Abstract
The interstitial lung diseases (ILDs) are a group of progressive disorders characterized by chronic inflammation and/or fibrosis in the lung. While some ILDs can be linked to specific environmental causes (i.e. asbestosis, silicosis), in many individuals no culprit exposure can be identified; these patients are deemed to have “idiopathic interstitial pneumonia”, or IIP. Family history is now recognized as the strongest risk factor for IIP, and IIP cases that run in families comprise a syndrome termed “Familial Interstitial Pneumonia” (FIP). Mutations in more than 10 different genes have been implicated as responsible for disease in FIP families. Diverse ILD clinical phenotypes can be seen within a family, and available evidence suggests underlying genetic risk is the primary determinant of disease outcomes. Together, these FIP studies have provided unique insights into the pathobiology of ILDs, and brought focus on the unique issues that arise in the care of patients with FIP.
Keywords: IPF, Pulmonary fibrosis, Genetics, Telomere, Surfactant, Alveolar epithelial cell
Introduction
The diffuse parenchymal lung diseases (DPLDs, also commonly referred to as interstitial lung diseases, ILDs) are a group of disorders characterized by subacute or chronic inflammation and/or fibrosis in the lung.1 Patients with ILD typically present with symptoms of cough and/or dyspnea, which frequently progresses over time. There are numerous different risk factors that have been linked to different forms of ILD, including occupational and environmental exposures, systemic diseases, and more recently, genetic risk factors.2 Family history is now recognized as the strongest risk factor for ILD,3 and candidate gene-based, linkage, and next-generation sequencing-based approaches have now implicated mutations in more than 10 different genes as responsible for disease families with ILD. Studies of families with ILD (Familial Interstitial Pneumonia, FIP) have provided unique insights into the pathobiology of ILDs, and brought focus on the unique issues that arise in the care of patients with FIP.
Epidemiology
The syndrome of FIP is defined by the presence of ILD among two or more closely related individuals, including at least one family member with an idiopathic interstitial pneumonias (IIPs).4,5 The earliest reports of interstitial lung disease clustering in families dates to the early 1950’s, with a report by Peabody and colleagues described idiopathic pulmonary fibrosis in twin sisters.6 Through the following several decades, there were scattered reports of familial ILD,7–10 but it was generally believed these families were quite rare. As of the early 2000’s, it was generally believed that FIP reflected a very small subset of patients with ILD, with most estimates ranging from 2–5%.11,12 Several more recent studies employing intensive ascertainment strategies have reported that approximately 20% of ILD patients report a family history of ILD at referral centers in Mexico,3 the United States,13 and Canada.14 An important limitation of studies to date is a focus on populations of European ancestry; while case reports of FIP in populations of non-European ancestry are common, there have been no large-scale studies estimating the prevalence of FIP in a non-European population.
Clinical considerations
Among unselected FIP patients, clinical features including age at diagnosis, histopathologic patterns, and disease progression are generally similar to those seen in patients with “sporadic” forms of ILD.4 While usual interstitial pnuemonia (UIP) is the most common ILD phenotype in FIP kindreds, 50% or more of families have more than one ILD phenotype among affected family members.4 In addition, imaging patterns may not conform to typical ILD patterns; in one report comparing HRCT images from FIP and sporadic ILD patients, more than 50% of FIP HRCT images did not fit UIP or nonspecific interstitial pnuemonia (NSIP) criteria, related in part to a relative increase in apical and mid-lung involvement.15
The evaluation of patients with suspected FIP should focus on features that may suggest a specific genetic diagnosis. While the majority of FIP cases occur in middle-aged and older adults, it has become clear through time that FIP presentations can occur across the lifetime, ranging from the neonatal period to late adulthood. As described below, mutations in genes related to surfactant biology in have been linked to diverse presentations of ILD in children and adults within a family.16,17 A focused family history specifically inquiring about neonatal respiratory distress or neonatal death is advised evaluating all suspected cases of FIP. A personal or family history of unexplained chronic liver disease, bone marrow disease, or early greying suggests a potential short-telomere syndrome.18,19 The presence of abnormal bleeding and/or ocular albinism in a family should raise suspicion for Hermansky-Pudlak Syndrome.20,21 Hypothyroidism alone or in combination with movement disorders may suggest “brain-lung-thyroid syndrome” associated with mutations in the transcription factor NKX2–1.22,23 Interstitial lung disease associated with pulmonary hemorrhage and/or highly elevated autoantibodies may suggest a COPA mutation.24
Genetics of FIP
Kindreds with FIP most commonly involve multiple affected individuals across successive generations, consistent with an autosomal dominant inheritance pattern with incomplete penetrance,4,25 although in rare cases, X-linked26 and recessive27,28 disease patterns have been described. An autosomal dominant inheritance pattern indicates that a single copy of a mutant allele (a heterozygous variant) may be sufficient to confer disease risk. Consequently, most familial studies investigating genetic risk for FIP have focused on rare variants (RVs) with very low frequency in the general population (minor allele frequency <0.001 or lower). Studies to date have implicated rare genetic variants in more than 10 genes in FIP kindreds (Table 1). FIP genes identified to date comprise into two distinct groups: 1) genes related to surfactant production or processing, and 2) telomere-related genes.
Table 1.
Surfactant-related genes
In 2001, Nogee and colleagues reported the first recognized genetic cause of FIP; in this case a heterozygous mutation in the gene encoding for surfactant protein-C (SFTPC) in a mother and infant with NSIP and desquamative interstitial pneumonia (DIP), respectively.17 This was soon followed by a report of a different SFTPC mutation in a very large multiplex kindred including adults with UIP pattern.16 Subsequent studies indicated that SFTPC mutations are a rare cause of FIP29–31 outside selected populations influenced by founder effects,14,32 and very uncommon among ILD patients outside the context of FIP.31,33–35
The implication of SFTPC mutations in FIP led to a search for mutations in other genes related to surfactant biology in FIP kindreds. These studies have led to reports of mutations in the gene encoding for surfactant protein A2 (SFTPA2)36,37 and surfactant protein A1 (SFTPA1)38,39 in FIP kindreds. There are also a number of reports of FIP kindreds heterozygous for rare genetic variants in the gene encoding for ATP-binding cassette subfamily member A3 (ABCA3).40,41 These findings are intriguing, although it is not yet certain that heterozygous ABCA3 variants alone are sufficient to confer FIP risk. Biallelic ABCA3 mutations cause severe and often fatal neonatal respiratory distress syndrome,42,43 with severity of neonatal phenotypes closely associated with degree of retained ABCA3 enzymatic activity.42 The reported ABCA3 variants are found at low, but detectable frequency in the general population, and studies to date have not yet been large enough to determine whether ABCA3 RVs are statistically overrepresented in FIP kindreds. It may be that penetrance of heterozygous ABCA3 variants is influenced by other genetic loci; for example, in one family, an ABCA3 RV appeared to influence disease natural history in a family carrying the I73T SFTPC mutation.44
Studies using heterologous cell lines and transgenic mouse models indicate that surfactant pathway mutations act predominantly through toxic gain-of-function mechanisms. The most common of the surfactant mutations, I73T SFTPC, leads to a trafficking defect wherein the proprotein is misprocessed, directed to the plasma membrane (rather than secreted), leads to alterations in protein quality-control associated with a block in the autophagy pathway.45 Transgenic mice inducibly expressing this mutation develop a macrophage-rich alveolitis46 and progressive parenchymal fibrosis.47 In contrast, mutations affecting the BRICHOS domain generally lead to failure of proprotein processing, retention and aggregation of the misfolded proprotein in the endoplasmic reticulum, activation of the unfolded protein response (UPR).48–53 Severe or prolonged UPR activation then leads to AT2 apoptosis, patchy parenchymal inflammation, and fibrotic remodeling.54 A similar phenotype has been described in mice homozygous for an Sftpa1 mutation.55 A mouse model expressing an Abca3 mutation (Abca3E292V) also develop alveolar inflammation, although surprisingly, with aging this is associated with an emphysema-phenotype.56
There is considerable heterogeneity among the clinical features and phenotypes observed in families with surfactant-related mutations, ranging from neonatal/pediatric presentations to disease detection late in adulthood. The factors responsible for this phenotypic variability are not well understood. While there are exceptions, compared to “sporadic” interstitial lung disease in adults, adults with surfactant-related mutations tend to have earlier disease onset, but progress somewhat more slowly - over the course of decades in some cases. Together, these available data indicate while surfactant-related mutations are a rare cause of FIP (comprising 1–3% of families), studying these mutations has yielded important insights into disease mechanisms with relevance to broad groups of patients with pulmonary fibrosis.25,51,57 It is not yet clear whether currently approved antifibrotic therapies58,59 have efficacy in patients with surfactant mutations, however recent studies demonstrating benefit of pirfenidone60 and nintedanib61 in broad groups of ILD patients suggest there is reason to consider antifibrotic therapy in the appropriate clinical context. Similar to the approach used in cystic fibrosis,62–64 in-vitro models indicate potential benefit of pharmacologic chaperones designed to improve trafficking of misfolded proproteins.53,65
Telomere-related genes
Dyskeratosis congenita (DC) is a syndrome characterized by abnormal skin pigmentation, oral leukoplakia, and nail dystrophy,66 often accompanied by bone marrow failure, chronic liver disease and other multisystem complications. Historically, it was recognized that pulmonary fibrosis occurred occasionaly in families with DC.67–70 In the late 1990’s into the early 2000’s, linkage studies identified mutations in the three genes related to telomere biology in families with DC: dyskerin (DKC1),71,72 telomerase RNA component (TERC),73,74 and telomerase reverse transcriptase (TERT).70,75
These observations led several groups to test for mutations in telomere-related genes in FIP kindreds. In 2007, two independent groups reported heterozygous TERT or TERC mutations in 6/7376 and 7/4677 FIP families. These mutations were associated with short peripheral blood telomeres and reduced telomerase enzymatic activity, but notably were not exclusively found in families with classic DC features. Subsequent studies using candidate gene and next-generation sequencing approaches have implicated mutations in the genes encoding for DKC1,26,78 TERF1 interacting factor 2 (TINF2),79,80 regulator of telomere elongation helicase 1 (RTEL1),29,30,81 poly-A-specific ribonuclease (PARN),30,82 nuclear assembly factor 1 (NAF1),83, and zinc finger CCHC-type containing 8 (ZCCHC8),84 in FIP kindreds. In contrast to the surfactant pathway, mutations in telomere-related genes generally lead to alterations in cellular homeostasis through a haploinsufficiency mechanism. To date, it appears that nearly all of the mutations in telomere related genes converge upon a phenotype of reduced functional telomerase activity through a variety of different mechanisms. TERT mutations directly lead to reduced catalytic activity of the reverse transcriptase enzyme,76,77 while TERC mutations reduce levels or impair the function of the RNA template required for addition of telomere repeats. DKC1,26,85 PARN,86 NAF1,83 and ZCCHC884 regulate RNA stability and are associated with reduced TERC levels. RTEL1 mutations do not directly affect telomerase enzymatic activity, but impair unwinding of T-loops, a crucial step required for telomerase to physically interact with telomere DNA-sequences.87 In aggregate, estimates from across studies suggest that mutations in telomere-related genes are found in 25–30% of FIP kindreds, and more than 50% of families with short peripheral blood telomeres.
While the genetic evidence implicating defects in telomerase to pulmonary fibrosis is strong, the mechanisms linking telomere dysfunction to chronic lung disease are not yet well understood. The prevailing hypothesis has been that telomerase dysfunction leads to stem cell failure in the lung, although direct evidence to support this hypothesis is limited. In the lung, telomerase expression and activity is low and cell turnover relatively slow under homeostatic conditions,18 and recent single-cell genomic studies have demonstrated only rare TERT expressing cells88–92 in control or disease lungs, consequently, it remains unclear which cell type(s) are central to mediating disease pathogenesis. Studies using global Tert and/or Terc null mice have reported contradictory results with regard to the development of spontaneous lung pathology and response to injury.93–98 Several groups have demonstrated that deletion of shelterin components (Trf1 or Trf2) in type II alveolar epithelial cells is sufficient to trigger a telomere DNA-damage response,95,99,100 leading to chronic inflammation and/or progressive fibrosis.
Compared to other FIP patients and ILD patients without a family history, families with mutations in telomere related genes appear to develop disease at a somewhat earlier age (5–10 years younger than IPF trials populations).29,76,77,101,102 A family history of premature greying, cryptogenic cirrhosis, or bone marrow dyscrasias (aplastic anemia, myelodysplastic syndrome, acute myeloid leukemia) is suggestive of a “short-telomere syndrome.” However, it should be noted that some families with mutations in telomere-related genes lack other systemic manifestations of short telomere syndromes. Genetic anticipation may be one mechanism driving the phenotypic heterogeneity of disease manifestations in these families,70 although genetic and/or environmental factors also likely contribute.
Diverse imaging and histopathologic disease patterns (ILD phenotypes) have been described in patients and families with mutations in telomere-related genes,101–104 and discordant ILD phenotypes are commonly observed within the same family.29,101,102 Importantly, the available data suggest that disease natural history among patients in these families is similar regardless of imaging or histopathologic ILD pattern.101 The efficacy of antifibrotic treatments in patients with telomere-related gene mutations is not known. A retrospective analysis of patients in the pirfenidone clinical trials suggested a similar, if not larger, effect size in IPF patients with the short-telomeres as estimated by whole-genome sequencing35 compared to those with longer telomeres, however no studies to date have reported genotype stratified treatment effects. Due to the risk of occult chronic liver disease, careful monitoring of liver function is required in antifibrotic therapies are initiated.
There are unique considerations around lung transplantation required for patients with telomere-related gene mutations. While lung transplant can be successfully performed, patients with mutations in telomere-related genes may be at increased risk for complications related to immunosuppresion,105,106 and at higher risk for acute rejection.106
Pulmonary fibrosis associated with other systemic disorders
In addition to short telomere syndromes, interstitial lung disease can be a feature of several other systemic disorders (Table 2).
Table 2.
Syndome | Genes associated | Key Clinical Features |
---|---|---|
Surfactant-related | SFTPC, SFTPA1, SFTPA2, ABCA3 | Neonatal respiratory distress syndrome |
Waxing-and-waning ground glass and reticulation | ||
Diagnosis at <1 year to >70 years | ||
Short telomere | TERT, RTEL1, PARN, TERC, NAF1, TINF2, DCK1, ZCCHC8 | Premature greying (early 20’s or younger) |
Cryptogenic cirrhosis | ||
Macrocytosis | ||
Bone marrow dyscrasias (aplastic anemia, MDS, AML) | ||
Brain-lung-thyroid | NKX2–1 | Movement disorders |
Hypothyroidism | ||
NEHI | ||
COPA | COPA | Inflammatory arthritis |
Elevated autoantibodies | ||
Pulmonary hemorrhage | ||
Interstitial and ground glass infiltrates | ||
Hermansky-Pudlak Syndrome | HPS1, AP3B1, HPS3, HPS4, HPS5, HPS6, DNTBP1, BLOC1S3, BLOC1S6, AP3D1 | Bleeding diathesis - platelet dysfunction |
Oculocutaneous albinism | ||
Pulmonary fibrosis (subtypes 1, 2, 4) |
Bold indicates HPS genes associated with pulmonary fibrosis
Hermansky-Pudlak Syndrome (HPS) is an autosomal-recessive multisystem disorder related to defects in lysosomal biogenesis,20 leading to clinical manifestations related to dysfunction of lysosomal-derived organelles. HPS is characterized by platelet dysfunction, oculocutaneous albinism, and pulmonary fibrosis.20 In the appropriate clinical context, a diagnosis of HPS required demonstration of absent dense granules by platelet electron microscopy. There are at least 10 different subtypes of HPS caused by mutations in different factors related to lysosomal biogenesis and trafficking. Pulmonary fibrosis is highly penetrant in patients with HPS1, HPS2, and HPS4, but not other HPS subtypes.20,21 As a result, in patients with HPS, genetic testing provides important information as to risk for interstitial lung disease, the most life-threatening complication of this syndrome. Two clinical trials have tested the antifibrotic pirfenidone in HPS patients;107,108 while these studies did not detect differences in their primary endpoints, it is recognized that these studies were likely underpowered to detect clinically important effects, and there are now reports demonstrating safety and tolerability of long-term pirfenidone therapy in HPS-associated pulmonary fibrosis.109 HPS has been considered a contraindication to treatment with nintedanib due to concern of excess bleeding risk.
“Brain-lung-thyroid syndrome”, a disorder caused by heterozygous mutations in the transcription factor NKX2–1,110 is characterized by hypothyroidism, chorea-form movement disorders, and respiratory disease. A spectrum of interstitial lung disease phenotypes have been described in families with NKX2–1 mutations. The majority of cases have been described in pediatric populations, and include neuroendocrine hyperplasia of infancy (NEHI),111 pathology similar to surfactant-related disease,112 and pulmonary alveolar proteinosis,22,112,113 however there are now multiple reports of interstitial lung disease in adults with NKX2–1 mutations.23,113 Corticosteroids, hydroxychloroquine, and azithromycin have been used to treat patients with NKX 2–1 mutations, but the efficacy of these therapies is not yet established.113
Diffuse lung disease has also been described in families with “COPA Syndrome”, a recently described disorder characterized by high-titer autoantibodies, inflammatory arthritis, and interstitial lung disease associated with mutations in the gene encoding for coatamer subunit alpha (COPA).24 While initial report described pulmonary hemorrhage and/or interstitial and ground-glass infiltrates primarily in children,24 several recent reports suggest there may be a broader spectrum of ILD-phenotypes associated with COPA mutations.114–117 Immunsuppressive treatment with corticosteroids and/or other agents is generally recommended and beneficial in these patients.24
Common genetic variation in FIP
Family-based cohort studies have focused primarily on rare genetic variants of large effect in families, however common genetic variation also contributes to FIP risk.118–120 The available data suggest that common genetic variants linked to IPF risk by linkage118 or genome-wide association studies,119 including the MUC5B promoter polymorphism and more than 15 additional loci119,121–123 have similar effect sizes in familial and sporadic IPF patients.119,120 As common genetic variants follow complex rather than Mendelian inheritance patterns, genetic testing for common genetic variants does not currently have a role in the clinical evaluation or care of patients with FIP.
Genetic testing
The role of genetic testing in the evaluation of patients and families with FIP is evolving, and at present there are no established guidelines for the use of genetic testing in adults with interstitial lung disease. There are two primary considerations when considering genetic testing in a patient with FIP: 1) What is the likelihood of finding a culprit variant, and 2) What, if any, action would result from a positive test?
Detection of a germline risk variant in a patient with FIP has implications not only for the patient, but also for siblings, children, and other family members, thus any consideration of genetic testing should be performed in coordination with experienced genetic counselors.19 If testing is performed, testing an affected patient (rather than an unaffected relative) is highly recommended in order to determine whether a potentially pathogenic mutation segregates with disease within the family. Current estimates suggest that likely or definitely pathogenic rare variants are found in 20–30% of unselected FIP patients, thus in the majority of patients with FIP, a culprit variant will not be found. As a result, when considering a genetic test, it is important to recognize that only a positive result is informative. A negative genetic test does not indicate absence of genetic risk. One approach that maximizes the likelihood of an informative genetic test is to use clinical features and family history to identify patients with a high pretest probability of identifying a culprit variant,19 and to restrict testing to genes in which a culprit variant is anticipated. For example, in a 35 year old with ILD who has a child with unexplained chronic lung disease, testing for a surfactant-related mutations would be appropriate. In contrast, this scenario is not suggestive of a short-telomere syndrome, so testing for mutations in telomere-related genes would likely have low utility.
If a genetic test is performed, the American College of Medical Genetics offers guidance as to interpretation of variants that are identified.124 In broad terms, a variant may be reported as likely pathogenic, a variant of uncertain significance, or likely benign. Variants that lead to truncated proteins or null alleles (nonsense variants), variants altering splicing, or insertions/deletions may be considered pathogenic in the absence of prior reports of the same variant in disease, however missense variations (leading to changes in a single amino acid) are more challenging to classify. Many FIP-associated mutations reported to date are private (reported in only a single family), thus these variants would be considered variants of uncertain significance in the absence of data demonstrating the mutation alters protein function. In some cases, functional assays or corroborative measurements (such as measurement of peripheral blood telomere length)19,125 can aid in the interpretation of variants of uncertain significance.
There are several scenarios where a genetic test is likely to yield clinically actionable results. Among patients with Hermansky-Pudlak Syndrome, genetic testing to determine HPS subtype can inform with high confidence whether or not a patient will develop pulmonary fibrosis. Patients with COPA mutations appear to respond to immunosuppression, which is not recommended for most ILD patients. Detection of a mutation in a telomere-related gene has prognostic implications, and may also inform immunosuppression strategies in patients who require lung transplantation. A positive genetic test also provides the opportunity for subsequent generations to undergo targeted testing. For children of FIP patients, knowledge of risk variant status may inform approaches to screening for ILD, and can also influence reproductive considerations.
Implications for relatives of FIP patients
It is clear that bloodline relatives of FIP patients are at markedly elevated risk for ILD compared to the general population4. With the availability of effective antifibrotic therapies58–61,126 that slow disease progression and appear to have similar efficacy in patients with relatively preserved pulmonary function,127–129 a compelling case can be made for screening and early disease detection strategies for individuals at high risk for disease. The timing, modality, and interval for such a strategy remains an area of active investigation.
Data from multiple studies have demonstrated that 10–25% of close relatives of FIP patients have detectable early interstitial changes on HRCT despite having no known lung disease,130–132 and this prevalence increases with age. Among these individuals with early interstitial changes, pulmonary function tests (PFTs) including diffusing capacity for carbon monoxide are typically normal,130 indicating that PFT-based screening alone is not sufficiently sensitive to detect early disease. The MUC5B promoter variant has been associated with interstitial lung abnormalities in both families132 and population-based cohorts.133–135 A number of peripheral blood biomarkers have been associated with early interstitial changes in families131 or general-population based cohorts,136–139 however none of these biomarkers are currently approved or available for clinical use. The role of environmental exposures as a determinant of disease penetrance and phenotype has not been well-studied; identifying modifiable risk factors could enable preventive strategies in high-risk individuals. Advanced machine-learning and artificial-intelligence-based image analysis techniques hold promise for defining high-risk features,132 but it remains to be seen whether the use of such techniques can detect subtle evidence of disease progression before symptoms or pulmonary function tests change. As longitudinal data become available, there will be opportunities to develop individualized models of risk for interstitial lung abnormaitiies (ILA) progression integrating genetic information, biomarkers, and physiologic measurements.
Conclusion
It has become clear through the past two decades that pulmonary fibrosis is a heritable syndrome, and studies of FIP have yielded new insights into the pathobiology of ILD with broad relevance. With increasing evidence indicating that known genetic risk factors for sporadic and familial ILD are much more similar than different,35,118–120 considerations that inform the approach to and care of patients and families with FIP are likely to become increasingly relevant to broad groups of patients with ILD and their families.
References
- 1.Travis WD, Costabel U, Hansell DM, et al. An official American Thoracic Society/European Respiratory Society statement: Update of the international multidisciplinary classification of the idiopathic interstitial pneumonias. Am J Respir Crit Care Med 2013;188(6):733–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Lederer DJ, Martinez FJ. Idiopathic Pulmonary Fibrosis. N Engl J Med 2018;378(19):1811–23. [DOI] [PubMed] [Google Scholar]
- 3.García-Sancho C, Buendía-Roldán I, Fernández-Plata MR, et al. Familial pulmonary fibrosis is the strongest risk factor for idiopathic pulmonary fibrosis. Respir Med 2011;105(12):1902–7. [DOI] [PubMed] [Google Scholar]
- 4.Steele MP, Speer MC, Loyd JE, et al. Clinical and pathologic features of familial interstitial pneumonia. Am J Respir Crit Care Med 2005;172(9):1146–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Talbert JL, Schwartz DA. Pulmonary Fibrosis, Familial In: Adam MP, Ardinger HH, Pagon RA, et al. , editors. GeneReviews®. Seattle (WA): University of Washington, Seattle; 2005. [Google Scholar]
- 6.Peabody JW, Peabody JW Jr, Hayes EW, Hayes EW Jr. Idiopathic pulmonary fibrosis; its occurrence in identical twin sisters. Dis Chest 1950;18(4):330–44. [DOI] [PubMed] [Google Scholar]
- 7.Beaumont F, Jansen HM, Elema JD, ten Kate LP, Sluiter HJ. Simultaneous occurrence of pulmonary interstitial fibrosis and alveolar cell carcinoma in one family. Thorax 1981;36(4):252–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Solliday NH, Williams JA, Gaensler EA, Coutu RE, Carrington CB. Familial chronic interstitial pneumonia. Am Rev Respir Dis 1973;108(2):193–204. [DOI] [PubMed] [Google Scholar]
- 9.Swaye P, Van Ordstrand HS, McCormack LJ, Wolpaw SE. Familial Hamman-Rich syndrome. Report of eight cases. Dis Chest 1969;55(1):7–12. [DOI] [PubMed] [Google Scholar]
- 10.Hughes EW. FAMILIAL INTERSTITIAL PULMONARY FIBROSIS. Thorax 1964;19:515–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Marshall RP, Puddicombe A, Cookson WO, Laurent GJ. Adult familial cryptogenic fibrosing alveolitis in the United Kingdom. Thorax 2000;55(2):143–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Hodgson U, Laitinen T, Tukiainen P. Nationwide prevalence of sporadic and familial idiopathic pulmonary fibrosis: evidence of founder effect among multiplex families in Finland. Thorax 2002;57(4):338–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Loyd JE. Pulmonary fibrosis in families. Am J Respir Cell Mol Biol 2003;29(3 Suppl):S47–50. [PubMed] [Google Scholar]
- 14.Fernandez BA, Fox G, Bhatia R, et al. A Newfoundland cohort of familial and sporadic idiopathic pulmonary fibrosis patients: clinical and genetic features. Respir Res 2012;13:64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Lee HY, Seo JB, Steele MP, et al. High-resolution CT scan findings in familial interstitial pneumonia do not conform to those of idiopathic interstitial pneumonia. Chest 2012;142(6):1577–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Thomas AQ, Lane K, Phillips J 3rd, et al. Heterozygosity for a surfactant protein C gene mutation associated with usual interstitial pneumonitis and cellular nonspecific interstitial pneumonitis in one kindred. Am J Respir Crit Care Med 2002;165(9):1322–8. [DOI] [PubMed] [Google Scholar]
- 17.Nogee LM, Dunbar AE 3rd, Wert SE, Askin F, Hamvas A, Whitsett JA. A mutation in the surfactant protein C gene associated with familial interstitial lung disease. N Engl J Med 2001;344(8):573–9. [DOI] [PubMed] [Google Scholar]
- 18.Armanios M Telomerase mutations and the pulmonary fibrosis-bone marrow failure syndrome complex. N. Engl. J. Med 2012;367(4):384; author reply 384. [DOI] [PubMed] [Google Scholar]
- 19.Kropski JA, Young LR, Cogan JD, et al. Genetic Evaluation and Testing of Patients and Families with Idiopathic Pulmonary Fibrosis. Am J Respir Crit Care Med 2017;195(11):1423–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.El-Chemaly S, Young LR. Hermansky-Pudlak Syndrome. Clin Chest Med 2016;37(3):505–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Vicary GW, Vergne Y, Santiago-Cornier A, Young LR, Roman J. Pulmonary Fibrosis in Hermansky-Pudlak Syndrome. Ann Am Thorac Soc 2016;13(10):1839–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Hamvas A, Deterding RR, Wert SE, et al. Heterogeneous pulmonary phenotypes associated with mutations in the thyroid transcription factor gene NKX2–1. Chest 2013;144(3):794–804. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Nevel RJ, Garnett ET, Worrell JA, et al. Persistent Lung Disease in Adults with NKX2.1 Mutation and Familial Neuroendocrine Cell Hyperplasia of Infancy. Ann Am Thorac Soc 2016;13(8):1299–304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Watkin LB, Jessen B, Wiszniewski W, et al. COPA mutations impair ER-Golgi transport and cause hereditary autoimmune-mediated lung disease and arthritis. Nat Genet 2015;47(6):654–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Kropski JA, Lawson WE, Young LR, Blackwell TS. Genetic studies provide clues on the pathogenesis of idiopathic pulmonary fibrosis. Dis Model Mech 2013;6(1):9–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Kropski JA, Mitchell DB, Markin C, et al. A novel dyskerin (DKC1) mutation is associated with familial interstitial pneumonia. Chest 2014;146(1):e1–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Zhang D, Zhou Z, Abu-Hijleh M, Batra K, Xing C, Garcia CK. Homozygous Rare PARN Missense Mutation in Familial Pulmonary Fibrosis. Am J Respir Crit Care Med 2019;199(6):797–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Al-Mutairy EA, Imtiaz FA, Khalid M, et al. An atypical pulmonary fibrosis is associated with co-inheritance of mutations in the calcium binding protein genes S100A3 and S100A13. Eur Respir J [Internet] 2019;54(1). Available from: 10.1183/13993003.02041-2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Cogan JD, Kropski JA, Zhao M, et al. Rare variants in RTEL1 are associated with familial interstitial pneumonia. Am J Respir Crit Care Med 2015;191(6):646–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Stuart BD, Choi J, Zaidi S, et al. Exome sequencing links mutations in PARN and RTEL1 with familial pulmonary fibrosis and telomere shortening. Nat Genet 2015;47(5):512–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Petrovski S, Todd JL, Durheim MT, et al. An Exome Sequencing Study to Assess the Role of Rare Genetic Variation in Pulmonary Fibrosis. Am J Respir Crit Care Med 2017;196(1):82–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.van Moorsel CHM, van Oosterhout MFM, Barlo NP, et al. Surfactant protein C mutations are the basis of a significant portion of adult familial pulmonary fibrosis in a dutch cohort. Am J Respir Crit Care Med 2010;182(11):1419–25. [DOI] [PubMed] [Google Scholar]
- 33.Lawson WE, Grant SW, Ambrosini V, et al. Genetic mutations in surfactant protein C are a rare cause of sporadic cases of IPF. Thorax 2004;59(11):977–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Markart P, Ruppert C, Wygrecka M, et al. Surfactant protein C mutations in sporadic forms of idiopathic interstitial pneumonias. Eur Respir J 2007;29(1):134–7. [DOI] [PubMed] [Google Scholar]
- 35.Dressen A, Abbas AR, Cabanski C, et al. Analysis of protein-altering variants in telomerase genes and their association with MUC5B common variant status in patients with idiopathic pulmonary fibrosis: a candidate gene sequencing study. Lancet Respir Med 2018;6(8):603–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.van Moorsel CHM, Ten Klooster L, van Oosterhout MFM, et al. SFTPA2 Mutations in Familial and Sporadic Idiopathic Interstitial Pneumonia. Am J Respir Crit Care Med 2015;192(10):1249–52. [DOI] [PubMed] [Google Scholar]
- 37.Wang Y, Kuan PJ, Xing C, et al. Genetic defects in surfactant protein A2 are associated with pulmonary fibrosis and lung cancer. Am J Hum Genet 2009;84(1):52–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Nathan N, Giraud V, Picard C, et al. Germline SFTPA1 mutation in familial idiopathic interstitial pneumonia and lung cancer. Hum Mol Genet 2016;25(8):1457–67. [DOI] [PubMed] [Google Scholar]
- 39.Doubková M, Staňo Kozubík K, Radová L, et al. A novel germline mutation of the SFTPA1 gene in familial interstitial pneumonia. Hum Genome Var 2019;6:12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Epaud R, Delestrain C, Louha M, Simon S, Fanen P, Tazi A. Combined pulmonary fibrosis and emphysema syndrome associated with ABCA3 mutations. Eur Respir J 2014;43(2):638–41. [DOI] [PubMed] [Google Scholar]
- 41.Campo I, Zorzetto M, Mariani F, et al. A large kindred of pulmonary fibrosis associated with a novel ABCA3 gene variant. Respir Res 2014;15:43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Wambach JA, Casey AM, Fishman MP, et al. Genotype-phenotype correlations for infants and children with ABCA3 deficiency. Am J Respir Crit Care Med 2014;189(12):1538–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Shulenin S, Nogee LM, Annilo T, Wert SE, Whitsett JA, Dean M. ABCA3 gene mutations in newborns with fatal surfactant deficiency. N Engl J Med 2004;350(13):1296–303. [DOI] [PubMed] [Google Scholar]
- 44.Crossno PF, Polosukhin VV, Blackwell TS, et al. Identification of early interstitial lung disease in an individual with genetic variations in ABCA3 and SFTPC. Chest 2010;137(4):969–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Hawkins A, Guttentag SH, Deterding R, et al. A non-BRICHOS SFTPC mutant (SP-CI73T) linked to interstitial lung disease promotes a late block in macroautophagy disrupting cellular proteostasis and mitophagy. Am J Physiol Lung Cell Mol Physiol 2015;308(1):L33–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Venosa A, Katzen J, Tomer Y, et al. Epithelial Expression of an Interstitial Lung Disease-Associated Mutation in Surfactant Protein-C Modulates Recruitment and Activation of Key Myeloid Cell Populations in Mice. J Immunol 2019;202(9):2760–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Nureki S-I, Tomer Y, Venosa A, et al. Expression of mutant Sftpc in murine alveolar epithelia drives spontaneous lung fibrosis. J Clin Invest 2018;128(9):4008–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Mulugeta S, Nguyen V, Russo SJ, Muniswamy M, Beers MF. A surfactant protein C precursor protein BRICHOS domain mutation causes endoplasmic reticulum stress, proteasome dysfunction, and caspase 3 activation. Am J Respir Cell Mol Biol 2005;32(6):521–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Bridges JP, Wert SE, Nogee LM, Weaver TE. Expression of a human surfactant protein C mutation associated with interstitial lung disease disrupts lung development in transgenic mice. J Biol Chem 2003;278(52):52739–46. [DOI] [PubMed] [Google Scholar]
- 50.Bridges JP, Xu Y, Na C-L, Wong HR, Weaver TE. Adaptation and increased susceptibility to infection associated with constitutive expression of misfolded SP-C. J Cell Biol 2006;172(3):395–407. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Lawson WE, Crossno PF, Polosukhin VV, et al. Endoplasmic reticulum stress in alveolar epithelial cells is prominent in IPF: association with altered surfactant protein processing and herpesvirus infection. Am J Physiol Lung Cell Mol Physiol 2008;294(6):L1119–26. [DOI] [PubMed] [Google Scholar]
- 52.Lawson WE, Cheng D-S, Degryse AL, et al. Endoplasmic reticulum stress enhances fibrotic remodeling in the lungs. Proc Natl Acad Sci U S A 2011;108(26):10562–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Stewart GA, Ridsdale R, Martin EP, et al. 4-Phenylbutyric acid treatment rescues trafficking and processing of a mutant surfactant protein-C. Am J Respir Cell Mol Biol 2012;47(3):324–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Katzen J, Wagner BD, Venosa A, et al. An SFTPC BRICHOS mutant links epithelial ER stress and spontaneous lung fibrosis. JCI Insight [Internet] 2019;4(6). Available from: 10.1172/jci.insight.126125 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Takezaki A, Tsukumo S-I, Setoguchi Y, et al. A homozygous SFTPA1 mutation drives necroptosis of type II alveolar epithelial cells in patients with idiopathic pulmonary fibrosis. J Exp Med [Internet] 2019;Available from: 10.1084/jem.20182351 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Beers MF, Knudsen L, Tomer Y, et al. Aberrant lung remodeling in a mouse model of surfactant dysregulation induced by modulation of the Abca3 gene. Ann Anat 2017;210:135–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Korfei M, Ruppert C, Mahavadi P, et al. Epithelial endoplasmic reticulum stress and apoptosis in sporadic idiopathic pulmonary fibrosis. Am J Respir Crit Care Med 2008;178(8):838–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.King TE Jr, Bradford WZ, Castro-Bernardini S, et al. A phase 3 trial of pirfenidone in patients with idiopathic pulmonary fibrosis. N Engl J Med 2014;370(22):2083–92. [DOI] [PubMed] [Google Scholar]
- 59.Richeldi L, du Bois RM, Raghu G, et al. Efficacy and safety of nintedanib in idiopathic pulmonary fibrosis. N Engl J Med 2014;370(22):2071–82. [DOI] [PubMed] [Google Scholar]
- 60.Maher TM, Corte TJ, Fischer A, et al. Pirfenidone in patients with unclassifiable progressive fibrosing interstitial lung disease: a double-blind, randomised, placebo-controlled, phase 2 trial. Lancet Respir Med [Internet] 2019;Available from: 10.1016/S2213-2600(19)30341-8 [DOI] [PubMed] [Google Scholar]
- 61.Flaherty KR, Wells AU, Cottin V, et al. Nintedanib in Progressive Fibrosing Interstitial Lung Diseases. N Engl J Med [Internet] 2019;Available from: 10.1056/NEJMoa1908681 [DOI] [PubMed] [Google Scholar]
- 62.Ramsey BW, Davies J, McElvaney NG, et al. A CFTR potentiator in patients with cystic fibrosis and the G551D mutation. N Engl J Med 2011;365(18):1663–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Heijerman HGM, McKone EF, Downey DG, et al. Efficacy and safety of the elexacaftor plus tezacaftor plus ivacaftor combination regimen in people with cystic fibrosis homozygous for the F508del mutation: a double-blind, randomised, phase 3 trial. Lancet 2019;394(10212):1940–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Middleton PG, Mall MA, Dřevínek P, et al. Elexacaftor-Tezacaftor-Ivacaftor for Cystic Fibrosis with a Single Phe508del Allele. N Engl J Med 2019;381(19):1809–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Jiang X, Fang G, Dong L, et al. Chemical chaperone 4-phenylbutyric acid alleviates the aggregation of human familial pulmonary fibrosis-related mutant SP-A2 protein in part through effects on GRP78. Biochim Biophys Acta Mol Basis Dis 2018;1864(10):3546–57. [DOI] [PubMed] [Google Scholar]
- 66.Dokal I Dyskeratosis congenita: an inherited bone marrow failure syndrome. Br J Haematol 1996;92(4):775–9. [DOI] [PubMed] [Google Scholar]
- 67.Safa WF, Lestringant GG, Frossard PM. X-linked dyskeratosis congenita: restrictive pulmonary disease and a novel mutation. Thorax 2001;56(11):891–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Kilic S, Kose H, Ozturk H. Pulmonary involvement in a patient with dyskeratosis congenita. Pediatr Int 2003;45(6):740–2. [DOI] [PubMed] [Google Scholar]
- 69.Utz JP, Ryu JH, Myers JL, Michels VV. Usual interstitial pneumonia complicating dyskeratosis congenita. Mayo Clin Proc 2005;80(6):817–21. [DOI] [PubMed] [Google Scholar]
- 70.Armanios M, Chen J-L, Chang Y-PC, et al. Haploinsufficiency of telomerase reverse transcriptase leads to anticipation in autosomal dominant dyskeratosis congenita. Proc Natl Acad Sci U S A 2005;102(44):15960–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Knight SW, Heiss NS, Vulliamy TJ, et al. X-linked dyskeratosis congenita is predominantly caused by missense mutations in the DKC1 gene. Am J Hum Genet 1999;65(1):50–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Heiss NS, Knight SW, Vulliamy TJ, et al. X-linked dyskeratosis congenita is caused by mutations in a highly conserved gene with putative nucleolar functions. Nat Genet 1998;19(1):32–8. [DOI] [PubMed] [Google Scholar]
- 73.Vulliamy T, Marrone A, Dokal I, Mason PJ. Association between aplastic anaemia and mutations in telomerase RNA. Lancet 2002;359(9324):2168–70. [DOI] [PubMed] [Google Scholar]
- 74.Vulliamy T, Marrone A, Szydlo R, Walne A, Mason PJ, Dokal I. Disease anticipation is associated with progressive telomere shortening in families with dyskeratosis congenita due to mutations in TERC. Nat Genet 2004;36(5):447–9. [DOI] [PubMed] [Google Scholar]
- 75.Savage SA, Stewart BJ, Weksler BB, et al. Mutations in the reverse transcriptase component of telomerase (TERT) in patients with bone marrow failure. Blood Cells Mol. Dis 2006;37(2):134–6. [DOI] [PubMed] [Google Scholar]
- 76.Armanios MY, Chen JJ-L, Cogan JD, et al. Telomerase mutations in families with idiopathic pulmonary fibrosis. N Engl J Med 2007;356(13):1317–26. [DOI] [PubMed] [Google Scholar]
- 77.Tsakiri KD, Cronkhite JT, Kuan PJ, et al. Adult-onset pulmonary fibrosis caused by mutations in telomerase. Proc Natl Acad Sci U S A 2007;104(18):7552–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Alder JK, Parry EM, Yegnasubramanian S, et al. Telomere phenotypes in females with heterozygous mutations in the dyskeratosis congenita 1 (DKC1) gene. Hum Mutat 2013;34(11):1481–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Du H, Guo Y, Ma D, et al. A case report of heterozygous TINF2 gene mutation associated with pulmonary fibrosis in a patient with dyskeratosis congenita. Medicine 2018;97(19):e0724. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Alder JK, Stanley SE, Wagner CL, Hamilton M, Hanumanthu VS, Armanios M. Exome sequencing identifies mutant TINF2 in a family with pulmonary fibrosis. Chest 2015;147(5):1361–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Kannengiesser C, Borie R, Ménard C, et al. Heterozygous RTEL1 mutations are associated with familial pulmonary fibrosis. Eur Respir J 2015;46(2):474–85. [DOI] [PubMed] [Google Scholar]
- 82.Kropski JA, Reiss S, Markin C, et al. Rare Genetic Variants in PARN Are Associated with Pulmonary Fibrosis in Families. Am J Respir Crit Care Med 2017;196(11):1481–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Stanley SE, Gable DL, Wagner CL, et al. Loss-of-function mutations in the RNA biogenesis factor NAF1 predispose to pulmonary fibrosis-emphysema. Sci Transl Med 2016;8(351):351ra107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Gable DL, Gaysinskaya V, Atik CC, et al. ZCCHC8, the nuclear exosome targeting component, is mutated in familial pulmonary fibrosis and is required for telomerase RNA maturation. Genes Dev 2019;33(19–20):1381–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Brault ME, Lauzon C, Autexier C. Dyskeratosis congenita mutations in dyskerin SUMOylation consensus sites lead to impaired telomerase RNA accumulation and telomere defects. Hum Mol Genet 2013;22(17):3498–507. [DOI] [PubMed] [Google Scholar]
- 86.Tummala H, Walne A, Collopy L, et al. Poly(A)-specific ribonuclease deficiency impacts telomere biology and causes dyskeratosis congenita. J Clin Invest 2015;125(5):2151–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Vannier J-B, Pavicic-Kaltenbrunner V, Petalcorin MIR, Ding H, Boulton SJ. RTEL1 dismantles T loops and counteracts telomeric G4-DNA to maintain telomere integrity. Cell 2012;149(4):795–806. [DOI] [PubMed] [Google Scholar]
- 88.Habermann AC, Gutierrez AJ, Bui LT, et al. Single-cell RNA-sequencing reveals profibrotic roles of distinct epithelial and mesenchymal lineages in pulmonary fibrosis [Internet]. bioRxiv. 2019. [cited 2019 Oct 14];753806 Available from: https://www.biorxiv.org/content/10.1101/753806v1.article-metrics [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Adams TS, Schupp JC, Poli S, et al. Single Cell RNA-seq reveals ectopic and aberrant lung resident cell populations in Idiopathic Pulmonary Fibrosis [Internet]. bioRxiv. 2019. [cited 2019 Oct 24];759902 Available from: https://www.biorxiv.org/content/10.1101/759902v3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Morse C, Tabib T, Sembrat J, et al. Proliferating SPP1/MERTK-expressing macrophages in idiopathic pulmonary fibrosis. Eur Respir J [Internet] 2019;54(2). Available from: 10.1183/13993003.02441-2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Reyfman PA, Walter JM, Joshi N, et al. Single-Cell Transcriptomic Analysis of Human Lung Provides Insights into the Pathobiology of Pulmonary Fibrosis. Am J Respir Crit Care Med 2019;199(12):1517–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Xu Y, Mizuno T, Sridharan A, et al. Single-cell RNA sequencing identifies diverse roles of epithelial cells in idiopathic pulmonary fibrosis. JCI Insight 2016;1(20):e90558. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Degryse AL, Xu XC, Newman JL, et al. Telomerase deficiency does not alter bleomycin-induced fibrosis in mice. Exp Lung Res 2012;38(3):124–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Liu T, Chung MJ, Ullenbruch M, et al. Telomerase activity is required for bleomycin-induced pulmonary fibrosis in mice. J Clin Invest 2007;117(12):3800–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Povedano JM, Martinez P, Flores JM, Mulero F, Blasco MA. Mice with Pulmonary Fibrosis Driven by Telomere Dysfunction. Cell Rep 2015;12(2):286–99. [DOI] [PubMed] [Google Scholar]
- 96.Chen R, Zhang K, Chen H, et al. Telomerase Deficiency Causes Alveolar Stem Cell Senescence-associated Low-grade Inflammation in Lungs. J Biol Chem 2015;290(52):30813–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Lee J, Reddy R, Barsky L, et al. Lung alveolar integrity is compromised by telomere shortening in telomerase-null mice. Am J Physiol Lung Cell Mol Physiol 2009;296(1):L57–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Jackson S-R, Lee J, Reddy R, et al. Partial pneumonectomy of telomerase null mice carrying shortened telomeres initiates cell growth arrest resulting in a limited compensatory growth response. Am J Physiol Lung Cell Mol Physiol 2011;300(6):L898–909. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Naikawadi RP, Disayabutr S, Mallavia B, et al. Telomere dysfunction in alveolar epithelial cells causes lung remodeling and fibrosis. JCI Insight 2016;1(14):e86704. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Alder JK, Barkauskas CE, Limjunyawong N, et al. Telomere dysfunction causes alveolar stem cell failure. Proc Natl Acad Sci U S A 2015;112(16):5099–104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Newton CA, Batra K, Torrealba J, et al. Telomere-related lung fibrosis is diagnostically heterogeneous but uniformly progressive. Eur Respir J 2016;48(6):1710–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Borie R, Bouvry D, Cottin V, et al. Regulator of telomere length 1 (RTEL1) mutations are associated with heterogeneous pulmonary and extra-pulmonary phenotypes. Eur Respir J [Internet] 2019;53(2). Available from: 10.1183/13993003.00508-2018 [DOI] [PubMed] [Google Scholar]
- 103.Newton CA, Batra K, Torrealba J, Meyer K, Raghu G, Garcia CK. Pleuroparenchymal fibroelastosis associated with telomerase reverse transcriptase mutations [Internet]. Eur. Respir. J 2017;49(5). Available from: 10.1183/13993003.00696-2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Juge P-A, Borie R, Kannengiesser C, et al. Shared genetic predisposition in rheumatoid arthritis-interstitial lung disease and familial pulmonary fibrosis. Eur Respir J [Internet] 2017;49(5). Available from: 10.1183/13993003.02314-2016 [DOI] [PubMed] [Google Scholar]
- 105.Silhan LL, Shah PD, Chambers DC, et al. Lung transplantation in telomerase mutation carriers with pulmonary fibrosis. Eur Respir J 2014;44(1):178–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Newton CA, Kozlitina J, Lines JR, Kaza V, Torres F, Garcia CK. Telomere length in patients with pulmonary fibrosis associated with chronic lung allograft dysfunction and post-lung transplantation survival. J Heart Lung Transplant 2017;36(8):845–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.O’Brien K, Troendle J, Gochuico BR, et al. Pirfenidone for the treatment of Hermansky-Pudlak syndrome pulmonary fibrosis. Mol Genet Metab 2011;103(2):128–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Gahl WA, Brantly M, Troendle J, et al. Effect of pirfenidone on the pulmonary fibrosis of Hermansky-Pudlak syndrome. Mol Genet Metab 2002;76(3):234–42. [DOI] [PubMed] [Google Scholar]
- 109.O’Brien KJ, Introne WJ, Akal O, et al. Prolonged treatment with open-label pirfenidone in Hermansky-Pudlak syndrome pulmonary fibrosis. Mol Genet Metab 2018;125(1–2):168–73. [DOI] [PubMed] [Google Scholar]
- 110.Patel NJ, Jankovic J. NKX2–1-Related Disorders In: Adam MP, Ardinger HH, Pagon RA, et al. , editors. GeneReviews®. Seattle (WA): University of Washington, Seattle; 2014. [PubMed] [Google Scholar]
- 111.Young LR, Deutsch GH, Bokulic RE, Brody AS, Nogee LM. A mutation in TTF1/NKX2.1 is associated with familial neuroendocrine cell hyperplasia of infancy. Chest 2013;144(4):1199–206. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.LeMoine BD, Browne LP, Liptzin DR, Deterding RR, Galambos C, Weinman JP. High-resolution computed tomography findings of thyroid transcription factor 1 deficiency (NKX2–1 mutations). Pediatr Radiol 2019;49(7):869–75. [DOI] [PubMed] [Google Scholar]
- 113.Nattes E, Lejeune S, Carsin A, et al. Heterogeneity of lung disease associated with NK2 homeobox 1 mutations. Respir Med 2017;129:16–23. [DOI] [PubMed] [Google Scholar]
- 114.Vece TJ, Watkin LB, Nicholas S, et al. Copa Syndrome: a Novel Autosomal Dominant Immune Dysregulatory Disease. J Clin Immunol 2016;36(4):377–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Patwardhan A, Spencer CH. An unprecedented COPA gene mutation in two patients in the same family: comparative clinical analysis of newly reported patients with other known COPA gene mutations. Pediatr Rheumatol Online J 2019;17(1):59. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Jensson BO, Hansdottir S, Arnadottir GA, et al. COPA syndrome in an Icelandic family caused by a recurrent missense mutation in COPA. BMC Med Genet 2017;18(1):129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Taveira-DaSilva AM, Markello TC, Kleiner DE, et al. Expanding the phenotype of COPA syndrome: a kindred with typical and atypical features. J Med Genet 2019;56(11):778–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Seibold MA, Wise AL, Speer MC, et al. A common MUC5B promoter polymorphism and pulmonary fibrosis. N Engl J Med 2011;364(16):1503–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Fingerlin TE, Murphy E, Zhang W, et al. Genome-wide association study identifies multiple susceptibility loci for pulmonary fibrosis. Nat Genet 2013;45(6):613–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Moore C, Blumhagen RZ, Yang IV, et al. Resequencing Study Confirms That Host Defense and Cell Senescence Gene Variants Contribute to the Risk of Idiopathic Pulmonary Fibrosis. Am J Respir Crit Care Med 2019;200(2):199–208. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Noth I, Zhang Y, Ma S-F, et al. Genetic variants associated with idiopathic pulmonary fibrosis susceptibility and mortality: a genome-wide association study. Lancet Respir Med 2013;1(4):309–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Allen RJ, Porte J, Braybrooke R, et al. Genetic variants associated with susceptibility to idiopathic pulmonary fibrosis in people of European ancestry: a genome-wide association study. Lancet Respir Med 2017;5(11):869–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Allen RJ, Guillen-Guio B, Oldham JM, et al. Genome-Wide Association Study of Susceptibility to Idiopathic Pulmonary Fibrosis. Am J Respir Crit Care Med [Internet] 2019;Available from: 10.1164/rccm.201905-1017OC [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Richards S, Aziz N, Bale S, et al. Standards and guidelines for the interpretation of sequence variants: a joint consensus recommendation of the American College of Medical Genetics and Genomics and the Association for Molecular Pathology. Genet Med 2015;17(5):405–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Alder JK, Hanumanthu VS, Strong MA, et al. Diagnostic utility of telomere length testing in a hospital-based setting. Proc Natl Acad Sci U S A 2018;115(10):E2358–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Distler O, Highland KB, Gahlemann M, et al. Nintedanib for Systemic Sclerosis-Associated Interstitial Lung Disease. N Engl J Med 2019;380(26):2518–28. [DOI] [PubMed] [Google Scholar]
- 127.Albera C, Costabel U, Fagan EA, et al. Efficacy of pirfenidone in patients with idiopathic pulmonary fibrosis with more preserved lung function. Eur Respir J 2016;48(3):843–51. [DOI] [PubMed] [Google Scholar]
- 128.Kolb M, Richeldi L, Behr J, et al. Nintedanib in patients with idiopathic pulmonary fibrosis and preserved lung volume. Thorax 2017;72(4):340–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Maher TM, Stowasser S, Nishioka Y, et al. Biomarkers of extracellular matrix turnover in patients with idiopathic pulmonary fibrosis given nintedanib (INMARK study): a randomised, placebo-controlled study. Lancet Respir Med 2019;7(9):771–9. [DOI] [PubMed] [Google Scholar]
- 130.Rosas IO, Ren P, Avila NA, et al. Early interstitial lung disease in familial pulmonary fibrosis. Am J Respir Crit Care Med 2007;176(7):698–705. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Kropski JA, Pritchett JM, Zoz DF, et al. Extensive phenotyping of individuals at risk for familial interstitial pneumonia reveals clues to the pathogenesis of interstitial lung disease. Am J Respir Crit Care Med 2015;191(4):417–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Mathai SK, Humphries S, Kropski JA, et al. MUC5B variant is associated with visually and quantitatively detected preclinical pulmonary fibrosis. Thorax [Internet] 2019;Available from: 10.1136/thoraxjnl-2018-212430 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Putman RK, Gudmundsson G, Araki T, et al. The MUC5B promoter polymorphism is associated with specific interstitial lung abnormality subtypes. Eur Respir J [Internet] 2017;50(3). Available from: 10.1183/13993003.00537-2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Hunninghake GM, Hatabu H, Okajima Y, et al. MUC5B promoter polymorphism and interstitial lung abnormalities. N Engl J Med 2013;368(23):2192–200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Hobbs BD, Putman RK, Araki T, et al. Overlap of Genetic Risk Between Interstitial Lung Abnormalities and Idiopathic Pulmonary Fibrosis. Am J Respir Crit Care Med [Internet] 2019;Available from: 10.1164/rccm.201903-0511OC [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Ho JE, Gao W, Levy D, et al. Galectin-3 Is Associated with Restrictive Lung Disease and Interstitial Lung Abnormalities. Am J Respir Crit Care Med 2016;194(1):77–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Madahar P, Duprez DA, Podolanczuk AJ, et al. Collagen biomarkers and subclinical interstitial lung disease: The Multi-Ethnic Study of Atherosclerosis. Respir Med 2018;140:108–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Kim JS, Anderson MR, Podolanczuk AJ, et al. Associations of Serum Adipokines With Subclinical Interstitial Lung Disease Among Community-Dwelling Adults: The Multi-Ethnic Study of Atherosclerosis (MESA). Chest [Internet] 2019;Available from: 10.1016/j.chest.2019.10.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.McGroder CF, Aaron CP, Bielinski SJ, et al. Circulating adhesion molecules and subclinical interstitial lung disease: the Multi-Ethnic Study of Atherosclerosis. Eur Respir J [Internet] 2019;54(3). Available from: 10.1183/13993003.00295-2019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Alder JK, Cogan JD, Brown AF, et al. Ancestral mutation in telomerase causes defects in repeat addition processivity and manifests as familial pulmonary fibrosis. PLoS Genet 2011;7(3):e1001352. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Ono S, Tanaka T, Ishida M, et al. Surfactant protein C G100S mutation causes familial pulmonary fibrosis in Japanese kindred. Eur Respir J 2011;38(4):861–9. [DOI] [PubMed] [Google Scholar]
- 142.Cottin V, Reix P, Khouatra C, Thivolet-Béjui F, Feldmann D, Cordier J-F. Combined pulmonary fibrosis and emphysema syndrome associated with familial SFTPC mutation. Thorax 2011;66(10):918–9. [DOI] [PubMed] [Google Scholar]