Download as pdf or txt
Download as pdf or txt
You are on page 1of 54

Interstitial Lung Disease 1st Edition

Edition Harold R Collard And Luca


Richeldi (Auth.) - eBook PDF
Go to download the full and correct content document:
https://ebooksecure.com/download/interstitial-lung-disease-ebook-pdf/
More products digital (pdf, epub, mobi) instant
download maybe you interests ...

3D Lung Models for Regenerating Lung Tissue 1st Edition


- eBook PDF

https://ebooksecure.com/download/3d-lung-models-for-regenerating-
lung-tissue-ebook-pdf/

Developing the Digital Lung: From First Lung CT to


Clinical AI 1st Edition - eBook PDF

https://ebooksecure.com/download/developing-the-digital-lung-
from-first-lung-ct-to-clinical-ai-ebook-pdf/

Developing the Digital Lung: From First Lung CT to


Clinical AI 1st Edition John D. Newell - eBook PDF

https://ebooksecure.com/download/developing-the-digital-lung-
from-first-lung-ct-to-clinical-ai-ebook-pdf-2/

Progress in Heterocyclic Chemistry Volume 29 1st


Edition - eBook PDF

https://ebooksecure.com/download/progress-in-heterocyclic-
chemistry-ebook-pdf/
Microalgae in Health and Disease Prevention 1st Edition
- eBook PDF

https://ebooksecure.com/download/microalgae-in-health-and-
disease-prevention-ebook-pdf/

Dry Eye Disease 1st Edition - eBook PDF

https://ebooksecure.com/download/dry-eye-disease-ebook-pdf/

Chronic Coronary Artery Disease: A Companion to


Braunwald's Heart Disease 1st Edition- eBook PDF

https://ebooksecure.com/download/chronic-coronary-artery-disease-
a-companion-to-braunwalds-heart-disease-ebook-pdf/

The Newborn Lung: Neonatology Questions and


Controversies 3rd Edition Eduardo Bancalari - eBook PDF

https://ebooksecure.com/download/the-newborn-lung-neonatology-
questions-and-controversies-ebook-pdf/

Epigenetics in Cardiovascular Disease 1st edition -


eBook PDF

https://ebooksecure.com/download/epigenetics-in-cardiovascular-
disease-ebook-pdf/
Interstitial Lung
Disease
HAROLD R. COLLARD, MD
Division of Pulmonary and Critical Care Medicine
Department of Medicine
University of California San Francisco
San Francisco, CA, United States

LUCA RICHELDI, MD, PhD


Division of Pulmonary Medicine
Agostino Gemelli University Hospital
Catholic University of the Sacred Heart
Rome, Italy
1600 John F. Kennedy Blvd.
Ste 1800
Philadelphia, PA 19103-2899

INTERSTITIAL LUNG DISEASE ISBN: 978-0-323-48024-6


Copyright © 2018 by Elsevier, Inc. All rights reserved.

No part of this publication may be reproduced or transmitted in any form or by any means, electronic
or mechanical, including photocopying, recording, or any information storage and retrieval system,
without permission in writing from the publisher. Details on how to seek permission, further infor-
mation about the Publisher’s permissions policies and our arrangements with organizations such
as the Copyright Clearance Center and the Copyright Licensing Agency, can be found at our website:
www.elsevier.com/permissions.

This book and the individual contributions contained in it are protected under copyright by the Publisher
(other than as may be noted herein).

Notices

Knowledge and best practice in this field are constantly changing. As new research and experience
broaden our understanding, changes in research methods, professional practices, or medical treat-
ment may become necessary.
Practitioners and researchers must always rely on their own experience and knowledge in evalu-
ating and using any information, methods, compounds, or experiments described herein. In using
such information or methods they should be mindful of their own safety and the safety of others,
including parties for whom they have a professional responsibility.
With respect to any drug or pharmaceutical products identified, readers are advised to check the
most current information provided (i) on procedures featured or (ii) by the manufacturer of each
product to be administered, to verify the recommended dose or formula, the method and duration
of administration, and contraindications. It is the responsibility of practitioners, relying on their
own experience and knowledge of their patients, to make diagnoses, to determine dosages and the
best treatment for each individual patient, and to take all appropriate safety precautions.
To the fullest extent of the law, neither the Publisher nor the authors, contributors, or editors,
assume any liability for any injury and/or damage to persons or property as a matter of products
liability, negligence or otherwise, or from any use or operation of any methods, products, instruc-
tions, or ideas contained in the material herein.

International Standard Book Number: 978-0-323-48024-6

Content Strategist: Patrick Manley


Content Development Specialist: Meredith Clinton
Design Direction: Renee Duenow

Printed in United States of America

Last digit is the print number: 9 8 7 6 5 4 3 2 1


Contributors

Danielle Antin-Ozerkis, MD Robert J. Homer, MD, PhD


Associate Professor of Medicine Professor of Pathology and Medicine (Pulmonary)
Director, Yale Interstitial Lung Disease Program Yale University School of Medicine
Pulmonary and Critical Care Medicine Section New Haven, CT, United States
Department of Internal Medicine
Yale University School of Medicine Kirk D. Jones, MD
New Haven, CT, United States Professor
Department of Pathology
Jürgen Behr, MD University of California, San Francisco, School of
Department of Internal Medicine V Medicine
University of Munich San Francisco, CA, United States
Asklepios Kliniken München-Gauting Comprehensive
Pneumology Center Dong Soon Kim, MD, PhD
Member, German Center for Lung Research Emeritus Professor
Munich, Germany University of Ulsan
Seoul, South Korea
Stefania Cerri, MD, PhD
Center for Rare Lung Disease Kathleen Oare Lindell, PhD, RN
University Hospital of Modena Research Assistant Professor of Medicine
Modena, Italy Clinical Nurse Specialist
Dorothy P. & Richard P. Simmons Center for
Sonye K. Danoff, MD, PhD Interstitial Lung Disease
Department of Medicine Pulmonary, Allergy and Critical Care Medicine
Pulmonary and Critical Care Medicine The University of Pittsburgh
Johns Hopkins University School of Medicine Pittsburgh, PA, United States
Baltimore, MD, United States
Fabrizio Luppi, MD, PhD
Janine Evans, MD Center for Rare Lung Disease
Associate Professor of Medicine University Hospital of Modena
Rheumatology Section Modena, Italy
Department of Internal Medicine
Yale University School of Medicine Richard A. Matthay, MD
New Haven, CT, United States Boehringer Ingelheim Emeritus Professor of Medicine
and Senior Research Scientist
Charlene D. Fell, MD, MSc, FRCPC Pulmonary and Critical Care Medicine Section
Division of Respirology Department of Internal Medicine
Department of Medicine Yale University School of Medicine
University of Calgary New Haven, CT, United States
Calgary, AB, Canada

Christine Kim Garcia, MD, PhD


McDermott Center for Human Growth and
Development
Department of Internal Medicine, Division of
Pulmonary and Critical Care
University of Texas Southwestern Medical Center
Dallas, TX, United States v
vi CONTRIBUTORS

Amy L. Olson, MD, MSPH Aditi Shah, MD


Assistant Professor Division of Respirology, Critical Care, and Sleep
Department of Medicine Medicine
Division of Pulmonary, Critical Care and Sleep Department of Medicine
Medicine University of Saskatchewan
National Jewish Health Saskatoon, SK, Canada
Denver, CO, United States;
Department of Medicine Leann L. Silhan, MD
Division of Pulmonary Sciences and Critical Care Department of Medicine
Medicine Pulmonary and Critical Care Medicine
University of Colorado, Denver Johns Hopkins University School of Medicine
Aurora, CO, United States Baltimore, MD, United States

Luca Paoletti, MD Paolo Spagnolo, MD, PhD


Assistant Professor Respiratory Disease Unit
Division of Pulmonary and Critical Care Medicine Department of Cardiac, Thoracic, and Vascular
Medical University of South Carolina Sciences
Charleston, SC, United States University of Padua
Padua, Italy
Silvia Puglisi, MD
Regional Referral Centre for Rare Lung Diseases Jeffrey J. Swigris, DO, MS
University of Catania Associate Professor
University-Hospital Policlinico–Vittorio Emanuele Department of Medicine
Catania, Italy Division of Pulmonary, Critical Care and Sleep
Medicine
Luca Richeldi, MD, PhD National Jewish Health
Division of Pulmonary Medicine Denver, CO, United States;
Agostino Gemelli University Hospital Department of Medicine
Catholic University of the Sacred Heart Division of Pulmonary Sciences and Critical Care
Rome, Italy Medicine
University of Colorado, Denver
Ami Rubinowitz, MD Aurora, CO, United States
Associate Professor of Diagnostic Radiology
Chief of Thoracic Imaging Anatoly Urisman, MD, PhD
Department of Diagnostic Radiology and Biomedical Assistant Professor
Imaging Department of Pathology
Yale University School of Medicine University of California, San Francisco, School of
New Haven, CT, United States Medicine
San Francisco, CA, United States
Jay H. Ryu, MD
Division of Pulmonary and Critical Care Medicine Carlo Vancheri, MD, PhD
Mayo Clinic Regional Referral Centre for Rare Lung Diseases
Rochester, MN, United States University of Catania
University-Hospital Policlinico–Vittorio Emanuele
Giacomo Sgalla, MD Catania, Italy
National Institute for Health Research
Respiratory Biomedical Research Unit
University Hospital Southampton
Southampton, United Kingdom
CONTRIBUTORS vii

Robert Vassallo, MD Zulma X. Yunt, MD


Division of Pulmonary and Critical Care Medicine Assistant Professor
Departments of Medicine, Physiology and Biomedical Department of Medicine
Engineering Division of Pulmonary, Critical Care and Sleep
Mayo Clinic Medicine
Rochester, MN, United States National Jewish Health
Denver, CO, United States;
Timothy P.M. Whelan, MD Department of Medicine
Professor of Medicine Division of Pulmonary Sciences and Critical Care
Division of Pulmonary and Critical Care Medicine Medicine
Medical University of South Carolina University of Colorado, Denver
Charleston, SC, United States Aurora, CO, United States
Preface

The interstitial lung diseases (ILDs) are a large and het- congestive heart failure, pneumothorax, or drugs. How-
erogeneous group of disease entities that differ signifi- ever, in many cases the etiology is not certain despite
cantly with respect to presentation, cause, prevention, rigorous searches. The current state of our understand-
therapy, and prognosis. Many of them have no clear eti- ing of this important manifestation of IPF is discussed.
ology. However, in recent years considerable progress Also, the mechanisms of action of current and potential
has been made in our understanding of these entities. IPF therapies are reviewed, with particular reference to
In Interstitial Lung Disease, we discuss the most impor- current disease understanding and to highlight areas of
tant of the ILDs and update the reader about their IPF disease biology that afford attractive targets for the
management. development of the new treatments. The general man-
Arriving at the correct diagnosis often requires cor- agement of a patient with IPF is described, especially
relation with clinical, radiologic, and pathologic find- the management of important comorbidities (e.g., pul-
ings. The diagnostic strategy that should be employed monary hypertension and gastroesophageal reflux) and
when faced with a patient with ILD is described. symptoms (e.g., dyspnea, exercise limitation, fatigue,
Given the important role of high-resolution computed anxiety, mood disturbance, sleep disorders) that dra-
tomography (HRCT) in the diagnosis of ILD, we intro- matically affect IPF patients’ lives. Furthermore, the
duce pulmonologists and clinicians to the imaging increasingly important role of lung transplantation in
appearances of ILDs by providing a pattern approach the treatment of patients with ILD is discussed, espe-
to the evaluation of HRCT. In addition, a focused dis- cially the evidence suggesting that patients with pul-
cussion of the HRCT findings in the common ILDs is monary fibrosis undergoing lung transplantation have
presented. a favorable long-term survival compared with other
Idiopathic pulmonary fibrosis (IPF) is the most disease indications.
common of the idiopathic interstitial pneumonias. Finally, several chapters review important individual
Over the past decade, our thinking about IPF has been ILD entities, including inherited fibrotic lung diseases,
reshaped and guidelines have been revised using an nonspecific interstitial pneumonia, ILD associated with
evidence-based approach. A number of epidemiologic connective tissue diseases, chronic hypersensitivity
studies have identified potential risk factors for IPF. We pneumonitis, and smoking-related ILDs.
summarize the approach to diagnosing IPF and review
its epidemiology. With improved understanding of IPF, Harold R. Collard, MD
we now recognize that there may also be distinct phe- Division of Pulmonary and Critical Care Medicine
notypes among this group of patients. Furthermore, we Department of Medicine
describe how the identification and management of University of California San Francisco
comorbidities may improve the overall quality of life San Francisco, CA, USA
and well-being of these patients. It has become clear
that the clinical course of individual patients with IPF is Luca Richeldi, MD, PhD
variable and that the sudden deterioration of a patient’s Division of Pulmonary Medicine
respiratory condition during a relatively stable course Agostino Gemelli University Hospital
is possible. This deterioration can result from well- Catholic University of the Sacred Heart
known causes such as infection, pulmonary embolism, Rome, Italy

ix
CHAPTER 1

Genetic Interstitial Lung Disease


CHRISTINE KIM GARCIA

KEY POINTS

• The interstitial lung diseases (ILDs), or diffuse parenchymal lung diseases, are a heterogeneous collection of more
than 100 different pulmonary disorders that affect the tissue and spaces surrounding the alveoli.
• Many of the genes involve pathways that lead to altered surfactant metabolism, increased ER stress signaling, and
telomere shortening.
• Genetics provides a molecular framework for explaining phenotypes and, sometimes, provides information that
directly affects patient care.

The interstitial lung diseases (ILDs) include a wide mechanism of disease. A detailed family history also
variety of relatively uncommon disorders. Technologic provides important information about personal and
advances in sequencing and genotyping have led to an family member phenotypes, providing important
explosion of genetic discoveries, shedding new light on clues that may suggest a certain genetic diagnosis. For
the underlying pathogenesis of ILD. New monogenic example, a personal or family history of bone marrow
syndromes have been described, often with clinically failure, early graying, or liver disease in a patient with
diverse and extreme phenotypes, based on the discov- adult-onset pulmonary fibrosis suggests a short telo-
ery of single gene mutations. In addition, many appar- mere syndrome. Younger and more severely affected
ently disparate clinical presentations have been linked individuals in later generations may reflect genetic
together through the discovery of mutations in the same anticipation, which can also be seen in short telomere
gene or mutations in multiple genes sharing a common syndromes. Some diseases show a predisposition for
pathway. These genetic discoveries have only further affecting a certain gender, for example, affected males
increased the number of discrete ILDs. The genetic eti- and asymptomatic carrier females suggest an X-linked
ology provides a molecular framework for the disease disorder.
and provides patients and their treating physicians All ILDs arise from the infiltration of inflamma-
with an explanation of phenotypes that are often seen tory and fibrotic mediators into the lung parenchyma.
across multiple organs. As more cohorts of patients are Very few cells normally reside within the interstitium,
described with these rare syndromes, more information which is the delicate space between the alveolar epi-
will be gleaned about the natural history of disease and thelial cells and the capillary vascular endothelial
best practices for surveillance and treatment. The goal cells. The filling of the interstitial space with inflam-
of this chapter is to summarize (1) genetic syndromes matory cells, activated fibroblasts, and extracellular
involving multiple organs, in which ILD is one of many matrix causes irreversible architectural distortion and
different phenotypes, and (2) genetic disorders in impairs gas exchange. Most ILDs share similar clinical
which ILD is the dominant phenotype. signs and symptoms, including respiratory distress and
As a rule, manifestations of ILD even within a cough. Pulmonary restriction and a decreased diffusion
single gene syndrome are generally characterized by capacity are frequently found, as well as radiographic
a spectrum of clinical presentations, a wide range evidence of parenchymal abnormalities. However, the
in age of onset, and incomplete penetrance. Thus, a radiographic and histopathologic features of ILDs vary
high level of suspicion is needed for many of these widely. Historically, the type of infiltrating cells, the
disorders. Detecting a pattern of inheritance in large, pattern of infiltration (nodular, reticular, alveolar), the
extended kindreds across multiple generations sepa- nature of extracellular protein deposits (collagen, elas-
rated by time and space strongly supports a genetic tin, periodic acid–Schiff [PAS]-positive), the location

1
2 Interstitial Lung Disease

of abnormalities (peripheral, alveolar, peribronchio- and extrathoracic lymphatic structures. The hallmark
lar), the pattern of the fibrotic response (fibroblastic feature of the disease is the radiographically apparent
foci, temporal/spatial homogeneity, or heterogeneity), numerous, round, thin-walled cysts, generally between
and the form of lung destruction (cysts, bronchiectasis, 2 and 60 mm in diameter, which are found throughout
honeycombing) have been used to describe different the lung. Extraparenchymal lung abnormalities include
clinical forms of ILD. Now that genetic underpinnings pneumothorax and chylous pleural effusions. Eighty
of some monogenic ILDs are being established, clas- percent of patients develop a pneumothorax at some
sification by genetic etiology may ultimately supplant time during the course of their disease. Pleural effusions
historical classification schemes. This will occur most are characterized by elevated triglyceride levels and an
readily for those disorders in which the genetic clas- abundance of chylomicrons. Obstructive and mixed
sification predicts specific treatments (e.g., sirolimus obstructive-restrictive ventilator defects are observed.
for tuberous sclerosis complex [TSC]–lymphangioleio- Renal angiomyolipomas occur in ∼30% of sporadic
myomatosis [LAM]). A genetic classification of ILD is LAM patients and in >90% of TSC-LAM patients.1
also advisable for those diseases in which the genetic LAM can occur either in patients with sporadic (non-
information provides information relevant to patient inherited) disease or in 1–3% of patients with TSC, an
care. For example, regular screening may lead to ear- autosomal dominant multisystem disorder character-
lier interventions to remove premalignant renal cancers ized by hamartomas in multiple organ systems, includ-
in patients with Birt-Hogg-Dubé syndrome (BHDS). ing the brain, skin, heart, kidneys, and lungs. The ILD
Similarly, knowledge of a monogenic short telomere manifestation of TSC patients is indistinguishable
syndrome provides prognostic information regarding from sporadic LAM.2 TSC patients who develop LAM
the rate of ILD progression and the nature of specific are often women over 30 years of age who have little
post–lung transplant complications. or no mental retardation. Extrapulmonary features
In this chapter, I focus primarily on disorders caused include facial angiofibromas (or adenoma sebaceum),
by rare mutations and include selected common vari- hypomelanotic macules (which are most easily visual-
ants that significantly increase susceptibility to ILD. ized with a Wood light), rough, yellow thickening of
The nomenclature of diseases follows the genetic clas- skin over the lumbosacral area (shagreen patch), ungual
sification system adopted by the Online Mendelian or periungual fibromas, renal angiomyolipomas, renal
Inheritance in Man (http://www.omim.org). Other pul- cysts, subependymal giant cell astrocytomas, brain
monary genetic diseases are not covered. I refer to other cysts, hamartomas, cardiac rhabdomyomas, dental pits,
excellent resources for reviews of alpha-1-antitrypsin epilepsy, learning difficulties, and mental retardation.
deficiency, cystic fibrosis and CFTR-related disorders, TSC is characterized by autosomal dominant inheri-
primary ciliary dyskinesia, pulmonary capillary or veno- tance. However, about two-thirds of patients have de novo
occlusive disease, pulmonary malformation syndromes, mutations. There are two genes associated with this syn-
and disorders that primarily affect the thoracic cage. drome. Mutations in TSC2 occur more frequently, account-
ing for 75–80% of all cases.3 Mutations in TSC1 are less
common.3 There are no mutational hot spots within these
GENETIC DISORDERS AFFECTING MULTIPLE two genes. In general, TSC2 mutations are associated with
ORGANS, INCLUDING THE LUNG more severe disease, more frequent and severe epilepsy,
Table 1.1 lists the defined genetic disorders that are mental retardation, and cortical tubers. Inactivation of
associated with ILDs, with the lung being only one of both alleles of TSC1 or TSC2 is needed for the develop-
the many different affected organs. In this broad cat- ment of tumors. Loss of heterozygosity is frequently found
egory that encompasses many different disorders, ILD in renal angiomyolipomas and supports Knudson’s two-
may be a more severe or life-threatening manifestation hit model tumor suppressor pathogenesis.4
of disease or one of the more common features associ- Most sporadic LAM results from two somatic muta-
ated with the disease. tions in the TSC2 gene, although rare cases are caused
by germline TSC1 mutations.5–7 In contrast with TSC
Lymphangioleiomyomatosis and Tuberous patients in whom germline mutations are present in
Sclerosis Complex tumor and normal tissue, the mutations in LAM patients
Pulmonary LAM is a rare disease that almost exclu- are generally not present in normal tissues. This sug-
sively affects women. It is caused by proliferating gests that two mutations arise in a precursor cell of ori-
smooth muscle–like cells, which can involve small gin and that the mutation-carrying smooth muscle–like
airways, the pulmonary vasculature, and intrathoracic cells spread to various organs (lungs, kidneys).
CHAPTER 1 Genetic Interstitial Lung Disease 3

TABLE 1.1
Genetic Interstitial Lung Disease (ILD): Disorders Affecting Multiple Organs, Including the Lung
Pulmonary ILD
Disease Inheritance Gene Pathogenesis Involvement Presentation
LAM/tuberous Sporadic, TSC1, TSC2 mTOR ∼100% Multiple cysts
sclerosis AD activation
Birt-Hogg-Dubé AD FLCN Loss of ∼90% Multiple cysts
syndrome folliculin
Dyskeratosis XLR, AD, DKC1, TERC, TERT, Telomere Second Inflammatory
congenita AR NOP10, NHP2, shortening most serious infiltrates and
TINF2, WRAP53, complication interstitial fibrosis
RTEL1, ACD, CTC1,
PARN
RIDDLE syndrome AR RNF168 DSB repair defect 50% Pulmonary fibrosis
Hermansky-Pudlak AR HPS1, AP3B1, Cytoplasmic or- Described Fibrosis, foamy
syndrome HPS3, HPS4, HPS5, ganelle defect for those type II
HPS6, DTNBP1, with HPS1, pneumocytes
BLOC1S3, BLOC1S6 HPS2, and
HPS4
NKX2-1 related AD NPX2-1 Transcription fac- ∼50% Respiratory
disorders tor defect distress, NEHI,
pulmonary fibrosis
Neurofibromatosis AD NF1 Loss of tumor ∼10% Pulmonary
suppressor fibrosis, bullae
Poikiloderma with tendon AD FAM111B Unknown >50% UIP
contractures and
pulmonary fibrosis
ILD, nephrotic AR ITGA3 Integrin defect 100% Abnormal
syndrome, and alveolarization
epidermolysis
bullosa
STING-associated AD TMEM173 Increased >50% Alveolitis with
vasculopathy interferon fibrosis
Autoimmune disease AR ITCH E3 ubiquitin ligase ∼90% NSIP
with facial defect
dysmorphism
Autoimmune interstitial AD COPA ER stress ∼100% Lymphocytic
lung, joint, and kidney infiltration
disease
GATA2 deficiency Sporadic, GATA2 Transcription fac- ∼60% PAP
AD tor defect
CVID syndromes Sporadic, ICOS, CD19, CD20, Variable 55–90% Bronchiectasis,
AR, AD CD21, CD81, TN- antibody GLILD, organizing
FRSF13B, TNFRS- deficiencies pneumonia
F13C, LRBA, IL21,
NFKB1, NFKB2,
IKZF1
Agammaglobulinemia X-linked, BTK, IGHM, IGLL1, B cell defect ∼25% Bronchiectasis
AR, AD CD79A, CD79B,
BLNK, TCF3
Continued
4 Interstitial Lung Disease

TABLE 1.1
Genetic Interstitial Lung Disease (ILD): Disorders Affecting Multiple Organs, Including the Lung—cont’d
Pulmonary ILD
Disease Inheritance Gene Pathogenesis Involvement Presentation
Hyper IgE syndrome Sporadic, STAT3 STAT3 defect, ∼70% Bronchiectasis,
AD Lack of Th17 cells pneumatoceles
Activated PI3K-δ AD PIK3CD Activation of PI3K ∼75% Lymphoid
syndrome aggregates
CTLA4 deficiency AD CTLA4 Activated T cells ∼75% GLILD
X-linked reticulate X-linked POLA1 Increased type 1 ∼100% Bronchiectasis
pigmentary interferons
Gaucher disease, type I AR GBA Deficiency of acid ∼65% Gaucher cell
β-glucosidase infiltration
Niemann-Pick disease, AR SMPD1, NPC1, Deficiency of acid Various GGO and septal
types A, B, C1, and C2 NPC2 sphingomyelinase; thickening
defective intracellu-
lar lipid movement
Lysinuric protein AR SLC7A7 Defect of cationic PAP, septal
intolerance amino acid thickening
transport

AD, autosomal dominant; AR, autosomal recessive; DSB, double-stranded DNA break; GGO, ground-glass opacities; GLILD, granulomatous
lymphocytic interstitial lung disease; mTOR, mammalian target of rapamycin; NEHI, neuroendocrine cell hyperplasia of infancy; NSIP, nonspe-
cific interstitial pneumonia; PAP, pulmonary alveolar proteinosis; UIP, usual interstitial pneumonia; XLR, X-linked recessive.

The proteins encoded by these two genes regulate and represent a forme fruste of BHDS.17,18 Nearly all
the mammalian target of rapamycin (mTOR) path- mutations, including the common mutation hot spot
way.8 Inhibition of the mTOR complex corrects the that introduces a frameshift mutation (c.1285insC
specific molecular defects underlying TSC. mTOR and c.1285delC), predict a truncated protein prod-
inhibitors cause shrinkage of renal or retroperito- uct.15 The pulmonary cysts are generally bilateral, are
neal angiomyolipomas and subependymal giant cell usually located in a subpleural distribution in the
astrocytomas in TSC patients.9–11 In addition, siro- mid- and lower-lung zones, and are of varying sizes,
limus stabilizes lung function, reduces respiratory ranging from round, oval, lentiform, to multisep-
symptoms, and improves the quality of life of LAM tated shapes.19,20 Because the skin findings typically
patients.12 appear in the fourth decade and the renal malignancy
can be a late finding (mean age of 48 years), a spon-
Birt-Hogg-Dubé Syndrome taneous pneumothorax in the second or third decade
This disorder, which is also known as Hornstein- of life is often the presenting manifestation.15,18
Knickenberg syndrome,13 is characterized by the The molecular mechanism of lung cyst formation is
autosomal dominant inheritance of multiple benign incompletely understood.
skin tumors, which are usually characterized as fibro- Genetic testing is indicated for all individuals with sus-
folliculomas. Approximately 90% of affected indi- pected BHDS because the presence of a mutation prompts
viduals have evidence of lung cysts,14 and up to 34% screening for renal cancer for which surgical resection may
develop kidney tumors.15 The disease is caused by be curative. As was described in a past case,21 the pheno-
loss-of-function mutations in the folliculin (FLCN) type of familial spontaneous pneumothorax may be the
gene.16 Two different studies, one using whole- presenting feature for the index case that then leads to the
genome linkage analysis and the other a candidate identification of an occult renal malignancy in a family
gene approach, independently found that kindreds member with a FLCN mutation. All first-degree relatives
presenting solely with familial spontaneous pneu- have a 50% chance of having the same germline FLCN
mothorax and/or lung cysts have mutations in FLCN mutation.
CHAPTER 1 Genetic Interstitial Lung Disease 5

Dyskeratosis Congenita with the catalytically active RNA and protein compo-
Dyskeratosis congenita (DC) is a rare multisystem dis- nent of telomerase.34 Mutations in both genes encod-
order characterized by the classic triad of a lacy reticular ing telomerase (hTR, encoded by TERC gene and TERT)
pigmentation on the upper chest and neck, nail dystro- have been found in patients with autosomal dominant
phy, and oral leukoplakia. The prevalence is approxi- DC35,36; biallelic mutations in TERT have been found
mately 1 in 1,000,000, with death occurring at a median in patients with autosomal recessive DC.37,38 Germ-
age of 16 years.22 Patients are usually healthy at birth line mutations in TINF2, a component of the shelterin
and then develop different organ dysfunction, includ- complex that protects the telomeric ends of chromo-
ing bone marrow failure, pulmonary fibrosis, and eye, somes, mostly occur de novo and are found in patients
tooth, gastrointestinal, endocrine, skeletal, urologic, and with autosomal dominant DC.39,40 Mutations in the
immunologic abnormalities. Significant developmental gene ACD, encoding another member of the shelterin
delay is found for the more severe clinical variants of DC. complex, TPP1, have been found in two kindreds with
There is a wide variation in the severity and spectrum of DC.41,42 Compound heterozygous mutations in CTC1
clinical findings, which is only partly explained by locus were first described in patients with Coats plus and in
heterogeneity. The mode of inheritance varies by gene the phenotypically similar disorder cranioretinal micro-
mutation. Most patients in a large international registry angiopathy with calcifications and cysts and then were
are male with X-linked recessive inheritance.23 Kindreds later described in patients with autosomal recessive
with autosomal recessive and autosomal dominant pat- DC.43,44 CTC1 is part of the trimeric telomere capping
terns of inheritance are less common. For affected male complex that cooperates with the shelterin complex to
patients, the classic skin and nail findings are present in protect telomeres. Homozygous mutations in NOLA2
∼90%. The abnormal skin findings have been described and NOLA3 account for less than 1% of DC mutations;
as poikiloderma vascularis atrophicans or a latticework these genes encode NHP2 and NOP10, respectively,
hypo- and hyperpigmentation defect found especially which maintain the stability and proper trafficking of
on the neck, upper chest, and proximal limbs.23,24 Bone a complex of H/ACA small nucleolar RNAs, including
marrow failure is very common (>85%) and is the lead- the telomerase RNA, hTR.45–47 Mutations in WRAP53
ing cause of death. have been described linked to DC; this gene encodes
After bone marrow failure, pulmonary fibrosis is TCAB1 that binds telomerase and directs its localiza-
the most serious and life-threatening complication of tion to nuclear Cajal bodies, an important step in telo-
DC. In most cases it presents either after hematopoietic mere maintenance.48 Whole exome sequencing has led
cell transplantation25,26 or as a later manifestation of to the identification of heterozygous or compound het-
disease in those over the age of 30 years.27–29 Patients erozygous mutations in RTEL1.49–51 This gene encodes
with pulmonary disease have rales, digital clubbing, a a DNA helicase that is crucial for telomere maintenance
restrictive pulmonary defect, a reduced diffusion capac- and DNA repair. Homozygous or compound hetero-
ity, and diffuse interstitial markings on high-resolution zygous mutations in PARN have most recently been
computed tomography (HRCT) imaging of the chest. linked to autosomal recessive DC.52–54 The PARN pro-
Lung histopathology generally features a mixture of tein belongs to a family of conserved exoribonucleases
cellular inflammatory infiltrates and interstitial fibro- that shorten the poly(A) tail of messenger RNA through
sis.30,31 Clinical survival of DC patients after the devel- deadenylation. One of the RNA species that is modi-
opment of ILD is poor as pulmonary disease is generally fied by PARN is hTR or the RNA component of telo­
rapidly progressive. Death is usually 12–40 months merase.54 Thus, mutations in PARN lead to defective
after the onset of clinical symptoms.28 hTR and telomerase biogenesis and telomere disease.
Mutations in 11 different genes have been described Genetic anticipation has been seen in DC kindreds
in DC patients. Regardless of the pattern of genetic with TERC and TERT mutations. Inheritance of pro-
inheritance or the individual genetic mutation, all gressively shorter telomere lengths in each subsequent
DC patients exhibit short telomere lengths for their generation of mutation carriers provides a molecular
age.32 Telomeres are specialized structures essential for explanation for the observation of an earlier age of
maintenance of integrity of chromosomal ends com- onset in more severely affected individuals with each
posed of nucleotide repeats (TTAGGG)n and telomere- successive generation.36,55 Siblings who do not inherit
specific accessory proteins. Positional cloning first led the mutated gene can inherit short telomere lengths
to the identification of mutations in the DKC1 gene from the affected parent.56 Thus, DC patients have to
in those with the X-linked recessive form of the dis- inherit both short telomere lengths and a deleterious
ease.33 Dyskerin is a nucleolar protein that copurifies telomerase mutation to demonstrate anticipation.
6 Interstitial Lung Disease

RIDDLE Syndrome a peripheral distribution of HRCT reticulations with a


Patients with this very rare autosomal recessive disorder trend toward increasing involvement of the central por-
have a syndrome of increased sensitivity to radiation, tions of the lungs with progressive severity as a result of
immunodeficiency, learning difficulties, dysmorphic peribronchovascular thickening and bronchiectasis.73
features, short stature, and pulmonary fibrosis in later Surgical lung biopsies demonstrate lung remodeling,
years.57,58 The syndrome is caused by homozygous or numerous chronic inflammatory cells, and distinctive
compound heterozygous mutations in the RNF168 clusters of clear vacuolated type II pneumocytes with
gene, an E3 ubiquitin ligase critical for double-stranded florid foamy swelling and degeneration (“giant lamel-
DNA break (DSB) repair. It promotes ubiquitination of lar body degeneration”).78,79 Despite an early positive
H2A and H2AX, which, in turn, mediates accumula- study,76 a follow-up clinical trial showed no benefit of
tion of 53BP1 and BRACA1 at DSBs to promote DNA pirfenidone for the treatment of Hermansky-Pudlak
repair.59,60 It also has a role in TRF2-mediated protec- pulmonary fibrosis.80
tion of telomeres.61 Pathogenesis of pulmonary fibrosis is incompletely
understood. Studies of a mouse model of disease that is
Hermansky-Pudlak Syndrome homozygous for both HPS1 and HPS2 mutations dem-
Hermansky-Pudlak syndrome (HPS) is an autosomal onstrate additive effects of the genetic defects toward
recessive disease first described in 1959.62 It is character- the development of spontaneous pulmonary fibrosis.81
ized by oculocutaneous albinism, a bleeding diathesis Subpleural reticulations begin at 3 months and exten-
resulting from platelet storage pool deficiency, and in sive fibrosis is seen by 9 months.82 The histology of
some cases, pulmonary fibrosis.63 It is now known that the mouse lung replicates the human phenotype with
HPS is caused by defects of multiple cytoplasmic organ- “giant lamellar body degeneration” of the type II cells
elles, including melanosomes, platelet-dense granules, and demonstrates decreased phospholipid and surfac-
and lysosomes leading to diverse clinical features. tant protein (SP)-B and C secretion.81,83 The underlying
Other clinical features of the disease include granulo- molecular mechanism of murine HPS-associated pul-
matous colitis, neutropenia, immunodeficiency, cuta- monary fibrosis seems related to endoplasmic reticu-
neous malignancies, cardiomyopathy, and renal failure. lum (ER) stress and chronic alveolar epithelial type II
Currently the diagnosis is confirmed by the absence of cell injury.82
dense bodies on whole-mount electron microscopy of
platelets.64 Mutations in nine different genes cause this NKX2-1–Related Disorders
disease. Each of these genes functions in trafficking of These disorders include benign hereditary chorea, neo-
vesicular cargo proteins to cytoplasmic organelles or in natal respiratory distress, and congenital hypothyroid-
organelle biogenesis and maturation.65–70 HPS is the ism; it is also known as brain-lung-thyroid syndrome.
most common single gene disorder in Puerto Rico with Childhood-onset chorea is the hallmark clinical feature
an estimated frequency of about 1 in 1800 and a carrier of this disorder. Pulmonary disease is the second most
frequency of 1 in 21.71 Mutations in HPS1 and HPS3 common presentation, with nearly half of the patients
are found in 75% and 25% of Puerto Rican patients, having some type of pulmonary manifestation.84 These
respectively. include neonatal respiratory distress with or without
ILD generally causes symptoms in the 30s and, pulmonary hypertension, neuroendocrine cell hyper-
when it occurs, is generally fatal within a decade. plasia, ILD in children between the ages of 4 months
Patients have progressive restrictive disease with a to 7 years, and pulmonary fibrosis in older individu-
highly variable course.63,72 Pulmonary fibrosis has been als.85–87 Defective surfactant homeostasis and recurrent
described most frequently in patients with HPS1 muta- respiratory infections have been found to be a promi-
tions, and more rarely in patients with HPS2 or HPS4 nent feature in some subjects.86,88 Mutations include
mutations.72–77 In this regard, molecular subtyping is missense, nonsense, and deletions of the NKX2-1 gene.
important to assess the risk of developing pulmonary
fibrosis. In one of the largest studies, the mean age of Neurofibromatosis
onset was 35 years with a range of 15–53 years.72 The Neurofibromatosis (NF1) is an autosomal dominant
variability of pulmonary findings was generally not disorder that affects all ethnic groups. Type I, von
attributable to prior environmental exposures. Over Recklinghausen disease or classic NF1, is character-
80% of patients had abnormalities on CT scans, which ized by multiple (>6) café au lait spots, axillary and
were generally predictive of the degree of physiologic inguinal freckling, multiple cutaneous neurofibromas,
impairment and mortality. Most patients demonstrate and iris Lisch nodules. Pulmonary manifestations
CHAPTER 1 Genetic Interstitial Lung Disease 7

are less common. The incidence of ILD in NF1 has Stimulator of Interferon Genes–Associated
been estimated at 6–12%.89–91 It is characterized by Vasculopathy, Infantile-Onset
lower lobe–predominant diffuse interstitial fibrosis Patients with this autosomal dominant disorder have
and honeycombing. Thin-walled bullae are present in an autoimmune vasculopathy that causes severe skin
almost all patients with ILD or may be seen in isola- lesions affecting the face, ears, nose, and digits, result-
tion; they are large, asymmetric, and typically involve ing in ulceration and necrosis beginning in infancy.
the upper lobes.91 Histologic evidence of an alveoli- Over 85% have an ILD apparent on HRCT chest imag-
tis and interstitial fibrosis has been found in patients ing. Lung biopsy samples demonstrate a scattered
with normal chest X-rays or those with only apical mix of lymphocytic inflammatory infiltrate, follicular
bullae.90 Although there is near complete penetrance hyperplasia and B-cell germinal centers, interstitial
of the disease after childhood, the ILD is not observed fibrosis, and emphysematous changes.97,98 Patients can
until adulthood, typically in patients older than have livedo reticularis, Raynaud phenomenon, myosi-
40 years of age.90 The disease is often progressive and tis, joint involvement, immune complex deposition,
may lead to pulmonary hypertension and right-sided hypergammaglobulinemia, leukopenia, and autoan-
heart failure.92 The pathogenesis of NF-associated ILD tibodies (antinuclear, antiphospholipid, anticardio-
is unknown. lipin). All cases are caused by heterozygous, missense,
gain-of-function mutations in the TMEM173 gene,
Poikiloderma, With Tendon Contractures, which encodes the STING (stimulator of interferon
Myopathy, and Pulmonary Fibrosis genes).97,98 Mutations lead to activation of STAT1.97
This rare disorder usually affects individuals from
early childhood; presenting features include thin hair, Autoimmune Disease With Facial
telangiectasias and pigmentary abnormalities on sun- Dysmorphism
exposed areas, and tendon contractures frequently This autosomal recessive disease has been described
involving the ankles and feet.93 There is incomplete only in children with features of failure to thrive, devel-
penetrance of pulmonary fibrosis; >50% of cases have opmental delay, dysmorphic features, autoantibodies,
lung involvement. When it is present, the pulmonary and inflammatory cell infiltration of the lungs, liver,
fibrosis generally develops during the second decade of and gut. Chronic lung disease was found in 9 of 10 chil-
life, is progressive, and can lead to death. The histopa- dren. Three had a chronic oxygen requirement and died
thology for at least one case was consistent with usual of respiratory failure. Surgical lung biopsy from one
interstitial pneumonia.93 Different missense mutations case showed a cellular, nonspecific interstitial pneumo-
in the FAM111B gene have been found in unrelated nitis (NSIP). All affected children in one large Amish
cases of different ancestry.94 The function of this gene kindred were homozygous for a frameshift mutation in
is unclear. the ITCH gene that is predicted to cause a truncation
of an E3 ubiquitin ligase.99 Of note, mice with a small
Interstitial Lung Disease, Nephrotic genomic inversion that disrupts the Itch and agouti
Syndrome, and Epidermolysis Bullosa genes develop a similar spectrum of immunologic dis-
Infants with this congenital autosomal recessive disor- ease of the spleen, lymph nodes, stomach, and skin,
der present with disease in multiple organs. The pul- as well as chronic pulmonary interstitial inflammation
monary and renal features cause significant morbidity and alveolar proteinosis.100,101
and mortality, although dermatologic features are
common (epidermolysis bullosa, thin hair, dystrophic Autoimmune Interstitial Lung, Joint, and
nails, onycholysis). The lung involvement is character- Kidney Disease
ized radiographically by diffuse ground-glass opacity This rare syndrome was recently elucidating by whole
and interlobular septal thickening. Histology demon- exome sequencing of patients from five unrelated
strates abnormal alveolarization with poorly septated families with autoimmune interstitial lung, joint, and
(“simplified”) alveolar spaces.95,96 All mutations are kidney disease.102 All patients had high-titer autoanti-
homozygous deletion/frameshift or missense muta- bodies, ILD, and inflammatory arthritis. The average age
tions in ITGA3, which encode an integrin alpha chain at presentation was 3.5 years, with a range of 6 months
belonging to a family of cell surface adhesion proteins. to 22 years. Some patients presented with pulmonary
One of the missense mutations prevents proper post- hemorrhage that required immunosuppression. Lung
translational modification of the integrin alpha chain biopsies showed lymphocytic interstitial infiltration
with defective cell surface expression. with lung-infiltrating CD4+ T cells and CD20+ B cells
8 Interstitial Lung Disease

within germinal centers. All mutations affect the same This genetically heterogeneous group of diseases
functional domain of the COPA protein, impair intra- includes those with variable antibody and B-cell,
cellular ER-Golgi transport, and lead to ER stress and T-cell, and NK-cell deficiencies, as well as defective
upregulation of cytokines. responses to vaccination. Mutations in multiple genes,
including ICOS,113 TNFRSF13B (encoding the trans-
GATA2 Deficiency membrane activator and CAML interactor TACI),114,115
Haploinsufficiency of the hematopoietic transcrip- TNFRSF13C (encoding the B-cell activating factor),116
tion factor GATA2 leads to a wide spectrum of diseases CD19,117 CD20,118 CD81,119 CD21,120 LRBA,121,122
including primary immunodeficiency syndromes such IL21,123 NFKB1,124 NFKB2,125 and IKZF1 (encoding the
as dendritic cell, monocyte, B lymphocyte, and NK hematopoietic zinc finger transcription factor),126,127
lymphocyte deficiency (DCML) and monocytopenia lead to sporadic, autosomal recessive, and autosomal
and mycobacterial infection syndrome (MONOMAC); dominant patterns of disease.
primary lymphedema with myelodysplasia (Emberger X-linked agammaglobulinemia (XLA) is an immuno-
syndrome); and susceptibility to myelodysplastic syn- deficiency disorder that occurs almost exclusively in
drome and acute myeloid leukemia. Besides immu- males from a failure of B-cell maturation. Although
nodeficiency (with predisposition especially to severe there is a high prevalence of pneumonia in XLA, there
viral, disseminated nontuberculous mycobacterial, and is a lower prevalence of chronic lung disease with 47%
invasive fungal infections), pulmonary disease, vascu- and 25% demonstrating abnormal pulmonary function
lar/lymphatic dysfunction, and hearing loss are com- studies and bronchiectasis, respectively.109 Although
mon.103–107 Most individuals have some manifestation the degree of immune dysregulation may be lower,109
of pulmonary disease; 79% have a diffusion defect, 63% the rate of progression of lung function decline is
with abnormal pulmonary function tests, 18% with greater for those with XLA.128 Between 85% and 90% of
biopsy-proved pulmonary alveolar proteinosis (PAP), cases are caused by mutations in BTK, the gene encod-
and 9% with pulmonary arterial hypertension.107 All ing Bruton tyrosine kinase and a key regulator in B-cell
lack anti–GM-CSF antibodies and are resistant to sub- development.129 The remaining cases include a geneti-
cutaneous or inhaled granulocyte-macrophage colony– cally heterogeneous group of patients with mutations
stimulating factor (GM-CSF) therapy. Chest CT findings in IGHM (encoding the mu heavy-chain),130 IGLL1
include reticulonodular opacities, “crazy paving,” and (encoding immunoglobulin lambda-like-1),130,131
paraseptal emphysema. Median age at first presentation CD79A,132 BLNK,133 CD79B,134 and TCF3.135
is 20 years, but is highly variable (5 months to 78 years). Autosomal dominant hyper-IgE syndrome is an immune
Most mutations are predicted to cause a loss of function deficiency disorder characterized by the triad of recur-
of the mutated allele, leading to haploinsufficiency and rent staphylococcal skin abscesses, pneumonias, and
autosomal dominant inheritance.106,108 Hematopoietic elevations of IgE (usually >2000 IU/mL). Survival is
stem cell transplantation is indicated for severe disease. typically into adulthood, but death is usually secondary
to infections. Diagnosis requires a high level of suspi-
Immunodeficiency Syndromes cion due to variability of phenotypic features. A clini-
Common variable immunodeficiency (CVID) syndromes cal scoring system has been developed that combines
are all associated with an increased susceptibility to immunologic (elevated IgE, eosinophilia >700/μL,
recurrent infections, especially sinopulmonary infec- decreased T-helper [Th]17 cells, recurrent infections)
tions. There is a high variability of clinical features. and nonimmune features (retained primary teeth, sco-
Respiratory complications are frequently responsible liosis, joint hyperextensibility, bone fractures following
for patient morbidity and mortality and include acute minimal trauma, a typical facial appearance, and vascu-
infections, sequelae of infections (bronchiectasis), lar abnormalities). Pneumatoceles and bronchiectasis,
noninfectious immune-mediated infiltrates such as which result from aberrant healing of the pneumonias,
granulomatous lymphocytic interstitial lung disease are seen in ∼70% of patients and lead to significant
(GLILD) and lymphoma, and progressive respiratory mortality.136,137 Patients have heterozygous missense,
failure.109,110 When severe, bronchiectasis is gener- splice site, or small deletions in the STAT3 gene.138 Most
ally associated with obstructive lung disease; GLILD mutations cluster in the DNA-binding domain of this
is typically associated with restrictive physiology and transcription factor, which regulates responses to many
decreased diffusion capacity.111 Lung disease is fre- different cytokines. Patients’ purified native T cells are
quently found in patients with CVID, as >90% have unable to differentiate in vitro into IL-17–producing
radiographic abnormalities apparent on chest CT.112 Th17 cells, which play a critical role in the clearance of
CHAPTER 1 Genetic Interstitial Lung Disease 9

fungal and extracellular bacterial infections of the lung and bone manifestations including osteopenia, lytic
and skin.139 Most patients represent sporadic cases, but lesions, bone crisis, and skeletal deformities. Infiltra-
autosomal dominant transmission has been seen in tion of Gaucher cells into the alveoli, interstitium,
some kindreds. and pulmonary capillaries can lead to lung involve-
Activated PI3K-delta syndrome is a monogenic auto- ment. Over 65% of patients with type I disease have
somal dominant disease that leads to overactivation pulmonary function abnormalities, but only a fraction
of the PI3K signaling pathway and lymphoprolifera- (<5%) have diffuse lower lobe linear infiltrates, restric-
tion. This autosomal dominant disease is character- tive physiologic impairment, and a reduced diffusion
ized by recurrent sinopulmonary infections, reduced capacity consistent with ILD.148,149 Enzyme replace-
IgG2, increased serum IgM, and impaired vaccine ment therapy can reduce organ volumes and improve
responses. In one series, 75% of patients had CT evi- the hematologic parameters and bone pain but is usu-
dence of bronchiectasis or mosaic attenuation.140 The ally poorly effective in treating the lung manifestations
lymphoid aggregates within the lung and can lead to of this disease.150 Other therapeutic treatments, such
the compression of nearby bronchi.110 Heterozygous as substrate reduction therapy as well as others, are in
missense mutations in PIK3CD result in increased development.151
phosphorylation of AKT, consistent with a gain of Niemann–Pick disease types A and B are caused by
function.140,141 an inherited deficiency of acid sphingomyelinase activ-
CTLA4 deficiency syndrome is an autosomal dom- ity. Type C is caused by defective movement of lipids,
inant immunodeficiency characterized by T-cell including cholesterol, from endosomes and lysosomes.
immune dysregulation. Pulmonary manifestations Patients demonstrate a range of ages and can have
mirror CVID with recurrent respiratory infections and diverse symptoms affecting the lung, liver, spleen, bone
GLILD. Lymphocytic infiltration is found for other marrow, skeleton, brain, muscle, mental ability, and
organs, including the bone marrow, kidney, brain, movement. The pulmonary manifestations include ILD
liver, spleen, and lymph nodes. Heterozygous loss- and recurrent lung infections. In some patients the ILD is
of-function mutations in CTLA4 have been recently predominantly in the bases with thickened interlobular
described.142,143 septa, interlobular lines, and ground-glass opacities.153
X-linked reticulate pigmentary disorder is a rare Lysinuric protein intolerance is an autosomal reces-
immunodeficiency disorder with features of recurrent sive disease caused by an inherited defect of cationic
infections and systemic sterile inflammation. Affected amino acid transport. There is excess urinary clear-
patients typically develop recurrent pneumonias in ance of these amino acids and deficient intestinal
the first few months of life. By childhood, affected absorption, which leads to depleted body pools.
males develop diffuse skin hyperpigmentation with Most patients present in infancy with failure to thrive,
a distinctive reticulate pattern, bronchiectasis, hypo- growth retardation, protein aversion, muscular hypo-
hidrosis, corneal scarring, enterocolitis, and recur- tonia, hepatosplenomegaly, and osteoporosis. In one
rent urethral strictures.144–146 A recurrent intronic study, all patients who developed a fatal respiratory
mutation has been found that disrupts the expression insufficiency, usually PAP, were children less than
of the catalytic subunit of DNA polymerase-alpha 15 years of age.154 Most adult patients have evidence of
(POLA1) and leads to increased production of type I an ILD on CT scans with interlobular and intralobular
interferons.147 septal thickening and subpleural cysts, but only a few
are symptomatic.154
Inborn Errors of Metabolism
Gaucher disease is an autosomal recessive lysosomal
storage disease characterized by the accumulation of GENETIC DISEASES IN WHICH
the glycolipid glucosylceramide due to the deficiency INTERSTITIAL LUNG DISEASE IS THE
of the enzyme acid-beta glucosidase. Diagnosis relies DOMINANT PHENOTYPE
on the demonstration of deficient enzyme activity in Table 1.2 lists the genetic disorders in which an ILD is
cells or the identification of two disease-causing muta- the dominant clinical feature. Included in this group
tions in the GBA gene. Patients can display a large are the surfactant disorders. Pulmonary surfactant is
variety of symptoms, ranging from patients who are a mixture of phospholipids and associated proteins
completely asymptomatic to those who present with that covers the alveolar surface at the gas-alveolus
perinatal lethality. The usual clinical findings include interface, where it functions to reduce surface tension
hepatosplenomegaly, anemia, thrombocytopenia, and prevent atelectasis. A group of genetic disorders
10 Interstitial Lung Disease

TABLE 1.2
Genetic Interstitial Lung Disease (ILD): Disorders in Which ILD Is the Dominant Phenotype
Disease Inheritance Gene Pathogenesis Presentation
Surfactant metabolism AR SFTPB Absent SP-B Neonatal respiratory failure
dysfunction 1
Surfactant metabolism AD SFTPC Lack of SP-C and Neonatal respiratory
dysfunction 2 ER stress distress, NSIP, UIP
Surfactant metabolism AR ABCA3 Phospholipid Neonatal respiratory
dysfunction 3 transport defect distress, PAP
Surfactant metabolism AR CSF2RA GM-CSF signaling PAP
dysfunction 4 CSF2RB defect
Interstitial lung and liver AR MARS Methionine tRNA PAP
disease ligase defect
Pulmonary fibrosis, AD TERT Short telomere Pulmonary fibrosis
telomere related, type 1 length
Pulmonary fibrosis, AD TERC Short telomere Pulmonary fibrosis
telomere related, type 2 length
Pulmonary fibrosis, AD RTEL1 Short telomere Pulmonary fibrosis
telomere related, type 3 length
Pulmonary fibrosis, AD PARN Short telomere Pulmonary fibrosis
telomere related, type 4 length
Pulmonary fibrosis, Various DKC1, TINF2, NAF1 Short telomere Pulmonary fibrosis
telomere related length
Pulmonary fibrosis and AD SFTPA1, SFTPA2 ER stress Pulmonary fibrosis,
adenocarcinoma adenocarcinoma, respiratory
distress in infancy
Pulmonary fibrosis Sporadic MUC5B ER stress? UIP, ILAs
susceptibility
Pulmonary fibrosis Sporadic TERT, TERC, OBFC1 Short telomere UIP
susceptibility length
Pulmonary alveolar AR SLC34A2 Phosphate trans- Microliths
microlithiasis port defect

ER, endoplasmic reticulum; GM-CSF, granulocyte-macrophage colony-stimulating factor; ILA, interstitial lung abnormalities; NSIP, nonspe-
cific interstitial pneumonia; PAP, pulmonary alveolar proteinosis; SP, surfactant protein; UIP, usual interstitial pneumonia.

involving the production, processing, and clearance surfactant dysfunction disorders, patients with muta-
of surfactant has been recognized as an important tions in SFTPC display the broadest range in age of
cause of neonatal and pediatric respiratory illness. onset, with affected neonates to older adults.
This group, collectively referred to as surfactant dys-
function disorders,155 encompasses a variety of muta- Surfactant Metabolism Dysfunction, Type 1:
tions involving the genes that encode SP-B (SFTPB), SFTPB Mutations
SP-C (SFTPC), the ATP-binding cassette transporter SP-B deficiency is inherited in an autosomal reces-
A3 (ABCA3), and the receptor for granulocyte-mac- sive manner, and the majority of affected patients
rophage colony–stimulating factor (CSF2RA and develop respiratory failure in the neonatal period
CSF2RB). ABCA3 deficiency is the most prevalent for with rapid progression of disease and death at
the surfactant disorders that present in the neonatal 3–6 months.156–161 A few mutations that seem to con-
period with severe respiratory distress. Mutations in fer a milder phenotype have been found. Children
SFTPB, SFTPC, and NKX2-1 are more rare. Of all the with these mutations have partial expression of the
CHAPTER 1 Genetic Interstitial Lung Disease 11

SP-B protein, survive longer, and go on to develop a in ABCA3 are the most common inherited defects in
chronic ILD.162,163 surfactant metabolism presenting either with severe
neonatal disease or as diffuse lung disease in infancy
Surfactant Metabolism Dysfunction, Type 2: or childhood.155,174,179,180 ABCA3 is an ATP-binding
SFTPC Mutations cassette transporter that localizes to the limiting mem-
Autosomal dominant lung disease due to mutations in brane of lamellar bodies where it functions in translo-
the gene encoding SP-C, SFTPC, was first described in cating phospholipids, primarily phosphatidylcholine,
2001 by Nogee and colleagues.164 The index case was a into these organelles for assembly and storage of surfac-
full-term infant who developed respiratory symptoms tant in type II AECs.181,182 The functional consequences
at 6 weeks of age. An open lung biopsy revealed his- of the ABCA3 mutations include decreased expression
tologic features of NSIP. The child’s mother had been of the protein, abnormal localization of the protein to
diagnosed with desquamative interstitial pneumonitis the lamellar membrane, production of surfactant that
at 1 year of age, and the child’s maternal grandfather is deficient in phosphatidylcholine, and increased alve-
had died from lifelong respiratory illness of unknown olar surface tension.183–185
cause. Both the mother and her infant had a hetero-
zygous mutation at the splice donor site in intron 4 Surfactant Metabolism Dysfunction, Types
(c.460+1G > A), which resulted in skipping of exon 4 4 and 5: CSF2RA and CSF3RB Mutations
and deletion of 37 amino acids.164 In the next year, a Protein alveolar proteinosis (PAP) is a rare form of
large five-generation kindred was described with 14 lung disease characterized by intraalveolar accumula-
affected family members.165 The age at diagnosis for the tion of surfactant, which results in respiratory insuffi-
affected individuals ranged from 4 months to 57 years ciency. Histopathology specimens from affected patients
and included four adults with surgical lung biopsy evi- demonstrate distal airspaces filled with foamy alveolar
dence of usual interstitial pneumonitis and three chil- macrophages and a granular, eosinophilic material that
dren with NSIP. Genomic sequencing revealed a rare stains positively with PAS reagent. In general, the under-
heterozygous missense SP-C mutation (L188Q) in all lying lung architecture is normal unless infection is pres-
analyzed individuals. Human type II alveolar epithelial ent.186 When the accumulated surfactant is removed, as
cells from one of the patients and mouse lung epithe- is done with whole-lung lavage, the gas exchange proper-
lial cells producing the mutant SP-C protein showed ties of the lung improve.186 Approximately 90% of PAP
abnormal lamellar bodies. Since these two studies, over cases are acquired and are referred to as primary PAP.187–
30 different mutations in SFTPC have been identified in 189 Circulating autoantibodies to GM-CSF can be found

children and adults. A missense mutation (c.218T > C) in over 92% of patients with primary PAP.190–193 These
that changes a conserved isoleucine at position 73 to antibodies block GM-CSF signaling in vivo, reduce
a threonine is one of the more common and accounts alveolar macrophage surfactant catabolism, and impair
for 25–35% of abnormal alleles.155,166–171 There is surfactant clearance.186,193–196 Secondary PAP occurs in
incomplete penetrance of the ILD phenotype and phe- several different clinical settings, such as in association
notypic heterogeneity. The disease ranges from severe with hematologic malignancies, immunosuppression,
respiratory distress in infants to IPF in older adults. inhalation of inorganic dusts, and certain infections.
Mutations of SFTPC are rare in individuals without a The GM-CSF receptor is composed of an α- and a
family history of pulmonary fibrosis.171–173 At present, β-chain, encoded by the CSF2RA and CSF2RB genes,
the disease phenotype is thought to be caused by aber- respectively.197 Mutations in both CSF2RA and CSF2RB
rant protein-folding, which elicits the unfolded protein have been identified in children with PAP.198,199 In
response, ER stress, and apoptosis of alveolar epithelial 2011, a case of adult-onset hereditary PAP was reported
cells.155,174–178 in Japan.200 Here, a 36-year-old woman with PAP had
elevated circulating levels of GM-CSF, no measurable
Surfactant Metabolism Dysfunction, Type 3: anti-GM-CSF antibodies, and reduced expression of the
ABCA3 Mutations GM-CSF receptor β-chain. Genetic sequencing revealed
There is a significant amount of phenotypic overlap a homozygous, single-base deletion at nucleotide 631 in
between patients with SFTPB mutations and those with exon 6 of CSF2RB. Both of the patient’s parents were het-
defects in ABCA3. Like SP-B deficiency, the disease is erozygous for the mutation. Hereditary PAP follows an
inherited in an autosomal recessive manner. Since autosomal recessive pattern. Mice with deletion of the
the first mutation was discovered in 2004, more than gene encoding GM-CSF or its receptor demonstrate a phe-
200 different mutations have been found. Mutations notype very similar to adults with primary PAP.186,201–204
12 Interstitial Lung Disease

Interstitial Lung and Liver Disease biopsies.210,218 While approximately half of patients
A specific and severe type of PAP has been described carry a diagnosis of idiopathic pulmonary fibrosis (IPF),
affecting infants and children on Réunion Island.205 others have diagnoses of another idiopathic interstitial
Since 1970, 34 children have been diagnosed and pneumonia, unclassifiable lung fibrosis, chronic hyper-
treated, giving rise to an incidence of disease of 1 in at sensitivity pneumonitis, pleuroparenchymal fibroelas-
least 10,000 newborns. The lung disease progresses to tosis, idiopathic fibrosis with autoimmune features, or,
lung fibrosis despite regular whole-lung lavage. It has rarely, a connective tissue disease–associated ILD.218
recently been found that patients have biallelic muta- Many TERT mutation carriers with pulmonary fibro-
tions in MARS, the gene encoding methionine tRNA sis report past cigarette smoking or an exposure to a
ligase.205 Compound heterozygous mutations in the fibrogenic environmental insult, suggesting that injuri-
same gene have been found in one patient with mul- ous environmental exposures in conjunction with the
tiorgan disease, predominated by liver failure.206 The underlying inherited genetic predisposition lead to the
increased prevalence of disease on Réunion Island and lung disease. Disease progression is inexorable, with a
in nearby Tunisia and France is due to founder muta- mean transplant-free survival of <3 years after diagno-
tions. A potential benefit of high-dose methionine sis. TERT mutation patients undergoing lung transplant
supplementation has not yet been studied in these generally have a higher risk of cytopenias and other
patients. extrapulmonary complications.219–221
Genetic anticipation is seen for TERT mutation
Pulmonary Fibrosis, Telomere Related, patients, with an earlier age of diagnosis of lung fibrosis
Type 1 in subsequent generations.218 Similar to DC, progres-
Heterozygous mutations in the gene encoding the pro- sively shorter telomere lengths are found in subse-
tein component (TERT) of telomerase have been found quent generations of TERT mutation carriers, and these
in ∼15% of patients with autosomal dominant famil- shorter telomere lengths explain genetic anticipation
ial pulmonary fibrosis.207,208 The frequency of TERT seen in kindreds.36,210,218
mutations in patients with sporadic adult-onset ILD is
less common.208,209 Telomerase is a multimeric ribo- Pulmonary Fibrosis, Telomere Related,
nucleoprotein enzyme that catalyzes the addition of a Type 2
repetitive DNA sequence to the ends of chromosomes Heterozygous mutations in the gene encoding the RNA
known as telomeres. Each of the mutations is rare and component (TERC) of telomerase have been found in
associated with decreased in vitro activity of telomer- ∼3% of patients with autosomal dominant familial pul-
ase and short leukocyte telomere lengths. TERT muta- monary fibrosis.207,208 Each of the mutations is rare and
tions are the most common genetic mutations found associated with decreased in vitro activity of telomerase
in adult patients with ILD.210 In addition, adult-onset and short leukocyte telomere lengths. A wide range of
pulmonary fibrosis is the most common manifestation progressive adult-onset pulmonary fibrosis subtypes
of TERT mutations.211 Other clinical manifestations of are seen in patients with TERC mutations.218 Patients
TERT mutations include DC (see earlier discussion), with TERC mutations are generally diagnosed with ILD
aplastic anemia and other forms of bone marrow fail- at an earlier age (51 years) than TERT mutation carri-
ure,212 liver disease including liver cirrhosis,213,214 early ers (58 years). In addition, they have a higher incidence
graying of hair,215 and an increased risk for myelodys- of hematologic comorbidities, especially leukopenia,
plastic syndrome and acute myeloid leukemia.216,217 thrombocytopenia, aplastic anemia, or myelodysplas-
Altogether, disparate clinical presentations and dis- tic syndrome.
eases linked together by defects in telomere-related
genes and characterized by short telomere lengths are Pulmonary Fibrosis, Telomere Related,
known as monogenic short telomere syndromes or Type 3
telomeropathies. Autosomal dominant pulmonary fibrosis with incom-
The penetrance of pulmonary fibrosis in TERT plete penetrance has been linked to rare mutations
mutation carriers is age and gender dependent.210 The in RTEL1.222–224 RTEL1 surpassed the threshold for
penetrance of pulmonary fibrosis for men aged 40–49, genomewide significance when comparing the num-
50–59, and >60 years is 14%, 38%, and 60%, respec- ber of observed versus expected novel damaging or
tively. Women show the same age-dependent increase conserved missense mutations in familial pulmonary
in penetrance. Microscopic honeycombing and fibro- fibrosis cases and controls.223 Across independent
blastic foci are commonly found in surgical lung studies, RTEL1 mutations are rare and associated with
CHAPTER 1 Genetic Interstitial Lung Disease 13

short leukocyte telomere lengths. This gene encodes Pulmonary Fibrosis, Other Telomere Related
the regulator of telomere elongation helicase 1 and has Genes found to be mutated in DC patients are candidate
a known role in telomere maintenance. Mutations in genes for patients with adult-onset pulmonary fibrosis and
RTEL1 were previously shown to cause Hoyeraal-Hre- short telomere lengths. Similar to the other short telomere
idarsson syndrome, a severe variant of DC (see earlier syndromes mentioned earlier, many individuals have clin-
discussion), in which affected children generally have ical features that overlap with DC, including bone marrow
very short telomere lengths and biallelic mutations. In abnormalities and skin changes. A missense mutation in
contrast, those affected with adult-onset pulmonary the DKC1, encoding dyskerin, has been reported in a kin-
fibrosis are heterozygous for rare missense or loss-of- dred with two affected older males with interstitial fibro-
function RTEL1 mutations. Similar to patients with sis, hyperpigmented skin changes, dyskeratotic nails, and
TERT mutations, ∼50% of heterozygous RTEL1 muta- macrocytic anemia.225 Blood leukocyte telomere lengths
tion carriers have hematologic manifestations such as were short. The expression of hTR in a lymphoblastoid
anemia or macrocytosis.218 A wide range of progressive cell line derived from the proband was lower than that
adult-onset pulmonary fibrosis subtypes are seen in of controls, even though expression of DKC1 was not
patients with RTEL1 mutations. Genetic anticipation is reduced. Female mutation carriers were unaffected.
also seen in RTEL1 kindreds. TINF2 encodes one of the shelterin complex pro-
teins that functions to protect the telomeric ends of
Pulmonary Fibrosis, Telomere Related, Type 4 chromosomes. Exome sequencing revealed a complex
Exome sequencing has linked autosomal dominant spice acceptor site and missense mutation on the same
pulmonary fibrosis with incomplete penetrance to TINF2 allele, with predominant expression of the mis-
mutations in PARN, the gene encoding a polyadenyl- sense mutation in lung-derived DNA from a female
ate-specific ribonuclease.223 PARN surpassed the thresh- patient with pulmonary fibrosis and infertility.226
old for genomewide significance when comparing the Heterozygous loss of function mutations in NAF1
number of observed versus expected novel damaging have most recently been linked to pulmonary fibrosis-
mutations in cases and controls.223 Most mutations are emphysema, short telomere lengths, bone marrow fail-
predicted to cause protein loss of function (splice site, ure, and liver disease.226a NAF1 has been shown to be
nonsense, and frameshift). One large kindred was iden- essential for the biogenesis of telomerase RNA.
tified by linking together two smaller and previously
unknowingly related kindreds with familial pulmonary Pulmonary Fibrosis and Adenocarcinoma,
fibrosis and an identical splice-site PARN mutation. The Related to Surfactant A Mutations
kinship of the two families was confirmed by demon- Novel missense mutations in the gene encoding SP-A2
strating that the two probands share ∼6% of their over- (SFTPA2) were discovered by genomewide linkage
all genome, including the genomic segment on which followed by sequencing candidate genes within the
the PARN mutation is located. As a further test for the linked region.227 Affected individuals have evidence of
relevance of the PARN mutations, the cosegregation of pulmonary fibrosis and/or lung adenocarcinoma with
the mutations with pulmonary fibrosis was compared features of bronchoalveolar cell carcinoma. A second
across extended kindreds. The overall backward LOD study reported three additional rare SFTPA2 mutations
score across all informative PARN kindreds was 3.6, in patients with adult-onset pulmonary fibrosis and
reflecting an odds ratio of 1:4096 in favor of linkage. a personal or family history of adenocarcinoma.228 A
Other clinical manifestations of PARN mutations third study has recently described the cosegregation
include DC (see earlier discussion), in which biallelic of ILD and lung adenocarcinoma in an extended kin-
mutations in the gene are frequently found.52–54 Patient dred with a germline mutation in SFTPA1 that is pre-
cells show deficiencies in trimming of small nucleolar dicted to change a conserved tryptophan at position
RNAs, including abnormally adenylated hTR.54 In general, 211 (Trp211Arg).229 While most of the affected indi-
leukocyte telomere lengths of PARN mutation patients are viduals in the SFTPA1 kindred were affected with an
shorter than in controls, but longer than in subjects with ILD after age 30 (range 31–69 years at diagnosis), one
TERT, TERC, or RTEL1 mutations.223 Patients with PARN was diagnosed at 7 months of age and died of respira-
mutations are generally diagnosed with ILD at an older tory failure 2 months later.229 Thus, a wide range of age
age (64 years) than TERT mutation carriers (58 years).218 at the time of diagnosis is observed, from respiratory
However, once pulmonary fibrosis is diagnosed, the respi- insufficiency in infancy to lung fibrosis and adenocar-
ratory disease progresses and patients have a mean trans- cinoma in adulthood. All known mutations in the sur-
plant-free survival of 5.7 years from diagnosis. factant A genes affect highly conserved residues in the
14 Interstitial Lung Disease

carbohydrate recognition domain of the proteins. Two lead to increased susceptibility to lung fibrosis through
of the mutations are predicted to alter the same glycine its effects on increased mucin expression and ER stress.
at amino acid position 231 (Gly231Val, Gly231Arg). The GWAS study confirmed the association of IPF
Cells expressing mutant proteins fail to secrete with variants in the TERT237 and TERC genes, with meta-
mature protein into the culture media.227,229 In addi- analysis p values of 1.7 × 10−19 and 4.5 × 10−8, respec-
tion, expression of these mutant proteins led to fewer tively.232 Overall, common variants in three telomere
intracellular oligomers, greater protein instability, and length–related genes (TERT, TERC, and OBFC1)238–240
increased markers of ER stress.230 Family members were found to be associated with lung fibrosis in this
with these heterozygous mutations secrete comparable study. The GWAS study also found seven new loci that
amounts of total SP-A into the alveolar space, as com- reached genomewide statistical significance.232 These
pared with control family members, suggesting that the loci are located near genes involved with host defense,
pathogenic mechanism may be related to ER stress of cell-cell adhesion, and DNA repair.
the resident epithelial alveolar cells.230
Pulmonary Alveolar Microlithiasis
Susceptibility to Pulmonary Fibrosis, Pulmonary alveolar microlithiasis (PAM) is a rare dis-
Common Variants order characterized by laminated calcium phosphate
Using a genomewide screen, a common variant located concretions within the alveoli. PAM is inherited in an
3 kb upstream of the MUC5B transcription start site autosomal recessive manner and is particularly prevalent
(rs35705950) was found to be present at a higher fre- in Turkey, Italy, the United States, and Japan.241 The chest
quency in IPF patients. This variant is found in ∼9% of radiographs of patients with PAM show diffuse, bilateral,
controls (an overall allele frequency of 0.10) and 38% of micronodular opacities, which obscure the heart border,
patients with IPF. The odds ratio for IPF disease among mediastinum, and diaphragmatic surfaces. Despite the
subjects who are heterozygous or homozygous for the “sandstormlike” appearance, the clinical presentation
minor allele of this single nucleotide polymorphism of PAM is variable.242–245 Early in the disease course,
(SNP) was 9.0 (CI 6.2–13.1) and 21.8 (5.1–93.5), patients are often asymptomatic, but over time develop
respectively.231 The variant allele was associated with cough, dyspnea, and a restrictive defect with reduced
upregulation of MUC5B expression (up to 37.4 times diffusion. In 2006, Corut et al. reported the discovery
as high when compared with those homozygous for the of several mutations in the gene encoding the type IIb
wild-type allele) in lung tissue of unaffected subjects.231 sodium-phosphate cotransporter protein (SCL34A2)
A genomewide association study (GWAS) was per- in individuals with PAM.246 The group used linkage
formed comparing the frequency of common SNPs in analysis of a large consanguineous family in which six
patients with fibrotic idiopathic interstitial pneumonia members were affected by PAM, and ultimately identi-
(n = 1616) with controls (n = 4683). A replication anal- fied six homozygous mutations in SCL34A2 that predict
ysis included 876 cases and 1890 controls. A MUC5B loss of protein function. A separate group in Japan used
promoter variant (rs868903) was again found to be sig- genomewide SNP mapping to identify candidate genes
nificantly associated with fibrotic lung disease, with a in six patients with PAM, and they independently identi-
metaanalysis P value of 9.2 × 10−26.232 A broad region fied SCL34A2 as a gene of interest.247 Two patients had
on 11p15, including the MUC5B, MUC2, and TOLLIP homozygous frameshift mutations and four had splice-
genes, was found to demonstrate genomewide signifi- site mutations of the gene. None of the mutations was
cance. After adjusting for the MUC5B promoter SNP identified in normal controls. Since these initial discov-
found in the earlier study (rs35705950),231 most of the eries, additional missense and frameshift mutations, as
variants in this broad region were no longer significantly well as intragenetic deletions, have been identified.248–250
associated, suggesting that the associations seen for the The gene is expressed in high levels in the lung, pre-
other SNPs may be due to linkage disequilibrium. The dominantly in type II epithelial cells.251–253 Epithelial
MUC5B promoter (rs35705950) has been found in the deletion of the mouse Npt2b gene in the lung leads to
Framingham Heart Study233 to be associated with inter- a progressive pulmonary process characterized by diffuse
stitial lung abnormalities, thus linking it to an early alveolar microliths, restrictive physiology, and alveolar
manifestation of IPF. Mucins undergo a complex matu- phospholipidosis.254 Microliths are readily dissolved by
ration process in airway cells, with glycosylation and whole-lung EDTA lavage. A low-phosphate diet prevents
disulfide multimerization, before secretion. As genetic microlith formation in young mice and decreases the
perturbations leading to increased mucin production burden of pulmonary calcium deposits in older mice.
cause elevated ER stress signaling,234–236 it is interest- The effectiveness of such a diet has not yet been studied
ing to speculate that the MUC5B promoter variant may in human patients.
CHAPTER 1 Genetic Interstitial Lung Disease 15

TABLE 1.3
Genetic Interstitial Lung Disease: Pathogenesis
Pathway Surfactant Metabolism ER Stress Telomere Shortening
Rare variants NKX2.1 SFTPC TERT, TERC
ABCA3 SFTPA1, SFTPA2 RTEL1
SFTPB HPS1, HPS2, HPS4 PARN
SFTPC COPA DKC1, TINF2
SFTPA1, SFTPA2 Other DC genes
CSF2RA, SFT2RB
Common variants MUC5B? TERT, TERC
OBFC1

DC, dyskeratosis congenita; ER, endoplasmic reticulum.

CONCLUSION identification of genes relevant to the molecular patho-


Discoveries of the genetic underpinnings of ILD have genesis of more common manifestations of ILD. How
demonstrated a few consistent themes. Many of the this knowledge leads to effective treatments across the
genes involve common pathways that lead to altered spectrum of genetic ILD will be the next grand opportu-
surfactant metabolism, increased ER stress signaling, nity to improve patient care.
and telomere shortening (Table 1.3). Mutations, or
pathologic ultrarare variants, have been found in genes
involved with the metabolism of pulmonary surfac- REFERENCES
tant. Given the central role surfactant has for reduc- 1. Avila NA, Dwyer AJ, Rabel A, Moss J. Sporadic lym-
ing surface tension of the lung, opsonizing respiratory phangioleiomyomatosis and tuberous sclerosis complex
pathogens, and regulating inflammatory responses with lymphangioleiomyomatosis: comparison of CT fea-
in the alveolar space, it is not surprising that patients tures. Radiology. 2007;242(1):277–285.
2. Lenoir S, Grenier P, Brauner MW, et al. Pulmonary lym-
with mutations in these genes have a wide range of ILD
phangiomyomatosis and tuberous sclerosis: comparison
presentations. of radiographic and thin-section CT findings. Radiology.
Another common pathway, increased ER stress sig- 1990;175(2):329–334.
naling, has been elucidated by genetic variants linked 3. Curatolo P, Bombardieri R, Jozwiak S. Tuberous sclero-
to various ILD subtypes.255,256 Production of misfolded sis. Lancet. 2008;372(9639):657–668.
(missense) surfactant proteins in type 2 alveolar epithe- 4. Henske EP, Wessner LL, Golden J, et al. Loss of tuber-
lial cells leads to increased ER stress. Mutations in the in in both subependymal giant cell astrocytomas and
HPS and COPA genes alter intracellular protein trans- angiomyolipomas supports a two-hit model for the
port and also upregulate ER stress pathways and lead pathogenesis of tuberous sclerosis tumors. Am J Pathol.
to lung fibrosis. As the common variant located in the 1997;151(6):1639–1647.
5. Carsillo T, Astrinidis A, Henske EP. Mutations in the tu-
MUC5B promoter increases gene expression, this SNP
berous sclerosis complex gene TSC2 are a cause of spo-
may lead to increased susceptibility to lung fibrosis radic pulmonary lymphangioleiomyomatosis. Proc Natl
through its effects on increased mucin expression and Acad Sci USA. 2000;97(11):6085–6090.
increased ER stress. Finally, many different rare and 6. Sato T, Seyama K, Fujii H, et al. Mutation analysis of
common variants linked to genetic ILD share a common the TSC1 and TSC2 genes in Japanese patients with
pathway of telomere shortening. Pulmonary fibrosis has pulmonary lymphangioleiomyomatosis. J Hum Genet.
been estimated to be the most common manifestation 2002;47(1):20–28.
of the monogenic short telomere syndromes, and SNPs 7. Smolarek TA, Wessner LL, McCormack FX, Mylet JC,
in three telomere-related genes reach genomewide sig- Menon AG, Henske EP. Evidence that lymphangiomy-
nificance in a large GWAS study. The link between telo- omatosis is caused by TSC2 mutations: chromosome
16p13 loss of heterozygosity in angiomyolipomas and
mere attrition and lung fibrosis is further strengthened
lymph nodes from women with lymphangiomyomato-
by the finding that telomere length predicts transplant- sis. Am J Hum Genet. 1998;62(4):810–815.
free survival in patients with sporadic (noninherited) 8. Henske EP, McCormack FX. Lymphangioleiomy-
IPF across independent cohorts.257,258 Thus, the study omatosis – a wolf in sheep’s clothing. J Clin Invest.
of rare human patients and families has led to the 2012;122(11):3807–3816.
16 Interstitial Lung Disease

9. Bissler JJ, McCormack FX, Young LR, et al. Siroli- 25. de la Fuente J, Dokal I. Dyskeratosis congenita: advances
mus for angiomyolipoma in tuberous sclerosis com- in the understanding of the telomerase defect and the
plex or lymphangioleiomyomatosis. N Engl J Med. role of stem cell transplantation. Pediatr Transplant.
2008;358(2):140–151. 2007;11(6):584–594.
10. Morton JM, McLean C, Booth SS, Snell GI, Whitford HM. 26. Giri N, Lee R, Faro A, et al. Lung transplantation for
Regression of pulmonary lymphangioleiomyomatosis pulmonary fibrosis in dyskeratosis congenita: Case Re-
(PLAM)-associated retroperitoneal angiomyolipoma port and systematic literature review. BMC Blood Disord.
post-lung transplantation with rapamycin treatment. J 2011;11:3.
Heart Lung Transplant. 2008;27(4):462–465. 27. Imokawa S, Sato A, Toyoshima M, et al. Dyskeratosis
11. Krueger DA, Care MM, Holland K, et al. Everolimus for congenita showing usual interstitial pneumonia. Intern
subependymal giant-cell astrocytomas in tuberous scle- Med. 1994;33(4):226–230.
rosis. N Engl J Med. 2010;363(19):1801–1811. 28. Verra F, Kouzan S, Saiag P, Bignon J, de Cremoux H.
12. McCormack FX, Inoue Y, Moss J, et al. Efficacy and safety Bronchoalveolar disease in dyskeratosis congenita. Eur
of sirolimus in lymphangioleiomyomatosis. N Engl J Respir J. 1992;5(4):497–499.
Med. 2011;364(17):1595–1606. 29. Parry EM, Alder JK, Qi X, Chen JJ, Armanios M. Syn-
13. Hornstein OP, Knickenberg M. Perifollicular fi- drome complex of bone marrow failure and pulmonary
bromatosis cutis with polyps of the colon–a cutaneo- fibrosis predicts germline defects in telomerase. Blood.
intestinal syndrome sui generis. Arch Dermatol Res. 2011;117(21):5607–5611.
1975;253(2):161–175. 30. Yabe M, Yabe H, Hattori K, et al. Fatal interstitial pul-
14. Toro JR, Pautler SE, Stewart L, et al. Lung cysts, spon- monary disease in a patient with dyskeratosis congenita
taneous pneumothorax, and genetic associations in 89 after allogeneic bone marrow transplantation. Bone Mar-
families with Birt-Hogg-Dube syndrome. Am J Respir Crit row Transplant. 1997;19(4):389–392.
Care Med. 2007;175(10):1044–1053. 31. Rocha V, Devergie A, Socie G, et al. Unusual complica-
15. Toro JR, Wei MH, Glenn GM, et al. BHD mutations, clin- tions after bone marrow transplantation for dyskeratosis
ical and molecular genetic investigations of Birt-Hogg- congenita. Br J Haematol. 1998;103(1):243–248.
Dube syndrome: a new series of 50 families and a review 32. Alter BP, Baerlocher GM, Savage SA, et al. Very short tel-
of published reports. J Med Genet. 2008;45(6):321–331. omere length by flow FISH identifies patients with Dys-
16. Nickerson ML, Warren MB, Toro JR, et al. Mutations in a keratosis Congenita. Blood. 2007;110(5):1439–1447.
novel gene lead to kidney tumors, lung wall defects, and 33. Heiss NS, Knight SW, Vulliamy TJ, et al. X-linked dys-
benign tumors of the hair follicle in patients with the keratosis congenita is caused by mutations in a highly
Birt-Hogg-Dube syndrome. Cancer Cell. 2002;2(2):157– conserved gene with putative nucleolar functions. Nat
164. Genet. 1998;19(1):32–38.
17. Painter JN, Tapanainen H, Somer M, Tukiainen P, Ait- 34. Cohen SB, Graham ME, Lovrecz GO, Bache N, Robin-
tomaki KA. 4-bp deletion in the Birt-Hogg-Dube gene son PJ, Reddel RR. Protein composition of catalytically
(FLCN) causes dominantly inherited spontaneous pneu- active human telomerase from immortal cells. Science.
mothorax. Am J Hum Genet. 2005;76(3):522–527. 2007;315(5820):1850–1853.
18. Graham RB, Nolasco M, Peterlin B, Garcia CK. Nonsense 35. Vulliamy T, Marrone A, Goldman F, et al. The RNA com-
mutations in folliculin presenting as isolated familial ponent of telomerase is mutated in autosomal domi-
spontaneous pneumothorax in adults. Am J Respir Crit nant dyskeratosis congenita. Nature. 2001;413(6854):
Care Med. 2005;172(1):39–44. 432–435.
19. Agarwal PP, Gross BH, Holloway BJ, Seely J, Stark P, 36. Armanios M, Chen JL, Chang YP, et al. Haploinsuf-
Kazerooni EA. Thoracic CT findings in Birt-Hogg-Dube ficiency of telomerase reverse transcriptase leads to
syndrome. AJR Am J Roentgenol. 2011;196(2):349–352. anticipation in autosomal dominant dyskeratosis con-
20. Tobino K, Gunji Y, Kurihara M, et al. Characteristics of genita. Proc Natl Acad Sci USA. 2005;102(44):15960–
pulmonary cysts in Birt-Hogg-Dube syndrome: thin- 15964.
section CT findings of the chest in 12 patients. Eur J 37. Du HY, Pumbo E, Manley P, et al. Complex inheritance
Radiol. 2011;77(3):403–409. pattern of dyskeratosis congenita in two families with
21. Devine MS, Garcia CK. Genetic interstitial lung disease. 2 different mutations in the telomerase reverse tran-
Clin Chest Med. 2012;33(1):95–110. scriptase gene. Blood. 2008;111(3):1128–1130.
22. Drachtman RA, Alter BP. Dyskeratosis congenita. Derma- 38. Aspesi A, Vallero S, Rocci A, et al. Compound heterozy-
tol Clin. 1995;13(1):33–39. gosity for two new TERT mutations in a patient with
23. Dokal I. Dyskeratosis congenita in all its forms. Br J Hae- aplastic anemia. Pediatr Blood Cancer. 2010;55(3):550–
matol. 2000;110(4):768–779. 553.
24. Zinsser F. Atrophia Cutis Reticularis cum Pigmentations, 39. Savage SA, Giri N, Baerlocher GM, Orr N, Lansdorp PM,
Dystrophia Unguium et Leukoplakis oris (Poikiooder- Alter BP. TINF2, a component of the shelterin telomere
mia atrophicans vascularis Jacobi). Ikonogr Dermatol. protection complex, is mutated in dyskeratosis congen-
1910;5:219–223. ita. Am J Hum Genet. 2008;82(2):501–509.
CHAPTER 1 Genetic Interstitial Lung Disease 17

40. Walne AJ, Vulliamy T, Beswick R, Kirwan M, Dokal I. 55. Vulliamy T, Marrone A, Szydlo R, Walne A, Mason PJ,
TINF2 mutations result in very short telomeres: analysis Dokal I. Disease anticipation is associated with pro-
of a large cohort of patients with dyskeratosis congen- gressive telomere shortening in families with dyskera-
ita and related bone marrow failure syndromes. Blood. tosis congenita due to mutations in TERC. Nat Genet.
2008;112(9):3594–3600. 2004;36(5):447–449.
41. Guo Y, Kartawinata M, Li J, et al. Inherited bone mar- 56. Goldman F, Bouarich R, Kulkarni S, et al. The effect of TERC
row failure associated with germline mutation of haploinsufficiency on the inheritance of telomere length.
ACD, the gene encoding telomere protein TPP1. Blood. Proc Natl Acad Sci USA. 2005;102(47):17119–17124.
2014;124(18):2767–2774. 57. Devgan SS, Sanal O, Doil C, et al. Homozygous deficien-
42. Kocak H, Ballew BJ, Bisht K, et al. Hoyeraal-Hreidarsson syn- cy of ubiquitin-ligase ring-finger protein RNF168 mim-
drome caused by a germline mutation in the TEL patch of ics the radiosensitivity syndrome of ataxia-telangiectasia.
the telomere protein TPP1. Genes Dev. 2014;28(19):2090– Cell Death Differ. 2011;18(9):1500–1506.
2102. 58. Stewart GS, Stankovic T, Byrd PJ, et al. RIDDLE immu-
43. Keller RB, Gagne KE, Usmani GN, et al. CTC1 Mutations nodeficiency syndrome is linked to defects in 53BP1-
in a patient with dyskeratosis congenita. Pediatr Blood mediated DNA damage signaling. Proc Natl Acad Sci
Cancer. 2012;59(2):311–314. USA. 2007;104(43):16910–16915.
44. Walne AJ, Bhagat T, Kirwan M, et al. Mutations in the 59. Doil C, Mailand N, Bekker-Jensen S, et al. RNF168 binds
telomere capping complex in bone marrow failure and and amplifies ubiquitin conjugates on damaged chro-
related syndromes. Haematologica. 2013;98(3):334–338. mosomes to allow accumulation of repair proteins. Cell.
45. Walne AJ, Vulliamy T, Marrone A, et al. Genetic heterogene- 2009;136(3):435–446.
ity in autosomal recessive dyskeratosis congenita with one 60. Stewart GS, Panier S, Townsend K, et al. The RIDDLE syn-
subtype due to mutations in the telomerase-associated pro- drome protein mediates a ubiquitin-dependent signaling
tein NOP10. Hum Mol Genet. 2007;16(13):1619–1629. cascade at sites of DNA damage. Cell. 2009;136(3):420–
46. Trahan C, Martel C, Dragon F. Effects of dyskeratosis 434.
congenita mutations in dyskerin, NHP2 and NOP10 61. Okamoto K, Bartocci C, Ouzounov I, Diedrich JK,
on assembly of H/ACA pre-RNPs. Hum Mol Genet. Yates 3rd JR, Denchi EL. A two-step mechanism for
2010;19(5):825–836. TRF2-mediated chromosome-end protection. Nature.
47. Vulliamy T, Beswick R, Kirwan M, et al. Mutations in the 2013;494(7438):502–505.
telomerase component NHP2 cause the premature age- 62. Hermansky F, Pudlak P. Albinism associated with hem-
ing syndrome dyskeratosis congenita. Proc Natl Acad Sci orrhagic diathesis and unusual pigmented reticular cells
USA. 2008;105(23):8073–8078. in the bone marrow: report of two cases with histochem-
48. Zhong F, Savage SA, Shkreli M, et al. Disruption of tel- ical studies. Blood. 1959;14(2):162–169.
omerase trafficking by TCAB1 mutation causes dyskera- 63. Gahl WA, Brantly M, Kaiser-Kupfer MI, et al. Genetic de-
tosis congenita. Genes Dev. 2011;25(1):11–16. fects and clinical characteristics of patients with a form
49. Walne AJ, Vulliamy T, Kirwan M, Plagnol V, Dokal I. of oculocutaneous albinism (Hermansky-Pudlak syn-
Constitutional mutations in RTEL1 cause severe dyskera- drome). N Engl J Med. 1998;338(18):1258–1264.
tosis congenita. Am J Hum Genet. 2013;92(3):448–453. 64. Witkop CJ, Krumwiede M, Sedano H, White JG. Reliabil-
50. Le Guen T, Jullien L, Touzot F, et al. Human RTEL1 de- ity of absent platelet dense bodies as a diagnostic cri-
ficiency causes Hoyeraal-Hreidarsson syndrome with terion for Hermansky-Pudlak syndrome. Am J Hematol.
short telomeres and genome instability. Hum Mol Genet. 1987;26(4):305–311.
2013;22(16):3239–3249. 65. Dell’Angelica EC, Shotelersuk V, Aguilar RC, Gahl WA,
51. Ballew BJ, Yeager M, Jacobs K, et al. Germline mutations Bonifacino JS. Altered trafficking of lysosomal proteins
of regulator of telomere elongation helicase 1, RTEL1, in in Hermansky-Pudlak syndrome due to mutations
Dyskeratosis congenita. Hum Genet. 2013;132(4):473– in the beta 3A subunit of the AP-3 adaptor. Mol Cell.
480. 1999;3(1):11–21.
52. Tummala H, Walne A, Collopy L, et al. Poly(A)-specific 66. Morgan NV, Pasha S, Johnson CA, et al. A germline mu-
ribonuclease deficiency impacts telomere biology and tation in BLOC1S3/reduced pigmentation causes a novel
causes dyskeratosis congenita. J Clin Invest. 2015;125(5): variant of Hermansky-Pudlak syndrome (HPS8). Am J
2151–2160. Hum Genet. 2006;78(1):160–166.
53. Dhanraj S, Gunja SM, Deveau AP, et al. Bone marrow 67. Martina JA, Moriyama K, Bonifacino JS. BLOC-3, a
failure and developmental delay caused by mutations protein complex containing the Hermansky-Pudlak
in poly(A)-specific ribonuclease (PARN). J Med Genet. syndrome gene products HPS1 and HPS4. J Biol Chem.
2015;52(11):738–748. 2003;278(31):29376–29384.
54. Moon DH, Segal M, Boyraz B, et al. Poly(A)-specific 68. Suzuki T, Li W, Zhang Q, et al. The gene mutated in
ribonuclease (PARN) mediates 3’-end maturation of the cocoa mice, carrying a defect of organelle biogenesis,
telomerase RNA component. Nat Genet. 2015;47(12): is a homologue of the human Hermansky-Pudlak syn-
1482–1488. drome-3 gene. Genomics. 2001;78(1–2):30–37.
18 Interstitial Lung Disease

69. Zhang Q, Zhao B, Li W, et al. Ru2 and Ru encode 83. Guttentag SH, Akhtar A, Tao JQ, et al. Defective surfactant
mouse orthologs of the genes mutated in human secretion in a mouse model of Hermansky-Pudlak syn-
Hermansky-Pudlak syndrome types 5 and 6. Nat Genet. drome. Am J Respir Cell Mol Biol. 2005;33(1):14–21.
2003;33(2):145–153. 84. Gras D, Jonard L, Roze E, et al. Benign hereditary chorea:
70. Li W, Zhang Q, Oiso N, et al. Hermansky-Pudlak syn- phenotype, prognosis, therapeutic outcome and long
drome type 7 (HPS-7) results from mutant dysbindin, term follow-up in a large series with new mutations in
a member of the biogenesis of lysosome-related orga- the TITF1/NKX2-1 gene. J Neurol Neurosurg Psychiatry.
nelles complex 1 (BLOC-1). Nat Genet. 2003;35(1): 2012;83(10):956–962.
84–89. 85. Young LR, Deutsch GH, Bokulic RE, Brody AS, Nogee LM.
71. Wildenberg SC, Oetting WS, Almodovar C, Krumwiede M, A mutation in TTF1/NKX2.1 is associated with famil-
White JG, King RA. A gene causing Hermansky-Pudlak ial neuroendocrine cell hyperplasia of infancy. Chest.
syndrome in a Puerto Rican population maps to chro- 2013;144(4):1199–1206.
mosome 10q2. Am J Hum Genet. 1995;57(4):755–765. 86. Hamvas A, Deterding RR, Wert SE, et al. Heterogene-
72. Brantly M, Avila NA, Shotelersuk V, Lucero C, Huizing ous pulmonary phenotypes associated with mutations
M, Gahl WA. Pulmonary function and high-resolution in the thyroid transcription factor gene NKX2-1. Chest.
CT findings in patients with an inherited form of pul- 2013;144(3):794–804.
monary fibrosis, Hermansky-Pudlak syndrome, due to 87. Carre A, Szinnai G, Castanet M, et al. Five new TTF1/
mutations in HPS-1. Chest. 2000;117(1):129–136. NKX2.1 mutations in brain-lung-thyroid syndrome:
73. Avila NA, Brantly M, Premkumar A, Huizing M, rescue by PAX8 synergism in one case. Hum Mol Genet.
Dwyer A, Gahl WA. Hermansky-Pudlak syndrome: radi- 2009;18(12):2266–2276.
ography and CT of the chest compared with pulmonary 88. Peca D, Petrini S, Tzialla C, et al. Altered surfactant ho-
function tests and genetic studies. AJR Am J Roentgenol. meostasis and recurrent respiratory failure secondary to
2002;179(4):887–892. TTF-1 nuclear targeting defect. Respir Res. 2011;12:115.
74. Anderson PD, Huizing M, Claassen DA, White J, Gahl 89. Burkhalter JL, Morano JU, McCay MB. Diffuse inter-
WA. Hermansky-Pudlak syndrome type 4 (HPS-4): stitial lung disease in neurofibromatosis. South Med J.
clinical and molecular characteristics. Hum Genet. 1986;79(8):944–946.
2003;113(1):10–17. 90. Massaro D, Katz S. Fibrosing alveolitis: its occurrence,
75. Bachli EB, Brack T, Eppler E, et al. Hermansky-Pudlak roentgenographic, and pathologic features in von Reck-
syndrome type 4 in a patient from Sri Lanka with pul- linghausen’s neurofibromatosis. Am Rev Respir Dis.
monary fibrosis. Am J Med Genet A. 2004;127A(2):201– 1966;93(6):934–942.
207. 91. Webb WR, Goodman PC. Fibrosing alveolitis in patients
76. Gahl WA, Brantly M, Troendle J, et al. Effect of pirfeni- with neurofibromatosis. Radiology. 1977;122(2):289–293.
done on the pulmonary fibrosis of Hermansky-Pudlak 92. Porterfield JK, Pyeritz RE, Traill TA. Pulmonary hyperten-
syndrome. Mol Genet Metab. 2002;76(3):234–242. sion and interstitial fibrosis in von Recklinghausen neu-
77. Gochuico BR, Huizing M, Golas GA, et al. Interstitial rofibromatosis. Am J Med Genet. 1986;25(3):531–535.
lung disease and pulmonary fibrosis in Hermansky- 93. Khumalo NP, Pillay K, Beighton P, et al. Poikilo-
Pudlak syndrome type 2, an adaptor protein-3 complex derma, tendon contracture and pulmonary fibrosis: a
disease. Mol Med. 2012;18:56–64. new autosomal dominant syndrome? Br J Dermatol.
78. Nakatani Y, Nakamura N, Sano J, et al. Interstitial pneu- 2006;155(5):1057–1061.
monia in Hermansky-Pudlak syndrome: significance of 94. Mercier S, Kury S, Shaboodien G, et al. Mutations in
florid foamy swelling/degeneration (giant lamellar body FAM111B cause hereditary fibrosing poikiloderma with
degeneration) of type-2 pneumocytes. Virchows Arch. tendon contracture, myopathy, and pulmonary fibrosis.
2000;437(3):304–313. Am J Hum Genet. 2013;93(6):1100–1107.
79. Pierson DM, Ionescu D, Qing G, et al. Pulmonary fibro- 95. Has C, Sparta G, Kiritsi D, et al. Integrin alpha3 muta-
sis in hermansky-pudlak syndrome. a case report and tions with kidney, lung, and skin disease. N Engl J Med.
review. Respiration. 2006;73(3):382–395. 2012;366(16):1508–1514.
80. O’Brien K, Troendle J, Gochuico BR, et al. Pirfenidone 96. Yalcin EG, He Y, Orhan D, Pazzagli C, Emiralioglu N,
for the treatment of Hermansky-Pudlak syndrome pul- Has C. Crucial role of posttranslational modifications of
monary fibrosis. Mol Genet Metab. 2011;103(2):128–134. integrin alpha3 in interstitial lung disease and nephrotic
81. Lyerla TA, Rusiniak ME, Borchers M, et al. Aberrant lung syndrome. Hum Mol Genet. 2015;24(13):3679–3688.
structure, composition, and function in a murine model 97. Liu Y, Jesus AA, Marrero B, et al. Activated STING in
of Hermansky-Pudlak syndrome. Am J Physiol Lung Cell a vascular and pulmonary syndrome. N Engl J Med.
Mol Physiol. 2003;285(3):L643–L653. 2014;371(6):507–518.
82. Mahavadi P, Korfei M, Henneke I, et al. Epithelial stress 98. Jeremiah N, Neven B, Gentili M, et al. Inherited STING-
and apoptosis underlie Hermansky-Pudlak syndrome- activating mutation underlies a familial inflammatory
associated interstitial pneumonia. Am J Respir Crit Care syndrome with lupus-like manifestations. J Clin Invest.
Med. 2010;182(2):207–219. 2014;124(12):5516–5520.
CHAPTER 1 Genetic Interstitial Lung Disease 19

99. Lohr NJ, Molleston JP, Strauss KA, et al. Human 114. Salzer U, Chapel HM, Webster AD, et al. Mutations in
ITCH E3 ubiquitin ligase deficiency causes syndromic TNFRSF13B encoding TACI are associated with com-
multisystem autoimmune disease. Am J Hum Genet. mon variable immunodeficiency in humans. Nat Genet.
2010;86(3):447–453. 2005;37(8):820–828.
100. Hustad CM, Perry WL, Siracusa LD, et al. Molecular ge- 115. Castigli E, Wilson SA, Garibyan L, et al. TACI is mutant
netic characterization of six recessive viable alleles of the in common variable immunodeficiency and IgA defi-
mouse agouti locus. Genetics. 1995;140(1):255–265. ciency. Nat Genet. 2005;37(8):829–834.
101. Perry WL, Hustad CM, Swing DA, O’Sullivan TN, Jenkins 116. Warnatz K, Salzer U, Rizzi M, et al. B-cell activating fac-
NA, Copeland NG. The itchy locus encodes a novel ubiq- tor receptor deficiency is associated with an adult-onset
uitin protein ligase that is disrupted in a18H mice. Nat antibody deficiency syndrome in humans. Proc Natl Acad
Genet. 1998;18(2):143–146. Sci USA. 2009;106(33):13945–13950.
102. Watkin LB, Jessen B, Wiszniewski W, et al. COPA mu- 117. van Zelm MC, Reisli I, van der Burg M, et al. An antibody-
tations impair ER-Golgi transport and cause hereditary deficiency syndrome due to mutations in the CD19 gene.
autoimmune-mediated lung disease and arthritis. Nat N Engl J Med. 2006;354(18):1901–1912.
Genet. 2015;47(6):654–660. 118. Kuijpers TW, Bende RJ, Baars PA, et al. CD20 deficiency
103. Ostergaard P, Simpson MA, Connell FC, et al. Mutations in humans results in impaired T cell-independent anti-
in GATA2 cause primary lymphedema associated with body responses. J Clin Invest. 2010;120(1):214–222.
a predisposition to acute myeloid leukemia (Emberger 119. van Zelm MC, Smet J, Adams B, et al. CD81 gene defect
syndrome). Nat Genet. 2011;43(10):929–931. in humans disrupts CD19 complex formation and leads
104. Hahn CN, Chong CE, Carmichael CL, et al. Heritable to antibody deficiency. J Clin Invest. 2010;120(4):1265–
GATA2 mutations associated with familial myelodysplastic 1274.
syndrome and acute myeloid leukemia. Nat Genet. 120. Thiel J, Kimmig L, Salzer U, et al. Genetic CD21 deficien-
2011;43(10):1012–1017. cy is associated with hypogammaglobulinemia. J Allergy
105. Dickinson RE, Griffin H, Bigley V, et al. Exome sequenc- Clin Immunol. 2012;129(3):801–810.e806.
ing identifies GATA-2 mutation as the cause of dendritic 121. Lopez-Herrera G, Tampella G, Pan-Hammarstrom Q,
cell, monocyte, B and NK lymphoid deficiency. Blood. et al. Deleterious mutations in LRBA are associated with
2011;118(10):2656–2658. a syndrome of immune deficiency and autoimmunity.
106. Hsu AP, Sampaio EP, Khan J, et al. Mutations in GATA2 Am J Hum Genet. 2012;90(6):986–1001.
are associated with the autosomal dominant and sporad- 122. Alangari A, Alsultan A, Adly N, et al. LPS-responsive
ic monocytopenia and mycobacterial infection (Mono- beige-like anchor (LRBA) gene mutation in a family with
MAC) syndrome. Blood. 2011;118(10):2653–2655. inflammatory bowel disease and combined immunode-
107. Spinner MA, Sanchez LA, Hsu AP, et al. GATA2 deficien- ficiency. J Allergy Clin Immunol. 2012;130(2):481–488.
cy: a protean disorder of hematopoiesis, lymphatics, and e482.
immunity. Blood. 2014;123(6):809–821. 123. Salzer E, Kansu A, Sic H, et al. Early-onset inflammatory
108. Hsu AP, Johnson KD, Falcone EL, et al. GATA2 haplo- bowel disease and common variable immunodeficiency-
insufficiency caused by mutations in a conserved in- like disease caused by IL-21 deficiency. J Allergy Clin Im-
tronic element leads to MonoMAC syndrome. Blood. munol. 2014;133(6):1651–1659.e1612.
2013;121(19):3830–3837. S3831–S3837. 124. Fliegauf M, Bryant VL, Frede N, et al. Haploinsufficiency
109. Aghamohammadi A, Allahverdi A, Abolhassani H, et al. of the NF-kappaB1 Subunit p50 in common variable im-
Comparison of pulmonary diseases in common variable munodeficiency. Am J Hum Genet. 2015;97(3):389–403.
immunodeficiency and X-linked agammaglobulinae- 125. Chen K, Coonrod EM, Kumanovics A, et al. Germline
mia. Respirology. 2010;15(2):289–295. mutations in NFKB2 implicate the noncanonical NF-
110. Verma N, Grimbacher B, Hurst JR. Lung disease in primary kappaB pathway in the pathogenesis of common variable
antibody deficiency. Lancet Respir Med. 2015;3(8):651–660. immunodeficiency. Am J Hum Genet. 2013;93(5):812–
111. Maarschalk-Ellerbroek LJ, de Jong PA, van Montfrans JM, 824.
et al. CT screening for pulmonary pathology in common 126. Goldman FD, Gurel Z, Al-Zubeidi D, et al. Congenital
variable immunodeficiency disorders and the correla- pancytopenia and absence of B lymphocytes in a neo-
tion with clinical and immunological parameters. J Clin nate with a mutation in the Ikaros gene. Pediatr Blood
Immunol. 2014;34(6):642–654. Cancer. 2012;58(4):591–597.
112. Gregersen S, Aalokken TM, Mynarek G, et al. High reso- 127. Kuehn HS, Boisson B, Cunningham-Rundles C,
lution computed tomography and pulmonary function et al. Loss of B cells in patients with heterozygous
in common variable immunodeficiency. Respir Med. mutations in IKAROS. N Engl J Med. 2016;374(11):
2009;103(6):873–880. 1032–1043.
113. Grimbacher B, Hutloff A, Schlesier M, et al. Homozygous 128. Chen Y, Stirling RG, Paul E, Hore-Lacy F, Thompson BR,
loss of ICOS is associated with adult-onset common Douglass JA. Longitudinal decline in lung function in
variable immunodeficiency. Nat Immunol. 2003;4(3): patients with primary immunoglobulin deficiencies. J
261–268. Allergy Clin Immunol. 2011;127(6):1414–1417.
20 Interstitial Lung Disease

129. Vetrie D, Vorechovsky I, Sideras P, et al. The gene in- 144. Partington MW, Prentice RS. X-linked cutaneous amyloi-
volved in X-linked agammaglobulinaemia is a member dosis: further clinical and pathological observations. Am
of the src family of protein-tyrosine kinases. Nature. J Med Genet. 1989;32(1):115–119.
1993;361(6409):226–233. 145. Ades LC, Rogers M, Sillence DO. An X-linked reticulate
130. Yel L, Minegishi Y, Coustan-Smith E, et al. Mutations in pigmentary disorder with systemic manifestations: report
the mu heavy-chain gene in patients with agammaglob- of a second family. Pediatr Dermatol. 1993;10(4):344–351.
ulinemia. N Engl J Med. 1996;335(20):1486–1493. 146. Anderson RC, Zinn AR, Kim J, Carder KR. X-linked retic-
131. Minegishi Y, Coustan-Smith E, Wang YH, Cooper MD, ulate pigmentary disorder with systemic manifestations:
Campana D, Conley ME. Mutations in the human lamb- report of a third family and literature review. Pediatr Der-
da5/14.1 gene result in B cell deficiency and agamma- matol. 2005;22(2):122–126.
globulinemia. J Exp Med. 1998;187(1):71–77. 147. Starokadomskyy P, Gemelli T, Rios JJ, et al. DNA
132. Minegishi Y, Coustan-Smith E, Rapalus L, Ersoy F, polymerase-alpha regulates the activation of type I
Campana D, Conley ME. Mutations in Igalpha (CD79a) interferons through cytosolic RNA: DNA synthesis. Nat
result in a complete block in B-cell development. J Clin Immunol. 2016;17(5):495–504.
Invest. 1999;104(8):1115–1121. 148. Kerem E, Elstein D, Abrahamov A, et al. Pulmonary func-
133. Minegishi Y, Rohrer J, Coustan-Smith E, et al. An essen- tion abnormalities in type I Gaucher disease. Eur Respir J.
tial role for BLNK in human B cell development. Science. 1996;9(2):340–345.
1999;286(5446):1954–1957. 149. Miller A, Brown LK, Pastores GM, Desnick RJ. Pulmonary
134. Dobbs AK, Yang T, Farmer D, Kager L, Parolini O, involvement in type 1 Gaucher disease: functional and
Conley ME. Cutting edge: a hypomorphic mutation exercise findings in patients with and without clinical in-
in Igbeta (CD79b) in a patient with immunodeficiency terstitial lung disease. Clin Genet. 2003;63(5):368–376.
and a leaky defect in B cell development. J Immunol. 150. Goitein O, Elstein D, Abrahamov A, et al. Lung involve-
2007;179(4):2055–2059. ment and enzyme replacement therapy in Gaucher’s dis-
135. Boisson B, Wang YD, Bosompem A, et al. A recur- ease. QJM. 2001;94(8):407–415.
rent dominant negative E47 mutation causes agam- 151. Jmoudiak M, Futerman AH. Gaucher disease: pathologi-
maglobulinemia and BCR(-) B cells. J Clin Invest. cal mechanisms and modern management. Br J Haema-
2013;123(11):4781–4785. tol. 2005;129(2):178–188.
136. Freeman AF, Holland SM. Clinical manifestations, etiol- 152. McGovern MM, Wasserstein MP, Giugliani R, et al. A
ogy, and pathogenesis of the hyper-IgE syndromes. Pedi- prospective, cross-sectional survey study of the natu-
atr Res. 2009;65(5 Pt 2):32R–37R. ral history of Niemann-Pick disease type B. Pediatrics.
137. Freeman AF, Kleiner DE, Nadiminti H, et al. Causes of 2008;122(2):e341–e349.
death in hyper-IgE syndrome. J Allergy Clin Immunol. 153. Mendelson DS, Wasserstein MP, Desnick RJ, et al. Type
2007;119(5):1234–1240. B Niemann-Pick disease: findings at chest radiography,
138. Minegishi Y, Saito M, Tsuchiya S, et al. Dominant- thin-section CT, and pulmonary function testing. Radiol-
negative mutations in the DNA-binding domain of STAT3 ogy. 2006;238(1):339–345.
cause hyper-IgE syndrome. Nature. 2007;448(7157): 154. Parto K, Svedstrom E, Majurin ML, Harkonen R, Simell O.
1058–1062. Pulmonary manifestations in lysinuric protein intolerance.
139. Milner JD, Brenchley JM, Laurence A, et al. Impaired Chest. 1993;104(4):1176–1182.
T(H)17 cell differentiation in subjects with autosomal 155. Nogee LM. Genetic basis of children’s interstitial lung
dominant hyper-IgE syndrome. Nature. 2008;452(7188): disease. Pediatr Allergy Immunol Pulmonol. 2010;23(1).
773–776. 156. Nogee LM, de Mello DE, Dehner LP, Colten HR. Brief
140. Angulo I, Vadas O, Garcon F, et al. Phosphoinositide report: deficiency of pulmonary surfactant protein
3-kinase delta gene mutation predisposes to respiratory B in congenital alveolar proteinosis. N Engl J Med.
infection and airway damage. Science. 2013;342(6160): 1993;328(6):406–410.
866–871. 157. Nogee LM, Garnier G, Dietz HC, et al. A mutation in the
141. Lucas CL, Kuehn HS, Zhao F, et al. Dominant-activating surfactant protein B gene responsible for fatal neonatal
germline mutations in the gene encoding the PI(3)K respiratory disease in multiple kindreds. J Clin Invest.
catalytic subunit p110delta result in T cell senescence and 1994;93(4):1860–1863.
human immunodeficiency. Nat Immunol. 2014;15(1): 158. Andersen C, Ramsay JA, Nogee LM, et al. Recurrent fa-
88–97. milial neonatal deaths: hereditary surfactant protein B
142. Kuehn HS, Ouyang W, Lo B, et al. Immune dysregu- deficiency. Am J Perinatol. 2000;17(4):219–224.
lation in human subjects with heterozygous ger- 159. Cole FS, Hamvas A, Rubinstein P, et al. Population-
mline mutations in CTLA4. Science. 2014;345(6204): based estimates of surfactant protein B deficiency. Pedi-
1623–1627. atrics. 2000;105(3 Pt 1):538–541.
143. Schubert D, Bode C, Kenefeck R, et al. Autosomal domi- 160. Somaschini M, Wert S, Mangili G, Colombo A, Nogee L.
nant immune dysregulation syndrome in humans with Hereditary surfactant protein B deficiency resulting from
CTLA4 mutations. Nature Med. 2014;20(12):1410–1416. a novel mutation. Intensive Care Med. 2000;26(1):97–100.
CHAPTER 1 Genetic Interstitial Lung Disease 21

161. Wegner DJ, Hertzberg T, Heins HB, et al. A major 176. Stevens PA, Pettenazzo A, Brasch F, et al. Nonspecific
deletion in the surfactant protein-B gene causing le- interstitial pneumonia, alveolar proteinosis, and abnor-
thal respiratory distress. Acta Paediatr. 2007;96(4): mal proprotein trafficking resulting from a spontaneous
516–520. mutation in the surfactant protein C gene. Pediatr Res.
162. Dunbar 3rd AE, Wert SE, Ikegami M, et al. Prolonged 2005;57(1):89–98.
survival in hereditary surfactant protein B (SP-B) defi- 177. Woischnik M, Sparr C, Kern S, et al. A non-BRICHOS
ciency associated with a novel splicing mutation. Pediatr surfactant protein c mutation disrupts epithelial cell
Res. 2000;48(3):275–282. function and intercellular signaling. BMC Cell Biol.
163. Ballard PL, Nogee LM, Beers MF, et al. Partial deficiency 2010;11:88.
of surfactant protein B in an infant with chronic lung 178. Bridges JP, Xu Y, Na CL, Wong HR, Weaver TE. Adapta-
disease. Pediatrics. 1995;96(6):1046–1052. tion and increased susceptibility to infection associated
164. Nogee LM, Dunbar 3rd AE, Wert SE, Askin F, Hamvas A, with constitutive expression of misfolded SP-C. J Cell
Whitsett JA. A mutation in the surfactant protein C gene Biol. 2006;172(3):395–407.
associated with familial interstitial lung disease. N Engl J 179. Garmany TH, Wambach JA, Heins HB, et al. Population
Med. 2001;344(8):573–579. and disease-based prevalence of the common muta-
165. Thomas AQ, Lane K, Phillips 3rd J, et al. Heterozy- tions associated with surfactant deficiency. Pediatr Res.
gosity for a surfactant protein C gene mutation as- 2008;63(6):645–649.
sociated with usual interstitial pneumonitis and 180. Somaschini M, Nogee LM, Sassi I, et al. Unexplained
cellular nonspecific interstitial pneumonitis in one neonatal respiratory distress due to congenital sur-
kindred. Am J Respir Crit Care Med. 2002;165(9): factant deficiency. J Pediatr. 2007;150(6):649–653.
1322–1328. 653.e641.
166. Abou Taam R, Jaubert F, Emond S, et al. Familial inter- 181. Weaver TE. Synthesis, processing and secretion of
stitial disease with I73T mutation: a mid- and long-term surfactant proteins B and C. Biochim Biophys Acta.
study. Pediatr Pulmonol. 2009;44(2):167–175. 1998;1408(2–3):173–179.
167. Brasch F, Griese M, Tredano M, et al. Interstitial lung dis- 182. Serrano AG, Perez-Gil J. Protein-lipid interactions and
ease in a baby with a de novo mutation in the SFTPC surface activity in the pulmonary surfactant system.
gene. Eur Respir J. 2004;24(1):30–39. Chem Phys Lipids. 2006;141(1–2):105–118.
168. Bullard JE, Nogee LM. Heterozygosity for ABCA3 mu- 183. Matsumura Y, Ban N, Inagaki N. Aberrant catalytic cycle
tations modifies the severity of lung disease associated and impaired lipid transport into intracellular vesicles
with a surfactant protein C gene (SFTPC) mutation. Pedi- in ABCA3 mutants associated with nonfatal pediatric in-
atr Res. 2007;62(2):176–179. terstitial lung disease. Am J Physiol Lung Cell Mol Physiol.
169. Cameron HS, Somaschini M, Carrera P, et al. A common 2008;295(4):L698–L707.
mutation in the surfactant protein C gene associated 184. Matsumura Y, Ban N, Ueda K, Inagaki N. Characteriza-
with lung disease. J Pediatr. 2005;146(3):370–375. tion and classification of ATP-binding cassette transport-
170. Soraisham AS, Tierney AJ, Amin HJ. Neonatal respira- er ABCA3 mutants in fatal surfactant deficiency. J Biol
tory failure associated with mutation in the surfactant Chem. 2006;281(45):34503–34514.
protein C gene. J Perinatol. 2006;26(1):67–70. 185. Cheong N, Madesh M, Gonzales LW, et al. Functional
171. van Moorsel CH, van Oosterhout MF, Barlo NP, and trafficking defects in ATP binding cassette A3 mu-
et al. Surfactant protein C mutations are the basis tants associated with respiratory distress syndrome. J Biol
of a significant portion of adult familial pulmonary Chem. 2006;281(14):9791–9800.
fibrosis in a dutch cohort. Am J Respir Crit Care Med. 186. Trapnell BC, Whitsett JA, Nakata K. Pulmonary alveolar
2010;182(11):1419–1425. proteinosis. N Engl J Med. 2003;349(26):2527–2539.
172. Lawson WE, Grant SW, Ambrosini V, et al. Genetic muta- 187. Prakash UB, Barham SS, Carpenter HA, Dines DE,
tions in surfactant protein C are a rare cause of sporadic Marsh HM. Pulmonary alveolar phospholipoproteino-
cases of IPF. Thorax. 2004;59(11):977–980. sis: experience with 34 cases and a review. Mayo Clin
173. Markart P, Ruppert C, Wygrecka M, et al. Surfactant Proc. 1987;62(6):499–518.
protein C mutations in sporadic forms of idiopathic 188. Goldstein LS, Kavuru MS, Curtis-McCarthy P, Christie HA,
interstitial pneumonias. Eur Respir J. 2007;29(1): Farver C, Stoller JK. Pulmonary alveolar proteinosis: clini-
134–137. cal features and outcomes. Chest. 1998;114(5):1357–1362.
174. Wert SE, Whitsett JA, Nogee LM. Genetic disorders of 189. Seymour JF, Presneill JJ. Pulmonary alveolar proteinosis:
surfactant dysfunction. Pediatr Dev Pathol. 2009;12(4): progress in the first 44 years. Am J Respir Crit Care Med.
253–274. 2002;166(2):215–235.
175. Mulugeta S, Maguire JA, Newitt JL, Russo SJ, Kotorash- 190. Kitamura T, Tanaka N, Watanabe J, et al. Idiopathic
vili A, Beers MF. Misfolded BRICHOS SP-C mutant pro- pulmonary alveolar proteinosis as an autoimmune
teins induce apoptosis via caspase-4- and cytochrome disease with neutralizing antibody against granulo-
c-related mechanisms. Am J Physiol Lung Cell Mol Physiol. cyte/macrophage colony-stimulating factor. J Exp Med.
2007;293(3):L720–L729. 1999;190(6):875–880.
22 Interstitial Lung Disease

191. Bonfield TL, Russell D, Burgess S, Malur A, Kavuru MS, 205. Hadchouel A, Wieland T, Griese M, et al. Biallelic muta-
Thomassen MJ. Autoantibodies against granulocyte tions of methionyl-tRNA synthetase cause a specific type
macrophage colony-stimulating factor are diagnostic for of pulmonary alveolar proteinosis prevalent on reunion
pulmonary alveolar proteinosis. Am J Respir Cell Mol Biol. island. Am J Hum Genet. 2015;96(5):826–831.
2002;27(4):481–486. 206. van Meel E, Wegner DJ, Cliften P, et al. Rare recessive
192. Lin FC, Chang GD, Chern MS, Chen YC, Chang SC. loss-of-function methionyl-tRNA synthetase mutations
Clinical significance of anti-GM-CSF antibodies in presenting as a multi-organ phenotype. BMC Med Genet.
idiopathic pulmonary alveolar proteinosis. Thorax. 2013;14:106.
2006;61(6):528–534. 207. Armanios MY, Chen JJ, Cogan JD, et al. Telomerase mu-
193. Inoue Y, Trapnell BC, Tazawa R, et al. Characteristics of tations in families with idiopathic pulmonary fibrosis. N
a large cohort of patients with autoimmune pulmonary Engl J Med. 2007;356(13):1317–1326.
alveolar proteinosis in Japan. Am J Respir Crit Care Med. 208. Tsakiri KD, Cronkhite JT, Kuan PJ, et al. Adult-onset pul-
2008;177(7):752–762. monary fibrosis caused by mutations in telomerase. Proc
194. Carey B, Trapnell BC. The molecular basis of pulmo- Natl Acad Sci USA. 2007;104(18):7552–7557.
nary alveolar proteinosis. Clin Immunol. 2010;135(2): 209. Cronkhite JT, Xing C, Raghu G, et al. Telomere shorten-
223–235. ing in familial and sporadic pulmonary fibrosis. Am J
195. Uchida K, Beck DC, Yamamoto T, et al. GM-CSF autoan- Respir Crit Care Med. 2008;178(7):729–737.
tibodies and neutrophil dysfunction in pulmonary al- 210. Diaz de Leon A, Cronkhite JT, Katzenstein AL, et al.
veolar proteinosis. N Engl J Med. 2007;356(6):567–579. Telomere lengths, pulmonary fibrosis and telomerase
196. Uchida K, Nakata K, Trapnell BC, et al. High-affinity (TERT) mutations. PLoS One. 2010;5(5):e10680.
autoantibodies specifically eliminate granulocyte- 211. Armanios M. Telomerase and idiopathic pulmonary fi-
macrophage colony-stimulating factor activity in the brosis. Mutat Res. 2012;730(1–2):52–58.
lungs of patients with idiopathic pulmonary alveolar 212. Yamaguchi H, Calado RT, Ly H, et al. Mutations in TERT,
proteinosis. Blood. 2004;103(3):1089–1098. the gene for telomerase reverse transcriptase, in aplastic
197. Hansen G, Hercus TR, McClure BJ, et al. The structure of anemia. N Engl J Med. 2005;352(14):1413–1424.
the GM-CSF receptor complex reveals a distinct mode 213. Calado RT, Brudno J, Mehta P, et al. Constitutional tel-
of cytokine receptor activation. Cell. 2008;134(3): omerase mutations are genetic risk factors for cirrhosis.
496–507. Hepatology. 2011;53(5):1600–1607.
198. Suzuki T, Sakagami T, Rubin BK, et al. Familial pulmo- 214. Hartmann D, Srivastava U, Thaler M, et al. Telomerase
nary alveolar proteinosis caused by mutations in CS- gene mutations are associated with cirrhosis formation.
F2RA. J Exp Med. 2008;205(12):2703–2710. Hepatology. 2011;53(5):1608–1617.
199. Suzuki T, Sakagami T, Young LR, et al. Hereditary pul- 215. Diaz de Leon A, Cronkhite JT, Yilmaz C, et al. Subclini-
monary alveolar proteinosis: pathogenesis, presenta- cal lung disease, macrocytosis, and premature graying
tion, diagnosis, and therapy. Am J Respir Crit Care Med. in kindreds with telomerase (TERT) mutations. Chest.
2010;182(10):1292–1304. 2011;140(3):753–763.
200. Tanaka T, Motoi N, Tsuchihashi Y, et al. Adult-onset 216. Calado RT, Regal JA, Hills M, et al. Constitutional hy-
hereditary pulmonary alveolar proteinosis caused pomorphic telomerase mutations in patients with
by a single-base deletion in CSF2RB. J Med Genet. acute myeloid leukemia. Proc Natl Acad Sci USA.
2011;48(3):205–209. 2009;106(4):1187–1192.
201. Dranoff G, Crawford AD, Sadelain M, et al. Involvement of 217. Kirwan M, Vulliamy T, Marrone A, et al. Defining the
granulocyte-macrophage colony-stimulating factor in pul- pathogenic role of telomerase mutations in myelodys-
monary homeostasis. Science. 1994;264(5159):713–716. plastic syndrome and acute myeloid leukemia. Hum Mu-
202. Stanley E, Lieschke GJ, Grail D, et al. Granulocyte/mac- tat. 2009;30(11):1567–1573.
rophage colony-stimulating factor-deficient mice show 218. Newton C, Batra K, Torrealba J, et al. Telomere-related
no major perturbation of hematopoiesis but develop a lung fibrosis is diagnostically heterogeneous but uni-
characteristic pulmonary pathology. Proc Natl Acad Sci formly progressive. Eur Respir J. 2016;48(6):1710–1720.
USA. 1994;91(12):5592–5596. 219. Silhan LL, Shah PD, Chambers DC, et al. Lung
203. Nishinakamura R, Nakayama N, Hirabayashi Y, et al. transplantation in telomerase mutation carriers
Mice deficient for the IL-3/GM-CSF/IL-5 beta c receptor with pulmonary fibrosis. Eur Respir J. 2014;44(1):
exhibit lung pathology and impaired immune response, 178–187.
while beta IL3 receptor-deficient mice are normal. Im- 220. Borie R, Kannengiesser C, Hirschi S, et al. Severe hema-
munity. 1995;2(3):211–222. tologic complications after lung transplantation in pa-
204. Robb L, Drinkwater CC, Metcalf D, et al. Hematopoi- tients with telomerase complex mutations. J Heart Lung
etic and lung abnormalities in mice with a null mu- Transplant. 2014.
tation of the common beta subunit of the receptors 221. Tokman S, Singer JP, Devine MS, et al. Clinical out-
for granulocyte-macrophage colony-stimulating fac- comes of lung transplantation in patients with telom-
tor and interleukins 3 and 5. Proc Natl Acad Sci USA. erase mutations. J Heart Lung Transplant. 2015;34(10):
1995;92(21):9565–9569. 1318–1324.
CHAPTER 1 Genetic Interstitial Lung Disease 23

222. Cogan JD, Kropski JA, Zhao M, et al. Rare variants 237. Mushiroda T, Wattanapokayakit S, Takahashi A, et al. A ge-
in RTEL1 are associated with familial interstitial nome-wide association study identifies an association of a
pneumonia. Am J Respir Crit Care Med. 2015;191(6): common variant in TERT with susceptibility to idiopathic
646–655. pulmonary fibrosis. J Med Genet. 2008;45(10):654–656.
223. Stuart BD, Choi J, Zaidi S, et al. Exome sequencing 238. Calado RT, Young NS. Telomere diseases. N Engl J Med.
links mutations in PARN and RTEL1 with familial pul- 2009;361(24):2353–2365.
monary fibrosis and telomere shortening. Nat Genet. 239. Levy D, Neuhausen SL, Hunt SC, et al. Genome-wide
2015;47(5):512–517. association identifies OBFC1 as a locus involved in hu-
224. Kannengiesser C, Borie R, Menard C, et al. Heterozygous man leukocyte telomere biology. Proc Natl Acad Sci USA.
RTEL1 mutations are associated with familial pulmo- 2010;107(20):9293–9298.
nary fibrosis. Eur Respir J. 2015. 240. Codd V, Nelson CP, Albrecht E, et al. Identification of
225. Kropski JA, Mitchell DB, Markin C, et al. A novel dysk- seven loci affecting mean telomere length and their as-
erin (DKC1) mutation is associated with familial inter- sociation with disease. Nat Genet. 2013;45(4):422–427.
stitial pneumonia. Chest. 2014;146(1):e1–e7. 427e421–e422.
226. Alder JK, Stanley SE, Wagner CL, Hamilton M, Hanu- 241. Castellana G, Lamorgese V. Pulmonary alveolar micro-
manthu VS, Armanios M. Exome sequencing identi- lithiasis. World cases and review of the literature. Respira-
fies mutant TINF2 in a family with pulmonary fibrosis. tion. 2003;70(5):549–555.
Chest. 2015;147(5):1361–1368. 242. Sosman MC, Dodd GD, Jones WD, Pillmore GU. The fa-
226a. Stanley SE, Gable DL, Wagner CL, et al. Loss-of-function milial occurrence of pulmonary alveolar microlithiasis. Am
mutations in the RNA biogenesis factor NAF1 predis- J Roentgenol Radium Ther Nucl Med. 1957;77(6):947–1012.
pose to pulmonary fibrosis–emphysema. Sci Transl Med. 243. Ucan ES, Keyf AI, Aydilek R, et al. Pulmonary alveo-
2016;8(351):351ra107. lar microlithiasis: review of Turkish reports. Thorax.
227. Wang Y, Kuan PJ, Xing C, et al. Genetic defects in 1993;48(2):171–173.
surfactant protein A2 are associated with pulmo- 244. Melamed JW, Sostman HD, Ravin CE. Interstitial thick-
nary fibrosis and lung cancer. Am J Hum Genet. ening in pulmonary alveolar microlithiasis: an underap-
2009;84(1):52–59. preciated finding. J Thorac Imaging. 1994;9(2):126–128.
228. van Moorsel CH, Ten Klooster L, van Oosterhout MF, 245. Helbich TH, Wojnarovsky C, Wunderbaldinger P,
et al. SFTPA2 mutations in familial and sporadic idio- Heinz-Peer G, Eichler I, Herold CJ. Pulmonary al-
pathic interstitial pneumonia. Am J Respir Crit Care Med. veolar microlithiasis in children: radiographic and
2015;192(10):1249–1252. high-resolution CT findings. AJR Am J Roentgenol.
229. Nathan N, Giraud V, Picard C, et al. Germline SFTPA1 1997;168(1):63–65.
mutation in familial idiopathic interstitial pneumonia 246. Corut A, Senyigit A, Ugur SA, et al. Mutations in
and lung cancer. Hum Mol Genet. 2016. SLC34A2 cause pulmonary alveolar microlithiasis and
230. Maitra M, Wang Y, Gerard RD, Mendelson CR, Garcia CK. are possibly associated with testicular microlithiasis. Am
Surfactant protein A2 mutations associated with pulmo- J Hum Genet. 2006;79(4):650–656.
nary fibrosis lead to protein instability and endoplasmic 247. Huqun, Izumi S, Miyazawa H, et al. Mutations in
reticulum stress. J Biol Chem. 2010;285(29):22103–22113. the SLC34A2 gene are associated with pulmonary
231. Seibold MA, Wise AL, Speer MC, et al. A common alveolar microlithiasis. Am J Respir Crit Care Med.
MUC5B promoter polymorphism and pulmonary fibro- 2007;175(3):263–268.
sis. N Engl J Med. 2011;364(16):1503–1512. 248. Wang H, Yin X, Wu D. Novel human pathological muta-
232. Fingerlin TE, Murphy E, Zhang W, et al. Genome-wide tions. SLC34A2. Pulmonary alveolar microlithiasis. Hum
association study identifies multiple susceptibility loci Genet. 2010;127(4):471.
for pulmonary fibrosis. Nat Genet. 2013;45(6):613–620. 249. Dogan OT, Ozsahin SL, Gul E, et al. A frame-shift muta-
233. Hunninghake GM, Hatabu H, Okajima Y, et al. MUC5B tion in the SLC34A2 gene in three patients with pulmo-
promoter polymorphism and interstitial lung abnormal- nary alveolar microlithiasis in an inbred family. Intern
ities. N Engl J Med. 2013;368(23):2192–2200. Med. 2010;49(1):45–49.
234. Schroeder BW, Verhaeghe C, Park SW, et al. AGR2 is 250. Ishihara Y, Hagiwara K, Zen K, Huqun, Hosokawa Y,
induced in asthma and promotes allergen-induced Natsuhara A. A case of pulmonary alveolar microlithiasis
mucin overproduction. Am J Respir Cell Mol Biol. with an intragenetic deletion in SLC34A2 detected by a
2012;47(2):178–185. genome-wide SNP study. Thorax. 2009;64(4):365–367.
235. Zhao F, Edwards R, Dizon D, et al. Disruption of 251. Hashimoto M, Wang DY, Kamo T, et al. Isolation and
Paneth and goblet cell homeostasis and increased en- localization of type IIb Na/Pi cotransporter in the devel-
doplasmic reticulum stress in Agr2-/- mice. Dev Biol. oping rat lung. Am J Pathol. 2000;157(1):21–27.
2010;338(2):270–279. 252. Feild JA, Zhang L, Brun KA, Brooks DP, Edwards RM.
236. Tsuru A, Fujimoto N, Takahashi S, et al. Negative Cloning and functional characterization of a sodium-
feedback by IRE1beta optimizes mucin production dependent phosphate transporter expressed in human
in goblet cells. Proc Natl Acad Sci USA. 2013;110(8): lung and small intestine. Biochem Biophys Res Commun.
2864–2869. 1999;258(3):578–582.
24 Interstitial Lung Disease

253. Traebert M, Hattenhauer O, Murer H, Kaissling B, Biber J. 256. Tanjore H, Blackwell TS, Lawson WE. Emerging evidence
Expression of type II Na-P(i) cotransporter in alveolar for endoplasmic reticulum stress in the pathogenesis of
type II cells. Am J Physiol. 1999;277(5 Pt 1):L868–L873. idiopathic pulmonary fibrosis. Am J Physiol Lung Cell Mol
254. Saito A, Nikolaidis NM, Amlal H, et al. Modeling pul- Physiol. 2012;302(8):L721–L729.
monary alveolar microlithiasis by epithelial deletion of 257. Stuart BD, Lee JS, Kozlitina J, et al. Effect of telomere
the Npt2b sodium phosphate cotransporter reveals pu- length on survival in patients with idiopathic pulmonary
tative biomarkers and strategies for treatment. Sci Transl fibrosis: an observational cohort study with independ-
Med. 2015;7(313):313ra181. ent validation. Lancet Respir Med. 2014;2(7):557–565.
255. Mulugeta S, Nureki S, Beers MF. Lost after transla- 258. Dai J, Cai H, Li H, et al. Association between telomere
tion: insights from pulmonary surfactant for under- length and survival in patients with idiopathic pulmo-
standing the role of alveolar epithelial dysfunction nary fibrosis. Respirology. 2015;20(6):947–952.
and cellular quality control in fibrotic lung disease.
Am J Physiol Lung Cell Mol Physiol. 2015;309(6):
L507–L525.
Another random document with
no related content on Scribd:
den volgenden morgen vlogen er een menigte papegaaien over zijn hut.
Zijn kinderen maakten hem er opmerkzaam op en hij had nog juist tijd,
de vogels toe te roepen, een zaad van zekeren boom te laten vallen,
van wiens bast de Indiaansche medicijnmeesters bij hunne genezingen
gebruik maken. De vogels voldeden onmiddellijk aan het verzoek en
hoewel de jongens het zaad hadden zien vallen, konden zij het nergens
vinden.

Hariwali had er echter dadelijk zijn voet op gezet. Daar kinderen er niets
meê te maken hadden, wat hij ging doen, zei hij, dat er geen zaad
gevallen was en dat zij zich zoo gauw mogelijk moesten verwijderen.
Het jonge volk mag immers niet zien wat de oude piaiman voor kunsten
uitvoert!

Zoodra Hariwali alleen was, legde hij het zaad in den grond, op
dezelfde plek, waar het was neêrgevallen, en nadat hij den zelfden
avond zijn tooverkunsten met den rammelaar herhaald had, stond den
volgenden morgen op de plek een statige boom. Zoodra Hariwali nu de
vrucht van zijn kunde had gezien, riep hij zijn moeder, wie hij gelastte,
alles wat zij bij elkander had gepakt, aan de takken van den boom te
hangen, en te wachten, tot hij met zijn broeder zou terugkeeren.

Zoodra hij nu op de woonplaats van Jawahoe was aangekomen, waar


hij de geheele familie afwezig vond, nam [102]hij zijn gevangen broeder
uit de hangmat, trok den bundel beenderen van het bladerdak der hut
en maakte, dat hij met zijn last wegkwam. Maar ongelukkig kwam de
Geest eerder terug, dan hij gedacht had, en deze, ziende dat de
hangmat leeg was en de beenderen verdwenen waren, begreep
dadelijk, wat er gebeurd was. Hij ontdekte spoedig het versche spoor en
zond zijn honden vooruit, om de vluchtelingen op te sporen. Arme
Hariwali, arme broêr! Deze hoorden het blaffen van de honden en het
fluiten van den Geest, maar zij hadden nog juist den tijd, om in het hol
van een armadil* te kruipen. Pas waren zij er in, of Jawahoe kwam
aanrennen, die hen toeriep, onmiddellijk te voorschijn te komen, want
anders zou hij hen met een stok doorboren. De vluchtelingen lagen
echter diep en laag en antwoordden niet. Jawahoe, daarop woedend
geworden, bracht een stok met een harden stoot in het hol, maar
Hariwali greep hem vast en veranderde het harde voorwerp onmiddellijk
in een kapassi-slang*.

Jawahoe nu, denkende, toen hij de slang zag, dat hij zich vergist had en
het spoor der vluchtelingen kwijt was geraakt, verwijderde zich nu en
ging elders zoeken. Toen Hariwali zich overtuigd had, dat het gevaar
voorbij was, plaatste hij de beenderen weêr in het lichaam van zijn
broeder en toog met hem op weg naar zijn dorp.

Hoe verheugd was zijn moeder, haar beide zoons weêr te zien! Zij had
inmiddels alles aan de takken van den zwaren boom opgehangen, en
had zich met haar dochter en kleinkinderen voor een lange reis gereed
gemaakt. Toen het avond begon te worden, klommen zij allen, groot en
klein in de onderste takken, waarop zij beschutting vonden onder het
dichte gebladerte, terwijl Hariwali nog hooger klom en hier met zijn
tooverrammelaar begon te werken.

Midden in den nacht voelde de familie beneden plotseling, [103]dat de


boom begon te schudden; zij hoorde rommelende geluiden, gevolgd
door trillingen, en kregen de gewaarwording, dat de boom zijn wortels
uit den zandachtigen bodem begon los te maken, en op het punt was,
er vandoor te gaan. 44 Juist waren ze voornemens de lange reis te
ondernemen, toen Hariwali’s zuster, haar oogen naar beneden richtend,
zich herinnerde, dat zij haar kwejoe* in de hut had achtergelaten. Zij
riep haar broeder boven in den boom toe: Dekeweyo-daiba (ik heb mijn
voorschoot vergeten) en toen zij ten antwoord kreeg, dat zij het moest
gaan halen, veranderde zij op het oogenblik, dat zij haar hut had
bereikt, in een wicissi-eend*. Nog altijd kan men deze eend „dekeweyo-
daiba” hooren roepen, hoewel niet zoo duidelijk, als wanneer de
woorden langzaam zouden worden uitgesproken.

Wat de rest van de familie betreft—wel, wij weten, dat de boom ergens
heen vloog, maar nooit hebben wij iets van de menschen vernomen, die
er op waren, toen hij van de plaats, waar hij zoo gauw gegroeid was,
vertrok.

No. 17. De legende van den Ouden man’s val.

In lang vervlogen tijden, zoo vertellen onze voorouders, bevonden zich


boven den Potaro-val* meerdere Indianen-dorpen. In een der hutten
woonde een oude man, die half blind en een ieder tot last was. Want,
hoewel hij vroeger sterk en vlug was geweest, werden zijn arme
lijdende voeten door een menigte zandvlooien* geplaagd, die er zich
venijnig hadden ingegraven. De oude, hulpelooze man had steeds meer
verzorging noodig—en dit verdroot den jongeren, die er ten slotte de
brui van gaven en uitriepen: „Wat helpt dat? Laten we hem laten liggen
en laat hem maar schreeuwen! Wie er lust in heeft, ga zijn gang. Hij is
een lastige, afzichtelijke oude man [104]geworden. Wat is er aan te
doen? Allen wenschen zijn dood”. Niemand wilde echter het hoofd van
den armen man klieven. „Laat hij gaan naar het land der Geesten”,
riepen sommigen, „de rivier zal hem er wel brengen; als zijn lijden nog
langer voortduurt, zal hij toch sterven. Wij kunnen hem niet meer
helpen; wij zondigen dus niet”.

Toen zei de kapitein: „Breng een korjaal, 45 leg er den oude in, maar
verzuim vooral niet, al hetgeen hij bezit er ook in te leggen; laat hier
niets van hem achterblijven, want wij wenschen geen voordeel, doch
willen alleen aan zijn lijden een einde maken. Wij zenden hem naar een
andere wereld, en laten hem niet met ledige handen gaan; want anders
zouden we zondigen”.

De jongeren gehoorzaamden. Zij brachten den gevraagden korjaal,


legden den ouden man met zijn bezittingen er in, duwden het vaartuigje
vooruit en zagen hem langzaam stroomafwaarts drijven. Het geraas van
den naderenden waterval moet voor hem een benauwende droom
geweest zijn, maar zijn angst duurde niet lang, want—slechts eenige
oogenblikken daarna stortte het bootje met het slachtoffer en al zijn
zandvlooien naar omlaag.
… slechts eenige oogenblikken daarna stortte het bootje met het
slachtoffer en al zijn zandvlooien omlaag.—Zie blz. 104.

Het kan zijn, dat het jonge volk, dat zoo wreed was geweest, spijt heeft
gehad, niet anders gehandeld te hebben; het is niet bekend, want het is
al heel, heel lang geleden. Maar wel weten we, dat de klokkenvogel*,
met zijn wit gevederte, in de buurt nog altijd voortgaat, des nachts zijn
klokkentonen te laten hooren.

Sommige Indianen weten echter te vertellen, dat toen de man de


afschuwelijke tuimeling over den val maakte, onzichtbare krachten
tusschenbeide gekomen zijn, die, om zijn arm gebeente te redden,
boot, met alles wat er in was, in steenen veranderd hebben. Ieder, die
de rotsen [105]daar nu ziet, zal de boot in de gedaante van een puntigen
rots duidelijk herkennen en, niet ver van daar, de in steen veranderde
pagaai. Het is echter niet zoo duidelijk, waar de man zelf gebleven is.
„Want”, beweren de Indianen, die beneden den val wonen, „daar een
sterke stroom de hardste rotsen afslijpt, zullen eerst zijn ooren en zijn
neus, vervolgens zijn vingers en zijn teenen afgeslepen zijn, zoodat zijn
lichaam misschien in het zand beneden den val is overgegaan”.

No. 18. Amanna en haar praatzieke man. (C.)

Verhit door den drank van een groot paiwarri-feest, nam een Indiaan
zijn weg naar het water, om zich te baden en wat op te frisschen. Toen
hij er aankwam, ontmoette hij Amanna, een van de geesten der
Okoyoemo’s, een aantrekkelijke jonge vrouw, die hem vroeg, haar in
het water te vergezellen. Eerst had hij er wel wat op tegen, maar toen
zij bleef aanhouden, kon hij ten slotte niet langer aan haar
bekoorlijkheden weêrstand bieden. Toen hij op het laatste oogenblik
toch niet nalaten kon, haar toe te voegen, dat hij er haast zeker van
was, dat men hem zou willen verdrinken, voegde zij hem toe: „Wees
niet bevreesd, ik zal goed op je passen en zorgen, dat er geen kwaad
met je gebeuren kan”. Zij doken nu samen onder, en toen zij op den
bodem van het water aankwamen, zag hij een aantal hutten, vol met
volk, waaronder vele jonge vrouwen. Hij voelde zich nu erg tevreden,
vooral toen deze hem drank aanboden. Amanna verbood hem er echter
van te gebruiken en nam hem meê naar haar ouden vader, die hem
hartelijk verwelkomde en zijn dochter op het hart drukte, goed op hem
te letten.

Dit deed zij ook.

In dien tusschentijd was de moeder over het lange wegblijven van haar
zoon ongerust geworden; zij volgde [106]het spoor, dat helaas naar de
rivier leidde. „Mijn arme zoon zal verdronken zijn”, dacht zij, en zij
begon naar zijn lijk te zoeken. Maar overal te vergeefs. Zij keerde
bedroefd naar haar dorp terug, in de vaste meening, dat haar zoon
dood was.

Geruimen tijd was er voorbijgegaan, toen de man het verlangen kreeg,


om zijn moeder een bezoek te brengen. Hij rees uit het water omhoog
en sloeg den weg in naar zijn oude woonplaats. De moeder, verheugd
haar zoon weêr te zien, vroeg hem, waar hij al dien tijd was geweest; en
hij vertelde, dat hij een verre wandeling had gemaakt. De oude vrouw
was echter met dit antwoord niet tevreden, en zei tot haar zoon: „Je
moet niet meer van me weggaan, want ik ben oud en lijd honger; je
jongere broêr kan mij niet ondersteunen en ik heb geen andere
kinderen dan jelui beiden”.

Maar haar zoon, die een wilden aard had, luisterde niet en vertrok nog
dienzelfden namiddag naar zijn vrouw, de Watergeest, terug. Ditmaal
bleef hij nog veel langer weg dan de eerste maal. Toen hij nu na zijn
tweede afwezigheid weêr eens naar zijn moeder kwam zien, vond hij
haar met zijn kleinen broêr weêr aan het drinken van paiwarri. De
jongen begon hem uit te vragen, waarom hij toch bij het Okoyoemo-volk
verblijf hield. Dit verontrustte hem, en onder het uitroepen van: „Hoe
durf je me zoo’n vraag te doen?” holde hij weg en verdween hij weêr
onder water, waar hij nog langer bleef dan den tweeden keer.

Er verliep nu weêr een lange tijd, gedurende welke Amanna hem drie
kinderen had geschonken, en toen de man weêr zijn wensch te kennen
gaf, zijn moeder te willen bezoeken, voegde hij er bij, dat hij graag zou
zien, dat Amanna hem vergezelde. Zij vond het goed, op voorwaarde,
dat de kinderen thuis zouden blijven. [107]

Toen het paar in de ouderlijke hut aankwam, was de oude moeder met
haar zoon weêr bezig met het drinken van paiwarri, maar ditmaal was
de broêr bepaald dronken, en weêr kon deze niet laten de vraag te
doen, of hij onder het Okoyoema-volk bleef wonen? Ja, dat doe ik, zei
de man geprikkeld, en hier heb je mijn vrouw Amanna, die tot die natie
behoort!

Zoodra Amanna dit hoorde, holde zij naar de waterkant, en plonste zij
onder water. Haar man vluchtte haar na, maar niet zoodra was hij ook
onder water gekomen, of zijn vrienden en verwanten vermoordden hem,
omdat hij aan zijn moeder haar naam had genoemd, en verteld had,
waar zij van daan kwam.

No. 19. De zon en zijn beide tweelingzoons. (C.)

Zeer lang geleden leefde er een vrouw, aan wie de Zon* een tweeling
schonk. Het waren jongens, Pia en Makoenaima* genoemd. Eens op
een dag zei de toen nog niet geboren Pia tot zijn moeder: „Laten wij
gaan en onzen vader gaan opzoeken. Wij zullen U den weg wijzen, en
als wij op pad zijn, moet gij voor ons mooie bloemen plukken.” De
moeder liep naar het Westen 46 om haar man te ontmoeten, en terwijl zij
aan het bloemen plukken was, struikelde zij, waardoor zij zich
verwondde en viel. Zij betichtte nu van dit ongeluk hare nog ongeboren
kinderen; en nadat zij hen beknord had en hun vroeg, welken weg zij
verder moest volgen, weigerden zij het te zeggen.

Zoo kwam het, dat zij een verkeerden weg insloeg en eindelijk met den
gewonden voet en doodmoe aan een [108]vreemdsoortige hut kwam. De
hut was het eigendom van Tijger’s moeder, Kono(bo)-aroe, de
Regenkikvorsch*, en toen de uitgeputte wandelaarster ontdekte waar zij
was, zei ze tot de oude vrouw, dat zij groote spijt had, hier gekomen te
zijn, daar zij gehoord had, hoe wreed haar zoon was. De huisvrouw, die
medelijden met haar had, beduidde haar, dat zij niet bang behoefde te
zijn, maar verborg haar voor alle veiligheid in den kassiripot en smeet
haastig het deksel er op.

Toen Tijger des nachts thuis kwam, begon hij overal rond te snuiven.
„Moeder”, zei hij, „ik ruik iemand, wie heb je hier?” En ofschoon zij
volhield, dat zij niemand voor hem had, om zijn maal te doen, was Tijger
niet tevreden en ging zelf op onderzoek uit. Eindelijk ontdekte hij het
angstige schepsel in den kassiri-pot.

Toen hij nu de arme vrouw met één slag gedood had, ontdekte hij de
nog niet geboren kinderen. Hij toonde ze aan zijn moeder, die hem
aanried, goed op ze te passen en ze zorgvuldig te verplegen. Tijger
deed wat hem gezegd was: hij wikkelde ze in een bundel katoen, om ze
warm te houden en ziet, den volgenden morgen waren ze al zóó groot
geworden, dat ze begonnen te kruipen. Nog een dag later waren ze nog
meer gegroeid, en dat ging zoo voort, tot dat ze reeds na verloop van
een maand volwassen waren.

Tijger’s moeder zei, dat zij nu wel pijl en boog zouden kunnen
hanteeren, waarmede zij den powies* moesten schieten, omdat het
deze vogel was geweest, die hun moeder gedood had.

Pia and Makoenaima gingen nu de volgenden dag op jacht en schoten


een powies. Dat ging lang goed; iederen dag brachten zij powies mede.
Maar toen zij weêr eens op een powies mikten, riep den vogel hen toe,
dat het niet een powies was geweest, die hun moeder gedood [109]had,
maar Tijger zelf, terwijl hij er de noodige inlichtingen bij gaf, hoe zij
Tijger moesten dooden.

De beide jongens schrokken hevig, toen zij dit hoorden. Zij spaarden
den vogel; maar toen zij met leêge handen van de jacht terugkwamen,
maakten zij hun moeder wijs, dat de powies hun pijlen en boog had
weggenomen. Deze leugen hadden ze maar verzonnen, om de kans te
hebben, nieuwe en betere wapens te maken. Zij hadden hun wapens
ergens in het bosch verborgen.

Nadat zij nu met hunne krachtiger wapens gereed waren, bouwden zij
een stellage tegen een boom, klommen er in en schoten, toen Tijger
voorbij kwam, op hem, zoodat hij morsdood neêrviel. Haastig daalden
zij naar beneden, renden naar de hut en schoten nu ook zijn moeder
neêr.

De beide jongens vervolgden nu hun weg en kwamen ten laatste aan


een groep Kankantries, bij een hut, waarvoor een stokoude vrouw zat,
die in werkelijkheid een kikvorsch* was. Zij namen bij haar hun intrek en
gingen iederen dag op jacht. Iederen keer, wanneer zij van de jacht
terugkeerden, vonden zij cassavebrood gereed staan, dat hun
gastvrouw gebakken had. „Dat is erg vreemd”, zei Pia tot zijn
tweelingbroeder, „wij zien hier geen kostgronden, en kijk nu eens,
hoeveel cassave de oude ons wel voorzet. Wij moeten eens goed op
haar letten.”
… bij een hut, waarvoor een stokoude vrouw zat, die in
werkelijkheid een kikvorsch was.—Zie blz. 109.

Den volgenden morgen bleven zij, in plaats van hun jachtterrein op te


zoeken, in de buurt en verscholen zij zich achter een boom, van waar zij
alles konden zien, wat er in en om de hut gebeurde. Zij zagen toen, dat
de oude een witte vlek op haar schouders had, dat zij telkens zich
voorover boog en in deze vlek beet, waaruit onmiddellijk cassavemeel
begon neêr te vallen.

Bij hun thuiskomst weigerden de jongens nu van de cassave te eten, nu


zij wisten, waar zij vandaan was gekomen. [110]

Den volgenden dag plukten zij nu een hoeveelheid katoen 47 van de


boomen en spreidden dit op den bodem uit. „Wat doen jelui daar?”
vroeg de oude. „Wel, we maken iets heel moois voor je. Die zachte
zitplaats zal je bevallen”, antwoordden de jongens. De oude was er erg
blij meê; maar niet zoodra was zij op het zachte bed gaan zitten, of de
jongens staken het in brand. De huid van de oude werd nu zóó
erbarmelijk geschroeid, dat haar huid vol met rimpels trok. Dit leelijke,
ruwe vel heeft de Regenkikvorsch altijd behouden.

Pia en Makoenaima zetten nu hun reis verder voort, om hun vader te


ontmoeten en kwamen al heel gauw aan de woning van Maipoeri*, den
Tapir, waar zij drie dagen bleven. Op den derden avond keerde
Maipoeri glanzend en vet van zijn dagelijksche wandeling terug. De
jongens, die wilden weten, hoe hij het wel aanlegde, er zoo doorvoed uit
te zien, volgden den anderen morgen vroeg zijn spoor, dat leidde naar
een pruim*. Hier aangekomen, begonnen ze zóó hard aan den boom te
schudden, dat alle vruchten, rijp en onrijp, op den grond vielen.

Toen Maipoeri zich den volgenden morgen weêr dacht te goed te


kunnen doen, maar bemerkte, dat al zijn voedsel voor hem verloren
was, keerde hij haastig naar huis terug, ranselde de beide jongens af en
verdween haastig in het bosch. Pia en Makoenaima liepen hem
achterna, volgden den geheelen dag zijn voetspoor en haalden hem
eindelijk in. Pia beduidde zijn broêr, achter Maipoeri om te gaan en het
dier naar hem toe te drijven, en toen hij het dier zag naderen, schoot hij
het een haapoena (harpoenpijl) door het lijf, maar de lijn kwam door het
nog een eind voortloopende dier langs den armen Makoenaima en
sneed hem een been af. [111]

Op een helderen nacht kan men ze nu nog zien: je ziet Maipoeri


(Hyaden), Makoenaima (de Pleiaden) en daaronder zijn afgesneden
been (de gordel van Orion).

No. 20. De Legende van den Vleermuis-berg. (M.)

Langen tijd geleden, leefde er in het gebergte 48 een reusachtige


vleermuis, die vrees en schrik verspreidde onder de Makoesi-Indianen*.
Niet zoodra was de zon in het Westen nedergedaald, of het groote
afschuwelijke dier verliet zijn onbekend verblijf, fladderde over hunne
vóór dien tijd zoo ongelukkige woonplaatsen en schoot pijlsnel naar
beneden, om den eerste den beste, dien het buiten de hut aantrof, van
den grond te lichten. Het wezen droeg daarop het slachtoffer in zijn
vervaarlijke klauwen naar zijn onbekend nest en verslond het daar.

Angstig bracht men den avond in de Indianen-kampen door, en de


morgen brak dikwijls met geweeklaag aan, wanneer twee, soms zelfs
drie dorpsgenooten gemist werden; geen nacht ging er voorbij zonder
een ontvoering en de stam verminderde dagelijks, zoodat zijn geheele
vernietiging nabij scheen.

De piaiman was steeds ijverig in de weêr, om den Geest uit te drijven,


maar telkens kwam het gevreesde geheimzinnige dier terug; mannen
trokken er op uit, om het verblijf van den vervloekten moordenaar op te
sporen, maar zij konden het nergens vinden—Makoenaima was
blijkbaar niet met hen.
Om den stam voor uitsterven te behoeden, verrees er nu een oude
vrouw uit haar hangmat en verklaarde aan het volk, dat zij zich wilde
opofferen ter wille van hare mede-stamgenooten. Nadat de zon was
ondergegaan, plaatste zij zich met een overdekten vuurstok 49 in het
[112]midden van het dorp, terwijl haar dorpsgenooten, gebukt en angstig
in hun hutten zaten af te wachten, wat gebeuren zou. Men hoorde het
fladderen van de reusachtige vleugels, en begreep, dat de heldin in zijn
vreeselijke klauwen was vastgegrepen, en meêgenomen werd naar het
geheimzinnige knekelhuis.

Nauwelijks was het slachtoffer omhoog geheven, of het ontdeed den


vuurstok van zijn omkleedsel, en deze wierp nu, evenals de Zon, zijn
vurige stralen achterwaarts 50 zoodat het volk beneden kon zien, welken
weg het zou moeten volgen, om het kerkhof der verloren broeders te
vinden.

De hooge vlammen, die den volgenden morgen uit het brandende nest
opstegen, wezen nu de verblijfplaats van het monster aan. Het volk
wachtte geen oogenblik, om er op uit te trekken, en het gelukte den
moordenaar te dooden. De geschiedenis vertelt niet, of de oude vrouw
voor haar heldhaftige daad heeft moeten boeten; maar wel is het
bekend, dat een stapel gebleekte beenderen de plaats aanwijst, waar
eenmaal het nest van den roover zich bevond.

No. 21. De Uil en zijn schoonbroeders vleermuis. (W.)

Bokoe-Bokoe, de nachtuil, was getrouwd met de zuster van de


vleermuis en nam des nachts haar broêrs meermalen mede, om in de
Indianen-hutten op roof uit te gaan. Eens op een nacht kwamen zij
langs een hut, waarin het volk juist bezig was, op den barbakot visch te
roosteren. Om de menschen schrik aan te jagen, zongen zij luidkeels:
bokoe! bokoe! bokoe! waardoor zij de bewoners op de vlucht joegen,
zoodat zij gelegenheid kregen, de visch te stelen. Het drietal haalde in
meerdere hutten denzelfden streek uit, totdat de uil op zekeren dag aan
zijn kornuiten [113]meêdeelde, dat hij voor eenigen tijd op reis moest en
dat zij zich tijdens zijne afwezigheid maar wat moesten behelpen en
goed deden, ’s nachts binnenshuis (d.i. in het bosch) te blijven, daar er
anders zeker iets minder prettigs met hen zou gebeuren. Maar niet
zoodra had Bokoe-bokoe zijn hielen gelicht, of zwager Vleermuis nam
nu in zijn plaats zijn broêr op den rooftocht meê.

Eens op een nacht kwamen de beide vleermuizen weêr aan een hut,
waar visch geroosterd werd, en daar zij nu den uil niet bij zich hadden
en dus niet zoo duidelijk bokoe! bokoe! bokoe! konden roepen als te
voren, werd het volk ook niet zoo bang en liep het niet zoo ver weg,
zoodat het in staat was te zien, dat de vleermuizen het waren, die hun
die poets bakten. Maar de vleermuizen, die zich niet lieten afschrikken
en dachten, dat zij ongestraft hun slag zouden kunnen slaan, kwamen
den volgenden avond terug, toen een deel van de menschen reeds in
hun hangmatten lag, anderen nog rustig bijeenzaten. De snoodaards
dachten zeker, dat er niets met hen gebeuren kon en lachten van
vreugde Chi! Chi! Chi! Maar de hutmeester nam zijn boog en schoot
een pijl, voorzien van een prop was 51, een der vleermuizen tegen het
lijf, waardoor deze bedwelmd neêr viel. Zijn broêr, die onmiddellijk naar
het bosch gevlucht was, ontmoette daar Bokoe-bokoe, die juist van zijn
reis teruggekeerd was en aan wien hij den vroegen dood van zijn broêr
vertelde. Maar het verlies schrikte hen niet af, zoodat zij er den
volgenden nacht met hun tweeën op uittrokken. Het geluid hunner
stemmen bracht, nu Uil er bij was, weêr zulk een opschudding onder
het Indianenvolk teweeg, dat allen weêr de vlucht naar het bosch
namen en Bokoe-bokoe met zijn schoonbroêr de visch kon wegnemen.
Toen zij nu op den barbakot ook [114]de doode vleermuis ontdekten,
namen zij het lichaam van hun verwante meê naar huis, waar zij flink op
de plek begonnen te kloppen, waar hij met den pijl geraakt was; dit
bracht het dier weêr bij, daar het vuur deze plek niet had aangedaan, en
als te voren begon de tot het leven weêr terug geroepen vleermuis te
lachen, Chi! chi! chi!
Hoewel nu Bokoe-bokoe den volgenden avond verhinderd was zijn
beide schoonbroêrs te vergezellen, herhaalden deze toch hun
nachtelijken strooptocht, en zooals van zelf sprak, was het volk weêr
minder bevreesd, zoodat een der vleermuizen een schot in zijn
achterste ontving.

Den volgenden nacht keerde nu Bokoe-bokoe met zijn schoonbroêr


terug en evenals de eerste keer vonden zij op den barbakot weêr de
gedoode vleermuis. Weêr namen zij het lichaam meê naar huis; maar
toen zij weêr op het lichaam klopten, wilde het leven niet terugkeeren;
het was te veel geroosterd boven het vuur.

De overlevende vleermuis echter gaat nu altijd voort met het volk en zijn
gevogelte uit te zuigen 52, of hen op andere wijze te hinderen, terwijl de
verschijning van Bokoe-bokoe voortaan altijd ongeluk beteekent.
Wanneer het bokoe! bokoe! bokoe! des nachts wordt gehoord,
beteekent dit steeds ziekte of dood.

No. 22. De Lichtkever en de verdwaalde Jager. (C.)

Er gingen eens vijf mannen gezamenlijk op jacht. Zij hadden zich echter
te diep het bosch in begeven, om nog vóór den nacht huiswaarts te
kunnen keeren, zoodat zij, toen de duisternis hen overviel, een banab*
bouwden. Den volgenden morgen trokken zij in verschillende richtingen
het bosch in. Laat in den avond waren allen in de tijdelijke verblijfplaats
teruggekeerd, behalve één. Drie van hen [115]beweerden, dat hij zeker
door een tijger verslonden was, maar toen zij de zaak nader gingen
overleggen, herinnerden zij zich, dat zij geen enkel tijgerspoor hadden
gezien. De hoofdman was dichter bij de waarheid, toen hij zei: „Neen!
Hij moet verdwaald zijn”. 53 Dit was ook werkelijk, wat er met den vijfden
man gebeurd was. Hij was al dieper en dieper het bosch ingedrongen,
en was door de duisternis overvallen, waardoor hij zijn weg niet terug
had kunnen vinden. Hij was toen in alle richtingen gaan zoeken en had
zich eindelijk onder een boom te slapen gelegd.

Nu en dan vloog er een Poe-yoe, een lichtkever* om zijn rustplaats


heen, die hem vroeg, wat hij daar zoo alleen uitvoerde. „Ik ben den weg
kwijt geraakt”, antwoordde hij, waarop het insekt aanbood, hem den
weg te wijzen. Maar de verdwaalde kon niet gelooven, dat zoo’n klein
ding in staat zou zijn, hem te helpen. Eerst toen de lichtkever hem
vertelde, dat hij voornemens was, zich te gaan warmen aan het echte
vuur, dat zijn vrienden bij de banab hadden ontstoken, stemde hij toe en
volgde hij het insekt.

Toen de beide vrienden het kampement in het oerwoud naderden,


hoorden zij in de verte reeds menschelijke stemmen. „Luister”, zei de
kleine kever, „hier zijn je gezellen. We zullen er heengaan”. Toen zij
eindelijk de banab hadden bereikt, vloog de lichtkever vooruit en
vertelde aan de jagers, dat hij hun verloren metgezel kwam brengen.
Onmiddellijk daarna trad deze binnen, en de vier vrienden waren
verheugd hem weêr te zien. [116]

No. 23. De bina 54, de weder in het leven geroepen vader en de


slechte vrouw. (W.)

In een der Warrau-dorpen leefde een man met zijn vrouw en zijn beide
kinderen. Zijn schoonbroêr woonde bij hem in. Eens op een morgen
begaven man en vrouw zich naar den kostgrond en droegen den
broeder op, om te gaan visschen, zoodat zij bij hun thuiskomst wat te
eten zouden hebben. Toen zij echter terugkwamen van hun werk, zagen
zij, dat de broêr geluierd had en dat hij niet eens buiten de hut was
geweest; ja, dat hij zelfs het weinige, dat er nog van den vorigen dag
was overgebleven, had opgegeten. Dit maakte hem boos en hij zei tot
zijn schoonbroêr: „Ik moet naar het land gaan, om het terrein in orde te
brengen. Ik moet in het bosch gaan, om mij wild te verschaffen, en in
mijn bootje de rivier opvaren, om visschen te vangen. Ik moet al het
werk doen, terwijl jij niets uitvoert en alle dagen maar ligt te luieren in je
hangmat. Al ben ik vermoeid, toch moet ik weêr uitgaan, om visch te
gaan halen.”

Terwijl hij dit op een alles behalve vriendelijken toon zei, nam hij zijn
harpoen-lans* en toog op weg naar de kreek. De schoonbroêr nam op
zijn beurt zijn kapmes, en na het gescherpt te hebben, volgde hij den
man in zijn korjaal. Beide mannen ontmoetten elkander juist op het
oogenblik, toen de echtgenoot met zijn boot terug wilde keeren, nu hij
een grooten visch had weten te bemachtigen. „Hallo! nu al terug!” zei de
schoonbroêr. „Ja”, antwoordde de ander, „ik heb een grooten visch
geschoten, hier is hij”. „Nu, leen mij dan je lans”, zei de eerste weêr, „ik
wil zien, of ik ook zoo’n grooten visch kan schieten als jij”. Toen beide
korjalen naast elkander lagen, hief de man zijn lans omhoog en wilde
het wapen juist naar de boot van zijn schoonbroêr richten, toen hij op
hetzelfde oogenblik door het kapmes werd getroffen, waarmeê hij
[117]twee sneden ontving, die hem dood deden neêrvallen.

De moordenaar probeerde nu het lijk kwijt te raken en het in het water


te werpen. Maar zijn zuster, die haar broêr zijn kapmes had zien
scherpen, was in toorn de hut uitgeloopen, en had, bevreesd dat hij er
iets kwaads meê voor had, tegen haar kinderen gezegd: „jelui oom is
zeker boos over iets; hij scherpt zijn houwer, en is jelui vader achterna
gegaan. Laten we gaan zien, wat hij wil gaan doen”.

Met haar beide kinderen was zij haar broêr gevolgd en zij kwam juist
aan bij de rivier op het oogenblik, dat hij probeerde, het doode lichaam
overboord te werpen. „Neen, doe dat niet, broêr”, zei ze. „Nu je hem
gedood hebt, moet je zijn lijk meê naar zijn hut nemen en hem daar
begraven”. Hij deed, wat hem werd gevraagd, nam het lijk meê in zijn
korjaal en begon een boom te vellen, om hout te hebben voor zijn
laatste rustplaats. In dien tusschentijd zond de vrouw van den
vermoorde hare kinderen naar diens broêr en oude moeder, met de
boodschap, dat zij niet ongerust moesten zijn, maar eens moesten
komen. Moeder en zoon lieten niet op zich wachten en toen zij naderbij
kwamen, zagen zij, dat hij juist gereed was gekomen met het uithollen
van den stam, de lijkkist in de hut had gebracht en nu bezig was een
graf te graven. De broêr van den vermoorde was woedend, maar zijn
moeder zei: „Hinder de man niet: wij willen eerst eens zien, wat de
weduwe denkt te doen”. Deze wachtte bij het graf met een kapmes in
haar hand en zei tot den moordenaar, wat haast te maken.

Toen de delver met het graf gereed was, liet hij de kist er in zakken,
legde hij er het lijk in, dat hij netjes gekleed had en met verf en sieraden
had opgetooid, en gaf het zijn mes, zijn vischhaken en andere
voorwerpen van zijn voormalig bezit meê. Toen hij eindelijk met zijn
[118]nog met bloed bevlekte handen begonnen was, het graf met aarde
dicht te werpen, trof zijn zuster, die achter hem stond, hem met haar
kapmes in den nek en na enkele oogenblikken bloedde de delver dood.

Er werd nu een tweede graf naast het eerst gegravene gedolven, en


daarin legde de zuster hem, zooals hij in zijn naaktheid was, neder,
zonder kleeding, zonder sieraden, zonder zijn bezittingen, want zij
voelde niet het minste medelijden of spijt jegens haar broêr.

Den zelfden dag vertrokken de moeder en de broeder weêr naar hun


woonplaats, na de weduwe te hebben overgehaald, bij hen haar intrek
te nemen en de kinderen mede te nemen. Dadelijk na aankomst in het
verblijf der oude moeder nam haar zoon de zorg voor de weduwe op
zich, legde haar in zijn hangmat en zei tegen zijn vrouw, dat hij liever
deze vrouw nam, omdat deze kinderen had, en zij hem geen kinderen
kon schenken.

In het nieuwe verblijf voelden de kinderen van de weduwe zich weinig


op hun gemak. Iederen morgen, nadat zij wat hadden gegeten,
begaven zij zich zoo gauw zij konden naar het graf van hun vader en
keerden zij eerst laat op den dag terug. Op den derden dag van hun
bezoek aan het graf ontmoetten zij een Heboe*, maar de kinderen
herkenden hem niet. De Heboe sprak tot hen: „Wanneer jelui je vader
weêr terug wilt hebben, moet je een blad van een zekeren boom (hij
noemde den boom) plukken en er mede over het graf heen en weêr
wrijven. Dan zal hij verschijnen”. 55 „Maar we kennen den boom niet”,
antwoordden zij, en toen de kleine man eenige bladen van den
bedoelden boom had geplukt, wees hij hun, hoe zij de bladeren over
den grond moesten wrijven, waar hun vader lag begraven, en drukte
hen op het hart, dit den volgenden morgen dadelijk te doen en dan
[119]’s middags terug te komen. Hun vader zouden zij dan zien.

Den anderen morgen volgden de kinderen op, wat de Heboe had


gezegd, en toen zij des middags terug kwamen, zat hun vader in de hut.
Hij ging naar hen toe en zei: „Haal wat water voor mij: ik heb dorst”.
Nadat hij wat gedronken had, vroeg hij: „Waar is jelui moeder?” Toen hij
hoorde, dat ze bij haar schoonmoeder woonde, droeg hij hen op, haar
te gaan halen. Toen de kinderen bij hun moeder kwamen en haar alles
hadden verteld, wat ze gezien hadden, riep zij uit: „Hoe kan dat nu?
Hoe kan jullie vader mij roepen, hij is immers dood?” Zij wilde er eerst
niets van gelooven, maar toen de jongens bleven aandringen en al
maar riepen: „Kom toch moeder, ga toch meê, alles is waar wat wij
vertelden”, volgde zij haar kinderen. Zij weigerde echter haar hangmat
meê te nemen, want zij kon nog altijd het verhaal niet gelooven.

Toen zij met de kinderen bij het graf gekomen was, kon zij haar oogen
niet gelooven. Want haar man zat zoowaar in levenden lijve in de hut.
Het was haar echte man, die daar tegenover haar zat. Het eerste wat hij
vroeg was: „Waar is je broêr?” waarop zij antwoordde: „Wat, ik doodde
hem en begroef zijn lijk naast het jouwe”. „Nu”, zei haar man, „hem zal
je nooit terug zien”.

You might also like