Immunopathogenesis of Pulmonary Granulomas

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 9

Immunopathogenesis of Pulmonary

Granulomas in the Guinea Pig after


Infection with Mycobacterium tuberculosis
Oliver C. Turner, Randall J. Basaraba and Ian M. Orme
Infect. Immun. 2003, 71(2):864. DOI:
10.1128/IAI.71.2.864-871.2003.

Updated information and services can be found at:


http://iai.asm.org/content/71/2/864

Downloaded from http://iai.asm.org/ on March 3, 2014 by guest


These include:
REFERENCES This article cites 43 articles, 19 of which can be accessed free
at: http://iai.asm.org/content/71/2/864#ref-list-1

CONTENT ALERTS Receive: RSS Feeds, eTOCs, free email alerts (when new
articles cite this article), more»

Information about commercial reprint orders: http://journals.asm.org/site/misc/reprints.xhtml


To subscribe to to another ASM Journal go to: http://journals.asm.org/site/subscriptions/
INFECTION AND IMMUNITY, Feb. 2003, p. 864–871 Vol. 71, No. 2
0019-9567/03/$08.00⫹0 DOI: 10.1128/IAI.71.2.864–871.2003
Copyright © 2003, American Society for Microbiology. All Rights Reserved.

Immunopathogenesis of Pulmonary Granulomas in the Guinea Pig


after Infection with Mycobacterium tuberculosis
Oliver C. Turner, Randall J. Basaraba, and Ian M. Orme*
Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology,
Colorado State University, Fort Collins, Colorado 80523
Received 4 April 2002/Returned for modification 12 September 2002/Accepted 31 October 2002

Pulmonary tuberculosis in guinea pigs is similar to the disease in humans and is accordingly widely used as
a model to test tuberculosis vaccines. The primary site of expression of acquired immunity and the hallmark
of tuberculosis is the granuloma. Granuloma morphology is well described, but there is limited information
regarding T-cell subset influx. We monitored the course of pulmonary tuberculosis in guinea pigs and observed

Downloaded from http://iai.asm.org/ on March 3, 2014 by guest


four distinct immunohistopathological stages. In all stages there were similar numbers and arrangement of
CD4 and CD8 T cells. There were only small numbers of apoptotic lymphocytes, scattered around and within
the necrotic core, and acid-fast bacilli were visible both within macrophages and free within airway debris. A
key finding of the study was the observation that the development of the necrotic core was an early event and
almost certainly preceded the emergence of the acquired immune response. This in turn suggests that innate
mechanisms are the basis of the early lesions and that subsequent acquired responses are unable to moderate
them. This hypothesis differs from the current dogma that excessive activity of T cells mediates delayed-type
hypersensitivity and that cellular cytolysis is the root cause of the necrosis.

Mycobacterium tuberculosis continues to kill approximately 2 cells into the granuloma and their retention in the granuloma
million people per year (12), more than any other single hu- (39).
man pathogen. In addition, it is estimated that in excess of 100 Because of the significant ways in which it resembles both
million people may carry the organism in a dormant or latent the normal human physiology and the pathophysiology of
state (14). This has prompted a concerted effort to develop pulmonary tuberculosis, the guinea pig is one of the more
new vaccines with which to prevent the disease in its various extensively studied animal models of tuberculosis (40). Fol-
forms (31, 33). lowing exposure to a small number of bacilli delivered via
There are now several different animal models available with aerosol, the infection in the guinea pig lung grows progres-
which to test new vaccines, ranging (in size and expense) from sively for about 30 days (41) before being contained by the
the mouse to primate models. It is widely accepted that per- host response (30). Similarly to other animal models how-
haps the best model is the guinea pig (Cavia porcellus), given
ever, this expression of host immunity is not completely
the similarities of this model to humans infected with tuber-
effective, and as a result, many bacilli survive, giving rise to
culosis in terms of the formation of the lung granuloma and its
a chronic state of disease.
subsequent necrosis and mineralization (13, 18, 24, 37).
Although it has long been realized that the protective
The formation of the lung granuloma is the classical hall-
mark of pulmonary tuberculosis. The morphology of the gran- process in the guinea pig lung involves the formation of
uloma is characterized by a caseous necrotic core which is granulomas, there is surprisingly little further description of
surrounded by a layer of epithelioid macrophages, which, in this event. For example, previous descriptions have charac-
turn, is surrounded by concentric layers of lymphocytes and terized “primary” and “secondary” lesions with reference to
fibrosis (6). Multinucleated giant cells and occasional plasma only the size and amount of necrosis in the lesions (42). The
cells are also seen. The influx of leukocytes and fibroblasts guinea pig is well known to be a susceptible animal model
forming these structures is an attempt to segregate the infec- that develops granulomas with prominent central caseation
tion, to prevent bacterial dissemination to the remainder of the and extensive connective tissue deposition, yet knowledge of
lung and other organs, and to focus the immune response the arrangement of these elements over time remains ob-
directly at the site of implantation. Its development involves an scure. In addition, the influx of T-cell subsets, only recently
elaborate production and interaction of chemokines and cyto- described in the mouse model (16), has yet to have been
kines by effector cells as well as local tissue cells. In addition, documented in the guinea pig.
the upregulation of receptors for these molecules as well as for Given the importance of the model to current vaccine de-
adhesion and integrin molecules on responder cells facilitates velopment work, the purpose of this study was to chronicle the
the proper coordination of the recruitment and migration of temporal and spatial arrangement of the cell influx, as well as
other aspects of the overall process, and to attempt to relate
these data to the known pathological process. The findings
* Corresponding author. Mailing address: Department of Microbi-
presented seriously challenge the current dogma that excessive
ology, Immunology and Pathology, Colorado State University, Fort
Collins, CO 80523. Phone: (970) 491-5777. Fax: (970) 491-5125. E- expression of the acquired T-cell response triggers the patho-
mail: iorme@lamar.colostate.edu. logic degeneration of the lung granuloma.

864
VOL. 71, 2003 PATHOLOGY OF TUBERCULOSIS IN GUINEA PIGS 865

MATERIALS AND METHODS


Animals. Female Hartley guinea pigs were obtained from Charles River Lab-
oratories (Willmington, Mass.). They were housed under biosafety level III
conditions at Colorado State University and fed standard guinea pig chow and
water ad libitum. After infection, the animals were assessed using a modified
Karnofsky scale (20) for pain and distress in guinea pigs.
Bacterial infections. M. tuberculosis Erdman (TMCC 107) was previously
grown to early mid-log phase from low-passage seed lots in Proskauer-Beck
liquid medium containing 0.05% Tween 80. Cultures were aliquoted into 1-ml
tubes and frozen at ⫺70°C until used. Thawed aliquots were diluted in double-
distilled sterile water to the desired inoculum concentrations. Guinea pigs were
infected via the aerosol route with a low dose of bacteria. Briefly, the nebulizer
compartment of a Middlebrook airborne-infection apparatus (Glas-Col, Terre
Haute, Ind.) was filled with 5 ml of distilled water containing a suspension of
bacteria known to deliver approximately 20 to 50 bacteria into the lungs.
Enumeration of bacteria. Groups of two guinea pigs were killed humanely at
11, 21, 31, 71, and 93 days after aerosol infection with M. tuberculosis strain
Erdman. Only one animal survived to and was examined at 99 days postinfection.
FIG. 1. Course of infection in guinea pigs exposed to aerosol in-

Downloaded from http://iai.asm.org/ on March 3, 2014 by guest


At necropsy, all lung lobes were removed from the thorax individually to enable
fection with M. tuberculosis. At the indicated time points, the right
separate manipulations with each lobe. The number of viable bacteria in the
cranial lung lobe was removed for enumeration of bacterial numbers.
lungs was monitored over time by plating serial 10-fold dilutions of right cranial
Data are expressed as the mean number of viable bacteria (CFU) and
lung lobe homogenates onto nutrient Middlebrook 7H11 agar. The bacterial
the standard error of the mean (where error is greater than 0.1).
colony formations were counted after 21 days of incubation at 37°C under 5%
CO2. The data were expressed as the log10 of the mean number of bacteria
recovered.
Histological testing. The right middle lung lobe was first slowly infused RESULTS
through the major vessels at the hilus with 10% neutral buffered formalin. This
was to prevent alveolar collapse and assist in morphometry. The lobe was then Course of M. tuberculosis infection in the lungs of infected
submerged in the formalin. After a minimum of 48 h, the tissue was prepared and guinea pigs. The course of the infection after aerosol exposure
sectioned for light microscopy, with lobe orientation designed to allow the is shown in Fig. 1. As anticipated, the infection increased
maximum surface area to be seen. Consecutive sections were stained with he- progressively for the first 3 weeks and then stabilized into an
matoxylin and eosin and with Ziehl-Neelsen stain for the detection of acid-fast
apparent chronic state of disease. In the later stages of the
bacilli. Sections were also stained by the Masson trichrome method for the
detection of collagen and by the Fraser-Lendrum method for the detection of
experiment, there was evidence of an increase in the bacterial
fibrin (23). Sections were examined by a veterinary pathologist who had no prior load, consistent with the worsening pathological condition de-
knowledge of the experimental groups and were evaluated at least twice to verify scribed below.
the reproducibility of the observations. Distinct cell populations are seen in guinea pig lung gran-
Immunohistochemistry. Mouse anti-guinea pig monoclonal antibodies specific ulomas after aerosol infection with M. tuberculosis. The lesions
for guinea pig CD4 (clone CT7) and CD8 (clone CT6) were purchased from
that formed in response to the M. tuberculosis infection over
Serotec (Oxford, England). In each case, the Serotec mouse immunoglobulin G1
(IgG1)-negative control (clone W3/25) was used as the isotype control. F(ab⬘)2
time could generally be resolved into four histological stages,
rabbit anti-mouse Ig-biotin, also from Serotec was used as the secondary anti- which are summarized in Table 1. Examples of each stage are
body. At necropsy, the left cranial lung lobe was first slowly infused with a 20% shown in Fig. 2. Detailed descriptions of the four stages are
OCT (Tissue-Tek, Inc. Torrance, Calif.)–phosphate-buffered saline solution listed below.
through the major vessels at the hilus. The samples were then placed in a tissue (i) Stage 1. By 11 days after infection, small (up to approx-
mold, completely surrounded by OCT, frozen in a bath of liquid nitrogen, and
imately 500 ␮m in diameter), discrete, round lesions composed
stored at ⫺70°C until used. Serial sections from each lung, 5 ␮m thick, were cut
in a cryotome (CM 1850; Leica, Bannockburn, Ill.) by employing the Instrumed- of tight accumulations of epithelioid macrophages admixed
ics Inc. (Hackensack, N.J.) tape transfer system, fixed in cold acetone for 5 to 10 with granulocytes could be seen (Fig. 2A and B). These cells
min, and then air dried. Next, the sections were washed in APK buffer solution have distinct eosinophilic cytoplasmic granules and are some-
(Vetana Medical Systems, Tucson, Ariz.) for 15 to 20 min and incubated in a 1:50 times referred to as pseudoeosinophils (28). Similar numbers
(CD4) or 1:100 (CD8) solution of Protein Block goat serum (Biogenex, San of lymphocytes were also present. The lesions were most com-
Ramon, Calif.) and primary antibody for 30 min. The sections were then placed
on a Nexus automated immunostainer (Ventana Medical Systems). The labeled
monly seen in the parenchyma, close to major airways and
avidin-alkaline phosphatase and Fast red/napthol detection kit was employed. blood vessels, effacing and expanding alveolar septa.
The secondary antibody was incubated for 30 min at room temperature. Sections (ii) Stage 2. In stage 2 (Fig. 2C and D), at 21 days postin-
were counterstained with Meyer’s hematoxylin. Sections of spleen were also fection, scattered discrete, round lesions with a similar distri-
examined to act as positive controls. bution to those in stage 1 infection were seen. The diameter of
Detection of apoptosis. The ApopTag Plus peroxidase in situ apoptosis detec-
each lesion was substantially increased (up to approximately
tion kit S7101 (Intergen, Purchase, N.Y.) was used to detect apoptotic cells in
paraffin-embedded tissue. Two or three 5-␮m sections of the same tissue used for
1 mm). While epithelioid macrophages made up the bulk of
histological analysis were analyzed per time point. these foci, neutrophils and lymphocytes were also seen scat-
Photomicroscopy and morphometry. Photomicroscopy was performed with an tered throughout the lesion except in the central core. Occa-
Olympus AH-2 microscope linked to a Sony SKC-DK5 digital camera and Adobe sionally, a thin band of lymphocytes was visible at the margin
Photoshop 6.0 software. In an attempt to further classify the pulmonary lesions, of the granuloma. The core of these lesions was characterized
the granuloma fraction was calculated from each of the hematoxylin-and-eosin-
by a small but distinct accumulation of karyorrhectic debris
stained histological slides. In brief, using Metamorph software (Metamorph
4.5r5; Universal Imaging Corp.), the perceived inflammatory area was deter- admixed with a fibrillar eosinophilic material. Similar eosino-
mined by outlining the affected tissue on the captured images and expressing this philic deposits were present randomly within the tight aggre-
as a percentage of the total area of the observed lung tissue. gate of cells surrounding the core. Additionally, granuloma-
866 TURNER ET AL. INFECT. IMMUN.

Downloaded from http://iai.asm.org/ on March 3, 2014 by guest

FIG. 2. Representative photomicrographs of the four stages of pulmonary granuloma development. (A) Stage 1 granuloma (day 11). A single
discrete round focus of macrophages admixed with granulocytes and small numbers of lymphocytes adjacent to a large blood vessel and airway.
a, artery; b, bronchiole; ca, cartilage. Bar, 100 ␮m. (B) Stage 1 granuloma; area delineated by the black rectangle in panel A. Note the abundance
of granulocytes (arrows) intermixed with epithelioid macrophages and the lack of normal alveolar structure. Bar, 10 ␮m. (C) Stage 2 granuloma
(day 21). A single discrete round focus of macrophages with an eosinophilic necrotic core (c) abutting a bronchiole (b). Bar, 100 ␮m. (D) Stage
2 granuloma; area delineated by the black rectangle in panel C. Note the abundance of neutrophils (arrows) intermixed with epithelioid macro-
phages and lymphocytes. Karyorrhectic debris is present in and around the core (c), and there are multiple small scattered islands of fibrin (ⴱ).
Bar, 10 ␮m. (E) Stage 3 granuloma (day 31). A discrete round lesion with a distinct necrotic core (c) is surrounded by a layer of epithelioid and
VOL. 71, 2003 PATHOLOGY OF TUBERCULOSIS IN GUINEA PIGS 867

TABLE 1. Four-stage categorization of the pulmonary lesions in infected guinea pigsa


Lesion Extent of lesions Macrophage
Lymphocyte organization Granulocyte localization Stage sequelae
stage (% GF)b types

1 Single (11: 0.1, Histiocytes, Few, scattered Prominent, scattered Alveolitis


0.1) epithelioid

2 Multifocal (21: Histiocytes, Few, scattered, with small Prominent, scattered around Karyorrhectic debris, fibrin
4.9, 19.1; 31: epithelioid aggregates at margin and in the core deposition in the core
7.1, 16.7) and surrounding mantle;
fibroplasia;
granulomatous
lymphadenitis

3 Multifocal (71: Epithelioid, Scattered plus a Prominent, scattered around “Classical” granuloma;
45.7, 51.8) foamy prominent halo and in the core necrosis and fibroplasia;
surrounding granulomatous
macrophages lymphadenitis

Downloaded from http://iai.asm.org/ on March 3, 2014 by guest


4 Coalescing (93: Epithelioid, Scattered aggregates or Prominent, scattered Necrosis, mineralization,
42.9, 52.1; 99: foamy individual cells throughout macrophage marked fibroplasia;
56.1) focus, around and in the multinucleated giant
core, and amid airway cells; airway epithelium
debris erosion and purulent
airway exudate;
granulomatous
lymphadenitis
a
Hematoxylin-and-eosin-stained lung sections were examined by standard light microscopy, and several parameters of the lesions were characterized. The parameters
included morphometric assessment of the extent of the pulmonary involvement (granuloma fraction), the predominant macrophage types present, and the overall
arrangement of lymphocytes and neutrophils in the lesions. Several sequelae of each stage were also noted in the infected lungs. Each lesion in a section was
characterized individually, and stages were defined which encompassed the different types of lesions that were present.
b
Bold values represent days postinfection. Values following these are percent granuloma fractions (GF) from individual animals.

tous lymphadenitis was present with increasing prominence bacilli were often detected both within the macrophages effac-
from this stage on. ing the lung parenchyma and free within the suppurative air-
(iii) Stage 3. By day 31, the lesion had taken the form of the way debris (data not shown).
“classical granuloma,” with a diameter of up to 2 mm (Fig. 2E Localization of lymphocyte subsets by immunohistochemis-
and F). There was a prominent central necrotic eosinophilic try. The distribution of CD4⫹ and CD8⫹ T cells in the devel-
core, which was packed with karyorrhectic debris. The core was oping lesions is shown in Fig. 3. On day 11, small numbers of
surrounded by epithelioid macrophages and neutrophils, and lymphocytes were evident. There were apparently similar num-
lymphocytes were also present. Beyond this, the lesion con- bers of CD4⫹ and CD8⫹ cells, and there was no evidence of
tained a prominent collar of lymphocytes admixed with cords cellular aggregation. This pattern was similar on days 21 and
of fibrous connective tissue. 31, as the lesions considerably increased in size. As the lesions
(iv) Stage 4. Stage 4 lesions (Fig. 2G and H) represented the degenerated, mineralized lymphocytes could still be detected
end stage spectrum of granuloma evolution (the example but were present in smaller numbers.
shown is from day 93). There was extensive coalescence of Characterization of the fibrotic response. The development
multiple granulomas into a dense sheet of inflammation, often of lesion fibrosis was monitored by using specific staining meth-
effacing more than half of the lung sections. The necrotic cores ods (data not shown). No response was seen until day 21, when
were typically mineralized and surrounded by epithelioid mac- small discrete regions peripheral to the core and extending
rophages. An elaborate fibrous web that had enveloped large toward the outer margin of the granuloma stained positive for
foamy macrophages marked the previous outer margin of the fibrin, indicating vascular damage. Collagen was also detected
granulomas. Neutrophils and lymphocytes were randomly forming irregular cords around cells adjacent to the core. By
spread at all levels of the inflammation. A few multinucleated day 31, the prominent lymphocyte collar now present was in-
giant cells were scattered throughout the lesions. Acid-fast terlaced by bands of collagen of variable thickness whereas

foamy macrophages. This in turn is surrounded by a prominent basophilic layer of lymphocytes (ⴱ), which are admixed with macrophages at the
extreme margins of the lesion. A bronchiole (b) is present at one margin. Bar, 100 ␮m. (F) Stage 3 granuloma; area delineated by the black
rectangle in panel E. Note the karyorrhectic debris in and around the core area to the right (c), the dense sheet of epithelioid and foamy
macrophages infiltrated by neutrophils surrounding it, and (on the left) a region rich in lymphocytes, between which there is considerable collagen
deposition (arrow). Bar, 10 ␮m. (G) Stage 4 granuloma (day 93). The core (c) has become mineralized and fractured (arrow). Epithelioid and
foamy macrophages admixed with lymphocytes surround it, enveloped in an elaborate network of fibrosis (f). A medium-sized artery (a) and
bronchiole (b) are present at the margins. Bar, 100 ␮m. (H) Stage 4 granuloma; area of the lesion showing a longitudinal section of a bronchiole
(b) which is blocked with neutrophilic and necrotic debris (arrows) filling the lumen. Dense sheets of macrophages with scattered lymphocytes and
interweaving fibrosis (f) surround the airway. Bar, 100 ␮m. All sections are stained with hematoxylin and eosin.
868 TURNER ET AL. INFECT. IMMUN.

Downloaded from http://iai.asm.org/ on March 3, 2014 by guest

FIG. 3. Representative photomicrographs showing immunohistochemical staining for CD4⫹ (left panels) and CD8⫹ (right panels) T cells
within the four stages of pulmonary granuloma formation: stage 1 (A and B), stage 2 (C and D), stage 3 (E and F), and stage 4 (G and H). For
the top four panels, the scale bar represents 10 ␮m; for the bottom four panels the scale bar represents 100 ␮m. c, necrotic core. Arrows indicate
individual positive cells. Labeled avidin-alkaline phosphatase with the naphthol red method was used for all sections, and hematoxylin counterstain
was used. CD4⫹ T cells were stained with Serotec clone CT7, and CD8⫹ T cells were stained with Serotec clone CT6. Biotin-labeled F(ab⬘)2 rabbit
anti-mouse Ig was used as the secondary antibody.
VOL. 71, 2003 PATHOLOGY OF TUBERCULOSIS IN GUINEA PIGS 869

Downloaded from http://iai.asm.org/ on March 3, 2014 by guest


FIG. 4. Representative photomicrographs of the distribution of apoptotic cells within the four stages of pulmonary granuloma formation.
(A) Stage 1 granuloma. A few scattered single apoptotic cells are indicated by the arrows. Bar, 10 ␮m. (B) Stage 2 granuloma. Bar, 100 ␮m.
(C) Stage 3 granuloma. At this time point, most apoptotic cells tended to be adjacent to the core (c). Bar, 100 ␮m. (D) Stage 4 granuloma. The
arrowheads point to mineral debris at the margin of the core. Bar, 10 ␮m. All sections were stained by the Apo-Tag TUNEL method, using
diamonbenzidine as the chromogen and methyl green as the counterstain.

fibrin deposits were spread throughout the granulomas, as well course of the infection, only a few cells stained positive for
as prominently focused around the core. apoptosis and appeared to be randomly distributed.
By day 93, the structure was dominated by thick bands of
collagen deposition, forming a thick web that appeared to trap DISCUSSION
large numbers of foamy macrophages in addition to completely
surrounding the mineralized core (Fig. 2G). In contrast, fibrin The guinea pig is widely considered the consummate animal
deposition was rarely seen at this stage. model of human tuberculosis, given the similarities in the
Distribution of apoptotic cells. It has been suggested that pathological responses to pulmonary infection; as a result, it is
apoptosis of infected cells may be a major defense mechanism an important tool in the search for new vaccines. The results
against M. tuberculosis. As shown in Fig. 4, very few apoptotic shown here are the first attempt to provide a comprehensive
cells could be detected at any of the four stages of the disease description of the stages of the disease in concert with immu-
process. On days 11 through 31, some apoptotic cells could be nohistochemical analysis documenting the influx of CD4⫹ and
seen close to the developing core lesion, but later during the CD8⫹ T cells into the developing lesions. In several subse-
870 TURNER ET AL. INFECT. IMMUN.

quent experiments, it was shown that this progression was 140 days after exposure. We propose that at least two mecha-
evident and reproducible. The results of these studies raise a nisms underlie this event. First, the size of the lesion consoli-
variety of issues that challenge the current dogma. dates the lung lobe and interferes with efficient gas exchange.
The current dogma to explain the pathological process in the Second, the dissemination of the infection via debris deposi-
guinea pig and rabbit is based on the historic studies by Lurie and tion in the airways results in fresh ingestion by monocytes and
more recently by Dannenberg (8–11). In that model, it is held that the subsequent triggering of gamma interferon production by
the initial presence of the bacilli causes no detectable tissue dam- memory T cells. As a result, activated macrophages secrete
age at first. Then, as cell-mediated immunity is triggered, “exces- large quantities of tumor necrosis factor alpha, leading to the
sive delayed-type hypersensitivity” mediating the cellular influx weight loss invariably seen for a week or so prior to death of
and inflammation, in concert with cytolytic T-cell activity, leads to the animal.
the necrotic degeneration of the center of the lesion. It was fur- Both M. bovis BCG and several new candidate vaccines are
ther proposed that the development of this caseous necrosis ini- highly protective in the guinea pig model, which seems to suggest
tially limits the growth of extracellular bacteria, until (in the rab- that this animal is capable of generating a potent memory T-cell
bit) the lesion liquefies and the animal dies. response (2, 19). This in turn explains the very rapid lymphocytic
In this paper we propose a new hypothesis, which differs con- response seen in vaccinated animals, which would be needed to
siderably from the Lurie-Dannenberg model. It is apparent to us prevent the development of the central necrotic core. As one of us

Downloaded from http://iai.asm.org/ on March 3, 2014 by guest


that there is a very rapid response to the infection despite the very has suggested elsewhere (32), this appears to be an important
low dose, and hence it is highly probable that this response is parameter of effective vaccination in this animal model.
innate in nature. Given the preponderance of macrophages, Another important finding here was the observation that
which are already present in significant numbers in the lesion by very few lymphocytes stained positive for apoptosis, suggesting
day 10, this may involve CD1- or Toll receptor-mediated mech- that this mechanism does not play significant role in the disease
anisms (3, 5, 25, 26, 35, 46). In addition, however, there are also process. This finding is rather contrary to the bulk of the
obvious and prominent pockets of granulocytes; these may rep- literature (7, 15, 17, 21, 22, 27, 29, 38, 47), which holds that
resent a double-edged sword in that they may be contributing to apoptosis of both T cells and infected macrophages is an im-
early protection, as suggested previously (1, 34), but their accu- portant defense mechanism, although we note that most of
mulation in response to local tissue damage and their own sub- these observations were all made in vitro.
sequent degranulation (perhaps merely due to their short life Finally, although the mouse and guinea pig are considered
span) may also contribute to the local pathological process. Given to be quite different models of tuberculosis, there are also
the presence of these eosinophil-like cells throughout the early some similarities. If one regards the course of the disease in
response, the data suggests that these cells are continuously ac- the guinea pig as having a stage of chronic disease followed by
cumulating in the lesions over this period. reactivation, then the fact that the latter stage is predisposed by
It is possible that this innate immune response, which happens much earlier events has certain similarities to the situation in
very early during the course of the infection, may be the trigger to various inbred mouse strains that are also prone to this event
the initial development of the characteristic central necrotic core. (45). Moreover, both animal models show evidence of a very
This structure is already very obvious by day 21, which seems to efficient fibrotic response leading to deposition of collagen to
indicate that it has begun development long before the emer- provide some degree of integrity to the granuloma (30, 36).
gence of the acquired response. This latter response seems to Where there appear to be some interesting differences are in
peak about 10 days later and is associated with a large number of terms of the lymphocyte response. We have previously pointed
lymphocytes (a mixture of CD4⫹ and CD8⫹ cells) entering the to differences in terms of the propensity of mouse lymphocytes
lesion and forming a layer of cells peripheral to the central core. to accumulate toward the center of the granuloma (16, 30), and
Moreover, in this region there are multiple scattered areas of it may be that lymphocytes in the guinea pig occupy a more
fibrin deposition, which are suggestive of vascular permeability, peripheral position simply because of the presence of the cen-
which in turn would create local hypoxic conditions and cause the tral necrosis. We have described clear distinctions between the
lesion to further increase in size. In contrast, there was no evi- CD4 and CD8 responses in mice, with CD4⫹ cells forming
dence for the hypothesis that the central core was promoted by large aggregations dominating the granulomas and with CD8⫹
cytolytic T cells, since there were no aggregates of either CD4⫹ or cells being more sparse and distributed more toward the pe-
CD8⫹ cells adjacent to the lesion. Accordingly, our new hypoth- riphery of the lesion (16). These data, when viewed with our
esis suggests that these very early mechanisms of innate immunity data on CD8 gene knockout mice (43), are consistent with an
resulted in a process of irreversible damage that the subsequent immunosurveillance role for the CD8 population.
emergence of the acquired response was too slow to prevent. In the guinea pig, the distributions of the two T-cell subsets
In a sense, moreover, the period of chronic disease seen seem to be very different. Although large numbers of lympho-
between about days 31 and 71 might also be controlled in part cytes entered the lesions by day 31, they remained a fairly even
by innate mechanisms. There is no evidence that the process of randomized mixture of CD4⫹ and CD8⫹ cells, with no evi-
mineralization has an immune basis, and the substantial fi- dence of any cellular aggregation. In addition, they did not
brotic response could also be regarded as a primitive event appear to collectively represent the entire population of lym-
designed to wall off the lesion. Unfortunately, however, this phocytes, suggesting that the additional cells might be B cells
process of consolidation is itself damaging, allowing erosion of and other cells, as previously seen in the mouse model (4, 16,
cellular debris (including extracellular bacteria) into surround- 44). Whether this implies that CD8⫹ cells play a more prom-
ing airways. inent role in the early protective response in the guinea pig
These guinea pigs succumb to the disease at about 100 to than in the mouse is impossible to say at this point.
VOL. 71, 2003 PATHOLOGY OF TUBERCULOSIS IN GUINEA PIGS 871

ACKNOWLEDGMENTS tuberculosis promotes human alveolar macrophage apoptosis. Infect. Immun.


65:298–304.
This work was supported by U.S. Public Health Service grants AI- 22. Klingler, K., K. M. Tchou-Wong, O. Brandli, C. Aston, R. Kim, C. Chi, and
44072 and AI-45707 from NIAID, NIH. W. N. Rom. 1997. Effects of mycobacteria on regulation of apoptosis in
We thank Andrea Cooper, Tony Frank, and Christopher Dascher mononuclear phagocytes. Infect. Immun. 65:5272–5278.
for their helpful advice on this project and David McMurray for his 23. Luna, L. G. 1968. Manual of histologic staining methods of the Armed
continued encouragement. Forces Institute of Pathology, 3rd ed. McGraw-Hill, New York, N.Y.
24. McMurray, D. N., F. M. Collins, A. M. Dannenberg, Jr., and D. W. Smith.
REFERENCES 1996. Pathogenesis of experimental tuberculosis in animal models. Curr.
Top. Microbiol. Immunol. 215:157–179.
1. Appelberg, R., A. G. Castro, S. Gomes, J. Pedrosa, and M. T. Silva. 1995.
25. Means, T. K., S. Wang, E. Lien, A. Yoshimura, D. T. Golenbock, and M. J.
Susceptibility of beige mice to Mycobacterium avium infection:role of neu-
Fenton. 1999. Human toll-like receptors mediate cellular activation by My-
trophils. Infect. Immun. 63:3381–3387.
cobacterium tuberculosis. J. Immunol. 163:3920–3927.
2. Baldwin, S., D. D’Souza, A. Roberts, B. Kelly, A. Frank, M. Lui, J. Ulmer, K.
26. Modlin, R. L., H. D. Brightbill, and P. J. Godowski. 1999. The toll of innate
Huygen, D. McMurray, and I. Orme. 1997. Evaluation of new vaccines in
immunity on microbial pathogens. N. Engl. J. Med. 340:1834–1835.
mouse and guinea pig models of tuberculosis. Infect. Immun. 66:2951–2959.
27. Molloy, A., P. Laochumroonvorapong, and G. Kaplan. 1994. Apoptosis, but
3. Bendelac, A., M. N. Rivera, S. H. Park, and J. H. Roark. 1997. Mouse
not necrosis, of infected monocytes is coupled with killing of intracellular
CD1-specific autoreactive T cells. Development, specificity and function.
bacillus Calmette-Guerin. J. Exp. Med. 180:1499–1509.
Annu. Rev. Immunol. 15:535–550.
28. Moore, D. M. 2000. Hematology of the guinea pig (Cavia porcellus), p. 1107.
4. Bosio, C. M., D. Gardner, and K. L. Elkins. 2000. Infection of B cell-deficient
In C. Feldman, M. Zinkl, and J. Jain (ed.), Schalm’s veterinary hematology,
mice with CDC 1551, a clinical isolate of Mycobacterium tuberculosis: delay
5th ed. Lippincot, Williams & Wilkins, Baltimore, Md.
in dissemination and development of lung pathology. J. Immunol. 164:6417–
29.

Downloaded from http://iai.asm.org/ on March 3, 2014 by guest


6425. Oddo, M., T. Renno, A. Attinger, T. Bakker, H. R. MacDonald, and P. R. A.
5. Brightbill, H. D., D. H. Libraty, S. R. Krutzik, R. B. Yang, J. T. Belisle, J. R. Meylan. 1998. Fas ligand-induced apoptosis of infected human macrophages
Bleharski, M. Maitland, M. V. Norgard, S. E. Plevy, S. T. Smale, P. J. reduces the viability of intracellular Mycobacterium tuberculosis. J. Immunol.
Brennan, B. R. Bloom, P. J. Godowski, and R. L. Modlin. 1999. Host defense 160:5448–5454.
mechanisms triggered by microbial lipoproteins through toll-like receptors. 30. Orme, I. M. 1998. The immunopathogenesis of tuberculosis: a new working
Science 285:732–736. hypothesis. Trends Microbiol. 6:94–97.
6. Cotran, R. S., V. Kumar, and T. Collins. 1999. Robbins pathologic basis of 31. Orme, I. M. 2001. The search for new vaccines against tuberculosis. J. Leu-
disease, 6th ed. The W. B. Saunders Co., Philadelphia, Pa. koc. Biol. 70:1–10.
7. Dalton, D. K., L. Haynes, C. Q. Chu, S. L. Swain, and S. Wittmer. 2000. 32. Orme, I. M. 1999. Vaccination against tuberculosis: recent progress. Adv.
Interferon gamma eliminates responding CD4 T cells during mycobacterial Vet. Med. 41:135–143.
infection by inducing apoptosis of activated CD4 T cells. J. Exp. Med. 33. Orme, I. M., D. N. McMurray, and J. T. Belisle. 2001. Tuberculosis vaccine
192:117–122. development: recent progress. Trends Microbiol. 9:115–118.
8. Dannenberg, A. M., Jr. 1991. Delayed-type hypersensitivity and cell-mediat- 34. Pedrosa, J., B. M. Saunders, R. Appelberg, I. M. Orme, M. T. Silva, and
ed immunity in the pathogenesis of tuberculosis. Immunol. Today 12:228–233. A. M. Cooper. 2000. Neutrophils play a protective, non-phagocytic, role in
9. Dannenberg, A. M., Jr. 1989. Immune mechanisms in the pathogenesis of systemic Mycobacterium tuberculosis infection of mice. Infect. Immun. 68:
pulmonary tuberculosis. Rev. Infect. Dis. 11(Suppl. 2):S369–S378. 577–583.
10. Dannenberg, A. M., Jr. 1993. Immunopathogenesis of pulmonary tubercu- 35. Porcelli, S. A., and R. L. Modlin. 1999. The CD1 system: antigen-presenting
losis. Hosp. Pract. (Off. Ed.) 28:51–58. molecules for T cell recognition of lipids and glycolipids. Annu. Rev. Immu-
11. Dannenberg, A. M., Jr. 1994. Roles of cytotoxic delayed-type hypersensitivity nol. 17:297–329.
and macrophage- activating cell-mediated immunity in the pathogenesis of 36. Rhoades, E. R., A. A. Frank, and I. M. Orme. 1997. Progression of chronic
tuberculosis. Immunobiology 191:461–473. pulmonary tuberculosis in mice aerogenically infected with virulent Myco-
12. Dye, C., S. Scheele, P. Dolin, V. Pathania, and M. C. Raviglione. 1999. bacterium tuberculosis. Tubercle Lung Dis. 78:57–66.
Consensus statement. Global burden of tuberculosis: estimated incidence, 37. Ridley, D. S., and M. J. Ridley. 1987. Rationale for the histological spectrum
prevalence, and mortality by country. WHO Global Surveillance and Mon- of tuberculosis. A basis for classification. Pathology 19:186–192.
itoring Project. JAMA 282:677–686. 38. Rojas, M., M. Olivier, P. Gros, L. F. Barrera, and L. F. Garcia. 1999. TNF-
13. Fenhalls, G., L. Stevens, J. Bezuidenhout, G. E. Amphlett, K. Duncan, P. alpha and IL-10 modulate the induction of apoptosis by virulent Mycobac-
Bardin, and P. T. Lukey. 2002. Distribution of IFN-gamma, IL-4 and TNF- terium tuberculosis in murine macrophages. J. Immunol. 162:6122–6131.
alpha protein and CD8 T cells producing IL-12p40 mRNA in human lung 39. Saunders, B. M., and A. M. Cooper. 2000. Restraining mycobacteria: role of
tuberculous granulomas. Immunology 105:325–335. granulomas in mycobacterial infections. Immunol. Cell Biol. 78:334–341.
14. Flynn, J. L., and J. Chan. 2001. Tuberculosis: latency and reactivation. 40. Smith, D. W., and G. E. Harding. 1977. Animal model of human disease.
Infect. Immun. 69:4195–4201. Pulmonary tuberculosis. Animal model: experimental airborne tuberculosis
15. Fratazzi, C., R. D. Arbeit, C. Carini, M. K. Balcewicz-Sablinska, J. Keane, H. in the guinea pig. Am. J. Pathol. 89:273–276.
Kornfeld, and H. G. Remold. 1999. Macrophage apoptosis in mycobacterial 41. Smith, D. W., D. N. McMurray, E. H. Wiegeshaus, A. A. Grover, and G. E.
infections. J. Leukoc. Biol. 66:763–764. Harding. 1970. Host-parasite relationships in experimental airborne tuber-
16. Gonzalez-Juarrero, M., O. C. Turner, J. Turner, P. Marietta, J. V. Brooks, culosis. IV. Early events in the course of infection in vaccinated and nonva-
and I. M. Orme. 2001. Temporal and spatial arrangement of lymphocytes ccinated guinea pigs. Am. Rev. Respir. Dis. 102:937–949.
within lung granulomas induced by aerosol infection with Mycobacterium 42. Smith, D. W., and E. H. Wiegeshaus. 1989. What animal models can teach us
tuberculosis. Infect. Immun. 69:1722–1728. about the pathogenesis of tuberculosis in humans. Rev. Infect. Dis. 11(Suppl.
17. Hirsch, C. S., Z. Toossi, J. L. Johnson, H. Luzze, L. Ntambi, P. Peters, M. 2):S385–S393.
McHugh, A. Okwera, M. Joloba, P. Mugyenyi, R. D. Mugerwa, P. Terebuh, 43. Turner, J., C. D. D’Souza, J. E. Pearl, P. Marietta, M. Noel, A. A. Frank, R.
and J. J. Ellner. 2001. Augmentation of apoptosis and interferon-gamma Appelberg, I. M. Orme, and A. M. Cooper. 2001. CD8- and CD95/95L-
production at sites of active Mycobacterium tuberculosis infection in human dependent mechanisms of resistance in mice with chronic pulmonary tuber-
tuberculosis. J. Infect. Dis. 183:779–788. culosis. Am. J. Respir. Cell Mol. Biol. 24:203–209.
18. Hopewell, P. C. 1994. Overview of clinical tuberculosis, p. 25–46. In B. R. 44. Turner, J., A. A. Frank, J. V. Brooks, M. Gonzalez-Juarrero, and I. M. Orme.
Bloom (ed.), Tuberculosis: pathogenesis, protection, and control. ASM 2001. The progression of chronic tuberculosis in the mouse does not require the
Press, Washington, D.C. participation of B lymphocytes or interleukin-4. Exp. Gerontol. 36:537–545.
19. Horwitz, M. A., G. Harth, B. J. Dillon, and S. Maslesa-Galic. 2000. Recom- 45. Turner, J., M. Gonzalez-Juarrero, B. M. Saunders, J. V. Brooks, P. Marietta,
binant bacillus calmette-guerin (BCG) vaccines expressing the Mycobacte- D. L. Ellis, A. A. Frank, A. M. Cooper, and I. M. Orme. 2001. Immunological
rium tuberculosis 30-kDa major secretory protein induce greater protective basis for reactivation of tuberculosis in mice. Infect. Immun. 69:3264–3270.
immunity against tuberculosis than conventional BCG vaccines in a highly 46. Underhill, D. M., A. Ozinsky, K. D. Smith, and A. Aderem. 1999. Toll-like
susceptible animal model. Proc. Natl. Acad. Sci. USA 97:13853–13858. receptor-2 mediates mycobacteria-induced proinflammatory signaling in
20. Karnofsky, D. A., W. H. Abelmann, L. F. Craver, and J. H. Burchenal. 1948. macrophages. Proc. Natl. Acad. Sci. USA 96:14459–14463.
The use of nitrogen mustards in the palliative treatment of cancer. Cancer 47. Watson, V. E., L. L. Hill, L. B. Owen-Schaub, D. W. Davis, D. J. McConkey,
1:634–656. C. Jagannath, R. L. Hunter, Jr., and J. K. Actor. 2000. Apoptosis in myco-
21. Keane, J., M. K. Balcewicz-Sablinska, H. G. Remold, G. L. Chupp, B. B. bacterium tuberculosis infection in mice exhibiting varied immunopathology.
Meek, M. J. Fenton, and H. Kornfeld. 1997. Infection by Mycobacterium J. Pathol. 190:211–220.

Editor: S. H. E. Kaufmann

You might also like