method. J. Histochem. Cytochem.
15. Shigenaga, T., A. M. Dannenberg, Jr., D. B. 14:254–259.
Lowrie, W. Said, M. J. Urist, H. Abbey, B. H. 26. Daoust, R. 1965. Histochemical localization of
Schofield, P. Mounts, and K. Sugisaki. 2001. enzyme activities by substrate film methods: ribonu-
Immune responses in tuberculosis: antibodies and cleases, deoxyribonucleases, proteases, amylase, and
CD4/CD8 lymphocytes with vascular adhesion hyaluronidase. Int. Rev. Cytol. 18:191–221.
molecules and cytokines (chemokines) cause a rapid 27. Cunningham, L. 1967. Histochemical observa-
antigen-specific cell infiltration at sites of bacillus Cal- tions of the enzymatic hydrolysis of gelatin films.
mette-Guérin reinfection. Immunology 102:466–479. J. Histochem. Cytochem. 15:292–298.
16. Dannenberg, A. M., Jr. 1968. Cellular hyper-
28. Oshima,Y.,T. Hagihara,Y. Mizuta, S. Hattori,
sensitivity and cellular immunity in the patho-
and J. Mishima. 1955. Histochemical study on the
genesis of tuberculosis: specificity, systemic and local
relationship between tuberculosis and lipase activ-
nature, and associated macrophage enzymes. Bacte-
ity. I. Lipase and lipids of tuberculous lesions.
riol. Rev. 32:85–102.
Kekkaku 30:716–720.
17. Weiss, C., and M. L. Boyar-Manstein. 1951. On
the mechanism of liquefaction of tubercles. I.The 29. Umezawa, H., T. Aoyagi, H. Morishima, M.
behavior of endocellular proteinases in tubercles Matsuzaki, M. Hamada, and T.Takeuchi. 1970.
developing in the lungs of rabbits. Am. Rev.Tuberc. Pepstatin, a new pepsin inhibitor produced by actin-
63:694–705. omycetes. J.Antibiot. 23:259–262.
18. Weiss, C., and F. M. Singer. 1953 Mechanism of 30. Aoyagi, T., S. Kunimoto, H. Morishima, T.
softening of tubercles. II. Behavior of desoxyri- Takeuchi, and H. Umezawa. 1971. Effect of
bonuclease in tubercles developing in the lungs of Pepstatin on acid proteases. J. Antibiot. (Tokyo)
rabbits. Arch. Pathol. 55:516–530. 24:687–694.
19. Weiss, C., J. Tabachnick, and H. P. Cohen. 31. Umezawa, H. 1972. Enzyme Inhibitors of Microbial
1954. Mechanism of softening of tubercles. III. Hy- Origin. University Park Press, Baltimore, Md.
drolysis of protein and nucleic acid during anaer- 32. McAdoo, M. H., A. M. Dannenberg, Jr., C. J.
obic autolysis of normal and tuberculous lung tis- Hayes, S. P. James, and J. H. Sanner. 1973. Inhi-
sue in vitro. Arch. Pathol. 57:179–193. bition of the cathepsin D-type proteinase of
20. Converse, P. J., A. M. Dannenberg, Jr., J. E. macrophages by pepstatin, a specific pepsin
Estep, K. Sugisaki,Y.Abe, B. H. Schofield, and inhibitor, and other substances. Infect. Immun. 7:655–
M. L. M. Pitt. 1996. Cavitary tuberculosis pro- 665.
duced in rabbits by aerosolized virulent tubercle 33. Dannenberg, A. M., Jr., P. C.Walter, and F. A.
bacilli. Infect. Immun. 64:4776–4787. Kapral. 1963. A histochemical study of phago-
21. Rojas-Espinosa, O., A. M. Dannenberg, Jr., cytic and enzymatic functions of rabbit mononu-
L. A. Sternberger, and T.Tsuda. 1974. Role of clear and polymorphonuclear exudate cells and
cathepsin D in the pathogenesis of tuberculosis. A alveolar macrophages. II. The effect of particle
histochemical study employing unlabeled antibod- ingestion on enzyme activity; two phases of in vi-
ies and the peroxidase-antiperoxidase complex. Am. tro activation. J. Immunol. 90:448–465.
J. Pathol. 74:1–17. 34. Grosset, J. 2003. Mycobacterium tuberculosis in the
22. Dannenberg, A. M., Jr., O. T. Meyer, J. R. extracellular compartment: an underestimated
Esterly, and T. Kambara. 1968.The local nature adversary. Antimicrob. Agents Chemother. 47:833–
of immunity in tuberculosis, illustrated histochem- 836.
4. LIQUEFACTION OF CASEOUS FOCI AND CAVITY FORMATION 䡵 93

35. Yoder, M. A., G. Lamichhane, and W. R. 47. Lurie, M. B. 1941. Heredity, constitution, and
Bishai. 2004. Cavitary tuberculosis: the “Holey tuberculosis, an experimental study. Am. Rev.Tuberc.
Grail” of disease transmission. Curr. Sci. 86:74–81. 44:1–125.
36. Helke, K. L., J. L. Mankowski, and Y. C. Man- 48. Lurie, M. B. 1932. The correlation between the
abe. 2005. Animal models of cavitation in pul- histological changes and the fate of living tubercle
monary tuberculosis. Tuberculosis (Edinburgh), in bacilli in the organs of tuberculous rabbits. J. Exp.
press. Med. 55:31–54.
37. Converse, P. J., A. M. Dannenberg, Jr.,T. Shi- 49. Yamori, T. 1964. On phagocytes; their structures
genaga, D. N. McMurray, S. W. Phalen, J. L. and participation in inflammation. Acta Pathol. Jpn.
Stanford, G. A. W. Rook, T. Koru-Sengul, H. 14:1–43.
Abbey, J. E. Estep, and M. L. M. Pitt. 1998. Pul- 50. Namba, M., A. M. Dannenberg, Jr., and F.
monary bovine-type tuberculosis in rabbits: bacil- Tanaka. 1983. Improvement in the histochemical
lary virulence, inhaled dose effects, tuberculin sen- demonstration of acid phosphatase, -galactosidase
sitivity, and Mycobacterium vaccae immunotherapy. and nonspecific esterase in glycol methacrylate tis-
Clin. Diagn. Lab. Immunol. 5:871–881. sue sections by cold temperature embedding. Stain
38. Lurie, M. B., P. Zappasodi, and C. Tickner. Technol. 58:207–213.
1955. On the nature of genetic resistance to tuber- 51. Ando, M., A. M. Dannenberg, Jr., M. Sugi-
culosis in the light of the host-parasite relationships moto, and B. S. Tepper. 1977. Histochemical
in natively resistant and susceptible rabbits. Am. studies relating the activation of macrophages to the
Rev.Tuberc. Pulm. Dis. 72:297–323. intracellular destruction of tubercle bacilli. Am. J.
39. Manabe, Y. C., A. M. Dannenberg, Jr., S. K. Pathol. 86:623–634.
Tyagi, C. L. Hatem, M.Yoder, S. C.Woolwine, 52. Cohn, Z. A., and B. Benson. 1965.The in vitro
B. C. Zook, M. L. M. Pitt, and W. R. Bishai. differentiation of mononuclear phagocytes. III.The
2003. Different strains of Mycobacterium tuberculosis reversibility of granule and hydrolytic enzyme for-
cause various spectrums of disease in the rabbit mation and the turnover of granule constituents.
model of tuberculosis. Infect. Immun. 71:6004–6011. J. Exp. Med. 122:455–466.
40. Suga, M., A. M. Dannenberg, Jr., and S. 53. Higuchi, S., M. Suga, A. M. Dannenberg, Jr.,
Higuchi. 1980. Macrophage functional hetero- and B. H. Schofield. 1979. Histochemical demon-
geneity in vivo: macrolocal and microlocal stration of enzyme activities in plastic- and paraffin-
macrophage activation, identified by double-stain- embedded tissue sections. Stain Technol. 54:5–12.
ing tissue sections of BCG granulomas for pairs of 54. Vogt, R. F., Jr., N. A. Hynes, A. M. Dannen-
enzymes. Am. J. Pathol. 99:305–324. berg, Jr., S. Castracane, and L. Weiss. 1983.
41. Dannenberg,A. M., Jr., and E. L. Smith. 1955. Improved techniques using Giemsa-stained glycol
Proteolytic enzymes of lung. J. Biol. Chem. 215:545– methacrylate tissue sections to quantitate basophils
554. and other leukocytes in inflammatory skin lesions.
42. Dannenberg,A. M., Jr., and E. L. Smith. 1955. Stain Technol. 58:193–205.
Action of proteinase I of bovine lung. Hydrolysis of 55. Simon, R. H., and R. Paine III. 1995. Partici-
the oxidized B chain of insulin; polymer formation pation of pulmonary alveolar epithelial cells in lung
from amino acid esters. J. Biol. Chem. 215:55–66. inflammation. J. Lab. Clin. Med. 126:108–118.
43. Rojas-Espinosa, O., A. M. Dannenberg, Jr., 56. Tsukamura, M. 1984. The “non-pathogenic”
P. A. Murphy, P. A. Straat, P. C. Huang, and species of mycobacteria: their distribution and
S. P. James. 1973. Purification and properties of the ecology in non-living reservoirs, p. 1339–1359.
cathepsin D-type proteinase from beef and rabbit In G. P. Kubica and L. G.Wayne (ed.), The Myco-
lung and its identification in macrophages. Infect. bacteria: a Sourcebook. Marcel Dekker, Inc., New
Immun. 8:1000–1008. York, N.Y.
44. Allison, M. J., P. Zappasodi, and M. B. Lurie. 57. Corlan, E., C. Marica, C. Macavei, J. L. Stan-
1962. Host-parasite relationships in natively resis- ford, and C. A. Stanford. 1997. Immunotherapy
tant and susceptible rabbits on quantitative inhala- with Mycobacterium vaccae in the treatment of tuber-
tion of tubercle bacilli. Am. Rev. Respir. Dis. 85:553– culosis in Romania. 2. Chronic or relapsed disease.
569. Respir. Med. 91:21–29.
45. Ratcliffe, H. L., and W. F.Wells. 1948.Tubercu- 58. Onyebujoh, P.,T.Abdulmumini, S. Robinson,
losis of rabbits induced by droplet nuclei infec- G. A. W. Rook, and J. L. Stanford. 1995.
tion. II. Response to reinfection. J. Exp. Med. Immunotherapy with Mycobacterium vaccae as an
87:585–594. addition to chemotherapy for the treatment of pul-
46. Ratcliffe, H. L., and W. F.Wells. 1948.Tubercu- monary tuberculosis under difficult conditions in
losis of rabbits induced by droplet nuclei infec- Africa. Respir. Med. 89:199–207.
tion. I. Initial response to infection. J. Exp. Med. 59. Stanford, J. L., C. A. Stanford, G. A.W. Rook,
87:575–584. and J. M. Grange. 1994. Immunotherapy for
94 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

tuberculosis: investigative and practical aspects. Clin. 68. Corlan, E., C. Marcia, C. Macacei, J. L. Stan-
Immunother. 1:430–440. ford, and C. A. Stanford. 1997. Immunotherapy
60. Stanford, J. L., G. A. W. Rook, G. M. Bahr,Y. with Mycobacterium vaccae in the treatment of tuber-
Dowlati, R. Ganapati, K. Ghazi-Saidi, S. culosis in Romania. 1. Newly-diagnosed pulmonary
Lucas, G. Ramu, P.Torres, H. Minh-Ly, and N. disease. Respir. Med. 91:13–19.
Anstey. 1990. Mycobacterium vaccae in immunopro- 69. Johnson, J. L., R. M. Kamya,A. Okwera,A. M.
phylaxis and immunotherapy of leprosy and tuber- Loughlin, S. Nyole, D. L. Hom, R. S. Wallis,
culosis. Vaccine 8:525–530. C. S. Hirsch, K. Wolski, J. Foulds, R. D.
61. Dlugovitzky, D., O. Bottasso, J. C. Dominino, Mugerwa, and J. J. Ellner. 2000. Randomized
E. Valentini, R. Hartopp, M. Singh, C. Stan- controlled trial of Mycobacterium vaccae immuno-
ford, and J. Stanford. 1999. Clinical and serolog- therapy in non-human immunodeficiency virus-
ical studies of tuberculosis patients in Argentina infected Ugandan adults with newly diagnosed pul-
receiving immunotherapy with Mycobacterium vaccae monary tuberculosis. J. Infect. Dis. 181:1304–1312.
(SRL 172). Respir. Med. 93:557–562. 70. Davies, P. D. O. 2001. New hope for treatment of
62. Grange, J. 1997. Immunotherapy: a new strategy multi-drug-resistant tuberculosis? Respir. Med.
for tuberculosis control? Respir. Med. 91:1–4. 95:435–436. (Editorial.)
63. Stanford, J., C. Stanford, and J. Grange. 2004. 71. Durban Immunotherapy Trial Group. 1999.
Immunotherapy with Mycobacterium vaccae in the Immunotherapy with Mycobacterium vaccae in
treatment of tuberculosis. Front. Biosci. 9:1701– patients with newly diagnosed pulmonary tuber-
1719. culosis: randomized controlled trial. Lancet 354:116–
64. von Reyn, C. F., R. D. Arbeit, G. Yeaman, 119.
R. D. Waddell, B. J. Marsh, P. Morin, J. F. 72. Fourie, P. B., J. J. Ellner, and J. L. Johnson.
Modlin, and H. G. Remold. 1997. Immunization 2002.Whither Mycobacterium vaccae—encore? Lancet
of healthy adult subjects in the United States with 360:1032–1033.
inactivated Mycobacterium vaccae administered in a 73. Medlar, E. M. 1955.The behavior of pulmonary
three-dose series. Clin. Infect. Dis. 24:843–848. tuberculous lesions: a pathological study. Am. Rev.
65. Stanford, J. L., and J. M. Grange. 1993. New Tuberc. 71(Part II):1–244.
concepts for the control of tuberculosis in the 74. Poole, J. C. F., and H.W. Florey. 1970. Chronic
twenty first century. J. R. Coll. Physicians (London) inflammation and tuberculosis, p. 1183–1224. In
27:218–223. H. W. Florey (ed.), General Pathology, 4th ed. The
66. Stanford, J. L., and C. A. Stanford. 1996. W. B. Saunders Co., Philadelphia, Pa.
Immunotherapy for tuberculosis with M. vaccae. 75. Sutinen, S. 1968. Evaluation of activity in tuber-
J. Med. Microbiol. 44:24–34. culous cavities of the lung: a histopathologic and
67. Stanford, J. L., C. A. Stanford, J. M. Grange, bacteriologic study of resected specimens with clin-
N. N. Lan, and A. Etemadi. 2001. Does ical and roentgenographic correlations. Scand. J.
immunotherapy with heat-killed Mycobacterium vac- Respir. Dis. Suppl. 67:1–78.
cae offer hope for the treatment of multi-drug- 76. Ackerman, N. R., and J. R. Beebe. 1974.
resistant pulmonary tuberculosis? Respir. Med. 95: Release of lysosomal enzymes by alveolar mononu-
444–447. clear cells. Nature 247:475–477.
Section 2.

IMMUNOLOGY OF TUBERCULOSIS
This page intentionally left blank
5
DELAYED-TYPE HYPERSENSITIVITY,
CELL-MEDIATED IMMUNITY, AND
ANTIBODIES IN TUBERCULOSIS

Overview of innate immunity in tuberculosis 98


Delayed-type hypersensitivity in tuberculosis 98
Cell-mediated immunity in tuberculosis 99
DTH and CMI similarities in tuberculosis 99
The most favorable CMI/DTH ratios in tuberculosis 100
CMI and DTH are both required to arrest tuberculous lesions 101
Acquired cellular resistance in tuberculosis 102
Local nature of DTH and CMI; organ resistance 102
Systemic immunity in tuberculosis 104
Macrophage activation 104
Synergism of DTH and CMI in arresting tuberculosis: does true dormancy
exist? 104
The size of the tuberculin skin reaction has no prognostic value 105
The “booster” phenomenon in repeated tuberculin testing 105
Lurie’s eye chamber experiments elucidating the factors involved in
CMI 107
Duration and specificity of ACR and its recall upon reinfection 111
Innate immunity and its relation to acquired (adaptive) immunity 112
Nonspecific and antigen-specific immune responses in innate and adaptive
immunity 114

Abstract. Both delayed-type hypersensitivity (DTH) and cell-mediated immunity (CMI)


are T-lymphocyte responses to bacillary antigens presented mainly by dendritic cells. In
tuberculous lesions, DTH kills (nonactivated) macrophages that contain numerous tuber-
cle bacilli when these bacilli release tissue-damaging local concentrations of tuberculin-
like products. In the resulting (solid) caseous necrosis, bacillary growth is inhibited and many
bacilli die because of low oxygen tension and other factors.Therefore, tissue-damaging DTH
has apparently evolved in mammals to stop continuing bacillary growth within the non-
activated macrophages that have permitted such growth.
In tuberculous lesions, CMI activates macrophages so that they can inhibit and destroy
ingested tubercle bacilli. DTH can also activate macrophages if only low local concentrations
of tuberculin-like products are present. In this respect, DTH (the host reaction to tuber-
culin-like products) is part of the CMI response.
Nonactivated macrophages continuously enter every tuberculous lesion and may ingest
tubercle bacilli (see chapter 10).To stop the progression of the disease, macrophages con-
taining a few bacilli must be activated by CMI to prevent further intracellular bacillary mul-
tiplication, and macrophages containing many bacilli must be killed by the DTH process.
The interplay of CMI and tissue-damaging DTH seems to explain the entire spectrum
of the disease found in tuberculous hosts.
Both DTH and CMI exert their control locally.Their main systemic manifestation is
to provide an expanded antigen-specific lymphocyte population to infiltrate local sites of
bacillary lodgement.
Antibodies that aid phagocytosis apparently play little or no role in the destruction
of the tubercle bacillus.The bacillus readily enters macrophages without being opsonized
by antibodies and evidently can multiply intracellularly within nonactivated macrophages
in the presence of antibodies. However, in immunized hosts, antibodies seem to be an
important host defense against the development of clinically apparent tuberculosis.

97
98 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

Antigen-antibody reactions at sites of bacillary lodgement result in the production of


chemotactic factors, including the C5a component of complement. In immunized
hosts, such chemotaxins cause a rapid local accumulation of dendritic cells, macrophages,
and antigen-specific T cells—all of which would accumulate more slowly without
the local antigen-antibody reaction. In other words, in immunized hosts the antigen-
antibody reaction enables the local cell-mediated immune response to occur so rapidly
that the bacillus is often inhibited before it multiplies extensively.

OVERVIEW OF INNATE IMMUNITY of hours and reaches a peak in 1 to 3 days.


IN TUBERCULOSIS Because of the slow development, this antigen-
In rabbits and humans, the innate immunity of specific reaction is called “delayed” (2). A low
pulmonary alveolar macrophages plays an concentration of tuberculin (as used in skin
important role in preventing the establishment testing) usually causes a local cell infiltration
of lesions by inhaled tubercle bacilli.The alve- without necrosis in the sensitized host, but high
olar macrophage population is nonspecifically concentrations will cause necrosis, especially if
(innately) activated. These macrophages can the host possesses high levels of DTH. Because
destroy many inhaled virulent human-type antigen-specific antibodies are almost always
tubercle bacilli before a tuberculous lesion is present in tuberculin-positive hosts, typical cell-
established. However, if the bacillus multiplies, mediated tuberculin reactions usually contain
an early lesion is established. Such a lesion con- an Arthus component (see glossary) caused by
tains dendritic cells, macrophages, and lympho- antigen-antibody complexes and complement
cytes. The dendritic cells have innate pattern activation.
recognition receptors (PRR) that induce co- Only a rough correlation exists between the
stimulatory factors for antigen presentation (to size of the dermal reaction to tuberculin and the
lymphocytes) and direct the type of adaptive amount of caseation found in the pulmonary
(acquired) immune response that results. Details tuberculous lesions. Caseous necrosis in the
on innate immunity and the relation of innate lesions is a response to the local concentration
immunity to acquired immunity are presented of tuberculin-like products of the bacilli.These
at the end of this chapter. products increase as the bacilli in the lesions
increase.The size of the dermal tuberculin reac-
DELAYED-TYPE HYPERSENSITIVITY tion represents the overall systemic sensitivity of
IN TUBERCULOSIS the host to a standard amount of tuberculin.
In tuberculosis, delayed-type hypersensitivity DTH is an important host mechanism for
(DTH) (see glossary) is characterized by an stopping the multiplication of tubercle bacilli
expanded Th1 lymphocyte population that within the nonactivated macrophages that con-
responds to extremely small concentrations of the tinually enter tuberculous lesions from the
tuberculin-like bacillary products (1). At sites bloodstream. DTH kills macrophages when-
where the bacilli lodge, these Th1 lymphocytes ever they contain more tubercle bacilli than
produce cytokines that cause mononuclear cell cell-mediated immunity (CMI) (i.e., the acti-
infiltration, followed (in humans, guinea pigs, and vation of macrophages) can control. Tubercle
rabbits) by caseous necrosis as soon as the local bacilli cannot multiply in the solid caseous
concentration of tuberculin-like products in- necrosis that forms from the dead macrophages
creases to damaging levels. and the surrounding tissues (see chapter 2).
When applied to tuberculosis, the terms Therefore, DTH is a fail-safe immune mecha-
DTH and tuberculin sensitivity are used inter- nism that apparently developed throughout
changeably. In hosts sensitized by a previous mammalian evolution to protect the host when-
exposure to the antigens of tubercle bacilli, the ever CMI was inadequate.
intradermal injection of tuberculin produces a The term tissue-damaging DTH is used for
skin reaction that slowly develops over a period the reaction that causes such necrosis.This form
5. INNATE AND ACQUIRED IMMUNITY IN TUBERCULOSIS 䡵 99

of DTH develops in tuberculous lesions when- Figure 1 illustrates the basic principles of
ever tuberculin-like antigens from the bacillus CMI. Macrophages are shown in various stages
reach cytotoxic concentrations (see reference of activation. Such activation is caused by the
3).The host could be sensitive to many bacillary cytokines of antigen-stimulated lymphocytes
antigens besides those in standard tuberculin (called lymphokines in this figure). Only the
preparations (4–12).Therefore, in this book, I use macrophages that are highly activated seem to be
the term tuberculin-like to include all antigens capable of inhibiting or killing fully virulent
that produce a DTH reaction in the host.The tubercle bacilli. Not included in the figure are
main characteristic of a tuberculin-like antigen antigen-presenting cells (APCs) (dendritic cells),
is its ability to cause macrophage and lympho- which process the antigens of tubercle bacilli
cyte infiltration in very low concentrations and and present them to lymphocytes with specific
to cause tissue necrosis at somewhat higher con- receptors for these antigens (see chapter 6).
centrations. One tuberculin unit contains Whether a nonactivated macrophage con-
0.00002 mg of purified protein derivative (PPD) taining only 1 to 4 bacilli can be activated suf-
(see glossary). ficiently to prevent further intracellular bacillary
In brief, DTH and CMI are similar immune growth is not known, but macrophages within
reactions to the antigens of the tubercle bacil- which the bacilli have multiplied to 10 or more
lus, but the concentrations of the antigens that could never do so. In this case, tissue-damaging
elicit DTH are far lower than those that elicit DTH is required to kill this macrophage. To
CMI. prevent the intracellular multiplication of viru-
lent tubercle bacilli, macrophages in tuberculous
CELL-MEDIATED IMMUNITY IN lesions may have to be activated prior to the
TUBERCULOSIS ingestion of such bacilli. If so, a major compo-
In tuberculosis, CMI, like DTH, is characterized nent of the CMI response would be the local
by an expanded Th1 lymphocyte population, accumulation of large numbers of highly acti-
but, as stated above, the CMI population is not vated macrophages that are ready to ingest
as sensitive as the DTH population to bacillary tubercle bacilli released from macrophages killed
products. In other words, the local concentra- by tissue-damaging DTH (Fig. 2 and 3).
tions of antigens that produce CMI rarely get
high enough to cause necrosis (1). DTH AND CMI SIMILARITIES IN
CMI-producing lymphocytes enter sites TUBERCULOSIS
where bacilli are located and, via their cyto- DTH is really a form of CMI (2). By conven-
kines, activate macrophages to inhibit or destroy tion, the ability of antigen-specific Th1 lym-
these bacilli. Gamma interferon (IFN-) and phocytes to cause a more rapid infiltration of
tumor necrosis factor alpha (TNF-) are major dendritic cells, macrophages, and lymphocytes is
macrophage-activating cytokines (reviewed in called delayed-type hypersensitivity, and the
references 13, 14, and 15). IFN- also induces ability of Th1 lymphocytes to activate macro-
interleukin-2 (IL-2) receptors in macrophages phages to inhibit and destroy microorganisms is
(16–18), after which IL-2 (from T lympho- called cell-mediated immunity.
cytes exposed to specific antigens) becomes an The relationship between DTH and CMI
additional activating cytokine for these phago- in the control of facultative intracellular micro-
cytes (17, 18). Activated macrophages produce organisms was a subject of debate for most of the
reactive oxygen intermediates (19–21) and reac- 20th century (33). DTH and CMI are usually
tive nitrogen (20–28) intermediates, lysosomal produced together, albeit in different proportions
enzymes (29), and other factors that kill and depending on the nature of the antigenic stim-
digest tubercle bacilli. (See references 30, 31, and ulus and the genetic characteristics of each host.
32 for overviews of the role of cytokines in CD8 lymphocytes (see glossary) probably play
inflammatory responses.) Chapter 6 discusses a significant role in the DTH-mediated killing
macrophage activation in more detail. of macrophages containing more than a few
100 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

FIGURE 1 Cell-mediated immunity producing local activation of macrophages (acquired cel-


lular resistance) in a tuberculous lesion. Dendritic cells, macrophages, and antigen-specific Th1
lymphocytes enter the site from the bloodstream. Dendritic cells (not shown) present the bacil-
lary antigens to the lymphocytes (originally in the draining lymph nodes).The lymphocytes pro-
duce lymphokines (LK) (now called cytokines), which activate the macrophages. Only highly acti-
vated macrophages are capable of inhibiting or destroying the virulent tubercle bacilli. Reproduced
with permission from reference 125.

tubercle bacilli (1). And CD4 lymphocytes are antigen has a concentration that best activates
known to play a major role in activating macrophages and a much higher concentration
macrophages so that ingested bacilli do not that causes tissue necrosis.With tuberculin-like
grow intracellularly. The percentage of CD4 (DTH) antigens, macrophage-activating con-
cells and CD8 cells in tissue sections of pro- centrations are low and easily exceeded, so necro-
gressing and regressing tuberculous lesions (pro- tizing concentrations are soon attained.
duced in rabbits by Mycobacterium bovis BCG) is
given in chapter 6. THE MOST FAVORABLE CMI/DTH
Many mycobacterial antigens cause CMI and RATIOS IN TUBERCULOSIS
DTH via antigen-specific T cells. Some of these To select better vaccines for tuberculosis, we
antigens may produce strong DTH (tuberculin need to know what antigenic composition pro-
sensitivity) with little CMI (activated macro- duces the most favorable ratio of CMI to DTH
phages), whereas others may produce weak DTH (5) (see chapter 15). Specifically, we would like
with strong CMI. In tuberculous lesions, each a vaccine to produce increased numbers of
5. INNATE AND ACQUIRED IMMUNITY IN TUBERCULOSIS 䡵 101

FIGURE 2 Highly activated macrophages surrounding the caseous center of a 12-


day (rabbit) dermal BCG lesion.This figure illustrates an important aspect of effective
cell-mediated immunity and acquired cellular resistance, namely, that large numbers of
activated (-galactosidase-positive) macrophages accumulate around a caseous focus,
so that bacilli released from dead and dying cells will be ingested by competent rather
than incompetent cells. Stained with 5-bromo-4-chloro-3-indolyl--D-galactoside,
counterstained with hematoxylin. Magnification, ⫻90. Reproduced with permission
from reference 45.

antigen-specific lymphocytes that activate CMI AND DTH ARE BOTH REQUIRED
macrophages (CMI) and decreased numbers of TO ARREST TUBERCULOUS LESIONS
antigen-specific lymphocytes that contribute From the bacillary growth curves presented in
to caseous necrosis (DTH). In other words, the chapter 3 and a histologic study of the lesions,
vaccine should increase the inhibition of tuber- it seems that both CMI and tissue-damaging
cle bacilli within macrophages (CMI) and DTH can inhibit the multiplication of tubercle
decrease the amount of tissue damage from bacilli (34–36). However, CMI does so by acti-
their tuberculin-like products. vating macrophages to kill or inhibit the bacilli
The same beneficial CMI/DTH ratio cre- they ingest, and tissue-damaging DTH does so
ated by the vaccine would occur in the memory by destroying bacilli-laden, nonactivated macro-
lymphocytes that infiltrate a developing pul- phages and nearby tissues, thereby eliminating
monary tubercle caused by the inhalation of the intracellular environment that is so favorable
virulent tubercle bacilli.Therefore, the progres- for bacillary growth (37–39; see also reference
sion of such a developing pulmonary tubercle 29). Thus, both tuberculin-positive hosts with
would be stopped with less damage to the tissues good CMI and tuberculin-positive hosts with
of the host. In other words, the memory T cells, poor CMI can arrest bacillary growth. But the
having the most beneficial CMI/DTH ratio, host with poor CMI does so with much dam-
would remain in the host after vaccination and age to its own tissues. Eventually, the host with
provide the most favorable protection against good CMI may recover, but the host with poor
the development of clinical tuberculosis. CMI will die from excessive tissue destruction.
102 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

become activated and develop their microbici-


dal ability only where the bacillary antigens are
located (Fig. 4).The greater the local accumu-
lation of highly activated macrophages (Fig. 2),
the greater the host’s ability to destroy or inhibit
the tubercle bacillus.
Since ACR is a local reaction at sites harbor-
ing the tubercle bacillus, it is not technically the
same as CMI, which is systemic. CMI is charac-
terized by an expanded antigen-specific T-cell
population that exists (as memory cells) through-
out the body even after the bacilli and their anti-
gens have been destroyed. In contrast, ACR is
FIGURE 3 Macrophages in a 21-day rabbit dermal
BCG lesion stained both for acid-fast bacilli (red) and manifested only at sites (containing tubercle
for -galactosidase (to show macrophage activation) bacilli) where macrophages have been activated
(bright blue). (Colors not shown here.) One of these by the cytokines of infiltrating (antigen-specific)
macrophages shows negligible -galactosidase activity. lymphocytes. In this book, I usually include ACR
It contains numerous bacilli and has ruptured.Another
in the definition of CMI, because ACR cannot
macrophage (adjacent) shows high -galactosidase activ-
ity. It contains no bacilli, but is apparently ingesting the develop locally without the existence of systemic
bacilli released from the ruptured cell.This figure illus- CMI.Additional properties of ACR are discussed
trates how acquired cellular resistance, produced by later in this chapter.
CMI, stops the local growth of tubercle bacilli, namely,
that highly activated macrophages accumulate at the
local site and ingest (and destroy) the bacilli released from LOCAL NATURE OF DTH AND CMI;
ineffectual macrophages. Stained with 5-bromo-4- ORGAN RESISTANCE
chloro-3-indolyl--D-galactoside, counterstained with Marked degrees of macrophage activation take
hematoxylin, then carbol-fuchsin. Magnification, ⫻600. place locally in tuberculous lesions, where
Reproduced with permission from reference 29. macrophages become epithelioid cells (see glos-
sary) extremely rich in lysosomal (Fig. 2 through
Tissue-damaging DTH causing local necrosis 4) and mitochondrial enzymes (29, 40). The
stops the initial bacillary growth within non- same degree of activation is not common in
activated macrophages (see chapters 2 and 15). macrophages found elsewhere in the body.
Such cytotoxic DTH can never take the place of Present in the local lesion are bacillary com-
CMI, because the bacilli that escape from the ponents as well as host defense cells (29), i.e., den-
edge of necrotic areas are ingested by perifocal dritic cells, macrophages, antigen-specific lym-
macrophages. If the perifocal macrophages have phocytes, and even plasma cells (41). Because of
been sufficiently activated by CMI, they can the local antigenic stimulus (to lymphocytes)
destroy the ingested bacilli. If these macrophages caused by bacillary components, the macrophages
have not been sufficiently activated by CMI, the often become highly activated. Resident cells,
bacilli will again grow intracellularly until the including fibroblasts and vascular endothelial cells
macrophages are killed by tissue-damaging DTH, (42), also become activated and produce cyto-
thereby enlarging the caseous necrotic area. kines. Cytokines are not antigen specific and
stimulate (or suppress) any cell containing the
ACQUIRED CELLULAR RESISTANCE appropriate receptor.
IN TUBERCULOSIS We demonstrated local macrophage activation
Acquired cellular resistance (ACR) is charac- histochemically with Pearson’s indolyl substrate
terized by a local population of microbicidal for -galactosidase (43, 44) (Fig. 2 through 4).
macrophages (Fig. 2 and 4) that have been acti- (-Galactosidase is a representative lysosomal
vated by the cytokines of accumulating antigen- hydrolase.) With the indolyl substrate, we could
specific Th1 lymphocytes. These macrophages stain for both -galactosidase and tubercle bacilli
5. INNATE AND ACQUIRED IMMUNITY IN TUBERCULOSIS 䡵 103

FIGURE 4 A group of activated macrophages (epithelioid cells) in a 21-day dermal BCG


lesion from a rabbit, stained darkly for the lysosomal enzyme -galactosidase (bright blue). (Color
not shown here.) We use -galactosidase activity as a histochemical marker for activated
macrophages that are capable of destroying tubercle bacilli (29, 40).Although the perifocal tuber-
culous granulation tissue contains thousands of macrophages, only those macrophages located
near tubercle bacilli (and their products) become activated and develop the power to destroy
the bacillus. In other words, CMI is mainly a local phenomenon.The darker the macrophage
is stained for -galactosidase, the more it resembles Lurie’s mature epithelioid cell (see glos-
sary), a cell that has destroyed tubercle bacilli (31, 43). Stained with 5-bromo-4-chloro-3-indolyl-
-D-galactoside, counterstained with hematoxylin. Magnification, ⫻200. Reproduced with per-
mission from reference 29.

in the same tissue section, because the indigo nase indicated that many hydrolases and oxi-
blue dye produced by -galactosidase was not dases were markedly elevated in mature epithe-
removed by the acid-fast staining procedure, in lioid cells (29, 40).
which the bacilli were stained red by carbol- Highly activated macrophages may be rather
fuchsin (29, 40, 45) (Fig. 3). toxic to host tissue because of their high levels of
The local presence of tubercle bacilli was hydrolases and oxidases. It makes sense that such
associated with the formation of partly and activated macrophages are produced only where
fully mature epithelioid cells. Such cells were the bacilli are located.The host would probably
shown by Lurie to have destroyed the tubercle not survive if such cells were ubiquitous.
bacilli that they once contained (34, 46). The The local nature of acquired resistance in
most mature epithelioid cells (i.e., those with tuberculosis is also evident clinically.A tubercu-
the most rounded appearance) stained the dark- lous lesion in one part of a patient’s lung may
est for -galactosidase (Fig. 3 and 4). Such cells progress while a lesion in another part of the
did not contain as many bacilli as immature same lung may regress, depending on the local
epithelioid cells, which stained more lightly concentration of tubercle bacilli at each site and
(47, 48) (Fig. 3) (see chapter 6). Histochemical the host’s reaction to them (discussed more fully
staining for acid phosphatase, -glucuronidase, in chapter 3). Such local immunity also occurs
cytochrome oxidase, and succinic dehydroge- in tuberculous rabbits and guinea pigs (49).
104 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

The importance of the local site in directing erates the local activation of macrophages. Sys-
the immune response was clearly brought forth temic immunity does not prevent the
by Matzinger (50–52).When irritated or injured establishment of a microscopic tubercu-
by trauma, toxins, or infectious agents, each site lous lesion by inhaled tubercle bacilli. It
evidently produces or releases factors (including only stops the progression of such a
cytokines) that influence the type of differenti- lesion by causing a rapid accumulation of
ation that occurs in the resident cells as well as macrophages and antigen-specific lym-
in the infiltrating dendritic cells, macrophages, phocytes at that site.
and lymphocytes. Such factors first affect APCs
(dendritic cells) so that the “appropriate” MACROPHAGE ACTIVATION
cytokines are produced and the “appropriate” Macrophages have many functions, e.g., phago-
costimulatory (or coinhibitory) ligands are cytosis, killing microorganisms, cytokine pro-
upregulated. These ligands then determine duction, immunostimulation (as well as
which type of T cell is favored. In each organ immunosuppression), and some antigen pre-
of the host, different coligands may be pro- sentation. Macrophages can be activated for one
duced (53), which could explain why tubercu- or more of these functions and remain inactive
lous lesions in the liver of rabbits rarely progress, for others (54–57).
whereas those in their lungs and kidneys usually Activated macrophages that inhibit and
do so (see chapters 6 and 7). destroy the tubercle bacillus and its compo-
nents have a greatly increased content of oxida-
SYSTEMIC IMMUNITY IN tive and hydrolytic enzymes (29, 40, 58). The
TUBERCULOSIS actual killing of the bacillus seems to be due to
Individuals infected with tubercle bacilli— reactive nitrogen and reactive oxygen interme-
whether the bacilli are virulent or avirulent— diates (19, 21–28), whereas the breakdown of
develop greater numbers of T lymphocytes with bacillary components seems to be due to vari-
specific receptors for mycobacterial antigens in ous hydrolases.
their blood and lymphoid tissues than do indi- We used the lysosomal enzyme -galactosi-
viduals who have never been infected with such dase as a marker for the type of macrophage acti-
bacilli (see references 29 and 47). In other words, vation that inhibits and destroys tubercle bacilli
such individuals are immunized. When these (29, 40, 58).This marker was correlated histo-
individuals inhale exogenous virulent tubercle chemically with other hydrolytic enzymes and
bacilli, greater numbers of antigen-specific T with certain oxidative enzymes (29, 40). See
cells will accumulate at sites where the bacilli are chapter 6 for a full discussion of macrophage
deposited; once there, these T cells will pro- activation in tuberculosis.
duce cytokines that hasten the local accumula-
tion and activation of macrophages.The bacilli SYNERGISM OF DTH AND CMI IN
are then inhibited or destroyed before they mul- ARRESTING TUBERCULOSIS: DOES
tiply appreciably, and the developing lesion TRUE DORMANCY EXIST?
remains small and heals rapidly.This acceleration Nonactivated macrophages continually enter
of the host’s immune response often protects the most tuberculous lesions from the bloodstream
immunized person from clinically recognizable (see chapter 10) and may ingest occasional bacilli
exogenous and endogenous reinfection. The that then multiply within these macrophages.
role of antibodies in accelerating the local accu- Such bacillary growth is stopped when tissue-
mulation of macrophages and antigen-specific T damaging DTH kills the macrophages (see
cells is discussed below. chapters 2 and 3). In hosts with good CMI, the
In brief, systemic immunity in tuberculosis bacilli released from the dead macrophages are
is characterized by an expanded antigen- often ingested by nearby activated macrophages
specific T-cell population that enters each site that can prevent further bacillary multiplica-
where tubercle bacilli are deposited and accel- tion (Fig. 3). In other words, even in nonpro-
5. INNATE AND ACQUIRED IMMUNITY IN TUBERCULOSIS 䡵 105

gressing lesions, at least a few nonactivated determined immune response that kept the
macrophages probably allow some bacillary mul- bacillary titers low. Therefore, the size of
tiplication to occur. the dermal tuberculin reaction does not
The sequence that passes tubercle bacilli from reflect the amount of disease in the host
a nonactivated macrophage into an activated or whether it is progressing or regressing
macrophage suggests that in many “arrested” (34). In fact, patients with extensive tuberculo-
tuberculous lesions at least some tubercle bacilli sis frequently show tuberculin anergy (61–63),
are not dormant, but are undergoing repeated but after they receive effective antimicrobial
cycles of growth and inhibition. These bacilli therapy and begin to recover, they again
would reside at the edge of solid caseous areas, become tuberculin positive (63). Possible causes
escape into nearby poorly activated macrophages, of such anergy are reviewed in references 1
and change from a metabolic state of extracel- and 62.
lular dormancy into a state of intracellular mul- A large tuberculin reaction remaining years
tiplication.To determine (by molecular biology after the primary lesion has been arrested prob-
techniques) which mycobacterial genes are active ably signifies that a few viable bacilli are still pre-
in each of these metabolic states, separation of sent in caseous foci that are often radiologically
solid caseum from the surrounding (still viable) unrecognizable. Evidently, such bacilli are
tissues is essential. released from time to time, but the new lesions
Because of such bacillary growth and destruc- that they produce are soon healed.This sequence
tion, we have concluded that at least some of the boosts the host’s immune system, including the
bacilli in arrested tuberculous lesions may not level of tuberculin sensitivity.
remain dormant. In fact, in healthy individuals,
tuberculin positivity is probably maintained THE “BOOSTER” PHENOMENON IN
because of occasional episodes of bacillary mul- REPEATED TUBERCULIN TESTING
tiplication and subsequent destruction. A positive second tuberculin skin reaction may
occur after a negative first skin reaction because
THE SIZE OF THE TUBERCULIN of boosting of DTH by antigens in the first test.
SKIN REACTION HAS NO Individuals who have been infected with the
PROGNOSTIC VALUE tubercle bacillus can, in time, become tuber-
The size of the tuberculin reaction depends on culin negative with or without antimicrobial
(i) the ability of the host to respond and (ii) the treatment. However, in many of these individu-
amounts of the appropriate bacillary antigens als a recall of tuberculin sensitivity can be pro-
that sensitize the host. duced by the antigens in tuberculin (PPD) that
A strong dermal tuberculin reaction could be is injected for skin testing (64, 65, 65a). When
caused by a small number of bacilli in a host retested with intermediate-strength PPD, a per-
innately capable of producing a strong DTH son who had a negative skin reaction 1 to 5
response, or it could be caused by a large num- weeks earlier may now be tuberculin positive as
ber of bacilli in a host innately capable of pro- a result of the booster effect of tuberculin itself
ducing a weaker DTH response. Lurie’s sus- (reviewed in reference 65).The first injection of
ceptible rabbits were genetically poor responders PPD expands the tuberculin-sensitive T-cell pop-
to many antigens (34, 59, 60). However, they ulation (i.e., the memory T cells) to a level where
often developed tuberculin skin reactions just as the number of these cells is now sufficient to pro-
strong as those of his resistant rabbits, because duce a positive skin reaction with a subsequent
numerous tubercle bacilli were growing in their PPD injection.Therefore, the conversion of the
bodies (59). skin test due to boosting by a previous skin test
Conversely, a weak dermal tuberculin reac- does not signify that a recent exogenous infec-
tion could signify a poor genetically deter- tion with tubercle bacilli or even a reactivation
mined immune response that allowed many of an arrested endogenous tuberculous lesion
bacilli to grow in the host, or a good genetically has occurred.
106 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

Improved Diagnostic Tests for Active positive patients (reviewed in references 75, 76,
Tuberculosis and 77), but, to date, antibody titers are not a
The tuberculin skin test cannot reliably differ- completely reliable way to distinguish between
entiate between DTH from a progressive tuber- progressive and inapparent disease (see reference
culous lesion and DTH from environmental 78). In mice, titers of the serum antibody levels
mycobacteria, BCG vaccination, or a nonpro- against mycobacterial glycolipids (diacyltrehaloses
gressive arrested lesion. Since the secreted anti- and sulfolipid I) paralleled the bacillary concen-
gen ESAT-6 is produced by virulent human- tration in the host tissues better than did antibody
type tubercle bacilli and not by BCG or many levels to protein antigens (73). In other words, the
environmental mycobacteria, an in vitro diag- levels of glycolipid antibodies indicated pro-
nostic test on human peripheral blood mononu- gression of the disease. In general, Lurie’s resis-
clear cells (PBMC) was developed and evaluated tant inbred rabbit strains produced higher anti-
in Ethiopia (66, 67). This test measures the body titers than his susceptible inbred strains
amount of IFN- produced by PBMC after 5 (59), but exceptions existed (60).
days in culture and restimulation with ESAT-6. Antibodies have always been thought to play
At the start of this study, the amount of IFN- little or no role in the immunity to tuberculo-
 produced by PBMC from household contacts sis, because the passive transfer of immune serum
of persons with active (sputum-positive) tuber- neither protected the host from contracting the
culosis was determined (68). Two years later disease nor appreciably reduced its progression
these contacts were evaluated to determine (75, 77, 79). Recent studies in my laboratory (69)
whether they had clinically active tuberculosis. suggest a role for antibodies that had previously
The household contacts whose PBMC pro- not been considered; i.e., in hosts with good
duced high levels of IFN- developed clini- cell-mediated immunity, an antigen-antibody
cally active tuberculosis, whereas those contacts reaction causes a rapid infiltration of DTH-
whose PBMC produced low levels of IFN- and CMI-producing cells into sites where tuber-
remained healthy. Parallel in vitro studies with cle bacilli are located. In other words, anti-
tuberculin (PPD) as the antigen showed no dif- bodies enhance the local cell-mediated
ference between these two groups. Evidently, the host response (69).
contacts with early progressive tuberculous We compared rabbits containing primary der-
lesions could be identified by an increase in the mal BCG lesions with rabbits containing rein-
number of circulating lymphocytes capable of fection dermal BCG lesions (Fig. 5A through
responding to ESAT-6. C). The reinfection lesions were begun when
This was the first study in which a laboratory the primary lesions were 24 days of age. In the
test could distinguish tuberculin-positive persons first 3 h, a marked infiltration of macrophages and
with early progressive tuberculous lesions (who lymphocytes (and probably dendritic cells)
should be treated with antimicrobials) from per- occurred in the reinfection group while very lit-
sons with nonprogressive lesions (who would tle infiltration occurred in the primary group
not need antimicrobial therapy). If further stud- (Fig. 5C) (69). It seems that only an antigen-
ies confirm these results, the ESAT-6 in vitro test antibody reaction could produce such an imme-
would be a major addition to the control of this diate, pronounced antigen-specific chemotactic
disease. effect,because very few lymphocytes are normally
present in the skin. Lymphocytes and antibodies
Antibodies Enhance Local DTH and are the only antigen-specific immunological
CMI Reactions defenses that the host possesses.
Although immunity in tuberculosis is primarily At 1 to 2 days, a 400- to 500-fold difference
cell mediated, numerous antibodies circulate in in size between primary and reinfection BCG
the hosts (9, 34, 69–74), and plasma cells are lesions was present (Fig. 5C) (69). By 4 to 5 days,
common in tuberculous lesions (41). Circulating the size of the reinfection lesions had declined,
antibodies are present in nearly all tuberculin- while the size of the primary lesions had
5. INNATE AND ACQUIRED IMMUNITY IN TUBERCULOSIS 䡵 107

increased, so that both types of lesion were role in preventing clinically apparent tuberculo-
grossly similar (Fig. 5A and C). sis.Antigens reacting with such antibodies rapidly
At 8 days in reinfection lesions and at 12 bring dendritic cells,macrophages,and antigen-
days in primary lesions, small secondary peaks specific memory T cells to sites of endogenous
occurred in the size of the BCG lesions (Fig. and exogenous reinfection.These cells can then
5A).These peaks were probably caused by the stop many developing microscopic tubercles
development of CMI in the primary lesions from reaching clinically apparent size.However,
and a boosting of the CMI already present in the the immune effects of antibodies can only
reinfection lesions. have a beneficial effect in tuberculosis
In rabbits with primary BCG lesions, skin when an expanded antigen-specific T-cell
tests with Old Tuberculin became positive by 9 population exists to enter developing lesions,
days (Fig. 5B), accompanied by a rise in the because the passive transfer of antibodies to non-
levels of antibodies to the secreted antigen PstS- immune hosts provides little or no benefit.
1 (38 kDa) (Fig. 6A) (69), which is one of sev- A variation of the usual passive transfer exper-
eral antigens secreted by live tubercle bacilli iment was recently published (74). SCID mice
(80, 81). In rabbits with primary BCG lesions, were aerosol infected with Mycobacterium tuber-
antibodies to the constitutive antigens HSP65 culosis (H37Rv).After 3 weeks, they were treated
and PPD did not increase appreciably until with isoniazid and rifampin for the next 5 weeks.
much later (Fig. 6B and C) (69). Constitutive Then, hyperimmune serum to Mycobacterium
antigens are released from tubercle bacilli after tuberculosis was administered intraperitoneally,
they have been killed. once every 3 days for 4 injections.The mice were
The almost immediate difference in size euthanized 2 days after the last injection of
between primary and reinfection BCG lesions serum.When compared with controls receiving
(Fig. 5C) seems to be caused by circulating and normal serum, the mice receiving the hyper-
cytophilic antibodies. Circulating antibodies immune serum had markedly reduced bacillary
rapidly combine with the infecting bacilli and titers and had reduced tissue destruction by the
their antigens, causing an Arthus-type reaction recurring disease. Since SCID mice have few, if
and activating complement (components C1 any,Th1 lymphocytes, the antibodies may have
to C9) (82).The released C3a and C5a compo- had some direct effect on the ability of the
nents cause vasodilation and activate connective tubercle bacillus to emerge from the dormant
tissue-type mast cells and other cell types, state created by the antimicrobial therapy.
including vascular endothelial cells (details pre-
sented in reference 69). C5a is a potent chemo- LURIE’S EYE CHAMBER
taxin (83).Antigen-antibody complexes induce EXPERIMENTS ELUCIDATING THE
dendritic cell maturation and promote antigen FACTORS INVOLVED IN CMI
presentation (84), both of which also hasten the Although this book mostly concerns tuberculo-
host’s immune response to reinfection. sis produced in vivo, a description of Lurie’s eye
Cytophilic antibodies (including IgE) bind to chamber experiments (performed over 60 years
the Fc receptors on many cell types, e.g., mast ago) is included here, because they represent a
cells, fibroblasts, natural killer cells, macrophages, combination of in vitro and in vivo studies (34,
and APCs. In the presence of antigen, these 94).These experiments showed that antibodies
cells become activated and secrete cytokines do not directly affect the ability of macrophages
(including chemokines) and even the chemo- to inhibit the growth of tubercle bacilli.
taxin leukotriene B4 (85–93). The role of In several experiments, Lurie immunized rab-
cytophilic antibodies in tuberculosis has not bits (by the subcutaneous or intravenous routes)
been adequately studied. with virulent human- or bovine-type tubercle
In brief, in BCG-vaccinated hosts and in bacilli, and, after 1.5 or more months, he injected
healthy hosts made tuberculin positive by viru- acacia (gum arabic) into their pleural cavities.
lent tubercle bacilli,antibodies play an important The resulting mononuclear cell exudate was
108 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

FIGURE 5 (A) Size of primary BCG lesions and reinfection BCG lesions from 3 h to 42 days
in rabbits of Experiment I.The reinfected rabbits had been sensitized intradermally by BCG 24
days previously. Note that the reinfection BCG lesions were many times larger than the primary
5. INNATE AND ACQUIRED IMMUNITY IN TUBERCULOSIS 䡵 109

collected 4 to 5 days later and washed by cen- in the chamber fluids; most of the bacilli were in
trifugation. The exudate cells were over 85% the iris preparations (94).
mononuclear cells—mostly macrophages, with In the eye chambers, the macrophages from
some lymphocytes, granulocytes, and probably the immunized rabbits inhibited the multipli-
dendritic cells.The macrophages were not puri- cation of tubercle bacilli in their cytoplasm
fied by density gradient centrifugation or by much more effectively than did macrophages
adherence to glass. from normal rabbits. Both normal serum and
In the presence of either normal or immune immune serum had no appreciable effects on the
serum, the macrophages were allowed to ingest bacillary multiplication. Adherent to (and
in vitro (for 30 min) carbon particles mixed with within) the irises, many of the carbon-labeled
low concentrations of human-type tubercle bacilli macrophages from the immunized rabbits had
(1 bacillus for 10 or more macrophages). The the appearance of mature epithelioid cells.The
carbon particles were used to label these phago- number of carbon particles that the macrophages
cytes. Combinations of normal cells with either contained had not decreased appreciably from
normal or immune serum and combinations of the number originally present. In other words,
immune cells with normal or immune serum, the macrophages from immunized rabbits had
were made. One of the four combinations was apparently undergone little or no cell division
injected into the anterior chamber of the right in the anterior eye chambers (see chapter 10).
eye of a normal rabbit, and another combination In the eye chambers, the macrophages from
was injected into the anterior chamber of the left the nonimmunized rabbits contained numerous
eye.After 14 to 20 days, the rabbits in this exper- tubercle bacilli and did not form mature epithe-
iment were sacrificed. Then, (i) the irises with lioid cells.These macrophages showed a reduc-
adherent macrophages (containing intracellular tion in the number of intracellular carbon par-
tubercle bacilli and carbon particles) and (ii) the ticles: the numerous tubercle bacilli may have
anterior chamber fluids (containing tubercle stimulated these cells to divide, or these bacilli
bacilli, which were mostly within carbon-labeled killed such cells (after local DTH developed) and
macrophages) were cultured to determine the several nearby macrophages ingested a portion
number of viable bacilli.The irises and cells in the of the carbon load.
chamber fluids were also processed and evaluated In the light of modern knowledge, these find-
microscopically. Relatively few bacilli were found ings indicate that the macrophages from the

BCG lesions at 3 h, 12 h, and 1, 2, and 3 days (panels A and C), apparently initiated by an
antigen-antibody reaction. Note also that the size of the reinfection BCG lesions reached a sec-
ond peak at 8 days, whereas the primary lesions reached a similar peak at 12 days.These second
peaks were apparently caused by an antigen-specific CMI/DTH reaction. After the second
peaks, the lesions slowly regressed. Each point represents the mean size of the lesions and its stan-
dard error. *P ⬍ 0.05; **P ⬍ 0.01.
(B) Size of 2-day tuberculin reactions in rabbits of Experiment I. In the reinfected host, tuber-
culin sensitivity was highest before challenge.This sensitivity declined thereafter, and no booster
effect from the second BCG injection was apparent. In contrast, hosts with primary BCG infec-
tions developed strong tuberculin sensitivity by 9 days, which tended to remain higher than that
present in the reinfected hosts, possibly because the infecting bacilli were not destroyed as read-
ily. Each point represents the mean size of the tuberculin reactions and its standard error.
(C) Size of primary and reinfection BCG lesions and tuberculin reactions in Experiment II,
each measured from 3 h to 5 days.As in Experiment I, the reinfected rabbits were sensitized intra-
dermally by BCG 24 days previously.
Note that the reinfection BCG lesions and the tuberculin reactions had a similar pattern, and
that the primary BCG lesions remained very small until DTH and CMI started to develop at 4
or 5 days. Each point represents the mean size of the lesions and its standard error. *P ⬍ 0.05;
**P ⬍ 0.01. Reproduced with permission from reference 69.
110 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

FIGURE 6 Antibody levels to the mycobacterial 38-kDa secreted antigens (A) PstS-1,
(B) HSP65, and (C) PPD in rabbits with primary and reinfection BCG lesions. In the rein-
fected hosts, the second injection of BCG enhanced all existing antibody levels. In hosts
with primary BCG infection, the antibody titer to the secreted 38-kDa antigen PstS-1
became substantial by day 9, but the titers to the constitutive antigens HSP65 and PPD rose
more slowly. Each point represents the mean and its standard error. *P ⬍ 0.05; **P ⬍ 0.01.
Reproduced with permission from reference 69.
5. INNATE AND ACQUIRED IMMUNITY IN TUBERCULOSIS 䡵 111

immunized rabbits were activated specifically by later with the same or a different type of intra-
tubercle bacilli and their products after the cellular bacillus. At various times after chal-
mononuclear cell preparations (containing lenge, they determined the number and type of
macrophages, dendritic cells, and lymphocytes) viable bacilli in the spleens. In this manner, they
were placed within the anterior eye chamber: the could assess the amount of antigen-specific
expanded antigen-specific lymphocyte popula- immunity produced by the primary infection,
tion (in the mononuclear cell preparation from because most of the antigens had no cross-
the immunized host) produced cytokines (includ- reactivity among the various bacillary types
ing IFN-) that activated the macrophages pres- evaluated.
ent, so that these macrophages could inhibit the After an intravenous immunization with
intracellular growth of tubercle bacilli during BCG, the acquired resistance (adaptive immu-
their 2- to 3-week residence within the anterior nity) of the host will decrease with time. How-
eye chamber. ever, a reinjection of BCG will rapidly recall
Similar results were obtained from other eye high levels of resistance, because the immunized
chamber experiments in which “immune” host retains increased numbers of clonally
mononuclear cells were collected from the expanded (memory) T cells with specific recep-
draining lymph nodes (or bone marrow) 2 days tors for BCG antigens. After the reinjection of
after a subcutaneous (or intravenous) injection BCG, these memory T cells rapidly produce
of virulent tubercle bacilli mixed with carbon cytokines, which cause local macrophage (and
particles. In other words, when compared with lymphocyte) accumulation and activation, espe-
“nonimmune” controls, “immune” mononu- cially in the spleen, which is an organ known to
clear cell preparations that had ingested tuber- retain a large percentage of intravenously
cle bacilli in vivo (in the lymph nodes or mar- injected bacteria.
row) inhibited tubercle bacilli in the anterior eye The acquired resistance produced by the first
chamber in a fashion similar to those that had injection of BCG was not recalled by the
ingested tubercle bacilli in vitro. intravenous injection of other types of
All of these experiments show that circulat- facultative intracellular bacilli (e.g., liste-
ing antibodies had little or no direct effect on ria or salmonella). However, after this acquired
the ability of macrophages (from either normal resistance (in the spleen) had been recalled by a
or immunized hosts) to inhibit the multiplica- second injection of BCG (the specific antigen),
tion of tubercle bacilli.Therefore, immunity in the host could nonspecifically destroy the other
tuberculosis is cell-mediated by (as we know types of facultative intracellular bacilli. In other
today) antigen-specific lymphocytes produc- words, macrophages in the spleen were rapidly
ing cytokines that locally activate macrophages. activated by the expanded antigen-specific T-cell
The many purely in vitro experiments con- population that responded to the second injec-
cerning the power of “immune” mononuclear tion of BCG. Once activated, these macrophages
cells to inhibit the growth of ingested tubercle could ingest and destroy the other types of
bacilli are reviewed by Lurie (34). bacilli.
The specificity of CMI resides entirely in
DURATION AND SPECIFICITY OF the T lymphocyte, not in the macrophage.
ACR AND ITS RECALL UPON Macrophages kill intracellular microorganisms
REINFECTION nonspecifically.
In the 1960s, Mackaness and his associates clar- In most humans infected with the tubercle
ified the role of T lymphocytes and macrophages bacillus, the number of antigen-specific T cells in
in a series of beautifully designed experiments their blood and tissues decreases over time.Their
(95–98, 98a, 99, 100).They injected mice intra- tuberculin skin test may even become negative,
venously with one type of facultative intracel- especially if the tubercle bacilli and their antigens
lular bacillus (e.g., BCG, listeria, salmonella, or have been eliminated. Nonetheless, increased
brucella) and challenged these mice some time numbers of antigen-specific (memory) T cells still
112 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

remain, so that upon reinfection with virulent or INNATE IMMUNITY AND ITS
avirulent mycobacteria, their tuberculin sensi- RELATION TO ACQUIRED
tivity is rapidly recalled, as is their ACR at sites (ADAPTIVE) IMMUNITY
of bacillary lodgement. The late Charles A. Janeway, Jr., initiated the
The principles that Mackaness and his asso- recent surge of research on innate immunity
ciates (95–98, 98a, 99, 100) have established can (Table 1) (2, 102–108). Such immunity can be
be briefly summarized as follows (29). defined as the early host reaction in which the
host recognizes and responds to the invading
1. ACR (acquired cellular resistance), pro- microorganisms. Innate immunity is present at
duced by many activated macrophages at all times and does not substantially increase
sites of bacillary lodgement, is not specific with repeated exposure to a given pathogen.
for the microorganism that induced it. Unlike acquired (adaptive) immunity, it shows
Once activated, macrophages can kill a broad specificity in that it recognizes as foreign
variety of microorganisms. many components of microorganisms. In
2. Such local ACR disappears when bacil- humans and rabbits (but not mice and guinea
lary antigens are eliminated, which might pigs), innate immunity (mainly the alveolar
take years for many of the antigens of macrophages) prevents the growth of most
tubercle bacilli (72; see also reference inhaled virulent human-type tubercle bacilli
101). ACR is rapidly and specifically (see chapters 2 and 15).
recalled locally (in the spleen in Mack- Once a tubercle bacillus multiplies in the
aness’s experiments) by homologous lungs, innate immunity is manifested by an
bacilli, but ACR is produced by heterol- inflammatory response in which local resident
ogous bacilli only at the rate found in a cells release phlogistic, chemotactic, and acti-
nonimmunized host. vating substances (such as histamine), various
3. The specificity of the rapid recall of ACR cytokines (including chemokines), and the lipid
is due to an expanded population of anti- mediators: prostaglandins, leukotrienes, and
gen-specific (Th1) lymphocytes, which platelet-activating factor. This initial nonspe-
circulate as the “memory” cells for DTH cific reaction is then continued by the release of
and CMI. these and other inflammatory mediators from
4. The amount of ACR and CMI remain- the cells infiltrating the site.
ing after recovery from the primary infec- The innate recognition of invading micro-
tion is at least partly dependent on the organisms falls into two categories: cellular and
persistence and type of bacillus. Mycobac- serological. Host cells recognize microorgan-
teria persist longer than listeria. isms by pattern recognition receptors (PRR),
5. CMI is relative, not absolute.A large num- such as Toll-like receptors (109–114). PRR
ber of reinfecting bacilli will usually cause enable the phagocytes (and dendritic cells) to
disease. However, the resulting disease is recognize (for ingestion) substances in the patho-
often less severe than that present in non- gen that are different from those in the host
immunized controls. itself, e.g., various peptidoglycans, lipoproteins,
6. The presence of tuberculin sensitivity and lipopolysaccharides (2,103,104).A complete
(DTH) means that the host has at least review of the roles ofToll-like receptors in tuber-
some CMI, but not necessarily ACR. culosis (and leprosy) was published in 2004 (110).
However, such a host can develop ACR Serological recognition is by the comple-
locally at a faster rate and to a greater ment system and various circulating opsonins
degree when reinfected with tubercle (see glossary) (2). Both the alternative pathway
bacilli, because the antigen-specific lym- and the mannan-binding lectin (collectin) path-
phocyte population was expanded by the ways of complement are innately activated by
first infection with tubercle bacilli. bacteria. The complement fragments C3a and
5. INNATE AND ACQUIRED IMMUNITY IN TUBERCULOSIS 䡵 113

TABLE 1 Comparisons of the innate and adaptive immune systemsa,b


Property Innate immune system Adaptive immune system
Receptors Fixed in genome Encoded gene segments
Rearrangement not necessary Rearrangements necessary
Distribution Nonclonal Clonal
All cells of a class identical All cells of a class distinct
Recognition Conserved molecular patterns Details of molecular structure (proteins,
(lipopolysaccharides, lipoteichoic acids, peptides, carbohydrates)
mannans, glycans)
Self-nonself Perfect: selected over evolutionary time Imperfect: selected in individual somatic
discrimination cells
Action time Immediate activation of effectors Delayed activation of effectors
Response Costimulatory molecules Clonal expansion or anergy
Cytokines, such as IL-1 and IL-6 IL-2 and others
Chemokines, such as IL-8 Effector cytokines, such as IL-4 and IFN-
a
Adapted from reference 106.
b
Lurie (34) used the terms native and acquired to describe resistance to tuberculosis. Because of Janeway’s contributions to
innate immunity (2), the terms innate and adaptive have now replaced Lurie’s terminology. In this book, I have used both desig-
nations interchangeably, with a preponderance of those used by Lurie.

C5a are vasodilatory and increase the local blood molecules, e.g., MHC-I, MHC-II, B7.1, and
supply, and the chemotaxin C5a brings leuko- B7.2 (2, 118), and group 1 CD1 proteins (119).
cytes into the local area. C3b opsonizes bacte- Costimulatory molecules are necessary for
ria for ingestion by phagocytes. (Phagocytes antigen presentation to T lymphocytes (103,
have C3b receptors.) The terminal components 104). Most self antigens do not cause APCs to
of the complement cascade create pores in bac- upregulate costimulatory molecules.Therefore,
terial membranes, causing them to lyse. In addi- autoimmune reactions are rare (see references
tion to complement, serum contains C-reactive 105 and 111).
protein and 2-macroglobulin, both of which APCs also contain coinhibitory surface mol-
are innate opsonins that are increased during ecules that control and limit the antigen-specific
microbial infections. immune response (118). Coinhibition seems to
Surfactant proteins are a third group of innate be defective in some autoimmune diseases (118).
host defense, distinct from the cellular and sero- The intricacies of APC,T-cell, and B-cell inter-
logical defenses. Surfactant proteins are secreted actions have been clearly presented by Matzinger
into the alveolar spaces by type II pulmonary (50, 51) and reviewed extensively in reference 2.
alveolar epithelial cells and by Clara cells (115). IL-12 is a major cytokine of the host’s innate
SP-A and SP-D are members of the collectin defense system (120). It is produced by mac-
family. Surfactant proteins enhance phagocyto- rophages and dendritic cells that have ingested
sis of inhaled bacteria by macrophages and neu- microorganisms and their products. IL-12 acti-
trophils, and can be directly microbicidal for vates both natural killer cells and Th1 lympho-
some bacteria (116, 117). cytes, causing them to produce IFN-, which,
The host’s innate response catalyzes the adap- in turn, increases the microbicidal powers of
tive (antigen-specific) immune response in the macrophages (121). Subsequently, IL-12 induces
following manner.After APCs innately recognize IL-10 production in lymphocytes and phago-
substances unique to microorganisms (often cytes, and the IL-10 then inhibits or regulates
through their Toll-like receptors [110]), the IL-12 production (120). IL-10 also activates
APCs upregulate their costimulatory surface Th2 lymphocytes. HIV/AIDS patients have a
114 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

deficiency in their ability to produce IL-12 because for the first 2 to 3 weeks the tubercle
(120), and this deficiency seems to play a role in bacillus multiplies well within the accumulating
the susceptibility of such patients to tuberculo- nonactivated macrophages by altering their nor-
sis (120). The roles of dendritic cells, macro- mal microbicidal functions (see chapter 1).
phages, and Th1 and Th2 lymphocytes are In contrast, the inflammation caused by the
described in chapter 6. tuberculin-like products of the bacillus is anti-
In brief, the innate immune response of pul- gen specific. It is called DTH and causes an
monary alveolar macrophages plays an impor- inflammatory reaction that is an order of mag-
tant role in preventing the establishment of both nitude greater than that caused nonspecifically
primary and reinfection tuberculous lesions. by bacillary irritants in a nonsensitized (non-
Once these lesions are established, the innate immune) host. Details are presented in chapters
PRR in APCs initiate and direct the acquired 19 and 20.
(adaptive) antigen-specific immune response by In mycobacteria-infected humans, rabbits,
upregulating various costimulating factors.The and guinea pigs, the stopping of the logarithmic
integration of the innate and acquired immune (symbiotic) stage of bacillary growth (at 2 to 3
responses ensures that most small established weeks) seems to be due to an antigen-specific
tuberculous lesions do not progress in humans tissue-damaging DTH reaction at sites where
and rabbits infected with M. tuberculosis. the bacilli are located (see chapter 2). At this
The basics of innate and acquired (adaptive) time, both CMI and DTH develop, many bacilli
immunity are summarized in Table 1. References are killed, and additional bacillary components
108, 122, 123, and 124 provide more details on are released.The host reacts both antigen specif-
the immunology of tuberculosis. ically and nonspecifically to the released bacil-
lary components. However, before CMI and
NONSPECIFIC AND ANTIGEN- DTH develop, only a few bacillary products are
SPECIFIC IMMUNE RESPONSES IN released.
INNATE AND ADAPTIVE IMMUNITY
In other words, the acquired (adaptive) anti-
Confusion exists in the literature between non-
gen-specific resistance of the host as well as the
specific and antigen-specific immunity (acquired
local (innate) nonspecific resistance of the host
resistance).All antigen-specific immune responses
are both greatly enhanced by DTH and CMI.
in tuberculosis have nonspecific components.
Without the antigen specificity of DTH and
In fact, once activated, the macrophage, which is
CMI, the intracellular multiplication of the
the main effector cell killing tubercle bacilli,
tubercle bacillus would not be controlled, and
kills other intracellular bacilli quite effectively (see
a disease would occur that resembles leproma-
Mackaness, reviewed above under “Duration
tous leprosy, Johne’s disease of cattle, and avian
and Specificity of ACR and Its Recall upon
tuberculosis in birds.
Reinfection”).
The antigen specificity of each lymphocyte
participating in a tuberculous granuloma has REFERENCES
never been determined. Lymphocytes are non- 1. Dannenberg, A. M., Jr. 1999. Pathophysiology:
specifically attracted into all inflammatory sites basic aspects. I. Pathogenesis of tuberculosis. II.
by chemotactic factors, including those from Immunology of tuberculosis, p. 17–47. In D. Schloss-
berg (ed.), Tuberculosis and Nontuberculous Mycobac-
microorganisms, as well as chemokines from terial Infections, 4th ed. The W. B. Saunders Co.,
local cells.The initial accumulation of lympho- Philadelphia, Pa.
cytes, macrophages, and dendritic cells at sites of 2. Janeway, C. A., Jr., P.Travers, M.Walport, and
bacillary lodgement is mainly an innate (non- M. J. Shlomchik. 2001. Immunobiology: the Immune
specific) host inflammatory response to any irri- System in Health and Disease, 5th ed. Garland Pub-
lishing, New York, N.Y.
tant. However, in the initial stages of pulmonary 3. Lewinsohn, D. A., A. S. Heinzel, J. M. Gard-
tuberculosis, this nonspecific response benefits ner, L. Zhu, M. R.Alderson, and D. M. Lewin-
the bacillus more than it benefits the host, sohn. 2003. Mycobacterium tuberculosis-specific
5. INNATE AND ACQUIRED IMMUNITY IN TUBERCULOSIS 䡵 115

CD8⫹ T cells preferentially recognize heavily gene expression and interleukin 2 augmentation
infected cells. Am. J. Respir. Crit. Care Med. of microbicidal activity. J. Immunol. 139:1342–
168:1346–1352. 1347.
4. Stanford, J. L., and J. M. Grange. 1974. The 18. Holter, W., C. K. Goldman, L. Casabo, D. L.
meaning and structure of species as applied to Nelson, W. C. Greene, and T. A. Waldmann.
mycobacteria. Tubercle 55:143–152. 1987. Expression of functional IL-2 receptors by
5. Dannenberg, A.M., Jr. 1990. Controlling tuber- lipopolysaccharide and interferon-gamma stimu-
culosis: the pathologist’s point of view. Res. Micro- lated human monocytes. J. Immunol. 138:2917–
biol. 141:192–196. 2922.
6. McManus, I. C., D. N. J. Lockwood, J. L. Stan- 19. Klebanoff, S. J. 1988. Phagocytic cells: products of
ford, M.A. Shaaban, M.Abdul-Ati, and G. M. oxygen metabolism, p. 391–444. In J. I. Gallin, I. M.
Bahr. 1988. Recognition of a category of respon- Goldstein, and R. Snyderman (ed.), Inflammation:
ders to group II, slow-grower associated antigens Basic Principles and Clinical Correlates. Raven Press,
amongst Kuwaiti senior school children, using a sta- New York, N.Y.
tistical model. Tubercle 69:275–281. 20. Beutler, B. 2004. Innate immunity: an overview.
7. Stanford, J. L. 1997. Newer tuberculins: profile in Mol. Immunol. 40:845–859.
developing countries, p. 58–72. In V. Seth (ed.), 21. Nathan, C., and M. U. Shiloh. 2000. Reactive
Essentials of Tuberculosis in Children. Jaypee Brothers, oxygen and nitrogen intermediates in the rela-
New Delhi, India. tionship between mammalian hosts and micro-
8. Kaufmann, S. H. E. 2001. How can immunology bial pathogens. Proc. Natl.Acad. Sci. USA 97:8841–
contribute to the control of tuberculosis? Nat. Rev. 8848.
Immunol. 1:20–30. 22. Liew, F.Y., and F. E. G. Cox. 1991. Nonspecific
9. Daniel,T. M., and B.W. Janicki. 1978. Mycobac- defence mechanism: the role of nitric oxide.
terial antigens: a review of their isolation, chemistry, Immunol.Today 12:A17–A21.
and immunological properties. Microbiol. Rev. 42:84– 23. Stuehr, D. J., and M. A. Marletta. 1987. Induc-
113. tion of nitrite/nitrate synthesis in murine mac-
10. Abou-Zeid, C., I. Smith, J. M. Grange, T. L. rophages by BCG infection, lymphokines, or
Ratliff, J. Steele, and G. A.W. Rook. 1988.The interferon-gamma. J. Immunol. 139:518–525.
secreted antigens of Mycobacterium tuberculosis and 24. Moncada, S., R. M. J. Palmer, and E. A.
their relationship to those recognized by the avail- Higgs. 1991. Nitric oxide: physiology, patho-
able antibodies. J. Gen. Microbiol. 134:531–538. physiology, and pharmacology. Pharmacol. Rev.
11. Andersen, P., D. Askgaard, L. Ljungqvist, 43:109–142.
J. Bennedsen, and I. Heron. 1991. Proteins 25. Green, S. J., C. A. Nacy, and M. S. Meltzer.
released from Mycobacterium tuberculosis during 1991. Cytokine-induced synthesis of nitrogen
growth. Infect. Immun. 59:1905–1910. oxides in macrophages: a protective host response
12. Long, E. R. 1958. The Chemistry and Chemother- to Leishmania and other intracellular pathogens.
apy of Tuberculosis, 3rd ed., p. 106–108, 122–124. J. Leukoc. Biol. 50:93–103.
Lippincott Williams & Wilkins, Philadelphia, Pa. 26. Weinberg, J. B. 1998. Nitric oxide production and
13. Nathan, C., and R. Yoshida. 1988. Cytokines: nitric oxide synthase type 2 expression by human
interferon-gamma, p. 229–251. In J. I. Gallin, I. M. mononuclear phagocytes: a review. Mol. Med. 4:557–
Goldstein, and R. Snyderman (ed.), Inflammation: 591.
Basic Principles and Clinical Correlates. Raven Press, 27. Bogdan, C. 2001. Nitric oxide and the immune
New York, N.Y. response. Nat. Immunol. 2:907–916.
14. Barnes, P. F., S.-J. Fong, P. J. Brennan, P. E. 28. Chan, J.,Y. Xing, R. S. Magliozzo, and B. R.
Twomey, A. Mazumder, and R. L. Modlin. Bloom. 1992. Killing of virulent Mycobacterium
1990. Local production of tumor necrosis factor and tuberculosis by reactive nitrogen intermediates pro-
IFN-gamma in tuberculous pleuritis. J. Immunol. duced by activated murine macrophages. J. Exp.
145:149–154. Med. 175:1111–1122.
15. Celada, A., and C. Nathan. 1994. Macrophage 29. Dannenberg, A. M., Jr. 1968. Cellular hyper-
activation revisited. Immunol.Today 15:100–102. sensitivity and cellular immunity in the patho-
16. Toossi, Z., J. R. Sedor, J. P. Lapurga, R. J. genesis of tuberculosis: specificity, systemic and local
Ondash, and J. J. Ellner. 1990. Expression of nature, and associated macrophage enzymes. Bacte-
functional interleukin 2 receptors by peripheral riol. Rev. 32:85–102.
blood monocytes from patients with active pul- 30. Nathan, C., and M. Sporn. 1991. Cytokines in
monary tuberculosis. J. Clin. Investig. 85:1777–1784. context. J. Cell Biol. 113:981–986.
17. Wahl, S. M., N. McCartney-Francis, D. A. 31. Remick, D. G., and J. S. Friedland (ed.). 1997.
Hunt, P. D. Smith, L. M. Wahl, and I. M. Cytokines in Health and Disease, 2nd ed. Marcel
Katona. 1987. Monocyte interleukin 2 receptor Dekker, Inc., New York, N.Y.
116 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

32. Dinarello, C.A., M. J. Kluger, M. C. Powanda, on -galactosidase, -glucuronidase and aminopep-


and J. J. Oppenheim (ed.). 1990. The Physiolog- tidase with indolyl and naphthyl substrates. J. Retic-
ical and Pathological Effects of Cytokines, p. 81–205. uloendothel. Soc. 4:390–408.
Wiley-Liss, New York, N.Y. 45. Shima, K., A. M. Dannenberg, Jr., M. Ando,
33. Rich, A. R. 1951. The Pathogenesis of Tuberculosis, S. Chandrasekhar, J. A. Seluzicki, and J. I.
2nd ed., p. 508–569. Charles C Thomas, Springfield, Fabrikant. 1972. Macrophage accumulation, divi-
Ill. sion, maturation, and digestive and microbicidal
34. Lurie, M. B. 1964. Resistance to Tuberculosis: Exper- capacities in tuberculous lesions. I. Studies involv-
imental Studies in Native and Acquired Defensive Mech- ing their incorporation of tritiated thymidine and
anisms, p. 77–104. Harvard University Press, Cam- their content of lysosomal enzymes and bacilli.
bridge, Mass. Am. J. Pathol. 67:159–180.
35. Lurie, M. B., P. Zappasodi, and C. Tickner. 46. Lurie, M. B. 1932. The correlation between the
1955. On the nature of genetic resistance to tuber- histological changes and the fate of living tubercle
culosis in the light of the host-parasite relationships bacilli in the organs of tuberculous rabbits. J. Exp.
in natively resistant and susceptible rabbits. Am. Med. 55:31–52.
Rev.Tuberc. 72:297–329. 47. Dannenberg, A. M., Jr. 1989. Immune mecha-
36. Allison, M., P. Zappasodi, and M. B. Lurie. nisms in the pathogenesis of pulmonary tubercu-
1962. Host-parasite relationships in natively resis- losis. Rev. Infect. Dis. 11(Suppl. 2):S369–S378.
tant and susceptible rabbits on quantitative inhala- 48. Ando, M., A. M. Dannenberg, Jr., M. Sugi-
tion of tubercle bacilli. Am. Rev. Respir. Dis. 85:553– moto, and B. S. Tepper. 1977. Histochemical
569. studies relating the activation of macrophages to the
37. Dannenberg, A. M., Jr. 1991. Delayed-type intracellular destruction of tubercle bacilli. Am. J.
hypersensitivity and cell-mediated immunity in the Pathol. 86:623–634.
pathogenesis of tuberculosis. Immunol.Today 12:228– 49. Lurie, M. B. 1939. Studies on the mechanism of
233. immunity in tuberculosis.The role of extracellular
38. Dannenberg, A. M., Jr. 1993. Immunopatho- factors and local immunity in fixation and inhibi-
genesis of pulmonary tuberculosis. Hosp. Pract. tion of growth of tubercle bacilli. J. Exp. Med.
28:51–58. 69:555–578.
39. Dannenberg, A. M., Jr., and G. A. W. Rook. 50. Matzinger, P. 2002.The danger model: a renewed
1994. Pathogenesis of pulmonary tuberculosis: an sense of self. Science 296:301–305.
interplay of tissue-damaging and macrophage- 51. Matzinger, P. 1994. Tolerance, danger, and the
activating immune responses—dual mechanisms extended family. Annu. Rev. Immunol. 12:991–1045.
that control bacillary multiplication, p. 459–483. 52. Gallucci, S., and P. Matzinger. 2001. Danger sig-
In B. R. Bloom (ed.), Tuberculosis: Pathogenesis, Pro- nals: SOS to the immune system. Curr. Opin.
tection, and Control. American Society for Micro- Immunol. 13:114–119.
biology,Washington, D.C. 53. Chen, L. 2004. Co-inhibitory molecules of the B7-
40. Dannenberg, A. M., Jr., O. T. Meyer, J. R. CD28 family in the control of T-cell immunity. Nat.
Esterly, and T. Kambara. 1968.The local nature Rev. Immunol. 4:336–347.
of immunity in tuberculosis, illustrated histochem- 54. Suga, M., A. M. Dannenberg, Jr., and S.
ically in dermal BCG lesions. J. Immunol. 100:931– Higuchi. 1980. Macrophage functional hetero-
941. geneity in vivo: macrolocal and microlocal
41. Converse, P. J., A. M. Dannenberg, Jr., J. E. macrophage activation, identified by double-stain-
Estep, K. Sugisaki, Y. Abe, B. H. Schofield, ing tissue sections of BCG granulomas for pairs of
M. L. M. Pitt. 1996. Cavitary tuberculosis pro- enzymes. Am. J. Pathol. 99:305–324.
duced in rabbits by aerosolized virulent tubercle 55. Nathan, C. F. 1987. Secretory products of
bacilli. Infect. Immun. 64:4776–4787. macrophages. J. Clin. Investig. 79:319–326.
42. Sugisaki, K., A. M. Dannenberg, Jr.,Y. Abe, J. 56. Grage-Griebenow, E., H.-D. Flad, and M.
Tsuruta, W.-J. Su, W. Said, L. Feng, T. Ernst. 2001. Heterogeneity of human peripheral
Yoshimura, P. J. Converse, and P. Mounts. blood monocyte subsets. J. Leukoc. Biol. 69:11–
1998. Nonspecific and immune-specific up-regu- 20.
lation of cytokines in rabbit dermal tuberculous 57. Ragno, S., M. Romano, S. Howell, D. J. C.
(BCG) lesions. J. Leukoc. Biol. 63:440–450. Pappin, P. J. Jenner, and M. J. Colston. 2001.
43. Pearson, B., P. L. Wolf, and J.Vazquez. 1963. Changes in gene expression in macrophages
A comparative study of a series of new indolyl infected with Mycobacterium tuberculosis: a combined
compounds to localize -galactosidase in tissues. transcriptomic and proteomic approach. Immunol-
Lab. Investig. 12:1249–1259. ogy 104:99–108.
44. Yarborough, D. J., O. T. Meyer, A. M. Dan- 58. Ando, M., A. M. Dannenberg, Jr., M. Sugi-
nenberg, Jr., and B. Pearson. 1967. Histo- moto, and B. S. Tepper. 1977. Histochemical
chemistry of macrophage hydrolases. III. Studies studies relating the activation of macrophages to the
5. INNATE AND ACQUIRED IMMUNITY IN TUBERCULOSIS 䡵 117

intracellular destruction of tubercle bacilli. Am. J. gens of Mycobacterium tuberculosis: immunodomi-


Pathol. 86:623–634. nance of high-molecular-mass antigens. Clin. Diagn.
59. Lurie, M. B., P. Zappasodi, E. Cardona-Lynch, Lab. Immunol. 4:49–56.
and A. M. Dannenberg, Jr. 1952.The response 71. Samanich, K. M., J. T. Belisle, M. G. Sonnen-
to the intracutaneous inoculation of BCG as an berg, M.A. Keen, S. Zolla-Pazner, and S. Laal.
index of native resistance to tuberculosis. J. Immunol. 1998. Delineation of human antibody responses to
68:369–387. culture filtrate antigens of Mycobacterium tuberculosis.
60. Lurie, M. B., and A. M. Dannenberg, Jr. 1965. J. Infect. Dis. 178:1534–1538.
Macrophage function in infectious disease with 72. Barksdale, L., and K.-S. Kim. 1977. Mycobac-
inbred rabbits. Bacteriol. Rev. 29:466–476. terium. Bacteriol. Rev. 41:217–372.
61. Ellner, J. J. 1997. Regulation of the human 73. Cardona, P. J., E. Julián, X.Vallès, S. Goradillo,
immune response during tuberculosis. J. Lab. Clin. M. Munõs, M. Luquin, and V. Ausina. 2002.
Med. 130:469–475. Production of antibodies against glycolipids from
62. Ellner, J. J. 1996. Immunosuppression in tubercu- the Mycobacterium tuberculosis cell wall in aerosol
losis. Infect.Agents Dis. 5:62–72. murine models of tuberculosis. Scand. J. Immunol.
63. Ellner, J. J. 1997. Review: the immune response in 55:639–645.
human tuberculosis—implications for tuberculosis 74. Guirado, E., I. Amat, O. Gil, J. Diaz,
control. J. Infect. Dis. 176:1351–1359. V. Arcos, N. Caceres,V. Ausina, and P. J. Car-
64. Thompson, N. J., J. L. Glassroth, D. E. Snider, dona. 2006. Passive-serum therapy with polyclonal
Jr., and L. S. Farer. 1979.The booster phenom- antibodies against Mycobacterium tuberculosis pro-
enon in serial tuberculin testing. Am. Rev. Respir. Dis. tects against post-chemotherapy relapse of tuber-
119:587–597. culosis infection in SCID mice. Microbes Infect.
65. Menzies, D. 1999. Interpretation of repeated [E-pub ahead of print.]
tuberculin tests. Boosting, conversion, and reversion. 75. Glatman-Freedman, A., and A. Casadevall.
Am. J. Respir. Crit. Care Med. 159:15–21. 1998. Serum therapy for tuberculosis revisited: reap-
65a.Comstock, G. W., and S. F. Woolpert. 1978. praisal of the role of antibody-mediated immunity
Tuberculin conversion: true or false? Am. Rev. Respir. against Mycobacterium tuberculosis. Clin. Microbiol.
Dis. 118:215–217. (Editorial.) Rev. 11:514–532.
66. Demissie,A., P. Ravn, J. Olobo,T. M. Doherty, 76. Glatman-Freedman, A. 2003.Advances in anti-
T. Eguale, M. Geletu, W. Hailu, P. Andersen, body-mediated immunity against Mycobacterium
and S. Britton. 1999. T-cell recognition of tuberculosis: implications for a novel vaccine strategy.
Mycobacterium tuberculosis culture filtrate fractions FEMS Immunol. Med. Microbiol. 39:9–16.
in tuberculosis patients and their household con-
77. Rich, A. R. 1951. The Pathogenesis of Tuberculosis,
tacts. Infect. Immun. 67:5967–5971.
2nd ed., p. 570–613. Charles C Thomas Publishers,
67. Ravn, P., A. Demissie, T. Eguale, H. Wond-
Springfield, Ill.
wosson, D. Lein, H.A.Amoudy,A. S. Mustafa,
A. K. Jensen, A. Holm, I. Rosenkrands, F. 78. Laal, S., and Y. A. W. Skeiky. 2005. Immune-
Oftung, J. Olobo, F. von Reyn, and P. Ander- based methods, p. 71–83. In S.T. Cole, K. D. Eise-
sen. 1999. Human T cell responses to the ESAT-6 nach, D. N. McMurray, and W. R. Jacobs, Jr. (ed.),
antigen from Mycobacterium tuberculosis. J. Infect. Dis. Tuberculosis and the Tubercle Bacillus. ASM Press,
179:637–645. Washington, D.C.
68. Doherty,T. M., A. Demissie, J. Olobo, D.Wol- 79. Reggiardo, Z., and G. Middlebrook. 1974.
day, S. Britton, T. Eguale, P. Ravn, and P. Failure of passive serum transfer of immunity against
Andersen. 2002. Immune responses to the Mycobac- aerogenic tuberculosis in rabbits. Proc. Soc. Exp. Biol.
terium tuberculosis-specific antigen ESAT-6 signal Med. 145:173–175.
subclinical infection among contacts of tuberculo- 80. Sethna, K. B., N. F. Mistry,Y. Dholakia, N. H.
sis patients. J. Clin. Microbiol. 40:704–706. Antia, and M. Harboe. 1998. Longitudinal trends
69. Shigenaga, T., A. M. Dannenberg, Jr., D. B. in serum levels of mycobacterial secretory (30 kD)
Lowrie, W. Said, M. J. Urist, H. Abbey, B. H. and cytoplasmic (65 kD) antigens during chemo-
Schofield, P. Mounts, and K. Sugisaki. 2001. therapy of pulmonary tuberculosis patients. Scand. J.
Immune responses in tuberculosis: antibodies and Infect. Dis. 30:363–369.
CD4-CD8 lymphocytes with vascular adhesion 81. Wiker, H. G., S. L. Michell, R. G. Hewinson,
molecules and cytokines (chemokines) cause a rapid E. Spierings, S. Nagai, and M. Harboe. 1999.
antigen-specific cell infiltration at sites of bacillus Cloning, expression and significance of MPT53
Calmette-Guérin reinfection. Immunology 102:466– for identification of secreted proteins of Mycobac-
479. terium tuberculosis. Microb. Pathog. 26:207–219.
70. Laal, S., K. M. Samanich, M. G. Sonnenberg, 82. Majno, G., and I. Joris. 1996. Cells,Tissues, and
S. Zolla-Pazner, J. M. Phadtare, and J. T. Disease: Principles of General Pathology, p. 530–532.
Belisle. 1997. Human humoral responses to anti- Blackwell Science, Cambridge, Mass.
118 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

83. Cooper, N. R. 1999. Biology of the complement normal and immunized animals. J. Exp. Med.
system, p. 281–315. In J. I. Gallin, R. Snyderman, 75:247–268.
D. T. Fearon, B. F. Haynes, and C. Nathan (ed.), 95. Mackaness, G. B. 1962. Cellular resistance to
Inflammation: Basic Principles and Clinical Correlates, 3rd infection. J. Exp. Med. 116:381–406.
ed. Lippincott Williams & Wilkins, Philadelphia, Pa. 96. Mackaness, G. B. 1964. The immunological
84. Regnault, A., D. Lankar, V. Lacabanne, A. basis of acquired cellular resistance. J. Exp. Med.
Rodriguez, C.Thery, M. Rescigno,T. Saito, S. 120:105–120.
Verbeek, C. Bonnerot, P. Ricciardi-Castagnoli, 97. Mackaness, G. B. 1967. The relationship of
and S. Amigorena. 1999. Fc gamma receptor- delayed hypersensitivity to acquired cellular resis-
mediated induction of dendritic cell maturation tance. Br. Med. Bull. 23:52–54.
and major histocompatibility complex class 1- 98. Mackaness, G. B., and R. V. Blanden. 1967.
restricted antigen presentation after immune com- Cellular immunity. Prog.Allergy 11:89–140.
plex internalization. J. Exp. Med. 189:371–380. 98a.Mackaness, G. B. 1968. The immunology of
85. Rumsaeng,V.,W.W. Cruikshank, B. Foster, C. antituberculous immunity. Am. Rev. Respir. Dis.
Prussin, A. S. Kirshenbaum, T. A. Davis, H. 97:337–344. (Editorial.)
Kornfeld, D. M. Center, and D. D. Metcalfe. 99. North, R. J. 1974. Cell-mediated immunity and
1997. Human mast cells produce the CD4⫹ T lym- the response to infection, p. 185–219. In R. T.
phocyte chemoattractant factor, IL-16. J. Immunol. McCluskey and S. Cohen (ed.), Mechanisms of
59:2904–2910. Cell-Mediated Immunity. John Wiley & Sons, Inc.,
86. Crooks, S. W., and R. A. Stockley. 1998. New York, N.Y.
Leukotriene B4. Int. J. Biochem. Cell Biol. 30:173–178. 100. Collins, F. M., and S. G. Campbell. 1982.
87. Lam, B. K., and K. F.Austen. 1992. Leukotrienes: Immunity to intracellular bacteria. Vet. Immunol.
biosynthesis, release, and actions, p. 139–47. In J. I. Immunopathol. 3:5–66.
Gallin, I. M. Goldstein, and R. Snyderman (ed.), 101. Higuchi, S., M. Suga,A. M. Dannenberg, Jr.,
Inflammation: Basic Principles and Clinical Correlates, L. F. Affronti, I. Azuma, T. M. Daniel, and
2nd ed. Raven Press, Ltd, New York, N.Y. J. P. Petrali. 1981. Persistence of protein, carbo-
88. Yuan, D., C. Y. Koh, and J. A. Wilder. 1994. hydrate and wax components of tubercle bacilli in
Interactions between B lymphocytes and NK cells. dermal BCG lesions. Am. Rev. Respir. Dis. 123:397–
FASEB J. 8:1012–1018. 401.
89. Smith, R. S.,T. J. Smith,T. M. Blieden, and R. 102. Luster, A. D. 2002. The role of chemokines in
P. Phipps. 1997. Fibroblasts as sentinel cells. Syn- linking innate and adaptive immunity. Curr. Opin.
thesis of chemokines and regulation of inflamma- Immunol. 14:129–135.
tion. Am. J. Pathol. 151:317–322. 103. Bendelac, A., and D. T. Fearon. 1997. Innate
90. Gosset, P., I.Tillie-Leblond, S. Oudin, O. Par- immunity: innate pathways that control acquired
mentier, B. Wallaert, M. Joseph, and A.-B. immunity. Curr. Opin. Immunol. 9:1–3.
Tonnel. 1999. Production of chemokines and 104. Medzhitov, R., and C. A. Janeway, Jr. 1997.
proinflammatory and antiinflammatory cytokines Innate immunity: impact on the adaptive immune
by human alveolar macrophages activated by IgE response. Curr. Opin. Immunol. 9:4–9.
receptors. J.Allergy Clin. Immunol. 103:289–297. 105. Medzhitov, R., and C. A. Janeway, Jr. 2002.
91. Ying, S., L.T. Barata, Q. Meng, J. A. Grant, J. Decoding the patterns of self and nonself by the
Barkans, S. R. Durham, and A. B. Kay. 1998. innate immune system. Science 296:298–300.
High-affinity immunoglobulin E receptor (Fc(RI)- 106. Janeway, C. A., Jr., and R. Medzhitov. 2002.
bearing eosinophils, mast cells, macrophages and Innate immune recognition. Annu. Rev. Immunol.
Langerhans’ cells in allergen-induced late-phase 20:197–216.
cutaneous reactions in atopic subjects. Immunology 107. Beutler, B., and J. Hoffmann. 2003. Innate
93:281–288. immunity. Editorial overview. Curr. Opin. Immunol.
92. Nilsson, G., J. J. Costa, and D. D. Metcalfe. 16:1–3.
1999. Mast cells and basophils, p. 97–117. In J. I. 108. van Crevel, R., T. H. M. Ottenhoff, and
Gallin, R. Snyderman, D.T. Fearon, B. F. Haynes, and J. W. M. van der Meer. 2002. Innate immunity
C. Nathan (ed.), Inflammation: Basic Principles and to Mycobacterium tuberculosis. Clin. Microbiol. Rev.
Clinical Correlates, 3rd ed. Lippincott Williams & 15:294–309.
Wilkins, Philadelphia, Pa. 109. Uronen-Hansson, H., J. Allen, M. Osman,
93. Bradding, P. 1996. Human mast cell cytokines. G. Squires, N. Klein, and R. E. Callard. 2004.
Clin. Exp.Allergy 26:13–19. Toll-like receptor 2 (TLR2) and TLR4 are present
94. Lurie, M. B. 1942. Studies on the mechanism of inside human dendritic cells, associated with
immunity in tuberculosis.The fate of tubercle bacilli microtubules and the Golgi apparatus but are not
ingested by mononuclear phagocytes derived from detectable on the cell surface: integrity of micro-
5. INNATE AND ACQUIRED IMMUNITY IN TUBERCULOSIS 䡵 119

tubules is required for interleukin-12 production 118. Lenschow, D. J., T. L. Walunas, and J. A.
in response to internalized bacteria. Immunology Bluestone. 1996. CD28/B7 system of T cell
111:173–178. costimulation. Annu. Rev. Immunol. 14:233–
110. Krutzik, S. R., and R. L. Modlin. 2004.The 258.
role of Toll-like receptors in combating mycobac- 119. Roura-Mir, C., L.Wang,T.-Y. Cheng, I. Mat-
teria. Semin. Immunol. 16:35–41. sunaga, C. C. Dascher, S. L. Peng, M. J. Fen-
111. Pasare, C., and R. Medzhitov. 2003. Toll ton, C. Kirschning, and D. B. Moody. 2005.
pathway-dependent blockade of CD4⫹CD8⫹ Mycobacterium tuberculosis regulates CD1 antigen
T cell-mediated suppression by dendritic cells. presentation pathways through TLR-2. J. Immunol.
Science 299:1033–1036. 175:1758–1766.
112. Kopp, E., and R. Medzhitov. 2003. Recogni- 120. Trinchieri, G. 1997. Cytokines acting on or
tion of microbial infection by Toll-like receptors. secreted by macrophages during intracellular infec-
Curr. Opin. Immunol. 15:396–401. tion (IL-10, IL-12, IFN-). Curr. Opin. Immunol.
113. Heine, H., and E. Lien. 2003.Toll-like receptors 9:17–23.
and their function in innate and adaptive immu- 121. Trinchieri, G. 1995. Interleukin-12: a proin-
nity. Int.Arch.Allergy Immunol. 130:180–192. flammatory cytokine with immunoregulatory
functions that bridge innate resistance and antigen-
114. Beutler, B., K. Hoebe, X. Du, and R. J. Ule-
specific adaptive immunity. Annu. Rev. Immunol.
vitch. 2003. How we detect microbes and respond
13:251–276.
to them: the Toll-like receptors and their trans-
122. Flynn, J. L., and J. Chan. 2001. Immunology
ducers. J. Leukoc. Biol. 74:479–485.
of tuberculosis. Annu. Rev. Immunol. 19:93–
115. Auten, R. L., R. H. Watkins, D. L. Shapiro, 129.
and S. Horowitz. 1990. Surfactant apoprotein A 123. Kaufmann, S. H. E. 2003. Immune response to
(SP-A) is synthesized in airway cells. Am. J. Respir. tuberculosis: experimental animal models. Tuber-
Cell Mol. Biol. 3:491–496. culosis 83:107–111.
116. Wu, H.,A. Kuzmenko, S.Wan, L. Schaffer,A. 124. Rook, G.A.W., G. Seah, and A. Ustianowski.
Weiss, J. H. Fisher, K. S. Kim, and F. X. 2001. M. tuberculosis: immunology and vaccination.
McCormack. 2003. Surfactant proteins A and D Eur. Respir. J. 17:537–557.
inhibit the growth of Gram-negative bacteria by 125. Dannenberg, A. M., Jr. 1978. Pathogenesis of
increasing membrane permeability. J. Clin. Investig. pulmonary tuberculosis in man and animals; pro-
111:1589–1602. tection of personnel against tuberculosis, p. 65–75.
117. Wright, J. R. 2003. Pulmonary surfactant: a front In R. J. Montali (ed.), Mycobacterial Infections of
line of lung host defense. J. Clin. Investig. 111:1453– Zoo Animals. Smithsonian Institution Press,Wash-
1455. ington, D.C.
6
MACROPHAGES AND OTHER
CELLS IN TUBERCULOUS LESIONS

Antigen-presenting cells 120


Role of the major histocompatibility complex and CD1 proteins in antigen
presentation 122
Phagocytosis of tubercle bacilli by macrophages and their fate within
these cells 123
Mycobacterial dormancy in mouse tuberculous granulomas 124
Macrophage activation 126
Hydrolytic enzymes of rabbit mononuclear and polymorphonuclear exudate
cells, pulmonary alveolar macrophages, and lung homogenates 128
Macrophage enzymes released extracellularly in BCG lesions 128
Macrophage heterogeneity in tissue sections of BCG lesions 129
Causes of macrolocal and microlocal macrophage heterogeneity 131
Epithelioid cells 133
Langhans’ giant cells 133
Mononuclear cell turnover 136
Overview of lymphocytes: their subsets and their functions 137
Th1, Th2, CD4, CD8, Treg, and  T cells 138
Memory T cells 140
NK cells, cytotoxic T lymphocytes, and apoptosis 140
Granulocytes in tuberculous lesions 143
Fibroblasts in tuberculous lesions 144
Lymphatics in tuberculous lesions 144

Abstract. The main types of cells participating in rabbit tuberculous lesions are dendritic
cells, macrophages, natural killer cells, lymphocytes, and granulocytes.The role of most of
these cells is discussed only briefly in this chapter because details are available in textbooks
of immunology.The role of macrophages, however, is presented more fully because their
rates of turnover, their states of activation, their extracellular and intracellular enzymes, and
their heterogeneity have been extensively studied in my laboratory in the rabbit model
of tuberculosis.

Many types of cells and subgroups of cell types in vitro in cell cultures and in vivo in transgenic
have been identified in inflammatory and or knockout mice.To my knowledge, no histo-
immune responses (1), and all of these cells par- chemical studies (except those in my laboratory)
ticipate in tuberculous lesions.Table 1 lists some have been made on rabbit tuberculous lesions.
of the major players (see also reference 2). As Information on dendritic cells and various
stated in the abstract, this chapter briefly describes types of lymphocytes, however, is accumulating
the attributes and functions of each of these cell so fast that even current textbooks are not fully
types. For more details, the reader is referred to up to date.Therefore, in this chapter, I added a
immunology textbooks, e.g., Immunobiology: the few items from the recent literature on these two
Immune System in Health and Disease (1). cell types.
Very few cell functions have been studied in
tissue sections by histochemical techniques to ANTIGEN-PRESENTING CELLS
elucidate how each cell influences its neighbors Dendritic cells (DCs) are the main antigen-
in vivo.Cell functions have mostly been studied presenting cells (APCs) (1, 3–10) (Table 1).They

120
6. MACROPHAGES AND OTHER CELLS IN TUBERCULOUS LESIONS 䡵 121

TABLE 1 Major cell types involved in specific and nonspecific host defense reactions against the tubercle
bacillusa
Macrophages
Macrophages are the effector cells of the mononuclear phagocyte system.They are produced in the bone
marrow, circulate as monocytes in the bloodstream, and are called macrophages when they emigrate from the
blood into the tissues. Nonactivated monocytes/macrophages allow tubercle bacilli to multiply within them,
whereas highly activated macrophages inhibit or destroy tubercle bacilli. Many pulmonary alveolar
macrophages are highly activated and therefore (in rabbits and humans) often prevent inhaled virulent human-
type tubercle bacilli from starting an infection.
Antigen-Presenting Cells (APCs)
Dendritic cells are the main APCs. Dendritic cells migrate from the site of infection to the draining lymph
nodes, where they initiate an immune response by presenting antigens to the recirculating lymphocytes that
enter these nodes.Within tuberculous lesions, dendritic cells (along with macrophages and B lymphocytes) can
also present antigens to lymphocytes, especially after the host has developed acquired (adaptive) immunity.
Lymphocytes (T Cells and B Cells)
T cells (from the thymus) and B cells (from the bone marrow in mammals and the bursa in birds) provide
immunologic specificity to the host’s defense against tubercle bacilli. In tuberculous lesions, antigen-activated
T cells produce cytokines that activate macrophages to inhibit or destroy the tubercle bacillus.
T cells have been subdivided in a variety of ways based on (i) their surface markers (CD4 and CD8 T cells), (ii)
their receptors (/ and /), (iii) the cytokines they produce (Th1 and Th2 cells), and (iv) their functions
(helper, regulatory, and cytotoxic T cells). See Fig. 11.
Antigen-activated B cells produce antibodies, especially when they differentiate into plasma cells. In tubercu-
lous lesions, antigen-antibody reactions hasten the local accumulation of dendritic cells, macrophages, and
antigen-specific T cells; i.e., antigen-antibody reactions enhance the local cell-mediated immune response
(see chapter 5).
Natural Killer (NK) Cells
NK cells (both local and circulating) provide an important early defense against intracellular microorganisms
(viruses, bacteria, fungi, and protozoa). In tuberculosis, NK cells participate in the killing of bacilli-laden
macrophages and can produce IFN-, which activates macrophages and stimulates a Th1 cytokine immune
response.
a
Adapted from reference 2.

are the “conductor of the whole immunologic (e.g., the capsular polysaccharides of bacteria) are
orchestra” (R. K. Gershon, personal communi- T-lymphocyte independent but nevertheless
cation modified by Noel R. Rose). DCs that seem to require some interaction with T cells
activateT cells are produced in the bone marrow, and/or DCs (reviewed in reference 11).
reside in various tissues, and mature by a multi-
step process for antigen processing and antigen DCs That Activate T Cells
presentation.DCs that activate B cells (called fol- The primary acquired immune response to the
licular DCs) are cells of uncertain origin. tubercle bacillus is initiated by DCs that activate
DCs in the outer paracortex of the lymph T cells. Macrophages and B cells evidently pre-
nodes (the most common type) present antigens sent antigens only after antigen-specific T cells
to T cells (10).These paracortical DCs are dis- have been expanded by the initial response. Also,
tinct from the follicular DCs in the germinal the type of antigen presented by macrophages
centers of the lymphoid follicles. and B cells probably differs from that presented
Follicular DCs and antigen-activated helper T by DCs (1). DCs are 2 to 3 logs more efficient
cells interact with B cells for antibody produc- than macrophages and B cells in presenting anti-
tion.Antigens that cause B lymphocytes to pro- gens to T cells, because mature DCs have high
liferate and produce antibodies are sometimes T- levels of costimulatory molecules (such as B7.1
lymphocyte dependent, whereas other antigens and B7.2). DCs also interact with other cells,
122 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

including natural killer (NK) cells and B cells immature DCs mature, and therefore deter-
(12, 13). mines the type of local immune response that
occurs in that organ (see chapters 5 and 7).
DC Kinetics How microbial components and inflamma-
DCs circulate in the blood (4) and are recruited tory cytokines affect DC phenotypes and func-
to sites of inflammation (14), such as tubercu- tions has been reviewed (12, 21), but much
lous lesions. Immature DCs at such sites capture more information is needed to understand the
antigens and carry them to the draining lymph process fully.
nodes (1). There, the DCs mature and present
antigens to antigen-specific T cells that recircu- Tolerogenic DCs
late through the nodes. Not only do DCs initiate and enhance the
In the draining lymph nodes, these antigen- immune response, but “tolerogenic” DCs exist
specific lymphocytes are activated, stimulated to that turn off or suppress the immune response
proliferate, and stimulated to differentiate into through several mechanisms, including the
cytokine-producing effector cells.These antigen- induction of regulatory (suppressor) cells (6, 9,
specific lymphocytes then reenter the circulation 17, 23–26).
and emigrate into sites of inflammation (includ-
ing tuberculous lesions), where infiltrating DCs, DCs as Adjuvants
as well as macrophages and B cells, continue the The use of DCs (expanded in vitro) as adjuvants
antigen-presenting process. to increase the host immune response is begin-
In brief, the primary acquired (adaptive) ning to find clinical application (5, 6, 9, 27).
immune response to tubercle bacilli is initiated
in the draining lymph nodes but continues at the
Conventional and Plasmacytoid DCs
sites of infection (see references 12 and 15).
DC populations have recently been divided
into conventional DCs and plasmacytoid DCs—
How Individual Organs Affect DCs
each with differing functions in both antigen
The following discussion offers some insight
presentation and host defense (28, 29). Plasma-
into why certain organs of the host are more re-
cytoid DCs produce type I (/) interferons
sistant to tuberculosis than other organs (see
that increase host resistance to viruses and pos-
chapters 7 and 25).
sibly other intracellular pathogens.They can be
When injured or irritated, each organ releases
immunostimulating, immunosuppressive, or even
factors that control both innate (nonspecific) and
tolerogenic. Plasmacytoid DCs usually favor a
acquired (adaptive) immune responses. Among
Th1 antigenic response but have a role in Th2
these factors are (i) cytokines (including
responses (29). Chronic tuberculous lesions con-
chemokines) (12, 16) that interact with one
tain many cells that resemble plasma cells (see
another, as well as with receptors on nearby
chapter 4). How many of these cells are typical
cells (12, 17), and (ii) uric acid that is released
antibody-producing cells and how many are
from dying cells and is a strong adjuvant to the
plasmacytoid DCs remains to be determined.
immune response (18). Its use as a potentiator of
vaccines remains to be developed.
ROLE OF THE MAJOR
Organ specificity determines how local and HISTOCOMPATIBILITY COMPLEX
infiltrating immature DCs differentiate into AND CD1 PROTEINS IN ANTIGEN
mature subsets, and these subsets determine the PRESENTATION
functional characteristics of the infiltrating lym- DCs present antigens to most T cells as specific
phocytes (19–21). Each DC subset produces peptides in the context of the major histocom-
different costimulatory molecules for antigen patibility complex (MHC) (1), which means
presentation, which determine the Th1/Th2 that a given T cell (with a clonally expressed 
lymphocyte balance (7, 8, 22). In other words, antigen receptor) will recognize the antigen
each organ determines the subset into which only when its peptide fragments are bound to
6. MACROPHAGES AND OTHER CELLS IN TUBERCULOUS LESIONS 䡵 123

the DC’s MHC molecule. The MHC class I References 12, 30 to 32, 35, and 47 review the
molecules of DCs present peptides (derived complexities of antigen processing and pre-
from proteins that gain access to the cytosol) to sentation and how they are regulated. Reference
cytotoxic CD8 T cells (1).This pathway recog- 48 reviews DC functions in inflammatory sites
nizes and eliminates host cells containing intra- and in organized lymphoid tissue.The section
cellular microorganisms (reviewed in references “Innate Immunity and Its Relation to Acquired
30, 31, and 32).The MHC class II molecules of (Adaptive) Immunity” in chapter 5 has addi-
DCs present peptides (from exogenous proteins tional information on how antigen-presenting
degraded in endosomes) to CD4 T cells (1). cells function.
This pathway produces the delayed-type hyper-
sensitivity (DTH) and cell-mediated immunity PHAGOCYTOSIS OF TUBERCLE
(CMI) that can stop the progression of tuber- BACILLI BY MACROPHAGES AND
culosis. (CD stands for clusters of differentiation, THEIR FATE WITHIN THESE CELLS
specifically, cell surface molecules recognized
Macrophage Receptors
by monoclonal antibodies with the designated
To recognize a microorganism, macrophages
number [1].)
must first recognize patterns on its surface.The
MHCs differ from host to host.Therefore, the
pattern recognition receptors of macrophages
response to various antigens also differs from
(and DCs) include mannose receptors, com-
host to host—in part depending on the affinity
plement receptors, fibronectin receptors, and
of the host’s own MHC molecules for each
Toll-like receptors—all of which are involved in
antigenic peptide.This may be one of the rea-
the host’s innate immune response (1). After
sons why some individuals are more resistant to
acquired immunity develops, macrophages rec-
tuberculosis than others.
ognize antibody-opsonized microorganisms by
CD1 molecules are an antigen-presentation
Fc receptors with help from complement C3b
system that is similar to (but independent of ) the
receptors (reviewed in references 1, 49, and 50).
MHC system (33–36). CD1 proteins on DCs
Extensive studies have recently been made on
have been divided into two groups: group 1
Toll-like receptors (TLRs) (1, 40, 51–53).This
(CD1a, CD1b, CD1c) in rabbits, guinea pigs, and
receptor family is composed of at least 10 mem-
humans, and group 2 (CD1d) in mice (37).The
bers that recognize various mycobacterial cell
characteristics of each group are reviewed in ref-
wall components, including lipoproteins and
erences 34 and 35. Insights into how CD1 mol-
glycolipids (e.g., lipoarabinomannan). Engage-
ecules function are clearly presented in refer-
ment of human macrophage TLRs by 19-kDa
ences 36, 38, and 39.
mycobacterial lipoproteins activates NF-B,
CD1 molecules bind and present (to T cells
induces interleukin 12 (IL-12) secretion, and
with the appropriate receptors) several lipid, gly-
increases the transcription of inducible nitric
colipid, and lipopeptide components of tubercle
oxide synthase (NOS) (54)—all of which par-
bacilli, such as mycolic acid, lipoarabinoman-
ticipate in killing and/or inhibiting tubercle
nan, and mycobactin (32, 35, 36, 38–44; see also
bacilli.TLR-knockout mice produced conflict-
reference 45). These T cells are then activated
ing results on the importance of TLRs in resis-
either to lyse macrophages infected with virulent
tance to tuberculosis (55–57).
Mycobacterium tuberculosis (42) or to produce high
levels of gamma interferon (IFN-) (42), which
is one of the main cytokines increasing the Fate of Mycobacteria within
power of macrophages to inhibit or destroy the Macrophages
bacillus. DCs expressing CD1 may play an Because of the receptors just mentioned, M.
important role in the host immune response to tuberculosis is easily bound to macrophages and
tubercle bacilli. In fact, a mycobacterial vaccine internalized into phagosomes. Ordinarily, such
containing an increased amount of lipid was phagosomes fuse with lysosomes, undergo acid-
more effective than the parent vaccine (46). ification, and receive an abundance of lysosomal
124 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

enzymes that can degrade the microorganisms. Macrophages can inhibit or kill intracellular
However, M. tuberculosis subverts phagosome- tubercle bacilli by means of reactive nitrogen
lysosome fusion and subsequent maturation of and oxygen intermediates (RNIs and ROIs), to
the phagosome into acidic, microbicidal, and which M. tuberculosis is exquisitely susceptible
hydrolytic compartments.M.tuberculosis also resists (67). In mice, chemical inhibition of NOS or
degradation by lysosomal enzymes.Although the the deletion of the NOS gene exacerbates
bacillus inhibits the entry of many microbicidal tuberculosis (68–70). The proteasomes of the
factors into the phagosome (58), it allows nutri- tubercle bacillus are also involved in its resis-
ents (such as iron carried by transferrin) to enter tance against host RNIs (71, 72). See chapters
(reviewed in references 59, 60, 61, and 62). 1 and 25 for additional information on viru-
The tubercle bacillus alters the signal trans- lence.
duction system of macrophages in which it
resides, decreasing the processes that are harm- MYCOBACTERIAL DORMANCY
ful to it. A mycobacterial eukaryotic-like ser- IN MOUSE TUBERCULOUS
ine/threonine protein kinase G modulates sig- GRANULOMAS
nal transduction pathways for trafficking of Tubercle bacilli can live for months in mouse
host-cell organelles (63), thereby inhibiting lyso- granulomas without multiplying and without
some-phagosome fusion.Tetrahydrobenzothio- dying (73, 74).The bacilli presumably are liv-
phene specifically inhibits protein kinase G and ing in activated macrophages (see next section)
is a promising candidate for developing another that inhibit their multiplication. Rees and Hart
class of antimicrobials to combat tuberculosis (73) made homogenates of lungs from albino P
(63). Also, M. tuberculosis inhibits IFN- tran- strain mice after the mice had been infected
scriptional responses without inhibiting activa- intravenously with virulent human-type tuber-
tion of STAT-1 (64). (IFN- is a major cytokine cle bacilli (H37Rv).These homogenates were
that activates macrophages to kill or inhibit cultured to determine the number of live tuber-
tubercle bacilli.) cle bacilli, and smears of these homogenates
In mice, isocitrate lyase is produced by my- were stained by the Ziehl-Neelsen acid-fast
cobacteria that are inhibited within activated procedure and then counted microscopically to
macrophages, but it is not produced by mycobac- determine the number of live-plus-dead bacilli.
teria that are multiplying within nonactivated In these untreated mice, live tubercle bacilli
macrophages (59, 65). This lyase enables the persisted (during the 20 weeks of this experi-
bacillus to subsist on the fatty acids released from ment) at only slightly lower titers than did live-
lipids and thereby avoid being destroyed by plus-dead tubercle bacilli (Fig. 1) (73).Therefore,
the microbicidins of activated macrophages most of the bacilli in the mouse lungs were
(reviewed in references 59 and 65). Isocitrate viable but not multiplying and being killed,
lyase is present in mycobacteria but not in mam- because, if they had multiplied and then were
malian hosts, and therefore is a potential target killed, the number of live-plus-dead bacilli
for new antimicrobials. would have increased.
Virulent tubercle bacilli can persist in a dor- To support this conclusion, Rees and Hart
mancy-like state in solid caseous tissue for many (73) treated mice with pyrazinamide and isoni-
years, and can persist in such a state within acti- azid from 2 months to about 6 months after
vated macrophages for a shorter time (see next infection (Fig. 1). The antimicrobial treatment
section).The accumulation of lipids that enable reduced the number of live bacilli in the lung
such persistence is carried out by a putative tri- homogenates over 10,000-fold (and to uncul-
acylglycerol synthase (66). Isocitrate lyase is turable levels at 6 months), but the number of
involved in the gradual metabolism of such live-plus-dead bacilli was reduced less than 10-
lipids. This metabolism evidently provides the fold (Fig. 1). Therefore, after being killed by
small amount of energy required to sustain via- antimicrobials, dead tubercle bacilli remained
bility in the dormant state. intact in the pulmonary granulomas for several
6. MACROPHAGES AND OTHER CELLS IN TUBERCULOUS LESIONS 䡵 125

FIGURE 1 Viable counts and total counts of virulent tubercle bacilli (H37Rv)
in the lungs of mice from 9 to 25 weeks after an intravenous infection.After the
lungs were homogenized, the viable counts were calculated from the CFU devel-
oping on plates containing solid culture medium.The total counts were calcu-
lated from the bacilli observed microscopically on spread-smears after acid-fast
staining. During this time period, one of the four groups of mice received
isoniazid-pyrazinamide (PZA/INH) daily to kill the bacilli.
Note that for untreated mice the average total counts were 0.3 to 0.4 logs
higher (2.0 to 2.5 times) than the average viable counts, and that PZA/INH treat-
ment markedly reduced the viable counts but had relatively little effect on the
total counts.These findings indicate that (i) most of the dead tubercle bacilli per-
sisted in mouse lungs for many weeks; (ii) most of the live bacilli were in a “dor-
mant” nonmultiplying state, because if they had been multiplying and then had
been killed, the total counts (including the dead bacilli) would have increased;
and (iii) the good CMI developed by mice activated macrophages sufficiently
to prevent the intracellular multiplication of most of the tubercle bacilli. How-
ever, at least some of the bacilli were not inhibited by this good CMI, because
the disease progressed until the mice succumbed. In other words, not all of the
bacilli were dormant, and some bacillary multiplication occurred.
Redrawn from reference 73.These results were confirmed in reference 74
using quantitative real-time PCR technology.
126 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

months with few, if any, cycles of multiplication macrophages for many months, dormancy peri-
and destruction. ods of many years probably only occur in solid
Other experiments reaching the same con- caseous tissue, in which there is no infiltration of
clusions were recently performed by the Mc- fresh macrophages from the circulation.Appar-
Kinney group (74).They used the quantitative ently, two categories of dormancy exist.The first
real-time PCR (polymerase chain reaction) to category (lasting for months) is a nonreplicating
enumerate the live-plus-dead bacilli.The DNA viable (but inhibited) existence within an acti-
of dead tubercle bacilli was found to persist for vated macrophage. This category of dormancy
months as mycobacterial chromosome equiva- ends whenever the macrophage (containing the
lents (74), similar to the acid-fast staining dormant bacillus) dies and the bacillus is ingested
described by Rees and Hart (73). by a nonactivated macrophage. Degenerating
The above experiments indicate that viru- macrophages containing tubercle bacilli are
lent tubercle bacilli persist in the activated sometimes found in older mouse granulomas
macrophages of mouse granulomas without (see chapter 15).
appreciable multiplication or destruction, but The second category (lasting for years) is a
these bacilli grow readily in nonactivated nonreplicating existence within solid caseum
macrophages (see above). Such persistence can- that often ends when and if the caseum lique-
not, however, be considered true dormancy, fies and the bacillus multiplies extracellularly.
because the granulomas progress and eventu- Such caseous necrosis occurs in rabbits, guinea
ally kill the mouse (see chapter 15). pigs, and humans, but not in mice.
Several factors involved in the nonreplicating Several active genes were recently found to be
persistence of virulent tubercle bacilli in mice associated with mycobacterial latency (and per-
have been identified (75–77). Evidently, the haps dormancy) in tuberculous rabbits (Y. C.
bacilli upregulate dormancy regulons that enable Manabe et al., unpublished data). Real-time
them to survive (without multiplying) for weeks reverse transcription-PCR was used to identify
within the phagosomes of macrophages. How- such genes at 5, 10, and 15 weeks after the inhala-
ever, a small proportion of the bacillary popu- tion of human-type tubercle bacilli (H37Rv), and
lation may be in a replicating state (74, 75), also at 15 and 20 weeks in a similar set of rabbits
especially if these bacilli have been ingested by that were treated with dexamethasone from 10 to
nonactivated macrophages that had recently 15 weeks to reactivate the healing lesions. Ten
entered the granuloma. such genes were identified. In rabbits, tuberculous
Live counts and live-plus-dead counts of lesions caused by virulent human-type tubercle
tubercle bacilli should be repeated in tubercu- bacilli begin healing at 5 weeks and are often
lous rabbits and guinea pigs, which develop completely healed by 15 and 20 weeks.
strong DTH and readily form caseous necrotic
tissue. In these animals, numerous persisting MACROPHAGE ACTIVATION
tubercle bacilli would probably be found in Macrophages are the major cells of the mononu-
solid caseum: some live and dormant and some clear phagocyte system (79).This system is com-
dead.Whether tubercle bacilli can persist (as in posed of promonocytes in the bone marrow,
mice) for many months within live macrophages monocytes in the circulation, and macrophages
in a dormant state without multiplying or dying in the tissues. Monocytes are called macrophages
remains to be investigated in rabbits and guinea after they enter the tissues.
pigs. Rabbits may not break down dead tuber- Macrophages in the tissues, especially in sites
cle bacilli any better than mice do, because the of inflammation, differentiate, i.e., become acti-
Wax D component of tubercle bacilli was found vated, for a variety of functions (80–84). Such
to persist in rabbit Mycobacterium bovis BCG differentiation is controlled by local conditions,
lesions for over 56 days (78). especially nearby cells and their cytokines. Cer-
In brief, although nonreplicating dormant tain activated macrophages are highly phago-
tubercle bacilli evidently can persist in activated cytic; others are highly microbicidal (due to
6. MACROPHAGES AND OTHER CELLS IN TUBERCULOUS LESIONS 䡵 127

RNIs and ROIs as well as other factors); others Within the tuberculous lesion, macrophages
are rich in digestive enzymes; others produce also adapt to local conditions (80–82). Macro-
cytokines, e.g., interleukins and tumor necrosis phages must be activated before they can destroy
factor, as well as many other secretory products tubercle bacilli (80, 81, 90). Nonactivated
(85–87); and still others are releasing and/or macrophages provide a very fertile “soil” for
presenting antigens from the tubercle bacilli the intracellular growth of tubercle bacilli. In tis-
within them. Macrophages may combine several sue sections of dermal BCG lesions, highly acti-
of these functions (82). Therefore, the term vated macrophages (staining ⫹⫹⫹ and ⫹⫹⫹⫹
“activated” is generally used rather loosely to with our activation marker -galactosidase)
describe macrophages in which one or more of contained fewer tubercle bacilli than poorly
these activities is enhanced. activated macrophages (Table 2) (80, 81, 90).This
How macrophage functions are affected by finding suggests that such highly activated
local conditions was clearly demonstrated by macrophages had destroyed some of the tuber-
the following experiment of Cohn and Benson cle bacilli they once contained.
(88). Mouse macrophages were cultured in 50% To confirm this conclusion, we produced der-
newborn calf serum for 24 h. After the mal BCG lesions in rabbits by injecting 14C-
macrophages pinocytosed the serum protein labeled BCG (90). In these lesions, we then
(and presumably digested it), their content of determined the percentage of ⫹,⫹⫹,⫹⫹⫹,and
various hydrolytic enzymes increased to high ⫹⫹⫹⫹ -galactosidase-positive macrophages
levels. On the next day, after the 50% serum was that contained the 14C label without any intact
replaced by 1% serum, the content of these bacilli, which shows that these macrophages once
enzymes decreased to low levels. On the third contained intact bacilli but had destroyed them.
day, after the 50% serum was replaced, the con- Table 3 documents our expected findings: highly
tent of these enzymes increased again to high activated (⫹⫹⫹ and ⫹⫹⫹⫹ -galactosidase-
levels.We confirmed this experiment with rab- positive) macrophages contained a much higher
bit macrophages with boiled yeast as the percentage of the 14C label without intact bacilli
digestible substrate (89). Therefore, a “good than did poorly activated (⫹ -galactosidase-
meal” in the pinocytic or phagocytic vacuoles positive) macrophages. This is direct proof that
of macrophages results in the production of activation of macrophages enables them to destroy
enzymes to digest that meal. In other words, the the tubercle bacilli they ingest.
local environment greatly influences how Activation of macrophages can be caused (i)
macrophages differentiate for various functions. by the ingestion of necrotic cells and tissues

TABLE 2 -Galactosidase (-Gal) activity and the number of acid-fast bacilli seen in immature and mature
epithelioid cells in nonnecrotic granulation tissue of dermal BCG lesionsa
Intensity of staining for -Gal
(a marker for macrophage activation) ⫹ ⫹⫹ ⫹⫹⫹ ⫹⫹⫹⫹
No. of -Gal-positive cells counted in each tissue 79 ⫾ 12 104 ⫾ 16 68 ⫾ 16 6.7 ⫾ 2.7
section
No. of tubercle bacilli in ⫹⫹, ⫹⫹⫹, and ⫹⫹⫹⫹ 100% 40 ⫾ 9% 22 ⫾ 5% 1.3 ⫾ 0.8%
-Gal-staining cells as a percentage of the no. in
⫹ staining cellsb
a
Cumulative data from six experiments. One tissue section from each of 29 biopsies was evaluated. Every ⫹ to ⫹⫹⫹⫹
-Gal-positive epithelioid cell in a given area of tissue section was counted. Then, for each tissue section, we calculated the
number of acid-fast tubercle bacilli (found microscopically) in 100 ⫹ -Gal cells, in 100 ⫹⫹ -Gal cells, in 100 ⫹⫹⫹ -Gal
cells, and in 100 ⫹⫹⫹⫹ -Gal cells, and listed it as a percentage of the number of bacilli in 100 ⫹ cells. Relatively few ⫹⫹⫹⫹
-Gal cells were present.The means and their standard errors are shown.The P values for the last row were ⬍ 0.001, except
for 40 vs. 22, which was 0.044.This table indicates that as epithelioid cells (macrophages) mature, they stain more strongly for
-galactosidase and destroy tubercle bacilli more efficiently. (Adapted from reference 90).
b
Immature epithelioid cells are partly activated macrophages, and mature epithelioid cells are fully activated.
128 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

TABLE 3 Macrophage activation and 14C-labeled bacillary components within dermal BCG lesions
% of Macrophages with no
intact bacilli in -Gal categorya
Age of lesions when
Lesions produced by: biopsied (days) ⫹ ⫹⫹ ⫹⫹⫹ ⫹⫹⫹⫹
14
C-labeled bacilli 7 0 0 32.4 21.4
Heat-killed 14C-labeled bacilli 7 0 0 34.6 33.3
a
Percent of macrophages containing only 14C-labeled bacillary components and no intact bacilli in each -galactosidase
(-Gal) category: [14C-pos, but bacilli-neg, cells] divided by [-Gal-pos cells] ⫻ 100).
Note that at 7 days many ⫹⫹⫹ and ⫹⫹⫹⫹ -Gal macrophages contained 14C-labeled bacillary components that were not
attached to intact bacilli, whereas at 7 days all ⫹ and ⫹⫹ -Gal macrophages contained 14C-labeled bacillary components that
were only attached to intact bacilli.
The 14C label produced silver grains in the photographic emulsion covering the tissue section. Ninety percent of the intact
tubercle bacilli (identified by acid-fast staining with carbol-fuchsin) contained silver grains.Therefore, the presence of 14C-labeled
silver grains in macrophages without the presence of intact bacilli indicates that ingested bacilli had been destroyed and broken
down into components that still remained within the macrophages.
In this experiment, heat-killed BCG were not destroyed more readily than live BCG, which is consistent with our finding
that the wax D component of tubercle bacilli can persist in rabbit BCG lesions for at least 56 days (77).
Adapted from reference 90, where complete data and statistics are presented.

and other digestible substances (89), (ii) by the son, Medical College of Virginia, Richmond
cytokines released by lymphocytes and other (103). Cathepsin D, RNase, and phospholipase A2
cells, and (iii) by ligands, such as the mannose were then visualized immunohistochemically in
and lipopolysaccharides, combining with specific tissue sections of BCG lesions (103, 104). The
macrophage receptors. In general, the greatest substrate-film technique was also used to iden-
number of activated macrophages is present tify proteinase, DNase, RNase, hyaluronidase,
when tuberculous lesions reach peak size. chondroitinase, and fibrinolytic activities in such
tissue sections (81, 105, 106). Each of these
HYDROLYTIC ENZYMES OF hydrolases was present in activated macrophages
RABBIT MONONUCLEAR AND within the lesions.
POLYMORPHONUCLEAR EXUDATE
CELLS, PULMONARY ALVEOLAR In vitro ingested tubercle bacilli downregu-
MACROPHAGES, AND LUNG lated cathepsin G and upregulated cathepsins B
HOMOGENATES and D in the human macrophage cell line THP-
Because intracellular hydrolytic enzymes play 1 (107).
major roles in how macrophages digest com-
ponents of tubercle bacilli, we identified and/or MACROPHAGE ENZYMES RELEASED
characterized the proteases, esterase, lipase, EXTRACELLULARLY IN BCG LESIONS
lysozyme, DNase, and/or RNase in rabbit peri- In addition to their ingestive and digestive func-
toneal mononuclear and polymorphonuclear tions, macrophages have extracellular functions
exudate cells, pulmonary alveolar macrophages, (reviewed in references 85, 86, 87, 105, and 108)
and/or lung tissue. References 84, 89, and 91 that appear to be important in tuberculosis.
through 102 provide specific details. Upon activation, macrophages secrete or release
The proteinase cathepsin D (91, 92, 99), a a variety of substances.These include (i) elastase,
chymotrypsin-like esterase (100), and RNase collagenase, and plasminogen activator, (ii)
(102) were purified from rabbit lungs, and anti- lysozyme, (iii) clotting factors, and (iv) interfer-
bodies to the purified cathepsin D and RNase ons, as well as (v) colony-stimulating factors,
were made (99, 102).Antibodies to rabbit phos- which enhance monocyte and granulocyte pro-
pholipase A2 were provided by Richard C. Fran- duction in the bone marrow, and (vi) a variety
6. MACROPHAGES AND OTHER CELLS IN TUBERCULOUS LESIONS 䡵 129

of cytokines, e.g., interleukins, chemokines, and exceeded the concentration of their inhibitors,
factors that stimulate fibroblasts (see chapters 19 and the pH was favorable (108).
and 20).
Most of these studies have been done in vi- MACROPHAGE HETEROGENEITY IN
tro with relatively pure populations of cells.The TISSUE SECTIONS OF BCG LESIONS
studies described below are among the few per- Color Plates 1 and 2 pictorially demonstrate
formed in vivo.They were done before most of macrophage heterogeneity in tuberculous
the cytokines were discovered, so only hydrolytic lesions produced by BCG. For Color Plate 1,
enzymes were measured. the tissue section was double-stained, first for
We placed plastic chambers over dermal BCG cathepsin D (rust color) and then for -galac-
lesions and tuberculin reactions (with the epithe- tosidase (blue). Note that macrophages in the
lium removed) (108–110). The hydrolytic periphery of the lesion contained cathepsin D,
enzymes released into the chamber were assayed, whereas macrophages near the caseous center
and a quantitative histochemical study was made contained -galactosidase. We called this type
of the number of activated (-galactosidase- of macrophage distribution “macrolocal” or
positive) macrophages in the chamber beds. regional (82).
Although a portion of the enzymes assayed For Color Plate 2, the tissue section was dou-
could have come from other cell types, ble-stained, first for acid phosphatase (red) and
macrophages were probably the major source, then for -galactosidase (blue). Macrophages
because they were the dominant cell in the containing acid phosphatase were sometimes
lesions and contained these enzymes. next to macrophages containing -galactosi-
At 18 days of age, the BCG lesions were dase.We called this type of distribution “microlo-
largest (Fig. 2B), and the activated macrophages cal,” i.e., individual macrophages could be acti-
in the chamber beds were most numerous (Fig. vated for different functions, even though they
2C and 3). Between 11 and 18 days, the levels were adjacent to each other in the tuberculous
of the five enzymes (assayed in the chamber lesion. Some macrophages (colored purple) were
fluids) reached their peaks (Fig. 4).At this time, activated for both acid phosphatase and -galac-
tuberculin hypersensitivity was well developed tosidase (Color Plate 2).
(Fig. 2A), and the bacilli and their components We quantitated the macrolocal and microlo-
were still present. Two-week-old polystyrene cal activation of macrophages in developing,
lesions (Fig. 5) served as controls. peak, and healing dermal BCG lesions. The
The enzyme levels in 2-day chamber fluids amounts of the four macrophage enzymes eval-
from tuberculin reactions (1, 2, and 3 days old) uated were highest when the lesions reached
were not statistically different from the levels in peak size (Fig. 6). Macrophages staining for -
BCG lesions (Fig. 4). galactosidase and esterase were always more
Active collagenase (an enzyme secreted but numerous near the caseous center of the lesions
not stored in macrophages) was only detected in than in the periphery (Color Plate 1 and Fig. 7
fluids from peak BCG lesions (108). Evidently, and 8). In contrast, macrophages staining for
the serum in the chamber fluids was sufficient cathepsin D and acid phosphatase were always
to inhibit the lower collagenase levels that were more numerous in the periphery than near the
probably released from smaller BCG lesions and caseous center (Color Plate 1 and Fig. 7 and 8).
tuberculin reactions. In tuberculous lesions, macrolocal and
These studies demonstrate that, in chronic microlocal heterogeneity should also occur in
inflammatory lesions produced by the tubercle macrophage populations producing various
bacillus, both acid-acting and neutral-acting cytokines, RNIs and ROIs, and many other
hydrolytic enzymes are released extracellularly. cell functions, including the upregulation of
Tissue components would be hydrolyzed locally different types of receptors.Therefore, the obser-
when the concentration of these enzymes vations just portrayed clearly establish a general
FIGURE 2 (A) Size of 48-h tuberculin reactions. (B) Size of BCG lesions. (C) Weighted num-
ber of activated -galactosidase-positive macrophages, just below the surface of the chamber beds
of BCG lesions (closed circles) and 72-h tuberculin reactions (open circles). To obtain the

130
6. MACROPHAGES AND OTHER CELLS IN TUBERCULOUS LESIONS 䡵 131

FIGURE 3 Chamber bed of a 32-day BCG lesion. Many darkly staining ⫹⫹⫹ and ⫹⫹⫹⫹
-galactosidase-positive macrophages are present. A thin proteinaceous layer covers the surface
of the chamber bed. Stained with 5-bromo-4-chloro-3-indolyl--D-galactoside, hematoxylin and
eosin. Magnification, ⫻30. Reproduced with permission from reference 108.

pattern for all inflammatory reactions, namely, CAUSES OF MACROLOCAL AND


that the functions of the participating cells are MICROLOCAL MACROPHAGE
locally controlled. HETEROGENEITY
Other types of macrophage heterogeneity were How and why such macrolocal and microlocal
recently reviewed by Mosser (83).There is het- distribution of macrophages occurs in tubercu-
erogeneity in their receptors,in the cytokines they lous lesions is a matter of conjecture (82).Vari-
produce (including chemokines), and in their ous possibilities exist, but many need experi-
microbicidal abilities. Some macrophages favor mental proof.
the Th2 response over the usual (IL-12-induced) 1. Macrophages probably differentiate locally
Th1 response. Some macrophages downregulate for various functions because of interac-
inflammatory and immune responses by pro- tions with other cells in their environ-
ducing IL-10 and IL-1-receptor antagonist. ment.Adjacent (or nearby) lymphocytes,
And some macrophages stimulate repair and plasma cells, granulocytes, vascular en-
healing by producing transforming growth dothelial cells, fibroblasts, and even other
factor . macrophages may determine the direction

weighted number of -galactosidase-positive macrophages, the number of ⫹ cells was multiplied


by 1, ⫹⫹ cells were multiplied by 2, ⫹⫹⫹ cells were multiplied by 3, and ⫹⫹⫹⫹ cells were
multiplied by 4, and then the products were added together. Reproduced with permission from
reference 108.
132 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS
6. MACROPHAGES AND OTHER CELLS IN TUBERCULOUS LESIONS 䡵 133

in which a given macrophage activates.


Macrophages may express different surface
receptors that determine how these cells
respond to stimuli from adjacent cells.
2. Macrophages may become activated for
one function when the lesion is young
and remain activated for the same function
when the lesion is older. In other words,
two adjacent macrophages can be acti-
vated for a different function at different
times.
3. Macrophages may be activated in one part
of the lesion and then migrate to another
part, possibly due to different chemokine FIGURE 5 Tissue section of a 14-day rabbit skin
lesion produced by the intradermal injection of 60 ⫻
receptors.
106 polystyrene latex particles. This lesion showed no
4. The hydrolytic enzyme content of each inflammation. All of the polystyrene particles were
macrophage may be determined by what within macrophages staining ⫹⫹ to ⫹⫹⫹⫹ for -
it ingests, e.g., the ingestion of lipids may galactosidase. Note that the dermal collagen fibers adja-
stimulate lipase production. If so, in the cent to the macrophages are intact, suggesting that no
effective collagenase was produced by these cells. A
tuberculous lesion, the location of each
small amount of collagenase was found at 48 h in flu-
type of activated macrophage would ids within chambers placed over the polystyrene lesions,
reflect the location of the lipids, proteins, but 2 to 3 times this amount of collagenase was found
carbohydrates and nucleic acids, which in chamber fluids placed over 14-day BCG lesions,
are probably not distributed evenly. where the collagen fibers were hydrolyzed (108). Stained
with 5-bromo-4-chloro-3-indolyl--D-galactoside,
5. The presence or absence of tubercle bacilli
hematoxylin and eosin. Magnification, ⫻350. Repro-
within a macrophage also affects its type duced with permission from reference 108.
of activation (64).

EPITHELIOID CELLS
Epithelioid cells are macrophages in various bicidins, and therefore are much more effective
states of activation that are sometimes orga- than immature epithelioid cells in destroying
nized into an epithelium-like pattern due to ingested bacilli (80–82, 111, 112). Some epithe-
adhesion molecules (see chapter 21). In tuber- lioid cells are secretory macrophages.
culosis, epithelioid cells have large vesicular
(euchromatic) nuclei, suggesting active DNA LANGHANS’ GIANT CELLS
transcription for synthetic functions. Mature In tuberculosis, Langhans’ giant cells occur more
epithelioid cells are highly activated cells (Fig. frequently in human lesions than in rabbit
9 and 10).They are rich in enzymes and micro- lesions (see chapter 3).A Langhans’ giant cell is

FIGURE 4 Lysozyme, RNase, DNase, and lactic dehydrogenase (LDH) activities in the cham-
ber fluids of BCG lesions of various ages (closed circles and line graph), 1-day tuberculin reac-
tions (open circles), and normal skin controls (shaded horizontal lines). Lysozyme is both secreted
and stored; RNase and DNase are released on cell death and possibly regurgitated, but not secreted;
and LDH is released only on cell death.
The chambers were glued to the still intact skin around the area where the epidermis was
removed.They were then filled with HEPES culture medium 199, and the fluid within the cham-
bers was collected 48 h later. Note that the highest level of these extracellular hydrolases occurred
when tuberculin sensitivity had developed, the BCG lesions were growing to peak size, and the
greatest number of activated macrophages was in the chamber bed (see Fig. 1). Reproduced with
permission from reference 108.
134 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

FIGURE 6 Estimates of the total amount


of each of four enzymes in tissue sections of
dermal BCG lesions during their develop-
ment and healing. The mononuclear cells
(mostly macrophages) in these tissue sections
were single-stained for acid phosphatase,
cathepsin D, esterase and -galactosidase and
were evaluated microscopically at both 35⫻
and 125⫻ magnifications. For quantitation,
both the distribution and intensity of the his-
tochemically produced color were taken into
account. Then, the total amount of staining
was given a rating on a 0 to 9 scale.The stan-
dard errors of the means are shown.
Note that the total amount of each enzyme
was greatest when the lesions peaked in size
(Fig. 2B), and the levels of each enzyme more
or less rose and fell in parallel during the devel-
opment and healing of the lesions. Repro-
duced with permission from reference 82.

FIGURE 7 Distribution of mononuclear cells (MN) (mostly macrophages) that single-stained


for acid phosphatase, cathepsin D, -galactosidase, or esterase in developing, peak, and healing der-
mal BCG lesions.The percentage of MN staining ⫹ to ⫹⫹⫹⫹ was evaluated microscopically
at the edge of the caseous necrotic center, in the viable tissue near this center, and more periph-
erally in representative high-power fields at ⫻500 magnification.The standard errors of the means
are shown.
Note that macrophages containing -galactosidase and esterase were more frequent near the
caseous center (A and B), and macrophages containing acid phosphatase and cathepsin D were
more frequent in the peripheral regions (C). These findings quantitatively demonstrate the
“macrolocal” distribution of activated macrophages within tuberculous lesions (see text). Repro-
duced with permission from reference 82.
6. MACROPHAGES AND OTHER CELLS IN TUBERCULOUS LESIONS 䡵 135

FIGURE 8 Distribution of activated mononuclear cells (mainly macrophages) in double-stained


tissue sections of developing, peak, and healing rabbit dermal BCG lesions.These sections were
stained histochemically for pairs of enzymes: one red and one blue.The red enzymes were acid
phosphatase, cathepsin D, or red esterase.The blue enzymes were -galactosidase or blue esterase.
The mononuclear cells were counted microscopically at the edge of the caseous necrotic cen-
ter (A), in the viable tissue near this necrosis (B), and in peripheral areas of the lesion (C).
The length of the bars represents 100% of the mononuclear cells in each area.The cells that
stained for only one enzyme of the pair were either red (stippled bars) or blue (hatched bars).
The cells that stained for both enzymes of the pair were purple (black bars).
The black bars in column 3 and the mirror-image patterns C:B:A and A:B:C in columns 1
and 2 show that the same macrophage population contained -galactosidase, red esterase, and blue
esterase. Macrophages containing cathepsin D and acid phosphatase had similar distributions (see
columns 1 and 2), but since they both stained red, they could not be differentiated in the same
tissue section. (The red esterase is from a diazo dye; the blue esterase is from an indolyl dye [82].)
These graphs confirm the microlocal mononuclear cell activation shown in Color Plate 2, i.e.,
that some macrophages almost always stain for an enzyme different from that of the majority of
macrophages in a given area.
Reproduced with permission from reference 82.
136 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

FIGURE 9 Tissue section of a 21-day rabbit dermal BCG lesion stained for -galactosidase,
our marker enzyme for macrophage activation.A group of epithelioid cells with high enzyme
activity is seen in the tuberculous granulation tissue that surrounds the lesion’s liquefied
caseous center (in the lower right corner of the photograph). Mature epithelioid cells (iden-
tified by their rounded appearance) stain the strongest (⫹⫹⫹ and ⫹⫹⫹⫹) for -galactosi-
dase.These activated macrophages cluster in the area of the BCG lesion where the bacilli (iden-
tified by acid-fast staining) are located. Stained with 5-bromo-4-chloro-3-indolyl--
D-galactoside, lightly counterstained with hematoxylin. Magnification, ⫻200. Reproduced
with permission from reference 80.
This picture clearly demonstrates the principle of local immunity, i.e., the bacilli and their
products stimulate local lymphocytes to produce cytokines that activate nearby macrophages.
Highly activated macrophages are known to contain high concentrations of reactive oxygen
and nitrogen intermediates and hydrolytic enzymes that kill or inhibit the tubercle bacillus. Such
highly activated macrophages may be harmful to tissues, especially if they die and release their
contents.Therefore, the host apparently limits the activation of macrophages to local sites of
bacillary lodgement where they are most needed to control the infection.

a multinucleated epithelioid cell. It is formed MONONUCLEAR CELL TURNOVER


when two cells fuse together (sometimes around Macrophages and lymphocytes entering an
a bit of caseous material), or when one cell fails established tuberculous lesion do not remain
to divide after duplicating its nucleus, or both there very long (113). Within 10 days, more
(112). Their nuclei are arranged about the than 90% of them have “turned over,” i.e., they
periphery of the cell, in contrast to the usual for- have died (because of apoptosis or tissue-dam-
eign-body type of giant cell, in which the nuclei aging DTH) or have left the lesion via the
are scattered throughout the cytoplasm. The lymphatics and have been replaced by new
presence of Langhans’ giant cells seems to be a macrophages. With the development of CMI
sign of a decrease in the progression of the dis- and DTH, the rate of entry of these cells, their
ease, as well as a sign of chronicity (111, 112). rate of loss, and their rate of activation are
These cells do not play a major role in the increased. In the developing lesion, the rate of
pathogenesis of tuberculosis (112). entry exceeds the rate of loss, so large numbers
6. MACROPHAGES AND OTHER CELLS IN TUBERCULOUS LESIONS 䡵 137

of macrophages and lymphocytes accumulate Common T-cell subsets


and activate. Chapter 10 presents details on CD4
macrophage turnover. • Th1 cells (producing IL-1, IL-2, TNF-␣, and
IFN-␥)
In almost-healed BCG lesions, macrophage • Th2 cells (producing IL-1, IL-2, TNF-␣, IL-4,
turnover still occurs but at reduced levels.This IL-5, IL 10, and IL-13)
suggests that, even in arrested tuberculous lesions, • Regulatory T cells (CD4⫹/CD25⫹) (produ-
a few nonactivated macrophages enter and some cing IL-10 and TGF-␤)
intracellular bacillary multiplication may occur CD8
• Cytotoxic T cells
(113).True bacillary dormancy may only exist in • Cytokine-producing cells (IFN-␥ and others)
solid caseous tissues and not in the viable tissues
that surround such necrotic foci (113). FIGURE 11 A generalized presentation of the com-
mon types of T cells, i.e., those with  antigen recep-
tors (see reference 1). Note that T cells with the CD4
OVERVIEW OF LYMPHOCYTES: and CD8 surface markers can produce similar cytokines.
THEIR SUBSETS AND THEIR
FUNCTIONS
Lymphocytes (Table 1) are the antigen-specific
have a receptor, they respond by clonal prolif-
effector cells of the host’s acquired (adaptive)
eration, which greatly increases their number.
immune response (1).Their role in the patho-
Antigen-specific B lymphocytes (called B
genesis of tuberculosis is described in chapter 5.
cells) come from the bone marrow and produce
Lymphocytes develop clonally expressed anti-
antibodies. Antibodies enhance the local accu-
gen-specific receptors by DNA rearrangements.
mulation of defense cells in tuberculous lesions
When exposed to the antigen for which they
(see chapter 5). B cells make greater amounts of
antibody when they differentiate into plasma
cells. Plasma cells are common in tuberculous
lesions (see chapter 4).
Antigen-specific T lymphocytes (called T cells)
come from the thymus.They produce the CMI
and DTH that develop in tuberculosis and other
infections. T cells recognize antigens that are
processed and bound to either MHC (class I or
II) or CD1 molecules (see “Antigen-Presenting
Cells” at the beginning of this chapter).
T lymphocytes have been divided into vari-
ous subsets by their CD4 and CD8 surface anti-
gens, by their functions (T helper [Th] cells, reg-
ulatory T cells, and cytotoxic T cells), and by the
FIGURE 10 Tissue section of a 33-day rabbit der-
mal BCG lesion stained for cytochrome oxidase, an cytokines they produce (Th1 and Th2) (Fig.
enzyme involved in oxygen metabolism. The large 11). For additional details, the reader should
cells are epithelioid cells, similar to those in Fig. 9.The consult reference 15 and textbooks of immunol-
most mature ones with the rounded appearance stain ogy (e.g., reference 1). Most of the studies on the
the darkest. In other words, the macrophages most types and functions of lymphocytes in tubercu-
effective in inhibiting the intracellular growth of the
tubercle bacillus contained the highest levels of both losis have been made in mice (40, 41, 114, 115).
hydrolytic and oxidative enzymes. The other cells Numerous lymphocytes accumulate in tuber-
(which we cannot differentiate) are probably small culous lesions that do not seem to have any
macrophages, dendritic cells, lymphocytes, and plasma receptor specific for antigens of tubercle bacilli.
cells. Stained with 8-amino-1,2,3,4-tetrahydroquino- The role of these bystander lymphocytes remains
line and p-aminodiphenylamine (84) with no coun-
terstain, so the cell nuclei stain lighter than the cyto- to be determined. They probably produce
plasm. Magnification, ⫻470. Reproduced with cytokines that participate in this chronic inflam-
permission from reference 81. matory reaction.
138 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

In the host’s circulation and in lymphoid tis- that the presence of tuberculin sensitivity
sues, the number of naive lymphocytes and the enhanced the accumulation of cytotoxic T cells,
number of memory lymphocytes seem to be which could kill macrophages in which tuber-
controlled independently (116). During any cle bacilli were growing. At 5 days, reinfection
infection, including tuberculosis, antigen-specific BCG lesions showed a higher percentage of
lymphocytes proliferate, and the entire lym- both CD4 and CD8 cells than did primary
phoid system adjusts accordingly. BCG lesions (Fig. 12). Cytotoxic M. tuberculosis-
The overall control of the number of lym- specific CD8 T cells preferentially recognize
phocytes in the host was recently reviewed in cells that are heavily infected with tubercle
reference 117. bacilli (120). CD8 functions in infectious diseases
are reviewed in reference 121.
TH1, TH2, CD4, CD8, TREG,
AND  T CELLS Regulatory T Cells
Th Cells Regulatory T cells (Treg), formerly called sup-
The Th1 subset produces IL-2 (which causes T- pressor T cells (122), downregulate antigen-
cell proliferation) and IFN- and tumor necro- specific immune responses, usually by produc-
sis factor beta (TNF-), both of which activate ing IL-10 (123, 124). These lymphocytes also
macrophages. IFN- downregulates the Th2 stimulate macrophages and other cells to pro-
response. By means of reverse transcription- duce prostaglandin E2 and other suppressor fac-
PCR, we evaluated changes in IFN- in devel- tors, including lipoxins (1, 87, 125). In addi-
oping and healing tuberculous lesions produced tion, many inflammatory mediators, such as the
by BCG in rabbits. IFN- production seemed cytokines TNF-, IFN-, and transforming
to occur later than the production of the growth factor , as well as oxygen and nitrogen
chemokine MCP-1 (see chapter 19). intermediates, can switch from being pro-
IL-12 is a major cytokine that specifically inflammatory to anti-inflammatory (87) and
expands the Th1 population and upregulates its thereby probably contribute to the regression
functions. Macrophages and DCs are the main and healing of tuberculous lesions. See references
producers of IL-12, especially when they con- 124 and 126 through 130 for details on Treg cell
tain intracellular microorganisms, such as M. function in vitro and in vivo.A substantial num-
tuberculosis (118).Therefore, IL-12 plays a major ber of these cells are CD4⫹CD25⫹ (131, 132).
role in acquired host resistance to tuberculosis. Treg cells keep the immune response from
It also plays a role in innate immunity by acti- becoming excessive and causing tissue damage.
vating NK cells. When infections are properly controlled, the
The Th2 subset produces IL-4, IL-5, IL-6, IL- proper balance between effector immunocytes
10, and IL-13, all of which promote antibody and Treg cells exists (133).
production by B cells. IL-4, IL-10, and IL-13
downregulate the Th1 response.  T Cells
Most T cells have alpha-beta () antigen recep-
CD4 and CD8 Lymphocytes in tors (Fig. 11). However, a small population of T
Tuberculous Lesions cells possessing gamma-delta () antigen recep-
The percentage of CD4 and CD8 cells in the tors also exists (reviewed in reference 134).
mononuclear cell population of rabbit dermal Since  T cells are activated before  T cells,
BCG lesions was determined (119). During they have been postulated to play a role in the
both the development and healing of the BCG early antigen-specific immune response against
lesions, CD4 cells were always more numerous tuberculosis (135, 136). Health care profession-
than CD8 cells (Fig. 12 and Table 4).At 2 days, als recently exposed to M. tuberculosis, as well as
reinfection BCG lesions and tuberculin reactions patients with active infection, have increased
showed a higher percentage of CD8 cells than numbers of  T cells in their blood (137–140;
did primary BCG lesions (Fig. 12), suggesting also see references 141–143).
6. MACROPHAGES AND OTHER CELLS IN TUBERCULOUS LESIONS 䡵 139

FIGURE 12 Percentage of mononuclear


cells (MN) (mostly lymphocytes) immuno-
stained for CD4 (A) or CD8 (B) in primary
and reinfection BCG lesions and in tuber-
culin reactions. At 2 days, the reinfection
BCG lesions and the tuberculin reactions
contained a higher percentage of CD8 cells
than did the primary lesions, suggesting that
tuberculin sensitivity increases the number
of cytotoxic CD8 cells in tuberculous
lesions. Note, however, that CD4 cells are
always much more numerous than CD8
cells (compare the y axes).
Each point represents the mean of four
lesions with its standard error. For reinfec-
tion BCG lesions versus primary BCG
lesions: *P ⬍ 0.05 and **P ⬍ 0.01; for
tuberculin reactions versus primary BCG
lesions: †P ⬍ 0.05; and for reinfection
BCG lesions versus tuberculin reactions:
‡P ⬍ 0.05 and ‡‡P ⬍ 0.01. Reproduced
with permission from reference 119.

 T cells seem to have a much wider influ-  T cells expressing the V2V2 T-cell
ence on the host’s response to microorganisms receptor constitute the majority of circulating
than was previously believed, because they affect  T cells in humans and nonhuman primates,
functions of NK cells, DCs, B cells, and other T but are absent in mice (32, 145).The V2V2 T
cells, in part by producing IFN- and other cells recognize small organic phosphate anti-
cytokines (134, 144).At least some of the  T gens that are present in M. tuberculosis. In
cells have receptors that combine directly with macaques after BCG reinfection, the V2V2⫹
intact antigens without the usual processing and T-cell subset expands 2 to 9 times. Similarly,
presentation by DCs. In other words, unlike BCG-vaccinated macaques have a rapid recall of
T cells with  receptors, this  group does V2V2 T cells in bronchoalveolar lavages after
not require processing of antigens and is a challenge with virulent M. tuberculosis (146).
neither class I nor class II MHC restricted (1). These findings indicate that V2V2⫹ T cells
140 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

TABLE 4 Percentage of CD4 and CD8 lymphocytes in the mononuclear cell population of primary BCG
lesions during their development and healinga
Age of BCG % of CD4 % of CD8 Ratio CD4/CD8 Ratio CD8/CD4
lesion (days) cells (A) cells (B) (A/B) (B/A)
9 13.2 ⫾ 1.2 8.7 ⫾ 2.1 1.5 0.66
23 17.8 ⫾ 1.1 5.4 ⫾ 0.9 3.3 0.30
37 15.7 ⫾ 2.4 6.3 ⫾ 1.6 2.5 0.40
a
Note that the number of CD4 cells was always higher than the number of CD8 cells (A/B) and that the ratio of CD8 to
CD4 cells (CD8/CD4 ⫽ B/A) was highest in 9-day lesions. At 9 days, many bacilli were multiplying in nonactivated macrophages,
and DTH developed that stopped such multiplication (see chapter 2). (Adapted from reference 119.)
In single-cell suspensions of rabbit tuberculous lungs and their hilar lymph nodes, similar ratios of CD4/CD8 cells were found
by flow cytometry (unpublished data).

are important contributors to the acquired morphology of large granular lymphocytes and
immune response to tubercle bacilli. are currently defined as cytotoxic cells that do
V2V2 T cells are absent in mice (32, 145), not express surface CD3 or T-cell antigen recep-
but they have V4⫹  T cells that regulate air- tors, but do express CD56.
way hyperreactivity (147). The NK cells have both MHC class I and
non-MHC-requiring receptors, whereas the
MEMORY T CELLS cytotoxic T (CD8) cells recognize only specific
Memory CD4 and CD8 T lymphocytes are of antigenic peptides in association with MHC
two types: central memory cells and effector class I molecules. In fact, engagement of surface
memory cells (15). Effector memory cells are receptors on NK cells with MHC class I mol-
located in peripheral tissues and in sites of ecules on host cells usually turns off, rather than
inflammation, where they provide immediate activates, the lytic machinery of the NK cells
protection against microorganisms (15). Central (148). Cells infected by many viruses and other
memory cells are located in the lymphoid intracellular microorganisms have little or no
organs, primarily the lymph nodes and spleen. MHC class I expression and will be lysed by NK
There, they can rapidly expand and differenti- cells, whereas healthy cells (expressing normal
ate to supply effector T cells to peripheral sites levels of MHC I) will not be lysed. Some viruses
(15).The two types can be recognized by their produce MHC class I homologs that prevent
CD surface markers (15).Vaccines that greatly NK cells from lysing host cells that contain
increase the number and availability of memory these viruses (148), but similar virulence mech-
cells should provide the best protection against anisms have yet to be investigated in mycobac-
clinical tuberculosis (see chapter 22). teria.
NK CELLS, CYTOTOXIC Since NK cells can kill infected host cells
T LYMPHOCYTES, AND APOPTOSIS without prior sensitization, they are considered
The killing of poorly activated macrophages in part of the innate immune system (149). In
which tubercle bacilli are multiplying intracel- tuberculosis, NK cells are an early source of
lularly is an important factor in the control of IFN-, which activates macrophages and en-
tuberculosis (1) (see chapter 2). NK cells and hances Th1 immunity. Also, macrophages can
cytotoxic T lymphocytes (CTLs) play major activate NK cells (150).
roles in this host defense reaction. NK1.1 T cells express  T-cell receptors
that recognize CD1 molecules, rather than
NK Cells MHC class I or MHC class II molecules (1; also
NK cells (see Table 1) compose 5 to 10% of the see references 36 and 151). Since they mostly
peripheral blood lymphocyte population.They respond to lipid and glycolipid antigens, they
proliferate in response to both macrophage and probably play a role in the early host response
T-cell-derived cytokines. NK cells have the to tubercle bacilli.
6. MACROPHAGES AND OTHER CELLS IN TUBERCULOUS LESIONS 䡵 141

Only a few studies have been made on NK sis (155). In humans, however, NK cells are
cells and mycobacteria. Human peripheral blood probably more important, because humans defi-
mononuclear cells kill more intracellular M. cient in NK cells are extremely susceptible to
tuberculosis when cultured with NK cells (152). intracellular infections such as that caused by
Also, human NK cells (stimulated in vitro with herpesvirus (156).
IL-12) activate macrophages to inhibit the intra- NK cells can directly lyse cells in which bac-
cellular growth of Mycobacterium avium (153). teria and viruses are multiplying. Therefore,
Macrophages containing intracellular bacte- NK cells are a major force in early antigen-
ria produce TNF- and IL-12, which activate independent host resistance to tuberculosis.
NK cells. NK cells, in turn, produce much IFN- NK cells also interact with DCs, presumably in
 (and many other cytokines) that (i) activate ways beneficial to the host (157, 158).
macrophages nonspecifically early during an NK cell functions are discussed more fully in
infection, and (ii) favor a subsequent Th1 spe- Immunobiology (1) and in reference 149.
cific immune response over a Th2 response
(154). However, NK cells can also produce IL- CTLs
5 and IL-13, which favor a Th2 response (154). Antigen-specific CTLs come from the thymus,
In mice, the removal of NK cells did not as do other antigen-specific T cells. Most CTLs
substantially alter host resistance to tuberculo- are CD8 cells. They recognize self-peptide

FIGURE 13 Mononuclear cells (MN) and granulocytes (PMN) per mm2 of tissue section in
BCG lesions at various times during their development and healing.The mononuclear cells were
mostly macrophages with some medium and large lymphocytes (and probably some dendritic
cells). In the BCG lesions, only the areas that were densely infiltrated with cells were counted.
These areas were usually found about one-third of the distance from the edge of the caseous cen-
ter to the edge of the lesion. PMN were even more numerous nearer the caseous center. At 37
days, the BCG lesions were much smaller, so the total number of cells present was much reduced.
Reproduced with permission from reference 163.
142 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

FIGURE 14 A tissue section of a 19-day rabbit dermal BCG lesion incubated on a film of RNA
for 1 h at 23°C and stained with toluidine blue. Note the “starry sky” appearance representing
RNase activity in a percentage of the granulocytes.The cells that have digested the substrate film
beneath the tissue section were probably eosinophils, because eosinophils are known to contain
high levels of RNase (181, 182), and because most of the PMN (recognized by their multilobed
nucleus) were inactive. Magnification, ⫻180. Reproduced with permission from reference 81.

antigens in the context of MHC class I. CTLs The perforin-independent (nonsecretory)


require activation by exogenous antigens, as pathway of apoptosis occurs by cross-linking
well as by cytokines (IL-2 and others), to (on the target cell’s surface) Fas (CD95) or
develop their full cytotoxic activity. Most (but TNF-receptor molecules with the Fas ligand on
not all) CD8 cells exhibit a Th1 cytokine pro- NK cells and CTLs. Fas is defined as a cell
file. References 159 and 160 provide more receptor that induces apoptosis. (In humans, Fas
details on CTLs.Their role in mouse tubercu- is also known as APO-1.) The cross-linking of
losis is reviewed in references 40 and 41. Fas molecules on cell membranes initiates pro-
grammed cell death via the sphingomyelin
Apoptosis (ceramide) signal transduction pathway.
Both NK cells and CTLs kill their target cells by Apoptosis keeps the immune response to var-
perforin-dependent and perforin-independent ious antigens from becoming excessive (161).
apoptotic pathways (1). Perforin is a protein that Also, the viability of monocytes/macrophages
can polymerize to form a hole (or pore) in the (and probably activated lymphocytes) depends
target cell’s membrane. Perforin, cytolysin, and on stimulatory cytokines, without which these
hydrolytic enzymes (called granzymes) are stored cells undergo apoptosis (162).
in secretory granules of NK cells and CTLs. In mice, apoptosis evidently plays a major
After entry through the perforin-produced hole role in killing macrophages containing numer-
in the target cell’s membrane, granzymes (espe- ous intracellular tubercle bacilli, because in
cially proteases) apparently contribute to the mouse tuberculosis very little necrosis occurs
DNA fragmentation that characterizes apopto- (see chapter 15).The relative roles of apoptosis
sis, which is programmed cell death. and caseous necrosis in controlling the numbers
6. MACROPHAGES AND OTHER CELLS IN TUBERCULOUS LESIONS 䡵 143

of bacilli in tuberculous humans, guinea pigs, and are major factors in recruiting PMN to sites of
rabbits remain to be determined. inflammation (166). The number of PMN in
an inflammatory lesion is determined not only
GRANULOCYTES IN by their rate of entry, but also by the rate at
TUBERCULOUS LESIONS which they undergo apoptosis (167).
Polymorphonuclear leukocytes (PMN) are pre- Eosinophils are also present in tuberculous
sent in tuberculous lesions (Fig. 13), especially lesions (168), but eosinophils are not easily dis-
near their caseous centers (Color Plate 2) (163). tinguished from PMN in rabbits. Rabbit eo-
They do not seem to be able to destroy tuber- sinophils often have large orange-brown egg-
cle bacilli and may, in fact, play a role in their dis- shaped granules, whereas rabbit PMN usually
semination (112). Rabbit PMN are eosinophilic, have smaller granules and stain a brighter orange
in contrast to human, mouse, and guinea pig (164). The eosinophils in rabbit BCG lesions
PMN, which are neutrophilic. Because of their seem to show higher ribonuclease activity than
eosin staining, rabbit PMN should be called het- any other cell present (Fig. 14). However, per-
erophils, not neutrophils (164). PMN evidently oxidase staining with diaminobenzidine would
recruit immature DCs to sites of inflammation probably distinguish eosinophils from PMN
and activate them (165). Platelet chemokines with more certainty (164). Reference 169
(e.g., platelet factor 4 and -thromboglobulins) reviews the beneficial and detrimental effects of
eosinophils on cells participating in inflamma-
tory processes. The role of eotaxin in mouse
mycobacterial granulomas was analyzed in ref-
erence 170.
We have made no studies on basophils and
mast cells in rabbit tuberculous lesions, but we
have studied basophils and mast cells in dermal
lesions produced by the chemical irritant dilute
sulfur mustard (164, 171–173). For these studies,
we prepared 1- to 2-m sections from tissues
embedded in glycol-methacrylate (GMA) (Fig.
15 and 16) (164, 174, 175). GMA preserves blue-
purple (i.e., basophilic) granules within these

FIGURE 15 A postcapillary venule in a guinea pig


contact-sensitivity reaction to dinitrochlorobenzene.
Note the preservation of three basophils (arrows): one
just outside the venule, one inside the venule, and one
between the endothelium and its basement membrane.
In this thin GMA tissue section, endothelial cells and
pericytes can be easily distinguished by their location.
In other words, these thin plastic-embedded tissue sec-
tions enable a resolution with light microscopy that
approaches the resolution with low-power electron
microscopy. In guinea pigs, rabbits, and humans, mast
cells and basophils can be easily distinguished by their
shape and staining characteristics (164).The tissue spec- FIGURE 16 Mast cells in a tissue section of normal
imen was embedded in GMA, cut at 1 to 2 m, and rabbit skin.The tissue specimen was “cold-embedded”
stained with Giemsa. Magnification, ⫻790. Reproduced in GMA (174), cut at 1 to 2 m, and stained with
with permission from reference 164. Giemsa. Magnification, ⫻900.
144 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

two cell types.These granules are often destroyed by the sulfur mustard and to correlate this
in paraffin-embedded preparations. In the mild, degranulation with the amount of histamine in
slowly developing acute dermal inflammatory the organ-culture fluids (173).
lesions produced by 1% sulfur mustard, basophils At sites of many infections, mast cell cytokines
in relatively large numbers (20 to 30% of the have been found to enhance the recruitment of
granulocyte infiltrate) entered during the first T cells (176, 177).They probably play a similar
2 h in both rabbits and guinea pigs (Fig. 15) role in lesions produced by the tubercle bacillus.
(171).After this time, the percentage of basophils Reference 178 is an extensive review of the
decreased (171). Perhaps, basophils (and the his- ultrastructure of human mast cells and basophils
tamine that they contain) enhance many early and how their organelles function.
inflammatory responses, including those pro-
duced by the tubercle bacillus. FIBROBLASTS IN
We also evaluated mast cells in dermal sulfur TUBERCULOUS LESIONS
mustard lesions produced both in vivo in rab- We were not always able to distinguish acti-
bits (Fig. 16) (172) and in vitro in organ-cultured vated fibroblasts in tissue sections from acti-
full-thickness human skin explants (173). The vated infiltrating macrophages and have not
thin GMA tissue sections enabled us to assess the studied them in tuberculous lesions, except in
amount of mast cell degranulation produced relation to fibrosis and healing (see chapter 4).
However, in tissue sections of rabbit dermal sul-
fur mustard lesions, the number of activated
fibroblasts (Fig. 17) was increased and the num-
ber of activated infiltrating macrophages was
decreased as the lesions healed (179).

LYMPHATICS IN TUBERCULOUS
LESIONS
Lymphatics have rarely been evaluated in tissue
sections, mainly because they are not readily
visible in standard paraffin-embedded sections.

FIGURE 17 Activated fibroblasts (⫹⫹⫹ to ⫹⫹


⫹⫹) between collagen fibers in the corium of a heal-
ing (6-day) rabbit dermal inflammatory lesion (pro-
duced by the topical application of 1% sulfur mus-
tard). These fibroblasts were stained histochemically
for the lysosomal enzyme acid phosphatase, which pro-
duces the bright red color that appears dark in this
black-and-white photograph. The high activation of
these fibroblasts is indicated by their large size and the
large amount of acid phosphatase that they contain.
Normal rabbit skin has relatively few activated fibro-
blasts (179).Activated fibroblasts produce the new col- FIGURE 18 A 1-day dermal inflammatory lesion that
lagen and ground substance associated with healing. was produced in the skin of a rabbit by topical 1% sulfur
A small blood vessel containing erythrocytes can be seen mustard in methylene chloride. Note the dilated lym-
in the lower half of the photograph. phatic vessel and two adjacent small blood vessels con-
Depicted is a 6-m “cold-embedded” GMA tissue taining erythrocytes.This acute chemically induced lesion
section, stained histochemically with naphthol AS-BI was grossly edematous, so the lymphatic vessel was dilated
phosphate and fast red violet LB, and counterstained from the excess tissue fluid that it was removing. Depicted
with hematoxylin. Magnification, ⫻540. Reproduced is a “cold-embedded” (174) 1- to 2-m GMA tissue sec-
with permission from reference 179. tion, stained with Giemsa (164). Magnification, ⫻600.
6. MACROPHAGES AND OTHER CELLS IN TUBERCULOUS LESIONS 䡵 145

In thin GMA-embedded sections, lymphatics are 12. Luster,A. D. 2002.The role of chemokines in link-
readily visible. Figure 18 shows a dilated lym- ing innate and adaptive immunity. Curr. Opin.
Immunol. 14:129–135.
phatic in a 1-day rabbit dermal sulfur mustard 13. Granucci, F., I. Zanoni, S. Feau, and P. Ric-
lesion that was grossly edematous (180). ciardi-Castagnoli. 2003. Dendritic cell regulation
Tuberculous lesions are never this edema- of immune responses: a new role for interleukin 2
tous. However, lymphatics carry tubercle bacilli at the intersection of innate and adaptive immunity.
from the lesions to the draining lymph nodes, EMBO J. 22:2546–2551.
14. Brown, K. A., P. Bedford, M. Macey, D. A.
and lymphatic endothelial cells probably become McCarthy, F. Leroy, A. J.Vora, A. J. Stagg, D.
activated and produce cytokines, similar to the C. Dumonde, and S. C. Knight. 1997. Human
endothelial cells of the microvasculature. blood dendritic cells: binding to vascular endothe-
lium and expression of adhesion molecules. Clin.
Exp. Immunol. 107:601–607.
REFERENCES 15. Esser, M. T., R. D. Marchese, L. S. Kierstead,
1. Janeway, C. A., Jr., P.Travers, M.Walport, and L. G.Tussey, F.Wang, N. Chirmule, and M.W.
M. J. Shlomchik. 2001. Immunobiology: the Immune Washabaugh. 2003. Memory T cells and vaccines.
System in Health and Disease, 5th ed. Garland Pub- Vaccine 21:419–430.
lishing, New York, N.Y. 16. Dong,V. M., D. H. McDermott, and R. Abdi.
2. Dannenberg,A. M., Jr., and J. F.Tomashefski, 2003. Chemokines and diseases. Eur. J. Dermatol.
Jr. 1998. Pathogenesis of pulmonary tuberculosis, 13:224–230.
p. 2447–2471. In A. P. Fishman (ed.), Fishman’s Pul- 17. Woltman, A. M., and C. van Kooten. 2003.
monary Diseases and Disorders, 3rd ed., vol. 2. Functional modulation of dendritic cells to suppress
McGraw-Hill Co., Inc, New York, N.Y. adaptive immune responses. J. Leukoc. Biol. 73:428–
3. Banchereau, J., and R. M. Steinman. 1998. 441.
Dendritic cells and the control of immunity. Nature 18. Shi,Y., J. E. Evans, and K. L. Rock. 2003. Mo-
392:245–252. lecular identification of a danger signal that alerts
4. Cella, M., F. Sallusto, and A. Lanzavecchia. the immune system to dying cells. Nature 425:516–
1997. Origin, maturation and antigen presenting 521.
function of dendritic cells. Curr. Opin. Immunol. 19. Matzinger, P. 2002.The danger model: a renewed
9:10–16. sense of self. Science 296:301–305.
5. Reid, S. D., G. Penna, and L. Adorini. 2000. 20. Galluci, S., and P. Matzinger. 2001. Danger sig-
The control of T cell responses by dendritic cell nals: SOS to the immune system. Curr. Opin.
subsets. Curr. Opin. Immunol. 12:114–121. Immunol. 13:114–119.
6. Schuler, G., B.Thurner, and N. Romani. 1997. 21. Reis e Sousa, C. 2004. Activation of dendritic
Dendritic cells: from ignored cells to major players cells: translating innate into adaptive immunity.
in T-cell-mediated immunity. Int. Arch. Allergy Curr. Opin. Immunol. 16:21–25.
Immunol. 112:317–322. 22. Chen, L. 2004. Co-inhibitory molecules of the B7-
7. Maldonado-Lopez, R., and M. Moser. 2001. CD28 family in the control of T-cell immunity. Nat.
Dendritic cell subsets and the regulation of Rev. Immunol. 4:336–347.
Th1/Th2 responses. Semin. Immunol. 13:275–282. 23. Min,W.-P., D. Zhou,T. E. Ichim, G. H. Stre-
8. Moser, M., and K. M. Murphy. 2000. Dendritic jan, X. Xia, J. Yang, X. Huang, B. Garcia,
cell regulation of Th1-Th2 development. Nat. D. White, P. Dutartre, A. M. Jevnikar, and
Immunol. 1:199–205. R. Zhong. 2003. Inhibitory feedback loop
9. Arina, A., I.Tirapu, C. Alfaro, M. Rodriguez- between tolerogenic dendritic cells and regulatory
Calvillo, G. Mazzolini, S. Inoges, A. Lopez, T cells in transplant tolerance. J. Immunol.
E. Feijoo, M. Bendandi, and I. Melero. 170:1304–1312.
2002. Clinical implications of antigen transfer 24. Steinman, R. M., D. Hawiger, and M. C.
mechanisms from malignant to dendritic cells: Nussenzweig. 2003. Tolerogenic dendritic cells.
exploiting cross-priming. Exp. Hematol. 30:1355– Annu. Rev. Immunol. 21:685–711.
1364. 25. Morel, P. A., M. Feili-Hariri, P.T. Coates, and
10. Bajénoff, M., S. Granjeaud, and S. Guerder. A.W.Thomson. 2003. Dendritic cells,T cell tol-
2003.The strategy of T cell antigen-presenting cell erance and therapy of adverse immune reactions.
encounter in antigen-draining lymph nodes Clin. Exp. Immunol. 133:1–10.
revealed by imaging of initial T cell activation. J. Exp. 26. Steptoe, R. J., and A.W.Thomson. 1996. Den-
Med. 198:715–724. dritic cells and tolerance induction. Clin. Exp.
11. Jeurissen, A., J. L. Ceuppens, and X. Bossuyt. Immunol. 105:397–402.
2004. T lymphocyte dependence of the antibody 27. Colino, J., and C. M. Snapper. 2003. Dendritic
response to “T lymphocyte independent type 2” cells, new tools for vaccination. Microbes Infect.
antigens. Immunology 111:1–7. 5:311–319.
146 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

28. Asselin-Paturel, C., and G. Trinchieri. 2005. 45. Sköld, M., and S. M. Behar. 2003.The role of
Production of type I interferons: plasmacytoid CD1d-restricted NKT cells in microbial immunity.
dendritic cells and beyond. J. Exp. Med. 202:461– Infect. Immun. 71:5447–5455.
465. 46. Dascher, C. C., K. Hiromatsu, X. Xiong, C.
29. Colonna, M., G.Trinchieri, and Y.-J. Liu. 2004. Morehouse, G.Watts, G. Liu, D. N. McMurray,
Plasmacytoid dendritic cells in immunity. Nat. K. P. LeClair, S. A. Porcelli, and M. B. Bren-
Immunol. 5:1219–1226. ner. 2003. Immunization with a mycobacterial
30. van den Elson, P. J., and A. Rudensky. 2004. lipid vaccine improves pulmonary pathology in the
Antigen presentation and recognition: recent devel- guinea pig model of tuberculosis. Int. Immunol.
opments—an editorial overview. Curr. Opin. 15:915–925.
Immunol. 16:63–66. 47. Granucci, F., S. Feau, I. Zanoni, N. Pavelka, C.
31. Gatti, E., and P. Pierre. 2003. Understanding Vizzardelli, G. Raimondi, and P. Ricciardi-
the cell biology of antigen presentation: the dendritic Castagnoli. 2003.The immune response is initi-
cell contribution. Curr. Opin. Cell Biol. 15:468–473. ated by dendritic cells via interaction with microor-
32. Kaufmann, S. H. E., and U. E. Schaible. 2005. ganisms and interleukin-2 production. J. Infect. Dis.
Antigen presentation and recognition in bacterial 187(Suppl. 2):S346–S350.
infections. Curr. Opin. Immunol. 17:79–87. 48. Germain, R. N., and M. K. Jenkins. 2004. In
33. Sugita, M., M. Cernadas, and M. B. Brenner. vivo antigen presentation. Curr. Opin. Immunol.
2004. New insights into pathways for CD1-mediated 16:120–125.
antigen presentation. Curr. Opin. Immunol. 16:90–95. 49. Majno, G., and I. Joris. 2004. Cells,Tissues, and
34. Moody, D. B., and S. A. Porcelli. 2003. Intra- Disease: Principles of General Pathology, 2nd ed. Oxford
cellular pathways of CD1 antigen presentation. University Press, New York, N.Y.
Nat. Rev. Immunol. 3:11–22. 50. Schlesinger, L. S. 1996. Role of mononuclear
35. Brigl, M., and M. B. Brenner. 2004. CD1: anti- phagocytes in M. tuberculosis pathogenesis. J. Inves-
gen presentation and T cell function. Annu. Rev. tig. Med. 44:312–323.
Immunol. 22:817–890. 51. North, R. J., and Y. J. Jung. 2004. Immunity to
36. Van Rhijn, I., D. M. Zajonc, I. A.Wilson, and tuberculosis. Annu. Rev. Immunol. 22:599–623.
D. B. Moody. 2005.T-cell activation by lipopep- 52. Means,T. K., S.Wang, E. Lien, A.Yoshimura,
tide antigens. Curr. Opin. Immunol. 17:222–229. D. T. Golenbock, and M. J. Fenton. 1999.
37. Calabi, F., J. M. Jarvis, L. Martin, and C. Mil- Human toll-like receptors mediate cellular activa-
stein. 1989. Two classes of CD1 genes. Eur. J. tion by Mycobacterium tuberculosis. J. Immunol.
Immunol. 19:285–292. 163:3920–3927.
38. Roura-Mir, C., L.Wang,T.-Y. Cheng, I. Mat- 53. Krutzik, S. R., and R. L. Modlin. 2004.The role
sunaga, C. C. Dascher, S. L. Peng, M. J. Fen- of Toll-like receptors in combating mycobacteria.
ton, C. Kirschning, and D. B. Moody. 2005. Semin. Immunol. 16:35–41.
Mycobacterium tuberculosis regulates CD1 antigen 54. Brightbill, H. D., D. H. Libraty, S. R. Krutzik,
presentation pathways through TLR-2. J. Immunol. R. B. Yang, J. T. Belisle, J. R. Bleharski, M.
175:1758–1766. Maitland, M. V. Norgard, S. E. Plevy, S. T.
39. Moody, D. B., D. C.Young,T.-Y. Cheng, J.-P. Smale, P. J. Brennan, B. R. Bloom, P. J.
Rosat, C. Roura-Mir, P. B. O’Connor, D. M. Godowski, and R. L. Modlin. 1999. Host
Zajonc, A. Walz, M. J. Miller, S. B. Levery, defense mechanisms triggered by microbial lipopro-
I. A. Wilson, C. E. Costello, and M. B. Bren- teins through toll-like receptors. Science 285:732–
ner. 2004.T cell activation by lipopeptide antigens. 736.
Science 303:527–531. 55. Shim, T. S., O. C. Turner, and I. M. Orme.
40. Flynn, J. L., and J. Chan. 2001. Immunology of 2003.Toll-like receptor 4 plays no role in suscep-
tuberculosis. Annu. Rev. Immunol. 19:93–129. tibility of mice to Mycobacterium tuberculosis infection.
41. Flynn, J. L. 2004. Immunology of tuberculosis Tuberculosis (Edinb.) 83:367–371.
and implications in vaccine development. Tubercu- 56. Reiling, N., C. Holscher, A. Fehrenbach, S.
losis 84:93–101. Kroger, C. J. Kirschning, S. Goyert, and S.
42. Jullien, D., S. Stenger, W. A. Ernst, and R. L. Ehlers. 2002. Toll-like receptor-2 (TLR2)- and
Modlin. 1997. CD1 presentation of microbial non- TLR4-mediated pathogen recognition in resistance
peptide antigens to T cells. J. Clin. Investig. 99:2071– to airborne infection with Mycobacterium tuberculo-
2074. sis. J. Immunol. 169:3480–3484.
43. Porcelli, S. A., and R. L. Modlin. 1999. The 57. Abel, B., N. Thieblemont, V. J. Quesniaux,
CD1 system: antigen-presenting molecules for T-cell N. Brown, J. Mpagi, K. Miyake, F. Bihl, and
recognition of lipids and glycolipids. Annu. Rev. B. Ryffel. 2002. Toll-like receptor 4 expression
Immunol. 17:297–329. is required to control chronic Mycobacterium tuber-
44. Hayes, S. M., and K. L. Knight. 2001. Group 1 culosis infection in mice. J. Immunol. 169:3155–
CD1 genes in rabbit. J. Immunol. 166:403–410. 3162.
6. MACROPHAGES AND OTHER CELLS IN TUBERCULOUS LESIONS 䡵 147

58. Armstrong, J. A., and P. D. Hart. 1971. Re- latent tuberculosis: variations on the Cornell murine
sponse of cultured macrophages to Mycobacter- model. Infect. Immun. 67:4531–4538.
ium tuberculosis, with observations on fusion of 71. Darwin, K. H., S. Ehrt, J. C. Gutierrez-Ramos,
lysosomes with phagosomes. J. Exp. Med. 134:713– N. Weich, and C. F. Nathan. 2003.The protea-
740. some of Mycobacterium tuberculosis is required for
59. Russell, D. G. 2001. Mycobacterium tuberculosis: resistance to nitric oxide. Science 302:1963–1966.
here today, and here tomorrow. Nat. Rev. Mol. Cell 72. Pieters, J., and H. Ploegh. 2003. Chemical war-
Biol. 2:569–577. fare and mycobacterial defense. Science 302:1900–
60. Pieters, J. 2001. Entry and survival of pathogenic 1902.
mycobacteria in macrophages. Microbes Infect. 3:249– 73. Rees, R. J.W., and P. D. Hart. 1961.Analysis of
255. the host-parasite equilibrium in chronic murine
61. Russell, D. G., H. C. Mwandumba, and E. E. tuberculosis by total and viable bacillary counts. Br.
Rhoades. 2002. Mycobacterium and the coat of J. Exp. Pathol. 42:83–88.
many lipids. J. Cell Biol. 158:421–426. 74. Muñoz-Elías, E. J., J. Timm, T. Botha, W.-T.
62. Russell, D. G., G. E. Purdy, R. M. Owens, Chan, J. E. Gomez, and J. D. McKinney. 2005.
K. H. Rohde, and R. M.Yates. 2005. Mycobac- Replication dynamics of Mycobacterium tuberculosis
terium tuberculosis and the four-minute phagosome. in chronically infected mice. Infect. Immun. 73:546–
ASM News 71:459–463. 551.
63. Walburger,A.,A. Koul, G. Ferrari, L. Nguyen, 75. Shi, L.,Y.-J. Jung, S.Tyagi, M. L. Gennaro, and
C. Prescianotto-Baschong, K. Huygen, B. R. J. North. 2003. Expression of Th1-mediated
Klebl, C.Thompson, G. Bacher, and J. Pieters. immunity in mouse lungs induces a Mycobacterium
2004. Protein kinase G from pathogenic mycobac- tuberculosis transcription pattern characteristic of
teria promotes survival within macrophages. Science nonreplicating persistence. Proc. Natl.Acad. Sci. USA
304:1800–1804. 100:241–246.
64. Ting, L. M., A. C. Kim, A. Cattamanchi, and 76. Schnappinger, D., S. Ehrt, M. I.Voskuil,Y. Liu,
J. D. Ernst. 1999. Mycobacterium tuberculosis inhibits J.A. Mangan, I. M. Monahan, G. Dolganov, B.
IFN-gamma transcriptional responses without Efron, P. D. Butcher, C. Nathan, and G. K.
inhibiting activation of STAT1. J. Immunol. Schoolnik. 2003. Transcriptional adaptation of
163:3898–3906. Mycobacterium tuberculosis within macrophages:
65. McKinney, J. D., K. Höner zu Bentrup, E. J. insights into the phagosomal environment. J. Exp.
Muñoz-Elias,A. Miczak, B. Chen,W.T. Chan, Med. 198:693–704.
D. Svenson, J. C. Sacchettini,W. R. Jacobs, Jr., 77. Voskuil, M. I., D. Schnappinger, K. C.Visconti,
and D. G. Russell. 2000. Persistence of Mycobac- M. I. Harrell, G. M. Dolganov, D. R. Sherman,
terium tuberculosis in macrophages and mice requires and G. K. Schoolnik. 2003. Inhibition of respi-
the glyoxylate shunt enzyme isocitrate lyase. Nature ration by nitric oxide induces a Mycobacterium tuber-
406:735–738. culosis dormancy program. J. Exp. Med. 198:705–713.
66. Daniel, J., C. Deb,V. S. Dubey,T. D. Sirakova, 78. Higuchi, S., M. Suga, A M. Dannenberg, Jr.,
B. Abomoelak, H. R. Morbidoni, and P. E. L. F. Affronti, I. Azuma, T. M. Daniel, and
Kolattukudy. 2004. Induction of a novel class of J. P. Petrali. 1981. Persistence of protein, carbohy-
diacylglycerol acyltransferase and triglycerol accu- drate and wax components of tubercle bacilli in der-
mulation in Mycobacterium tuberculosis as it goes into mal BCG lesions. Am. Rev. Respir. Dis. 123:397–401.
a dormancy-like state in culture. J. Bacteriol. 79. Langewoort, H. L., Z.A. Cohn, J. G. Hirsch, J.
186:5017–5030. H. Humphrey,W. G. Spector, and R. van Furth.
67. Long, R., B. Light, and J. A. Talbot. 1999. 1970. The nomenclature of mononuclear phago-
Mycobacteriocidal action of exogenous nitric oxide. cytic cells, p.1–6. In R. van Furth (ed.), Mononuclear
Antimicrob.Agents Chemother. 43:403–405. Phagocytes. F.A. Davis Co., Philadelphia, Pa.
68. MacMicking, J. D., R. J. North, R. LaCourse, 80. Dannenberg, A. M., Jr. 1968. Cellular hypersen-
J. S. Mudgett, S. K. Shah, and C. F. Nathan. sitivity and cellular immunity in the pathogenesis of
1997. Identification of nitric oxide synthase as a pro- tuberculosis: specificity, systemic and local nature, and
tective locus against tuberculosis. Proc. Natl.Acad. Sci. associated macrophage enzymes. Bacteriol. Rev.
USA 94:5243–5248. 32:85–102.
69. Scanga, C. A., V. P. Mohan, K. Tanaka, D. 81. Dannenberg, A. M., Jr., O. T. Meyer, J. R.
Alland, J. L. Flynn, and J. Chan. 2001. The Esterly, and T. Kambara. 1968.The local nature
inducible nitric oxide synthase locus confers pro- of immunity in tuberculosis, illustrated histochem-
tection against aerogenic challenge of both clinical ically in dermal BCG lesions. J. Immunol. 100:931–
and laboratory strains of Mycobacterium tuberculosis in 941.
mice. Infect. Immun. 69:7711–7717. 82. Suga, M., A. M. Dannenberg, Jr., and S.
70. Scanga, C. A.,V. P. Mohan, H. Joseph, K.Yu, Higuchi. 1980. Macrophage functional hetero-
J. Chan, and J. L. Flynn. 1999. Reactivation of geneity in vivo: macrolocal and microlocal
148 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

macrophage activation, identified by double-staining 96. Mizunoe, K., and A. M. Dannenberg, Jr. 1965.
tissue sections of BCG granulomas for pairs of Hydrolases of rabbit macrophages. III. Effect of
enzymes. Am. J. Pathol. 99:305–324. BCG vaccination, tissue culture, and ingested tuber-
83. Mosser, D. M. 2003. The many faces of cle bacilli. Proc. Soc. Exp. Biol. Med. 120:284–290.
macrophage activation. J. Leukoc. Biol. 73:209–212. 97. Carson, M. E., and A. M. Dannenberg, Jr.
84. Dannenberg, A. M., Jr., M. S. Burstone, P. C. 1965. Hydrolytic enzymes of rabbit mononuclear
Walter, and J. W. Kinsley. 1963.A histochemical exudate cells. II. Lysozyme: properties and quan-
study of phagocytic and enzymatic functions of titative assay in tuberculous and control inbred rab-
rabbit mononuclear and polymorphonuclear exu- bits. J. Immunol. 94:99–104.
date cells and alveolar macrophages. I. Survey and 98. Meyer, O. T., A. M. Dannenberg, Jr., and K.
quantitation of enzymes, and states of cellular acti- Mizunoe. 1970. Hydrolytic enzymes of rabbit
vation. J. Cell Biol. 17:465–486. mononuclear and polymorphonuclear exudate
85. Nathan, C., and M. Sporn. 1991. Cytokines in cells and pulmonary alveolar macrophages. III.
context. J. Cell Biol. 113:981–986. Deoxyribonuclease and ribonuclease: properties
86. Nathan, C. F. 1987. Secretory products of and quantitative assay in macrophages from tuber-
macrophages. J. Clin. Investig. 79:319–326. culous and control inbred rabbits. J. Reticuloen-
87. Nathan, C. 2002. Points of control in inflamma- dothel. Soc. 7:15–31.
tion. Nature 420:846–852. 99. Rojas-Espinosa, O., A. M. Dannenberg, Jr.,
88. Cohn, Z. A., and B. Benson. 1965.The in vitro P. A. Murphy, P. A. Straat, P. C. Huang, and
differentiation of mononuclear phagocytes. III.The S. P. James. 1973. Purification and properties of
reversibility of granule and hydrolytic enzyme for- the cathepsin D-type proteinase from beef and
mation and the turnover of granule constituents. J. rabbit lung and its identification in macrophages.
Exp. Med. 122:455–466. Infect. Immun. 8:1000–1008.
89. Dannenberg, A. M., Jr., P. C.Walter, and F. A. 100. Rojas-Espinosa, O., P. Arce-Paredez, A. M.
Kapral. 1963. A histochemical study of phago- Dannenberg, Jr., and R. L. Kamenetz. 1975.
cytic and enzymatic functions of rabbit mononu- Macrophage esterase: identification, purification
clear and polymorphonuclear exudate cells and and properties of a chymotrypsin-like esterase
alveolar macrophages. II. The effect of particle from lung that hydrolyzes and transfers nonpolar
ingestion on enzyme activity; two phases of in vi- amino acid esters. Biochim. Biophys.Acta 403:161–
tro activation. J. Immunol. 90:448–465. 179.
90. Ando, M., A. M. Dannenberg, Jr., M. Sugi- 101. McAdoo, M. H., A. M. Dannenberg, Jr.,
moto, and B. S. Tepper. 1977. Histochemical C. J. Hayes, S. P. James, and J. H. Sanner.
studies relating the activation of macrophages to the 1973. Inhibition of the cathepsin D-type pro-
intracellular destruction of tubercle bacilli. Am. J. teinase of macrophages by pepstatin, a specific
Pathol. 86:623–634. pepsin inhibitor, and other substances. Infect.
91. Dannenberg,A. M., Jr., and E. L. Smith. 1955. Immun. 7:655–665.
Proteolytic enzymes of lung. J. Biol. Chem. 215:45– 102. Hastie, A. T. 1981. Monospecific antibodies to
54. rabbit lung ribonucleases. J. Biol. Chem.
92. Dannenberg,A. M., Jr., and E. L. Smith. 1955. 256:12553–12560.
Action of proteinase I of bovine lung. Hydrolysis of 103. Namba, M., M. Suga, F.Tanaka, A. M. Dan-
the oxidized B chain of insulin; polymer formation nenberg, Jr.,A.T. Hastie, and R. C. Franson.
from amino acid esters. J. Biol. Chem. 215:55–66. 1983. Immunocytochemical demonstration of
93. Dannenberg, A. M., Jr., and W. E. Bennett. rabbit ribonuclease and phospholipase A2 by the
1964. Hydrolytic enzymes of rabbit mononuclear peroxidase-antiperoxidase technique in profes-
exudate cells. I. Quantitative assay and properties of sional phagocytes (pulmonary alveolar macro-
certain proteases, nonspecific esterases and lipases of phages and granulocytic and mononuclear peri-
mononuclear and polymorphonuclear cells and toneal exudate cells) and in glycol methacrylate
erythrocytes. J. Cell Biol. 21:1–13. sections of dermal tuberculous (BCG) lesions. J.
94. Dannenberg, A. M., Jr., and W. E. Bennett. Reticuloendothel. Soc. 34:425–435.
1963. Hydrolases of mononuclear exudate cells and 104. Rojas-Espinosa, O., A. M. Dannenberg, Jr.,
tuberculosis. I. Exudate characteristics, esterases, L.A. Sternberger, and T.Tsuda. 1974. Role of
proteinases and lipase. Arch. Pathol. 76:581–591. cathepsin D in the pathogenesis of tuberculosis.A
95. Yarborough, D. J., O. T. Meyer, A. M. Dan- histochemical study employing unlabeled anti-
nenberg, Jr., and B. Pearson. 1967. Histo- bodies and the peroxidase-antiperoxidase com-
chemistry of macrophage hydrolases. III. Studies on plex. Am. J. Pathol. 74:1–17.
-galactosidase, -glucuronidase and aminopep- 105. Tsuda, T., A. M. Dannenberg, Jr., M. Ando,
tidase with indolyl and naphthyl substrates. J. Retic- O. Rojas-Espinosa, and K. Shima. 1974.
uloendothel. Soc. 4:390–408. Enzymes in tuberculous lesions hydrolyzing pro-
6. MACROPHAGES AND OTHER CELLS IN TUBERCULOUS LESIONS 䡵 149

tein, hyaluronic acid and chondroitin sulfate: a 118. Trinchieri, G. 1997. Cytokines acting on or
study of isolated macrophages in developing and secreted by macrophages during intracellular infec-
healing rabbit BCG lesions with substrate film tion (IL-10, IL-12, IFN-). Curr. Opin. Immunol.
techniques; the shift of enzyme pH optima towards 9:17–23.
neutrality in “intact” cells and tissues. J. Reticu- 119. Shigenaga, T., A. M. Dannenberg, Jr., D. B.
loendothel. Soc. 16:220–231. Lowrie,W. Said, M. J. Urist, H. Abbey, B. H.
106. Smokovitis, A., M. Sugimoto, A. M. Dan- Schofield, P. Mounts, and K. Sugisaki. 2001.
nenberg, Jr., and T. Astrup. 1976. A histo- Immune responses in tuberculosis: antibodies and
chemical study of the fibrinolytic activity in der- CD4/CD8 lymphocytes with vascular adhesion
mal tuberculous lesions produced by BCG in molecules and cytokines (chemokines) cause a
rabbits. Exp. Mol. Pathol. 25:236–241. rapid antigen-specific cell infiltration at sites of
107. Rivera-Marrero, C. A., J. Stewart, W. M. bacillus Calmette-Guérin reinfection. Immunol-
Shafer, and J. Roman. 2004. Down-regulation ogy 102:466–479.
of cathepsin G in THP-1 monocytes after infec- 120. Lewinsohn, D. A., A. S. Heinzel, J. M. Gard-
tion with Mycobacterium tuberculosis is associated ner, L. Zhu, M. R. Alderson, and D. M.
with increased intracellular survival of bacilli. Lewinsohn. 2003. Mycobacterium tuberculosis-
Infect. Immun. 72:5712–5721. specific CD8⫹ T cells preferentially recognize
108. Sugimoto, M., A. M. Dannenberg, Jr., L. M. heavily infected cells. Am. J. Respir. Crit. Care Med.
Wahl, W. H. Ettinger, Jr., A. T. Hastie, D. C. 168:1346–1352.
Daniels, C. R. Thomas, and L. Demoulin- 121. Harty, J. T., A. R. Tvinnereim, and D. W.
Brahy. 1978. Extracellular hydrolytic enzymes of White. 2000. CD8⫹ T cell effector mechanisms
rabbit dermal tuberculous lesions and tuberculin in resistance to infection. Annu. Rev. Immunol.
reactions collected in skin chambers. Am. J. Pathol. 18:275–308.
90:583–608. 122. Janeway, C. A., Jr. 2002.A trip through my life
109. Sheldon, W. H., D. Mildvan, and J. C. Allen. with an immunological theme. Annu. Rev.
1967. Some serum protein and cellular constituents Immunol. 20:1–28.
of inflammatory lesions. Collection of exudates in 123. Wakkach, A., N. Fournier, V. Brun, J.-P.
a chamber adhered over skin wounds of rabbits. Breittmayer, F. Cottrez, and H. Groux. 2003.
Johns Hopkins Med. J. 121:113–133. Characterization of dendritic cells that induce
110. Edlow, D. W., and W. H. Sheldon. 1971.The tolerance and T-regulatory-1 cell differentiation in
pH of inflammatory exudates. Proc. Soc. Exp. Biol. vivo. Immunity 18:605–617.
Med. 137:1328–1332. 124. von Boehmer, H. 2003. Dynamics of suppres-
111. Lurie, M. B. 1932.The correlation between the sor T cells: in vivo veritas. J. Exp. Med. 198:845–
histological changes and the fate of living tuber- 849.
cle bacilli in the organs of tuberculous rabbits. J. 125. Levy, B. D., C. B. Clish, B. Schmidt, K.
Exp. Med. 55:31–54. Gronert, and C. N. Serhan. 2001. Lipid medi-
112. Lurie, M. B. 1964. Resistance to Tuberculosis: Exper- ator class switching during acute inflammation:
imental Studies in Native and Acquired Defensive signals in resolution. Nat. Immunol. 2:612–619.
Mechanisms. Harvard University Press, Cambridge, 126. Walker, L. S. K. 2003. CD4⫹ CD25⫹ Treg:
Mass. divide and rule? Immunology 111:129–137.
113. Dannenberg, A. M., Jr. 2003. Macrophage 127. Akbar,A. N., L. S.Taams, M. Salmon, and M.
turnover, division and activation within develop- Vukmanovic-Stejic. 2003.The peripheral gen-
ing, peak and “healed” tuberculous lesions pro- eration of CD4⫹ CD25⫹ regulatory T cells.
duced in rabbits by BCG. Tuberculosis 83:251– Immunology 109:319–325.
260. 128. Rubin, B.,Y. D. de Durana, N. Li, and E. E.
114. Mustafa, T., S. Phyu, R. Nilsen, R. Jonsson, Sercarz. 2003. Regulator T cells: specific for anti-
and G. Bjune. 2000. In situ expression of cyto- gen and/or antigen receptors? Scand. J. Immunol.
kines and cellular phenotypes in the lungs of mice 57:399–409.
with slowly progressive primary tuberculosis. 129. Baecher-Allan, C., V. Viglietta, and D. A.
Scand. J. Immunol. 51:548–556. Hafler. 2004. Human CD4⫹CD25⫹ regulatory
115. Orme, I. M. 1993. Immunity to mycobacteria. T cells. Semin. Immunol. 16:89–97.
Curr. Opin. Immunol. 5:497–502. 130. Horwitz, D. A., S. G. Zheng, and J. D. Gray.
116. Khaled, A. R., and S. K. Durum. 2002. Lym- 2003. The role of the combination of IL-2 and
phocide: cytokines and the control of lymphoid TGF- or IL-10 in the generation and function
homeostasis. Nat. Rev. Immunol. 2:817–830. of CD4⫹CD25⫹ and CD8⫹ regulatory T cell
117. Marleau,A. M., and N. Sarvetnick. 2005.T cell subsets. J. Leukoc. Biol. 74:471–478.
homeostasis in tolerance and immunity. J. Leukoc. 131. Piccirillo, A. C., and E. M. Shevach. 2004.
Biol. 78:575–584. Naturally-occurring CD4⫹CD25⫹ immunoreg-
150 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

ulatory T cells: central players in the arena of 144. Boismenu, R., and W. L. Havran. 1997. An
peripheral tolerance. Semin. Immunol. 16:81–88. innate view of  T cells. Curr. Opin. Immunol.
132. Shevach, E. M. 2004. Regulatory/suppressor T 9:57–63.
cells in health and disease. Arthritis Rheum. 145. Kaufmann, S. H. E. 2003. Immunity to intra-
50:2721–2724. cellular bacteria, p. 1229–1261. In W. E. Paul (ed.),
133. Belkaid,Y., and B.T. Rouse. 2005. Natural reg- Fundamental Immunology. Lippincott Williams &
ulatory T cells in infectious disease. Nat. Immunol. Wilkins, Philadelphia, Pa.
6:353–360. 146. Shen,Y., D. Zhou, L. Qiu, X. Lai, M. Simon,
134. Hayday, A. C. 2000.  cells: a right time and a L. Shen, Z. Kou, Q.Wang, L. Jiang, J. Estep,
right place for a conserved third way of protection. R. Hunt, M. Clagett, P. K. Sehgal, Y. Li, X.
Annu. Rev. Immunol. 18:975–1026. Zeng, C. T. Morita, M. B. Brenner, N. L.
135. Ladel, C. H., C. Blum, A. Dreher, K. Reifen- Letvin, and Z.W. Chen. 2002.Adaptive immune
berg, and S. H. Kaufmann. 1995. Protective response of Vgamma2Vdelta2⫹ T cells during
role of gamma/delta T cells and alpha/beta T cells mycobacterial infections. Science 295:2255–2258.
in tuberculosis. Eur. J. Immunol. 25:2877–2881. 147. Hahn, Y.-S., C. Taube, N. Jin, K. Takeda,
136. D’Souza, C. D., A. M. Cooper, A. A. Frank, J.-W. Park, J. M. Wands, M. K. Aydintug,
R. J. Mazzaccaro, B. R. Bloom, and I. M. C. L. Roark, M. Lahn, R. L. O’Brien, E. W.
Orme. 1997. An anti-inflammatory role for Gelfand, and W. K. Born. 2003.V4⫹  T
gamma delta T lymphocytes in acquired immunity cells regulate airway hyperreactivity to metha-
to Mycobacterium tuberculosis. J. Immunol. 158:1217– choline in ovalbumin-sensitized and challenged
1221. mice. J. Immunol. 171:3170–3178.
137. Balbi, B., M. T. Valle, S. Oddera, D. Giunti, 148. Robbins, S. H., and L. Brossay. 2002. NK cell
F. Manca, G. A. Rossi, and L. Allegra. 1993. receptors: emerging roles in host defense against
T-lymphocytes with gamma delta⫹ V delta 2⫹ infectious agents. Microbes Infect. 4:1523–1530.
antigen receptors are present in increased pro- 149. Raulet, D. H. 2003.Natural killer cells,p.365–391.
portions in a fraction of patients with tuberculo- In W. E. Paul (ed.), Fundamental Immunology. Lip-
sis or with sarcoidosis. Am. Rev. Respir. Dis. pincott Williams & Wilkins, Philadelphia, Pa.
148:1685–1690. 150. Atochina, O., and D. Harn. 2005. LNF-
138. Chen, Z. W., and N. L. Letvin. 2003. PIII/LeX-stimulated macrophages activate nat-
Vgamma2Vdelta2⫹ T cells and anti-microbial ural killer cells via CD40-CD40L interaction.
immune responses. Microbes Infect. 5:491–498. Clin. Diagn. Lab. Immunol. 12:1041–1049.
139. Ito, M., N. Kojiro, T. Ikeda, T. Ito, J. Funada, 151. Kronenberg, M., and L. Gapin. 2002. The
and T. Kokubu. 1992. Increased proportions of unconventional lifestyle of NKT cells. Nat. Rev.
peripheral blood gamma-delta T cells in patients Immunol. 2:557–568.
with pulmonary tuberculosis. Chest 102:195–197. 152. Yoneda,T., and J. J. Ellner. 1998. CD4(⫹) T cell
140. Ueta, C., I. Tsuyuguchi, H. Kawasumi, T. and natural killer cell-dependent killing of Myco-
Takashima, H.Toba, and S. Kishimoto. 1994. bacterium tuberculosis by human monocytes. Am. J.
Increase of gamma/delta T cells in hospital work- Respir. Crit. Care Med. 158:395–403.
ers who are in close contact with tuberculosis 153. Bermudez, L. E., M. Wu, and L. S. Young.
patients. Infect. Immun. 62:5434–5441. 1995. Interleukin-12-stimulated natural killer cells
141. Li, B., M. D. Rossman, T. Imir, A. F. Oner- can activate human macrophages to inhibit growth
Eyuboglu, C. W. Lee, R. Biancaniello, and of Mycobacterium avium. Infect. Immun. 63:4099–
S. R. Carding. 1996. Disease-specific changes in 4104.
gamma-delta T cell repertoire and function in 154. Lieberman, L. A., and C. A. Hunter. 2002.
patients with pulmonary tuberculosis. J. Immunol. Regulatory pathways involved in the infection-
157:4222–4229. induced production of IFN- by NK cells.
142. Carvalho, A. C., A. Matteelli, P. Airo, S. Microbes Infect. 4:1531–1538.
Tedoldi, C. Casalini, L. Imberti, G. P. Cadeo, 155. Junqueira-Kipnis, A. P., A. Kipnis, A.
A. Beltrame, and G. Carosi. 2002. Gamma- Jamieson, M. Gonzales Juarrero, A. Diefen-
delta T lymphocytes in the peripheral blood of bach, D. H. Raulet, J. Turner, and I. M.
patients with tuberculosis with and without HIV Orme. 2003. NK cells respond to pulmonary
co-infection. Thorax 57:357–360. infection with Mycobacterium tuberculosis, but play
143. Gioia, C., C. Agrati, R. Casetti, C. Cairo, G. a minimal role in protection. J. Immunol. 171:6039–
Borsellino, L. Battistini, G. Mancino, D. 6045.
Goletti,V. Colizzi, L. P. Pucillo, and F. Poc- 156. Orange, J. S. 2002. Human natural killer cell defi-
cia. 2002. Lack of CD27-CD45RA-V gamma 9V ciencies and susceptibility to infection. Microbes
delta 2⫹ T cell effectors in immunocompromised Infect. 4:1545–1558.
hosts and during active pulmonary tuberculosis. J. 157. Andrews, D. M., A. A. Scalzo, W. M.
Immunol. 168:1484–1489. Yokoyama, M. J. Smyth, and M. A. Degli-
6. MACROPHAGES AND OTHER CELLS IN TUBERCULOUS LESIONS 䡵 151

Esposti. 2003. Functional interactions between (Type 1) and schistosomal (Type 2) antigen-elicited
dendritic cells and NK cells during vital infection. granuloma formation. J. Immunol. 161: 4276–
Nat. Immunol. 4:175–181. 4282.
158. Moretta, A. 2002. Natural killer cells and den- 171. Vogt, R. F., Jr., A. M. Dannenberg, Jr., B. H.
dritic cells: rendezvous in abused tissues. Nat. Rev. Schofield, N. A. Hynes, and B. Papirmeister.
Immunol. 2:957–964. 1984. Pathogenesis of skin lesions caused by sulfur
159. Barry, M., and R. C. Bleackley. 2002. Cyto- mustard. Fundam.Appl.Toxicol. 4:S71–S83.
toxic T lymphocytes: all roads lead to death. Nat. 172. Dannenberg, A. M., Jr., P. J. Pula, L. Liu, S.
Rev. Immunol. 2:401–409. Harada, F. Tanaka, R. F. Vogt, Jr., A. Kajiki,
160. Rock, K. L. 2003. The ins and outs of cross- and K. Higuchi. 1985. Inflammatory mediators
presentation. Nat. Immunol. 4:941–943. and modulators released in organ culture from
161. van Parijs, L., and A. K. Abbas. 1996. Role of rabbit skin lesions produced in vivo by sulfur
Fas-mediated cell death in the regulation of mustard. I. Quantitative histopathology; PMN,
immune responses. Curr. Opin. Immunol. 8:355– basophil and mononuclear cell survival; and
361. unbound (serum) protein content. Am. J. Pathol.
162. Mangan, D. F., and S. M. Wahl. 1991. Differ- 121:15–27.
ential regulation of human monocyte programmed 173. Rikimaru, T., M. Nakamura, T. Yano, G.
cell death (apoptosis) by chemotactic factors and Beck, G. S. Habicht, L. L. Rennie, M.Widra,
pro-inflammatory cytokines. J. Immunol. 147:3408– C. A. Hirshman, M. G. Boulay, E. W.
3412. Spannhake, G. S. Lazarus, P. J. Pula, and
163. Sugisaki, K., A. M. Dannenberg, Jr.,Y. Abe, A. M. Dannenberg, Jr. 1991. Mediators, initi-
J. Tsuruta, W.-J. Su, W. Said, L. Feng, T. ating the inflammatory response, released in organ
Yoshimura, P. J. Converse, and P. Mounts. culture by full-thickness human-skin explants
1998. Nonspecific and immune-specific up- exposed to the irritant, sulfur mustard. J. Investig.
regulation of cytokines in rabbit dermal tubercu- Dermatol. 96:888–897.
lous (BCG) lesions. J. Leukoc. Biol. 63:440–450. 174. Namba, M., A. M. Dannenberg, Jr., and
164. Vogt, R. F., Jr., N. A. Hynes, A. M. Dannen- F. Tanaka. 1983. Improvement of the histo-
berg, Jr., S. Castracane, and L. Weiss. 1983. chemical demonstration of acid phosphatase,
Improved techniques using Giemsa-stained glycol -galactosidase and nonspecific esterase in gly-
methacrylate tissue sections to quantitate basophils col methacrylate tissue sections by cold temper-
and other leukocytes in inflammatory skin lesions. ature embedding. Stain Technol. 58:207–213.
Stain Technol. 58:193–205. 175. Higuchi, S., M. Suga,A. M. Dannenberg, Jr.,
165. Bennouna, S., S. K. Bliss, T. J. Curiel, and and B. H. Schofield. 1979. Histochemical
E.Y. Denkers. 2003. Cross-talk in the innate sys- demonstration of enzyme activities in plastic- and
tem: neutrophils instruct recruitment and activa- paraffin-embedded tissue sections. Stain Technol.
tion of dendritic cells during microbial infection. 54:5–12.
J. Immunol. 171:6052–6058. 176. Galli, S. J., and S. Nakae. 2003. Mast cells to the
166. Brandt, E., F. Petersen, A. Ludwig, J. E. defense. Nat. Immunol. 4:1160–1162.
Ehlert, L. Bock, and H.-D. Flad. 2000.The - 177. McLachlan, J. B., J. P. Hart, S.V. Pizzo, C. P.
thromboglobulins and platelet factor 4: blood Shelburne, H. F. Staats, M. D. Gunn, and
platelet-derived CXC chemokines with diver- S. N. Abraham. 2003. Mast cell-derived tumor
gent roles in early neutrophil regulation. J. Leukoc. necrosis factor induces hypertrophy of draining
Biol. 67:471–478. lymph nodes during infection. Nat. Immunol.
167. Simon, H.-U. 2003. Neutrophil apoptosis path- 4:1199–1205.
ways and their modifications in inflammation. 178. Dvorak, A. M. 2005. Ultrastructural studies of
Immunol. Rev. 193:101–110. human basophils and mast cells. J. Histochem.
168. Lasco, T. M., O. C. Turner, L. Cassone, I. Cytochem. 53:1043–1070.
Sugawara, H.Yamada, D. N. McMurray, and 179. Kajiki, A., K. Higuchi, M. Nakamura, L. H.
I. M. Orme. 2004. Rapid accumulation of Liu, P. J. Pula, and A. M. Dannenberg, Jr.
eosinophils in lung lesions in guinea pigs infected 1988. Sources of extracellular lysosomal enzymes
with Mycobacterium tuberculosis. Infect. Immun. released in organ culture by developing and heal-
72:1147–1149. ing inflammatory lesions. J. Leukoc. Biol. 43: 104–
169. Munitz, A., and F. Levi-Schaffer. 2004. 116.
Eosinophils: ‘new’ roles for ‘old’ cells. Allergy 180. Harada, S.,A. M. Dannenberg, Jr.,A. Kajiki,
59:268–275. K. Higuchi, F. Tanaka, and P. J. Pula. 1985.
170. Ruth, J. H., N.W. Lukacs, K. S.Warmington, Inflammatory mediators and modulators released
T. J. Polak, M. Burdick, S. L. Kunkel, R. M. in organ culture from rabbit skin lesions produced
Strieter, and S. W. Chensue. 1998. Expression in vivo by sulfur mustard. II. Evans blue dye exper-
and participation of eotaxin during microbacterial iments that determined the rates of entry and
152 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

turnover of serum protein in developing and heal- 182. Gleich, G. J., C. R. Adolphson, and K. M.
ing lesions. Am. J. Pathol. 121:28–38. Leiferman. 1992. Eosinophils, p. 663–700. In
181. Moqbel, R., and P. Lacy. 1998. Eosinophils, p. J. I. Gallin, I. M. Goldstein, and R. Snyderman
139–165. In J.A. Denburg (ed.), Allergy and Aller- (ed.), Inflammation: Basic Principles and Clinical
gic Diseases: the New Mechanisms and Therapeutics. Correlates, 2nd ed. Raven Press, Ltd., New
Humana Press,Totowa, N.J. York, N.Y.
Section 3.

TUBERCULOUS LESIONS
This page intentionally left blank
7
STRUCTURAL COMPONENTS
OF TUBERCULOUS LESIONS

Overview 155
Tuberculous granulation tissue 155
Caseous tissues 155
Liquefied caseum and cavities 157
Healing tuberculous lesions 158
Persisting viable tubercle bacilli 158
Organ and species resistance 158
Resistance to bovine-type and human-type tubercle bacilli 159

Abstract. This chapter describes the structural components of tuberculous lesions: the sur-
rounding granulation tissue, solid caseous necrosis, liquefied caseum, cavities, and the
fibrosis and calcification of healing lesions.

OVERVIEW reticular fibers, and, in time, fibroblasts and the


In humans, rabbits, and guinea pigs, typical collagen and ground substance which they pro-
tubercles often have a caseous necrotic center. duce (see chapters 4 and 6). See chapters 8 and
Surrounding this caseous center is tuberculous 21 for details on the role of the vasculature in
granulation tissue, which is rich in macrophages these lesions.
and lymphocytes and contains dendritic cells, In humans, tuberculous granulation tissue
plasma cells, polymorphonuclear leukocytes, often contains ill-defined granulomas and areas
eosinophils, fibroblasts, and many capillaries (see of caseous necrosis, along with variable numbers
chapter 6).The caseum is initially solid, but in of tubercle bacilli.The cells in tuberculous gran-
time may liquefy. In solid caseum, many tuber- ulomas produce a variety of cytokines, e.g.,
cle bacilli are dead, but some are only dormant. interleukin 1 (IL-1), IL-2, and IL-12, monocyte
In liquefied caseum, the bacilli may multiply chemoattractant protein-1, tumor necrosis fac-
extracellularly, sometimes extensively. If they tor, gamma interferon, and transforming growth
multiply, their tuberculin-like products can cause factor  (see chapters 19 and 20) (2–4).
necrosis of a bronchial wall so that a cavity
forms.The tubercle bacilli then spread via the CASEOUS TISSUES
bronchial tree to other parts of the lung and to In rabbits, guinea pigs, and humans, the necrotic
the environment. Healing with fibrosis can occur tissue in tuberculous lesions is described as
if the number of bacilli remains small. caseous, because it resembles cheese, being rather
homogeneous, yellow-white, and rich in lipids
TUBERCULOUS GRANULATION and proteins derived from tubercle bacilli and
TISSUE dead cells. It is caused by tissue-damaging
The active part of all lesions in tuberculosis con- delayed-type hypersensitivity (DTH). (See chap-
sists of tuberculous granulation tissue (1). It con- ters 3 and 4 for photographs of caseous tissue.)
tains young macrophages, activated macrophages No connective tissue fibers are usually found
(now called epithelioid cells), natural killer cells, within the caseum, because they were hydrolyzed
antigen-presenting cells (dendritic cells), lym- by the collagenases and elastases of macrophages
phocytes, plasma cells, granulocytes (usually near and granulocytes in the tubercle before caseation
areas of caseous necrosis), capillaries, lymphatics, took place. In time, caseous foci in humans may

155
156 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

calcify or even ossify.Tubercle bacilli may survive Intact bacilli seem rather nontoxic to
in solid caseous material, but they are usually macrophages (9). DTH is definitely involved,
inhibited there by the reduced oxygen tension because caseous necrosis occurs in tuberculous
and relatively low pH (5), as well as by the local lesions at about the same time that the tuber-
accumulation of toxic fatty acids (6, 7). culin skin test becomes positive. Table 1 lists
In rabbits, guinea pigs, and humans, caseous some of the possibilities. (i) Natural killer cells
necrosis first occurs at the end of the logarith- and cytotoxic T cells seem to kill bacilli-laden
mic (symbiotic) stage of tuberculosis when macrophages and may also injure nearby tis-
tuberculin sensitivity develops (see chapter 2). sues. (ii) Clotting factors from macrophages, as
The nonactivated macrophages in which the well as from necrotic cells and tissues, may acti-
bacilli reside are killed by the tissue-damaging vate the clotting system, impairing the local
DTH reaction, so the intracellular growth of blood supply and causing additional tissue injury
tubercle bacilli is stopped. Whether or not a (8) (see chapter 8). (iii) Reactive oxygen and
true caseous center develops and enlarges nitrogen intermediates (e.g., superoxide and
depends on the sensitivity of the host and the nitric oxide produced by activated macrophages)
amount of tuberculin products in the sur- may kill cells and tissues as well as bacilli. (iv)
rounding tuberculous granulation tissue. Con- Certain cytokines produced by macrophages
siderable caseation is produced when the host and T lymphocytes (such as tumor necrosis fac-
develops high degrees of tuberculin sensitivity tor) are probably toxic at higher than normal
and/or large amounts of tuberculin-like prod- concentrations. (v) Hydrolytic enzymes, e.g.,
ucts are released from numerous bacilli. proteinases, nucleases, and lipases, released from
When macrophages surrounding the caseous live and disintegrating macrophages and gran-
center are poorly activated, the bacillus again ulocytes may injure tissues directly. (vi) Antigen-
grows intracellularly, and again the DTH reaction antibody reactions, or even aggregated proteins
kills such cells. The lesion progresses, and the in the necrotic tissues, may locally activate the
caseous center enlarges. However, when most of complement system, which may injure cells.
the macrophages surrounding the caseous center
are strongly activated, they will inhibit or destroy
the bacilli that they ingest, and the lesion will TABLE 1 Causes of tissue damage and caseous
regress or at least stabilize. All early lesions in necrosisa
humans apparently develop a caseous center (see Natural killer cells and cytotoxic T cells
chapter 3), but only some of the lesions develop Involving perforin, apoptosis and other mechanisms
such a center in rabbits infected by aerosol with Anoxia
virulent (human-type) tubercle bacilli. Produced by thrombosis (macrophages produce
Macrophages cannot penetrate very far into clotting factors)
the caseous center.The center is avascular and Cell products that may be toxic at high local
concentrations
the adjacent blood vessels are thrombosed (8). Reactive oxygen and nitrogen intermediates; certain
In addition to the relatively low oxygen tension cytokines, such as tumor necrosis factor; hydrolytic
and pH mentioned above, the caseous material enzymes; and complement
probably contains toxic metabolic products and Bacillary products that may be toxic at high
relatively high concentrations of tuberculin-like local concentrations
products. Thus, the host’s defense cells cannot Intact tubercle bacilli are nontoxic, but when they are
broken down, toxic products, such as “cord factor”
eliminate tubercle bacilli within caseous foci, (trehalose dimycolate), may be released.
but, fortunately, such bacilli cannot multiply Overview
appreciably in solid caseum because of these Caseous necrosis is initiated by a tissue-damaging
same adverse conditions. DTH reaction to high local concentrations of
What causes the death of macrophages that tuberculin-like bacillary products.Th1-type
have ingested tubercle bacilli and the death of lymphocytes impart specificity to this reaction.
a
nearby tissues is not precisely known (Table 1). Reprinted with permission from reference 1.
7. STRUCTURAL COMPONENTS OF TUBERCULOUS LESIONS 䡵 157

(vii) After tubercle bacilli are fragmented, toxic TABLE 2 Causes and results of liquefactiona
components, such as “cord factor” (trehalose
Causes
dimycolate), may be released. The most likely DTH to tuberculin-like bacillary products
cause of the tissue damage, however, is a com- Hydrolytic enzymes, especially proteases, DNases and
bination of many of these possibilities (10). RNases, and probably lipases
Regardless of the exact mechanisms involved, Results
it is generally agreed that almost all of the cell Extracellular bacillary multiplication (that is
death and tissue damage found in tuberculosis sometimes tremendous), which may allow
are due to DTH, either directly or indirectly, and antimicrobial drug-resistant mutants to develop
that thrombosis of the vasculature is a major Caseous necrosis of bronchial walls and cavity
formation, caused by the high concentrations of
cause of resulting necrosis. tuberculin-like products
Spread of bacilli through the air passages to other
LIQUEFIED CASEUM AND CAVITIES parts of the lungs and to the environment, where
Liquefaction occurs when solid caseous mate- they may infect other people
rial softens (see chapter 4) (6, 9, 11–14, 14a). In a
Adapted from reference 1.
contrast to pyogenic abscesses, which liquefy
soon after they form, the caseous foci of tuber-
culosis may not liquefy for months, if ever. For probably lipases (17, 18) released from these
photographs of liquefied caseum and cavities see macrophages liquefy the solid caseous material
Fig. 1 and chapters 3 and 4. (see chapter 4). The breakdown products of
The exact cause of liquefaction is not known, caseous material are osmotically active, so that
but the following pathophysiological sequence is fluid is absorbed from the surrounding tissues.
consistent with present knowledge (Table 2). This liquefied material, rich in nutrients, is often
The antigens of the tubercle bacillus, especially an excellent culture medium for tubercle bacilli
tuberculin-like antigens (after the host becomes (5, 9, 13, 14), which now may grow profusely and
tuberculin positive) (15, 16), cause an accumu- extracellularly for the first time during the course
lation of macrophages at the site (see chapter 10). of this disease (Fig. 1) (1, 9).The wall of a nearby
Then, the proteinases and nucleases (14a) and bronchus is eroded by the tissue-damaging DTH

FIGURE 1 The wall of a cavity from a


genetically resistant rabbit, 8 weeks after
the inhalation of human-type tubercle
bacilli.The liquefied caseous tissue (right)
and liquefying caseous tissue (left) are
both swarming with (rod-shaped) acid-fast
bacilli. Cavities can be produced by both
virulent human-type tubercle bacilli (illus-
trated here) and virulent bovine-type
tubercle bacilli (illustrated in chapter 4);
the bacilli can grow profusely in cavities
produced by either type. Magnification,
⫻620. Reproduced with permission from
reference 25.
158 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

reaction to the large amount of tuberculin-like ical tuberculosis may have progressing and heal-
products present. The bronchus ruptures, and ing lesions within the same lung (see chapter 3).
the liquefied caseum, often containing numer-
ous bacilli, is discharged into the air passages, PERSISTING VIABLE
thereby forming a cavity.Then air enters, and the TUBERCLE BACILLI
high oxygen content provides an additional stim- In humans, after even a clinically inapparent
ulus for the extracellular bacillary growth. tuberculous infection heals, the lungs may con-
Current knowledge on the cause of lique- tain small encapsulated, solid, or semisolid caseous
faction is presented in chapter 4. Detailed char- foci. In such foci, tubercle bacilli may persist in
acteristics of human and rabbit tuberculous a dormant (almost nonmetabolizing) state (see
cavities are presented in chapters 3 and 4, chapters 1 and 6) and are therefore resistant to
respectively. killing by antimicrobial agents. (Many bacteria,
including the tubercle bacillus, have dormancy
HEALING TUBERCULOUS LESIONS genes that enable such persistence [23].)
In humans, caseous centers occur more readily This bacillary persistence is the reason why
than in rabbits because of the high human tuber- any infection with virulent tubercle bacilli is
culin sensitivity.Arrested calcified caseous lesions potentially dangerous. The bacilli may remain
as small as 0.5 mm can still be found years after viable for decades and may cause active disease
they were formed (see chapter 3). when the host resistance is lowered by advanced
Larger foci may reactivate many years later if age, corticosteroids, immunosuppressants, HIV,
the resistance of the person is lowered, or if liq- or other factors.
uefaction occurs (see chapters 1, 3, and 4). Persistence of viable tubercle bacilli is prob-
Whether calcified foci less than 1 mm in diam- ably the reason why a positive tuberculin reac-
eter can reactivate remains to be determined. tion is often maintained for life. Each time the
In rabbits infected with virulent Mycobacterium bacillus multiplies in a primary or secondary
tuberculosis, enlargement of lesions with or with- focus, the immune system is stimulated.
out solid caseous centers is prevented by the local Although in humans (and rabbits), new metasta-
accumulation and activation of macrophages (see tic foci usually heal as microscopic lesions, suf-
chapters 2 and 5) (19, 20).Tuberculous lesions 0.1 ficient bacillary antigen is almost always pro-
to 2 mm in diameter may heal completely and duced to maintain the tuberculin reaction.
disappear.Larger lesions (2 to 5 mm) with caseous One of the arguments in favor of vaccina-
centers may eventually be absorbed. Still larger tion with effective Mycobacterium bovis BCG is
caseous and liquefied foci (5 to 20 mm) may that the host is safer harboring BCG than har-
undergo fibrous encapsulation and are thereby boring virulent tubercle bacilli.Although both
isolated within the host (see chapter 4). attenuated and virulent strains increase resis-
In both humans and rabbits, the healing of tance to exogenous reinfection, hosts harbor-
small endogenous metastatic tubercles (and small ing virulent strains have a greater risk of pro-
tubercles of exogenous reinfection) involves gressive infection should their resistance be
basically the same mechanisms as healing of impaired.
primary foci, i.e., the local accumulation and
activation of macrophages at the site of infection. ORGAN AND SPECIES RESISTANCE
However, because of the already existing cell- Different organs of the host have different sus-
mediated immunity and DTH in the host, ceptibilities to tubercle bacilli. In rabbits, tuber-
macrophage accumulation and activation occur cles in the liver usually remain small and heal,
more rapidly in metastatic tubercles (see chap- whereas in their kidneys they enlarge and cause
ters 5 and 10) and often stop bacillary multipli- renal disease (9). In guinea pigs, the reverse
cation in the early stages of lesion development. situation occurs: tubercles enlarge in the liver,
Since the interplay of local factors determines but they remain small in the kidneys and usu-
the fate of each lesion (21, 22), hosts with clin- ally heal (9).
7. STRUCTURAL COMPONENTS OF TUBERCULOUS LESIONS 䡵 159

In each animal species and in each organ, molecules and cytokines (chemokines) cause a
macrophages from the blood enter the devel- rapid antigen-specific cell infiltration at sites of
bacillus Calmette-Guérin reinfection. Immunol-
oping lesion, but once there, they are evidently ogy 102:466–479.
influenced by local factors present in the specific 5. Long, E. R. 1958. The Chemistry and Chemother-
organ and by genetic factors present in the apy of Tuberculosis, 3rd ed., p. 163–182. Lippincott
species. To date, these local factors have not Williams & Wilkins, Baltimore, Md.
been identified, but they probably involve 6. Poole, J. C. F., and H.W. Florey. 1970. Chronic
cytokines (produced by resident cells) that affect inflammation and tuberculosis, p. 1183–1224. In H.
W. Florey (ed.), General Pathology, 4th ed. The
the infiltrating dendritic cells, macrophages, and W. B. Saunders Co., Philadelphia, Pa.
lymphocytes (see chapters 5 and 6). 7. Hemsworth, G. R., and I. Kochan. 1978.
Secretion of anti-mycobacterial fatty acids by nor-
RESISTANCE TO BOVINE-TYPE AND mal and activated macrophages. Infect. Immun.
HUMAN-TYPE TUBERCLE BACILLI 19:170–177.
In general, bovine-type tubercle bacilli are more 8. Courtade, E. T., T. Tsuda, C. R. Thomas,
virulent than human-type bacilli in most animal and A. M. Dannenberg, Jr. 1975. Capillary
species, especially in the rabbit, where the dif- density in developing and healing tuberculous
lesions produced by BCG in rabbits. A quantita-
ference is marked (see chapter 13). (In humans, tive study. Am. J. Pathol. 78:243–260.
the difference in virulence is not readily appar- 9. Lurie, M. B. 1964. Resistance to Tuberculosis: Exper-
ent.) In both guinea pigs and rabbits, live as imental Studies in Native and Acquired Defensive
well as dead bovine-type bacilli are more diffi- Mechanisms. Harvard University Press, Cambridge,
cult for the host to destroy than the human Mass.
10. Laskin, D. L., and K. J. Pendino. 1995.
type, and the bovine type usually produces larger Macrophages and inflammatory mediators in tis-
lesions (9). sue injury. Annu. Rev. Pharmacol.Toxicol. 35:655–
Live bovine-type bacilli can apparently sur- 677.
vive at a lower oxygen tension than can human- 11. Canetti, G. 1955. The Tubercle Bacillus in the Pul-
type bacilli (9), which explains some of the dif- monary Lesion of Man. Springer-Verlag, New York,
ferences found in the disease produced by the N.Y.
12. Rich, A. R. 1951. The Pathogenesis of Tuberculosis,
live strains. However, the main difference prob- 2nd ed. Charles C Thomas Publisher, Springfield,
ably is in their lipid compositions (see references Ill.
5, 9, and 24). 13. Converse, P. J., A. M. Dannenberg, Jr., J. E.
Estep, K. Sugisaki, Y. Abe, B. H. Schofield,
REFERENCES and M. L. M. Pitt. 1996. Cavitary tuberculosis
1. Dannenberg,A. M., Jr., and J. F.Tomashefski, produced in rabbits by aerosolized virulent tuber-
Jr. 1998. Pathogenesis of pulmonary tuberculosis, cle bacilli. Infect. Immun. 64:4776–4787.
p. 2447–2471. In A. P. Fishman (ed.), Fishman’s Pul- 14. Converse, P. J., A. M. Dannenberg, Jr., T.
monary Diseases and Disorders, 3rd ed., vol. 2. Shigenaga, D. N. McMurray, S.W. Phalen, J.
McGraw-Hill Co., Inc., New York, N.Y. L. Stanford, G. A.W. Rook,T. Koru-Sengul,
2. Chensue, S. W., K. Warmington, J. Ruth, H. Abbey, J. E. Estep, and M. L. M. Pitt.
P. Lincoln, M.-C. Kuo, and S. L. Kunkel. 1994. 1998. Pulmonary bovine-type tuberculosis in rab-
Cytokine responses during mycobacterial and schis- bits: bacillary virulence, inhaled dose effects, tuber-
tosomal antigen-induced pulmonary granuloma culin sensitivity, and Mycobacterium vaccae
formation. Am. J. Pathol. 145:1105–1113. immunotherapy. Clin. Diagn. Lab. Immunol. 5:871–
3. Sugisaki, K., A. M. Dannenberg, Jr., Y. Abe, 881.
J. Tsuruta, W.-J. Su, W. Said, L. Feng, 14a. Dannenberg, A. M., Jr., and M. Sugimoto.
T.Yoshimura, P. J. Converse, and P. Mounts. 1976. Liquefaction of caseous foci in tuberculosis.
1998. Nonspecific and immune-specific up- Am. Rev. Respir. Dis. 113:257–259. (Editorial.)
regulation of cytokines in rabbit dermal tubercu- 15. Yamamura,Y. 1958.The pathogenesis of tuber-
lous (BCG) lesions. J. Leukoc. Biol. 63:440–450. culous cavities. Adv.Tuberc. Res. 9:13–37.
4. Shigenaga, T., A. M. Dannenberg, Jr., D. B. 16. Yamamura, Y., Y. Ogawa, H. Maeda, and
Lowrie, W. Said, M. J. Urist, H. Abbey, B. H. Y. Yamamura. 1974. Prevention of tubercu-
Schofield, P. Mounts, and K. Sugisaki. 2001. lous cavity formation by desensitization with
Immune responses in tuberculosis: antibodies and tuberculin-active peptide. Am. Rev. Respir. Dis.
CD4-CD8 lymphocytes with vascular adhesion 109:594–601.
160 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

17. Dannenberg, A. M., Jr., and W. E. Bennett. genesis of tuberculosis: specificity, systemic and local
1964. Hydrolytic enzymes of rabbit mononuclear nature, and associated macrophage enzymes. Bacte-
exudate cells. I. Quantitative assay and properties of riol. Rev. 32:85–102.
certain proteases, nonspecific esterases and lipases of 22. Suga, M., A. M. Dannenberg, Jr., and
mononuclear and polymorphonuclear cells and S. Higuchi. 1980. Macrophage functional hetero-
erythrocytes. J. Cell Biol. 21:1–13. geneity in vivo: macrolocal and microlocal macro-
18. Dannenberg, A. M., Jr., and W. E. Bennett. phage activation, identified by double-staining
1963. Hydrolases of mononuclear exudate cells and tissue sections of BCG granulomas for pairs of
tuberculosis. I. Exudate characteristics, esterases, enzymes. Am. J. Pathol. 99:305–324.
proteinases and lipase. Arch. Pathol. 76: 581–591. 23. Gangadharam, P. R. J. 1995. Mycobacterial dor-
19. Dannenberg, A. M., Jr. 1991. Delayed-type mancy. Tuberc. Lung Dis. 76:477–479.
hypersensitivity and cell-mediated immunity in the
pathogenesis of tuberculosis. Immunol.Today 12:228– 24. Raffel, S. 1961. Immunity, 2nd ed., p. 412-441.
233. Appleton, New York, N.Y.
20. Dannenberg, A. M., Jr. 1993. Immunopatho- 25. Lurie, M. B., P. Zappasodi, and C. Tickner.
genesis of pulmonary tuberculosis. Hosp. Pract. 28: 1955. On the nature of genetic resistance to tuber-
33–40 (Off. ed. 51–58). culosis in the light of the host-parasite relationships
21. Dannenberg, A. M., Jr. 1968. Cellular hyper- in natively resistant and susceptible rabbits. Am.
sensitivity and cellular immunity in the patho- Rev.Tuberc. Pulm. Dis. 72:297–329.
8
MICROVASCULAR DENSITY
IN TUBERCULOUS LESIONS

Overview 161
Production of developing and healing BCG lesions and 2-day tuberculin
reactions 161
Capillary density determined by gelatin-colloidal carbon perfusion 162
Histopathology: microvasculature-cell interactions 163
Regulators of angiogenesis 167
Pathophysiology of blood supply 167
Microvascular thrombosis 168

Abstract. The vasculature plays an important role in the pathogenesis of tuberculous


lesions. Blood vessels bring in the host defense cells, and vascular thrombosis is a major
cause of caseous necrosis. This chapter describes the study of microvascular density in
tissue sections of developing and healing dermal BCG lesions and in 48-h dermal tuber-
culin reactions.
Rabbits were placed under deep terminal anesthesia, and their entire vasculature was
perfused (via the aorta) with a gelatin-colloidal carbon suspension.Then, serial 250-µm-
thick tissue sections of the dermal BCG lesions were prepared, and the total length of the
microvasculature in the whole BCG lesion was calculated from measurements made with
a microscope containing an ocular grid.
By 3 days, the vascular density in BCG lesions had increased to roughly 1.6 times that
found in normal skin. It remained at this level for at least 6 to 7 weeks.The vascular den-
sity in tuberculin reactions showed a similar increase.
It was concluded that the local microvasculature increases relatively little during the course
of this slowly healing infection.A greater blood flow through existing capillaries evidently
provides most of the nourishment needed by the infiltrating cells.
These studies also demonstrated that microvascular thrombosis is a major cause of the
caseous necrosis that occurs during the course of this disease.

OVERVIEW The study described in this chapter on der-


The microvasculature plays a major role in the mal BCG lesions seems to be the only one
inflammatory response to infectious agents (see ever done on the total amount of microvas-
reference 1). It supplies defense cells, antibodies, culature of tuberculous lesions as they progress
and other blood components so that the offend- and heal. See chapter 21 for our studies on the
ing agent is destroyed (or detoxified), and microvascular adhesion molecules in such
necrotic debris is removed. For convenience, in lesions.
this chapter I often use the word “capillaries” to
represent the entire microvascular network (2), PRODUCTION OF DEVELOPING
i.e., the precapillary arterioles that control the AND HEALING BCG LESIONS AND
amount of blood entering, the capillaries them- 2-DAY TUBERCULIN REACTIONS
selves, which supply oxygen and nutrients to the In Experiment I, each rabbit had either 21-day
tissues and remove waste products, and the post- or 42-day dermal BCG lesions when eutha-
capillary venules through which leukocytes nized. In Experiment II, each rabbit, when euth-
enter the tissues (2, 3). anized, had 49-, 35-, 15-, and 3-day lesions;

161
162 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

i.e., lesions of different ages were present on venules, with the gelatinized colloidal carbon
their flanks at the same time (1). particles. After formalin fixation, serial tissue
Tuberculin reactions were produced by the sections (250 m thick) of the BCG lesions
intradermal injection of a 1:10 dilution of Old and tuberculin reactions were prepared in a
Tuberculin and evaluated 2 days later. cryostat, stained with hematoxylin, and mounted
serially in Permount.Then, in the nonnecrotic
CAPILLARY DENSITY DETERMINED tissues of the lesions, the average capillary den-
BY GELATIN-COLLOIDAL CARBON sity was determined in millimeters of capillary
PERFUSION
length per mm2 of 250-m tissue section (see
Procedure reference 1).
Rabbits under deep terminal anesthesia were
heparinized, exsanguinated, and then perfused Results and Conclusions
via the aorta with a warm solution of 2% gelatin, The capillary density in BCG lesions of various
10% Pelikan India ink (a colloidal carbon sus- ages increased roughly 1.6 times when com-
pension), and 1% detergent in 0.9% pyrogen-free pared with the density in the normal skin nearby
NaCl (1).This procedure filled the entire vas- (Fig. 1).This increase was evident as early as 3
culature of the animal, including capillaries and days, which suggests that vascular proliferation

FIGURE 1 Capillary density in the BCG


lesions of Experiment II and in the normal
skin nearby. In this experiment, the skin of
each of the six rabbits contained 3-, 15-,
35-, and 49-day BCG lesions when they
were perfused with the gelatin-colloidal
carbon suspension. The means and their
standard errors are depicted.
The capillary density of BCG lesions
(in lengths per mm2) was increased as early
as 3 days. This density was highest at 15
days, when tuberculin sensitivity was devel-
oping, and decreased relatively little as the
lesions healed. Reproduced with permission
from reference 1.
8. MICROVASCULAR DENSITY IN TUBERCULOUS LESIONS 䡵 163

occurred rather rapidly. The perfusion of the HISTOPATHOLOGY:


vasculature with the warm colloidal carbon sus- MICROVASCULATURE-CELL
pension seemed to dilate every blood vessel in INTERACTIONS
the entire host.Therefore, none of the microvas- BCG Lesions
culature was closed by precapillary sphincters, The vascular network plays a role in the for-
and every vessel in both the BCG lesions and mation, progression, and resolution of BCG
normal skin was apparently filled with the col- lesions. Following the injection of tubercle
loidal carbon suspension. bacilli, granulocytes and mononuclear cells
In 15-day BCG lesions, the capillary density (mostly macrophages) emigrate from the sub-
tended to be maximally different from that in papillary vascular plexus that nourishes the epi-
normal skin (Fig. 1). At that time tuberculin dermis and hair follicles. The hydrolases from
sensitivity was developing (1). Otherwise, the cells in the exudate are probably responsible for
capillary density changed rather little as the the dissolution of ground substance, collagen
BCG lesions developed and then healed. In fibers (4), and the fibrin that had formed (5).
other words, the 60% increase in the density of Some of the cells in the exudate died and
the capillary network seemed adequate to con- formed microabscesses (Fig. 2), which soon
tinuously supply the local needs throughout the fused to create the caseous center. Presumably
course of this infection. by looping, budding, and anastomosing (6), the
The capillary densities in the 2-day tuberculin capillary network increased around the hair fol-
reactions resembled those found in the BCG licles and formed a plexus around the caseous
lesions (1). center (Fig. 3, 4, and 5). The capillaries were

FIGURE 2 Dermal BCG lesion, 3 days of age.An increased number of vessels are present in
the subpapillary vascular plexus surrounding the hair follicles (sometimes called the peritrichous
plexus). In the center and on the right of the photograph, the hair follicles were damaged by
microabscesses developing between them. In the lower part of the photograph, the deep cuta-
neous vascular plexus is prominent. It supplies the intrinsic muscles of the skin and helps regu-
late body heat. Gelatin-colloidal carbon perfusion, counterstained with hematoxylin. Magnifi-
cation, ⫻16. Reproduced with permission from reference 1.
164 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

FIGURE 3 Dermal BCG lesion, 15 days of age, cut near the edge of the necrotic center. At
this location, the vessels of the subpapillary vascular plexus form a rich anastomosing network
with the vessels of the deep cutaneous vascular plexus. Magnification, ⫻23.
We used an ocular grid to count (at 44⫻ magnification) the lengths of capillaries within
grid squares, each measuring 0.45 by 0.45 mm. In the grid square shown centrally in the inset,
the 23 pieces of microvasculature totaled 8.3 grid widths. Gelatin-colloidal carbon perfusion,
counterstained with hematoxylin. Magnification, ⫻3.7. Reproduced with permission from ref-
erence 1.

thrombosed next to the caseous center. Such (Fig. 6), and the rest was gradually ingested,
thrombosis is thought to be a major cause of broken down, or sequestered by the infiltrating
caseous necrosis. macrophages. The capillaries surrounding the
The capillaries of the deep cutaneous plexus now shrunken caseous center apparently
are situated in the dermis just above the muscle remained fully functional and served as the
fibers that move the skin (Fig. 2 and 3).This vas- source of new phagocytes, as well as nutrients for
cular plexus contains many arterial-venous shunts fibroblasts.The capillary network was reduced
that regulate the flow of blood to the skin for to normal size only after most of the necrotic
heat exchange. As the caseous center enlarged, material had been removed and new connective
this deep cutaneous plexus also proliferated, sup- tissue had formed (Fig. 7).
plying branches that surrounded the lower half
of the caseous center and fused with similar Tuberculin Reactions
branches from the subpapillary plexus. Some The tuberculin reactions usually showed no
capillary plexuses became buried in exudate cells necrosis, although microabscesses were occa-
(Fig. 4), some were injured, some leaked (Fig. 6), sionally found.The capillaries in these reactions
and some thrombosed. were mainly from the subpapillary plexus, with
After the bacilli were inhibited or destroyed little contribution from the deep cutaneous
by the macrophages (7–11), the BCG lesions plexus (Fig. 8). In other respects, the capillary net-
regressed and began to heal. Much of the work resembled that found in the nonnecrotic
necrotic material was discharged by ulceration tissues of fully developed BCG lesions (1).
FIGURE 4 Dermal BCG lesion, 35 days of age.This section was cut from the outer regions
of the caseous necrotic center (but more central than that in Fig. 3). Here, the cellular infiltrate
is not completely necrotic, and the capillaries are still patent or only partly thrombosed. Gelatin-
colloidal carbon perfusion, counterstained with hematoxylin. Magnification, ⫻22. Reproduced
with permission from reference 1.

FIGURE 5 Dermal BCG lesion, about 21 days of age.This section was cut through the caseous
necrotic center.The intact epidermis is shown above.The liquefied caseous necrotic center is shown
below. Part of this center fell out during the preparation of this tissue section.
Note that in the necrotic area the capillaries were not perfused with carbon, which indicates
that thrombosis and the resulting ischemia are major causes of tissue necrosis. Gelatin-colloidal car-
bon perfusion, counterstained with hematoxylin. Magnification, ⫻23.
165
166 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

FIGURE 6 Dermal BCG lesion, 15 days of age, with ulceration and discharge of some of the
necrotic material.The vascular plexus at the base of the ulcer is richly developed. Some leakage
of the colloidal carbon into the necrotic area is evident. Gelatin-colloidal carbon perfusion, coun-
terstained with hematoxylin. Magnification, ⫻20. Reproduced with permission from reference 1.

FIGURE 7 Healing dermal BCG lesion, 49 days of age. The total vascular network has
decreased, and most of the necrotic material has been discharged or absorbed. Fibroblasts, pro-
ducing new collagen fibers and ground substance, are present.The intrinsic muscles of the skin
(the panniculus carnosis) can be seen at the bottom of the photograph. Gelatin-colloidal carbon
perfusion, counterstained with hematoxylin. Magnification, ⫻17. Reproduced with permission
from reference 1.
8. MICROVASCULAR DENSITY IN TUBERCULOUS LESIONS 䡵 167

FIGURE 8 A positive 2-day tuberculin reaction.The subpapillary vascular plexus surround-


ing the hair follicles has proliferated, but no necrosis is present. Gelatin-colloidal carbon perfu-
sion, counterstained with hematoxylin. Magnification, ⫻23. Reproduced with permission from
reference 1.

REGULATORS OF ANGIOGENESIS thrombospondin, platelet factor 4, interferons,


Angiogenesis is controlled by a balance between interleukin 12, and tissue inhibitors of metallo-
positive and negative regulators (2). Most of proteinases (2, 12).
these regulators are produced by macrophages,
lymphocytes, granulocytes, mast cells, fibroblasts, PATHOPHYSIOLOGY
pericytes, and platelets (see references 2 and 12 OF BLOOD SUPPLY
through 21). An adequate blood supply is necessary for cells,
The major stimulatory regulators of angio- tissues, and organs to function.The amount of
genesis are hypoxia, extracellular matrix and blood received by a tissue depends both on the
connective tissue components (13), angiogenin, number of microvessels and the amount of blood
angiopoietin-1, nitric oxide, fibroblast growth flowing through them.Visualization of capillar-
factors, vascular endothelial growth factor, tumor ies by microangiography with colloidal carbon
necrosis factor alpha, transforming growth fac- (or radio-opaque suspensions) demonstrates the
tor , interleukin-8, epidermal growth factor, number of functional microvessels, but not the
granulocyte colony-stimulating factor, platelet- amount of blood flow through them.
activating factor, urokinase, plasminogen activa- In acute inflammation, blood flow is often at
tor, and prostaglandins E1 and E2 (12). Metal- its maximum within a few hours and is limited
loproteinases degrade the nearby extracellular by the number of capillaries already in the tis-
matrix, thereby enabling endothelial cell migra- sue. In delayed-type inflammation, blood flow
tion to establish new microvasculature. can be further enhanced by capillary prolifera-
The major inhibitory regulators of angio- tion.Therefore, even though microangiographic
genesis are angiostatin, endostatin, vasostatin, techniques do not measure total blood flow
168 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

into the tissues, they do measure the total capac- genase by macrophages exposed to lymphocyte
ity of the local vasculature.The density of this products. Fed. Proc. 33:618.
5. Unkeless, J. C., S. Gordon, and E. Reich. 1974.
vasculature in dermal BCG lesions was increased Secretion of plasminogen activator by stimulated
an average of 1.6 times over that of normal skin macrophages. J. Exp. Med. 139:834–850.
and remained at this capacity as the lesions pro- 6. Schoefl, G. I. 1963. Studies on inflammation.
gressed and healed.This moderate increase was III. Growing capillaries: their structure and per-
apparently adequate to stop the progression of meability. Virchows Arch. Pathol.Anat. 337:97–141.
7. Lurie, M. B. 1965. Resistance to Tuberculosis: Exper-
the BCG infection. imental Studies in Native and Acquired Defensive Mech-
Note that the skin and lungs are endowed anisms. Harvard University Press, Cambridge, Mass.
with much more vasculature than is required to 8. Dannenberg, A. M., Jr. 1968. Cellular hyper-
nourish these tissues. The skin has excess vas- sensitivity and cellular immunity in the patho-
culature to dissipate the excess body heat and genesis of tuberculosis: specificity, systemic and local
nature, and associated macrophage enzymes. Bacte-
nourish the hair follicles.The lungs have excess riol. Rev. 32:85–102.
vasculature to supply oxygen and remove car- 9. Mackaness, G. B. 1971. Delayed hypersensitivity
bon dioxide from the blood. In each case, the and the mechanism of cellular resistance to infec-
total vasculature should be ample to supply tion, p. 413–124. In B. Amos (ed.), Progress in
most of the needs of acute and chronic inflam- Immunology (1st International Congress of Immunology).
Academic Press, Inc., New York, N.Y.
matory processes without substantial increases. 10. Ando, M., A. M. Dannenberg, Jr., M. Sugi-
moto, and B. S. Tepper. 1977. Histochemical
MICROVASCULAR THROMBOSIS studies relating the activation of macrophages to the
Our studies also demonstrate the importance of intracellular destruction of tubercle bacilli. Am. J.
vascular thrombosis in the formation of caseous Pathol. 86:623–634.
necrosis in tuberculosis (see Fig. 5 and 6). 11. Lurie, M. B., P. Zappasodi, E. Cardona-Lynch,
and A. M. Dannenberg, Jr. 1952.The response
Thrombosis not only prevents hemorrhage but to the intracutaneous inoculation of BCG as an
also plays a role in killing nonactivated index of native resistance to tuberculosis. J. Immunol.
macrophages in which numerous tubercle bacilli 68:369–387.
are growing (see chapters 7 and 21). 12. Talks, K. L., and A. L. Harris. 2000. Current
The clotting of blood is under tight control status of antiangiogenic factors. Br. J. Haematol.
109:477–489.
and is subject to many interacting mechanisms 13. Inber, D. E., and J. Folkman. 1989. How does
(2), including cytokines (22). extracellular matrix control capillary morphogen-
esis? Cell 58:803–805.
REFERENCES 14. Folkman, J., and Y. Shing. 1992. Angiogenesis.
1. Courtade, E. T., T. Tsuda, C. R. Thomas, and J. Biol. Chem. 267:10931–10934.
A. M. Dannenberg, Jr. 1975. Capillary density in 15. Polverini, P. J., R. S. Cotran, and M. M. Shol-
developing and healing tuberculous lesions pro- ley. 1977. Endothelial proliferation in the delayed
duced by BCG in rabbits.A quantitative study. Am. hypersensitivity reaction: an autoradiographic study.
J. Pathol. 78:243–260. J. Immunol. 118:529–532.
2. Majno, G., and I. Joris. 2004. Cells,Tissues, and 16. Cashin, C. H., B. Dodge, and G. P. Harper.
Disease. Principles of General Pathology, 2nd ed. Oxford 1991. Angiogenesis and chronic inflammation.
University Press, New York, N.Y. Agents Actions 34:332–338.
3. Tanaka, F.,A. M. Dannenberg, Jr., K. Higuchi, 17. Keatinge,W. R., and M. C. Harmon. 1980. Local
M. Nakamura, P. J. Pula, T. E. Hugli, R. G. Mechanisms Controlling Blood Vessels. Monographs of the
DiScipio, and D. L. Kreutzer. 1997. Chemo- Physiological Society, vol. 37. Academic Press, Inc.,
tactic factors are continuously released by cultured London, England.
intact developing and healing skin lesions produced 18. Folkman, J., and M. Klagsbrun. 1987. Angio-
in rabbits by sulfur mustard. Inflammation 21:251– genic factors. Science 235:442–447.
267. 19. Folkman, J. 1995.Angiogenesis in cancer, vascu-
4. Wahl, L. M., S. M. Wahl, G. R. Martin, and lar, rheumatoid and other diseases. Nat. Med. 1:
S. E. Mergenhagen. 1974. Production of colla- 27–31.
8. MICROVASCULAR DENSITY IN TUBERCULOUS LESIONS 䡵 169

20. Hammersen, F., and O. Hudlicka (ed.). 1984. growth factor by endothelial cells and monocytes
Angiogenesis. Progress in Applied Microcirculation, vol. and promotes angiogenesis in vivo. Blood 96:3801–
4. S. Karger, Basel, Switzerland. 3808.
21. Melter, M., M. E. J. Reinders, M. Sho, S. Pal, 22. Joseph, L., L. M. Fink, and M. Hauer-Jensen.
C. Geehan, M. D. Denton, D. Mukhopadhyay, 2002. Cytokines in coagulation and thrombosis: a
and D. M. Briscoe. 2000. Ligation of CD40 preclinical and clinical review. Blood Coagul. Fibri-
induces the expression of vascular endothelial nolysis 13:105–116.
9
EARLY PULMONARY
LESIONS IN RABBITS

Advantages of producing early pulmonary lesions by the intravenous


injection of tubercle bacilli 170
Activation and division of blood-borne macrophages in
the early lesions 170
Division of activated pulmonary alveolar macrophages 175

Abstract. In rabbits, 1- to 7-day pulmonary tuberculous lesions produced by aerosols are


difficult to find because the inhaled dose of tubercle bacilli cannot be made large enough.
A large intravenous dose, however, readily produces many such tubercles. This chapter
describes their characteristics and provides information on the activation and multiplica-
tion of macrophages within such lesions.
To produce these early lesions, we injected rabbits intravenously with 108 to 109 tuber-
cle bacilli (BCG). The blood-borne macrophages that entered the developing tubercles
became partly activated during the first day.These entering macrophages retained their abil-
ity to divide, i.e., incorporate [3H]thymidine ([3H]TdR), even though they had ingested
tubercle bacilli. In contrast, fully activated macrophages within tuberculous lesions lose their
ability to divide (see chapter 10).
Pulmonary alveolar macrophages did not seem to participate in early pulmonary lesions
produced by the intravenous route, but accumulated in the surrounding alveolar spaces.
However, even though these alveolar macrophages were highly activated, they retained their
ability to divide.

ADVANTAGES OF PRODUCING microscopic lesions.Therefore, early pulmonary


EARLY PULMONARY LESIONS BY lesions produced by intravenous tubercle bacilli
THE INTRAVENOUS INJECTION OF should resemble lesions produced by inhaled
TUBERCLE BACILLI
virulent tubercle bacilli in the early symbiotic
In rabbits, 1- to 7-day pulmonary lesions pro-
(logarithmic) stage (see chapter 2), and also
duced by the inhalation of tubercle bacilli are
resemble beginning metastatic lesions caused
usually too few to find microscopically in tissue
by hematogenously spread tubercle bacilli.
sections. The inhaled dose is always too small
In clinically active human tuberculosis, early
compared to the number of alveolar spaces, and
tubercles of hematogenous origin often occur.
the highly activated alveolar macrophage pop-
They should be similar to those described here,
ulation often prevents the inhaled bacilli from
except that their progress would be acceler-
establishing lesions.
ated, because they develop in a person who is
An intravenous injection of tubercle bacilli
already tuberculin positive.
eliminates these difficulties.A much larger bacil-
lary dose can be deposited in the lungs, and the ACTIVATION AND DIVISION OF
pulmonary alveolar macrophages are bypassed. BLOOD-BORNE MACROPHAGES IN
Following an intravenous injection, the bacilli THE EARLY LESIONS
(lodging in capillaries) are ingested directly by In each rabbit an uncentrifuged suspension of
nonactivated blood-borne monocytes/macro- BCG containing 108 to 109 viable bacilli was
phages, in which they grow and produce early injected intravenously (1).The suspensions con-

170
9. EARLY PULMONARY LESIONS IN RABBITS 䡵 171

FIGURE 1 One-day pulmonary lesion produced by intravenously injected BCG.A small blood
vessel (part of which is cut tangentially) is depicted with an embolus of tubercle bacilli in its cen-
ter (arrow). Some of the macrophages in this lesion stain ⫹⫹ for -galactosidase. In the alveo-
lar spaces (on the left) are six or seven (dividing) [3H]TdR-positive mononuclear cells (proba-
bly lymphocytes and young macrophages). In the lower right corner are three [3H]TdR-negative
alveolar macrophages, identified by their large size and ⫹⫹⫹ staining for -galactosidase. In vi-
tro [3H]TdR, 5-bromo-4-chloro-3-indolyl--D-galactoside, carbol-fuchsin, and hematoxylin. Mag-
nification, ⫻760. Reproduced with permission from reference 1.

tained many units of 2 to 5 bacilli, as well as In 1-day pulmonary BCG lesions, mononu-
some larger units. Many of these units lodged in clear cells (mostly macrophages with some lym-
the microvasculature of the lung and formed phocytes and probably dendritic cells) accumu-
microscopic lesions (Fig. 1). lated around a central bacillary core (Fig. 1) (1).
To assess the percentage of dividing cells in At this stage within the lesions, a few mononu-
the developing lesions, we incubated pieces of clear cells showed ⫹ and ⫹⫹ -galactosidase
pulmonary tissue (3 to 4 mm in diameter) with activity, but [3H]TdR-labeled mononuclear cells
[3H]TdR in vitro under hyperbaric oxygen (1) were rarely found there (Fig. 1). Most rabbit
(see chapter 10). Then, frozen tissue sections lymphocytes do not stain for -galactosidase.
were prepared, stained for -galactosidase (our In 2- and 3-day pulmonary BCG lesions,
lysosomal marker for macrophage activation [1– more macrophages were activated, i.e., had
4]) and sometimes for cytochrome oxidase (our increased their levels of -galactosidase. Some of
mitochrondrial marker for macrophage activa- these macrophages died, and some of them
tion [1, 2, 5]), and autoradiographed as described divided (Fig. 2 and 3A and B). Perifocally, many
in reference 1. This in vitro method labeled new mononuclear cells accumulated, and these
only about half as many mononuclear cells as did cells divided more frequently (Fig. 2) (1). At
an intravenous injection of [3H]TdR during about this time, typical tuberculous lesions
the first hour (1) (see chapter 10). became established. They contained a central
172 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

FIGURE 2 Two-day pulmonary lesions produced by intravenously injected BCG. A few


acid-fast tubercle bacilli (arrows) can be seen in their small necrotic centers. Mononuclear cells
from the bloodstream surround the centers.The lesions contained bright-blue ⫹⫹ -galactosi-
dase-positive macrophages, some of which were necrotic. In the alveolar areas outside the lesions
are about a dozen young -galactosidase-negative mononuclear cells showing [3H]TdR incor-
poration. In vitro [3H]TdR, 5-bromo-4-chloro-3-indolyl--D-galactoside, carbol-fuchsin, and
hematoxylin. Magnification, ⫻520. Reproduced with permission from reference 1.

core of dead and dying cells and a more periph- In summary, the activation of macrophages in
eral region of accumulating and proliferating rabbit pulmonary tuberculous lesions produced
cells (Fig. 2). by intravenous BCG began within 24 h. By 6
In 6-day pulmonary BCG lesions, -galac- days, many macrophages had died and formed
tosidase had disappeared from the dead cells in the distinct necrotic center of the developing
the necrotic centers, but cytochrome oxidase tubercle. Only recently arriving macrophages,
sometimes remained there (Fig. 4). By 10 and 12 i.e., those in 2- and 3-day lesions and those in
days, neither enzyme was present in the necrotic the peripheral areas of older lesions, seemed to
areas (Fig. 5). divide, i.e., incorporate [3H]TdR. Macrophages
In rabbits, 13-day pulmonary lesions pro- close to the necrotic center no longer divided.
duced by an aerosol inhalation of 5.6 ⫻ 106 vir- They had apparently entered the lesions earlier,
ulent bovine-type tubercle bacilli and 19-day and some of them were probably dying.
pulmonary lesions produced by an aerosol Data presented in reference 1 indicate that
inhalation of 4.2 ⫻ 107 virulent human-type intracellular tubercle bacilli do not stimulate
bacilli showed percentages of [3H]TdR-labeled macrophage division and that tissue macrophages
macrophages similar to those produced by intra- do not divide when they are strongly activated
venous BCG (1). for -galactosidase (see chapter 10).
9. EARLY PULMONARY LESIONS IN RABBITS 䡵 173

FIGURE 3 Three-day pulmonary lesions produced by intravenously injected BCG.


(A) In the upper part of the photograph is a [3H]TdR-positive macrophage staining ⫹⫹
for -galactosidase containing over 10 bacilli (upper arrow). Below this cell is a [3H]TdR-
negative, ⫹ -galactosidase-positive macrophage containing four bacilli (lower arrow). A
⫹⫹⫹ -galactosidase-positive macrophage with two bacilli is at the far right.The lowest
“black” cell is probably a [3H]TdR-positive, -galactosidase-negative lymphocyte. (B) A
[3H]TdR-positive, ⫹ -galactosidase-positive macrophage containing two bacilli is marked
with an arrow. Two ⫹⫹⫹ -galactosidase-positive ([3H]TdR-negative) macrophages are
also present. [3H]TdR-positive mononuclear cells containing bacilli were rare and could
be found with assurance only in early pulmonary lesions. In vitro [3H]TdR, 5-bromo-4-
chloro-3-indolyl--D-galactoside, carbol-fuchsin, and hematoxylin. Magnification, ⫻720.
Reproduced with permission from reference 1.
174 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

FIGURE 4 Six-day pulmonary lesion produced by intravenously injected BCG and stained
for cytochrome oxidase (CO). In the center are epithelioid cells rich in CO. Surrounding them
are young infiltrating macrophages, most of which are still poor in CO. At the periphery are
numerous alveolar macrophages, rich in CO, that were apparently attracted to the site by chemo-
taxins released from the lesion. Some of the epithelioid cells in the center are already necrotic,
but their CO activity is still present. 1,2,3,4-Tetrahydroquinoline and p-aminodiphenylamine;
no counterstain. Magnification, ⫻180. Reproduced with permission from reference 1.
9. EARLY PULMONARY LESIONS IN RABBITS 䡵 175

FIGURE 5 Ten-day pulmonary lesion produced by an intravenous injec-


tion of BCG. In the caseous center of the tubercle are disintegrating -galac-
tosidase-negative epithelioid cells containing more than 10 faintly staining
tubercle bacilli.Around the center are still-viable young -galactosidase-neg-
ative mononuclear cells, several of which show [3H]TdR incorporation.Alve-
olar macrophages, staining ⫹⫹⫹ and ⫹⫹⫹⫹ for -galactosidase, have
accumulated in the surrounding alveoli below the tubercle.These alveolar
macrophages did not contain tubercle bacilli, but several had incorporated
[3H]TdR (not clearly seen in this photograph). In vitro [3H]TdR, 5-bromo-
4-chloro-3-indolyl--D-galactoside, carbol-fuchsin, and hematoxylin. Mag-
nification, ⫻320. Reproduced with permission from reference 1.

DIVISION OF ACTIVATED highly activated tissue macrophages within the


PULMONARY ALVEOLAR lesions rarely do so (see chapter 10).
MACROPHAGES
These in vitro [3H]TdR studies also provided
information on pulmonary alveolar macro- REFERENCES
phages.Alveolar macrophages (as a population) 1. Shima, K., A. M. Dannenberg, Jr., M. Ando,
are highly activated (5) with a high content of S. Chandrasekhar, J. A. Seluzicki, and J. I.
-galactosidase (4) and other enzymes (4–11). Fabrikant. 1972. Macrophage accumulation, divi-
They frequently migrated from neighboring sion, maturation, and digestive and microbicidal
alveoli into the alveoli surrounding the young capacities in tuberculous lesions. I. Studies involv-
ing their incorporation of tritiated thymidine and
tuberculous lesions (Figs. 1, 4, and 5), but no their content of lysosomal enzymes and bacilli.
tubercle bacilli were found in these alveoli.The Am. J. Pathol. 67:159–180.
highly activated alveolar macrophages adjacent 2. Dannenberg, A. M., Jr., O. T. Meyer, J. R.
to the lesions could limit the local spread of Esterly, and T. Kambara. 1968.The local nature
tubercle bacilli if it occurred. of immunity in tuberculosis, illustrated histochem-
ically in dermal BCG lesions. J. Immunol. 100:931–
Alveolar macrophages with ⫹⫹⫹ to ⫹⫹⫹⫹ 941.
-galactosidase activity were often labeled with 3. Ando, M., A. M. Dannenberg, Jr., M. Sugi-
[3H]TdR (Fig. 5). Therefore, highly activated moto, and B. S. Tepper. 1977. Histochemical
alveolar macrophages can still divide, whereas studies relating the activation of macrophages to the
176 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

intracellular destruction of tubercle bacilli. Am. J. Macrophage esterase: identification, purification


Pathol. 86:623–634. and properties of a chymotrypsin-like esterase
4. Yarborough, D. J., O. T. Meyer, A. M. Dan- from lung that hydrolyzes and transfers nonpolar
nenberg, Jr., and B. Pearson. 1967. Histochem- amino acid esters. Biochim. Biophys.Acta 403:161–
istry of macrophage hydrolases. III. Studies on 179.
-galactosidase, -glucuronidase and aminopeptidase 9. Rojas-Espinosa, O., A. M. Dannenberg, Jr.,
with indolyl and naphthyl substrates. J. Reticuloen- L. A. Sternberger, and T.Tsuda. 1974. Role of
dothel. Soc. 4:390–408. cathepsin D in the pathogenesis of tuberculosis.
5. Dannenberg, A. M., Jr., M. S. Burstone, P. C. A histochemical study employing unlabeled anti-
Walter, and J.W. Kinsley. 1963.A histochemical bodies and the peroxidase-antiperoxidase complex.
study of phagocytic and enzymatic functions of Am. J. Pathol. 74:1–17.
rabbit mononuclear and polymorphonuclear exu- 10. Namba, M., M. Suga, F. Tanaka, A. M. Dan-
date cells and alveolar macrophages. I. Survey and nenberg, Jr., A.T. Hastie, and R. C. Franson.
quantitation of enzymes, and states of cellular acti- 1983. Immunocytochemical demonstration of rab-
vation. J. Cell Biol. 17:465–486. bit ribonuclease and phospholipase A2 by the per-
6. Carson, M. E., and A. M. Dannenberg, Jr. oxidase-antiperoxidase technique in professional
1965. Hydrolytic enzymes of rabbit mononuclear phagocytes (pulmonary alveolar macrophages and
exudate cells. II. Lysozyme: properties and quanti- granulocytic and mononuclear peritoneal exudate
tative assay in tuberculous and control inbred rab- cells) and in glycol methacrylate sections of dermal
bits. J. Immunol. 94:99–104. tuberculous (BCG) lesions. J. Reticuloendothel. Soc.
7. Meyer, O. T., A. M. Dannenberg, Jr., and 34:425–435.
K. Mizunoe. 1970. Hydrolytic enzymes of rabbit 11. Tsuda, T., A. M. Dannenberg, Jr., M. Ando,
mononuclear and polymorphonuclear exudate cells O. Rojas-Espinosa, and K. Shima. 1974.
and pulmonary alveolar macrophages. III. Deoxyri- Enzymes in tuberculous lesions hydrolyzing pro-
bonuclease and ribonuclease: properties and quan- tein, hyaluronic acid and chondroitin sulfate: a
titative assay in macrophages from tuberculous study of isolated macrophages and developing and
and control inbred rabbits. J. Reticuloendothel. Soc. healing rabbit BCG lesions with substrate film
7:15–31. techniques; the shift of enzyme pH optima towards
8. Rojas-Espinosa, O., P. Arce-Paredez, A. M. neutrality in “intact” cells and tissues. J. Reticu-
Dannenberg, Jr., and R. L. Kamenetz. 1975. loendothel. Soc. 16:220–231.
10
MACROPHAGE TURNOVER,
DIVISION, AND ACTIVATION
IN TUBERCULOUS LESIONS

Overview 178
In vitro [3H]TdR methods and their efficacy 178
Percentage of MN labeled in vitro with [3H]TdR in BCG lesions 179
In vitro division of activated macrophages in BCG lesions 179
In vitro division of macrophages containing tubercle bacilli in
BCG lesions 180
In vivo [3H]TdR studies of BCG lesions of various ages 180
MN entry into BCG lesions 180
MN disappearance from BCG lesions 182
Rates of MN division within BCG lesions 185
[3H]TdR-labeled MN in 1-day “tuberculin traps” 185
[3H]TdR-labeled MN in the blood 185
Rates of activation of MN in primary and reinfection BCG lesions 187
Macrophage turnover, activation, and division in healing dermal
BCG lesions 189
MN activation in tuberculin reactions and in lesions caused by nonspecific
irritants 192
Macrophage turnover in mouse tuberculous granulomas 193
Insights into DTH and CMI provided by our kinetic studies in
rabbits 193
Recapitulation of our kinetic studies on macrophages in tuberculous lesions
produced by BCG in rabbits 194

Abstract. In rabbit BCG lesions, the turnover of mononuclear cells was most rapid in BCG
lesions at 2 to 3 weeks, when the lesion size peaked and tuberculin sensitivity and acquired
cellular resistance were well developed. (The mononuclear cells were mostly macrophages,
with some medium and large lymphocytes and probably some dendritic cells.) At this 2-
to 3-week peak, more macrophages entered, more died or left, more remained at the site,
and more became activated than before or afterward. Before this time, the host had neither
delayed-type hypersensitivity nor cell-mediated immunity, so no antigen-specific enhance-
ment of the inflammatory response occurred.After this time, the bacilli and their antigenic
products had decreased, so antigen-specific stimuli for cell infiltration and activation were
reduced. In “healed” lesions, the mononuclear cell turnover still occurred but was decreased.
The continuous entry of live nonactivated macrophages into the viable parts of tuber-
culous lesions provides fresh intracellular sites where tubercle bacilli can multiply before
they are again inhibited by the delayed-type hypersensitivity and cell-mediated immunity
of the host. In tuberculosis, bacillary dormancy of long duration can only be present in
caseous necrotic tissue where no live host cells exist.

To my knowledge, our papers on macrophage these nine papers and provides new insights and
kinetics in rabbits (references 1 through 9) are interpretations that were not presented in the
the only ones in which macrophage entry, divi- original publications. (Most of the chapter was
sion, activation, accumulation, and death were published as a review in reference 10.)
evaluated in tuberculous lesions as they devel- I realize that these kinetic studies are complex
oped and healed.This chapter is a synthesis of and difficult to understand. Yet, they provide

177
178 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

insights into cell turnover within tuberculous TABLE 1 Comparison of in vitro and in vivo
lesions that are not obtainable in any other way. [3H]TdR-labeling of MN in dermal BCG lesionsa
For a quick review of this chapter and its rele- In vitro (%) In vivo (%)
vance, see the last two sections.
1.3 2.6
The mononuclear cells (MN) that we
1.4 3.0
counted in our Giemsa-stained tissue sections
1.4 3.8
were mostly macrophages, with some medium
1.7 4.1
and large lymphocytes and probably some den-
1.9
dritic cells. Cells staining for -galactosidase
2.0
and cells containing tubercle bacilli were almost
Mean:
always macrophages.Therefore, we usually used
1.6 ⫾ 0.1b 3.4 ⫾ 0.3
the term macrophages rather than mononu-
clear cells (or MN) when cells containing - a
The percentages of in vitro and in vivo [3H]TdR-labeled
MN in rabbit 10-day dermal BCG lesions were compared. For
galactosidase or tubercle bacilli were evaluated. the in vitro experiments, MN were labeled by incubating thin
Lymphocytes do not usually stain for -galac- BCG lesion biopsy specimens in [3H]TdR (1.0 Ci/ml) for
tosidase, or stain only weakly.We do not know 1 h under hyperbaric oxygen. For the in vivo experiments, the
rabbits were euthanized 30 or 60 min after an intravenous
the percentage of dendritic cells in the mac- [3H]TdR pulse (0.5 Ci/g of body weight). The in vitro
rophage population that was evaluated, but we [3H]TdR labeled only about half as many MN as the in vivo
expect that it is relatively small. [3H]TdR labeled.Adapted from reference 1, where additional
data are presented.
b
P ⬍0.001.
OVERVIEW
We performed both in vitro (1) and in vivo
(2–9) experiments. In the in vitro experiments,
biopsy samples of dermal BCG lesions of vari- macrophages into the lesions and their disap-
ous ages were incubated with tritiated thymidine pearance from the lesions, as well as their rates
([3H]TdR) for 1 h under hyperbaric oxygen. of division and their rates of activation as the
The in vitro experiments clearly showed that BCG lesions developed, peaked, and regressed.
only a small percentage (1.6 ⫾ 0.1%) of MN
divided locally within the lesions (Table 1).
These results were confirmed by in vivo exper- IN VITRO [3H]TdR METHODS
iments in which the rabbits were euthanized 30 AND THEIR EFFICACY
or 60 min after an intravenous injection of Cross sections (2 to 3 mm thick) of these lesions
[3H]TdR (Table 1). were incubated under three atmospheres of
In vivo experiments described herein pro- oxygen at 37°C for 1 h in culture medium 199,
vide information on macrophage turnover in containing 1.0 Ci/ml of [3H]TdR in 20%
tuberculous lesions. In these experiments, fetal calf serum (1, 14). Frozen tissue sections of
[3H]TdR was injected intravenously once dur- these explants (prepared in a cryostat) were (i)
ing the development and healing of the dermal stained for the lysosomal enzyme -galactosi-
BCG lesions. Macrophages came from a rapidly dase, (ii) autoradiographed to detect [3H]TdR
dividing precursor population in the bone incorporation, (iii) stained for acid-fast bacilli,
marrow (11–13), where they became [3H]TdR and (iv) counterstained with hematoxylin (1, 2).
labeled. They then entered the bloodstream We only evaluated MN in the viable areas sur-
and subsequently entered the BCG lesions rounding the caseous centers (10).At these sites,
along with unlabeled macrophages. Five days some activated MN were usually present, as
after the intravenous [3H]TdR pulse, we found were the bacilli and/or their products.The MN
that 19 to 35% of MN were labeled in the were mostly macrophages (10).
lesions (2).Therefore, in vivo [3H]TdR-label- The in vitro method labeled about half as
ing experiments (in contrast to those in vitro) many MN (1) as did a 1-h in vivo [3H]TdR
enabled us to study the rates of entry of labeled pulse (Table 1).This discrepancy may be due to
10. MACROPHAGE TURNOVER, DIVISION,AND ACTIVATION 䡵 179

mild cell injury (from anoxia or trauma) during labeled in vitro with [3H]TdR had arrived
the biopsy procedure. It does, however, provide recently from the bloodstream (9).
an approximate measure of the number of cells
undergoing local division within the tubercu- IN VITRO DIVISION OF ACTIVATED
lous lesion. The in vitro method is much less MACROPHAGES IN BCG LESIONS
costly than the in vivo method, because relatively In viable areas of BCG lesions that contain
little [3H]TdR is used. bacilli, many macrophages develop into epithe-
lioid cells, rich in mitochondrial and lysosomal
PERCENTAGE OF MN LABELED enzymes (15, 16). Our stain for -galactosidase
IN VITRO WITH [3H]TdR IN BCG is a marker for such activated cells. The more
LESIONS mature the epithelioid cell, the darker it stains
In vitro [3H]TdR incubation labeled 0.8 to 4.0% for -galactosidase (15, 16). Such darkly stained
of the mononuclear cells (Tables 1 and 2) (1).This macrophages/epithelioid cells can effectively
low percentage remained fairly constant through- inhibit or destroy tubercle bacilli (15–17).
out the growth and regression of these lesions In the in vitro [3H]TdR experiments, some
(Table 2). This low percentage of in vitro macrophages staining ⫹ and ⫹⫹ for -galac-
[3H]TdR-labeled MN should be compared to tosidase incorporated [3H]TdR, but macrophages
the 19 to 35% in vivo [3H]TdR-labeled MN staining ⫹⫹⫹ and ⫹⫹⫹⫹ for -galactosidase
found in the lesions 5 days after an intravenous did not (1). Apparently, the strongly stained
[3H]TdR pulse (2).The [3H]TdR-labeled MN at macrophages had become so completely differ-
5 days were labeled in the bone marrow and entiated for digestive and microbicidal functions
entered the lesions during the 5-day period. that they no longer continued to divide.
From these studies we concluded that relatively In the in vivo [3H]TdR experiments on der-
few MN divide locally in the lesions,and the MN mal BCG lesions (described below), some

TABLE 2 In vitro[3H]TdR labeling of dermal BCG lesions indicates that intracellular tubercle bacilli do not
stimulate mononuclear cell (MN) divisiona
Percent Total no. of MN with both
3
Age of lesion Total no. of Percent of of MN [H]TdR label and intracellular
3
when biopsied MN counted [H]TdR- containing tubercle bacilli
(days) (A) positive tubercle
MN (B) bacilli (C) Expectedb Observed
Experiment I
7 14,100 2.0 ⫾ 0.02 3.5 ⫾ 0.8 8.1 0
12 13,800 4.0 ⫾ 1.3 2.7 ⫾ 0.8 7.5 5
19 31,500 2.3 ⫾ 0.6 0.4 ⫾ 0.2 5.7 0
Experiment II
7 17,900 1.0 ⫾ 0.3 3.4 ⫾ 0.7 4.9 0
12 8,300 1.6 ⫾ 0.3 4.4 ⫾ 0.5 4.3 0
19 22,400 0.8 ⫾ 0.2 0.8 ⫾ 0.4 2.4 0
Experiment III
7 27,500 1.6 ⫾ 0.3 4.6 ⫾ 1.2 16.7 5
14 29,600 2.6 ⫾ 0.5 3.5 ⫾ 1.1 24.0 7
a
The observed number of “doubly marked” MN was less than the expected number, which suggests that intracellular tuber-
cle bacilli did not stimulate macrophage division. Means and their standard errors are listed. More details are in footnote a of Table
1.Adapted from reference 1.
b
The expected numbers of “doubly marked” MN were not obtained by multiplying the means listed in columns A, B, and C:
they were obtained by multiplying A ⫻ B ⫻ C from individual rabbits and then averaging them. For example, in Experiment III,
the mean A ⫻ B ⫻ C: 27,500 ⫻ 1.6% ⫻ 4.6% ⫽ 20.2, whereas the average of A ⫻ B ⫻ C from individual rabbits was 16.7. Both
20.2 and 16.7 for the expected “doubly marked” MN were much higher than the 5 that were found.
180 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

[3H]TdR-labeled macrophages staining ⫹⫹⫹ lesions about 5 days after the intravenous
and ⫹⫹⫹⫹ for -galactosidase were found. [3H]TdR injection (2).This injection had appar-
These local macrophages had probably incor- ently labeled most of the dividing MN (mono-
porated [3H]TdR before they became highly cyte/macrophages) while they were still in the
activated for -galactosidase, because at least 5 bone marrow, and 5 days was enough time for
days (rather than 1 h) had elapsed after exposure these cells to leave the marrow and accumulate
to [3H]TdR. in the lesions.
[3H]TdR is rapidly incorporated into cells
IN VITRO DIVISION OF that replicate their DNA prior to cell division,
MACROPHAGES CONTAINING and the excess (unincorporated) [3H]TdR is
TUBERCLE BACILLI IN BCG LESIONS degraded and/or excreted within a few hours
[3H]TdR-positive macrophages containing (18). Once [3H]TdR is incorporated into DNA,
tubercle bacilli could occasionally be found in it will remain there for the life of the cell (18)
1- to 3-week BCG lesions (Table 2). These (unless it is diluted out by cell division).
findings indicate that a few macrophages con- In these experiments, [3H]TdR (0.5 Ci/g
taining tubercle bacilli were still able to divide. of body weight) was injected intravenously.
The observed number of MN “doubly When [3H]TdR of either 2 Ci/mmol or 20
marked” with [3H]TdR and intracellular bacilli Ci/mmol specific activity was used, no differ-
was lower than expected (Table 2). The lower ences were found in the percentage of MN
observed number could have been caused by labeled or in their [3H]TdR grain counts (7).
the shorter residence of [3H]TdR-labeled [3H]TdR cutoffs of 3 or 5 grains per cell were
macrophages in the dermal lesions. (Macrophages used, depending on the background silver grains
labeled in vitro with [3H]TdR were recently produced in each batch of Kodak photographic
entering cells with little opportunity to ingest emulsion.
bacilli.) The lower observed number of doubly The analysis of these in vivo [3H]TdR ex-
marked MN could also have been caused by periments is complex, so only the main princi-
the lymphocytes in the [3H]TdR-labeled MN ples will be presented here. If more details
population. Lymphocytes never ingest bacilli, (including the actual numerical data) are de-
but they may divide more frequently than sired, the reader should consult references 1
macrophages within BCG lesions, where the through 9.
antigens of the bacillus are located.
From these findings, we conclude that MN ENTRY INTO BCG LESIONS
macrophage division is not appreciably stimu- Figure 1 depicts the total number of [3H]TdR-
lated by intracellular bacilli, and that macrophage labeled MN entering the BCG lesions during
division is probably not a major mechanism by the 5-day period between the [3H]TdR pulse
which these phagocytes reduce their bacillary and lesion biopsy. Large numbers of [3H]TdR-
load. Lurie came to the same conclusion in his labeled MN entered primary BCG lesions when
eye chamber experiments (see chapter 5). the host became tuberculin positive.
Rabbits with reinfection BCG lesions were
IN VIVO [3H]TdR STUDIES OF BCG already tuberculin positive.Therefore, the num-
LESIONS OF VARIOUS AGES ber of labeled MN in reinfection lesions 4 days
The remainder of this chapter describes exper- of age was similar to the number in primary
iments in which [3H]TdR was injected intra- lesions at 11, 19, and 32 days (Fig. 1).
venously into rabbits 1 day before or 6, 14, 27, As BCG lesions were developing and healing,
41, or 63 days after dermal BCG lesions were the percentage of [3H]TdR-positive MN (5 days
begun. These lesions peaked in size at 2 to 3 after the [3H]TdR pulse) remained between 19
weeks and then slowly regressed. and 35% of the total MN population (Fig.2) (data
The highest percentage of [3H]TdR-labeled in reference 2). During the final stages of healing
MN was found in tissue sections of the BCG in BCG lesions 41 and 63 days of age, this per-
10. MACROPHAGE TURNOVER, DIVISION,AND ACTIVATION 䡵 181

FIGURE 1 The total number of [3H]TdR-labeled MN in primary BCG lesions of various


ages 5 days after an intravenous injection of [3H]TdR (3HT in figure). (A, age of BCG lesions
when [3H]TdR was injected intravenously; B, age of BCG lesions when biopsied. For example,
biopsies for the ⫹6 days on the bar graph were taken when the BCG lesions were 11 days of
age.)
Most of the [3H]TdR-labeled MN had incorporated [3H]TdR in the bone marrow before
they entered the lesions.Therefore, the number of [3H]TdR-labeled MN should be proportional
to the number of labeled plus unlabeled MN that entered during the previous 5 days.The num-
ber of [3H]TdR-labeled MN was 19 to 34% of the total number of MN within the lesions (2).
Note that many more MN entered after DTH was present. Reproduced with permission from
reference 2.
The total number of [3H]TdR-labeled MN in BCG lesions was calculated from the lesion
size (when measured with calipers), the number of MN per mm2 (counted microscopically), and
the percentage of these MN that were [3H]TdR labeled. After the caseous center developed in
1 to 2 weeks, the total number of [3H]TdR-labeled MN in the lesions was less than the num-
ber calculated, because caseous necrosis was present (3, 5).
182 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

FIGURE 2 Rate of appearance and disappearance of [3H]TdR-labeled MN in primary BCG


lesions of various ages. [3H]TdR (3HT in figure) was given as a single intravenous pulse to dif-
ferent groups of rabbits at the times indicated at the top of the graph, i.e., when their BCG lesions
were –1, ⫹6, ⫹14, or ⫹27 days of age. Note that in each case the percentage of [3H]TdR-labeled
MN in the lesions reached its peak about 5 days later, and that about 10 days after the peak these
labeled MN had largely disappeared from the lesions (see text). Reproduced with permission from
reference 3.

centage of [3H]TdR-labeled MN was reduced lesions had a rather high turnover rate and did not
but was still about 12% (data in reference 4). apparently persist there for long periods of time.
From these studies, we concluded that delayed- Peak [3H]TdR labeling of MN in the lesions
type hypersensitivity (DTH) and cell-mediated occurred around 5 days after the [3H]TdR pulse,
immunity (CMI) caused a high rate of entry of and most of these labeled MN had disappeared
new MN into BCG lesions throughout their from the lesions 10 days later (Fig. 2).The rate
development and regression. of disappearance of [3H]TdR-labeled MN was
most rapid when both acquired resistance
MN DISAPPEARANCE (DTH/CMI) and bacillary antigens were locally
FROM BCG LESIONS present (Fig. 2 and 3) (3, 4).
The continual entry of new MN into the BCG Before DTH and CMI developed, about 40%
lesions must be offset by a continual disappear- of the [3H]TdR-labeled MN remained in the
ance of MN from the lesions by dying or by leav- lesions during the 10 days between 5 and 15
ing via the lymphatics,because the lesions reached days after the [3H]TdR pulse (Fig. 3). When
their peak size in 14 to 24 days and then regressed the lesions peaked in size, only about 5% of the
(2, 3, 5). In other words, most MN in BCG MN remained for these 10 days. In the final
10. MACROPHAGE TURNOVER, DIVISION,AND ACTIVATION 䡵 183

FIGURE 3 Percentage of [3H]TdR-labeled MN that survived for 10 days in primary dermal


BCG lesions at various times during their growth and regression, specifically, the number of
[3H]TdR-labeled MN in the lesions at 15 days (after the intravenous [3H]TdR injection) divided
by the number at 5 days, multiplied by 100. (3HT is [3H]TdR.) In this experiment 5 days after
the intravenous [3H]TdR pulse, 11 to 33% of the MN in the lesions were labeled with [3H]TdR
(3).Ten days later many of these cells had disappeared (died).The percentage that survived dur-
ing this 10-day period is plotted in this graph (see text).The P values between the upper and lower
points are statistically significant (see references 3 and 4). Reproduced with permission from ref-
erence 4.
184 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

FIGURE 4 Changes in [3H]TdR grain counts in [3H]TdR-labeled MN over 22 days.An intra-


venous pulse of [3H]TdR (3HT in figure) was given when the primary BCG lesions were either
6 or 14 days of age.The grain count made 1 day after the intravenous pulse of [3H]TdR was con-
sidered 100%. Halving of this grain count should indicate one cell division.This figure suggests
(i) that MN in BCG lesions divided only once (or twice), and (ii) that the presence of DTH
reduced the time for this single division to occur, but see the text for another interpretation. Repro-
duced with permission from reference 2.
Grain counts on MN in BCG lesions of reinfection (with [3H]TdR given at –1 day) resem-
bled the lower curve. Grain counts on MN in primary lesions (with [3H]TdR given at –1 day)
resembled the upper curve, but the 1-day primary lesions were small with few cells to count. Grain
counts on MN in regressing primary BCG lesions (with [3H]TdR given at ⫹27 days) fell
between these two curves.

stages of healing, when most of the bacilli and tion confirmed that the rate of disappearance of
their tuberculin-like products had been [3H]TdR-labeled MN was increased in hosts
destroyed, about 40% again remained (Fig. 3). possessing DTH and CMI (3).
These percentages are only approximate, because This disappearance was largely due to cell
during this 10-day period, MN containing death, because the size of the caseous centers
decreasing numbers of [3H]TdR-labeled cells increased after the hosts became tuberculin pos-
were continually entering the lesions from the itive (3, 5). High local concentrations of tuber-
bloodstream (Fig. 2). culin are known to produce necrosis in tuber-
Nevertheless, this approximation clearly culin-positive rabbits, guinea pigs, and humans.
showed that [3H]TdR-labeled MN disappeared Spector’s group also found that the highest
from primary BCG lesions at an increased rate macrophage turnover rates occurred in granu-
when the host first developed DTH and CMI. lomas produced by agents that were most inju-
Similar studies with BCG lesions of reinfec- rious to tissues (19, 20).
10. MACROPHAGE TURNOVER, DIVISION,AND ACTIVATION 䡵 185

RATES OF MN DIVISION were evaluated (5) (Fig. 5). One day before
WITHIN BCG LESIONS each BCG biopsy was taken, we injected Old
To assess MN division, we counted the silver Tuberculin intradermally as an MN “trap” to
grains in the [3H]TdR-labeled MN. Halving of collect unlabeled and [3H]TdR-labeled MN
[3H]TdR grain counts indicates one cell divi- from the blood. The 1-day traps and BCG
sion after allowance is made for grains in the lesions were biopsied at the same time. The
background. At 2 and 5 days after the pulse of traps monitored the percentage of MN labeled
[3H]TdR, MN in the 14-day BCG lesions with [3H]TdR that entered the BCG lesions
showed lower [3H]TdR grain counts than did from the bloodstream during the 24 h before
MN in the 6-day lesions (Fig. 4), which may sug- the biopsy.
gest that DTH and CMI increased the rate of the At each biopsy time, the percentage of
local MN division. However, a more likely [3H]TdR-labeled MN in the 1-day trap was
explanation would be that by 2 and 5 days a about the same as the percentage in the BCG
larger number of young dividing (bone marrow- lesions (Fig. 5), which was consistent with a
derived) MN with lower grain counts apparently high turnover rate for MN within the lesions.
had infiltrated the 14-day lesions than had infil- The mean percentage of MN labeled with
trated the 6-day lesions. (MN apparently divide [3H]TdR in the traps had decreased from 68%
frequently in the bone marrow and less fre- to 5% by 12 days after the [3H]TdR injection
quently in the lesions.) The [3H]TdR grain (5), suggesting that the bone marrow’s supply of
counts in this experiment did not drop below [3H]TdR-labeled MN was mostly depleted by
50% of the initial grain count, which suggests this time.
that MN remaining in the lesions did not divide The [3H]TdR grain counts of MN in the 1-
more than once.A comparison of primary and day traps were also about the same as those
reinfection BCG lesions confirmed that the within the BCG lesions (5), which suggests that
presence of DTH reduces the [3H]TdR-grain the major reduction in MN grain counts
counts of MN in the lesions (Table 3). occurred in the bone marrow (and possibly the
blood) before the MN entered the lesions.
[3H]TdR-LABELED MN IN 1-DAY
“TUBERCULIN TRAPS” [3H]TdR-LABELED
Six rabbits were injected intradermally with MN IN THE BLOOD
BCG in multiple sites, and a single intravenous The [3H]TdR-labeled MN in the blood itself
[3H]TdR pulse was given 4 days later. At var- generally resembled those in the traps and in the
ious times thereafter, a lesion from each rabbit BCG lesions—both in the percentage of MN
was biopsied, and the [3H]TdR-labeled MN that were [3H]TdR labeled (Fig. 5) and in the

TABLE 3 [3H]TdR grain counts in [3H]TdR-positive -galactosidase-positive macrophages in primary and


reinfection dermal BCG lesionsa

Days after No. of [3H]TdR grains in macrophages that stained


[3H]TdR ⴙⴙ, ⴙⴙⴙ, and ⴙⴙⴙⴙ for -galactosidase in: P value
injection Primary BCG lesions BCG lesions of reinfection
5 12.9 ⫾ 1.8 9.7 ⫾ 0.5 0.056
8 11.9 ⫾ 1.6 6.7 ⫾ 0.3 0.006
a 3
[ H]TdR was injected intravenously 1 day before the dermal BCG lesions were begun. Note that macrophages activated for
-galactosidase had lower [3H]TdR grain counts in the early reinfection BCG lesions than in the early primary BCG lesions. In
reinfection BCG lesions, there is a more rapid infiltration of macrophages from the bone marrow (where their precursors are rapidly
dividing) and a more rapid activation of these macrophages (for -galactosidase) (see Fig.7).Adapted from reference 2.
186 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

FIGURE 5 Percentage of cells labeled with [3H]TdR (3HT in figure) (A) in blood and (B) in
1-day tuberculin traps and in BCG lesions at various times after a single intravenous pulse of
[3H]TdR when the BCG lesions were 4 days old.The percentages of [3H]TdR-labeled MN in
the blood, traps, and BCG lesions showed similar patterns, which indicates that [3H]TdR incor-
poration occurred before the MN entered the BCG lesions or soon afterward. It also indicates
that the drop in percentage of [3H]TdR-labeled MN in the lesions was at least in part due to a
drop in [3H]TdR-labeled MN that continued to enter the BCG lesions from the bloodstream.
These rabbits became tuberculin positive when the BCG lesions were 9 days old. Reproduced
with permission from reference 5.

reduction in their [3H]TdR grain counts over inflammatory lesions produced by the 1-day
time (5).At 12 days and thereafter, the percent- tuberculin traps.
age of [3H]TdR-labeled MN in the blood was In brief, these studies indicate that most of the
somewhat higher than it was in the tuberculin MN in BCG lesions recently came from the
traps (Fig. 5), and probably represented long- bone marrow via the bloodstream and that they
lived lymphocytes that did not migrate into the have a high turnover rate within these lesions.
10. MACROPHAGE TURNOVER, DIVISION,AND ACTIVATION 䡵 187

FIGURE 5 Continued.

RATES OF ACTIVATION OF MN reinfection BCG lesions were started when their


IN PRIMARY AND REINFECTION primary BCG lesions were 27 days of age [3]).
BCG LESIONS
Biopsies were made periodically thereafter, sec-
The activation of MN in BCG lesions was
tioned, stained for -galactosidase, and autoradi-
assessed histochemically with Pearson’s indolyl
ographed. The approximate time in which the
method (15, 21, 22) for the lysosomal enzyme
-galactosidase. The levels of this “marker” [3H]TdR-labeled macrophages entered the BCG
enzyme in macrophages increase as they become lesions was known from the time of the [3H]TdR
mature epithelioid cells (15, 16). Mature epithe- injection, and their degree of activation was indi-
lioid cells are capable of destroying tubercle cated by their -galactosidase activity.
bacilli (17, 23–25). Figure 6 shows the percentage of [3H]TdR-
Rabbits with either primary or reinfection labeled MN that stained ⫹⫹ to ⫹⫹⫹⫹ for
dermal BCG lesions were given an intravenous -galactosidase 5 and 8 days after a single
pulse of [3H]TdR 1 day before or 6, 14, or 27 [3H]TdR pulse (given when primary BCG
days after those lesions were begun (3). (The lesions were 6, 14, and 27 days of age). The
188 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

FIGURE 6 Percentage of recently entering MN (i.e., [3H]TdR-labeled MN) in primary der-


mal BCG lesions of various ages that were activated for -galactosidase. MN enter BCG lesions
in an unactivated state.They become activated at a rate that depends on the intensity of the BCG
stimulus and on the sensitivity of the host to this stimulus.
The highest percentage of entering MN became activated when [3H]TdR (3HT in figure)
was injected intravenously into rabbits with BCG lesions 14 days of age. At this time both the
DTH and CMI of the host and the antigenic products of the bacilli were at their height. Means
and their standard errors are shown. Reproduced with permission from reference 3.
10. MACROPHAGE TURNOVER, DIVISION,AND ACTIVATION 䡵 189

FIGURE 7 Percentage of [3H]TdR-labeled MN in early primary dermal BCG


lesions and in early lesions of reinfection. (3HT, [3H]TdR.) A higher percentage of
[3H]TdR-labeled MN staining ⫹⫹ and ⫹⫹⫹ to ⫹⫹⫹⫹ for -galactosidase was pre-
sent in reinfection lesions than in primary lesions, indicating that young (recently enter-
ing) MN (that incorporated [3H]TdR in the bone marrow) were activated more
rapidly when DTH and CMI were present. Reproduced with permission from refer-
ence 3.The -galactosidase substrates used for Fig. 6 and Fig. 7 came from different lots.

largest percentage of MN was activated for [3H]TdR-labeled macrophages that the pri-
-galactosidase in 14-day lesions, which was mary BCG lesions contained (Fig. 7), but by 14
the time when the size of the lesions and the days this difference between the reinfection and
amount of DTH and CMI were at their peaks. primary lesions was no longer present (3).
The [3H]TdR-labeled -galactosidase popula-
tion consisted almost entirely of macrophages, MACROPHAGE TURNOVER,
because few rabbit lymphocytes stain ⫹⫹ to ACTIVATION, AND DIVISION IN
⫹⫹⫹⫹ for -galactosidase. HEALING DERMAL BCG LESIONS
At 4 and 7 days of age, the BCG lesions of Rabbit dermal BCG lesions 27, 41, and 63 days
reinfection contained about twice the percent- of age were in their early, intermediate, and
age of ⫹⫹ to ⫹⫹⫹⫹ -galactosidase– late stages of healing, respectively. Five days
190 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

FIGURE 8 Total number of [3H]TdR-labeled MN (see Fig. 1) that entered early-healing and
late-healing dermal BCG lesions in 5 days. The number of [3H]TdR-labeled MN in these
lesions, 5 days after the [3H]TdR (3HT in figure) pulse, is proportional to the number of new
MN that entered during these 5 days.This figure shows that, even in the final stages of healing,
an appreciable number of new MN still enter tuberculous lesions.The difference between the
27-day experiment and the 41- and 63-day experiments was statistically significant. Reproduced
with permission from reference 4. (The 27-day experiment presented here was different from the
one presented in Fig. 1.Also, the lesion size here was calculated with a 0.52 factor, not the 0.20
factor used in Fig. 1 [see reference 3].)
10. MACROPHAGE TURNOVER, DIVISION,AND ACTIVATION 䡵 191

FIGURE 9 Percentage of recently entering MN (i.e., [3H]TdR-labeled MN) that were acti-
vated for -galactosidase in 5 and 8 days in dermal BCG lesions during early, intermediate, and
late stages of healing. (3HT, [3H]TdR.) In the late stages of healing, a lower percentage of
[3H]TdR-labeled MN was activated, apparently because most of the antigenic bacillary prod-
ucts had been destroyed. Reproduced with permission from reference 4. (The substrate for -
galactosidase used for Fig. 7 and 9 came from the same lot.)

after a [3H]TdR pulse, the percentage of lesions (Fig. 8), a reduced percentage disap-
[3H]TdR-labeled MN in the 41- and 63-day peared between 5 and 15 days after the
lesions was about two-thirds that in the 27-day [3H]TdR pulse (Fig. 3), and a reduced per-
lesions (data in reference 4), indicating that centage became activated (Fig. 9).These find-
MN turnover in these late lesions was still ings indicate that these almost healed lesions
appreciable. (The late lesions were about half the were still quite active and that some bacillary
size of 27-day lesions [4].) A reduced number growth and destruction probably occurred
of [3H]TdR-labeled MN entered these late within them.
192 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

FIGURE 10 Size of the tuberculin reactions (top) and percentage of the MN in these tuber-
culin reactions that stained ⫹⫹ and ⫹⫹⫹ for -galactosidase in primary and reinfection
dermal BCG lesions (bottom). (3HT, [3H]TdR.) Note that (i) Old Tuberculin (OT) itself acti-
vates considerable numbers of MN, and (ii) after DTH develops, the tuberculin reactions in
rabbits with primary BCG lesions are comparable to those in rabbits with reinfection BCG
lesions.
We do not know the amount of tuberculin and the amount of other bacillary compo-
nents within BCG lesions.Therefore, comparisons of the percentages of MN staining for
-galactosidase in these tuberculin reactions with those in the BCG lesions are meaning-
less. Reproduced with permission from reference 6.

MN ACTIVATION IN TUBERCULIN
REACTIONS AND IN LESIONS tuberculin reactions were biopsied 1 and 2 days
CAUSED BY NONSPECIFIC later. On both days rather similar results were
IRRITANTS obtained (6).
Tuberculin reactions were produced with Old Rabbits reinfected with BCG (and rabbits
Tuberculin in rabbits with primary BCG lesions with primary BCG infection after they became
and in those with reinfection BCG lesions.The tuberculin positive) showed large tuberculin
10. MACROPHAGE TURNOVER, DIVISION,AND ACTIVATION 䡵 193

reactions (Fig. 10). Such antigen-specific reac- produce none (26).With more necrosis, a greater
tions contained a greater percentage of both number of macrophages should enter and a
[3H]TdR-labeled and unlabeled -galactosi- greater number should die.
dase-positive MN than did the small nonspecific With greater macrophage turnover, more
reactions in unsensitized rabbits (Fig. 10). nonactivated macrophages would enter the
Vaccination with BCG had little or no effect tuberculous granulomas. Such macrophages
on the host’s reaction to nonspecific irritants. would permit tubercle bacilli to multiply within
Both tuberculin-positive BCG-vaccinated rab- them until they are controlled by the DTH-
bits and nonvaccinated control rabbits showed CMI process. In other words, the greater
similar degrees of cell infiltration and MN acti- turnover of macrophages in the lesions of rab-
vation in response to the intradermal injection bits and guinea pigs should allow more growth
of dilute phenol and carrageenan (6). In other and destruction of tubercle bacilli than in the
words, BCG vaccination enhanced the host lesions of mice where the bacilli appear to be
response to the tuberculin-like products of the more “dormant” (see chapter 6).
bacillus, but not to nonspecific irritants. These conclusions must be confirmed exper-
imentally, because macrophage turnover rates in
MACROPHAGE TURNOVER IN tuberculous lesions have been studied only in
MOUSE TUBERCULOUS rabbits.
GRANULOMAS
Tuberculous mice develop only low levels of INSIGHTS INTO DTH AND CMI
DTH.Therefore, mouse granulomas show little PROVIDED BY OUR KINETIC
or no necrosis, and the blood supply within STUDIES IN RABBITS
them remains intact (see chapter 15). Like rab- Our [3H]TdR-labeling studies clearly estab-
bits and guinea pigs, mice allow aerosolized vir- lished that in tuberculous lesions the presence of
ulent tubercle bacilli to grow logarithmically in DTH and CMI increased the turnover of MN
their lungs until DTH and CMI develop. Mice (mostly macrophages). Prior to these studies,
evidently use apoptosis to stop this logarithmic we knew that the presence of DTH increased
growth (see chapter 15).Then, a stationary stage MN accumulation, but we did not know much
occurs, in which the number of viable bacilli no about their rates of disappearance, except that
longer increases. some MN died in the caseous centers.Therefore,
When apoptosis kills macrophages, the sur- the rates of MN entry and disappearance from
rounding tissues are not killed and no inflam- tuberculous lesions were much higher than we
matory reaction occurs (26). Comparisons previously thought.
between the apoptosis found in mice and the Similarly, we knew that CMI increased the
caseous necrosis found in rabbits and guinea accumulation of activated macrophages (15, 16),
pigs have never been made. In fact, rabbits and but we did not know the rate at which enter-
guinea pigs may use both methods to kill ing macrophages became activated. Our
macrophages that contain more than a few [3H]TdR studies showed that macrophages
tubercle bacilli.The apoptosis method may be became activated within 5 to 8 days.
less efficient, because in C57BL/6 mice (a rather We found that relatively little local MN
resistant strain), virulent human-type tubercle (macrophage) division took place.The reduction
bacilli grow (during the logarithmic stage) to in their [3H]TdR grain counts seemed to be
higher titers than they do in rabbits and guinea mostly caused by the entry (into the lesions) of
pigs (see chapter 15). new macrophages with reduced grain counts.
Macrophage turnover in tuberculous granu- In sensitized hosts, the intradermal injection
lomas of rabbits and guinea pigs is probably of low concentrations of Old Tuberculin not
greater than that found in the granulomas of only caused cell infiltration (a classic DTH reac-
mice. Cells undergoing necrosis produce many tion) but also activated an appreciable number
chemotaxins (26), and cells undergoing apoptosis of macrophages in the infiltrating population (6).
194 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

These findings indicate that the tuberculin-like viable tuberculous granulation tissue of large or
products of the bacillus contribute to CMI, even small “arrested” tuberculous lesions, because
because activated macrophages can inhibit a continuing influx of nonactivated macrophages
and/or destroy ingested tubercle bacilli. occurs even in almost-healed BCG lesions.These
High concentrations of tuberculin-like bacil- nonactivated macrophages provide an opportu-
lary products are known to cause necrosis in sen- nity for renewed intracellular bacillary growth.
sitized hosts.To control the growth of tubercle Populations of tubercle bacilli may only be truly
bacilli in the host, such necrosis (and/or apop- dormant within solid caseous necrotic tissue
tosis) is apparently required to kill macrophages where macrophages cannot survive.
within which numerous bacilli are multiplying Therefore, solid caseous tissue would be the
(27–29).Any future tuberculosis vaccine that is best source of bacilli for the identification of
more effective than BCG in human popula- dormancy genes. Such studies were begun in
tions will most likely produce some degree of healing tuberculous lesions of rabbits infected by
DTH—if not to tuberculin itself, at least to aerosol with virulent human-type tubercle bacilli
other bacillary components that activate (H37Rv) (Y. C. Manabe et al., in preparation).
macrophages in low concentrations and cause Ten such genes were identified, but the location
necrosis in high concentrations. (within the lesions) of the bacilli containing
these genes remains to be identified. See chap-
ter 6 for more details.
RECAPITULATION OF OUR KINETIC
STUDIES ON MACROPHAGES IN
TUBERCULOUS LESIONS PRODUCED
BY BCG IN RABBITS REFERENCES
In tuberculous lesions produced by BCG in rab- 1. Shima, K., A. M. Dannenberg, Jr., M. Ando,
bits (and probably those in guinea pigs and S. Chandrasekhar, J. A. Seluzicki, and J. I.
humans), macrophages have a continual turnover. Fabrikant. 1972. Macrophage accumulation, divi-
sion, maturation, and digestive and microbicidal
Numerous macrophages enter and many die or capacities in tuberculous lesions. I. Studies involv-
leave via lymphatics. However, despite this high ing their incorporation of tritiated thymidine and
turnover, many macrophages accumulate in the their content of lysosomal enzymes and bacilli.
lesions and become activated. Am. J. Pathol. 67:159–180.
In an early lesion (produced by BCG), local 2. Ando, M., A. M. Dannenberg, Jr., and
K. Shima. 1972. Macrophage accumulation, divi-
macrophage activation does not develop rapidly sion, maturation and digestive and microbicidal
enough to be effective, but after DTH and capacities in tuberculous lesions. II. Rate at which
CMI develop, the rate of macrophage activation mononuclear cells enter and divide in primary
increases considerably, which enables the host BCG lesions and those of reinfection. J. Immunol.
to control the BCG infection. Activated 109:8–19.
3. Dannenberg, A. M., Jr., M. Ando, and
macrophages ingest and inhibit the bacilli
K. Shima. 1972. Macrophage accumulation, divi-
released from killed macrophages. sion, maturation and digestive and microbicidal
If numerous activated macrophages are pre- capacities in tuberculous lesions. III.The turnover
sent, tuberculous lesions will regress and even- of macrophages and its relation to their activation
tually heal. However, even in almost-healed and antimicrobial immunity in primary BCG
lesions and those of reinfection. J. Immunol.
lesions, nonactivated macrophages continuously
109:1109–1121.
enter and may ingest bacilli escaping from the 4. Ando, M., and A. M. Dannenberg, Jr. 1972.
edges of the caseous center.Therefore, a few of Macrophage accumulation, division, maturation
the bacilli may undergo many cycles of intra- and digestive and microbicidal capacities in tuber-
cellular multiplication before the number of culous lesions. IV. Macrophage turnover, lysosomal
activated macrophages becomes sufficient to enzymes and division in healing lesions. Lab. Inves-
tig. 27:466–472.
stop the progression of the disease. 5. Tsuda, T., A. M. Dannenberg, Jr., M. Ando,
In rabbits, and most likely in humans, the pop- H.Abbey, and A. R. Corrin. 1976. Mononuclear
ulation of tubercle bacilli is not dormant in the cell turnover in chronic inflammation. Studies on
10. MACROPHAGE TURNOVER, DIVISION,AND ACTIVATION 䡵 195

tritiated-thymidine-labeled cells in blood, tuberculin 17. Ando, M., A. M. Dannenberg, Jr., M. Sugi-
traps and dermal BCG lesions of rabbits. Am. moto, and B. S. Tepper. 1977. Histochemical
J. Pathol. 83:255–268. studies relating the activation of macrophages to the
6. Ando, M. 1973. Macrophage activation in tuber- intracellular destruction of tubercle bacilli. Am. J.
culin reactions of rabbits with primary BCG infec- Pathol. 86:623–634.
tion and reinfection. J. Reticuloendothel. Soc. 14:132– 18. Cleaver, J. E. 1967. Thymidine Metabolism and
145. Cell Kinetics. Frontiers of Biology, vol. 6. North-
7. Dannenberg, A. M., Jr., M. Sugimoto, L. P. Holland Publishing Company, Amsterdam, The
Fay, and A. L. Massaquoi. 1976. In vivo labeling Netherlands.
effectiveness of tritiated thymidine of high and low 19. Spector,W. G. 1969.The granulomatous inflam-
specific activities in rabbits. Radiat. Res. 67:98–103. matory exudate. Int. Rev. Exp. Pathol. 8:1–55.
8. Ando, M.,A. M. Dannenberg, Jr., E. Courtade, 20. Ryan, G. B., and W. G. Spector. 1970.
and K. Shima. 1976.Turnover of tritiated-thymi- Macrophage turnover in inflamed connective tissue.
dine-labeled mononuclear cells in tuberculous Proc. R. Soc. (Biol.) 175:269–292.
lesions of rabbits.A comparison of primary dermal 21. Pearson, B., P. L. Wolf, and J.Vazquez. 1963.
BCG lesions and those of reinfection. Proc. Soc. A comparative study of a series of new indolyl
Exp. Biol. Med. 151:491–494. compounds to localize -galactosidase in tissues.
9. Chandrasekhar, S., K. Shima, A. M. Dannen- Lab. Investig. 12:1249–1259.
berg, Jr., T. Kambara, J. I. Fabrikant, and 22. Yarborough, D. J., O. T. Meyer, A. M. Dan-
W. G. Roessler. 1971. Radiation, infection and nenberg, Jr., and B. Pearson. 1967. Histo-
macrophage function. IV.The effect of radiation on chemistry of macrophage hydrolases. III. Studies
the proliferative abilities of mononuclear phagocytes on -galactosidase, -glucuronidase and aminopep-
in tuberculous lesions of rabbits. Infect. Immun. tidase with indolyl and naphthyl substrates. J. Retic-
3:254–259. uloendothel. Soc. 4:390–408.
10. Dannenberg, A. M., Jr. 2003. Macrophage 23. Lurie, M. B., P. Zappasodi, E. Cardona-Lynch,
turnover, division and activation within developing, and A. M. Dannenberg, Jr. 1952.The response
peak and “healed” tuberculous lesions produced in to the intracutaneous inoculation of BCG as an
rabbits by BCG. Tuberculosis 83:251–260. index of native resistance to tuberculosis. J. Immunol.
11. van Furth, R. 1970. Origins and kinetics of 68:369–387.
monocytes and macrophages. Semin. Hematol. 24. Lurie, M. B. 1932. The correlation between the
7:125–141. histological changes and the fate of living tubercle
12. Virolainen, M. 1968. Hematopoietic origin of bacilli in the organs of tuberculous rabbits. J. Exp.
macrophages as studied by chromosome markers in Med. 55:31–54.
mice. J. Exp. Med. 127:943–951. 25. Lurie, M. B. 1964. Resistance to Tuberculosis: Exper-
13. Volkman, A. 1970. The origin and fate of the imental Studies in Native and Acquired Defensive Mech-
monocyte. Ser. Haematol. 3:62–92. anisms. Harvard University Press, Cambridge, Mass.
14. Fabrikant, J. I., C. L. Wisseman III, and M. 26. Majno, G., and I. Joris. 2004. Cells,Tissues, and
J.Vitak. 1969.The kinetics of cellular prolifera- Disease. Principles of General Pathology, 2nd ed. Oxford
tion in normal and malignant tissues. II. An in University Press, New York, N.Y.
vitro method for incorporation of tritiated thymi- 27. Dannenberg, A. M., Jr. 1993. Immunopatho-
dine in human tissues. Radiology 92:1309– genesis of pulmonary tuberculosis. Hosp. Pract.
1320. 28:33–40. (Off. ed. 51–58.)
15. Dannenberg, A. M., Jr., O. T. Meyer, J. R. 28. Dannenberg,A. M., Jr. 2001. Pathogenesis of pul-
Esterly, and T. Kambara. 1968.The local nature monary Mycobacterium bovis infection: basic princi-
of immunity in tuberculosis, illustrated histochem- ples established by the rabbit model. Tuberculosis
ically in dermal BCG lesions. J. Immunol. 100:931– 81:87–96.
941. 29. Dannenberg, A. M., Jr., and F. M. Collins.
16. Dannenberg, A. M., Jr. 1968. Cellular hyper- 2001. Progressive pulmonary tuberculosis is not
sensitivity and cellular immunity in the patho- due to increasing numbers of viable bacilli in rab-
genesis of tuberculosis: specificity, systemic and local bits, mice and guinea pigs, but is due to a contin-
nature, and associated macrophage enzymes. Bacte- uous host response to mycobacterial products. Tuber-
riol. Rev. 32:85–102. culosis 81:229–242.
11
LURIE’S PULMONARY
TUBERCLE-COUNT METHOD

Tubercle development in humans [197]


Lurie’s tubercle-count method [197]
Advantages of the rabbit model of tuberculosis [199]
Role of alveolar macrophages in the establishment of grossly visible primary
pulmonary tubercles [200]
Tubercle counts to determine bacillary virulence [200]
Tubercle counts to determine the genetic resistance of the host [204]
Tubercle counts to determine the degree of immunization produced by a
given vaccine [204]
A comparison of tubercle counts and “the ratios” [204]
Effect of inhaled dosage on the ratio [206]
Variability of tubercle counts and ratios [206]
Comparison of aerosol mycobacterial infection with intravenous
infection [206]
Pulmonary tubercle counts in guinea pigs and mice [207]
Tubercle counting in mice—methodology [210]

Abstract. Lurie’s tubercle-count method consists of counting the number of grossly vis-
ible primary pulmonary tubercles, present 5 weeks after an aerosol infection of rabbits with
virulent human-type tubercle bacilli. It is a quantitative measure of clinically apparent dis-
ease.At 5 weeks, the grossly visible primary tubercles are easily recognized, and many micro-
scopic tubercles have regressed. Since human-type tubercle bacilli are not fully virulent
for rabbits, the pulmonary-count method has a sensitivity that is not possible with fully
virulent strains.
The number of grossly visible pulmonary tubercles produced by human-type bacilli
decreases (i) when rabbits are infected with bacilli of reduced virulence, (ii) when rabbits
of high genetic (innate) resistance are used, and (iii) when rabbits are effectively immu-
nized, so that they can rapidly activate macrophages and stop the development of early tuber-
cles while they are still microscopic in size.Therefore, the pulmonary tubercle-count method
can be used to assess (i) bacillary virulence, (ii) the genetic resistance of the host, and (iii)
the efficacy of vaccines for tuberculosis.

In rabbits, the number of primary tubercles bits, and remains to be developed for mice and
present in the lung following the inhalation of guinea pigs.
virulent human-type tubercle bacilli is a precise The majority of virulent human-type tuber-
measure of both the ability of the bacillus to cle bacilli inhaled by rabbits (and probably by
produce clinically recognizable disease and the humans) are immediately destroyed or inhibited
ability of the host to prevent such clinical dis- by pulmonary alveolar macrophages (2) (see
ease. To date, tubercle counting in rabbits has chapter 2). Also, the majority of the bacilli
been used in references 1 through 13, as well as inhaled by rabbits that do manage to grow (and
in Lurie’s studies on the effects of adrenal and form microscopic tubercles) are inhibited after
thyroid hormones (described in chapters 16 the host acquires immunity in 2 or 3 weeks. In
and 17).The method has been used only in rab- other words, the visible pulmonary tubercles

196
11. LURIE’S PULMONARY TUBERCLE-COUNT METHOD 䡵 197

(found at 5 weeks in rabbits) are produced only ately destroyed (or inhibited) without converting
by bacilli that have survived the forces of both the tuberculin skin test. However, a small pro-
native and acquired host resistance. portion of the inhaled bacilli may multiply in a
When fully virulent bovine-type tubercle weakly activated alveolar macrophage and even-
bacilli are inhaled by rabbits, differences in host tually cause a single primary tubercle.The host
resistance will not be readily apparent, because then becomes tuberculin positive.The exact num-
one grossly visible pulmonary tubercle is pro- ber of inhaled bacilli that produces a detectable
duced for every unit (of 1 to 3 bacilli) that is tubercle is not known for humans.A mean of 20
inhaled into the alveoli by either resistant or sus- to 200 is a reasonable estimate. Most of these early
ceptible hosts (2, 13, 14) (Table 1). However, tubercles never progress beyond a millimeter or
when human-type tubercle bacilli (which are so in size (see Lindgren’s studies described at the
only semivirulent for rabbits) are inhaled, there end of chapter 3).A few, however, may progress
is a wide range in the number of bacilli required to clinically apparent tuberculosis.
to produce one grossly visible tubercle.There- Human tuberculosis (and tuberculosis in rab-
fore, small differences in host resistance or in bits and guinea pigs exposed naturally to low
bacillary virulence are easily recognized. concentrations of tubercle bacilli in room air)
To develop the tubercle-count method for begins as only one primary pulmonary lesion
mice and guinea pigs, a semivirulent strain of (see chapter 12). However, multiple grossly vis-
tubercle bacilli must be used. Fully virulent ible primary pulmonary tubercles are usually
human strains of tubercle bacilli in these hosts produced in laboratory animals following an
(like fully virulent bovine strains in the rabbit) aerosol infection. Each of these tubercles devel-
produce one grossly visible tubercle for every ops whenever (i) an alveolar macrophage allows
unit inhaled into the alveoli, and therefore would intracellular bacillary multiplication and (ii) a
not easily detect variations in host resistance by weak acquired immunity does not prevent the
the tubercle-count method (see chapter 15). resulting microscopic tubercle from reaching
visible size. Many tubercles in multiple sites
TUBERCLE DEVELOPMENT more accurately reflects the overall resistance of
IN HUMANS a given host better than does only one tubercle.
Human pulmonary tuberculosis begins as a sin-
gle lesion, because bacilli are inhaled gradually, LURIE’S TUBERCLE-COUNT
usually over periods of months. Most of the METHOD
inhaled bacillary units seem to be ingested by With Lurie’s tubercle-count method, rabbits are
activated alveolar macrophages and immedi- infected by aerosol with a known number of

TABLE 1 The number of inhaled bovine-type tubercle bacilli (Ravenel) required to produce one primary
pulmonary tubercle in Lurie’s inbred rabbitsa
No. of bacilli
Rabbit No. of rabbits No. of isolated required to form
Resistance bacilli estimated as
strain exposed one visible tubercle
inhaled (range) (the ratio)
CaC Susceptible 6 40–462 2.8 ⫾ 0.2
AD Intermediate 22 90–688 3.4 ⫾ 0.3
III Resistant 6 216–306 2.9 ⫾ 0.3
C Susceptible 4 29–546 3.3 ⫾ 0.7
a
Note that 3 small units of inhaled fully virulent bovine-type tubercle bacilli (Ravenel) generate one grossly visible primary
pulmonary tubercle in every rabbit strain, despite differences in their genetic resistance. Means and their standard errors are listed.
The mean ratios were calculated from the ratios of individual rabbits by the Lurie method (see text and Tables 2, 3, and 5).Adapted
from references 2 and 13.
198 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

virulent human-type bacilli—Lurie used H37Rv 10–12). Similar methods were used by Lurie to
(1, 3, 15). Five weeks later, the animals are euth- determine the inhaled bacillary dose (1, 14). He
anized. Then, the grossly visible primary pul- calculated the inhaled dose for each rabbit from
monary tubercles (Fig. 1 and 2) are counted and Kleiber’s formula based on the weight of each
their diameters are measured, either at the time rabbit (1, 14). We calculated the inhaled dose
of necropsy (4, 12) or in formalin-fixed lungs at from the volume of air breathed by each rabbit
a later time (1, 2, 7–9, 15). in a plethysmograph (3, 4, 10–12).When all of the
In our experiments, an aliquot of the air in the exposed rabbits were of similar size, the variations
exposure chamber was collected by means of an in the number of bacilli inhaled by each rabbit
impinger into a 1:10 dilution (in 0.9% NaCl) of were insignificant when compared to the much
Oleic Albumin Complex (Dubos) (Becton Dick- larger variations in the number of tubercles caused
inson, Co., Sparks, Md.) containing an antifoam by differences in the innate and acquired resis-
agent.Then, the impinger fluids were cultured (4, tance of each rabbit.

FIGURE 1 Formalin-fixed lungs of a commercial rabbit that inhaled


about 33,000 virulent human-type tubercle bacilli (H37Rv) 5 weeks pre-
viously. Upon dissection, these lungs contained 131 grossly visible pri-
mary tubercles, with no apparent secondary tubercles.The ratio of the
number of bacilli estimated as inhaled to the number of grossly visible
primary tubercles produced was 250. Effective BCG (and other vaccines
for tuberculosis) should increase this ratio at least fivefold (9, 12). Small
areas of caseous necrosis are visible in many of the tubercles. On the left,
this photograph shows the ventral surface of the right upper, middle, and
azygous lobes; on the right, the entire left lung (upper and lower lobes)
is shown.The right lower lobe had been removed for culture.This right
lower lobe contained 23 grossly visible tubercles and 1.35 ⫻ 105 cultur-
able tubercle bacilli. Magnification, ⫻1.1. Reproduced with permission
from reference 3.
11. LURIE’S PULMONARY TUBERCLE-COUNT METHOD 䡵 199

FIGURE 2 A tissue section of a primary lesion similar to the lesions shown in Fig. 1. From
left to right are (i) one of the small sites of caseous necrosis, (ii) a surrounding area of large epithe-
lioid macrophages, and (iii) an outside area that is densely infiltrated by smaller macrophages, lym-
phocytes, and plasma cells.Azure-eosin stain. Magnification, ⫻350. Reproduced with permission
from reference 3.

ADVANTAGES OF THE RABBIT Cavities never form in mice and in Lurie’s


MODEL OF TUBERCULOSIS inbred susceptible rabbits, and they rarely form
Most scientists in tuberculosis research at the in guinea pigs.The extracellular multiplication
present time evaluate both bacillary virulence of tubercle bacilli in cavities is not affected by
and vaccine efficacy in mice, and a few scientists the native or acquired resistance of the host.
evaluate them in guinea pigs.Aliquots of infected Tuberculous humans usually have strong
lungs and spleens are usually cultured, and the tuberculin sensitivity and considerable caseation,
time of death is usually recorded. Although and rabbits have moderate amounts of each,
bacillary titers in the lungs and spleen reflect especially if virulent bovine-type bacilli are used
both the virulence of the bacilli and the native to infect the animals. Caseation reduces the
and acquired resistance of the host (just as tuber- number of viable tubercle bacilli that can be cul-
cle counts do), certain aspects of host-parasite tured from lungs and other organs (see chapters
interactions are not taken into account by such 2 and 5).Tuberculous guinea pigs usually have
measurements. good delayed-type hypersensitivity (DTH) and
For example, in humans and in rabbits, the much caseation, and their lesions contain rela-
bacilli may multiply extracellularly to tremen- tively few bacilli (16) (see chapter 15). Tuber-
dous numbers in liquefied caseum, especially in culous mice usually have weak DTH and little
the liquefied caseum of the inner part of a cav- or no caseation, and their lesions contain more
ity wall. Cavities readily form in commercial bacilli (16) (see chapter 15).Yet, despite lower
rabbits and in Lurie’s inbred resistant rabbits. numbers of bacilli present, the disease often
200 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

progresses more rapidly in guinea pigs than it intestinal mucosa are rather resistant to infection
does in mice. In other words, the number of cul- by tubercle bacilli (1).
turable bacilli in the lungs of different species Alveolar macrophages are the first cells of the
does not always parallel their native and acquired host to ingest the inhaled tubercle bacilli that enter
resistance. the alveoli of the lung.The alveolar macrophage
Tubercle counting in rabbits has additional population is highly activated (20–22), apparently
advantages over tubercle counting in mice and because of the continuous ingestion of dust par-
guinea pigs. In New Zealand White rabbits ticles and microorganisms.A “good meal” in the
inhaling virulent human-type bacilli (H37Rv), “stomach”of macrophages upregulates their diges-
only 1 in 500 to 1,000 inhaled bacillary units tive and probably their oxidative enzymes (23).The
produces a grossly visible tubercle at 5 weeks alveolar macrophages of Lurie’s resistant rabbits
(see below and chapter 14).As stated above, the seemed to be more highly activated than those of
vast majority of these bacilli are destroyed or his susceptible rabbits (1, 24–29).
inhibited by the alveolar macrophages and by In rabbits, and probably in humans, most
the developing immune response. Therefore, inhaled virulent human-type bacilli are soon
small changes in host resistance can be readily destroyed or inhibited by activated alveolar
detected. macrophages and do not multiply. If no bacillary
Tubercle counting in mice and guinea pigs multiplication occurs, the antigenic load remains
is not as sensitive to such host defenses. In these below that required to call forth an immune
hosts, as in rabbits inhaling fully virulent response, and the host remains tuberculin neg-
bovine-type bacilli (Table 1), only about 3 ative. Following the inhalation of virulent
inhaled bacillary units are required to produce human-type tubercle bacilli, an initial micro-
a grossly visible tubercle. Therefore, in mice scopic tubercle becomes established only after a
and guinea pigs, the tubercle-count method strong bacillus is ingested by a relatively weak
will not as easily detect small changes in bacil- alveolar macrophage (1, 26–28): weak tubercle
lary virulence and host resistance. Tubercle bacilli are usually destroyed by the alveolar
counting in mice and guinea pigs is discussed macrophages in rabbits and probably in humans.
further at the end of this chapter. A full discussion of the role of alveolar
Because of the differences in caseation and macrophages in the establishment of tuberculosis
cavity formation among rabbits, mice, and is presented in chapter 12.
guinea pigs, we recommend that rabbits be
included in experiments assessing (i) the viru-
TUBERCLE COUNTS TO DETERMINE
lence of the infecting human-type tubercle BACILLARY VIRULENCE
bacilli, (ii) the genetic resistance of the host,
and (iii) the efficacy of vaccines. Overview
Among the laboratory animals commonly used
to study tuberculosis, rabbits are unique.
ROLE OF ALVEOLAR MACROPHAGES
IN THE ESTABLISHMENT OF Immunocompetent rabbits usually recover when
GROSSLY VISIBLE PRIMARY infected with fully virulent human-type tuber-
PULMONARY TUBERCLES cle bacilli (1,15, 30), and usually die when
Only one-third of the bacillary units of 1 to 3 infected with fully virulent bovine-type tuber-
bacilli remain suspended in the airstream and cle bacilli (1, 31). In fact, in the early part of the
reach the alveolar spaces (1, 14, 17–19). Units of 20th century, rabbits were used to determine
more than about 3 bacilli are so heavy that they whether the bovine or the human type of tuber-
do not remain in the airstream, but impinge cle bacillus had infected a given patient.
upon the walls of the bronchial tree, often at its Lurie’s tubercle-count method in rabbits can
bifurcations. They are then carried by the be used to assess the virulence of mycobacterial
bronchial mucociliary escalator to the pharynx isolates from human cases (4), and to measure the
and are swallowed. Both the bronchial and virulence of human-type bacilli that were altered
11. LURIE’S PULMONARY TUBERCLE-COUNT METHOD 䡵 201

by molecular biological techniques.The method ratios with those presented in Table 2.) There-
provided a quantitative measure of virulence, fore, variations in virulence of human-type
namely, “the ratio,” which is the number of tubercle bacilli can have profound effects on
inhaled bacillary units (of 1 to 3 bacilli) divided the number of primary pulmonary tubercles
by the number of grossly visible primary tuber- generated.*
cles present 5 weeks after infection (1, 3), i.e., the Note that the greater the virulence of the
number of inhaled bacilli that produces one inhaled bacillary strain, the smaller the difference
primary pulmonary tubercle. in tubercle counts between the resistant and
susceptible inbred rabbits. In other words,
Virulence of Bovine-Type Tubercle genetic (and immunological) differences are
Bacilli most apparent in the tubercle counts when
With fully virulent bovine-type bacilli (Ravenel aerosolized virulent human-type tubercle bacilli
S), about 3 bacillary units must be inhaled to of somewhat reduced virulence are used to
produce one grossly visible primary pulmonary infect the rabbits.
tubercle (a ratio of 3) (1, 2, 13, 14) (Table 1). This principle was recently confirmed with
With bovine-type bacilli of somewhat reduced Thorbecke inbred rabbits. which have reduced
virulence, this “ratio” varied from 18 to 107 resistance to tuberculosis (see Tables 3 and 5 in
(see Table 3 in chapter 4).The large number of chapter 14).The more virulent Erdman strain of
inhaled bovine-type bacilli seemed to decrease human-type tubercle bacilli did not produce
the host’s ability to prevent grossly visible pri- statistically different tubercle counts when Thor-
mary tubercles (10, 11). (However, a large num- becke rabbits were compared to outbred rabbits,
ber of inhaled human-type bacilli seemed to but the less virulent H37Rv strain did so (6).
increase the host’s ability to prevent grossly vis-
ible primary tubercles [5].) Virulence of a Clinical Mycobacterium
tuberculosis Isolate
Virulence of Human-Type Tubercle In 1994 and 1995, a miniepidemic of tubercu-
Bacilli (H37Rv) losis occurred at the Kentucky-Tennessee bor-
With H37Rv, Lurie’s resistant strain III rabbits der where the Oshkosh clothing factory is
required an average of 1,065 inhaled bacillary located. The human-type bacillary strain that
units to produce one visible tubercle, but his sus- caused this epidemic was designated CDC1551
ceptible strain C rabbits required only an aver- by the Centers for Disease Control and Pre-
age of 70 bacillary units to do so, i.e., ratios of vention in Atlanta, Ga. Because it caused many
1,065 and 70, respectively (Table 2) (1, 2).With
H37Rv in commercial New Zealand White *Part of the apparent increased virulence of H37Rv
rabbits, we obtained ratios ranging from 765 to between 1956 and 1965 may have been due to a reduced
1,946 (Table 3) (12).These ratios were similar to ability to grow in vitro after aerosolization. If fewer
bacilli were culturable from the aerosol, the ratios (bacilli
those in Lurie’s resistant strain III rabbits. Ratios inhaled divided by number of tubercles produced)
for Lurie’s other inbred rabbit strains are listed would be decreased—even though the ability of these
in Table 2. bacilli to grow in the rabbit host (i.e., virulence) was
By 1965, Lurie evidently had an even more unchanged. About 1958, Lurie moved his laboratory
virulent H37Rv strain (8).The ratios of four of from the University of Pennsylvania’s old Henry Phipps
Institute building on 7th Street to new quarters on the
his inbred rabbit strains were IIIR (TR) ⫽ 182 main university campus on 36th Street.When incubated
⫾ 36, AD ⫽ 56 ⫾ 4, and C ⫽ 48 ⫾ 8. (IIIR in the new location in an electric incubator, the bacilli
was a more resistant substrain of III rabbits.) In obtained after aerosolization may not have been able to
other words, in each of these inbred rabbit start as many colonies (on Lowenstein’s agar slants) as
groups, this more virulent H37Rv strain pro- they did in the gas-heated incubator in the old location.
Gas burners release CO2 into the air of the incubator.
duced more tubercles per inhaled dose than When Lurie increased the CO2 (using a compressed-
did the H37Rv bacilli used in all of his previ- CO2 cylinder), many more colonies developed from the
ously reported experiments. (Compare these same inoculum.
202
TABLE 2 The relative resistance of inbred rabbit strains as determined by their response to quantitative inhalation of human-type bacilli (H37Rv)a


E G
C No. of bacilli F Average diameter
A B D Variability in of tubercles (mm)

PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS


No. of bacilli required to form
Rabbit strain No. of rabbits estimated as Range of tubercles one visible tubercle column E in and number of
and resistance evaluated inhaled (range) generated (the ratio) percent rabbits in which
(Lurie method) SE/ratio ⫻ 100 they were
measured
C (suscept.) 30 1,100–45,000 14–1,500 70 ⫾ 14 20% 4.0 ⫾ 0.27 (11)
(14/70)
FC (suscept.)b 45 700–54,000 8–1,400 79 ⫾ 9 11% 3.9 ⫾ 0.15 (33)
(9/79)
CaC (suscept.) 7 9,200–15,000 100–290 97 ⫾ 12 12% 3.9 ⫾ 0.20 (7)
(12/97)
A (intermed.) 11 14,900–21,000 51–230 230 ⫾ 24 10% 3.8 ⫾ 0.12 (11)
(24/230)
AD (intermed.) 12 25,000–35,000 25–370 362 ⫾ 103 28% 3.2 ⫾ 0.06 (12)
(103/362)
III (resistant)b 71 570–40,000 0–150 1,065 ⫾ 138 13% 3.1 ⫾ 0.11 (52)
(138/1,065)
FC (suscept.) 12 700–7,200 8–200 107 ⫾ 20 19%
(20/107)
Ca (suscept.) 9 520–3,100 3–440 49 ⫾ 19 39%
(19/49)
III (resistant) 8 570–4,300 0–11 ⬎642 ⫾ 107
a
Lurie determined the resistance of each rabbit by calculating the number of inhaled human-type bacilli required to form one grossly visible tubercle (the ratio), i.e., the number of
bacillary units inhaled by each rabbit divided by the number of visible tubercles produced at 5 weeks after infection. He then averaged the individual ratios to obtain the ratios listed above
for each inbred rabbit strain.
The average size of these tubercles also reflected host resistance; e.g., the tubercles of resistant strain III were smaller on average than those of susceptible strains C, FC, and CaC.
Data for strains C, FC,A,AD, and III are from reference 2; data for strain CaC are from reference 1 (p. 241); data for strains Ca and the second FC and III are from reference 15. Means
and their standard errors are listed.
b
The large number of rabbits in the III and FC groups (71 and 45, respectively) would, of course, have smaller standard errors than the 11 strain III and 13 strain FC rabbits listed in
Table 5.
11. LURIE’S PULMONARY TUBERCLE-COUNT METHOD 䡵 203

TABLE 3 Number of inhaled virulent human-type bacilli (H37Rv) required to produce one grossly visible
primary pulmonary tubercle (the ratio) in commercial outbred New Zealand White rabbitsa
A. Experiment no. (no. of rabbits) I (6) II (6) III (6) IV (8)
B. Mean no. of viable bacilli 370,000 60,000 230,000 38,000
inhaled by each rabbit
C. Mean no. of visible tubercles 278 ⫾ 65 81 ⫾ 21 217 ⫾ 53 65 ⫾ 15
in lungs of each rabbit
D. Mean diameter of these tubercles 2.3 ⫾ 0.1 1.6 ⫾ 0.2 1.6 ⫾ 0.2 2.2 ⫾ 0.2
E. No. of bacilli inhaled by each 1,946 ⫾ 622 1,058 ⫾ 270 1,535 ⫾ 432 765 ⫾ 123
rabbit divided by the number of
tubercles produced by each rabbit:
average of individual ratios
(Lurie method)b
F.Average no. of inhaled bacilli 1,330 740 1060 585
divided by average number of
tubercles produced
(ratios of averages: B/C)
G.Variability in row E in percent: 32% (622/1,946) 26% (270/1,058) 28% (432/1,535) 16% (123/765)
SE/ratio ⫻ 100 (Lurie method)c
H.Variability in row C in percent: 23% (65/278) 26% (21/81) 24% (53/217) 23% (15/65)
SE/no. of tubercles ⫻ 100c
a
Note that the ratios tended to be higher when more bacilli were inhaled, perhaps because acquired immunity developed sooner.
Tubercle size, however, seemed independent of the bacillary dose inhaled. Means and their standard errors are listed. Data are from
reference 12.
b
In row E, the standard errors were calculated from the ratios of each rabbit in a given experiment.
c
Unexpectedly, the standard errors as percent of the means found in these outbred rabbits were not significantly larger than
those found in Lurie’s inbred rabbits (see Table 5), indicating that Lurie’s inbreeding did not appreciably reduce the variability within
each rabbit strain.

clinical cases after short exposure times, Virulence for Rabbits of Erdman,
CDC1551 was thought to be more virulent H37Rv, and CDC1551 (Oshkosh)
than most clinical M. tuberculosis isolates. Strains of Human-Type Bacilli
Using Lurie’s tubercle-count method, we Erdman was more virulent for rabbits than
found that, on the contrary, CDC1551 produced H37Rv, and H37Rv was more virulent than
about the same number of primary tubercles as CDC1551 (5). Genetic analysis of these strains
did the laboratory strain H37Rv in commercially identified some of the factors responsible for the
available New Zealand White rabbits (4) (Table differences (5).
4); and, unexpectedly, the tubercles produced by
CDC1551 were significantly smaller and con- Effect of Freezing and Thawing on
tained fewer viable bacilli (4) (Table 4).There- Infectivity
fore, the rabbits controlled the progress of the dis- In rabbits, log-phase human-type tubercle bacilli
ease produced by CDC1551 better than they produced more grossly visible tubercles than a
controlled that produced by H37Rv (4). This similar inhaled dose of viable frozen-and-
finding was supported by a careful evaluation of thawed bacilli of the same strain (5).The frozen-
CDC1551 in mice (32). Other studies in mice and-thawed bacilli were probably more easily
also showed that CDC1551 was not more vir- destroyed by pulmonary alveolar macrophages.
ulent than other strains of M. tuberculosis, includ- This finding supports the concept that the phe-
ing Erdman (33) and H37Rv (33, 34) and sev- notypic state of a viable tubercle bacillus affects
eral clinical isolates (35). its ability to initiate the disease: a dormant
204 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

TABLE 4 Virulence of two strains of human-type tubercle bacilli—H37Rv and CDC1551 (Oshkosh)—
assessed in rabbits by Lurie’s tubercle-count methoda

Strain of aerosolized bacilli No. of visible tubercles produced Average diameter of


5 weeks after aerosol infection tubercles (mm)
H37Rv 8.0 ⫾ 4.0 1.9 ⫾ 0.1
CDC1551 (Oshkosh) 5.2 ⫾ 1.5 1.2 ⫾ 0.1
a
About 8,000 viable units of 1 to 3 bacilli were inhaled by each commercial outbred rabbit.The number of primary tubercles
produced by the strain CDC1551 was similar to that produced by H37Rv, but the CDC1551 tubercles were smaller (P ⫽ 0.007)
and contained fewer bacilli when cultured. Means and their standard errors are shown. Data are from reference 4.

bacillus is probably less infectious than an resistance is recalled only after the inhaled bacil-
actively multiplying bacillus, possibly because its lus has multiplied and produced enough anti-
metabolism is inactive at the time of inhalation. genic material to react with the memory lym-
phocytes entering the developing tubercle.These
TUBERCLE COUNTS TO DETERMINE antigen-specific lymphocytes rapidly accumulate
THE GENETIC RESISTANCE OF THE in the lesions and produce gamma interferon and
HOST
other cytokines that rapidly activate nearby
Lurie’s tubercle-count method is useful in assess-
macrophages and other cells.Therefore, tubercle
ing the innate (genetic) resistance of the host (1,
bacilli are killed or inhibited sooner in the immu-
2, 7) (Table 2).About 1,000 bacillary units of 1
nized host, so fewer tubercles reach grossly visi-
to 3 H37Rv bacilli were required to produce
ble size.Chapter 23 describes experiments that use
one such tubercle in the resistant strain III rab-
the tubercle-count method to assess vaccine effi-
bits, whereas only about 50 to 100 such units
cacy.Tables 5 and 6 summarize the experiments
were required in the highly susceptible strains C,
made to date with various live vaccines in Lurie’s
FC, Ca, and CaC (1, 2) (Table 2). In addition,
inbred rabbits and in commercial outbred rabbits.
the average diameters of the tubercles in rabbits
of resistant strain III were smaller than those in
the susceptible strains C, FC, and CaC (1, 2) A COMPARISON OF TUBERCLE
COUNTS AND “THE RATIOS”
(Table 2). Among the intermediate strains, A
In general, larger rabbits inhale more bacilli and
had relatively large tubercles, and AD had rela-
therefore have higher tubercle counts than
tively small tubercles (Table 2). Chapter 14
smaller rabbits do, if both have similar resis-
describes the use of the tubercle-count method
tance to tuberculosis. Because the supply of
to determine the resistance of crossbred resistant
Lurie’s inbred rabbits was limited, both small and
and susceptible rabbits.
large rabbits of the same inbred strain sometimes
After the tubercle-count method was devel-
had to be used in a given experiment. Also,
oped, Lurie used it to select the most resistant and
major differences in size existed among the var-
the most susceptible rabbits for breeding: when
ious inbred rabbit strains themselves, e.g., the
rabbits (at necropsy) showed the desired tuber-
strain III rabbits were much larger than the
cle counts, their nearest relatives were chosen as
strain FC and CaC rabbits.
breeders to continue the desired characteristic.
Lurie compensated for such variations in
TUBERCLE COUNTS TO DETERMINE inhaled dose by dividing the number of bacilli
THE DEGREE OF IMMUNIZATION inhaled (by each rabbit) by the number of grossly
PRODUCED BY A GIVEN VACCINE visible tubercles developed in order to obtain a
Vaccines have no effect on whether an inhaled ratio. Strains III and FC rabbits had ratios that dif-
tubercle bacillus establishes a microscopic lesion fered substantially (Table 5). In addition to adjust-
in the host,because alveolar macrophages have no ing for variations in rabbit size, ratios enable the
immunological specificity. Effective acquired pooling of data from several experiments in
11. LURIE’S PULMONARY TUBERCLE-COUNT METHOD 䡵 205

TABLE 5 Variability in tubercle counts in two of Lurie’s inbred rabbit strainsa

No. of No. of No. of inhaled bacilli Average


Rabbit strain bacilli tubercles required to produce diameter
and numberb inhaled formed one tubercle (ratios) (mm)
(Lurie method)c
III 5⫽27 6,006 27 1,222 3.4
III 5⫽66 6,468 5 1,293 3.9
III 5⫽69 7,014 9 1,779 3.3
III 5⫽76 7,392 6 1,232 2.9
III 5⫽81 7,392 2 3,696 1.5
III 5⫽74 5,273 13 1,406 2.7
III 6⫽2 5,397 18 1,300 3.9
III 6⫽3 5,366 20 1,268 3.8
III 6⫽11 5,366 9 1,596 2.3
III 6⫽14 5,086 4 1,272 2.3
III 6⫽21 4,930 5 1,986 2.3

Mean and SE 6,209 ⫾ 340 10 ⫾ 3 1,004 ⫾ 296 2.9 ⫾ 0.2


Mean ratio from 1,621
the averages listed (6,209/10)

FC 2-9 4,215 107 1,039


FC 3-13 4,700 14 1,050
FC 2-46 4,470 55 1,081
FC 2-47 5,960 63 1,095
FC 3-18 5,960 201 1,030
FC 2-50 6,647 72 1,092
FC 3-29 7,233 32 1,226
FC 3-31 1,796 17 1,106
FC 4-3 2,120 8 1,265
FC 3-37 1,590 36 1,044
FC 3-36 1,440 9 1,160
FC 2-56 1,080 11 1,098

Mean and SE 3,601 ⫾ 695 52 ⫾ 23 107 ⫾ 20


Mean ratio from 69
the averages listed (3,601/52)
a
Variability is seen in the individual values listed.This variability is represented by the SE.The SE can also be represented as
a percentage of the mean: SE/mean ⫻ 100; e.g., for strain III, 296/1,004 ⫻ 100 ⫽ 29%, and for strain FC, 20/107 ⫻ 100 ⫽ 19%.
Commercial New Zealand White rabbits showed a similar amount of variation (see Table 3, row G). Adapted from references 2
and 9.
b
Lurie’s code for labeling his rabbits is best described with the following examples: III 5⫽27 means the 27th rabbit of resis-
tant strain III rabbits in the fifth generation with at least one backcross (double dash) to one of the parents. FC 2-9 means the 9th
rabbit of susceptible strain FC rabbits in the second generation with no backcrosses (single dash) to any parent.
c
The ratio is the number of inhaled human-type tubercle bacilli (H37Rv) required to produce one grossly visible primary pul-
monary tubercle 5 weeks after infection. It is the number of inhaled bacilli divided by the number of visible tubercles produced.
The Lurie method was to divide the dose inhaled by each rabbit by the number of primary tubercles present in each rabbit. He
then averaged the ratios (see text).
206 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

which the number of bacilli in the aerosol was each grossly visible tubercle (11) (see Table 3
markedly different.Although ratios have all these in chapter 4). In contrast, with human-type
advantages, they increase the difference between bacilli (which are always of reduced virulence
rabbits with few grossly visible tubercles and for rabbits), a high inhaled dose increased the
those with many tubercles. Specifically, when a effectiveness of the host’s immune system, i.e.,
series of tubercle counts is compared with a series a greater number of bacilli were required to
of ratios on the same group of rabbits, the rabbits produce each visible tubercle (5). In other
with few tubercles show proportionally higher words, the high inhaled dose of the more vir-
ratios than do the rabbits with many tubercles. In ulent bacillus (the bovine strain in rabbits)
other words, the mean ratio, obtained by divid- reduced acquired (adaptive) host resistance,
ing the number of bacilli inhaled by each rabbit whereas the high inhaled dose of the less vir-
by the number of primary tubercles in each rab- ulent bacillus (the human strain in rabbits)
bit, is inflated by ratios derived from rabbits with enhanced acquired host resistance (discussed
low numbers of tubercles. further in chapter 4).
For example, in Table 5 the inflated ratio
obtained by averaging the ratios from individ- VARIABILITY OF TUBERCLE
ual strain III rabbits was 1,004, whereas the COUNTS AND RATIOS
ratio obtained by dividing the average inhaled The standard error of the mean (SE) repre-
dose by the average number of tubercles was sents the variability within a given strain of
621. Similarly, for the FC rabbits, the inflated rabbits. With Lurie’s strain III rabbits, the
ratio was 107, and the ratio of averages was 69. SE/mean for the ratios (expressed as a per-
Similarly, in Table 3, the averages of individual centage of the mean) was 29% (296/1,004)
ratios compared to ratios of the averages were (Table 5). (The relatively high variability may be
1,946 versus 1,330, 1,058 versus 740, 1,535 ver- partly due to the low number of tubercles pro-
sus 1,060, and 765 versus 585; i.e., averaging duced; low numbers of tubercles exaggerate
ratios from each rabbit inflated the means from ratio.) With Lurie’s strain FC rabbits, the SE was
131% to 162%. 19% (20/107) (Table 5), and with commer-
We recommend that comparisons of all cially outbred New Zealand White rabbits, it
experimental and control groups be made by was 16 to 32% (Table 3). In other words, Lurie’s
first averaging the dose inhaled by each group inbreeding did not always reduce the variabil-
and dividing it by the average number of tuber- ity over that found among outbred New
cles produced in all of the rabbits in that group. Zealand White rabbits. However, the recently
When calculated in this manner, the ratios of the available Thorbecke inbred rabbits (see chapter
two groups being compared will not be inflated 14) did show less variability than outbred rab-
as much by individual rabbits with fewer pri- bits in their resistance to tuberculosis (6; unpub-
mary tubercles than the mean. lished data).
In brief, Lurie’s use of ratios from individual
rabbits exaggerated the differences between his
COMPARISON OF AEROSOL
resistant and susceptible rabbits but did not alter MYCOBACTERIAL INFECTION
the conclusions he derived from them. WITH INTRAVENOUS
INFECTION
EFFECT OF INHALED DOSAGE After an aerosol infection, acquired immunity
ON THE RATIO develops mostly in bronchus-associated lym-
In rabbits, the dose of inhaled tubercle bacilli phoid tissues and the hilar lymph nodes (see
seemed to have an effect on the ratio. When reference 36). After an intravenous infection,
compared to a low inhaled dose, a high inhaled acquired immunity develops mostly in the
dose of virulent bovine-type tubercle bacilli spleen. Therefore, the route of infection may
reduced the effectiveness of the host’s immune affect measurements of (i) bacillary virulence, (ii)
system; i.e., fewer bacilli were required to produce host genetic resistance, and (iii) vaccine efficacy.
11. LURIE’S PULMONARY TUBERCLE-COUNT METHOD 䡵 207

Bacillary Virulence case, the vaccine increases the number of anti-


When aerosols of two bacillary strains of dif- gen-specific lymphocytes, so that the acquired
fering virulence are inhaled, the alveolar (adaptive) host response develops more rapidly
macrophage population (which is highly acti- after challenge with virulent tubercle bacilli.
vated) would immediately destroy or inhibit However, as stated at the beginning of this sec-
greater numbers of the less virulent strain.When tion, the immunity recalled after an aerosol
such bacillary strains are given intravenously, challenge may be somewhat different from the
the two strains would initially grow at about the immunity recalled after an intravenous chal-
same rate in the lungs, because they are ingested lenge, in which the spleen plays a major role. In
by the nonactivated blood-borne macrophages addition, different organs have different resis-
within the pulmonary vasculature. tances to the growth of tubercle bacilli within
For example, C57BL/6 mice injected intra- them (see chapter 15).
venously with wild-type and attenuated tuber-
cle bacilli showed the same bacillary titers at 2 Other Routes of Infection
weeks, and then the titers in the attenuated Bacilli injected by the intraperitoneal route
group gradually declined (37; see reference 38) rapidly enter the bloodstream. They travel
(also see chapter 15). Yet, mice receiving the through the diaphragm into major lymphatic
wild-type bacilli died, and those receiving the trunks that drain into the great veins.Therefore,
attenuated bacilli lived (38). If the mice had been the intraperitoneal route resembles the intra-
infected by aerosol, markedly different bacillary venous route. Bacilli injected subcutaneously,
titers might have been found at 2 weeks, because intramuscularly, or intradermally first drain to the
the alveolar macrophage population is already local lymph nodes (where acquired immunity
activated and kills or inhibits many attenuated develops), and only relatively few bacilli rapidly
inhaled tubercle bacilli before one is allowed to enter the bloodstream.All of these routes bypass
multiply. Therefore, to compare the virulence the alveolar macrophages and measure mainly
of two strains of tubercle bacilli, an aerosol infec- the resistance (immunity) acquired by the host
tion is the most sensitive assay. during the course of the infection.

Host Genetic Resistance PULMONARY TUBERCLE COUNTS IN


Lurie’s resistant and susceptible inbred rabbits GUINEA PIGS AND MICE
apparently differed in their ability to activate Tubercle counts in guinea pigs and mice will not
macrophages—both nonspecifically in the pul- be as useful as they are in rabbits. In these two
monary alveolar macrophage population and laboratory species, each inhaled fully virulent
immune-specifically in their acquired resistance human-type tubercle bacillus reaching the alve-
(see chapters 2 and 5).As with bacillary virulence, olar spaces produces a primary microscopic
infection by aerosol would measure a combina- tubercle that progresses until the host dies (39,
tion of the nonspecific resistance of alveolar 40) (see chapter 15). In guinea pigs and mice, the
macrophages and the acquired resistance devel- pulmonary alveolar macrophages (a highly acti-
oped by expanding antigen-specific lympho- vated cell population) slow (but do not stop) the
cyte populations. Infection by the intravenous intracellular multiplication of the inhaled bacilli.
route would measure only the acquired type. Similarly, macrophages activated by antigen-
specific lymphocytes in tuberculous lesions of
Vaccine Efficacy immunized guinea pigs and mice only slow the
Immunization has little effect on innate immu- progression of pulmonary lesions.
nity, i.e., the activation of the alveolar When euthanized at the appropriate time,
macrophage population after the nonspecific immunized guinea pigs had fewer grossly visible
effects have disappeared.Therefore, either aerosol pulmonary tubercles than did nonimmunized
or intravenous routes of challenge are satisfac- controls, and the tubercles were smaller than
tory for measuring vaccine efficacy. In each those in controls (39). If the mice had been
208

PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS
TABLE 6 Vaccine efficacy: number of inhaled human tubercle bacilli (H37Rv) required to produce one grossly visible primary pulmonary tubercle in unvacci-
nated and vaccinated rabbitsa

B C
Unvaccinated Vaccinated
D E
No. of rabbits No. of bacilli No. of rabbits No. of bacilli Vaccine Vaccine
evaluated inhaled by each evaluated inhaled by each efficacy: no. of efficacy: no. of
rabbit divided rabbit divided tubercles in tubercles in F
A by the no. of by the no. of vaccinated vaccinated P values:
Inbred or tubercles tubercles rabbits as rabbits as vaccinated vs.
commercial produced by produced by percent of the percent of the unvaccinated
rabbit and each rabbit each rabbit no. in control no. in control (using ratios in
vaccine used (avg. of (avg. of rabbits from rabbits from column D)
individual individual ratios of averages of
ratios [Lurie ratios [Lurie averages individual
method]) method]) (B/C ⫻ 100)b ratiosb
Inbred rabbits
Inbred resistant
strain III rabbits
BCG (Phipps) 8 (1,642 ⫾ 107 8 2,948 ⫾ 914 22% P ⫽ 0.013
Inbred susceptible
strain FC rabbits
BCG (Phipps) 12 (1,107 ⫾ 20 9 1,124 ⫾ 18 86% NS

(continued)
Commercial rabbits
BCG (Danish) 6 (1,946 ⫾ 622 6 4,841 ⫾ 2,281 54% 40% P ⫽ 0.08
6 (1,535 ⫾ 432 6 2,458 ⫾ 863 69% 62% NS
BCG (Japanese) 6 (1,946 ⫾ 622 6 5,587 ⫾ 1,480 48% 35% P ⫽ 0.06
6 (1,535 ⫾ 432 6 1,813 ⫾ 154 61% 85% P ⫽ 0.08
BCG (Tice) 6 (1,535 ⫾ 432 6 2,167 ⫾ 441 65% 71% NS
BCG (Pasteur) 8 (1,765 ⫾ 123 8 5,870 ⫾ 1,246 13% 13% P ⫽ 0.001

Mycobacterium 6 1,946 ⫾ 622 6 3,257 ⫾ 753 56% 60% P ⫽ 0.07


microti NCO

11.
8712
M. microti ATCC 6 1,058 ⫾ 270 6 1,427 ⫾ 302 62% 74% NS

LURIE’S PULMONARY TUBERCLE-COUNT METHOD


35781
M. microti ATCC 6 1,058 ⫾ 270 6 3,586 ⫾ 1,026 65% 30% NS
35782
M. microti OV 254 6 1,535 ⫾ 432 6 5,233 ⫾ 859 40% 29% P ⫽ 0.03
6 1,058 ⫾ 270 6 3,440 ⫾ 1,732 25% 31% P ⫽ 0.007
8 1,765 ⫾ 123 8 5,254 ⫾ 2,091 22% 15% P ⫽ 0.002
M. microti ATCC 6 1,058 ⫾ 270 6 3,076 ⫾ 969 29% 34% P ⫽ 0.01
11152 8 1,765 ⫾ 123 8 7,805 ⫾ 44,15 20% 10% P ⫽ 0.002
a
In addition to ratios from Lurie’s inbred strain III and strain FC rabbits, this table lists the mean ratios from each experiment containing 6 to 8 vaccinated commercial rabbits and 6 to 8
control commercial rabbits (see text). NS, not significant, using P ⫽ 0.10.We realize that the P values listed between 0.05 and 0.10 are not usually considered significant, but they indicate
a trend. Data are from references 2, 9, and 12.
b
The percentages listed in columns D and E are a comparison of the average tubercle count in each experiment, which is more precise than a comparison of individual ratios (see text).
Insufficient numbers of rabbits were evaluated to compare the efficacy of the various vaccines (see text). However, the vaccines with the best P values seem most promising.


209
210 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

euthanized at the appropriate time, tubercle 3. Dannenberg, A. M., Jr. 1998. Lurie’s tubercle-
counts could have been made. count method to test TB vaccine efficacy in rabbits.
Front. Biosci. 3:c27–33. Available at http://www
In brief, tubercle counting is most valuable for .bioscience.org/1998/v3/c/dannenbe/list.htm.
testing vaccine efficacy in rabbits challenged by 4. Bishai, W. R., A. M. Dannenberg, Jr.,
aerosol with human-type tubercle bacilli N. Parrish, R. Ruiz, P. Chen, B. C. Zook,
(H37Rv),because rabbits have sufficient resistance W. Johnson, J. W. Boles, and M. L. M. Pitt.
to destroy or inhibit most of these bacilli. How- 1999. Virulence of Mycobacterium tuberculosis
CDC1551 and H37Rv in rabbits evaluated by
ever, in guinea pigs and mice, tubercle counting
Lurie’s pulmonary tubercle-count method. Infect.
could be an excellent way to quantitate vaccine Immun. 67:4931–4934.
efficacy—especially if they are challenged by 5. Manabe, Y. C., A. M. Dannenberg, Jr., S. K.
aerosol with tubercle bacilli of somewhat reduced Tyagi, C. L. Hatem, M.Yoder, S. C.Woolwine,
virulence and they are euthanized before many B. C. Zook, M. L. M. Pitt, and W. R. Bishai.
(but not all) microscopic pulmonary lesions reach 2003. Different strains of Mycobacterium tuberculosis
cause various spectrums of disease in the rabbit
countable size. model of tuberculosis. Infect. Immun. 71:6004–6011.
6. Dorman, S., C. L. Hatem, S. K. Tyagi,
TUBERCLE COUNTING IN MICE— K. Aird, J. Lopez-Molina, M. L. M. Pitt, B. C.
METHODOLOGY Zook,A. M. Dannenberg, Jr.,W. R. Bishai, and
We did not make pulmonary tubercle counts in Y. C. Manabe. 2003. Susceptibility to tuberculo-
mice inhaling M. tuberculosis, but we did so in sis: clues from studies with inbred and outbred New
Zealand White rabbits. Infect. Immun. 72:1700–1705.
mice inhaling the virulent gram-negative bacil- 7. Lurie, M. B., P. Zappasodi,A. M. Dannenberg,
lus Burkholderia pseudomallei, which produces Jr., and G. H.Weiss. 1953. On the mechanism of
melioidosis (41–44). (This bacillus was formerly genetic resistance to tuberculosis and its mode of
called Malleomyces pseudomallei and Pseudomonas inheritance. Am. J. Hum. Genet. 4:302–314.
pseudomallei.) We usually made pulmonary lesion 8. Dannenberg,A. M., Jr., K. Mizunoe, M. Peace,
and P. Zappasodi. 1965. Dermal response to the
counts by dissecting the lungs after they were liposaccharide PmKo from tubercle bacilli as an
fixed.When the lesions were too numerous to index of resistance to tuberculosis. Bull. Johns Hop-
count directly, we counted the surface lesions kins Hosp. 117:174–194.
and prepared a graph that enabled us to estimate 9. Lurie, M. B., P. Zappasodi, E. Cardona-Lynch,
the total number of pulmonary lesions from and A. M. Dannenberg, Jr. 1952.The response
to the intracutaneous inoculation of BCG as an
the number of surface lesions (41). We also
index of native resistance to tuberculosis. J. Immunol.
counted in tissue sections the number of micro- 68:369–387.
scopic pulmonary lesions produced by avirulent 10. Converse, P. J., A. M. Dannenberg, Jr., J. E.
B. pseudomallei 2 days after their inhalation, Estep, K. Sugisaki,Y.Abe, B. H. Schofield, and
before they regressed (43). M. L. M. Pitt. 1996. Cavitary tuberculosis pro-
In brief, the methodology developed in mice duced in rabbits by aerosolized virulent tubercle
bacilli. Infect. Immun. 64:4776–4787.
for counting lesions produced by inhaled B. 11. Converse, P. J., A. M. Dannenberg, Jr.,
pseudomallei could be used for counting tuber- T. Shigenaga, D. N. McMurray, S. W. Phalen,
cles produced by inhaled M. tuberculosis. J. L. Stanford, G. A.W. Rook,T. Koru-Sengul,
H. Abbey, J. E. Estep, and M. L. M. Pitt. 1998.
Pulmonary bovine-type tuberculosis in rabbits:
REFERENCES bacillary virulence, inhaled dose effects, tuberculin
1. Lurie, M. B. 1964. Resistance to Tuberculosis: Exper- sensitivity, and Mycobacterium vaccae immunotherapy.
imental Studies in Native and Acquired Defensive Clin. Diagn. Lab. Immunol. 5:871–881.
Mechanisms. Harvard University Press, Cambridge, 12. Dannenberg, A. M., Jr., W. R. Bishai, N. Par-
Mass. rish, R. Ruiz, W. Johnson, B. C. Zook, J. W.
2. Lurie, M. B., P. Zappasodi, and C. Tickner. Boles, and M. L. M. Pitt. 2000. Efficacies of
1955. On the nature of genetic resistance to tuber- BCG and vole bacillus (Mycobacterium microti) vac-
culosis in the light of the host-parasite relationships cines in preventing clinically apparent pulmonary
in natively resistant and susceptible rabbits. Am. tuberculosis in rabbits: a preliminary report. Vaccine
Rev.Tuberc. 72:297–329. 19:796–800.
11. LURIE’S PULMONARY TUBERCLE-COUNT METHOD 䡵 211

13. Allison, M. J., P. Zappasodi, and M. B. Lurie. 26. Dannenberg, A. M., Jr., and G. A. W. Rook.
1962. Host-parasite relationships in natively resistant 1994. Pathogenesis of pulmonary tuberculosis: an
and susceptible rabbits on quantitative inhalation interplay of tissue-damaging and macrophage-
of tubercle bacilli. Am. Rev. Respir. Dis. 85:553–569. activating immune responses—dual mechanisms
14. Lurie, M. B., A. G. Heppleston, S. Abramson, that control bacillary multiplication, p. 459–483. In
and I. B. Swartz. 1950. An evaluation of the B. R. Bloom (ed.), Tuberculosis: Pathogenesis, Protec-
method of quantitative airborne infection and its use tion, and Control.ASM Press,Washington, D.C.
in the study of the pathogenesis of tuberculosis. Am. 27. Dannenberg,A. M., Jr., and J. F.Tomashefski,
Rev.Tuberc. 61:765–797. Jr. 1998. Pathogenesis of pulmonary tuberculosis,
15. Lurie, M. B., S. Abramson, and A. G. Hep- p. 2447–2471. In A. P. Fishman (ed.), Fishman’s Pul-
pleston. 1952. On the response of genetically re- monary Diseases and Disorders, 3rd ed., vol. 2.
sistant and susceptible rabbits to the quantitative McGraw-Hill Co., Inc, New York, N.Y.
inhalation of human-type tubercle bacilli and the 28. Dannenberg, A. M., Jr. 1999. Pathophysiology:
nature of resistance to tuberculosis. J. Exp. Med. basic aspects. I. Pathogenesis of tuberculosis.
95:119–134. II. Immunology of tuberculosis, p. 17–47. In
16. Francis, J. 1958. Tuberculosis in Animals and Man. D. Schlossberg (ed.), Tuberculosis and Nontuberculous
A Study in Comparative Pathology, p. 293–318. Cas- Mycobacterial Infections, 4th ed. The W. B. Saunders
sell and Company, Ltd., London, United Kingdom. Co., Philadelphia, Pa.
17. Brown, J. H., K. M. Cook, F. G. Ney, and 29. Henderson, H. J., A. M. Dannenberg, Jr., and
T. Hatch. 1950. Influence of particle size upon the M. B. Lurie. 1963. Phagocytosis of tubercle bacilli
retention of particulate matter in the human lung. by rabbit pulmonary alveolar macrophages and its
Am. J. Public Health 40:450–458. relation to native resistance to tuberculosis.
18. Goldberg, L. J., and W. R. Leif. 1950.The use J. Immunol. 91:553–556.
of radioactive isotopes in determining the retention 30. Heppleston, A. G. 1949. Quantitative air-borne
and initial distribution of airborne bacteria in the tuberculosis in the rabbit.The course of human-type
mouse. Science 112:299–300. infection. J. Exp. Med. 89:597–610.
19. Sonkin, L. S. 1951. The role of particle size in 31. Lurie, M. B. 1941. Heredity, constitution and
experimental airborne infection. Am. J. Public Health tuberculosis.An experimental study. Am. Rev.Tuberc.
53:337–354. 44(Suppl. 3):1–125.
20. Dannenberg, A. M., Jr., M. Burstone, P. C. 32. Manca, C., L. Tsenova, C. E. Barry III,
Walter, and J.W. Kinsley. 1963.A histochemical A. Bergtold, S. Freeman, P. A. J. Haslett, J. M.
study of phagocytic and enzymatic functions of Musser,V. H. Freedman, and G. Kaplan. 1999.
rabbit mononuclear and polymorphonuclear exu- Mycobacterium tuberculosis CDC1551 induces a more
date cells and alveolar macrophages. I. Survey and vigorous host response in vivo and in vitro, but is
quantitation of enzymes, and states of cellular acti- not more virulent than other clinical isolates.
vation. J. Cell Biol. 17:465–486. J. Immunol. 162:6740–6746.
21. Myrvik, Q. N., E. Soto Leake, and D. Gonza- 33. Kelley, C. L., and F. M. Collins. 1999. Growth
lez-Ojeda. 1962. Enzymatic differences between of a highly virulent strain of Mycobacterium tubercu-
normal rabbit alveolar macrophages and oil-induced losis in mice of differing susceptibility to tubercu-
peritoneal macrophages. Bacteriol. Proc. 62:79. lous challenge. Tuber. Lung Dis. 79:367–370.
22. Myrvik, Q. N., E. Soto Leake, and B. Fariss. 34. North, R. J., L. Ryan, R. LaCource,T. Mogues,
1961. Lysozyme content of alveolar and peritoneal and M. E. Goodrich. 1999. Growth rate of
macrophages from the rabbit. J. Immunol. 86:133– mycobacteria in mice as an unreliable indicator of
136. mycobacterial virulence. Infect. Immun. 67:5483–
23. Dannenberg, A. M., Jr., P. C. Walter, and 5485.
F. A. Kapral. 1963. A histochemical study of 35. Orme, I. M. 1999.Virulence of recent notorious
phagocytic and enzymatic functions of rabbit Mycobacterium tuberculosis isolates. Tuber. Lung Dis.
mononuclear and polymorphonuclear exudate cells 79:379–381.
and alveolar macrophages. II.The effect of particle 36. Strober,W., B. Kelsall, and T. Marth. 1998. Oral
ingestion on enzyme activity; two phases of in tolerance. J. Clin. Immunol. 18:1–30.
vitro activation. J. Immunol. 90:448–465. 37. McKinney, J. D., K. Höner zu Bentrup, E. J.
24. Dannenberg,A. M., Jr. 1991. Delayed-type hyper- Muñoz-Elías,A. Miczak, B. Chen,W.-T. Chan,
sensitivity and cell-mediated immunity in the patho- D. Svenson, J. C. Sacchettini,W. R. Jacobs, Jr.,
genesis of tuberculosis. Immunol.Today 12:228–233. and D. G. Russell. 2000. Persistence of Mycobac-
25. Dannenberg, A. M., Jr. 1993. Immunopatho- terium tuberculosis in macrophages and mice requires
genesis of pulmonary tuberculosis. Hosp. Pract. the glyoxylate shunt enzyme isocitrate lyase. Nature
28:33–40 (Off. ed. 51–58). 406:735–738.
212 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

38. Glickman, M. S., J. S. Cox, and W. R. Jacobs, primary lesions as illustrated by melioidosis. Proc. Soc.
Jr. 2000. A novel mycolic acid cyclopropane syn- Exp. Biol. Med. 92:571–575.
thetase is required for cording, persistence, and 42. Dannenberg,A. M., Jr., and E. M. Scott. 1958.
virulence of Mycobacterium tuberculosis. Mol. Cell 5: Melioidosis: pathogenesis and immunity in mice and
717–727. hamsters. I. Studies with virulent strains of
39. Horowitz, M. A., G. Harth, B. J. Dillon, and Malleomyces pseudomallei. J. Exp. Med. 107:153–166.
S. Maslesa-Galic. 2000. Recombinant bacillus
Calmette-Guérin (BCG) vaccines expressing the 43. Dannenberg,A. M., Jr., and E. M. Scott. 1958.
Mycobacterium tuberculosis 30-kDa major secretory Melioidosis: pathogenesis and immunity in mice and
protein induce greater protective immunity against hamsters. II. Studies with avirulent strains of
tuberculosis than conventional BCG vaccines in a Malleomyces pseudomallei. Am. J. Pathol. 34:1099–1121.
highly susceptible animal model. Proc. Natl.Acad. Sci. 44. Dannenberg,A. M., Jr., and E. M. Scott. 1960.
USA 97:13853–13858. Melioidosis: pathogenesis and immunity in mice
40. Medina, E., and R. J. North. 1999. Genetically and hamsters. III. The effect of vaccination with
susceptible mice remain proportionally more sus- avirulent strains of Pseudomonas pseudomallei on the
ceptible to tuberculosis after vaccination. Immunol- resistance to the establishment and the resistance to
ogy 96:16–21. the progress of respiratory melioidosis caused by
41. Dannenberg,A. M., Jr., and E. M. Scott. 1956. virulent strains: all-or-none aspects of this disease.
Determination of respiratory LD50 from number of J. Immunol. 84:233–246.
Section 4.

TUBERCULOSIS IN RABBITS AND OTHER


COMMON LABORATORY ANIMALS
This page intentionally left blank
12
NATURAL AIRBORNE INFECTION

Lurie’s natural airborne infection [215]


Resistance to the establishment of tuberculous lesions produced by virulent
bovine-type bacilli in Lurie’s rabbits [216]
Resistance to the progress of pulmonary tuberculosis produced by virulent
bovine-type bacilli in Lurie’s rabbits [218]
Conversion of the tuberculin skin test with no grossly visible primary
pulmonary lesion at necropsy [223]
Only a single primary tuberculous lesion develops under natural
conditions [223]
Resistance to the establishment of tuberculous lesions produced by virulent
human-type bacilli in Lurie’s rabbits [223]
Resistance to the progress of pulmonary tuberculosis produced by virulent
human-type bacilli in Lurie’s rabbits [224]
Establishment and progress of tuberculosis in mice and guinea pigs [224]
Riley’s experiments with natural airborne infection [224]
Other methods of room air sampling [226]
Comparisons of tuberculosis in rabbits and humans [226]
Effect of immunization on resistance to the establishment and resistance to
the progress of an acute septicemic disease (melioidosis) and a chronic,
slowly progressing disease (tuberculosis) [226]

Abstract. Using natural airborne infection of virulent bovine-type tubercle bacilli over
many months, Lurie showed that resistance to the establishment of tuberculosis and resis-
tance to its progress are separate phenomena: his inbred resistant rabbits converted their
tuberculin skin tests an average of 2.7 months sooner than did his inbred susceptible rab-
bits.Yet, once established, the disease progressed slowly in the resistant rabbits and rapidly
in the susceptible rabbits.The separation of the establishment and progress of tuberculo-
sis is only applicable to experiments in which occasional fully virulent tubercle bacilli are
inhaled over many months. It does not seem applicable to rabbits or humans inhaling
human-type tubercle bacilli, which are never fully virulent in these hosts.
Airborne infection of laboratory animals over many months has, however, established
other concepts directly applicable to tuberculosis in humans. (i) Only a single grossly vis-
ible primary pulmonary lesion will be produced, despite the continuous presence of vir-
ulent tubercle bacilli in the air.The immunity developed in response to the primary lesion
is evidently sufficient to prevent other occasionally inhaled tubercle bacilli from causing
grossly visible lesions. (ii) Some animals (and perhaps a few humans) may convert their der-
mal tuberculin reactions, and yet show no grossly visible primary lesions in their lungs at
necropsy.This occurrence may be due to the early spread of inhaled bacilli out of the lungs
to the hilar lymph nodes, where the growth of tubercle bacilli can be more easily con-
trolled.These concepts are consistent with what Riley found when he exposed guinea pigs
for months to air from a ward containing sputum-positive tuberculous patients.

LURIE’S NATURAL AIRBORNE mesh metal screen from the cages containing
INFECTION uninfected inbred rabbits (Fig. 1). When the
Lurie’s natural airborne experiments (1–5) were infected commercial rabbits (shedding virulent
performed in a room where the runs contain- bovine-type tubercle bacilli in their urine)
ing the source rabbits were separated by a fine- moved around in the bedding on the floor of

215
216 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

their runs, the tubercle bacilli became airborne. monary lesion usually produces enough acquired
The approximate time at which a primary immunity to prevent subsequently inhaled
(microscopic) tuberculous lesion was established tubercle bacilli from causing lesions that reach
in the lungs of the uninfected inbred rabbits was grossly visible size (2, 3) (discussed below).
indicated by the conversion of their tuberculin
skin tests, which were made every 2 to 4 weeks RESISTANCE TO THE
(Tables 1 and 2) (1, 3). ESTABLISHMENT OF TUBERCULOUS
LESIONS PRODUCED BY VIRULENT
Lurie performed three natural airborne BOVINE-TYPE BACILLI IN LURIE’S
experiments, each with a different concentration RABBITS
of virulent bovine-type tubercle bacilli in the air Resistance to the establishment of an infection
of the room (Table 2) (3). He compared one is to be contrasted with resistance to its progress
genetically resistant strain of rabbits with two (1, 3, 5). In tuberculosis, lesions are established
genetically susceptible strains.Table 1 is a com- when the bacillus initially multiplies in the host,
posite of all three experiments. It shows the and lesions progress if such multiplication con-
variations among the individual rabbit strains. tinues.
These experiments established two basic prin- In Lurie’s natural airborne experiments,
ciples: (i) Resistance to the establishment of a almost every unit of virulent bovine-type tuber-
tuberculous lesion is distinct from resistance to cle bacilli that is retained in the alveoli should
its progress, and (ii) a single active primary pul- cause progressive disease in both his resistant and

FIGURE 1 A diagram of the rabbit cages that Lurie used for natural airborne infection.Thirty
commercial rabbits, many of which were shedding virulent bovine-type tubercle bacilli (Ravenel)
in their urine, were distributed into the three rabbit runs (rear of the diagram).As the rabbits moved
throughout the runs, they caused tubercle bacilli in the urine-dampened bedding to become air-
borne.
Noninfected inbred resistant and susceptible rabbits were placed in 30 cages (front of the dia-
gram).These rabbits inhaled occasional tubercle bacilli over a period of many months.The cages
were moved daily, so that during the course of 30 days, each cage resided in each position for 1
day. Reproduced with permission from reference 2.
12. NATURAL AIRBORNE INFECTION 䡵 217

TABLE 1 Resistance to the establishment of tuberculosis and resistance to its progress with a natural airborne
contagion of virulent bovine-type tubercle bacillia
Preallergic period Duration of disease
Rabbit strain Rabbit no.
(months) (months)
Strain A, high resistance A8⫽19 0.9 4.7
A8⫽29 1.3 10.4
A7⫽31 1.8 No visible lesions
A8⫽43 1.8 No visible lesions
A4-4 2.0 No visible lesions
A8⫽51 2.3 4.1
A7⫽26 2.3 5.0
A3⫽3 3.2 11.5
A6⫽21 3.3 No visible lesions
A5⫽3 3.3 12.9
A5⫽4 3.3 No visible lesions
A5⫽21 3.3 10.7
A7⫽5 3.5 No visible lesions
A7⫽36 3.6 8.0
A5⫽2 4.3 No visible lesions
A7⫽10 4.9 No visible lesions
A2⫽11 5.0 7.5
A7⫽4 6.0 No visible lesions
Average 3.1 ⫾ 1.3 8.3 ⫾ 3.1
Strain F, low resistance F4-25 1.9 3.8
F4-30 2.4 3.0
F2-15 3.0 6.2
F5-2 4.6 No visible lesions
F6-25 5.1 3.7
F2-3 6.0 5.0
F5-14 6.0 No visible lesions
F4-33 6.2 3.0
F3-9 6.5 8.0
F6-14 8.3 2.1
F3-7 11.0 3.5
F4-11 11.7 4.0
Average 6.1 ⫾ 3.0 4.2 ⫾ 1.7
Strain C, low resistance C6-9 1.3 3.9
C2-8 2.0 9.5
C6-32 2.1 3.6
C5-50 2.3 3.9
C4R-2 2.7 4.3
C5-30 3.0 3.3
C4S-9 3.3 Tuberc. ⫹ snuffles
C2-6 3.7 3.7
C4S-11 3.8 Tuberc. ⫹ snuffles
C4R-6 4.7 3.8
C4S-30 4.9 No visible lesions

(continued next page)


218 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

TABLE 1 Resistance to the establishment of tuberculosis and resistance to its progress with a natural airborne
contagion of virulent bovine-type tubercle bacillia (continued)
Preallergic period Duration of disease
Rabbit strain Rabbit no.
(months) (months)
Strain C, low resistance C2-1 5.0 3.0
(continued) C6-1 5.5 No visible lesions
C2-7 7.0 5.0
C6-26 8.3 Tuberc. ⫹ snuffles
C5-14 9.0 2.6
C2-18 11.9 No visible lesions
Average 4.7 ⫾ 2.8 4.2 ⫾ 1.8
a
This table lists data on all of the rabbits with positive tuberculin reactions that were used in Lurie’s natural airborne infec-
tion experiments over a 10-year period. Note that the resistant A rabbits converted their tuberculin reactions sooner, but lived
longer, than did the susceptible F and C rabbits.This table was reproduced in full, because these rabbit strains are now extinct,
which makes these experiments unique. Preallergic period: A vs. F: P ⫽ 0.001; A vs. C: P ⫽ 0.024. Duration of disease: A vs. F:
P ⫽ 0.001;A vs. C: P ⫽ 0.001. Means and standard errors are listed. Data from reference 3.

susceptible rabbits. Fully virulent bovine-type the resistant group (7). Perhaps, more bacilli in
bacilli may be somewhat inhibited by pul- the susceptible group adhered to the bronchial
monary alveolar macrophages, but these bacilli walls, because of the increased mucus there, and
are never inactivated (6, 7). did not reach the alveolar spaces.
In Lurie’s high-intensity experiment, the re- Did the resistant rabbits breathe more air
sistant rabbits converted their tuberculin skin than the susceptible ones? The resistant rabbits
tests several months earlier than did the sus- were usually larger, but the susceptible rabbits
ceptible rabbits—an average of 2.0 versus 4.7 were usually more active.
months (Table 2 and Fig. 2) (3).Therefore, with In brief, many possibilities exist for why, in
virulent bovine-type tubercle bacilli, a primary these natural airborne experiments, the resistant
lesion was apparently established sooner in the rabbits (on average) converted their tuberculin
resistant group. reactions sooner than did the susceptible rabbits,
In the resistant rabbits, could the alveolar but none of these possibilities has been investi-
macrophages (which are highly activated [8]) gated further.
trap inhaled bacilli more effectively than the
alveolar macrophages of the susceptible rabbits?
In one experiment, alveolar macrophages that
RESISTANCE TO THE PROGRESS
were washed out of the lungs of resistant rabbits OF PULMONARY TUBERCULOSIS
phagocytized twice as many tubercle bacilli in PRODUCED BY VIRULENT BOVINE-
vitro than did those washed out of the lungs of TYPE BACILLI IN LURIE’S RABBITS
susceptible rabbits (9). If fewer tubercle bacilli In the highest-dose natural airborne experiment,
were phagocytized in vivo in the susceptible the inbred resistant rabbits lived an average of 6.4
rabbits, then more bacilli would ascend the months after they developed tuberculin sensi-
bronchial escalator. (The bronchial tree is rather tivity (3) (Table 2, Fig. 2).They died of a slowly
resistant to tuberculosis.) progressing cavitary disease with bronchial spread
Could the bronchial trees of the two rabbit (Fig. 3), similar to that found in adult humans.The
groups have been of such a configuration that susceptible rabbits, in contrast, only lived an aver-
more bacilli reached the alveoli in the resistant age of 3.5 months (3) (Table 2, Fig. 2) and devel-
group? We do not know.The susceptible group oped a hematogenously spread disease (Fig.4) (see
had more overt snuffles (infection with Pas- chapters 13 and 14), similar to that found in
teurella sp. or Bordetella bronchisepticum) than did infants and immunosuppressed individuals (see
TABLE 2 Effect of the intensity of natural airborne contagion on tuberculosis acquired by rabbits of various levels of natural resistancea
C E
Avg. No. (%) of rabbits
D G
B duration F
A No. (%) of that developed Duration of H
No. of of Preallergic (or
Intensity of rabbits that positive tuberculin fatal Type of
Rabbit rabbits exposure pretuberculous)
contagion developed reactions without tuberculosis tuberculosis
strain exposed before period (months)
tuberculosis demonstrable (months)
death tuberculosis
(months)
A (resistant) Low 7 14.6 0 4 (57%) 4.4 – None
Medium 6 10.4 3 (50%) 2 (33%) 3.7 11.4 Slowly progressive,
localizing
High 8 9.3 5 (63%) 2 (25%) 2.0 6.4 3 intermediate;

12.
2 slowly

NATURAL AIRBORNE INFECTION


progressive,
localizing
C, F
(susceptible) Low 11 9.8 2 (18%) 3 (27%) 5.5 3.0 Rapidly
progressive,
disseminating
Medium 11 8.6 10 (91%) 0 4.3 3.8 Rapidly
progressive,
disseminating
High 7 8.1 6 (86%) 1 (14%) 4.7 3.5 Rapidly
progressive,
disseminating
a
Note that a higher percentage of resistant A rabbits than susceptible C and F rabbits developed a positive tuberculin reaction without evidence of grossly visible tuberculosis at necropsy
(column E). Note also that (as expected) a higher intensity of contagion caused a higher percentage of rabbits to develop tuberculosis (column D). Adapted from reference 3.


219
220 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

FIGURE 2 Resistance to attack (the preallergic period) and resistance to the progress (dura-
tion) of tuberculosis in the resistant A and susceptible C and F strains of rabbits.The graph on
the left shows the average time (in months) in which these rabbits converted their tuberculin reac-
tions.The graph on the right shows the average time (in months) that these rabbits lived after
they became tuberculin positive.
The difference in the preallergic period between the resistant A strain and the susceptible
F strain was highly significant, but that between the A strain and the susceptible C strain was not
(3). However, the duration of the disease in the resistant A strain was always significantly longer
than that in both susceptible strains (C and F) (3).
Experiments were performed with relatively low, medium, and high numbers of tubercle bacilli
in the air of the room.This figure represents data from the experiment with the highest num-
ber of tubercle bacilli. Reproduced with permission from reference 3.

chapters 1 and 3).Therefore, the resistant rabbits In the resistant rabbits, tubercle bacilli spread
lived about twice as long as susceptible rabbits to the hilar lymph nodes more readily than they
after infection with virulent bovine-type tuber- did in susceptible rabbits,but once in the lymph
cle bacilli (2, 3), which confirmed that the resis- nodes,the bacilli are inhibited much more effec-
tant group in this experiment was truly resistant tively in the resistant rabbits (2,7,10).After infec-
and that the susceptible group was truly suscep- tion with virulent bovine-type tubercle bacilli,
tible. These findings were supported by other the hilar lymph nodes of the resistant rabbits
natural airborne experiments (Table 2). remained small and showed little caseation, and
The tubercles in resistant hosts contained the growth of bacilli was well controlled.In con-
fewer bacilli than those in susceptible hosts (see trast, the hilar nodes of the susceptible rabbits
chapters 2, 13, and 14), because of the more were large and showed considerable caseation,
rapid development of macrophages into mature and bacillary growth was poorly controlled (2).
epithelioid cells (1, 10, 11).A mature epithelioid With the bovine-type infection, the bacilli
cell is one that is highly activated, i.e., rich in invaded the bloodstream in both resistant and
oxidative and hydrolytic enzymes (e.g., -galac- susceptible rabbits, especially after caseous necro-
tosidase) and therefore capable of inhibiting the sis had developed. The resistant rabbits usually
intracellular growth of tubercle bacilli and developed relatively few metastatic lesions in
digesting their components (12–14). their lungs, spleen, and kidneys, and such lesions
12. NATURAL AIRBORNE INFECTION 䡵 221

FIGURE 3 Lungs and other


organs of resistant rabbit A2-6. It
converted its tuberculin reaction at
9 months and died of tuberculosis 9
months later.The primary lesion was
the single (well-encapsulated) cavity
in the right lung.There was tuber-
culosis of the larynx and intestines,
as well as a tuberculous ulcer of the
colon—all from the spread of large
numbers of virulent bovine-type
tubercle bacilli up the bronchial tree
and then into the alimentary canal.
The hilar and mesenteric lymph
nodes were grossly normal, as were
the kidneys. Several nonprogressive
tubercles of hematogenous origin
were present in both lungs. Re-
produced with permission from ref-
erence 2.

had relatively few bacilli and relatively small of bacilli from the primary pulmonary lesion. In
amounts of caseation (2, 7). In contrast, the sus- the resistant rabbits during the first weeks, the
ceptible rabbits developed numerous metastatic greater interstitial inflammation increased the
lesions, and such lesions had many bacilli and spread of bacilli to the hilar lymph nodes, but
large amounts of caseation (2, 7).The disease in the bacilli were inhibited once there. In the
the susceptible rabbits resembled the childhood- susceptible rabbits, the greater caseation
type tuberculosis found in humans (15–17) (see increased the lymphogenous and hematoge-
chapter 1). nous spread of the bacilli, and the bacilli grew
In summary, attributes of both resistance and rather well in the metastatic sites.The decisive
susceptibility contributed to the dissemination factor in the development of secondary lesions
222 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

FIGURE 4 Lungs and other organs of susceptible rabbit F4-33. It converted its
tuberculin reaction at 6.2 months and died of tuberculosis 3.0 months later
(Table 1).The primary lesion is the large, completely caseous lesion in the middle
of the left lung.The caseum in this lesion did not liquefy, so no cavity formed.The
homolateral draining hilar lymph nodes show extensive enlargement and massive
caseation. Numerous large progressive caseous tubercles of hematogenous origin are
present in both lungs.The kidneys, pleura, and knee joint show rapidly progressive
caseous lesions. Reproduced with permission from reference 2.
12. NATURAL AIRBORNE INFECTION 䡵 223

was the fate of the bacilli in their new locations: monary lesion remained small and healed by the
In metastatic sites, the resistant rabbits could time of necropsy; and (ii) (as discussed above) the
inhibit bacillary growth much more effectively inhaled bacilli may not have multiplied in the
than could the susceptible rabbits. alveoli, but were carried in the lymph (proba-
bly within macrophages) to the hilar lymph
nodes, where in Lurie’s resistant rabbits bacillary
CONVERSION OF THE TUBERCULIN
SKIN TEST WITH NO GROSSLY multiplication was inhibited (2, 3), and in Riley’s
VISIBLE PRIMARY PULMONARY guinea pigs bacillary multiplication usually
LESION AT NECROPSY caused visible tuberculous lesions (18, 19).
In Lurie’s natural airborne infection of rabbits
with virulent bovine-type bacilli, some resis- ONLY A SINGLE PRIMARY
tant and susceptible animals became tuber- TUBERCULOUS LESION DEVELOPS
culin positive but showed no grossly visible UNDER NATURAL CONDITIONS
disease at necropsy (Table 2) (2, 3).This occur- In Lurie’s experiments, the rabbits inhaled rel-
rence was more common in the natively resis- atively few tubercle bacilli over many months,
tant rabbits and more common in the experi- so that the initial primary pulmonary lesion
ment with the lowest concentrations of had time to produce sufficient immunity to
tubercle bacilli in the room air (Table 2) (3). In prevent subsequently inhaled tubercle bacilli
the tuberculin-positive rabbits without pul- from producing grossly visible lesions (1, 2).
monary tuberculous lesions, no grossly visible Such immunity cannot, however, prevent the
tuberculous lesions were found in the hilar development of microscopic lesions from the
lymph nodes. Such lesions probably had devel- subsequently inhaled bacilli, because immu-
oped (at least microscopically) and then healed nization does not increase the microbicidal
(see chapter 13). power of pulmonary alveolar macrophages (see
In Riley’s natural airborne experiments (see chapters 11 and 22).
below), guinea pigs inhaled (over many months) In Riley’s experiments (see below), the guinea
air containing virulent human-type tubercle pigs also inhaled relatively few tubercle bacilli
bacilli from sputum-positive tuberculous patients over many months, and also developed only
in a hospital ward. In Riley’s Experiment I, 34% one primary pulmonary tubercle (18, 19). How-
(26 of 77) of the animals became tuberculin ever, these guinea pigs were removed from the
positive without any grossly visible pulmonary infectious air and necropsied soon after they
tuberculous lesions (18, 19). In Riley’s Experi- converted their tuberculin skin test, so there
ment II, 16% (10 of 63) did the same. However, was little opportunity for exogenous reinfection
in contrast to rabbits, most of these guinea pigs to occur. In contrast, Lurie’s rabbits were allowed
had grossly visible tuberculous lesions in their to remain in the infectious air for many months,
hilar lymph nodes, and over half of these guinea and usually for the course of the disease.
pigs also had lesions in their spleens.
One inhaled unit of 1 to 3 fully virulent RESISTANCE TO THE
ESTABLISHMENT OF TUBERCULOUS
bovine-type (Ravenel) tubercle bacilli in the LESIONS PRODUCED BY VIRULENT
alveolar spaces causes progressive disease in rab- HUMAN-TYPE BACILLI IN LURIE’S
bits (1), and one such unit of fully virulent RABBITS
human-type or bovine-type bacilli causes pro- Human-type tubercle bacilli are never fully vir-
gressive disease in guinea pigs. If this is true, then ulent in rabbits (or humans) (1, 3, 5). Rabbits
how does a rabbit or guinea pig become tuber- inactivate many inhaled units of virulent human-
culin positive from inhaled tubercle bacilli with- type bacilli for every unit that produces a grossly
out developing grossly visible pulmonary dis- visible pulmonary lesion (7, 10, 20). In con-
ease? There are two likely possibilities. (i) The trast, every inhaled unit of fully virulent bovine-
inhaled bacillary unit may have been weak and type tubercle bacilli reaching the pulmonary
not fully virulent, so that the primary pul- alveoli of rabbits produces a progressive disease.
224 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

From lung culture data (10), we know that a bits did not progress, but showed fibrosis, which
high proportion of inhaled human-type bacilli indicated that they would eventually heal.
are destroyed by rabbit alveolar macrophages In brief, resistance to the establishment of
during the first 7 days. From studies in BCG- tuberculosis and resistance to its progress would
immunized rabbits (21, 22), we know that not be separable phenomena in rabbits or
acquired immunity is also able to decrease the humans exposed to virulent human-type tuber-
number of grossly visible pulmonary tubercles. cle bacilli. In both of these hosts, most inhaled
However, we do not know what proportion of human-type tubercle bacilli seem to be imme-
the grossly visible tubercles are prevented initially diately destroyed by alveolar macrophages (pre-
by alveolar macrophages and what proportion venting establishment), but once established,
are prevented subsequently by stopping the pro- many of these lesions do not progress.
gression of microscopic tubercles.
A much greater number of human-type
bacilli are destroyed or inhibited by the alveo- ESTABLISHMENT AND PROGRESS
lar macrophages of Lurie’s resistant rabbits than OF TUBERCULOSIS IN MICE AND
by the alveolar macrophages of his susceptible GUINEA PIGS
rabbits (7, 10, 23, 24). Therefore, although the Mice and guinea pigs are so susceptible to viru-
alveolar macrophages of the resistant group may lent human-type and bovine-type tubercle bacilli
trap these bacilli more readily, fewer micro- that their alveolar macrophages kill few, if any, of
scopic lesions would form, because these alve- these bacilli. Also, tuberculosis once established
olar macrophages are highly activated. In other usually progresses in mice and in guinea pigs and
words, following the inhalation of human-type kills these hosts. It is possible that some mice or
bacilli, the resistant rabbits would prevent the guinea pigs have alveolar macrophages that can
establishment of a microscopic lesion more trap occasional airborne human-type tubercle
effectively than would the susceptible rabbits— bacilli (when inhaled over many months) more
which is not true for inhalation of virulent effectively than do other mice or guinea pigs. If
bovine-type bacilli (described above). so, then these laboratory animals would show
the same separation of establishment and progress
of tuberculosis that was found with Lurie’s rab-
RESISTANCE TO THE PROGRESS
OF PULMONARY TUBERCULOSIS bits inhaling occasional bovine-type bacilli.
PRODUCED BY VIRULENT HUMAN- Of course, in all hosts, immunization slows the
TYPE BACILLI IN LURIE’S RABBITS progress of the disease and has no effect on its
The progress of tuberculosis produced by viru- establishment: alveolar macrophage functions
lent human-type tubercle bacilli is described in have no immunologic specificity (see chapters
chapter 14 and in references 10 and 20. In brief, 5 and 22).
primary pulmonary lesions in Lurie’s resistant
rabbits usually healed within a few months, but
if they liquefied and cavitated, such healing would RILEY’S EXPERIMENTS WITH
probably take over a year.We would expect such NATURAL AIRBORNE INFECTION
primary lesions to have a similar course in com- Riley performed two major experiments (each
mercial New Zealand White rabbits. lasting 2 years) with outbred guinea pigs (18, 19,
Primary pulmonary lesions in Lurie’s sus- 25–27).The source of infection was air from six
ceptible rabbits did not heal as quickly, and usu- single-bed hospital rooms, each housing a patient
ally secondary pulmonary lesions occurred.The producing sputum that contained virulent
bacilli causing these secondary lesions probably human-type tubercle bacilli. The guinea pigs
came from the hilar nodes via efferent lym- were housed on the floor above that of the
phatics draining into the great veins (see chap- patients in a vertical cylindrical chamber aero-
ter 1). Nevertheless, the secondary lesions caused dynamically engineered by Wells (28). This
by human-type bacilli in Lurie’s susceptible rab- chamber had six levels that held cages contain-
12. NATURAL AIRBORNE INFECTION 䡵 225

ing 4 to 5 guinea pigs per cage, so that an mutations), and (iii) test the efficacy of novel
average of about 150 guinea pigs were con- interventions (such as aerosolized drugs) on
tinuously exposed at one time. The animals transmissibility.
were skin-tested with tuberculin every month, Each month the tuberculin-positive guinea
at which time each tuberculin-positive guinea pigs will be removed from the source of expo-
pig was removed from the chamber, necrop- sure. Some of these animals will be euthanized
sied, and replaced by an uninfected guinea immediately, but many will be allowed to
pig. This procedure eliminated the possibility develop extensive disease over the next 6
of the guinea pigs’ infecting each other, months. In every animal, the virulence of the
because early lesions would not shed any bacilli infecting tubercle bacilli will be assessed at
into the environment. Individual patients were necropsy for both the extent of the disease and
identified by the antimicrobial susceptibility the type of pathology present.
of the tubercle bacilli in their sputum (18, These natural airborne experiments will pro-
19). Also, various sputum samples were aero- vide information on the infectivity of tubercu-
solized, and their infectivity for guinea pigs was lous patients that is not obtainable in any other
correlated with the infectivity of the air from manner.The bacilli aerosolized by tuberculous
the ward (18). patients are surrounded by bronchial secretions
These experiments clearly established that and usually by components of the liquefied
tuberculous patients can transmit the disease by caseum. Bacilli grown in culture medium (and
the airborne route and that patients with numer- subsequently aerosolized) are not surrounded by
ous bacilli in their sputum are more infectious these components and spend less time in the air
than those with fewer bacilli. Only one pri- before they are inhaled by animals in the expo-
mary pulmonary lesion was found in each sure chamber.Therefore, the infectivity of bacilli
guinea pig. However, as mentioned above, the in natural airborne experiments is probably dif-
guinea pigs were removed from the source of ferent from the infectivity of bacilli in aerosol
contagion as soon as they converted their tuber- experiments.
culin reaction, so few if any would have had the Also, the infectivity of human-type tubercle
opportunity to develop another lesion from bacilli for guinea pigs is probably greater than
inhaling a second infectious particle. their infectivity for humans. Guinea pigs must
Guided by Edward A. Nardell (Harvard inhale 3 to 6 units of 1 to 3 bacilli to convert
School of Public Health), who was Riley’s col- their tuberculin skin test, whereas humans must
league, and by Paul Jensen (Centers for Disease inhale an estimate of 20 to 200 bacillary units
Control and Prevention), Bernard Fourie and to do so. (Rabbits must inhale 600 to 1,200
Karen Weyer of the South African Medical units.) Therefore, small changes in bacillary
Research Council (Witbank, South Africa) have infectivity for humans might not be detected by
built an experimental tuberculosis ward similar guinea pigs for reasons presented in chapter 11.
to the one built by Riley about 50 years ago.The However, if rabbits were substituted for guinea
patients in their study have multidrug-resistant pigs in these experiments, almost none of the
tuberculosis, and about 50% of them are HIV animals would contract the disease or convert
infected. The long-term goals of this project their tuberculin skin tests, because too large an
are to (i) investigate the principal determinants inhaled dose of human-type tubercle bacilli
of transmission of tuberculosis and (ii) test infec- would be required to do so.
tion control interventions suitable for use in A similar experiment with different logistics
countries with a high burden of the disease, is being performed in Lima, Peru, by Roderick
such as upper room germicidal irradiation.The Escombe with Carlton Evans of the Johns Hop-
short-term goals are to (i) compare the infec- kins Bloomberg School of Public Health in
tiousness of HIV-positive and HIV-negative Baltimore, Md. To date, they have found that
patients, (ii) study the transmissibility of opening doors and windows is the most efficient
mycobacterial strain types (including those with method of reducing the concentration of the
226 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

infectious airborne particles in the rooms of humans with positive tuberculin skin tests
these tuberculous patients. develop progressive tuberculosis (see chapter 1
and see reference 34 for details).
OTHER METHODS OF Humans are more susceptible to human-type
ROOM AIR SAMPLING tubercle bacilli than are rabbits. Rabbits usually
Another experiment that detected tubercle recover from active lesions caused by human-
bacilli in hospital room air was published in 1999 type bacilli, whereas many humans with clinically
(29).Air near the patient’s bed was forced through active tuberculosis will die of the disease, unless
a micropore membrane filter, and PCR (poly- effective antimicrobial agents are given. On the
merase chain reaction) was used to detect the other hand, humans are somewhat more resistant
DNA of tubercle bacilli collected on the filter. than rabbits to fully virulent bovine-type bacilli,
This experiment does not, however, distinguish because tuberculous lesions produced by such
between live and dead tubercle bacilli or between bacilli are fatal in rabbits, but not always fatal in
large and small particle sizes (30).When inhaled, humans. Except for these slight differences, how-
bacillary particles of large size do not reach the ever, the disease in rabbits and the disease in
alveolar spaces to cause pulmonary disease. humans resemble each other quite well.
Nevertheless, this method can aid the diagnosis
of tuberculosis by sometimes detecting tubercle EFFECT OF IMMUNIZATION ON
bacilli when they are not detected in sputum. RESISTANCE TO THE
ESTABLISHMENT AND RESISTANCE
TO THE PROGRESS OF AN ACUTE
COMPARISONS OF TUBERCULOSIS SEPTICEMIC DISEASE (MELIOIDOSIS)
IN RABBITS AND HUMANS AND A CHRONIC, SLOWLY
Human tuberculosis usually resembles the disease PROGRESSING DISEASE
produced in rabbits following the inhalation of (TUBERCULOSIS)
human-type bacilli in that resistance to its estab- Melioidosis is caused by Burkholderia pseudomal-
lishment and resistance to its progress are not sep- lei, a gram-negative bacillus that was formerly
arable. Persons evidently must inhale numerous called Malleomyces pseudomallei and Pseudomonas
virulent tubercle bacilli before they develop a pseudomallei. When inhaled by mice, virulent
microscopic tubercle that converts their tuber- B. pseudomallei produce pulmonary lesions that
culin skin test (see chapter 3), because they do usually progress to an acute septicemic form of
not convert their tuberculin reaction for many the disease followed by death (35–37). These
months, even though they live in households bacilli grow both intracellularly in phagocytes
with infectious tuberculous patients. This fact and extracellularly in both viable and damaged
indicates that most human alveolar macrophages host tissues (35, 38).Therefore, once a lesion was
are able to destroy many inhaled human-type established, the host was usually unable to stop
tubercle bacilli and thereby prevent the estab- its progression, because the endotoxins and exo-
lishment of a primary lesion. Rabbit alveolar toxins of the bacillus kill host defense cells.
macrophages destroy human-type tubercle bacilli Nevertheless, an average of 17 to 75 inhaled
even more effectively. virulent B. pseudomallei bacilli were required to
Unlike rabbits, humans evidently cannot pre- produce one grossly visible lesion (39; also see
vent an already established microscopic pul- reference 37).This finding suggests that before
monary lesion from reaching 0.5 to 1.0 mm in the bacilli could establish a microscopic pul-
size (31–33): humans are highly sensitive to the monary lesion, most of the bacilli were destroyed
tuberculin-like products of the bacillus, so that by the highly activated alveolar macrophage
early lesions rapidly develop a small caseous population normally existing in the lungs.
center (see chapter 3). Nevertheless, in 90% of In mouse melioidosis, after an aerosol infec-
humans these 0.5- to 1.0-mm lesions usually tion with B. pseudomallei, immunized mice had
become arrested and remain inactive for the fewer primary pulmonary lesions than did the
person’s lifetime. An estimated 5 to 10% of nonimmunized controls: their resistance was
12. NATURAL AIRBORNE INFECTION 䡵 227

increased 4- to 17-fold (37). In other words, bits while they are still microscopic, and even
immunization could prevent the establishment grossly apparent lesions eventually heal.
of many microscopic lesions.Apparently, immu- Although antibodies have no effect on the
nization was beneficial, because it produced establishment of pulmonary tuberculous lesions,
antibodies that opsonized B. pseudomallei, thereby they apparently reduce the progress of such
facilitating their ingestion by the alveolar lesions by accelerating the local accumulation of
macrophages. Since most alveolar macrophages macrophages and antigen-specific lymphocytes
are activated, they evidently killed inhaled B. (see chapter 5).
pseudomallei before these bacilli could multiply A comparison of the characteristics of acute
to cause an uncontrollable progressive lesion. and chronic bacterial infections is presented in
In rabbit tuberculosis, immunization has no chapter 24.
effect on the establishment of primary micro-
scopic lesions that convert the tuberculin skin REFERENCES
test, because the pulmonary alveolar macrophage 1. Lurie, M. B. 1964. Resistance to Tuberculosis: Exper-
population has no immunologic “memory.” imental Studies in Native and Acquired Defensive Mech-
Memory resides in an expanded antigen-specific anisms. Harvard University Press, Cambridge, Mass.
2. Lurie, M. B. 1941. Heredity, constitution and
lymphocyte population (see chapters 5 and 22). tuberculosis: an experimental study. Am. Rev.Tuberc.
Antibodies have little or no effect on the growth 44(Suppl. to no. 3):1–125.
of tubercle bacilli within any macrophage. 3. Lurie, M. B. 1944. Experimental epidemiology of
Tubercle bacilli are readily phagocytized by tuberculosis. Hereditary resistance to attack by
alveolar macrophages without opsonization by tuberculosis and to the ensuing disease and the
effect of the concentration of tubercle bacilli upon
antibodies (40), and the passive transfer of anti- these two phases of resistance. J. Exp. Med. 79:573–
bodies does not increase the resistance of non- 589.
immunized hosts (41). Therefore, immuniza- 4. Lurie, M. B. 1944. Experimental epidemiology of
tion against tuberculosis has no effect on the tuberculosis. The prevention of natural air-borne
establishment of a microscopic pulmonary lesion contagion of tuberculosis in rabbits by ultraviolet
irradiation. J. Exp. Med. 79:559–572.
produced by tubercle bacilli, but can prevent the 5. Lurie, M. B., and A. M. Dannenberg, Jr. 1965.
progress of such lesions into clinically active Macrophage function in infectious disease with
tuberculosis. inbred rabbits. Bacteriol. Rev. 29:466–476.
In brief, resistance to the establishment of 6. Lurie, M. B., A. G. Heppleston, S. Abramson,
pulmonary melioidosis in mice is determined by and I. B. Swartz. 1950. An evaluation of the
method of quantitative airborne infection and its use
the alveolar macrophages and is increased by in the study of the pathogenesis of tuberculosis. Am.
immunization because of opsonizing antibodies. Rev.Tuberc. 61:765–797.
Resistance to the progress of melioidosis is only 7. Allison, M. J., P. Zappasodi, and M. B. Lurie.
slightly reduced by immunization, and the dis- 1962. Host-parasite relationships in natively resistant
ease continues rapidly to death. and susceptible rabbits on quantitative inhalation
of tubercle bacilli. Am. Rev. Respir. Dis. 85:553–569.
Resistance to the establishment of pulmonary 8. Dannenberg, A. M., Jr., M. S. Burstone, P. C.
tuberculosis caused by virulent human-type bacilli Walter, and J.W. Kinsley. 1963.A histochemical
in rabbits is also determined by the alveolar study of phagocytic and enzymatic functions of
macrophages, but is not increased by immu- rabbit mononuclear and polymorphonuclear exu-
nization, because alveolar macrophages readily date cells and alveolar macrophages. I. Survey and
quantitation of enzymes, and states of cellular acti-
phagocytize tubercle bacilli without opsoniza- vation. J. Cell Biol. 17:465–486.
tion by antibodies. However, resistance to the 9. Henderson, H. J., A. M. Dannenberg, Jr., and
progress of tuberculosis is greatly reduced by the M. B. Lurie. 1963. Phagocytosis of tubercle bacilli
delayed-type hypersensitivity and cell-mediated by rabbit pulmonary alveolar macrophages and its
immunity that develop during the infection, relation to native resistance to tuberculosis.
J. Immunol. 91:553–556.
and is also reduced by prior vaccination. In fact, 10. Lurie, M. B., P. Zappasodi, and C. Tickner.
most tuberculous lesions (caused by human- 1955. On the nature of genetic resistance to tuber-
type tubercle bacilli) are usually arrested in rab- culosis in the light of the host-parasite relationships
228 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

in natively resistant and susceptible rabbits. Am. and vole bacillus (Mycobacterium microti) vaccines in
Rev.Tuberc. 72:297–323. preventing clinically apparent pulmonary tubercu-
11. Lurie, M. B. 1932. The correlation between the losis in rabbits: a preliminary report. Vaccine 19:796–
histological changes and the fate of living tubercle 800.
bacilli in the organs of tuberculous rabbits. J. Exp. 23. Dannenberg, A. M., Jr. 1991. Delayed-type
Med. 55:31–54. hypersensitivity and cell-mediated immunity in the
12. Dannenberg, A. M., Jr., O. T. Meyer, J. R. pathogenesis of tuberculosis. Immunol.Today 12:228–
Esterly, and T. Kambara. 1968.The local nature 233.
of immunity in tuberculosis, illustrated histochem- 24. Dannenberg, A. M., Jr. 1993. Immunopatho-
ically in dermal BCG lesions. J. Immunol. 100:931– genesis of pulmonary tuberculosis. Hosp. Pract.
941. 28:51–58.
13. Dannenberg, A. M., Jr. 1968. Cellular hyper- 25. Riley, R. L., W. F. Wells, C. C. Mills, W. Nyka,
sensitivity and cellular immunity in the patho- and R. L. McLean. 1957. Air hygiene in tuber-
genesis of tuberculosis: specificity, systemic and local culosis: quantitative studies of infectivity and con-
nature, and associated macrophage enzymes. Bacte- trol in a pilot ward.A cooperative study between the
riol. Rev. 32:85–102. Veterans Administration,The Johns Hopkins Uni-
14. Ando, M., A. M. Dannenberg, Jr., M. Sugi- versity School of Hygiene and Public Health, and
moto, and B. S. Tepper. 1977. Histochemical the Maryland Tuberculosis Association. Am. Rev.
studies relating the activation of macrophages to the Respir. Dis. 75:420–431.
intracellular destruction of tubercle bacilli. Am. J. 26. Riley, R. L. 2001. How it really happened.What
Pathol. 86:623–634. nobody needs to know about airborne infection.
15. Rich, A. R. 1951. The Pathogenesis of Tuberculosis, Am. J. Respir. Crit. Care Med. 163:7–8.
2nd ed. Charles C Thomas Publisher, Springfield, 27. Permutt, S. 2002. Richard Lord Riley, 1911–
Ill. 2001.An appreciation. Am. J. Respir. Crit. Care Med.
16. Canetti, G. 1955. The Tubercle Bacillus in the Pul- 166:257.
monary Lesion of Man. Springer Publishing Com- 28. Wells, W. F. 1955. Airborne Contagion and Air
pany, Inc., New York, N.Y. Hygiene. Harvard University Press, Cambridge,
17. Dannenberg,A. M., Jr., and J. F.Tomashefski, Mass.
Jr. 1998. Pathogenesis of pulmonary tuberculosis, 29. Mastorides, S. M., R. L. Oehler, J. N. Greene,
p. 2447–2471. In A. P. Fishman (ed.), Fishman’s J.T. Sinnott IV, M. Kranik, and R. L. Sandin.
Pulmonary Diseases and Disorders, 3rd ed., vol. 2. 1999.The detection of airborne Mycobacterium tuber-
McGraw-Hill Co., Inc., New York, N.Y. culosis using micropore membrane air sampling and
18. Riley, R. L., C. C. Mills, W. Nyka, N. Wein- polymerase chain reaction. Chest 115:19–25.
stock, P. B. Storly, L. U. Sultan, M. C. Riley, 30. Nardell, E. A. 1999. Air sampling for tuberculo-
and W. F. Wells. 1959. Aerial dissemination of sis—homage to the lowly guinea pig. Chest
pulmonary tuberculosis.A two-year study of con- 116:1143–1145.
tagion in a tuberculosis ward. Am. J. Epidemiol. 31. Sutherland, I., and I. Lindgren. 1979.The pro-
142:3–14. tective effect of BCG vaccination as indicated by
19. Riley, R. L., C. C. Mills, F. O’Grady, L. U. Sul- autopsy studies. Tubercle 60:225–231.
tan, F. Wittstadt, and D. N. Shivpuri. 1962. 32. Lindgren, I. 1961. Anatomical and roentgeno-
Infectiousness of air from a tuberculosis ward. Ultra- logic studies of tuberculosis infection in BCG-
violet irradiation of infected air: comparative infec- vaccinated and non-vaccinated subjects, with bio-
tiousness of different patients. Am. Rev. Respir. Dis. physical investigations of calcified foci. Acta Radiol.
85:511–525. Suppl. 209:1–101.
20. Lurie, M. B., S. Abramson, and A. G. Hep- 33. Lindgren, I. 1965.The pathology of tuberculous
pleston. 1952. On the response of genetically re- infection in BCG—vaccinated humans. Adv.Tuberc.
sistant and susceptible rabbits to the quantitative Res. 14:202–234.
inhalation of human-type tubercle bacilli and the 34. Comstock, G. W. 1982. Epidemiology of tuber-
nature of resistance to tuberculosis. J. Exp. Med. culosis. Am. Rev. Respir. Dis. 125(Suppl.):8–16.
95:119–134. 35. Dannenberg,A. M., Jr., and E. M. Scott. 1958.
21. Lurie, M. B., P. Zappasodi, E. Cardona-Lynch, Melioidosis: pathogenesis and immunity in mice and
and A. M. Dannenberg, Jr. 1952.The response hamsters. I. Studies with virulent strains of
to the intracutaneous inoculation of BCG as an Malleomyces pseudomallei. J. Exp. Med. 107:153–166.
index of native resistance to tuberculosis. J. Immunol. 36. Dannenberg,A. M., Jr., and E. M. Scott. 1958.
68:369–387. Melioidosis: pathogenesis and immunity in mice
22. Dannenberg, A. M., Jr., W. R. Bishai, N. Par- and hamsters. II. Studies with avirulent strains of
rish, R. Ruiz, W. Johnson, B. C. Zook, J. W. Malleomyces pseudomallei. Am. J. Pathol. 34:1099–
Boles, and L. M. Pitt. 2001. Efficacies of BCG 1121.
12. NATURAL AIRBORNE INFECTION 䡵 229

37. Dannenberg,A. M., Jr., and E. M. Scott. 1960. 39. Dannenberg,A. M., Jr., and E. M. Scott. 1956.
Melioidosis: pathogenesis and immunity in mice and Determination of respiratory LD50 from a number
hamsters. III.The effect of vaccination with aviru- of primary lesions as illustrated by melioidosis. Proc.
lent strains of Pseudomonas pseudomallei on the re- Soc. Exp. Biol. Med. 92:571–575.
sistance to the establishment and the resistance to 40. Schlesinger, L. S. 1996. Role of mononuclear
the progress of respiratory melioidosis caused by phagocytes in M. tuberculosis pathogenesis. J. Inves-
virulent strains: all-or-none aspects of this disease. tig. Med. 44:312–323.
J. Immunol. 84:233–246. 41. Reggiardo, Z., and G. Middlebrook. 1974.
38. Jones, A. L.,T. J. Beveridge, and D. E.Woods. Failure of passive serum transfer of immunity against
1996. Intracellular survival of Burkholderia pseudo- aerogenic tuberculosis in rabbits. Proc. Soc. Exp. Biol.
mallei. Infect. Immun. 64:782–790. Med. 145:173–175.
13
RESPONSE OF RABBITS TO
INHALED TUBERCLE BACILLI

Tuberculosis in rabbits caused by inhaled virulent bovine-type tubercle


bacilli [231]
Tuberculosis in rabbits caused by inhaled virulent human-type tubercle
bacilli [233]
Tubercles in rabbits caused by inhaled BCG [233]

Abstract.Virulent bovine-type and human-type tubercle bacilli and BCG are of decreas-
ing virulence for rabbits, in that order.The host uses the same type of immune response
to control each of these infections, but the response is more effective with bacillary strains
of reduced virulence.
With fully virulent bovine-type tubercle bacilli, only 3 bacillary units of 1 to 3 bacilli
must be inhaled to cause one grossly visible tubercle at 5 weeks. In Lurie’s inbred susceptible
rabbits, bovine-type bacilli produced the childhood form of tuberculosis with hemato-
genous dissemination. In Lurie’s resistant rabbits, bovine-type bacilli produced the adult
form of tuberculosis with pulmonary cavities and bronchial dissemination.
Human-type tubercle bacilli are not fully virulent in rabbits. In Lurie’s inbred resistant
rabbits and in commercially available New Zealand White rabbits, 300 to 1,900 bacillary
units must be inhaled to produce one grossly visible primary pulmonary tubercle at 5 weeks,
depending on the virulence of the infecting strain and on variations among the individ-
ual rabbits. In Lurie’s susceptible rabbits, such tubercles often gave rise to secondary
tubercles. In Lurie’s resistant rabbits and commercial New Zealand White rabbits, human-
type tubercle bacilli usually healed in a few months, except when they formed cavities,
which persisted much longer. However, in all rabbits, lesions produced by human-type bacilli
eventually heal and are never fatal.
BCG is avirulent in all common laboratory species. In commercial rabbits, a large inhaled
dose of aerosolized BCG produces few, if any, tiny nonprogressive tubercles. Most of the
inhaled BCG bacilli are apparently destroyed by the alveolar macrophages before they can
multiply appreciably.Without such multiplication, the degree of immunization would be
negligible. Similar to rabbits, humans should be less immunized by the inhalation of BCG
than by parenteral administration, in which higher doses of BCG can be injected and greater
bacillary multiplication can occur (because the alveolar macrophages are bypassed). In mice
and guinea pigs (which are more susceptible to Mycobacterium tuberculosis), inhaled BCG
would immunize more effectively, because their alveolar macrophages do not destroy inhaled
BCG as readily (see chapter 22).

The purpose of this chapter is to assemble in one in most cases. However, human-type bacilli are
place the characteristics of the disease produced somewhat more virulent for humans than they
in rabbits by the inhalation of virulent bovine are for rabbits, because in 5 to 10% of human
and human types of tubercle bacilli and by the cases, the primary lesion progresses or reacti-
inhalation of BCG. Other chapters in this book vates. When it does, the disease resembles that
contain the pertinent graphs and photographs. produced in rabbits by virulent bovine-type
Tuberculosis in humans usually resembles bacilli. Cavity formation with bronchial spread
that produced by virulent human-type tubercle of the disease followed by death is produced
bacilli in rabbits in that the primary lesion heals in rabbits only by virulent bovine-type bacilli.

230
13. RESPONSE TO INHALED TUBERCLE BACILLI INCLUDING BCG 䡵 231

(Cavities occasionally form in rabbits with bacillary growth stopped when tissue-damaging
human-type bacilli, but they are nonprogressive.) delayed-type hypersensitivity (DTH) to the
In brief, nonprogressive human tuberculosis tuberculin-like products of the bacillus devel-
has many of the characteristics of rabbit tuber- oped (8, 9) (see chapters 2 and 5). DTH killed
culosis caused by virulent human-type tubercle the nonactivated macrophages in which the
bacilli, and clinically progressive human tuber- bacillus was proliferating and, in doing so,
culosis has many of the characteristics of rabbit destroyed the intracellular environment that was
tuberculosis caused by virulent bovine-type so favorable to bacillary growth. In the solid
bacilli.* caseous tissue that resulted from DTH killing,
tubercle bacilli did not multiply.
TUBERCULOSIS IN RABBITS CAUSED
BY INHALED VIRULENT BOVINE- Tuberculosis in Lurie’s Susceptible
TYPE TUBERCLE BACILLI Rabbits
In Lurie’s susceptible rabbits, virulent bovine-
Overview
type tubercle bacilli grew in the poorly activated
Fully virulent bovine-type bacilli (1, 2) usually
macrophages that surrounded the caseum.Again,
produced one grossly visible pulmonary tuber-
DTH killed these parasitized host cells and
cle for each inhaled unit of 1 to 3 bacilli that
extended the area of caseous necrosis. This
reached the alveolar spaces in both susceptible
sequence continued. The bacilli entered lym-
and resistant inbred Lurie rabbits (3–5). This
phatics and spread to the hilar lymph nodes,
conclusion was derived from quantitative air-
entered blood vessels, and spread throughout
borne infection of rabbits, in which the number
the body. In the lungs, secondary tubercles devel-
of bacilli inhaled was divided by the number of
oped and were enlarged by the same process
grossly visible primary pulmonary tubercles gen-
until the host died.
erated (3–5).
Large caseous hilar lymph nodes developed in
This conclusion, however, needs some qual-
these susceptible rabbits, and tubercles formed
ification. During natural airborne infection
in the kidneys, spleen, bone marrow, and other
(when a single unit of 1 to 3 virulent bovine-
sites (see chapters 1, 2, 12, and 14). Systemic
type tubercle bacilli was inhaled over a period
DTH, an endotoxin-like syndrome, probably
of many weeks), some resistant rabbits became
contributed to the cause of death. Cavity for-
tuberculin positive and had no visible pulmonary
mation was rare.
lesions at necropsy (6, 7). In these rabbits, the sin-
gle inhaled bacillary unit was probably carried
Tuberculosis in Lurie’s Resistant
by the lymphatics from the lungs to the hilar
Rabbits
lymph nodes, where bacillary growth could be
In Lurie’s resistant rabbits, virulent bovine-type
more easily controlled (see chapter 12).
tubercle bacilli did not multiply to as high a titer
Virulent bovine-type bacilli multiplied to
as they did in Lurie’s susceptible rabbits (Fig. 1)
higher titers in susceptible rabbits than they did
(4). The logarithmic bacillary growth was
in resistant rabbits (Fig. 1) (4).The logarithmic
stopped by the same immune mechanism as
that employed by susceptible rabbits: tissue-
*Note that Lurie’s rabbit strains are now extinct (see damaging DTH to the tuberculin-like prod-
chapter 14).Therefore, some immunosuppression may be ucts of the bacillus (8, 9) (see chapters 2 and 5).
needed to reproduce the childhood form of tuberculo- The bacillary titer at the end of the logarithmic
sis in commercial adult rabbits. To our knowledge, no stage was lower than that in susceptible rabbits,
study of the susceptibility of infant rabbits to aerosolized because bacillary growth had initially been
virulent tubercle bacilli has been done. Adult guinea
pigs are natively very susceptible to tuberculosis and inhibited to a greater degree by the alveolar
would be the animal of choice for reproducing the macrophages of the resistant host (Fig. 1) (8, 9)
childhood form of tuberculosis. (see chapter 2), and these resistant rabbits more
232 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

FIGURE 1 Number of virulent human-type and bovine-type tubercle bacilli in the lungs of
inbred resistant and susceptible rabbits on various days after airborne infection. Note that bovine-
type bacilli multiplied to higher titers than did human-type bacilli, and that both types multi-
plied to higher titers in susceptible strain C rabbits than they did in resistant strain III rabbits. Repro-
duced with permission from reference 4.

quickly developed effective cell-mediated Because of the good CMI, the hilar lymph
immunity (CMI). nodes showed minimal disease. Secondary lym-
The good CMI of the resistant rabbits acti- phogenous and hematogenous lesions in the
vated the macrophages surrounding the solid lungs and other organs occurred but were usu-
caseous centers of the tubercles (8–10) (see ally nonprogressive.
chapter 2). Therefore, when tubercle bacilli Frequently, however, the caseous centers in the
escaped from the edge of the caseum, they were resistant host underwent liquefaction, a process
often engulfed by activated macrophages that in which fluid was absorbed and the caseous
inhibited their intracellular growth (in contrast material softened. In the liquefied menstruum,
to the situation with susceptible rabbits).These the bacilli sometimes multiplied extracellularly to
tubercles contained mature epithelioid cells, tremendous numbers (1, 6). The host’s DTH
which are activated macrophages capable of reaction to the tuberculin-like products of such
inhibiting tubercle bacilli. In the resistant rabbits, bacilli was able to cause much tissue destruction
caseation progressed slowly and was less exten- (see chapters 2 and 12). When this occurred,
sive than that in the susceptible rabbits. the walls of the bronchial tree were frequently
Histologically, the lesions usually contained a eroded; the bacilli entered the air passages and
caseous center, surrounded by tuberculous gran- spread to other parts of the lungs, where they
ulation tissue composed of macrophages, lym- sometimes produced areas of caseous pneumo-
phocytes, plasma cells, fibroblasts, capillaries, and nia, as well as new lesions that underwent liq-
lymphatics (1, 6, 11; see also references 12 and uefaction (see chapter 4).The large number of
13). In time, the lesions became encapsulated by virulent bovine-type bacilli in the airways over-
fibrous tissue (1; see also references 12 and 13). whelmed the resistant rabbit’s ability to control
13. RESPONSE TO INHALED TUBERCLE BACILLI INCLUDING BCG 䡵 233

the disease, so it progressed until the host suc- tible rabbits (5).The tubercles in the resistant ani-
cumbed. But for liquefaction and cavity forma- mals were usually smaller and contained fewer
tion, Lurie’s inbred resistant rabbits would prob- bacilli, and their caseous centers were more
ably have arrested the infection. mature; i.e., the nuclear debris present was more
completely disintegrated. The cells present in
Tuberculosis in Commercial New these tubercles were located more interstitially,
Zealand White Rabbits i.e., within the alveolar walls (5). In time, some
In commercial New Zealand White rabbits, of the caseous centers liquefied and even formed
aerosolized virulent bovine-type tubercle bacilli cavities (5), but healing was relatively rapid (16),
produce a disease that resembles that found in calcification sometimes occurred (14), and sec-
Lurie’s resistant rabbits (12, 13) (see chapter 4). ondary lesions were rare (16). (See Table 2 in
chapter 14.)
TUBERCULOSIS IN RABBITS CAUSED When compared with tubercles in the resis-
BY INHALED VIRULENT HUMAN- tant rabbits, tubercles in the susceptible rabbits
TYPE TUBERCLE BACILLI were usually larger and contained more bacilli.
See references 5, 11, 14, and 15. Human-type Their caseous centers were less mature; i.e., the
tubercle bacilli are never fully virulent in rabbits. nuclear debris present was incompletely disin-
After an aerosol infection with strain H37Rv (a tegrated.The cells present in the tubercles of the
standard laboratory strain of virulent human- susceptible rabbits were located more intra-
type tubercle bacillus), the lesions produced alveolarly; i.e., many were within the air spaces
even in Lurie’s genetically susceptible rabbits (5). None of the caseous centers liquefied, and
eventually regressed (1). cavities never formed (5). Healing was slow,
The use of such aerosolized human-type bacilli and secondary lesions of lymphogenous and
enabled Lurie to develop his tubercle-count hematogenous origin were common (1, 16).
method, which is the most quantitative method (See Table 2 in chapter 14.)
for assaying innate and acquired resistance to Commercial New Zealand White rabbits
tuberculosis in rabbits as well as the virulence of responded to the inhalation of virulent human-
the infecting tubercle bacillus (see chapter 11). In type tubercle bacilli in a manner similar to that
brief, rabbits were exposed to a known quantity of Lurie’s inbred resistant rabbits (15).The Erd-
of aerosolized bacilli, and the number of pri- man strain was more virulent than H37Rv (15),
mary tubercles in the lungs was counted 5 weeks but not nearly as virulent as the virulent bovine-
later.Then, the number of inhaled human-type type Ravenel strain. The Thorbecke strain of
tubercle bacilli required to produce one visible inbred rabbits (Covance Inc., Denver, Pa.) was
primary pulmonary tubercle (called “the ratio”) more susceptible than standard New Zealand
was calculated. In Lurie’s inbred rabbits, 50 to White rabbits (17),but not as susceptible as Lurie’s
1,500 inhaled virulent human-type bacilli inbred susceptible rabbits (see chapter 14).
(H37Rv) had to be inhaled to produce such a
tubercle,depending on the innate resistance of the TUBERCLES IN RABBITS
host.Therefore, a wide range of ratios was pro- CAUSED BY INHALED BCG
vided by using human-type bacilli in rabbits. BCG is much less virulent for rabbits than
With fully virulent bovine-type bacilli, a ratio of even human-type tubercle bacilli (1, 2). The
about 3 always occurred, regardless of the host’s inhalation of several million BCG bacilli was
innate resistance. Therefore, the tubercle count required to produce a single primary pul-
method with bovine-type bacilli is not recom- monary tubercle, which was quite small and
mended for assaying the immunity produced by should have healed rapidly (S. Abramson,
new vaccines (see chapter 11). unpublished data; quoted on page 223 of ref-
Following the inhalation of virulent human- erence 1). In mice and guinea pigs, the inhala-
type bacilli (H37Rv), Lurie’s resistant rabbits tion of BCG produces tubercles more readily
developed fewer tubercles than did his suscep- (see chapters 15 and 22).
234 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

The lungs were not cultured for tubercle 9. Dannenberg, A. M., Jr. 1993. Immunopatho-
bacilli. Therefore, we do not know whether genesis of pulmonary tuberculosis. Hosp. Pract.
28:33–40 (Off. ed. 51–58).
BCG would multiply (in nonactivated
10. Dannenberg, A. M., Jr. 1968. Cellular hyper-
macrophages) during the logarithmic phase at sensitivity and cellular immunity in the patho-
the same rate as did the virulent bovine and genesis of tuberculosis: specificity, systemic and local
human types shown in Fig. 1. However, we nature, and associated macrophage enzymes. Bacte-
would predict from Fig. 1 that the logarithmic riol. Rev. 32:85–102.
phase of BCG growth would be shorter than the 11. Lurie, M. B. 1932. The correlation between the
histological changes and the fate of living tubercle
logarithmic phase of the two more virulent bacilli in the organs of tuberculous rabbits. J. Exp.
strains, and that BCG would not reach as high Med. 55:31–54.
a titer.The characteristics of the disease produced 12. Converse, P. J., A. M. Dannenberg, Jr., J. E.
in rabbits by intravenous BCG are presented in Estep, K. Sugisaki,Y.Abe, B. H. Schofield, and
references 1 and 18. M. L. M. Pitt. 1996. Cavitary tuberculosis pro-
duced in rabbits by aerosolized virulent tubercle
bacilli. Infect. Immun. 64:4776–4787.
REFERENCES 13. Converse, P. J., A. M. Dannenberg, Jr.,
1. Lurie, M. B. 1964. Resistance to Tuberculosis: Exper- T. Shigenaga, D. N. McMurray, S. W. Phalen,
imental Studies in Native and Acquired Defensive Mech- J. L. Stanford, G. A. W. Rook, T. Koru-
anisms. Harvard University Press, Cambridge, Mass. Sengul, H. Abbey, J. E. Estep, and M. L. M.
2. Lurie, M. B., and A. M. Dannenberg, Jr. 1965. Pitt. 1998. Pulmonary bovine-type tuberculosis
Macrophage function in infectious disease with in rabbits: bacillary virulence, inhaled dose effects,
inbred rabbits. Bacteriol. Rev. 29:466–476. tuberculin sensitivity, and Mycobacterium vaccae
3. Lurie, M. B., A. G. Heppleston, S. Abramson, immunotherapy. Clin. Diagn. Lab. Immunol. 5:871–
and I. B. Swartz. 1950. An evaluation of the 881.
method of quantitative airborne infection and its use 14. Heppleston, A. G. 1949. Quantitative air-borne
in the study of the pathogenesis of tuberculosis. Am. tuberculosis in the rabbit.The course of human-type
Rev.Tuberc. 61:765–797. infection. J. Exp. Med. 89:597–610.
4. Allison, M. J., P. Zappasodi, and M. B. Lurie. 15. Manabe, Y. C., A. M. Dannenberg, Jr., S. K.
1962. Host-parasite relationships in natively resistant Tyagi, C. L. Hatem, M.Yoder, S. C.Woolwine,
and susceptible rabbits on quantitative inhalation B. C. Zook, M. L. M. Pitt, and W. R. Bishai.
of tubercle bacilli. Am. Rev. Respir. Dis. 85:553–569. 2003. Different strains of Mycobacterium tuberculosis
5. Lurie, M. B., P. Zappasodi, and C. Tickner. cause various spectrums of disease in the rabbit
1955. On the nature of genetic resistance to tuber- model of tuberculosis. Infect. Immun. 71:6004–6011.
culosis in the light of the host-parasite relationships 16. Lurie, M. B., S. Abramson, and A. G. Hep-
in natively resistant and susceptible rabbits. Am. pleston. 1952. On the response of genetically re-
Rev.Tuberc. 72:297–323. sistant and susceptible rabbits to the quantitative
6. Lurie, M. B. 1941. Heredity, constitution and inhalation of human-type tubercle bacilli and the
tuberculosis: an experimental study. Am. Rev.Tuberc. nature of resistance to tuberculosis. J. Exp. Med.
44(Suppl. to no. 3):1–125. 95:119–134.
7. Lurie, M. B. 1944. Experimental epidemiology of 17. Dorman, S., C. L. Hatem, S. Tyagi, K. Aird,
tuberculosis. Hereditary resistance to attack by J. Lopez-Molina, M. L. M. Pitt, B. C. Zook,
tuberculosis and to the ensuing disease and the A. M. Dannenberg, Jr.,W. R. Bishai, and Y. C.
effect of the concentration of tubercle bacilli upon Manabe. 2004. Susceptibility to tuberculosis: clues
these two phases of resistance. J. Exp. Med. 79:573– from studies with inbred and outbred New Zealand
589. White rabbits. Infect. Immun. 72:1700–1705.
8. Dannenberg,A. M., Jr. 1991. Delayed-type hyper- 18. Lurie, M. B. 1934.The fate of BCG and associ-
sensitivity and cell-mediated immunity in the patho- ated changes in organs of rabbits. J. Exp. Med.
genesis of tuberculosis. Immunol.Today 12:228–233. 60:163–178.
14
CHARACTERISTICS OF RESISTANCE AND
SUSCEPTIBILITY TO TUBERCULOSIS
IN LURIE’S INBRED RABBITS

History and description of Lurie’s inbred rabbits [235]


Resistance and susceptibility to tuberculosis in Lurie’s inbred
rabbits [237]
Genetic experiments with Lurie’s inbred rabbits [238]
Rate of healing of dermal BCG lesions as a method of determining the
native resistance of inbred rabbit strains [241]
Inbred rabbits today [241]
Use of rabbits for studies other than tuberculosis [244]

Abstract. Lurie’s rabbits were inbred for either susceptibility or resistance to the progress
of tuberculosis.When infected with virulent bovine-type tubercle bacilli, the susceptible
rabbits developed a rapidly progressing, hematogenously spreading “childhood type” of
tuberculosis, and the resistant rabbits developed a slowly progressing, cavitary, bronchial-
spreading “adult type” of tuberculosis.
In lesions produced by virulent bovine-type bacilli, by human-type bacilli, and by BCG,
the same manifestations of genetic resistance to tuberculosis were evident histologically:
mature epithelioid cells (now known as highly activated macrophages) were always more
numerous in the lesions of resistant rabbits than in the lesions of susceptible rabbits, irre-
spective of the differences in virulence of the infecting bacillary strain.
The genetic resistance of these rabbits resides in their ability to activate macrophages
to control the growth of tubercle bacilli, both nonspecifically and immune-specifically.
Crossbreeding showed that the genetic resistance to tuberculosis is multifactorial, with genes
associated with resistance being dominant over susceptibility genes.
The commercially available outbred New Zealand White rabbits seem almost as resistant
as Lurie’s inbred resistant strain III rabbits.Thorbecke inbred rabbits were distinctly more sus-
ceptible than commercial outbred rabbits, but apparently not as susceptible as Lurie’s inbred
C and FC rabbits. van Zutphen’s inbred rabbits (which are hypo- and hyperresponsive to
dietary cholesterol,respectively) have not been adequately studied for resistance to tuberculosis.

HISTORY AND DESCRIPTION OF the specifics of the inbreeding, are presented in


LURIE’S INBRED RABBITS his book (1) and his monograph (2).Table 1 lists
The rabbit is the only available laboratory ani- the origins and the degree of resistance to tuber-
mal species in which tuberculous lesions readily culosis of each of Lurie’s rabbit strains.
liquefy, form cavities, and spread bacilli through Between 1948 and 1962, the most important
the bronchial tree (1). In fact, depending on the strains in Lurie’s rabbit colony were the resistant
genetic resistance of the host and the virulence strain III and the susceptible strains C and FC.
of the infecting bacilli, tuberculosis in rabbits Strain III needed to inhale about 1,070 virulent
can mimic all forms of the human disease. human-type bacilli (H37Rv) to generate one
For this reason, in the 1930s, Lurie began grossly visible primary pulmonary tubercle 5
inbreeding different rabbit strains for either re- weeks after infection; strain C needed about
sistance or susceptibility to tuberculosis, and he 70; strain FC needed about 80; and strain AD (of
continued this inbreeding throughout the rest of intermediate resistance) needed about 360
his professional career (1, 2). How he began, and (Table 1) (3). The inbreeding of these rabbit

235
236 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

TABLE 1 Relative resistance of Lurie’s inbred rabbit strains, determined by their survival after a standard
intracutaneous inoculation of virulent bovine-type tubercle bacilli (Ravenel), by their response to the quantita-
tive inhalation of human-type tubercle bacilli (H37Rv), or by both
Average survival No. of inhaled human References for
after infection bacilli required to resistance to
Derivation with bovine form one tubercle tuberculosis
Strain bacilli (the “ratio”)
and reference(s)
No. of No. of Bovine Human
Days rabbits Ratios rabbits bacilli bacilli
Ca Carworth farms (26) — — 49 ⫾ 19 9 — (26)
Ca Swift stock (2) 121 5 70 ⫾ 14 30 (2) (3)
CaC Ca ⫻ C (3, 27) — — 97 ⫾ 12 7 — (1)
F Swift stock (2) 141 4 — (2) —
FCa F ⫻ C (26) 132 8 79 ⫾ 9 45 (26) (3)
FCCab FC ⫻ CaC (28) — — 44 ⫾ 3 6 — —
Ac Stubbs (2) 539 6 — (2) —
ADc,d A ⫻ D (29) 197 10 362 ⫾ 103 12 (3) (3)
III (T)a Sawin (26) 422 8 1,065 ⫾ 138 71 (26) (3)
TAc T ⫻ A (30) — — 154 ⫾ 27 9 — (30)
TTC TC ⫻ T (14) — — 315 ⫾ 67 12 — (30)
NZWe Covance, Inc. 500–1,500 26 (31)
Thorbeckef,g Covance, Inc. 100–500 6 (24)
a
The majority of Lurie’s experiments were done with these strains. Strain III was formerly called strain T. The mean ratios
(by Lurie’s method; see chapter 11) and their standard errors are listed. Data from reference 4.
b
See references 9 and 4 for other information on this strain.
c
The resistance of strains A and AD has declined since the ninth and seventh generations, respectively (1).
d
The D strain of Swift stock was of intermediate resistance (2).
e
NZW are commercially available New Zealand White rabbits.
f
Thorbecke rabbits were inbred by the late G. Jeanette Thorbecke, Professor of Pathology, New York University, School of Med-
icine.They were available from Covance, Inc., Denver, PA.
g
Estimated ratios.

strains was not complete, when evaluated by for size and pelt characteristics. Upon speaking
rejection of skin grafts, blood types, and other with a member of this association, I was left with
criteria (see reference 4). the impression that these strains were far from
The susceptible strain C and resistant strain III inbred, and that some were even crossbred with
(formerly called T) were albino rabbits.The sus- other strains to maintain the desired size and coat
ceptible strains Ca and FC and the strain AD of characteristics. No one, to date, has measured the
intermediate resistance were Dutch-belted rab- overall resistance to tuberculosis of rabbit strains
bits. Strain A was English in its coat markings; certified by the American Rabbit Breeders Asso-
it was initially resistant, but had become inter- ciation, or even determined whether their
mediate over the years. response to the inhalation of virulent human-
Whether any correlation exists between coat type tubercle bacilli is more uniform than that
markings and native resistance to tuberculosis found with commercially available New Zealand
remains to be determined but seems worthy of White rabbits. Increased uniformity in resis-
investigation, because more than 50 registered tance to tuberculosis would simplify the evalu-
rabbits strains are available today.These strains are ation of new tuberculosis vaccines in the rabbit
described in a publication by the American species, because fewer animals would be required
Rabbit Breeders Association (5).They were bred to obtain significant results.
14. CHARACTERISTICS OF RESISTANCE AND SUSCEPTIBILITY 䡵 237

RESISTANCE AND SUSCEPTIBILITY dendritic cells, macrophages, lymphocytes, and


TO TUBERCULOSIS IN LURIE’S other leukocytes (see chapters 19 and 20).
INBRED RABBITS The primary pulmonary lesions of the resis-
Characteristics of resistance and susceptibility tant rabbits show more interstitial inflammation
to tuberculosis in Lurie’s inbred rabbits are listed (3), which is apparently the cause of the increased
in Table 2, and the details are presented in chap- drainage of tubercle bacilli to their hilar lymph
ters 2, 11, 12, and 13.The basic tissue responses nodes.The susceptible rabbits show less intersti-
of Lurie’s resistant and susceptible rabbits to tial inflammation and more pneumonic inflam-
tubercle bacilli were independent of the viru- mation, i.e., cells in the alveolar spaces.
lence of the infecting bacilli, because tuberculous Mature epithelioid cells are highly activated
lesions produced by highly virulent bovine-type macrophages (1, 6–8).They are more numerous
bacilli, semivirulent human-type bacilli, and in the lesions of the resistant host. Lurie recog-
BCG vaccines showed similar characteristics. nized mature epithelioid cells in tissue sections
In brief, the main inbred characteristic of by their rounded appearance and homogeneous
Lurie’s resistant rabbits was their ability to activate cytoplasm (1, 10). Our -galactosidase histo-
macrophages to such a high degree that these chemical procedure to identify mature epithe-
macrophages could inhibit and/or destroy tuber- lioid cells (11, 12) was not available when Lurie
cle bacilli. His resistant rabbits nonspecifically was alive.
activated their pulmonary alveolar macrophages Maturation of the caseous process seems to
to a greater degree than did his susceptible rab- be due to greater hydrolysis of the necrotic
bits (1, 6–8).Within tuberculous lesions, his resis- material—probably due to increased levels of
tant rabbits also immunologically activated their hydrolases in the macrophages of the resistant
blood-borne macrophages to a greater degree hosts.
than did his susceptible rabbits (1, 6–8). (Blood- Increased numbers of lymphocytes and plasma
borne macrophages enter tuberculous lesions cells are in the lesions of the resistant rabbits (13).
and become activated by cytokines, mainly from The plasma cells there and in the lymph nodes are
antigen-specific lymphocytes.) the source of the greater antibody titers in the re-
In Table 2, note (as detailed in the following sistant rabbits (13).These rabbits may have more
paragraphs) how many characteristics of the re- efficient antigen presentation by dendritic cells
sistant rabbits were due to the greater activation and macrophages (see chapters 5 and 6).
developed by their macrophages. Fewer and smaller primary tubercles, fewer
The increased trapping of inhaled bacilli in secondary tubercles, and more rapid healing are
the lungs by resistant rabbits (see chapter 12) is due to the decrease in the number of bacilli
possibly due to their alveolar macrophages’ being caused by the highly activated macrophages of
more actively phagocytic for inhaled bacilli (9). the resistant group.
The increased inhibition of bacillary multi- Liquefaction of solid caseous tissues and cav-
plication seems to be directly related to the ity formation in the resistant rabbits seem related
degree of activation of alveolar macrophages, and to high levels of hydrolytic enzymes in dead and
the subsequent inhibition in tuberculous lesions live activated macrophages surrounding the
seems to be directly related to the degree of acti- lesion’s caseous center.
vation of blood-borne macrophages. However, The faster rate of healing of the lesions, the
tubercle bacilli grow at equal rates in nonacti- stronger acquired immunity, and the increased
vated macrophages of both resistant and sus- longevity (after infection with virulent bovine-
ceptible rabbits (see chapters 2 and 15). See type bacilli) in the resistant host are mostly due
below for drainage of bacilli to lymph nodes. to many highly activated macrophages capable
Mononuclear cells accumulate (and activate) of destroying or inhibiting tubercle bacilli.
more rapidly in the early pulmonary lesions of The increased antibody titers in the resistant
resistant rabbits. Activated macrophages (and rabbits hasten the local acquired cell-mediated
other cells) produce chemokines that attract immune response (see chapter 5).
238 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

TABLE 2 Characteristics of resistance and susceptibility to tuberculosis in Lurie’s inbred rabbitsa


Resistance Susceptibility
Characteristic Rabbit strain c
Rabbit strain
Degreeb and reference(s) Degreeb and reference(s)
Bacilli
Trapping of tubercle bacilli in lung ⫹⫹⫹⫹ A (33); III (3, 9, 32) ⫹⫹ F(7); C(3, 9, 32, 33); FCCa (9)
Initial inhibition and/or destruction ⫹⫹⫹ III (3, 32) ⫹ C (3, 32)
of inhaled bacilli
Subsequent inhibition of bacillary ⫹⫹⫹⫹ III (3, 32) ⫹⫹ C (3, 32)
multiplication
Drainage of bacilli to ⫹⫹⫹ III (3, 32) ⫹ C (3, 32)
tracheobronchial lymph nodes
Histopathology
Rate of accumulation of ⫹⫹⫹ III (3) ⫹ C (3)
mononuclear cells in early
pulmonary lesions
Interstitial inflammation ⫹⫹⫹ III (3) ⫹ C (3)
Pneumonic inflammation ⫹ III (3) ⫹⫹⫹ C (3)
Rate of epithelioid
cell maturation ⫹⫹⫹⫹ A (2); III (3, 13, 26) ⫹ C (2, 3, 13, 26); F (2)
Maturation of caseous process ⫹⫹⫹⫹ III (3) ⫹⫹ C (3)
Number of lymphocytes and ⫹⫹⫹ III (3, 13) ⫹ C (3, 13)
plasma cells
Gross pathology
Number of gross tubercles 5 weeks ⫹ III (3, 13, 14, 26) ⫹⫹⫹⫹ C (3, 14); Ca (26); FC (3, 13, 26)
after inhalation of human bacilli
Size of tubercles and their caseous ⫹⫹ III (3) ⫹⫹⫹⫹ C (3); FC (3)
centers
Spread of disease to kidneys and ⫹ A (2); III (1, 32) ⫹⫹⫹ F (2); C (2)
other organs
Liquefaction and cavity formation ⫹⫹ A (2); III (3) ⫾ F (2); C (2,3)
Rate of healing of lesionsd ⫹⫹⫹⫹ A (2); III (26) ⫹ F (2); C (2); FC (26)
Absorption of tubercles and ⫹⫹⫹⫹ III (3) ⫹ C (3)
fibroplasiasd
Other factors
Amount of acquired immunity ⫹⫹⫹⫹ A (2); III (13) ⫹ F (2); C (2); FC (13)
Longevity after infection with ⫹⫹⫹⫹ A (2, 33); III (26) ⫹ F (2, 33); C (2, 33); FC (1, 32)
virulent bovine-type bacilli
Antibody titer ⫹⫹⫹⫹ III (13) ⫹⫹ FC (13)
a
Adapted from reference 4, where other characteristics of Lurie’s inbred rabbits are described.
b
⫹, low degree; ⫹⫹ and ⫹⫹⫹, intermediate degrees; ⫹⫹⫹⫹, high degree.
c
Strain III was formerly called strain T.
d
Virulent human-type tubercle bacilli are never fully virulent for rabbits, because pulmonary lesions caused by these bacilli
eventually heal. However, pulmonary lesions caused by fully virulent bovine-type tubercle bacilli never heal.

GENETIC EXPERIMENTS WITH Backcrossing the F1 generation (III ⫻ C) to


LURIE’S INBRED RABBITS resistant strain III produced rabbits (IIIC ⫻ III)
Crossbreeding resistant strain III with suscepti- that were just as resistant as the original strain III
ble strain C produced an F1 generation (III ⫻ rabbits (14) (Fig. 1 and 2). However, backcross-
C) of intermediate resistance (14) (Fig. 1 and 2). ing the F1 generation (III ⫻ C) to susceptible
14. CHARACTERISTICS OF RESISTANCE AND SUSCEPTIBILITY 䡵 239

FIGURE 1 Lungs of (top) an F1 hybrid (IIIC ⫽


III ⫻ C), (middle) its backcross to III (IIIC ⫻ III),
and (bottom) its backcross to C (IIIC ⫻ C) 5
weeks after the inhalation of 75,000, 79,000, and
64,000 human-type tubercle bacilli (H37Rv).The
lungs of IIIC 1-31 (top), IIIC III 1-16 (middle),
and IIIC C 1-4 (bottom) contained 497, 142, and
772 tubercles, respectively; the ratios for these rab-
bits were 131, 557, and 82, respectively. See Fig. 2
for the means for each of these combinations.
Reproduced with permission from reference 14.

strain C did not regain appreciable susceptibil- This hybrid was more resistant than either par-
ity: this backcross (IIIC ⫻ C) had intermediate ent: C12⫽3 (with a ratio of 41) crossed with
resistance to tuberculosis (14) (Fig. 1 and 2). Ca5⫽41 (with a ratio of 37) produced CaC
Lurie also crossed two susceptible strains (C hybrids with a mean ratio of 97 ⫾ 12 (P ⫽
with Ca) to produce a first-generation hybrid. 0.001) (see reference 1, p. 240–241).
240 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

FIGURE 2 Ratios of individual rabbits from resistant strain III, susceptible strain C, and
hybrids III ⫻ C, IIIC ⫻ C, and IIIC ⫻ III.The ratio is the number of inhaled human-type bacilli
(H37Rv) required to generate one grossly visible primary tubercle; i.e., the ratio is the number
of bacilli inhaled by each rabbit divided by the number of primary tubercles found in its lungs
5 weeks later.The mean ratios (by the Lurie method; see chapter 11) with their standard errors
(and log10) are listed on the right.
The original resistant and susceptible rabbit strains were III and C, respectively.The F1 hybrids
were III ⫻ C, and the backcrosses were IIIC ⫻ C and IIIC ⫻ III, respectively.The resistance of
the F1 generation backcrossed to resistant III rabbits was equal to that of the original III group
(ratios of 620 ⫾ 110 and 640 ⫾ 170, respectively). However, the resistance of the F1 generation
backcrossed to susceptible C rabbits was far greater than that of the original C group (ratios of
243 ⫾ 51 and 48 ⫾ 10, respectively). In other words, many of the factors controlling good re-
sistance appeared to be dominant.
This figure also shows the ratio as a log10 (and its arithmetic mean) for the rabbits in this exper-
iment; each subsection represents one rabbit. For example, the IIIC ⫻ C contained 21 rabbits
with ratios of 101.6 to 103.4, and the IIIC ⫻ III group contained 26 rabbits with ratios of 101.9
to 103.7. Note that the resistance of the F1 generation seemed to be less variable than that of the
other groups; i.e., it ranged over five rather than six 0.3 log10 increments (but this result may have
been a chance phenomenon).The shaded boxes represent the rabbits shown in Fig. 1.
Adapted from reference 14.
Note the variation in tubercle counts among the individual rabbits.
14. CHARACTERISTICS OF RESISTANCE AND SUSCEPTIBILITY 䡵 241

From these crossbreeding experiments, Lurie by standard radiographs, or even by computer-


concluded that resistance to tuberculosis is mul- analyzed tomography.
tifactorial or polygenic, with the resistance genes
dominating the susceptibility genes (1, 14). INBRED RABBITS TODAY
Alternatively, susceptibility may be an absence of
certain resistance factors (1, 14).Therefore, re- Fate of Lurie’s Inbred Rabbits
sistance to tuberculosis is “multiple, complex Unfortunately, Lurie’s rabbit strains became
and additive in nature” (14). infertile with time. Strain III rabbits readily
A similar dominance of resistance over sus- became pregnant, but the fetuses failed to com-
ceptibility may exist in humans and would partly plete development and usually were absorbed.
explain why immunocompetent humans have The last viable strain III (white) rabbits were
apparently become more resistant to tuberculosis produced in commercial black rabbits by trans-
over the centuries. planting early-stage strain III embryos (unpub-
lished data).Two- to 8-cell embryos were col-
lected from the fallopian tubes of a female strain
RATE OF HEALING OF DERMAL III rabbit within a few days after mating with a
BCG LESIONS AS A METHOD OF male strain III rabbit. The embryos were then
DETERMINING THE NATIVE RESIS-
TANCE OF INBRED RABBIT STRAINS placed in the fallopian tubes of a chorionic
About 1950, Lurie used the time of healing of gonadotropin-treated female black rabbit. The
dermal BCG lesions, instead of the tubercle- black surrogate mother gave birth to white
count method, to select for breeding the most strain III rabbits. Since the black color is dom-
resistant and the most susceptible rabbits and inant, the origin of the offspring could not be
thereby maintain these characteristics in the questioned.
offspring (1, 13).When BCG was used for such
selection in Lurie’s rabbit colony, there was no van Zutphen Rabbits
apparent spread of the BCG infection to other L. F. M. van Zutphen, at the Faculty of Veteri-
rabbits in the colony, because every unvaccinated nary Medicine of the University of Utrecht in
rabbit tested remained tuberculin negative. In The Netherlands, has inbred a relative
other words, even though the BCG lesions (IIIVO/JU) of Lurie’s resistant strain III rabbits
would ulcerate and spread occasional BCG (8, 15, 16). It is hyporesponsive to dietary choles-
bacilli into the air of the room, the other rab- terol (16). van Zutphen also maintains another
bits in the breeding colony did not inhale a inbred rabbit strain (AX/JU) that is hyperre-
sufficient quantity of these bacilli to establish a sponsive to dietary cholesterol (16). Neither of
primary lesion in which the BCG multiplied these inbred rabbit strains has been evaluated for
(see chapter 13). (Lurie’s rabbits were housed resistance to tuberculosis by Lurie’s tubercle-
individually in cages with grates that allowed count method with aerosolized virulent human-
urine and feces to pass through.) type tubercle bacilli (17) (see chapter 11). Strain
Because the time of healing of BCG lesions IIIVO/JU resembled Lurie’s inbred strain III
cannot be precisely determined, it provides only in its resistance to tuberculosis produced by vir-
a rough measure of host resistance. Healing is too ulent bovine-type tubercle bacilli (18, 19), but
gradual a process and is in part dependent on the susceptibility of strain AX/JU to tuberculosis
whether or not the lesion ulcerates and dis- with bovine-type bacilli was never tested.
charges much of its liquefied contents contain-
ing bacilli.Therefore, the tubercle-count method
(see chapter 11) is a more precise way of mea- Bar Harbor Rabbits
suring the genetic resistance of different strains The Jackson Laboratories (Bar Harbor, Me.) had
of rabbits.Tubercle counting, however, requires several inbred rabbit strains of unknown resistance
euthanizing the animal, because not all grossly to tuberculosis (20–22) but has stopped inbreed-
visible pulmonary tubercles can be identified ing rabbits. Richard R. Fox of these laboratories
242 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

preserved a few of these inbred strains in a frozen produce the resistant form of tuberculosis with
state as early embryos in the 4- to 8-cell stage (8). cavity formation (18, 19).
These embryos can be made to continue their
development by placing them into the fallopian Thorbecke Rabbits
tubes of living female rabbits primed with chori- Another New Zealand White inbred rabbit
onic gonadotropin (see above).This frozen stock strain was developed by the late G. Jeanette
contains embryos of the original resistant strain Thorbecke at New York University (unpub-
III rabbits from which both Lurie and van lished) and maintained by Covance Research
Zutphen developed their inbred colonies. Products, Inc. These Thorbecke rabbits had
stunted facies and groomed themselves poorly,
Commercial Rabbits and they tended to be more excitable (similar to
With the tubercle-count method (23, 24) (see Lurie’s susceptible FC and CaC rabbit strains).
chapter 11), we evaluated the resistance of out- The group headed by Yukari C. Manabe
bred commercial New Zealand White rabbits tested the resistance of these Thorbecke inbred
(from Covance Research Products, Inc., Denver, rabbits by the tubercle-count method with
Pa.) (Tables 1 and 3).They seem to be highly re- human-type tubercle bacilli (24).When com-
sistant to tuberculosis, perhaps just as resistant as pared with commercial outbred New Zealand
Lurie’s strain III rabbits (18, 19). However, the White rabbits, the Thorbecke inbred rabbits
H37Rv strain of human-type tubercle bacilli were found to be more susceptible to tubercu-
used by Lurie and by us was never evaluated for losis (24); i.e., they had more numerous primary
virulence in the same experiment.Therefore, we pulmonary tubercles 5 weeks after aerosol infec-
do not know whether Lurie’s strain III rabbits tion with human-type tubercle bacilli (H37Rv)
were more resistant than the commercial out- (Table 3) (24). These tubercles were larger,
bred rabbits available today.We do know, how- showed more caseous necrosis and surrounding
ever, that, when exposed to bovine-type tuber- pneumonitis, and contained more bacilli (Tables
cle bacilli, commercial outbred rabbits readily 3 and 4).Their hilar lymph nodes were larger

TABLE 3 Tuberculosis in inbred Thorbecke rabbits and outbred commercial rabbits 5 weeks after aerosol
infection with Mycobacterium tuberculosis H37Rv (luciferase)a
Inbred Outbred P values
No. of rabbits 12 6
No. of tubercles 98 ⫾ 12 33 ⫾ 13 0.007
Average diameter of tubercles (mm) 2.73 ⫾ 0.13 1.43 ⫾ 0.09 0.001
No. of bacilli inhaled divided by the number of 128b 323b
tubercles produced (the ratio)
Dermal tuberculin reactions (mm3) 289 ⫾ 35 1,152 ⫾ 348 0.009
No. of viable bacilli cultured from hilar lymph nodes 697 ⫾ 192 26 ⫾ 20 0.004
No. of viable bacilli cultured from each representative 809 ⫾ 210 215 ⫾ 115 0.027
tubercle
No. of viable bacilli cultured from entire right upper 11,475 ⫾ 9,448 1,154 ⫾ 986 0.003
lobe of lungs
a
Two-day dermal reactions to Old Tuberculin were measured at 5 weeks after the aerosol infection. Data from reference 24.
b
Calculated from the means.
H37Rv (luciferase) is a genetically engineered strain of H37Rv. It seemed to have the same virulence (for rabbits) as the par-
ent H37Rv strain by the tubercle count method (24), but it and the parent strain were not compared in the same aerosol exper-
iment.The means and their standard errors are listed.
14. CHARACTERISTICS OF RESISTANCE AND SUSCEPTIBILITY 䡵 243

TABLE 4 Histologic characteristics of tubercles from inbred Thorbecke rabbits and outbred commercial rab-
bits 5 weeks after aerosol infection with M. tuberculosis (Erdman)a
Inbred rabbits Outbred rabbits P values
Size of tubercles, mm 2b
2.8 ⫾ 0.2 1.8 ⫾ 0.2 0.020
Percentage of tubercles showing caseous necrosis 89 ⫾ 3 60 ⫾ 11 0.045
Area of necrosis in tubercles, mm2 2.6 ⫾ 0.5 0.6 ⫾ 0.2 0.004
No. of neutrophils in tubercles (1⫹ to 4⫹) 2.2 ⫾ 0.3 1.7 ⫾ 0.4 NS
Percentage of mature epithelioid macrophages 58 ⫾ 5 72 ⫾ 3 0.066
No. of lymphocytes and plasma cells in tubercles (1⫹ to 4⫹) 1.3 ⫾ 0.2 3.7 ⫾ 0.2 0.004
Peripheral fibrosis in tubercles (1⫹ to 4⫹) 2.8 ⫾ 0.5 1.8 ⫾ 0.4 NS
Perigranuloma pneumonitis (1⫹ to 4⫹) 3.2 ⫾ 0.3 1.2 ⫾ 0.2 0.005
No. of acid-fast bacilli/mm2 of caseumc 1.04 ⫾ 0.40 0.43 ⫾ 0.15 0.010
a
The means and their standard errors are listed.The P values were determined by Mann-Whitney U nonparametric comparison.
NS, not significant.Adapted from reference 24.
b
The average size of the tubercles listed here is only from those in the tissue sections.This size differs from the average size in
Table 5, which was determined at necropsy from all lesions in the lungs.
c
The number of acid-fast bacilli per mm2 of caseous lung tissue was counted in 3 representative tubercles from each rabbit.

and also contained more bacilli (Table 3), but The increased number of tubercles and the
their dermal reactions to tuberculin were decreased size of the dermal tuberculin reactions
decreased (Table 3).The tubercle counts among suggest that these inbred rabbits developed less
these inbred rabbits were less variable than acquired immunity than did the outbred rab-
those found in the outbred rabbits (see the bits.The innate immunity of the inbred rabbits
standard errors in Tables 3 and 5). With the also may be reduced, because in vitro peritoneal
Erdman strain of human-type bacilli, similar macrophages from uninfected inbred rabbits pro-
differences between the inbred and outbred duced less tumor necrosis factor alpha than did
rabbits were found. However, with this more uninfected outbred rabbits when both groups
virulent strain, more rabbits would be required were stimulated by endotoxin or tubercle bacilli
to obtain tubercle counts that were statistically (24). Unfortunately, a fire destroyed all of the
significant (Table 5) (24). Thorbecke rabbits that Covance Laboratories

TABLE 5 Tuberculosis in inbred Thorbecke rabbits and outbred commercial rabbits 5 weeks after aerosol
infection with M. tuberculosis (Erdman)a
Rabbit group Inbred Outbred P values
No. of rabbits 6 6
No. of tubercles 351 ⫾ 41 305 ⫾ 107 0.170
Average diameter (mm) 3.80 ⫾ 0.30 2.13 ⫾ 0.22 0.004
No. of bacilli inhaled divided by the number of tubercles
produced (the “ratio”) 296b 684b
Dermal tuberculin reactions (mm3) 471 ⫾ 68 1,342 ⫾ 488 0.055
a
Two-day dermal reactions to Old Tuberculin were measured at 5 weeks.The means and their standard errors are listed.Adapted
from reference 24.
b
Calculated from the means.
244 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

was breeding, so they are no longer commercially 11. Dannenberg, A. M., Jr., O. T. Meyer, J. R.
available. Esterly, and T. Kambara. 1968.The local nature
of immunity in tuberculosis, illustrated histochem-
ically in dermal BCG lesions. J. Immunol. 100:931–
USE OF RABBITS FOR STUDIES 941.
OTHER THAN TUBERCULOSIS
12. Dannenberg, A. M., Jr. 1968. Cellular hyper-
Rabbits have been used for studies of arte- sensitivity and cellular immunity in the patho-
riosclerosis, various eye diseases, arthritis, micro- genesis of tuberculosis: specificity, systemic and local
bial diseases (including syphilis, tularemia, can- nature, and associated macrophage enzymes. Bacte-
didiasis, cryptococcosis, coccidioidomycosis, riol. Rev. 32:85–102.
anthrax, meningitis, and endocarditis), cardiac 13. Lurie, M. B., P. Zappasodi, E. Cardone-Lynch,
and A. M. Dannenberg, Jr. 1952.The response
allograph rejection, and cardiomyopathy. In addi- to the intracutaneous inoculation of BCG as an
tion, rabbits are often used for testing various index of native resistance to tuberculosis. J. Immunol.
vaccines, for antibody production, and for var- 68:369–387.
ious toxicity tests (including those for pyro- 14. Lurie, M. B., P. Zappasodi,A M. Dannenberg,
gens) (25). Jr., and G. H.Weiss. 1952. On the mechanism of
genetic resistance to tuberculosis and its mode of
inheritance. Am. J. Hum. Genet. 4:302–314.
REFERENCES
15. van Zutphen, L. F. M., and R. R. Fox. 1977.
1. Lurie, M. B. 1964. Resistance to Tuberculosis: Exper- Strain differences in response to dietary cholesterol
imental Studies in Native and Acquired Defensive Mech- by JAX rabbits: correlation with esterase patterns.
anisms. Harvard University Press, Cambridge, Mass. Atherosclerosis 28:435–446.
2. Lurie, M. B. 1941. Heredity, constitution and 16. Beynen, A. C., G.W. Meijer, A. G. Lemmens,
tuberculosis: an experimental study. Am. Rev.Tuberc. J. F. C. Glatz, A. Versluis, M. B. Katan, and
44(Suppl. to no. 3):1–125. L. F. M. van Zutphen. 1989. Sterol balance and
3. Lurie, M. B., P. Zappasodi, and C. Tickner. cholesterol absorption in inbred strains of rabbits
1955. On the nature of genetic resistance to tuber- hypo- or hyperresponsive to dietary cholesterol.
culosis in the light of the host-parasite relationships Atherosclerosis 77:151–157.
in natively resistant and susceptible rabbits. Am.
17. Dannenberg, A. M., Jr. 1998. Lurie’s tubercle-
Rev.Tuberc. Pulm. Dis. 72:297–329.
count method to test TB vaccine efficiency in rab-
4. Lurie, M. B., and A. M. Dannenberg, Jr. 1965.
bits. Front. Biosci. 3:c27–33. Available at
Macrophage function in infectious disease with
http://www.bioscience.org/1998/v3/c/dannenbe/
inbred rabbits. Bacteriol. Rev. 29:466–476.
list.htm.
5. American Rabbit Breeders Association, Inc.
1996. Standard Bred Rabbits and Cavies, 1996 through 18. Converse, P. J., A. M. Dannenberg, Jr., J. E.
2000.American Rabbit Breeders Association, Inc, Estep, K. Sugisaki,Y.Abe, B. H. Schofield, and
P.O. Box 426, Bloomington, Ill. 61702. M. L. M. Pitt. 1996. Cavitary tuberculosis pro-
6. Dannenberg, A. M., Jr. 1991. Delayed-type duced in rabbits by aerosolized virulent tubercle
hypersensitivity and cell-mediated immunity in the bacilli. Infect. Immun. 64:4776–4787.
pathogenesis of tuberculosis. Immunol.Today 12:228– 19. Converse, P. J., A. M. Dannenberg, Jr.,T. Shi-
233. genaga, D. N. McMurray, S. W. Phalen, J. L.
7. Dannenberg, A. M., Jr. 1993. Immunopatho- Stanford, G. A. W. Rook, T. Koru-Sengul,
genesis of pulmonary tuberculosis. Hosp. Pract. 28: H.Abbey, J. E. Estep, and M. L. .M. Pitt. 1998.
33–40 (Off. ed. 51–58). Pulmonary bovine-type tuberculosis in rabbits:
8. Dannenberg, A. M., Jr. 1994. Rabbit model of bacillary virulence, inhaled dose effects, tuberculin
tuberculosis, p. 149–156. In B. R. Bloom (ed.), sensitivity, and Mycobacterium vaccae immunotherapy.
Tuberculosis: Pathogenesis, Protection, and Control. Clin. Diagn. Lab. Immunol. 5:871–881.
ASM Press,Washington, D.C. 20. Fox, R. R. 1974.Taxonomy and genetics, p. 1–22.
9. Henderson, H. J., A. M. Dannenberg, Jr., and In S. H.Weisbroth, R. E. Flatt, and A. L. Kraus (ed.),
M. B. Lurie. 1963. Phagocytosis of tubercle bacilli The Biology of the Laboratory Rabbit.Academic Press,
by rabbit pulmonary alveolar macrophages and its Inc., New York, N.Y.
relation to native resistance to tuberculosis. 21. Fox, R. R. (ed.). 1975. Handbook on Genetically
J. Immunol. 91:553–556. Standardized JAX Rabbits.The Jackson Laboratory,
10. Lurie, M. B. 1932. The correlation between the Bar Harbor, Me.
histological changes and the fate of living tubercle 22. Altman, P. L., and D. D. Katz. 1979. Rabbit, p.
bacilli in the organs of tuberculous rabbits. J. Exp. 565–606. In Inbred and Genetically Defined Strains of
Med. 55:31–54. Laboratory Animals, vol. 3, pt. 2, Hamster, Guinea Pig,
14. CHARACTERISTICS OF RESISTANCE AND SUSCEPTIBILITY 䡵 245

Rabbit, and Chicken. Federation of American Soci- lipopolysaccharide PmKo from tubercle bacilli as an
eties for Experimental Biology, Bethesda, Md. index of resistance to tuberculosis. Bull. Johns Hop-
23. Manabe, Y. C., A. M. Dannenberg, Jr., S. K. kins Hosp. 117:174–194.
Tyagi, C. L. Hatem, M.Yoder, S. C.Woolwine, 29. Lurie, M. B., A. G. Heppleston, S. Abramson,
B. C. Zook, M. L. M. Pitt, and W. R. Bishai. and I. B. Swartz. 1950. An evaluation of the
2003. Different strains of Mycobacterium tuberculosis method of quantitative airborne infection and its use
cause various spectrums of disease in the rabbit in the study of the pathogenesis of tuberculosis.
model of tuberculosis. Infect. Immun. 71:6004–6011. Am. Rev.Tuberc. 61:765–797.
24. Dorman, S., C. L. Hatem, S. Tyagi, 30. Lurie, M. B., P. Zappasodi, R. S. Levy, and
K. Aird, J. Lopez-Molina, M. L. M. Pitt, B. C. R. G. Blaker. 1959. On the role of the thyroid
Zook,A. M. Dannenberg, Jr.,W. R. Bishai, and in native resistance to tuberculosis. I.The effect of
Y. C. Manabe. 2004. Susceptibility to tuberculo- hyperthyroidism. Am. Rev. Respir. Dis. 79:152–
sis: clues from studies with inbred and outbred New 179.
Zealand White rabbits. Infect. Immun. 72:1700–1705. 31. Dannenberg, A. M., Jr., W. R. Bishai, N. Par-
25. Manning, P. J., D. H. Ringler, and C. E. New- rish, R. Ruiz, W. Johnson, B. C. Zook, J. W.
comer (ed.). 1994. The Biology of the Laboratory Boles, and M. L. M. Pitt. 2000. Efficacies of
Rabbit, 2nd ed., p. 367. Academic Press, Inc., New BCG and vole bacillus (Mycobacterium microti) vac-
York, N.Y. cines in preventing clinically apparent pulmonary
26. Lurie, M. B., S. Abramson, and A. G. Hep- tuberculosis in rabbits: a preliminary report. Vaccine
pleston. 1952. On the response of genetically re- 19:796–800.
sistant and susceptible rabbits to the quantitative 32. Allison, M. J., P. Zappasodi, and M. B. Lurie.
inhalation of human-type tubercle bacilli and the 1962. Host-parasite relationships in natively resis-
nature of resistance to tuberculosis. J. Exp. Med. tant and susceptible rabbits on quantitative inhala-
95:119–134. tion of tubercle bacilli: their significance for the
27. Allison, M. J., P. Zappasodi, and M. B. Lurie. nature of genetic resistance. Am. Rev. Respir. Dis.
1961. Metabolic studies of mononuclear cells from 85:553–569.
rabbits of varying genetic resistance to tuberculo- 33. Lurie, M. B. 1944. Experimental epidemiology of
sis. I. Studies on cells of normal non-infected ani- tuberculosis: hereditary resistance to attack by tuber-
mals. Am. Rev. Respir. Dis. 84:364–370. culosis and to the ensuing disease and the effect of
28. Dannenberg,A. M., Jr., K. Mizunoe, M. Peace, the concentration of tubercle bacilli upon these two
and P. Zappasodi. 1965. Dermal response to the phases of resistance. J. Exp. Med. 79:573–589.
15
COMPARISONS OF TUBERCULOSIS
IN RABBITS, MICE, AND GUINEA PIGS

Overview [247]
Tuberculosis in rabbits [247]
Tuberculosis in mice [247]
Tuberculosis in guinea pigs [254]
Tuberculosis in monkeys [257]
Comparison of tuberculosis in mice with that in rabbits, humans, and guinea
pigs [259]
Virulence of inhaled tubercle bacilli in rabbits, mice, and guinea
pigs [260]
Bacillary titers in the stationary stage after aerosol infection of rabbits, mice,
and guinea pigs [261]
Fate of aerosolized BCG in rabbits, mice, and guinea pigs [262]
Tuberculin sensitivity among the three common laboratory species [262]
Organ resistance in rabbits and guinea pigs [262]
Early tuberculous lesions in humans [263]
Cavity formation in various species [263]
Role of DTH and CMI in mouse, guinea pig, and rabbit tuberculosis [263]

Abstract. In recent times, mice have been by far the most frequently used animal for the
study of tuberculosis. Guinea pigs and rabbits are used less often, and monkeys are used
only occasionally.
Rabbits are highly susceptible to bovine-type tubercle bacilli and are the only common
laboratory species in which chronic cavitary tuberculosis with bronchial spread is readily
produced. Rabbits develop grossly visible pulmonary tubercles following the inhalation of
virulent human-type tubercle bacilli, but these tubercles usually regress, as they do in most
humans. Rabbit pulmonary lesions caused by virulent human-type tubercle bacilli some-
times form nonprogressive cavities.Rabbits die after an infection with virulent bovine-type
tubercle bacilli, but eventually heal an infection with virulent human-type tubercle bacilli.
Mice develop slowly progressing pulmonary tubercles with both bovine and human
strains of tubercle bacilli, but the disease progresses more rapidly with the bovine strain.
With virulent human-type bacilli, the tubercles of mice contain a larger number of viable
bacilli than do the tubercles of rabbits and guinea pigs. Apparently, the low levels of
delayed-type hypersensitivity (DTH) in mice and the rarity of caseous necrosis allow the
bacillus to grow to higher titers in the logarithmic stage. After these titers are reached, the
good cell-mediated immunity (CMI) developed by mice reduces the intracellular multi-
plication of the majority of the bacilli to almost a dormant state. However, the low per-
centage of nondormant bacilli causes the disease to slowly progress until the animal dies.
Guinea pigs are highly susceptible to both human and bovine strains of tubercle bacilli.
They usually develop a rapidly progressing, hematogenously spread form of tuberculosis,
similar to that developed by infants and immunosuppressed individuals. Because guinea
pigs develop relatively high sensitivity to tuberculin, which causes considerable caseous
necrosis, their lesions contain a rather low number of viable bacilli. In other words, despite
the rather poor CMI developed by guinea pigs, their good DTH effectively reduces the
number of viable bacilli in their lesions. However, because of the extensive lung destruc-
tion caused by tissue-damaging DTH, guinea pigs often die in less time than do rabbits
and mice.
Rhesus monkeys are very susceptible to tuberculosis, but cynomolgus monkeys are more
resistant. Some cynomolgus monkeys can even stop the progression of the disease.

246
15. TUBERCULOSIS IN RABBITS, MICE,AND GUINEA PIGS 䡵 247

OVERVIEW duced by the virulent bovine type progresses


In all three species of common laboratory ani- until the host dies, whereas tuberculosis pro-
mals, if the tubercle bacillus is fully virulent it duced by the virulent human type uniformly
will cause progressive disease terminating in heals, although it may take over a year to do so.
death. However, after aerosol infection, despite The pathogenesis of tuberculosis in rabbits
the progression of the disease, the number of with both bovine and human bacillary types is
viable bacilli in the lungs of all three species stops described in chapter 13. In brief, 5 weeks after
increasing at 3 to 4 weeks (Fig. 1, 2, and 3). At the inhalation of either type of virulent tuber-
this time, the host has developed both delayed- cle bacillus, typical tubercles are usually present,
type hypersensitivity (DTH) to the tuberculin- often with small caseous centers.These tubercles
like products of the bacillus and cell-mediated progress or regress, depending on the amount of
immunity (CMI), which together keep the immunity (acquired resistance) developed by the
number of viable bacilli rather constant (see host and the virulence of the infecting strain. In
chapter 2). hosts that develop good acquired resistance, liq-
The number of viable bacilli present in the uefaction and cavity formation may occur (6),
lungs does not necessarily parallel increases in even with human-type bacilli (7). However, only
antigen levels. Antigen levels can increase with the bovine type produces progressive tuberculosis
no change in the number of viable bacilli if with spread through the bronchial airways.
their rate of multiplication and their rate of
death are equal. In this case, bacillary secretory TUBERCULOSIS IN MICE
products, carcasses (1, 2), and other components
Bacillary Growth Curves and Type of
(3) can increase if they are not easily destroyed.
Disease
In fact, viable bacilli persisting at constant levels often
Similar to rabbits and guinea pigs, the number
cause progressive disease (see Fig. 1, 2, and 3) (4).
of virulent bovine- or human-type bacilli in
Only after the number of viable bacilli has
the lungs of mice stops increasing at 3 to 4
been substantially reduced do the antigen con-
weeks after an aerosol infection (Fig. 2).Then,
centrations actually decrease and the lesions
the bacillary titers remain more or less constant
begin to heal. However, when tubercle bacilli are
until near death (8–11).The pulmonary gran-
fully virulent for the host, the number of viable
ulomas enlarge progressively (often with nearby
bacilli in the lungs never decreases, although it
satellite lesions [12]) until the mouse succumbs
may remain constant for months.
(4, 13, 14).The lesions show little or no necro-
In mice, which are animals with little or no
sis (Table 1) before the final stages of the disease.
tissue-damaging DTH, the majority of tubercle
bacilli persist intracellularly in macrophages
Ending the Logarithmic (Symbiotic)
without multiplying or dying (1, 2), but some
Stage of Bacillary Growth
bacilli multiply and are killed, adding to the
The logarithmic stage ends in 3 to 4 weeks
antigenic load (discussed more fully below.)
when CMI and DTH develop (Fig. 2). Mice
Table 1 presents the main characteristics of
develop only low levels of tuberculin sensitivity,
tuberculosis in humans and in laboratory ani-
which explains why there is little or no necro-
mals. Reference 5 reviews the literature on pul-
sis. They probably use apoptosis to kill bacilli-
monary tuberculous cavities in rabbits, mice,
laden macrophages and stop the logarithmic
guinea pigs, humans, and nonhuman primates.
stage of bacillary growth. Cells killed by apop-
tosis are rapidly ingested and destroyed by
TUBERCULOSIS IN RABBITS macrophages without any nearby tissue reac-
The disease in rabbits is unique, because in this tion (15–17). Apoptosis is apparently caused at
species virulent human-type tubercle bacilli and least in part by cytotoxic T lymphocytes, because
virulent bovine-type bacilli produce disease of the multiplication of Mycobacterium tuberculosis
markedly different severity. Tuberculosis pro- is uncontrolled in knockout mice (see glossary)
248 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

FIGURE 1 The number of viable human- and bovine-type tubercle bacilli in the lungs of
natively resistant and susceptible rabbits at different intervals following quantitative airborne infec-
tion (6, 7, 75).This graph shows the increase in the number of viable bacilli relative to the ini-
tial number deposited into the pulmonary alveoli (see explanation below). All human-type
tubercle bacilli are of reduced virulence for rabbits.The means are shown (see references 7 and
75 for the standard errors).
By 1 week after infection, the resistant rabbits had inhibited the growth of the human-type
bacilli in their lungs about 25 times more effectively than did the susceptible rabbits, and the re-
sistant rabbits had inhibited the growth of bovine-type bacilli about 5 times more effectively. From
then on, the bacillary growth curves in both strains of rabbit were parallel. At 4 weeks, similar
comparisons of the number of bacilli in the susceptible and resistant rabbits were about 20 times
for the human type and about 2.5 times for the bovine type.
These findings indicate that bovine-type bacilli are harder to inhibit than human-type bacilli—
both by native resistance (pulmonary alveolar macrophages) and by acquired resistance (T lym-
phocytes activating macrophages).These findings also indicate that both native and acquired re-
sistance are much more effective in the genetically resistant group. Commercially available New
Zealand White rabbits resemble Lurie’s resistant strain of rabbits (68, 69, 94–97).
The number of bacilli in the lungs of the resistant group failed to decrease during the period
illustrated, because liquefaction with extracellular multiplication of the bacillus readily occurs in
these rabbits (6). Liquefaction usually does not occur in the susceptible rabbits (6), because their
macrophages probably develop only low levels of hydrolytic enzymes. Reproduced with permission
from reference 75.
Note that this figure was derived from the results of many experiments, each using a some-
what different inhaled dosage. For this reason, the y axis represents the number of viable tuber-
cle bacilli in the entire lung divided by one-third the number of bacilli inhaled.This ratio shows
increases (or decreases) in the number of viable pulmonary bacilli at various times after infec-
tion.At 8 weeks, the lungs of the resistant and susceptible rabbits contained, respectively, an aver-
age of 2.2 ⫻ 108 and 3.4 ⫻ 108 viable bovine-type bacilli, and 2.2 ⫻ 106 and 25.5 ⫻ 106 human-
type bacilli (see references 7 and 75).
The number of bacilli was estimated by culturing lung homogenates.The number of inhaled
bacilli was calculated by sampling and culturing the aerosols with an impinger and determining
the amount of air breathed by each rabbit calculated from its weight (6, 7, 75).Two-thirds of the
small bacillary particles breathed by each rabbit impinge on the mucosa of the bronchial tree and
are eventually swallowed. Only one-third reach the alveoli where the infection begins (6). In the
graph, the number of tubercle bacilli inhaled into the alveoli is therefore one-third the number
of bacilli that the rabbit breathed.
15. TUBERCULOSIS IN RABBITS, MICE,AND GUINEA PIGS 䡵 249

FIGURE 2 The number of viable viru-


lent human-type (H37Rv) and bovine-
type (Ravenel) tubercle bacilli in the lungs
of C57BL/6 mice at different intervals fol-
lowing quantitative airborne infection (8).
Note that the in vivo bacillary growth
curves in mice resembled those found in
rabbits (see Fig. 1), but in contrast to rab-
bits, H37Rv and Ravenel tubercle bacilli
multiplied to the same levels in C57BL/6
mice (a relatively resistant strain). Repro-
duced with permission from reference 8.

that lack such lymphocytes (18–21).The killing pigs. After immunity develops in mice, many
of bacilli-laden macrophages is necessary to stop macrophages in the lesions are probably activated
the logarithmic stage of bacillary multiplication, and do not allow the ingested bacilli to multi-
because during this stage many macrophages ply appreciably.The good CMI evidently keeps
contain too many actively growing bacilli to be tubercle bacilli in a rather dormant state in
controlled by CMI alone (see chapter 2).* which their multiplication is inhibited (Fig. 4)
(1, 2). In other words, during the stationary
Maintaining the Stationary Stage of stage relatively few tubercle bacilli multiply
Bacillary Growth within the macrophages, so the number of dead
In maintaining the stationary stage, mice appar- bacilli does not increase. Dead bacilli are only
ently differ from humans, rabbits, and guinea slowly destroyed over many weeks (Fig. 4).
Nevertheless, a small amount of bacillary
replication probably occurs in mouse macro-
*The logarithmic stage of bacillary growth apparently phages during the stationary stage, followed by
ends sooner after an intravenous infection with tuber-
cle bacilli than after an aerosol infection (10) for the fol-
apoptosis of these macrophages (1, 2).This small
lowing reasons. (i) A major portion of the intravenous amount of bacillary multiplication causes the
dose directly enters the spleen, which is a lymphoid disease to progress slowly until the mouse suc-
organ that can rapidly produce acquired immunity (10). cumbs.
(ii) The dose of bacilli given intravenously is usually In brief, the CMI evidently activates sufficient
higher than that given by aerosol, thereby providing a
greater immunological stimulus. And (iii) the dose of
macrophages in the tuberculous lesions of mice
bacilli given by aerosol is lowered even further, because to keep most intracellular tubercle bacilli from
the pulmonary alveolar macrophage population is highly multiplying appreciably. Unfortunately, this inhi-
activated and immediately inhibits or slows the multi- bition of intracellular bacillary growth is incom-
plication of many inhaled bacilli. Bacilli entering the plete and is insufficient to stop the progression
lungs following an intravenous injection bypass the
alveolar macrophage population and are ingested by
of the disease, which eventually kills the host.
nonactivated macrophages. Following an aerosol infec-
tion, some dissemination of bacilli from the lungs to the Tissue Necrosis in Mice
lymph nodes, liver, and spleen occurs in mice, but the Tissue necrosis will occur in mice when large
disease in other organs is more readily produced by an numbers of bacilli grow within macrophages
intravenous infection. Since the inhalation of tubercle (22). The exact cause of such necrosis has not
bacilli is the most common way that humans contract
tuberculosis, we have limited our comparisons of tuber- been identified. Some of the necrosis in mice
culosis in rabbits, mice, and guinea pigs to this method may be due to their low level of DTH that
of infection. kills macrophages when the tuberculin-like
250 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

FIGURE 3 The number of viable virulent human-type tubercle bacilli (H37Rv) in the lungs
of BCG-vaccinated and control guinea pigs at different intervals following quantitative airborne
infection (40). Note that the in vivo bacillary growth curves in guinea pigs resemble those found
in rabbits (Fig. 1) and in mice (Fig. 2). Other experiments showed that the stationary phase in
guinea pigs continues for at least 18 weeks (41). Note also that in the BCG-vaccinated guinea
pigs, the logarithmic growth stage ends soon and that the number of viable tubercle bacilli in the
stationary stage is markedly reduced (see text).The guinea pigs were immunized intradermally
with live BCG 6 weeks before the aerosol challenge. Reproduced with permission from refer-
ence 40.
The number of tubercle bacilli in the stationary stage in guinea pigs is lower than the num-
ber found in mice (compare Fig. 2 and 3), because guinea pigs probably end the logarithmic stage
sooner (although insufficient time points were plotted in these two figures to show this differ-
ence).Therefore, the good DTH of guinea pigs is probably more effective than the good CMI
of mice in ending the logarithmic stage, which supports the principle that tissue-damaging DTH
is an important host defense mechanism in controlling intracellular bacillary multiplication.

products of the bacilli reach a high local con- The intragranulomatous necrosis found in
centration. Some of the necrosis may be merely the more chronic stages of pulmonary tuber-
degeneration of macrophages when they con- culosis in mice is somewhat different from the
tain numerous bacilli (11, 18, 22–26). Some of typical caseous necrosis found in rabbits, guinea
the necrosis may be a local Shwartzman phe- pigs, and humans. Mice show little damage to
nomenon (see glossary) (27, 28) (characterized the lung tissue surrounding the granulomas,
by intravascular fibrin deposits that impair the and they show little vascular thrombosis, which
local blood supply [29]) and/or an antigen- is a major factor in producing caseous necro-
antibody reaction causing an Arthus reaction sis in the other three species (see below).
(see glossary). In mice, such large numbers of Mouse intragranulomatous necrosis (22, 27)
tubercle bacilli often occur in various types of does not seem as “mature” as the typical
knockout mice (22, 27, 30, 31) and may occur caseous necrosis found in other species. Mature
terminally in wild-type mice (22, 27). caseum is a homogeneous necrosis with the
nuclear debris of the dead cells finely dispersed
15. TUBERCULOSIS IN RABBITS, MICE,AND GUINEA PIGS 䡵 251

TABLE 1 Characteristics of tuberculosis in humans and in laboratory animalsa,b

Species Tuberculin-type Caseous necrosis Cell-mediated Cavity


allergy (DTH) (due to DTH)c immunity (CMI) formation
Isolated human populationsd ⫹⫹⫹⫹⫹ ⫹⫹⫹⫹⫹ ⫹⫹ ⫹⫹⫹
Rhesus monkeyse ⫹⫹ ⫹⫹⫹⫹⫹ ⫹⫹ ⫹⫹⫹
Guinea pigs ⫹⫹⫹ ⫹⫹⫹⫹⫹ ⫹⫹ ⫹
Modern humans
Immunocompetent ⫹⫹⫹⫹⫹ ⫹⫹⫹⫹⫹ ⫹⫹⫹⫹ ⫹⫹⫹⫹⫹
Immunosuppressed ⫹ ⫹⫹⫹⫹⫹ ⫹ ⫹
Rabbits
Resistant ⫹⫹ ⫹⫹⫹ ⫹⫹⫹⫹ ⫹⫹⫹⫹
Susceptible ⫹⫹ ⫹⫹⫹⫹⫹ ⫹⫹ 0
Mice ⫹ ⫹ ⫹⫹⫹ 0
a
Reproduced with permission from reference 4 and adapted from a table by Francis (43). References 60 and 93 reproduce
the entire Francis table, which contains many animal species, including elephants.
b
The assignment of ⫹ to ⫹⫹⫹⫹⫹ is a generalization of a given species as a whole, because much variation exists within a
species. Good CMI and DTH are both needed to arrest the disease. Guinea pigs have good DTH but relatively poor CMI, and
mice have good CMI but poor DTH. Both species show progressive disease leading to their death. Most humans and rabbits (infected
with virulent human-type tubercle bacilli) have adequate amounts of DTH and CMI to arrest the disease.
c
Tissue-damaging DTH is the cause of caseous necrosis.Tubercle bacilli are dormant in solid caseum, and many do not sur-
vive there. In arrested tuberculous lesions, solid caseum is often encapsulated by fibrous tissue.
d
Isolated human populations are those that have never before been exposed to tuberculosis, such as the Senegalese troops brought
from Africa to Europe during World War I (65).
e
Cynomolgus monkeys are more resistant to tuberculosis than rhesus monkeys and may even arrest the disease (see text).

or absent (probably due to proteases and nucle- amounts of various antigens (or more reactive
ases) (6, 7). antigens) than do live virulent human-type
bacilli. Bovine-type bacilli may also stimulate a
Tuberculosis Produced in Mice by greater chemokine response in the host. An
Virulent Bovine-Type and Virulent increase in chemokines would cause a more
Human-Type Tubercle Bacilli rapid growth of the pulmonary lesions and
In some mouse strains, the virulent bovine-type therefore cause earlier death.
and the virulent human-type bacilli multiplied In other mouse strains, virulent bovine-type
in the lungs to the same titers (Fig. 2) (8). How- bacilli multiply in the lung to higher titers dur-
ever, the bovine type (Ravenel) killed mice ing the logarithmic stage than do virulent
faster than did the human type (H37Rv): by 160 human-type bacilli (33; K. Bosio, personal com-
days, all the mice inhaling Ravenel were dead, munication). Similarly, the virulent H37Rv
whereas all the mice inhaling H37Rv were alive strain multiplies in mouse lungs to higher titers
(8). By 200 days, only 20% of the mice inhaling than does the relatively avirulent R1Rv strain
H37Rv had died (8). In this experiment, since (33, 34).
the number of live pulmonary bacilli of each
strain was the same, components of the dead Genetic Differences in Resistance to
bovine-type bacilli evidently persisted longer Tuberculosis among Mouse Strains
in the lungs than components of the dead As in rabbits, genetic differences among various
human type, causing the granulomas to increase inbred mouse strains influence the course of
in size more rapidly (8, 26, 32) and kill the host the disease. During the stationary phase after an
sooner (see references 8, 26, and 33). Perhaps, live intravenous injection, the number of viable
bovine-type bacilli produce and/or secrete larger H37Rv bacilli in the lungs of BALB/c mice was
252 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

FIGURE 4 Viable counts and total counts of virulent tubercle bacilli (H37Rv)
in the lungs of mice from 9 weeks to 25 weeks after an intravenous infection.
After the lungs were homogenized, the viable counts were calculated from the
CFU developing on plates containing solid culture medium.The total counts
were calculated from the bacilli observed microscopically after acid-fast stain-
ing of spread smears made from the homogenates. During this time period, one
of the four groups of mice received isoniazid-pyrazinamide (PZA/INH) daily
to kill the bacilli.
Note that for untreated mice the average total counts were 0.3 to 0.4 logs
higher (2.0 to 2.5 times) than the average viable counts, and that PZA/INH treat-
ment markedly reduced the viable counts but had relatively little effect on the
total counts.These findings indicate that (i) most of the dead tubercle bacilli per-
sisted in mouse lungs for many weeks, (ii) most of the live bacilli in the untreated
mice were in a dormant state, because if they had been multiplying and then
had been killed, the total counts (including the dead bacilli) would have
increased, and (iii) the good CMI developed by mice activated macrophages suf-
ficiently to prevent the intracellular multiplication of most of the tubercle
bacilli.
Note also that the 2.0- to 2.5-fold mean difference in total and viable counts
suggests that at least some bacillary growth and destruction were occurring. In
other words, at least some of the bacilli were not inhibited by the good CMI,
which is probably the reason for the progression of the disease in mice until they
succumb.
These results were confirmed in reference 2 using quantitative real-time PCR.
Reproduced with permission from reference 1.The same figure appears as
Fig. 1 in chapter 6 of this volume.
15. TUBERCULOSIS IN RABBITS, MICE,AND GUINEA PIGS 䡵 253

FIGURE 5 A primary pulmonary tuberculous lesion in a C57BL/6


mouse 8 weeks after an aerosol inhalation of virulent human-type tubercle
bacilli. On the right is the lesion’s center containing large viable epithelioid
macrophages, some of which probably inhibited (or destroyed) the tubercle
bacilli that they had ingested during the earlier stages of the infection.A few
polymorphonuclear leukocytes have accumulated where some of these
macrophages have disintegrated (far right). In the middle of the photograph
is part of the mantle of compact lymphocytes, which surrounds the center
of epithelioid macrophages. On the far left are a few foamy macrophages
within the surrounding alveolar spaces. Many of these foamy macrophages
are pulmonary alveolar macrophages that tend to accumulate around pul-
monary tuberculous lesions (see Fig. 5 of chapter 9). Pulmonary alveolar
macrophages often ingest surfactant, which gives them the foamy appearance.
Stained with hematoxylin and eosin. Magnification, ⫻275. Photograph pro-
vided by P.-J. Cardona.

about two logs lower than the number in the reference 25). SCID mice are so susceptible that
lungs of DBA/2 mice (35).Also, BALB/c mice virtually no stationary phase occurs before the
and C57BL/6 mice lived about twice as long as animal dies (36).
the DBA/2 and C3H/HeJ mice: their mean
survival times were about 8 and 4 months, Histopathology of Mouse Pulmonary
respectively (14). Tuberculous Lesions
Among the mouse strains tested, aerosol Lesion progression in mice occurs locally (12, 37).
infection produced results similar to the intra- Macrophages in which tubercle bacilli are grow-
venous infection: C57BL/6 and BALB/c mice ing evidently migrate from the primary lesions
are more resistant than DBA/2, 129/SvJ, CBA, into neighboring alveoli and cause secondary
C3H/HeN, and C3H/HeJ mice (12, 14, 27). lesions (Fig.5 and 6).Therefore,the secondary and
Also, knockout mice (with one or more com- tertiary lesions are often nearby, and confluence
ponents of innate or acquired immune response occurs in the terminal stages of the disease.
removed from their genome by molecular biol- Bacilli-laden foamy macrophages are present
ogy techniques) show higher bacillary titers in bronchoalveolar lavages (12, 37).The vacuoles
during the stationary stage (22, 24, 31; see also causing their foamy appearance are probably
254 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

FIGURE 6 The outer layer of the 8-week lesion shown in Fig. 5. Note
how these foamy cells fill the alveolar spaces.Tubercle bacilli (not shown) are
occasionally present in some of these foamy macrophages.The bacilli were
probably carried to the lesion’s periphery within macrophages that had
migrated at an earlier time from the center. Such bacilli sometimes cause sec-
ondary lesions in nearby alveoli. Stained with hematoxylin and eosin. Mag-
nification, ⫻275. Photograph provided by P.-J. Cardona.
In mice, few, if any, tubercle bacilli remain free in viable tissues or even
in lymphatics, because bacilli released from disintegrating macrophages are
soon ingested by other (still viable) macrophages. In animal species where
cavities form, many free tubercle bacilli enter the airspaces, and some of these
bacilli may be ingested by the alveolar macrophages. Mouse lesions do not
liquefy and form cavities, so extracellular tubercle bacilli are rarely present.

filled with the lipids from both dead tubercle caseous necrosis (6, 38; see also reference 39).
bacilli and dead apoptotic host cells, as well as Such thrombosis does not usually occur in the
the surfactants that these cells ingest. tubercles of mice, and functional capillaries and
It is not known how the bacilli get from the venules, as well as viable lymphocytes, dendritic
necrotic center to the periphery. They could cells, and macrophages, are usually present
travel in lymphatics in a free state or within throughout the tubercles.
macrophages. In mouse pulmonary granulomas, See references 12, 22, 27, and 35 for other
lymphatics may or may not develop along with photomicrographs, and see references 23, 30, and
the vasculature. (The walls of the alveoli have no 31 for reviews of factors regulating tubercu-
lymphatics. Lymphatics first appear in the lungs lous granulomas in mice, including CD4 and
at the level of the respiratory bronchioles.) CD8 lymphocytes and various cytokines.

Microvascular Patency in Mouse


Pulmonary Lesions
TUBERCULOSIS IN GUINEA PIGS
The absence of caseous necrosis in the pul-
monary tubercles of mice is partly due to intact Bacillary Growth Curves
microvasculature (Fig. 7). In the tubercles of In guinea pigs, as in rabbits and mice, inhaled
other laboratory animal species, microvascular virulent tubercle bacilli show a logarithmic
thrombosis is a major factor contributing to the growth phase followed by a stationary phase in
15. TUBERCULOSIS IN RABBITS, MICE,AND GUINEA PIGS 䡵 255

FIGURE 7 A tuberculous lesion in a mouse similar to those shown in Fig. 5 and


6. Little or no necrosis occurs in mouse lesions, mainly because there is no throm-
bosis of the microvasculature. This photograph shows patent microvessels (white
arrows) within the dense lymphocyte layer that surrounds the center, containing
numerous viable epithelioid macrophages.The microvessels are easily identified by
the intact erythrocytes that they contain. Stained with hematoxylin and eosin. Mag-
nification, ⫻295. Photograph provided by P.-J. Cardona.

which multiplication and killing of bacilli appar- sue-damaging DTH, but in each case a
ently occur at equal rates (Fig. 3) (40–42). proportion of this inhibition (or killing) is
due to activation of macrophages by CMI.
4. The considerable caseous necrosis (43) is
Gross Pathology
apparently due to thrombosed microvas-
Tuberculosis in guinea pigs, produced by both
culature (see reference 44)—in contrast to
virulent human-type bacilli and virulent bovine-
mouse tuberculous lesions, in which such
type bacilli, has many of the characteristics of
thrombosis is usually absent.
tuberculosis caused by virulent bovine-type
5. Acquired (adaptive) immunity, i.e., DTH
bacilli in Lurie’s susceptible strains of rabbits
and CMI, slows the progression of the
(Table 1) (also see chapters 2, 13, and 14).
disease, but not enough to arrest it.
1. One grossly visible primary pulmonary 6. Fatal progressive destruction of the lungs
tubercle is usually produced by every and hematogenous spread of this disease
inhaled virulent tubercle bacillary unit occur, mainly due to the tissue-damaging
that reaches the alveolar spaces. DTH. In guinea pigs, the hilar nodes and
2. The “childhood type” of tuberculosis with spleen are major sites of the metastatic dis-
large caseous hilar lymph nodes usually ease. (The lymphogenous and hematoge-
develops. nous spread of tubercle bacilli from the ini-
3. Both Lurie’s susceptible rabbits and guinea tial site of infection occurs throughout the
pigs develop relatively weak CMI.Tuber- course of the disease. Initially, the bacilli are
cle bacilli multiply within the poorly acti- probably in macrophages that transport
vated macrophages that surround the them to the hilar nodes and then [via the
caseous center. Killing or inhibition of efferent lymph and great veins] back to the
these bacilli seems to be largely due to tis- lungs. Later in the disease, the caseous
256 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

necrotic process destroys the adjacent Unfortunately, this acquired immunity is rarely
microvasculature, and free bacilli in the strong enough to stop the eventually fatal
caseum can directly enter the bloodstream progress of the disease.
[see reference 6]).
Cavity Formation
Two factors may change the typical suscep- When guinea pigs inhale such low numbers of
tible type of tuberculosis just described for bacilli that only 1 to 3 primary pulmonary
guinea pigs into a more resistant type (see ref- lesions are produced, cavities may form after
erence 45). The first is the virulence of the several months (53–55). However, as stated
infecting strain: Isoniazid-resistant catalase- above, tuberculosis in guinea pigs is primarily a
negative tubercle bacilli are often, but not always, hematogenously spread disease, similar to that
of reduced virulence for guinea pigs (46–48). In described in Lurie’s susceptible strains of rabbits.
fact, guinea pigs infected with such resistant
strains may even arrest the disease, depending on Inbred Guinea Pigs
the degree of attenuation (46, 48).The second Two inbred guinea pig strains exist: strain 2 and
is the number of tubercle bacilli inhaled: a very strain 13 (56).These strains did not significantly
low inhaled dose sometimes converted the differ in their response to aerosol challenge with
tuberculin skin test with no gross evidence of virulent human-type tubercle bacilli (H37Rv)
disease at necropsy.This sometimes occurred in (57).When a protein deficit existed in their diet,
Riley’s experiments in which guinea pigs BCG immunization no longer reduced the
inhaled an occasional bacillus over periods of growth of inhaled H37Rv bacilli in the lungs of
many weeks (see chapter 12). strain 13, and reduced the growth of inhaled
Note also that fully virulent tubercle bacilli H37Rv bacilli in strain 2 only slightly (57).
may be present in the sputum of isoniazid-
resistant patients, because not all bacilli in spu- Fibrin Meshworks
tum are identical (46–48). After DTH develops, fibrin readily forms in
guinea pig tuberculous lesions (6, 44).The asso-
Histopathology ciated thrombosis not only increases the amount
Figures 8 and 9 show typical guinea pig lesions of necrosis, but also partly obstructs the lym-
4 weeks after an aerosol infection with virulent phatics draining the lesions (6, 44). Therefore,
human-type tubercle bacilli. Each has a caseous tuberculin positivity in guinea pigs reduces the
necrotic center surrounded by tuberculous gran- spread of tubercle bacilli from tuberculous
ulation tissue. lesions to the draining lymph nodes, whereas
Reference 49 presents a detailed histological tuberculin positivity in rabbits enhances the
description of guinea pig tuberculous granulo- spread of bacilli from lesions to the draining
mas during various stages of their development, nodes. Rabbits produce relatively little fibrin, and
including the identification of CD4 and CD8 the inflammatory processes increase the flow
cells and the cells undergoing apoptosis. Refer- of lymph (6, 44).
ence 50 presents the effects of BCG on these After an aerosol infection, the number of
granulomas. tubercle bacilli reaching the hilar lymph nodes
has relatively little effect on the amount of
Immunization tuberculosis produced there, which depends on
In effectively immunized guinea pigs (Fig. 3), the whether the host inhibits the growth of the
growth of tubercle bacilli in both primary and bacilli in these nodes (6). Guinea pigs (and
metastatic lesions is decreased, and fewer bacilli Lurie’s susceptible rabbits) usually inhibit bacil-
spread to other organs (40, 44, 51, 52). In other lary growth in the nodes poorly, whereas
words, guinea pigs are able to develop some Lurie’s resistant rabbits (and commercial rab-
effective CMI, i.e., activated macrophages that bits) usually inhibit such bacillary growth
prevent the growth of ingested tubercle bacilli. effectively.
15. TUBERCULOSIS IN RABBITS, MICE,AND GUINEA PIGS 䡵 257

FIGURE 8 A guinea pig tuberculous lesion 4 weeks after an aerosol infection with virulent
human-type tubercle bacilli. Note (from left to right) the pronounced caseous center, the typi-
cal tuberculous granulation tissue with many lymphocytes, and the nearby alveoli with slightly
thickened walls. Stained with hematoxylin and eosin. Magnification, ⫻80. Photograph provided
by P.-J. Cardona.

Koch Phenomenon in rabbits (44) (and possibly in humans) may not


The Koch phenomenon is classically defined as be as effective as those in guinea pigs in reduc-
the local necrotic reaction caused by an intra- ing the spread of bacilli to the nodes. Details and
dermal injection of tubercle bacilli (or their possible mediators of the Koch phenomenon are
culture filtrate) into a tuberculous guinea pig. presented in reference 58.
Because such guinea pigs have strong tuber-
culin sensitivity, the developing dermal lesion TUBERCULOSIS IN MONKEYS
soon caseates, ulcerates, and discharges much Rhesus monkeys (Macaca mulatta) are highly
of its necrotic contents.An intradermal challenge susceptible to virulent M. tuberculosis and viru-
may also increase the amount of necrosis in the lent Mycobacterium bovis (43, 59–64).They resem-
existing (pulmonary) lesions. ble isolated human populations that had never
The Koch phenomenon is thought to be been previously exposed to the tubercle bacil-
protective, because many bacilli are discharged lus (Table 1) (6), such as the Senegalese troops
from the ulcer, and, in guinea pigs, few bacilli taken from Africa to Europe to fight in World
reach the draining lymph nodes due to the fib- War I (65).
rin meshworks in the draining lymphatics. How- Tuberculosis in rhesus monkeys is usually a
ever, as mentioned above, the fibrin meshworks rapidly progressive pulmonary disease, frequently
258 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

FIGURE 9 Another guinea pig tuberculous lesion 4 weeks after an aerosol infection with vir-
ulent human-type tubercle bacilli. Note (from left to right) the pronounced caseous center, the
typical tuberculous granulation tissue, the surrounding fibroblasts, and the intact pleura. Stained
with hematoxylin and eosin. Magnification, ⫻80. Photograph provided by P.-J. Cardona.

with both bronchial and hematogenous spread virulent tubercle bacilli, it usually dies within 6
of the bacilli. Extensive areas of caseous necro- months (67). Because of such a rapid course,
sis occur, as well as liquefaction of the caseum tuberculosis in rhesus monkeys is usually a poor
and cavities (43, 59, 61, 63, 64, 66).The cavities model for the slowly progressive fibrotic cavi-
often have ragged linings harboring numerous tary disease found in modern adult humans
tubercle bacilli (63) that spread through the air- and in commercially available rabbits infected
ways.Areas of tuberculous bronchopneumonia with virulent bovine-type bacilli (68, 69).The
are common (43, 61, 63, 66), as are enlarged disease in cynomolgus monkeys is a better
caseous hilar nodes (61, 63, 66). model.
BCG immunization of rhesus monkeys Cynomolgus monkeys (Macaca fascicularis)
reduces the severity of the disease (59, 61, 66) appear to be more resistant than rhesus monkeys
and can prevent grossly visible pulmonary lesions to infection with virulent human-type tubercle
in some animals if only a few virulent M. tuber- bacilli (70, 71). When infected intratracheally
culosis cells are initially inhaled (66). with 10 to 100 bacilli (Erdman strain), cynomol-
Since monkeys cough, they can readily infect gus monkeys often develop a chronic, slowly
handlers and other monkeys, if appropriate pre- progressive granulomatous pulmonary disease
cautions are not taken. Once a rhesus monkey with hilar lymph node involvement and variable
becomes tuberculin positive from infection with degrees of tuberculin skin test positivity (70). In
15. TUBERCULOSIS IN RABBITS, MICE,AND GUINEA PIGS 䡵 259

addition, a significant proportion of cynomol- tible rabbits benefited little from BCG vaccina-
gus monkeys contain their infection in a non- tion (73) (see chapter 23).
progressive subclinical state (70). See references 71, 72, and 74 for reviews of the
When 17 cynomolgus monkeys were infected tuberculosis literature in nonhuman primates.
bronchoscopically with about 25 CFU of viable
tubercle bacilli (Erdman strain) (71), no clinically COMPARISON OF TUBERCULOSIS IN
apparent disease was evident in about 40% of the MICE WITH THAT IN RABBITS,
animals, even though they were radiographed HUMANS, AND GUINEA PIGS
periodically for 15 to 20 months. They were, Mouse tuberculosis is quite different from that
however, clearly infected, as most were tuber- found in rabbits, guinea pigs, and humans.Table
culin positive. One of these animals with arrested 2 lists the various characteristics of pulmonary
tuberculosis was necropsied at 17 weeks. It con- tuberculous lesions in the three laboratory ani-
tained a few 1-mm fibrotic granulomas with mal species. Not all of these characteristics have
caseous centers and also a hilar caseous lymph been directly observed, but they are listed in
node with calcification and peripheral fibrosis. Table 2 because they seem to be consistent with
Among the remaining 60% of the cynomol- the histopathology.
gus monkeys with active tuberculosis, one had Mice have poor DTH, good CMI, and no
a rapidly progressive disease lasting about 2.5 true caseous necrosis.
months, and one had reactivated tuberculosis Mouse lesions contain a greater number of
with cavity formation and bronchial spread (71). viable virulent human-type tubercle bacilli than
The majority of these monkeys with active dis- do noncavitary lesions of the other species.
ease, however, showed slowly progressive chronic In mouse lesions, cycles of bacillary growth
tuberculosis, characterized by granulomas with and destruction (bacillary turnover) seem to be
caseous centers and outer fibrosis, accompanied reduced, because (i) most of these bacilli are in
by caseous hilar lymph nodes. a semidormant state within (probably activated)
In brief, cynomolgus monkeys varied con- macrophages (Fig. 4) (1, 2), and (ii) macrophage
siderably in the type of tuberculosis produced by turnover is probably reduced; i.e., fewer new
infection with virulent human-type bacilli. A macrophages apparently enter from the blood-
substantial number (40%) of the infected animals stream and fewer are killed in the lesions (see
developed no clinically apparent disease, and a chapter 10). New macrophages are nonacti-
substantial number (60%) developed a chronic vated and provide an opportunity for renewed
fibrotic granulomatous type similar to that found intracellular bacillary multiplication. DTH
in human populations. Cavities occurred, but increases macrophage turnover, but mice have
apparently not as frequently as cavities occur in only low levels of DTH.
humans. Mouse lesions show little or no thrombosis of
When both cynomolgus and rhesus mon- the microvasculature, whereas in lesions of the
keys were infected intratracheally with 3,000 other species, microvascular thrombosis proba-
virulent human-type bacilli (Erdman strain) bly causes most of the caseous necrosis.
(72), the disease was less severe in the cynomol- Mouse lesions never undergo liquefaction
gus group.With this large infecting dose, prior and cavity formation, whereas the tuberculous
BCG vaccination substantially reduced the lesions of rabbits and humans frequently do so.
amount of disease in the cynomolgus group, but Extracellular multiplication of the bacillus often
had little effect in the rhesus group (72).There- occurs in liquefied caseum and cavities.
fore, cynomolgus monkeys, rather than rhesus Despite having a much higher number of
monkeys, would be the preferred nonhuman viable tubercle bacilli in their lesions (see Table
primate for the evaluation of newly developed 3), mice are actually more resistant than guinea
tuberculosis vaccines.These findings were sim- pigs. The progression of the disease is deter-
ilar to those found with Lurie’s susceptible and mined by the total number of live-plus-dead
resistant inbred rabbits, namely, that the suscep- bacilli, not just the number of live bacilli (see
260 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

TABLE 2 Characteristics of pulmonary tuberculous lesions in rabbits, mice, and guinea pigs
Lurie’s
resistant rabbits Lurie’s Mice Guinea
Characteristica (including susceptible (resistant pigs
commercial rabbits strains)
rabbits)
Delayed-type hypersensitivity ⫹⫹ ⫹⫹ ⫹ ⫹⫹⫹
(DTH)
Cell-mediated immunity (CMI) ⫹⫹⫹⫹ ⫹⫹ ⫹⫹⫹⫹ ⫹⫹
Caseous necrosis (solid, not liquefied)* ⫹⫹ ⫹⫹⫹ ± ⫹⫹⫹
Number of tubercle bacilli in noncavitary ⫹ ⫹⫹ ⫹⫹⫹ ⫹
lesions*
Turnover (growth and destruction) of ⫹ ⫹⫹⫹⫹ ⫹ ⫹⫹⫹⫹
tubercle bacilli in lesions*
Dormancy of tubercle bacilli in ⫹⫹⫹ ⫹ ⫹⫹⫹ ⫹
macrophages of lesions
Dormancy of tubercle bacilli in solid ⫹⫹⫹ ⫹⫹⫹ ⫹⫹⫹
caseum of lesions
Turnover of macrophages in lesions ⫹⫹ ⫹⫹⫹⫹ ⫹ ⫹⫹⫹⫹
Thrombosis of the microvasculature ⫹⫹⫹ ⫹⫹⫹ 0 ⫹⫹⫹
causing caseous necrosis*
Cavity formation and extracellular growth ⫹⫹ 0 0 ⫾
of tubercle bacilli*
a
This table is derived from our knowledge of tuberculous lesions produced by virulent human tubercle bacilli. Bovine-type
tubercle bacilli are more virulent for rabbits than the human type.The categories marked with an asterisk (*) would be increased
in rabbit lesions caused by the bovine type, especially the amount of caseation and number of cavities.

reference 1). Dead tubercle bacilli and live tuber- readily form around arrested tuberculous lesions
cle bacilli produce almost identical tissue reac- of rabbits and humans.
tions (6). Mouse lesions probably contain a
lower number of live-plus-dead tubercle bacilli VIRULENCE OF INHALED TUBERCLE
than do guinea pig lesions.Therefore, the disease BACILLI IN RABBITS, MICE, AND
progresses more slowly in mice than in guinea GUINEA PIGS
pigs. In immunocompetent rabbits, fully virulent
Mice never arrest pulmonary lesions caused Ravenel strains of M. bovis produce one grossly
by virulent human-type tubercle bacilli, but visible tubercle for each 3 units (of 1 to 3 bacilli)
rabbits and humans readily do so. The CMI inhaled (6), whereas fully virulent H37Rv strains
and apoptotic mechanisms (used to kill mac- of M. tuberculosis produce one such tubercle for
rophages overloaded with bacilli) found in mice each 300 to 3,000 bacillary units inhaled (Table
are probably not as effective in stopping the 3) (6,7,75).The Ravenel strain eventually kills the
progression of the disease as the CMI and rabbit,but the H37Rv strain rarely does so (6,75).
tissue-damaging DTH found in rabbits and In both mice and guinea pigs, 3 to 6 inhaled
humans. bacillary units of either fully virulent human- or
Mouse lesions are never encapsulated, i.e., bovine-type tubercle bacilli apparently produces
completely surrounded by a fibrous tissue. In one grossly visible tubercle (Table 3) (see refer-
fact, in mouse lesions, collagen fibers are actu- ences 32, 76, and 77). In these animals, the
ally scattered centrally among bacilli-laden bovine type produces a more rapidly progressive
macrophages (22, 27, 28). In contrast, capsules disease than the human type (8, 26, 32). Rabbits
15. TUBERCULOSIS IN RABBITS, MICE,AND GUINEA PIGS 䡵 261

are unique in that they uniformly survive infec- than do human-type bacilli (Fig. 1). The loga-
tion with the virulent human type (6, 78). rithmic stage ends when the host kills
Unless the infecting bacillary strain is macrophages that harbor numerous intracellular
markedly attenuated, the doubling times of the bacilli (see chapters 2 and 5).Why the killing of
infecting bacilli should not vary greatly during virulent bovine-type bacilli is delayed in rabbits
their logarithmic stage of growth, whether this is not known.The secreted and constitutive anti-
growth directly follows an intravenous infection gens of the bovine type may be different from
(79) or whether it follows an aerosol infection those of the human type, and/or they may be
after the blood-borne nonactivated macrophages released more slowly.When discovered, the prop-
prevail in the lesion. However, markedly atten- erty of bovine-type bacilli that causes the delay
uated bacilli do not multiply very well, even in in host acquired immunity will add yet another
nonactivated macrophages (79). virulence characteristic to those already known.
After the inhalation of attenuated tubercle
bacilli, the bacillary titers would be lower in the
stationary stage, because many of these bacilli
BACILLARY TITERS IN THE
would be killed by the alveolar macrophages, and STATIONARY STAGE AFTER
DTH and CMI would develop sooner. Molec- AEROSOL INFECTION OF RABBITS,
ular biologists are producing genetically altered MICE, AND GUINEA PIGS
tubercle bacilli of reduced virulence (see refer- As stated above, after inhalation, the number of
ences 80 and 81). Such bacilli should multiply to viable virulent tubercle bacilli increases loga-
substantially lower levels in vivo, similar to the rithmically in the lungs of these three laboratory
semivirulent M. tuberculosis R1Rv (33, 34). animal species for about 3 weeks and then fails
In rabbits, virulent bovine-type bacilli reach to increase further until the host approaches
higher titers in the logarithmic stage of growth the terminal stages of the disease.The bacillary

TABLE 3 Number of inhaled tubercle bacilli required to produce one primary pulmonary tubercle and the
amount of multiplication during the logarithmic growth phase in rabbits, mice, and guinea pigsa
Lurie’s
resistant rabbitsb
(including Mice Guinea pigs
commercial (C57BL/6)c
rabbits)
Average no. of inhaled virulent bovine-type 3 3–6 3–6
bacilli (Ravenel) required to produce one
pulmonary tubercle
Average no. of inhaled virulent human-type 300–3,000 3–6 3–6
bacilli (H37Rv) required to produce one
pulmonary tubercle
Increase in the number of virulent bovine-type 1,000,000 ⫻ 10,000 ⫻ No data
bacilli (Ravenel) before the stationary phase (from Fig. 1) (from Fig. 2)
Increase in the number of virulent human-type 1,000 ⫻ 10,000 ⫻ 1,000 ⫻
bacilli (H37Rv) before the stationary phase (from Fig. 1) (from Fig. 2) (from Fig. 3)
a
Adapted from reference 4. Note that, although many factors determine the amount of bacillary multiplication in the lung (see
text), species differences and genetic differences within species play important roles. This table clearly shows the importance of
tissue-damaging DTH in limiting bacillary titers: guinea pigs (with strong DTH) have lower titers than mice (with weak DTH).
Yet, despite having fewer viable bacilli, guinea pigs seem to have a more rapid progression of the disease.
b
Lurie’s resistant rabbits (e.g., strain III) (6, 7, 75). Commercially available New Zealand White rabbits seem to be similar (68, 69).
c
Data from reference 8. Bacillary multiplication in other mouse strains may be different from those listed here for C57BL/6
(see references 14 and 35). C57BL/6 is a relatively resistant strain of mice.
262 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

titer during the stationary stage is determined by (1 mm or less) was visible in their lungs 5 weeks
several factors: (i) the species of laboratory ani- later (S. Abramson, unpublished data, 1964 [as
mal that inhaled the bacilli, (ii) the native cited in reference 6]). Evidently, the alveolar
(genetic) resistance of the host (6, 35), (iii) the macrophage population of rabbits and their
virulence of the infecting tubercle bacilli, (iv) the acquired immunity readily destroy this attenu-
degree of immunization provided by a vaccine, ated bacillus before it multiplies extensively.
and (v) perhaps the number of bacilli initially In contrast, BCG inhaled by mice multiplies
inhaled (69) (see chapter 11). readily (82) and produces many pulmonary
Bacillary titers in the stationary stage often tubercles that heal only after a period of many
differ among rabbits, mice, and guinea pigs weeks (83).Therefore, aerosol immunization of
(Table 3 and Fig. 1, 2, and 3) (4). In rabbits, mice should be quite effective, whereas aerosol
bovine-type tubercle bacilli (Ravenel) multi- immunization of rabbits, and probably humans,
plied the most of any strain of tubercle bacillus might not be (discussed further in chapter 22).
(1,000,000-fold), and human-type tubercle In guinea pigs, aerosolized BCG provided
bacilli (H37Rv) multiplied the least (1,000- substantial immunization against aerosol chal-
fold). In C57BL/6 mice, both types of tubercle lenge with virulent tubercle bacilli (40, 77).
bacilli multiplied 10,000-fold, and such multi-
plication may be even higher in more suscep-
tible strains of mice (14, 35). In guinea pigs, TUBERCULIN SENSITIVITY
AMONG THE THREE COMMON
H37Rv multiplied 1,000-fold, similar to H37Rv LABORATORY SPECIES
in rabbits. Therefore, guinea pigs (which are The amount of DTH and CMI developed dur-
generally considered the most susceptible of ing infection with tubercle bacilli differs among
these three laboratory species) stop the bacillary these laboratory species (Table 1). Humans are
increase in the logarithmic (symbiotic) stage by far the most sensitive to tuberculin, followed
quite effectively (Fig. 3)—probably because in descending order by guinea pigs, rabbits, and
guinea pigs develop rather high DTH to the mice (see DTH and PPD in the glossary).
tuberculin-like products of the bacillus. The positive skin test is a DTH response to
Vaccination with BCG produces both DTH antigens present in Old Tuberculin, or PPD (a
and CMI. The stopping of the logarithmic purified tuberculin). Caseous necrosis is a DTH
stage requires some DTH, because tubercle response to these and other antigens of the
bacilli in nonactivated macrophages are often tubercle bacillus. Usually, no necrosis is pro-
too numerous for the developing CMI to con- duced by the small amount of tuberculin
trol. In immunized guinea pigs, the logarithmic injected into the skin. However, in pulmonary
stage ends sooner (Fig. 3), initially due to pre- lesions of humans, guinea pigs, and rabbits, the
existing DTH. However, once the stationary local concentrations of bacillary antigens may
stage is reached, both CMI and DTH would reach such high levels that considerable necro-
play more equal roles: the CMI would prevent sis occurs. Therefore, the size of the dermal
renewed intracellular bacillary growth, and the tuberculin reaction does not necessarily reflect
DTH would be effective whenever CMI failed the amount of caseation within the pulmonary
to do so. lesions (see chapter 5).
FATE OF AEROSOLIZED BCG IN
RABBITS, MICE, AND GUINEA PIGS ORGAN RESISTANCE IN
It takes 300 to 3,000 inhaled virulent human- RABBITS AND GUINEA PIGS
type bacilli to produce one grossly visible pri- The livers of rabbits and guinea pigs show oppo-
mary pulmonary tubercle in commercial rab- site susceptibilities to mycobacteria. In rabbits,
bits, and BCG is even less virulent (6). After the liver is resistant and the kidneys are suscep-
commercial rabbits inhaled several million BCG tible. In guinea pigs, the liver is susceptible and
bacilli by aerosol, only a rare primary tubercle the kidneys are resistant (see chapters 7 and 25).
15. TUBERCULOSIS IN RABBITS, MICE,AND GUINEA PIGS 䡵 263

EARLY TUBERCULOUS LESIONS ROLE OF DTH AND CMI IN MOUSE,


IN HUMANS GUINEA PIG, AND RABBIT
Humans are more resistant than mice and guinea TUBERCULOSIS
pigs to virulent human-type tubercle bacilli, Definitions: Similarities and
because only about 5% of recently infected Differences between CMI and DTH
humans continue to progress to clinically active In brief, CMI and DTH are both Th1 lympho-
disease (see chapter 3). In humans, each estab- cyte responses to antigens of the tubercle bacil-
lished microscopic pulmonary tubercle develops lus, and both can activate macrophages to kill or
a small caseous center and reaches grossly visi- inhibit ingested mycobacteria at the appropriate
ble size. However, its progression is usually concentration. The difference between DTH
stopped while it is only 0.5 to 3 mm in diam- and CMI resides in the concentration of the
eter (84–86) (reviewed in chapter 3). antigen that produces each reaction: with DTH,
the tuberculin-like products of the bacillus acti-
CAVITY FORMATION IN vate macrophages at an extremely low concen-
VARIOUS SPECIES tration, and at a slightly higher concentration
Rabbits are the main laboratory animal species kill bacilli-laden macrophages and cause caseous
that readily forms chronic fibrotic cavities simi- necrosis. With CMI, other antigenic products
lar to those found today in adult humans (87–89) of the bacillus activate macrophages to kill or
(Table 1). Guinea pigs do not readily form cavi- inhibit ingested mycobacteria at higher con-
ties, probably because they die rather soon of a centrations and do not cause caseous necrosis at
hematogenously disseminated (often miliary- concentrations usually present in the host.There-
type) tuberculosis.When infected by aerosol with fore, the difference between DTH and CMI is
only 1 to 10 bacillary units, however, guinea pigs merely the host’s sensitivity to the antigen that
may develop cavities (53–55). Mice never develop elicits these reactions. More details are presented
true cavities, apparently because of their low sen- in chapter 5.
sitivity to the tuberculin-like products of the
bacillus (see references 90 and 91 and chapter 4). Species Differences
In both mice and guinea pigs, some local Mice, guinea pigs, rabbits, and humans control
bronchial spread of virulent human-type the progress of tuberculosis by using DTH and
(H37Rv) and bovine-type (Ravenel) tubercle CMI in differing proportions (Table 1 and Fig.
bacilli does occur 7 to 12 weeks after an aerosol 10).Yet, each DTH/CMI proportion seems to
infection (12, 26). Such spread is apparently due be effective in preventing the number of viable
to a discharge of semisolid caseous necrotic ma- tubercle bacilli in the lungs from increasing or
terial into bronchioles and smaller bronchi. How- decreasing for many months.
ever, in both mice and guinea pigs, more distant
spread of the disease via the bronchial tree is rare. Apoptosis and Tissue Necrosis
Rhesus monkeys form cavities but rarely Both apoptosis and tissue necrosis probably play
develop a slowly progressive fibrotic version of the a role in all species. However, mice seem to
disease. Cynomolgus monkeys do develop such have a high proportion of apoptosis; guinea pigs
a chronic disease, but cavities apparently do not and humans (with progressing disease) seem to
occur as frequently as they do in humans and in have a high proportion of necrosis; and rabbits
rabbits infected with virulent bovine-type bacilli. seem to have intermediate levels of necrosis.
In the human population, cavities are the The role of DTH in apoptosis has never been
major reason why the tubercle bacillus spreads investigated.
from person to person. Drugs to prevent the liq-
uefaction of solid caseum and the formation Intracellular and Extracellular
and progression of cavities are greatly needed, Bacillary Dormancy
both for therapy and for reducing the number Mice apparently develop so much CMI that
of new cases of this disease (92) (see chapter 25). tubercle bacilli can remain dormant within
264 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

FIGURE 10 Overview of the characteristics of tuberculosis in laboratory animals and humans.


Note that these groups overlap because of variations among individual members. Liquefaction, cav-
ity formation, extracellular bacillary multiplication, and bronchial dissemination can overwhelm the
hosts in the middle group. CMI, cell-mediated immunity (highly activated macrophages). Caseation,
caseous tissue necrosis.Adapted from reference 4. See Table 1 for additional information.

presumably activated macrophages for months. Tuberculosis in Guinea Pigs


This type of intracellular dormancy must occur When infected with moderate or large numbers
in all species, but whether it occurs to the same of virulent tubercle bacilli, guinea pigs develop
extent as in mice remains to be investigated. In a progressive tissue-destructive type of disease,
other words, the relative amounts of intracellular associated with strong tissue-damaging DTH.
dormancy in macrophages and extracellular They develop appreciable CMI, but not enough
dormancy in solid caseum among the various to arrest the disease. As in Lurie’s susceptible
laboratory species are still unknown. strains of rabbits, bacillary multiplication in
guinea pigs seems to be mostly controlled by
Tuberculosis in Mice DTH, which produces extensive caseous necro-
Mice develop a progressive granulomatous dis- sis. Bacilli are spread by the hematogenous route,
ease with generalized thickening and eventual secondary lesions form, and the disease pro-
fibrosis of the lung parenchyma (11, 33).They gresses until the animal dies.
develop only low sensitivity to tuberculin, and
their lesions usually show relatively little necro- Tuberculosis in Rabbits
sis. Since the vasculature remains patent, lym- Rabbits (and present-day humans) develop a
phocytes, macrophages, and antigen-presenting spectrum of tuberculosis that varies from that
cells are functional throughout each lesion. Mice found in mice to that found in guinea pigs,
evidently develop good CMI and seem to kill depending on the virulence of the bacillus and
bacilli-laden macrophages by apoptosis, rather the native and acquired resistance expressed by
than by tissue-damaging DTH. the host. In commercially available New Zealand
15. TUBERCULOSIS IN RABBITS, MICE,AND GUINEA PIGS 䡵 265

White rabbits (and in immunocompetent adult 7. Lurie, M. B., P. Zappasodi, and C. Tickner.
humans), the disease produced by human-type 1955. On the nature of genetic resistance to tuber-
culosis in the light of the host-parasite relationships
tubercle bacilli is usually controlled rather well, in natively resistant and susceptible rabbits. Am.
due to an effective balance of tissue-damaging Rev.Tuberc. 72:297–323.
DTH and CMI. However, with bovine-type 8. North, R. J., L. Ryan, R. LaCource,T. Mogues,
bacilli in rabbits (and with both types in and M. E. Goodrich. 1999. Growth rate in mice
humans), a solid caseous focus may liquefy and is an unreliable indicator of mycobacterial virulence.
Infect. Immun. 67:5483–5485.
form a cavity. Then, the bacilli may multiply 9. Collins, F. M. 1991. Antituberculous immunity:
extracellularly (for the first time during the new solutions to an old problem. Rev. Infect. Dis.
course of the disease) and may reach such large 13:940–950.
numbers that even the strong CMI and DTH 10. Cardona, P.-J., A. Cooper, M. Luquín,
developed by these hosts cannot prevent a fatal A. Ariza, F. Filipo, I. M. Orme, and V. Ausina.
1999.The intravenous model of murine tubercu-
progression of the disease. losis is less pathogenic than the aerogenic model
Because of the differences in the pathogenesis owing to a more rapid induction of systemic immu-
of tuberculosis in rabbits, mice, and guinea pigs, nity. Scand. J. Immunol. 49:362–366.
all three species should be used for testing new 11. Rhoades, E. R., A. A. Frank, and I. M. Orme.
vaccines prior to their trials in humans. The 1997. Progression of chronic pulmonary tubercu-
losis in mice aerogenically infected with virulent
aerosol route of challenge is preferable to other Mycobacterium tuberculosis. Tuberc. Lung Dis. 78:57–
routes, because it measures both the microbici- 66.
dal efficiency of the alveolar macrophage pop- 12. Cardona, P.-J., S. Gordillo, J. Díaz, G. Tapia,
ulation (a component of innate resistance) and I.Amat, Á. Pallarés, C.Vilaplana,A.Ariza, and
the acquired immunity (DTH and CMI) pro- V. Ausina. 2003. Widespread bronchogenic dis-
semination makes DBA/2 mice more susceptible
duced by the vaccine. than C57BL/6 mice to experimental aerosol infec-
tion with Mycobacterium tuberculosis. Infect. Immun.
71:5845–5954.
REFERENCES 13. Kelley, C. L., and F. M. Collins. 1999. Growth
1. Rees, R. J.W., and P. D. Hart. 1961.Analysis of of a highly virulent strain of Mycobacterium tubercu-
the host-parasite equilibrium in chronic murine losis in mice of differing susceptibility to tubercu-
tuberculosis by total and viable bacillary counts. lous challenge. Tuberc. Lung Dis. 79:367–370.
Br. J. Exp. Pathol. 42:83–88. 14. Medina, E., and R. J. North. 1998. Resistance
2. Muñoz-Elías, E. J., J. Timm, T. Botha, W.-T. ranking of some common inbred mouse strains to
Chan, J. E. Gomez, and J. D. McKinney. 2005. Mycobacterium tuberculosis and relationship to major
Replication dynamics of Mycobacterium tuberculosis histocompatibility complex haplotype and Nramp1
in chronically infected mice. Infect. Immun. 73:546– genotype. Immunology 93:270–274.
551. 15. Kornfeld, H., G. Mancino, and V. Colizzi.
3. Higuchi, S., M. Suga, A. M. Dannenberg, Jr., 1999.The role of macrophage cell death in tuber-
L. F. Affronti, I. Azuma, T. M. Daniel, and culosis. Cell Death Differ. 6:71–78.
J. P. Petrali. 1981. Persistence of protein, carbo- 16. Mustafa,T., S. Phyu, R. Nilsen, G. Bjune, and
hydrate and wax components of tubercle bacilli in R. Jonsson. 1999. Increased expression of Fas lig-
dermal BCG lesions. Am. Rev. Respir. Dis. 123:397– and on Mycobacterium tuberculosis infected macro-
401. phages: a potential novel mechanism of immune
4. Dannenberg, A. M., Jr., and F. M. Collins. evasion by Mycobacterium tuberculosis? Inflammation
2001. Progressive pulmonary tuberculosis is not 23:507–521.
due to increasing numbers of viable bacilli in rab- 17. Moss, J. E., A. O. Aliprantis, and A. Zychlin-
bits, mice and guinea pigs, but is due to a contin- sky. 1999.The regulation of apoptosis by microbial
uous host response to mycobacterial products. Tuber- pathogens. Int. Rev. Cytol. 187:203–259.
culosis 81:229–242. 18. Flynn, J. L., M. M. Goldstein, K. J. Triebold,
5. Helke, K., J. L. Mankowski, and Y. C. Manabe. B. O. Koller, and B. R. Bloom. 1992. Major
13 December 2005.Animal models of cavitation in histocompatibility complex class I-restricted T
pulmonary tuberculosis. Tuberculosis (Edinb.) [Epub cells are required for resistance to Mycobacterium
ahead of print]. tuberculosis infection. Proc. Natl. Acad. Sci. USA
6. Lurie, M. B. 1964. Resistance to Tuberculosis, Exper- 89:12013–12017.
imental Studies in Native and Acquired Defensive Mech- 19. Behar, S. M., C. C. Dascher, M. J. Grusby,
anisms. Harvard University Press, Cambridge, Mass. C.-R. Wang, and M. B. Brenner. 1999.
266 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

Susceptibility of mice deficient in CD1D or TAP1 32. Ratcliffe, H. L. 1952. Tuberculosis induced by
to infection with Mycobacterium tuberculosis. J. Exp. droplet nuclei infection: pulmonary tuberculosis
Med. 189:1973–1980. of predetermined initial intensity in mammals. Am.
20. Orme, I. M., and F. M. Collins. 1994. Mouse J. Hyg. 55:36–48.
model of tuberculosis, p. 113–134. In B. R. Bloom 33. Dunn, P. L., and R. J. North. 1995.Virulence
(ed.), Tuberculosis: Pathogenesis, Protection, and Control. ranking of some Mycobacterium tuberculosis and
ASM Press,Washington, D.C. Mycobacterium bovis strains according to their abil-
21. McMurray, D. N., F. M. Collins, A. M. Dan- ity to multiply in the lungs, induce lung pathology,
nenberg, Jr., and D. W. Smith. 1996. Patho- and cause mortality in mice. Infect. Immun. 63:3428–
genesis of experimental tuberculosis in animal mod- 3437.
els, p. 157–179. In T. M. Shinnick (ed.), Tuberculosis. 34. Dunn, P. L., and R. J. North. 1996. Persistent
Springer Publishing Co., Berlin, Germany. infection with virulent but not avirulent Myco-
22. Gil, O., E. Guirado, S. Gordillo, J. Díaz, bacterium tuberculosis in the lungs of mice causes
G.Tapia, C.Vilaplana, A. Ariza,V. Ausina, and progressive pathology. J. Med. Microbiol. 45:103–
P.-J. Cardona. 2006. Intragranulomatous necrosis 109.
in lungs of mice infected by aerosol with Mycobac- 35. Medina, E., and R. J. North. 1999. Genetically
terium tuberculosis is related to bacterial load, rather susceptible mice remain proportionally more sus-
than to any one cytokine or T cell type. Microbes ceptible to tuberculosis after vaccination. Immunol-
Infect. 8:628–636. ogy 96:16–21.
23. Cardona, P.-J. 2006. RUTI: a new chance to 36. North, R. J., and A. A. Izzo. 1993. Granuloma
shorten the treatment of latent tuberculosis infec- formation in severe combined immunodeficient
tion. Tuberculosis (Edinb.) 86:273–289. (SCID) mice in response to progressive BCG infec-
24. Cooper, A. M., D. K. Dalton, T. A. Stewart, tion. Am. J. Pathol. 142:1959–1966.
J. P. Griffin, D. G. Russell, and I. M. Orme. 37. Cardona, P.-J., R. Llatjós, S. Gordillo, J. Díaz,
(1993) Disseminated tuberculosis in interferon- I. Ojanguren, A. Ariza, and V. Ausina. 2000.
gene-disrupted mice. J. Exp. Med. 178:2243–2247. Evolution of granulomas in lungs of mice infected
25. Flynn, J. L., J. Chan, K. J.Triebold, D. K. Dal- aerogenically with Mycobacterium tuberculosis. Scand.
ton,T. A. Stewart, and B. R. Bloom. 1993.An J. Immunol. 52:156–163.
essential role for interferon in resistance to Mycobac- 38. Courtade, E. T., T. Tsuda, C. R. Thomas, and
terium tuberculosis infection. J. Exp. Med. 178:2249– A. M. Dannenberg, Jr. 1975. Capillary density in
2254. developing and healing tuberculous lesions pro-
26. Ratcliffe, H. L., and V. S. Palladino. 1953. duced by BCG in rabbits.A quantitative study. Am.
Tuberculosis induced by droplet nuclei infection. J. Pathol. 78:243–260.
Initial homogeneous response of small mammals 39. Abe,Y., K. Sugisaki, and A. M. Dannenberg,
(rats, mice, guinea pigs, and hamsters) to human and Jr. 1996. Rabbit vascular endothelial adhesion
to bovine bacilli and the rate and pattern of tuber- molecules, ELAM-1 is most elevated in acute
cle development. J. Exp. Med. 97:61–68. inflammation, whereas VCAM-1 and ICAM-1 pre-
27. Guirado, E., S. Gordillo, O. Gil, J. Díaz, dominate in chronic inflammation. J. Leukoc. Biol.
G.Tapia, C.Vilaplana,V.Ausina, P.-J. Cardona. 60:692–703.
2006. Intragranulomatous necrosis in pulmonary 40. Smith, D.W., D. N. McMurray, E. H.Wieges-
granulomas is not related to resistance against haus, A. A. Grover, and G. E. Harding. 1970.
Mycobacterium tuberculosis infection in experimental Host-parasite relationships in experimental air-
murine models induced by aerosol. Int. J. Exp. Pathol. borne tuberculosis. IV. Early events in the course of
87:139–149. infection in vaccinated and nonvaccinated guinea
28. Cardona, P.-J., R. Llatjós, S. Gordillo, J. Díaz, pigs. Am. Rev. Respir. Dis. 102:937–949.
B. Viñado, A. Ariza, and V. Ausina. 2001. 41. Alsaadi, A.-I., and D.W. Smith. 1973.The fate
Towards a ‘human-like’ model of tuberculosis: of virulent and attenuated mycobacteria in guinea
Intranasal inoculation of LPS induces intragranu- pigs infected by the respiratory route. Am. Rev.
lomatous lung necrosis in mice infected aerogeni- Respir. Dis. 107:1041–1046.
cally with Mycobacterium tuberculosis. Scand. J. 42. Balasubramanian, V., E. H. Wiegeshaus, and
Immunol. 53:65–71. D. W. Smith. 1992. Growth characteristics of
29. Majno, G., and I. Joris. 1996. Cells,Tissues, and recent sputum isolates of Mycobacterium tuberculosis
Disease, p. 509–556. Blackwell Science, Cambridge, in guinea pigs infected by the respiratory route.
Mass. Infect. Immun. 60:4762–4767.
30. Saunders, B. M., and A. M. Cooper. 2000. 43. Francis, J. 1958. Tuberculosis in Animals and Man.
Restraining mycobacteria: role of granulomas in A Study in Comparative Pathology. Cassell & Com-
mycobacterial infections. Immunol. Cell Biol. 78:334– pany Ltd., London, U.K.
341. 44. Lurie, M. B. 1939. Studies on the mechanism of
31. Flynn, J. L., and J. Chan. 2001. Immunology of immunity in tuberculosis: the role of extracellular
tuberculosis. Annu. Rev. Immunol. 19:93–129. factors and local immunity in the fixation and inhi-
15. TUBERCULOSIS IN RABBITS, MICE,AND GUINEA PIGS 䡵 267

bition of growth of tubercle bacilli. J. Exp. Med. In B. R. Bloom (ed.), Tuberculosis: Pathogenesis, Pro-
69:555–577. tection, and Control.ASM Press,Washington, D.C.
45. Mills, C. C., F. O’Grady, and R. L. Riley. 1960. 59. Barclay, W. R., R. L. Anacker, W. Brehmer,
Tuberculin conversion in the “naturally infected” W. Leif, and E. Ribi. 1970. Aerosol-induced
guinea pig. Bull. Johns Hopkins Hosp. 106:36–45. tuberculosis in subhuman primates and the course
46. Middlebrook, G., and M. L. Cohn. 1953. Some of the disease after intravenous BCG vaccination.
observations on the pathogenicity of isoniazid- Infect. Immun. 2:574–582.
resistant variants of tubercle bacilli. Science 118:297– 60. Thoen, C. O. 1994. Tuberculosis in wild and
299. domestic mammals, p. 157–162. In B. R. Bloom
47. Steenkin, W., and E. Wolinsky. 1953.Virulence (ed.), Tuberculosis: Pathogenesis, Protection, and Control.
of tubercle bacilli recovered from patients treated ASM Press,Washington, D.C.
with isoniazid. Am. Rev.Tuberc. 68:548–556. 61. Good, R. C. 1968. Biology of the mycobacterioses.
48. Rist, N. 1956. Pathogenicity of isoniazid-resistant Simian tuberculosis: immunologic aspects. Ann. N.
tubercle bacilli and prophylaxis of tuberculosis in Y.Acad. Sci. 154:200–213.
children. Am. Rev.Tuberc. 74(Aug. Pt. 2):75–99. 62. Good, R. C. 1973. Tuberculosis and bacterial
49. Turner, O. C., R. J. Basaraba, and I. M. Orme. infection, p. 39–60. In G. H. Bourne (ed.), Non-
2003. Immunopathogenesis of pulmonary granu- human Primates and Medical Research.Academic Press,
lomas in the guinea pig after infection with Mycobac- Inc., New York, N.Y.
terium tuberculosis. Infect. Immun. 71:864–871. 63. Schmidt, L. H. 1956. Some observations on the
50. Baldwin, S. L., C. D’Souza, A. D. Roberts, utility of simian pulmonary tuberculosis in defin-
B. P. Kelly,A.A. Frank, M.A. Lui, J. B. Ulmer, ing the therapeutic potentialities of isoniazid. Am.
K. Huygen, D. McMurray, and I. M. Orme. Rev.Tuberc. 74(Pt. II):138–159.
1998. Evaluation of new vaccines in the mouse 64. Francis, J. 1956. Natural and experimental tuber-
and guinea pig model of tuberculosis. Infect. Immun. culosis in monkeys. J. Comp. Pathol. 66:123–135.
66:2951–2959. 65. Borrel, A. 1920. Pneumonie et tuberculose chez
51. McMurray, D. N. 1994. Guinea pig model of les troupes noires. Ann. Inst. Pasteur 34:105–148.
tuberculosis, p. 135–147. In B. R. Bloom (ed.), 66. Barclay, W. R., W. M. Busey, D. W. Dalgard,
Tuberculosis: Pathogenesis, Protection, and Control. R. C. Good, B.W. Janicki, J. E. Kasik, E. Ribi,
ASM Press,Washington, D.C. C. E. Ulrich, and E.Wolinsky. 1973. Protection
52. Fok, J. S., R. S. Ho, P. K. Arora, G. E. Hard- of monkeys against airborne tuberculosis by aerosol
ing, and D. W. Smith. 1976. Host-parasite rela- vaccination with Bacillus Calmette-Guérin. Am.
tionships in experimental airborne tuberculosis. Rev. Respir. Dis. 107:351–358.
V. Lack of hematogenous dissemination of Mycobac- 67. Habel, K. 1947.Tuberculosis in a laboratory mon-
terium tuberculosis to the lungs in animals vaccinated key colony: its spread and control. Am. Rev.Tuberc.
with Bacille Calmette-Guérin. J. Infect. Dis. 133: 55:77–92.
137–144. 68. Converse, P. J., A. M. Dannenberg, Jr., J. E.
53. Lurie, M. B. 1930. Experimental epidemiology of Estep, K. Sugisaki,Y.Abe, B. H. Schofield, and
tuberculosis: the effect of eliminating exposure to M. L. M. Pitt. 1996. Cavitary tuberculosis pro-
enteric infection on the incidence and course of duced in rabbits by aerosolized virulent tubercle
tuberculosis acquired by normal guinea pigs con- bacilli. Infect. Immun. 64:4776–4787.
fined with tuberculous cage mates. J. Exp. Med. 69. Converse, P. J., A. M. Dannenberg, Jr.,T. Shi-
51:753–768. genaga, D. N. McMurray, S. W. Phalen, J. L.
54. Lurie, M. B. 1930. Experimental epidemiology of Stanford, G. A. W. Rook, T. Koru-Sengul,
tuberculosis: airborne contagion of tuberculosis in H. Abbey, J. E. Estep, and M. L. M. Pitt. 1998.
an animal room. J. Exp. Med. 51:743–751. Pulmonary bovine-type tuberculosis in rabbits:
55. Smith, D. W., and G. E. Harding. 1977. Pul- bacillary virulence, inhaled dose effects, tuberculin
monary tuberculosis.Animal model: experimental sensitivity, and Mycobacterium vaccae immunotherapy.
airborne tuberculosis in the guinea pig. Am. J. Clin. Diagn. Lab. Immunol. 5:871–881.
Pathol. 89:273–276. 70. Walsh, G. P., E.V.Tan, E. C. dela Cruz, R. M.
56. Wright, S., and P.A. Lewis. 1921. Factors in the Abalos, L. G. Villahermosa, L. J. Young,
resistance of guinea pigs to tuberculosis, with espe- R.V. Cellona, J. B. Nazareno, and M. A. Hor-
cial regard to inbreeding and heredity. Am. Nat. witz. 1996. The Philippine cynomolgus monkey
55:20–50. (Macaca fasicularis) provides a new nonhuman pri-
57. Cohen, M. K., R. A. Bartow, C. L. Mintzer, mate model of tuberculosis that resembles human
and D. N. McMurray. 1987. Effects of diet and disease. Nat. Med. 2:430–436.
genetics on Mycobacterium bovis BCG vaccine effi- 71. Capuano, S. V., III, D. A. Croix, S. Pawar,
cacy in inbred guinea pigs. Infect. Immun. 55:314– A. Zinovik, A. Myers, P. L. Lin, S. Bissel,
319. C. Fuhrman, E. Klein, and J. L. Flynn. 2003.
58. Rook, G.A.W., and B. R. Bloom. 1994. Mech- Experimental Mycobacterium tuberculosis infection
anisms of pathogenesis in tuberculosis, p. 485–501. of cynomolgus macaques closely resembles the
268 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

various manifestations of human M. tuberculosis 83. Nuermberger, E. L., T.Yoshimatsu, S. Tyagi,


infection. Infect. Immun. 71:5831–5844. W. R. Bishai, and J. H. Grosset. 2004. Pau-
72. Langermans, J.A., P.Andersen, D. van Soolin- cibacillary tuberculosis in mice after prior aerosol
gen, R. A. Vervenne, P. A. Frost, T. van der immunization with Mycobacterium bovis BCG. Infect.
Laan, L. A. van Pinxteren, J. van den Immun. 72:1065–1071.
Hombergh, S. Kroon, I. Peekel, S. Florquin, 84. Lindgren, I. 1961. Anatomical and roentgeno-
and A. W. Thomas. 2001. Divergent effect of logic studies of tuberculous infections in BCG-
Bacillus Calmette-Guérin (BCG) vaccination on vaccinated and non-vaccinated subjects, with
Mycobacterium tuberculosis infection in highly related biophysical investigations of calcified foci. Acta
macaque species: implications for primate models Radiol. Suppl. 209:1–101.
in tuberculosis vaccine research. Proc. Natl.Acad. Sci. 85. Sutherland, I., and I. Lindgren. 1979.The pro-
USA 98:11497–11502. tective effect of BCG vaccination as indicated by
73. Lurie, M. B., P. Zappasodi, E. Cardona-Lynch, autopsy studies. Tubercle 60:225–231.
and A. M. Dannenberg, Jr. 1952.The response 86. Lindgren, I. 1965.The pathology of tuberculous
to the intracutaneous inoculation of BCG as an infection in BCG-vaccinated humans. Adv.Tuberc.
index of native resistance to tuberculosis. J. Immunol. Res. 14:202–234.
68:369–387. 87. Rich, A. R. 1951. The Pathogenesis of Tuberculosis,
74. McMurray, D. N. 2000. A nonhuman primate 2nd ed. Charles C Thomas Publisher, Springfield,
model for preclinical testing of new tuberculosis Ill.
vaccines. Clin. Infect. Dis. 30(Suppl. 3):S210–S212. 88. Canetti, G. 1955. The Tubercle Bacillus in the Pul-
75. Allison, M. J., P. Zappasodi, and M. B. Lurie. monary Lesion of Man. Springer Publishing Co.,
1962. Host-parasite relationships in natively resis- New York, N.Y.
tant and susceptible rabbits on quantitative inhala- 89. Dannenberg,A. M., Jr., and J. F.Tomashefski,
tion of tubercle bacilli. Their significance for the Jr. 1998. Pathogenesis of pulmonary tuberculosis,
nature of genetic resistance. Am. Rev. Respir. Dis. p. 2447–2471. In A. P. Fishman (ed.), Fishman’s Pul-
85:553–569. monary Diseases and Disorders, 3rd ed., vol. 2.
76. Ho, R. S., J. S. Fok, G. E. Harding, and D. W. McGraw-Hill Co., New York, N.Y.
Smith. 1978. Host-parasite relationships in exper- 90. Yamamura,Y. 1958.The pathogenesis of tuber-
imental airborne tuberculosis. VII. Fate of Myco- culous cavities. Adv.Tuberc. Res. 9:13–37.
bacterium tuberculosis in primary lung lesions and in 91. Yamamura, Y., Y. Ogawa, H. Maeda, and
primary lesion-free lung tissue infected as a result Y. Yamamura. 1974. Prevention of tuberculous
of bacillemia. J. Infect. Dis. 138:237–241. cavity formation by desensitization with tuberculin-
77. Middlebrook, G. 1961. Immunological aspects active peptide. Am. Rev. Respir. Dis. 109:594–601.
of airborne infection: reactions to inhaled antigens. 92. Grosset, J. 2003. Mycobacterium tuberculosis in the
Bacteriol. Rev. 25:331–346. extracellular compartment: an underestimated
78. Dannenberg, A. M., Jr. 1994. Rabbit model of adversary. Antimicrob. Agents Chemother. 47:833–
tuberculosis, p. 149–156. In B. R. Bloom (ed.), 836.
Tuberculosis: Pathogenesis, Protection, and Control. 93. Dannenberg, A. M., Jr. 1984. Pathogenesis of
ASM Press,Washington, D.C. tuberculosis: native and acquired resistance in animals
79. North, R. J., and A. A. Izzo. 1993. Mycobacte- and humans, p. 344–354. In Microbiology—1984.
rial virulence.Virulent strains of Mycobacterium tuber- American Society for Microbiology, Washington,
culosis have faster in vivo doubling times and are bet- D.C.
ter equipped to resist growth-inhibiting functions 94. Bishai, W. R., A. M. Dannenberg, Jr., N. Par-
of macrophages in the presence and absence of rish, R. Ruiz, P. Chen, B. C. Zook, W. John-
specific immunity. J. Exp. Med. 177:1723– son, J. W. Boles, and M. L. M. Pitt. 1999.
1733. Virulence of Mycobacterium tuberculosis CDC1551
80. Jacobs, W. R. 1992. Advances in mycobacterial and H37Rv in rabbits evaluated by Lurie’s pul-
genetics: new promises for old diseases. Immunobi- monary tubercle count method. Infect. Immun.
ology 184:147–156. 67:4931–4934.
81. Bloom, B. R. 1989. New approaches to vaccine 95. Dannenberg, A. M., Jr., W. R. Bishai, N. Par-
development. Rev. Infect. Dis. 11(Suppl. 2):S460– rish, R. Ruiz, W. Johnson, B. C. Zook, J. W.
S466. Boles, and M. L. M. Pitt. 2000. Efficacies of
82. Morrison, N. E., and F. M. Collins. 1975. BCG and vole bacillus (Mycobacterium microti) vac-
Immunogenicity of an aerogenic BCG vaccine in cines in preventing clinically apparent pulmonary
T-cell-depleted and normal mice. Infect. Immun. tuberculosis in rabbits: a preliminary report. Vaccine
11:1110–1121. 19:796–800.
15. TUBERCULOSIS IN RABBITS, MICE,AND GUINEA PIGS 䡵 269

96. Manabe, Y. C., A. M. Dannenberg, Jr., S. K. 97. Dorman, S., C. L. Hatem, S. Tyagi, K. Aird,
Tyagi, C. L. Hatem, M.Yoder, S. C.Woolwine, J. Lopez-Molina, M. L. M. Pitt, B. C. Zook,
B. C. Zook, M. L. M. Pitt, and W. R. Bishai. A. M. Dannenberg, Jr.,W. R. Bishai, and Y. C.
2003. Different strains of Mycobacterium tuberculosis Manabe. 2004. Susceptibility to tuberculosis: clues
cause various spectrums of disease in the rabbit from studies with inbred and outbred New Zealand
model of tuberculosis. Infect. Immun. 71:6004–6011. White rabbits. Infect. Immun. 72:1700–1705.
This page intentionally left blank
Section 5.

EFFECTS OF HORMONES
AND X-IRRADIATION
ON TUBERCULOSIS
This page intentionally left blank
16
EFFECTS OF CORTISONE AND
ADRENOCORTICOTROPIC
HORMONE ON TUBERCULOSIS

Effects of pharmacological concentrations of cortisone on the development


of tuberculosis [273]
Effects of cortisone withdrawal [275]
Reactivation by glucocorticoids of healing pulmonary tubercles [279]
Effects of physiological concentrations of glucocorticoids, illustrated by
studies with adrenocorticotropic hormone (ACTH) in inbred rabbits
with small adrenals [280]
Effects of dehydroepiandrosterone (DHEA) and its derivative
3,17-androstenediol (AED) [282]
Addendum [283]

Abstract. Pharmacological amounts of glucocorticoids are frequently given as therapy for


a variety of allergic, autoimmune, and inflammatory conditions, such as asthma and
rheumatoid arthritis.When such drugs are continued for long periods of time, latent tuber-
culosis may reactivate.
In tuberculous rabbits infected with human-type tubercle bacilli, pharmacological
amounts of glucocorticoids decreased cell-mediated immunity and delayed-type hyper-
sensitivity. Macrophages were poorly activated, and tubercle bacilli grew to large numbers
within these phagocytes.After glucocorticoid administration was stopped, the tuberculin
sensitivity returned, and (because of the large numbers of bacilli) liquefaction, cavity for-
mation, tuberculous bronchopneumonia, and hematogenous dissemination occurred in
some of the rabbits.
One of Lurie’s inbred rabbit strains was evidently deficient in glucocorticoid produc-
tion. In these rabbits, the administration of physiological doses of adrenocorticotropic hor-
mone increased their resistance to tuberculosis.

Glucocorticoids are hormones that physiolog- tisone acetate (2 mg/kg) every 2 days for 38
ically enable the host to adapt to stress (1). In days. This dosage produced pharmacological
fact, rabbits with tuberculosis show hyperplasia glucocorticoid effects, many of which are listed
of the adrenal cortices (2), which helps them in Table 1: (i) a decrease in the number of cir-
control the progress of this disease. Only phar- culating lymphocytes and in the size of the
macological concentrations of glucocorticoids spleen (from both bone marrow suppression
have an adverse effect on tuberculosis (see and from lympholysis) (3, 4), (ii) an increase in
below). References 3 through 5 review their fasting blood sugar and a decrease in blood
effects on inflammatory processes. ascorbic acid, (iii) an increase in liver size (from
an increase in glycogen and fat content), (iv) a
decrease in the size of the adrenals, thyroid, and
EFFECTS OF PHARMACOLOGICAL
CONCENTRATIONS OF CORTISONE testicles (from suppression of various pituitary
ON THE DEVELOPMENT OF hormones), (v) an increase in liver and spleen
TUBERCULOSIS phagocytosis of carbon particles (India ink)
In a series of experiments in the 1950s, Lurie (2, injected intravenously, and (vi) a decrease in the
6–9) injected rabbits intramuscularly with cor- dermal reaction to nonspecific irritants (2, 9).

273
274 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

TABLE 1 Effects of cortisone on tuberculosis produced by the inhalation of human-type tubercle bacilli
(H37Rv) 5 weeks previouslya
Rabbits receiving cortisone Controls P values
No. of inhaled tubercle bacilli required to 35 ⫾ 2.9 120 ⫾ 22 0.002
produce one primary pulmonary tubercle
(“the ratio”)b
Average diameter of tubercles (mm) 2.5 ⫾ 0.33 5.6 ⫾ 0.24 0.000
Adrenal weight (mg/kg body weight) 66 ⫾ 6 128 ⫾ 10 0.000
Liver weight (g/kg body weight) 48 ⫾ 2.9 20 ⫾ 0.9 0.000
Spleen weight (mg/kg body weight) 310 ⫾ 20 530 ⫾ 30 0.000
Thyroid weight (mg/kg body weight) 40 ⫾ 0.5 47 ⫾ 1.3 0.000
Testes weight (mg/kg body weight) 714 ⫾ 96 1480 ⫾ 60 0.000
a
The pharmacological effects of cortisone are shown by the reduction in weight of the adrenals, spleen, thyroid, and testicles,
and by the increase in weight of the liver (see text).Adapted from reference 2.
b
“The ratio” is the number of inhaled human-type tubercle bacilli (H37Rv) divided by the number of grossly visible primary
pulmonary tubercles, usually counted at 5 weeks after infection, i.e., the number of inhaled bacilli required to produce one such
tubercle.The ratios in this table were calculated by the Lurie method (see chapter 11). In the cortisone-treated rabbits, the num-
ber of such primary tubercles was increased, i.e., fewer inhaled tubercle bacilli were needed to produce one such tubercle, but the
size of these tubercles was decreased (an anti-inflammatory effect).

On the third day after the second cortisone treatment had the expected pharmacological
injection, the rabbits were exposed to an aerosol effects, because at 5 weeks the number of cir-
of virulent human-type tubercle bacilli (H37Rv) culating white blood cells as well as the weights
(6). Five weeks later, the rabbits were euthanized, of the spleens and adrenal glands were decreased.
and the number of primary pulmonary tuber- The dexamethasone-treated rabbits also showed
cles was counted.The cortisone-treated rabbits a decreased percentage of lung CD4 cells and
had 3 to 4 times the number of tubercles in their splenic CD4 and CD8 cells (10). However,
lungs when compared with the untreated rab- unlike Lurie’s rabbits treated with cortisone,
bits (Table 1 and Fig. 1). Although they were the number of grossly visible pulmonary tuber-
more numerous, the tubercles in the cortisone- cles in the dexamethasone-treated group was not
treated group were much smaller (due to the significantly increased over the number in the
anti-inflammatory effects of the steroid) (Table controls, and the size of these tubercles was not
1 and Fig. 1) and contained many more tuber- significantly smaller. The difference in results
cle bacilli within live and dead macrophages, from those found by Lurie was probably because
which formed intra-alveolar plugs (Fig. 2) (2, 9). commercial rabbits are more resistant to tuber-
In the cortisone-treated rabbits, the alveolar culosis than Lurie’s susceptible rabbits and
walls remained more or less normal, whereas because a somewhat less virulent strain of
those in the control rabbits were thickened by human-type tubercle bacillus (CDC1551) was
cell infiltration and often destroyed by the used to infect the commercial rabbits.
inflammatory process (Fig. 2). In the steroid- In 1978, our laboratory studied the effects of
treated group, fewer bacilli drained (via lym- cortisone acetate (2 mg/kg every 2 days) on rab-
phatics) to the hilar lymph nodes (because of bit dermal BCG lesions (11), because we had
reduced perifocal inflammation) (2, 9). The recently developed the -galactosidase histo-
steroid also reduced the size of the tuberculin chemical test for macrophage activation (12–14).
skin test reaction (2, 9). This steroid greatly reduced the size of the BCG
In an experiment similar to Lurie’s cortisone lesions (Fig.3),the amount of cell infiltration (Fig.
experiment, commercial rabbits were treated 4), the number of activated macrophages present
with dexamethasone (10). Dexamethasone (Fig. 4), and the size of their caseous centers
16. CORTISONE AND ANDRENOCORTICOTROPIC HORMONE 䡵 275

FIGURE 1 Lungs of control rabbit FC 2-46 (left) and cortisone-treated rabbit FC 2⫽1
(right) 5 weeks after the inhalation of aerosolized virulent human-type tubercle bacilli (H37Rv).
The lungs are black, because shortly before the rabbits were euthanized, an intravenous injection
of carbon particles (India ink) was made to assess the phagocytic abilities in the liver and spleen.
Note that (i) fewer grossly visible primary pulmonary tubercles are present in the corticosteroid-
treated animal, (ii) their size is smaller, (iii) their caseous centers are whiter, and (iv) little perifocal
inflammation exists around the caseous centers. Reproduced with permission from reference 6.

(Fig. 5).Their dermal tuberculin reactions were EFFECTS OF CORTISONE


also reduced (Fig. 3). WITHDRAWAL
In brief, pharmacological amounts of gluco- Lurie’s susceptible Ca rabbits were infected with
corticosteroids decreased the ability of the aerosolized human-type tubercle bacilli
macrophages to destroy tubercle bacilli by (H37Rv) and similarly treated with cortisone for
decreasing both the inflammatory and immune 6.5 weeks (2). Then, the glucocorticoid treat-
responses of the host (3–5). ment was stopped. Five weeks later, these ani-
Despite these adverse effects of glucocorti- mals died of massive caseous pneumonia, liq-
coids on host resistance to tuberculosis, they uefaction, and spread of bacilli by both bronchial
may be lifesaving in patients with either tuber- and hematogenous routes (Fig. 6). Such large
culous meningitis or caseous pneumonia when numbers of bacilli had accumulated in the lungs
combined with effective antimicrobial therapy of the cortisone-treated host that the disease
(15–18; see also reference 19). progressed to death. As expected, the control
276 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

FIGURE 2 A tissue section of a


primary tubercle from the lungs of
a cortisone-treated rabbit (A) and
a control rabbit (B), 5 weeks after
the inhalation of an aerosol of viru-
lent human-type tubercle bacilli
(H37Rv). These specimens were
from the same experiment as that
depicted in Fig. 1. (A) The tubercle
of the cortisone-treated rabbit shows
intra-alveolar plugs of necrotic
macrophages containing very large
numbers of tubercle bacilli.The alve-
olar walls are thin with little or no
cell infiltration, because the gluco-
corticoids reduced the inflamma-
tory response. (B) The tubercle of the
control rabbit (which received no
glucocorticoids) shows extensive
perifocal inflammation and more
mature (i.e., more homogeneous)
caseation. Relatively few bacilli were
present (not visible at this magnifi-
cation). Reproduced with permis-
sion from reference 6.

(untreated) susceptible rabbits in this experi- The liquefaction of tuberculous lesions and
ment showed regressive pulmonary tubercles bronchial spread of the disease never occurred
with evidence of healing (Fig. 6).All untreated before in Lurie’s susceptible rabbits.These rab-
rabbits, including inbred susceptible rabbits, usu- bits apparently do not form cavities.That these
ally recover from infection with virulent human- phenomena occurred in susceptible rabbits after
type tubercle bacilli. withdrawal of corticosteroids suggests that the
16. CORTISONE AND ANDRENOCORTICOTROPIC HORMONE 䡵 277

FIGURE 3 Size of BCG lesions and size of dermal tuberculin reactions in cortisone-treated
and control rabbits at various times after the BCG lesions were produced. Cortisone treatment
markedly reduced the size of both the BCG lesions and the tuberculin reactions, because there
was less cell infiltration in each (see Fig. 4). Reproduced with permission from reference 11.

local presence of large numbers of tubercle The high resistance of these rabbits enabled them
bacilli plays some role in the liquefaction process to survive during the period of study, despite the
(see chapter 4). fact that glucocorticoid treatment had caused
In a similar experiment, after corticosteroid large numbers of human-type tubercle bacilli
withdrawal, Lurie’s resistant rabbits showed a to be present in their lungs. In this experiment,
chronic cavitary form of the disease (Fig. 7) (2). the control (untreated) resistant rabbits showed
FIGURE 4 Tissue sections of 12-day BCG lesions from a cortisone-treated rabbit (A) and a
control rabbit (B) stained for -galactosidase, our marker enzyme for macrophage activation (12–
14). Near the necrotic center of the control lesion are large -galactosidase-positive epithelioid
cells (activated macrophages). Such mature epithelioid cells are almost absent in the lesion of the
cortisone-treated rabbit (P ⫽ 0.001). Stained with 5-bromo-4-chloro-3-indolyl--D-galactoside,
hematoxylin, and carbol-fuchsin.
Both photographs are at the same magnification (⫻155).When macrophages become activated,
they greatly increase in size. Reproduced with permission from reference 11.

278
16. CORTISONE AND ANDRENOCORTICOTROPIC HORMONE 䡵 279

FIGURE 5 Tissue sections of 18-day BCG lesions from a cortisone-treated rabbit (A) and a
control rabbit (B), showing the size of their necrotic liquefied centers. Note that cortisone treat-
ment reduced the lesion size, the amount of cell infiltration, the amount of necrosis, and the -
galactosidase activity (recognized here by the dark-staining cells in the infiltrate). Stained with
5-bromo-4-chloro-3-indolyl--D-galactoside, hematoxylin, and carbol-fuchsin. Magnification,
⫻10. Reproduced with permission from reference 11.
The volumes of the necrotic centers illustrated were 16 mm3 in the cortisone-treated rabbit
and 46 mm3 in the control rabbit.The average volume of these centers for the six cortisone-treated
rabbits (in the 18-day BCG group of this experiment) was 5.0 ⫾ 2.5 mm3, and that for the six
control rabbits was 28 ⫾ 7 mm3 (P ⫽ 0.01) (11).

only a few minute regressive pulmonary tuber- Commercial rabbits were infected by aerosol
cles (Fig. 7). with human-type tubercle bacilli (H37Rv) and
divided into experimental and control groups.
REACTIVATION BY Ten weeks after infection, when the pulmonary
GLUCOCORTICOIDS OF HEALING lesions were healing, the rabbits in the experi-
PULMONARY TUBERCLES mental group were injected intramuscularly with
The following data are as yet unpublished (Y. C. dexamethasone (0.15 mg/kg on 6 days per
Manabe et al., in preparation). week) for the next 5 weeks.Twenty weeks after
280 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

FIGURE 6 Lungs of a control rabbit of the inbred susceptible Ca strain (Ca 5-23) (left)
and a cortisone-treated rabbit of the same strain (Ca 4-7) (right).These rabbits had inhaled
aerosols of virulent human-type tubercle bacilli (H37Rv) 11 weeks previously.The corti-
sone treatment was started 4 days before infection and continued for 6 weeks during infec-
tion. Five weeks after the cortisone treatment was stopped, the rabbit died of massive
caseous pneumonia with liquefaction (shown on the right). It had both bronchial and
hematogenous spread of the disease.At this time, the control rabbit (which had received dilu-
ent alone) had regressive pulmonary tubercles with evidence of healing (shown on the left).
These studies clearly demonstrate the dangers of pharmacological glucocorticoid ther-
apy in tuberculous hosts, i.e., a marked decrease in the host’s ability to control bacillary growth
and, after hormone withdrawal, an extensive delayed-type hypersensitivity reaction to accu-
mulated bacillary products (see text).
Reproduced with permission from reference 2.

infection (5 weeks after the glucocorticoid treat- EFFECTS OF PHYSIOLOGICAL


ment was stopped), the rabbits were euthanized. CONCENTRATIONS OF
In four of the six rabbits in the experimen- GLUCOCORTICOIDS,
ILLUSTRATED BY STUDIES
tal group, dexamethasone had allowed many WITH ADRENOCORTICOTROPIC
more bacilli to persist in the lesions and then HORMONE (ACTH) IN INBRED
multiply after the glucocorticoid was stopped. RABBITS WITH SMALL ADRENALS
The host’s reaction to these numerous bacilli Lurie’s susceptible FC rabbits had relatively small
made these lesions much larger than the lesions adrenals, which apparently produced suboptimal
in the controls. amounts of glucocorticoids (6).To stimulate the
Real-time PCR identified several genes that adrenals of these FC rabbits, sheep ACTH in
seem to be involved in mycobacterial survival in gelatin (0.5 mg/kg intramuscularly) was given
vivo in rabbit tuberculous lesions. daily (Table 2).The rabbits were then infected
16. CORTISONE AND ANDRENOCORTICOTROPIC HORMONE 䡵 281

FIGURE 7 Lungs of control rabbits of the inbred resistant strains (IIIFIII 1-16 and III IIIC
1-5) on the left, and lungs of cortisone-treated rabbits of the same strains (IIIFIII 1-6 and III IIIC
1-4) on the right. These rabbits had inhaled aerosols of virulent human-type tubercle bacilli
(H37Rv) 13 weeks previously.The cortisone treatment was started 9 days before infection and
continued for 8 weeks during infection. Four and one-half weeks after the treatment was
stopped, the rabbits were euthanized.
The cortisone-treated rabbit IIIFIII 1-6 (right) had two large thick-walled cavities in the right
lung, one of which is shown in the photograph.The cortisone-treated rabbit III IIIC 1-4 (right)
had numerous active tuberculous lesions in both lungs.The two control rabbits, however, had only
a few minute regressing tubercles.
Although this experiment was similar to the one portrayed in Fig. 6, the inbred IIIFIII and
III IIIC rabbits, which were genetically much more resistant than the inbred Ca rabbits in
Fig. 6, developed fewer primary tubercles.These two factors, especially the native resistance of
the host, evidently made the adverse effects of the glucocorticoids less severe.
Reproduced with permission from reference 2.

by aerosol with virulent human-type bacilli rather than increased them. The diameters of
(H37Rv), and the ACTH injections continued these tubercles were not recorded, but apparently
for 5 weeks, at which time the rabbits were they were similar to those of the controls,
euthanized.The number of primary pulmonary because the ACTH produced no pharmaco-
tubercles found in the lungs of the ACTH- logical glucocorticoid effects (Table 2) (6).
treated rabbits was about half that found in the This ACTH regimen had no appreciable
untreated controls (Table 2). In other words, effect on another inbred susceptible strain of rab-
the ACTH reduced the number of tubercles bits (strain C), which innately had normal-size
282 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

TABLE 2 Effects of adrenocorticotropic hormone (ACTH) on tuberculosis produced by the inhalation of


human-type tubercle bacilli (H37Rv)a
Sheep ACTH in Porcine ACTH
FC rabbits of low Sheep ACTH in C in FC rabbits of
resistance (with small rabbits of low resistance low resistance (with
adrenals) (with normal adrenals) small adrenals)
Treated Control Treated Control Treated Control
Number of inhaled 110 ⫾ 11* 62 ⫾ 14 50 ⫾ 7 47 ⫾ 4 70 ⫾ 19 82 ⫾ 14
tubercle bacilli
required to
produce one
primary
pulmonary
tubercle
(the ratio)b
Average diameter 2.9 ⫾ 0.2** 3.7 ⫾ 0.2
of tubercles (mm)
Adrenal weight 147 ⫾ 11 120 ⫾ 19 259 ⫾ 13 245 ⫾ 14 160 ⫾ 23 133 ⫾ 12
(mg/kg body
weight)
Liver weight 23 ⫾ 1 21 ⫾ 1 33 ⫾ 2† 25 ⫾ 1 25 ⫾ 2‡ 19 ⫾ 1
(g/kg body
weight)
Adapted from reference 6.Values significantly different from control: *P ⫽ 0.01; **P ⫽ 0.007; †P ⫽ 0.004; ‡P ⫽ 0.008.
a
b
The ratio is explained in Table 1.The larger the number of inhaled bacilli required to produce one tubercle, the fewer the
number of tubercles produced in the lung by a given inhaled dose.

adrenals (Table 2). It seems, therefore, that to destroy or inhibit ingested tubercle bacilli,
additional glucocorticoids will only be benefi- whereas a pharmacological concentration only
cial in hosts that are deficient in glucocorticoid decreases cytokine production.This dose-effect
production. principle has been established for systemic hor-
To determine the physiological amount of mones (19, 20) as well as for cytokines (21)
glucocorticoids is difficult. A porcine ACTH (which are local hormones). See the addendum
in gelatin that was given to another set of FC below.
rabbits at the same dosage as the sheep ACTH
was not beneficial (Table 2).This porcine ACTH
EFFECTS OF
preparation caused pharmacological glucocor- DEHYDROEPIANDROSTERONE
ticoid effects, such as an increase in liver weight (DHEA) AND ITS DERIVATIVE
from glycogen and fat, and a decrease in the size 3,17-ANDROSTENEDIOL (AED)
of the primary pulmonary tubercles—an anti- This chapter would not be complete without
inflammatory effect (Table 2). mention of DHEA and AED. These cortico-
In brief, a slight increase in glucocorticoids in steroids counteract the adverse effects of gluco-
a corticosteroid-deficient host increased its re- corticoids on tuberculosis (22). Unfortunately,
sistance to tuberculosis, but additional gluco- DHEA and AED have only been evaluated in
corticoids had no effect, or even an adverse the mouse model (22). Briefly, these antigluco-
effect, on hosts with normal adrenal function.A corticoids reduced the number of mycobacte-
physiological concentration of glucocorticoids rial titers in the lungs of BALB/c mice, reduced
probably increases rather than decreases the local the extent of the disease, and shifted the
cytokine production that activates macrophages Th1/Th2 lymphocyte ratio toward Th1 (22).
16. CORTISONE AND ANDRENOCORTICOTROPIC HORMONE 䡵 283

ADDENDUM cortisone and ACTH on the pathogenesis of tuber-


The physiological and pharmacological effects of culosis. Ann. N.Y.Acad. Sci. 56:779–792.
7. Lurie, M. B., P. Zappasodi,A. M. Dannenberg,
glucocorticoids are well understood (reviewed in
Jr., and I. B. Swartz. 1951. Constitutional factors
references 19 and 20). Physiologically, they keep in resistance to infection: the effect of cortisone on
normal inflammatory and immune responses the pathogenesis of tuberculosis. Science 113:234–
from becoming excessive. In acute septicemic 237.
infections, the host produces such high levels of 8. Lurie, M. B., and P. Zappasodi. 1955. On the
mode of action of cortisone on the pathogenesis of
endogenous glucocorticoids that shock and death
tuberculosis and its implications for the nature of
may sometimes be prevented.The high physio- genetic resistance to the disease, p. 246–258. In
logical levels of glucocorticoids evidently reduce Ciba Foundation Symposium on Experimental Tuber-
the production of proinflammatory and fever- culosis. Churchill, London, United Kingdom.
producing cytokines (e.g., interleukin-1, tumor 9. Lurie, M. B. 1964. Resistance to Tuberculosis: Exper-
imental Studies in Native and Acquired Defensive
necrosis factor , and interleukin-6) that enter the
Mechanisms, p. 244–264. Harvard University Press,
circulation (18, 19). Pharmacologically, gluco- Cambridge, Mass.
corticoids reduce both local and systemic inflam- 10. Kesavan, A. K., S. Mendez, C. L. Hatem,
matory responses (again by reducing cytokine J. Lopez-Molina, M. Brooks, R. Fujiwara,
production) and are helpful in diseases such as K. Aird, M. L. M. Pitt, A. M. Dannenberg, Jr.,
and Y. C. Manabe. 2005. Effects of dexamethasone
asthma, arthritis, and dermatoses whenever other
and transient malnutrition on rabbits infected with
therapies are ineffectual. aerosolized Mycobacterium tuberculosis CDC1551.
The adrenals of different animal species Infect. Immun. 73:7056–7060.
secrete different proportions of glucocorticoids: 11. McCue, R. E., A. M. Dannenberg, Jr.,
e.g., rabbits and rats have low ratios of 17- S. Higuchi, and M. Sugimoto. 1978.The effect
of cortisone on the accumulation, activation, and
hydroxycorticosterone (hydrocortisone) to cor- necrosis of macrophages in tuberculous lesions.
ticosterone, whereas monkeys and humans have Inflammation 3:159–176.
high ratios (23). Five weeks after infection with 12. Yarborough, D. J., O. T. Meyer, A. M. Dan-
human-type tubercle bacilli, Lurie’s resistant nenberg, Jr., and B. Pearson. 1967. Histo-
strain III rabbits apparently produced higher chemistry of macrophage hydrolases. III. Studies
on -galactosidase, -glucuronidase and aminopep-
levels of both hormones than did his suscepti- tidase with indolyl and naphthyl substrates. J. Retic-
ble strain FC rabbits (23). uloendothel. Soc. 4:390–408.
13. Dannenberg, A. M., Jr., O. T. Meyer, J. R.
Esterly, and T. Kambara. 1968.The local nature
REFERENCES of immunity in tuberculosis, illustrated histochem-
1. Seyle, H. 1946.The general adaptation syndrome ically in dermal BCG lesions. J. Immunol. 100:931–
and the diseases of adaptation. J. Clin. Endocrinol. 941.
6:117–230. 14. Dannenberg, A. M., Jr. 1968. Cellular hyper-
2. Lurie, M. B., P. Zappasodi,A. M. Dannenberg, sensitivity and cellular immunity in the patho-
Jr., and E. Cardona-Lynch. 1953. Constitu- genesis of tuberculosis: specificity, systemic and local
tional factors in resistance to infection: the effect of nature, and associated macrophage enzymes. Bacte-
cortisone on the pathogenesis of tuberculosis, p. 84– riol. Rev. 32:85–102.
99. In G. Shwartzman (ed.), The Effect of ACTH and 15. Johnson, J. R., and W. N. Davey. 1954. Corti-
Cortisone upon Infection and Resistance. Columbia sone, corticotropin, and antimicrobial therapy in
University Press, New York, N.Y. tuberculosis in animals and man: a review. Am. Rev.
3. Claman, H. N. 1983. Glucocorticosteroids I: anti- Tuberc. 70:623–636.
inflammatory mechanisms. Hosp. Pract. 18:123–126, 16. Spink, W. W. 1957. ACTH and adrenocortico-
131–134. steroids as therapeutic adjuncts in infectious diseases.
4. Claman, H. N. 1983. Glucocorticosteroids II: the N. Engl. J. Med. 257:979–983 (continued).
clinical responses. Hosp. Pract. 18:143–146, 149– 17. Spink, W. W. 1957. ACTH and adrenocortico-
151. steroids as therapeutic adjuncts in infectious diseases.
5. Dannenberg, A. M., Jr. 1979. The anti- N. Engl. J. Med. 257:1031–1035 (concluded).
inflammatory effects of glucocorticosteroids: a brief 18. Silverstein, R., and D. C. Johnson. 2003.
review of the literature. Inflammation 3:329–343. Endogenous vs exogenous glucocorticoid responses
6. Lurie, M. B., P. Zappasodi,A. M. Dannenberg, to experimental bacterial sepsis. J. Leukoc. Biol.
Jr., and E. Cardone-Lynch. 1953.The effect of 73:417–427.
284 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

19. Webster, J. I., and E. M. Sternberg. 2004. Role 22. Hernandez-Pando, R., M. de la Luz Streber,
of the hypothalamic-pituitary-adrenal axis, gluco- H. Orozco, K. Arriaga, L. Pavon, S. A.
corticoids and glucocorticoid receptors in the toxic Al-Nakhli, and G.A.W. Rook. 1998.The effects
sequelae of exposure to bacterial and viral products. of androstenediol and dehydroepiandrosterone on
J. Endocrinol. 181:207–221. the course and cytokine profile of tuberculosis in
BALB/c mice. Immunology 95:234–241.
20. Webster, J. I., L.Tonelli, and E. M. Sternberg.
23. Kass, E. H., O. Hechter,T.W. Mou, and M. B.
2002. Neuroendocrine regulation of immunity.
Lurie. 1955. Effects of adrenal steroids on resistance
Annu. Rev. Immunol. 20:125–163.
to infection. Differences in the relative amounts of
21. Nathan, C. 2002. Points of control in inflamma- corticosterone and hydrocortisone secreted and their
tion. Nature 420:846–852. biologic effects. Arch. Intern. Med. 96:397–402.
17
EFFECTS OF ESTROGEN, CHORIONIC
GONADOTROPIN, AND THYROID
HORMONES ON TUBERCULOSIS

Effects of estrogen and chorionic gonadotropin [285]


Effects of thyroid hormones [286]

Abstract. Since estrogen increased the hyaluronic acid and water content of the skin, it
decreased the spread of intradermally injected virulent bovine-type tubercle bacilli in rab-
bits. Chorionic gonadotropin had the reverse effect.These two sex hormones had no appre-
ciable effect on the innate or acquired ability of the host to control the progression of tuber-
culosis, because neither hormone appreciably changed the number of primary pulmonary
tubercles generated by aerosols of virulent human-type tubercle bacilli. However, estro-
gen markedly suppressed the development of amyloid in the spleens of rabbits dying of
the more chronic form of tuberculosis caused by bovine-type bacilli.
In rabbits, triiodothyronine or thyroxine increased host resistance in that they decreased
the number of grossly visible primary pulmonary tubercles produced by the inhalation of
virulent human-type tubercle bacilli, whereas thyroidectomy or propylthiouracil treatment
increased the number of such primary tubercles.Thyroid hormones were most beneficial
in inbred rabbits of intermediate resistance.The resistance of the most susceptible inbred
C rabbits and that of the most resistant III(r) rabbits were not appreciably increased by thy-
roid hormones.

EFFECTS OF ESTROGEN AND extracellular matrix (reviewed in references 3


CHORIONIC GONADOTROPIN and 4). Estrogen also enlarged the nipples,
The effects of estrogen and chorionic go- increased the size and congestion of the vulva,
nadotropin on tuberculosis in rabbits were stud- and increased the weight of the uterus and
ied by Lurie (1–4) because of the inferred effects vagina (observed at necropsy) (1).
of these hormones on the disease in humans. Soon after the effects of estrogen became
Latent tuberculosis tends to reactivate during apparent, the rabbits were injected intrader-
adolescence (especially in females) and also dur- mally with virulent bovine-type tubercle bacilli
ing the first 4 months of pregnancy, when the (Ravenel).When compared with control rabbits
levels of chorionic gonadotropin in the blood receiving only the diluent, estrogen markedly
are high (4). During the last 5 months of preg- reduced the size of the resulting dermal tuber-
nancy, blood estrogen levels are high, and reac- culous lesion and usually reduced the spread of
tivation of tuberculosis rarely occurs. disease to other sites in the body (1–3). Specif-
Lurie injected -estradiol dipropionate (0.5 ically, the lungs and kidneys often showed less
mg in sesame oil) subcutaneously into inbred extensive tuberculosis, and the other organs
rabbits (usually his resistant strain A and his sus- often showed fewer metastatic lesions.
ceptible strain C) once a week throughout the Estrogen did not affect the power of the host
experiment (1, 4). By the second week, this to inhibit the growth of tubercle bacilli (3, 4),
estrogen had increased the turgidity of the skin, nor did it affect the basic immunity that the host
apparently by enhancing its hyaluronic acid developed (3, 4) (Table 1). After the inhalation
content and the amount of water bound to the of virulent human-type bacilli (H37Rv), the

285
286 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

TABLE 1 Effects of estrogen on tuberculosis produced by the inhalation of human-type tubercle bacilli
(H37Rv) in female susceptible CaC rabbitsa
Estrogen-treated Controls P value
Weight of uterus 29.9 ⫾ 1.4 g 18.0 ⫾ 0.8 g 0.001
Weight of ovaries 380 ⫾ 24 mg/kg 500 ⫾ 57 mg/kg 0.038
4-h spread of India ink 687 ⫾ 44 mm2 846 ⫾ 80 mm2 0.045
4-h spread of Evans Blue dye 728 ⫾ 44 mm2 1,290 ⫾ 251 mm2 0.012
Maximum tuberculin sensitivity 78 ⫾ 13 mm3 264 ⫾ 35 mm3 0.001
Number of inhaled tubercle bacilli
required to produce one primary
pulmonary tubercle (“the ratio”)b 27 ⫾ 3.6 28 ⫾ 2.2 No difference
Average diameter of tubercles 2.6 ⫾ 1.4 mm 3.8 ⫾ 0.1 mm 0.01
a
Estrogen was given weekly, starting 2 weeks before the airborne infection, and was continued throughout the experiment.
Note that estrogen increased the weight of the uterus, decreased the weight of the ovaries, and reduced the spread in the skin of
both India ink particles and a solution of Evans Blue dye.Therefore, the hormone was active in the dosage given.The reduced
spread of these markers was apparently due to increased dermal turgidity (produced by increased hyaluronic acid and the water
it binds).Adapted from reference 4.
b
“The ratio,” a measure of both the native and acquired resistance of the host, was calculated by the Lurie method (see chap-
ter 11).The absence of a significant change in the ratio indicates that estrogen had no appreciable effect on the resistance of these
rabbits to tuberculosis.

same number of primary pulmonary tubercles larger than those in control rabbits, and the
was produced in the estrogen-treated group as spread of the tubercle bacilli from an intrader-
in the control group (Table 1) (4). mal site of injection was increased (1).
Estrogen’s main effect on tuberculosis seemed Reference 5 reviews the older literature on
to be a reduction in the spread of the bacilli the effects of these and several other hormones
through the connective tissue and a reduction on tuberculosis.
in the inflammatory response of the skin to
irritants, such as heat-killed tubercle bacilli and EFFECTS OF THYROID HORMONES
pertussis endotoxin. In tuberculous hosts, estro- The effects of thyroid hormones on tuberculo-
gen reduced the size of the tuberculin reaction sis in rabbits are presented in references 6, 7, and
(Table 1) (2, 4). 8. The thyroid hormones L-triiodothyronine
Unexpectedly, the estrogen treatment (40 to 50 g) or L-thyroxine (100 g) were
almost eliminated the amount of amy- injected intramuscularly daily (or on alternate
loid in the spleens of rabbits dying of days) into several strains of inbred rabbits
chronic tuberculosis caused by the bovine- throughout the experiment (5). After about 4
type bacilli (3, 4). weeks, the rabbits were exposed to aerosols of
The effects of chorionic gonadotropin were virulent human-type tubercle bacilli (H37Rv).
the reverse of those produced by estrogen. Lurie The rabbits were euthanized 5 weeks later, and
injected chorionic gonadotropin (0.02 to 0.2 the grossly visible primary pulmonary tubercles
mg) intravenously (usually into his resistant were counted.
strain A rabbits) every 10th day throughout the In inbred rabbits of intermediate resistance, a
experiment.This pituitary hormone causes the significant reduction in the number of grossly
ovaries to form corpora lutea, which produce visible tubercles was produced by treatment with
progesterone, a hormone that depolymerizes thyroid hormones (Table 2) (7, 8). These hor-
hyaluronic acid and thereby reduces the turgid- mones had much less effect on the resistance of
ity of the skin (3, 4). Lurie’s susceptible rabbits (strain C) (7) and on
In the chorionic gonadotropin-treated rab- the resistance of his resistant rabbits (strain III) (8).
bits, lesions caused by the intradermal injection The thyroid hormones apparently increased
of virulent bovine-type tubercle bacilli were both the native and the acquired resistance of the
17. ESTROGEN, CHORIONIC GONADOTROPIN,THYROID HORMONES 䡵 287

TABLE 2 Effects of L-triiodothyronine, dinitrophenol (DNP), and L-thyroxine on tuberculosis produced by


the inhalation of human-type tubercle bacilli (H37Rv) in inbred rabbitsa
L-Triiodothyronine- DNP-treated Thyroxine-treated
treated AD rabbits AD rabbits (of IIIA rabbits
(of intermediate intermediate (of intermediate
resistance) resistance) resistance)
Treated Control Treated Control Treated Control
Change in basal 41% 11% 28% 11% 65% 1%
metabolic
rate during
infection
Maximal 206 ⫾ 50 mm3 809 ⫾ 62 mm3
tuberculin P ⬍ 0.001
sensitivity
Number of 273 ⫾ 55 155 ⫾ 22 89 ⫾ 27 155 ⫾ 22 659 ⫾ 99 154 ⫾ 27
inhaled P ⫽ 0.03 P ⫽ 0.038 P ⬍ 0.001
tubercle
bacilli
required to
produce
one primary
pulmonary
tubercle
(the ratio)b
Average 2.5 ⫾ 0.2 3.3 ⫾ 0.3 3.2 ⫾ 0.2 3.3 ⫾ 0.3 2.7 ⫾ 0.1 3.7 ⫾ 0.2
diameter of P ⫽ 0.02 P ⬍ 0.001
tubercles
(mm)
Number of Few Moderate Many Moderate Few Moderate
bacilli
Type of Interstitial More More Interstitial Interstitial More
inflammation pneumonic pneumonic pneumonic
Amount of None to Moderate Extensive Moderate None to Moderate to
caseation in slight slight extensive
the tubercles
a
Note that both thyroid hormones and DNP increased the basal metabolism of these rabbits. However, only the hormones
increased their resistance to tuberculosis. In fact, DNP decreased their resistance. Data from Table 4 in reference 8 and Table 7 in
reference 7.
b
The higher the ratio, the fewer the number of grossly visible tubercles (see footnote b in Table 1).

responding rabbits.The hormone-treated rabbits reduced in size in the hormone-treated group


had smaller tubercles that showed greater inter- (Table 2) (7).
stitial inflammation, fewer bacilli, and less In another experiment, with triiodothyro-
caseation than tubercles in the control rabbits nine treatment beginning 3 weeks after the
(Table 2) (7). The thyroid hormones decreased inhalation of virulent human-type tubercle
the number of tubercle bacilli present in the bacilli, the number of grossly visible primary
lungs (Fig. 1) but increased the number of tuber- tubercles found at necropsy 7 weeks later (10
cle bacilli reaching the hilar nodes.These find- weeks after infection) was decreased to 36%
ings were similar to those observed in the tuber- that of the control rabbits (Table 3) (7). This
culosis of Lurie’s resistant rabbits (9) (see chapter result suggests that thyroid hormones may have
14). The tuberculin skin test reactions were a therapeutic effect on tuberculosis.
288 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

FIGURE 1 Effect of thyroxine on the number of human-type tubercle bacilli


(H37Rv) cultured from the lungs of inbred III IIIC rabbits (which are of rather high
resistance) at various times after aerosol infection.The points represent the mean
number of viable bacilli at weekly intervals divided by the number on the first day.
Reproduced from reference 7.
Note that thyroxine treatment of these rabbits inhibited bacillary growth, espe-
cially during the first 7 days. Evidently, the pulmonary alveolar macrophages of these
hyperthyroid rabbits had an increased ability to destroy or inhibit the inhaled
bacilli.

TABLE 3 Effect on tuberculosis of triiodothyronine treatment begun 3 weeks after the inhalation of human-
type tubercle bacilli (H37Rv) and continued for 7 subsequent weeks in III IIIC rabbits of rather high
resistancea
Triiodothyronine-treated Controls P value
Number of inhaled tubercle bacilli required to
produce one primary pulmonary tubercle
(the ratio)b 1,004 ⫾ 371 359 ⫾ 64 0.05
Average diameter of tubercles (mm) 2.7 ⫾ 0.2 3.3 ⫾ 0.3 0.05
a
Note that treatment with triiodothyronine that began after pulmonary tubercles were well established at 3 weeks still reduced
the number of grossly visible tubercles found 7 weeks later to 36% of those found in controls (calculated from ratios).These find-
ings suggest that thyroid hormones increased the acquired (adaptive) resistance of the host. Data from Table 5 of reference 7.
b
The higher the ratio, the fewer the number of grossly visible tubercles (see footnote b of Table 1).
TABLE 4 Effects of thyroidectomy (Thyx) and propylthiouracil treatment (PtuRx) on tuberculosis produced by the inhalation of human-type tubercle
bacilli (H37Rv)a
Thyroidectomy
Propylthiouracil-treated
IIIA rabbits III rabbits CaC rabbits CaC rabbits

17.
(of intermediate (of high (of low (of low
resistance) resistance) resistance) resistance)

ESTROGEN, CHORIONIC GONADOTROPIN,THYROID HORMONES


Thyx Control Thyx Control Thyx Control PtuRx Control
Change in basal ⫺20% 0% ⫺20% 0% ⫺22% 0% ⫺19% 0%
metabolic P ⫽ 0.001
rate before
infection
Intradermal 1,551 ⫾ 3,050 ⫾ 2,152 ⫾ 3,359 ⫾
turpentine- 157 mm3 312 mm3 381 mm3 267 mm3
induced P ⫽ 0.001 P ⫽ 0.009
inflammation
Maximal 54 ⫾ 9 mm3 306 ⫾ 103 mm3 86 ⫾ 23 mm3 303 ⫾ 48 mm3 167 ⫾ 23 mm3 303 ⫾ 48 mm3
tuberculin P ⫽ 0.01 P ⫽ 0.001 P ⫽ 0.01
sensitivity
Weight of spleen 305 ⫾ 39 mg 461 ⫾ 35 mg 185 ⫾ 10 mg 300 ⫾ 20 mg 119 ⫾ 11 mg 339 ⫾ 24 mg 321 ⫾ 11 mg 339 ⫾ 24 mg
(mg/kg) P ⫽ 0.004 P ⫽ 0.001 P ⫽ 0.001
Weight of 12 ⫾ 0.8 mg 9 ⫾ 0.2 mg
pituitary P ⫽ 0.001
(mg/kg)
Weight of 184 ⫾ 23 mg 64 ⫾ 3 mg
thyroid P ⫽ 0.001
(mg/kg)
No. of inhaled 16 ⫾ 3.4 26 ⫾ 2.2 1,500 ⫾ 260 5,100 ⫾ 1,200 9⫾1 18 ⫾ 2 9⫾1 18 ⫾ 2
tubercle P ⫽ 0.01 P ⫽ 0.004 P ⫽ 0.001 P ⫽ 0.001
bacilli
required to
produce one


primary
pulmonary
tubercle

289
(the ratio)b (continued next page)
290

PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS
TABLE 4 Effects of thyroidectomy (Thyx) and propylthiouracil treatment (PtuRx) on tuberculosis produced by the inhalation of human-type tubercle
bacilli (H37Rv)a (continued)
Thyroidectomy
Propylthiouracil-treated
IIIA rabbits III rabbits CaC rabbits CaC rabbits
(of intermediate (of high (of low (of low
resistance) resistance) resistance) resistance)
Thyx Control Thyx Control Thyx Control PtuRx Control
Average 3.4 ⫾ 0.1 mm 3.0 ⫾ 0.3 mm 3.1 ⫾ 0.3 2.5 ⫾ 0.3 2.7 ⫾ 0.2 3.4 ⫾ 0.1 2.9 ⫾ 0.1 3.4 ⫾ 0.1
diameter of P ⫽ 0.11 P ⫽ 0.07 P ⫽ 0.001 P ⫽ 0.001
tubercles
Percent of 29 ⫾ 6 7⫾2 61.1 ⫾ 1.8 11.3 ⫾ 2.7 51.2 ⫾ 8.2 11.3 ⫾ 2.7
tubercles with P ⫽ 0.002 P ⫽ 0.001 P ⫽ 0.001
white caseous
centers
Percent of 24 ⫾ 7.8 6 ⫾ 2.4
tubercles P ⫽ 0.02
liquefied, with
or without a
cavity
a
Note that thyroidectomy had a glucocorticoid-like effect on turpentine-induced inflammation, tuberculin sensitivity, and the lymphoid tissues of the spleen. Note also that in resis-
tant strain III rabbits, a large percentage of the pulmonary lesions liquefied and/or formed cavities in the thyroidectomized group, probably because the lesions contained so many more
bacilli.This experiment lasted 15 weeks (see reference 6 for details).Adapted from Tables 1, 2, and 3 of reference 8.
b
The higher the ratio, the fewer the number of grossly visible tubercles (also see Table 1).
17. ESTROGEN, CHORIONIC GONADOTROPIN,THYROID HORMONES 䡵 291

Decreasing thyroid function by thyroidec- gonadotropin on the course of tuberculosis in


tomy or the administration of propylthiouracil highly inbred rabbits. Am. Rev.Tuberc. 59:168–185.
2. Lurie, M. B., T. N. Harris, S. Abramson, and
(averaging 100 mg subcutaneously each day M. J. Allison. 1949. Constitutional factors in re-
throughout the experiment) markedly decreased sistance to infection. II. The effect of estrogen on
host resistance to tuberculosis produced by the tuberculin skin sensitivity and on the allergy of the
inhalation of H37Rv (Table 4) (8).The effects internal tissues. Am. Rev.Tuberc. 59:186–197.
of the lowered metabolic rate resembled those 3. Lurie, M. B., S. Abramson, A. G. Heppleston,
and M. J. Allison. 1949. Constitutional factors in
produced by pharmacological doses of gluco- resistance to infection. III. On the mode of action
corticoids, i.e., increased numbers of bacilli, of estrogen and gonadotropin on the progress of
decreased perifocal inflammation, decreased tuberculosis. Am. Rev.Tuberc. 59:198–218.
drainage of bacilli to the hilar lymph nodes, 4. Lurie, M. B. 1964. Resistance to Tuberculosis: Exper-
decreased size of dermal tuberculin reactions, imental Studies in Native and Acquired Defensive
Mechanisms, p. 181–191. Harvard University Press,
and decreased weight of the spleen (except in Cambridge, Mass.
the propylthiouracil-treated rabbits) (Table 4) 5. Lurie, M. B. 1955. On the role of hormones in
(8).These hypothyroid effects were also similar experimental tuberculosis. Adv.Tuberc. Res. 6:18–48.
to those found innately in rabbits with low re- 6. Lurie, M. B. 1964. Resistance to Tuberculosis: Exper-
sistance to tuberculosis (8). imental Studies in Native and Acquired Defensive
Mechanisms, p. 265–301. Harvard University Press,
2,4-Dinitrophenol (roughly 10 mg/kg in Cambridge, Mass.
peanut oil subcutaneously each day throughout 7. Lurie, M. B., P. Zappasodi, R. S. Levy, and
the experiment) increased the host’s metabolic R. G. Blaker. 1959. On the role of the thyroid in
rate but reduced (rather than increased) host native resistance to tuberculosis. I. The effect of
resistance (Table 2) (8).Therefore, the beneficial hyperthyroidism. Am. Rev.Tuberc. 79:152–179.
8. Lurie, M. B., P. Zappasodi, R. G. Blaker, and
effects of thyroid hormones were due not R. S. Levy. 1959. On the role of the thyroid in
merely to increased oxygen consumption by native resistance to tuberculosis. II. The effect of
the host, but also to other metabolic effects. hypothyroidism: the mode of action of thyroid hor-
mones. Am. Rev.Tuberc. 79:180–203.
9. Lurie, M. B., P. Zappasodi, and C. Tickner.
REFERENCES 1955. On the nature of genetic resistance to tuber-
1. Lurie, M. B., S. Abramson, and M. J. Allison. culosis in the light of the host-parasite relationships
1949. Constitutional factors in resistance to infec- in natively resistant and susceptible rabbits. Am.
tion. I. The effect of estrogen and chorionic Rev.Tuberc. 72:297–329.
18
EFFECTS OF WHOLE-BODY
X-IRRADIATION ON
TUBERCULOSIS

Effects of X-irradiation on dermal BCG lesions [293]


Effects of X-irradiation on pulmonary alveolar macrophage
populations [293]
Effects of X-irradiation on the number of AM from granulomatous
lungs [294]
Recovery from the effects of X-irradiation [296]
Effects of X-irradiation on tuberculosis produced by the inhalation of
virulent human-type bacilli (H37Rv) [296]
Conclusions concerning the effects of 400 rads of whole-body X-irradiation
on tuberculosis in rabbits [297]
Comment on the public health consequences of medical irradiation [298]

Abstract. Commercial rabbits were irradiated with 400 rads of whole-body X-irradia-
tion—a sublethal dose. At 2 or 10 days thereafter, they were injected intradermally with
BCG. Between 2 and 4 weeks after irradiation, the BCG lesions and 48-h tuberculin reac-
tions in the irradiated group were smaller than those of the nonirradiated controls.The
BCG lesions in the irradiated group also contained more bacilli.
This dose of whole-body X-irradiation evidently decreased the supply of macrophages
and lymphocytes from “cell factories” in the bone marrow and lymphoid tissues, so that
fewer cells were available to infiltrate the BCG lesions.These cell factories had apparently
recovered 4 to 5 weeks after irradiation, because BCG lesions starting at this time were
the same size as those in the nonirradiated controls and contained the same number of
bacilli.
Pulmonary alveolar macrophages (AM) recovered by bronchoalveolar lavage (BAL) from
irradiated rabbits contained higher levels of hydrolytic enzymes than did AM from non-
irradiated controls.The AM from the irradiated group were apparently an older (more acti-
vated) cell population, because they had ingested inhaled particles for a longer period of
time. The irradiation evidently had reduced the young macrophage population that
replenishes the AM population.
When heat-killed tubercle bacilli in oil were injected intravenously 1 day after irradi-
ation, the BAL specimens obtained 9 and 10 days later (from the resulting granulomatous
lungs) contained about half as many macrophages as did those from nonirradiated con-
trols.This finding indicated that the irradiation reduced the supply of macrophages from
the bone marrow.When the heat-killed tubercle bacilli were injected 4 weeks after irra-
diation, BAL specimens from the granulomatous lungs of the irradiated and the nonirra-
diated animals contained similar numbers of macrophages, which indicated that the sup-
ply of these macrophages had recovered.
Rabbits were infected by aerosol with virulent human-type tubercle bacilli (H37Rv)
at 12 or 30 days after irradiation. In each case, 5 weeks after infection, the number of pri-
mary pulmonary tubercles in their lungs was the same in both the irradiated and the non-
irradiated groups. Also, the number of viable bacilli in these tubercles was the same.
Therefore, this sublethal dose of irradiation had no appreciable effect on the development
and progress of primary pulmonary tubercles in rabbits.
In brief,X-irradiation reduces the bone marrow’s capacity to provide defense cells to pro-
tect the host against infection.When the host is challenged by inhaled virulent human-type
tubercle bacilli,an adequate supply of defense cells is available.However,with acute infections
(requiring many more defense cells), the irradiated host would have an inadequate supply.

292
18. WHOLE-BODY X-IRRADIATION 䡵 293

After a single dose of whole-body X-irradi- circulating leukocytes per mm3 of blood was
ation in the 50% lethal dose (LD50) range (i.e., markedly reduced, mainly due to decreases in
600 to 700 rads for rabbits and probably lymphocytes and granulocytes (1, 3).The BCG
humans), the recipient may live relatively well for lesions and tuberculin reactions were smaller in
7 to 10 days and then develop a progressive the irradiated group than in the controls (Fig.
systemic infection that ends in death (reviewed 2), because evidently fewer cells were available
in reference 1). In most cases, this infection is to infiltrate them (4). In the irradiated group, the
caused by microorganisms that normally inhabit total number of bacilli present in the entire
the gastrointestinal or respiratory tract. BCG lesion, as well as the number within each
In unirradiated hosts, these microorganisms parasitized macrophage therein, was increased.
frequently gain access to host tissues, but they are This observation suggests that the ability of the
rapidly destroyed by mononuclear phagocytes macrophages to destroy the bacillus was impaired
(primarily those in the liver and spleen) and (1).The bone marrow and lymphoid tissues had
also by granulocytes that are circulating and apparently recovered in 4 to 5 weeks, because
sequestered (mainly in the lung). the blood counts had returned to normal (Fig.
In hosts irradiated in the LD50 range, serum 1), and the BCG lesions and tuberculin reac-
and leukocyte microbicidins decline after 7 to 10 tions, begun at this late time, showed no appre-
days, and the bone marrow’s supply of granulo- ciable difference in size from those in the non-
cytes, macrophages, and lymphocytes becomes irradiated controls (3).
low.At this time,endogenous microorganisms are
no longer destroyed, and septicemia results.Also, EFFECTS OF X-IRRADIATION
at this time,or shortly afterward,the macrophages ON PULMONARY ALVEOLAR
of the host seem to be deficient in microbicidal MACROPHAGE POPULATIONS
and other abilities.These effects are partly due to Alveolar macrophages (AM) are the host’s ini-
the direct effects of irradiation and partly due to tial defense against inhaled microorganisms.
indirect effects, such as increased intestinal per- They are highly activated cells (5, 6) because
meability to bacterial endotoxins, partial starva- they continuously ingest and digest a variety of
tion, decreased microbicidal factors in the serum airborne particles (6). (The AM population eval-
(including antibodies), increased adrenal gluco- uated were roughly 90% macrophages with
corticoid production, and decreased granulo- some lymphocytes, a few granulocytes, and pos-
cytes, which, live or dead, may synergistically aid sibly a few dendritic cells.)
macrophage function (reviewed in reference 1). Ten to 11 days after whole-body irradiation
We studied the effects of sublethal whole- with 400 rads, the rabbits were euthanized and
body irradiation in rabbits injected with tuber- bronchoalveolar lavage (BAL) specimens were
cle bacilli, and reported our findings in a series obtained (2).Although a comparable number of
of four papers (1–4). They are summarized in pulmonary AM were found in the unirradiated
this chapter. and irradiated animals (75 and 67 million, respec-
tively) (Table 1), the pulmonary AM obtained
EFFECTS OF X-IRRADIATION from the irradiated hosts showed increased lev-
ON DERMAL BCG LESIONS els of a variety of hydrolytic enzymes, namely,
Rabbits were irradiated with 400 rads of whole- lysozyme, -glucuronidase, -galactosidase, acid
body X-irradiation and injected intradermally phosphatase, RNase, DNase, and a benzoyl-
with BCG 2 or 10 days thereafter (1, 3). This phenylalanine--napththol esterase.
amount of irradiation is about 60% of the LD50. The reason for this effect on enzyme content
Therefore, all of the rabbits in these experi- is not known. It was probably due to the presence
ments survived. of older, more mature macrophages in the AM
The effect of such irradiation was clearly population. Irradiation reduces the number of
demonstrated by the peripheral blood counts young unstimulated macrophages from the bone
(Fig. 1). During the first 2 weeks, the number of marrow that migrate via the bloodstream into the
294 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

FIGURE 1 Blood counts (cells


per mm3) in control rabbits (shaded
areas) and rabbits irradiated with 400
rads on day 0 (line graphs). The
means and their standard errors are
shown for each group.
Note the differences in the total
leukocyte count, the granulocyte
(PMN) count, and the lymphocyte
count 2 days after irradiation. PMN
were high at 2 days, probably because
the sequestered PMN (mainly in the
lungs) were released into the circula-
tion. Lymphocytes are very radiosen-
sitive, so their number markedly
decreased soon after irradiation.
During the first 2 weeks, the bone
marrow could not maintain normal
circulating leukocyte levels, but then
it gradually recovered so that the
leukocyte counts were nearly normal
at 4 to 5 weeks.
Reproduced with permission
from reference 3.

alveolar spaces (see reference 4). Therefore, the reside in the alveolar spaces, the longer they are
AM population, although reduced in size 10 to exposed to such stimuli and the higher these
11 days after whole-body irradiation, should con- enzyme levels should become.The percentage
tain a high proportion of older AM with a high of older (highly activated) AM in the population
enzyme content, and this is what we found (2). is increased if the entry of new young (poorly
Other stimuli contributing to these increased activated) AM is reduced by irradiation (see ref-
enzyme levels following irradiation would include erence 4).
lymphocytic debris, and endotoxin and bacteria
from the gut (see above). EFFECTS OF X-IRRADIATION
AM are continually ingesting dust particles, ON THE NUMBER OF AM FROM
inhaled microorganisms, and extravasated eryth- GRANULOMATOUS LUNGS
rocytes. Such stimuli activate these cells and Commercial rabbits were irradiated with 400
increase their content of hydrolytic (and meta- rads (see Table 1). On the next day, they were
bolic) enzymes (5–7).The longer macrophages injected intravenously with 0.2 mg of heat-killed
18. WHOLE-BODY X-IRRADIATION 䡵 295

FIGURE 2 Size of the BCG


lesions and 2-day tuberculin reac-
tions in control rabbits and in rabbits
that received 400 rads of whole-body
radiation 2 days before 0.1 ⫻ 106
viable BCG were injected intrader-
mally in each of several sites. The
means and their standard errors are
shown.
Note that between 2 and 3
weeks the size of the BCG lesions
and the size of the tuberculin reac-
tions were reduced in the irradiated
rabbits, apparently because the bone
marrow could not supply the lesions
with the usual number of leuko-
cytes (mainly macrophages and lym-
phocytes).
Reproduced with permission
from reference 1.
296 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

TABLE 1 Effect of whole-body irradiation on the number of rabbit pulmonary alveolar macrophages (AM)
obtained by bronchoalveolar lavage (BAL) after an intravenous injection of heat-killed tubercle bacilli in oila
Experiment Rabbit group No. of AM obtained (106)
Effect of irradiation Normal noninjected 75 ⫾ 13
Irradiated noninjected 67 ⫾ 21 (P ⫽ 0.375)
Normal injected 317 ⫾ 54
Irradiated injected 161 ⫾ 16 (P ⫽ 0.007)
Effect of bacillary injection Normal noninjected 75 ⫾ 13
Normal injected 317 ⫾ 54 (P ⬍ 0.001)
Irradiated noninjected 67 ⫾ 21
Irradiated injected 161 ⫾ 16 (P ⫽ 0.001)
a
Eight rabbits in each group received 400 rads of whole-body X-irradiation on day 0.They were injected intravenously with
lyophilized heat-killed tubercle bacilli (BCG) in mineral oil on day 1 and killed for BAL on day 10 or 11.The means and their
standard errors are listed.Adapted from reference 2.

lyophilized tubercle bacilli (BCG) in mineral oil. RECOVERY FROM THE EFFECTS
They were euthanized 10 or 11 days after radi- OF X-IRRADIATION
ation and then bronchoalveolar lavaged (usually By 4 to 7 weeks, the rabbits had apparently
three times) (2).At 10 and 11 days the lungs con- recovered from the effects of whole-body radi-
tained many small granulomatous lesions. ation (1–3). (i) Their peripheral blood counts had
Irradiation had no effect on the number of returned to normal (Fig. 1) (1, 3). (ii) Their
AM obtained by BAL from rabbits that did not BCG lesions, begun 4 to 5 weeks after the irra-
receive the intravenous bacillary injection (Table diation, were similar in size to the controls and
1) (2). However, irradiation reduced to half the histologically contained comparable numbers
larger number of macrophages obtained after the of bacilli (3). And (iii) the number of their
bacillary injection (Table 1) (2). macrophages obtained by BAL from the gran-
In the unirradiated rabbits, the intravenous ulomatous lungs was also similar to that of con-
bacillary injection increased 4.2-fold the num- trols (2).
ber of macrophages in the lavages (Table 1) (2). Recovery from the effects of X-irradiation in
In contrast, in the irradiated rabbits, the bacil- the LD50 range in other laboratory animals is
lary injection increased only 2.4-fold the num- reviewed in reference 3. The recovery times
ber of macrophages in the lavages (Table 1) (2). varied from 3 weeks to more than 8 weeks.
Therefore, the number of macrophages available
to migrate (from the bone marrow) into the
EFFECTS OF X-IRRADIATION ON
lungs in response to this bacterial stimulus was TUBERCULOSIS PRODUCED BY THE
markedly reduced by irradiation. INHALATION OF VIRULENT HUMAN-
In the unirradiated group, the intravenous TYPE BACILLI (H37Rv)
injection of heat-killed tubercle bacilli increased Three experiments were performed. In each, the
the levels of several enzymes per million AM, but rabbits were irradiated with 400 rads. In Exper-
in the irradiated group, the intravenous bacillary iment I, the rabbits were infected by aerosol
injection caused no further enzyme increase (2). with virulent human-type tubercle bacilli
Apparently, the AM enzymes in the irradiated (H37Rv) 8 days after irradiation. Six to 8 days
group were at maximal levels before the bacillary after infection, the lungs were cultured for viable
injection. tubercle bacilli. No appreciable difference was
18. WHOLE-BODY X-IRRADIATION 䡵 297

found in the number of tubercle bacilli cul- cocci (8).The delayed detrimental effect of irra-
tured from the lungs of the control and the diation on the AM population of mice was not,
irradiated rabbits (3). however,evident in rabbits inhaling (slowly grow-
In Experiment II, the rabbits were infected by ing) virulent human-type tubercle bacilli (3).
aerosol with H37Rv 12 days after irradiation (3).
Five weeks later, the number of grossly visible
CONCLUSIONS CONCERNING THE
tubercles in the lungs was the same in both the EFFECTS OF 400 RADS OF WHOLE-
control and irradiated groups, as were the size of BODY X-IRRADIATION ON
these tubercles and the number of culturable TUBERCULOSIS IN RABBITS
bacilli in them. 1. Whole-body irradiation with 400 rads
In Experiment III, the rabbits were infected decreases the production of macrophages,
by aerosol 30 days after irradiation (3). Five lymphocytes, and other cells by the bone
weeks later, the tubercle count, tubercle size, and marrow and lymphoid tissues, so that
number of culturable bacilli per tubercle were fewer defense cells are available to infiltrate
also the same in both the control and irradiated early dermal BCG lesions (4).
groups. 2. By 30 days after this whole-body irradi-
These findings suggest that resident AM are ation, the production of macrophages,
rather resistant to X-radiation (2, 3).This pop- lymphocytes, and other cells had recovered
ulation is composed of highly activated (differ- almost completely, with few, if any, resid-
entiated) macrophages (5, 6), which in rabbits ual effects.
can destroy many inhaled virulent human-type 3. X-irradiation with 400 rads on 8, 12, or 30
tubercle bacilli (3). days before aerosol infection with virulent
In our inhalation experiments, the highly human-type bacilli (H37Rv) had little or
activated AM population ingested the bacilli in no effect on the establishment and progress
both the irradiated and control rabbits. In our of the resulting pulmonary lesions. (i) The
intravenous experiments with heat-killed tuber- AM population was rather radioresistant.
cle bacilli, the AM population was bypassed, (ii) Early pulmonary lesions contained rel-
because the lesions began in the pulmonary atively few bacilli, compared with dermal
capillaries where only nonactivated macrophages BCG lesions. And (iii) after this dose of
exist. Also, in our intravenous experiments, sublethal X-irradiation, macrophages and
numerous pulmonary granulomas were pro- lymphocytes were probably still produced
duced, whereas in the inhalation experiments, in sufficient numbers by the bone marrow
relatively few pulmonary lesions were produced. and lymphoid tissues.
Therefore, the absence of any recognizable effect 4. In rabbits, one of the most sensitive indi-
of 400 rads of X-irradiation on tuberculosis cators of radiation injury was the bone
(produced in rabbits by aerosols of virulent marrow’s capacity to provide large
human-type tubercle bacilli) could be explained quantities of macrophages.These large
(i) by the high activation of the AM population quantities were required to produce the
initially ingesting the inhaled bacilli, and (ii) by multiple pulmonary granulomas that fol-
the relatively small number of pulmonary lesions lowed an intravenous injection of numer-
in such rabbits. ous heat-killed tubercle bacilli. One to 2
In mice, 2 weeks after irradiation, most of the weeks after such an intravenous injection,
radioresistant AM population had apparently died the number of macrophages obtained by
off and was not appreciably replaced by new BAL was reduced 50% in the irradiated
macrophages from the bone marrow (reviewed in rabbits (when compared with the number
reference 3). At this time, but not before, the in the nonirradiated controls).When lungs
mice had a reduced ability to control the growth containing no granulomas were lavaged,
of inhaled (rapidly growing) virulent strepto- similar numbers of macrophages were
298 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

obtained from both the control and the Effect of whole body radiation on the number of
irradiated groups. In other words, after pulmonary alveolar macrophages and their levels of
hydrolytic enzymes. J. Reticuloendothel. Soc. 7:79–90.
400 rads of whole-body radiation, the 3. Dannenberg, A. M., Jr., W. G. Roessler, O. T.
bone marrow function should be quite Meyer, S. Chandrasekhar, and T. Kambara.
adequate if the host is not challenged by 1970. Radiation, infection and macrophage func-
numerous microorganisms. tion. III. Recovery from the effects of radiation
illustrated by dermal BCG lesions: resistance of
pulmonary alveolar macrophages to radiation illus-
COMMENT ON THE PUBLIC HEALTH trated by tuberculosis produced by the airborne
CONSEQUENCES OF MEDICAL route. J. Reticuloendothel. Soc. 7:91–108.
IRRADIATION 4. Chandrasekhar, S., K. Shima, A. M. Dannen-
Cells differ in their sensitivity to irradiation. berg, Jr., T. Kambara, J. I. Fabrikant, and
Sperm production and egg production in the W. G. Roessler. 1971. Radiation, infection and
reproductive organs are affected by a few rads. macrophage function. IV.The effect of radiation on
the proliferative abilities of mononuclear phagocytes
Lymphocytes in the lymphoid tissues are injured in tuberculous lesions of rabbits. Infect. Immun.
by 50 rads. Myeloblasts (producing macrophages 3:254–259.
and granulocytes in the bone marrow) are injured 5. Dannenberg, A. M., Jr., M. S. Burstone, P. C.
by 500 rads. Walter, and J.W. Kinsley. 1963.A histochemical
Because of these differing sensitivities, a lower study of phagocytic and enzymatic functions of
rabbit mononuclear and polymorphonuclear exu-
GI series, i.e., gastrointestinal X-irradiation (usu- date cells and alveolar macrophages. I. Survey and
ally 5 rads) following barium installation, should quantitation of enzymes, and states of cellular acti-
be avoided if possible in young people hoping vation. J. Cell Biol. 17:465–486.
eventually to have children. However, the 6. Mizunoe, K., and A. M. Dannenberg, Jr. 1965.
adverse effects of such irradiation on dormant Hydrolases of rabbit macrophages. III. Effect of
BCG vaccination, tissue culture, and ingested tuber-
ova (in contrast to the adverse effects on devel- cle bacilli. Proc. Soc. Exp. Biol. Med. 120:284–290.
oping ova [9] and fetuses) have not been eval- 7. Dannenberg, A. M., Jr., P. C.Walter, and F. A.
uated in human populations. Kapral. 1963. A histochemical study of phago-
cytic and enzymatic functions of rabbit mononu-
clear and polymorphonuclear exudate cells and
REFERENCES alveolar macrophages. II. The effect of particle
1. Kambara, T., S. Chandrasekhar, A. M. Dan- ingestion on enzymes activity; two phases of in vi-
nenberg, Jr., and O.T. Meyer. 1970. Radiation, tro activation. J. Immunol. 90:448–465.
infection and macrophage function. I. Effects of 8. Shechmeister, I. L., V. P. Bond, and M. N.
whole body radiation on dermal tuberculous lesions Swift. 1952.The susceptibility of irradiated mice
in rabbits: development, histology and histochem- to infection as a function of post-irradiation time.
istry. J. Reticuloendothel. Soc. 7:53–78. J. Immunol. 68:87–95.
2. Meyer, O.T., and A. M. Dannenberg, Jr. 1970. 9. Pearson, R. 1989. Radiography in women of
Radiation, infection and macrophage function. II. childbearing ability. Br. Med. J. 299:1175–1176.
Section 6.

CYTOKINES AND VASCULAR ADHESION


MOLECULES IN TUBERCULOUS LESIONS
This page intentionally left blank
19
CYTOKINE PRODUCTION IN
PRIMARY BCG LESIONS

Cytokine mRNA in cells within tissue sections of BCG lesions [304]


Cell types containing MCP-1, IL-1, IL-8, and TNF- mRNAs [304]
Immunohistochemical studies for cytokine proteins [305]
IFN- mRNA identified by RT-PCR [305]
Comparisons among the five cytokines evaluated [306]
Studies on cytokines produced by tuberculosis in humans and
in mice [307]
Conclusions: nonspecific and antigen-specific cytokine production [309]

Abstract. A sequential histochemical study of cytokines in developing and healing rab-


bit tuberculous (BCG) lesions is described. In tissue sections, interleukin-1 (IL-1), tumor
necrosis factor alpha (TNF-), macrophage chemoattractant (activating) protein 1 (MCP-
1), and IL-8 were evaluated for cytokine mRNA by in situ hybridization techniques and
for cytokine protein by immunohistochemical techniques. In tissue homogenates, gamma
interferon (IFN-) mRNA was evaluated by reverse transcription-PCR.
In the BCG lesions, the percentage of mononuclear cells that contained the mRNAs
of these cytokines showed a biphasic pattern.At 1 to 3 days, a peak occurred, which was
apparently a nonspecific inflammatory response caused by the tubercle bacilli in the BCG
vaccine.At 5 days, the percentage of mononuclear cells containing the cytokine mRNAs
was significantly reduced, but by 9 days, the percentage had again increased, and the rab-
bits had become tuberculin positive.This second peak was apparently antigen specific.With
IFN-, the two mRNA peaks were delayed by 2 days.
Mononuclear cells containing IL-1 and IL-8 mRNAs were more numerous sur-
rounding the caseous center.These cytokines evidently recruited the polymorphonuclear
leukocytes that were common in this location. Mononuclear cells containing MCP-1
mRNA were more numerous in the outer third of the lesion where new macrophages and
lymphocytes were being recruited.
Both the nonspecific and antigen-specific cytokine responses of BCG vaccines are evi-
dently synergistic.The early nonspecific cytokine (chemokine) response causes a local accu-
mulation of antigen-presenting cells and lymphocytes, which explains, at least in part, why
tubercle bacilli are good immunological adjuvants.This adjuvant effect should be consid-
ered in developing improved vaccines for the prevention of tuberculosis, because vaccines
producing a strong early nonspecific cytokine (chemokine) response should be more
immunogenic than vaccines with similar antigens producing a weak response.

Cytokines from macrophages, lymphocytes, and Interleukin 1 (IL-1) and tumor necrosis factor
other cells (including vascular endothelial cells alpha (TNF-) are primary cytokines that up-
and local fibroblasts) play major roles in the regulate the production of other cytokines (4–6).
pathogenesis of tuberculosis.A large number of The chemokine macrophage chemoattractant
cytokines exist (listed in references 1 to 3), and (activating) protein 1 (MCP-1) (7–13) attracts
more are being discovered every year.This chap- mononuclear cells (macrophages and lympho-
ter describes sequential changes in the mRNAs cytes) into tuberculous lesions, and the chemo-
and/or proteins of five major cytokines during kine interleukin 8 (IL-8) attracts granulocytes.
the 5-week period when rabbit dermal BCG Gamma interferon (IFN-) (14–18) and TNF-
lesions developed and healed. (4, 5, 19–21) play major roles in activating

301
302 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

FIGURE 1 The percentage of mononuclear cells labeled for IL-1 (A),TNF- (B), MCP-1
(C), and IL-8 (D) mRNAs in BCG lesions at various times during their development and regres-
sion. Note the biphasic pattern of these cytokine mRNAs.The first peak is due to nonspecific
irritation by the injected BCG.The second peak is probably due to the development of antigen-
specific immunity.The means and their standard errors are shown. Reproduced with permission
from reference 11.
19. CYTOKINE PRODUCTION IN PRIMARY BCG LESIONS 䡵 303

FIGURE 1 Continued.

macrophages (22), so that the macrophages can Our findings are presented as a percentage of
inhibit and/or kill tubercle bacilli (16, 18, 24). See mononuclear cells (MN) (mainly macrophages
glossary for additional information on various with some lymphocytes and probably some
cytokines. dendritic cells) that are labeled for cytokine
Tuberculosis is a locally controlled disease, i.e., mRNA (or for cytokine protein) in the densely
the host arrests the disease locally at the sites cell-infiltrated areas of the BCG lesions.A local
where bacilli are present (see chapter 5) (25). increase in the percentage of cells labeled for
304 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

chemokines indicates that more MN will be opment of cell-mediated immunity and delayed-
attracted to the area and that the lesion will type hypersensitivity.
enlarge. Conversely, a local decrease in the per-
centage of cells labeled for chemokines indicates The Distribution of Labeled Cells
that fewer MN will be attracted to the area and At 3 days, all of the MN labeled for four cytokine
that the lesion will enlarge more slowly or even mRNAs were located in the exudate between the
regress, depending on the rate of turnover of the collagen fibers.However,from 5 days on,after dis-
MN (see chapter 10). tinct areas of necrosis were established, the MN
Cytokines are local hormones that primar- labeled for each of the four cytokine mRNAs had
ily act on nearby cells. In tuberculous lesions, their own distribution patterns. (i) Cells con-
two adjacent macrophages may even produce taining IL-1 and IL-8 mRNAs were found
different cytokines (see reference 26 and chap- near the necrotic centers and sometimes even
ter 6). In tissue sections, in situ hybridization within such centers (Fig. 2). Therefore, IL-1
and immunohistochemical methods for identi- and IL-8 were probably involved in the recruit-
fying cytokine mRNA and protein allow us to ment of the polymorphonuclear leukocytes
evaluate the distribution of cytokines to dif- (PMN) that accumulated near the caseous cen-
ferent parts of tuberculous lesions as they ter. (ii) MN containing TNF- mRNAs were
develop and regress. In other words, the cells more widely distributed than those containing
producing certain cytokines can be identified IL-1 and IL-8 mRNAs, and were most numer-
along with the nearby cells they have recruited ous among the infiltrating cells of 3-day BCG
and activated. In homogenates of tuberculous lesions. (iii) The majority of the MN containing
lesions, such cell-cell interaction would not be MCP-1 mRNA were found in the outer third of
recognized. the areas densely infiltrated with cells. These
labeled MN were closely associated with the
CYTOKINE mRNA IN CELLS WITHIN microvasculature, a propitious location for cells
TISSUE SECTIONS OF BCG LESIONS producing chemoattractants. Cells containing
Methods MCP-1 mRNA were also present more periph-
In tissue sections of rabbit BCG lesions of vari- erally in the highly vascularized areas located
ous ages, cells were identified with cytokine 35S- between the hair follicles (see chapter 8).
labeled antisense mRNAs by in situ hybridization
techniques followed by autoradiography (6, 11). CELL TYPES CONTAINING MCP-1,
Cytokine 35S-labeled sense RNAs were used as IL-1, IL-8, AND TNF- mRNAs
(negative) controls. Cells were considered labeled In BCG lesions, MCP-1 mRNA was frequently
when they contained three or more silver grains found in MN and occasionally in vascular
above the number found in the background. endothelial cells, but never in PMN (6, 11). IL-
Cytokine proteins were identified by immuno- 1 and IL-8 mRNAs were found mainly in
histochemical techniques (6, 11). MN and occasionally in PMN.TNF- mRNA
was found only in MN (6, 11).
The Biphasic Pattern In healing (37-day) lesions, large MN were
The percentage of MN labeled for IL-1,TNF- often labeled for MCP-1 mRNA. However, in
, MCP-1, and IL-8 mRNAs showed a bipha- the 37-day lesions, mature epithelioid cells were
sic pattern as these lesions developed (Fig. 1A to not labeled for MCP-1 (or IL-1, TNF-, or
D), i.e., a large initial peak at 1 to 3 days, fol- IL-8) mRNA. In other words, fully activated
lowed by a trough at 5 days, and then another, macrophages were apparently programmed to
smaller peak.The initial MCP-1 mRNA peak destroy or inhibit tubercle bacilli rather than to
indicated that this MN-attracting chemokine produce cytokines.
was a major mediator of the early nonspecific The keratinocytes of the epidermis and hair
cell infiltration caused by BCG. The smaller follicles were occasionally labeled for IL-8
second peak was apparently caused by the devel- mRNA, but not for the other three cytokines.
19. CYTOKINE PRODUCTION IN PRIMARY BCG LESIONS 䡵 305

FIGURE 2 A 16-day dermal BCG lesion


in situ hybridized for IL-8 mRNA. Lique-
fied caseum is in the left lower part of the
photograph. Solid caseum is shown at the far
right of the photograph. Large macrophages
in the liquefied caseum adjacent to the solid
caseum are heavily labeled for IL-8 mRNA.
IL-8 is a major chemokine that attracts
PMN, and numerous PMN are present
throughout the liquefied material. Fixed-
frozen tissue section hybridized with anti-
sense IL-8 35S-labeled RNA, autoradi-
ographed, and counterstained with Giemsa.
Magnification, ⫻350. Reproduced with
permission from reference 11.

IMMUNOHISTOCHEMICAL STUDIES the cytokine proteins are stabilized by compo-


FOR CYTOKINE PROTEINS nents of the extracellular matrix (27).
BCG lesions of various ages were stained by The cells stained for MCP-1 protein were
immunohistochemical techniques for MCP-1 mainly large macrophages (Fig. 3A and B).At 23
and TNF- proteins (11) (Table 1). The per- and 37 days, some microvascular endothelial
centage of cells containing the proteins of these cells stained for MCP-1 protein, but none
two cytokines was highest at 1, 3, and 5 days and stained for TNF- protein.
reduced at 9 days. No decrease occurred in the In tissue sections of BCG lesions, the MN
percentage of MN staining positive for these staining positive for MCP-1 and TNF- pro-
cytokine proteins at 5 days, even though the per- teins showed the same distribution pattern as the
centage of MN containing the mRNAs was MN containing MCP-1 and TNF- mRNAs.
decreased at that time (see Fig. 1). Evidently, This finding further supports the conclusion
cytokine proteins have a slower turnover rate that cells containing cytokine mRNA were
than the mRNAs that produce them. Perhaps, actively producing cytokine protein.
IFN- mRNA IDENTIFIED BY RT-PCR
We were unable to demonstrate rabbit IFN- in
tissue sections of dermal BCG lesions by in situ
TABLE 1 Percentage of mononuclear cells stained hybridization techniques.Therefore, the reverse
for MCP-1 and TNF-a transcription-polymerase chain reaction (RT-
Age of BCG MCP-1 TNF- PCR) (see glossary) was used to measure IFN-
lesions (days) protein protein mRNA in BCG lesions of various ages (11).
1 ⫹⫹⫹ ⫹
The ratio of the IFN- mRNA to the “house-
3 ⫹⫹⫹ ⫹⫹
keeping” GAPDH (glyceraldehyde-3-phosphate
5 ⫹⫹⫹ ⫹⫹
dehydrogenase) mRNA was highest at 5 days,was
9 ⫹⫹ ⫾
decreased at 7 days, and was increased again at
23 ⫾ ⫺
9 days (Fig. 4).The 9-day level was maintained at
37 ⫹ ⫺
23 days, which is when the BCG lesions were
largest in size (11).Thus, similar to the mRNA
a
Symbols: ⫺ ⫽ 0–0.5%; ⫾ ⫽ 0.5–1%; ⫹ ⫽ 1–2%; ⫹⫹ ⫽ levels of the four cytokines represented in Fig. 1,
2–5%; ⫹⫹⫹ ⫽ ⬎5%.Adapted from reference 11.The absence
of a biphasic response suggests that these cytokine proteins have the IFN- mRNAs showed a biphasic pattern,
a slower turnover rate than their mRNAs. but it was somewhat delayed.
306 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

FIGURE 3 (A) An unfixed-frozen


tissue section of a 3-day dermal
BCG lesion stained immunohisto-
chemically for MCP-1 protein.
Large mononuclear cells in the
partly necrotic exudate between
the remaining collagen fibers are
darkly stained for MCP-1 protein.
Anti-rabbit MCP-1 polyclonal anti-
body, biotinylated anti-guinea pig
IgG, and the avidin-biotin-peroxi-
dase procedure, counterstained with
Giemsa. Magnification, ⫻200.
Reproduced with permission from
reference 11. (B) A lightly fixed
tissue section of a 3-day dermal
BCG lesion embedded in glycol
methacrylate and stained with
Giemsa. This photograph shows
some of the histologic detail that is
absent in frozen sections. In the cen-
ter and to the left are a few large
mononuclear cells that are similar
to those stained histochemically in
panel A. Note the frequent occur-
rence of PMN in the 3-day BCG
lesions. Magnification, ⫻460. Repro-
duced with permission from refer-
ence 11.

COMPARISONS AMONG THE FIVE However,the changes in the mRNA levels of


CYTOKINES EVALUATED each cytokine can be compared as the BCG
With in situ hybridization, the antisense RNA lesions developed and healed (Fig. 1, 4, and 5).
probes for one type of cytokine do not necessarily With all five cytokines, the highest levels of
hybridize with the same affinity as do the anti- mRNA seemed to occur early.Subsequently,the
sense probes for another type of cytokine.There- mRNAs decreased, but, during the immune
fore, in Fig. 1, the percentages of MN containing phase,the number of MN containing IFN- and
IL-1,TNF-, MCP-1, and IL-8 mRNAs can- IL-8 mRNAs seemed to remain higher (relative
not be precisely compared with one another, to the early peak) than the number containing IL-
nor can the in situ hybridization technique be 1,TNF-,and MCP-1 mRNAs (Fig.1 and 4).
precisely compared with the RT-PCR technique As stated above, IFN- and TNF- activate
used to measure lesion IFN- mRNA. macrophages to destroy or inhibit the tubercle
19. CYTOKINE PRODUCTION IN PRIMARY BCG LESIONS 䡵 307

FIGURE 4 Changes in IFN- mRNA


in rabbit BCG lesions, relative to those of
GAPDH mRNA (see text). (The amount of
GAPDH mRNA in cells is known to
remain rather constant without wide fluc-
tuations, as the cells up- and downregulate
their various activities.) Note that the bipha-
sic response was somewhat delayed when
compared with that of the other cytokine
mRNAs shown in Fig. 1.This delay suggests
that the initial mononuclear cells produced
cytokines (including chemokines) that sub-
sequently upregulated the IFN- produc-
tion in other cells (mostly in nearby lym-
phocytes). ***P ⬍ 0.01 for day 5 vs. day 7.
The means and their standard errors are
shown. Reproduced with permission from
reference 11.

bacilli that they phagocytize. IL-8 is produced by are consistent with the early nonspecific in vivo
macrophages, but its major function is to attract cytokine response found in the macrophages
and activate granulocytes (mainly PMN) (7). of rabbit BCG lesions described above.
PMN are known to accumulate in areas of bac- Several investigators evaluated the cytokines
terial invasion and around areas of necrosis (28). at the sites of active tuberculosis in humans
Therefore, the high levels of IL-8 mRNA on day (reviewed in references 30, 31, and 32). In tuber-
3, when the BCG bacilli are numerous, and on culous pleuritis (a self-healing form of human
days 9 and 23, when necrosis is present, would tuberculosis), cytokine proteins and mRNAs
be expected (Fig. 1D). At 37 days, when tissue were found in the pleural exudates and/or in
necrosis was much reduced and the ulcer (in pleural tissue biopsies. The cytokines present
the center of the lesion) was becoming epithe- were TNF (33), IFN- (33, 34), IL-10 (34), IL-
lialized, the levels of IL-8 seemed to be decreased. 12 (35), and others. In tuberculous granulomas
within human lungs, IL-1 protein (36) and
STUDIES ON CYTOKINES TGF- (37) were present. None of these stud-
PRODUCED BY TUBERCULOSIS ies, however, evaluated changes in cytokine
IN HUMANS AND IN MICE mRNA or protein as the lesions progressed and
The role of cytokines in tuberculosis is a very regressed.
active field of investigation. Here, we list a few Other investigators evaluated the cytokines in
of these studies with comments on their appli- peripheral blood mononuclear cells (PBMC)
cation to the pathogenesis of the human disease. from tuberculous patients. With the RT-PCR
technique, IL-1, IL-8, TNF-, and IFN-
Humans mRNAs were found in PBMC of these patients
The human monocytic-leukemia cell line THP1 but were absent or reduced in PBMC from
was infected with Mycobacterium tuberculosis healthy tuberculin-negative persons (38).When
(H37Rv) at an average of 10 bacilli per cultured 24 h in the presence of tubercle bacilli,
macrophage (29). In these cells, the mRNAs of the PBMC of tuberculous patients produced less
numerous chemokines were increased several- IFN- mRNA and IFN- protein than did the
fold during the first 6 to 12 h after infection. PBMC of healthy tuberculin-positive controls
These in vitro results with human macrophages (39). Also, more TGF-, an immunosuppressive
308 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

FIGURE 5 Summary of the histologic characteristics and cytokines in early and peak rabbit
dermal BCG lesions. Note that the cytokines upregulated in the early lesions were downregulated
by the time the lesions peaked at 14 to 23 days. Reproduced with permission from reference 11.

cytokine, was produced by the PBMC of patients Mice


with tuberculosis than by the PBMC of healthy Cytokines participating in mouse tuberculosis
tuberculin-positive controls (32, 37, 40). are described in references 41 through 49.
Although analyses of human PBMC may be Mouse lungs, infected by aerosol with virulent
prognostically useful, they do not always reflect M. tuberculosis, contained the mRNAs of IL-1,
the situation in the tuberculous lesions them- TNF-, IL-2, IFN- and the chemokines
selves. For example, in hosts with poor resistance, RANTES, MCP-1, MCP-3, MIP-1, MIP-2,
an increase in suppressor cytokines may reflect and IP-10. To measure the mRNAs of these
(and be the cause of) the progression of the cytokines, lung homogenates were prepared,
local disease. On the other hand, in hosts with the RT-PCR was used.The amount of cytokine
good resistance, an increase in such suppressors mRNA was compared with the amount of a
may reflect the regression or control of the local “housekeeping” gene.The highest levels of most
disease, because the further accumulation of cytokine mRNAs in the mouse tuberculous
host defense cells may no longer be needed and lungs occurred between 3 and 9 weeks after
may even be detrimental. onset of the infection (47). However, the exact
19. CYTOKINE PRODUCTION IN PRIMARY BCG LESIONS 䡵 309

role of each cytokine and its effect on neigh- 11. Sugisaki, K., A. M. Dannenberg, Jr., Y. Abe,
boring cells in tuberculous lesions remain to be J. Tsuruta, W.-J. Su, W. Said, L. Feng,
T.Yoshimura, P. J. Converse, and P. Mounts.
elucidated. 1998. Nonspecific and immune-specific up-
regulation of cytokines in rabbit dermal tubercu-
CONCLUSIONS: NONSPECIFIC AND lous (BCG) lesions. J. Leukoc. Biol. 63:440–450.
ANTIGEN-SPECIFIC CYTOKINE 12. Shigenaga, T., A. M. Dannenberg, Jr., D. B.
PRODUCTION Lowrie, W. Said, M. J. Urist, H. Abbey, B. H.
Schofield, P. Mounts, and K. Sugisaki. 2001.
Cytokines (including chemokines) play a major Immune responses in tuberculosis: antibodies and
role in the development, progression, and regres- CD4-CD8 lymphocytes with vascular adhesion
sion of tuberculous lesions. They cause host molecules and cytokines (chemokines) cause a rapid
defense cells to enter and become activated, so antigen-specific cell infiltration at sites of bacillus
that those cells can destroy or inhibit the tuber- Calmette-Guérin reinfection. Immunology 102:466–
479.
cle bacillus. Early cytokine production is non- 13. Apella, E., K. Matsushima, J. J. Oppenheim,
specific, caused by the irritant and adjuvant T.Yoshimura, E. J. Leonard, G. M. Clore, and
effects of the bacillus, but subsequent cytokine A. M. Gronenborn. 1990. Determination of the
production is mostly antigen specific and caused primary and secondary structure of NAP-1/IL-8
by the host’s immune response. and a monocyte chemoattractant protein, MCP-
1/MCAF, p. 405–417. In M. Melli and L. Parente
(ed.), Cytokines and Lipocortins in Inflammation and
REFERENCES Differentiation.Wiley-Liss, Inc., New York, N.Y.
14. Nathan, C., and R. Yoshida. 1988. Cytokines:
1. Janeway, C. A., Jr., P.Travers, M.Walport, and
interferon-gamma, p. 229–251. In J. I. Gallin, I. M.
M. J. Shlomchik. 2001. Immunobiology: the Immune
Goldstein, and R. Snyderman (ed.), Inflammation:
System in Health and Disease, 5th ed. Garland Pub-
Basic Principles and Clinical Correlates. Raven Press,
lishing, New York, N.Y.
New York, N.Y.
2. Callard, R. E., and A. J. H. Gearing. 1994. The
15. Nathan, C. F., H.W. Murray, M. E.Wiebe, and
Cytokine FactsBook.Academic Press, Inc., San Diego,
B. Y. Rubin. 1983. Identification of interferon-
Calif.
gamma as the lymphokine that activates human
3. Luger,T. A., and T. Schwarz. 1995. The role of
macrophage oxidative metabolism and antimicro-
cytokines and neuro-endocrine hormones in cuta-
bial activity. J. Exp. Med. 158:670–689.
neous immunity and inflammation. Allergy 50:292–
302. 16. Murray, H.W. 1988. Interferon-gamma, the acti-
4. Kupper, T. S. 1990. Immune and inflammatory vated macrophage, and host defense against micro-
processes in cutaneous tissues: mechanisms and bial challenge. Ann. Intern. Med. 108:595–608.
speculations. J. Clin. Investig. 86:1783–1789. 17. Denis, M. 1991. Interferon-gamma-treated murine
5. (Reference deleted.) macrophages inhibit growth of tubercle bacilli via
6. Tsuruta, J., K. Sugisaki, A. M. Dannenberg, the generation of reactive nitrogen intermediates.
Jr.,T.Yoshimura,Y. Abe, and P. Mounts. 1996. Cell. Immunol. 132:150–157.
The cytokines NAP-1 (IL-8), MCP-1, IL-1 and 18. Flynn, J. L., J. Chan, K. J.Triebold, D. K. Dal-
GRO in rabbit inflammatory skin lesions produced ton, T. A. Stewart, and B. R. Bloom. 1993.
by the chemical irritant sulfur mustard. Inflammation An essential role for interferon- in resistance to
20:293–318. Mycobacterium tuberculosis infection. J. Exp. Med.
7. Vaddi, K., M. Keller, and R. C. Newton. 1997. 178:2249–2254.
The Chemokine FactsBook.Academic Press, Inc., San 19. Beutler, B. 1995.TNF, immunity and inflamma-
Diego, Calif. tory disease: lessons of the past decade. J. Investig.
8. Furie, M. B., and G. J. Randolph. 1995. Med. 43:227–235.
Chemokines and tissue injury. Am. J. Pathol. 146: 20. Hernandez-Pando, R., and G. A. W. Rook.
1287–1301. 1994. The role of TNF-alpha in T-cell mediated
9. Kunkel, S. L., R. M. Strieter, I. J. D. Lindley, inflammation depends on the Th1/Th2 cytokine
and J. Westwick. 1995. Chemokines: new li- balance. Immunology 82:591–595.
gands, receptors and activities. Immunol.Today 16: 21. Hirsch, C. S., J. J. Ellner, D. G. Russell, and
559–561. E.A. Rich. 1994. Complement receptor-mediated
10. Carr, M. W., S. J. Roth, E. Luther, S. S. Rose, uptake and tumor necrosis factor alpha-mediated
and T. A. Springer. 1994. Monocyte chemoat- growth inhibition of Mycobacterium tuberculosis by
tractant protein acts as a T-lymphocyte chemoat- human alveolar macrophages. J. Immunol. 152:743–
tractant. Proc. Natl.Acad. Sci. USA 91:3652–3656. 753.
310 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

22. Nathan, C. 1991. Mechanisms and modulations of 35. Zhang, M., M. K. Gately, E. Wang, J. Gong,
macrophage activation. Behring Inst. Mitt. 88:200– S. F.Wolf, S. Lu, R. L. Modlin, and P. F. Barnes.
207. 1994. Interleukin-12 at the site of disease in tuber-
23. (Reference deleted.) culosis. J. Clin. Investig. 93:1733–1739.
24. Orme, I. M., E. S. Miller, A. D. Roberts, S. K. 36. Chensue, S. W., K. S. Warmington, A. E.
Furney, J. P. Griffin, K. M. Dobos, D. Chi, Berger, and D. E. Tracey. 1992. Immunohisto-
B. Rivoire, and P. J. Brennan. 1992.T lympho- chemical demonstration of interleukin-1 receptor
cytes mediating protection and cellular cytolysis antagonist protein and interleukin-1 in human
during the course of Mycobacterium tuberculosis infec- lymphoid tissue and granulomas. Am. J. Pathol.
tion. Evidence for different kinetics and recognition 140:269–275.
of a wide spectrum of protein antigens. J. Immunol. 37. Toossi, Z., P. Gogate, H. Shiratsuchi,
148:189–196. T.Young, and J. J. Ellner. 1995. Enhanced pro-
25. Dannenberg, A. M., Jr. 1968. Cellular hyper- duction of TGF- by blood monocytes from
sensitivity and cellular immunity in the patho- patients with active tuberculosis and presence of
genesis of tuberculosis: specificity, systemic and local TGF- in tuberculous granulomatous lung lesions.
nature, and associated macrophage enzymes. Bacte- J. Immunol. 154:465–473.
riol. Rev. 32:85–102. 38. Schauf, V., W. N. Rom, K. A. Smith, E. P.
26. Suga, M., A. M. Dannenberg, Jr., and Sampaio, P. A. Meyn, J. M. Tramontana,
S. Higuchi. 1980. Macrophage functional hetero- Z.A. Cohn, and G. Kaplan. 1993. Cytokine gene
geneity in vivo: macrolocal and microlocal activation and modified responsiveness to inter-
macrophage activation, identified by double- leukin-2 in the blood of tuberculosis patients.
staining tissue sections of BCG granulomas for J. Infect. Dis. 168:1056–1059.
pairs of enzymes. Am. J. Pathol. 99:305–324. 39. Zhang, M., Y. Lin, D. V. Iyer, J. Gong, J. S.
27. Smith, R. E., C. M. Hogaboam, R. M. Strieter, Abrams, and P. F. Barnes. 1995.T-cell cytokine
N. W. Lukacs, and S. L. Kunkel. 1997. Cell- responses in human infection with Mycobacterium
to-cell and cell-to-matrix interactions mediate tuberculosis. Infect. Immun. 63:3231–3234.
chemokine expression: an important component 40. Ellner, J. J. 1997. Regulation of the human
of the inflammatory lesion. J. Leukoc. Biol. 62:612– immune response during tuberculosis. J. Lab. Clin.
619. Med. 130:469–475.
28. Majno, G., and I. Joris. 1996. Cells,Tissues, and 41. Orme, I. M. 1996. Immune responses in animal
Disease: Principles of General Pathology, p. 385–427. models, p. 181–196. In T. M. Shinnick (ed.), Tuber-
Blackwell Science, Inc, Cambridge, Mass. culosis. Springer Verlag, Berlin, Germany.
29. Ragno, S., M. Romano, S. Howell, D. J. C. 42. Flynn, J. L., and J. Chan. 2001. Immunology of
Pappin, P. J. Jenner, and M. J. Colston. 2001. tuberculosis. Annu. Rev. Immunol. 19:93–129.
Changes in gene expression in macrophages 43. Orme, I. M., and F. M. Collins. 1994. Mouse
infected with Mycobacterium tuberculosis: a combined model of tuberculosis, p. 113-134. In B. R. Bloom
transcriptomic and proteomic approach. Immunol- (ed.), Tuberculosis: Pathogenesis, Protection and Control.
ogy 104:99–108. ASM Press,Washington, D.C.
30. Barnes, P. F., and R. L. Modlin. 1996. Human 44. Rhoades, E. R.,A. M. Cooper, and I. M. Orme.
cellular immune responses to Mycobacterium tuber- 1995. Chemokine response in mice infected with
culosis, p. 197–219. In T. M. Shinnick (ed.), Tubercu- Mycobacterium tuberculosis. Infect. Immun. 63:3871–
losis. Springer Verlag, Berlin, Germany. 3877.
31. Barnes, P. F., R. L. Modlin, and J. J. Ellner. 45. Saunders, B. M., and A. M. Cooper. 2000.
1994.T-cell responses and cytokines, p. 417–435. In Restraining mycobacteria: role of granulomas in
B. R. Bloom (ed.), Tuberculosis: Pathogenesis, Protec- mycobacterial infections. Immunol. Cell Biol. 78:334–
tion and Control.ASM Press,Washington, D.C. 341.
32. Wallis, R. S., and J. J. Ellner. 1994. Cytokines 46. Cardona, P.-J., R. Llatjós, S. Gordillo, J. Díaz,
and tuberculosis: a review. J. Leukoc. Biol. 55:676– I. Ojanguren, A. Ariza, and V. Ausina. 2000.
681. Evolution of granulomas in lungs of mice infected
33. Barnes, P. F., S.-J. Fong, P. J. Brennan, P. E. aerogenically with Mycobacterium tuberculosis. Scand.
Twomey, A. Mazumder, and R. L. Modlin. J. Immunol. 52:156–163.
1990. Local production of tumor necrosis factor and 47. Cardona, P.-J., S. Gordillo, J. Díaz, G. Tapia,
IFN- in tuberculous pleuritis. J. Immunol. 145:149– I.Amat, Á. Pallarés, C.Vilaplana,A.Ariza, and
154. V. Ausina. 2003. Widespread bronchogenic dis-
34. Barnes, P. F., S. Lu, J. S. Abrams, E. Wang, semination makes DBA/2 mice more susceptible
M. Yamamura, and R. L. Modlin. 1993. than C57BL/6 mice to experimental aerosol infec-
Cytokine production at the site of disease in human tion with Mycobacterium tuberculosis. Infect. Immun.
tuberculosis. Infect. Immun. 61:3482–3489. 71:5845–5954.
19. CYTOKINE PRODUCTION IN PRIMARY BCG LESIONS 䡵 311

48. Guirado, E., S. Gordillo, O. Gil, J. Díaz, 49. Gil, O., E. Guirado, S. Gordillo, J. Díaz,
G. Tapia, C. Vilaplana, V. Ausina, and P.-J. G.Tapia, C.Vilaplana, A. Ariza,V. Ausina, and
Cardona. 2006. Intragranulomatous necrosis in P.-J. Cardona. 2006. Intragranulomatous necrosis
pulmonary granulomas is not related to resistance in lungs of mice infected by aerosol with Mycobac-
against Mycobacterium tuberculosis infection in exper- terium tuberculosis is related to bacterial load rather
imental murine models induced by aerosol. Int. J. than to any one cytokine or T cell type. Microbes
Exp. Pathol. 87:139–149. Infect. 8:628–636.
20
CYTOKINE PRODUCTION IN
REINFECTION BCG LESIONS
AND IN TUBERCULIN REACTIONS

Production of primary and reinfection BCG lesions and tuberculin


reactions [313]
Local cell infiltration [313]
Lesion size, ulceration, and healing [313]
Tuberculin reactions [313]
Mononuclear cells (MN) and granulocytes (PMN) in primary and reinfection
BCG lesions [315]
Number of tubercle bacilli in early primary and reinfection BCG
lesions [319]
Cytokines in primary and reinfection BCG lesions and in tuberculin
reactions [319]
IL-1 and IL-8 mRNAs in PMN [321]
Causes of cytokine downregulation [321]
Cytokine networks [323]
Conclusions [323]

Abstract. Reinfection BCG lesions provide a simple model of how tuberculosis vaccines
would affect an exogenous infection with virulent tubercle bacilli.Therefore, rabbit der-
mal primary and reinfection BCG lesions were produced and evaluated during the first
5 days of their existence.Tissue sections of the lesions were prepared, and the types of cells
and their cytokine mRNAs and proteins were analyzed by histochemical methods.The
cytokines studied were interleukin-1, macrophage chemoattractant (activating) protein
(MCP-1), interleukin-8, and tumor necrosis factor alpha (see chapter 19).
Our most informative findings were with MCP-1, one of the main chemokines attract-
ing mononuclear cells (MN). (In tuberculous lesions, MN are mostly macrophages but also
contain dendritic cells and lymphocytes.) At 3 h, both the reinfection lesions and the pri-
mary lesions contained the same percentage of MN labeled for MCP-1 mRNA. How-
ever, the reinfection lesions were 400 to 500 times larger and therefore contained many
more of these MN.This high cell number alone would cause the total chemokine pro-
duction to exceed by far that occurring in the primary lesions.
By 1 day, the percentage of MN containing MCP-1 mRNA (and protein) had markedly
decreased in the reinfection lesions, but remained high for at least 2 days in the primary
lesions, which were beginning to increase in size.This finding suggests that chemokine pro-
duction is turned off when sufficient MN have accumulated. In other words, the local accu-
mulation of MN is carefully regulated, so that excessive cell infiltration into the lesions is
prevented.
The rapid local accumulation of MN (macrophages, dendritic cells, and antigen-
specific lymphocytes) in the early reinfection BCG lesions seemed to be due to the
presence of antibodies that developed during the first BCG infection (see chapter 5).
The antigen-antibody complexes formed at the site of reinfection evidently produced
chemotactic factors that markedly hastened the cell infiltration.
In general, cytokine production in tuberculin reactions showed the same pattern as that
found in the early reinfection BCG lesions.

312
20. CYTOKINE PRODUCTION IN REINFECTION BCG LESIONS 䡵 313

Rabbits were immunized with BCG.Then, the By 4 and 5 days, the size of the reinfection
effects of this immunization on the cell compo- BCG lesions had markedly decreased, and the
sition and the cytokines of BCG lesions of rein- size of the primary lesions had moderately
fection (made 24 days later) were evaluated. increased, so that the two types of lesion became
These studies provide insight into how vaccines almost the same size (Fig. 1A and C). From 5
would affect the development and progress of a days on, the sizes of the primary and reinfection
challenging infection with virulent bacilli. BCG lesions followed a similar pattern. How-
Chapter 19 describes briefly the functions ever, a second (immune-mediated) peak in size
of the cytokines that we evaluated and the occurred in the reinfection lesions at 8 days
methodology used. More details are presented (Fig. 1A) and in the primary lesions at 12 days.
in references 1, 2, and 3. The second peak occurred earlier in the rein-
fection group, because the second BCG injec-
PRODUCTION OF PRIMARY AND tion boosted the already existing CMI and
REINFECTION BCG LESIONS AND DTH. In both groups, a gradual decline in lesion
TUBERCULIN REACTIONS size followed the second peak.
New Zealand White rabbits (about 2.6 kg) were
immunized by the intradermal injection of LESION SIZE, ULCERATION,
about 5 ⫻ 106 (log-phase) Tice BCG bacilli on AND HEALING
the right and left flanks (2, 3).Twenty-four days No apparent difference was found in the times
later, they were reinfected with 5 ⫻ 106 (log- when the primary and reinfection BCG lesions
phase) Tice BCG bacilli in two similar sites (2, ulcerated and when they healed—probably
3).At that time, for comparison, primary BCG because of the variations present among these
lesions were also produced in nonimmunized outbred rabbits.The ulceration was followed by
rabbits. Two experiments were performed. In discharge of the lesion’s necrotic contents, which
Experiment I, the size of the BCG lesions was was a major contributor to the decrease in lesion
followed for 42 days (Fig. 1). In Experiment II, size associated with healing. Sometimes, however,
tissue sections of the lesions were made and the discharge of necrotic material was incomplete,
evaluated for only 5 days, when the difference which slowed the healing process.At 6 weeks, the
in the size of primary and reinfection BCG sites of the BCG injections still could be read-
lesions was most striking (see Fig. 1). ily discerned, but at 14 weeks, grossly visible
evidence of the lesions could not usually be
LOCAL CELL INFILTRATION found, either on the skin surface or, at necropsy,
Figures 1A and C show the size of primary and on the underside of the skin.
reinfection BCG lesions at various times in
Experiments I and II, respectively. Within the TUBERCULIN REACTIONS
first 3 h, the reinfection lesions had substantially In Experiment I, the rabbits were injected intra-
increased in size, and peaked at 1 or 2 days. At dermally at various times with a 1:30 dilution of
that time, the primary lesions were still very 4⫻ Old Tuberculin, and the induration of the
small; in fact, the primary lesions and reinfection resulting tuberculin reaction was measured with
lesions showed a 400- to 500-fold difference in calipers 2 days later (Fig. 1B) (3). In the primary
size.The cell infiltration, which caused this dif- BCG group, tuberculin sensitivity was not pre-
ference in size, was antigen specific. It could sent initially, but became strong between 9 and 23
only have been initiated by an antigen- days, and then slowly declined. In the reinfection
antibody reaction, because few, if any, memory group, tuberculin sensitivity was rather strong
T cells are present in skin. (See chapter 5 for a before the second injection of BCG and declined
discussion of the role of antibodies in acceler- thereafter. If a booster effect of BCG on the 2-
ating local cell-mediated immunity [CMI] and day tuberculin reactions occurred in these rabbits,
delayed-type hypersensitivity [DTH]). it was no longer apparent 10 days later (3).
FIGURE 1 (A) Size of primary and reinfection BCG lesions from 3 h to 42 days in rabbits of
Experiment I (3).The reinfected rabbits had been sensitized intradermally by BCG 24 days pre-
viously. Note that the reinfection BCG lesions were many times larger than the primary BCG
lesions at 3, 12, 24, and 48 h, a fact that was apparently initiated by an antigen-antibody reaction
(see chapter 5). Note also that the size of the reinfection BCG lesions reached a second peak at

314
20. CYTOKINE PRODUCTION IN REINFECTION BCG LESIONS 䡵 315

In Experiment II (which is the main one pro- macrophages, dendritic cells, medium and large
viding the data for this chapter), we injected Old lymphocytes, and some activated fibroblasts.
Tuberculin at the same time that we injected Figure 2 also shows the number of PMN
BCG to produce lesions in the primary and rein- (including some eosinophils) per mm2. In the
fection groups of rabbits.The resulting skin lesions early BCG lesions, small areas of necrosis
were then measured over the next 5 days (Fig.1C) occurred, probably at sites containing the
(3). In the reinfected host, the size of lesions pro- most bacilli. PMN were present near these
duced by both BCG and tuberculin increased and necroses, but PMN were rare in more periph-
decreased in parallel (Fig. 1C). By 5 days, how- eral areas. The cell counts were made in the
ever, the local amount of tuberculin had proba- large intermediate area between the more
bly decreased substantially and no longer provided centrally located necroses and the relatively
the stimulus that it had provided earlier. Note that cell-poor areas in the lesion periphery. This
in this experiment, no pre-reinfection tuberculin intermediate area was densely infiltrated with
tests were given, so we could not determine MN along with some PMN.
whether BCG and tuberculin had a booster effect Many more MN per mm2 were present in the
on tuberculin sensitivity. reinfection BCG lesions than in primary BCG
During the same 5-day period of study, var- lesions until day 5, at which time both types of
ious cell types and their cytokines were evalu- lesion contained the same number of MN per
ated (Fig. 2 through 6). The cell types and mm2 (Fig. 2A).The number of MN per mm2 was
cytokines in the tuberculin reactions and those usually much greater than the number of PMN
in the reinfection BCG lesions increased and per mm2 (Fig. 2 and Fig. 13 of chapter 6) (2, 3).
decreased in a rather similar manner (3), despite The early PMN response depicted in Fig. 2B
the fact that the antigenic compositions of Old was probably due to the nonspecific irritants in
Tuberculin and intact tubercle bacilli are some- the vaccine (see chapter 19) (2). PMN are
what different (see reference 4). thought to play only a minimal role in the devel-
opment and healing of tuberculous lesions (5).
MONONUCLEAR CELLS (MN) AND Eosinophils were included in the PMN
GRANULOCYTES (PMN) IN PRIMARY group, because PMN are eosinophilic in the
AND REINFECTION BCG LESIONS rabbit (see chapter 6). Rabbit eosinophils can
Figure 2 shows the number of MN per mm2 of sometimes be differentiated from PMN by their
tissue section of BCG lesions. MN include large oval granules (6). By this criterion,

8 days, whereas the primary lesions reached a similar peak at 12 days.These second peaks were
apparently caused by an antigen-specific CMI/DTH reaction (see chapter 5). After the second
peaks, the lesions slowly regressed. Each point represents the mean size of the lesions and its stan-
dard error. (B) Size of 2-day tuberculin reactions in rabbits of Experiment 1 (3). In the reinfected
host, tuberculin sensitivity was highest before challenge.This sensitivity declined thereafter, and
no booster effect from the second BCG injection was apparent. In contrast, hosts with primary
BCG infections had strong tuberculin sensitivity by 9 days, which tended to remain higher than
that present in the reinfected hosts, possibly because the infecting bacilli were not destroyed as
readily. Each point represents the mean size of the tuberculin reactions and its standard error.
(C) Size of primary and reinfection BCG lesions and tuberculin reactions, each measured from
3 h to 5 days in Experiment II (3).As in Experiment I, the reinfected rabbits were sensitized intra-
dermally by BCG 24 days previously. Note that the reinfection BCG lesions and the tuberculin
reactions showed the same pattern, and that the primary BCG lesions remained very small until
DTH and CMI started to develop at 4 or 5 days.The tuberculin reactions in the rabbits that had
only primary BCG lesions are not shown, because they were small or absent (see panel B). Each
point represents the mean size of the lesions and its standard error.
In panels A and C, reinfection BCG lesions versus primary BCG lesions: *P ⬍ 0.05 and **P
⬍ 0.01. Reproduced with permission from reference 3. Note that this figure also appears as Fig.
5 in chapter 5.
316 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

FIGURE 2 Number of mononuclear cells (MN) (A) and PMN (B) per mm2 of tissue section
at various times in reinfection BCG lesions, tuberculin reactions, and primary BCG lesions (3).
The reinfection BCG lesions and tuberculin reactions show no differences in the density of either
cell type, but the primary lesions contain fewer MN per mm2 than the other two at 12 h and
at 1 and 2 days.The total number of MN and PMN in each type of lesion can be estimated by
combining the data per mm2 (this figure) with the lesion size (Fig. 1). Each point represents the
mean and its standard error (see reference 3 for P values). Reproduced with permission from ref-
erence 3.
The number of MN and PMN per mm2 in primary BCG lesions at later time periods is shown
in Fig. 13 in chapter 6.
20. CYTOKINE PRODUCTION IN REINFECTION BCG LESIONS 䡵 317

FIGURE 3 Percentage of MN immunostained for CD4 (A) and CD8 (B) in the three types
of lesion (3).At 2 days, the reinfection BCG lesions and tuberculin reactions contained a higher
percentage of CD8 cells per mm2 than did the primary lesions, suggesting that tuberculin sen-
sitivity favors the production of cytotoxic T lymphocytes. Note that CD4 cells are much more
numerous than CD8 cells (compare the scales on the y axes).The means and their standard errors
are shown (see reference 3 for P values).The CD4/CD8 ratios in primary BCG lesions at later
times are shown in Table 4 of chapter 6. Reproduced with permission from reference 3.This fig-
ure also appears as Fig. 12 in chapter 6.
FIGURE 4 Percentage of mononuclear cells (MN) labeled for IL-1 mRNA (A), MCP-1
mRNA (B), and IL-8 mRNA (C) in the three types of lesion (3).The percentage of MN con-
taining the three cytokine mRNAs shows early peak levels at 3 h.Then, this percentage of MN
rapidly declines in BCG lesions of reinfection and in tuberculin reactions, but remains relatively

318
20. CYTOKINE PRODUCTION IN REINFECTION BCG LESIONS 䡵 319

eosinophils seemed to compose less than 10% of the mRNAs and/or the proteins of the cyto-
the granulocyte population in BCG lesions. kines interleukin-1 (IL-1), tumor necrosis
CD4 MN (mainly lymphocytes) were more factor alpha (TNF-), macrophage chemoat-
numerous than CD8 MN in both primary and tractant-activating protein-1 (MCP-1), and
reinfection BCG lesions (Fig. 3). Both CD4 IL-8 (Fig. 4 and 5) as described in chapter 19
and CD8 cell types increased more rapidly in the and reference 3.
reinfection lesions. See chapter 6 for other stud-
ies and a more complete discussion of CD4 Cytokine mRNAs
and CD8 lymphocytes in the rabbit model of At 3 h, the percentage of MN labeled for IL-1
tuberculosis. mRNA, MCP-1 mRNA, and IL-8 mRNA
In brief, the cells mainly responsible for the showed peak levels in all three types of lesion:
fate of rabbit tuberculous lesions are macro- primary BCG lesions, reinfection BCG lesions,
phages and lymphocytes. (The percentage of and tuberculin reactions (Fig. 4). Since the rab-
dendritic cells in such lesions has not been bits with primary lesions were not immunized,
determined.) Within the lymphocyte popula- their 3-h reaction was nonspecific (2), whereas
tion, CD4 cells predominate. that in the reinfection lesions was probably a
combination of a nonspecific response and an
NUMBER OF TUBERCLE BACILLI IN antigen-antibody response (see chapter 5). In
EARLY PRIMARY AND REINFECTION reinfection lesions, the percentage of MN con-
BCG LESIONS
taining the MCP-1 mRNAs rapidly declined
In tissue sections, we were unable to accurately
between 3 and 12 h (Fig. 4), but in the primary
assess the total number of tubercle bacilli in the
lesions this decline was delayed (Fig. 4).
primary and reinfection BCG lesions by count-
At 2 days, the percentages of MN labeled for
ing the number of acid-fast bacilli in the central
IL-1, MCP-1, and IL-8 mRNAs in primary
areas of the lesions (3). Before day 5, the lesions
lesions were higher than those in the reinfection
of the two groups differed so markedly in size
lesions (Fig. 4).This finding does not mean that
(Fig. 1) that differences in the number of bacilli
the primary BCG lesions contained a greater
per mm2 were rather meaningless:the same num-
number of MN labeled for cytokine mRNAs
ber of bacilli would be distributed over markedly
than did the reinfection lesions, because the
different volumes. On day 5, however, when the
primary lesions were so much smaller than the
primary and reinfection BCG lesions were
reinfection lesions (see Fig. 1C).
approximately the same size (Fig.1),similar num-
Changes in the percentage of cells labeled for
bers of tubercle bacilli seemed to be present.
cytokine mRNA foretell the course of the
Homogenizing BCG skin lesions is difficult
lesion. For example, an increase in the per-
because of the collagen present.Therefore,no cul-
centage of cells labeled for MCP-1 mRNA
tures were made to determine the effect of immu-
indicates that more MCP-1 protein will be
nization on the number of viable bacilli present.
produced and that more MN will infiltrate the
CYTOKINES IN PRIMARY AND lesion because of being attracted there by this
REINFECTION BCG LESIONS AND IN chemokine. Conversely, a decrease in the per-
TUBERCULIN REACTIONS centage of cells labeled for MCP-1 mRNA
In tissue sections of primary and reinfection indicates that MN infiltration will be down-
BCG lesions and tuberculin reactions, we regulated. Similarly, increases or decreases in
determined the percentage of cells containing the percentage of cells containing a cytokine

high in primary BCG lesions at 2 days, especially the percentage containing MCP-1 mRNA.At
2 days, the primary lesions are growing in size, whereas the reinfection lesions and tuberculin reac-
tions are regressing (see Fig. 1C).The means and their standard errors are shown (see reference
3 for P values). Reproduced with permission from reference 3.
320 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

FIGURE 5 Percentage of mononuclear cells (MN) labeled for MCP-1 protein (A) and
TNF- protein (B) in the three types of lesion (3).The percentage of MN containing these
two cytokine proteins shows the same pattern as the percentage of MN containing MCP-
1 mRNA in Fig. 4, but the early peak at 3 h in the tuberculin-sensitive hosts is much less
pronounced. (We did not label TNF- mRNA in these lesions.) The means and their stan-
dard errors are shown (see reference 3 for P values). Reproduced with permission from
reference 3.
20. CYTOKINE PRODUCTION IN REINFECTION BCG LESIONS 䡵 321

protein indicate that the lesion is progressing or of the PMN and 7% of the MN were labeled for
regressing, respectively. IL-8 mRNA.This difference in the percentages
of cytokine-labeled PMN and MN was also
Cytokine Proteins present at a later time. In other words, a high
The MCP-1 and TNF- proteins were visual- proportion of PMN were engaged in IL-1
ized immunohistochemically with specific anti- and IL-8 production, and only a relatively small
bodies (Fig. 5) (2, 3). In primary and reinfection proportion of the MN were so engaged. Most
BCG lesions, the percentage of MN stained for of the MN were probably involved in numer-
MCP-1 and TNF- proteins followed roughly ous other activities (11, 12).
the same pattern as the percentage of MN
CAUSES OF CYTOKINE
stained for MCP-1 mRNA (compare Fig. 4 DOWNREGULATION
with Fig. 5).This finding indicates that the cells The literature identifies many factors that seem
containing MCP-1 mRNA were actively pro- to be involved in the downregulation of cyto-
ducing MCP-1 protein. (TNF mRNA was not kines.
evaluated in these experiments.)
During the first 5 days, the cytokines in tuber- 1. High local concentrations of both anti-
culin reactions resembled those in reinfection gens and certain cytokines no longer
BCG lesions (Fig. 4 and 5). By 5 days, however, stimulate nearby cells and can even induce
most of the tuberculin at the site had probably their apoptosis (13)—called active apop-
been removed by hydrolysis or by diffusion into tosis (14, 15). Fas ligand and TNF are
lymphatics. involved (14, 16).
2. Very low antigen concentrations (caused
IL-1 AND IL-8 mRNAs IN PMN by a decrease in bacilli and their products)
IL-1 is a primary cytokine that tends to upreg- stop the production of certain cytokines
ulate other cytokines (7–9), and IL-8 is a and result in apoptosis of nearby cells—
chemokine that attracts PMN into the local called passive apoptosis (14). The with-
site and activates them (10). (We did not find drawal of IL-2 induces this type of apop-
MCP-1 mRNA in rabbit PMN [1, 2]). tosis (14).
PMN were evaluated in the densely infiltrated 3. Regulatory T cells (17–25) (including T
areas of BCG lesions that were not adjacent to suppressor cells [23, 24, 26]) and regulatory
areas of necrosis (see chapter 19). In reinfection dendritic cells (25, 27–29) play distinct
BCG lesions,the percentage of PMN labeled for roles (also see chapter 6).
IL-1 mRNA and IL-8 mRNA showed a peak 4. The TIM gene family (30–32) stops both
at 3 h and then remained at lower levels (Fig. 6). excessive T-cell proliferation and excessive
In primary BCG lesions,the 3-h peak was smaller, cytokine production.
and less regression occurred (Fig.6).In tuberculin 5. Decreases in the number and type of
reactions, the PMN labeled for IL-1 and IL-8 chemokine receptors also play a role (33–
mRNAs peaked at 3 h and were low at 1, 2, and 35), as do increases in soluble cytokine
5 days (Fig. 6). receptors (36–38) and increases in soluble
In all lesions, the intensity of the PMN stain- inhibitors, such as IL-1 receptor antago-
ing for the IL-1 and IL-8 mRNAs indicates nist (39).
the amounts present.The intensity was strongest 6. In addition, the production of various
at 3 h.Thereafter, few PMN stained, and those cytokines is downregulated by transform-
that did stained only faintly. ing growth factor  (17, 40), IL-10 (17, 28,
A higher percentage of PMN than MN were 41), IL-4 (17), IL-6 (41), prostaglandin E2
labeled for IL-1 and IL-8 mRNAs (compare (40),lipoxygenase-derived eicosanoids (e.g.,
Fig. 4 and 6). At the 3-h peak of reinfection 15-hydroperoxy-eicosatetraenoic acid)
BCG lesions, 36% of the PMN and 5% of the (42a, 43), inducible NO (44–46), platelet-
MN were labeled for IL-1 mRNA, and 48% activating factor (40), adenosine (47),
322 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

FIGURE 6 Percentage of PMN labeled for IL-1 mRNA (A) and IL-8 mRNA (B) in
the three types of lesion (3). Rabbit PMN do not contain MCP-1 mRNA (1–3). Similar
to the MN in Fig. 4, the percentage of PMN containing these cytokine mRNAs showed
a 3-h peak.Then, the percentage of labeled PMN declined in the reinfection BCG lesions
and tuberculin reactions, but remained somewhat elevated in the primary BCG lesions, again
similar to the MN in Fig. 4.
20. CYTOKINE PRODUCTION IN REINFECTION BCG LESIONS 䡵 323

adenosine triphosphate (48), catecho- the type and stage of the inflammatory lesions
lamines (49), immune complexes (50), involved.
annexin I (lipocortin 1) (51), suppressors of The initial exposure to one cytokine evi-
cytokine signaling (52, 53), and, at times, dently determines the type of macrophage acti-
even gamma interferon (IFN-) (54). vation and renders the macrophage temporar-
ily unresponsive to activation of another type by
The exact role of each of these factors in the
another cytokine (58). Such studies may explain
early downregulation of chemokines in rein-
the heterogeneity of macrophage functions
fection BCG lesions is not known. It is clear,
within lesions caused by tubercle bacilli (11,
however, that when sufficient numbers of MN
12). Of interest is that IL-12, IFN-, and IL-2
are present, the chemokine production causing
bind to heparin and heparan sulfate, which keep
further MN infiltration is turned off.
these cytokines localized near the cells that pro-
CYTOKINE NETWORKS duce them (reviewed in reference 59).
Our studies describe the rise and fall of various An immunohistochemical study of cytokines
cytokines within primary and reinfection rab- in tuberculous mice is described in reference 60,
bit BCG lesions during the first 5 days. Multi- and the effects of BCG vaccination of guinea
ple mechanisms are probably involved, because pigs on their cytokines are described in refer-
cytokines are enhanced and suppressed by host ences 61 and 62.
factors, as well as by factors from the microor-
ganisms themselves (55). We may never fully CONCLUSIONS
understand the in vivo cytokine network in Our studies suggest that BCG vaccination pro-
tuberculous lesions or, in fact, in any inflamma- tects in the following manner. After the host
tory lesion (see reference 56).We can, however, inhales virulent tubercle bacilli, chemotaxins
visualize (as we have done here) the overall would initially be produced when bacillary anti-
cytokine pattern and thereby gain some insight gens combine with circulating and cytophilic
into which cytokines are participating. antibodies (see chapter 5). These chemotaxins
Chemokine production is influenced in vivo would cause a rapid accumulation of dendritic
by cell-cell, cell-adhesion molecule, and cell- cells, macrophages, and antigen-specific (mem-
extracellular matrix interactions, and by other ory) T lymphocytes at the sites of bacillary lodge-
cytokines (57). For example, cocultures of ment.The infiltrating cells would then produce
monocytes with endothelial cells produce more cytokines, such as MCP-1, IL-1,TNF-, and
IL-8, MCP-1, and macrophage inflammatory IFN-, that enhance cell infiltration and activate
protein-1 than do cultures of either cell type the accumulating macrophages (and lympho-
alone (57). Similarly, monocyte-fibroblast cocul- cytes).After activation, the macrophages would
tures produce more MCP-1 and macrophage kill or inhibit the tubercle bacillus and thereby
inflammatory protein-1 than do cultures of prevent many microscopic tubercles from
either cell type alone (57). Such cell-cell inter- becoming clinically apparent (see chapter 11)
actions can also downregulate the production of (63; http://www.bioscience.org/1998/v3/c/
certain chemokines (57), probably depending on dannenbe/list.htm).

Note that at 3 h in reinfection BCG lesions, a much higher percentage of PMN (this figure) than
MN (Fig. 4) contained IL-1 mRNA and IL-8 mRNA. At their peaks, IL-1 mRNA was 36%
for PMN and 5% for MN, and IL-8 mRNA was 48% for PMN and 8% for MN. Evidently, many
PMN produce cytokines as soon as they enter sites of inflammation.
The means and their standard errors are shown (see reference 3 for P values). Reproduced with
permission from reference 3.
324 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

REFERENCES 13. Nathan, C. 2002. Points of control in inflamma-


1. Tsuruta, J., K. Sugisaki, A. M. Dannenberg, tion. Nature 420:846–852.
Jr.,T.Yoshimura,Y. Abe, and P. Mounts. 1996. 14. Lenardo, M., F. K. Chan, F. Hornung,
The cytokines NAP-1 (IL-8), MCP-1, IL-1 and H. McFarland, R. Siegel, J. Wang, and
GRO in rabbit inflammatory skin lesions produced L. Zheng. 1999. Mature T lymphocyte apoptosis.
by the chemical irritant sulfur mustard. Inflammation Immune regulation in a dynamic and unpredictable
20:293–318. antigenic environment. Annu. Rev. Immunol. 17:221–
2. Sugisaki, K., A. M. Dannenberg, Jr., Y. Abe, 253.
J. Tsuruta, W.-J. Su, W. Said, L. Feng, 15. Green, D. R. 2003. Overview: apoptotic signaling
T.Yoshimura, P. J. Converse, and P. Mounts. pathways in the immune system. Immunol. Rev.
1998. Nonspecific and immune-specific up- 193:5–9.
regulation of cytokines in rabbit dermal tubercu- 16. Zganiacz, A., M. Santosuosso, J. Wang,
lous (BCG) lesions. J. Leukoc. Biol. 63:440–450. T.Yang, L. Chen, M. Anzulovic, S. Alexander,
3. Shigenaga, T., A. M. Dannenberg, Jr., D. B. B. Gicquel,Y.Wan, J. Bramson, M. Inman, and
Lowrie, W. Said, M. J. Urist, H. Abbey, B. H. Z. Xing. 2004.TNF- is a critical negative regu-
Schofield, P. Mounts, and K. Sugisaki. 2001. lator of type 1 immune activation during intra-
Immune responses in tuberculosis: antibodies and cellular bacterial infection. J. Clin. Investig. 113:401–
CD4-CD8 lymphocytes with vascular adhesion 413.
molecules and cytokines (chemokines) cause a rapid 17. Mason, D., and F. Powrie. 1998. Control of
antigen-specific cell infiltration at sites of Bacillus immune pathology by regulatory T cells. Curr. Opin.
Calmette-Guérin reinfection. Immunology 102:466– Immunol. 10:649–655.
479. 18. Read, S., and F. Powrie. 2001. CD4⫹ regulatory
4. Pais, T. F., R. A. Silva, B. Smedegaard, cells. Curr. Opin. Immunol. 13:644–649.
R. Appelberg, and P. Andersen. 1998. Analysis 19. Akbari, O., P. Stock, R. H. DeKruyff, and
of T cells recruited during delayed-type hypersen- D.T. Umetsu. 2003. Role of regulatory T cells in
sitivity to purified protein derivative (PPD) versus allergy and asthma. Curr. Opin. Immunol. 15:627–
challenge with tuberculosis infection. Immunology 633.
95:69–75. 20. Umetsu, D.T., O.Akbari, and R. H. DeKruyff.
5. Lurie, M. B. 1964. Resistance to Tuberculosis: Exper- 2003. Regulatory T cells control the development
imental Studies in Native and Acquired Defensive Mech- of allergic disease and asthma. J. Allergy Clin.
anisms. Harvard University Press, Cambridge, Mass. Immunol. 112:480–487.
6. Vogt, R. F., Jr., N. A. Hynes, A. M. Dannen- 21. Fisson, S., G. Darrasse-Jèze, E. Litvinova,
berg, Jr., S. Castracane, and L. Weiss. 1983. F. Septier, D. Klazmann, R. Liblau, and B. L.
Improved techniques using Giemsa-stained glycol Salomon. 2003. Continuous activation of autore-
methacrylate tissue sections to quantitate basophils active CD4⫹CD25⫹ regulatory T cells in the steady
and other leukocytes in inflammatory skin lesions. state. J. Exp. Med. 198:737–746.
Stain Technol. 58:193–205. 22. Mittrücker, H.-W., and S. H. E. Kaufmann.
7. Kupper, T. S. 1990. Immune and inflammatory 2004. Regulatory T cells and infection: suppression
processes in cutaneous tissues: mechanisms and revisited. Eur. J. Immunol. 34:306–312.
speculations. J. Clin. Investig. 86:1783–1789. 23. Shevach, E. M. 2004. Regulatory/suppressor
8. Luger,T. A., and T. Schwarz. 1995.The role of T cells in health and disease. Arthritis Rheum.
cytokines and neuroendocrine hormones in cuta- 50:2721–2724.
neous immunity and inflammation. Allergy 50:292– 24. Piccirillo, A. C., and E. M. Shevach. 2004.
302. Naturally-occurring CD4⫹CD25⫹ immuno-
9. Kupper, T. S., and R. W. Groves. 1995. The regulatory T cells: central players in the arena of
interleukin-1 axis and cutaneous inflammation. peripheral tolerance. Semin. Immunol. 16:81–88.
J. Investig. Dermatol. 105:62S–66S. 25. Belkaid,Y., and B.T. Rouse. 2005. Natural reg-
10. Vaddi, K., M. Keller, and R. C. Newton. 1997. ulatory T cells in infectious disease. Nat. Immunol.
The Chemokine FactsBook.Academic Press, Inc., San 6:353–360.
Diego, Calif. 26. Shreedhar, V. K., M. W. Pride, Y. Sun, M. L.
11. Suga, M., A. M. Dannenberg, Jr., and Kripke, and F. M. Strickland. 1998. Origin and
S. Higuchi. 1980. Macrophage functional hetero- characteristics of ultraviolet-B radiation-induced
geneity in vivo: macrolocal and microlocal macro- suppressor T lymphocytes. J. Immunol. 161:1327–
phage activation, identified by double-staining tissue 1335.
sections of BCG granulomas for pairs of enzymes. 27. Sato, K., N.Yamashita, M. Baba, and T. Mat-
Am. J. Pathol. 99:305–324. suyama. 2003. Modified myeloid dendritic cells act
12. Nathan, C. F. 1987. Secretory products of macro- as regulatory dendritic cells to induce anergic and
phages. J. Clin. Investig. 79:319–326. regulatory T cells. Blood 101:3581–3589.
20. CYTOKINE PRODUCTION IN REINFECTION BCG LESIONS 䡵 325

28. Wakkach, A., N. Fournier, V. Brun, J.-P. 39. Arend,W. P., M. Malyak, C. J. Guthridge, and
Breittmayer, F. Cottrez, and H. Groux. 2003. C. Gabay. 1998. Interleukin 1 receptor antagonist:
Characterization of dendritic cells that induce tol- role in biology. Annu. Rev. Immunol. 16:27–55.
erance and T regulatory-1 cell differentiation in 40. Fadok,V. A., D. L. Bratton, A. Konowal, P.W.
vivo. Immunity 18:605–617. Freed, J.Y.Westcott, and P. M. Henson. 1998.
29. Mellor, A. L., and D. H. Munn. 2004. IDO Macrophages that have ingested apoptotic cells in
expression by dendritic cells: tolerance and trypto- vitro inhibit proinflammatory cytokine production
phan catabolism. Nat. Rev. Immunol. 4:762–774. through autocrine/paracrine mechanisms involving
30. Kuchroo,V. K., D.T. Umetsu, R. H. DeKruyff, TGF-, PGE2, and PAF. J. Clin. Investig. 101:890–
and G. J. Freeman. 2003.The TIM gene family: 898.
emerging roles in immunity and disease. Nat. Rev. 41. Conti, P., D. Kempuraj, K. Kandere, M. di
Immunol. 3:454–462. Gioacchino, R. C. Barbacane, M. L. Castellani,
31. Sabatos, C. A., S. Chakravarti, E. Cha, M. Felaco, W. Boucher, R. Letourneau, and
A. Schubart,A. Sánchez-Fueyo, X. X. Zheng, T. C.Theoharides. 2003. IL-10, an inflammatory/
A. J. Coyle, T. B. Strom, G. J. Freeman, and inhibitory cytokine, but not always. Immunol. Lett.
V. K. Kuchroo. 2003. Interaction of Tim-3 and 86:123–129.
Tim-3 ligand regulates T helper type 1 responses 42. Barton, B. E. 1997. IL-6: insights into novel bio-
and induction of peripheral tolerance. Nat. Immunol. logical activities. Clin. Immunol. Immunopathology
4:1102–1110. 85:16–20.
32. Meyers, J. H., S. Chakravarti, D. Schlesinger, Z. 42a.Serhan, C. N., and J. M. Drazen. 1997. Antiin-
Illes, H. Waldner, S. E. Umetsu, J. Kenny, flammatory potential of lipoxygenase-derived
X. X. Zheng, D. T. Umetsu, R. H. DeKruyff, eicosanoids: a molecular switch at 5 and 15 posi-
T. B. Strom, and V. K. Kuchroo. 2005.TIM-4 is tions? J. Clin. Invest. 99:1147–1148. (Editorial.)
the ligand for TIM-1, and the TIM-1—TIM-4 inter- 43. Ferrante, J. V., Z. H. Huang, M. Nandoskar,
action regulates T-cell proliferation. Nat. Immunol. C. S. T. Hii, B. S. Robinson, D. A. Rathjen,
6:455–464. A. Poulos, C. P. Morris, and A. Ferrante. 1997.
Altered responses of human macrophages to
33. Mantovani, A., P. Allavena, A. Vecchi, and S.
lipopolysaccharide by hydroperoxy eicosatetraenoic
Sozzani. 1998. Chemokines and chemokine
acid, hydroxy eicosatetraenoic acid, and arachi-
receptors during activation and deactivation of
donic acid: inhibition of tumor necrosis factor pro-
monocytes and dendritic cells and in amplifica-
duction. J. Clin. Investig. 99:1445–1452.
tion of Th1 versus Th2 responses. Int. J. Clin. Lab.
44. Peng, H.-B., M. Spiecker, and J. K. Liao. 1998.
Res. 28:77–82.
Inducible nitric oxide: an autoregulatory feedback
34. Penton-Rol, G., N. Polentarutti, W. Luini, inhibitor of vascular inflammation. J. Immunol.
A. Borsatti, R. Mancinelli,A. Sica, S. Sozzani, 161:1970–1976.
and A. Mantovani. 1998. Selective inhibition of 45. Taylor-Robinson, A. W. 1997. Inhibition of
expression of the chemokine receptor CCR2 in IL-2 production by nitric oxide: a novel self-
human monocytes by IFN-. J. Immunol. 160:3869– regulatory mechanism for Th1 cell proliferation.
3873. Immunol. Cell Biol. 75:167–175.
35. Fantuzzi, L., P. Borghi,V. Ciolli, G. Pavlakis, 46. Brüne, B. 2003. Nitric oxide: NO apoptosis or
F. Belardelli, and S. Gessani. 1999. Loss of turning it ON? Cell Death Differ. 10:864–869.
CCR2 expression and functional response to 47. Zidek, Z. 1999.Adenosine–cyclic AMP pathways
monocyte chemotactic protein (MCP-1) during the and cytokine expression. Eur. Cytokine Netw.
differentiation of human monocytes: role of secreted 10:319–328.
MCP-1 in the regulation of the chemotactic 48. La Sala,A., D. Ferrari, F. Di Virgilio, M. Idzko,
response. Blood 94:875–883. J. Norgauer, and G. Girolomoni. 2003. Alert-
36. Heaney, M. L., and D. W. Golde. 1998. Soluble ing and tuning the immune response by extracel-
receptors in human disease. J. Leukoc. Biol. 4:135– lular nucleotides. J. Leukoc. Biol. 73:339–343.
146. 49. Bergquist, J., A. Tarkowski, A. Ewing, and
37. Mantovani, A., R. Bonecchi, F. O. Martinez, R. Ekman. 1998. Catecholaminergic suppression
E. Galliera, P. Perrier, P. Allavena, and of immunocompetent cells. Immunol.Today 19:562–
M. Locati. 2003.Tuning of innate immunity and 567.
polarized responses by decoy receptors. Int. Arch. 50. Clynes, R., J. S. Maizes, R. Guinamard,
Allergy Immunol. 132:109–115. M. Ono,T.Takai, and J.V. Ravetch. 1999. Mod-
38. Takahashi, S., Y. Setoguchi, T. Nukiwa, and ulation of immune complex-induced inflamma-
S. Kira. 1991. Soluble interleukin-2 receptor in sera tion in vivo by the coordinated expression of acti-
of patients with pulmonary tuberculosis. Chest vation and inhibitory Fc receptors. J. Exp. Med.
99:310–314. 189:179–185.
326 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

51. Euzger, H. S., R. J. Flower, N. J. Goulding, and mines function of macrophages and renders them
M. Perritti. 1999. Differential modulation of unresponsive to other cytokines. J. Immunol. 161:
annexin I binding sites on monocytes and neu- 1983–1988.
trophils. Mediators Inflamm. 8:53–62. 59. Hasan, M., S. Najjam, M. Y. Gordon, R. V.
52. Kubo, M.,T. Hanada, and A.Yoshimura. 2003. Gibbs, and C. C. Rider. 1999. IL-12 is a heparin-
Suppressors of cytokine signaling and immunity. binding cytokine. J. Immunol. 162:1064–1070.
Nat. Immunol. 4:1169–1176. 60. Mustafa,T., S. Phyu, R. Nilsen, R. Jonsson, and
53. Alexander, W. S. 2002. Suppressors of cytokine G. Bjune. 2000. In situ expression of cytokines and
signalling (SOCS) in the immune system. Nat. Rev. cellular phenotypes in the lungs of mice with slowly
Immunol. 2:1–7. progressive primary tuberculosis. Scand. J. Immunol.
54. Mühl, H., and J. Pfeilschifter. 2003. Anti- 51:548–556.
inflammatory properties of pro-inflammatory inter- 61. Kawahara, M., T. Nakasone, and M. Honda.
feron-. Int. Immunopharmacol. 3:1247–1255. 2002. Dynamics of gamma-interferon, interleukin-
55. Wilson, M., R. Seymour, and B. Henderson. 12 (IL-12), IL-10 and transforming growth factor
1998. Bacterial perturbation of cytokine networks.  mRNA expression in primary Mycobacterium
Infect. Immun. 66:2401–2409. bovis BCG infection in guinea pigs measured by a
56. D’Ambrosio, D., P. Panina-Bordignon, and real-time fluorogenic reverse transcription-PCR
F. Sinigaglia. 2003. Chemokine receptors in assay. Infect. Immun. 70:6614–6620.
inflammation: an overview. J. Immunol. Methods 273: 62. Jeevan, A.,T.Yoshimura, K. E. Lee, and D. N.
3–13. McMurray. 2003. Differential expression of gamma
57. Smith, R. E., C. M. Hogaboam, R. M. Strieter, interferon mRNA induced by attenuated and vir-
N.W. Lukacs, and S. L. Kunkel. 1997. Cell-to- ulent Mycobacterium tuberculosis in guinea pig cells
cell and cell-to-matrix interactions mediate after Mycobacterium bovis BCG vaccination. Infect.
chemokine expression: an important component of Immun. 71:354–364.
the inflammatory lesion. J. Leukoc. Biol. 62:612– 63. Dannenberg, A. M., Jr. 1998. Lurie’s tubercle-
619. count method to test TB vaccine efficacy in rabbits.
58. Erwig, L.-P., D. C. Kluth, G. M. Walsh, and Front. Biosci. 3:c27–33. [Online.] http://www.
A. J. Rees. 1998. Initial cytokine exposure deter- bioscience.org/1998/v3/c/dannenbe/list.htm.
21
VASCULAR ADHESION MOLECULES
IN TUBERCULOUS LESIONS

Identification of microvessels in tissue sections of BCG lesions [329]


Quantitation of vascular adhesion molecules in tissue sections [329]
Vascular adhesion molecules in primary BCG lesions [329]
Vascular ICAM, ELAM, and VCAM in reinfection BCG lesions [331]
Vascular adhesion molecules in acute inflammatory lesions [331]
Leukocyte ligands for adhesion molecules in BCG lesions [333]
Adhesion molecules in epithelioid cells [333]
Overview of ICAM, ELAM, and VCAM functions [333]
How activation of microvascular endothelium may contribute to caseous
necrosis [334]
Questions to be answered [335]

Abstract.Vascular adhesion molecules enable host defense cells to leave the bloodstream
and enter tuberculous lesions.After the inhalation of tubercle bacilli, Lurie’s resistant rab-
bits had a larger number of mononuclear cells within developing lesions than did his sus-
ceptible rabbits.Therefore, the rapid local accumulation of mononuclear cells seems to be
one of the factors associated with resistance to the progress of this disease.Vascular adhe-
sion molecules enable such an accumulation to occur.
With immunohistochemical techniques, we evaluated the rise and fall of three major
vascular adhesion molecules as rabbit dermal BCG lesions developed and healed. ICAM-
1 (intercellular adhesion molecule 1) is important for the adherence of polymorphonu-
clear leukocytes (PMN), monocytes, and lymphocytes to activated vascular endothelium
before they emigrate from the bloodstream into sites of inflammation and infection.
VCAM-1 (vascular cell adhesion molecule 1) is a major factor in monocyte, lymphocyte,
and eosinophil emigration. ELAM-1 (endothelial-leukocyte adhesion molecule 1, now
called E-selectin) aids the emigration of granulocytes (and some monocytes and T lym-
phocytes).
In primary BCG lesions, ICAM and VCAM peaked at 1 to 2 weeks and decreased as
the lesions healed. In reinfection BCG lesions, ICAM and VCAM were upregulated
much sooner, beginning at 3 to 12 h and peaking at 1 to 2 days.The upregulation of these
two adhesion molecules apparently caused the rapid infiltration of mononuclear cells into
sites of BCG reinfection. ELAM-1 seemed to be less involved.
In tuberculosis, epithelioid cells are macrophages that adhere to one another in an epithe-
lial-like pattern.This adherence seems to be due in part to the ICAM-1 of one macrophage’s
binding to its ligand LFA-1 (lymphocyte function-associated antigen 1) (CD11a/CD18)
on a neighboring macrophage.Whether or not this epithelial-like pattern benefits the host
remains to be determined.
From these studies of vascular adhesion molecules, we developed a theory of why so much
tissue destruction occurs in tuberculosis: ICAM,VCAM, and ELAM are markers for acti-
vated vascular endothelial cells. In tuberculous lesions, such activated endothelial cells can
capture and present local mycobacterial antigens and therefore may be killed by antigen-
specific cytotoxic T lymphocytes.When the vascular endothelium is no longer intact, throm-
bosis occurs, and the local tissues (now lacking a blood supply) undergo caseous necrosis.

327
328 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

Vascular adhesion molecules, such as ICAM-1, adhesion molecules (13, 14) and then diapedese
VCAM-1, and ELAM-1 (1–4), enable the dia- into the tissues (5).
pedesis of leukocytes. These adhesion mole- The local vascular endothelium is a major
cules are upregulated in the postcapillary venules participant in the inflammatory process (2–4, 13,
of all inflammatory reactions (5, 6), including 14). It produces cytokines, eicosanoids, nitric
those produced in rabbits by BCG (1). The oxide, vasoactive peptides, procoagulants and
upregulation is produced by the primary anticoagulants, and fibrinolytic factors, all of
cytokines interleukin-1 (IL-1) and tumor which probably play important roles in devel-
necrosis factor alpha (TNF-) (7–10) as well as opment (and regression) of tuberculous lesions.
by chemokines (11, 12).The leukocytes in these This chapter describes the upregulation and
venules upregulate their ligands for these vascular downregulation of three major vascular adhesion

FIGURE 1 Microvessels in a 9-
day rabbit BCG lesion, stained
immunohistochemically for ICAM-
1 (A) and for VCAM-1 (B).Vessels
immunostained for ELAM were
similar in appearance. Mouse mon-
oclonal antibodies to rabbit ICAM-
1 and VCAM-1, rabbit anti-mouse
IgG, counterstained with Giemsa.
Magnification, ⫻475. Reproduced
with permission from reference 1.
21. VASCULAR ADHESION MOLECULES IN TUBERCULOUS LESIONS 䡵 329

molecules in rabbit dermal BCG lesions as they included in these measurements. Stained areas
developed and healed. the size of a single cell were also not included,
because such cells may or may not have been
IDENTIFICATION OF MICROVESSELS part of the microvasculature.
IN TISSUE SECTIONS OF BCG From these measurements, we calculated the
LESIONS area of functional (vW-staining) microvascula-
The size of the BCG lesions as they developed ture per mm2 of lesion in the tissue section, and
and regressed is shown in Fig. 1A. In general, the we calculated the area per mm2 that stained for
larger the lesion, the greater the number of each of the three vascular adhesion molecules
microvessels it will contain. (1).The percentage of the vW-positive microvas-
The total number of microvessels in the lesions culature that stained for each vascular adhesion
is best measured by the gelatin–colloidal car- molecule could then be determined (see below).
bon perfusion method in thick (250-m) tissue
sections (15, 16) (see chapter 8). However, the VASCULAR ADHESION MOLECULES
sections prepared by this method are too thick IN PRIMARY BCG LESIONS
for the evaluation of individual cells, including The size of the BCG lesions as they developed
vascular endothelial cells.We therefore immuno- and healed is shown in Fig. 2A.They peaked in
stained the microvessels for von Willebrand (vW) size at 2 to 3 weeks, when the host became
factor in cryostat-prepared tissue sections 5 m tuberculin positive. Such lesions showed much
thick. (In standard tissue sections, the microvas- cellular infiltration, but little edema. The cells
culature is rarely sectioned lengthwise. The present apparently hydrolyzed the ground sub-
microvessels are usually cut crosswise [Fig. 2].) stance and collagen, so that the microvessels
vW factor is present in almost all functional were not spread apart as they would have been
vascular endothelial cells (17, 18) and therefore if frank edema had been present.
is an excellent marker for the total number of The microvessels that stained for both vW
these microvessels in a tissue section. vW factor factor and adhesion molecules reached an early
is both stored and secreted by endothelial cells peak and then declined as the lesions healed
(17, 18). When secreted, it binds to vascular (Fig. 2B, C, and D). The peaks for ICAM and
basement membrane, where it helps platelets VCAM seemed to be earlier than the peak for
initiate the clotting cascade if overlying endothe- ELAM (Fig. 2C) (21).This increase in microvas-
lium is no longer intact (17, 18). vW factor cir- culature was due partly to an increase in the
culates bound to Factor VIII (19, 20) and also in number of vessels per unit area (see chapter 8)
free form (20a). and partly to thickening (activation) of the ves-
sel endothelium.
QUANTITATION OF VASCULAR A similar pattern was found in the percentage
ADHESION MOLECULES IN TISSUE of vW-positive microvessels that stained for these
SECTIONS three adhesion molecules (Fig. 2D).Therefore,
Vascular endothelial cells in tissue sections of the increase in the adhesion molecules per unit
BCG lesions were stained for vW factor and for area was due not only to greater vascular density
ICAM, ELAM, and VCAM by immunohisto- but also to the upregulation of ICAM, ELAM,
chemical methods (Fig. 1) (1).The areas occu- and VCAM within the functional microvessels.
pied by the stained endothelial cells in the walls A comparison of Fig. 2A with Fig. 2B, C, and
of the microvessels were measured using the D shows that the vascular adhesion molecules
probe of a computerized image analyzer. (The peaked before the number of cells in the BCG
inflammatory process activates microvascular lesions peaked, and they started to decline before
endothelial cells causing them to increase in the number of cells in the BCG lesions declined.
size, so that their area in cross section is easily In other words, the upregulation of vascular
measured with a computerized image analyzer.) adhesion molecules increases BCG lesion size,
The lumens of the microvessels were not and their downregulation decreases lesion size.
330 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS
21. VASCULAR ADHESION MOLECULES IN TUBERCULOUS LESIONS 䡵 331

VASCULAR ICAM, ELAM, AND VCAM ulated in tuberculin reactions of both humans
IN REINFECTION BCG LESIONS (23) and rhesus monkeys (24).
The percentages of vascular adhesion molecules
in primary BCG lesions and those in reinfection VASCULAR ADHESION MOLECULES
BCG lesions and in tuberculin reactions during IN ACUTE INFLAMMATORY LESIONS
the first 5 days are plotted in Fig. 3 (21, 22). We also studied these vascular adhesion molecules
Reinfection BCG lesions showed an ELAM in acute inflammatory lesions produced in rab-
peak at 3 h, an ICAM peak at about 2 days, and bits by placing a drop of 1% sulfur mustard on the
a possible VCAM peak at 5 days.Therefore, in skin (1).These acute lesions reached their peak
reinfection lesions, the upregulation of ELAM size in 1 day and were almost healed in 6 days.
(which is associated with granulocyte emigra- The sulfur mustard lesions showed higher levels
tion) was rapid, and the upregulation of ICAM of ELAM-1 than VCAM-1, whereas the more
and VCAM (which are associated with a chronic BCG lesions showed the reverse (data in
mononuclear cell response) was slower and more reference 1).These findings are consistent with the
prolonged. In primary BCG lesions, the upreg- high numbers of granulocytes found in the crust
ulation of all these adhesion molecules was more of sulfur mustard lesions and the high numbers
delayed (Fig. 3). of mononuclear cells (macrophages and lym-
In tuberculin reactions produced in the rein- phocytes) found in the tuberculous granulation
fected rabbits, the vascular adhesion molecules tissue of BCG lesions. Of interest is that dilated
rose and fell in a pattern similar to that found in lymphatics are easily seen in the acute edematous
their BCG lesions (Fig. 3). The slight differ- sulfur mustard lesions (Fig.4).In the more chronic
ences were probably due to the antigens in the BCG lesions, the lymphatics are not nearly as
tuberculin reactions and the BCG lesions leav- dilated and are hard to recognize (see Fig. 18 in
ing at different rates.Vascular VCAM was upreg- chapter 4).

FIGURE 2 (A) Size (in mm3) of rabbit dermal BCG lesions at various times after their onset.
(B) Area of stained microvasculature in such lesions as a percentage of a 1-mm2 area of tissue sec-
tion.The tissue sections were stained immunohistochemically for von Willebrand (vW) factor (a
measure of the total functional microvasculature) and for the adhesion molecules ICAM-1, ELAM-
1, and VCAM-1 (1) (see text).The microvasculature was evaluated only in the intact areas of the
BCG lesions that were densely infiltrated with inflammatory cells. Normal skin values are shown
at zero time.The means and their standard errors are shown. Note that peak levels of adhesion
molecules preceded the peak size of the BCG lesions, because the upregulation of these mole-
cules enabled the cell infiltration that caused the BCG lesions to grow in size.To produce this
panel, each stained microvessel was circled with the probe of a computerized image analyzer, and
its lumen was also circled.The two circled areas were subtracted to provide the area (in mm2) of
the vessel wall.Then, the areas occupied by vessel walls in 1 mm2 of tissue section were com-
puted. Reproduced with permission from reference 1. (C) Microvasculature stained for ICAM-
1, ELAM-1, and VCAM-1 in BCG lesions as a percentage of a 1-mm2 area of tissue section.This
panel confirms the data in panel B, with important 3-day measurements added. Note that ICAM
and VCAM peaked at 3 days, suggesting that these adhesion molecules enabled the initial cell infil-
tration of mononuclear cells (macrophages and lymphocytes) into the BCG lesions. ELAM upreg-
ulation was more delayed.The means and their standard errors are shown. Reproduced with per-
mission from reference 21. (D) Vasculature in BCG lesions stained for adhesion molecules as a
percentage of the total functional vasculature (vW factor-stained vessels). Presenting the data in
this way identifies the upregulation and downregulation of the adhesion molecules more precisely,
because changes in the total functional vasculature have been factored out.The means and their
standard errors are shown. Reproduced with permission from reference 1.
332 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS
21. VASCULAR ADHESION MOLECULES IN TUBERCULOUS LESIONS 䡵 333

LEUKOCYTE LIGANDS FOR fibroblasts, and epithelial cells, including ker-


ADHESION MOLECULES IN BCG atinocytes (4). ICAM makes these cells adhere
LESIONS to their surroundings and plays an important role
For leukocytes to emigrate from the bloodstream, in the adherence of PMN, monocytes, and lym-
they must contain ligands on their surface for the phocytes to activated vascular endothelium.This
adhesion molecules of the vascular endothelium. adherence is followed by the emigration of these
The leukocyte ligands for ICAM-1 are LFA-1 leukocytes from the circulation into the tissues.
(CD11a/CD18) and Mac-1 (CD11b/CD18) ELAM plays a similar role in the emigration of
(25).We therefore immunostained the cells in the granulocytes, monocytes, and a subpopulation of
BCG lesions with monoclonal antibodies to these T cells (4, 9, 23, 25, 35–41), and VCAM plays a
ligands.Throughout these lesions, mononuclear similar role in monocyte, lymphocyte, and
cells (mostly macrophages) stained for LFA-1. eosinophil emigration (4, 42).
Close to areas of caseous necrosis, granulocytes Details of the mechanisms involved—rolling,
(PMN) stained for Mac-1. adherence, penetration, and migration toward
VCAM is monocyte/lymphocyte selective chemoattractants—are reviewed in references
(26–31) because its ligand,VLA-4, is present on 25, 35, 40, 43, 44, and 45, and the various lym-
monocytes and lymphocytes, but not on PMN. phocyte adhesion molecules are reviewed in
VCAM and VLA-4 are especially important references 13 and 46.
factors involved in the accumulation of lym- In most chronic inflammatory lesions,
phocytes in tissues (28, 32–34). Unfortunately, mononuclear cells predominate over PMN. Our
we were able to immunostain only a few studies suggest that vascular VCAM is a major
mononuclear cells for VLA-4 in BCG lesions, player in this selection (43, 47–49). However, in
apparently because the only available antibody inflammatory processes, interactions between
was for human VLA-4, rather than for rabbit leukocytes, platelets, and endothelial cells are
VLA-4, and did not appreciably cross-react. complex and even reciprocal (47), involving (i)
vascular ICAM, ELAM, VCAM, PADGEM
ADHESION MOLECULES IN
EPITHELIOID CELLS (GMP-140, P-selectin) (50), and others; (ii)
Both ICAM-1 and its ligand, LFA-1, were leukocyte ligands for each of these adhesion
strongly expressed by epithelioid cells of the molecules; (iii) various cytokines, including IL-
BCG granuloma (Fig. 5).This fact suggests that 1 and tumor necrosis factor alpha (51, 52); (iv)
the ICAM–LFA-1 pair plays a major role in chemokines, such as macrophage chemoattrac-
the binding of these mature macrophages to tant (activating) protein-1 and IL-8 (44); (v) his-
one another. Such binding is the cause of their tamine (53) and platelet-activating factor (53);
epithelial-like appearance (1). (vi) thrombin (53, 54), fibrinogen (47), and
fibrin (55); and, finally, (vii) elements of extra-
OVERVIEW OF ICAM, ELAM, AND cellular matrix (56), e.g., fibronectin (57, 57a),
VCAM FUNCTIONS laminin, and collagen. Details on many of these
ICAM-1 is produced by many cells in the body: interactions are reviewed in references 35, 43,
lymphocytes, dendritic cells, endothelial cells, and 56.

FIGURE 3 Areas of microvasculature stained immunohistochemically for von Willebrand (vW)


factor, ICAM-1,VCAM-1, and ELAM-1, as a percentage of 1-mm2 areas of tissue sections of der-
mal BCG lesions during the first 5 days.The functional microvasculature (per mm2) (recognized
by vW staining) and the three adhesion molecules increased much more rapidly in reinfection
lesions (and in tuberculin reactions) than in primary lesions. Most of the functional microvascu-
lature stained for ICAM, but only about half as much stained for VCAM and ELAM (compare
the scales on the y axes). In the primary BCG lesions, ELAM-1 was highest at 5 days, but in the
reinfection BCG lesions, ELAM was highest at 3 h.The means and their standard errors are shown.
Reproduced with permission from reference 22 (where P values can be found).
334 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

FIGURE 4 A dilated lymphatic vessel in a 1-day acute dermal inflammatory lesion produced
in a rabbit by the topical application of 1% sulfur mustard (1).These lesions are markedly ede-
matous at 1 and 2 days, but they are healed in 6 to 10 days. Note the valve in the center of the
lymphatic, the nerve on the upper left of the photograph, and the capillary with three erythro-
cytes below. Numerous leukocytes, especially PMN, are present in the connective tissue. Lym-
phatics remain patent in rabbit inflammatory lesions (74). In fact, extravasated serum proteins in
1-day sulfur mustard lesions are replaced every 8 h (75). Glycol methacrylate-embedded tissue
section (1 to 2 m thick) stained with Giemsa. Magnification, ⫻475.

Overviews of the various roles of adhesion delayed-type hypersensitivity reactions to the


molecules in inflammatory processes have been tuberculin-like products of the bacilli are a
published (1, 2, 4, 58–61).The roles of rabbit vas- major cause of such necrosis (70, 71). If this
cular adhesion molecules in arteriosclerosis and immune reaction is directed only against
in cardiac allograph rejection are presented in macrophages containing bacilli (see references 70
references 62, 63, 64, and 65. 71), why is there so much damage to adjacent
A rather complete list of animal models in tissue? The answer seems to be injury to nearby
which anti-adhesion therapy in vivo decreased vascular endothelial cells, followed by clot for-
granulocyte or mononuclear cell infiltration mation (see reference 15), ischemia, and local
into inflammatory sites appears in reference 35. necrosis.
The sequence of events would be as follows.
Vascular endothelial cells are activated by the
HOW ACTIVATION OF
MICROVASCULAR ENDOTHELIUM inflammatory process. Since activated endothe-
MAY CONTRIBUTE TO CASEOUS lial cells upregulate MHC class I and MHC
NECROSIS class II (2, 66–69), they could present the tuber-
Vascular endothelium is activated in both acute culin-like antigens released by nearby tubercle
and chronic inflammatory reactions. Such acti- bacilli (1, 72). Cytotoxic T lymphocytes could
vation upregulates various adhesion molecules then kill these endothelial cells, initiating the
and causes the endothelial cells to produce var- clotting cascade. Local thrombosis would follow,
ious cytokines and express major histocompat- causing ischemia and death of the surrounding
ibility complex class I (MHC-I) and MHC-II tissues (see reference 15).
molecules (2, 66–69). The dead endothelial cells cause blood clot-
In humans, rabbits, and guinea pigs, well- ting and subsequent thrombosis by two mech-
developed tuberculous lesions have solid caseous anisms (73): (i) they no longer produce heparin-
or liquefied necrotic centers.Tissue-damaging like glycoproteins that prevent platelet adherence,
21. VASCULAR ADHESION MOLECULES IN TUBERCULOUS LESIONS 䡵 335

FIGURE 5 (A and B) Both pho-


tographs show a group of rather
mature epithelioid cells (macro-
phages) surrounding a small necrotic
area in a 37-day BCG lesion. The
cells in panel A were immunostained
for ICAM-1, whereas those in panel
B were immunostained for CD11a,
which is a component of LFA-1, the
ligand on macrophages that binds to
ICAM-1.These findings suggest that
the ICAM-1–LFA-1 pair is a major
contributor to the rather firm cell-cell
adherence that characterizes epithe-
lioid cells. Mouse monoclonal anti-
body to rabbit ICAM and CD11a,
biotinylated rabbit anti-mouse IgG,
counterstained with methyl green.
Magnification, ⫻200. Reproduced
with permission from reference 1.

and (ii) the collagen in the vascular basement QUESTIONS TO BE ANSWERED


membrane is no longer covered by endothe- This theory on the role of vascular endothelial
lium, so platelets become activated, and throm- cells in the tissue necrosis found in tuberculo-
bin is formed from prothrombin. sis raises several questions that need to be
In tuberculous lesions, it has always been puz- answered experimentally. (i) Do activated vas-
zling why cytotoxic reactions directed against cular endothelial cells capture and present local
bacilli-laden macrophages cause so much necro- mycobacterial antigens? (ii) Are the concentra-
sis of nearby tissues (see references 70 and 71). tions of tuberculin-like antigens on vascular
The killing of activated endothelial cells (con- endothelial cells high enough to be recognized
taining MHC-associated bacillary antigens) by by cytotoxic T cells? (iii) Does the death of
cytotoxic T cells and the resulting thrombosis these cells result in a change in the endothelial
could explain much of the tissue necrosis. lining so that thrombosis occurs? Endothelial
336 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

cells may just undergo apoptosis, and the gap in 12. Bacon, K. B., and T. J. Schall. 1996. Chemokines
the endothelial lining may be quickly repaired. as mediators of allergic inflammation. Int. Arch.
Allergy Immunol. 109:97–109.
(iv) Is antigen-induced endothelial damage the
13. Springer, T. A. 1990. Adhesion receptors of the
main cause of the extensive tissue necrosis found immune system. Nature 346:425–434.
in many cases of progressive human tuberculo- 14. Wawryk, S. O., J. R. Novotny, I. P. Wicks,
sis, or are other mechanisms operating? D.Wilkinson, D. Maher, E. Salvaris, K.Welch,
The study of vascular endothelial cell activa- J. Fecondo, and A. W. Boyd. 1989.The role of
tion must be more extensively pursued before LFA-1/ICAM-1 interaction in human leukocyte
homing and adhesion. Immunol. Rev. 108:135–161.
we can fully understand the role of endothelial
15. Courtade, E. T., T. Tsuda, C. R. Thomas, and
cells in the pathogenesis of tuberculosis. A. M. Dannenberg, Jr. 1975. Capillary density in
developing and healing tuberculous lesions pro-
duced by BCG in rabbits.A quantitative study. Am.
REFERENCES J. Pathol. 78:243–260.
1. Abe,Y., K. Sugisaki, and A. M. Dannenberg, 16. Eddy, H. A., and G.W. Casarett. 1968. Intestinal
Jr. 1996. Rabbit vascular endothelial adhesion vascular changes in the acute radiation intestinal
molecules: ELAM-1 is most elevated in acute syndrome, p. 385–395. In M. F. Sullivan (ed.), Mono-
inflammation, whereas VCAM-1 and ICAM-1 pre- graphs on Nuclear Medicine and Biology, no. 1. Gas-
dominate in chronic inflammation. J. Leukoc. Biol. trointestinal Radiation Injury. Excerpta Medica Foun-
60:692–703. dation,Amsterdam,The Netherlands.
2. Cotran, R. S., and T. Mayadas-Norton. 1998. 17. Wagner, D. D., and R. Bonfanti. 1991. von
Endothelial adhesion molecules in health and dis- Willebrand factor and the endothelium. Mayo Clin.
ease. Pathol. Biol. (Paris) 46:164–170. Proc. 66:621–627.
3. Pober, J. S., and R. S. Cotran. 1990.The role of 18. Jaffe, E. A. 1987. Cell biology of endothelial cells.
endothelial cells in inflammation. Transplantation Hum. Pathol. 18:234–239.
50:537–544. 19. Lollar, P., D. C. Hill-Eubanks, and C. G.
4. Pigott, R., and C. Power. 1993. The Adhesion Parker. 1988. Association of the Factor VIII light
Molecule Facts Book.Academic Press, Inc., San Diego, chain with von Willebrand factor. J. Biol. Chem.
Calif. 263:10451–10455.
5. Majno, G., and I. Joris. 2004. Cells,Tissues, and Dis- 20. Blann, A. D. 1993. Is raised von Willebrand fac-
ease: Principles of General Pathology, 2nd ed., p. 403– tor a marker of endothelial cell damage? Med.
428. Oxford University Press, New York, N.Y. Hypotheses 41:419–424.
6. Norris, P., R. N. Poston, D. S. Thomas, 20a.Blann, A. D. 1993. von Willebrand factor as a
M. Thornhill, J. Hawk, and D. O. Haskard. marker of injury to the endothelium in inflamma-
1991.The expression of endothelial leukocyte adhe- tory vascular disease. J. Rheumatol. 20:1469–1471.
sion molecule-1 (ELAM-1), intercellular adhesion (Editorial.)
molecule-1 (ICAM-1), and vascular cell adhesion 21. Sugisaki, K., A. M. Dannenberg, Jr., Y. Abe,
molecule-1 (VCAM-1) in experimental cutaneous J. Tsuruta, W.-J. Su, W. Said, L. Feng,
inflammation: a comparison of ultraviolet B ery- T.Yoshimura, P. J. Converse, and P. Mounts.
thema and delayed hypersensitivity. J. Investig. Der- 1998. Nonspecific and immune-specific up-
matol. 96:763–770. regulation of cytokines in rabbit dermal tubercu-
7. Kupper, T. S. 1990. Immune and inflammatory lous (BCG) lesions. J. Leukoc. Biol. 63:440–450.
processes in cutaneous tissues: mechanisms and 22. Shigenaga, T., A. M. Dannenberg, Jr., D. B.
speculations. J. Clin. Investig. 86:1783–1789. Lowrie, W. Said, M. J. Urist, H. Abbey, B. H.
8. Tsuruta, J., K. Sugisaki, A. M. Dannenberg, Schofield, P. Mounts, and K. Sugisaki. 2001.
Jr.,T.Yoshimura,Y. Abe, and P. Mounts. 1996. Immune responses in tuberculosis: antibodies and
The cytokines NAP-1 (IL-8), MCP-1, IL-1, and CD4-CD8 lymphocytes with vascular adhesion
GRO in rabbit inflammatory skin lesions produced molecules and cytokines (chemokines) cause a rapid
by the chemical irritant sulfur mustard. Inflammation antigen-specific cell infiltration at sites of bacillus
20:293–318. Calmette-Guérin reinfection.Immunology 102:466–
9. Bevilacqua, M. P., and R. M. Nelson. 1993. 479.
Selectins. J. Clin. Investig. 91:379–387. 23. Norris, P., R. N. Poston, D. S. Thomas,
10. McEver, R. P. 1994. Selectins. Curr. Opin. Immunol. M. Thornhill, J. Hawk, and D. O. Haskard.
6:75–84. 1991.The expression of endothelial leukocyte adhe-
11. Zack, O. M.,A. Ben-Baruch, and J. J. Oppen- sion molecule-1 (ELAM-1), intercellular adhesion
heim. 1996. Chemokines: progress toward identi- molecule-1 (ICAM-1), and vascular cell adhesion
fying molecular targets for therapeutic agents. Trends molecule-1 (VCAM-1) in experimental cutaneous
Biotechnol. 14:46–51. inflammation: a comparison of ultraviolet B ery-
21. VASCULAR ADHESION MOLECULES IN TUBERCULOUS LESIONS 䡵 337

thema and delayed hypersensitivity. J. Investig. Der- 36. Adams, D. H., and S. Shaw. 1994. Leucocyte-
matol. 96:763–770. endothelial interactions and regulation of leucocyte
24. Silber, A., W. Newman, K. A. Reimann, migration. Lancet 343:831–836.
E. Hendricks, D. Walsh, and D. J. Ringler. 37. Olofsson, A. M., K.-E. Arfors, L. Ramezani,
1994. Kinetic expression of endothelial adhesion B. A. Wolitzky, E. C. Butcher, and U. H. von
molecules and relationship to leukocyte recruitment Andrian. 1994. E-selectin mediates leukocyte
in two cutaneous models of inflammation. Lab. rolling interleukin-1-treated rabbit mesentery
Investig. 70:163–175. venules. Blood 84:2749–2758.
25. Schleiffenbaum, B., and J. Fehr. 1996. Regu- 38. Munro, J. M., J. S. Pober, and R. S. Cotran.
lation and selectivity of leukocyte emigration. J. Lab. 1991. Recruitment of neutrophils in the local endo-
Clin. Med. 127:151–168. toxin response: association with de novo endothe-
26. Chuluyan, H. E., L. Osborn, R. Lobb, and lial expression of endothelial leukocyte adhesion
A. C. Issekutz. 1995. Domains 1 and 4 of vascu- molecule-1. Lab. Investig. 64:295–299.
lar cell adhesion molecule-1 (CD106) both support 39. Butcher, E. C. 1991. Leukocyte-endothelial cell
very late activation antigen-4 (CD49d/CD29)- recognition: three (or more) steps to specificity and
dependent monocyte transendothelial migration. diversity. Cell 67:1033–1036.
J. Immunol. 155:3135–3144. 40. Luscinskas, F. W., G. S. Kansas, H. Ding,
27. Rice, G. E., J. M. Munro, and M. P. Bevilac- P. Pizcueta, B. E. Schleiffenbaum,T. F.Tedder,
qua. 1990. Inducible cell adhesion molecule 110 and M.A. Gimbrone, Jr. 1994. Monocyte rolling,
(INCAM-110) is an endothelial receptor for lym- arrest and spreading on IL-4-activated vascular
phocytes. A CD11/CD18-independent adhesion endothelium under flow is mediated via sequential
mechanism. J. Exp. Med. 171:1369–1374. action of L-selectin, 1-integrins, and 2-integrins.
28. Carlos, T. M., B. R. Schwartz, N. L. Kovach, J. Cell Biol. 125:1417–1427.
41. Picker, L. J., T. K. Kishimoto, C. W. Smith,
E. Yee, M. Rosso, L. Osborn, G. Chi-Rosso,
R. A. Warnock, and E. C. Butcher. 1991.
B. Newman, R. Lobb, and J. M. Harlan. 1990.
ELAM-1 is an adhesion molecule for skin-homing
Vascular cell adhesion molecule-1 mediates lym-
T cells. Nature 349:796–799.
phocyte adherence to cytokine-activated cultured
42. Schleimer, R. P., M. Ebisawa, S. N. Georas,
human endothelial cells. Blood 76:965–970. and B. S. Bochner. 1994. The role of adhesion
29. Kuijpers,T.W., and J. M. Harlan. 1993. Mono- molecules and cytokines in eosinophil recruitment,
cyte-endothelial interactions: insights and ques- p. 99–114. In G. J. Gleich and A. B. Kay (ed.),
tions. J. Lab. Clin. Med. 122:641–651. Eosinophils in Allergy and Inflammation. Marcel
30. Chuluyan, H. E., and A. C. Issekutz. 1993. Dekker, Inc., New York, N.Y.
VLA-4 integrin can mediate CD11/CD18-inde- 43. Sharar, S. R., R. K. Winn, and J. M. Harlan.
pendent transendothelial migration of human 1995.The adhesion cascade and anti-adhesion ther-
monocytes. J. Clin. Investig. 92:2768–2777. apy: an overview. Springer Semin. Immunopathol.
31. Verdegaal, E. M. E., H. Beekhuizen, I. Blok- 16:359–378.
land, and R. van Furth. 1993. Increased adhesion 44. Lukacs, N. W., R. M. Strieter, V. Elner, H. L.
of human monocytes to IL-4-stimulated human Evanoff, M. D. Burdick, and S. L. Kunkel.
venous endothelial cells via CD11/CD18, and very 1995. Production of chemokines, interleukin-8 and
late antigen-4 (VLA-4)/vascular cell adhesion mol- monocyte chemoattractant protein-1, during
ecule-1 (VCAM-1)-dependent mechanisms. Clin. monocyte:endothelial cell interactions. Blood
Exp. Immunol. 93:292–298. 86:2767–2773.
32. Silber, A., W. Newman, V. G. Sasseville, 45. Albelda, S. M., C. W. Smith, and P. A. Ward.
D. Pauley, D. Beall, D. G. Walsh, and D. J. 1994.Adhesion molecules and inflammatory injury.
Ringler. 1995. Recruitment of lymphocytes dur- FASEB J. 8:504–512.
ing cutaneous delayed hypersensitivity in nonhuman 46. Hogg, N., and C. Berlin. 1995. Structure and
primates is dependent on E-selectin and VCAM-1. function of adhesion receptors in leukocyte traf-
J. Clin. Investig. 93:1554–1563. ficking. Immunol.Today 16:327–330.
33. Alon, R., P. D. Kassner, M.W. Carr, E. B. Fin- 47. Duperray, A., A. Mantovani, M. Introna, and
ger, M. E. Hemler, and T. A. Springer. 1995. E. Dejana. 1995. Endothelial cell regulation of
The integrin VLA-4 supports tethering and rolling leukocyte infiltration in inflammatory tissues.
in flow on VCAM-1. J. Cell Biol. 128:1243–1253. Mediat. Inflamm. 4:322–330.
34. Issekutz, T. B., and A. C. Issekutz. 1991. 48. Chuluyan, H. E., and A. C. Issekutz. 1995.
T lymphocyte migration to arthritic joints and Alpha 4-integrin-dependent emigration of mono-
dermal inflammation in the rat: differing migration cytes. Springer Semin. Immunopathol. 16:391–404.
patterns and the involvement of VLA-4. Clin. 49. Issekutz,A. C., H. E. Chuluyan, and N. Lopes.
Immunol. Immunopathol. 61:436–447. 1995. CD11/CD18-independent transendothelial
35. Carlos,T. M., and J. M. Harlan. 1994. Leukocyte- migration of human polymorphonuclear leuko-
endothelial adhesion molecules. Blood 84:2068– cytes and monocytes: involvement of distinct and
2101. unique mechanisms. J. Leukoc. Biol. 57:553–561.
338 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

50. Murphy, J. F., and J. L. McGregor. 1994.Two ing acute rejection of rabbit cardiac allografts. Am.
sites on P-selectin (the lectin and epidermal growth J. Pathol. 144:938–951.
factor-like domains) are involved in the adhesion of 65. Molossi, S., M. Elices, T. Arrhenius, R. Diaz,
monocytes to thrombin-activated endothelial cells. C. Coulber, and M. Rabinovitch. 1995. Block-
Biochem. J. 303:619–624. ade of very late antigen-4 integrin binding to
51. Meng, H., M. G. Tonnesen, M. J. Marchese, fibronectin with connecting segment-1 peptide
R. A. F. Clark,W. F. Bahou, and B. L. Gruber. reduces accelerated coronary arteriopathy in rabbit
1995. Mast cells are potent regulators of endothe- cardiac allografts. J. Clin. Investig. 95:2601–2610.
lial cell adhesion molecule ICAM-1 and VCAM-1 66. Pober, J. S., M.A. Gimbrone, Jr., R. S. Cotran,
expression. J. Cell. Physiol. 165:40–53. C. S. Reiss, S. J. Burakoff,W. Fiers, and K. A.
52. Pugin, J., R. J. Ulevitch, and P. S.Tobias. 1995. Ault. 1983. Ia expression by vascular endothelium
Tumor necrosis factor- and interleukin-1 medi- is inducible by activated T cells and by human
ate human endothelial cell activation in blood at low interferon gamma. J. Exp. Med. 157:1339–1353.
endotoxin concentrations. J. Inflamm. 45:49–55. 67. Collins, T., A. J. Korman, C. T. Wake, J. M.
53. McEver, R. P. 1991. Selectins: novel receptors Boss, D. J. Kappes,W. Fiers, K. A. Ault, M. A.
that mediate leukocyte adhesion during inflam- Gimbrone, Jr., J. L. Strominger, and J. S.
mation. Thromb. Haemost. 65:223–228. Pober. 1984. Immune interferon activates multiple
54. Garcia, J. G. N., F. M. Pavalko, and C. E. Pat- class II major histocompatibility complex genes
terson. 1995.Vascular endothelial cell activation and and the associated invariant chain gene in human
permeability responses to thrombin. Blood Coagul. endothelial cells and dermal fibroblasts. Proc. Natl.
Fibrinolysis 6:609–626. Acad. Sci. USA 81:4917–4921.
55. Qi, J., and D. L. Kreutzer. 1995. Fibrin activa- 68. Pober, J. S., T. Collins, M. A. Gimbrone, Jr.,
tion of vascular endothelial cells: induction of P. Libby, and C. S. Reiss. 1986. Overview:
IL-8 expression. J. Immunol. 155:867–876. inducible expression of class II major histocompat-
56. Nathan, C., and M. Sporn. 1991. Cytokines in ibility complex antigens and the immunogenicity of
context. J. Cell Biol. 113:981–986. vascular endothelium. Transplantation 41:141–146.
57. Kuijpers,T.W. 1995. Pathophysiological aspects of
69. Turner, R. R., J. H. Beckstead, R. A.Warnke,
VLA-4 interactions and possibilities for therapeu-
and G. S.Wood. 1987. Endothelial cell phenotypic
tical interventions. Springer Semin. Immunopathol.
diversity: in situ demonstration of immunologic
16:379–389.
and enzymatic heterogeneity that correlates with
57a.Buck, C. A. 1995.What’s potentially more effec-
specific morphologic subtypes. Am. J. Clin. Pathol.
tive than a Roto-Rooter and less toxic than Drano?
87:569–575.
Anti-adhesions mimetics! J. Clin. Invest. 95:2431–
2432. 70. Dannenberg,A. M., Jr. 1993. Pathogenesis of pul-
58. Hamman, A. (ed.). 1997. Adhesion Molecules and monary tuberculosis. Hosp. Pract. 28:33–40 (Off.
Chemokines in Lymphocyte Trafficking. Harwood Aca- ed. 51–58).
demic Publishers,Amsterdam,The Netherlands. 71. Dannenberg, A. M., Jr., and G. A. W. Rook.
59. Pober, J. S., and R. S. Cotran. 1991. Immuno- 1994. Pathogenesis of pulmonary tuberculosis: an
logic interactions of T lymphocytes with vascular interplay of tissue-damaging and macrophage-
endothelium. Adv. Immunol. 50:261–302. activating immune responses—dual mechanisms
60. Smith, R. E., C. M. Hogaboam, R. M. Strieter, that control bacillary multiplication, p. 459–483. In
N. W. Lukacs, and S. L. Kunkel. 1997. Cell- B. R. Bloom (ed.), Tuberculosis: Pathogenesis, Protec-
to-cell and cell-to-matrix interactions mediate tion, and Control.ASM Press,Washington, D.C.
chemokine expression: an important component of 72. Pasqualini, R., D. M. McDonald, and W.Arap.
the inflammatory lesion. J. Leukoc. Biol. 62:612–619. 2001.Vascular targeting and antigen presentation.
61. Bochner, B. S. 1997. Cellular adhesion and its Nat. Immunol. 2:567–568.
antagonism. J.Allergy Clin. Immunol. 100:581–585. 73. Weiss, H. J. 1975. Platelet physiology and abnor-
62. Cybulsky, M. I., and M.A. Gimbrone, Jr. 1991. malities of platelet function. N. Engl. J. Med.
Endothelial expression of a mononuclear leukocyte 293:531–541.
adhesion molecule during atherogenesis. Science 74. Lurie, M. B. 1964. Resistance to Tuberculosis: Exper-
251:788–791. imental Studies in Native and Acquired Defensive Mech-
63. Richardson, M., S. J. Hadcock, M. DeReske, anisms. Harvard University Press, Cambridge, Mass.
and M. I. Cybulsky. 1994. Increased expression 75. Harada, S., A. M. Dannenberg, Jr., A. Kajiki,
in vivo of VCAM-1 and E-selectin by the aortic K. Higuchi, F. Tanaka, and P. J. Pula. 1985.
endothelium of normolipemic and hyperlipemic Inflammatory mediators and modulators released in
diabetic rabbits. Arterioscler.Thromb. 14:760–769. organ culture from rabbit skin lesions produced in
64. Tanaka, H., G. K. Sukhova, S. J. Swanson, vivo by sulfur mustard. II. Evans blue dye experi-
M. I. Cybulsky, F. J. Schoen, and P. Libby. ments that determined the rates of entry and
1994. Endothelial and smooth muscle cells express turnover of serum protein in developing and heal-
leukocyte adhesion molecules heterogeneously dur- ing lesions. Am. J. Pathol. 121:28–38.
Section 7.

TUBERCULOSIS VACCINES
This page intentionally left blank
22
PRINCIPLES AND GUIDELINES
FOR DEVELOPING BETTER
TUBERCULOSIS VACCINES

Effects of vaccines on the establishment of a microscopic pulmonary


tubercle [342]
Effects of vaccines on the progress of a microscopic pulmonary
tubercle [342]
Effective routes of vaccine administration [343]
Immunization of mice and guinea pigs [343]
Benefits of vaccines containing few, if any, tuberculin-like antigens [344]
Composition of improved vaccines against tuberculosis [345]
Hosts do not need high levels of tuberculin sensitivity to control
the disease [346]
Live tuberculosis vaccines should be standardized before comparing their
relative efficacies [346]
Immunotherapy [347]
Maximal effectiveness of tuberculosis vaccines in humans [347]
Maximal effectiveness of tuberculosis vaccines in rabbits [348]
Other vaccine possibilities [349]
BCG must multiply in the host to be most effective [350]
BCG as a carrier of antigens from other pathogens [350]

Abstract. Tuberculosis vaccines have little or no effect on the establishment of a micro-


scopic pulmonary lesion produced by the inhalation of a virulent tubercle bacillus. Such
a lesion is established only when the pulmonary alveolar macrophages fail to destroy the
inhaled bacillus.Alveolar macrophages do not expand their population in response to spe-
cific antigens. Therefore, the establishment of a microscopic pulmonary tubercle is not
affected by vaccination. Effective tuberculosis vaccines may, however, stop the progression
of a tiny established lesion, because the vaccination has expanded antigen-specific lym-
phocyte populations.These lymphocytes enter the early lesion, where they cause a rapid
local delayed-type hypersensitivity (DTH) and cell-mediated immunity (CMI) response
that often prevents progression of the disease.
When comparing their relative efficacies, two or more live vaccines should be stan-
dardized for equal numbers of live and dead bacilli, equal numbers of log-phase and dor-
mant bacilli, and equal numbers of clumps and isolated bacilli.
Vaccines will probably never be 100% effective in preventing active tuberculosis in
humans, because humans with arrested tuberculous lesions are able to be reinfected with
exogenous tubercle bacilli. However, the efficacy of BCG could be improved by recom-
binant constructs that contain major protective antigens.Also, the immunity provided by
BCG could be increased by booster injections of such antigens.
If vaccines more effective than BCG are ever developed, they would probably produce
in the host a higher CMI/DTH ratio, i.e., an expanded antigen-specific lymphocyte pop-
ulation capable of producing increased numbers of activated macrophages and decreased
amounts of tissue necrosis.To do this, the improved vaccine would probably contain increased
bacillary glycolipid-protein components and decreased tissue-damaging tuberculin-like pro-
tein components.The vaccine should also contain components that increase the Th1/Th2
ratio.

341
342 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

EFFECTS OF VACCINES ON distribution—weakly activated ones at the left,


THE ESTABLISHMENT OF A extremely activated ones at the right, and the
MICROSCOPIC PULMONARY majority (highly activated) in the middle. Immu-
TUBERCLE
nization does not change this distribution after
Role of Pulmonary Alveolar the nonspecific effects of the vaccine have sub-
Macrophages sided.
When first inhaled, 1 bacillary unit of 1 to 3
bacilli (which is the only size that can stay sus- EFFECTS OF VACCINES ON THE
pended in the airstream long enough to reach PROGRESS OF A MICROSCOPIC
PULMONARY TUBERCLE
the alveolar spaces) does not contain enough
antigen to stimulate the immune system. In Progress of Early Pulmonary Tubercles
both humans and rabbits, pulmonary alveolar in Nonvaccinated Rabbits
macrophages apparently ingest and destroy most After an inhaled bacillary unit multiplies in a
inhaled virulent human-type tubercle bacilli, weak alveolar macrophage, chemotactic factors
and the tuberculin skin test remains negative.A are produced that attract dendritic cells,
microscopic primary tubercle begins in the lung macrophages, and antigen-specific lymphocytes
only if a phenotypically more virulent bacillary to the site (see chapters 5 and 6). In nonvacci-
unit is ingested by a relatively weak alveolar nated rabbits challenged with virulent human-
macrophage (1, 2) (see chapters 2 and 11). type tubercle bacilli, these host defense cells
In humans and rabbits, the alveolar macro- prevent most of the microscopic lesions from
phage population is highly activated (non- reaching grossly visible size.Those lesions that
specifically) and therefore is capable of destroy- do reach visible size form the basis of Lurie’s
ing many inhaled human-type tubercle bacilli tubercle count method for assessing vaccine
before they multiply (see chapter 11).This con- efficacy (see chapter 11).
clusion is supported in rabbits by quantitative
airborne infection (3) and in humans by the Progress of Early Pulmonary Tubercles
absence of conversion of the tuberculin skin in Vaccinated Rabbits
test in many persons (including nurses) living in The sequence just described for nonvaccinated
an environment containing low numbers of air- rabbits also occurs in vaccinated rabbits, but in
borne tubercle bacilli. However, the average the vaccinated group the number of recirculat-
number of inhaled virulent tubercle bacilli ing antigen-specific lymphocytes has been
required to convert the tuberculin skin test in highly expanded.Therefore, the local immune
humans is not known.This number could not response is greatly accelerated, i.e., macrophages
be determined in Riley’s studies of a hospital and antigen-specific lymphocytes rapidly accu-
ward containing tuberculous patients (4) (where mulate (see chapter 20).The macrophages soon
the concentration of tubercle bacilli in the air activate and keep many microscopic lesions
was calculated), because the exposure times of from reaching grossly visible size (8–10).A good
personnel entering and leaving the ward was not vaccine will prevent over 80% of the primary
known (see chapter 12). tubercles from becoming visible at necropsy 5
Tuberculosis vaccines cannot prevent the weeks after aerosol challenge (9, 11).
establishment of an infection with the tubercle
bacillus, because alveolar macrophages (a highly Progress of Early Pulmonary Tubercles
activated population) show no immunologic in Humans
specificity. Alveolar macrophages readily ingest Humans are much more sensitive than rabbits to
tubercle bacilli nonspecifically (via mannose the tuberculin-like products of tubercle bacilli.
and other receptors [5–7]) and do not require Therefore, each microscopic pulmonary tuber-
opsonization of these bacilli by antibodies. cle that converts the tuberculin reaction in non-
In rabbits and humans, the alveolar macro- vaccinated persons soon forms a tiny caseous
phage population shows a bell-shaped activation center. However, in 95% of cases, these early
22. DEVELOPING BETTER TUBERCULOSIS VACCINES 䡵 343

lesions are arrested when they are 0.5 to 2.0 present in (unpasteurized) milk. In neonates,
mm in size, and the person has no clinical evi- the gut is more permeable to antigens, so they
dence of the disease, except for the conversion are absorbed intact more readily than in older
of the tuberculin skin test (12–14). These tiny individuals. Also, in all neonates (because of
inapparent lesions often calcify, so that they are their low resistance to mycobacteria), BCG can
detectable at necropsy many years later (12–14). multiply to higher titers. For both reasons, oral
In BCG-vaccinated individuals, such lesions are immunization should be more effective in
arrested sooner and therefore are smaller in size neonates than in older hosts. However, in recent
than those in unvaccinated individuals (12–14) times, infants are immunized with BCG only by
(see chapter 3). the intradermal route. In children and adults,
antigens administered by the oral route may
EFFECTIVE ROUTES OF VACCINE elicit tolerance to these antigens (reviewed in
ADMINISTRATION reference 19).
The most effective route of immunization The intradermal, subcutaneous, and intra-
remains to be determined. No comparisons of muscular routes deliver a large number of viable
oral, intradermal, subcutaneous, intramuscular, BCG (and their antigens) to the draining lymph
intranasal, and aerosol routes of immunization of nodes, where they clonally expand the antigen-
rabbits—followed by aerosol challenge and specific lymphocyte population that recircu-
tubercle counts—have yet been made.Therefore, lates through these nodes.
we can only mention several theoretical possi- The intravenous route delivers the BCG
bilities. throughout the body—especially to the spleen,
The aerosol route can deliver only a relatively which contains much lymphoid tissue.The intra-
small number of live BCG into the lungs,whereas venous route, therefore, should be quite effective
intradermal and parenteral routes can deliver in laboratory animals, but should not be used in
thousands.If the live BCG multiplies extensively human populations because of the greater pos-
in the lungs and throughout the body (as in mice sibility that live BCG will cause minute lesions
[15, 16] and evidently in guinea pigs [17, 18]), in critical sites, such as the eye and brain.
then the route of vaccine administration would
matter little. However, if BCG multiplies rela- IMMUNIZATION OF MICE
tively little (as in rabbits [S.Abramson, discussed AND GUINEA PIGS
by Lurie in 1964; see reference 35, below] and The multiplication of a live vaccine and the
possibly in humans), then the intradermal and degree of its persistence have marked effects on
parenteral routes would be preferable,because the its efficacy (reviewed in reference 21). BCG
dose administered could be so much higher. multiplies extensively in mice (15, 16) and
The intranasal and oral routes deliver antigens apparently in guinea pigs (18), but less so in
in the vaccine to bronchial-associated and gut- rabbits and probably humans, which are more
associated lymphoid tissues (BALT and GALT, resistant species.Yet, to our knowledge, live vac-
respectively). Lymphocytes from BALT and cine multiplication has not been carefully com-
GALT home mainly to mucosal surfaces (19), pared among these species.
and these surfaces are not easily infected by In mice and guinea pigs, the aerosol route of
inhaled tubercle bacilli. However, some anti- BCG immunization was equally effective as
gen-specific lymphocytes from BALT and other routes (and sometimes more effective) in
GALT do not home to mucosal surfaces (19) reducing the multiplication of inhaled virulent
and should be able to enter early tubercles in the human-type bacilli in the lungs (reviewed in ref-
peripheral lung. erences 15 and 18). In mice (15, 16) and prob-
In the 1930s, infants were immunized with ably in guinea pigs (18), inhaled BCG multiplies
BCG by the oral route during the first 10 days readily, apparently because their alveolar
of life (20).The oral route was chosen because macrophages cannot inhibit inhaled BCG as
bovine-type tubercle bacilli were sometimes easily as can rabbit alveolar macrophages.
344 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

In mice, aerosolized BCG persisted in the persist, and (ii) the susceptibility of each species
lungs for at least 12 weeks in relatively high to the virulent tubercle bacilli used to assess
titers (15, 16). Such aerosol-immunized mice the degree of immunization: the more virulent
reduced the growth of subsequently inhaled vir- the challenge strain of tubercle bacillus used for
ulent tubercle bacilli (H37Rv) by about 2 logs such assessment, the less effect the immunization
(15, 18). would appear to have on the disease. There-
In guinea pigs, the aerosol, subcutaneous, and fore, the tubercle-count method (see chapter 11)
intradermal routes of BCG immunization were would measure vaccine efficacy in rabbits more
equally effective in reducing tuberculosis after precisely than it would in mice and guinea pigs.
challenge by aerosol with virulent human-type (Human-type tubercle bacilli are much less vir-
tubercle bacilli (H37Rv)—even though the ulent for rabbits.) On the other hand, if mice and
BCG in the subcutaneous and intradermal guinea pigs were challenged with tubercle bacilli
routes was at much higher dose levels (18). Evi- of reduced virulence, a precision comparable to
dently, aerosolized BCG multiplies extensively that found in rabbits could be achieved.
in the lungs of guinea pigs (see Table 16 in ref-
erence 18). BENEFITS OF VACCINES
In rabbits, aerosolized BCG only rarely pro- CONTAINING FEW, IF ANY,
TUBERCULIN-LIKE ANTIGENS
duced any visible pulmonary lesion (S.Abram-
There are many benefits in reducing or elimi-
son, discussed by Lurie, 1964), apparently because
nating tuberculin-like antigens from BCG vac-
aerosolized BCG did not grow or persist in
cine (Table 1) (2).
titers as high as those in mice and guinea pigs.
In human beings, the efficacy of the aerosol 1. Vaccinated individuals with little or no
route of immunization has never been evaluated. sensitivity to tuberculin would not
However, humans were immunized by aero- become appreciably tuberculin positive.
solized BCG (22, 23), and they sometimes, but Therefore, tuberculin testing of such indi-
not always, converted their tuberculin skin tests viduals would still be useful for detecting
(22, 23). infection with virulent tubercle bacilli.
In brief, vaccine efficacy varies greatly among 2. As with all effective vaccines, such a vac-
laboratory animal species because of differences cine would prevent the development of
in (i) their ability to allow the live attenuated clinically apparent tuberculosis.
tubercle bacilli in the vaccine to multiply and 3. Such a vaccine could be used as
immunotherapy for active tuberculosis,
because it would expand the T-cell pop-
TABLE 1 Benefits of tuberculosis vaccines produc- ulation responding to protective antigens
ing strong CMI and weak DTH, especially those pro-
of the tubercle bacillus and not expand
ducing little or no tuberculin sensitivitya
the T-cell population responding to the
1. Continued usefulness of the tuberculin skin test tuberculin-like antigens that are involved
for diagnosing infection with virulent tubercle in caseous necrosis, liquefaction, and cav-
bacilli
ity formation.
2. Prevention of clinical tuberculosis (prophylaxis)
4. Such a vaccine could be given without
3. Treatment of clinical tuberculosis
(immunotherapy) harm to tuberculin-positive individuals
4. Vaccination of tuberculin-positive individuals with inapparent Mycobacterium tuberculosis
without harm infection to reduce the likelihood of reac-
5. Preventive therapy, as a replacement for tivation. Reference 24 emphasizes the
chemoprophylaxis with isoniazid or rifampin need for a vaccine to increase the immu-
6. Further enhancement of existing acquired nity of individuals who are already tuber-
immunity by repeated vaccinations (which are not culin positive and harbor virulent tuber-
advised with current BCG vaccines) cle bacilli. Such individuals constitute a
a
Adapted from reference 2. large proportion of the world’s population.
22. DEVELOPING BETTER TUBERCULOSIS VACCINES 䡵 345

5. Such a vaccine could replace isoniazid lesion will also have this favorable CMI/DTH
(INH) as preventive therapy in persons ratio, and the lesion would probably have less
who had recently become tuberculin pos- caseation and undergo liquefaction less readily.
itive. Substituting such a vaccine for INH Some antigenic fractions of the tubercle bacil-
would eliminate the danger of hepato- lus, i.e., certain tuberculoproteins (or peptides)
toxicity and the development of drug re- complexed with certain carbohydrates and lipids
sistance to INH. (28–34), seem to stimulate CMI with minimal
6. Because it produces negligible levels of tissue-damaging DTH (28, 34).To improve cur-
delayed-type hypersensitivity (DTH), such rently available strains of BCG, their content of
a vaccine could be given more than once, such antigens should probably be increased.
to create a higher level of immunity. Other antigenic fractions (i.e., tuberculopro-
teins that produce the tuberculin reaction) seem
Boosting immunity with a better BCG or
to stimulate tissue-damaging DTH with less
another live vaccine in individuals who are
CMI (28, 35, 36). These tuberculoproteins are
tuberculin positive from an arrested virulent
not protective as a vaccine (28, 35, 36). In fact,
infection may temporarily lower resistance to
tuberculoproteins can produce severe necrosis in
that endogenous infection (25), because a pro-
the tuberculin-positive host if their concentra-
portion of the recirculating antigen-specific
tion exceeds that which is safe for that host
lymphocytes now enter the sites of BCG (or live
(37). To improve currently available strains of
vaccine) multiplication.Therefore, fewer defense
BCG, their content of tuberculoproteins should
cells would be available to keep the virulent
probably be decreased.
infection arrested. (See comments after reference
The engineering of vaccines to produce an
58 of Appendix C.) However, the negative
increase in the CMI/DTH ratio seems logical,
effects of the boosting with important antigens
but still remains to be established. Also, the
would not be long lasting, and the subsequent
immunogenic role of proteins secreted by live
positive effects should far outweigh any tem-
tubercle bacilli (32, 38, 39) and the immuno-
porarily harmful effects that may have occurred.
genic role of carbohydrate fractions of the bacil-
For reasons discussed in chapters 2 and 5,
lus (28, 34) need further investigation.
DTH to some of the antigens of tubercle bacilli
is required to effectively control this disease.
Adjuvant Composition
However, the amount of tuberculin sensitivity
The intradermal injection of BCG apparently
had no relation to the amount of protection
initiates an early, pronounced nonspecific
against clinical tuberculosis that was produced by
cytokine response, both in the resident cells and
vaccination with BCG or vole bacilli (Mycobac-
in the mononuclear cells that soon infiltrate the
terium microti) (26, 27).Therefore, antigens caus-
site (40). Mononuclear cells at sites of BCG
ing low levels of tuberculin sensitivity could be
injection have upregulated their IL-1,TNF-,
just as effective as vaccines producing high lev-
MCP-1, and IL-8 mRNAs by the third day,
els (discussed further below).
which is long before the time when the immune
response occurs (40) (see chapter 19). These
COMPOSITION OF IMPROVED cytokines, as well as the accompanying upreg-
VACCINES AGAINST TUBERCULOSIS
ulation of vascular adhesion molecules (41) (see
Antigen Composition chapter 21), play important roles in the accu-
The antigens in vaccines causing the most mulation of dendritic cells, macrophages, lym-
favorable cell-mediated immunity (CMI)/DTH phocytes, and granulocytes at the site of BCG
ratio will produce an expanded (memory) T- injection and in the draining lymph nodes.
lymphocyte population with the same favorable Incorporation into live vaccines of genes that
CMI/DTH ratio. Then, when the vaccinated produce cytokines and/or costimulatory mole-
individual inhales virulent tubercle bacilli, the cules for antigen presentation is a field of active
memory lymphocytes infiltrating the resulting investigation (42–46).
346 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

Lurie showed that acquired resistance to that produce little or no sensitivity to tuber-
tuberculosis was superimposed on, and deter- culin still be able to stop such logarithmic intra-
mined by, the native (innate) resistance of his cellular growth? The answer seems to be “yes.”
inbred rabbits (9, 35). The early tuberculous In the British Medical Research Council tri-
lesions (and probably their draining lymph als on human populations, one lot of vole bacil-
nodes) of Lurie’s resistant rabbits contained lus vaccine produced a high incidence of tuber-
greater numbers of lymphocytes and macro- culin positivity and another lot did not. Yet,
phages (and probably dendritic cells) than did both lots showed equally high protective
the early lesions of his susceptible rabbits (3). potency against tuberculosis (27).This and other
Such large numbers of local mononuclear cells studies with different strains of BCG (51) clearly
contributed to the high levels of acquired demonstrate that the antigens causing appre-
immunity to tuberculosis that developed in his ciable tuberculin positivity were not required to
resistant rabbits. By analogy, a vaccine that protect human beings.
recruited a large number of these cells into sites When applied to the pathogenesis of tuber-
where antigens are located should produce more culosis, these findings indicate that the killing of
immunity than a vaccine that recruited a small nonactivated macrophages, in which the bacilli
number of these cells. are growing intracellularly in a logarithmic fash-
Therefore, a more effective vaccine than the ion, is not uniquely due to the antigens in tuber-
currently available BCG requires not only a culin preparations. It can apparently be pro-
more appropriate mycobacterial antigen com- duced by a DTH response to other antigens of
position, but also an adjuvant composition that the bacillus, especially when these antigens are
recruits the same or a larger number of dendritic in sufficiently high concentrations. In fact, in the
cells, macrophages, and lymphocytes to sites mouse model, lymphocytes that transfer pro-
where the vaccine is deposited. tective immunity can be dissociated from those
Additional aspects of designing an improved that transfer tuberculin sensitivity (56).
vaccine for tuberculosis have been reviewed (24,
44,47–54).Of considerable importance is increas- LIVE TUBERCULOSIS VACCINES
ing the number of memory lymphocytes that SHOULD BE STANDARDIZED
remain in the host for many years following BEFORE COMPARING THEIR
RELATIVE EFFICACIES
immunization (see chapter 6). During the first
Two types of live vaccines should be standard-
year after the initial immune response, up to 90%
ized before we can conclude that the antigens
of the antigen-specific T lymphocytes are elim-
in one are more effective than the antigens in the
inated by apoptosis—a phenomenon that seems
other.
to prevent autoimmunity (42).The remaining T
cells become memory cells (42). New (recom- 1. They should be administered in the same
binant) vaccines that reduce cell death in the manner. Intradermal administration is
initial expanded antigen-specific T-cell population more accurate than the percutaneous
should have greatly increased efficacy (42). (multiple puncture) method. Oral and
intranasal administration may perhaps be
HOSTS DO NOT NEED HIGH LEVELS the least accurate because of variations in
OF TUBERCULIN SENSITIVITY TO antigen absorption.
CONTROL THE DISEASE 2. The two vaccines should contain the same
In humans and rabbits, tissue-damaging DTH number of live bacilli and also the same
seems to be required to stop the initial logarith- number of dead bacilli. Some antigens are
mic growth of tubercle bacilli within nonacti- only secreted by live bacilli (57). Immu-
vated macrophages (see chapters 2 and 15), as nization of mice by live, but not dead,
well as the subsequent logarithmic growth within tubercle bacilli produces antigen-specific
nonactivated macrophages that continuously lymphocytes (in the spleen) that can adop-
enter established lesions from the bloodstream tively transfer immunity to naive mice
(55) (see chapter 10).Therefore, would vaccines (50).An increased percentage of live bacilli
22. DEVELOPING BETTER TUBERCULOSIS VACCINES 䡵 347

may also increase the Th1/Th2 lympho- tuberculous hosts, because it elicits minimal
cyte ratio. This ratio seems to play an tuberculin sensitivity. Due to the presence of
important role in host resistance to tuber- common mycobacterial antigens, M. vaccae
culosis (57, 58). increases the Th1/Th2 ratio (58), thereby
3. The bacilli in each vaccine should be in the enhancing the host’s CMI.
same stage of their growth cycle. Actively Repeated immunization with M. vaccae appar-
growing log-phase bacilli may multiply to ently reduced the required length of antimi-
higher titers in recipient hosts than dormant crobial therapy and helped in the treatment of
(frozen or lyophilized) bacilli, because dor- multidrug-resistant tuberculosis (67). Since the
mant bacilli may be slow in starting to host’s immune forces work in synergy with
grow in the host and may not reach as antimicrobials, studies on the use of this and
high a titer before acquired (adaptive) other immunotherapeutic agents should be
immunity develops (see chapter 11 and actively pursued. (In the two experiments that
reference 59). we did on immunotherapy with M. vaccae in
4. The two vaccines should contain the same rabbits with cavitary tuberculosis, the variation
number of clumped and isolated bacilli. among the rabbits was too large to detect any
Clumps are more common in thawed significant effect [see chapter 4].)
frozen bacilli and in reconstituted lyo-
philized bacilli.When injected parenter-
Dendritic Cells as Vaccine Carriers
ally, such clumps may resist destruction by
The use of dendritic cells as vaccine carriers is
the host better than do isolated bacilli.
being explored (68). Dendritic cell-vaccine car-
We prepared fresh 100% live nonclumped riers may find their best use as immunotherapy
and 100% viable (log-phase) bacilli (11) for the in immunocompetent patients with multidrug-
rabbit experiments with BCG and M. microti resistant tuberculosis.
(the vole bacillus) described in chapter 23. How-
ever, for trials in human populations, reconsti- Surgical Removal of a Cavitary Lesion
tuted lyophilized vaccines, which usually con- Most multidrug-resistant tubercle bacilli arise
tain many clumps and many dead bacilli (60), are in cavities where the growth of extracellular
more practical. Reference 61 describes how to bacilli may be tremendous. If antimicrobial ther-
obtain nonclumped BCG preparations that are apy with second-line drugs is unsuccessful,
90 to 99% viable after freezing and storing at surgery to remove the cavitary lesion may be
⫺70°C, even after a year. indicated. Doing so would remove the major
Excellent reviews of the factors just listed (as source of multidrug-resistant bacilli. In immuno-
well as others) have been published (60–64). competent patients, noncavitary lesions con-
Included were vaccine stability, the growth of taining multidrug-resistant bacilli (originating
BCG in the organs of mice, and the degree of metastatically from cavities) would probably be
protection produced, as well as standardization controlled by the host’s immune forces, but if
of how the vaccine was injected. Because of such lesions were not controlled, boosting immu-
such variables, different laboratories were unable nity with immunotherapy could be beneficial.
to rank the efficacy of different BCG prepara-
tions in animals in any consistent order (65).
MAXIMAL EFFECTIVENESS
IMMUNOTHERAPY OF TUBERCULOSIS VACCINES
IN HUMANS
Mycobacterium vaccae In theory, humans should have maximal immu-
Heat-killed M. vaccae, a nonpathogenic soil bacil- nity after they arrest an infection with virulent
lus, is being used as immunotherapy in patients tubercle bacilli. In most of these tuberculin-
with active tuberculosis (reviewed in references positive persons, there is a tiny caseous calcified
58, 66, and 67). Heat-killed M. vaccae does not lesion (0.5 to 2.0 mm in size) (see chapter 3).
produce adverse systemic DTH reactions in If such persons remain tuberculin positive,
348 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

tubercle bacilli may occasionally escape from Environmental mycobacteria may also lessen
the lesion, enter nonactivated blood-borne the difference between vaccinated and control
macrophages, multiply, and then be inhibited by groups (26, 27). Constant exposure to mycobac-
the DTH and CMI response. This sequence teria in the soil (and in dust arising from the soil)
maintains immunity and tuberculin sensitivity. can increase host resistance to tuberculosis
Nevertheless, (i) careful epidemiological studies (reviewed in reference 26).
(69), (ii) restriction fragment length polymor-
phism with DNA fingerprinting (70), (iii) phage MAXIMAL EFFECTIVENESS OF
typing (71), and (iv) antimicrobial-resistant spec- TUBERCULOSIS VACCINES IN
RABBITS
tra (71, 72) have shown that tuberculin-positive
individuals are able to develop active reinfection Lurie’s Experiments with Virulent
pulmonary tuberculosis when the exposure to Human-Type Tubercle Bacilli
exogenous virulent tubercle bacilli is high (70, Rabbits should have maximal immunity while
73a). However, active reinfection tuberculosis is they are arresting an infection with human-
rare when the exposure is low (73, 73a, 74). type tubercle bacilli (which are never fully vir-
Because such natural reinfection occurs, immu- ulent in this animal species). Lesions produced
nity in humans apparently is never complete. by the human type may caseate, liquefy, and
Therefore, one should not expect immunity cavitate, but do not progress to death, but lesions
produced by any vaccine to be complete. produced by the bovine type almost always
Active tuberculosis produced by a different progress to death (35).
strain from the one causing the primary infec- Therefore, to test their immunogenicity, Lurie
tion is most frequently due to a reinfection with (78, 79) infected rabbits intravenously with vir-
an exogenously inhaled bacillus. However, dif- ulent human-type bacilli (H37Rv), and 6
ferent strains of tubercle bacilli may be found in months later he reinfected them intravenously
the same sputum specimen (75), because of bacil- with virulent bovine-type bacilli.After the rein-
lary mutations occurring within the cavities pro- fection, Lurie euthanized the rabbits at various
duced by the original bacillary strain (76). intervals (up to 2 months) and cultured the
lungs for tubercle bacilli. He found that the
Factors Influencing Vaccine bacilli were mostly the human type.The mul-
Effectiveness tiplication of the fully virulent bovine type had
Vaccines are less effective in human immuno- been markedly reduced by the presence of the
deficiency virus (HIV)-infected persons and residual human type. (The human-type bacilli
others with decreased immune function.These were identified by their eugonic colonies on
individuals develop active tuberculosis more solid culture media, and the bovine-type bacilli
readily after inhaling exogenous tubercle bacilli. were identified by their dysgonic colonies [78].)
They also develop active tuberculosis more read- This experiment indicates that residual pri-
ily from endogenous tubercle bacilli that had mary lesions caused by human-type bacilli
been latent in primary tuberculous lesions.They (which are only semivirulent in rabbits) offer
are more susceptible to tuberculosis because considerable protection against reinfection by
they have a reduced Th1 lymphocyte popula- bovine-type bacilli (which are highly virulent in
tion. Helminth infection and/or environmental rabbits).
mycobacteria can also shift a Th1 lymphocyte
response toward a Th2 response (reviewed in ref- Proposed Studies in Rabbits with the
erence 77). Tuberculosis vaccines have often Same Strain of Tubercle Bacilli
been ineffective in populations in sub-Saharan With modern recombinant technology, virulent
Africa because of parasitic infections (77). Every human-type bacilli can be made to produce
tuberculosis vaccine trial in developing countries green or red fluorescent proteins. Rabbits can
should attempt to eliminate helminth infection then be exposed to “green” bacilli by aerosol and
before the vaccine is administered. challenged with “red” bacilli by aerosol weeks
22. DEVELOPING BETTER TUBERCULOSIS VACCINES 䡵 349

later (suggested by William R. Bishai). The Postinfection Immunization


tubercles produced can then be counted (see A substantial percentage of the world’s human
chapter 11) and cultured to determine whether population is already tuberculin positive from
they arose from the primary or challenge inhaling virulent tubercle bacilli. Therefore,
aerosol. Controls of rabbits that had not been postinfection immunization to prevent endoge-
previously infected, but were challenged with nous (and exogenous) reinfection would be
“red” bacilli, should be included.Also, the exper- especially useful. Subunit vaccines (including
iment should be repeated with the infection by DNA vaccines) are probably the safest type to use
“green” and “red” bacilli in reverse order. for this purpose (reviewed in references 90 and
91).Antigens should be chosen that do not elicit
OTHER VACCINE POSSIBILITIES tuberculin sensitivity, because systemic tuber-
culin can exacerbate existing tuberculous lesions
Recombinant Technology To Improve and even induce cavities to form (see chapter 4).
Vaccines
Protection of rabbits against reinfection by vir- Combination Vaccines
ulent human-type bacilli assumes that these Combination vaccines will probably provide
bacilli contain a full complement of antigens and the most effective protection against active
that a host recovering from infection with such tuberculosis (see references 24, 44, and 90–92).
bacilli will have the most effective immune Such an immunization often consists of an ini-
response. However, certain antigens may be tial BCG vaccination followed by one or more
more protective than others. If such antigens are booster immunizations with important my-
increased substantially in BCG by recombinant cobacterial antigens (including those produced
technology (see references 44 and 80–84), it is by DNA vaccines [93]). The antigens in such
possible that the resulting BCG vaccine will boosters would expand the appropriate T-
immunize more effectively than even virulent lymphocyte population almost as well as if these
human-type tubercle bacilli. The BCG strain antigens had been incorporated into the live
should be chosen carefully, because differences vaccine itself, and would avoid the possibility of
among various BCG strains exist (64, 85, 86). an increase in virulence that a direct incorpo-
It is also possible that the additional antigens ration into the BCG genome might produce.
will increase the virulence of BCG and make it However, booster immunization would proba-
unsafe to use as a vaccine. Such a vaccine should bly have to be given more than once to provide
be tested in mice, guinea pigs, rabbits, and per- the best results.
haps monkeys before human trials are per-
formed (see references 87–89). M. tuberculosis Mutants
as Possible New Vaccines
DNA Vaccines Reference 94 lists the in vivo growth charac-
DNA vaccines are those in which the DNAs of teristics in mice of several M. tuberculosis mutants
one or more mycobacterial antigens are incor- that lack specific genes. (See reference 95 for a
porated into a plasmid and usually injected more detailed discussion of the production of
intramuscularly. Such DNA vaccines increased such vaccines and their use.) These mutants
the immunity of mice and guinea pigs to viru- often do not grow or persist in the host. Some
lent tubercle bacilli, but no more than BCG did, of these mutants may eventually find use as an
and the immunity produced was shorter lived improved vaccine for tuberculosis (especially in
(reviewed in references 24 and 90). HIV/AIDS individuals) or as a booster for
DNA vaccines or vaccines incorporating immunity after BCG vaccination.
mycobacterial protein, lipid, and/or glycolipid
antigens (in adjuvants) may be useful in HIV Reviews of Possible New Vaccines
patients, because such vaccines cannot cause References 24, 44, 45, 58, 87, 90, 91, and 94
tuberculosis in immunosuppressed individuals. briefly review some of the newly developed
350 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

potential vaccines for tuberculosis that were A upregulates activity of the mannose receptor, a
evaluated in mice and/or guinea pigs. pattern recognition receptor expressed on human
macrophages. J. Immunol. 169:3565–3573.
8. Dannenberg, A. M., Jr. 1998. Lurie’s tubercle-
BCG MUST MULTIPLY IN THE HOST count method to test TB vaccine efficacy in rabbits.
TO BE MOST EFFECTIVE Front. Biosci. 3:c27–33. Available at http://www
We favor the use of live vaccines for immuno- .bioscience.org/1998/v3/c/dannenbe/list.htm.
competent persons.The bacilli in live vaccines 9. Lurie, M. B., P. Zappasodi, E. Cardona-Lynch,
multiply in the host and produce (and even and A. M. Dannenberg, Jr. 1952.The response
secrete) certain antigens that are not present in to the intracutaneous inoculation of BCG as an
index of native resistance to tuberculosis. J. Immunol.
killed preparations. Live bacilli also persist longer 68:369–387.
in the host than do killed bacilli. Reference 96 10. Dannenberg, A. M., Jr. 1999. Pathophysiology:
reviews the older literature on nonliving vac- basic aspects. I. Pathogenesis of tuberculosis. II.
cines against tuberculosis. Immunology of tuberculosis, p. 17–47. In D. Schloss-
berg (ed.), Tuberculosis and Nontuberculous Mycobac-
BCG AS A CARRIER OF ANTIGENS terial Infections, 4th ed. The W. B. Saunders Co.,
FROM OTHER PATHOGENS Philadelphia, Pa.
11. Dannenberg, A. M., Jr., W. R. Bishai, N. Par-
Because BCG tends to persist in the host for rish, R. Ruiz, W. Johnson, B. C. Zook, J. W.
many years, recombinant BCG vaccines con- Boles, and M. L. M. Pitt. 2000. Efficacies of
taining specific antigens from various viral, bac- BCG and vole bacillus (Mycobacterium microti) vac-
terial, and parasitic microorganisms are currently cines in preventing clinically apparent pulmonary
being evaluated (reviewed in reference 83). tuberculosis in rabbits: a preliminary report. Vaccine
19:796–800.
12. Lindgren, I. 1961. Anatomical and roentgeno-
REFERENCES logic studies of tuberculosis infection in BCG-
1. Lurie, M. B. 1944. Experimental epidemiology of vaccinated and non-vaccinated subjects, with
tuberculosis: hereditary resistance to attack by tuber- biophysical investigations of calcified foci. Acta
culosis and to the ensuing disease and the effect of Radiol. Suppl. 209:1–101.
the concentration of tubercle bacilli upon these 13. Lindgren, I. 1965.The pathology of tuberculous
two phases of resistance. J. Exp. Med. 79:573–589. infection in BCG-vaccinated humans. Adv.Tuberc.
2. Dannenberg,A. M., Jr. 1990. Controlling tuber- Res. 14:202–234.
culosis: the pathologist’s point of view. Res. Micro- 14. Sutherland, I., and I. Lindgren. 1979.The pro-
biol. 141:192–196, 262–263. tective effect of BCG vaccination as indicated by
3. Lurie, M. B., P. Zappasodi, and C. Tickner. autopsy studies. Tubercle 60:225–231.
1955. On the nature of genetic resistance to tuber- 15. Nuermberger, E. L., T.Yoshimatsu, S. Tyagi,
culosis in the light of the host-parasite relationships W. R. Bishai, and J. H. Grosset. 2004. Pau-
in natively resistant and susceptible rabbits. Am. cibacillary tuberculosis in mice after prior aerosol
Rev.Tuberc. 72:297–329. immunization with Mycobacterium bovis BCG. Infect.
4. Riley, R. L., C. C. Mills, F. O’Grady, L. U. Sul- Immun. 72:1065–1071.
tan, F. Wittstadt, and D. N. Shivpuri. 1962. 16. Morrison, N. E., and F. M. Collins. 1975.
Infectiousness of air from a tuberculosis ward. Ultra- Immunogenicity of an aerogenic BCG vaccine in
violet irradiation of infected air: comparative infec- T-cell-depleted and normal mice. Infect. Immun.
tiousness of different patients. Am. Rev. Respir. Dis. 11:1110–1121.
85:511–525. 17. Cohn, M. L., C. L. Davis, and G. Middle-
5. Schlesinger, L. S. 1996. Role of mononuclear brook. 1958.Airborne immunization against tuber-
phagocytes in M. tuberculosis pathogenesis. J. Inves- culosis. Science 128:1282–1283.
tig. Med. 44:312–323. 18. Middlebrook, G. 1961. Immunological aspects
6. Fenton, M. J., L.W. Riley, and L. S. Schlesinger. of airborne infection: reactions to inhaled antigens.
2005. Receptor-mediated recognition of Mycobac- Bacteriol. Rev. 25:331–346.
terium tuberculosis in host cells, p. 405–426. In S. T. 19. Strober,W., B. Kelsall, and T. Marth. 1998. Oral
Cole, K. D. Eisenach, D. N. McMurray, and W. R. tolerance. J. Clin. Immunol. 18:1–30.
Jacobs, Jr. (ed.), Tuberculosis and the Tubercle Bacillus. 20. Aronson, J. D., and A. M. Dannenberg. 1935.
ASM Press,Washington, D.C. Effect of vaccination with BCG on tuberculosis in
7. Beharka, A., C. D. Gaynor, B. K. Kang, D. R. infancy and in childhood: correlation of reactions
Voelker, F. X. McCormack, and L. S. to tuberculin tests, roentgenologic diagnosis and
Schlesinger. 2002. Pulmonary surfactant Protein mortality. Am. J. Dis. Child. 50:1117–1130.
22. DEVELOPING BETTER TUBERCULOSIS VACCINES 䡵 351

21. Ulrichs, T., and S. H. E. Kaufmann. 2002. aerogenic tuberculosis in guinea pigs. Infect. Immun.
Mycobacterial persistence and immunity. Front. 9:815–820.
Biosci. 7:d458–d469. 37. Rich, A. R. 1951. The Pathogenesis of Tuberculosis,
22. Troisier, J., J. Le Melletier, and J. Sifferlen. 2nd ed. Charles C Thomas Publisher, Springfield,
1944. La vaccination antituberculeuse par les voies Ill.
respiratoires.Aérosols et brouillards de BCG. Ann. 38. Harboe, M. 1992. The significance of proteins
Inst. Pasteur 70:33–36. actively secreted by Mycobacterium tuberculosis in
23. Rosenthal, S. R., J.T. McEnery, and N. Raisys. relation to immunity and complications of myco-
1968. Aerogenic BCG vaccination against tuber- bacterial diseases. Int. J. Leprosy 60:470–476.
culosis in animal and human subjects. J. Asthma 39. Doherty,T. M., and P. Andersen. 2002.Tuber-
Res. 5:309–323. culosis vaccine development. Curr. Opin. Pulm. Med.
24. Andersen, P. 2001. TB vaccines: progress and 8:183–187.
problems. Trends Immunol. 22:160–168. 40. Sugisaki, K., A. M. Dannenberg, Jr., Y. Abe,
25. Allison, M. J., P. Zappasodi, and M. B. Lurie. J. Tsuruta, W.-J. Su, W. Said, L. Feng,
1962. The correlation of a biphasic metabolic T.Yoshimura, P. J. Converse, and P. Mounts.
response with a biphasic response in resistance to 1998. Nonspecific and immune-specific upregula-
tuberculosis in rabbits. J. Exp. Med. 115:881–890. tion of cytokines in rabbit dermal tuberculous
26. Comstock, G.W. 1994. Field trials of tuberculo- (BCG) lesions. J. Leukoc. Biol. 63:440–450.
sis vaccines: how could we have done them better? 41. Abe,Y., K. Sugisaki, and A. M. Dannenberg,
Control. Clin.Trials 15:247–276. Jr. 1996. Rabbit vascular endothelial adhesion
27. Comstock, G.W. 1988. Identification of an effec- molecules: ELAM-1 is most elevated in acute
tive vaccine against tuberculosis. Am. Rev. Respir. Dis. inflammation, whereas VCAM-1 and ICAM-1 pre-
138:479–480. dominate in chronic inflammation. J. Leukoc. Biol.
28. Crowle, A. J. 1988. Immunization against tuber- 60:692–703.
culosis: what kind of vaccine? Infect. Immun. 42. Esser, M. T., R. D. Marchese, L. S. Kierstead,
56:2769–2773. L. G.Tussey, F.Wang, N. Chirmule, and M.W.
29. Chan, J., X. Fan, S. W. Hunter, P. J. Brennan, Washabaugh. 2003. Memory T cells and vaccines.
and B. R. Bloom. 1991. Lipoarabinomannan, a Vaccine 21:419–430.
possible virulence factor involved in persistence of 43. Spickler, A. R., and J. A. Roth. 2003.Adjuvants
Mycobacterium tuberculosis within macrophages. Infect. in veterinary medicine: modes of action and adverse
Immun. 59:1755–1761. effects. J.Vet. Intern. Med. 17:273–281.
30. Barnes, P. F., V. Mehra, G. R. Hirschfield,
44. Kaufmann, S. H. E. 2001. How can immunology
S.-J. Fong, C.Abou-Zeid, G.A.W. Rook, S.W.
contribute to the control of tuberculosis? Nat. Rev.
Hunter, P. J. Brennan, and R. L. Modlin. 1989.
Immunol. 1:20–30.
Characterization of T cell antigens associated with
the cell wall protein peptidoglycan complex of 45. Young, D. B., and G. R. Stewart. 2002.Tuber-
Mycobacterium tuberculosis. J. Immunol. 143:2656– culosis vaccines. Br. Med. Bull. 62:73–86.
2662. 46. Murray, P. J., A. Aldovini, and R. A. Young.
31. Brennan, P. J. 1989. Structure of mycobacteria: 1996. Manipulation and potentiation of antibacte-
recent developments in defining cell wall carbohy- rial immunity using recombinant Bacille Calmette-
drates and proteins. Rev. Infect. Dis. 11(Suppl. Guérin strains that secrete cytokines. Proc. Natl.
2):S420–S430. Acad. Sci. USA 93:934–939.
32. Wiker, H. G., S. Nagai, M. Harboe, and 47. Kaufmann, S. H. E. 1989. Leprosy and tubercu-
L. Ljungqvist. 1992. A family of cross-reacting losis vaccine design. Trop. Med. Parasitol. 40:251–257.
proteins secreted by Mycobacterium tuberculosis. Scand. 48. Nossal, G. J. V. 1990. Immunogenicity. UCLA
J. Immunol. 36:307–319. Symp. Mol. Cell Biol. 13:309–319.
33. Young, D. B., S. H. E. Kaufmann, P. W. M. 49. Grange, J. M. 1988. Molecular biology: new hopes
Hermans, and J. E. R. Thole. 1992. Mycobac- and challenges. Tubercle 69:1–4.
terial protein antigens: a compilation. Mol. Microbiol. 50. Orme, I. M. 1988. Induction of nonspecific
6:133–145. acquired resistance and delayed-type hypersensi-
34. Crowle,A. J. 1972.Trypsin-extracted immunizing tivity, but not specific acquired resistance, in mice
antigen of the tubercle bacillus: a practical vaccine? inoculated with killed mycobacterial vaccines. Infect.
Adv.Tuberc. Res. 18:31–102. Immun. 56:3310–3312.
35. Lurie, M. B. 1964. Resistance to Tuberculosis: Exper- 51. Fine, P. E. M. 1989.The BCG story: lessons from
imental Studies in Native and Acquired Defensive Mech- the past and implications for the future. Rev. Infect.
anisms. Harvard University Press, Cambridge, Mass. Dis. 11(Suppl. 2):S353–S359.
36. Reggiardo, A., and G. Middlebrook. 1974. 52. Orme, I. M. 2001. The search for new vaccines
Delayed-type hypersensitivity and immunity against against tuberculosis. J. Leukoc. Biol. 70:1–10.
352 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

53. Wang, J., and Z. Xing. 2002. Tuberculosis vac- treatment of tuberculosis. Front. Biosci. 9:1701–
cines: the past, present and future. Expert Rev.Vaccines 1719.
1:341–354. 68. Moll, H. 2003. Dendritic cells as a tool to combat
54. Young, D. B., and G. R. Stewart. 2002.Tuber- infectious diseases. Immunol. Lett. 85:153–157.
culosis vaccines. Br. Med. Bull. 62:73–86. 69. Romeyn, J. A. 1970. Exogenous reinfection in
55. Dannenberg, A. M., Jr. 2003. Macrophage tuberculosis. Am. Rev. Respir. Dis. 101:923–927.
turnover, division and activation within developing, 70. van Rie, A., R. Warren, M. Richardson, T. C.
peak and “healed” tuberculous lesions produced in Victor, R. P. Gie, D.A. Enarson, N. Beyers, and
rabbits by BCG. Tuberculosis 83:251–260. P. D. van Helden. 1999. Exogenous reinfection as
56. Orme, I. M., and F. M. Collins. 1984.Adoptive a cause of recurrent tuberculosis after curative treat-
protection of the Mycobacterium tuberculosis-infected ment. N. Engl. J. Med. 341:1174–1179.
lung. Dissociation between cells that passively trans- 71. Nardell, E., B. McInnis, B. Thomas, and
fer protective immunity and those that transfer S. Weidhaas. 1986. Exogenous reinfection with
delayed-type hypersensitivity to tuberculin. Cell. tuberculosis in a shelter for the homeless. N. Engl.
Immunol. 84:113–120. J. Med. 315:1570–1575.
57. Barnes, P. F., R. L. Modlin, and J. J. Ellner. 72. Small, P. M., R. W. Shafer, P. C. Hopewell,
1994.T-cell responses and cytokines, p. 417–435. In S. P. Singh, M. J. Murphy, E. Desmond, M. F.
B. R. Bloom (ed.), Tuberculosis: Pathogenesis, Protec- Sierra, and G. K. Schoolnik. 1993. Exogenous
tion, and Control.ASM Press,Washington, D.C. reinfection with multidrug-resistant Mycobacterium
58. Rook, G. A.W., G. Seah, and A. Ustianowski. tuberculosis in patients with advanced HIV infection.
2001. M. tuberculosis: immunology and vaccination. N. Engl. J. Med. 328:1137–1144.
Eur. Respir. J. 17:537–557. 73. Fine, P. E. M., and P. M. Small. 1999. Exoge-
59. Manabe, Y. C., A. M. Dannenberg, Jr., S. K. nous reinfection in tuberculosis. N. Engl. J. Med.
Tyagi, C. L. Hatem, M.Yoder, S. C.Woolwine, 341:1226–1227.
B. C. Zook, M. L. M. Pitt, and W. R. Bishai. 73a.Letters to the Editor. 2000. Recurrent tubercu-
2003. Different strains of Mycobacterium tuberculosis losis due to exogenous infection. N. Engl. J. Med.
cause various spectrums of disease in the rabbit 342:1050–1051.
model of tuberculosis. Infect. Immun. 71:6004–6011. 74. de Boer, A., M. W. Borgdorff, P. E. W. de
60. Gheorghiu, M., and P. H. Lagrange. 1983.Via- Haas, N. J. D. Hagelkerke, J. D. A. van Emb-
bility, heat stability, and immunogenicity of four den, and D. van Soolingen. 1999.Analysis of rate
BCG vaccines prepared from four different BCG of change of IS6110 RFLP patterns of Mycobac-
strains. Ann. Immunol. (Paris) 134C:125–147. terium tuberculosis based on serial patient isolates.
61. Dietrich, G., H.-J. Mollenkopf, H. Weber, J. Infect. Dis. 180:1238–1244.
B. Knapp, K.-D. Diehl, J. Hess, F. Blackkolb, 75. Warren, R. M., T. C. Victor, E. M. Streicher,
M. Bröker, S. H. E. Kaufmann, and E. Hundt. M. Richardson, N. Beyers, N. C. Gey van Pit-
2002. Cultivation of Mycobacterium bovis BCG in tius, and P. D. van Helden. 2004. Patients with
bioreactors. J. Biotechnol. 96:259–270. active tuberculosis often have different strains in the
62. Dietrich, G., J.-F. Viret, and J. Hess. 2003. same sputum specimen. Am. J. Respir. Crit. Care
Mycobacterium bovis BCG-based vaccines against Med. 169:610–614.
tuberculosis: novel developments. Vaccine 21:667– 76. Behr, M. A. 2004. Tuberculosis due to multiple
670. strains: a concern for the patient? a concern for
63. Milstien, J. B., and J. J. Gibson. 1990. Quality tuberculosis control? Am. J. Respir. Crit. Care Med.
control of BCG vaccine by WHO: a review of fac- 169:554–555.
tors that may influence vaccine effectiveness and 77. Rook, G. A. W., K. Dheda, and A. Zumla.
safety. Bull.W. H. O. 68:93–108. 2005. Do successful tuberculosis vaccines need to
64. Behr, M. A. 2002. BCG—different strains, differ- be immunoregulatory rather than Th1-boosting?
ent vaccines? Lancet Infect. Dis. 2:86–92. Vaccine 23:2115–2120.
65. Wiegeshaus, E. H., G. E. Harding, D. N. 78. Lurie, M. B. 1929.The fate of tubercle bacilli in
McMurray, A. A. Grover, and D. W. Smith. the organs of reinfected rabbits. J. Exp. Med. 50:747–
1971.A cooperative evaluation of test systems used 765.
to assay tuberculosis vaccines. Bull.W. H. O. 45:543– 79. Lurie, M. B. 1933.A correlation between the his-
550. tological changes and the fate of living tubercle
66. Fourie, P. B., J. J. Ellner, and J. L. Johnson. bacilli in the organs of reinfected rabbits. J. Exp. Med.
2002.Whither Mycobacterium vaccae—encore. Lancet 57:181–201.
360:1032–1033. 80. Horowitz, M. A., G. Harth, B. J. Dillon, and
67. Stanford, J., C. Stanford, and J. Grange. 2004. S. Maslesa-Galic. 2000. Recombinant bacillus
Immunotherapy with Mycobacterium vaccae in the Calmette-Guérin (BCG) vaccines expressing the
22. DEVELOPING BETTER TUBERCULOSIS VACCINES 䡵 353

Mycobacterium tuberculosis 30-kDa major secretory 88. McMurray, D. N. 2001. Determinants of vaccine-
protein induce greater protective immunity against induced resistance in animal models of pulmonary
tuberculosis than conventional BCG vaccines in a tuberculosis. Scand. J. Infect. Dis. 33:175–178.
highly susceptible animal model. Proc. Natl.Acad. Sci. 89. McMurray, D. N. 2003. Recent progress in the
USA 97:13853–13858. development and testing of vaccines against human
81. Ohara, N., and T.Yamada. 2001. Recombinant tuberculosis. Int. J. Parasitol. 33:547–554.
BCG vaccines. Vaccine 19:4089–4098. 90. Britton,W. J., and U. Palendira. 2003. Improv-
82. Agger, E. M., and P. Andersen. 2002. A novel ing vaccines against tuberculosis. Immunol. Cell Biol.
TB vaccine; towards a strategy base on our under- 81:34–45.
standing of BCG failure. Vaccine 21:7–14. 91. Collins, H. L., and S. H. E. Kaufmann. 2001.
83. O’Donnell, M. A. 1997.The genetic reconstitu- Prospects for better tuberculosis vaccines. Lancet
tion of BCG as a new immunotherapeutic tool. Infect. Dis. 1:21–28.
Trends Biotechnol. 15:512–517. 92. Brooks, J. V., A. A. Frank, M. A. Keen, J. T.
84. Dhar, N., V. Rao, and A. K. Tyagi. 2004. Belisle, and I. M. Orme. 2001. Boosting vaccine
Immunogenicity of recombinant BCG strains over- for tuberculosis. Infect. Immun. 69:2714–2717.
expressing components of the antigen 85 complex
93. Lowrie, D. B. 2003. DNA vaccination: an update.
of Mycobacterium tuberculosis. Med. Microbiol. Immunol.
Methods Mol. Med. 87:377–390.
193:19–25.
85. Behr, M. A., M. A.Wilson,W. P. Gill, H. Sala- 94. Glickman, M. S., and W. R. Jacobs, Jr. 2001.
mon, G. K. Schoolnik, S. Rane, and P. M. Microbial pathogenesis of Mycobacterium tuberculosis:
Small. 1999. Comparative genomics of BCG vac- dawn of a new discipline. Cell 104:477–485.
cines by whole-genome DNA microarray. Science 95. Braunstein, M., S. S. Bardarov, and W. R.
284:1520–1523. Jacobs, Jr. 2002. Genetic methods for deciphering
86. Mostowy, S., A. G. Tsolaki, P. M. Small, and virulence determinants of Mycobacterium tuberculo-
M. A. Behr. 2003.The in vitro evolution of BCG sis. Methods Enzymol. 358:67–99.
vaccines. Vaccine 21:4270–4274. 96. Weiss, D.W. 1959.Vaccination against tuberculo-
87. Orme, I. M., D. N. McMurray, and J.T. Belisle. sis with nonliving vaccines. I.The problem and its
2001. Tuberculosis vaccine development: recent historical background. Am. Rev. Respir. Dis. 80:340–
progress. Trends Microbiol. 9:115–118. 358, 495–509, 676–688.
23
CHARACTERISTICS OF RABBIT BCG
LESIONS AND EFFICACIES OF BCG AND
MYCOBACTERIUM MICROTI VACCINES

Characteristics of dermal BCG lesions in commercial New Zealand White


rabbits [354]
Comparison of dermal BCG lesions and pulmonary lesions produced by
virulent tubercle bacilli in rabbits [358]
Healing of dermal BCG lesions as a measure of host resistance [359]
Lurie’s intravenous BCG experiments in rabbits [359]
Systemic nature of BCG vaccination in humans [360]
Intradermal BCG vaccination in Lurie’s inbred resistant and susceptible
rabbits [361]
Efficacies of BCG and M. microti vaccines in commercial New Zealand
White rabbits [363]

Abstract. In rabbits (and humans), dermal BCG lesions have the same components as
lesions caused by virulent tubercle bacilli, i.e., a caseous liquefied center surrounded by
tuberculous granulation tissue.Therefore, BCG lesions can be used as a model in which
to study the host response to this disease.
In immunocompetent hosts, BCG lesions do not progress and always heal.The rate of
healing of dermal BCG lesions reflected the native and acquired resistance of Lurie’s sus-
ceptible and resistant inbred rabbit strains. Similarly, the rate of healing of BCG lesions has
been shown to reflect the resistance to tuberculosis of human populations. However, the
healing of BCG lesions in individual rabbits and humans may vary considerably from the
mean.
In Lurie’s natively resistant rabbits, immunization with BCG was quite effective in pre-
venting many grossly visible primary pulmonary tubercles (i.e., clinically apparent tuber-
culosis). In his natively susceptible rabbits, BCG was hardly effective at all. In other words,
the rabbits that needed it the least were helped by BCG vaccination the most, and the
rabbits that needed it the most were helped the least. This principle also applies to
human populations: persons with poor resistance to clinical tuberculosis would develop
less immunity from BCG administration than persons with strong resistance.
The efficacies of various BCG and Mycobacterium microti (the vole bacillus) vaccines were
evaluated in commercial outbred rabbits by the tubercle-count method. Both of these vac-
cine types were effective. However, the variations in resistance among outbred rabbits pre-
vented our rating any one vaccine above another. Recombinant BCG vaccines that con-
tain additional antigenic components may be more promising than currently used BCG
vaccines (see chapter 22).

CHARACTERISTICS OF DERMAL BCG Jensen slants, so that, when harvested, the BCG
LESIONS IN COMMERCIAL NEW colonies were still in the log phase of growth.
ZEALAND WHITE RABBITS
They were then ground in an agate mortar with
BCG Preparation 10 to 20 µl of Tween 80, suspended in a solution
In our laboratory, strains of BCG and M. microti of Becton-Dickinson’s BBL Oleic Albumin
were usually maintained on solid Lowenstein- Complex (diluted 1:10 in 0.9% NaCl), and cen-
Jensen slants with periodic transplantation to trifuged for 10 min at 2,000 rpm to remove
fresh slants. Seven to 10 days before use, the clumps containing more than a few bacilli (1).
BCG was again transplanted to fresh Lowenstein- Then, the bacilli in the supernatant fluids were

354
23. RABBIT DERMAL BCG LESIONS 䡵 355

counted microscopically in a hemacytometer were almost healed at 5 to 7 weeks (Fig. 1) (3).


(to obtain the number of live plus dead bacilli in Variations in size among the lesions were often
the preparation) and adjusted to 106 to 107 bacilli due to the time of ulceration and the quantity
per 0.1 ml. Every bacillary unit was found to be of necrotic material discharged from the lique-
viable when cultured.This method of prepara- fied centers. The rabbits were slightly tuber-
tion standardized the vaccines to be compared culin positive 9 days after the injection of BCG
(discussed in chapter 22). In tissue sections, tuber- and strongly tuberculin positive from 14 days on
cle bacilli were identified by acid-fast staining (Fig. 1). At 3 to 5 days, the transected lesions
with carbol-fuchsin (2). showed small foci of necrosis on gross exami-
nation. Between 9 and 23 days, most of these
Dermal BCG Lesion Development necrotic foci had fused to form a relatively large
and Healing caseous center, which liquefied and usually
In rabbits, BCG lesions (produced by the Tice ulcerated, discharging some of its contents.At 37
strain) reached peak size in 2 to 3 weeks and days, the lesions were almost healed: they were

FIGURE 1 Size of BCG lesions in rabbits at various times after receiving 8 intrader-
mal injections of about 5 ⫻ 106 BCG bacilli on each flank (3).The biphasic nature of the
curve is due to a decline in the initial nonspecific inflammatory response to the vaccine
followed by the development of an antigen-specific response.The size of 2-day tuberculin
reactions in these rabbits is shown below the curve: ⫺ ⫽ 0 to 100 mm3, ⫹ ⫽ 100 to 200
mm3, ⫹⫹ ⫽ 200 to 400 mm3, and ⫹⫹⫹ ⫽ greater than 400 mm3.The rabbits had sig-
nificant tuberculin reactions from 9 days on.The means and their standard errors are shown
along with the statistical significance between adjacent points: *P ⬍ 0.05 and ***P ⬍ 0.01.
Reproduced with permission from reference 3.
356 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

FIGURE 2 Mononuclear cells (MN) and granulocytes (PMN) per mm2 of tissue sec-
tion in rabbit BCG lesions at various times during their development and healing. PMN
accumulated around the caseous necrotic areas. Note that, although the number of MN
per mm2 remained high during healing (at 37 days), the volumes of the BCG lesions were
diminished (see Fig. 1). ***P ⬍ 0.01. Reproduced with permission from reference 3.

much reduced in size, the ulcer had usually peripheral areas contained more dispersed cells,
epithelialized, and little or no necrotic contents as well as more collagen fibers, including newly
remained inside. formed collagen as the lesions healed.
The lesions that had discharged most of their The cell infiltrate was mainly composed of
liquefied necrotic contents seemed to heal the mononuclear cells (mostly macrophages and
fastest. We purposely selected such healing lymphocytes, but probably some dendritic cells)
lesions for our 37-day evaluations. Compar- (Fig. 2) (3). Polymorphonuclear leukocytes were
isons of primary and reinfection dermal BCG present in the initial infiltrate and near the
lesions are presented in chapter 20. caseous and liquefied areas of the older lesions,
but elsewhere they were only a minor compo-
Histopathology nent of the cell infiltrate (Fig. 2). Eosinophils
At 1 to 3 days, the BCG lesions showed scattered were rare, but plasma cells and fibroblasts were
areas of intact and fragmented leukocytes among common as the BCG lesions healed.
the collagen fibers.At 9, 23, and 37 days, few, if The ratio of CD4 to CD8 mononuclear cells
any, collagen fibers were present in the necrotic (mainly lymphocytes) in primary BCG lesions
areas or in the adjoining areas densely infiltrated did not change appreciably during their devel-
with cells. (Both granulocytes and macrophages opment and healing (Table 1) (5).A larger per-
contain collagenases [see reference 4].) The centage of CD4 cells than CD8 cells was always
peripheral areas of BCG lesions were not present (5). (In rabbits, 5 weeks after an aerosol
included in our quantitative evaluations. Such inhalation of Mycobacterium tuberculosis strain
23. RABBIT DERMAL BCG LESIONS 䡵 357

TABLE 1 CD4 and CD8 cells in mononuclear cell populations within primary BCG lesions during their
development and healinga
Age of BCG lesion (days) % of CD4 cells % of CD8 cells Ratio CD4/CD8
9 13.2 ⫾ 1.2 8.7 ⫾ 2.1 1.7 ⫾ 0.5
23 17.8 ⫾ 1.1 5.4 ⫾ 0.9 3.9 ⫾ 0.5
37 15.7 ⫾ 2.4 6.3 ⫾ 1.6 2.9 ⫾ 0.6
a
Listed are the percentages (mean ⫾ standard error) of CD4 and CD8 cells identified in the tissue sections by immunohis-
tochemical techniques. Note that CD4 cells are more numerous than CD8 cells.Adapted from reference 5.

CDC1551 [6] or strain H37Rv [Y. C. Manabe, variety of functions, e.g., secretion, digestion of
unpublished data], single-cell suspensions of ingested materials, and/or cytokine production
lung homogenates also contained [by flow (9, 10) (see chapter 6).
cytometry] a larger percentage of CD4 cells Vasculature-cell interactions in developing
than CD8 cells.) and healing BCG lesions are presented in chap-
In 23-day primary BCG lesions, epithelioid ter 21 and in references 1 and 5.A complete his-
cells were present in the tuberculous granulation tological description of developing and healing
tissue surrounding the caseous/liquefied center, BCG lesions in Lurie’s resistant and susceptible
and fibroblasts were present in the periphery as inbred rabbits is published in reference 11.
the lesions healed. Epithelioid cells are macro-
phages that adhere to one another, so they Number of Bacilli in BCG Lesions
resemble epithelium (see chapter 6).They have Acid-fast bacilli were always more numerous
often ingested and digested tubercle bacilli where necrosis was present.At 1 to 3 days, many
(7, 8). In addition, they have differentiated for a bacilli were present in both intact macrophages

FIGURE 3 Number of bacilli in


central areas of rabbit BCG lesions of
various ages, visualized by acid-fast
staining with carbol-fuchsin. As the
lesions progressed, a shift occurred from
bacilli located intracellularly (mostly in
intact and necrotic macrophages) to
bacilli located extracellularly in areas
of (liquefied) caseous necrosis. By 23
days, the number of bacilli was greatly
reduced. Reproduced with permission
from reference 3.
358 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

FIGURE 4 Liquefied caseous


center of a 5-day dermal BCG lesion
stained for tubercle bacilli. Many
bacilli appear as clumps within
darkly stained macrophages. Other
bacilli are located extracellularly in
the liquefied debris. All darkly
stained tiny particles are bacilli. Since
this is necrotic material, the macro-
phages are not distinctly recognizable
in the photograph, but their rounded
outline and size clearly identify their
cell type. Fixed-frozen tissue sec-
tions stained by carbol-fuchsin and
counterstained with methylene blue.
Magnification, ⫻800. Reproduced
with permission from reference 3.

(and polymorphonuclear leukocytes) and in the When skin tested, the rabbits were tuber-
necrotic cells (Fig. 3).At 5 to 9 days, the number culin negative at 3 days after the BCG injection,
of bacilli seen in intact cells was much reduced, but at 9 days, they had become tuberculin pos-
but many bacilli remained in necrotic foci that itive (Fig. 1).At 14 to 23 days, tuberculin sensi-
were coalescing and liquefying (Fig. 3 and 4).At tivity frequently reached a peak and was some-
23 days, very few bacilli could be seen in intact what reduced as the BCG lesions healed (5)
tissues, and the number of bacilli in the caseous (see Fig. 5B).
necrotic center was greatly reduced (Fig. 3).At 37 After the rabbits became tuberculin positive,
days, the BCG lesions were almost healed. the BCG lesions developed distinct caseous
Homogenized dermal BCG lesions were cul- centers, which soon liquefied. In hosts possess-
tured by Lurie for viable bacilli (11). These ing delayed-type hypersensitivity, caseous necro-
bacilli multiplied for 2 weeks in the susceptible sis and liquefaction are caused by the high local
rabbits but did not multiply appreciably in the concentrations of tuberculin-like products (7)
resistant rabbits (Table 2). We did not culture from the large number of bacilli injected (see
BCG lesions produced in commercial New chapter 4).
Zealand White rabbits, but we would expect the
number of viable bacilli in them to follow the
COMPARISON OF DERMAL BCG
pattern found in Lurie’s resistant rabbits. LESIONS AND PULMONARY LESIONS
PRODUCED BY VIRULENT TUBERCLE
Tuberculin Reactions BACILLI IN RABBITS
For the tuberculin tests, rabbits were injected Dermal BCG lesions can serve as a model of
intradermally (0.1 ml per site) with an equiva- pulmonary lesions caused by fully virulent
lent to a 1:10 dilution of standard Old Tuber- tubercle bacilli. In rabbits, both types of lesion
culin (7; see also reference 3) (see glossary).The usually develop tuberculous granulation tissue,
resulting tuberculin reactions were measured caseous necrosis, liquefaction, ulceration or cav-
with calipers, as described in reference 12. Old ities, and encapsulation by fibrous tissue.Also, the
Tuberculin contains more tuberculin-like anti- same host factors control their progression and
gens than purified protein derivative and was regression.
preferred by Lurie for testing the tuberculin The main differences between dermal BCG
sensitivity of rabbits (7). lesions and those caused by fully virulent bacilli
23. RABBIT DERMAL BCG LESIONS 䡵 359

TABLE 2 Relative numbers of tubercle bacilli in dermal BCG lesions of resistant strain III rabbits and suscep-
tible strain C rabbits at various times after infectiona
Avg. no. of BCG bacilli cultured from the
No. rabbits of each Interval after BCG injection same aliquot of the entire BCG lesion
strain tested
Resistant strain III Susceptible strain C
2 1 day 48 30
2 3 days — 54
2 1 week 65 155
2 2 weeks 40 575
2 4 weeks 6 42
a
On “day zero,” 12 ⫻ 106 viable BCG bacilli were injected intradermally into each of the 20 rabbits. On the days and weeks
listed, the entire lesion was surgically removed, ground in a mortar, and cultured for viable bacilli. Note that the BCG bacilli in
the dermal lesions of the resistant strain III rabbits were greater on day 1, did not increase appreciably over time, and declined more
rapidly than the BCG bacilli in the susceptible C rabbits.These findings were apparently due to a greater initial localization of
the bacilli at the dermal site of injection, a more rapid development of delayed-type hypersensitivity and cell-mediated immu-
nity, and a more rapid ulceration (with discharge of liquefied caseum containing bacilli) in the resistant group.Adapted from ref-
erence 11.

in the lungs is that dermal BCG lesions, usually Palmer (17) did a study that supported this
started by the injection of over a million tuber- premise.They found that smaller BCG lesions
cle bacilli in one site, soon heal, whereas pul- and the rapid development of tuberculin sensi-
monary lesions, caused by inhalation of virulent tivity were, in general, associated with a greater
tubercle bacilli, begin with only 1 unit of 1 to resistance to clinical tuberculosis.
3 bacilli in a given site.
In addition, in rabbits, virulent human- and LURIE’S INTRAVENOUS BCG
bovine-type tubercle bacilli multiply to a greater EXPERIMENTS IN RABBITS
extent than BCG, and the virulent bovine-type Following the intravenous injection of 1 mg
causes progressive disease (see chapter 13). wet weight of BCG (about 5 ⫻ 106), rabbits
Mycobacteria are currently being studied by developed small lesions in many organs of the
molecular biology techniques to identify their body, including the lungs, liver, spleen, and
virulence factors (13–16). lymph nodes (7, 18, 19). By 4 weeks most of the
BCG had been destroyed, and because of the
HEALING OF DERMAL BCG development of cell-mediated immunity and
LESIONS AS A MEASURE delayed-type hypersensitivity, the BCG lesions
OF HOST RESISTANCE soon healed.
Lurie (11) used the rapid healing of dermal Among the various organs, bacillary multi-
BCG lesions to select breeders for his resistant plication varied greatly. In general, similar to
rabbits (Fig. 5A) and used the delayed healing of more virulent mycobacteria, the bacilli in the
dermal BCG lesions to select breeders for his liver multiplied the least, and those in the lung
susceptible rabbits. Resistance determined by (and spleen) multiplied the most (7, 18, 19).
this method of selection paralleled that deter- Some caseous necrosis was even present in the
mined by his tubercle-count method (see chap- tracheobronchial lymph nodes, and viable BCG
ter 11), but did not always match on an indi- occasionally persisted in these (and other) nodes
vidual basis (11). for many months.
In human populations, people whose BCG In one of these studies (7, 18), the BCG
lesions healed quickly should usually be more strain injected intravenously was somewhat
resistant to clinical tuberculosis than those whose more virulent than the strains available today,
BCG lesions healed more slowly. Meyer and but in a subsequent study, the standard BCG was
360 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

FIGURE 5 Size of dermal BCG lesions and tuberculin reactions in Lurie’s inbred re-
sistant and susceptible rabbits at various times after infection. Note that at 1 and 2 weeks,
the BCG lesions (A) were largest in resistant strain III rabbits, because their tuberculin sen-
sitivity (B) developed more quickly and to a greater degree.Then, the BCG lesions in this
resistant strain III declined rapidly and healed sooner than did those in the susceptible strain
C. Strain A rabbits were of intermediate resistance when this experiment was performed.
Reproduced with permission from reference 11.

used (7, 19). The results from each of these 6 to 40 months after intradermal BCG vacci-
experiments were similar, although, as expected, nation (usually of trauma) (20). Over half of
the more virulent strain multiplied more exten- these individuals had granulomas, most often in
sively in the various organs. the liver and/or lungs, and sometimes in the
bronchial lymph nodes, spleen, or kidneys.
SYSTEMIC NATURE OF BCG Therefore, following BCG vaccination in
VACCINATION IN HUMANS humans, the BCG bacilli are apparently widely
In Scandinavia, necropsies were performed on disseminated throughout the body and produce
20 infants, children, and young adults who died nonprogressing lesions in many organs. These
23. RABBIT DERMAL BCG LESIONS 䡵 361

studies indicate that BCG vaccination is not


confined to the intradermal site of injection
and the draining lymph nodes, but is systemic in
nature (reviewed in references 20 and 21).

INTRADERMAL BCG VACCINATION


IN LURIE’S INBRED RESISTANT AND
SUSCEPTIBLE RABBITS
The BCG Vaccine
The vaccine used in these rabbit experiments
was Phipps BCG produced by Joseph D.Aron-
son (22, 23). It was freshly prepared each week
(by techniques similar to those described by
Calmette and Guérin [24]) and used within 3
days. Each rabbit received an intradermal injec-
tion of 106 to 107 viable bacilli, the exact dose
depending on the experiment (11).
FIGURE 6 Antibody titers in inbred resistant strain
The BCG Lesions III rabbits and susceptible strain FC rabbits at various
The lesions of the resistant strain III rabbits times after an intradermal injection of BCG.The anti-
enlarged more rapidly, reached a peak more bodies were determined by the hemagglutination
method of Middlebrook (31) with Old Tuberculin as the
quickly, ulcerated more frequently, and healed antigen. Note that such antibodies reached much higher
sooner than did those of the susceptible strain titers in the resistant group.Adapted from reference 11.
C rabbits (Fig. 5A) (11). In resistant rabbits, The FC strain rabbits were similar to the C strain rab-
tuberculin sensitivity developed earlier (Fig. 5B) bits in their resistance to tuberculosis (see chapter 14).
(11), the bacilli multiplied for a shorter period
of time, the bacilli were more rapidly elimi-
nated (Table 2) (11), and antibodies were pro-
duced in higher titers (Fig. 6) (11). Histologically, ber of tubercles in the BCG-vaccinated rabbits
in resistant hosts, the tissue responses to BCG was 22% of the number of tubercles in the
were accelerated, i.e., mature epithelioid cells unvaccinated controls, if no allowance was made
(which are highly activated macrophages [9, 25, for “ratio inflation” by the Lurie method (see
26]) and plasma cells appeared sooner and in chaper 11).
greater numbers, as did fibroblasts and collagen In susceptible strain FC rabbits, the ratio was
bundles (11). increased by BCG from 107 to 124. In other
words, the average number of tubercles (similarly
The Amount of Protection determined) in the BCG-vaccinated rabbits was
Ten to 11 weeks after the BCG vaccination, 86% of the number of tubercles in the unvac-
Lurie’s resistant and susceptible rabbits were cinated controls (which was not statistically sig-
challenged by aerosol with virulent human- nificant) (Table 3) (11).
type tubercle bacilli (H37Rv) (11). Five weeks The difference in the ability of inbred resis-
after this aerosol challenge, the rabbits were tant and susceptible rabbits to be immunized
euthanized, and the grossly visible primary against tuberculosis should have been expected.
tubercles in the lungs were counted. Resistant rabbits, which are known to develop
In resistant strain III rabbits, the number of good acquired resistance during an infection
inhaled human-type bacilli (H37Rv) required to with virulent tubercle bacilli, should also develop
produce one grossly visible pulmonary tubercle good acquired resistance from immunization
(“the ratio”) was increased by BCG from 642 to with BCG.This acquired resistance from BCG
2,948 (by the Lurie method of calculating ratios) would then protect them substantially against an
(Table 3) (11). In other words, the average num- aerosol challenge with virulent tubercle bacilli.
362 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

TABLE 3 Number of inhaled human-type tubercle bacilli producing one grossly visible primary pulmonary
tubercle in unvaccinated and BCG-vaccinated inbred resistant and susceptible rabbits (“the ratio” by the Lurie
method)a
A B A B
Unvaccinated Vaccinated Unvaccinated Vaccinated
resistant resistant susceptible susceptible
strain III strain III strain FC strain FC
No. of rabbits 8 8 12 9
No. of inhaled bacilli 642 ⫾ 107 2,948 ⫾ 914 107 ⫾ 200 124 ⫾ 18
producing one visible (536) (1,073) (69) (98)
tubercle (ratios)
Amount of protection: 22% 86%
no. of visible tubercles P ⫽ 0.013 P ⫽ 0.270
in the vaccinated rabbits
compared to the no.
in the controls
a
The rabbits were vaccinated intradermally with 1.6 ⫻ 106 Phipps strain BCG.Ten to 11 weeks later, they were challenged
by aerosol with 4,000 to 12,000 virulent human-type tubercle bacilli (H37Rv) inhaled per rabbit. Five weeks after challenge, the
rabbits were euthanized, and the grossly visible primary pulmonary tubercles were counted.The vaccinated strain III rabbits were
paired with vaccinated strain FC rabbits and infected with H37Rv at the same times. However, the data for the unvaccinated rab-
bits were assembled mostly from experiments in which the resistant and susceptible rabbits were infected with H37Rv at differ-
ent times (11). Note that BCG vaccination was much more effective in resistant strain III rabbits than in the susceptible strain FC
rabbits (see text).The ratios were determined by the Lurie method, which inflates the higher ratios but is the best way to pool
data from experiments differing in inhaled bacillary dosages (see chapter 11).The numbers in parentheses are noninflated ratios
derived from averages (see chapter 11).Adapted from reference 11.

TABLE 4 Vaccine efficacy: number of grossly visible tubercles in vaccinated rabbits and their diameters as per-
centages of those found in controlsa
No. of grossly visible tubercles as a Diameters of tubercles as a percentage
Vaccine percentage of the no. in controls of those in controls
BCG
Danish 54, 69, 64 74, 86, 78
Japanese 48, 61 77, 83
Tice 65 77
Pasteur 13, 52 85, 76
M. microti
OV 254 40, 25, 22, 61 81, 62, 80, 69
NCO 8712 56 65
ATCC 35781 62 77
ATCC 35782 65 79
ATCC 11152 29, 20 96, 81
ATCC 19422 73 72
a
This table summarizes the results of numerous experiments, each containing 6 to 8 commercially available outbred New Zealand
White rabbits in the control and vaccinated groups. Five weeks after these rabbits inhaled virulent human-type tubercle bacilli
(H37Rv), the animals were euthanized, and the grossly visible primary tubercles in their lungs were counted. Note that every vac-
cine evaluated decreased both the tubercle count and the tubercle size. However, because of the variability among outbred rab-
bits, no vaccine proved consistently better than the others.Adapted from reference 29, with some unpublished experiments added
from Yukari C. Manabe.
23. RABBIT DERMAL BCG LESIONS 䡵 363

In contrast, susceptible rabbits, which are known these rabbits (rather than 6 to 8) in each cate-
to develop poor acquired resistance during an gory of a given experiment would be required
infection with virulent bacilli, should also to obtain statistical significance, if any exists.
develop poor acquired resistance from BCG, The reduction in tubercle size did not seem to
and therefore, benefit less from the immuniza- be correlated with the reduction in the number
tion. In other words, BCG vaccination helped of tubercles (29). Also, the number of bacilli
the most the hosts that needed it the least; con- inhaled seemed to have no effect on the size of
versely, it helped the least the hosts that needed the resulting tubercles (29).
it the most. In human populations, no one BCG prepa-
These findings are just another example of ration was found to be more efficacious than any
Lurie’s cardinal principle: “Acquired resistance other (30). However, human trials cannot be
is superimposed upon and determined by native standardized for variables as carefully as those
resistance” (7). Recent studies, especially those performed in laboratory animals.
by the Janeway group (27, 28), provide insight
into some of the mechanisms by which innate
REFERENCES
resistance affects the development of acquired re-
1. Abe,Y., K. Sugisaki, and A. M. Dannenberg,
sistance (see chapter 5). Jr. 1996. Rabbit vascular endothelial adhesion mol-
If these results are applied to human popula- ecules: ELAM-1 is most elevated in acute inflam-
tions, tuberculosis vaccines would be most effec- mation, whereas VCAM-1 and ICAM-1 predom-
tive in people who are able to produce a good inate in chronic inflammation. J. Leukoc. Biol.
immune response,which fortunately is the major- 60:692–703.
2. Bartholomew, J.W. 1981. Stains for microorgan-
ity of the immunocompetent persons living today. isms in sections, p. 441–473. In G. Clark (ed.), Stain-
Such vaccines would be less effective in people ing Procedures, 4th ed. Lippincott Williams & Wilkins,
who are unable to mount a good immune Philadelphia, Pa.
response, e.g., those treated with immunosup- 3. Sugisaki, K., A. M. Dannenberg, Jr., Y. Abe,
pressive agents and those infected with J. Tsuruta, W.-J. Su, W. Said, L. Feng,
T.Yoshimura, P. J. Converse, and P. Mounts.
HIV/AIDS.Such persons need the protection the 1998. Nonspecific and immune-specific up-
most, but vaccination with BCG would proba- regulation of cytokines in rabbit dermal tubercu-
bly help them relatively little. In fact, live vaccines lous (BCG) lesions. J. Leukoc. Biol. 63:440–450.
may be dangerous to give to such persons. 4. Woessner, J. F., Jr., A. M. Dannenberg, Jr.,
P. J. Pula, M. G. Selzer, C. L. Ruppert,
K. Higuchi, A. Kajiki, M. Nakamura, N. M.
EFFICACIES OF BCG AND M. MICROTI Dahms, J. S. Kerr, and G.W. Hart. 1990. Extra-
VACCINES IN COMMERCIAL NEW cellular collagenase, proteoglycanase and products
ZEALAND WHITE RABBITS of their activity, released in organ culture by intact
We have recently used the tubercle-count dermal inflammatory lesions produced by sulfur
method (see chapter 11) to evaluate the efficacy mustard. J. Investig. Dermatol. 95:717–726.
5. Shigenaga, T., A. M. Dannenberg, Jr., D. B.
of various strains of BCG and M. microti (the vole Lowrie, W. Said, M. J. Urist, H. Abbey, B. H.
bacillus) in commercial New Zealand White Schofield, P. Mounts, and K. Sugisaki. 2001.
rabbits (29). All of the vaccines tested so far Immune responses in tuberculosis: antibodies and
reduced the average number of pulmonary CD4-CD8 lymphocytes with vascular adhesion
tubercles found 5 weeks after the inhalation of molecules and cytokines (chemokines) cause a rapid
antigen-specific cell infiltration at sites of bacillus
virulent human-type bacilli (H37Rv) and Calmette-Guérin reinfection. Immunology 102:466–
decreased the average diameter of these tuber- 479.
cles (Table 4) (29). However, the number of 6. Kesavan, A. K., S. Mendez, C. L. Hatem,
tubercles produced in these outbred rabbits was J. Lopez-Molina, M. Brooks, R. Fujiwara,
so variable that we could not determine which K. Aird, M. L. M. Pitt, A. M. Dannenberg, Jr.,
and Y. C. Manabe. 2005. Effects of dexamethasone
vaccine was significantly better than the others. and transient malnutrition on rabbits infected with
The late Professor Helen Abbey of our Biosta- aerosolized Mycobacterium tuberculosis CDC1551.
tistics Department calculated that about 18 of Infect. Immun. 73:7056–7060.
364 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

7. Lurie, M. B. 1964. Resistance to Tuberculosis: Exper- bacilli in the organs of tuberculous rabbits. J. Exp.
imental Studies in Native and Acquired Defensive Mech- Med. 55:31–54.
anisms. Harvard University Press, Cambridge, Mass. 19. Lurie, M. B. 1934.The fate of BCG and associ-
8. Ando, M., A. M. Dannenberg, Jr., M. Sugi- ated changes in organs of rabbits. J. Exp. Med.
moto, and B. S. Tepper. 1977. Histochemical 60:163–178.
studies relating the activation of macrophages to the 20. Gormsen, H. 1956. On the occurrence of epithe-
intracellular destruction of tubercle bacilli. Am. J. lioid cell granulomas in the organs of BCG-
Pathol. 86:623–634. vaccinated human beings. Acta Pathol. Microbiol.
9. Suga, M., A. M. Dannenberg, Jr., and Scand. Suppl. 111:117–120.
S. Higuchi. 1980. Macrophage functional hetero- 21. Lindgren, I. 1961. Anatomical and roentgeno-
geneity in vivo: macrolocal and microlocal logic studies of tuberculous infections in BCG-
macrophage activation, identified by double-staining vaccinated and non-vaccinated subjects, with
tissue sections of BCG granulomas for pairs of biophysical investigations of calcified foci. Acta
enzymes. Am. J. Pathol. 99:305–324. Radiol. 209(Suppl):1–101.
10. Nathan, C. F. 1987. Secretory products of 22. Aronson, J. D., E. I. Parr, and R. M. Saylor.
macrophages. J. Clin. Investig. 79:319–326. 1940. BCG vaccine. Its preparation and the local
11. Lurie, M. B., P. Zappasodi, E. Cardona-Lynch, reaction to its injection. Am. Rev.Tuberc. 42:651–666.
and A. M. Dannenberg, Jr. 1952.The response 23. Aronson, J. D., C. F. Aronson, and H. C.Tay-
to the intracutaneous inoculation of BCG as an lor. 1958. A twenty-year appraisal of BCG vacci-
index of native resistance to tuberculosis. J. Immunol. nation in the control of tuberculosis. Arch. Intern.
68:369–387. Med. 101:881–893.
12. Dannenberg, A. M., Jr., M. Ando, and 24. Calmette, A., and C. Guérin. 1924.Vaccination
K. Shima. 1972. Macrophage accumulation, divi- des bovides contre la tuberculose et méthode nou-
sion, maturation and digestive and microbicidal velle de prophylaxie de la tuberculose bovine. Ann.
capacities in tuberculous lesions. III.The turnover Inst. Pasteur 38:371–398.
of macrophages and its relation to their activation 25. Dannenberg, A. M., Jr., O. T. Meyer, J. R.
and antimicrobial immunity in primary BCG Esterly, and T. Kambara. 1968.The local nature
lesions and those of reinfection. J. Immunol. of immunity in tuberculosis, illustrated histochem-
109:1109–1121. ically in dermal BCG lesions. J. Immunol. 100:931–
13. Manabe, Y. C., A. M. Dannenberg, Jr., and 941.
W. R. Bishai. 2000.What can we learn from the 26. Dannenberg, A. M., Jr. 1968. Cellular hyper-
Mycobacterium tuberculosis genome sequencing pro- sensitivity and cellular immunity in the patho-
jects? Int. J.Tuberc. Lung Dis. 4:S18–S23. genesis of tuberculosis: specificity, systemic and local
14. Brodin, P., C. Demangel, and S.T. Cole. 2005. nature, and associated macrophage enzymes. Bacte-
Introduction to functional genomics of the Mycobac- riol. Rev. 32:85–102.
terium tuberculosis complex, p. 143–153. In S.T. Cole, 27. Janeway, C. A., Jr., P.Travers, M.Walport, and
K. D. Eisenach, D. N. McMurray, and W. R. Jacobs, M. J. Shlomchik. 2001. Immunobiology: the Immune
Jr. (ed.), Tuberculosis and the Tubercle Bacillus. ASM System in Health and Disease, 5th ed. Garland Pub-
Press,Washington, D.C. lishing, New York, N.Y.
15. Brosch, R., and M. A. Behr. 2005. Comparative 28. Medzhitov, R., and C. Janeway, Jr. 2000. Innate
genomics and evolution of Mycobacterium bovis immunity. N. Engl. J. Med. 343:338–344.
BCG, p. 155–164. In S. T. Cole, K. D. Eisenach, 29. Dannenberg, A. M., Jr., W. R. Bishai, N. Par-
D. N. McMurray, and W. R. Jacobs, Jr. (ed.), Tuber- rish, R. Ruiz, W. Johnson, B. C. Zook, J. W.
culosis and the Tubercle Bacillus.ASM Press,Washing- Boles, and M. L. M. Pitt. 2000. Efficacies of
ton, D.C. BCG and vole bacillus (Mycobacterium microti) vac-
16. Goulding, C. W., D. Pal, and D. Eisenberg. cines in preventing clinically apparent pulmonary
2005.TB or not TB: a structural genomics mission, tuberculosis in rabbits: a preliminary report. Vaccine
p. 165–179. In S. T. Cole, K. D. Eisenach, D. N. 19:796–800.
McMurray, and W. R. Jacobs, Jr. (ed.), Tuberculosis and 30. Milstien, J. B., and J. J. Gibson. 1990. Quality
the Tubercle Bacillus.ASM Press,Washington, D.C. control of BCG vaccine by WHO: a review of fac-
17. Meyer, S. N., and C. E. Palmer. 1954. Response tors that may influence vaccine effectiveness and
to BCG vaccination as an index of familial suscep- safety. Bull.W. H. O. 68:93–108.
tibility to tuberculosis. Am. Rev. Tuberc. 69:383– 31. Middlebrook, G. 1950.A hemolytic modification
395. of the hemagglutination test for antibodies against
18. Lurie, M. B. 1932. The correlation between the tubercle bacillus antigens. J. Clin. Investig. 29:1480–
histological changes and the fate of living tubercle 1485.
Section 8.

PAST, PRESENT, AND FUTURE


This page intentionally left blank
24
SUMMARY AND CONCLUSIONS

Part I. Summary [367]


Host-parasite interactions [367]
Stages in the pathogenesis of tuberculosis [367]
Role of DTH and CMI in the pathogenesis of tuberculosis [368]
Part II. Conclusions [370]
Principles of host-parasite interaction established by this
disease [370]
Application to other infectious diseases [372]

PART I. SUMMARY The vulnerabilities of the host are:


HOST-PARASITE INTERACTIONS 1. Nonactivated macrophages that provide a
The pathogenesis of tuberculosis can be con- favorable environment for the intracellu-
sidered a series of battles between the host and lar growth of the bacillus
the tubercle bacillus, each of which has its own 2. Liquefied caseous material, the only men-
weapons that can be used against the other. In struum in the host that can support the
addition, both the host and the bacillus have sites extracellular growth of the bacillus
of vulnerability that can be exploited by the
adversary (1). The vulnerabilities of the bacillus are:
The weapons of the host are: 1. The inability to survive within a fully
1. Cell-mediated immunity (CMI), which activated macrophage
activates macrophages so that they can 2. The inability to multiply in solid caseous
kill or inhibit tubercle bacilli that they tissue
ingest Table 1 correlates the multiplication of the
2. Delayed-type hypersensitivity (DTH), bacilli with the type of disease and the immune
which stops the intracellular growth of response of the host.
bacilli in nonactivated macrophages by
killing these macrophages. DTH thereby STAGES IN THE PATHOGENESIS
transforms an environment that is favor- OF TUBERCULOSIS
able for the bacillus into an environment See chapter 2 and reference 2.An inhaled bacil-
that is inhibitory, i.e., solid caseous tissue. lary unit of 1 to 3 bacilli will reach the alveo-
lar spaces and will be ingested by an alveolar
The weapons of the bacillus are:
macrophage (Stage I). In humans and rabbits,
1. The ability to multiply intracellularly in most of these macrophages seem to be acti-
nonactivated macrophages, i.e., within the vated sufficiently to destroy the bacillary unit
monocytes/macrophages that emigrate before any lesion is produced. However, if the
from the bloodstream into the tissues at alveolar macrophage is not sufficiently activated,
the sites of the infection the ingested bacilli multiply and disrupt the
2. The ability to multiply extracellularly in alveolar macrophage.
the liquefied caseum of a cavity wall, The released bacilli are then ingested by non-
sometimes reaching tremendous numbers activated monocytes/macrophages that are

367
368 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

TABLE 1 Multiplication of tubercle bacilli, the type of disease, and the host immune responsea
No multiplication of virulent tubercle bacilli occurs in the majority of human and rabbit pulmonary
alveolar macrophages, because they are already highly activated.
Disease: No disease.
Immune response: No cell-mediated immunity (CMI) and no delayed-type hypersensitivity (DTH) or
conversion of the tuberculin skin test.
Bacilli are dormant in the solid caseous center of tuberculous lesions. (In humans the bacilli may remain
dormant in solid caseum for over 30 years.)
Disease: Arrested disease. Bacilli that escape from the edge of the caseum are soon ingested and, in resistant
hosts, are inhibited by nearby highly activated macrophages.
Immune response: Good CMI and (in humans) strong or weak DTH, depending on the extent of residual
disease and the amount of bacillary growth and destruction at the edge of the caseum.
Intracellular multiplication of bacilli occurs in poorly activated macrophages surrounding the solid caseous
center.Tissue-damaging DTH kills these macrophages. (Intracellular bacillary multiplication also occurs in
nonactivated macrophages before DTH and CMI develop.)
Disease: Progressive disease. If only poorly activated macrophages are nearby, the solid caseous center
enlarges, and hematogenous and lymphatic spread of bacilli occurs.
Immune response: Poor CMI, but often good DTH, i.e., tuberculin sensitivity (except terminally).
Extracellular multiplication of bacilli occurs in liquefied caseum, especially in the liquefied caseum lining the
walls of cavities. (Coughing spreads these extracellular bacilli into the environment.)
Disease: Bronchial spread of bacilli, causing new pulmonary lesions and sometimes bronchopneumonia.
Immune response: Good CMI (which may be overwhelmed by a large number of bacilli), and usually good
DTH (except terminally).
a
Revised from reference 1.

chemotactically attracted there from the blood- more, in the same lung, some lesions may
stream. In these phagocytes the bacilli grow progress while other lesions may regress.Tuber-
logarithmically (Stage II). Lurie called this the culosis is a locally controlled disease that depends
stage of symbiosis, because both macrophages on the growth of bacilli in nonactivated
and tubercle bacilli increase in the developing macrophages or in liquefied caseum, or on the
lesion, with no apparent injury to either. inhibition of bacilli in activated macrophages or
When the host becomes tuberculin positive, in solid caseum.
a caseous center may develop in the lesion
(Stage III).A lesion with a small caseous center ROLE OF DTH AND CMI IN THE
(up to 2 mm in diameter) may heal or stabilize PATHOGENESIS OF TUBERCULOSIS
before it is detectable by radiography. A larger See chapter 5. Tissue-damaging DTH will
caseous lesion may enlarge and shed bacilli into directly or indirectly kill nonactivated macro-
the blood and lymph (Stage IVa), or it may heal phages that have permitted more than a few
or stabilize (Stage IVb). tubercle bacilli to grow in their cytoplasm.
Alternatively, a caseous lesion may liquefy (These bacilli apparently release high local con-
and form a cavity (Stage V). Now, the bacilli can centrations of tuberculin-like products.) The
grow extracellularly (for the first time), enter the killing of such macrophages eliminates the
bronchial tree, and spread to other parts of the intracellular environment that is favorable to
lung and to the environment. bacillary growth and replaces it with the extra-
In general, each stage of tuberculosis is won cellular environment of solid caseous tissue that
by the host with increasing difficulty. Further- is inhibitory to bacillary growth. The bacillus
24. SUMMARY AND CONCLUSIONS 䡵 369

itself is nontoxic; it damages host tissue mainly The resistant rabbits developed strong immunity
by means of the host’s own immune response (CMI) that activated macrophages to destroy
to its tuberculin-like products. or inhibit tubercle bacilli.The susceptible rab-
CMI activates macrophages so that they can bits developed only weak CMI that often was
kill or inhibit ingested tubercle bacilli.The bacil- incapable of activating macrophages sufficiently
lus does not multiply within highly activated to control the growth of the bacilli.Therefore,
macrophages.Therefore, to arrest this disease, the in Fig. 1, the complete parallelism found in the
host must produce many activated macrophages bacillary growth curves between the two strains
at sites where the bacilli lodge. of rabbits was totally unexpected.
To control bacillary multiplication, both CMI This riddle was solved by the realization that
and tissue-damaging DTH are required. This two methods of controlling bacillary growth
statement was proved by correlating bacillary exist: CMI and tissue-damaging DTH. After
growth curves (Fig. 1) with the observed gross tubercle bacilli are inhaled, they are ingested
pathology and histopathology (see chapter 2). by alveolar macrophages. The alveolar macro-
Lurie produced these growth curves by infect- phages of resistant rabbits are apparently more
ing susceptible and resistant inbred rabbits with highly (nonspecifically) activated and therefore
aerosolized virulent tubercle bacilli and cultur- destroy or inhibit tubercle bacilli more effec-
ing the lungs for viable bacilli at different times tively than those of susceptible rabbits.Therefore,
during the course of the resulting disease (3, 4). at 7 days after infection, there is about a 20- to

FIGURE 1 Changes in the number of human-type tubercle bacilli in the lungs of


Lurie’s natively resistant and natively susceptible rabbits at different intervals after aerosol
infection. By 7 days after infection, the resistant rabbits had inhibited the growth of the
bacilli 20 to 30 times more effectively than did the susceptible rabbits, but, from then
on, the two curves were parallel.At 4 to 5 weeks, the lungs of susceptible rabbits con-
tained about 13 times the number of primary pulmonary tubercles that the lungs of re-
sistant rabbits contained.The means and their standard errors are shown. (The y axis of
this figure is on a log scale.) Reproduced with permission from reference 3.
The number of tubercle bacilli in the lungs of the resistant rabbits failed to decrease
during the period illustrated, because liquefaction with extracellular multiplication of
the bacillus readily occurred in these rabbits. Liquefaction did not occur in the susceptible
rabbits, possibly because only low levels of hydrolytic enzymes developed in their
macrophages.
The logarithmic and stationary phases of this bacillary growth curve also occur in mice
and guinea pigs, following the inhalation of virulent tubercle bacilli (see chapter 15).
370 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

30-fold difference in the number of bacilli in the bits), liquefaction may begin 8 weeks after an
lungs of the two strains of rabbits (Fig. 1). aerosol infection with fully virulent bovine-
From this point on, however, the bacillary type tubercle bacilli, whereas in the susceptible
growth curves in resistant and susceptible rabbits rabbits, liquefaction does not occur.
follow a parallel course. In both rabbit strains, dur- All of these principles developed in the rab-
ing the logarithmic stage of symbiosis, the bacilli bit model seem applicable to the pathogenesis
apparently multiply in nonactivated macrophages of tuberculosis in humans. The type of tuber-
without any inhibition. Also, the logarithmic culosis described in the resistant rabbits resem-
stage stops in both rabbit strains at the same time, bles that found in immunocompetent adults.The
because each rabbit strain develops tuberculin type of tuberculosis described in the susceptible
sensitivity (DTH), which kills the nonactivated rabbits resembles that found in infants and
macrophages in which the bacilli are multiplying. immunocompromised adults.
The susceptible rabbits often had tuberculin reac- The various chapters of this book provide
tions comparable in size to those of the resistant more details on the pathogenesis and immunol-
rabbits, because the immune system of the sus- ogy of tuberculosis, on the role of antigen-
ceptible rabbits (although weaker) had been stim- presenting cells, macrophages, and lymphocytes,
ulated by 20 to 30 times the quantity of bacillary and on the role of antibodies. Chapters on the
antigens. Such antigens are released by the bacilli vascular endothelium, cytokines, and hormones
growing within poorly activated macrophages. are included, as well as chapters on improving
Such antigens would also stimulate CMI, but vaccines. Chapter 15 compares the characteris-
the bacillary load is too high in the susceptible tics of tuberculosis found in rabbits, mice, and
rabbits, and their ability to produce CMI (acti- guinea pigs.
vated macrophages) is too weak, to effectively For a quick overview of the entire book, the
arrest the progress of the lesion. reader is referred to appendix F, where the
Subsequently, the two bacillary growth curves abstracts of these chapters are collected.
still follow a parallel course (Fig. 1), but for dif-
ferent reasons. In the susceptible rabbits, bacilli PART II. CONCLUSIONS
escape from the edge of the caseum and are
ingested by poorly activated macrophages.Again, PRINCIPLES OF HOST-PARASITE
intracellular multiplication of the bacilli occurs, INTERACTION ESTABLISHED BY
THIS DISEASE
and, again, the poorly activated macrophages
The following list succinctly presents some of
harboring these bacilli are killed by tissue-
the principles described above and adds addi-
damaging DTH. The process is repeated, and
tional ones.
much of the surrounding lung tissue is
destroyed. Nevertheless, this strategy is effec- 1. Tuberculosis is controlled locally at each
tive for months, because the total number of site where the bacillus resides in the host.
viable bacilli in the lung does not increase. 2. Local DTH and CMI are both needed to
In the resistant rabbits, bacilli escaping from arrest the progress of this disease.
the edge of the caseum are often ingested by 3. Tissue-damaging DTH kills overloaded
highly activated macrophages that surround the macrophages in which tubercle bacilli are
caseum.These activated macrophages ingest and multiplying intracellularly when the
inhibit or destroy the bacilli, so that the disease tuberculin-like products locally reach high
would be arrested at this time were it not for the concentrations.The bacillus does not mul-
process of liquefaction and cavity formation. tiply in the resulting solid caseous tissue.
In the liquefied caseum, tubercle bacilli can 4. CMI (via the cytokines of Th1 lympho-
grow extracellularly for the first time, and at such cytes) activates local macrophages, so that
sites they cannot be reached, even by highly these macrophages can now inhibit
activated macrophages. In the resistant rabbits and/or kill the tubercle bacilli that they
(and in commercial New Zealand White rab- phagocytize.
24. SUMMARY AND CONCLUSIONS 䡵 371

TABLE 2 General characteristics of acute and chronic bacterial infectionsa


Acute septicemic infections, Chronic granulomatous
Characteristics such as pneumococcal infections, such as
pneumonia tuberculosis
1. Rate of accumulation of ⫹⫹⫹⫹ ⫹
bacteria in tissues
2. Endotoxins (lipopolysaccharides) ⫹⫹⫹⫹ ⫾
from gram-negative bacteria
and similarly acting components
of gram-positive bacteriab that
can cause local necrosis, blood
vessel damage, and systemic
shock, as well as terminal
irreversibility of the
disease process
3. DTH, which can cause local ⫾ ⫹⫹⫹⫹
necrosis and systemic toxicity
4a. Bacterial intracellular None in PMN and macrophages Only in nonactivated macrophages
multiplication
4b. Bacterial extracellular In tissues and blood In the liquefied caseum of
multiplication pulmonary cavities
5. Predominant type of
phagocyte:
granulocytes (PMN) ⫹⫹⫹⫹ ⫾
mononuclear cells ⫹ ⫹⫹⫹
(lymphocytes and macrophages)
6. Importance of circulating ⫹⫹⫹⫹ ⫾
antibodies in recovery of the
host
7. Degree of resistance to Completely resistant to reinfection Incompletely resistant to
exogenous or endogenous with the same strain of reinfection with M. tuberculosis
reinfection pneumococcus
8. Frequency of reactivation of ⫾ ⫹⫹⫹
the primary infection
9. Frequency of symptom-free ⫾ ⫹⫹⫹
inapparent infection that
increases antigen-specific
acquired resistance
a
Exceptions exist.
b
For example, peptidoglycans, teichoicated peptidoglycans, lipoteichoic acid, and phosphorylcholine attached to teichoic acids
(5, 6).

5. Antigen-antibody reactions (by produc- reinfection may be stopped at an earlier


ing chemotactic complement components) time, i.e., while they are still microscopic
hasten the local accumulation of both in size. However, without an expanded
macrophages and antigen-specific Th1 cells Th1 cell population (produced by vac-
at sites of reinfection. In this manner, anti- cines or a primary infection), antibodies
gen-antibody reactions greatly enhance would have little effect on the progress of
local cell-mediated immune response (i.e., the disease.
DTH and CMI) wherever reinfecting 6. The amount of cell infiltration is under
tubercle bacilli lodge. Because of circulat- strict control. Apparently, after sufficient
ing antibodies, the progression of lesions of lymphocytes and macrophages have
372 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

entered the site of bacillary multiplica- mycotic infections. Most infectious diseases are
tion, the production of chemokines (and intermediate between these two extremes,
probably other chemotaxins) is down- because they possess some of the characteristics
regulated. of both the acute and the chronic types.
7. Liquefaction of solid caseous tissues Of course, disease produced by any given
enables the bacillus to multiply extracel- microorganism is not always typical and may
lularly for the first time, often reaching vary across the spectrum. However, the structure
such large numbers that even strong presented in Table 2 can serve as a format to
acquired resistance may be overwhelmed. which many of the attributes of most infec-
Bronchial walls are eroded by tissue- tious diseases can be assigned.
damaging DTH, and the bacilli spread
through the airways to other parts of the
lung and into the environment, where REFERENCES
they may be inhaled by other persons. 1. Dannenberg, A. M., Jr. 1999. Pathophysiology:
basic aspects. I. Pathogenesis of tuberculosis. II.
8. In humans, antimicrobial-resistant tuber- Immunology of tuberculosis, p. 17–47. In D. Schloss-
cle bacilli mostly occur within the bacil- berg (ed.), Tuberculosis and Nontuberculous Mycobac-
lary population that grows profusely in the terial Infections, 4th ed. The W. B. Saunders Co.,
liquefied caseum of the cavity wall. Philadelphia, Pa.
9. The prevention of liquefaction and cav- 2. Dannenberg, A. M., Jr. 1993. Immunopatho-
genesis of pulmonary tuberculosis. Hosp. Pract.
ity formation should greatly reduce the 28:33–40 (Off. ed. 51–58).
progress of tuberculosis in immunocom- 3. Lurie, M. B., P. Zappasodi, and C. Tickner.
petent hosts and greatly reduce the spread 1955. On the nature of genetic resistance to tuber-
of tuberculosis to other individuals. culosis in the light of the host-parasite relationships
in natively resistant and susceptible rabbits. Am.
APPLICATION TO OTHER Rev.Tuberc. Pulmon. Dis. 72:297–329.
INFECTIOUS DISEASES 4. Allison, M. J., P. Zappasodi, and M. B. Lurie.
1962. Host-parasite relationships in natively resistant
Acute and chronic bacterial infections show a and susceptible rabbits on quantitative inhalation
spectrum of host responses. At one end of the of tubercle bacilli: their significance for the nature
spectrum is typical pneumococcal pneumonia, a of genetic resistance. Am. Rev. Respir. Dis. 85:553–
rapidly progressing acute disease somewhat like 569.
anthrax and plague (Table 2).At the other end of 5. Weber, J. R., P. Moreillon, and E. I.Tuomanen.
2003. Innate sensors for Gram-positive bacteria.
the spectrum is typical adult-type tuberculosis, a Curr. Opin. Immunol. 15:408–415.
slowly progressing chronic disease somewhat like 6. McCullers, J. A., and E. I. Tuomanen. 2001.
chronic brucellosis and some of the chronic Molecular pathogenesis of pneumococcal pneu-
monia. Front. Biosci. 6:d877–d889.
25
SUGGESTED FUTURE RESEARCH
AND UNANSWERED QUESTIONS

Part I. Suggested research on the host [373]


Local control of the host response [373]
Organ resistance and its implications [374]
Ideal ratio of DTH to CMI [374]
Role of vascular thrombosis in causing caseous necrosis and
inhibition of bacillary growth [375]
Role of granulocytes in tuberculosis [376]
Formation of liquefied caseum and cavities and their
prevention [376]
Prognostic tests that reflect disease progression or
regression [377]
Comparisons of tuberculous lesions produced by live and dead
tubercle bacilli [378]
Role of antibodies in host resistance to reinfection [378]
New resistant and susceptible rabbit strains [378]
Part II. Suggested research on the bacillus [378]
Bacillary survival and virulence [378]
Part III. Suggested research on prophylactic vaccines and
immunotherapy [380]
More effective vaccines [380]
Immunotherapy to supplement antimicrobial therapy and to treat
multidrug-resistant tuberculosis [380]
Other vaccine possibilities [380]
Part IV. Development of new drugs for the treatment and prevention of
tuberculosis [380]
Drugs acting on the host [380]
Drugs acting on the bacillus [380]
Part V. Sources of additional information [382]

PART I. SUGGESTED RESEARCH differentiation? How many macrophages carry


ON THE HOST out multiple functions, and how many are spe-
LOCAL CONTROL OF THE cialized for only a single function? What cells
HOST RESPONSE direct the local differentiation of macrophages?
The cells participating in tuberculous lesions How do the parenchymal cells in different organs
show a heterogeneity of functions, even among and the type of infiltrating cell affect macrophage
cells of identical appearance (see chapter 6). One differentiation?
macrophage may differentiate in one direction, What cytokines are produced by each type of
e.g., to produce reactive oxygen intermediates infiltrating cell? How are such cytokines upreg-
(ROIs) and reactive nitrogen intermediates ulated and downregulated? How do these
(RNIs) to kill bacilli, while a nearby macrophage cytokines affect each other? What cells have
may differentiate in another direction, e.g., to receptors for these cytokines, and how are these
secrete interleukin-12 that stimulates lymphocytes receptors upregulated and downregulated?
to produce Th1-type cytokines. What factors These questions on macrophage differentia-
control each of the many types of macrophage tion would apply to the dendritic cells (DCs) in

373
374 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

tuberculous lesions. In addition, we do not know tors for antigen presentation) that activate the
how many DCs are actually present there, how infiltrating cells in different ways (see chapter 5).
they interact with the other cells, and how they The cytokines and cytokine receptors that
differentiate into their various functional types are involved in the immune response within
that determine the type of immune response. See different organs should be identified by molec-
chapter 6 for more details on dendritic cells. ular biology techniques. Liver and kidney cells
The interactions that occur within a tuber- from rabbits and guinea pigs could be cultured.
culous lesion are complex. For example, These cells could be stimulated with lipopolysac-
macrophages and lymphocytes can be stimula- charides, or tuberculin, or live tubercle bacilli
tory or inhibitory, depending on their local with unstimulated cells as controls. Cytokine
environment (discussed in reference 1).We can proteins could be assayed by immunological
get some insights from cell culture experiments, techniques, and cytokine mRNAs could be
but such experiments are not equivalent to assayed by reverse transcription-PCR techniques.
those made in vivo. Cell cultures have no con- Immunohistochemical and/or in situ hybridiza-
stant influx and egress of participating cells, no tion technology could also be applied to tissue
continuous fresh blood supply to provide nutri- sections of tuberculous lesions from each organ.
ents and removal of waste products, and no nat- Knowledge of the cytokines and the cofactors
ural cell–extracellular matrix interactions. His- for antigen presentation that prevent or enhance
tochemistry (including immunocytochemical the development of tuberculous lesions in each
and in situ hybridization procedures) can pro- organ might lead to new therapeutic agents for
vide some answers, but histochemistry is limited the control of this disease. For example, if the kid-
by the availability of reagents and by what fac- ney cytokines of the guinea pig can prevent the
tors remain localized in the tissue section and progress of tuberculous lesions, then pharmaco-
are not destroyed during its preparation. Are logical agents to increase the production of such
frozen sections necessary? Would cold-embed- inhibitory cytokines might be found.
ding in glycol methacrylate (2, 3) preserve fac-
tors not preserved in paraffin sections? Would
IDEAL RATIO OF DTH TO CMI
lyophilization (4) of tuberculous lesions before
embedding preserve more of these factors? A (i) Definitions and Goals. As discussed
variety of fixation and embedding techniques in chapters 2, 5, and 15, DTH and CMI play
exist (4). Some of these techniques may preserve major roles in the pathogenesis of tuberculosis.
certain enzymes, cytokines, and other tissue With DTH, macrophages containing too many
and bacillary components better than other tubercle bacilli for CMI to control are killed.
techniques. With CMI, lymphocytes produce cytokines that
activate macrophages to destroy or inhibit tuber-
ORGAN RESISTANCE AND cle bacilli. Both DTH and CMI are mediated by
ITS IMPLICATIONS Th1 lymphocytes and their cytokines.
In rabbits, tubercles in the liver rarely progress to Within tuberculous lesions, the main differ-
grossly visible size, but tubercles in the kidneys ence between DTH and CMI is the amount of
do so (5). In guinea pigs, tubercles in the liver host sensitivity to the bacillary products that
continue to progress, but tubercles in their kid- elicit each response.The local tissue-damaging
neys never do so (5).These facts are puzzling, but DTH (which kills macrophages containing
insight into their causes may provide new numerous tubercle bacilli) is caused by low con-
approaches to the control of tuberculosis. The centrations of tuberculin-like products, whereas
local conditions surrounding the infiltrating den- the local activation of macrophages (CMI) is
dritic cells, macrophages, and lymphocytes deter- apparently caused by higher concentrations of
mine how these cells function. Specifically, liver other bacillary products (see chapter 5).Tuber-
cells and kidney cells in various species may culin in very low concentrations within tuber-
produce different cytokines (and different cofac- culous lesions can activate macrophages without
25. SUGGESTED FUTURE RESEARCH 䡵 375

necrosis (6).Therefore, the sensitivity of the host cle bacilli multiply in nonactivated macrophages
to each bacillary antigen determines whether and in weakly activated macrophages, but tuber-
that antigen produces a local CMI or a local tis- cle bacilli are inhibited (and even killed) both in
sue-damaging DTH response. strongly activated macrophages and in solid
An ideal ratio of DTH to CMI would be one caseous tissues.However,we do not know the rel-
that produces the least amount of tissue damage ative importance of activated macrophages (CMI)
with the greatest destruction (or inhibition) of and solid caseum (DTH) in controlling the
the tubercle bacillus. Our goal would be to progress of tuberculosis in either rabbits or guinea
identify the antigenic composition that pro- pigs.
duces such an ideal ratio and then use this How does solid caseous tissue inhibit bacil-
knowledge to make a more effective vaccine. lary growth and even reduce the number of
Tissue necrosis in clinical tuberculosis could live tubercle bacilli? Do anoxia, toxic fatty acids,
be reduced by vaccines that cause little or no high osmolarity, and lower pH all play a role (see
sensitivity to tuberculin (see chapter 22). Such chapters 2 and 7 and reference 9)?
vaccines would expand the Th1 lymphocyte In brief, in the various laboratory animal
population that does not react to tuberculin-like species, the roles of apoptosis and necrosis need
products without expanding the population that to be investigated. Both processes kill macro-
does react to these products. In other words, this phages that contain too many tubercle bacilli for
vaccine would create a population of lympho- CMI to control. In each species, the roles of
cytes that could respond with a high CMI/ intracellular dormancy within activated macro-
DTH ratio. When such a vaccinated person phages (produced by CMI) and extracellular
inhales virulent tubercle bacilli, the expanded dormancy in solid caseous tissues (produced by
lymphocyte population that infiltrates the result- DTH) also need to be investigated.
ing pulmonary lesions would produce more
acquired cellular resistance (CMI, i.e., activated (iii) DTH and CMI and Turnover of
macrophages capable of inhibiting the growth Macrophages. The turnover of macrophages
of ingested tubercle bacilli) but show less tissue in tuberculous lesions provides a constant sup-
damage from DTH.After challenge with inhaled ply of nonactivated macrophages in which the
virulent tubercle bacilli, the recipient of this bacillus may multiply. Such turnover has been
low-DTH-producing vaccine should have documented in rabbit tuberculous lesions (see
reduced lung destruction and even reduced cav- chapter 10) but has not been studied in mice or
ity formation if the disease progresses. guinea pig lesions. Species variations probably
exist. In guinea pigs, many lesions show caseous
(ii) Species Variations in CMI/DTH necrosis, and they should have high macrophage
Ratios. In mice, exactly what stops the loga- turnover. In mice, few, if any, lesions have caseous
rithmic stage remains to be evaluated. DTH in necrosis, and they should have the least macro-
mice (although weak) probably causes apopto- phage turnover. Comparisons of macrophage
sis, which kills macrophages containing too many turnover in various laboratory species would
bacilli. Alternatively, macrophages overloaded provide insights into what might be occurring
with tubercle bacilli may simply burst. Then, in various types of human tuberculous lesions.
most of the released bacilli would be ingested by
ROLE OF VASCULAR THROMBOSIS
the highly activated macrophages that CMI in IN CAUSING CASEOUS NECROSIS
mice produces. Highly activated macrophages AND INHIBITION OF BACILLARY
must play a major role in mice, because in the sta- GROWTH
tionary stage the majority of bacilli are dormant In rabbits, guinea pigs, and humans, microvas-
for weeks within macrophages (7, 8). cular thrombosis causes much of the tissue dam-
In rabbits and guinea pigs, the CMI that causes age found in tuberculosis (see chapters 8 and 21).
bacillary dormancy within macrophages has never However, the amount of tissue necrosis pro-
been studied. In rabbit tuberculous lesions, tuber- duced by vascular thrombosis and the amount
376 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

produced by other DTH mechanisms still need FORMATION OF LIQUEFIED


to be compared. In rabbits, guinea pigs, and CASEUM AND CAVITIES AND
humans, thrombosis (with subsequent caseous THEIR PREVENTION
necrosis) seems to be a major factor in killing Liquefaction of solid caseum often provides an
macrophages that contain numerous bacilli, but ideal “culture medium” for the extracellular
killing such macrophages by other mechanisms growth of tubercle bacilli. (Before liquefaction
(including cytotoxic T cells) may be sufficient occurs, tubercle bacilli only grow intracellu-
without the additional local tissue damage pro- larly in nonactivated macrophages.) The large
duced by vascular thrombosis. numbers of actively growing bacilli in liquefied
What role does activation of vascular caseum release high levels of tuberculin-like
endothelial cells play in presenting mycobacte- products, which cause a tissue-damaging DTH
rial antigens and in the thrombosis present in reaction. If the walls of bronchi are destroyed by
caseous necrosis (described in chapter 21)? this reaction, a cavity is formed, and tubercle
Would decreasing the ability of the blood to clot bacilli are able to spread into the patient’s airways
be beneficial in tuberculous hosts, as it seems to and into the outside environment, where they
be in the prevention and treatment of myocar- may infect other people (see chapter 4).
dial infarctions and strokes? Studies on the formation of cavities and
their progression should be a major pri-
ority in tuberculosis research.Yet, at present,
ROLE OF GRANULOCYTES
IN TUBERCULOSIS few, if any, laboratory groups seem to be engaged
Polymorphonuclear leukocytes (PMN) (neu- in such work. Understanding the host factors
trophils in humans, mice, and guinea pigs, but involved would require:
eosinophilic heterophils in rabbits) are com-
mon near solid and liquefied caseum. Evidently, 1. Identifying the inhibitors that keep solid
they are attracted to all dying tissues (10). Are caseum from liquefying. Numerous
PMN able to destroy or inhibit the tubercle enzyme inhibitors should exist in solid
bacilli that they ingest? Do they carry the bacilli caseum.Yet, none has been identified and
to other sites and spread the disease? Do PMN characterized.These inhibitors may be of
produce factors (e.g., hydrolytic enzymes and host or bacillary origin.
cytokines) that aid the absorption of necrotic tis- 2. Determining the mechanism by which
sue and promote healing (see reference 11)? inhibitors are removed during the lique-
In dermal lesions produced in rabbits by faction of solid caseum.These inhibitors
the topical application of dilute sulfur mustard, may slowly diffuse from the caseum, or
PMN migrate right through the lesion and they may be hydrolyzed or otherwise
accumulate on the surface where the epithe- inactivated.
lium and previous PMN are dying (12).There, 3. Identifying and characterizing the spe-
they form a crust, or scab (12). In this sulfur cific enzymes that make solid caseum liq-
mustard model, the PMN attracted by the uefy. For the absorption of fluid into the
dying tissues seem to prevent bacterial invasion caseum, certain enzymes must be acti-
and aid the healing process. PMN may have vated, components of the caseum must
similar roles in caseous and liquefied tubercu- be hydrolyzed, and an osmotic gradient
lous lesions. must be produced. Proteases, nucleases,
The role of eosinophils, basophils, and mast lipases, and glycolytic enzymes seem to be
cells has yet to be evaluated in tuberculosis (see involved (see chapter 4). Some of these
chapter 6). Do they play any role in DTH reac- hydrolytic enzymes may exist as proen-
tions? Do any of their products activate zymes that require activation. After the
macrophages? Are they able to kill or inhibit enzymes causing liquefaction are identi-
tubercle bacilli? fied, specific pharmacological inhibitors
25. SUGGESTED FUTURE RESEARCH 䡵 377

could then be developed to stop this detri- 1. When many bacilli grow extracellularly in
mental process. liquefied caseum and disseminate in the
4. Determining why some types of liquefied lungs, the tuberculin skin test reaction may
caseum are more supportive of bacillary increase because of the boosting effect of
growth than other types. The liquefied the bacilli and their antigens. Tuberculin
caseum may require a maturation stage to sensitivity may also decrease, or even dis-
develop the right composition to support appear, when the host’s immune defenses
bacillary growth.Therefore, the composi- become exhausted by extensive disease (see
tion of caseum at various stages of the chapter 5). Changes in the tuberculin reac-
liquefying process should be analyzed. tion might be useful in monitoring the
Nutrients different from those present in course of the disease. However, careful clin-
laboratory culture medium may be found. ical correlations, including roentgeno-
In fact, liquefied caseum may contain spe- graphic studies, would have to be made
cific growth factors derived from host tis- before any prognostic meaning can be
sues or even from the bacilli themselves. attached to tuberculin skin reactivity.
5. Determining the role of DTH (tuber- 2. Similarly, analyses of peripheral blood lym-
culin sensitivity) in liquefaction and cav- phocyte functions (including cell types
ity formation (see chapter 4). DTH appar- and their cytokines) will have to be cor-
ently plays a role in cavity formation both related with clinical status to be meaning-
before and after the bacilli multiply extra- ful. Monitoring such peripheral changes to
cellularly. Before bacillary multiplication, reflect disease progression or regression is
DTH increases the peripheral accumula- further complicated by the fact that some
tion of macrophages and their hydrolytic tuberculous lesions progress while others
enzymes (and possibly alters the ratio of regress in the same patient, and that each
acid-acting to neutral-acting proteinases), lesion may have periods of exacerbation
so that solid caseum becomes liquid.After and regression.Therefore, cells assayed in
bacillary multiplication, enough antigen is the peripheral blood may not always reflect
produced that the tissue-damaging DTH the state of the entire disease.
reaction erodes a bronchial wall and forms 3. During liquefaction, the blood may show
the cavity. an increase in proteases or a decrease in
6. Developing a simple and convenient assay protease inhibitors. If so, assays for such
system to measure the efficacy of new inhibitors might have prognostic value.
methods that attempt to reduce liquefaction 4. Antigen-antibody complexes in the blood
and cavity formation. One such assay sys- (13, 13a) may increase as tubercle bacilli
tem is described at the end of chapter 4. and their antigens increase in the host.
That method uses the liquefaction and Assays of antigen-antibody complexes in
ulceration of dermal lesions produced by blood may be useful in determining the
BCG (and/or virulent tubercle bacilli) as extent of the disease and its prognosis, as
surrogates for liquefaction and cavity for- well as the effectiveness of antimicrobial
mation in the lungs. therapy (13, 13a). However, additional
studies of antigen-antibody complexes
PROGNOSTIC TESTS THAT must still be made before such assays are
REFLECT DISEASE PROGRESSION adopted for general clinical use.
OR REGRESSION 5. Chapter 5 discusses the progress made to
Tests for identifying increases and decreases in date in distinguishing tuberculin-positive
the number of tubercle bacilli in the host would persons with early progressive tubercu-
be quite useful in the care of tuberculosis lous lesions from persons with non-
patients. progressive lesions. Only those with
378 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

progressive lesions need to be treated with contains numerous bacilli? Answers to these
antimicrobial agents.The most promising questions would provide new insights into host-
of these studies (14) measured the amount parasite interaction.
of gamma interferon produced by periph-
eral blood mononuclear cells after 5 days NEW RESISTANT AND SUSCEPTIBLE
in culture and restimulation with the RABBIT STRAINS
Mycobacterium tuberculosis-specific antigen Since Lurie’s inbred rabbits are now extinct, it
ESAT-6. behooves us to develop replacements. Existing
6. Computed tomography scans might be possibilities are described in chapter 14, but
developed for rabbits so that the progress they are not commercially available.
of the disease could be followed without No one to date has evaluated the resistance
killing the animal. Perhaps even tubercle of any of the over 50 strains listed by the Amer-
counts could be made by this method, but ican Rabbit Breeders Association (16). Some of
our few attempts to do so have been these strains may have more uniform resistance
unsatisfactory (Y. C. Manabe, personal to tuberculosis than New Zealand White rab-
communication). bits have. Some may be more susceptible than
New Zealand White rabbits. A few of these
COMPARISONS OF TUBERCULOUS strains are smaller than New Zealand White
LESIONS PRODUCED BY LIVE AND rabbits, i.e., the size of large guinea pigs. The
DEAD TUBERCLE BACILLI smaller size would reduce the cost of caging,
Dead tubercle bacilli can produce lesions quite food, and care.
similar to those produced by live bacilli (5). We recommend that Lurie’s tubercle-count
Therefore, the number of dead bacilli, as well as method (described in chapter 11) be used to
the number of live bacilli, determines the course evaluate the resistance of many of these rabbit
of the disease. Dead tubercle bacilli contain strains. If any were to prove more valuable than
some of the same antigens as live bacilli. How- New Zealand White rabbits, they could be
ever, secreted antigens from live tubercle bacilli developed commercially.
play a definite role in the development of DTH
and CMI (see chapter 22). More insight into the PART II. SUGGESTED RESEARCH
immunology and pathology of the host response ON THE BACILLUS
to dead and live tubercle bacilli should be a BACILLARY SURVIVAL
fertile field of investigation (see reference 15). AND VIRULENCE
The dormancy of tubercle bacilli in the host is Virulent mycobacteria can survive (i) drying
discussed in chapters 6 and 10. in the outside environment, (ii) the effects of
ROIs (reactive oxygen intermediates), RNIs
ROLE OF ANTIBODIES IN HOST (reactive nitrogen intermediates), and hydrolytic
RESISTANCE TO REINFECTION enzymes within activated macrophages, and (iii)
As discussed in chapter 5, an antigen-antibody the toxic fatty acids, low oxygen tension, and
reaction can produce the chemotactic C5a reduced pH within solid caseum (see chapters
component of complement (and probably other 2 and 7) (9). They can multiply in liquefied
chemotactic factors) that hastens the accumu- caseum.What genes are expressed by the tuber-
lation of an expanded antigen-specific Th1 cle bacillus to enable it to survive (or multiply)
lymphocyte population at the site of bacillary in each of these different local environments?
lodgement. In other words, antibodies enhance
the local CMI response. Cytophilic antibodies (i) Bacillary Survival in Air. A thick wax-
may attach to macrophages. If so, do they like glycolipid coat enables the bacilli to resist
enhance macrophage activation whenever anti- desiccation in dry air. Is this coat altered when
gen is present? Do cytophilic antibodies cause the bacilli grow in macrophages? Are the thick-
macrophage apoptosis when the macrophage ness and composition of this wax coat changed
25. SUGGESTED FUTURE RESEARCH 䡵 379

when the bacilli grow extracellularly in the liq- laboratory cultures? See chapters 1, 6, and 10 for
uefied caseum within cavities? Do bacilli grow- additional information on bacillary persistence
ing in liquefied caseum have properties different and dormancy.
from bacilli growing in laboratory culture media? A recent experiment identified 10 such genes
Sigma factors (17) aid bacillary survival (see associated with mycobacterial persistence in rab-
chapter 1). Inhaled bacilli also resist the detergent bit tuberculous lesions (Y. C. Manabe et al.,
effects of surfactants in the airways of the lungs. unpublished data) (see chapter 6), but more work
needs to be done to identify the part of the
(ii) Bacillary Survival within Macro- lesion in which such bacilli reside.The rabbit is
phages. Tubercle bacilli produce virulence the only common laboratory animal in which
factors that interfere with the microbicidal func- pulmonary lesions produced by M. tuberculosis
tions of macrophages (reviewed in chapter 6). actually heal (similar to most early lesions in
They impair the maturation of the macrophage humans).This latency (dormancy) model should
phagosome into its normal acidic, microbicidal, be further investigated in order to develop drugs
and hydrolytic compartments, but allow nutri- to prevent and/or treat the reactivation of tuber-
ents to enter it (15). The bacilli also slow the culous lesions.
development of the Th1 lymphocyte acquired
immune response. (iv) Bacillary Growth in Liquefied
What are the various microbial factors enabling Caseum. Some of the mycobacterial genes
the bacillus to act in this way? Do tubercle bacilli enabling bacillary growth in liquefied caseum are
growing within macrophages produce antioxi- probably different from those enabling growth
dants that reduce the microbicidal effects of the within macrophages.Are any of these genes dif-
host’s ROIs and RNIs, or do such bacilli merely ferent from those enabling bacillary growth in
inhibit the macrophage production of ROIs and laboratory culture medium? Are the genes that
RNIs, or both? How do they resist and/or pre- are active in liquefied caseum before a cavity
vent the lower pH normally found in lysosomes? forms different from the genes that are active
Are some of the genes activated for bacillary after a cavity forms when the oxygen tension is
growth within macrophages not active during higher and stimulates bacillary growth? (See
bacillary growth in laboratory culture media? Fig. 20 in chapter 4.)
How are these various virulence mechanisms
integrated? (v) Other Virulence Considerations.
The multiplication of virulent tubercle bacilli How do the genes (and their products) of highly
within macrophages is currently an active field virulent tubercle bacilli differ from those of
of investigation (see chapter 6). However, much bacilli of reduced virulence (see references 15,
more insight is needed in order to develop new 20, and 20a)? How do such differences enable
drugs to inhibit such multiplication. the bacillus to live in each of the environments
found in the host and in the outside environ-
(iii) Bacillary Survival in Solid Caseum. ment? Fully virulent and semivirulent tubercle
Dormancy genes and their products, e.g., sigma bacilli may differ in cell wall composition (see
factors (17–19), may be required for the bacil- reference 21) and/or in the type and amount of
lus to survive for many years in solid caseum. tuberculin-like substances that they produce.
What activates these genes to enable the bacil- Many virulence factors exist (discussed in
lus to do so, and what turns dormancy genes off chapters 1 and 6). If a single gene is lacking, oth-
when liquefaction occurs? Are dormancy genes ers may compensate for it (21, 22). Also, the
enabling survival in solid caseum different from virulence of the various mycobacterial strains is
those enabling survival within macrophages not always the same in different animal species
and survival in the outside environment? Are (23, 24); e.g., rabbits are much more susceptible
dormancy genes that are active in solid caseous to Mycobacterium bovis than to M. tuberculosis (5,
tissue the same as those that are active in old 25) (see chapter 15). Despite the great progress
380 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

that has been made in explaining the virulence ratios. Are such ratios a major factor in vaccine
of pathogenic tubercle bacilli, many additional efficacy? If so, should Th1/Th2 ratios be used in
attributes remain to be discovered because of the selecting vaccines for clinical trials?
complexity of host-parasite interactions.
IMMUNOTHERAPY TO SUPPLEMENT
PART III. SUGGESTED RESEARCH ON ANTIMICROBIAL THERAPY AND TO
PROPHYLACTIC VACCINES AND TREAT MULTIDRUG-RESISTANT
IMMUNOTHERAPY TUBERCULOSIS
Immunotherapy should be pursued more exten-
MORE EFFECTIVE VACCINES sively, because it would be of great benefit in
The prevention of clinical tuberculosis by a treating multidrug-resistant tuberculosis, and
more effective vaccine than currently available because it could shorten the treatment of drug-
BCG vaccines would be the most efficient way susceptible tuberculosis.
to reduce the incidence of this disease in the The main recent immunotherapy trials were
world. The protection by improved vaccines carried out with Mycobacterium vaccae (reviewed
may never be 100%. In fact, even the 80% effi- in reference 27). These trials showed much
cacy of BCG and Mycobacterium microti vaccines, promise.Very few laboratory studies have been
used in the 1950s, does not seem to be repro- made on the immunotherapy of cavitary tuber-
duced by the vaccines available today (26).We culosis with M. vaccae or any other immunizing
should try to regain this efficacy and attempt to agent (see chapters 4 and 22)—probably because
increase it to 90 or 95%. the rabbit is the only common laboratory ani-
Perhaps adding genes for certain antigens to mal that readily produces cavities, and very few
BCG by molecular biology techniques would investigators work with rabbits.
increase its effectiveness. Such antigens might be
best given as a booster immunization, because OTHER VACCINE POSSIBILITIES
if they are incorporated into BCG, they might Other vaccines, including DNA vaccines, are
substantially increase its virulence. (See chapter discussed in chapter 22.
22 for other possibilities.)
Fortunately, once an effective vaccine has PART IV. DEVELOPMENT OF NEW
caused a major decline in the number of new DRUGS FOR THE TREATMENT AND
cases of active tuberculosis per year, the decline PREVENTION OF TUBERCULOSIS
would continue (because of decreased sources of DRUGS ACTING ON THE HOST
contagion) until the disease is no longer a major Drugs to inhibit the hydrolytic enzymes that
concern. cause liquefaction and cavity formation should be
To produce the most effective vaccine, certain evaluated in the rabbit model of tuberculosis.
questions remain to be answered.Are live vaccines Liquefied caseum and cavities are not readily
required because they multiply, secrete, and release produced in other common laboratory animals.
antigens and persist longer in the host? Does Anticoagulants might also be a useful addition
protection disappear if the vaccinating bacilli and to antimicrobial therapy. As discussed above,
their antigens are eliminated from the host? Is they might decrease the amount of caseous
revaccination with the same or a different vaccine necrosis by reducing the thrombosis in the sur-
required to boost immunity? Do effective vac- rounding vasculature.
cines need to produce a positive tuberculin skin
test reaction? What role does the adjuvanticity of DRUGS ACTING ON THE BACILLUS
BCG play in its effectiveness (see chapter 19)? Are New antimicrobial drugs are needed (i) to
recombinant BCG vaccines that produce shorten the present 6-month course of therapy
cytokines or costimulatory molecules more effec- for drug-susceptible tuberculosis and (ii) to treat
tive than the parent BCG? How can the number multidrug-resistant disease. Chemotherapeutic
of memory cells be increased? Different vaccines strategies that reduce the time needed for the
may produce different Th1/Th2 lymphocyte treatment of drug-susceptible tuberculosis would
25. SUGGESTED FUTURE RESEARCH 䡵 381

have important effects on cost, compliance, drug (iii) extracellular growth in liquefied caseum
toxicity, and emergence of drug-resistant strains (often within cavities), and (iv) survival in the
of tubercle bacilli (18, 19, 28, 29). Most antimi- outside environment, where the composition of
crobial agents for tuberculosis are effective the protecting coat plays an important role.The
against actively multiplying bacilli (18, 19, 30). contagiousness of multidrug-resistant patients
Tubercle bacilli in a dormant state are often might be reduced if a drug is developed that
not killed by such antimicrobials, especially reduces the thickness of the lipid coat of the
bacilli in solid caseous tissues, which is the rea- bacillus, even though the drug may have little
son why a long period of administration is effect on intracellular bacillary multiplication
required. Such persistence is reviewed in refer- in vivo.
ences 18, 19, and 31.Table 1 relates the effec- Drugs that target dormancy are especially
tiveness of antimicrobials to the in vivo meta- needed, e.g., drugs that inhibit DosR, RelA,
bolic states of the tubercle bacillus. sigma factors, triacylglycerol synthase (33), and
Pyrazinamide (PZA) is a prodrug that is con- isocitrate lyase (8) (reviewed in references 18, 19,
verted by the tubercle bacillus into pyrazinoic and 31; also see chapter 1). New drugs that
acid, which is microbicidal (18). It is most active affect the viability and virulence of dormant
at acid pH, at low oxygen concentrations, and tubercle bacilli seem particularly pertinent. To
on nonreplicating (rather than actively growing) identify new drugs that are active on nonmul-
tubercle bacilli (18). Therefore, unlike other tiplying, persistent tubercle bacilli, drug screens
antimycobacterial drugs, it can kill a population need to be developed that mimic dormancy in
of persistent tubercle bacilli residing in solid vivo, including low oxygen, reduced pH, and
caseous tissues, thereby shortening the duration toxic fatty acids (18).
of antimicrobial therapy. Since such caseum is Three new drugs that have apparently differ-
rich in lipid components, alteration of the PZA ent targets from previous antimycobacterial drugs
structure (to make it more lipid soluble) might are now in clinical trials.The first is a diarylquino-
increase its effectiveness against such dormant line (R207910, developed by Janssen Pharma-
bacilli. If so, one of the major difficulties in ceutica in Beerse, Belgium, a subsidiary of John-
controlling tuberculosis would be surmounted. son and Johnson). It inhibits the ATP synthase in
Perhaps incorporation into liposomes (see ref- the mycobacterial membrane (34–36).The sec-
erence 32) would increase the penetration of the ond is a nitroimidazopyran (PA-824, developed
drug into caseous areas. by PathoGenesis, Inc., a division of Chiron, Inc.,
New types of drugs are needed to prevent the Emeryville, Calif.) (36–38).The third is a pyrrole
ability of the bacillus to enter each of the four derivative (LL-3858, developed by Lupin Labo-
stages of its life cycle: (i) intracellular growth in ratories, Ltd., Pune, India) (S.Arora, Int. J.Tuberc.
macrophages, (ii) dormancy in solid caseum, Lung Dis. 8[Suppl. 1]:S29 [abstract], 2004). All

TABLE 1 In vivo effectiveness of principal antituberculous agents on tubercle bacilli in various metabolic
statesa
Bacilli are actively Bacilli are slowly Bacilli are slowly
Drug metabolizing metabolizing at acid pH metabolizing at neutral pH
Streptomycin ⫹⫹⫹ 0 0
Isoniazid ⫹⫹⫹ ⫹ 0
Rifampin ⫹⫹ ⫹ ⫹
Ethambutol ⫾ ⫾ 0
Pyrazinamide 0 ⫹⫹ 0
a
⫹, ⫹⫹, and ⫹⫹⫹ represent increasing degrees of bactericidal activity; ⫾ represents bacteriostatic activity; 0 represents no
activity.Adapted from reference 30.
382 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

three have mycobacterial sterilizing activity in paraffin-embedded tissue sections. Stain Technol.
mice. 54:5–12.
3. Namba, M., A. M. Dannenberg, Jr., and
However, it remains to be determined F.Tanaka. 1983. Improvement of the histochem-
whether (i) any of these new drugs will replace ical demonstration of acid phosphatase, -galac-
one or more existing drugs in the standard treat- tosidase and nonspecific esterase in glycol methacry-
ment of tuberculosis, (ii) the new drugs will late tissue sections by cold temperature embedding.
shorten the duration of therapy, e.g., from 6 to Stain Technol. 58:207–213.
4. Burstone, M. S. 1962. Enzyme Histochemistry and
3 months, or (iii) the new drugs will prevent Its Application to the Study of Neoplasms, p. 8–63.
relapses caused by formerly dormant tubercle Academic Press, Inc., New York, N.Y.
bacilli located in solid caseous tissues. 5. Lurie, M. B. 1964. Resistance to Tuberculosis: Exper-
In mice, when moxifloxacin (8-methoxy- imental Studies in Native and Acquired Defensive
fluoroquinolone) (MXF) replaced isoniazid Mechanisms. Harvard University Press, Cambridge,
Mass.
(INH) in combinations with PZA and rifampin 6. Ando, M. 1973. Macrophage activation in tuber-
(RIF), a greater number of tubercle bacilli were culin reactions of rabbits with primary BCG infec-
killed than when all four drugs were present tion and reinfection. J. Reticuloendothel. Soc. 14:132–
(39). This suggests that INH had inhibitory 145.
effects on one or more of the other drugs. In fact, 7. Rees, R. J.W., and P. D. Hart. 1961.Analysis of
the host-parasite equilibrium in chronic murine
in 4 months, a regimen of PZA, RIF, and MXF tuberculosis by total and viable bacillary counts. Br.
produced a stable cure of mice that were infected J. Exp. Pathol. 42:83–88.
by aerosol with virulent human-type tubercle 8. McKinney, J. D., K. Höner zu Bentrup, E. J.
bacilli (H37Rv) (40). In contrast, the standard Muñoz-Elias,A. Miczak, B. Chen,W.T. Chan,
regimen of PZA, RIF, and INH required 6 D. Svenson, J. C. Sacchettini,W. R. Jacobs, Jr.,
and D. G. Russell. 2000. Persistence of Mycobac-
months of therapy. MXF is in clinical use in the terium tuberculosis in macrophages and mice requires
treatment of multidrug-resistant tuberculosis and the glyoxylate shunt enzyme isocitrate lyase. Nature
seems to be well tolerated (41).These and other 406:735–738.
new drug candidates for the treatment of tuber- 9. Long, E. R. 1958. The Chemistry and Chemother-
culosis are reviewed in reference 36. apy of Tuberculosis.The Williams & Wilkins Co., Bal-
timore, Md.
10. Majno, G., and I. Joris. 1996. Cells,Tissues, and
PART V. SOURCES OF ADDITIONAL
Disease. Blackwell Science, Cambridge, Mass.
INFORMATION
11. Kajiki, A., K. Higuchi, M. Nakamura, L. H.
Structure of mycobacterial cell walls (42). Liu, P. J. Pula, and A. M. Dannenberg, Jr.
1988. Sources of extracellular lysosomal enzymes
Genomics of the M. tuberculosis complex (42 released in organ culture by developing and heal-
and 43). ing inflammatory lesions. J. Leukoc. Biol. 43:104–116.
Details on various antimicrobial agents and 12. Dannenberg, A. M., Jr., P. J. Pula, L. Liu,
S. Harada, F.Tanaka, R. F.Vogt, Jr., A. Kajiki,
strategies for developing new ones (28).
and K. Higuchi. 1985. Inflammatory mediators
Mechanisms of drug resistance in M. tubercu- and modulators released in organ culture from rab-
losis (44). bit skin lesions produced in vivo by sulfur mustard.
I. Quantitative histopathology; PMN, basophil and
Currently approved regimen for treating
mononuclear cell survival; and unbound (serum)
tuberculosis (45). protein content. Am. J. Pathol. 121:15–27.
Preventing the transmission of tuberculosis 13. Brostoff, J., L. Lenzini, P. Rottoli, and L. Rot-
(46) (see Appendix E). toli. 1981. Immune complexes in the spectrum of
tuberculosis. Tubercle 62:169–173.
13a.Daniel, T. M. 1986. Circulating immune com-
REFERENCES plexes in tuberculosis. Am. Rev. Respir. Dis. 134:199–
1. Nathan, C. 2002. Points of control in inflamma- 200. (Editorial.)
tion. Nature 420:846–852. 14. Doherty,T. M.,A. Demissie, J. Olobo, D.Wol-
2. Higuchi, S., M. Suga, A. M. Dannenberg, Jr., day, S. Britton, T. Eguale, P. Ravn, and
and B. H. Schofield. 1979. Histochemical P. Andersen. 2002. Immune responses to the
demonstration of enzyme activities in plastic- and Mycobacterium tuberculosis-specific antigen ESAT-6
25. SUGGESTED FUTURE RESEARCH 䡵 383

signal subclinical infection among contacts of tuber- 27. Stanford, J., C. Stanford, and J. C. Grange.
culosis patients. J. Clin. Microbiol. 40:704–706. 2004. Immunotherapy with Mycobacterium vaccae in
15. Russell, D. G., G. E. Purdy, R. M. Owens, the treatment of tuberculosis. Front. Biosci. 9:1701–
K. H. Rohde, and R. M.Yates. 2005. Mycobac- 1719.
terium tuberculosis and the four-minute phagosome. 28. Walsh, C. 2003. Antibiotics: Actions, Origins, Resis-
ASM News 71:459–463. tance.ASM Press,Washington, D.C.
16. American Rabbit Breeders Association, Inc. 29. O’Brien, R. J., and P. P. Nunn. 2001.The need
1996. Standard Bred Rabbits and Cavies, 1996 through for new drugs against tuberculosis. Obstacles, oppor-
2000.American Rabbit Breeders Association, Inc., tunities, and next steps. Am. J. Respir. Crit. Care
P.O. Box 426, Bloomington, IL 61702. Med. 162:1055–1058.
17. Smith, I. 2003. Mycobacterium tuberculosis patho- 30. Grosset, J. 2003. Mycobacterium tuberculosis in the
genesis and molecular determinants of virulence. extracellular compartment: an underestimated
Annu. Rev. Microbiol. 16:463–496. adversary. Antimicrob.Agents Chemother. 47:833–836.
18. Zhang,Y. 2005.The magic bullets and tuberculo- 31. Gomez, J. E., and J. D. McKinney. 2004.
sis drug targets. Annu. Rev. Pharmacol.Toxicol. 45:529– M. tuberculosis persistence, latency, and drug toler-
564. ance. Tuberculosis 84:29–44.
19. Zhang,Y. 2005. Persistent and dormant tubercle 32. Adams, L. B., I. Sinha, S. G. Franzblau, and
bacilli and latent tuberculosis. Front. Biosci. 9:1136– J. L. Krahenbuhl. 1999. Effective treatment of
1156. acute and chronic murine tuberculosis with lipo-
20. Russell, D. G. 2001. Mycobacterium tuberculosis: some-encapsulated clofazimine. Antimicrob. Agents
here today, and here tomorrow. Nat. Rev. Mol. Cell Chemother. 43:1638–1643.
Biol. 2:569–577. 33. Daniel, J., C. Deb,V. S. Dubey,T. D. Sirakova,
20a.Manabe, Y. C., A. M. Dannenberg, Jr., S. K. B. Abomoelak, H. R. Morbidoni, and P. E.
Tyagi, C. L. Hatem, M.Yoder, S. C.Woolwine, Kolattukudy. 2004. Induction of a novel class of
B. C. Zook, M. L. M. Pitt, and W. R. Bishai. diacylglycerol acyltransferase and triglycerol accu-
2003. Different strains of Mycobacterium tuberculosis mulation in Mycobacterium tuberculosis as it goes into
cause various spectrums of disease in the rabbit a dormancy-like state in culture. J. Bacteriol.
model of tuberculosis. Infect. Immun. 71:6004– 186:5017–5030.
6011. 34. Cole, S.T., and P. M. Alzari. 2005. Perspectives:
21. Constant, P., E. Perez, W. Malaga, M. A. TB—a new target drug. Science 307:214–215.
Lanéelle, O. Saurel, M. Daffé, and C. Guilhot. 35. Andries, K., P.Verhasselt, J. Guillemont, H.W.
2002. Role of the pks15/1 gene in the biosynthe- H. Göhlmann, J.-M. Neefs, H.Winkler, J. van
sis of phenolglycolipids in the Mycobacterium tuber- Gestel, P. Timmerman, M. Zhu, E. Lee,
culosis complex. Evidence that all strains synthesize P. Williams, D. de Chaffoy, E. Huitric,
glycosylated p-hydroxybenzoic methyl esters and S. Hoffner, E. Cambau, C. Truffot-Pernot,
that strains devoid of phenolglycolipids harbor a N. Lounis, and V. Jarlier. 2005.A diarylquinoline
frameshift mutation in the pks15/1 gene. J. Biol. drug active on ATP synthase of Mycobacterium tuber-
Chem. 277:38148–38158. culosis. Science 307:223–227.
22. Steyn,A. J. C., D. M. Collins, M. K. Hondalus, 36. Zhang, Y., K. Post-Martens, and S. Denkin.
W. R. Jacobs, Jr., R. P. Kawakami, and B. R. 2006. New drug candidates and therapeutic targets
Bloom. 2002. Mycobacterium tuberculosis WhiB3 for tuberculosis therapy. Drug Discov.Today 11:21–
interacts with RpoV (which is Sig A) to affect host 27.
survival but is dispensable for in vivo growth. Proc. 37. Stover, C. K., P.Warrener, D. R.VanDevanter,
Natl.Acad. Sci. USA 99:3147–3152. D. R. Sherman, T. M. Arain, M. H. Lang-
23. Francis, J. 1958. Tuberculosis in Animals and Man. horne, S. W. Anderson, J. A. Towell, Y. Yuan,
A Study in Comparative Pathology. Cassell and Co., D. N. McMurray, B. N. Kreiswirth, C. E. Barry,
London, United Kingdom. and W. R. Baker. 2000.A small-molecule nitroim-
24. Dannenberg, A. M., Jr. 1984. Pathogenesis of idazopyran drug candidate for the treatment of
tuberculosis: native and acquired resistance in ani- tuberculosis. Nature 405:962–966.
mals and humans, p. 344–354. In D. Schlessinger 38. Tyagi, S., E. Nuermberger, T. Yoshimatsu,
(ed.), Microbiology—1984. American Society for K.Williams, I. Rosenthal, N. Lounis,W. Bishai,
Microbiology,Washington, D.C. and J. Grosset. 2005. Bactericidal activity of the
25. Lurie, M. B., and A. M. Dannenberg, Jr. 1965. nitroimidazopyran, PA-824, in the murine model of
Macrophage function in infectious disease with tuberculosis. Antimicrob.Agents Chemother. 49:2289–
inbred rabbits. Bacteriol. Rev. 29:466–476. 2293.
26. Comstock, G.W. 1994. Field trials of tuberculo- 39. Nuermberger, E. L., T.Yoshimatsu, S. Tyagi,
sis vaccines: how could we have done them better? R. J. O’Brien, A. N.Vernon, R. E. Chaisson,
Control. Clin.Trials 15:247–276. W. R. Bishai, and J. H. Grosset. 2004.
384 䡵 PATHOGENESIS OF HUMAN PULMONARY TUBERCULOSIS

Moxifloxacin-containing regimen greatly reduces 43. Cole, S. T. 2002. Comparative and functional
time to culture conversion in murine tuberculosis. genomics of the Mycobacterium tuberculosis complex.
Am. J. Respir. Crit. Care Med. 169:421–426. Microbiology 148:2919-2928.
40. Nuermberger, E. L., T.Yoshimatsu, S. Tyagi, 44. Wade, M. M., and Y. Zhang. 2004. Mechanisms
K.Williams, I. Rosenthal, R. J. O’Brien, A. A.
of drug resistance in Mycobacterium tuberculosis. Front.
Vernon, R. E. Chaisson,W. R. Bishai, and J. H.
Grosset. 2004. Moxifloxacin-containing regimens Biosci. 9:975–994.
of reduced duration produce a stable cure in murine 45. American Thoracic Society, Centers for Dis-
tuberculosis. Am. J. Respir. Crit. Care Med. 170:1131– ease Control and Prevention, and Infectious
1134. Disease Society of America. 2003. Official joint
41. Valerio, G., P. Bracciale, V. Manisco, statement for the treatment of tuberculosis. Am. J.
M. Quitadamo, G. Legari, and S. Bellanova. Respir. Crit. Care Med. 167:603–662.
2003. Long-term tolerance and effectiveness of 46. Centers for Disease Control and Prevention.
moxifloxacin therapy for tuberculosis: preliminary
2005. Guidelines for preventing the transmission of
results. J. Chemother. 15:66–70.
42. Cole, S. T., K. D. Eisenach, D. N. McMurray, Mycobacterium tuberculosis in health-care settings,
and W. R. Jacobs, Jr. (ed.). 2005. Tuberculosis and 2005. Morb. Mortal. Wkly. Rep. 54(RR-17):1–
the Tubercle Bacillus.ASM Press,Washington, D.C. 141.
APPENDIX A
AWARD OF THE TRUDEAU MEDAL FOR 1955†

The Trudeau Medal for 1955 was awarded to Dr. Max B. ambition was to study philosophy, but he soon real-
Lurie at the annual meeting of the National Tuberculosis ized that this highly respected discipline offered lit-
Association, held in NewYork, N.Y., May 21–24, 1956.The tle that would provide a means of livelihood in the
presentation was made by Dr.William H. Feldman.
immediate or near future.“In despair [and I use his
words] I determined to study medicine.” This
DR. FELDMAN decision was influenced by “the idea that the most
In 1925 the Board of Directors of the National one can do is to alleviate the pain of human life;
Tuberculosis Association approved a suggestion of medicine offers that opportunity.”
the Committee on Medical Research that there be He entered Cornell University Medical College
awarded not oftener than once yearly a medal to and served in the Student Army Training Corps.
one who had made meritorious contributions to During his medical training, he became distressed
the knowledge of the cause, prevention, or treat- at the thought of practicing medicine because of
ment of tuberculosis.This symbol of recognition what he considered would be his incompetence.
was to be known as the Trudeau Medal.A year later Always conscientious to a fault, he feared that his
this suggestion was accepted by resolution and lack of knowledge would endanger the lives of
adopted by the Executive Committee. those whom he was expected to treat. Finally, his
At the 23rd annual meeting of the National dilemma was resolved. Fate intervened and a highly
Tuberculosis Association held in Washington, sensitive, reluctant young physician was spared the
October, 1926, the award was first presented to Dr. agony and vicissitudes of practicing medicine,
Theobald Smith. Dr. Smith had been president of eventually to become a highly productive and
the National Tuberculosis Association during the brilliant investigator. In the words of our medal-
preceding year and was one of the true giants of ist,“It was my good fortune to diagnose my own
American science.Thus was initiated a yearly cus- tuberculosis in the fourth year of medical school.”
tom, which has become an important tradition. (He probably would have made a good physi-
Today, we pause in our preoccupation with sci- cian.) He attempted to “take the cure” and at the
entific sessions, committee meetings, and the many same time carry on an internship, with the pre-
items that make a visit to New York exciting, to dictable result, i.e., he finally had to spend full
honor one of our peers by making him the 30th time as a patient in a tuberculosis sanatorium. It
recipient of this enviable symbol of our respect and was there that he became interested in what was
esteem. The one who has been chosen unani- to be his future career: research in tuberculosis. So
mously to receive the Trudeau Award this year, and much for the “clinical” aspects of this gifted man.
whose name I shall disclose in a few moments, is What are some of the attributes that have char-
known to all of you, and his contributions to the acterized him as a person and as a scientist? Above
knowledge of tuberculosis are highly regarded all, he has a great compassion, a real concern for the
throughout the world. unfortunate, and an intense aversion to all that
Before proceeding further with the scientific denies freedom and justice to man.While he can be
accomplishments that merited this award, I wish militantly indignant concerning social evils, he
to digress briefly and give a short “clinical history” believes in and practices good will. He is a man of
of our recipient. He was born in Lithuania, much humility and utterly without pretense. He
endowed with little else than a priceless scholarly places a high value on integrity, believing as he has
heritage. He came to America at the age of fifteen. expressed it:“Of what avail is the acclaim of others
After high school at Townsend Harris Hall, he if you cannot countenance yourself.” He has a sense
entered the College of the City of New York. His of humor, enjoys an argument and, parenthetically
I would say, is a skillful debater. He considers dif-

Reprinted from Am. Rev.Tuberc. Pulm. Dis. 74:647–649, ferences of opinion healthy and desirable. His extra-
1956, with slight modifications. scientific interests are social justice, philosophy and

385
386 䡵 APPENDIX A

music. The motivation of his labors has been to over the years would be an understatement. Indeed,
seek a meaning and a justification for life. In his I see in it a vindication of my hope that my life has
quest he has succeeded admirably—to the envy of not been altogether in vain. Even more, I take it as
us all. evidence of the great generosity of spirit that char-
He brought to his professional tasks a scholarly acterizes our democracy. For a poor immigrant
insight, an ingenious talent for experimentation, a boy to have received the necessary training, to be
masterful technique, and a fiery enthusiasm. Impor- given the opportunity to pursue his craving to try
tantly, he has a fine concept of scientific evidence. to understand even an iota of the mystery of life,
He has stated the underlying philosophy of his is a triumph of the human spirit.
research in the following words: “I have always Most of all, for the Trudeau Society to award this
thought that it is better to attempt the solution of distinguished honor to one who has achieved no
a fundamental problem and fail than to undertake immediately practical solution to the problems on
the elaboration of minor details with great success.” hand—but to him who has endeavoured by tenac-
These words are worth remembering. ity and devotion to penetrate ever so little into the
The scientific contributions of the recipient of intricacies of the host-parasite relationships which
the Trudeau Medal for 1955 are sufficiently well constitute native and acquired resistance to tuber-
known to require no enumeration. Briefly, he has culosis—is both a humbling thought and a warm-
been the sole or senior author of nearly 70 pub- ing of the heart. It is humbling because we all
lications, including several of monographic dimen- know the ephemeral nature of even the loftiest and
sion; and his name appears in joint authorship of most penetrating of scientific ideas.The centuries-
many more. His scientific writing, especially his old sway of Newton’s laws over the universe has
reviews, reveal a capacity to establish continuity of yielded to the relativistic concepts of Einstein. It is
new facts with those previously reported. This a sacrilege to mention these giants of the human
bringing together of the new with the old enables mind in the same breath with our feeble efforts.
the reader to obtain a composite understanding of But, the condition of human existence is the same
the interrelationships of the various facets of the in the great and in the small; absolute knowledge
subject discussed. is beyond our capacity. It is in increasing under-
Although the main theme of his many investi- standing that the salvation of man lies. Not only in
gations has been resistance to tuberculosis, of out- the mastery of his environment and in the subju-
standing importance is his brilliant conception gation of the elements to his will (as we are wit-
and successful demonstration of genetic factors nessing daily in ever increasing crescendo), but
in such resistance. One who knows him well has even more in the control over himself and the
said, “His early genetic work opened up a new moral growth of mankind.The more man under-
world.” He has continued to explore this world stands himself, the more truly human will he
successfully and with results that have brought become. It is heartening that the Trudeau Society
him international renown. deems it worthy to encourage these labors, assailed
The recipient of the Trudeau Medal for 1955 as they often are by failure and doubt.
has, in a word, accomplished work that has been I wish to take this opportunity to express my
magnificent in its purpose.This work has, I am cer- gratitude to the Henry Phipps Institute of the
tain, been deeply satisfying and has brought this University of Pennsylvania, to my teachers and
devoted and dedicated servant of science a touch inspirers within and outside its walls, to my wife
of immortality. who has stood unflinchingly by me in times of
On behalf of the Trudeau Award Medal Com- trial, to the organizations that have supported
mittee, I consider it an honor and a happy privi- these endeavours over the years, especially to the
lege to present the Trudeau Medal Award for 1955 Commonwealth Fund, the Trudeau Society and
to Max B. Lurie. the Public Health Service, and above all to my col-
laborators and assistants, without whose devotion
DR. LURIE even the little we did could not have been accom-
Mr. President, the Committee on the Trudeau plished. On behalf of all those whose greatest joy
Award, Dr. Feldman, ladies and gentlemen. in life is the glimmer of understanding that comes
To merely say that I am profoundly moved by after searching and critical analysis, allow me to
the recognition that you have given to our labors thank you for your generosity.
APPENDIX B
OBITUARY OF MAX B. LURIE, M.D. (1893–1966)†
Esmond R. Long, M.D., Ph.D., Sc.D.
Director of the Henry Phipps Institute for the Cure and Prevention
of Tuberculosis, University of Pennsy.lvania, Philadelphia

Max Bernard Lurie, experimental pathologist become a Fellow in Pathology under Eugene L.
and cultured scholar, well known for his investiga- Opie at the Henry Phipps Institute of the Uni-
tions on heredity and constitutional factors in versity of Pennsylvania in Philadelphia. Here, he
tuberculosis, died in Philadelphia September 23, stayed for forty years attaining the rank of full
1966. He had been in failing health for many professor in 1955, maintaining an unbroken line of
months, with increasing disability of the heart, but research from early studies on the fate of tubercle
maintained his studies and productivity to the end. bacilli in different organs of the animal body, to a
Lurie was born on September 12, 1893, in masterly comprehension of tuberculosis as a whole
Telshee, Lithuania, during the period of Russian and a position of eminence among investigators of
domination of the country. He emigrated with his that disease.
family to the United States in 1907. His parents The late 1920’s represented a period of general
were of distinguished Jewish ancestry, with origins and deep interest in pathologic and immunologic
traceable to Talmudic scholars in the eleventh cen- differences between first infection and reinfec-
tury. His early years in Lowell, Massachusetts, and tion tuberculosis. Lurie’s chief, Dr. Opie, who
New York City were steeped in Jewish culture. In inspired Lurie in many ways, was much concerned
spite of family tradition, however, he was not with the mechanism of transmission of the infec-
inclined to enter his father’s profession, the rab- tion in man. Lurie became a pioneer in the devel-
binate. He was deeply interested in philosophy, opment of laboratory methods for study of the
and would have liked to specialize in it, but for problem. Beginning shortly before 1930, he set up
practical reasons undertook the study of medi- experimental models for air-borne infection in
cine, which promised a more certain livelihood and, guinea pigs, with ingenious arrangements of cages,
more importantly, an opportunity for effective ser- which not only demonstrated the mechanisms
vice to mankind. He entered the medical school and frequency of air-borne contagion, but even led
of Cornell University, worked his way through, to determination of the particulate size of infect-
often at poorly paying tasks, and graduated in ing units.
1921. In 1932, Lurie acquired a colony of highly
In his medical school days, it was recognized inbred rabbits from a stock raised for experiments
that he had contracted tuberculosis, the disease at the University of Chicago, from which he soon
from which his mother died. Soon after graduation, separated out, by litter matings, two strains of ani-
he entered the National Jewish Hospital for Con- mals with sharply differing degrees of resistance to
sumptives in Denver, Colorado, for a residency and tuberculosis. These, and subsequent families of
what was intended to be a period of rest, but soon intermediate character, proved invaluable for later
proved to be one of work and preparation for a life investigations on the heredity of resistance and
of research on the disease from which he suffered. susceptibility to tuberculosis and the cellular mech-
After significant studies at the National Jewish anisms concerned.
Hospital, first with H. J. Corper and soon inde- In the ensuing years, Lurie steadily developed
pendently, he accepted an invitation in 1926 to the possibilities in this line of investigation, demon-
strating the varying role of the body’s macrophages

Reprinted from Am. Rev. Respir. Dis. 95:694–696, in the destruction of tubercle bacilli, a field in
1967. Dr. Lurie was at the Henry Phipps Institute from which he was preeminent, and throwing light on
1926 until his death. Dr. Long was director there dur- numerous aspects of the host-parasite relation,
ing much of that time. extracellular factors in resistance, the role of age and

387
388 䡵 APPENDIX B

sex, and many other elements concerned in one Many honors were conferred on Dr. Lurie,
way or another in intrinsic resistance. including the Dearholt lectureship in 1952,
A practical application of his studies came about the Claude Bernard Medal of the University of
through a cooperative investigation, in association Montreal in 1953, and the Trudeau Medal of the
with W. F.Wells, of the germicidal effect of ultra- National Tuberculosis Association in 1956. Over
violet light in the prevention of air-borne infection. many years, the Association furnished financial
This has had extensive development since their support for his work. In 1960, the American Tho-
original reports. racic Society of the National Tuberculosis Associ-
In his later years, Lurie was much concerned ation honored him with senior membership.
with the role of endocrine factors in resistance to Throughout those years, Lurie took part faithfully,
tuberculosis. His research was significant in demon- and with a vigor all his own, in both the presen-
strating the effect of cortisone in reducing pro- tation of reports and assessment of research in the
tective inflammation and of thyroid hormones in meetings of the National Tuberculosis Associa-
enhancing it. tion. He was an excellent teacher, and a debater of
The later years of Lurie’s life were devoted to stature, in spite of his small physical frame, on all
reflection and orderly arrangement of facts learned problems in his chosen field. His published work
during his years of research.The Commonwealth is widely quoted, and his influence will be lasting.
Fund, which had been one of the major organi- It was of great importance in transferring some
zations among the several sources of support for his emphasis from other fields to the large one of
work, made it possible for him to devote a pro- intrinsic constitution.
longed period to preparation of a comprehensive Lurie is survived by his wife, Rose Gamoran
book on Resistance to Tuberculosis, which covered his Lurie, herself an educator and author of attain-
experimental studies on native and acquired ment in Jewish history, and by his son Abraham
defense mechanisms in relation to those of others. A. Lurie, an Associate Professor of Anesthesiology
This book is now a highly influential monograph at the School of Medicine of the University of
in the field. Rochester.
APPENDIX C
PUBLICATIONS OF MAX B. LURIE

These publications are grouped somewhat chrono- The importance of the growth of tubercle
logically by subject. Comments follow many of the bacilli as determined by gaseous tension. Am.
publications to explain their contributions, which Rev.Tuberc. 15:65–87.
are often not evident from their titles.These pub- 5. Corper, H. J., and M. B. Lurie. 1927.The
lications are mainly the ones listed and summarized variability of localization of tuberculosis in
in Lurie’s book (see publication 68). Chapter 1 in the organs of different animals. IV.The cellu-
Lurie’s book reviews the basic pathogenesis of lar factor in the susceptibility of the various
tuberculosis, specifically, the cell and tissue response organs. Am. Rev.Tuberc. 15:237–269.
to this infection, and his chapter 15 reviews the 6. Corper, H. J., M. B. Lurie, and N. Uyei.
entire book. 1927.The variability of localization of tuber-
culosis in the organs of different animals.V.
CULTIVATION OF TUBERCLE BACILLI The significance of localization and devel-
1. Lurie, M. B. 1923. A comparison of the opment of the bacilli and of the cellular reac-
sodium hydroxide and anti-formin methods tion in man and animals. Am. Rev. Tuberc.
for cultivating tubercle bacilli. Am. Rev.Tuberc. 15:389–398.
7:332–343. 7. Lurie, M. B. 1928. The fate of human and
Sodium hydroxide treatment preserved the
bovine tubercle bacilli in various organs of the
viability of tubercle bacilli in sputum samples bet- rabbit. J. Exp. Med. 48:155–182.
ter than did anti-formin treatment. In rabbits, after an intravenous injection, both bacil-
lary strains are essentially cleared in 2 to 4 months
from the liver, spleen, and bone marrow, but may
ORGAN RESISTANCE IN PRIMARY AND grow extracellularly in the lungs (and kidneys)
REINFECTION TUBERCULOSIS where cavitation (ulceration) occurs.
Various organs in the body differ in their ability to 8. Lurie, M. B. 1929.The fate of tubercle bacilli
stop the progression of tuberculous lesions. In rab- in the organs of reinfected rabbits. J. Exp. Med.
bits, lesions progress in the lungs and kidneys but 50:747–765.
heal in the liver, whereas in guinea pigs, lesions After an intravenous injection of virulent human-
progress in the lungs and liver but heal in the kid- type tubercle bacilli, the persisting infection (mostly
neys. Residual primary infection provides strong in cavitary lesions in the lungs and in ulcerated
acquired resistance against reinfection. Reviewed lesions in the kidneys) markedly reduced the mul-
in Lurie’s chapter 1 and, briefly, in chapter 7 in the tiplication of virulent bovine-type tubercle bacilli
given intravenously 6 months later. Multiplication
present volume. of virulent human-type tubercle bacilli given intra-
2. Corper, H. J., and M. B. Lurie. 1926.The venously 6 months later was reduced even fur-
variability of localization of tuberculosis in ther. (The bovine type is much more virulent for
the organs of different animals. I. Quantitative rabbits than the human type.) See chapter 2 in
relations in the rabbit, guinea pig, dog and Lurie’s book for more details.
monkey. Am. Rev.Tuberc. 14:662–679. 9. Lurie, M. B. 1932.The correlation between
3. Corper, H. J., and M. B. Lurie. 1926.The histological changes and fate of living tuber-
variability of localization of tuberculosis in cle bacilli in the organs of tuberculous rabbits.
the organs of different animals. II.The impor- J. Exp. Med. 55:31–54.
tance of the distribution of tubercle bacilli as Lesions in the lungs, liver, spleen, bone marrow, and
concerns differences of susceptibility of the kidneys were produced in rabbits by an intravenous
injection of (i) BCG (an early strain that was more
organs. Am. Rev.Tuberc. 14:680–705. virulent than current BCG),(ii) virulent human-type
4. Corper, H. J., M. B. Lurie, and N. Uyei. bacilli, or (iii) virulent bovine-type bacilli.The rab-
1927.The variability of localization of tuber- bits responded to each type of tubercle bacillus in a
culosis in the organs of different animals. III. similar manner, but the bovine type was harder to

389
390 䡵 APPENDIX C

destroy and caused extensive progressive disease. was almost eliminated by 4 weeks, whereas some
Lesions produced by all three types of tubercle bacil- of the more virulent BCG persisted in the lungs
lus healed in the liver. and kidneys for at least 4 months. Lurie quoted
Mature epithelioid cells (described in detail) from the literature that BCG persisted in the lymph
were found to be associated with the destruction nodes of monkeys for at least 7 months and in the
of tubercle bacilli—a correlation that Lurie con- lymph nodes of guinea pigs for at least 19 months.
sidered to be one of his major contributions. Monkeys and guinea pigs are more susceptible to
Langhans’ giant cells occurred in the lesions virulent tubercle bacilli than are rabbits. See chap-
after the local multiplication of tubercle bacilli had ter 1 in Lurie’s book and chapters 15 and 23 in the
ceased. Giant cells frequently formed around bits present volume.
of caseous material.
Caseous necrosis was present when the host
became tuberculin positive and, therefore, delayed-
type hypersensitivity was considered to be the SPREAD OF TUBERCULOSIS FROM
cause of caseous necrosis. INFECTED TO UNINFECTED ANIMALS
Lymphocytes did not (directly) destroy tuber- This series of reports documents the respiratory
cle bacilli, whereas macrophages (epithelioid cells) and alimentary routes of infection among guinea
did so. pigs and rabbits housed in an animal room.
In expanding tuberculous lesions containing 12. Lurie, M. B. 1930. Experimental epidemi-
many tubercle bacilli, some mononuclear cells were
multiplying (shown by the presence of mitotic
ology of tuberculosis: the effect of crowding
figures). upon tuberculosis in guinea pigs, acquired by
Chapter 1 in Lurie’s book reviews these subjects. contact and by inoculation. J. Exp. Med.
Chapter 10 in the present volume provides insights 51:729–741.
concerning mononuclear cell division. 13. Lurie, M. B. 1930. Experimental epidemi-
10. Lurie, M. B. 1933.A correlation between the ology of tuberculosis: air-borne contagion in
histological changes and the fate of living tuberculosis in an animal room. J. Exp. Med.
tubercle bacilli in the organs of reinfected 51:743–751.
rabbits. J. Exp. Med. 57:181–201. 14. Lurie, M. B. 1930. Experimental epidemi-
Primary infection was produced by intravenous vir- ology of tuberculosis: the effect of eliminat-
ulent human-type tubercle bacilli in 30 rabbits. Six ing exposure to enteric infection on the inci-
months later, the bacilli had almost disappeared dence and course of tuberculosis acquired by
from the liver, spleen, and bone marrow, and their
normal guinea pigs confined with tuberculous
number was usually reduced in the lungs and kid-
neys. At that time, 15 of these rabbits were rein- cage mates. J. Exp. Med. 51:753–768.
fected intravenously with virulent human-type 15. Lurie, M. B. 1930. Experimental epidemi-
bacilli, and 15 were reinfected intravenously with ology of tuberculosis: the route of infection in
virulent bovine-type bacilli. At that time, as con- naturally acquired tuberculosis of the guinea
trols, 13 uninfected rabbits were injected intra- pig. J. Exp. Med. 51:769–776.
venously with virulent human-type bacilli, and
13 uninfected rabbits were similarly injected with
virulent bovine-type tubercle bacilli.
In the reinfected rabbits, the formation of new NATURAL AIRBORNE INFECTION AND
tubercles was accelerated, and a faster maturation of REINFECTION WITH TUBERCLE BACILLI
epithelioid cells occurred. Also, both the human- In this series of reports (and those in the next two
and bovine-type bacilli of reinfection were often sections), uninfected rabbits inhaled air in an ani-
destroyed, while the human-type bacilli of the pri- mal room where other rabbits (caged separately)
mary infection were still multiplying (extracellularly) were shedding virulent bovine-type tubercle bacilli
within cavitary lesions in the lungs and kidneys.
in their urine from infected kidneys.These bacilli
11. Lurie, M. B. 1934. The fate of BCG and
became airborne as the rabbits moved about on the
associated changes in organs of rabbits. J. Exp.
bedding.The following principles were established:
Med. 60:163–178.
(i) Residual primary infection prevented or
In these experiments, BCG of normal virulence
was given intravenously.This BCG increased in the
reduced the severity of reinfection.These exper-
organs of the rabbits for about 2 weeks and then iments tell us how much immunity might be
declined, similar to the more virulent BCG expected in rabbits from a truly effective vaccine
(described in publication 9). The normal BCG (see chapters 22 and 25 in the present volume).
APPENDIX C 䡵 391

(ii) Continuous exposure to low numbers of This is Lurie’s main report on the efficacy of UV
tubercle bacilli had no effect on the course of the light. See publication 21 for more experimental
primary infection.These experiments suggest that details.
hosts with active tuberculosis are well immunized 19. Lurie, M. B. 1946. Control of air-borne
against low-dose exogenous reinfection. See pub- contagion of tuberculosis. Am. J. Nurs. 46:808–
lications 22, 23, 24, and 26 for additional insights 810.
on immunity to reinfection. This report discusses additional measures to prevent
the spread of the tubercle bacillus to human beings,
16. Lurie, M. B. 1933. Experimental epidemi-
such as wearing masks and oiling the bed covers.
ology of tuberculosis: the effect of a primary
20. Lurie, M. B. 1947. Experimental air-borne
infection on contact tuberculosis in rabbits. J.
tuberculosis and its control. Am. Rev. Tuberc.
Exp. Med. 58:305–327.
55:124–127.
During the first 6 months, subcutaneous vaccina-
This is a review of the subject.
tion with virulent human-type bacilli reduced the
incidence of natural infection with virulent bovine-
type bacilli in an animal room: 64% of the unvac- RESISTANCE TO THE ESTABLISHMENT
cinated rabbits, whereas 37% of vaccinated rabbits OF TUBERCULOSIS VERSUS RESISTANCE
developed tuberculosis, and the disease was less TO ITS PROGRESSION
severe in the vaccinated group. During the next 6 21. Lurie, M. B. 1944. Experimental epidemi-
months, no appreciable difference between the ology of tuberculosis: hereditary resistance to
two groups was found. attack by tuberculosis and to the ensuing dis-
17. Lurie, M. B., and J. Becker. 1946. Effect of ease, and the effect of the concentration of
exogenous superinfection on naturally tubercle bacilli upon these two phases of resis-
acquired bovine tuberculosis in inbred rabbits. tance. J. Exp. Med. 79:573–589.
Proc. Soc. Exp. Biol. Med. 63:465–469. In these experiments, virulent bovine-type tuber-
The disease produced in inbred rabbits removed cle bacilli were shed in the urine of infected rab-
from the source of natural contagion in the ani- bits, and uninfected inbred rabbits in nearby cages
mal room as soon as they became tuberculin pos- contracted tuberculosis over a period of months.
itive was compared to the disease in rabbits As a group, the inbred resistant rabbits established
remaining with the source. No appreciable dif- a primary pulmonary lesion (indicated by a con-
ference was found. In other words, continuous version of their tuberculin skin test) a few months
exogenous inhalation of low doses of bovine-type sooner than did the inbred susceptible rabbits.
bacilli did not affect the course of an active pri- However, the resistant rabbits controlled the
mary infection with virulent bovine-type bacilli. progress of their disease better and lived twice as
Infection with the bovine type is uniformly fatal long. The alveolar macrophages of the resistant
in rabbits. strains apparently trapped inhaled bacilli more
effectively than those of susceptible strains (see
publication 61). Also, the resistant rabbits were
larger and therefore probably breathed more air.
USE OF ULTRAVIOLET LIGHTS See chapter 6 in Lurie’s book and chapter 12 in the
TO PREVENT TUBERCULOSIS present volume.
These studies prove that UV lights are highly
effective in preventing tuberculosis. These lights SPREAD OF TUBERCULOSIS
must be shielded to prevent damage to the eyes of FROM PRIMARY SITE OF
personnel from UV radiation.The shielding should INFECTION IN THE SKIN
permit these lights to irradiate only the ceiling of This series of reports covers many aspects of
the room, and to irradiate both the ceiling and the acquired resistance to tuberculosis. Publication 22
floor directly beneath doorways. See chapter 6 in is the primary study. It describes the fate of virulent
Lurie’s book (p. 159–164) and chapter 1 in the pre- bovine-type tubercle bacilli injected subcutaneously
sent volume. in agar into rabbits that (i) were immunized with
18. Lurie, M. B. 1944. Experimental epidemi- BCG intravenously, (ii) were immunized by a slowly
ology of tuberculosis: the prevention of nat- progressive tuberculosis produced by intravenous
ural air-borne contagion of tuberculosis in bovine-type tubercle bacilli (Ravenel), or (iii) were
rabbits by ultraviolet irradiation. J. Exp. Med. nonimmunized controls. Publication 24 describes
79:559–572. similar studies in guinea pigs.
392 䡵 APPENDIX C

In addition, it describes studies in rabbits in coarse and allowed the bacilli to pass into the
which the reinfecting bacilli in agar were contained lymphatics. Therefore, many reinfecting bacilli
in colloidin-coated silk bags placed intraperitoneally. reached the draining lymph nodes in rabbits,
whereas only a few bacilli reached these lymph
The walls of these bags allowed body fluids to enter nodes in guinea pigs.
but prevented host cells, such as macrophages, from Tuberculous humans are very tuberculin sensi-
doing so. tive (even more so than guinea pigs), so clotting of
The subcutaneous agar (not in bags) had allowed the lymph should readily occur. However, the fib-
some cells to reach the bacilli in the agar. Both rin meshwork in humans is coarse (similar to that
experiments, however, produced the same results: in rabbits) and may not stop the spread of bacilli as
acellular body fluids from immunized hosts inhib- completely as does the fibrin of guinea pigs.
What matters, however, is the fate of these
ited bacillary growth. See chapter 3 in Lurie’s book. bacilli after they reach the draining lymph nodes.
22. Lurie, M. B. 1936. On the mechanism of The bacilli in the draining lymph nodes of immu-
immunity in tuberculosis: the host-parasite nized guinea pigs and rabbits were inhibited much
relationship under the conditions of a localized more than those in the draining lymph nodes of
agar focus of infection and the generalization the nonimmunized controls.
Immunity in tuberculosis is a local phenomenon,
of the disease in normal and immunized rab- with each lesion developing and healing (or pro-
bits. J. Exp. Med. 63:923–946. gressing) somewhat independently of the other
The acellular body fluids of the immunized rab- lesions in the host (see chapter 5 in the present vol-
bits were markedly bacteriostatic in vivo when ume). Tubercle bacilli can be inhibited at sites of
compared with those of the nonimmunized con- reinfection and yet multiply in a lesion produced
trols.The reason is not known, but antibodies were by the primary infection.
probably not responsible. 25. Lurie, M. B. 1939. Relative spread of par-
The localization of tubercle bacilli at the portal ticulate and diffusible substances in the skin of
of entry is a minor component of acquired cellu-
lar resistance in tuberculosis. It occurred only with male and female rabbits. Proc. Soc. Exp. Biol.
low-dose reinfection, whereas with high-dose rein- Med. 42:741–744.
fection a more rapid dissemination of bacilli to 26. Lurie, M. B. 1950. Mechanisms affecting
the draining lymph nodes occurred. spread in tuberculosis. Ann. N.Y.Acad. Sci. 52:
The activation of macrophages to destroy tuber- 1074–1090.
cle bacilli (shown histologically by the presence of This is a review of the publications in this section.
mature epithelioid cells) is the major method by
which the host controls this disease.
Polymorphonuclear leukocytes were ineffective
at inhibiting the growth of tubercle bacilli and
ROLE OF MONONUCLEAR
soon died.
PHAGOCYTES IN TUBERCULOSIS
The immunity to reinfection produced by a 27. Lurie, M. B. 1939. Studies on the mechanism
slowly progressive primary infection with viru- of immunity in tuberculosis: the mobilization
lent bovine-type tubercle bacilli (Ravenel) was of mononuclear phagocytes in normal and
greater than that produced by BCG. immunized animals and their relative capaci-
23. Lurie, M. B. 1936. Further studies on the ties for division and phagocytosis. J. Exp. Med.
mechanism of immunity to tuberculosis. J. 69:579–609.
Bacteriol. 32:671–672. Exudate macrophages from tuberculous rabbits and
24. Lurie, M. B. 1939. Studies on the mechanism guinea pigs were more “activated” than were those
of immunity in tuberculosis.The role of extra- from normal controls: the rate of mobilization of
cellular factors and local immunity in fixation macrophages and their phagocytic abilities were
and inhibition of growth of tubercle bacilli. measured. Macrophages from tuberculous hosts also
responded to nonspecific irritants more vigorously
J. Exp. Med. 69:555–578. than did macrophages from uninfected hosts. Dis-
In guinea pigs, fibrin formed readily in tuberculous cussed in chapter 3 in Lurie’s book.
lesions of reinfection.This fibrin was a fine mesh- 28. Lurie, M. B. 1942. Studies on the mechanism
work that partly obstructed the lymphatics and
impeded the spread of tubercle bacilli to the drain-
of immunity in tuberculosis.The fate of tuber-
ing lymph nodes. cle bacilli ingested by mononuclear phago-
In rabbits, relatively little fibrin formed in the cytes derived from normal and immunized
lesions of reinfection. The fibrin meshwork was animals. J. Exp. Med. 75:247–268.
APPENDIX C 䡵 393

Mononuclear exudate cells that had ingested tuber- resistant strains of rabbits developed tuberculin
cle bacilli in vitro were transferred into the ante- sensitivity and antibodies more rapidly than did the
rior eye chambers of a normal rabbit. The cells susceptible strains of rabbits. Local immunity (fol-
(macrophages, lymphocytes, and probably den- lowing the intradermal injection of virulent
dritic cells) were from immunized (tuberculous) bovine-type tubercle bacilli) was most effective in
and control rabbits in the presence of immune the resistant rabbits. Heat-killed virulent bovine-
and control serum. The macrophages from the type tubercle bacilli could immunize resistant
immune host inhibited the growth of phagocytized strains of rabbits, but could not immunize suscep-
tubercle bacilli, whereas those from the normal tible strains to any appreciable degree.
host did not. Immune serum had no appreciable IV. Heredity Constitution and Tuberculosis.
effect. Described in chapter 4 in Lurie’s book and Summary and Discussion (p. 112–125).All of these
interpreted in chapter 5 in the present volume. studies support the principle that, in tuberculosis,
acquired immunity is superimposed on and deter-
mined by the native resistance of the host. Host
TUBERCULOSIS IN INBRED RESISTANT
resistance can be overwhelmed by large doses of
AND SUSCEPTIBLE RABBITS
virulent tubercle bacilli.
29. Lurie, M. B. 1938. Role of inherited natural
resistance to tuberculosis. Proc. Soc. Exp. Biol.
Med. 39:176–181. EFFECTS OF ESTROGEN AND
CHORIONIC GONADOTROPIN
30. Lurie, M. B. 1938. Nature of inherited nat- ON TUBERCULOSIS
ural resistance to tuberculosis. Proc. Soc. Exp. These hormones affected the spread of the disease
Biol. Med. 39:181–187. from the site of bacillary injection in the skin: estro-
31. Lurie, M. B. 1941. Heredity, constitution gens retarded the spread,and chorionic gonadotropin
and tuberculosis.An experimental study. Am. enhanced it.However,these hormones had no effect
Rev.Tuberc. 44(Suppl. 3):1–125. on the overall resistance of the host to tuberculosis.
This monograph describes the development of Reviewed in chapter 7 in Lurie’s book and in chap-
Lurie’s inbred rabbit strains and the characteristics
of the tuberculosis produced in them. It is sum-
ter 17 in the present volume.
marized in chapter 6 in Lurie’s book, and its four 32. Lurie, M. B., and P. Zappasodi. 1942.
parts are: Effect of chorionic gonadotropin on the
I. Inherited Native Resistance and Natural Res- spread of particulate substances in the skin of
piratory Contagion (p. 1–67). The origins and rabbits. Arch. Pathol. 34:151–166.
breeding of Lurie’s susceptible and resistant strains
33. Lurie, M. B., S. Abramson, and M. J.
of rabbits are described, as well as the characteris-
tics of tuberculosis produced in each strain by vir- Allison. 1949. Constitutional factors in resis-
ulent bovine-type bacilli inhaled in an animal room tance to infection. I.The effect of estrogen and
over a period of months. The disease in the sus- chorionic gonadotropin on the course of
ceptible rabbits was the “childhood type,” with tuberculosis in highly inbred rabbits. Am. Rev.
caseous hilar lymph nodes and hematogenous spread Tuberc. 59:168–185.
of the disease.The disease in the resistant rabbits was 34. Lurie, M. B.,T. N. Harris, S. Abramson,
the “adult type,” with cavity formation and
bronchial spread of the disease and minimal hilar
and M. J. Allison. 1949. Constitutional fac-
lymph node involvement. tors in resistance to infection. II.The effect of
II. Inherited Native Resistance and Artificial estrogen on tuberculin skin sensitivity and on
Respiratory Contagion (p. 68–79).Tuberculosis was the allergy of the internal tissues. Am. Rev.
produced in the inbred resistant and susceptible Tuberc. 59:186–197.
strains of rabbits by a known dose of virulent 35. Lurie, M. B., S. Abramson, A. G. Hep-
bovine-type bacilli given subcutaneously, intra- pleston, and M. J. Allison. 1949. Constitu-
venously, or by aerosol.After the aerosol infection,
multiple primary pulmonary lesions were produced. tional factors in resistance to infection. III. On
In contrast, after natural contagion, only a single pri- the mode of action of estrogen and
mary pulmonary lesion was produced. Otherwise, gonadotropin on the progress of tuberculosis.
the disease in each rabbit strain resembled that pro- Am. Rev.Tuberc. 59:198–218.
duced by natural contagion in the animal room. This report analyzes other effects of these hor-
III. Experimental Study of Certain Factors mones on the host. Unexpectedly, estrogen markedly
Related to Hereditary Constitutional Resistance to reduced the amount of amyloid degeneration in the
Tuberculosis (with P. Zappasodi) (p. 80–111).The spleens of rabbits dying of chronic tuberculosis.
394 䡵 APPENDIX C

CHEMOTHERAPY OF TUBERCULOSIS prepare suspensions containing only particles of 1


36. Lurie, M. B., and J. Stokes, Jr. 1943.The to 3 bacilli.A diagram of the modified Wells aerosol
effect of promin on experimental tuberculo- apparatus that Lurie used is shown here and in pub-
lication 41.
sis in the rabbit. J. Bacteriol. 45:194–195.
37. Lurie, M. B. 1949.The use of the rabbit in 41. Lurie, M. B., and S. Abramson. 1948.
experimental chemotherapy of tuberculosis. Reproduction of human ulcerative pulmonary
Ann. N.Y.Acad. Sci. 52:627–636. tuberculosis in rabbits by quantitative natural
airborne contagion. Proc. Soc. Exp. Biol. Med.
In humans, antimicrobial-resistant strains of tuber-
cle bacilli develop during their inordinate extra- 69:531–537.
cellular growth in the liquefied caseum in the walls Cavities were produced in inbred strain III rab-
of pulmonary tuberculous cavities. Rabbits exposed bits, which had recently been obtained from the
to aerosols of virulent bovine-type tubercle bacilli Jackson Laboratory in Bar Harbor, Me. These
readily form such cavities.Therefore, the rabbit is rabbits were sensitized with heat-killed virulent
the best commonly available laboratory animal in bovine-type tubercle bacilli (Ravenel) and chal-
which to study the development of antimicrobial lenged by aerosol with living tubercle bacilli of the
resistance within such cavities. same type. Such sensitization hastened the for-
mation of cavities (see chapter 4 in the present
volume).
BOOK REVIEW
38. Lurie, M. B. 1944. Review of The Pathogen-
FATE OF INHALED HUMAN-TYPE
esis of Tuberculosis by Arnold R. Rich (Charles
TUBERCLE BACILLI IN INBRED
C Thomas, Springfield, Ill.). Science 100:407– RABBITS; THE TUBERCLE-COUNT
408. METHOD TO ASSESS HOST
Lurie praises Rich’s book but admits that some of RESISTANCE
Rich’s conclusions are not shared by others. How- 42. Lurie, M. B., S. Abramson, and A. G.
ever, there is wide acceptance of Rich’s claim that Heppleston. 1952. On the response of
tuberculin sensitivity causes caseous necrosis and that
genetically resistant and susceptible rabbits to
tubercle bacilli are nontoxic before tuberculin sen-
sitivity develops. the quantitative inhalation of human-type
tubercle bacilli and the nature of resistance to
tuberculosis. J. Exp. Med. 95:119–134.
QUANTITATIVE AIRBORNE INFECTION
Human-type tubercle bacilli are never fully vir-
Lurie and William F.Wells, a brilliant aerodynamic ulent for rabbits. Rabbits usually recover from
engineer,constructed an apparatus that could expose infection with human-type bacilli, but not from
six rabbits (simultaneously and quantitatively) to infection with bovine-type bacilli. To produce
aerosols of tubercle bacilli.After 1941,Lurie used this one grossly visible primary tubercle 5 weeks after
apparatus in almost all of his studies. infection, inbred susceptible rabbits must inhale 50
to 100 units of 1 to 3 human-type bacilli,
39. Wells,W. F., and M. B. Lurie. 1941. Exper-
whereas inbred resistant rabbits must inhale 600
imental airborne diseases: quantitative natural to over 1,000 units. At 24 weeks after infection
respiratory contagion of tuberculosis. Am. J. with human-type bacilli, inbred resistant rabbits
Hyg. 34(Sect. B):21–40. usually showed no evidence of disease, but the
The original quantitative aerosol apparatus for inbred susceptible rabbits showed many residual
infecting rabbits with tubercle bacilli is described. lesions.
The number of bacilli estimated to be inhaled by In contrast, fully virulent bovine-type tubercle
the rabbits into the alveolar spaces was the same as bacilli produced one such grossly visible tubercle
the number obtained by culturing their lungs 1 day for every 3 bacillary units inhaled, in both resistant
after infection. and susceptible inbred rabbits. Two-thirds of the
40. Lurie, M. B.,A. G. Heppleston, S.Abram- inhaled bacilli impinge on the bronchial mucosa
son, and I. B. Swartz. 1950. An evaluation (which is quite resistant to tuberculosis).
The inhalation of human-type bacilli is the basis
of the method of quantitative airborne infec- of Lurie’s tubercle-count method to measure
tion and its use in the study of the pathogen- mycobacterial virulence, genetic host resistance,
esis of tuberculosis.Am. Rev.Tuberc. 61:765–797. and vaccine efficacy.The number of grossly visible
This report provides details on how these aerosol pulmonary tubercles is dependent on the power of
experiments were performed, including how to pulmonary alveolar macrophages to initially destroy
APPENDIX C 䡵 395

the inhaled bacillus, and is also dependent on the See chapter 8 in Lurie’s book and chapter 11 in the
acquired immunity that prevents early microscopic present volume.
lesions from reaching grossly visible size. See chap- The following publications contain some redun-
ter 8 in Lurie’s book and chapter 11 in the present
volume. dancies, because Lurie participated in several sym-
posia on glucocorticosteroids that were subse-
quently published.
THE TIME OF HEALING OF BCG LESIONS
MEASURES THE NATIVE RESISTANCE OF 44. Lurie, M. B., P. Zappasodi, A. M. Dan-
THE HOST; THE IMMUNITY OF HOSTS nenberg, Jr., and I. B. Swartz. 1951. Con-
WITH POOR NATIVE RESISTANCE IS stitutional factors in resistance to infection: the
NOT APPRECIABLY INCREASED BY BCG effect of cortisone on the pathogenesis of
VACCINATION tuberculosis. Science 113:234–237.
43. Lurie, M. B., P. Zappasodi, E. Cardona- 45. Lurie, M. B., P. Zappasodi, A. M. Dan-
Lynch, and A. M. Dannenberg, Jr. 1952. nenberg, Jr., and E. Cardona-Lynch.
The response to the intracutaneous inocula- 1952. Constitutional factors in resistance to
tion of BCG as an index of native resistance infection: the effect of cortisone on the patho-
to tuberculosis. J. Immunology 68:369–387. genesis of tuberculosis and its implications
The rate of healing of dermal BCG lesions reflected for nonspecific and allergic inflammations
how Lurie’s inbred rabbit strains controlled the
progress of infection with virulent tubercle bacilli; and infectious diseases in general, p. 247–253.
i.e., it measured their innate and acquired resistance In Advances in Medicine and Surgery (Graduate
to this disease.These studies apply to groups of rab- School of Medicine of the University of
bits but were not precise for individuals. Rabbit Pennsylvania). The W. B. Saunders Co.,
strains with poor innate resistance did not develop Philadelphia, Pa.
as much immunity from BCG vaccination as did 46. Lurie, M. B., P. Zappasodi, A. M. Dan-
rabbit strains with good innate resistance. See chap-
ter 9 in Lurie’s book and chapter 23 in the present nenberg, Jr., and E. Cardona-Lynch. 1953.
volume. The effect of cortisone and ACTH on the
pathogenesis of tuberculosis. Ann. N.Y.Acad. Sci.
56:779–792.
EFFECTS OF GLUCOCORTICOSTEROIDS
ON TUBERCULOSIS 47. Lurie, M. B., P. Zappasodi, A. M. Dan-
Following the inhalation of virulent human-type nenberg, Jr., and E. Cardona-Lynch. 1953.
tubercle bacilli (H37Rv), rabbits treated with cor- Constitutional factors in resistance to infection:
tisone developed 3 to 4 times as many grossly vis- the effect of cortisone on the pathogenesis of
ible primary pulmonary tubercles as did the tuberculosis, p. 84–99. In G. Shwartzman (ed.),
untreated controls. However, the tubercles in the The Effect of ACTH and Cortisone upon Infection
cortisone-treated host were smaller and had more and Resistance. Columbia University Press, New
compact caseous centers.These centers contained York, N.Y.
dead macrophages just teeming with tubercle 48. Lurie, M. B., and P. Zappasodi. 1955. On
bacilli.The dead macrophages were mostly within the mode of action of cortisone on the
alveoli, and very little perifocal inflammation pathogenesis of tuberculosis and its implica-
occurred in the surrounding alveolar walls.There- tions for the nature of genetic resistance to
fore, the spread of bacilli to the hilar nodes was the disease, p. 246–258. In Ciba Foundation
reduced. The tuberculin sensitivity of the corti- Symposium on Experimental Tuberculosis.
sone-treated group was also reduced. Churchill, London, U.K.
When the cortisone treatment was stopped after 49. Lurie, M. B. 1955. On the role of hormones
6 to 9 weeks, massive pulmonary tuberculosis and in experimental tuberculosis. Adv.Tuberc. Res.
death occurred in Lurie’s inbred susceptible rab- 6:18–48.
bits, and progressive disease with cavity forma- 50. Kass, E. H., O. Hechter, T. W. Mou, and
tion occurred in Lurie’s inbred resistant rabbits. In M. B. Lurie. 1955. Effects of adrenal steroids
untreated susceptible and resistant rabbits, human- on resistance to infection: differences in
type tubercle bacilli (H37Rv) never cause death. the relative amounts of corticosterone and
396 䡵 APPENDIX C

hydrocortisone secreted and in their biologic quantitative inhalation of tubercle bacilli: their
effects. Arch. Intern. Med. 96:397–402. significance for the nature of genetic resis-
Corticosterone is the predominant adrenal steroid tance. Am. Rev. Respir. Dis. 85:553–569.
secreted by normal rabbits. After adrenocorti- The growth curves of viable virulent Mycobac-
cotropin treatment, they secrete predominantly terium tuberculosis (H37Rv) and Mycobacterium bovis
hydrocortisone. Hydrocortisone decreases resis- (Ravenel) in rabbit lungs showed the same pattern,
tance to infection, whereas corticosterone does i.e., a logarithmic phase followed by a stationary
not have this effect. phase. M. bovis, however, grew for a longer period
of time in the logarithmic stage and reached higher
CROSSBREEDING INBRED RESISTANT titers before the stationary stage occurred. See
WITH INBRED SUSCEPTIBLE RABBITS chapter 15 in the present volume.
51. Lurie, M. B., P. Zappasodi, A. M. Dan-
nenberg, Jr., and G. H.Weiss. 1952. On the TISSUE RESPONSES TO MYCO-
mechanism of genetic resistance to tubercu- BACTERIA: M.TUBERCULOSIS,
losis and its mode of inheritance. Am. J. Hum. M. BOVIS, AND MYCOBACTERIUM
Genet. 4:302–314. LEPRAE
54. Lurie, M. B. 1955.A pathogenetic relation-
Crossbreeding of resistant and susceptible rabbits
produced an F1 hybrid of intermediate resistance. ship between tuberculosis and leprosy: the
Backcrossing this F1 hybrid with susceptible rab- common denominators in the tissue response
bits did not restore susceptibility in the offspring. to mycobacteria, p. 340–343. In Ciba Founda-
However, backcrossing this F1 hybrid with resistant tion Symposium on Experimental Tuberculosis.
rabbits did restore high resistance in the offspring. Churchill, London, U.K.
Therefore, resistant genes were dominant. See chap-
ter 10 in Lurie’s book and chapter 14 in the pre-
sent volume. EFFECTS OF THYROID HORMONES ON
TUBERCULOSIS
GROWTH CURVES OF VIRULENT Hyperthyroidism increased host resistance in only
HUMAN-TYPE TUBERCLE BACILLI certain inbred rabbit strains, but hypothyroidism
IN RABBIT LUNGS FOLLOWING decreased host resistance in every rabbit strain
AEROSOL INHALATION evaluated. See chapter 17 in the present volume.
52. Lurie, M. B., P. Zappasodi, and C.Tick- 55. Lurie, M. B., and G. S. Ninos. 1956.The
ner. 1955. On the nature of genetic resistance effect of triiodothyronine and propylthiouracil
to tuberculosis in the light of the host-parasite on native resistance to tuberculosis. Am. Rev.
relationships in natively resistant and suscepti- Tuberc. 73:434–437.
ble rabbits. Am. Rev.Tuberc. 72:297–329. 56. Lurie, M. B., P. Zappasodi, R. S. Levy,
Rabbit lung homogenates were cultured at various and R. G. Blaker. 1959. On the role of the
times after the inhalation of virulent human-type thyroid in native resistance to tuberculosis. I.
tubercle bacilli (H37Rv).There was an early log-
The effect of hyperthyroidism. Am. Rev.Tuberc.
arithmic increase in the number of viable tubercle
bacilli in the lungs. The logarithmic phase was 79:152–179.
then followed by a stationary (plateau) phase.These 57. Lurie, M. B., P. Zappasodi, R. S. Levy,
studies formed the basis of my concept that tissue- and R. G. Blaker. 1959. On the role of the
damaging delayed-type hypersensitivity has a ben- thyroid in native resistance to tuberculosis.
eficial role in tuberculosis. See chapters 2, 5, and 15 II.The effect of hypothyroidism: the mode of
in the present volume. action of thyroid hormones. Am. Rev.Tuberc.
79:180–203.
COMPARISON OF TUBERCULOSIS
PRODUCED IN RABBITS BY AN
AEROSOL OF VIRULENT HUMAN- BIPHASIC IMMUNE RESPONSE
TYPE TUBERCLE BACILLI WITH TO SUBCUTANEOUS BCG
THAT PRODUCED BY VIRULENT 58. Allison, M. J., P. Zappasodi, and M. B.
BOVINE-TYPE TUBERCLE BACILLI Lurie. 1962. The correlation of a biphasic
53. Allison, M. J., P. Zappasodi, and M. B. metabolic response with a biphasic response
Lurie. 1962. Host-parasite relationships in in resistance to tuberculosis in rabbits. J. Exp.
natively resistant and susceptible rabbits on Med. 115:881–890.
APPENDIX C 䡵 397

Rabbits were given a primary intradermal injection 60. Allison, M. J., P. Zappasodi, and M. B.
of BCG (30 ⫻ 106) 6 weeks before the same dose Lurie. 1962. Metabolic studies of mono-
was given subcutaneously as a booster.When chal- nuclear cells from rabbits of varying genetic
lenged by aerosol with virulent human-type tuber-
cle bacilli (H37Rv) 4 days after the BCG booster,
resistance to tuberculosis. II. Studies on cells
the rabbits had twice as many grossly visible pri- from BCG-vaccinated animals. Am. Rev. Respir.
mary tubercles (5 weeks after challenge) than did Dis. 85:364–372.
the unvaccinated controls.When challenged with
H37Rv 28 days after the BCG booster, the rabbits Note:Allison continued these studies as parts III
had half as many visible tubercles (5 weeks later) and IV of his series: III. Studies on variation of thy-
than did the controls. In other words, the BCG roid function. Am. Rev. Respir. Dis. 86:513–517,
booster made the host more susceptible to exoge- 1962. IV. Studies on cortisone-treated animals.
nous infection during the first week and more Am. Rev. Respir. Dis. 87:384–388, 1963.
resistant thereafter.
These experiments should be repeated with In general, compared to controls, hyperthy-
only a single intradermal primary BCG injection roid rabbits had more activated cells in the
and no booster—similar to what is commonly
used in humans. The negative phase of resistance oil-induced peritoneal exudates, whereas
to challenge by virulent bacilli (just described) cortisone-treated rabbits had fewer activated
was at a time when the host was tuberculin pos- cells in these exudates.
itive and the immune forces were actively elim-
inating the rather large dose of BCG in the vac-
cine booster. After a single primary intradermal
BCG injection, the host immune forces are PHAGOCYTIC ABILITIES OF
mobilized more gradually. Therefore, the negative ALVEOLAR MACROPHAGES
phase of host resistance would probably not FROM INBRED RESISTANT
occur. AND SUSCEPTIBLE RABBITS
61. Henderson, H. J., A. M. Dannenberg,
Jr., and M. B. Lurie. 1963. Phagocytosis of
METABOLIC STUDIES ON INTACT tubercle bacilli by rabbit pulmonary alveolar
OIL-INDUCED PERITONEAL MONO- macrophages and its relation to native resis-
NUCLEAR CELLS (MACROPHAGES) tance to tuberculosis. J. Immunol. 91:553–556.
FROM INBRED RESISTANT AND
SUSCEPTIBLE RABBITS Alveolar macrophages from inbred resistant rabbits
ingested twice as many tubercle bacilli in vitro than
Various metabolic activities of oil-induced peri- did those from inbred susceptible rabbits.Perhaps,the
toneal macrophages were sometimes higher in greater phagocytic ability of alveolar macrophages
Lurie’s inbred resistant rabbit strain III than in his from the resistant rabbits enabled them to trap more
susceptible strain CaC. However, peritoneal bacilli from inhaled air in the animal room:they con-
macrophages induced by mineral oil are only verted their tuberculin skin test several months
weakly activated when compared with pulmonary sooner than did the susceptible rabbits. See chapter
6 in Lurie’s book, publication 21 in this appendix,
alveolar macrophages and mature epithelioid cells
and chapter 12 in the present volume.
within tuberculous lesions (see publications 17, 26,
32, and 96 in appendix D).Alveolar macrophages
become highly activated by the continuous inges-
tion of dust and microorganisms, and macrophages VALY MENKIN: A BIOGRAPHICAL
(epithelioid cells) within tuberculous lesions SKETCH
become highly activated by cytokines from anti- 62. Lurie, M. B. 1961. On the work of Valy
gen-specific lymphocytes. Evidently, mineral oil is Menkin: a biographical sketch. Anat. Rec. 140:
a rather poor macrophage activator. 234–236.
59. Allison, M. J., P. Zappasodi, and M. B. Valy Menkin pioneered the cytokine field, but the
Lurie. 1961.Metabolic studies of mononuclear purification methods available at that time were not
sufficient to identify individual cytokines. Menkin
cells from rabbits of varying genetic resistance called the chemotactic fraction of his inflammatory
to tuberculosis. I. Studies on cells of normal exudates “leukotaxin.” See V. Menkin, Biochemical
non-infected animals. Am. Rev. Respir. Dis. 84: Mechanisms in Inflammation, 2nd ed. Charles C
364–370. Thomas, Springfield, Ill., 1956.
398 䡵 APPENDIX C

REVIEW ARTICLES ON THE EFFECTS 66. Lurie, M. B. 1950. Native and acquired resis-
OF VARIOUS HORMONES ON THE tance to tuberculosis. Am. J. Med. 9:591–610.
PATHOGENESIS OF TUBERCULOSIS This is an excellent review of the subject up to
63. Lurie, M. B. 1960. The reticuloendothelial 1950.
system, cortisone, and thyroid function: their 67. Lurie, M. B., and A. M. Dannenberg, Jr.
relation to native resistance to infection. Ann. 1965. Macrophage function in infectious dis-
N.Y.Acad. Sci. 88:83–98. ease with inbred rabbits. Bacteriol. Rev. 29:466–
64. Lurie, M. B. 1965. Immunological aspects of 476.
steroid therapy. Arch. Environ. Health 11:235– This review briefly describes the characteristics of
241. innate and acquired resistance to tuberculosis,as well
This review covers some of the literature that does as other characteristics, found in Lurie’s inbred rab-
not appear in Lurie’s other reports on this subject. bits. See chapters 13 and 14 of the present volume.

LURIE’S BOOK
REVIEW ARTICLES ON NATIVE
68. Lurie, M. B. 1964. Resistance to Tuberculosis.
AND ACQUIRED RESISTANCE
TO TUBERCULOSIS Experimental Studies in Native and Acquired
65. Lurie, M. B. 1950. The Cyclopedia of Medicine, Defensive Mechanisms. Harvard University
Surgery and Specialties, vol. 7. Immunology of Press, Cambridge, Mass.
Tuberculosis, p. 1–30. F. A. Davis Company, This book summarizes and interprets most of
Lurie’s publications.
Philadelphia, Pa.
The literature on immunology of tuberculosis up
to 1950 is reviewed. Unfortunately, the reader must BIOGRAPHY OF MAX B. LURIE
refer to Lurie’s article in the 1945 edition of the 69. Fahy,A. 1966. Scientists at work: Max Lurie,
Cyclopedia for all references before 1940. M.D. Bull. Natl.Tuberc.Assoc. 52:10.
APPENDIX D
PUBLICATIONS OF ARTHUR M. DANNENBERG, JR.

These publications are grouped somewhat chrono- the University of Pennsylvania). The W. B.
logically by subject. Comments follow many of the Saunders Co., Philadelphia, Pa.
publications to explain their contributions, which 4. Lurie, M. B., P. Zappasodi, A. M. Dan-
are often not evident from their titles. In this appen- nenberg, Jr., and E. Cardona-Lynch.
dix,“Lurie’s book” refers to his 1964 text (publica- 1953. The effect of cortisone and ACTH
tion 68 in appendix C). Chapters not attributed to on the pathogenesis of tuberculosis. Ann.
Lurie’s book refer to the present volume. Publica- N.Y.Acad. Sci. 56:779–792.
tion numbers refer to this appendix (D) unless 5. Lurie, M. B., P. Zappasodi, A. M. Dan-
stated otherwise. nenberg, Jr., and E. Cardona-Lynch.
1953. Constitutional factors in resistance to
PART I. RESEARCH ARTICLES infection: the effect of cortisone on the
RHEUMATOID ARTHRITIS pathogenesis of tuberculosis, p. 84–99. In G.
1. Harris,T. N., S. Harris, A. M. Dannen- Shwartzman (ed.), The Effect of ACTH and
berg, Jr., and J. L. Hollander. 1950. Strep- Cortisone upon Infection and Resistance. Colum-
tococcal antihyaluronidase titers in the sera bia University Press, New York, N.Y.
of patients with rheumatoid arthritis and 6. Lurie, M. B., and P. Zappasodi. 1955. On
glomerular nephritis. Ann. Intern. Med. 32: the mode of action of cortisone on the
917–922. pathogenesis of tuberculosis and its implica-
I helped the Harrises with this study during my tions for the nature of genetic resistance to
1-year research residency at the Children’s Hos- the disease, p. 246–258. In Ciba Foundation
pital of Philadelphia (University of Pennsylvania). Symposium on Experimental Tuberculosis.
Churchill, London, U.K.
STUDIES ON THE PATHOGENESIS I participated in many of the studies described
OF TUBERCULOSIS WITH LURIE’S here.
INBRED RABBIT STRAINS 7. Lurie, M. B., P. Zappasodi, E. Cardona-
Comments on the following glucocorticosteroid Lynch, and A. M. Dannenberg, Jr. 1952.
studies (publications 2 through 6) are in Lurie’s The response to the intracutaneous inocu-
publication list (appendix C, publications 44
lation of BCG as an index of native resistance
through 48).These studies are reviewed in chap-
to tuberculosis. J. Immunol. 68:369–387.
ter 11 in Lurie’s book and in chapter 16 in the pre-
The rate of healing of dermal BCG lesions
sent volume. reflected how inbred rabbits controlled the
2. Lurie, M. B., P. Zappasodi, A. M. Dan- progress of infection with virulent tubercle bacilli;
nenberg, Jr., and I. B. Swartz. 1951. Con- i.e., it indicated the amount of their innate resis-
stitutional factors in resistance to infection: the tance to this disease.These studies apply to groups
effect of cortisone on the pathogenesis of of rabbits, but were not precise for individual
rabbits.
tuberculosis. Science 113:234–237. Hosts with poor innate resistance did not
3. Lurie, M. B., P. Zappasodi, A. M. Dan- develop the amount of immunity from the vac-
nenberg, Jr., and E. Cardona-Lynch. cine that hosts with good innate resistance did.Yet,
1952. Constitutional factors in resistance to hosts with poor resistance need such immuniza-
infection: the effect of cortisone on the tion the most. See chapter 23.
pathogenesis of tuberculosis and its implica- 8. Lurie, M. B., P. Zappasodi, A. M. Dan-
tions for nonspecific and allergic inflamma- nenberg, Jr., and G. H. Weiss. 1953. On
tions and infectious diseases in general, the mechanism of genetic resistance to
p. 247–253. In Advances in Medicine and tuberculosis and its mode of inheritance.
Surgery (Graduate School of Medicine of Am. J. Hum. Genet. 4:302–314.

399
400 䡵 APPENDIX D

Crossbreeding of resistant and susceptible rabbits hydrolyzed by pepsin. However, another protease
produced an F1 hybrid of intermediate resistance. was present in the original proteinase I prepara-
Backcrossing this F1 hybrid with susceptible rab- tion (see publication 43). This second protease
bits did not regain susceptibility in the offspring. polymerized amino acid esters that were sub-
However, backcrossing this F1 hybrid with resis- strates for chymotrypsin.
tant rabbits did regain high resistance in the off-
spring.Therefore, resistance was dominant, or sus- RESPIRATORY MELIOIDOSIS
ceptibility was lacking some of the resistant genes. These studies were performed at the Naval Bio-
See chapter 10 in Lurie’s book and chapter 14 in
the present volume.
logical Laboratories in Oakland, Calif. (under Dr.
9. Lurie, M. B., and A. M. Dannenberg, Jr. Clara Nigg) while I was a naval officer attached to
Naval Medical Research Unit #1 in Berkeley, Calif.
1965. Macrophage function in infectious
13. Dannenberg,A. M., Jr., and E. M. Scott.
disease with inbred rabbits. Bacteriol. Rev.
29:466–476. 1956. Determination of respiratory LD50
from a number of primary lesions as illus-
This report briefly reviews the native and acquired
characteristics of Lurie’s inbred rabbits. I wrote it trated by melioidosis. Proc. Soc. Exp. Biol.
at Johns Hopkins while Dr. Lurie was still alive, Med. 92:571–575.
and he checked its accuracy. See chapters 13 and With virulent strains of Malleomyces pseudomallei,
14 in the present volume. every established pulmonary lesion was always
10. Colberg, J. E., and A. M. Dannenberg, fatal in mice.Therefore, if mice inhaled sufficient
Jr. 1965.An evaluation of inbred rabbit pop- bacilli to produce 10 to 50 primary pulmonary
lesions, one could calculate the inhaled dose
ulations by skin homotransplantation. Nature required to produce one lesion (the LD50), using
207:777–778. Poisson’s distribution.This method could actually
Lurie’s C and AD rabbit strains were only partly determine an LD50 by exposing only one animal.
inbred: the C strain to a greater extent than the However, more than one animal was usually used,
AD strain.The C strain rejected a skin graft from to allow for variations between individual mice.
an outbred Flemish strain of rabbits in a normal Pulmonary lesion counts can greatly reduce the
fashion, despite its poor immune response to the number of animals needed to obtain an LD50,
tubercle bacillus. because such counts provide a mean value of
many lesions in each animal, whereas a single
(lethal) lesion might not represent the mean.
IDENTIFICATION AND 14. Dannenberg,A. M., Jr., and E. M. Scott.
CHARACTERIZATION 1958. Melioidosis: pathogenesis and immu-
OF LUNG PROTEINASES
nity in mice and hamsters. I. Studies with
I performed these studies as a postdoctoral fellow
virulent strains of Malleomyces pseudomallei.
in the laboratory of Professor Emil L. Smith at the
J. Exp. Med. 107:153–166.
University of Utah, Salt Lake City.
The characteristics of peracute, acute, and chronic
11. Dannenberg,A. M., Jr., and E. L. Smith. melioidosis in mice are described. In general, the
1955. Proteolytic enzymes of lung. J. Biol. type of disease produced depended on the dose
Chem. 215:545–554. of the bacilli inhaled.The acute form of melioi-
Macrophage cathepsin D (proteinase I) was partly dosis was a septicemic disease similar to the acute
purified from beef lung. (A typographical error forms of plague and anthrax.
appeared in this report. It should read:“Two vol- 15. Dannenberg,A. M., Jr., and E. M. Scott.
umes of 50% acetone in water [not 100% acetone] 1958. Melioidosis: pathogenesis and immu-
was used in the Waring blender.”) Publication
nity in mice and hamsters. II. Studies with
42 describes the complete purification of this
enzyme from rabbit and beef lungs. avirulent strains of Malleomyces pseudomallei.
12. Dannenberg,A. M., Jr., and E. L. Smith. Am. J. Pathol. 34:1099–1121.
1955.Action of proteinase I of bovine lung. These avirulent strains cannot multiply well in the
host. However, if very large numbers of avirulent
Hydrolysis of the oxidized B chain of insulin; strains were administered by any route, the disease
polymer formation from amino acid esters. produced was similar to that produced by fully
J. Biol. Chem. 215:55–66. virulent strains.With these avirulent strains, the
Macrophage proteinase I hydrolyzed the oxi- disease was exacerbated in mice by cortico-
dized B chain of insulin at sites similar to those steroids or by fasting.
APPENDIX D 䡵 401

16. Dannenberg,A. M., Jr., and E. M. Scott. HYDROLASES OF CULTURED


1960. Melioidosis: pathogenesis and immunity MACROPHAGES
in mice and hamsters. III.The effect of vac- 19. Dannenberg, A. M., Jr., P. C. Walter,
cination with avirulent strains of Pseudomonas and F. A. Kapral. 1963. A histochemical
pseudomallei on the resistance to the estab- study of phagocytic and enzymatic func-
lishment and the resistance to the progress of tions of rabbit mononuclear and polymor-
respiratory melioidosis caused by virulent phonuclear exudate cells and alveolar
strains: all-or-none aspects of this disease. macrophages. II.The effect of particle inges-
J. Immunol. 84:233–246. tion on enzyme activity; two phases of in
Vaccination could prevent the establishment of vitro activation. J. Immunol. 90:448–465.
melioidosis after the inhalation of virulent strains “Protoplasmic excitation” is the first phase of
of Pseudomonas pseudomallei but had little or no phagocyte activation. It causes an increase in the
effect on its progress. See chapter 12. phagocyte’s oxygen and glucose consumption,
lipid turnover, pseudopod formation, and, prob-
ably, motility.With excitation, cell enzymes are not
increased but are utilized to nearer capacity.After
ENZYME HISTOCHEMISTRY excitation, “protoplasmic adaptation” occurs:
OF MACROPHAGES, POLY- many of the initial changes remain, and many of
MORPHONUCLEAR LEUKOCYTES the enzymes utilized in excitation are increased
(PMN), AND PULMONARY in amount, so that the phagocyte can remain in
ALVEOLAR MACROPHAGES the activated state.
17. Dannenberg,A. M., Jr., M. S. Burstone, 20. Mizunoe, K., and A. M. Dannenberg, Jr.
P. C.Walter, and J.W. Kinsley. 1963.A his- 1965. Hydrolases of rabbit macrophages. III.
tochemical study of phagocytic and enzy- Effect of BCG vaccination, tissue culture, and
matic functions of rabbit mononuclear and ingested tubercle bacilli. Proc. Soc. Exp. Biol.
polymorphonuclear exudate cells and alve- Med. 120:284–290.
olar macrophages. I. Survey and quantitation Rabbit mineral oil-induced peritoneal macro-
of enzymes, and states of cellular activation. phages were assayed for proteinase, chymotrypsin-
J. Cell Biol. 17:465–486. like esterase, lipase, lysozyme, and acid phosphatase.
This report illustrates that pulmonary alveolar These macrophages came from BCG-vaccinated
macrophages contain higher levels of hydrolytic rabbits and controls, with or without added tuber-
and oxidative enzymes than do macrophages and cle bacilli. The macrophages were cultured for
PMN. Cytochrome oxidase, aminopeptidase, suc- 2 days.
cinic dehydrogenase, acid phosphatase, esterase, Differences in macrophage enzymes occurred
and alkaline phosphatase were evaluated histo- in culture among these various groups. How-
chemically with respect to stability, activators, ever, cell function in culture cannot be considered
inhibitors, and pH optimum. Only PMN con- equivalent to cell function in vivo. Tuberculous
tained alkaline phosphatase. lesions in vivo have a continuous blood supply,
18. Yarborough, D. J., O. T. Meyer, A. M. enabling the local accumulation of dendritic cells,
Dannenberg, Jr., and B. Pearson. 1967. macrophages and lymphocytes, and serological
components, none of which can be adequately
Histochemistry of macrophage hydrolases.
reproduced in vitro.
III. Studies on -galactosidase, -glu-
curonidase and aminopeptidase with indolyl PHAGOCYTOSIS BY PULMONARY
and naphthyl substrates. J. Reticuloendothel. ALVEOLAR MACROPHAGES OF
Soc. 4:390–408. LURIE’S INBRED RABBITS
These enzymes, in addition to those in publica- 21. Henderson, H. J., A. M. Dannenberg,
tion 17, confirmed that pulmonary alveolar Jr., and M. B. Lurie. 1963. Phagocytosis of
macrophages are highly activated cells. tubercle bacilli by rabbit pulmonary alveo-
The possible role of enzymes that remove
lar macrophages and its relation to native
only one sugar or one amino acid at a time is dis-
cussed. Such enzymes may downregulate cell- resistance to tuberculosis. J. Immunol. 91:553–
receptor functions and may also alter the surfaces 556.
of invading microorganisms so that they are less Alveolar macrophages from inbred resistant rab-
virulent. bits ingested twice as many tubercle bacilli in
402 䡵 APPENDIX D

vitro than did those from inbred susceptible rab- 25. Carson, M. E., and A. M. Dannenberg,
bits. Perhaps, the greater phagocytic ability of Jr. 1965. Hydrolytic enzymes of rabbit
alveolar macrophages from the resistant rabbits mononuclear exudate cells. II. Lysozyme:
enabled them to trap more bacilli from inhaled air
in the animal room, because they converted their
properties and quantitative assay in tubercu-
tuberculin skin test several months sooner than lous and control inbred rabbits. J. Immunol.
did the susceptible rabbits. See chapter 6 (p. 164– 94:99–104.
168) in Lurie’s book and chapter 12 in the pre- 26. Meyer, O. T., A. M. Dannenberg, Jr.,
sent volume. and K. Mizunoe. 1970. Hydrolytic
enzymes of rabbit mononuclear and poly-
INFLAMMATION CAUSED BY A morphonuclear exudate cells and pulmonary
LIPOPOLYSACCHARIDE FROM
alveolar macrophages. III. Deoxyribonucle-
TUBERCLE BACILLI
ase and ribonuclease: properties and quanti-
22. Dannenberg, A. M., Jr., K. Mizunoe,
tative assay in macrophages from tuberculous
M. Peace, and P. Zappasodi. 1965. Der-
and control inbred rabbits. J. Reticuloendothel.
mal response to the lipopolysaccharide
Soc. 7:15–31.
PmKo from tubercle bacilli as an index of
Some of these assays were made on the peri-
resistance to tuberculosis. Bull. Johns Hopkins
toneal exudates, described in publication 23, that
Hosp. 117:174–194. had been stored at –20°C for 5 to 7 years with lit-
PmKo is a lipopolysaccharide extracted and puri- tle or no apparent loss of activity.
fied from heat-killed virulent tubercle bacilli.
Used as a skin test in tuberculous humans, it was RADIATION AND MACROPHAGE
meant to predict whether the disease was pro- FUNCTION
gressing or regressing. However, in several strains These reports are summarized in chapter 18.
of Lurie’s inbred resistant and susceptible rabbits
27. Kambara, T., S. Chandrasekhar, A. M.
with active tuberculosis, we could not show any
prognostic differences between the PmKo and Dannenberg, Jr., and O.T. Meyer. 1970.
tuberculin skin tests. Radiation, infection and macrophage func-
tion. I. Effects of whole body radiation on
HYDROLYTIC ENZYMES OF RABBIT dermal tuberculous lesions in rabbits: devel-
MACROPHAGES (AND PMN) opment, histology and histochemistry. J. Retic-
These reports describe the properties of various uloendothel. Soc. 7:53–78.
hydrolytic enzymes in rabbit peritoneal exudate 28. Meyer, O.T., and A. M. Dannenberg, Jr.
cells (not containing tubercle bacilli). 1970. Radiation, infection and macrophage
The major activation of macrophages occurs function. II. Effect of whole body radiation
locally within tuberculous lesions where the bacilli on the number of pulmonary alveolar
and their products exist. Therefore, the macro- macrophages and their levels of hydrolytic
phages in peritoneal exudates from tuberculous enzymes. J. Reticuloendothel. Soc. 7:79–90.
and control rabbits (where no tubercle bacilli are 29. Dannenberg, A. M., Jr.,W. G. Roessler,
present) showed little or no difference in their O. T. Meyer, S. Chandrasekhar, and T.
enzyme content (publications 23 and 26), except Kambara. 1970. Radiation, infection and
for lysozyme (publications 20 and 35). macrophage function. III. Recovery from
23. Dannenberg, A. M., Jr., and W. E. Ben- the effects of radiation illustrated by dermal
nett. 1963. Hydrolases of mononuclear BCG lesions: resistance of pulmonary alve-
exudate cells and tuberculosis. I. Exudate olar macrophages to radiation illustrated by
characteristics, esterases, proteinases and tuberculosis produced by the airborne route.
lipase. Arch. Pathol. 76:581–591. J. Reticuloendothel. Soc. 7:91–108.
24. Dannenberg, A. M., Jr., and W. E. Ben- 30. Chandrasekhar, S., K. Shima,A. M. Dan-
nett. 1964. Hydrolytic enzymes of rabbit nenberg, Jr.,T. Kambara, J. I. Fabrikant,
mononuclear exudate cells. I. Quantitative and W. G. Roessler. 1971. Radiation, infec-
assay and properties of certain proteases, tion and macrophage function. IV.The effect
nonspecific esterases and lipases of mononu- of radiation on the proliferative abilities of
clear and polymorphonuclear cells and ery- mononuclear phagocytes in tuberculous
throcytes. J. Cell Biol. 21:1–13. lesions of rabbits. Infect. Immun. 3:254–259.
APPENDIX D 䡵 403

MACROPHAGE PROTEINASE Dermal lesions were produced by 14C-labeled


ACTIVATES COMPLEMENT BCG.The most mature epithelioid cells (which
COMPONENTS Lurie associated with bacillary destruction) stained
31. Snyderman, R., H. S. Shin, and A. M. histochemically the darkest for -galactosidase
Dannenberg, Jr. 1972. Macrophage pro- (see publication 32).This publication (number 33)
teinase and inflammation: the production of showed that these mature epithelioid cells con-
chemotactic activity from the fifth compo- tained 14C-labeled bacillary components but few
intact bacilli, confirming that these cells had
nent of complement by macrophage pro- destroyed the bacilli that they once contained.
teinase. J. Immunol. 109:896–898.
Both partly purified beef lung cathepsin D and
rabbit macrophage homogenates hydrolyzed the MACROPHAGE KINETICS: THEIR
C5 component of complement to produce the TURNOVER AND ACTIVATION IN
active chemotaxin C5a.Therefore, complement, TUBERCULOUS LESIONS
in addition to chemokines, apparently plays a
role in the accumulation of mononuclear cells in A continuous turnover of macrophages occurs in
tuberculous lesions. See chapter 5. the viable tuberculous granulation tissue that sur-
rounds the caseous centers of the lesions. This
turnover occurs even in arrested lesions, although
THE LOCAL NATURE OF at a reduced rate.
CELL-MEDIATED IMMUNITY The continuous entry of nonactivated mono-
32. Dannenberg, A. M., Jr., O. T. Meyer, cytes/macrophages into viable areas around the
J. R. Esterly, and T. Kambara. 1968.The caseous center provides an opportunity for
local nature of immunity in tuberculosis, renewed intracellular bacillary growth.The caseous
illustrated histochemically in dermal BCG center contains dormant tubercle bacilli. In arrested
lesions. J. Immunol. 100:931–941. lesions, such bacilli could escape from the edges of
This seemed to be the first histochemical demon- the caseum and grow in nonactivated macrophages
stration of the local nature of acquired cellular
resistance (macrophage activation). Macrophages before they are destroyed by activated macro-
were most activated (for -galactosidase, - phages. Therefore, even in arrested lesions, some
glucuronidase, succinic dehydrogenase, and bacilli are probably not dormant. True bacillary
cytochrome oxidase) within the tuberculous dormancy may only occur within solid caseous tis-
lesion at sites where the bacillus and its antigens sue itself.
are located. Elsewhere in the lesion, macrophages This series of reports is summarized in publi-
were less activated.
cation 119 and in chapter 10.
This report also describes other important
contributions. (i) Macrophages staining darkest 34. Shima, K., A. M. Dannenberg, Jr.,
for -galactosidase contained fewer tubercle M.Ando, S. Chandrasekhar, J.A. Seluz-
bacilli than those staining lighter, and their shape icki, and J. I. Fabrikant. 1972. Macro-
resembled that of Lurie’s mature epithelioid cells.
(ii) DNase and RNase (demonstrated by the
phage accumulation, division, maturation,
Daoust substrate film technique) were active in and digestive and microbicidal capacities in
intact and necrotic cells within the liquefying tuberculous lesions. I. Studies involving their
areas of the lesions.This technique also showed incorporation of tritiated thymidine and
that rabbit eosinophils had very high RNase their content of lysosomal enzymes and
activity. bacilli. Am. J. Pathol. 67:159–180.
Our review of local and systemic immunity in 35. Ando, M., A. M. Dannenberg, Jr., and
tuberculosis (publication 96) was published shortly
after this report. Chapter 5 presents the concepts K. Shima. 1972. Macrophage accumula-
therein. Macrophages are locally activated for tion, division, maturation and digestive and
many different functions (see chapter 6 and pub- microbicidal capacities in tuberculous lesions.
lication 54). II. Rate at which mononuclear cells enter
33. Ando, M., A. M. Dannenberg, Jr., M. and divide in primary BCG lesions and those
Sugimoto, and B. S. Tepper. 1977. His- of reinfection. J. Immunol. 109:8–19.
tochemical studies relating the activation of 36. Dannenberg, A. M., Jr., M. Ando, and
macrophages to the intracellular destruction K. Shima. 1972. Macrophage accumula-
of tubercle bacilli. Am. J. Pathol. 86:623–634. tion, division and immunity in primary BCG
404 䡵 APPENDIX D

lesions and in those of reinfection. J. Immunol. Kamenetz. 1975. Macrophage esterase:


109:1109–1121. identification, purification and properties of
37. Ando, M., and A. M. Dannenberg, Jr. a chymotrypsin-like esterase from lung that
1972. Macrophage accumulation, division, hydrolyzes and transfers nonpolar amino acid
maturation and digestive and microbicidal esters. Biochim. Biophys.Acta 403:161–179.
capacities in tuberculous lesions. IV. Macro-
phage turnover, lysosomal enzymes and VISUALIZATION OF MACROPHAGE
division in healing lesions. Lab. Investig. 27: PROTEINASE, RIBONUCLEASE,
466–472. AND PHOSPHOLIPASE A2 IN TISSUE
38. Ando, M. 1973. Macrophage activation in SECTIONS OF TUBERCULOUS LESIONS
tuberculin reactions of rabbits with primary BY PEROXIDASE–ANTIPEROXIDASE
BCG infection and reinfection. J. Reticu- TECHNIQUES
loendothel. Soc. 14:132–145. 44. Rojas-Espinosa, O.,A. M. Dannenberg,
From our laboratory. Jr., L. A. Sternberger, and T. Tsuda.
39. Ando, M., A. M. Dannenberg, Jr., E. 1974. Role of cathepsin D in the patho-
Courtade, and K. Shima. 1976.Turnover genesis of tuberculosis.A histochemical study
of tritiated-thymidine-labeled mononuclear employing unlabeled antibodies and the per-
cells in tuberculous lesions of rabbits.A com- oxidase-antiperoxidase complex. Am. J.
parison of primary dermal BCG lesions and Pathol. 74:1–17.
those of reinfection. Proc. Soc. Exp. Biol. Med. Cathepsin D was visualized in rabbit peritoneal
and pulmonary macrophages and in dermal BCG
151:491–494. lesions. It was present at the border of the lique-
40. Dannenberg, A. M., Jr., M. Sugimoto, fied caseous centers of these lesions. See publica-
L. P. Fay, and A. L. Massaquoi. 1976. In tion 54 for additional histochemical studies.
vivo labeling effectiveness of tritiated thymi- 45. Namba, M., M. Suga, F.Tanaka, A. M.
dine of high and low specific activities in Dannenberg, Jr., A. T. Hastie, and R.
rabbits. Radiat. Res. 67:98–103. C. Franson. 1983. Immunocytochemical
41. Tsuda, T., A. M. Dannenberg, Jr., M. demonstration of rabbit ribonuclease and
Ando, H.Abbey, and A. R. Corrin. 1976. phospholipase A 2 by the peroxidase-
Mononuclear cell turnover in chronic antiperoxidase technique in professional
inflammation. Studies on tritiated-thymi- phagocytes (pulmonary alveolar macro-
dine-labeled cells in blood, tuberculin traps phages and granulocytic and mononuclear
and dermal BCG lesions of rabbits. Am. J. peritoneal exudate cells) and in glycol
Pathol. 83:255–268. methacrylate sections of dermal tubercu-
lous (BCG) lesions. J. Reticuloendothel. Soc.
PURIFICATION OF MACROPHAGE 34:425–435.
PROTEASES FROM RABBIT LUNGS Ribonuclease and phospholipase A2 (and cathep-
42. Rojas-Espinosa, O.,A. M. Dannenberg, sin D, publication 44) are increased together as
Jr., P. A. Murphy, P. A. Straat, P. C. macrophages become activated.
Huang, and S. P. James. 1973. Purification Anti-RNase serum stained alveolar macro-
and properties of the cathepsin D-type pro- phages more intensely than PMN and macro-
teinase from beef and rabbit lung and its phages in oil-induced exudates, whereas
anti-RNase serum stained PMN and macro-
identification in macrophages. Infect. Immun.
phages more intensely than alveolar macrophages
8:1000–1008. (see publication 56). RNase evidently increases
Cathepsin D was purified from rabbit and beef with macrophage activation.
lungs. Four isoenzymes were obtained from each
source.An antibody to one rabbit isoenzyme was
produced in goats for the immunohistochemical PEPSTATIN: A HIGHLY SPECIFIC
studies described in publication 44. The anti- INHIBITOR OF MACROPHAGE
body inhibited the proteolytic activity of rabbit CATHEPSIN D
peritoneal and pulmonary macrophages. 46. McAdoo, M. H., A. M. Dannenberg,
43. Rojas-Espinosa, O., P. Arce-Paredez, Jr., C. J. Hayes, S. P. James, and J. H.
A. M. Dannenberg, Jr., and R. L. Sanner. 1973. Inhibition of the cathepsin
APPENDIX D 䡵 405

D-type proteinase of macrophages by pep- CAPILLARY DENSITY IN DEVELOPING


statin, a specific pepsin inhibitor, and other AND HEALING TUBERCULOUS
substances. Infect. Immun. 7:655–665. LESIONS
Pepstatin, discovered in H. Umezawa’s laboratory 49. Courtade, E.T.,T.Tsuda, C. R.Thomas,
( J.Antibiot. [Japan] 23:259–262, 1970), is a highly and A. M. Dannenberg, Jr. 1975. Capillary
specific inhibitor of macrophage cathepsin D. density in developing and healing tuberculous
Other inhibitors were identified, but they were not lesions produced by BCG in rabbits.A quan-
nearly as specific as pepstatin. The structure of titative study. Am. J. Pathol. 78:243–260.
pepstatin, how it works, and the other proteinases This is the only study of this type on tuberculous
that it inhibits (including pepsin) are reviewed. Its lesions.The entire microvasculature in the rabbit
in vivo applications to date have been disap- was made visible by injecting (under deep ter-
pointing, even for the treatment of peptic ulcers. minal anesthesia) the aorta with a warm gela-
Perhaps an analog of pepstatin would be more tinized colloidal carbon suspension. After the
effective. See chapter 4 for the possible use of gelatin had solidified in 10% formalin, thick, 250-
such inhibitors in preventing liquefaction and the m tissue sections of the BCG lesions were made.
formation of cavities in tuberculosis. The microvasculature density seemed to peak in
15-day BCG lesions and remained about 60%
above normal throughout the healing process.
HISTOCHEMICAL VISUALIZATION OF Chapter 8 provides more details on this experi-
HYDROLYTIC ENZYMES IN ment, including photographs.
TUBERCULOUS LESIONS BY
SUBSTRATE FILM TECHNIQUES THE COLLECTION AND
47. Tsuda, T., A. M. Dannenberg, Jr., M. MEASUREMENT OF EXTRACELLULAR
Ando, O. Rojas-Espinosa, and K. HYDROLYTIC ENZYMES IN
Shima. 1974. Enzymes in tuberculous DEVELOPING AND HEALING BCG
lesions hydrolyzing protein, hyaluronic acid LESIONS BY USING SKIN CHAMBERS
and chondroitin sulfate: a study of isolated 50. Sugimoto, M., A. M. Dannenberg, Jr.,
macrophages in developing and healing rab- L. M. Wahl, W. H. Ettinger, Jr., A. T.
bit BCG lesions with substrate film tech- Hastie, D. C. Daniels, C. R.Thomas, and
niques; the shift of enzyme pH optima L. Demoulin-Brahy. 1978. Extracellular
towards neutrality in “intact” cells and tissues. hydrolytic enzymes of rabbit dermal tuber-
J. Reticuloendothel. Soc. 16:220–231. culous lesions and tuberculin reactions col-
In cryostat-prepared tissue sections of rabbit lected in skin chambers. Am. J. Pathol.
tuberculous lesions (produced by BCG), these 90:583–608.
enzymes were most active after delayed-type Macrophages are known to secrete collagenase,
hypersensitivity (DTH) and cell-mediated immu- secrete and store lysozyme, and release DNase,
nity (CMI) had developed.They remained active RNase, and lactic dehydrogenase after they die.
long after the cells containing them had died We measured these enzymes in skin chambers
(similar to the RNase and DNase described in placed over developing and healing BCG lesions
publication 32). (with the epidermis removed).The concentration
The pH optimum of these “acid-acting” hydro- of all enzymes in the chamber fluids peaked
lases in these tissue sections was closer to neutral- between 11 and 18 days—a time when CMI and
ity than their pH optimum in tissue homogenates. DTH produced the highest percentage of acti-
48. Smokovitis, A., M. Sugimoto, A. M. vated macrophages in the chamber beds.
Dannenberg, Jr., and T. Astrup. 1976.
A histochemical study of the fibrinolytic EFFECT OF GLUCOCORTICOSTEROIDS
IN DEVELOPING AND HEALING BCG
activity in dermal tuberculous lesions pro-
LESIONS
duced by BCG in rabbits. Exp. Mol. Pathol.
51. McCue, R. E.,A. M. Dannenberg, Jr., S.
25:236–241.
Higuchi, and M. Sugimoto. 1978. The
The microvasculature in tuberculous granulation
effect of cortisone on the accumulation,
tissue of BCG lesions showed high fibrinolytic
activity, probably due to plasminogen activator. activation, and necrosis of macrophages in
The fibrinolytic activity was highest between 11 tuberculous lesions. Inflammation 3:159–176.
and 18 days when the microvasculature was pro- In rabbit dermal BCG lesions, cortisone reduced
liferating. See publication 49. the number of infiltrating mononuclear cells, the
406 䡵 APPENDIX D

amount of caseous necrosis and ulceration, and the they were next to each other.Why and how this
percentage of the macrophage population that occurs are matters of conjecture.This report and
became activated (i.e., became positive for - chapter 6 review the possibilities.
galactosidase).These effects readily explain why
glucocorticosteroids reduce host resistance to PERSISTENCE OF MYCOBACTERIAL
tuberculosis.The effects of glucocorticosteroids on ANTIGENS IN TISSUES
the inflammatory response are reviewed in pub- 55. Higuchi, S., M. Suga, A. M. Dannen-
lication 85. (See chapter 16.) berg, Jr., L. F. Affronti, I. Azuma,T. M.
Daniel, and J. P. Petrali. 1981. Persistence
EFFECTS OF BCG VACCINATION ON
THE PATHOGENESIS OF SYPHILIS of protein, carbohydrate and wax compo-
52. Hardy, P. H., Jr., D. J. Graham, E. E. nents of tubercle bacilli in dermal BCG
Nell, and A. M. Dannenberg, Jr. 1979. lesions. Am. Rev. Respir. Dis. 123:397–401.
Macrophages in immunity to syphilis: sup- Antibodies were used to visualize these bacillary
components in tissue sections with the peroxi-
pressive effect of concurrent infection with
dase–antiperoxidase technique. These compo-
Mycobacterium bovis BCG on the develop- nents disappeared from the BCG lesions at dif-
ment of syphilitic lesions and growth of ferent rates: protein B was undetectable at 21
Treponema pallidum in tuberculin-positive days, polysaccharide I was undetectable at 35
rabbits. Infect. Immun. 26:751–763. days, but wax D could still be found at 56 days if
This report clearly demonstrates the nonspecific the lesions were not completely healed.Antisera
nature of local acquired resistance (see chapter 5). against intact tubercle bacilli produced results
BCG was injected into intradermal sites where similar to antibodies to wax D.
Treponema pallidum had been injected a few min-
utes earlier. In rabbits sensitized by BCG 3 weeks PURIFICATION AND PROPERTIES OF
earlier, these mixed lesions showed more activated RIBONUCLEASE FROM LUNG
macrophages and fewer treponemes. 56. Hastie, A.T. 1981. Monospecific antibod-
ies to rabbit lung ribonucleases. J. Biol. Chem.
EFFECTS OF THE IRRITANT SULFUR 256:12553–12560.
DIOXIDE ON THE AIRWAYS OF (From our laboratory.) RNases were purified
CHICKENS from rabbit lungs by chromatographic techniques
53. Okuyama, H., Y. Majima, A. M. Dan- and antibodies to them made in goats. These
nenberg, Jr., M. Suga, B. G. Bang, and antibodies were then used to visualize the RNases
F. B. Bang. 1979. Quantitative histological in macrophage suspensions and in tuberculous
changes produced in the tracheal mucosa of lesions (see publication 45). The anti-RNase
young chickens by the inhalation of sulfur and anti-RNase sera were obtained by immu-
dioxide in low concentrations. J. Environ. noelectrophoresis and immunoabsorbent chro-
matography.These sera inhibited their respective
Sci. Health C13(4):267–300. RNase activities.
Following the inhalation of SO2, the mucosa of the
trachea was thickened and contained more infil- HISTOLOGY AND HISTOCHEMISTRY
trating leukocytes, more acid phosphatase-con- IN PLASTIC-EMBEDDED TISSUE
taining mononuclear cells, and more cells secret- SECTIONS
ing sialidase-sensitive sialomucins (when compared 57. Higuchi, S., M. Suga, A. M. Dannen-
with controls).The change in the mucus compo-
sition was apparently a protective mechanism. berg, Jr., and B. H. Schofield. 1979. His-
tochemical demonstration of enzyme activ-
HETEROGENEITY OF MACROPHAGES ities in plastic- and paraffin-embedded tissue
IN TUBERCULOUS LESIONS sections. Stain Technol. 54:5–12.
54. Suga, M.,A. M. Dannenberg, Jr., and S. 58. Namba, M.,A. M. Dannenberg, Jr., and
Higuchi. 1980. Macrophage functional het- F. Tanaka. 1983. Improvement of the his-
erogeneity in vivo: macrolocal and microlocal tochemical demonstration of acid phos-
macrophage activation, identified by double- phatase, -galactosidase and nonspecific
staining tissue sections of BCG granulomas esterase in glycol methacrylate tissue sec-
for pairs of enzymes. Am. J. Pathol. 99:305– tions by cold temperature embedding. Stain
324. Technol. 58:207–213.
Many macrophages within tuberculous lesions Polymerization of glycol methacrylate generates
were activated for different functions, even though heat equivalent to boiling.When polymerization
APPENDIX D 䡵 407

was performed in cracked ice, the structure of cells (serum) protein content. Am. J. Pathol.
in the 1- to 2-m tissue sections was markedly 121:15–27.
improved. See publication 60. These dermal lesions contained mostly mononu-
59. Vogt, R. F., Jr., N.A. Hynes,A. M. Dan- clear cells, because as soon as the epithelium died,
nenberg, Jr., S. Castracane, and L.Weiss. the PMN migrated in large numbers through
1983. Improved techniques using Giemsa- the tissues to form the crust, i.e., the scab.Within
stained glycol methacrylate tissue sections to the deep parts of the lesion, basophils and PMN
quantitate basophils and other leukocytes in were about equal in number. Perhaps PMN are
attracted into the crust by the bacteria that
inflammatory skin lesions. Stain Technol.
invaded the area after surface epithelium died.
58:193–205. The full-thickness rabbit skin explants survived
The granules of basophils and mast cells are beau- rather well in culture for at least 3 days. In fact,
tifully preserved in these thin plastic-embedded the epithelium migrating under the crust in 3-,
tissue sections (see publication 60)—so much so 6- and 10-day lesions continued to do so in cul-
that the amount of degranulation can be quan- ture. Publication 121 reviews this organ culture
titated (see publication 67). series.
62. Harada, S., A. M. Dannenberg, Jr., A.
PATHOGENESIS AND SERUM Kajiki, K. Higuchi, F. Tanaka, and P. J.
TURNOVER OF ACUTE DERMAL Pula. 1985. Inflammatory mediators and
INFLAMMATORY LESIONS
PRODUCED IN RABBITS BY
modulators released in organ culture from
DILUTE SULFUR MUSTARD rabbit skin lesions produced in vivo by sul-
60. Vogt, R. F., Jr., A. M. Dannenberg, Jr., fur mustard. II. Evans blue dye experiments
B. H. Schofield, N. A. Hynes, and B. that determined the rates of entry and
Papirmeister. 1984. Pathogenesis of skin turnover of serum protein in developing
lesions caused by sulfur mustard. Fund.Appl. and healing lesions. Am. J. Pathol. 121:28–38.
Toxicol. 4:S71–S83. One-day sulfur mustard lesions are quite ede-
matous due to extravasated serum. The serum
In rabbits, basophils constituted 32% of the gran-
that was unbound, i.e., extractable into the cul-
ulocyte population that infiltrated (during the
ture fluids, had a turnover rate of once every 8 h.
first 2 h) skin lesions produced by dilute sulfur
In other words, the extravasated serum in ede-
mustard. In guinea pigs, basophils were 21% of this
matous lesions was not static. In 3- and 6-day
population. In guinea pigs, eosinophils were as
sulfur mustard lesions that were not grossly ede-
common as basophils at that time, but eosinophils
matous, the unbound serum had a turnover rate
cannot always be distinguished from heterophils
of once every 35 h. In normal skin, the unbound
(PMN) in rabbits, because both are eosinophilic.
serum had a turnover rate of once every 80 h.
These results suggest that basophils and
eosinophils play an important role in the early
inflammation that follows mild or slowly devel- MEDIATORS AND MODULATORS OF
oping epidermal injury. Basophils would release ACUTE DERMAL INFLAMMATORY
histamine, and eosinophils would destroy the LESIONS PRODUCED IN RABBITS BY
released histamine. Light abrasion, UV light, oxa- DILUTE SULFUR MUSTARD
zolone, and sometimes mild heat produced a sim- 63. Harada, S.,A. M. Dannenberg, Jr., R. F.
ilar early basophil infiltration.
The role of basophils in the initiation of inflam- Vogt, Jr., J. E. Myrick, F. Tanaka, L. C.
mation needs further study. The granules of Redding, R. M. Merkhofer, P. J. Pula,
basophils are not well preserved in paraffin- and A. L. Scott. 1987. Inflammatory medi-
embedded tissue sections but are very well pre- ators and modulators released in organ
served and easily recognized in cold-embedded culture from rabbit skin lesions produced
glycol methacrylate sections stained with Giemsa. in vivo by sulfur mustard. III. Electro-
61. Dannenberg, A.M., Jr., P. J. Pula, L. phoretic protein fractions, trypsin-inhibitory
Liu, S. Harada, F.Tanaka, R. F.Vogt, Jr., capacity, 1-proteinase inhibitor, and 1- and
A. Kajiki, and K. Higuchi. 1985. Inflam- 2-macroglobulin proteinase inhibitors of
matory mediators and modulators released in culture fluids and serum. Am. J. Pathol.
organ culture from rabbit skin lesions pro- 126:148–163.
duced in vivo by sulfur mustard. I. Quanti- In acute inflammatory reactions, the proteinase
tative histopathology; PMN, basophil and inhibitors in serum are probably major factors in
mononuclear cell survival; and unbound reducing tissue damage from the proteinases of
408 䡵 APPENDIX D

extravasated serum, infiltrating leukocytes, and Boulay, E.W. Spannhake, G. S. Lazarus,


activated fibroblasts. P. J. Pula, and A. M. Dannenberg, Jr.
64. Kajiki, A., K. Higuchi, M. Nakamura, 1991. Mediators, initiating the inflamma-
L. H. Liu, P. J. Pula, and A. M. Dan- tory response, released in organ culture by
nenberg, Jr. 1988. Sources of extracellular full-thickness human skin explants exposed
lysosomal enzymes released in organ cul- to the irritant, sulfur mustard. J. Investig. Der-
ture by developing and healing inflammatory matol. 96:888–897.
lesions. J. Leukoc. Biol. 43:104–116. Angiotensin-converting enzyme, trypsin-like and
We evaluated acid phosphatase, -glucuronidase, chymotrypsin-like proteases, acid phosphatase,
-galactosidase, lysozyme, and lactic dehydroge- -glucuronidase, -galactosidase, lysozyme,
nase in organ culture fluids from rabbit dermal deoxyribonuclease, ribonuclease, interleukin 1,
sulfur mustard lesions.The enzyme levels increased and lactic dehydrogenase were released in simi-
as the lesions healed. Many fibroblasts in the heal- lar amounts (into the culture fluids) from sulfur
ing lesions were shown histochemically to be mustard-exposed full-thickness skin explants and
activated for acid phosphatase and -galactosidase. from controls. However, histamine, prostaglandin
These cells and the large number of dead PMN E2, and plasminogen activators (which are medi-
in the crust seemed to be the source of the ators of the early inflammatory response) were
increased enzyme levels in the culture fluids of increased in the culture fluids from the toxicant-
healing lesions. exposed explants.
65. Higuchi, K.,A. Kajiki, M. Nakamura, S. The amount of mast-cell degranulation was
Harada, P. J. Pula,A. L. Scott, and A. M. correlated with the amount of histamine released.
Dannenberg, Jr. 1988. Proteases released in 68. Tanaka, F., A. M. Dannenberg, Jr., K.
organ culture by acute dermal inflamma- Higuchi, M. Nakamura, P. J. Pula,T. E.
tory lesions produced in vivo in rabbit skin Hugli, R. G. DiScipio, and D. L.
by sulfur mustard: hydrolysis of synthetic Kreutzer. 1997. Chemotactic factors are
peptide substrates for trypsin-like and chy- continuously released by cultured intact
motrypsin-like enzymes. Inflammation 12: developing and healing skin lesions pro-
311–334. duced in rabbits by sulfur mustard. Inflam-
Again, higher levels of these enzymes were found mation 21:251–267.
in culture fluids from healing lesions. The culture fluids were chemotactic for both
66. Woessner, J. F., Jr., A. M. Dannenberg, PMN and mononuclear cells and contained
Jr., P. J. Pula, M. G. Selzer, C. L. Rup- leukotriene B4, interleukin 8, and proteases pro-
pert, K. Higuchi, A. Kajiki, M. Naka- ducing the complement fragment C5a. Fluids
from cultured (as well as frozen-and-thawed)
mura, N. M. Dahms, J. S. Kerr, and macrophages, granulocytes (PMN), and fibroblasts
G.W. Hart. 1990. Extracellular collagenase, were also chemotactic for both macrophages and
proteoglycanase and products of their activ- PMN.The chemokines MCP-1 and GRO were
ity, released in organ culture by intact dermal probably also present; see publication 75.
inflammatory lesions produced by sulfur Note: In vivo, dying cells release chemotaxins,
mustard. J. Investig. Dermatol. 95:717–726. whereas living cells and dead cells do not (see
Majno, G., and I. Joris. 1996. Cells,Tissues, and
Collagenase and proteoglycanase and products
Disease: Principles of General Pathology, p. 390.
of their hydrolytic activities (hydroxyproline-
Blackwell Science, Cambridge, Mass.). Perhaps
containing peptides and glycosaminoglycans)
frozen and thawed cells are similar to dying cells.
were measured in culture fluids as these lesions
developed and healed. The activity of these
enzymes was highest during healing when
remodeling of the connective tissue occurred. IN VITRO TOXICITY TO SKIN,
At that time, there was a decrease in the extrava- ASSESSED IN ORGAN CULTURE BY
sation of serum with a concomitant decrease in PARANUCLEAR VACUOLIZATION IN
serum proteinase inhibitors (such as 1-proteinase 1- TO 2-␮m TISSUE SECTIONS AND BY
inhibitor and the -macroglobulins; see publica- [14C]LEUCINE INCORPORATION
tion 63). Fibroblasts seemed to be a major source BY LIVE CELLS IN THE EXPLANT
of both collagenase and proteoglycanase. 69. Moore, K. G., B. H. Schofield,
67. Rikimaru, T., M. Nakamura, T. Yano, K. Higuchi, A. Kajiki, K.-W. Au, P. J.
G. Beck, G. S. Habicht, L. L. Rennie, Pula, D. P. Bassett, and A. M. Dannen-
M. Widra, C. A. Hirshman, M. G. berg, Jr. 1986.Two sensitive in vitro mon-
APPENDIX D 䡵 409

itors of chemical toxicity to human and After 7 days of storage at 4°C, these explants
animal skin (in short-term organ culture): were still viable and could be used in this test.
I. Paranuclear vacuolization in glycol meth- Electron microscope studies suggest that the
paranuclear vacuoles were invaginations of
acrylate tissue sections. II. Interference with the cell’s plasma membrane.
14
C-leucine incorporation. J. Toxicol. Cuta-
neous Ocular Toxicol. 5:285–302.
Two sensitive in vitro methods were developed to
determine the toxicity of chemicals applied to STUDIES ON CONTACT
full-thickness human, rabbit, and guinea pig skin SENSITIVITY OF THE SKIN
explants. In both methods, the toxicants were 72. Moore, K. G., and A. M. Dannenberg,
topically applied to the outer surface of each Jr. 1992. Antigen-specific IgG1-mediated
explant. Then, the explant was organ cultured epidermal cell injury: a component of con-
for 24 h in shallow petri dishes. tact hypersensitivity reactions in guinea pigs,
For the first method, cold-embedded plastic tis-
measurable in vitro in full-thickness skin
sue sections were prepared (see publication 58),
and the paranuclear vacuoles in the basal epider- explants. J. Investig. Dermatol. 98:929–935.
mal cells were counted microscopically.The num- Full-thickness skin explants were used with our
ber of these vacuoles increased with the toxicant paranuclear vacuolization test (see publication
concentration. Electron microscopy suggested 69) to measure contact sensitivity reactions to
that the vacuoles were a bleb in the cell’s nuclear dinitrochlorobenzene and oxazolone. Studies on
membrane. mast cell degranulation, microblistering, and
For the second method, the explants were cul- [14C]leucine incorporation were also made.The
tured in the presence of [14C]leucine.The incor- intradermal injection of IgG1 from sensitized
poration of the [14C]leucine into the proteins of guinea pigs could passively sensitize normal skin
the explants decreased as the toxicant concentra- explants to these contact sensitizers.
tion increased. These studies support the “threshold hypoth-
Both methods indicate injury to the cells in the esis” that chemical sensitizers must be applied in
explant. slightly toxic or irritating concentrations to elicit
Organ culture itself enables studies on the ini- an antigen-specific reaction in the skin of a sen-
tiation of the inflammatory response by the res- sitized host. Nonspecific irritants can lower the
ident cells in the explant, which include epider- threshold of antigen-specific irritants. Publica-
mal cells, mast cells, vascular endothelial cells, tion 121 reviews these studies.
histiocytes, and fibroblasts. Intradermal DTH reactions in cattle infected
70. Dannenberg,A. M., Jr., K. G. Moore, B. with virulent Mycobacterium bovis showed a sim-
H. Schofield, K. Higuchi, A. Kajiki, K.- ilar threshold response to antigens, such as ESAT-
W.Au, P. J. Pula, and D. P. Bassett. 1987. 6. In this case, the lipopeptide Pam3CSK4 was used
as the nonspecific irritant (Whelan et al. 2003.
Two new in vitro methods for evaluating Infect. Immun. 71:6420–6425).
toxicity to skin (employing short-term organ 73. Moore, K. G., and A. M. Dannenberg,
culture): I. Paranuclear vacuolization, seen Jr. 1993. Immediate and delayed (late-phase)
in glycol methacrylate tissue sections; II. dermal contact sensitivity reactions in guinea
Inhibition of 14C-leucine incorporation, p. pigs: passive transfer by IgG1 antibodies, ini-
115–127. In A. M. Goldberg (ed.), Alternative tiation by mast cell degranulation, and sup-
Methods in Toxicology, vol. 4 (Proceedings of pression by soybean proteinase inhibitor. Int.
the 1986 Symposium of the Center for Arch.Allergy Immunol. 101:72–81.
Alternatives to Animal Testing). Mary Ann
This report describes the pathogenesis of imme-
Liebert, Inc., New York, N.Y.
diate and delayed (late-phase) dermal contact
This report is similar to publication 69. sensitivity reactions. The immediate phase seems
71. Nakamura, M., T. Rikimaru, T. Yano, to be due to mast cells releasing histamine and
K. G. Moore, P. J. Pula, B. H. Schofield, other quick-acting mediators. The late phase
and A. M. Dannenberg, Jr. 1990. Full- seems to be due to mast cells releasing cyto-
thickness human skin explants for testing kines, including chemokines. The late phase has
some components of a cutaneous basophil hyper-
the toxicity of topically applied chemicals. sensitivity reaction. Soybean proteinase inhibitor
J. Investig. Dermatol. 95:325–332. (which inhibits mast cell degranulation) sup-
Details of the paranuclear vacuolization test on pressed both the immediate and delayed skin
full-thickness human skin explants are described. reactivities mediated by antigen-specific IgG1.
410 䡵 APPENDIX D

HISTOCHEMISTRY OF HYDROGEN Cytokine production in dermal BCG lesions is


PEROXIDE biphasic: an early adjuvant-type response within
74. Dannenberg,A. M., Jr., B. H. Schofield, 2 days followed by an immune-specific response
J. B. Rao,T.T. Dinh, K. Lee, M. Boulay, at 9 days. See chapter 19.
Y. Abe, J.Tsuruta, and M. J. Steinbeck.
1994. Histochemical demonstration of ROLE OF ANTIBODIES,
hydrogen peroxide production by leuko- LYMPHOCYTES, CYTOKINES,
cytes in fixed-frozen tissue sections of inflam- AND VASCULAR ADHESION
MOLECULES IN TUBERCULOUS
matory lesions. J. Leukoc. Biol. 56:436–443. LESIONS OF REINFECTION
In lightly fixed frozen tissue sections of dermal 78. Shigenaga, T., A. M. Dannenberg, Jr.,
inflammatory reactions, PMN and eosinophils
still produced H2O2, which did not injure adja-
D. B. Lowrie, W. Said, M. J. Urist,
cent tissues.The H2O2 was demonstrated histo- H. Abbey, B. H. Schofield, P. Mounts,
chemically by the diaminobenzidine reaction. and K. Sugisaki. 2001. Immune responses
in tuberculosis: antibodies and CD4-CD8
lymphocytes with vascular adhesion mole-
CYTOKINES AND VASCULAR cules and cytokines (chemokines) cause a
ADHESION MOLECULES IN rapid antigen-specific cell infiltration at sites
ACUTE AND CHRONIC of Bacillus Calmette-Guérin reinfection.
INFLAMMATORY LESIONS
Immunology 102:466–479.
Cytokines (including chemokines) are major
This report clearly documents that antibodies play
mediators of the inflammatory response.Vascular an important role in hosts already possessing
adhesion molecules enable leukocytes to leave the acquired resistance to the tubercle bacillus: antigen-
bloodstream and enter inflammatory lesions. antibody reactions produce chemotactic factors
75. Tsuruta, J., K. Sugisaki, A. M. Dannen- that rapidly bring the expanded antigen-specific
berg, Jr., T. Yoshimura, Y. Abe, and lymphocyte population to sites of bacillary lodge-
ment.Therefore, antibodies increase the effective-
P. Mounts. 1996. The cytokines NAP-1 ness of CMI and DTH by hastening the local
(IL-8), MCP-1, IL-1 beta, and GRO in rab- accumulation of cells that produce the antigen-spe-
bit inflammatory skin lesions produced by cific Th1 response. The rapidity of the cellular
the chemical irritant sulfur mustard. Inflam- response to the bacilli of reinfection is a major fac-
mation 20:293–318. tor in the ability of an immunized host to prevent
In tissue sections of these lesions, the mRNAs of clinically apparent disease. See chapter 20.
these cytokines were hybridized with 35S-radio- After sufficient cells had accumulated at sites of
labeled antisense riboprobes and autoradiographed. bacillary deposition, the chemokines that attracted
This technique demonstrated histochemically the them were downregulated. In other words, the
“factories” for cytokine production. In these tissue number of cells in an inflammatory site is evi-
sections, the distribution of cells producing dently under tight control.
chemokines was correlated with the distribution
of cells that these chemokines are known to attract. PATHOGENESIS OF PULMONARY
76. Abe, Y., K. Sugisaki, and A. M. Dan- TUBERCULOSIS, PRODUCED BY
nenberg, Jr. 1996. Rabbit vascular endothe- INHALED BACILLI
lial adhesion molecules: ELAM-1 is most These two reports seem to be the first in over 30
elevated in acute inflammation, whereas years to produce chronic progressive cavitary
VCAM-1 and ICAM-1 predominate in tuberculosis in rabbits by the aerosol method.
chronic inflammation. J. Leukoc. Biol. 60:692– Chapter 4 presents the details.
703. 79. Converse, P. J., A. M. Dannenberg, Jr.,
Summarized in chapter 21. J. E. Estep, K. Sugisaki, Y. Abe, B. H.
77. Sugisaki, K., A. M. Dannenberg, Jr., Schofield, and M. L. M. Pitt. 1996. Cav-
Y. Abe, J. Tsuruta, W.-J. Su, W. Said, itary tuberculosis produced in rabbits by
L. Feng, T. Yoshimura, P. J. Converse, aerosolized virulent tubercle bacilli. Infect.
and P. Mounts. 1998. Nonspecific and Immun. 64:4776–4787.
immune-specific up-regulation of cytokines 80. Converse, P. J., A. M. Dannenberg, Jr.,
in rabbit dermal tuberculous (BCG) lesions. T. Shigenaga, D. N. McMurray, S. W.
J. Leukoc. Biol. 63:440–450. Phalen, J. L. Stanford, G. A. W. Rook,
APPENDIX D 䡵 411

T. Koru-Sengul, H. Abbey, J. E. Estep, 82. Bishai, W. R., A. M. Dannenberg, Jr.,


and M. L. M. Pitt. 1998. Pulmonary N. Parrish, R. Ruiz, P. Chen, B. C.
bovine-type tuberculosis in rabbits: bacillary Zook,W. Johnson, J.W. Boles, and M. L.
virulence, inhaled dose effects, tuberculin M. Pitt. 1999.Virulence of Mycobacterium
sensitivity, and Mycobacterium vaccae immuno- tuberculosis CDC1551 and H37Rv in rabbits
therapy. Clin. Diagn. Lab. Immunol. 5:871–881. evaluated by Lurie’s pulmonary tubercle
This report analyzes more deeply the data pre- count method. Infect. Immun. 67:4931–4934.
sented in publication 79.We could not detect any See chapter 11 and publication 128.
effect of Mycobacterium vaccae immunotherapy,
because the disease varied too much between
83. Dannenberg, A. M., Jr.,W. R. Bishai, N.
individual rabbits. Parrish, R. Ruiz,W. Johnson, B. C. Zook,
J.W. Boles, and M. L. M. Pitt. 2000. Effi-
BACILLARY VIRULENCE AND VACCINE cacies of BCG and vole bacillus (Mycobac-
EFFICACY EVALUATED IN RABBITS BY terium microti) vaccines in preventing clini-
LURIE’S TUBERCLE-COUNT METHOD cally apparent pulmonary tuberculosis in
81. Dannenberg, A. M., Jr. 1998. Lurie’s rabbits: a preliminary report. Vaccine 19:796–
tubercle-count method to test TB vaccine 800.
efficiency in rabbits. Front. Biosci. 3:c27–33. Reviewed in chapter 23.
http://www.bioscience.org/1998/v3/c/
dannenbe/list.htm. PART II. REVIEW ARTICLES
This article presents the details on the tubercle- ENVIRONMENTAL EFFECTS
count method and its use in assessing bacillary ON THE LUNG
virulence, host genetic resistance, and vaccine 84. Dannenberg,A. M., Jr. 1977. Influence of
efficacy. Since its publication, we have gained environmental factors on the respiratory
further insight into this method’s applicability to
other laboratory species. See chapter 11.
tract. Summary and perspectives. J. Reticu-
Major insights into primary tuberculous lesions loendothel. Soc. 22:273–290.
in human beings came from Lindgren’s necropsy This review briefly describes the effects of inhaled
studies (see chapter 3). In Finland at a time when environmental and industrial substances on the
infection with Mycobacterium tuberculosis was com- airways and peripheral lung, including clearance
mon, he found with a special roentgenographic mechanisms, carcinogenesis, asbestosis, silicosis,
technique that every human who became tuber- and allergic pulmonary reactions.
culin positive from a natural infection with M.
tuberculosis had at least a minute grossly visible cal- EFFECTS OF
cified primary pulmonary tuberculous lesion at GLUCOCORTICOSTEROIDS ON
death, even though death usually occurred years INFLAMMATION
later from other causes. BCG immunization did
not reduce the number of such lesions found,
85. Dannenberg, A. M., Jr. 1979. The anti-
but did reduce lesion size and lymph node inflammatory effects of glucocorticosteroids:
involvement. a brief review of the literature. Inflammation
Humans infected with virulent tubercle bacilli 3:329–343.
develop high tuberculin sensitivity. Therefore, This article describes the effects of glucocorti-
each primary tubercle develops a caseous center costeroids on (i) infections, (ii) lymphocytes,
that absorbs calcium and becomes grossly visible macrophages, and other leukocytes, (iii) cell recep-
before its progress is arrested. tors and transduction pathways, (iv) host metab-
In mice and guinea pigs, each microscopic pri- olism, (v) cell membranes, (vi) connective tissue,
mary pulmonary lesion usually progresses until the and (vii) inflammation.To obtain a more complete
animal dies. In these animals, because of their high orientation in this field, consult the two classic
susceptibility, immunization with BCG would articles by Henry N. Claman in Hospital Practice
probably not reduce the number of grossly visible (18:123–134 and 143–151, 1983).
primary lesions, but would reduce the progression
of the lesions and increase the survival time of the
host. Because of these considerations, the tubercle- MACROPHAGES AND THEIR
count method is best used in rabbits, a species that FUNCTIONS
is more resistant than mice and guinea pigs to vir- 86. Dannenberg, A. M., Jr., M. Ando,
ulent human-type tubercle bacilli. See chapters 11 O. Rojas-Espinosa, K. Shima, and
and 15. T. Tsuda. 1974. Macrophage activation in
412 䡵 APPENDIX D

tuberculous lesions, p. 223–235. In W. H. tell, Jr. (ed.), Clinical Medicine, vol. 5 (revised
Wagner and H. Hahn (ed.), Activation of ed.). Harper & Row, Publishers, Philadelphia.
Macrophages. Excerpta Medica International This article reviews many aspects of macrophage
Congress Series 325. Excerpta Medica, function, including (i) chemotaxis, (ii) phagocy-
Amsterdam,The Netherlands. tosis, (iii) their origin, life span, differentiation, and
87. Dannenberg, A. M., Jr., M. Ando, turnover, (iv) their heterogeneity, (v) their secre-
K. Shima, and T. Tsuda. 1975. tory and microbicidal functions, and (vi) their
roles in bacterial and viral infections, neoplasia,
Macrophage turnover and activation in and endotoxin inactivation. This review is an
tuberculous granulomata, p. 959–980. In update of publication 90.
R. van Furth (ed.), Mononuclear Phagocytes in 95. Dannenberg, A. M., Jr. 1984. Macro-
Immunity, Infection and Pathology. Blackwell phages and monocytes, p. 153–173. In J. L.
Scientific Publications, Oxford, U.K. Spivak (ed.), Fundamentals of Clinical Hema-
88. Dannenberg, A. M., Jr. 1975. Macro- tology, 2nd ed. Harper & Row, Publishers,
phages in inflammation and infection. Inc., New York, N.Y.
N. Engl. J. Med. 293:489–493.
This review is a republication of publication 94.
89. Dannenberg,A. M., Jr., and S. Higuchi.
1979. Chronic inflammation involving cel-
lular hypersensitivity. Chest 75(Suppl. 2):265– PATHOGENESIS OF TUBERCULOSIS
266. 96. Dannenberg, A. M., Jr. 1968. Cellular
90. Dannenberg, A. M., Jr. 1980. Macro- hypersensitivity and cellular immunity in
phages and monocytes, p. 1–17. In J.A. Spit- the pathogenesis of tuberculosis: specificity,
tell, Jr. (ed.), Clinical Medicine, vol. 5. Harper systemic and local nature, and associated
& Row, Publishers, Inc., New York, N.Y. macrophage enzymes. Bacteriol. Rev. 32:85–
See publication 94 for the update in the next 102.
edition. This review puts local and systemic immunity to
91. Dannenberg, A. M., Jr. 1980. Macro- tuberculosis in their proper perspective. See
chapter 5.
phages and monocytes, p. 137–153. In J. L.
Spivak (ed.), Fundamentals of Clinical Hema- 97. Dannenberg, A. M., Jr., and M. Sugi-
tology. Harper & Row, Publishers, Inc., New moto. 1976. Liquefaction of caseous foci in
York, N.Y. tuberculosis. Editorial. Am. Rev. Respir. Dis.
113:257–259.
Republication of publication 90. See publication
95 for the update in the next edition. This review briefly describes what little is known
about the causes of liquefaction and cavity for-
92. Dannenberg, A. M., Jr., and M. Suga. mation, including some of our own unpublished
1981. Histochemical stains for macro- experiments. It is summarized in chapter 4.
phages in cell smears and tissue sections: 98. Dannenberg, A. M., Jr. 1978. Pathogen-
-galactosidase, acid phosphatase, nonspe- esis of pulmonary tuberculosis in man and
cific esterase, succinic dehydrogenase, and animals; protection of personnel against
cytochrome oxidase, p. 375–396. In D. O. tuberculosis, p. 65–75. In J. L. Montali (ed.),
Adams, P. J. Edelson, and H. S. Koren (ed.), Mycobacterial Infections of Zoo Animals. Smith-
Methods for Studying Mononuclear Phagocytes. sonian Institution Press,Washington, D.C.
Academic Press, Inc., New York, N.Y. This review compares the disease caused by var-
This article describes the methodology we used. ious mycobacteria in different animal species,
93. Dannenberg, A. M., Jr., M. Suga, and including birds.Also see publication 103.
J. E. Garcia-Gonzalez. 1981. Macrophages 99. Dannenberg, A. M., Jr. 1980. Pathogene-
in granulomas: histochemical evidence sug- sis of tuberculosis, p. 1264–1281. In A. P. Fish-
gesting local control of heterogeneous func- man (ed.), Pulmonary Diseases and Disorders.
tions. Haematol. Blood Transfusion 27:109–119. McGraw-Hill Book Co., New York, N.Y.
This is a concise review of various macrophage This is the first edition of this textbook. See pub-
functions. lications 105 and 115 for updated versions.
94. Dannenberg, A. M., Jr. 1983. Macro- 100. Dannenberg, A. M., Jr., M. Suga, and
phages and monocytes, p. 1–21. In J.A. Spit- J. E. Garcia-Gonzalez. 1980. Cellular
APPENDIX D 䡵 413

composition of the tuberculous (BCG) gran- 107. Dannenberg,A. M., Jr. 1990. Controlling
uloma: local differentiation and turnover of tuberculosis: the pathologist’s point of view.
macrophages, p. 21–32. In D. L. Boros and Fifth Forum in Microbiology on “Killing
T.Yoshida (ed.), Basic and Clinical Aspects of Intracellular Mycobacteria: Dogmas and
Granulomatous Diseases. Elsevier/North- Realities.” Res. Microbiol. 141:192–196,
Holland, Inc.,Amsterdam,The Netherlands. 262–263.
See chapters 5 and 10. Here, we describe some of the areas in patho-
101. Dannenberg, A. M., Jr. 1982. Pathogen- genesis of tuberculosis that need further research.
See chapter 25 for an update.
esis of pulmonary tuberculosis. Am. Rev.
Respir. Dis. 125(no. 3 Suppl.):25–29. 108. Dannenberg, A. M., Jr. 1991. Delayed-
type hypersensitivity and cell-mediated
Koch Centennial Issue (by invitation).
immunity in the pathogenesis of tuberculo-
102. Dannenberg, A. M., Jr. 1982. Pathogen-
sis. Immunol.Today 12:228–233.
esis of pulmonary tuberculosis: basic princi-
This report describes for the first time the ben-
ples. Indian J. Chest Dis. 24:68–77.
eficial role of caseation in tuberculosis. It explains
Koch Centennial Issue. why DTH developed throughout mammalian
103. Dannenberg, A. M., Jr. 1984. Pathogen- evolution, despite the tissue damage it produces.
esis of tuberculosis: native and acquired resis- Included are photomicrographs of various
tance in animals and humans, p. 344–354. In stages in the pathogenesis of this disease. Publi-
Microbiology—1984. American Society for cation 109 contains our revised numbering sys-
Microbiology,Washington, D.C. tem for these stages.
109. Dannenberg, A. M., Jr. 1993. Immuno-
This review is similar to publication 98. It repro- pathogenesis of pulmonary tuberculosis.
duces (with slight modifications) Francis’s table Hosp. Pract. 28:51–58.
that compares the characteristics of tuberculosis
in 23 different animal species. (Francis, J. 1958. This review was written for clinicians. It contains
Tuberculosis in Animals and Man.A Study in Com- innovative drawings of the various stages in the
parative Pathology, p. 294. Cassell and Co., Ltd., pathogenesis of tuberculosis in a revised num-
London, U.K.). bering sequence. We should have used “tissue-
damaging DTH” or “cytotoxic DTH” in many
104. Dannenberg, A.M., Jr. 1984. Chemical
places of the report, but it was too late when this
and enzymatic host factors in resistance to was called to our attention. See publication 112
tuberculosis, p. 721–760. In G. P. Kubica and and chapter 2.
L. G.Wayne (ed.), The Mycobacteria: a Source- DTH is not damaging when the local con-
book. Marcel Dekker, Inc., New York, N.Y. centration of tuberculin is low. DTH damages tis-
sues only when the tuberculin concentration is
Our chapter (with 290 references) reviews our high, e.g., when many tubercle bacilli are rapidly
understanding of the pathogenesis of tuberculo- multiplying intracellularly within macrophages.
sis, with special emphasis on macrophage func- By killing such macrophages, bacillary multipli-
tions and their enzymes. cation is stopped, because tubercle bacilli do not
The Kubica and Wayne book is an excellent multiply in the solid caseous tissue that results.
source of information on mycobacteria up to
the time of its publication. 110. Dannenberg, A. M., Jr. 1994. Pathogen-
105. Dannenberg, A. M., Jr., and J. F. esis and immunology: basic aspects, p. 17–39.
Tomashefski, Jr. 1988. Pathogenesis of pul- In D. Schlossberg (ed.), Tuberculosis, 3rd ed.
monary tuberculosis, p. 1821–1842. In A. P. Springer-Verlag, New York, N.Y.
Fishman (ed.), Pulmonary Diseases and Disor- This is our first chapter in Schlossberg’s Tubercu-
losis. It was updated in the next two editions,
ders, 2nd ed., vol. 3. McGraw-Hill Book Co., publications 116 and 120.
New York, N.Y. 111. Dannenberg, A. M., Jr. 1994. Rabbit
See publication 115 for the updated version of this model of tuberculosis, p. 149–156. In B. R.
chapter in the next edition. Bloom (ed.), Tuberculosis: Pathogenesis, Protec-
106. Dannenberg, A. M., Jr. 1989. Immune tion, and Control. ASM Press, Washington,
mechanisms in the pathogenesis of pul- D.C.
monary tuberculosis. Rev. Infect. Dis. Here, we reviewed the pathology of the disease
11(Suppl. 2):S369–S378. produced in rabbits by virulent bovine-type
414 䡵 APPENDIX D

tubercle bacilli, by virulent human-type tubercle This is a rather complete review of the patho-
bacilli, and by BCG. It is similar to publication 9. genesis and immunology of tuberculosis. It was
See chapter 13. updated for the next edition. See publication 120.
112. Dannenberg, A. M., Jr., and G. A. W. 117. Dannenberg, A. M., Jr. 2001. Pathogen-
Rook. 1994. Pathogenesis of pulmonary esis of pulmonary Mycobacterium bovis infec-
tuberculosis: an interplay of tissue-damaging tion: basic principles established by the rab-
and macrophage-activating immune bit model. Presented at Third International
responses—dual mechanisms that control Conference on Mycobacterium bovis, Cam-
bacillary multiplication, p. 459–483. In bridge, U.K., July 13–16, 2000. Tuberculosis
B. R. Bloom (ed.), Tuberculosis: Pathogenesis, 81:87–96.
Protection, and Control. ASM Press,Washing- This review is a succinct summary of the patho-
ton, D.C. genesis of tuberculosis.
Our review presents the five stages of pulmonary 118. Dannenberg, A. M., Jr., and F. M.
tuberculosis with cartoons (see chapter 2) as well Collins. 2001. Progressive pulmonary tuber-
as with photographs. culosis is not due to increasing numbers of
Bloom’s Tuberculosis was written by numerous viable bacilli in rabbits, mice and guinea
authorities in the field. It is one of the most com-
plete books on this subject in existence. It cov- pigs, but is due to a continuous host response
ers tuberculosis history, epidemiology, diagnosis, to mycobacterial products. Tuberculosis
pathogenesis, immunology, prevention, and treat- (Edinb.) 81:229–242.
ment, as well as animal models, the cultivation of This review compares the disease in rabbits, mice,
the bacillus, mycobacterial physiology and genet- guinea pigs, and monkeys. See chapter 15.
ics, and laboratory safety. Cole et al. (Cole, S.T., 119. Dannenberg,A. M., Jr. 2003. Macrophage
K. D. Eisenach, D. N. McMurray, W. R.
Jacobs, Jr. [ed.]. 2005. Tuberculosis and the Tuber-
turnover, division and activation within
cle Bacillus.ASM Press,Washington, D.C.) update developing, peak and “healed” tuberculous
a lot of it. lesions produced in rabbits by BCG. Tuber-
113. Dannenberg, A. M., Jr. 1994. Roles of culosis (Edinb.). 83:251–260.
cytotoxic delayed-type hypersensitivity and This review gathers in one place our many reports
macrophage-activating cell-mediated immu- on macrophage kinetics in tuberculous lesions
nity in the pathogenesis of tuberculosis. (produced by BCG) and offers additional inter-
pretations of the data. See publications 34 to 41.
Immunobiology 191:461–473.
Chapter 10 is essentially the same.
114. McMurray, D. N., F. M. Collins, A. M. In brief, nonactivated monocytes/macrophages
Dannenberg, Jr., and D.W. Smith. 1996. still enter arrested “healed” lesions and provide an
Pathogenesis of experimental tuberculosis opportunity for renewed intracellular bacillary
in animal models. Curr. Top. Microbiol. growth. Such growth is then stopped by the
Immunol. 215:157–179. immune forces of the host.Tubercle bacilli may
I wrote the section on rabbit tuberculosis. only be truly dormant in solid caseous tissue, but
they are probably not fully dormant in the tuber-
115. Dannenberg, A. M., Jr., and J. F. culous granulation tissue that surrounds this
Tomashefski, Jr. 1998. Pathogenesis of pul- caseum.These studies explain why immunosup-
monary tuberculosis, p. 2447–2471. In A. P. pression, such as in HIV/AIDS, often activates a
Fishman (ed.), Fishman’s Pulmonary Diseases formerly well-controlled and inapparent tuber-
and Disorders, 3rd ed., vol. 2. McGraw-Hill culous lesion.
Co., Inc., New York, N.Y. 120. Manabe,Y. C., and A. M. Dannenberg,
Our chapter in this edition was updated and con- Jr. 2006. Pathophysiology: basic aspects. Part
tains additional references. I. Pathogenesis of tuberculosis. Part II.
Our chapters in previous editions are publica- Immunology of tuberculosis, p. 18–51. In
tions 99 and 105. D. Schlossberg (ed.), Tuberculosis and Nontu-
116. Dannenberg, A. M., Jr. 1999. Pathophys- berculous Mycobacterial Infections, 5th ed.,
iology: basic aspects. I. Pathogenesis of tuber- McGraw-Hill, New York, N.Y.
culosis. II. Immunology of tuberculosis, p. Our chapter in publications 110 and 116 was
17–47. In D. Schlossberg (ed.), Tuberculosis and updated again. Much of the recent understand-
Nontuberculous Mycobacterial Infections, 4th ed. ing about the immunology of tuberculosis appears
The W. B. Saunders Co., Philadelphia, Pa. in chapters 5 and 6 in the present volume.
APPENDIX D 䡵 415

OUR STUDIES ON IRRITANT 1986. Interstitial fibrosis and collateral ven-


INFLAMMATION AND CONTACT tilation. J.Appl. Physiol. 61:300–303.
SENSITIVITY 126. Feldman, G. M., A. M. Dannenberg,
121. Dannenberg,A. M., Jr., and K. G. Moore. Jr., and J. L. Seed. 1990. Physiologic oxy-
1994.Toxic and allergic skin reactions, evalu- gen tensions limit oxidant-mediated killing
ated in organ-cultured full-thickness human of schistosome eggs by inflammatory cells
and animal skin explants, p. 351–366. In A. and isolated granulomas. J. Leukoc. Biol.
Rougier,A. M. Goldberg, and H. I. Maibach 47:344–354.
(ed.),In Vitro Skin Toxicology (Alternative Meth- 127. Manabe, Y. C., A. M. Dannenberg, Jr.,
ods in Toxicology Series, vol. 10). Mary Ann and W. R. Bishai. 2000.What we can learn
Liebert, Inc, New York, N.Y. from the Mycobacterium tuberculosis genome
This review briefly summarizes most of these sequencing projects. Int. J.Tuberc. Lung Dis.
studies and includes the pertinent literature on 4:S18–S23.
(i) dermal inflammatory reactions produced by
the chemical toxicant dilute sulfur mustard, and The complete genome of M. tuberculosis (H37Rv)
(ii) dermal contact sensitivity reactions produced is now known. It is being used to identify viru-
by dinitrochlorobenzene and oxazolone. See pub- lence factors for the development of new drugs
lications 60 through 73. and new vaccines.
In many of these studies, full-thickness skin 128. Manabe,Y. C.,A. M. Dannenberg, Jr., S.
explants of inflammatory lesions in various stages K. Tyagi, C. L. Hatem, M.Yoder, S. C.
of development and healing were organ cultured, Woolwine, B. C. Zook, M. L. M. Pitt,
and the locally produced inflammatory mediators
and W. R. Bishai. 2003. Different strains of
(extracted by the culture fluids) were correlated
with the histological changes found in the lesion. Mycobacterium tuberculosis cause various spec-
In this review, unfortunately, some section head- trums of disease in the rabbit model of tuber-
ings were not printed correctly. Specifically, the culosis. Infect. Immun. 71:6004–6011.
summaries of parts 4 and 5 were not so designated. With the tubercle-count method, Erdman,
H37Rv, and CDC1551 (Oshkosh strain) had
decreasing virulence for rabbits, in that order.
PART III. COLLABORATIVE
PROJECTS MAINLY FROM 129. Dorman, S., C. L. Hatem, S. Tyagi, K.
OTHER LABORATORIES Aird, J. Lopez-Molina, M. L. M. Pitt, B.
C. Zook, A. M. Dannenberg, Jr., W. R.
122. Gertner, A., B. Bromberger-Barnea, Bishai, and Y. C. Manabe. 2004. Suscep-
A. M. Dannenberg, Jr., R. Traystman, tibility to tuberculosis: clues from studies
and H. Menkes. 1983. Responses of the with inbred and outbred New Zealand
lung periphery to 1.0 ppm ozone. J. Appl. White rabbits. Infect. Immun. 72:1700–1705.
Physiol. 55:770–776.
The Thorbecke inbred New Zealand White rab-
123. Emmett, E. A., P. G. Lewis, F. Tanaka, bits were more susceptible to tuberculosis than
M. Bleecker, R. Fox, A. C. Darlington, commercial outbred rabbits. Following the inhala-
D. R. Synkowski,A. M. Dannenberg, Jr., tion of virulent human-type tubercle bacilli
W. J. Taylor, and M. S. Levine. 1985. (H37Rv), the inbred rabbits produced 3 times as
Industrial exposure to organophosphorus many grossly visible tubercles, and these tubercles
compounds: studies of a group of workers were larger, showed more caseous necrosis, and
contained more bacilli.The tubercle counts in the
with a decrease in esterase-staining mono-
inbred rabbits also showed less variation. The
cytes. Occup. Med. 27:905–914. inbred rabbits had reduced tuberculin sensitivity,
124. Kleeberger, S. R., E. M. Wagner, G. K. and their macrophages produced decreased
Adams III, A. M. Dannenberg, Jr., and amounts of tumor necrosis factor alpha (a primary
E.W. Spannhake. 1985. Effect of repeated cytokine).
antigen exposure on antigen- and mediator- These were the only inbred rabbits that were
induced bronchospasm in sheep. J.Appl. Phys- commercially available in 2003, but unfortunately
(because of a fire), they were no longer available
iol. 59:1866–1873. in 2006 (see chapter 14).
125. Berzon, D. M., H. Menkes, A. M. Dan- The photographs of the granulomas in Fig. 3
nenberg, Jr., A. Gertner, P. Terry, in this publication were inadvertently published
D. Plump, and B. Bromberger-Barnea. vertically instead of horizontally.They should be
416 䡵 APPENDIX D

rotated clockwise and counterclockwise (respec- Pitt, D. N. McMurray, J.-A. Tufariello,


tively) to make the figure descriptions match J. Chan, A. M. Dannenberg, Jr., W. R.
what is portrayed. Bishai, and S. Mendez. Mycobacterial
130. Kesavan,A. K., S. Mendez, C. L. Hatem, gene expression in the resistant rabbit model
J. Lopez-Molina, K.Aird, M. L. M. Pitt, of latency, reactivation and immune recon-
A. M. Dannenberg, Jr., and Y. C. Man- stitution tuberculosis. In preparation.
abe. 2005. Effects of dexamethasone and Rabbits were aerosol infected with M. tuberculosis
transient malnutrition on rabbits infected (H37Rv). From 10 to 15 weeks after infection,
with aerosolized M. tuberculosis CDC1551. they were given dexamethasone.Then, the dex-
Infect. Immun. 73:7056–7060. amethasone was discontinued, and the rabbits
Transient malnutrition decreased the resistance of were euthanized 5 weeks later. This regimen
these rabbits in a manner similar to that of dex- reproduced (i) the paucibacillary stage of this dis-
amethasone. ease, (ii) the latent stage, and (iii) the early stage of
131. Manabe,Y. C., A. K. Kesavan, J. Lopez- reactivation, which are commonly found in
Molina, C. L. Hatem, M. Brooks, humans. Some of the mycobacterial genes that are
R. Fujiwara, K. Hochstein, M. L. M. upregulated in each of these stages were identified.
APPENDIX E
GUIDELINES FOR PREVENTING THE
TRANSMISSION OF MYCOBACTERIUM
TUBERCULOSIS IN HEALTH-CARE SETTINGS†
Department of Health and Human Services, Centers for
Disease Control and Prevention,Atlanta, Georgia

CONTENTS
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1
Overview . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1
HCWs Who Should Be Included in a TB Surveillance Program . . . . . . . . . 3
Risk for Health-Care-Associated Transmission of M. tuberculosis . . . . . . . . . . 6
Fundamentals of TB Infection Control . . . . . . . . . . . . . . . . . . . . . . . . . . . 6
Relevance to Biologic Terrorism Preparedness . . . . . . . . . . . . . . . . . . . . . . 8
Recommendations for Preventing Transmission of M. tuberculosis in
Health-Care Settings . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
TB Infection-Control Program . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
TB Risk Assessment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9
Risk Classification Examples . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 11
Managing Patients Who Have Suspected or Confirmed TB Disease:
General Recommendations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 16
Managing Patients Who Have Suspected or Confirmed Disease:
Considerations for Special Circumstances and Settings . . . . . . . . . . . 19
Training and Educating HCWs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 27
TB Infection-Control Surveillance . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 28
Problem Evaluation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 32
Collaboration with the Local or State Health Department . . . . . . . . . . . . 36
Environmental Controls . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 36
Respiratory Protection . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 38
Cough-Inducing and Aerosol-Generating Procedures . . . . . . . . . . . . . . . . 40


From Recommendations and Reports, Morb. Mortal.Wkly. Rep. 54(RR-17):1-141, December
30, 2005. Available on the Internet at http://www.cdc.gov/mmwr/PDF/rr/rr5417.pdf or
http://www.cdc.gov/mmwr/preview/mmwrhtml/rr5417a1.htm.

417
418 䡵 APPENDIX E

Supplements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 42
Estimating the Infectiousness of a TB Patient . . . . . . . . . . . . . . . . . . . . . . 42
Diagnostic Procedures for LTBI and TB Disease . . . . . . . . . . . . . . . . . . . . 44
Treatment Procedures for LTBI and TB Disease . . . . . . . . . . . . . . . . . . . . 53
Surveillance and Detection of M. tuberculosis Infections in
Health-Care Settings . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 56
Environmental Controls . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 60
Respiratory Protection . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 75
Cleaning, Disinfecting, and Sterilizing Patient-Care Equipment
and Rooms . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 79
Frequently Asked Questions (FAQs) . . . . . . . . . . . . . . . . . . . . . . . . . . . . 80
Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 89
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 89
Terms and Abbreviations Used in This Report . . . . . . . . . . . . . . . . . . . . . . 103
Glossary of Definitions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 107
Appendices . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 121
Continuing Education Activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . CE-1
APPENDIX F
COLLECTED ABSTRACTS IN THIS VOLUME

SECTION 1. PATHOGENESIS following stages. In humans, the disease begins


OF TUBERCULOSIS with the establishment of only a single primary
CHAPTER 1. OVERVIEW pulmonary tubercle.
Tuberculosis is still one of the major diseases of the Stage 1: Ingestion and often destruction of
world, especially in developing countries. It kills bacilli by pulmonary alveolar macrophages.
over 2 million people each year, more than any Stage 2: Logarithmic growth of bacilli within
other infectious disease. nonactivated macrophages that entered the devel-
Childhood-type pulmonary tuberculosis is a oping tubercle from the bloodstream.
disease of susceptible hosts, such as infants and Stage 3: Arrest of the logarithmic bacillary
immunosuppressed individuals. From the primary growth by delayed-type hypersensitivity, which
parenchymal lesion, the bacilli frequently spread via kills the bacilli-laden macrophages and often forms
lymphatics and cause caseous lesions of the hilar a solid caseous center in the tubercle.
lymph nodes.The bacilli may also spread via the Stage 4a: In hosts with weakly developed cell-
bloodstream and cause lesions elsewhere in the mediated immunity, enlargement of the tubercle
host.The primary lesion, as well as the metastatic and its caseous center with hematogenous dis-
lesions, often progresses until the host succumbs. semination of the bacilli.
Adult-type pulmonary tuberculosis is a disease Stage 4b: In hosts with strongly developed cell-
of innately resistant hosts, a category that includes mediated immunity, stabilization or regression of
most immunocompetent persons. The active the tubercle.
parenchymal lesion (often subapical) frequently Stage 5: Liquefaction of the caseous center,
forms a cavity, in which the bacilli may multiply extracellular bacillary growth, cavity formation, and
extracellularly. If so, the bacilli may spread via the bronchial dissemination of the bacilli.
bronchial tree to other parts of the lung. In such These stages are not distinct but blend into
a resistant host, many metastatic microscopic lesions each other.Also, stages 3, 4, and 5 may occur in the
do not progress, but those that do may form new same lung and even in different parts of the same
cavities. Also, caseous bronchopneumonia may lesion, depending on the local concentration of
occur when an appreciable amount of liquefied bacilli and their tuberculin-like products. Native
caseum enters the bronchial tree. Cavity formation and acquired resistance is never absolute, because
perpetuates tuberculosis in humankind because a large number of tubercle bacilli (which have
coughing spreads bacilli from the lungs into the grown extracellularly in a cavity) can overwhelm
environment, where they may infect other people. even the best-developed host resistance and cause
Contracting clinical tuberculosis depends on secondary pulmonary lesions.
(i) the size and physiological state of the bacillary
particle, (ii) its virulence, and (iii) the native and CHAPTER 3. TYPES OF HUMAN
acquired resistance of the host. PULMONARY TUBERCULOSIS
How to protect personnel against tuberculosis is Human tuberculosis most frequently occurs as a
also discussed in this chapter. UV lights (shielded tiny inapparent lesion that stays dormant through-
to protect people’s eyes) or HEPA-filtered air puri- out the life of the host. If clinical disease is pro-
fiers should be used more frequently in hospital duced, it varies from the rapidly progressing,
areas where tubercle bacilli are likely to be present. hematogenously spread disease that occurs in
infants and immunosuppressed individuals to a
CHAPTER 2. STAGES IN THE chronic, slowly progressing cavitary disease that is
PATHOGENESIS OF HUMAN commonly found in immunocompetent adults.
AND RABBIT TUBERCULOSIS A brief overview of both the childhood and adult
After the inhalation of tubercle bacilli by rabbits types of pulmonary tuberculosis is presented in
and humans, the disease may progress through the chapter 1.

419
420 䡵 APPENDIX F

In this chapter, the gross and histopathological mainly by dendritic cells. In tuberculous lesions,
characteristics of each type are described in more DTH kills (nonactivated) macrophages that con-
detail. Included are the possible causes of subapi- tain numerous tubercle bacilli when these bacilli
cal localization in adult-type tuberculosis, the loca- release tissue-damaging local concentrations of
tion of multidrug-resistant tubercle bacilli in the tuberculin-like products. In the resulting (solid)
lungs, the characteristics of tuberculosis in caseous necrosis, bacillary growth is inhibited and
immunocompromised individuals, and a compar- many bacilli die because of low oxygen tension
ison of inapparent arrested primary lesions in and other factors. Therefore, tissue-damaging
humans and rabbits. For further discussion of many DTH has apparently evolved in mammals to stop
of the topics in this chapter, see reference 1. continuing bacillary growth within the nonacti-
vated macrophages that have permitted such
CHAPTER 4. LIQUEFACTION growth.
OF CASEOUS FOCI AND In tuberculous lesions, CMI activates macro-
CAVITY FORMATION phages so that they can inhibit and destroy ingested
Part I is a review of the literature on liquefaction tubercle bacilli. DTH can also activate macro-
and cavity formation. Liquefaction seems to be a phages if only low local concentrations of tuber-
delayed-type hypersensitivity reaction to the tuber- culin-like products are present. In this respect,
culin-like products of the bacillus. It seems to be DTH (the host reaction to tuberculin-like prod-
carried out by hydrolytic enzymes from the sur- ucts) is part of the CMI response.
rounding host cells and possibly by enzymes within Nonactivated macrophages continuously enter
the caseum after inhibitors have dissipated. Liq- every tuberculous lesion and may ingest tubercle
uefied caseum and cavities occur frequently in bacilli (see chapter 10).To stop the progression of
rabbits inhaling virulent bovine-type tubercle the disease, macrophages containing a few bacilli
bacilli. They also occur occasionally in rabbits must be activated by CMI to prevent further intra-
inhaling virulent human-type tubercle bacilli. Part cellular bacillary multiplication, and macrophages
II describes experiments of long duration in which containing many bacilli must be killed by the
pulmonary cavities were produced in commercial DTH process. The interplay of CMI and tissue-
New Zealand White rabbits by aerosolized viru- damaging DTH seems to explain the entire spec-
lent bovine-type bacilli (Mycobacterium bovis, trum of the disease found in tuberculous hosts.
Ravenel strain).After the inhalation of a low num- Both DTH and CMI exert their control locally.
ber of these bacilli, liquefied caseum and cavities Their main systemic manifestation is to provide an
occurred in 6 to 10 weeks.After the inhalation of expanded antigen-specific lymphocyte popula-
a high number of these bacilli, liquefied caseum tion to infiltrate local sites of bacillary lodgement.
and cavities occurred sooner. Details on the gross Antibodies that aid phagocytosis apparently play
pathology and histopathology of these pulmonary little or no role in the destruction of the tubercle
lesions are presented. Part III describes two attempts bacillus. The bacillus readily enters macrophages
to reduce liquefaction and cavity formation in without being opsonized by antibodies and evi-
tuberculous rabbits. One was immunotherapy dently can multiply intracellularly within nonacti-
with Mycobacterium vaccae. The other was therapy vated macrophages in the presence of antibodies.
with Ritonavir, a proteinase inhibitor. Neither However, in immunized hosts, antibodies seem to
treatment (as administered) had any observable be an important host defense against the develop-
effect. ment of clinically apparent tuberculosis. Antigen-
antibody reactions at sites of bacillary lodgement
SECTION 2. IMMUNOLOGY result in the production of chemotactic factors,
OF TUBERCULOSIS including the C5a component of complement. In
CHAPTER 5. DELAYED-TYPE immunized hosts, such chemotaxins cause a rapid
HYPERSENSITIVITY, CELL- local accumulation of dendritic cells, macrophages,
MEDIATED IMMUNITY, AND and antigen-specific T cells—all of which would
ANTIBODIES IN TUBERCULOSIS accumulate more slowly without the local antigen-
Both delayed-type hypersensitivity (DTH) and antibody reaction. In other words, in immunized
cell-mediated immunity (CMI) are T-lympho- hosts the antigen-antibody reaction enables the
cyte responses to bacillary antigens presented local cell-mediated immune response to occur so
APPENDIX F 䡵 421

rapidly that the bacillus is often inhibited before it It was concluded that the local microvasculature
multiplies extensively. increases relatively little during the course of this
slowly healing infection. A greater blood flow
through existing capillaries evidently provides most
CHAPTER 6. MACROPHAGES AND
OTHER CELLS IN TUBERCULOUS of the nourishment needed by the infiltrating cells.
LESIONS These studies also demonstrated that microvas-
The main types of cells participating in rabbit cular thrombosis is a major cause of the caseous
tuberculous lesions are dendritic cells, macrophages, necrosis that occurs during the course of this
natural killer cells, lymphocytes, and granulocytes. disease.
The role of most of these cells is discussed only
briefly in this chapter because details are available CHAPTER 9. EARLY PULMONARY
in textbooks of immunology.The role of macro- LESIONS IN RABBITS
phages, however, is presented more fully because In rabbits, 1- to 7-day pulmonary tuberculous
their rates of turnover, their states of activation, their lesions produced by aerosols are difficult to find
extracellular and intracellular enzymes, and their because the inhaled dose of tubercle bacilli cannot
heterogeneity have been extensively studied in my be made large enough. A large intravenous dose,
laboratory in the rabbit model of tuberculosis. however, readily produces many such tubercles.
This chapter describes their characteristics and
provides information on the activation and mul-
SECTION 3. TUBERCULOUS LESIONS
tiplication of macrophages within such lesions.
CHAPTER 7. STRUCTURAL To produce these early lesions, we injected rab-
COMPONENTS OF bits intravenously with 108 to 109 tubercle bacilli
TUBERCULOUS LESIONS (BCG).The blood-borne macrophages that entered
This chapter describes the structural components the developing tubercles became partly activated
of tuberculous lesions: the surrounding granulation during the first day. These entering macrophages
tissue, solid caseous necrosis, liquefied caseum, retained their ability to divide, i.e., incorporate
cavities, and the fibrosis and calcification of heal- [3H]thymidine ([3H]TdR), even though they had
ing lesions. ingested tubercle bacilli. In contrast, fully activated
macrophages within tuberculous lesions lose their
CHAPTER 8. MICROVASCULAR ability to divide (see chapter 10).
DENSITY IN TUBERCULOUS LESIONS Pulmonary alveolar macrophages did not seem
The vasculature plays an important role in the to participate in early pulmonary lesions produced
pathogenesis of tuberculous lesions. Blood vessels by the intravenous route, but accumulated in the
bring in the host defense cells, and vascular throm- surrounding alveolar spaces. However, even though
bosis is a major cause of caseous necrosis. This these alveolar macrophages were highly activated,
chapter describes the study of microvascular den- they retained their ability to divide.
sity in tissue sections of developing and healing
dermal BCG lesions and in 48-h dermal tuberculin CHAPTER 10. MACROPHAGE
reactions. TURNOVER, DIVISION, AND
Rabbits were placed under deep terminal anes- ACTIVATION IN TUBERCULOUS
thesia,and their entire vasculature was perfused (via LESIONS
the aorta) with a gelatin-colloidal carbon suspen- In rabbit BCG lesions, the turnover of mono-
sion.Then, serial 250-µm-thick tissue sections of nuclear cells was most rapid in BCG lesions at
the dermal BCG lesions were prepared, and the 2 to 3 weeks, when the lesion size peaked and
total length of the microvasculature in the whole tuberculin sensitivity and acquired cellular resis-
BCG lesion was calculated from measurements tance were well developed. (The mononuclear
made with a microscope containing an ocular grid. cells were mostly macrophages, with some medium
By 3 days, the vascular density in BCG lesions and large lymphocytes and probably some den-
had increased to roughly 1.6 times that found in dritic cells.) At this 2- to 3-week peak, more
normal skin. It remained at this level for at least 6 macrophages entered, more died or left, more
to 7 weeks. The vascular density in tuberculin remained at the site, and more became activated
reactions showed a similar increase. than before or afterward. Before this time, the
422 䡵 APPENDIX F

host had neither delayed-type hypersensitivity nor Lurie showed that resistance to the establishment
cell-mediated immunity, so no antigen-specific of tuberculosis and resistance to its progress are sep-
enhancement of the inflammatory response arate phenomena: his inbred resistant rabbits con-
occurred.After this time, the bacilli and their anti- verted their tuberculin skin tests an average of 2.7
genic products had decreased, so antigen-specific months sooner than did his inbred susceptible
stimuli for cell infiltration and activation were rabbits.Yet, once established, the disease progressed
reduced. In “healed” lesions, the mononuclear cell slowly in the resistant rabbits and rapidly in the sus-
turnover still occurred but was decreased. ceptible rabbits.The separation of the establishment
The continuous entry of live nonactivated and progress of tuberculosis is only applicable to
macrophages into the viable parts of tuberculous experiments in which occasional fully virulent
lesions provides fresh intracellular sites where tubercle bacilli are inhaled over many months. It
tubercle bacilli can multiply before they are again does not seem applicable to rabbits or humans
inhibited by the delayed-type hypersensitivity and inhaling human-type tubercle bacilli, which are
cell-mediated immunity of the host. In tubercu- never fully virulent in these hosts.
losis, bacillary dormancy of long duration can Airborne infection of laboratory animals over
only be present in caseous necrotic tissue where no many months has, however, established other
live host cells exist. concepts directly applicable to tuberculosis in
humans. (i) Only a single grossly visible primary
CHAPTER 11. LURIE’S PULMONARY pulmonary lesion will be produced, despite the
TUBERCLE-COUNT METHOD continuous presence of virulent tubercle bacilli
Lurie’s tubercle-count method consists of counting in the air.The immunity developed in response
the number of grossly visible primary pulmonary to the primary lesion is evidently sufficient to
tubercles, present 5 weeks after an aerosol infection prevent other occasionally inhaled tubercle bacilli
of rabbits with virulent human-type tubercle bacilli. from causing grossly visible lesions. (ii) Some
It is a quantitative measure of clinically apparent dis- animals (and perhaps a few humans) may convert
ease.At 5 weeks, the grossly visible primary tuber- their dermal tuberculin reactions, and yet show
cles are easily recognized, and many microscopic no grossly visible primary lesions in their lungs
tubercles have regressed. Since human-type tuber- at necropsy.This occurrence may be due to the
cle bacilli are not fully virulent for rabbits, the pul- early spread of inhaled bacilli out of the lungs to
monary-count method has a sensitivity that is not the hilar lymph nodes, where the growth of
possible with fully virulent strains. tubercle bacilli can be more easily controlled.
The number of grossly visible pulmonary tuber- These concepts are consistent with what Riley
cles produced by human-type bacilli decreases (i) found when he exposed guinea pigs for months
when rabbits are infected with bacilli of reduced to air from a ward containing sputum-positive
virulence, (ii) when rabbits of high genetic (innate) tuberculous patients.
resistance are used, and (iii) when rabbits are effec-
tively immunized, so that they can rapidly activate CHAPTER 13. RESPONSE OF RABBITS
macrophages and stop the development of early TO INHALED TUBERCLE BACILLI
tubercles while they are still microscopic in size. Virulent bovine-type and human-type tubercle
Therefore, the pulmonary tubercle-count method bacilli and BCG are of decreasing virulence for
can be used to assess (i) bacillary virulence, (ii) the rabbits, in that order.The host uses the same type
genetic resistance of the host, and (iii) the efficacy of immune response to control each of these infec-
of vaccines for tuberculosis. tions, but the response is more effective with bacil-
lary strains of reduced virulence.
SECTION 4. TUBERCULOSIS IN With fully virulent bovine-type tubercle bacilli,
RABBITS AND OTHER COMMON only 3 bacillary units of 1 to 3 bacilli must be
LABORATORY ANIMALS inhaled to cause one grossly visible tubercle at 5
CHAPTER 12. NATURAL AIRBORNE weeks. In Lurie’s inbred susceptible rabbits, bovine-
INFECTION type bacilli produced the childhood form of tuber-
Using natural airborne infection of virulent culosis with hematogenous dissemination. In
bovine-type tubercle bacilli over many months, Lurie’s resistant rabbits, bovine-type bacilli pro-
APPENDIX F 䡵 423

duced the adult form of tuberculosis with pul- culosis were evident histologically: mature epithe-
monary cavities and bronchial dissemination. lioid cells (now known as highly activated
Human-type tubercle bacilli are not fully viru- macrophages) were always more numerous in the
lent in rabbits. In Lurie’s inbred resistant rabbits and lesions of resistant rabbits than in the lesions of sus-
in commercially available New Zealand White ceptible rabbits, irrespective of the differences in vir-
rabbits, 300 to 1,900 bacillary units must be inhaled ulence of the infecting bacillary strains.
to produce one grossly visible primary pulmonary The genetic resistance of these rabbits resides in
tubercle at 5 weeks, depending on the virulence of their ability to activate macrophages to control the
the infecting strain and on variations among the growth of tubercle bacilli, both nonspecifically
individual rabbits. In Lurie’s susceptible rabbits, and immune-specifically. Crossbreeding showed
such tubercles often gave rise to secondary tuber- that the genetic resistance to tuberculosis is mul-
cles. In Lurie’s resistant rabbits and commercial tifactorial, with genes associated with resistance
New Zealand White rabbits, human-type tubercle being dominant over susceptibility genes.
bacilli usually healed in a few months, except when The commercially available outbred New
they formed cavities, which persisted much longer. Zealand White rabbits seem almost as resistant as
However, in all rabbits, lesions produced by human- Lurie’s inbred resistant strain III rabbits. Thor-
type bacilli eventually heal and are never fatal. becke inbred rabbits were distinctly more suscep-
BCG is avirulent in all common laboratory tible than commercial outbred rabbits, but appar-
species. In commercial rabbits, a large inhaled dose ently not as susceptible as Lurie’s inbred C and FC
of aerosolized BCG produces few, if any, tiny non- rabbits.Van Zutphen’s inbred rabbits (which are
progressive tubercles. Most of the inhaled BCG hypo- and hyperresponsive to dietary cholesterol,
bacilli are apparently destroyed by the alveolar respectively) have not been adequately studied for
macrophages before they can multiply appreciably. resistance to tuberculosis.
Without such multiplication, the degree of immu-
nization would be negligible. Similar to rabbits, CHAPTER 15. COMPARISONS OF
humans should be less immunized by the inhala- TUBERCULOSIS IN RABBITS, MICE,
tion of BCG than by parenteral administration, in AND GUINEA PIGS
which higher doses of BCG can be injected and In recent times, mice have been by far the most fre-
greater bacillary multiplication can occur (because quently used animal for the study of tuberculosis.
the alveolar macrophages are bypassed). In mice Guinea pigs and rabbits are used less often, and
and guinea pigs (which are more susceptible to monkeys are used only occasionally.
Mycobacterium tuberculosis), inhaled BCG would Rabbits are highly susceptible to bovine-type
immunize more effectively, because their alveolar tubercle bacilli and are the only common labora-
macrophages do not destroy inhaled BCG as read- tory species in which chronic cavitary tuberculo-
ily (see chapter 22). sis with bronchial spread is readily produced. Rab-
bits develop grossly visible pulmonary tubercles
CHAPTER 14. CHARACTERISTICS OF following the inhalation of virulent human-type
RESISTANCE AND SUSCEPTIBILITY tubercle bacilli, but these tubercles usually regress,
TO TUBERCULOSIS IN LURIE’S as they do in most humans.Rabbit pulmonary
INBRED RABBITS lesions caused by virulent human-type tubercle
Lurie’s rabbits were inbred for either susceptibil- bacilli sometimes form nonprogressive cavities.
ity or resistance to the progress of tuberculosis. Rabbits die after an infection with virulent bovine-
When infected with virulent bovine-type tuber- type tubercle bacilli, but eventually heal an infec-
cle bacilli, the susceptible rabbits developed a tion with virulent human-type tubercle bacilli.
rapidly progressing, hematogenously spreading Mice develop slowly progressing pulmonary
“childhood type” of tuberculosis, and the resistant tubercles with both bovine and human strains of
rabbits developed a slowly progressing, cavitary, tubercle bacilli, but the disease progresses more
bronchial-spreading “adult type” of tuberculosis. rapidly with the bovine strain. With virulent
In lesions produced by virulent bovine-type human-type bacilli, the tubercles of mice contain
bacilli, by human-type bacilli, and by BCG, the a larger number of viable bacilli than do the tuber-
same manifestations of genetic resistance to tuber- cles of rabbits and guinea pigs.Apparently, the low
424 䡵 APPENDIX F

levels of delayed-type hypersensitivity (DTH) in bers of bacilli) liquefaction, cavity formation, tuber-
mice and the rarity of caseous necrosis allow the culous bronchopneumonia, and hematogenous dis-
bacillus to grow to higher titers in the logarithmic semination occurred in some of the rabbits.
stage.After these titers are reached, the good cell- One of Lurie’s inbred rabbit strains was evi-
mediated immunity (CMI) developed by mice dently deficient in glucocorticoid production. In
reduces the intracellular multiplication of the these rabbits, the administration of physiological
majority of the bacilli to almost a dormant state. doses of adrenocorticotropic hormone increased
However, the low percentage of nondormant their resistance to tuberculosis.
bacilli causes the disease to slowly progress until the
animal dies.
Guinea pigs are highly susceptible to both CHAPTER 17. EFFECTS OF ESTROGEN,
human and bovine strains of tubercle bacilli.They CHORIONIC GONADOTROPIN, AND
THYROID HORMONES ON
usually develop a rapidly progressing, hematoge-
TUBERCULOSIS
nously spread form of tuberculosis, similar to that
Since estrogen increased the hyaluronic acid and
developed by infants and immunosuppressed indi-
water content of the skin, it decreased the spread
viduals. Because guinea pigs develop relatively
of intradermally injected virulent bovine-type
high sensitivity to tuberculin, which causes con-
tubercle bacilli in rabbits. Chorionic gonadotropin
siderable caseous necrosis, their lesions contain a
had the reverse effect.These two sex hormones had
rather low number of viable bacilli. In other
no appreciable effect on the innate or acquired
words, despite the rather poor CMI developed by
ability of the host to control the progression of
guinea pigs, their good DTH effectively reduces
tuberculosis, because neither hormone apprecia-
the number of viable bacilli in their lesions. How-
bly changed the number of primary pulmonary
ever, because of the extensive lung destruction
tubercles generated by aerosols of virulent human-
caused by tissue-damaging DTH, guinea pigs
type tubercle bacilli. However, estrogen markedly
often die in less time than do rabbits and mice.
suppressed the development of amyloid in the
Rhesus monkeys are very susceptible to tuber-
spleens of rabbits dying of the more chronic form
culosis, but cynomolgus monkeys are more resis-
of tuberculosis caused by bovine-type bacilli.
tant. Some cynomolgus monkeys can even stop the
In rabbits, triiodothyronine or thyroxine
progression of the disease.
increased host resistance in that they decreased
the number of grossly visible primary pulmonary
tubercles produced by the inhalation of virulent
SECTION 5. EFFECTS OF HORMONES human-type tubercle bacilli, whereas thyroidec-
AND X-IRRADIATION ON
TUBERCULOSIS tomy or propylthiouracil treatment increased the
number of such primary tubercles.Thyroid hor-
CHAPTER 16. EFFECTS OF CORTISONE mones were most beneficial in inbred rabbits of
AND ADRENOCORTICOTROPIC intermediate resistance.The resistance of the most
HORMONE ON TUBERCULOSIS
susceptible inbred C rabbits and that of the
Pharmacological amounts of glucocorticoids are most resistant III(r) rabbits were not appreciably
frequently given as therapy for a variety of aller- increased by thyroid hormones.
gic, autoimmune, and inflammatory conditions,
such as asthma and rheumatoid arthritis. When
such drugs are continued for long periods of time, CHAPTER 18. EFFECTS OF WHOLE-
latent tuberculosis may reactivate. BODY X-IRRADIATION ON
In tuberculous rabbits infected with human-type TUBERCULOSIS
tubercle bacilli, pharmacological amounts of glu- Commercial rabbits were irradiated with 400 rads
cocorticoids decreased cell-mediated immunity and of whole-body X-irradiation—a sublethal dose.At
delayed-type hypersensitivity. Macrophages were 2 or 10 days thereafter, they were injected intra-
poorly activated, and tubercle bacilli grew to large dermally with BCG. Between 2 and 4 weeks after
numbers within these phagocytes.After glucocor- irradiation, the BCG lesions and 48-h tuberculin
ticoid administration was stopped, the tuberculin reactions in the irradiated group were smaller than
sensitivity returned, and (because of the large num- those of the nonirradiated controls. The BCG
APPENDIX F 䡵 425

lesions in the irradiated group also contained more bacilli, an adequate supply of defense cells is avail-
bacilli. able. However, with acute infections (requiring
This dose of whole-body X-irradiation evi- many more defense cells), the irradiated host would
dently decreased the supply of macrophages and have an inadequate supply.
lymphocytes from “cell factories” in the bone
marrow and lymphoid tissues, so that fewer cells
were available to infiltrate the BCG lesions.These SECTION 6. CYTOKINES AND
cell factories had apparently recovered 4 to 5 VASCULAR ADHESION MOLECULES
weeks after irradiation, because BCG lesions start- IN TUBERCULOUS LESIONS
ing at this time were the same size as those in the CHAPTER 19. CYTOKINE PRODUCTION
nonirradiated controls and contained the same IN PRIMARY BCG LESIONS
number of bacilli. A sequential histochemical study of cytokines in
Pulmonary alveolar macrophages (AM) recov- developing and healing rabbit tuberculous (BCG)
ered by bronchoalveolar lavage (BAL) from irra- lesions is described. In tissue sections, interleu-
diated rabbits contained higher levels of hydrolytic kin-1 (IL-1), tumor necrosis factor alpha
enzymes than did AM from nonirradiated controls. (TNF-), macrophage chemoattractant (activating)
The AM from the irradiated group were apparently protein 1 (MCP-1), and IL-8 were evaluated for
an older (more activated) cell population, because cytokine mRNA by in situ hybridization tech-
they had ingested inhaled particles for a longer niques and for cytokine protein by immunohis-
period of time. The irradiation evidently had tochemical techniques. In tissue homogenates,
reduced the young macrophage population that gamma interferon (IFN-) mRNA was evaluated
replenishes the AM population. by reverse transcription-PCR.
When heat-killed tubercle bacilli in oil were In the BCG lesions, the percentage of mononu-
injected intravenously 1 day after irradiation, the clear cells that contained the mRNAs of these
BAL specimens obtained 9 and 10 days later (from cytokines showed a biphasic pattern. At 1 to 3
the resulting granulomatous lungs) contained about days, a peak occurred, which was apparently a non-
half as many macrophages as did those from non- specific inflammatory response caused by the tuber-
irradiated controls.This finding indicated that the cle bacilli in the BCG vaccine.At 5 days, the per-
irradiation reduced the supply of macrophages centage of mononuclear cells containing the
from the bone marrow.When the heat-killed tuber- cytokine mRNAs was significantly reduced, but by
cle bacilli were injected 4 weeks after irradiation, 9 days, the percentage had again increased, and the
BAL specimens from the granulomatous lungs rabbits had become tuberculin positive.This second
of the irradiated and the nonirradiated animals peak was apparently antigen specific.With IFN-,
contained similar numbers of macrophages, which the two mRNA peaks were delayed by 2 days.
indicated that the supply of these macrophages Mononuclear cells containing IL-1 and
had recovered. IL-8 mRNAs were more numerous surrounding
Rabbits were infected by aerosol with virulent the caseous center. These cytokines evidently
human-type tubercle bacilli (H37Rv) at 12 or 30 recruited the polymorphonuclear leukocytes that
days after irradiation. In each case, 5 weeks after were common in this location. Mononuclear cells
infection, the number of primary pulmonary containing MCP-1 mRNA were more numerous
tubercles in their lungs was the same in both the in the outer third of the lesion where new
irradiated and the nonirradiated groups. Also, macrophages and lymphocytes were being re-
the number of viable bacilli in these tubercles cruited.
was the same. Therefore, this sublethal dose of Both the nonspecific and antigen-specific
irradiation had no appreciable effect on the devel- cytokine responses of BCG vaccines are evidently
opment and progress of primary pulmonary synergistic. The early nonspecific cytokine
tubercles in rabbits. (chemokine) response causes a local accumulation
In brief, X-irradiation reduces the bone mar- of antigen-presenting cells and lymphocytes,which
row’s capacity to provide defense cells to protect explains, at least in part, why tubercle bacilli are
the host against infection.When the host is chal- good immunological adjuvants. This adjuvant
lenged by inhaled virulent human-type tubercle effect should be considered in developing
426 䡵 APPENDIX F

improved vaccines for the prevention of tuber- chapter 5). The antigen-antibody complexes
culosis, because vaccines producing a strong early formed at the site of reinfection evidently produced
nonspecific cytokine (chemokine) response should chemotactic factors that markedly hastened the
be more immunogenic than vaccines with simi- cell infiltration.
lar antigens producing a weak response. In general, cytokine production in tuberculin
reactions showed the same pattern as that found in
the early reinfection BCG lesions.
CHAPTER 20. CYTOKINE PRODUCTION
IN REINFECTION BCG LESIONS AND
IN TUBERCULIN REACTIONS CHAPTER 21.VASCULAR ADHESION
Reinfection BCG lesions provide a simple model MOLECULES IN TUBERCULOUS
of how tuberculosis vaccines would affect an LESIONS
exogenous infection with virulent tubercle bacilli. Vascular adhesion molecules enable host defense
Therefore, rabbit dermal primary and reinfection cells to leave the bloodstream and enter tubercu-
BCG lesions were produced and evaluated during lous lesions.After the inhalation of tubercle bacilli,
the first 5 days of their existence.Tissue sections Lurie’s resistant rabbits had a larger number of
of the lesions were prepared, and the types of cells mononuclear cells within developing lesions than
and their cytokine mRNAs and proteins were did his susceptible rabbits. Therefore, the rapid
analyzed by histochemical methods.The cytokines local accumulation of mononuclear cells seems
studied were interleukin-1, macrophage to be one of the factors associated with resis-
chemoattractant (activating) protein (MCP-1), tance to the progress of this disease.Vascular adhe-
interleukin-8, and tumor necrosis factor alpha (see sion molecules enable such an accumulation to
chapter 19). occur.
Our most informative findings were with MCP- With immunohistochemical techniques, we
1, one of the main chemokines attracting mononu- evaluated the rise and fall of three major vascular
clear cells (MN). (In tuberculous lesions, MN are adhesion molecules as rabbit dermal BCG lesions
mostly macrophages but also contain dendritic developed and healed. ICAM-1 (intercellular adhe-
cells and lymphocytes.) At 3 h, both the reinfection sion molecule 1) is important for the adherence of
lesions and the primary lesions contained the same polymorphonuclear leukocytes (PMN), mono-
percentage of MN labeled for MCP-1 mRNA. cytes, and lymphocytes to activated vascular
However, the reinfection lesions were 400 to 500 endothelium before they emigrate from the blood-
times larger and therefore contained many more of stream into sites of inflammation and infection.
these MN. This high cell number alone would VCAM-1 (vascular cell adhesion molecule 1) is a
cause the total chemokine production to exceed by major factor in monocyte, lymphocyte, and
far that occurring in the primary lesions. eosinophil emigration. ELAM-1 (endothelial-
By 1 day, the percentage of MN containing leukocyte adhesion molecule 1, now called E-
MCP-1 mRNA (and protein) had markedly selectin) aids the emigration of granulocytes (and
decreased in the reinfection lesions, but remained some monocytes and T lymphocytes).
high for at least 2 days in the primary lesions, In primary BCG lesions, ICAM and VCAM
which were beginning to increase in size. This peaked at 1 to 2 weeks and decreased as the lesions
finding suggests that chemokine production is healed. In reinfection BCG lesions, ICAM and
turned off when sufficient MN have accumu- VCAM were upregulated much sooner, beginning
lated. In other words, the local accumulation of at 3 to 12 h and peaking at 1 to 2 days.The upreg-
MN is carefully regulated, so that excessive cell ulation of these two adhesion molecules apparently
infiltration into the lesions is prevented. caused the rapid infiltration of mononuclear cells
The rapid local accumulation of MN (macro- into sites of BCG reinfection. ELAM-1 seemed to
phages, dendritic cells, and antigen-specific lym- be less involved.
phocytes) in the early reinfection BCG lesions In tuberculosis, epithelioid cells are macrophages
seemed to be due to the presence of antibodies that that adhere to one another in an epithelial-like
developed during the first BCG infection (see pattern.This adherence seems to be due in part to
APPENDIX F 䡵 427

the ICAM-1 of one macrophage’s binding to its improved by recombinant constructs that contain
ligand LFA-1 (lymphocyte function-associated major protective antigens.Also, the immunity pro-
antigen 1) (CD11a/CD18) on a neighboring vided by BCG could be increased by booster
macrophage. Whether or not this epithelial-like injections of such antigens.
pattern benefits the host remains to be determined. If vaccines more effective than BCG are ever
From these studies of vascular adhesion mole- developed, they would probably produce in the
cules, we developed a theory of why so much tis- host a higher CMI/DTH ratio, i.e., an expanded
sue destruction occurs in tuberculosis: ICAM, antigen-specific lymphocyte population capable of
VCAM, and ELAM are markers for activated vas- producing increased numbers of activated macro-
cular endothelial cells. In tuberculous lesions, such phages and decreased amounts of tissue necrosis.To
activated endothelial cells can capture and present do this, the improved vaccine would probably con-
local mycobacterial antigens and therefore may be tain increased bacillary glycolipid-protein compo-
killed by antigen-specific cytotoxic T lympho- nents and decreased tissue-damaging tuberculin-like
cytes.When the vascular endothelium is no longer protein components.The vaccine should also con-
intact, thrombosis occurs, and the local tissues (now tain components that increase the Th1/Th2 ratio.
lacking a blood supply) undergo caseous necrosis.

CHAPTER 23. CHARACTERISTICS OF


SECTION 7. TUBERCULOSIS RABBIT BCG LESIONS AND EFFICACIES
VACCINES OF BCG AND MYCOBACTERIUM
CHAPTER 22. PRINCIPLES AND MICROTI VACCINES
GUIDELINES FOR DEVELOPING In rabbits (and humans), dermal BCG lesions have
BETTER TUBERCULOSIS VACCINES the same components as lesions caused by virulent
Tuberculosis vaccines have little or no effect on the tubercle bacilli, i.e., a caseous liquefied center sur-
establishment of a microscopic pulmonary lesion rounded by tuberculous granulation tissue.There-
produced by the inhalation of a virulent tubercle fore, BCG lesions can be used as a model in which
bacillus. Such a lesion is established only when the to study the host response to this disease.
pulmonary alveolar macrophages fail to destroy the In immunocompetent hosts, BCG lesions do
inhaled bacillus. Alveolar macrophages do not not progress and always heal.The rate of healing
expand their population in response to specific of dermal BCG lesions reflected the native and
antigens.Therefore, the establishment of a micro- acquired resistance of Lurie’s susceptible and resis-
scopic pulmonary tubercle is not affected by vac- tant inbred rabbit strains. Similarly, the rate of
cination. Effective tuberculosis vaccines may, how- healing of BCG lesions has been shown to reflect
ever, stop the progression of a tiny established the resistance to tuberculosis of human popula-
lesion, because the vaccination has expanded anti- tions. However, the healing of BCG lesions in
gen-specific lymphocyte populations.These lym- individual rabbits and humans may vary consid-
phocytes enter the early lesion, where they cause erably from the mean.
a rapid local delayed hypersensitivity (DTH) and In Lurie’s natively resistant rabbits, immuniza-
cell-mediated immunity (CMI) response that often tion with BCG was quite effective in preventing
prevents progression of the disease. many grossly visible primary pulmonary tubercles
When comparing their relative efficacies, two or (i.e., clinically apparent tuberculosis). In his natively
more live vaccines should be standardized for susceptible rabbits, BCG was hardly effective at all.
equal numbers of live and dead bacilli, equal num- In other words, the rabbits that needed it the least
bers of log-phase and dormant bacilli, and equal were helped by BCG vaccination the most, and the
numbers of clumps and isolated bacilli. rabbits that needed it the most were helped the
Vaccines will probably never be 100% effective least.This principle also applies to human popu-
in preventing active tuberculosis in humans, lations: persons with poor resistance to clinical
because humans with arrested tuberculous lesions tuberculosis would develop less immunity from
are able to be reinfected with exogenous tubercle BCG administration than persons with strong
bacilli. However, the efficacy of BCG could be resistance.
428 䡵 APPENDIX F

The efficacies of various BCG and Mycobacterium rabbits prevented our rating any one vaccine above
microti (the vole bacillus) vaccines were evaluated in another. Recombinant BCG vaccines that contain
commercial outbred rabbits by the tubercle-count additional antigenic components may be more
method. Both of these vaccine types were effective. promising than currently used BCG vaccines (see
However,the variations in resistance among outbred chapter 22).
APPENDIX G
ACKNOWLEDGMENTS

I wish to acknowledge the many associates who Safety; Edward J. Bernacki, M.D., M.P.H., Execu-
have carried out our laboratory’s experiments dur- tive Director of Health Safety and Environment;
ing their two-year sojourns in my laboratory. and John A. Schaefer, M.S., Associate Director of
Their names appear as coauthors in my publica- Health Safety and Environment.The entire chap-
tion list in Appendix D. Rena Ashworth and Ilse ter was reviewed by Carlton Evans, M.D., Ph.D.,
Moeller Harrop have assisted faithfully in the Associate in the Dept. of International Health.All
preparation of this entire book. Many friends have of these reviewers are at the Johns Hopkins Med-
read and improved various chapters.Their help is ical Institutions, Baltimore, Md. Edward A. Nardell,
acknowledged below. M.D. (Associate Professor, Harvard School of Pub-
Most of the photographs in this book were lic Health, Boston, Mass.), also reviewed this entire
carefully reproduced by Jon R. Christofersen in the chapter and provided the information on his cur-
Photography Laboratory of the Dept. of Pathol- rent studies of the transmission and infectiousness
ogy, Johns Hopkins Medical Institutions, and some of tubercle bacilli aerosolized by patients with
photographs were similarly reproduced by Nor- tuberculosis.
man J. Barker, director of this laboratory.
Finally, I wish to express my deep appreciation CHAPTER 3. TYPES OF HUMAN
to the editorial staff of ASM Press for the dedicated PULMONARY TUBERCULOSIS
care that they gave to the production of this book, I am indebted to the late Walter H. Sheldon, M.D.
particularly to Eleanor Tupper, Senior Production (Professor of Pathology, Johns Hopkins School of
Editor; Mary McKenney, the copyeditor; and Jen- Medicine), and to Joseph F.Tomashefski, Jr., M.D.
nifer Adelman, Marketing Director. This book (Professor of Pathology, Case-Western Reserve
could never have been published in its present School of Medicine), for their help in preparing
form without the faith in its value of Jeff Holt- the original publication of reference 1 from which
meier (Director,ASM Press) and Ellie Tupper and most of this chapter was derived. Dr.Tomashefski
their many concessions to variations from the supplied the text and figures for the part of this
usual ASM format. chapter describing tuberculosis in immunocom-
Partial financial support for the preparation of this promised persons. Richard E. Chaisson, M.D., and
book came from the Aeras (formerly Sequella) Jacques H. Grosset, M.D., Professors of Medicine;
Global TB Vaccine Foundation; from Ellison Med- Ralph H. Hruban, M.D., Professor of Pathology;
ical Foundation; from grant HL-71554 from the Robert Frank, M.D., Professor of Physiology; and
National Institutes of Health (NIH) to William R. Noreen A. Hynes, M.D., Assistant Professor of
Bishai and Yukari C. Manabe; and from Johns Hop- Medicine—all at Johns Hopkins Medical Institu-
kins Environmental Health Science Center, Dept. tions—reviewed parts or all of this chapter.
of Environmental Health Sciences, Bloomberg
School of Public Health, Johns Hopkins University CHAPTER 4. LIQUEFACTION OF
(grant ES-03819 from the National Institute of CASEOUS FOCI AND CAVITY
Environmental Health Sciences, NIH). FORMATION
Raymond E. Lund, Norman J. Barker, Jon R.
CHAPTER 1. OVERVIEW Christofersen, and Rick M.Tracey (Johns Hopkins
The Virulence section of this chapter was reviewed Medical Institutions, Baltimore, Md.) made many
by William R. Bishai, M.D., Ph.D., Professor of of the photographs in this chapter.Walter Johnson
Medicine at the Johns Hopkins Center for Tuber- (George Washington University School of Medi-
culosis Research. The Prevention section of this cine,Washington, D.C.) administered the Riton-
chapter was reviewed by Byron S.Tepper, Ph.D., avir orally to the rabbits.Yukari C. Manabe, M.D.,
former Director of Environmental Health and Paul J. Converse, Ph.D., and Norman E. Morrison,

429
430 䡵 APPENDIX G

Ph.D. (Johns Hopkins Center for Tuberculosis School of Medicine, Baltimore, Md.). David N.
Research), reviewed this chapter and offered sev- McMurray, Ph.D. (Dept. of Microbiology,Texas A
eral important suggestions for its improvement. and M University, College Station), reviewed the
parts concerning tuberculosis in guinea pigs. Dr.
CHAPTER 5. DELAYED-TYPE Flynn also reviewed the section on tuberculosis in
HYPERSENSITIVITY, CELL-MEDIATED monkeys. Dr. Cardona kindly provided Fig. 5, 6,
IMMUNITY, AND ANTIBODIES IN 7, 8, and 9.
TUBERCULOSIS
I am indebted to Alan L. Scott, Ph.D. (Professor of CHAPTER 19. CYTOKINE PRODUCTION
Immunology, School of Public Health), and Mark IN PRIMARY BCG LESIONS
J. Soloski, Ph.D. (Professor of Immunology, School I am indebted to Pere-Joan Cardona, M.D., Ph.D.
of Medicine), both at Johns Hopkins University, (Unitat de Tuberculosi Experimental, Dept. of
for reviewing the immunology in this chapter. Microbiology, Hospital Universitari “Germans
Trias i Pujol” and Universitat Autònoma de
CHAPTER 6. MACROPHAGES Barcelona, Catalonia, Spain), and to JoAnne L.
AND OTHER CELLS IN Flynn, Ph.D. (Dept. of Molecular Genetics and
TUBERCULOUS LESIONS Biochemistry, University of Pittsburgh School of
I am indebted to Alan L. Scott, Ph.D. (Professor of Medicine, Pittsburgh, Pa.), for reviewing this chap-
Immunology, School of Public Health), and Mark ter, especially the parts concerning tuberculosis
J. Soloski, Ph.D. (Professor of Immunology, School in mice.
of Medicine), both at Johns Hopkins University,
for reviewing the immunology in this chapter, CHAPTER 22. PRINCIPLES AND
and also to Yukari C. Manabe, M.D. (Assistant GUIDELINES FOR DEVELOPING
Professor, Division of Infectious Diseases, Johns BETTER TUBERCULOSIS VACCINES
Hopkins University School of Medicine), for her Jacques H. Grosset, M.D.,William R. Bishai, M.D.,
contributions to some sections of this chapter. Richard E. Chaisson, M.D., and Carlton Evans,
M.D., reviewed parts of this chapter and provided
CHAPTER 11. LURIE’S PULMONARY helpful suggestions.
TUBERCLE-COUNT METHOD
Edward A. Nardell, M.D. (Associate Professor, Har- CHAPTER 25. SUGGESTED FUTURE
vard School of Public Health, Boston, Mass.), pro- RESEARCH AND UNANSWERED
vided the information on current studies of the QUESTIONS
transmission and infectiousness of tubercle bacilli Mark A. Chambers, Ph.D. (Veterinary Laboratories
from patients with active tuberculosis. Agency,Weybridge, United Kingdom), suggested
that this chapter be written. He also suggested
that anticoagulants might decrease the amount of
CHAPTER 15. COMPARISONS OF
TUBERCULOSIS IN RABBITS, MICE, caseous necrosis in tuberculous lesions.William R.
AND GUINEA PIGS Bishai, M.D., Ph.D., Jacques H. Grosset, M.D.,
For reviewing the parts of this chapter concern- Eric L. Nuermberger, M.D., and Ying Zhang,
ing tuberculosis in mice, I am indebted to Pere- M.D., Ph.D. (all at Johns Hopkins Center for
Joan Cardona, M.D., Ph.D. (Unitat de Tuberculosi Tuberculosis Research), reviewed this chapter.
Experimental, Dept. of Microbiology, Hospital
Universitari “Germans Trias i Pujol” and Univer- GLOSSARY
sitat Autònoma de Barcelona, Catalonia, Spain), to I am indebted to Alan L. Scott, Ph.D., Professor of
Robert J. North, Ph.D. (Trudeau Institute, Saranac Immunology in the Johns Hopkins Bloomberg
Lake, N.Y.), to JoAnne L. Flynn, Ph.D. (Dept. of School of Public Health, and Mark J. Soloski,
Molecular Genetics and Biochemistry, University Ph.D., Professor of Immunology in the Johns
of Pittsburgh School of Medicine, Pittsburgh, Pa.), Hopkins School of Medicine, for reviewing the
and to Jacques H. Grosset, M.D. (Johns Hopkins immunology in this glossary.
GLOSSARY

This glossary includes definitions and abbreviations antigens are located, these lymphocytes produce
and is arranged in two parts: first, a listing of the cytokines that activate macrophages so that they
immunology terms most frequently used in this can inhibit or destroy ingested tubercle bacilli.
book, and then an alphabetical listing of the Delayed-type hypersensitivity (DTH). In
remaining terms. For details on cytokines, the field of tuberculosis, the terms DTH and tuber-
chemokines, and adhesion molecules, see refer- culin sensitivity are used interchangeably. DTH
ences 1, 2, and 3, respectively. Some of the defin- gets its name because the skin reaction (to tuber-
itions are from references 4 and 5. culin) usually peaks in 1 to 3 days rather than in the
minutes or hours that characterize the more imme-
PART I. FREQUENTLY USED diate hypersensitivities. Like CMI, DTH is char-
IMMUNOLOGY TERMS
acterized by an expanded population of recircu-
Acquired cellular resistance (ACR). ACR lating antigen-specific Th1 lymphocytes. The
occurs locally in tuberculous lesions. It is charac- difference from CMI is the amount of antigen that pro-
terized by a population of macrophages that have duces each reaction. In a highly tuberculin-positive
been activated by cytokines from nearby antigen- person, a positive skin test can be elicited with an
specific Th1 lymphocytes. Such activated macro- intradermal injection of 1 tuberculin unit of puri-
phages can inhibit and/or destroy ingested tuber- fied protein derivative, i.e., 0.1 ml containing 0.02
cle bacilli.Cell-mediated immunity (CMI) produces g (see “PPD”). If greater concentrations are
local acquired cellular resistance (ACR), and both injected intradermally into such a person, necrosis
terms are frequently used interchangeably. However, may develop at the site of injection. In immunized
we prefer to use ACR for macrophages that have hosts, antigens eliciting CMI usually require a
been already activated in tuberculous lesions, and higher concentration than antigens eliciting DTH.
to use CMI for an expanded antigen-specific Innate and adaptive (acquired) immunity.
T-lymphocyte population that is able to activate Innate immunity is present before the host is
macrophages if the antigen is present, whether or infected by the tubercle bacillus. The host will
not it actually does so. recognize the foreignness of an invader, but will not
Antigen-presenting cells (APCs). Dendritic recognize its exact nature. For example, the
cells (DCs) (see chapter 6) are the most effective lipopolysaccharides (endotoxins) of many differ-
antigen-presenting cells, but macrophages and B ent bacteria are recognized as foreign by receptors
lymphocytes can also present antigens. DCs are on the host’s phagocytes.Adaptive immunity to the
present both in the tissues and in the circulation. tubercle bacillus is specific for its antigens. It
Tissue DCs migrate from sites of infection to the involves the clonal expansion of lymphocytes that
draining lymph nodes, where they present antigens have specific receptors for these antigens. Lurie and
to lymphocytes that continuously circulate from I use the term “acquired immunity” for what is
the blood to lymph nodes and back to the blood now called adaptive immunity, and the term
(via efferent lymphatics). In the lymph nodes “native resistance” for what is now called innate
draining tuberculous lesions, antigen-specific lym- immunity.
phocytes clonally expand their population, enter Lymphocytes (T cells and B cells). T cells
the bloodstream, and then enter tuberculous and B cells provide immunologic specificity to the
lesions. Once there, these antigen-specific lym- host defense against tubercle bacilli.
phocytes produce macrophage-activating cytokines T cells originate in the bone marrow and enter
and continue to proliferate. the thymus, where they develop antigen-specific
Cell-mediated immunity (CMI). CMI in receptors (by DNA rearrangements). They then
tuberculosis is characterized by an expanded pop- enter the bloodstream and recirculate in and out of
ulation of recirculating antigen-specific Th1 lym- peripheral lymphoid tissues, including the spleen.
phocytes. In sites where tubercle bacilli and their T cells are the immunocytes responsible for the

431
432 䡵 GLOSSARY

cell-mediated immunity and delayed-type hyper- not grow. It kills macrophages and the surrounding
sensitivity found in tuberculosis.Antigen-activated tissues, because multiplying intracellular tubercle
T cells produce cytokines that activate nearby bacilli produce high local concentrations of
macrophages. T cells have been subdivided in a tuberculin-like products. In brief, tissue-damaging
variety of ways based on (i) their surface markers DTH is a fail-safe immune response to stop intra-
(CD4 and CD8 T cells), (ii) their antigenic recep- cellular bacillary growth when CMI cannot do so.
tors (/, /, and CD1), (iii) the cytokines they DTH carries out this function at the expense of host
produce (Th1 and Th2 cells), and (iv) their func- tissues, and is the cause of almost all of the tissue
tions (helper, regulatory, and cytotoxic T cells). damage found in tuberculosis.Tubercle bacilli do not
Th1 cells produce gamma interferon and other usually damage host tissues before DTH develops.
cytokines that activate macrophages, so that these Tissue-damaging DTH also damages vascular
macrophages can inhibit the growth of the tuber- endothelial cells, and the resulting thrombosis causes
cle bacilli that they ingest.Th2 cells produce inter- much of the tissue necrosis found in humans, rab-
leukin 4 (IL-4), IL-5, and other cytokines that bits, and guinea pigs with tuberculosis.
activate B lymphocytes (B cells) to produce anti-
body. PART II. ALPHABETICAL LISTINGS
B cells also originate in the bone marrow of
mammals and recirculate in a manner similar to Acquired cellular resistance (ACR). See
that of T cells. Antigen-activated B cells produce definition in part I.
antibodies, especially when they terminally dif- Acute-phase reaction. An early system host
ferentiate into plasma cells.Antibodies enhance the response to infection in which the liver increases
cell-mediated immune response in a variety of or decreases several proteins in the blood. These
ways (see chapter 5). changes aid the host in combating the infection.
Macrophages. Macrophages are the effector The acute-phase reaction is mediated by IL-1,
cells of the mononuclear phagocyte system.They IL-6, tumor necrosis factor alpha (TNF-), and
are produced in the bone marrow, circulate as probably other cytokines. (See references 4 and 6).
monocytes in the bloodstream, and are called Antibody-dependent cell-mediated cyto-
macrophages after they emigrate from the blood toxicity (ADCC). The killing of cells (that
into the tissues. Virulent tubercle bacilli readily bind antibody) by cells recognizing that antibody’s
multiply in nonactivated macrophages, whereas Fc region, especially natural killer (NK) cells.
they are inhibited (and often destroyed) in highly Antigen-presenting cells (APC). See defin-
activated macrophages. ition in part I.
Pulmonary alveolar macrophages (AM). Apoptosis (programmed cell death). An
Pulmonary alveolar macrophages possess high lev- internal cell death program whereby cells eliminate
els of innate immunity, because most are strongly themselves without causing an inflammatory
activated nonspecifically by a variety of inhaled par- response.Apoptosis removes cells that are no longer
ticles that the AM ingest and digest.AM are usu- needed during embryological development and
ally capable of destroying numerous inhaled viru- repair processes. It also keeps the immune responses
lent tubercle bacilli. In humans and rabbits, only a from becoming excessive. In apoptosis, a series of
rare alveolar macrophage—probably one that has caspases (cysteine-aspartic acid proteases) are acti-
recently migrated there from the bloodstream—is vated, DNA is degraded and condensed, and the
so poorly activated it will allow an ingested human- cell residue is phagocytized by macrophages. (See
type tubercle bacillus to multiply and establish an reference 6).
infection (thereby converting the tuberculin skin In necrosis, cells are killed by external (not inter-
test). Human-type tubercle bacilli do not cause nal) forces (e.g., microorganisms and their products,
progressive disease in most humans and rabbits. trauma, or thrombosis of their blood supply), and
Tissue-damaging DTH. Tissue-damaging an inflammatory response occurs. During necrosis,
DTH kills macrophages in which more than a few some cells may die from apoptosis.Therefore, these
tubercle bacilli are multiplying, thereby causing two types of cell death can exist together. (See
solid caseous necrosis in which tubercle bacilli can- reference 6).
GLOSSARY 䡵 433

Arthus reaction. An inflammatory reaction such as tubercle bacilli. Most CTLs are major his-
commonly produced by injecting an antigen intra- tocompatibility complex 1-restricted CD8 T cells,
dermally into a host that has circulating antigen- but some are CD4 cells.
specific (IgG) antibodies. The resulting antigen- Delayed-type hypersensitivity (DTH). See
antibody reaction activates complement, which definition in part I.
causes a slowly developing cell infiltration that Dendritic cells (DC). See “Antigen-present-
usually peaks in 1 day. In contrast, DTH reactions ing cells” in part I.
are caused by antigen-specific T lymphocytes. Endothelial-leukocyte adhesion molecule
However, because antibodies usually circulate in (ELAM). ELAM,or E-selectin,enables polymor-
tuberculin-positive individuals, the typical skin phonuclear leukocytes (and some monocytes) to
tests produced by tuberculin almost always have an emigrate from the microvasculature. (See refer-
Arthus component. ence 3).
B cells, B lymphocytes. See “Lymphocytes (T Epithelioid cells. Large macrophages within
cells and B cells)” in part I. tuberculous lesions in various states of activation (⫹
B7.1 and B7.2. Cell surface molecules on to ⫹⫹⫹⫹). They received their name because
antigen-presenting cells that costimulate antigen- they frequently adhere to one another to form
specific T cells. sheets that resemble epithelium. In rabbits, mature
BCG. Bacille Calmette-Guérin, the live atten- epithelioid cells have rounded borders with rather
uated strain of Mycobacterium bovis used world- homogeneous cytoplasm caused by finely dispersed
wide as a vaccine for tuberculosis. lipid material. Immature epithelioid cells have irreg-
C5a. A chemotactic peptide released when the ular borders with a vacuolated cytoplasm.
fifth component of complement is activated. Fas and Fas ligand. Fas is a receptor on cer-
C-C and C-X-C chemokines. See “Inter- tain cells that makes such cells undergo apoptosis
leukin 8.” (programmed cell death) when Fas ligand on a
CD (cluster of differentiation). CD numbers “killer” cell combines with this receptor. Fas is a
are the names given to cell surface markers that member of the same receptor family as the recep-
have been identified by specific monoclonal or tor for TNF, and Fas ligand is a member of the
polyclonal antibodies. Over 200 CD antigens have TNF family itself.
so far been identified, and many are listed in ref- Gamma interferon (IFN-). IFN- is a major
erences 1 and 7. Most helper T lymphocytes are macrophage-activating cytokine produced by T
CD4 positive, and most cytotoxic T lymphocytes lymphocytes and natural killer cells. IFN- inhibits
are CD8 positive (see chapter 6). the multiplication of tubercle bacilli within
Cell-mediated immunity (CMI). See defin- macrophages.All interferons enable cells to inhibit
ition in part I. viral replication.
Chemokines. Chemotactic cytokines that Gamma interferon-inducible protein 10 (IP-
attract various types of leukocytes into sites of 10). IP-10 is a C-X-C chemokine (see “Inter-
inflammation. See “Interleukin 8” and “Mono- leukin 8”) that attracts monocytes and NK cells.
cyte chemoattractant (activating) protein 1.” (See Glycol methacrylate (GMA). Tissues embed-
references 2 and 4). ded in polymerized GMA can be cut at 1 to 2 µm.
Colony-forming units (CFU). CFU arise Heat shock protein 65 (Hsp 65). A consti-
from bacteria deposited on solid culture medium. tutive antigen released from dead tubercle bacilli
Cytokines. Locally produced proteins that affect and other microorganisms. Heat shock proteins
the behavior of cells in their vicinity. Some enable microorganisms and cells of higher organ-
cytokines (e.g., IL-1, IL-6, and TNF-) enter the isms to survive adverse environmental conditions.
bloodstream and cause an acute-phase reaction, HEPA filters. High-efficiency particle air
including fever. (See reference 6). (HEPA) filters will not let airborne bacteria pass
Cytotoxic T lymphocytes (CTLs). CTLs through.
can kill other cells, especially those containing HIV/AIDS. Human immunodeficiency virus/
viruses and other intracellular microorganisms, acquired immunodeficiency syndrome.
434 䡵 GLOSSARY

[3H]TdR. Tritiated thymidine. Koch phenomenon. Classically, the Koch


IFN-. See “Gamma interferon.” phenomenon is the local necrotic skin reaction
produced in a tuberculous guinea pig by the intra-
Immunoglobulin G (IgG). IgG is the most
dermal injection of tubercle bacilli. Details are pre-
abundant class of circulating immunoglobulins
sented in chapter 15.
(antibodies).
Inducible nitric oxide synthase (iNOS). Macrophage inflammatory protein 1 (MIP-
iNOS is upregulated as macrophages become acti- 1). MIPs are chemokines attracting mono-
vated.This synthase produces nitric oxide (NO), cytes and T lymphocytes.
which in turn forms other reactive nitrogen inter- Major histocompatibility complex (MHC).
mediates. Reactive nitrogen intermediates and A set of genes encoding glycoproteins in cell
reactive oxygen intermediates (ROIs) enable membranes called MHC molecules (4). These
macrophages to kill intracellular tubercle bacilli. molecules are major histocompatibility antigens
Intercellular adhesion molecules (ICAMs). that enable the host to distinguish self from non-
ICAMS of vascular endothelium bind to inte- self tissues, and they are involved in antigen pro-
grins on leukocytes (e.g., the leukocyte functional cessing and host defense functions. MHC class I
antigen 1 [LFA-1]). This binding enables leuko- molecules present peptides generated in the cytosol
cytes to leave the circulation and enter sites of to CD8 T cells, and MHC class II molecules pre-
inflammation. ICAMs also play a role in all cell- sent peptides degraded in intracellular vesicles to
cell interactions, including the binding of lym- CD4 T cells (4).
phocytes to dendritic cells. (See reference 3). Miliary tubercles. Tubercles of hematogenous
Interleukin 1-beta (IL-1). IL-1 is a primary origin, often 1 to 2 mm in diameter, or about the
cytokine that upregulates the production of other size of a millet seed.
cytokines. Monocyte chemoattractant (activating) pro-
Interleukin 2 (IL-2). IL-2 causes antigen- tein 1 (MCP-1). MCP-1 is a C-C chemokine
specific T lymphocytes to clonally expand their (see “Interleukin 8”). It attracts into sites of inflam-
population in tuberculous lesions. It is produced mation monocytes/macrophages,T lymphocytes,
by the T lymphocytes in this population. dendritic cells, and NK cells, as well as eosinophils,
Interleukin 8 (IL-8). A C-X-C chemokine basophils, and mast cells.
(same as neutrophil attractant-activating protein 1). Mononuclear cells (MN). Histological term
C ⫽ cysteine; X ⫽ any single amino acid that sep- for a cell infiltrate containing macrophages, den-
arates the two cysteines. IL-8 attracts all granulo- dritic cells, and medium to large lymphocytes, in
cytes and a subpopulation of lymphocytes into which each cell type is not specifically identified
sites of inflammation. by immunohistochemical methods. In general, the
Interleukin 10 (IL-10). IL-10 is an inhibitor MN population of tuberculous lesions consists
of the Th1 cytokine response and an inducer of mostly of macrophages, a substantial proportion of
the Th2 cytokine response. It is produced by lymphocytes, and some dendritic cells.We did not
Th2 lymphocytes and other cells (including include small lymphocytes as members of the
macrophages). IL-10 suppresses macrophage mononuclear cell population that we counted in
functions. tuberculous lesions, because their size enabled us
Interleukin 12 (IL-12). IL-12 is the major to distinguish them from macrophages. In this
cytokine that induces natural killer cells and lym- book, I often use the term macrophages rather than
phocytes to produce IFN-. Macrophages that mononuclear cells to describe the cell population
have ingested tubercle bacilli produce large in tuberculous lesions, which is only precise when
amounts of IL-12. Dendritic cells exposed to bacil- we have identified the macrophages histochemi-
lary antigens also do so. cally by their -galactosidase (or acid phosphatase)
Knockout mice. Mice in which portions of content.The percentage of the macrophage pop-
their DNA have been removed or inactivated. For ulation in the lesions that are really dendritic cells
example, a TNF- knockout mouse does not pro- is not known. Both cell types have the same ori-
duce the cytokine TNF-, and a CD8 knockout gin and each may possibly convert to the other
mouse does not produce CD8 lymphocytes. type.
GLOSSARY 䡵 435

Natural killer (NK) cells. NK cells (both local [0.1 g]) is commonly used in the United States.
and circulating) are large, granular, non-T, non-B The worldwide standard today is PPD-RT23,
lymphocytes.They are an important early defense produced by the Statens Serum Institut in Copen-
against intracellular microorganisms (viruses, bac- hagen, Denmark. PPD-RT23 is used as 2 TU
teria, fungi, and protozoa). In tuberculosis, NK (0.04 g) for first strength and 10 TU (0.20 g)
cells can kill bacilli-laden macrophages and can for second strength.
produce IFN-, which activates macrophages and The dose of PPD for laboratory animals has not
stimulates a Th1-cytokine immune response. been established. P.-J. Cardona’s laboratory uses
Neutrophil attractant-activating protein 1 0.04 g of PPD-RT23 (2 TU) in humans, 0.50 g
(NAP-1). Same as IL-8. (25 TU) in guinea pigs, and 5.0 g (250 TU) in
Old Tuberculin. An unfractionated filtrate of mice. D. N. McMurray’s laboratory uses 100 TU
autoclaved Mycobacterium tuberculosis cultures. It of PPD-RT23 in rabbits and 50 TU of PPD-
contains more antigens than PPD does. RT23 in guinea pigs. (The PPD from Mycos
Research LLC, Loveland, Calif., is used in similar
Opsonins. Molecules, such as antibodies, the
concentrations.) Lurie used Old Tuberculin, diluted
C3b component of complement, mannose-binding
1:10 in rabbits, and diluted 1:20 in guinea pigs. Our
protein, and fibronectin, that attach to the surface
laboratory found that 250 TU of PPD-S gave a
of microorganisms so that they can be phagocytized
skin test reaction in rabbits comparable to the
(usually by granulocytes and macrophages).
1:10 dilution of Old Tuberculin.
PCR (polymerase chain reaction). PCR is The above concentrations can only serve as
a procedure that amplifies a specific sequence in guidelines, because the size of the dermal tuber-
DNA by repeated cycles of synthesis driven by culin reaction does not decrease in direct propor-
pairs of reciprocally oriented primers. (See tion to the tuberculin concentration. In BCG-
“RT-PCR,” below.) vaccinated rabbits, the size of the tuberculin
Peripheral blood mononuclear cells (PBMC). reactions produced by a 1:20 dilution of Old
Mostly lymphocytes, but they include mono- Tuberculin was only slightly smaller than the size
cytes/macrophages, dendritic cells, and NK cells. of those produced by a 1:10 dilution, not half the
Phosphate-specific transport protein 1 (PstS- size.
1). An antigen secreted from live tubercle bacilli. Pulmonary alveolar macrophages (AM).
Plasma cells. Antibody-producing cells of B- See definition in Part I.
lymphocyte origin. See “Lymphocytes (T cells RANTES (regulated upon activation, nor-
and B cells)” in part I. mal T cell expressed and secreted). A
Plethismograph. The apparatus that we used chemokine attracting monocytes, T cells, den-
to measure the volume of air breathed by a given dritic cells, natural killer cells, eosinophils, and
rabbit.This measurement enabled us to calculate basophils.
the volume of the aerosol of tubercle bacilli sub-
Reactive oxygen intermediates (ROIs) and
sequently breathed by this rabbit (8).
reactive nitrogen intermediates (RNIs).
PPD (purified protein derivative). PPD is ROIs and RNIs are produced by activated
the purified protein derivative of M. tuberculosis macrophages. These reactive intermediates enable
used for intradermal tuberculin testing. It con- macrophages to kill (or inhibit) the tubercle bacilli
tains several of the many mycobacterial antigens. within them.
The original PPD standard (PPD-S) was pro-
duced by Florence B. Seibert of the Henry Phipps Regulatory (suppressor) T lymphocytes (Treg).
Institute of the University of Pennsylvania (9). Treg produce cytokines and probably other factors
First-strength PPD-S is 1 TU (tuberculin unit), an that keep the T-cell immune response from becom-
injection of 0.02 g. It is used when the patient ing excessive. They also prevent autoimmune
is expected to have a positive reaction. Second- diseases.
strength PPD-S is 250 TU (5.0 g). It can be Reverse transcription-polymerase chain
used in persons testing negative with the first reaction (RT-PCR). Reverse transcription
strength. As a general test for human tuberculin enables mRNA to form the complementary
positivity, intermediate-strength PPD-S (5 TU DNA. Then, DNA polymerase is used with
436 䡵 GLOSSARY

specific primers in a series of heating and cool- organs of the tuberculous host.Tubercles range in
ing steps to make multiple copies of comple- size from microscopic to almost a centimeter and
mentary sense and antisense DNAs. RT-PCR is may or may not contain caseous centers.
used to identify genes from the mRNA that such Tumor necrosis factor alpha (TNF-). A
genes produce. In time, the genes of tubercle primary cytokine,like IL-1,that upregulates the pro-
bacilli that enable (i) their multiplication in non- duction of other cytokines.TNF- and IFN- are
activated macrophages, (ii) their dormancy in major macrophage activators.TNF- is the defin-
solid caseous tissue, and (iii) their multiplication ing member of the TNF family of cytokines (see
in liquefied caseum might be identified by this “Fas and Fas ligand”). It is both cell associated and
method. Real-time PCR is a more quantitative secreted.
version of the polymerase chain reaction, because von Willebrand factor (vW factor). vW fac-
it monitors the time course of the DNA accu- tor is stored in vascular endothelial cells and
mulation. released when they are injured. Then, vW factor
Shwartzman phenomenon. Produced by binds to the exposed vascular basement mem-
endotoxins, i.e., bacterial lipopolysaccharides (LPS), brane, where it helps platelets stick and initiate the
either locally in the skin or systemically in many clotting cascade.
organs. It occurs only after a second injection of Vascular cell adhesion molecule 1 (VCAM-1).
endotoxin (usually at 24 h), but not after the first A member of the immunoglobulin supergene fam-
injection.The systemic reaction is characterized by ily. It helps monocytes/macrophages, lymphocytes,
intravascular thrombosis that may cause ischemic and eosinophils emigrate from the bloodstream
necrosis (6, 10). The first LPS injection was evi- into sites of inflammation.
dently detoxified well, but the second injection was
Vascular endothelial growth factor (VEGF).
not detoxified, apparently because the detoxifica-
A cytokine, produced by monocytes/macrophages,
tion mechanisms were exhausted by the first LPS
keratinocytes, and other cells, that stimulates vas-
injection (6, 11, 12). The second LPS injection
cularization.
probably activates the clotting cascade, so that
thrombosis occurs. Other possibilities have also
been described (6, 10, 11). Mycobacterial tuber- REFERENCES
culin-like antigens have an endotoxin-like effect 1. Callard, R. E., and A. J. H. Gearing. 1994. The
in hosts with active tuberculosis (13).Therefore, the Cytokine FactsBook. Academic Press, San Diego,
Shwartzman phenomenon may cause some of the Calif.
2. Vaddi, K., M. Keller, and R. C. Newton. 1997.
thrombosis produced by tissue-damaging DTH in
The Chemokine FactsBook. Academic Press, San
this disease. Diego, Calif.
T cells (Th1 and Th2). T lymphocytes [see 3. Pigott, R., and C. Power. 1993. The Adhesion
“Lymphocytes (T cells and B cells)” in part I].Th1 Molecule FactsBook. Academic Press, San Diego,
cells are T-helper lymphocytes that produce IFN- Calif.
 and other cytokines that activate macrophages. 4. Janeway, C. A., Jr., P.Travers, M.Walport, and
M. J. Shlomchik. 2001. Immunobiology: the Immune
Th2 cells are T-helper lymphocytes that produce System in Health and Disease, 5th ed. Garland Pub-
IL-4, IL-5, and other cytokines that activate B lishing, New York, N.Y.
lymphocytes (B cells) to produce antibody. 5. Herbert,W. J., P. C.Wilkinson, and D. I. Stott.
T-cell immunoglobulin mucin gene (TIM) 1995. The Dictionary of Immunology, 4th ed. Acad-
family. TIM proteins are expressed by T cells emic Press, San Diego, Calif.
6. Majno, G., and I. Joris. 2004. Cells,Tissues, and
and regulate their function. Disease. Principles of General Pathology, 2nd ed. Oxford
Transforming growth factor beta (TGF-). University Press, New York, N.Y.
A cytokine that is a major downregulator of 7. Barclay, A. N., M. H. Brown, S. K. A. Law, A.
inflammatory and immune processes. It is also a J. McKnight, M. G. Tomlinson, and P. A. van
major cytokine stimulating fibroblasts to produce der Merwe. 1997. The Leucocyte Antigen FactsBook,
2nd ed.Academic Press, San Diego, Calif.
connective tissue for repair and healing. 8. Converse, P. J., A. M. Dannenberg, Jr., J. E.
Tubercles. Accumulations of lymphocytes and Estep, K. Sugisaki,Y.Abe, B. H. Schofield, and
macrophages (along with some other cell types) in M. L. M. Pitt. 1996. Cavitary tuberculosis pro-
GLOSSARY 䡵 437

duced in rabbits by aerosolized virulent tubercle 12. Good, R. A., and L. Thomas. 1952. Studies on
bacilli. Infect. Immun. 64:4776–4787. the generalized Shwartzman reaction. II.The pro-
9. Seibert, F. B. 1950. Progress in the chemistry of duction of bilateral cortical necrosis of the kidneys
tuberculin, p. 1–29. In Advances in Tuberculosis by a single injection of bacterial toxin in rabbits pre-
Research, vol. 3. S. Karger, New York, N.Y. viously treated with thorotrast or trypan blue. J. Exp.
10. Stetson, C. A., Jr. 1951. Similarities in the mech-
Med. 96:625–641.
anisms determining the Arthus and Shwartzman
phenomena. J. Exp. Med. 94:347–358. 13. Stetson, C. A., Jr. 1955. Studies on the mecha-
11. Thomas, L., and R. A. Good. 1952. Studies on nism of the Shwartzman phenomenon. Similari-
the generalized Shwartzman reaction. I. General ties between the reactions to endotoxins and cer-
observations concerning the phenomenon. J. Exp. tain reactions of bacterial allergy. J. Exp. Med. 101:
Med. 96:605–624. 421–436.
This page intentionally left blank
INDEX

A hemoptysis, 80, 81
Abstracts, of text chapters,Appendix F, 419–428 lymphocytes and plasma cells, 82–83, 88
Acknowledgments,Appendix G, 429–430 metaplastic alveolar epithelium and chemotaxis
Acquired cellular resistance (ACR) of alveolar macrophages, 83–84, 88, 89
definition, 431 number of bacilli, 81, 83, 84, 85
duration and specificity, 102 AIDS/HIV, 59–60, 433
overview, 102 Airborne infections, natural, 215–227
recall upon reinfection, 111–112 Lurie’s experiments, 215–224
ACTH (adrenocorticotropic hormone), 280–282 resistance to establishment of lesions by bovine-
Acute infections, general characteristics of, 371 type bacilli, 216–218
Acute inflammatory lesions, vascular adhesion mole- resistance to establishment of lesions by human-
cules in, 331 type bacilli, 223–224
Acute-phase reaction, 432 resistance to progress of pulmonary tuberculosis
Adaptive immunity by bovine-type bacilli, 218–223
definition, 431 resistance to progress of pulmonary tuberculosis
innate immunity relation to, 112–114 by human-type bacilli, 224
nonspecific and antigen-specific immune responses Riley’s experiments, 224–226
in, 114 Alveolar macrophages
ADCC (antibody-dependent cell-mediated cytotoxi- activation, 54–55
city), 432 chemotaxis, 83–84
Adjuvant, vaccine, 345–346 definition, 432
Adrenocorticotropic hormone (ACTH), 280–282 division of activated in early lesions in rabbits, 175
Adult-type tuberculosis, 11, 12, 13, 14, 15, 51–52 ingestion of tubercle bacilli, 16, 22–23, 36
AED (androstenediol), 282 innate immunity and, 98, 114
Aerosolized infection, see also Inhalation of bacilli location in lesion, 37
bacillary titers in stationary stage in rabbits, mice, logarithmic growth of bacilli within nonactivated,
and guinea pigs, 261–262 24–25
fate of BCG in rabbits, mice, and guinea pigs, 262 microbicidal power of, 16
histopathology of liquefaction of caseous foci and role in establishment of grossly visible primary
cavity formation by virulent M. bovis in rab- tubercles, 200
bits, 79–84 susceptibility versus resistance, 37–38
cavity formation, 78, 79–80 vaccine effects, 342
epithelioid cells, 80–81 X-irradiation, effects of, 293–296
fibroblasts, 82, 87, 88 Androstenediol (AED), 282
granulated macrophages, 80, 82 Angiogenesis, regulators of, 167

439
440 䡵 INDEX

Antibody macrophages
enhancement of local DTH and CMI reactions, activation, 127–128
106–107, 108, 110 enzymes released extracellularly, 128–129
host resistance to reinfection, role in, 378 heterogeneity in lesions, 129, 131, 133
role, as elucidated by Lurie’s eye chamber experi- turnover, activation, and division in healing der-
ments, 107, 109, 111 mal lesions, 189, 191
Antibody-dependent cell-mediated cytotoxicity in vitro division of activated in BCG lesions, 179
(ADCC), 432 in vitro division of cells containing bacilli in
Antigen-presenting cells (APCs), 99, 113–114, 121– BCG lesions, 179
123; see also Dendritic cells in vivo [3H]TdR labeling, 179, 180
definition, 431 microvasculature of lesions
published reviews on presentation, 123 histopathology, 163–166
T cell activation, 121–122 production of developing and healing lesions,
Antimicrobials, resistance to, 31, 43, 57–59 161–162
Apoptosis, 142–143, 193, 263, 432 mononuclear cells
Arrested lesions, 28, 37, 38, 60–62 activation in tuberculin reactions and in lesions
CMI and DTH required for, 101–102 caused by nonspecific irritants, 192–193
Arthus reaction, 432–433 in blood, labeled, 185–186
Atelectasis, 44 disappearance from lesions, 182–185
Azathioprine, cavity prevention by, 66 division rate within lesions, 184–185
entry into lesions, 180, 182
B rates of activation in primary and reinfection
B cells, 433 lesions, 187, 189
BAL (bronchoalveolar lavage), 296 pulmonary lesions in rabbits, production by intra-
BALT (bronchial-associated lymphoid tissue), 343 venous injection, 170–175
Bar Harbor rabbits, 241–242 rabbit response to inhaled bacilli, 233–234
Basophils, 144 vaccination
B7.1/B7.2, 433 combination vaccines, 349
BCG, see also Mycobacterium bovis BCG inapparent lesions and, 61–62
cytokines in primary lesions, 301–309 in mice and guinea pigs, 343–344
cell types containing MCP-1, IL-1, IL-8, and in monkeys, 258, 260
TNF- mRNAs, 304 nonspecific irritants, effect on, 193
comparison among cytokines, 306–307 recombinant technology improved, 349
cytokine mRNA in cells within tissue sections, strong CMI and weak DTH, 344–345
301–304 systemic nature in humans, 360–361
IFN- mRNA identified by RT-PCR, 305 vascular adhesion molecules, 327–336
immunohistochemical studies, 305 activation of microvascular endothelium and
overview, 301–304 caseous necrosis, 334–335
cytokines in reinfection lesions, 312–323 in acute inflammatory lesions, 331
lesion size, ulceration, and healing, 313 in epithelioid cells, 333
local cell infiltration, 313 functions, 333–334
mononuclear cells and granulocytes in lesions, identification of microvessels in tissue sections of
315–319 BCG lesions, 329
number of tubercle bacilli in, 319 leukocyte ligands in BCG lesions, 333
definition, 433 overview in, 328–329
dermal lesions in rabbits in primary BCG lesions, 329–330
BCG preparations for, 354–355 quantitation in tissue sections, 329
development and healing, 355–356 questions to be answered, 335–336
healing as measure of host resistance, 359 in reinfection BCG lesions, 331, 332
histopathology, 356–357 virulence, 13, 14
number of bacilli in lesions, 357–358 X-irradiation effects on dermal BCG lesions,
pulmonary lesions compared, 358–359 293
fate of aerosolized BCG in rabbits, mice, and Blood supply
guinea pigs, 262 activation of microvascular endothelium and
intradermal injection to produce caseous necrosis caseous necrosis, 334–335
and liquefaction, 68 angiogenesis, regulators of, 167
intravenous in rabbits, Lurie’s experiments with, capillary density, determined by gelatin-colloidal
359–360 carbon perfusion, 162–163
INDEX 䡵 441

microvascular patency, in mouse pulmonary lesions, granulated macrophages, 80, 82


254 hemoptysis, 80, 81
microvessels in tissue sections of BCG lesions, 329 lymphocytes and plasma cells, 82–83, 88
pathophysiology of, 167–168 metaplastic alveolar epithelium and chemotaxis
vascular thrombosis role in causing caseous necro- of alveolar macrophages, 83–84, 88, 89
sis, 375–376 number of bacilli, 81, 83, 84, 85
Bronchial spread hydrolytic enzymes, role of, 66
adult-type tuberculosis, 11, 14 literature review, 66–71
cavity formation and liquefaction, 29, 30–31, bronchoscope production of cavities in rabbits,
42–46 68
contagiousness, 11–12 dermal BCG lesions in pilot studies on caseation
Bronchial-associated lymphoid tissue (BALT), 343 and liquefaction, 68
Bronchoalveolar lavage (BAL), 296 distinction between caseation and liquefaction,
Burkholderia pseudomallei, 226–227 67–68
effects of large numbers of bacilli on liquefac-
C tion, 68, 71
C5a, 24, 433 role of delayed-type hypersensitivity, 66
Calcification of nodules, 37, 38, 45 role of hydrolytic enzymes, 66–67
Capillary density, determined by gelatin-colloidal measuring factors affecting, proposed method for,
carbon perfusion, 162–163 90–91
Caseous necrosis in mice, 263
activation of microvascular endothelium, 334–335 in monkeys, 263
advanced fibrocaseous tuberculosis, 46–47 pathogenesis, 29, 30–31, 42–46, 157–158
caseous tissue as structural component of lesions, in rabbits, 263
155–156 recent experiments attempting to reduce, 84–87
causes Mycobacterium vaccae, studies with, 84, 86
table of, 156 ritonavir, studies with, 86–87
vascular thrombosis, role of, 375–376 research, suggested future, 375–376
cavity formation, 65–91 summary, 87, 90–91
healed lesions, 45, 158 tuberculin sensitivity in rabbits and, 78–79
liquefaction, 29, 30–31, 42–46, 65–91 C-C chemokines, 433
progressive lesions, 46 CD4, 138, 139, 140, 319, 356
radiography of encapsulated nodules, 38 CD8, 99–100, 138, 139, 140, 319, 356
research, suggested future, 375–376 CD (cluster of differentiation), 433
bacillary growth in liquefied caseum, 379 CD1 proteins, 122–123
bacillary survival in solid caseum, 379 Cell-mediated immunity (CMI)
stages of tuberculosis and acquired cellular resistance (ACR) compared, 102
stage 3, early stage, 25–28 antibody enhancement of local reactions, 106–107
stage 4, progression or arrest of lesion, 28 arrested lesions, required for, 101–102
stage 5, cavity formation, 29, 30–31 CMI/DTH ratio
Caseous pneumonia, 31, 45, 53, 57 favorable, 100–101
Cathepsin D, 68, 128 ideal, 374
Cavity formation, 65–91; see also Liquefaction research, suggested future, 374–375
aerosolized virulent M. bovis in rabbits, 71–84 species variations, 375
background, 71 control of bacillary growth by, 26–28, 247, 249
high-dose experiments, 73, 74–78 definition, 99, 431
histopathology, 79–84 in guinea pigs, 263–264
low-dose experiments, 71–72, 74 kinetic studies in rabbits, insights on provided by,
tuberculin sensitivity, 78–79 193–194
analysis of lesions, 59 local nature of, 102–104
bronchoscope production in rabbits, 68 macrophage turnover, 375
delayed-type hypersensitivity (DTH), role of, 66 in mice, 259–260, 263–264
in guinea pigs, 256, 263 Mycobacterium vaccae, effect of, 84
histopathology, rabbit, from aerosolized virulent M. overview, 99, 100
bovis in rabbits, 263–265
cavity formation, 78, 79–80 similarities to DTH in tuberculosis, 99–100, 263
epithelioid cells, 80–81 summary of role in pathogenesis of tuberculosis,
fibroblasts, 82, 87, 88 369–370
442 䡵 INDEX

Cell-mediated immunity (CMI) (Continued) ideal, 374


synergism with DTH, 27, 104–105 research, suggested future, 374–375
vaccines producing strong CMI, 344–345 species variations, 375
Chemokines, 433; see also Cytokines control of bacillary growth by, 26–28, 247
Chemotaxins, 24 definition, 98, 431
Chemotaxis, of alveolar macrophages, 83–84 in guinea pigs, 263–264
Childhood-type tuberculosis, 8, 9–11, 49–51 kinetic studies in rabbits, insights on provided by,
Chorionic gonadotropin, effects of, 286 193–194
Chronic infections, general characteristics of, 371 liquefaction and cavity formation, role in, 66
CMI, see Cell-mediated immunity (CMI) local nature of, 102–104
Complement system, 112–113 macrophage turnover, 375
Contagiousness, 11–12 in mice, 259–260, 263–264
Cord factor, 157 Mycobacterium vaccae, effect of, 84
Cortisone overview, 98–99
development of disease, effects on, 273–275 in rabbits, 193–194, 263–265
withdrawal, effects of, 275–279 similarities to CMI in tuberculosis, 99–100, 263
CTLs (cytotoxic T lymphocytes), 141–142, 433 summary of role in pathogenesis of tuberculosis,
C-X-C chemokines, 433 368–370
Cynomolgus monkeys, tuberculosis in, 258–259, 263 synergism with CMI, 27, 104–105
Cytokines tissue-damaging, 26–27, 98–99, 155–157, 432
from CMI-producing lymphocytes, 99 tuberculin sensitivity, 98
definition, 433 tuberculin skin reaction and, 105–106
downregulation, causes of, 321, 323 vaccines producing weak DTH, 344–345
in humans, 307–308 Dendritic cells
in mice, 308–309 as adjuvants, 122
mRNAs, 319, 321 as antigen-presenting cells (APCs), 99, 121–123
networks, 323 CD1 proteins, 122–123
non-antigen-specific nature of, 102 conventional, 122
nonspecific and antigen-specific production, 309 major histocompatibility complex (MHC),
in primary BCG lesions, 301–309 122–123
cell types containing MCP-1, IL-1, IL-8, and organ effects on, 122
TNF- mRNAs, 304 pattern recognition receptors (PRR), 98
comparison among cytokines, 306–307 plasmacytoid, 122
cytokine mRNA in cells within tissue sections, published reviews on antigen presentation, 123
301–304 T-cell activation, 121–122
IFN- mRNA identified by RT-PCR, 305 tolerogenic, 122
immunohistochemical studies, 305 as vaccine carriers, 347
overview, 301–304 Dermal lesions, in rabbits
in reinfection BCG lesions, 312–323 BCG preparations for, 354–355
lesion size, ulceration, and healing, 313 development and healing, 355–356
local cell infiltration, 313 healing as measure of host resistance, 359
mononuclear cells and granulocytes in lesions, histopathology, 356–357
315–319 number of bacilli in lesions, 357–358
number of tubercle bacilli in, 319 pulmonary lesions compared, 358–359
in tuberculin reactions, 312–323 Dexamethasone, reactivation of healing pulmonary
Cytotoxic T lymphocytes (CTLs), 141–142, 433 tubercles by, 279–280
DHEA (dehydroepiandrosterone), 282
D Diapedesis, 328
Dannenberg,Arthur M., Jr. DNA vaccines, 349
contributions of, 2–3 Dormancy, 104–105, 124, 126, 263–264
publications,Appendix D, 399–416 Drugs, suggested future research
Dehydroepiandrosterone (DHEA), 282 acting on bacillus, 380–381
Delayed-type hypersensitivity (DTH) acting on host, 380
antibody enhancement of local reactions, 106–107 DTH, see Delayed-type hypersensitivity (DTH)
arrested lesions, required for, 101–102
caseous necrosis and, 155–157 E
cavity formation, role in, 30–31 ELAM (endothelial-leukocyte adhesion molecule),
CMI/DTH ratio 327–333, 433; see also Vascular adhesion
favorable, 100–101 molecules
INDEX 䡵 443

Emphysematous bleb or bulla, 45 Granulation tissue, 44, 59, 155


Empyema, 49 Granulocytes, 143–144, 376
Encapsulated nodules, 38, 40 in primary and reinfection BCG lesions, 315–319
Eosinophils, 144 Granuloma, macrophage turnover in mouse, 193
Epithelioid cells, 133; see also Langhans’ giant cells Growth curves, bacillary
adhesion molecules in, 333 guinea pig, 254–255
definition, 433 mouse, 247
histopathology in aerosolized virulent M. bovis in Guidelines for Preventing the Transmission of Mycobac-
rabbits, 80–81 terium tuberculosis in Health-Care Settings, CDC,
immature, 28, 103, 133 Appendix E, 417–418
mature, 26, 38, 49, 80–81, 103, 133 Guinea pigs
in miliary tubercles, 42 bacillary titers in stationary stage after aerosol
ESAT-6 antigen, 106 infection, 261–262
Establishment of infection, 35–38 establishment and progress of tuberculosis, 224
airborne infections, natural immunization of, 256, 343–344
resistance to establishment of lesions by bovine- inbred strains, 256
type bacilli, 216–218 organ resistance, 262
resistance to establishment of lesions by human- pulmonary tubercle counts in, 207, 210
type bacilli, 223–224 tuberculin sensitivity, 262
factors influencing, 15–16 tuberculosis in, 254–257
subapical localization, 54–55 bacillary growth curves, 254–255
vaccine, effects of cavity formation, 256, 263
macroscopic tubercle, 342–343 cell-mediated immunity (CMI), 263–264
microscopic tubercle, 342 characteristics of lesions, 260
Estrogen, effects of, 285–286 comparison to mice, humans, and rabbits,
Extracellular dormancy, 263–264 259–260
Exudative lesions, 39, 41, 42 delayed-type hypersensitivity (DTH), 263–264
fate of aerosolized BCG, 262
F fibrin meshworks, 256
Fas, 433 gross pathology, 255–256
Fas ligand, 433 histopathology, 256
Fatty acids, toxic, 31 immunization, 256
Feldman,William H., 385–386 Koch phenomenon, 257
Fibrin meshworks, 256 virulence of inhaled bacilli, 260–261
Fibroblasts, 144 Gut-associated lymphoid tissue (GALT), 343
Fibrocaseous tuberculosis, advanced, 46–47, 54
Fibrosis, 45 H
Filters, HEPA, 16, 17, 18, 19 Head cover, 17, 18, 19
N-Formyl-methionyl-leucyl-phenylalanine, 24 Healing
Freezing and thawing, effect on infectivity, 203–204 caseous necrosis, 45, 158
cytokines in reinfection BCG lesions, 313
G dermal BCG lesions in rabbits, 355–356
-Galactosidase, 67, 102–104, 127 macrophage turnover, activation, and division in
GALT (gut-associated lymphoid tissue), 343 BCG lesions, 189, 191
Gamma interferon (IFN-), 99, 106, 124, 141, 305– as measure of host resistance, 241, 359
308, 433 reactivation of healing pulmonary tubercles by
Gelatin-colloidal carbon perfusion, 162–163 glucocorticoids, 279–280
Genetic resistance, 37–38 Heat shock protein 65, 433
in mice, 251, 253 Hematogenous spread
tubercle-count method, determination by, 204, 207 in childhood-type tuberculosis, 8, 9–11
Ghon complex, 11, 37, 49–52, 58 miliary tuberculosis, 40–42
Glossary, 431–436 Hemoptysis
Glucocorticoids cavity formation and, 44
ACTH effects, 280–282 histopathology, 80, 81
development of disease, effects of cortisone on, HEPA filters, 16, 17, 18, 19
273–275 Histopathology
reactivation of healing pulmonary tubercles by, aerosolized virulent M. bovis lesions in rabbits,
279–280 79–84
withdrawal, effects of, 275–279 dermal BCG lesions in rabbits, 356–357
444 䡵 INDEX

Histopathology (Continued) Immunocompromised hosts, 59–60, 123


guinea pig, tuberculosis in, 256 Immunoglobulin G (IgG), 433
hemoptysis, 80, 81 Immunohistochemical studies for cytokine proteins,
microvascular-cell interactions, 163–167 305
BCG lesions, 163–166 Immunology of tuberculosis, 95–145
tuberculin reactions, 164, 167 acquired cellular resistance (ACR)
mouse pulmonary tuberculosis lesions, 253–254 definition, 431
HIV/AIDS, 59–60, 433 duration and specificity, 102
Hoods, 17, 18, 19 overview, 102
Hormones recall upon reinfection, 111–112
ACTH effects, 280–282 adaptive immunity
androstenediol (AED), 282 definition, 431
chorionic gonadotropin, effects of, 286 innate immunity relation to, 112–114
dehydroepiandrosterone (DHEA), 282 nonspecific and antigen-specific immune
estrogen, effects of, 285–286 responses in, 114
glucocorticoids antibody enhancement of local DTH and CMI
ACTH effects, 280–282 reactions, 106–107, 108, 110
development of disease, effects of cortisone on, antibody role, as elucidated by Lurie’s eye chamber
273–275 experiments, 107, 109, 111
reactivation of healing pulmonary tubercles by, antigen-presenting cells (APCs), 99, 113–114,
279–280 121–123
withdrawal, effects of, 275–279 arrested lesions, CMI and DTH required for,
thyroid hormones, effects of, 286–291 101–102
Host-parasite interactions cell-mediated immunity (CMI)
host response, suggested research on local control acquired cellular resistance (ACR) compared,
of, 373 102
principles of, 370–372 antibody enhancement of local reactions,
summary of, 367 106–107
Human disease, types of, 9–11 arrested lesions, required for, 101–102
adult-type tuberculosis, 11, 12, 13, 14, 15 control of bacillary growth by, 26–28, 247, 249
childhood-type tuberculosis, 8, 9–11 definition, 99, 431
Hydrolytic enzymes, 31 in guinea pigs, 263–264
in activated macrophages, 128 kinetic studies in rabbits, insights on provided by,
liquefaction and cavity formation, role in, 66 193–194
local nature of, 102–104
I macrophage turnover, 375
ICAM (intercellular adhesion molecule), 327–333, in mice, 259–260, 263–264
433–434; see also Vascular adhesion molecules Mycobacterium vaccae, effect of, 84
IFN-, 99, 106, 124, 141, 305–308, 433 overview, 99, 100
Immunity, see also Cell-mediated immunity (CMI) in rabbits, 263–265
adaptive similarities to DTH in tuberculosis, 99–100, 263
definition, 431 summary of role in pathogenesis of tuberculosis,
innate immunity relation to, 112–114 369–370
nonspecific and antigen-specific immune synergism with DTH, 27, 104–105
responses in, 114 vaccines producing strong CMI, 344–345
innate cells involved, 120–145
definition, 431 antigen-presenting cells, 99, 113–114, 121–123
overview, 98 epithelioid cells, 133
relation to acquired (adaptive) immunity, granulocytes, 143–144
112–114 Langhans’ giant cells, 133, 136
systemic, 104 lymphocytes, 137–140
Immunization, see also Vaccine macrophages, 104, 123–133
degree produced, tubercle-count method for deter- mononuclear cell turnover, 136–137
mining, 204 NK cells, 140–141, 142
guinea pig, 256 table of, 121
monkey, 258, 259 CMI/DTH ratios, 100–101
postinfection, 349 favorable, 100–101
resistance to melioidosis and tuberculosis com- ideal, 374
pared, 226–227 research, suggested future, 374–375
INDEX 䡵 445

species variations, 375 L


delayed-type hypersensitivity, 98–99 Labeling of mononuclear cells
DTH and CMI similarities, 99–100 in vitro [3H]TdR labeling, 178–179
DTH and CMI synergism, 27, 104–105 in vivo [3H]TdR labeling, 179, 180
innate immunity Langhans’ giant cells, 38, 39, 40, 42, 133, 136
definition, 431 Laryngitis, tuberculous, 56, 71
overview, 98 Latency, 126; see also Dormancy
relation to acquired (adaptive) immunity, Leukocyte ligands in BCG lesions, 333
112–114 Leukocytes, diapedesis of, 328
macrophage activation, 104 Leukotriene B4, 24
systemic immunity, 104 LFA-1, 333, 335
tuberculin skin test Liquefaction, 65–91
booster phenomenon in repeated testing, aerosolized virulent M. bovis in rabbits, 71–84
105–106 background, 71
size of reaction, prognostic value of, 105 high-dose experiments, 73, 74–78
Immunosuppression, effects of, 59–60 histopathology, 79–84
Immunotherapy low-dose experiments, 71–72, 74
dendritic cells, 347 tuberculin sensitivity, 78–79
Mycobacterium vaccae, 17, 77, 347 bronchogenic spread and, 42–46
research, suggested future, 380 caseation compared, 67–68
Inapparent lesions causes and results, 67, 157–158
in humans, 60–62 histopathology, rabbit, with aerosolized virulent
in rabbits, 62 M. bovis
Inducible nitric oxide synthase (iNOS), 433 cavity formation, 78, 79–80
Infectivity, freezing and thawing effect on, 203–204 epithelioid cells, 80–81
Inhalation of bacilli fibroblasts, 82, 87, 88
bacillary titers in stationary stage after aerosol granulated macrophages, 80, 82
infection, 261–262 hemoptysis, 80, 81
fate of BCG in rabbits, mice, and guinea pigs, lymphocytes and plasma cells, 82–83, 88
262 metaplastic alveolar epithelium and chemotaxis
Lurie’s tubercle-count, 206, 208–209 of alveolar macrophages, 83–84, 88, 89
number inhaled, 15–16 number of bacilli, 81, 83, 84, 85
rabbit response to, 233–234 literature review, 66–71
virulence of bacilli in rabbits, mice, and guinea bronchoscope production of cavities in rabbits,
pigs, 260–261 68
X-irradiation effects on virulent human-type dermal BCG lesions in pilot studies on caseation
bacilli, 296–297 and liquefaction, 68
Innate immunity distinction between caseation and liquefaction,
definition, 431 67–68
overview, 98 effects of large numbers of bacilli on liquefac-
relation to acquired (adaptive) immunity, 112–114 tion, 68, 71
iNOS (inducible nitric oxide synthase), 433 role of delayed-type hypersensitivity, 66
Intercellular adhesion molecule (ICAM), 327–333, role of hydrolytic enzymes, 66–67
433–434; see also Vascular adhesion molecules measuring factors affecting, proposed method for,
Interleukins 90–91
definition, 434 in pathogenesis process, 29, 30–31
IL-2, 307, 434 recent experiments attempting to reduce,
IL-8, 304, 306, 307, 319, 321–323, 434 84–87
IL-10, 113, 307, 434 Mycobacterium vaccae, studies with, 84, 86
IL-12, 113–114, 141, 307, 434 ritonavir, studies with, 86–87
IL-1, 304, 306, 307, 319, 321–323, 434 research, suggested future, 376–377
Th cell production of, 138 summary, 87, 90–91
Intracellular dormancy, 263–264 Logarithmic growth
Isocitrate lyase, 124 end of by DTH and CMI, 25–28, 247, 249
within macrophages, 24–25
K Long, Esmond R., 387–388
Knockout mice, 434 Lurie, Max B.
Koch phenomenon, 257, 434 antibody role, as elucidated by eye chamber experi-
Koch, Robert, 26 ments, 107, 109, 111
446 䡵 INDEX

Lurie, Max B. (Continued) Macaca mulatta, 257–258


award of Trudeau medal for 1955,Appendix A, Macrophage inflammatory proteins (MIPs), 434
385–386 Macrophages, 123–133; see also Epithelioid cells
contributions of, 1–2 activation, 54–55, 99, 100, 104, 126–128
intravenous BCG experiments, 359–360 in healing dermal BCG lesions, 189, 191
natural airborne infection experiments, 215–224 in vitro division of activated in BCG lesions, 179
obituary,Appendix B, 387–388 alveolar
publications,Appendix C, 389–398 activation, 54–55
pulmonary tubercle-count method, 196–210 chemotaxis, 83–84
Lurie’s inbred rabbits definition, 432
adrenocorticotropic hormone (ACTH), 280–282 division of activated in early lesions in rabbits,
airborne infections, natural, 215–224 175
resistance to establishment of lesions by bovine- ingestion of tubercle bacilli, 16, 22–23, 36
type bacilli, 216–218 innate immunity and, 98, 114
resistance to establishment of lesions by human- location in lesion, 37
type bacilli, 223–224 logarithmic growth of bacilli within nonacti-
resistance to progress of pulmonary tuberculosis vated, 24–25
by bovine-type bacilli, 218–223 microbicidal power of, 16
resistance to progress of pulmonary tuberculosis role in establishment of grossly visible primary
by human-type bacilli, 224 tubercles, 200
characteristics of lesions in, 260 susceptibility versus resistance, 37–38
fate of, 241 vaccine effects, 342
genetic experiments, 238–241 X-irradiation, effects of, 293–296
healing rate of dermal lesions as method of deter- bacillary survival within, 379
mining resistance, 241 blood-borne, 24–25, 36–37, 170–175
history and description of, 235–236 in caseous tissues, 155–156
intradermal BCG vaccine in Lurie’s inbred rabbit chemotaxins, 24
strains definition, 432
lesions, 361 delayed-type hypersensitivity (DTH) killing of,
protection, amount of, 361–363 26–28
vaccine, 361 division
relative resistance of strains, 236 of alveolar activated in early lesions in rabbits,
resistance and susceptibility to tuberculosis, 175
237–238 in early pulmonary lesions in rabbits, 155–156
Lymphatic spread, in childhood-type tuberculosis, 9, in healing dermal BCG lesions, 189, 191
49–51 in vitro division of activated in BCG lesions,
Lymphatics in tuberculous lesions, 144–145 179
Lymphocytes in vitro division of cells containing bacilli in
apoptosis and, 142 BCG lesions, 180
CD4, 138, 139, 140 enzymes released extracellularly in BCG lesions,
CD8, 99–100, 138, 139, 140 128–129
cell-mediated immunity and, 99 fate of mycobacteria within, 123–124
cytotoxic T cells, 141–142, 433 granulated
definition, 431–432 histopathology of aerosolized virulent M. bovis in
delayed-type hypersensitivity and, 98 rabbits, 80, 82
gamma-delta () T cells, 138–140 heterogeneity
histopathology of aerosolized virulent M. bovis in in BCG lesions, 129, 131, 133
rabbits, 82–83, 88 causes, 131, 133
memory T cells, 140 in vitro division
NK cells, 140–141, 142, 434 of activated in BCG lesions, 179
overview, 137–138 of cells containing bacilli in BCG lesions, 179
regulatory T cells, 138, 435 labeling of
Th cells, 138, 435–436 in vitro [3H]TdR labeling, 178–179
turnover, 136–137 in vivo [3H]TdR labeling, 180
Lysosomes, 123–124 location in lesion, 37, 59
nonspecific activation, 22–23, 54
M in pulmonary lesion in rabbits, early
Mac-1, 333 activation and division of blood-borne, 170–175
Macaca fascicularis, 258–259 division of activated pulmonary alveolar, 175
INDEX 䡵 447

receptors, 123 blood supply, pathophysiology of, 167–168


turnover, 136–137 capillary density, determination of, 162–163
in healing dermal BCG lesions, 189, 191 histopathology of microvascular-cell interactions,
in mouse tuberculous granulomas, 193 163–167
research, suggested future, 375 BCG lesions, 163–166
Major histocompatibility complex (MHC) tuberculin reactions, 164, 167
activation of microvascular endothelium contribu- overview, 161
tion to caseous necrosis, 334–335 production of BCG lesions and tuberculin reac-
cytotoxic T cells and, 142 tions, 161–162
dendritic cells, 122–123 Microvascular endothelium, activation and caseous
NK cells, 140 necrosis, 334–335
Masks, 16, 19 Microvascular patency, in mouse pulmonary lesions,
Mast cells, 144 254
MCP-1 (monocyte chemoattractant [activating] pro- Microvessels, identification in tissue sections of BCG
tein 1), 304–306, 308, 319, 321–323, 434 lesions, 329
Medical radiation, public health consequences of, Miliary tuberculosis, 40–42, 43, 44, 61
297–298 MIPs (macrophage inflammatory proteins), 434
Melioidosis, 226–227 Monkeys, tuberculosis in, 257–259, 263
Meningitis, tuberculous, 58 Monocyte chemoattractant protein 1, 24
Metaplastic alveolar epithelium Monocyte chemoattractant (activating) protein 1
histopathology of aerosolized virulent M. bovis in (MCP-1), 304–306, 308, 319, 321–323, 434
rabbits, 83–84, 88, 89 Monocytes, symbiotic growth of bacilli within non-
Metaplastic bronchial epithelium, 45 activated, 24–25
MHC, see Major histocompatibility complex (MHC) Mononuclear cells, see also Macrophages
Mice BCG lesions
cytokines, 308–309 activation in tuberculin reactions and in lesions
establishment and progress of tuberculosis, 224 caused by nonspecific irritants, 192–193
immunization of, 343–344 in blood, labeled, 185–186
melioidosis, immunization and resistance to, disappearance from lesions, 182–184
226–227 division rate within lesions, 185
pulmonary lesions entry into lesions, 180, 182
histopathology, 253–254 rates of activation in primary and reinfection
microvascular patency, 254 lesions, 187, 189
pulmonary tubercle counts in, 207, 210 definition, 434
tuberculin sensitivity, 262 labeling
tuberculosis in, 247–254 in vitro [3H]TdR labeling, 178–179
apoptosis, 263 in vivo [3H]TdR labeling, 180
bacillary growth curves and type of disease, in primary and reinfection BCG lesions, 315–319
247 turnover, 136–137
bacillary titers in stationary stage after aerosol Mycobacterium bovis, aerosolized virulent in rabbits,
infection, 261–262 79–84
bovine-type bacilli, 251 histopathology
cavity formation, 263 cavity formation, 78, 79–80
cell-mediated immunity (CMI), 263–264 epithelioid cells, 80–81
characteristics of lesions, 260 fibroblasts, 82, 87, 88
comparison to rabbits, humans, and guinea pigs, hemoptysis, 80, 81
259–260 lymphocytes and plasma cells, 82–83, 88
delayed-type hypersensitivity (DTH), 263–264 metaplastic alveolar epithelium and chemotaxis
fate of aerosolized BCG, 262 of alveolar macrophages, 83–84, 88, 89
genetic differences in resistance, 251, 253 number of bacilli, 81, 83, 84, 85
human-type bacilli, 251 Mycobacterium bovis BCG
logarithmic stage of growth, ending, 247, 249 intradermal injection to produce caseous necrosis
stationary stage of growth, maintaining, 249 and liquefaction, 68
tissue necrosis, 249–251, 263 macrophage activation, 127–128
virulence of inhaled bacilli, 260–261 macrophage enzymes released extracellularly,
Microbicidal power, of pulmonary alveolar 128–129
macrophages, 16 macrophage heterogeneity in lesions, 129, 131,
Microvascular density in tuberculous lesions, 161–168 133
angiogenesis, regulators of, 167 virulence, 13, 14
448 䡵 INDEX

Mycobacterium microti role of delayed-type hypersensitivity, 66


vaccine efficacy in New Zealand White rabbits, role of hydrolytic enzymes, 66–67
363 summary, 87, 90–91
virulence, 14 overview, 7–19
Mycobacterium tuberculosis contagiousness, 11–12
Guidelines for Preventing the Transmission of Mycobac- establishment of pulmonary lesion, factors influ-
terium tuberculosis in Health-Care Settings, CDC, encing, 15–16
Appendix E, 417–418 prevention of clinical disease, 17–19
mutants as possible new vaccines, 349 resistance, factors influencing, 16–17
strain virulence, 13–15 size of infectious particles, 12–13
virulence by Lurie tubercle-count method types of human disease, 9–11
CDC1551 (Oshkosh) strain, 201, 203, 204 virulence of bacillary strains, 13–15
Erdman strain, 203 stages, 22–31
H37Rv strain, 201, 202, 203, 204 stage 1, ingestion of bacilli by macrophages,
Mycobacterium vaccae 22–23
effect on cavity formation, 84, 86 stage 2, logarithmic growth of bacilli within
immunotherapy, 17, 347 macrophages, 24–25
for active tuberculosis from inhalation of viru- stage 3, end of logarithmic growth by DTH and
lent bovine-type bacilli, 77 CMI, 25–28
stage 4, progression or arrest of caseous lesions,
N 28
Necropsies, safety precautions for, 18 stage 5, cavity formation and bronchial spread of
Necrosis, see Caseous necrosis disease, 29, 30–31
Neutrophil attractant-activating protein 1 (NAP-1), summary, 367–368
434 table of, 30
Nitric oxide synthase (NOS), 123, 124 types of human pulmonary tuberculosis, 9–11,
NK (natural killer) cells, 140–141, 142, 434 34–62
adult-type tuberculosis, 11, 12, 13, 14, 15, 51–52
O advanced fibrocaseous, 46–47
Old tuberculin, 434 cavitary lesions, analyses of, 59
Opsonins, 434 childhood-type tuberculosis, 8, 9–11, 49–51
Organ resistance, 102–104, 158–159 early primary lesion, 38
guinea pigs, 262 encapsulated caseous or calcified nodules, 38
rabbits, 262 establishment of infection, 35–38
research, suggested future, 373–374 exudative lesions, 39, 41, 42
Ossification, 38, 45 in immunocompromised host, 59–60
inapparent lesions, 60–62
P liquefied caseous lesions, 42–46
Parasite-host interactions miliary, 39–42
host response, suggested research on local control multidrug-resistant bacilli, 57–59
of, 373 pneumonia and pleurisy, 47–49
principles of, 370–372 progressive, locally destructive lesions, 46
summary of, 367 proliferative lesions, 38–39, 40, 42
Pathogenesis subapical localization, 51–57
liquefaction of caseous foci and cavity formation, table, 35
65–91 Pathogenicity, virulence of bacillary strain and,
aerosolized virulent M. bovis in rabbits, 71–84 13–15, 24
bronchoscope production of cavities in rabbits, Pattern recognition receptors (PRR), 98, 113–114
68 PBMC, see Peripheral blood mononuclear cells
dermal BCG lesions in pilot studies on caseation Pepstatin, 67
and liquefaction, 68 Perforin, 142
distinction between caseation and liquefaction, Pericarditis, 48
67–68 Peripheral blood mononuclear cells (PBMC), 307–
effects of large numbers of bacilli on liquefac- 308, 434
tion, 68, 71 Persistence, of tubercle bacilli, 158
literature review, 66–71 Phagosomes, 123–124
measuring factors affecting, proposed method Phosphate-specific transport protein 1 (PstS-1), 435
for, 90–91 Plasma cells
recent experiments attempting to reduce, 84–87 definition, 435
INDEX 䡵 449

histopathology of aerosolized virulent M. bovis in R


rabbits, 82–83, 88 Rabbits
Plasmacytoid dendritic cells, 122 airborne infections, natural, 215–227
Plethismograph, 435 Lurie’s experiments, 215–224
Pleurisy, 48–49 Riley’s experiments, 224–226
Pneumonia Bar Harbor inbred, 241–242
caseous pneumonia, 31, 45, 53, 57 BCG, intravenous, 359–360
multidrug-resistant, 59 BCG dermal lesions
overview, 47–49 BCG preparations for, 354–355
rupture of liquefied lesion and, 44 development and healing, 355–356
Polymorphonuclear cells (PMNs), 143–144 healing as measure of host resistance, 359
interleukin mRNAs in, 321 histopathology, 356–357
in primary and reinfection BCG lesions, 315–319 number of bacilli in lesions, 357–358
PPD (purified protein derivative), 435; see also Tuber- pulmonary lesions compared, 358–359
culin skin test commercial inbred, 242
Prevention of clinical disease, 17–19 comparison of tuberculosis in humans, 226
Primary lesion, early, 38 histopathology of lesions from aerosolized virulent
Prognostic tests, 377–378 M. bovis, 79–84
Programmed cell death, see Apoptosis Lurie’s inbred
Progression, prognostic tests for, 377–378 adrenocorticotropic hormone (ACTH),
Progressive lesions, 46 280–282
Proliferative lesions, 38–39, 40, 42 airborne infections, natural, 215–224
Propylthiouracil, 289–291 characteristics of lesions in, 260
PRR, see Pattern recognition receptors (PRR) fate of, 241
PstS-1 (phosphate-specific transport protein 1), genetic experiments, 238–241
435 healing rate of dermal lesions as method of
Public health consequences of medical radiation, determining resistance, 241
297–298 history and description of, 235–236
Pulmonary alveolar macrophage, see Alveolar intradermal BCG vaccine in, 361–363
macrophages relative resistance of strains, 236
Pulmonary lesion resistance and susceptibility to tuberculosis,
caseous necrosis 237–238
cavity formation, 29, 30–31 Lurie’s tubercle-count method, 196–210
early stage, 25–28 model of tuberculosis, advantages of, 199–200
progression or arrest of lesion, 28 organ resistance, 262
dermal BCG lesions compared in rabbits, 358–359 pulmonary lesion, early, 170–175
factors influencing establishment of, 15–16 macrophages, activation and division of blood-
establishment of infection, 16 borne, 170–175
microbicidal power of pulmonary alveolar macrophages, division of activated pulmonary
macrophages, 16 alveolar, 175
number of bacilli inhaled, 15–16 production by intravenous injection of bacilli,
mouse advantages of, 170
histopathology of, 253–254 response to inhaled bacilli
microvascular patency of, 254 BCG, 233–234
in rabbits, early, 170–175 bovine-type bacilli, 231–233
macrophages, activation and division of blood- human-type bacilli, 233
borne, 170–175 Lurie’s resistant rabbits, 231–233
macrophages, division of activated pulmonary Lurie’s susceptible rabbits, 231
alveolar, 175 New Zealand White rabbits, 233
production by intravenous injection of bacilli, strains, new resistant and susceptible, 378
advantages of, 170 Thorbecke, 242–244
reactivation of healing pulmonary tubercles by glu- tuberculin reactions, 358
cocorticoids, 279–280 tuberculin sensitivity, 262
resistance to progress in Lurie’s rabbits tuberculosis in, 247, 248
bovine-type bacilli, 218–223 bacillary titers in stationary stage after aerosol
human-type bacilli, 224 infection, 261–262
Pulmonary lesion-count method, Lurie’s, 196–210 cavity formation, 263
Purified protein derivative (PPD), 435; see also Tuber- cell-mediated immunity (CMI), 263–265
culin skin test characteristics of lesions, 260
450 䡵 INDEX

Rabbits (Continued) antibody role in host resistance to reinfection,


comparison to mice, humans, and guinea pigs, 378
259–260 CMI/DTH ratio, 374–375
delayed-type hypersensitivity (DTH), 263–265 DTH and CMI and macrophage turnover, 375
fate of aerosolized BCG, 262 granulocytes, role of, 376
virulence of inhaled bacilli, 260–261 host response, local control of, 373
X-irradiation effects, 297 lesions produced by live and dead tubercle
vaccine bacilli, comparisons of, 378
efficacy of BCG in New Zealand White rabbits, liquefaction and cavity formation and preven-
363 tion, 375–376
efficacy of M. microti in New Zealand White organ resistance and its implications, 373–374
rabbits, 363 prognostic tests that reflect disease progression
maximal effectiveness, 348–349 and regression, 377–378
van Zutphen, 241 rabbit strains, new resistant and susceptible, 378
Radiation (X-irradiation), 292–298 vascular thrombosis, role in caseous necrosis and
alveolar macrophage from granulomatous lungs, inhibition of bacillary growth, 375–376
effects on, 294–296 on immunotherapy, 380
dermal BCG lesions, effects on, 293 on vaccines, 380
inhalation of virulent human-type bacilli, effects Resistance
on, 296–297 acquired cellular resistance (ACR), 102, 111–112
lethal dose, 293 in adults versus children, 51
public health consequences of medical radiation, airborne infections, natural
297–298 to establishment of lesions by bovine-type
pulmonary alveolar macrophage populations, bacilli, 216–218
effects on, 293–294 to establishment of lesions by human-type
in rabbits, 297 bacilli, 223–224
recovery from effects of, 296 to progress of pulmonary tuberculosis by
Radiographs bovine-type bacilli, 218–223
of encapsulated nodules, 38 to progress of pulmonary tuberculosis by
lesion size, 38 human-type bacilli, 224
Reactivation, 52, 59, 279–280 antibody role in, 378
Reactive oxygen intermediates (ROIs), 435 bovine-type and human-type bacilli, 159
Recombinant technology improved vaccines, 349 factors influencing, 16–17
Regression, prognostic tests for, 377–378 genetic, 37–38, 204, 207, 251, 253
Regulatory (suppressor) T lymphocytes, 138, 435 healing as measure of host resistance, 241, 359
Reinfection, 51–52 host
acquired cellular resistance (ACR) recall, 111–112 healing as measure of, 241, 359
antibody role in resistance to, 378 tubercle-count to determine, 204
cytokines in BCG lesions, 312–323 to melioidosis, 226–227
lesion size, ulceration, and healing, 313 mouse strains, 251, 253
local cell infiltration, 313 new rabbit strains, 378
mononuclear cells and granulocytes in lesions, organ, 102–104, 158–159, 263, 373–374
315–319 species, 158–159
number of tubercle bacilli in, 319 versus susceptibility, 24–25, 37–38
rates of activation of mononuclear cells in BCG symbiotic growth of bacilli within macrophages,
lesions, 187, 189 24–25, 37–38
vascular adhesion molecules in BCG lesions, 331, of tubercle bacilli to antimicrobials, 31, 57–59
332 Respirators, 16, 17
Research, suggested future, 373–384 Reverse transcription-polymerase chain reaction, see
on the bacillus RT-PCR
growth in liquefied caseum, 379 Rhesus monkeys, tuberculosis in, 257–258, 259, 263
survival in air, 378–379 Riley, R. L., 224–226
survival in solid caseum, 379 Ritonavir, studies with, 86–87
survival within macrophages, 379 RNase, 68, 128
virulence, 379–380 ROIs (reactive oxygen intermediates), 435
on drugs Room air sampling, 226
acting on bacillus, 380–381 RT-PCR (reverse transcription-polymerase chain
on the host reaction)
INDEX 䡵 451

definition, 435 Titers, in stationary stage in rabbits, mice, and guinea


IFN- mRNA identified by, 305 pigs, 261–262
TNF- (tumor necrosis factor alpha), 99, 141, 304–
S 308, 320–321, 323, 436
Satellite lesions, 46 Tolerogenic dendritic cells, 122
Shwartzman phenomenon, 435 Toll-like receptors, 112, 113, 123
Simon foci, 53 Transforming growth factor beta (TGF-), 436
Size, of infectious particles, 12–13 Triiodothyronine, effects of, 286–291
Stages in pathogenesis, 22–31 Trudeau medal for 1955, 385–386
stage 1, ingestion of bacilli by macrophages, Tubercle bacilli
22–23 dormancy, 124, 126, 263–264
stage 2, logarithmic growth of bacilli within ending of logarithmic growth by DTH and CMI,
macrophages, 24–25 25–28
stage 3, end of logarithmic growth by DTH and ingestion by macrophages, 16, 22–23, 36, 123–124
CMI, 25–28 inhaled, number, 15–16
stage 4, progression or arrest of caseous lesions, 28 inhibition of growth, 375–376
stage 5, cavity formation and bronchial spread of intracellular versus extracellular growth, 43
disease, 29, 30–31 logarithmic growth within macrophages, 24–25
table of, 30 number in dermal rabbit BCG lesions, 357–358
STAT-1, 124 persistence, 158
Strain, virulence of, 13–15 research, suggested future
Subapical localization growth in liquefied caseum, 379
in adult-type tuberculosis, 51–52 survival in air, 378–379
causes of, 52–54 survival in solid caseum, 379
physiology of apical versus basal pulmonary survival within macrophages, 379
regions, 53–54 virulence, 379
source of bacilli, 52–53 resistance, 31, 43
establishment of lesions, effect on, 54–55 virulence
progress of lesions, effect on, 55–57 research, suggested future, 378–380
Summary strain, 13–15, 24
host-parasite interactions, 367 Tubercle-count method, Lurie’s, 196–210
pathogenesis, role of DTH and CMI in, 368–370 advantages of rabbit model of tuberculosis,
pathogenesis, stages, 367–368 199–200
Suppressor T-cells, 138 aerosol infections versus intravenous infections,
Surfactant proteins, 113 comparison of, 206–207, 210
Symbiotic growth of bacilli within nonactivated bacillary virulence, 207
macrophages, 24–25 host genetic resistance, 207
Systemic immunity, 104 vaccine efficacy, 207
alveolar macrophage role in establishment of
T grossly visible primary tubercles, 200
T cell comparison of counts, 204–206
activation by dendritic cells, 121–122 genetic resistance of host, determination of, 204
cytotoxic, 141–142, 433 in guinea pigs and mice, 207, 210
gamma-delta (), 138–140 immunization degree, determination of, 204
helper, 138, 435–436 inhaled dose, effect of, 206, 208–209
memory, 140 overview, 197–198
regulatory, 138, 435 variability of counts, 205, 206
TGF- (transforming growth factor beta), 436 virulence, determination of
Th cells, 138, 435–436 bovine-type bacilli, 201
Thorbecke rabbits, 242–244 clinical Mycobacterium tuberculosis isolate, 201,
Thyroid hormones, effects of, 286–291 203, 204
Thyroidectomy, 289–291 freezing and thawing, effect on infectivity, 203–
Thyroxine, effects of, 286–291 204
Tissue damage human-type bacilli (H37Rv), 201, 202, 203, 204
causes, 156–157 overview, 200–201
delayed-type hypersensitivity (DTH), 26–27, 98– Tubercles
99, 155–157 alveolar macrophage role in establishment of
Tissue necrosis in mice, 249–251, 263 grossly visible primary tubercles, 200
452 䡵 INDEX

Tubercles (Continued) production by intravenous injection of bacilli,


definition, 436 advantages of, 170
development in humans, 197 resistance to establishment in Lurie’s rabbits
in rabbits caused by inhaled BCG, 233–234 bovine-type bacilli, 216–218
reactivation of healing pulmonary tubercles by glu- human-type bacilli, 223–224
cocorticoids, 279–280 structural components, 155–159
Tuberculin reactions caseous tissues, 155–157
cytokines in, 312–323 granulation tissue, 155
microvasculature of lesions healing, 158
histopathology, 164, 167 liquefied caseum and cavities, 157–158
production of developing and healing lesions, organ and species resistance, 158–159
161–162 overview, 155
mononuclear cell activation in, 192–193 persisting viable bacilli, 158
in rabbits, 358 vascular adhesion molecules in, 327–336
Tuberculin sensitivity; see also Delayed-type hypersen- activation of microvascular endothelium and
sitivity (DTH) caseous necrosis, 334–335
cavity formation in rabbits and, 78–79 in acute inflammatory lesions, 331
disease control and, 346 in epithelioid cells, 333
in laboratory animals, 262 functions, 333–334
Tuberculin skin test identification of microvessels in tissue sections of
anergy, 105 BCG lesions, 329
booster phenomenon in repeated testing, 105–106 leukocyte ligands in BCG lesions, 333
early positives, 38 overview in, 328–329
size of reaction, prognostic value of, 105 in primary BCG lesions, 329–330
Tuberculin traps, 185 quantitation in tissue sections, 329
Tuberculin unit, 99 questions to be answered, 335–336
Tuberculous lesions, 155–210; see also Pathogenesis in reinfection BCG lesions, 331, 332
arrested, 28, 37, 38, 60–62 Tumor necrosis factor alpha (TNF-), 99, 141, 304–
CMI and DTH required for, 101–102 308, 320–321, 323, 436
early in humans, 263 Types of human disease, 9–11
inapparent, 60–62 adult-type tuberculosis, 11, 12, 13, 14, 15
Lurie’s pulmonary lesion-count method, 196–210 childhood-type tuberculosis, 8, 9–11
lymphatics in, 144–145 Types of human pulmonary tuberculosis, 9–11,
macrophages, 177–194 34–62
activation, 189, 191 adult-type tuberculosis, 11, 12, 13, 14, 15, 51–52
division, 189, 191 advanced fibrocaseous, 46–47
in vitro division of activated in BCG lesions, 179 cavitary lesions, analyses of, 59
in vitro [3H]TdR labeling, 178–179 childhood-type tuberculosis, 8, 9–11, 49–51
in vivo [3H]TdR labeling, 180 early primary lesion, 38
overview, 178 encapsulated caseous or calcified nodules, 38
turnover in healing dermal BCG lesions, 189, establishment of infection, 35–38
191 exudative lesions, 39
turnover in mouse tuberculous granulomas, 193 in immunocompromised host, 59–60
microvascular density, 161–168 inapparent lesions, 60–62
angiogenesis, regulators of, 167 liquefied caseous lesions, 42–46
blood supply, pathophysiology of, 167–168 miliary, 39–42
capillary density, determination of, 162–163 multidrug-resistant bacilli, 57–59
histopathology of microvascular-cell interactions, pneumonia and pleurisy, 47–49
163–167 progressive, locally destructive lesions, 46
overview, 161 proliferative lesions, 38–39
production of BCG lesions and tuberculin reac- subapical localization, 51–57
tions, 161–162 table, 35
produced by live and dead bacilli, comparisons of,
378 U
pulmonary lesions in rabbits, early, 170–175 Uric acid, 24
macrophages, activation and division of blood- UV (ultraviolet) lights, for disease prevention, 18–19
borne, 170–175
macrophages, division of activated pulmonary V
alveolar, 175 Vaccine, 341–350
INDEX 䡵 453

BCG in acute inflammatory lesions, 331


combination vaccines, 349 in epithelioid cells, 333
inapparent lesions and, 61–62 functions, 333–334
in mice and guinea pigs, 343–344 identification of microvessels in tissue sections of
in monkeys, 258, 260 BCG lesions, 329
nonspecific irritants, effect on, 193 leukocyte ligands in BCG lesions, 333
recombinant technology improved, 349 overview in, 328–329
strong CMI and weak DTH, 344–345 in primary BCG lesions, 329–330
systemic nature in humans, 360–361 quantitation in tissue sections, 329
combination, 349 questions to be answered, 335–336
composition of improved in reinfection BCG lesions, 331, 332
adjuvant composition, 345–346 Vascular endothelial growth factor (VEGF), 436
antigen composition, 345 Vascular thrombosis, role in caseous necrosis and
degree of immunization produced, tubercle-count inhibition of bacillary growth, 375–376
method for determining, 204 VCAM-1 (vascular cell adhesion molecule-1), 327–
DNA vaccines, 349 333, 436
efficacy VEGF (vascular endothelial growth factor), 436
in aerosol infections versus intravenous infec- Virulence
tions, comparison of, 207 of bacillary strain, 13–15, 24, 207
BCG in New Zealand White rabbits, 363 inhaled bacilli in rabbits, mice, and guinea pigs,
M. microti in New Zealand White rabbits, 363 260–261
standardization for comparing relative, 346–347 tubercle-count method, determination by
future possibilities, 349–350 bovine-type bacilli, 201
intradermal BCG in Lurie’s inbred rabbit strains clinical Mycobacterium tuberculosis isolate, 201,
lesions, 361 203, 204
protection, amount of, 361–363 freezing and thawing, effect on infectivity,
vaccine, 361 203–204
intradermal in Lurie’s inbred rabbit strains, human-type bacilli (H37Rv), 201, 202, 203,
361–363 204
M. tuberculosis mutants, 349 overview, 200–201
maximal effectiveness VLA-4, 333
factors influencing, 348
in humans, 347–348
W
in rabbits, 347–348
Whole-body irradiation, see X-irradiation
mice and guinea pigs, immunization of, 343–344
postinfection immunization, 349
pulmonary tubercle establishment, effects on X
macroscopic tubercle, 342–343 X-irradiation, 292–298
microscopic tubercle, 342 alveolar macrophage from granulomatous lungs,
recombinant technology improved vaccines, 349 effects on, 294–296
research, suggested future, 380 dermal BCG lesions, effects on, 293
reviews of possible new, 349–350 inhalation of virulent human-type bacilli, effects
routes of administration, 343 on, 296–297
systemic nature of BCG vaccination in humans, lethal dose, 293
360–361 public health consequences of medical radiation,
van Willebrand factor, 436 297–298
van Zutphen rabbits, 241 pulmonary alveolar macrophage populations,
Vascular adhesion molecules, 327–336 effects on, 293–294
activation of microvascular endothelium and in rabbits, 297
caseous necrosis, 334–335 recovery from effects of, 296

You might also like