201
|
Furini F, Carnevale A, Casoni GL, Guerrini G, Cavagna L, Govoni M, Sciré CA. The Role of the Multidisciplinary Evaluation of Interstitial Lung Diseases: Systematic Literature Review of the Current Evidence and Future Perspectives. Front Med (Lausanne) 2019; 6:246. [PMID: 31750308 PMCID: PMC6842981 DOI: 10.3389/fmed.2019.00246] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 10/15/2019] [Indexed: 12/02/2022] Open
Abstract
The opportunity of a multidisciplinary evaluation for the diagnosis of interstitial pneumonias highlighted a major change in the diagnostic approach to diffuse lung disease. The new American Thoracic Society, European Respiratory Society, Japanese Respiratory Society, and Latin American Thoracic Society guidelines for the diagnosis of idiopathic pulmonary fibrosis have reinforced this assumption and have underlined that the exclusion of connective tissue disease related lung involvement is mandatory, with obvious clinical and therapeutic impact. The multidisciplinary team discussion consists in a moment of interaction among the radiologist, pathologist and pulmonologist, also including the rheumatologist when considered necessary, to improve diagnostic agreement and optimize the definition of those cases in which pulmonary involvement may represent the first or prominent manifestation of an autoimmune systemic disease. Moreover, the proposal of classification criteria for interstitial lung disease with autoimmune features (IPAF) represents an effort to define lung involvement in clinically undefined autoimmune conditions. The complexity of autoimmune diseases, and in particular the lack of classification criteria defined for pathologies such as anti-synthetase syndrome, makes the involvement of the rheumatologist essential for the correct interpretation of the autoimmune element and for the application of classification criteria, that could replace clinical pictures initially interpreted as IPAF in defined autoimmune disease, minimizing the risk of misdiagnosis. The aim of this review was to evaluate the available evidence about the efficiency and efficacy of different multidisciplinary team approaches, in order to standardize the professional figures and the core set procedures that should be necessary for a correct approach in diagnosing patients with interstitial lung disease.
Collapse
Affiliation(s)
- Federica Furini
- Section of Rheumatology, Department of Medical Sciences, University of Ferrara and Azienda Ospedaliero-Universitaria Sant'Anna di Ferrara, Cona, Italy
| | - Aldo Carnevale
- Department of Radiology, Azienda Ospedaliero-Universitaria Sant'Anna di Ferrara, Cona, Italy
| | - Gian Luca Casoni
- Department of Medical Sciences, Research Centre on Asthma and COPD, University of Ferrara, Ferrara, Italy
| | - Giulio Guerrini
- Section of Rheumatology, Department of Medical Sciences, University of Ferrara and Azienda Ospedaliero-Universitaria Sant'Anna di Ferrara, Cona, Italy
| | - Lorenzo Cavagna
- Division of Rheumatology, University and IRCCS Policlinico S. Matteo Foundation, Pavia, Italy
| | - Marcello Govoni
- Section of Rheumatology, Department of Medical Sciences, University of Ferrara and Azienda Ospedaliero-Universitaria Sant'Anna di Ferrara, Cona, Italy
| | - Carlo Alberto Sciré
- Section of Rheumatology, Department of Medical Sciences, University of Ferrara and Azienda Ospedaliero-Universitaria Sant'Anna di Ferrara, Cona, Italy
| |
Collapse
|
202
|
Collins BF, Raghu G. Antifibrotic therapy for fibrotic lung disease beyond idiopathic pulmonary fibrosis. Eur Respir Rev 2019; 28:28/153/190022. [PMID: 31578210 DOI: 10.1183/16000617.0022-2019] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Accepted: 05/24/2019] [Indexed: 11/05/2022] Open
Abstract
Two antifibrotic medications (nintedanib and pirfenidone) were recommended (conditionally) for the treatment of patients with idiopathic pulmonary fibrosis (IPF) in the 2015 IPF evidence-based guidelines. These medications have been shown to reduce the rate of decline in forced vital capacity among patients with IPF over time and are the only two disease-modulating pharmacological agents approved by regulatory agencies and available for clinical use worldwide. With the evolved standard of care for interstitial lung disease evaluation including routine use of high-resolution computed tomography, fibrotic lung diseases other than IPF are increasingly recognised. In addition, it is becoming evident that genetic and pathophysiological mechanisms as well as disease behaviour in patients manifesting other "non-IPF progressive fibrotic interstitial lung diseases" (non-IPF-PF) may be similar to those in patients with IPF. Thus, it is biologically plausible that pharmacological agents with antifibrotic properties may be efficacious in non-IPF-PF. Indeed, studies are underway or planned to assess the safety and efficacy of nintedanib or pirfenidone among patients with several non-IPF fibrotic lung diseases. In this review, we briefly summarise the use of pirfenidone and nintedanib in IPF as well as the rationale and potential for use of these medications in non-IPF-PF that are being investigated in ongoing and upcoming clinical trials.
Collapse
Affiliation(s)
- Bridget F Collins
- Center for Interstitial Lung Diseases, University of Washington Medical Center, Seattle, WA, USA
| | - Ganesh Raghu
- Center for Interstitial Lung Diseases, University of Washington Medical Center, Seattle, WA, USA
| |
Collapse
|
203
|
Borie R, Le Guen P, Ghanem M, Taillé C, Dupin C, Dieudé P, Kannengiesser C, Crestani B. The genetics of interstitial lung diseases. Eur Respir Rev 2019; 28:28/153/190053. [PMID: 31554702 PMCID: PMC9488931 DOI: 10.1183/16000617.0053-2019] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 08/01/2019] [Indexed: 12/21/2022] Open
Abstract
Interstitial lung diseases (ILDs) are a set of heterogeneous lung diseases characterised by inflammation and, in some cases, fibrosis. These lung conditions lead to dyspnoea, cough, abnormalities in gas exchange, restrictive physiology (characterised by decreased lung volumes), hypoxaemia and, if progressive, respiratory failure. In some cases, ILDs can be caused by systemic diseases or environmental exposures. The ability to treat or cure these ILDs varies based on the subtype and in many cases lung transplantation remains the only curative therapy. There is a growing body of evidence that both common and rare genetic variants contribute to the development and clinical manifestation of many of the ILDs. Here, we review the current understanding of genetic risk and ILD. Common and rare genetic variants contribute to the development and clinical manifestation of many interstitial lung diseaseshttp://bit.ly/31loHLh
Collapse
Affiliation(s)
- Raphael Borie
- Service de Pneumologie A, Hôpital Bichat, AP-HP, Paris, France.,INSERM U1152, Paris, France
| | - Pierre Le Guen
- Service de Pneumologie A, Hôpital Bichat, AP-HP, Paris, France.,INSERM U1152, Paris, France
| | - Mada Ghanem
- Service de Pneumologie A, Hôpital Bichat, AP-HP, Paris, France.,INSERM U1152, Paris, France
| | - Camille Taillé
- Service de Pneumologie A, Hôpital Bichat, AP-HP, Paris, France.,INSERM U1152, Paris, France
| | - Clairelyne Dupin
- Service de Pneumologie A, Hôpital Bichat, AP-HP, Paris, France.,INSERM U1152, Paris, France
| | - Philippe Dieudé
- INSERM U1152, Paris, France.,Département de Génétique, Hôpital Bichat, AP-HP, Paris, France
| | - Caroline Kannengiesser
- INSERM U1152, Paris, France.,Service de Rhumatologie, Hôpital Bichat, AP-HP, Paris, France
| | - Bruno Crestani
- Service de Pneumologie A, Hôpital Bichat, AP-HP, Paris, France .,INSERM U1152, Paris, France
| |
Collapse
|
204
|
Dodson LM, Baldan A, Nissbeck M, Gunja SMR, Bonnen PE, Aubert G, Birchansky S, Virtanen A, Bertuch AA. From incomplete penetrance with normal telomere length to severe disease and telomere shortening in a family with monoallelic and biallelic PARN pathogenic variants. Hum Mutat 2019; 40:2414-2429. [PMID: 31448843 DOI: 10.1002/humu.23898] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 07/24/2019] [Accepted: 08/19/2019] [Indexed: 12/21/2022]
Abstract
PARN encodes poly(A)-specific ribonuclease. Biallelic and monoallelic PARN variants are associated with Hoyeraal-Hreidarsson syndrome/dyskeratosis congenita and idiopathic pulmonary fibrosis (IPF), respectively. The molecular features associated with incomplete penetrance of PARN-associated IPF have not been described. We report a family with a rare missense, p.Y91C, and a novel insertion, p.(I274*), PARN variant. We found PARN p.Y91C had reduced deadenylase activity and the p.(I274*) transcript was depleted. Detailed analysis of the consequences of these variants revealed that, while PARN protein was lowest in the severely affected biallelic child who had the shortest telomeres, it was also reduced in his mother with the p.(I274*) variant but telomeres at the 50th percentile. Increased adenylation of telomerase RNA, human telomerase RNA, and certain small nucleolar RNAs, and impaired ribosomal RNA maturation were observed in cells derived from the severely affected biallelic carrier, but not in the other, less affected biallelic carrier, who had less severely shortened telomeres, nor in the monoallelic carriers who were unaffected and had telomeres ranging from the 1st to the 50th percentiles. We identified hsa-miR-202-5p as a potential negative regulator of PARN. We propose one or more genetic modifiers influence the impact of PARN variants on its targets and this underlies incomplete penetrance of PARN-associated disease.
Collapse
Affiliation(s)
- Lois M Dodson
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
| | - Alessandro Baldan
- Department of Pediatrics, Hematology/Oncology, Baylor College of Medicine and Texas Children's Hospital, Houston, Texas
| | - Mikael Nissbeck
- Department of Cell and Molecular Biology, Uppsala University, Uppsala, Sweden
| | - Sethu M R Gunja
- Department of Cell and Molecular Biology, Uppsala University, Uppsala, Sweden
| | - Penelope E Bonnen
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
| | - Geraldine Aubert
- Repeat Diagnostics Inc., North Vancouver, British Columbia, Canada
| | - Sherri Birchansky
- Department of Radiology, Baylor College of Medicine and Texas Children's Hospital, Houston, Texas
| | - Anders Virtanen
- Department of Cell and Molecular Biology, Uppsala University, Uppsala, Sweden
| | - Alison A Bertuch
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas.,Department of Pediatrics, Hematology/Oncology, Baylor College of Medicine and Texas Children's Hospital, Houston, Texas
| |
Collapse
|
205
|
Raghu G, Remy-Jardin M, Myers JL, Richeldi L, Ryerson CJ, Lederer DJ, Behr J, Cottin V, Danoff SK, Morell F, Flaherty KR, Wells A, Martinez FJ, Azuma A, Bice TJ, Bouros D, Brown KK, Collard HR, Duggal A, Galvin L, Inoue Y, Jenkins RG, Johkoh T, Kazerooni EA, Kitaichi M, Knight SL, Mansour G, Nicholson AG, Pipavath SNJ, Buendía-Roldán I, Selman M, Travis WD, Walsh S, Wilson KC. Diagnosis of Idiopathic Pulmonary Fibrosis. An Official ATS/ERS/JRS/ALAT Clinical Practice Guideline. Am J Respir Crit Care Med 2019; 198:e44-e68. [PMID: 30168753 DOI: 10.1164/rccm.201807-1255st] [Citation(s) in RCA: 2596] [Impact Index Per Article: 432.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND This document provides clinical recommendations for the diagnosis of idiopathic pulmonary fibrosis (IPF). It represents a collaborative effort between the American Thoracic Society, European Respiratory Society, Japanese Respiratory Society, and Latin American Thoracic Society. METHODS The evidence syntheses were discussed and recommendations formulated by a multidisciplinary committee of IPF experts. The evidence was appraised and recommendations were formulated, written, and graded using the Grading of Recommendations, Assessment, Development, and Evaluation approach. RESULTS The guideline panel updated the diagnostic criteria for IPF. Previously defined patterns of usual interstitial pneumonia (UIP) were refined to patterns of UIP, probable UIP, indeterminate, and alternate diagnosis. For patients with newly detected interstitial lung disease (ILD) who have a high-resolution computed tomography scan pattern of probable UIP, indeterminate, or an alternative diagnosis, conditional recommendations were made for performing BAL and surgical lung biopsy; because of lack of evidence, no recommendation was made for or against performing transbronchial lung biopsy or lung cryobiopsy. In contrast, for patients with newly detected ILD who have a high-resolution computed tomography scan pattern of UIP, strong recommendations were made against performing surgical lung biopsy, transbronchial lung biopsy, and lung cryobiopsy, and a conditional recommendation was made against performing BAL. Additional recommendations included a conditional recommendation for multidisciplinary discussion and a strong recommendation against measurement of serum biomarkers for the sole purpose of distinguishing IPF from other ILDs. CONCLUSIONS The guideline panel provided recommendations related to the diagnosis of IPF.
Collapse
|
206
|
Niewisch MR, Savage SA. An update on the biology and management of dyskeratosis congenita and related telomere biology disorders. Expert Rev Hematol 2019; 12:1037-1052. [PMID: 31478401 DOI: 10.1080/17474086.2019.1662720] [Citation(s) in RCA: 120] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Introduction: Telomere biology disorders (TBDs) encompass a group of illnesses caused by germline mutations in genes regulating telomere maintenance, resulting in very short telomeres. Possible TBD manifestations range from complex multisystem disorders with onset in childhood such as dyskeratosis congenita (DC), Hoyeraal-Hreidarsson syndrome, Revesz syndrome and Coats plus to adults presenting with one or two DC-related features.Areas covered: The discovery of multiple genetic causes and inheritance patterns has led to the recognition of a spectrum of clinical features affecting multiple organ systems. Patients with DC and associated TBDs are at high risk of bone marrow failure, cancer, liver and pulmonary disease. Recently, vascular diseases, including pulmonary arteriovenous malformations and gastrointestinal telangiectasias, have been recognized as additional manifestations. Diagnostics include detection of very short leukocyte telomeres and germline genetic testing. Hematopoietic cell transplantation and lung transplantation are the only current therapeutic modalities but are complicated by numerous comorbidities. This review summarizes the pathophysiology underlying TBDs, associated clinical features, management recommendations and therapeutic options.Expert opinion: Understanding TBDs as complex, multisystem disorders with a heterogenous genetic background and diverse phenotypes, highlights the importance of clinical surveillance and the urgent need to develop new therapeutic strategies to improve health outcomes.
Collapse
Affiliation(s)
- Marena R Niewisch
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Sharon A Savage
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
207
|
Callahan S, Pal K, Gomez D, Stoler M, Mehrad B. Two Siblings With Interstitial Lung Disease. Chest 2019; 153:e75-e79. [PMID: 29626972 DOI: 10.1016/j.chest.2017.12.020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Revised: 11/24/2017] [Accepted: 12/23/2017] [Indexed: 10/17/2022] Open
Abstract
A 52-year-old white woman and her 61-year-old white brother separately presented with gradually worsening dyspnea on exertion and cough, and evidence of interstitial lung disease on chest imaging.
Collapse
Affiliation(s)
- Sean Callahan
- Division of Pulmonary and Critical Care Medicine, University of Virginia, Charlottesville, VA
| | - Kavita Pal
- Division of Pulmonary and Critical Care Medicine, University of Virginia, Charlottesville, VA
| | - Diana Gomez
- Division of Pulmonary and Critical Care Medicine, University of Virginia, Charlottesville, VA
| | - Mark Stoler
- Department of Pathology, University of Virginia, Charlottesville, VA
| | - Borna Mehrad
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of Florida, Gainesville, FL.
| |
Collapse
|
208
|
Swaminathan AC, Neely ML, Frankel CW, Kelly FL, Petrovski S, Durheim MT, Bush E, Snyder L, Goldstein DB, Todd JL, Palmer SM. Lung Transplant Outcomes in Patients With Pulmonary Fibrosis With Telomere-Related Gene Variants. Chest 2019; 156:477-485. [DOI: 10.1016/j.chest.2019.03.030] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 01/10/2019] [Accepted: 03/01/2019] [Indexed: 01/21/2023] Open
|
209
|
Shaban MM, Elhefny RA, Hussein SH, Badr AA, Nour ZA. Role of telomerase expression in interstitial lung diseases. THE EGYPTIAN JOURNAL OF BRONCHOLOGY 2019. [DOI: 10.4103/ejb.ejb_71_18] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
|
210
|
Morisset J, Lee JS. New trajectories in the treatment of interstitial lung disease: treat the disease or treat the underlying pattern? Curr Opin Pulm Med 2019; 25:442-449. [PMID: 31365378 PMCID: PMC11008767 DOI: 10.1097/mcp.0000000000000600] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
PURPOSE OF REVIEW A subset of patients with interstitial lung diseases (ILDs), such as rheumatoid arthritis (RA)-associated ILD and chronic hypersensitivity pneumonitis, will experience a disease course similar to patients with idiopathic pulmonary fibrosis (IPF). They also often have a usual interstitial pneumonia (UIP) pattern of fibrosis. Although the standard of care for patients with RA-ILD and chronic hypersensitivity pneumonitis is immunosuppression, the optimal treatment for patients with progressive disease and a UIP pattern remains unknown. RECENT FINDINGS Recent research has highlighted shared risk factors, disease behavior and pathobiology between RA-ILD, chronic hypersensitivity pneumonitis and IPF. The presence of a UIP pattern, in both RA-ILD and chronic hypersensitivity pneumonitis, is associated with a worse prognosis. Moreover, genetic risk factors, previously well characterized in IPF, are increasingly being linked to RA-ILD and chronic hypersensitivity pneumonitis. The MUC5B promoter variant rs5705950, telomerase complex mutations and short telomere lengths are also linked to an increased susceptibility to pulmonary fibrosis in RA and chronic hypersensitivity pneumonitis. SUMMARY IPF shares several clinical, genetic and biological features with other ILDs exhibiting the UIP pattern. The optimal pharmacologic management of these patients remains uncertain. Several ongoing trials are evaluating the efficacy of antifibrotic medications in these other diagnoses and may change how we approach ILD treatment.
Collapse
Affiliation(s)
- Julie Morisset
- Département de Médecine, Centre Hospitalier de l’Université de Montréal, Québec, Canada
| | - Joyce S Lee
- Department of Medicine, University of Colorado, Denver, Colorado, USA
| |
Collapse
|
211
|
Bilgili H, Białas AJ, Górski P, Piotrowski WJ. Telomere Abnormalities in the Pathobiology of Idiopathic Pulmonary Fibrosis. J Clin Med 2019; 8:jcm8081232. [PMID: 31426295 PMCID: PMC6723768 DOI: 10.3390/jcm8081232] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 08/12/2019] [Accepted: 08/13/2019] [Indexed: 12/14/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) occurs primarily in older adults and the incidence is clearly associated with aging. This disease seems to be associated with several hallmarks of aging, including telomere attrition and cellular senescence. Increasing evidence suggests that abnormalities involving telomeres and their proteome play a significant role in the pathobiology of IPF. The aim of this study is to summarize present knowledge in the field, as well as to discuss its possible clinical implications. Numerous mutations in genes associated with telomere functioning were studied in the context of IPF, mainly for Telomerase Reverse Transcriptase (TERT) and Telomerase RNA Component (TERC). Such mutations may lead to telomere shortening, which seems to increase the risk of IPF, negatively influence disease progression, and contribute to worse prognosis after lung transplantation. Some evidence indicates the possibility for the use of telomerase activators as potential therapeutic agents in pulmonary fibrosis. To sum up, increasing evidence suggests the role of telomere abnormalities in the pathobiology of IPF, natural history and prognosis of the disease. There are also possibilities for telomerase targeting in the potential development of new treatment agents. However, all these aspects require further research.
Collapse
Affiliation(s)
- Hasancan Bilgili
- Department of Pneumology and Allergy, Medical University of Lodz, 90-154 Lodz, Poland
| | - Adam J Białas
- Department of Pneumology and Allergy, Medical University of Lodz, 90-154 Lodz, Poland.
| | - Paweł Górski
- Department of Pneumology and Allergy, Medical University of Lodz, 90-154 Lodz, Poland
| | - Wojciech J Piotrowski
- Department of Pneumology and Allergy, Medical University of Lodz, 90-154 Lodz, Poland
| |
Collapse
|
212
|
Newton CA, Zhang D, Oldham JM, Kozlitina J, Ma SF, Martinez FJ, Raghu G, Noth I, Garcia CK. Telomere Length and Use of Immunosuppressive Medications in Idiopathic Pulmonary Fibrosis. Am J Respir Crit Care Med 2019; 200:336-347. [PMID: 30566847 PMCID: PMC6680304 DOI: 10.1164/rccm.201809-1646oc] [Citation(s) in RCA: 98] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 12/18/2018] [Indexed: 12/17/2022] Open
Abstract
Rationale: Immunosuppression was associated with adverse events for patients with idiopathic pulmonary fibrosis (IPF) in the PANTHER-IPF (Evaluating the Effectiveness of Prednisone, Azathioprine and N-Acetylcysteine in Patients with IPF) clinical trial. The reason why some patients with IPF experience harm is unknown.Objectives: To determine whether age-adjusted leukocyte telomere length (LTL) was associated with the harmful effect of immunosuppression in patients with IPF.Methods: LTL was measured from available DNA samples from PANTHER-IPF (interim analysis, n = 79; final analysis, n = 118). Replication cohorts included ACE-IPF (Anticoagulant Effectiveness in Idiopathic Pulmonary Fibrosis) (n = 101) and an independent observational cohort (University of Texas Southwestern Medical Center-IPF, n = 170). LTL-stratified and medication-stratified survival analyses were performed using multivariable Cox regression models for composite endpoint-free survival.Measurements and Main Results: Of the subjects enrolled in the PANTHER-IPF and ACE-IPF, 62% (49/79) and 56% (28/50) had an LTL less than the 10th percentile of normal, respectively. In PANTHER-IPF, exposure to prednisone/azathioprine/N-acetylcysteine was associated with a higher composite endpoint of death, lung transplantation, hospitalization, or FVC decline for those with an LTL less than the 10th percentile (hazard ratio, 2.84; 95% confidence interval, 1.02-7.87; P = 0.045). This finding was replicated in the placebo arm of ACE-IPF for those exposed to immunosuppression (hazard ratio, 7.18; 95% confidence interval, 1.52-33.84; P = 0.013). A propensity-matched University of Texas Southwestern Medical Center IPF cohort showed a similar association between immunosuppression and composite endpoints (death, lung transplantation, or FVC decline) for those with an LTL less than the 10th percentile (hazard ratio, 3.79; 95% confidence interval, 1.73-8.30; P = 0.00085). An interaction was found between immunosuppression and LTL for the combined PANTHER-IPF and ACE-IPF clinical trials (Pinteraction = 0.048), and the University of Texas Southwestern Medical Center IPF cohort (Pinteraction = 0.00049).Conclusions: LTL is a biomarker that may identify patients with IPF at risk for poor outcomes when exposed to immunosuppression.
Collapse
Affiliation(s)
| | - David Zhang
- Department of Medicine and
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Justin M. Oldham
- Department of Medicine, University of California at Davis, Davis, California
| | - Julia Kozlitina
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Shwu-Fan Ma
- Department of Medicine, University of Virginia, Charlottesville, Virginia
| | | | - Ganesh Raghu
- Department of Medicine, University of Washington Medical Center, Seattle, Washington
| | - Imre Noth
- Department of Medicine, University of Virginia, Charlottesville, Virginia
| | - Christine Kim Garcia
- Department of Medicine and
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
213
|
Molina-Molina M. Telomere Shortening Is behind the Harm of Immunosuppressive Therapy in Idiopathic Pulmonary Fibrosis. Am J Respir Crit Care Med 2019; 200:274-275. [PMID: 30624965 PMCID: PMC6680301 DOI: 10.1164/rccm.201812-2330ed] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Affiliation(s)
- Maria Molina-Molina
- 1Respiratory DepartmentBellvitge University Hospital-Bellvitge Institute for Biomedical ResearchBarcelona, Spainand
- 2CIBER of Respiratory DiseasesMadrid, Spain
| |
Collapse
|
214
|
Krauss E, Gehrken G, Drakopanagiotakis F, Tello S, Dartsch RC, Maurer O, Windhorst A, von der Beck D, Griese M, Seeger W, Guenther A. Clinical characteristics of patients with familial idiopathic pulmonary fibrosis (f-IPF). BMC Pulm Med 2019; 19:130. [PMID: 31319833 PMCID: PMC6637501 DOI: 10.1186/s12890-019-0895-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 07/11/2019] [Indexed: 11/29/2022] Open
Abstract
Background The aim of this study was to analyze the relative frequency, clinical characteristics, disease onset and progression in f-IPF vs. sporadic IPF (s-IPF). Methods Familial IPF index patients and their family members were recruited into the European IPF registry/biobank (eurIPFreg) at the Universities of Giessen and Marburg (UGMLC). Initially, we employed wide range criteria of f-IPF (e.g. relatives who presumably died of some kind of parenchymal lung disease). After narrowing down the search to occurrence of idiopathic interstitial pneumonia (IIP) in at least one first grade relative, 28 index patients were finally identified, prospectively interviewed and examined. Their family members were phenotyped with establishment of pedigree charts. Results Within the 28 IPF families, overall 79 patients with f-IPF were identified. In the same observation period, 286 f-IIP and s-IIP patients were recruited into the eurIPFreg at our UGMLC sites, corresponding to a familial versus s-IPF of 9.8%. The both groups showed no difference in demographics (61 vs. 79% males), smoking history, and exposure to any environmental triggers known to cause lung fibrosis. The f-IPF group differed by an earlier age at the onset of the disease (55.4 vs. 63.2 years; p < 0.001). On average, the f-IPF patients presented a significantly milder extent of functional impairment at the time point of inclusion vs. the s-IPF group (FVC 75% pred. vs. FVC 62% pred., p = 0.011). In contrast, the decline in FVC was found to be faster in the f-IPF vs. the s-IPF group (4.94% decline in 6 months in f-IPF vs. 2.48% in s-IPF, p = 0.12). The average age of death in f-IPF group was 67 years vs. 71.8 years in s-IPF group (p = 0.059). The f-IIP group displayed diverse inheritance patterns, mostly autosomal-dominant with variable penetrance. In the f-IPF, the younger generations showed a tendency for earlier manifestation of IPF vs. the older generation (58 vs. 66 years, p = 0.013). Conclusions The 28 f-IPF index patients presented an earlier onset and more aggressive natural course of the disease. The disease seems to affect consecutive generations at a younger age. Trial registration Nr. NCT02951416http://www.www.clinicaltrials.gov
Collapse
Affiliation(s)
- Ekaterina Krauss
- Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Universities of Giessen and Marburg Lung Center (UGMLC), European IPF Registry (eurIPFreg), Klinikstrasse 36, 35392, Giessen, Germany.,European IPF Registry & Biobank (eurIPFreg), Giessen, Germany
| | - Godja Gehrken
- Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Universities of Giessen and Marburg Lung Center (UGMLC), European IPF Registry (eurIPFreg), Klinikstrasse 36, 35392, Giessen, Germany.,European IPF Registry & Biobank (eurIPFreg), Giessen, Germany
| | - Fotios Drakopanagiotakis
- Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Universities of Giessen and Marburg Lung Center (UGMLC), European IPF Registry (eurIPFreg), Klinikstrasse 36, 35392, Giessen, Germany.,European IPF Registry & Biobank (eurIPFreg), Giessen, Germany
| | - Silke Tello
- Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Universities of Giessen and Marburg Lung Center (UGMLC), European IPF Registry (eurIPFreg), Klinikstrasse 36, 35392, Giessen, Germany.,European IPF Registry & Biobank (eurIPFreg), Giessen, Germany
| | - Ruth C Dartsch
- Agaplesion Lung Clinic Waldhof-Elgershausen, Greifenstein, Germany
| | - Olga Maurer
- Agaplesion Lung Clinic Waldhof-Elgershausen, Greifenstein, Germany
| | - Anita Windhorst
- Department of Medical Statistics, Justus-Liebig-University of Giessen, Giessen, Germany
| | - Daniel von der Beck
- Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Universities of Giessen and Marburg Lung Center (UGMLC), European IPF Registry (eurIPFreg), Klinikstrasse 36, 35392, Giessen, Germany.,European IPF Registry & Biobank (eurIPFreg), Giessen, Germany
| | - Matthias Griese
- Children University Hospital, Campus Hauner, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Werner Seeger
- Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Universities of Giessen and Marburg Lung Center (UGMLC), European IPF Registry (eurIPFreg), Klinikstrasse 36, 35392, Giessen, Germany.,European IPF Registry & Biobank (eurIPFreg), Giessen, Germany.,Cardio-Pulmonary Institute, Giessen, Germany
| | - Andreas Guenther
- Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Universities of Giessen and Marburg Lung Center (UGMLC), European IPF Registry (eurIPFreg), Klinikstrasse 36, 35392, Giessen, Germany. .,European IPF Registry & Biobank (eurIPFreg), Giessen, Germany. .,Cardio-Pulmonary Institute, Giessen, Germany. .,Agaplesion Lung Clinic Waldhof-Elgershausen, Greifenstein, Germany.
| |
Collapse
|
215
|
Tanaka T, Ishida K. Update on Rare Idiopathic Interstitial Pneumonias and Rare Histologic Patterns. Arch Pathol Lab Med 2019; 142:1069-1079. [PMID: 30141991 DOI: 10.5858/arpa.2017-0534-ra] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
CONTEXT - In 2013, the revised American Thoracic Society and European Respiratory Society classification of idiopathic interstitial pneumonias (IIPs) described 2 rare IIPs and 2 rare histologic patterns. Because of the rarity of the disease, there is limited evidence related to the histology. Because the rare histologic patterns are provisional criteria, no unanimous consensus on histologic diagnostic criteria has yet been reached. OBJECTIVE - To review the histologic features for rare IIPs and rare histologic patterns, and to provide diagnostic aids and discuss the differential diagnosis. DATA SOURCES - Published peer-reviewed literature and the authors' personal experience. CONCLUSIONS - Following the publication of the international consensus classification, evidence regarding rare IIPs and rare histologic patterns has accumulated to some extent, although to date the amount remains insufficient and further evidence is required. Because the diagnosis is sometimes challenging, a multidisciplinary approach represents the gold standard in reaching an accurate diagnosis for these rare disorders.
Collapse
Affiliation(s)
| | - Kaori Ishida
- From the Department of Pathology, Faculty of Medicine, Kindai University, Osaka, Japan (Dr Tanaka); and the Department of Pathology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan (Dr Ishida)
| |
Collapse
|
216
|
The Role of Telomerase and Telomeres in Interstitial Lung Diseases: From Molecules to Clinical Implications. Int J Mol Sci 2019; 20:ijms20122996. [PMID: 31248154 PMCID: PMC6627617 DOI: 10.3390/ijms20122996] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 06/12/2019] [Accepted: 06/15/2019] [Indexed: 02/07/2023] Open
Abstract
Telomeres are distal chromosome regions associated with specific protein complexes that protect the chromosome against degradation and aberrations. Telomere maintenance capacity is an essential indication of healthy cell populations, and telomere damage is observed in processes such as malignant transformation, apoptosis, or cell senescence. At a cellular level, telomere damage may result from genotoxic stress, decreased activity of telomerase enzyme complex, dysfunction of shelterin proteins, or changes in expression of telomere-associated RNA such as TERRA. Clinical evidence suggests that mutation of telomerase genes (Tert/Terc) are associated with increased risk of congenital as well as age-related diseases (e.g., pneumonitis, idiopathic pulmonary fibrosis (IPF), dyskeratosis congenita, emphysema, nonspecific interstitial pneumonia, etc.). Thus, telomere length and maintenance can serve as an important prognostic factor as well as a potential target for new strategies of treatment for interstitial lung diseases (ILDs) and associated pulmonary pathologies.
Collapse
|
217
|
Abstract
PURPOSE OF REVIEW Telomere attrition has been proposed as one of the aging hallmarks in pulmonary fibrosis. Telomere shortening and telomerase gene mutations have been widely evaluated in recent years. Reduced telomere length may be identified in a quarter of patients with sporadic idiopathic pulmonary fibrosis (IPF) and half of those cases with family aggregation. However, telomere studies have not transferred from the research field to the clinic. This review is focused on our current understanding of the pathogenic implication of telomere dysfunction in lung fibrosis and its relevance in the clinical setting. RECENT FINDINGS The most prevalent clinical expression of telomere dysfunction is IPF. Disease onset is usually seen at a younger age and family aggregation is frequently present. Short telomere syndrome is associated in a minority of cases and includes premature hair greying, bone marrow failure and liver cirrhosis. However, patients often present with some extrapulmonary associated telomeric features and related comorbidities that may help to suspect telomere defects. Telomere shortening confers a poor prognosis and reduced lung-transplant free survival time in IPF and other nonidiopathic pulmonary fibrotic entities. SUMMARY Telomere dysfunction associates some common clinical features that could modify patient management in pulmonary fibrosis.
Collapse
|
218
|
Barros A, Oldham J, Noth I. Genetics of Idiopathic Pulmonary Fibrosis. Am J Med Sci 2019; 357:379-383. [PMID: 31010464 PMCID: PMC10538522 DOI: 10.1016/j.amjms.2019.02.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 01/28/2019] [Accepted: 02/10/2019] [Indexed: 01/02/2023]
Abstract
Idiopathic pulmonary fibrosis (IPF) is the most common form of interstitial lung disease. IPF is a complex disease, with environmental and genetic factors variably contributing to disease susceptibility and outcomes. A host of common gene variants with modest effect size impart disease risk in patients with sporadic IPF, while rare variants with large effect size influence disease risk in those with familial interstitial pneumonia. In this review, we highlight several common and rare variants underpinning IPF risk and call attention to recently published studies informing our understanding of this risk.
Collapse
Affiliation(s)
- Andrew Barros
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Virginia Health System, Charlottesville, Virginia.
| | - Justin Oldham
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, The University of California at Davis, Sacramento, California.
| | - Imre Noth
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Virginia Health System, Charlottesville, Virginia.
| |
Collapse
|
219
|
Tanizawa K, Ley B, Vittinghoff E, Elicker BM, Henry TS, Wolters PJ, Brownell R, Liu S, Collard HR, Jones KD. Significance of bronchiolocentric fibrosis in patients with histopathological usual interstitial pneumonia. Histopathology 2019; 74:1088-1097. [PMID: 30742318 DOI: 10.1111/his.13840] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 02/07/2019] [Indexed: 01/01/2023]
Abstract
AIMS To evaluate the clinical significance of bronchiolocentric fibrosis (BCF) in patients with a histopathological pattern of usual interstitial pneumonia (UIP). METHODS AND RESULTS Two hundred and fifty-two patients with pathological UIP pattern were identified. Two hundred and fifteen of these patients (215 of 252) had the multidisciplinary diagnosis of idiopathic pulmonary fibrosis (IPF). Prospectively defined clinical, radiological and pathological features (including BCF) were recorded, and peripheral blood MUC5B genotype and telomere length were measured. BCF was observed in 38% (96 of 252) of all patients and 33% (72 of 215) of IPF patients; its presence was associated with a non-IPF diagnosis on multivariate analysis (odds ratio = 3.71, 95% confidence interval = 1.68-8.19). BCF was not significantly associated with environmental exposures, gastroesophageal reflux, cigarette smoking or radiological patterns. There was no significant association of BCF with MUC5B genotype or telomere length. BCF has no significant impact on survival time. CONCLUSIONS Most patients with BCF and a histopathological pattern of UIP have IPF. However, this combined fibrotic pattern is associated with a non-IPF multidisciplinary diagnosis, with approximately one-quarter of these patients being diagnosed as chronic hypersensitivity pneumonia or unclassifiable interstitial fibrosis. The presence of BCF in these patients is not significantly associated with presumed clinical risk factors for bronchiolocentric involvement, radiological findings, MUC5B genotype, telomere length or survival time.
Collapse
Affiliation(s)
- Kiminobu Tanizawa
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA.,Kyoto Central Clinic, Clinical Research Center, Kyoto, Japan
| | - Brett Ley
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Eric Vittinghoff
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA
| | - Brett M Elicker
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Travis S Henry
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Paul J Wolters
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Robert Brownell
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Shuo Liu
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA.,Department of Respiratory Medicine, The Fourth Affiliated Hospital of China Medical University, Shenyang, People's Republic of China
| | - Harold R Collard
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Kirk D Jones
- Department of Pathology, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
220
|
Newton CA, Oldham JM, Ley B, Anand V, Adegunsoye A, Liu G, Batra K, Torrealba J, Kozlitina J, Glazer C, Strek ME, Wolters PJ, Noth I, Garcia CK. Telomere length and genetic variant associations with interstitial lung disease progression and survival. Eur Respir J 2019; 53:13993003.01641-2018. [PMID: 30635297 DOI: 10.1183/13993003.01641-2018] [Citation(s) in RCA: 120] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 12/27/2018] [Indexed: 01/08/2023]
Abstract
Leukocyte telomere length (LTL), MUC5B rs35705950 and TOLLIP rs5743890 have been associated with idiopathic pulmonary fibrosis (IPF).In this observational cohort study, we assessed the associations between these genomic markers and outcomes of survival and rate of disease progression in patients with interstitial pneumonia with autoimmune features (IPAF, n=250) and connective tissue disease-associated interstitial lung disease (CTD-ILD, n=248). IPF (n=499) was used as a comparator.The LTL of IPAF and CTD-ILD patients (mean age-adjusted log-transformed T/S of -0.05±0.29 and -0.04±0.25, respectively) is longer than that of IPF patients (-0.17±0.32). For IPAF patients, LTL <10th percentile is associated with faster lung function decline compared to LTL ≥10th percentile (-6.43% per year versus -0.86% per year; p<0.0001) and worse transplant-free survival (hazard ratio 2.97, 95% CI 1.70-5.20; p=0.00014). The MUC5B rs35705950 minor allele frequency (MAF) is greater for IPAF patients (23.2, 95% CI 18.8-28.2; p<0.0001) than controls and is associated with worse transplant-free IPAF survival (hazard ratio 1.92, 95% CI 1.18-3.13; p=0.0091). Rheumatoid arthritis (RA)-associated ILD (RA-ILD) has a shorter LTL than non-RA CTD-ILD (-0.14±0.27 versus -0.01±0.23; p=0.00055) and higher MUC5B MAF (34.6, 95% CI 24.4-46.3 versus 14.1, 95% CI 9.8-20.0; p=0.00025). Neither LTL nor MUC5B are associated with transplant-free CTD-ILD survival.LTL and MUC5B MAF have different associations with lung function progression and survival for IPAF and CTD-ILD.
Collapse
Affiliation(s)
- Chad A Newton
- Dept of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Justin M Oldham
- Dept of Internal Medicine, University of California at Davis, Davis, CA, USA
| | - Brett Ley
- Dept of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Vikram Anand
- Dept of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | | | - Gabrielle Liu
- Dept of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Kiran Batra
- Dept of Radiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jose Torrealba
- Dept of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Julia Kozlitina
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Craig Glazer
- Dept of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Mary E Strek
- Dept of Medicine, University of Chicago, Chicago, IL, USA
| | - Paul J Wolters
- Dept of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Imre Noth
- Dept of Medicine, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Christine Kim Garcia
- Dept of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
221
|
Abstract
PURPOSE OF REVIEW Genomic mutations in telomere-related genes have been recognized as a cause of familial forms of idiopathic pulmonary fibrosis (IPF). However, it has become increasingly clear that telomere syndromes and telomere shortening are associated with various types of pulmonary disease. Additionally, it was found that also single nucleotide polymorphisms (SNPs) in telomere-related genes are risk factors for the development of pulmonary disease. This review focuses on recent updates on pulmonary phenotypes associated with genetic variation in telomere-related genes. RECENT FINDINGS Genomic mutations in seven telomere-related genes cause pulmonary disease. Pulmonary phenotypes associated with these mutations range from many forms of pulmonary fibrosis to emphysema and pulmonary vascular disease. Telomere-related mutations account for up to 10% of sporadic IPF, 25% of familial IPF, 10% of connective-tissue disease-associated interstitial lung disease, and 1% of COPD. Mixed disease forms have also been found. Furthermore, SNPs in TERT, TERC, OBFC1, and RTEL1, as well as short telomere length, have been associated with several pulmonary diseases. Treatment of pulmonary disease caused by telomere-related gene variation is currently based on disease diagnosis and not on the underlying cause. SUMMARY Pulmonary phenotypes found in carriers of telomere-related gene mutations and SNPs are primarily pulmonary fibrosis, sometimes emphysema and rarely pulmonary vascular disease. Genotype-phenotype relations are weak, suggesting that environmental factors and genetic background of patients determine disease phenotypes to a large degree. A disease model is presented wherever genomic variation in telomere-related genes cause specific pulmonary disease phenotypes whenever triggered by environmental exposure, comorbidity, or unknown factors.
Collapse
|
222
|
Borie R, Bouvry D, Cottin V, Gauvain C, Cazes A, Debray MP, Cadranel J, Dieude P, Degot T, Dominique S, Gamez AS, Jaillet M, Juge PA, Londono-Vallejo A, Mailleux A, Mal H, Boileau C, Menard C, Nunes H, Prevot G, Quetant S, Revy P, Traclet J, Wemeau-Stervinou L, Wislez M, Kannengiesser C, Crestani B. Regulator of telomere length 1 ( RTEL1) mutations are associated with heterogeneous pulmonary and extra-pulmonary phenotypes. Eur Respir J 2019; 53:13993003.00508-2018. [PMID: 30523160 DOI: 10.1183/13993003.00508-2018] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 11/06/2018] [Indexed: 01/10/2023]
Abstract
Regulator of telomere length 1 (RTEL1) mutations have been evidenced in 5-9% of familial pulmonary fibrosis; however, the phenotype of patients with interstitial lung disease (ILD) and RTEL1 mutations is poorly understood.Whole exome sequencing was performed in 252 probands with ILD and we included all patients with ILD and RTEL1 mutation. RTEL1 expression was evaluated by immunochemistry in the lungs of controls, as well as in RTEL1 and telomerase reverse transcriptase (TERT) mutation carriers.We identified 35 subjects from 17 families. Median age at diagnosis of ILD was 53.1 years (range 28.0-80.6). The most frequent pulmonary diagnoses were idiopathic pulmonary fibrosis (n=20, 57%), secondary ILD (n=7, 20%) and unclassifiable fibrosis or interstitial pneumonia with autoimmune features (n=7, 20%). The median transplant-free and overall survival periods were 39.2 months and 45.3 months, respectively. Forced vital capacity at diagnosis was the only factor associated with decreased transplant-free survival. Extra-pulmonary manifestations were less frequent as compared to other telomere-related gene mutation carriers. A systematic analysis of the literature identified 110 patients with ILD and RTEL1 mutations (including this series) and confirmed the heterogeneity of the pulmonary phenotype, the prevalence of non-idiopathic diseases and the low prevalence of extra-pulmonary manifestations.Immunohistochemistry showed that RTEL1 was expressed by bronchial and alveolar epithelial cells, as well as by alveolar macrophages and lymphocytes, but not by fibroblasts.
Collapse
Affiliation(s)
- Raphael Borie
- Service de Pneumologie A, Hôpital Bichat, AP-HP, DHU FIRE, Paris, France.,Unité 1152, INSERM, Université Paris Diderot, Paris, France
| | - Diane Bouvry
- Service de Pneumologie, Hôpital Avicenne, AP-HP, Bobigny, France
| | - Vincent Cottin
- Service de Pneumologie, Hôpital Louis Pradel, Université Claude Bernard Lyon 1, Lyon, France
| | | | - Aurélie Cazes
- Unité 1152, INSERM, Université Paris Diderot, Paris, France.,Service d'Anatomopathologie, Hôpital Bichat, AP-HP, Paris, France
| | - Marie-Pierre Debray
- Unité 1152, INSERM, Université Paris Diderot, Paris, France.,Service de Radiologie, Hôpital Bichat, AP-HP, Paris, France
| | | | - Philippe Dieude
- Unité 1152, INSERM, Université Paris Diderot, Paris, France.,Service de Rhumatologie, Hôpital Bichat, AP-HP, Paris, France.,Université Paris Diderot, Paris, France
| | - Tristan Degot
- Service de Pneumologie, CHU Strasbourg, Strasbourg, France
| | | | | | | | | | - Arturo Londono-Vallejo
- UMR 3244 (Telomere and Cancer Lab), CNRS, Institut Curie, PSL Research University, Sorbonne Universités, Paris, France
| | | | - Hervé Mal
- Unité 1152, INSERM, Université Paris Diderot, Paris, France.,Service de Pneumologie B, Hôpital Bichat, AP-HP, Paris, France
| | - Catherine Boileau
- Université Paris Diderot, Paris, France.,Laboratoire de Génétique, Hôpital Bichat, AP-HP, Paris, France
| | | | - Hilario Nunes
- Service de Pneumologie, Hôpital Avicenne, AP-HP, Bobigny, France
| | | | | | - Patrick Revy
- UMR 1163 (Laboratory of Genome Dynamics in the Immune System), INSERM, Imagine Institute, Paris Descartes-Sorbonne Paris Cité University, Paris, France
| | - Julie Traclet
- Service de Pneumologie, Hôpital Tenon, AP-HP, Paris, France
| | - Lidwine Wemeau-Stervinou
- Service de Pneumologie, Centre de Compétence des Maladies Pulmonaires Rares, CHRU de Lille, Lille, France
| | - Marie Wislez
- Service de Pneumologie, Unité d'Oncologie Thoracique, Hôpital Cochin, Hôpitaux Universitaires Paris Centre, AP-HP, Paris, France
| | - Caroline Kannengiesser
- Unité 1152, INSERM, Université Paris Diderot, Paris, France.,Université Paris Diderot, Paris, France.,Laboratoire de Génétique, Hôpital Bichat, AP-HP, Paris, France
| | - Bruno Crestani
- Service de Pneumologie A, Hôpital Bichat, AP-HP, DHU FIRE, Paris, France.,Unité 1152, INSERM, Université Paris Diderot, Paris, France.,Université Paris Diderot, Paris, France
| |
Collapse
|
223
|
Courtwright AM, El-Chemaly S. Telomeres in Interstitial Lung Disease: The Short and the Long of It. Ann Am Thorac Soc 2019; 16:175-181. [PMID: 30540921 PMCID: PMC6376948 DOI: 10.1513/annalsats.201808-508cme] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 09/25/2018] [Indexed: 01/01/2023] Open
Abstract
Telomeres are repetitive nucleotide sequences that cap linear chromosomes, thereby limiting progressive chromosomal shortening during cell replication. In conjunction with environmental factors, common single-nucleotide polymorphisms and rare and ultra-rare telomere-related mutations are associated with accelerated telomere shortening resulting in organ dysfunction, including interstitial lung disease (ILD). The most common telomere-related mutation-associated ILD is idiopathic pulmonary fibrosis (IPF). Up to one-third of individuals with familial IPF have shortened telomeres and/or carry a telomere-related mutation, and 1 in 10 individuals with sporadic IPF have telomere-related mutations. Regardless of ILD phenotype, individuals with short telomeres and/or known telomere-related mutations have more rapid disease progression and shorter lung transplant-free survival. Management should include initiation of antifibrotic agents for those with an IPF phenotype and early referral to a transplant center. Patients with ILD being considered for transplant should be screened for short telomeres if there is a significant family history of pulmonary fibrosis or evidence of extrapulmonary organ dysfunction associated with a short telomere syndrome. Post-transplant management of recipients with telomere-related mutations should include careful adjustment of immunosuppression regimens on the basis of bone marrow reserve. Data on the impact of shortened telomeres on post-transplant outcomes, however, remain mixed.
Collapse
Affiliation(s)
- Andrew M. Courtwright
- Division of Pulmonary and Critical Care Medicine, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania; and
| | - Souheil El-Chemaly
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
| |
Collapse
|
224
|
Abstract
More than 100 different conditions are grouped under the term interstitial lung disease (ILD). A diagnosis of an ILD primarily relies on a combination of clinical, radiological, and pathological criteria, which should be evaluated by a multidisciplinary team of specialists. Multiple factors, such as environmental and occupational exposures, infections, drugs, radiation, and genetic predisposition have been implicated in the pathogenesis of these conditions. Asbestosis and other pneumoconiosis, hypersensitivity pneumonitis (HP), chronic beryllium disease, and smoking-related ILD are specifically linked to inhalational exposure of environmental agents. The recent Global Burden of Disease Study reported that ILD rank 40th in relation to global years of life lost in 2013, which represents an increase of 86% compared to 1990. Idiopathic pulmonary fibrosis (IPF) is the prototype of fibrotic ILD. A recent study from the United States reported that the incidence and prevalence of IPF are 14.6 per 100,000 person-years and 58.7 per 100,000 persons, respectively. These data suggests that, in large populated areas such as Brazil, Russia, India, and China (the BRIC region), there may be approximately 2 million people living with IPF. However, studies from South America found much lower rates (0.4–1.2 cases per 100,000 per year). Limited access to high-resolution computed tomography and spirometry or to multidisciplinary teams for accurate diagnosis and optimal treatment are common challenges to the management of ILD in developing countries.
Collapse
|
225
|
Integrating Genomics Into Management of Fibrotic Interstitial Lung Disease. Chest 2019; 155:1026-1040. [PMID: 30660786 DOI: 10.1016/j.chest.2018.12.011] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 11/30/2018] [Accepted: 12/20/2018] [Indexed: 12/31/2022] Open
Abstract
Fibrotic interstitial lung diseases (ILDs) have a high mortality rate with an unpredictable disease course and clinical features that frequently overlap. Recent data indicate important roles for genomics in the mechanisms underlying susceptibility and progression of pulmonary fibrosis. The impact of these genomic markers on pharmacotherapy and their contribution to outcomes is increasingly recognized. Interstitial lung abnormalities, frequently considered representative of early ILD, have been consistently associated with the MUC5B promoter polymorphism, a common gene variant. Other rare gene variant mutations, including TERT, TERC, SFTPC, and DKC1, may be present in patients with familial interstitial pneumonia and are frequently associated with a usual interstitial pneumonia pattern of fibrosis. The minor allele of the MUC5B rs35705950 genotype is prevalent in several sporadic forms of ILD, including idiopathic pulmonary fibrosis and chronic hypersensitivity pneumonitis. Gene mutations that characterize familial pulmonary fibrosis may be present in patients with connective tissue disease-related ILD, such as rheumatoid arthritis-ILD. Additionally, shorter telomere lengths and mutations in telomere biology-related genes have been demonstrated in both familial and sporadic ILD, with significant implications for disease progression, lung function, and survival. An improved understanding of the impact of genetic and genomic risk factors on disease progression would better guide personalized therapeutic choices in persons with fibrotic ILD.
Collapse
|
226
|
Meyer KC. Classification and Nomenclature of Interstitial Lung Disease. Respir Med 2019. [DOI: 10.1007/978-3-319-99975-3_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
227
|
|
228
|
Inchingolo R, Varone F, Sgalla G, Richeldi L. Existing and emerging biomarkers for disease progression in idiopathic pulmonary fibrosis. Expert Rev Respir Med 2018; 13:39-51. [DOI: 10.1080/17476348.2019.1553620] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Riccardo Inchingolo
- Pulmonary Medicine Unit, Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Francesco Varone
- Pulmonary Medicine Unit, Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Giacomo Sgalla
- Pulmonary Medicine Unit, Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Luca Richeldi
- Pulmonary Medicine Unit, Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| |
Collapse
|
229
|
Planas-Cerezales L, Arias-Salgado EG, Buendia-Roldán I, Montes-Worboys A, López CE, Vicens-Zygmunt V, Hernaiz PL, Sanuy RL, Leiro-Fernandez V, Vilarnau EB, Llinás ES, Sargatal JD, Abellón RP, Selman M, Molina-Molina M. Predictive factors and prognostic effect of telomere shortening in pulmonary fibrosis. Respirology 2018; 24:146-153. [PMID: 30320420 DOI: 10.1111/resp.13423] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Revised: 09/11/2018] [Accepted: 09/12/2018] [Indexed: 01/04/2023]
Abstract
BACKGROUND AND OBJECTIVE The abnormal shortening of telomeres is a mechanism linking ageing to idiopathic pulmonary fibrosis (IPF) that could be useful in the clinical setting. The objective of this study was to identify the IPF patients with higher risk for telomere shortening and to investigate the outcome implications. METHODS Consecutive Spanish patients were included at diagnosis and followed up for 3 years. DNA blood samples from a Mexican cohort were used to validate the results found in Spanish sporadic IPF. Prior to treatment, telomere length was measured through quantitative polymerase chain reaction (qPCR) and Southern blot. Outcome was assessed according to mortality or need for lung transplantation. A multivariate regression logistic model was used for statistical analysis. RESULTS Family aggregation, age of <60 years and the presence of non-specific immunological or haematological abnormalities were associated with a higher probability of telomere shortening. Overall, 66.6% of patients younger than 60 years with telomere shortening died or required lung transplantation, independent of functional impairment at diagnosis. By contrast, in patients older than 60 years with telomere shortening, the negative impact of telomere shortening in outcome was not significant. CONCLUSION Our data indicate that young sporadic IPF patients (<60 years) with some non-specific immunological or haematological abnormalities had higher risk of telomere shortening, and furthermore, they presented a poorer prognosis.
Collapse
Affiliation(s)
- Lurdes Planas-Cerezales
- Centro de Investigación en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain.,Unidad Funcional de Intersticio Pulmonar, Servicio Neumología, Hospital Universitario de Bellvitge, Instituto de Investigación Biomédica de Bellvitge (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
| | - Elena G Arias-Salgado
- Advanced Medical Projects, Madrid, Spain.,Instituto de Investigaciones Biomédicas, Consejo Superior de Investigaciones Científicas (CSIC)/Universidad Autónoma de Madrid (UAM), Centro de Investigación en Red de Enfermedades Raras (CIBERER), Madrid, Spain
| | - Ivette Buendia-Roldán
- Instituto Nacional de Enfermedades Respiratorias, "Ismael Cosío Villegas", México City, Mexico
| | - Ana Montes-Worboys
- Centro de Investigación en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain.,Unidad Funcional de Intersticio Pulmonar, Servicio Neumología, Hospital Universitario de Bellvitge, Instituto de Investigación Biomédica de Bellvitge (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
| | - Cristina Esquinas López
- Centro de Investigación en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain.,Servicio de Neumología, Hospital Vall d'Hebron, Barcelona, Spain
| | - Vanesa Vicens-Zygmunt
- Unidad Funcional de Intersticio Pulmonar, Servicio Neumología, Hospital Universitario de Bellvitge, Instituto de Investigación Biomédica de Bellvitge (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
| | - Patricio Luburich Hernaiz
- Unidad Funcional de Intersticio Pulmonar. Servicio Radiodiagnóstico, Hospital Universitario de Bellvitge, Instituto de Investigación Biomédica de Bellvitge (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
| | - Roger Llatjós Sanuy
- Unidad Funcional de Intersticio Pulmonar, Servicio de Anatomía Patológica, Hospital Universitario de Bellvitge, Instituto de Investigación Biomédica de Bellvitge (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
| | - Virginia Leiro-Fernandez
- Servicio de Neumología, Complexo Hospitalario Universitario de Vigo (CHUVI), Vigo, Spain.,Grupo de Investigación en Respiratorio, Instituto de Investigación Biomédica de Vigo, Vigo, Spain
| | - Eva Balcells Vilarnau
- Centro de Investigación en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain.,Servicio de Neumología, Hospital del Mar, Instituto Hospital del Mar de Investigaciones Médicas (IMIM), Universidad Pompeu Fabra (UPF), Barcelona, Spain
| | - Ernest Sala Llinás
- Centro de Investigación en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain.,Servicio de Neumología, Hospital Son Espases, Instituto de Investigación Sanitaria Islas Baleares (IdISBa), Palma de Mallorca, Spain
| | - Jordi Dorca Sargatal
- Centro de Investigación en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain.,Unidad Funcional de Intersticio Pulmonar, Servicio Neumología, Hospital Universitario de Bellvitge, Instituto de Investigación Biomédica de Bellvitge (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
| | - Rosario Perona Abellón
- Instituto de Investigaciones Biomédicas, Consejo Superior de Investigaciones Científicas (CSIC)/Universidad Autónoma de Madrid (UAM), Centro de Investigación en Red de Enfermedades Raras (CIBERER), Madrid, Spain
| | - Moisés Selman
- Instituto Nacional de Enfermedades Respiratorias, "Ismael Cosío Villegas", México City, Mexico
| | - Maria Molina-Molina
- Centro de Investigación en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain.,Unidad Funcional de Intersticio Pulmonar, Servicio Neumología, Hospital Universitario de Bellvitge, Instituto de Investigación Biomédica de Bellvitge (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
| |
Collapse
|
230
|
Multiple Solid Organ Transplantation in Telomeropathy: Case Series and Literature Review. Transplantation 2018; 102:1747-1755. [DOI: 10.1097/tp.0000000000002198] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
231
|
A DNA-Methylated Sight on Autoimmune Inflammation Network across RA, pSS, and SLE. J Immunol Res 2018; 2018:4390789. [PMID: 30159339 PMCID: PMC6109517 DOI: 10.1155/2018/4390789] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 07/08/2018] [Indexed: 12/14/2022] Open
Abstract
Methylation variabilities of inflammatory cytokines play important roles in the development of systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), and primary Sjögren's syndrome (pSS). With heightened focus on personalized and precise medicine, it is necessary to compare and contrast the difference and similarity of cytokine methylation status between the 3 most classic autoimmune diseases (AIDs). In this study, we integrated 5 Cytokine-Chips from genome-wide DNA methylation datasets of the 3 kind of AIDs, delta-beta value was calculated for intergroup difference, and comprehensive bioinformatics analyses of cytokine genes with aberrant methylations were performed. 125 shared differential methylation variabilities (DMVs) were identified. There were 102 shared DMVs with similar methylation status; 3 hypomethylated differential methylation regions (DMRs) across the AIDs were found, and all 3 DMRs were hypomethylated. DMRs (AZU1, LTBR, and RTEL1) were likely to serve as activator in the inflammatory process. Particularly, AZU1 and LTBR with hypomethylated TSS and first exon located in the promoter regions were able to trigger inflammation signaling cascades and play critical roles in autoimmune tautology. Moreover, functional epigenetic module (FEM) algorithm showed that different inflammatory networks are involved in different AIDs; 5 hotspots were identified as biologically plausible pathways in inducing or perpetuating of inflammation which are epigenetically deregulated in AIDs. We concluded methylation variabilities among the same cytokines can greatly impact the perpetuation of inflammatory process or signal pathway of AIDs. Differentiating the cytokine methylation status will serve as valuable resource for researchers alike to gain better understanding of the epigenetic mechanisms of the three AIDs. Even more importantly, better understanding of cytokine methylation variability existing between the three classic AIDs will aid in identification of potential epigenetic biomarkers and therapeutic targets. This trial is registered with ChiCTR-INR-16010290, a clinical trial for the treatment of rheumatoid arthritis with Warming yang and Smoothening Meridians.
Collapse
|
232
|
Barratt SL, Creamer A, Hayton C, Chaudhuri N. Idiopathic Pulmonary Fibrosis (IPF): An Overview. J Clin Med 2018; 7:jcm7080201. [PMID: 30082599 PMCID: PMC6111543 DOI: 10.3390/jcm7080201] [Citation(s) in RCA: 236] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 07/23/2018] [Accepted: 07/31/2018] [Indexed: 02/07/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is an interstitial lung disease characterised by chronic, progressive scarring of the lungs and the pathological hallmark of usual interstitial pneumonia. Current paradigms suggest alveolar epithelial cell damage is a key initiating factor. Globally, incidence of the disease is rising, with associated high morbidity, mortality, and economic healthcare burden. Diagnosis relies on a multidisciplinary team approach with exclusion of other causes of interstitial lung disease. Over recent years, two novel antifibrotic therapies, pirfenidone and nintedanib, have been developed, providing treatment options for many patients with IPF, with several other agents in early clinical trials. Current efforts are directed at identifying key biomarkers that may direct more customized patient-centred healthcare to improve outcomes for these patients in the future.
Collapse
Affiliation(s)
- Shaney L Barratt
- Bristol Interstitial Lung Disease Service, North Bristol NHS Trust, Bristol BS10 5NB, UK.
- Academic Respiratory Unit, University of Bristol, Bristol BS16 1QY, UK.
| | - Andrew Creamer
- Bristol Interstitial Lung Disease Service, North Bristol NHS Trust, Bristol BS10 5NB, UK.
| | - Conal Hayton
- North West Interstitial Lung Disease Unit, Manchester University NHS Foundation Trust, Wythenshawe, Manchester M23 9LT, UK.
| | - Nazia Chaudhuri
- North West Interstitial Lung Disease Unit, Manchester University NHS Foundation Trust, Wythenshawe, Manchester M23 9LT, UK.
| |
Collapse
|
233
|
Kiryluk K, Groopman E, Rasouly H, Garcia CK, Gharavi AG. Whole-Exome Sequencing in Adults With Chronic Kidney Disease. Ann Intern Med 2018; 169:132-133. [PMID: 30014107 DOI: 10.7326/l18-0207] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Affiliation(s)
| | - Emily Groopman
- Columbia University, New York, New York (K.K., E.G., H.R., A.G.G.)
| | - Hila Rasouly
- Columbia University, New York, New York (K.K., E.G., H.R., A.G.G.)
| | | | - Ali G Gharavi
- Columbia University, New York, New York (K.K., E.G., H.R., A.G.G.)
| |
Collapse
|
234
|
Mangaonkar AA, Ferrer A, Pinto E Vairo F, Cousin MA, Kuisle RJ, Klee EW, Kennedy CC, Peters SG, Scott JP, Utz JP, Baqir M, Sekiguchi H, Khan SP, Rodriguez V, Simonetto DA, Kamath PS, Abraham RS, Wylam ME, Patnaik MM. Clinical Correlates and Treatment Outcomes for Patients With Short Telomere Syndromes. Mayo Clin Proc 2018; 93:834-839. [PMID: 29976374 PMCID: PMC7646091 DOI: 10.1016/j.mayocp.2018.05.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 05/11/2018] [Accepted: 05/18/2018] [Indexed: 10/28/2022]
Abstract
Short telomere syndromes (STSs) are accelerated aging syndromes with multisystemic manifestations that present complex management challenges. In this article, we discuss a single-institution experience in diagnosing and managing patients with inherited STSs. In total, we identified 17 patients with short telomeres, defined by flow-fluorescence in-situ hybridization telomere lengths of less than first centile in granulocytes/lymphocytes OR the presence of a characteristic germline pathogenic variant in the context of a highly suggestive clinical phenotype. Genetic variations in the telomere complex were identified in 6 (35%) patients, with 4 being known pathogenic variants involving TERT (n=2), TERC (n=1), and DKC1 (n=1) genes, while 2 were variants of uncertain significance in TERT and RTEL1 genes. Idiopathic interstitial pneumonia (IIP) (n=12 [71%]), unexplained cytopenias (n=5 [29%]), and cirrhosis (n=2 [12%]) were most frequent clinical phenotypes at diagnosis. At median follow-up of 48 (range, 0-316) months, Kaplan-Meier estimate of overall survival, median (95% CI), was 182 (113, not reached) months. Treatment modalities included lung transplantation for IIP (n=5 [29%]), with 3 patients developing signs of acute cellular rejection (2, grade A2; 1, grade A1); danazol therapy for cytopenias (n=4 [24%]), with only 1 out of 4 patients showing a partial hematologic response; and allogeneic hematopoietic stem cell transplant for progressive bone marrow failure (n=2), with 1 patient dying from transplant-related complications. In summary, patients with STSs present with diverse clinical manifestations and require a multidisciplinary approach to management, with organ-specific transplantation capable of providing clinical benefit.
Collapse
Affiliation(s)
| | - Alejandro Ferrer
- Center for Individualized Medicine, Health Sciences Research, Mayo Clinic, Rochester, MN
| | - Filippo Pinto E Vairo
- Center for Individualized Medicine, Health Sciences Research, Mayo Clinic, Rochester, MN
| | - Margot A Cousin
- Center for Individualized Medicine, Health Sciences Research, Mayo Clinic, Rochester, MN
| | - Ryan J Kuisle
- Center for Individualized Medicine, Health Sciences Research, Mayo Clinic, Rochester, MN
| | - Eric W Klee
- Center for Individualized Medicine, Health Sciences Research, Mayo Clinic, Rochester, MN
| | - Cassie C Kennedy
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN
| | - Steve G Peters
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN
| | - J P Scott
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN
| | - James P Utz
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN
| | - Misbah Baqir
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN
| | - Hiroshi Sekiguchi
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN
| | - Shakila P Khan
- Division of Pediatric Hematology/Oncology, Mayo Clinic, Rochester, MN
| | | | | | | | - Roshini S Abraham
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Mark E Wylam
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN.
| | | |
Collapse
|
235
|
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. THE LANCET RESPIRATORY MEDICINE 2018; 6:603-614. [PMID: 29891356 DOI: 10.1016/s2213-2600(18)30135-8] [Citation(s) in RCA: 129] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 03/14/2018] [Accepted: 03/16/2018] [Indexed: 12/22/2022]
Abstract
BACKGROUND Idiopathic pulmonary fibrosis (IPF) risk has a strong genetic component. Studies have implicated variations at several loci, including TERT, surfactant genes, and a single nucleotide polymorphism at chr11p15 (rs35705950) in the intergenic region between TOLLIP and MUC5B. Patients with IPF who have risk alleles at rs35705950 have longer survival from the time of IPF diagnosis than do patients homozygous for the non-risk allele, whereas patients with shorter telomeres have shorter survival times. We aimed to assess whether rare protein-altering variants in genes regulating telomere length are enriched in patients with IPF homozygous for the non-risk alleles at rs35705950. METHODS Between Nov 1, 2014, and Nov 1, 2016, we assessed blood samples from patients aged 40 years or older and of European ancestry with sporadic IPF from three international phase 3 clinical trials (INSPIRE, CAPACITY, ASCEND), one phase 2 study (RIFF), and US-based observational studies (Vanderbilt Clinical Interstitial Lung Disease Registry and the UCSF Interstitial Lung Disease Clinic registry cohorts) at the Broad Institute (Cambridge, MA, USA) and Human Longevity (San Diego, CA, USA). We also assessed blood samples from non-IPF controls in several clinical trials. We did whole-genome sequencing to assess telomere length and identify rare protein-altering variants, stratified by rs35705950 genotype. We also assessed rare functional variation in TERT exons and compared telomere length and disease progression across genotypes. FINDINGS We assessed samples from 1510 patients with IPF and 1874 non-IPF controls. 30 (3%) of 1046 patients with an rs35705950 risk allele had a rare protein-altering variant in TERT compared with 34 (7%) of 464 non-risk allele carriers (odds ratio 0·40 [95% CI 0·24-0·66], p=0·00039). Subsequent analyses identified enrichment of rare protein-altering variants in PARN and RTEL1, and rare variation in TERC in patients with IPF compared with controls. We expanded our study population to provide a more accurate estimation of rare variant frequency in these four loci, and to calculate telomere length. The proportion of patients with at least one rare variant in TERT, PARN, TERC, or RTEL1 was higher in patients with IPF than in controls (149 [9%] of 1739 patients vs 205 [2%] of 8645 controls, p=2·44 × 10-8). Patients with IPF who had a variant in any of the four identified telomerase component genes had telomeres that were 3·69-16·10% shorter than patients without a variant in any of the four genes and had an earlier mean age of disease onset than patients without one or more variants (65·1 years [SD 7·8] vs 67·1 years [7·9], p=0·004). In the placebo arms of clinical trials, shorter telomeres were significantly associated with faster disease progression (1·7% predicted forced vital capacity per kb per year, p=0·002). Pirfenidone had treatment benefit regardless of telomere length (p=4·24 × 10-8 for telomere length lower than the median, p=0·0044 for telomere length greater than the median). INTERPRETATION Rare protein-altering variants in TERT, PARN, TERC, and RTEL1 are enriched in patients with IPF compared with controls, and, in the case of TERT, particularly in individuals without a risk allele at the rs35705950 locus. This suggests that multiple genetic factors contribute to sporadic IPF, which might implicate distinct mechanisms of pathogenesis and disease progression. FUNDING Genentech, National Institutes of Health, Francis Family Foundation, Pulmonary Fibrosis Foundation, Nina Ireland Program for Lung Health, US Department of Veterans Affairs.
Collapse
|
236
|
Merck SJ, Armanios M. Shall we call them "telomere-mediated"? Renaming the idiopathic after the cause is found. Eur Respir J 2018; 48:1556-1558. [PMID: 27903687 DOI: 10.1183/13993003.02115-2016] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 10/28/2016] [Indexed: 01/10/2023]
Affiliation(s)
- Samantha J Merck
- Dept of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mary Armanios
- Dept of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA .,McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
237
|
Radiological pleuroparenchymal fibroelastosis associated to limited cutaneous systemic sclerosis: a case report. BMC Pulm Med 2018; 18:73. [PMID: 29776440 PMCID: PMC5960118 DOI: 10.1186/s12890-018-0641-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 05/04/2018] [Indexed: 12/18/2022] Open
Abstract
Background Pleuroparenchymal fibroelastosis (PPFE) is a very rare interstitial lung disease (ILD) characterized by progressive fibrotic lesions of the visceral pleura and the sub-pleural parenchyma, affecting predominantly the upper lobes. PPFE may occur in different contextes like bone marrow or lung transplantations, but also in the context of telomeropathy with mutations of telomerase reverse transcriptase (TERT), telomerase RNA component (TERC) or regulator of telomere elongation helicase 1 (RTEL1) genes. PPFE-like lesions have recently been described in patients with connective tissue disease (CTD)-related ILD. We report here the first detailed case of PPFE associated to systemic sclerosis (SSc) in a woman free of telomeropathy mutations. Case presentation A caucasian 46 year old woman was followed for SSc in a limited form with anti-centromere Ab since 1998, and seen in 2008 for a routine visit. Her SSc was stable, and she had no respiratory signs. Pulmonary function tests showed an isolated decreased cTLCO at 55.9% (of predicted value). Cardiac ultrasonography was normal. Thoracic CT-scan showed upper lobes predominant mild and focal pleural and subpleural thickenings, suggestive of PPFE, with a slight worsening at 8 years of follow-up. She remained clinically stable. Biology only found a moderate and stable peripheral thrombocytopenia, and sequencing analysis did not find any mutations in TERT and TERC genes. Conclusions ILD is frequent in SSc but isolated PPFE has never been described so far. In our case, PPFE is not related to telomeropathy, has indolent outcome and seems to have good prognosis. PPFE might be an extremely rare form of SSc-related ILD, although a fortuitous association remains possible.
Collapse
|
238
|
Newton CA, Molyneaux PL, Oldham JM. Clinical Genetics in Interstitial Lung Disease. Front Med (Lausanne) 2018; 5:116. [PMID: 29755982 PMCID: PMC5932165 DOI: 10.3389/fmed.2018.00116] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 04/06/2018] [Indexed: 12/14/2022] Open
Abstract
Interstitial lung disease (ILD) comprises a heterogeneous group of diffuse parenchymal lung processes with overlapping clinical, radiographic, and histopathologic features. Among the most common and deadly ILDs are idiopathic pulmonary fibrosis (IPF) and chronic hypersensitivity pneumonitis (CHP). As the name implies, the cause of IPF remains elusive, but a variety of genetic and infectious risk factors have been identified. CHP results from chronic inhalation of an organic antigen, usually of avian or mold origin, and may occur in patients with a genetic predisposition. While IPF is treated with anti-fibrotic compounds, CHP is generally treated by suppression of the immune system and elimination of the causative antigen. Despite advances in our understanding of IPF and CHP, there exists substantial variability in the diagnosis and treatment of these disease processes. Furthermore, IPF and CHP natural history and treatment response remain far from uniform, leaving it unclear which patients derive the most benefit from disease-specific therapy. While clinical prediction models have improved our understanding of outcome risk in patients with various forms of ILD, recent advances in genomic technology provides a valuable opportunity to begin understanding the basis for outcome variability. Such advances will ultimately allow for the incorporation of genomic markers into risk stratification and clinical decision-making. In this piece, we highlight recent advances in our understanding of the genomic factors that influence susceptibility and outcome risk among patients with IPF and CHP. Genomic modalities used to identify these genomic markers include genome-wide association studies, analyses of gene expression, drug–gene interaction testing, telomere length determination, telomerase mutation analysis, and studies of the lung microbiome. We then identify gaps in knowledge that should be addressed to help facilitate the incorporation of these genomic technologies into ILD clinical practice.
Collapse
Affiliation(s)
- Chad A Newton
- Eugene McDermott Centre for Human Growth and Development, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, United States.,Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Philip L Molyneaux
- Fibrosis Research Group, National Heart and Lung Institute, Imperial College, London, United Kingdom.,National Institute for Health Research Respiratory Biomedical Research Unit, Royal Brompton Hospital, London, United Kingdom
| | - Justin M Oldham
- Division of Pulmonary, Department of Medicine, Critical Care and Sleep Medicine, University of California at Davis, Davis, CA, United States
| |
Collapse
|
239
|
Scahill CM, Digby Z, Sealy IM, White RJ, Wali N, Collins JE, Stemple DL, Busch-Nentwich EM. The age of heterozygous telomerase mutant parents influences the adult phenotype of their offspring irrespective of genotype in zebrafish. Wellcome Open Res 2018; 2:77. [PMID: 29568807 PMCID: PMC5840683 DOI: 10.12688/wellcomeopenres.12530.2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/30/2017] [Indexed: 12/03/2022] Open
Abstract
Background: Mutations in proteins involved in telomere maintenance lead to a range of human diseases, including dyskeratosis congenita, idiopathic pulmonary fibrosis and cancer. Telomerase functions to add telomeric repeats back onto the ends of chromosomes, however non-canonical roles of components of telomerase have recently been suggested. Methods: Here we use a zebrafish telomerase mutant which harbours a nonsense mutation in
tert to investigate the adult phenotypes of fish derived from heterozygous parents of different ages. Furthermore we use whole genome sequencing data to estimate average telomere lengths. Results: We show that homozygous offspring from older heterozygotes exhibit signs of body wasting at a younger age than those of younger parents, and that offspring of older heterozygous parents weigh less irrespective of genotype. We also demonstrate that
tert homozygous mutant fish have a male sex bias, and that clutches from older parents also have a male sex bias in the heterozygous and wild-type populations. Telomere length analysis reveals that the telomeres of younger heterozygous parents are shorter than those of older heterozygous parents. Conclusions: These data indicate that the phenotypes observed in offspring from older parents cannot be explained by telomere length. Instead we propose that Tert functions outside of telomere length maintenance in an age-dependent manner to influence the adult phenotypes of the next generation.
Collapse
Affiliation(s)
| | - Zsofia Digby
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, UK.,Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Ian M Sealy
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Richard J White
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Neha Wali
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - John E Collins
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Derek L Stemple
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Elisabeth M Busch-Nentwich
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, UK.,Department of Medicine, University of Cambridge, Cambridge, UK
| |
Collapse
|
240
|
Justet A, Thabut G, Manali E, Molina Molina M, Kannengiesser C, Cadranel J, Cottin V, Gondouin A, Nunes H, Magois E, Tromeur C, Prevot G, Papiris S, Marchand-Adam S, Gamez AS, Reynaud-Gaubert M, Wemeau L, Crestani B, Borie R. Safety and efficacy of pirfenidone in patients carrying telomerase complex mutation. Eur Respir J 2018; 51:13993003.01875-2017. [PMID: 29449422 DOI: 10.1183/13993003.01875-2017] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 01/04/2018] [Indexed: 11/05/2022]
Affiliation(s)
- Aurélien Justet
- APHP, Hôpital Bichat, Service de Pneumologie A, DHU FIRE, Paris, France.,INSERM, Unité 1152, Université Paris Diderot, Paris, France
| | - Gabriel Thabut
- INSERM, Unité 1152, Université Paris Diderot, Paris, France.,APHP, Hôpital Bichat, Service de Pneumologie B, Paris, France
| | - Effrosyni Manali
- Respiratory Medicine Dept, 'Attikon' University Hospital, Athens Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Maria Molina Molina
- Unit of Interstitial Lung Diseases, Dept of Pneumology, University Hospital of Bellvitge, Barcelona, Spain
| | | | - Jacques Cadranel
- APHP, Service de Pneumologie, maladies pulmonaires rares, Hôpital Tenon, Paris, France
| | - Vincent Cottin
- Service de Pneumologie, Hôpital Louis Pradel, Université Claude Bernard Lyon 1, Lyon, France
| | - Anne Gondouin
- CHU de Besançon, Service de Pneumologie, Besançon, France
| | - Hilario Nunes
- APHP, Service de Pneumologie, maladies pulmonaires rares, Hôpital Avicenne, Bobigny, France
| | - Eline Magois
- Service de Pneumologie, Hôpital d'Amiens, Université de Picardie Jules Verne, Amiens, France
| | - Cécile Tromeur
- Université Européenne de Bretagne, UBO, EA3878 (GETBO) IFR 148, CHU de la Cavale Blanche, Département de médecine interne et de pneumologie, Brest, France
| | | | - Spyros Papiris
- Respiratory Medicine Dept, 'Attikon' University Hospital, Athens Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Sylvain Marchand-Adam
- CHU de Tours, Service de Pneumologie et Explorations Fonctionnelles Respiratoires, Tours, France
| | - Anne Sophie Gamez
- Département de Pneumologie et Addictologie, Hôpital Arnaud de Villeneuve, CHU Montpellier, Montpellier, France
| | | | | | - Bruno Crestani
- APHP, Hôpital Bichat, Service de Pneumologie A, DHU FIRE, Paris, France.,INSERM, Unité 1152, Université Paris Diderot, Paris, France
| | - Raphael Borie
- APHP, Hôpital Bichat, Service de Pneumologie A, DHU FIRE, Paris, France .,INSERM, Unité 1152, Université Paris Diderot, Paris, France
| |
Collapse
|
241
|
El-Chemaly S, Cheung F, Kotliarov Y, O'Brien KJ, Gahl WA, Chen J, Perl SY, Biancotto A, Gochuico BR. The Immunome in Two Inherited Forms of Pulmonary Fibrosis. Front Immunol 2018; 9:76. [PMID: 29445374 PMCID: PMC5797737 DOI: 10.3389/fimmu.2018.00076] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 01/11/2018] [Indexed: 02/01/2023] Open
Abstract
The immunome (immune cell phenotype, gene expression, and serum cytokines profiling) in pulmonary fibrosis is incompletely defined. Studies focusing on inherited forms of pulmonary fibrosis provide insights into mechanisms of fibrotic lung disease in general. To define the cellular and molecular immunologic phenotype in peripheral blood, high-dimensional flow cytometry and large-scale gene expression of peripheral blood mononuclear cells and serum proteomic multiplex analyses were performed and compared in a cohort with familial pulmonary fibrosis (FPF), an autosomal dominant disorder with incomplete penetrance; Hermansky-Pudlak syndrome pulmonary fibrosis (HPSPF), a rare autosomal recessive disorder; and their unaffected relatives. Our results showed high peripheral blood concentrations of activated central memory helper cells in patients with FPF. Proportions of CD38+ memory CD27- B-cells, IgA+ memory CD27+ B-cells, IgM+ and IgD+ B-cells, and CD39+ T helper cells were increased whereas those of CD39- T helper cells were reduced in patients affected with either familial or HPSPF. Gene expression and serum proteomic analyses revealed enrichment of upregulated genes associated with mitosis and cell cycle control in circulating mononuclear cells as well as altered levels of several analytes, including leptin, cytokines, and growth factors. In conclusion, dysregulation of the extra-pulmonary immunome is a phenotypic feature of FPF or HPSPF. Further studies investigating the blood immunome are indicated to determine the role of immune system dysregulation in the pathogenesis of pulmonary fibrosis. Clinical Trial Registration www.ClinicalTrials.gov, identifiers NCT00968084, NCT01200823, NCT00001456, and NCT00084305.
Collapse
Affiliation(s)
- Souheil El-Chemaly
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, United States
| | - Foo Cheung
- Trans-NIH Center for Human Immunology, Autoimmunity, and Inflammation (CHI), National Institutes of Health, Bethesda, MD, United States
| | - Yuri Kotliarov
- Trans-NIH Center for Human Immunology, Autoimmunity, and Inflammation (CHI), National Institutes of Health, Bethesda, MD, United States
| | - Kevin J O'Brien
- Office of the Clinical Director, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States
| | - William A Gahl
- Office of the Clinical Director, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States.,Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States
| | - Jinguo Chen
- Trans-NIH Center for Human Immunology, Autoimmunity, and Inflammation (CHI), National Institutes of Health, Bethesda, MD, United States
| | - Shira Y Perl
- Trans-NIH Center for Human Immunology, Autoimmunity, and Inflammation (CHI), National Institutes of Health, Bethesda, MD, United States
| | - Angélique Biancotto
- Trans-NIH Center for Human Immunology, Autoimmunity, and Inflammation (CHI), National Institutes of Health, Bethesda, MD, United States
| | - Bernadette R Gochuico
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
242
|
Lata S, Marasa M, Li Y, Fasel DA, Groopman E, Jobanputra V, Rasouly H, Mitrotti A, Westland R, Verbitsky M, Nestor J, Slater LM, D'Agati V, Zaniew M, Materna-Kiryluk A, Lugani F, Caridi G, Rampoldi L, Mattoo A, Newton CA, Rao MK, Radhakrishnan J, Ahn W, Canetta PA, Bomback AS, Appel GB, Antignac C, Markowitz GS, Garcia CK, Kiryluk K, Sanna-Cherchi S, Gharavi AG. Whole-Exome Sequencing in Adults With Chronic Kidney Disease: A Pilot Study. Ann Intern Med 2018; 168:100-109. [PMID: 29204651 PMCID: PMC5947852 DOI: 10.7326/m17-1319] [Citation(s) in RCA: 140] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND The utility of whole-exome sequencing (WES) for the diagnosis and management of adult-onset constitutional disorders has not been adequately studied. Genetic diagnostics may be advantageous in adults with chronic kidney disease (CKD), in whom the cause of kidney failure often remains unknown. OBJECTIVE To study the diagnostic utility of WES in a selected referral population of adults with CKD. DESIGN Observational cohort. SETTING A major academic medical center. PATIENTS 92 adults with CKD of unknown cause or familial nephropathy or hypertension. MEASUREMENTS The diagnostic yield of WES and its potential effect on clinical management. RESULTS Whole-exome sequencing provided a diagnosis in 22 of 92 patients (24%), including 9 probands with CKD of unknown cause and encompassing 13 distinct genetic disorders. Among these, loss-of-function mutations were identified in PARN in 2 probands with tubulointerstitial fibrosis. PARN mutations have been implicated in a short telomere syndrome characterized by lung, bone marrow, and liver fibrosis; these findings extend the phenotype of PARN mutations to renal fibrosis. In addition, review of the American College of Medical Genetics actionable genes identified a pathogenic BRCA2 mutation in a proband who was diagnosed with breast cancer on follow-up. The results affected clinical management in most identified cases, including initiation of targeted surveillance, familial screening to guide donor selection for transplantation, and changes in therapy. LIMITATION The small sample size and recruitment at a tertiary care academic center limit generalizability of findings among the broader CKD population. CONCLUSION Whole-exome sequencing identified diagnostic mutations in a substantial number of adults with CKD of many causes. Further study of the utility of WES in the evaluation and care of patients with CKD in additional settings is warranted. PRIMARY FUNDING SOURCE New York State Empire Clinical Research Investigator Program, Renal Research Institute, and National Human Genome Research Institute of the National Institutes of Health.
Collapse
Affiliation(s)
- Sneh Lata
- From Columbia University, New York, New York; VU University Medical Center, Amsterdam, the Netherlands; Nephrology Associates, Newark, Delaware; Krysiewicza Children's Hospital, Poznań, Poland; Poznań University of Medical Sciences and Center for Medical Genetics GENESIS, Poznań, Poland; IRCCS Giannina Gaslini Children's Hospital, Genova, Italy; IRCCS San Raffaele Scientific Institute, Milan, Italy; New York University School of Medicine, New York, New York; University of Texas Southwestern Medical Center, Dallas, Texas; and French Institute of Health and Medical Research (INSERM) U1163, Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, and Necker Hospital, Paris, France
| | - Maddalena Marasa
- From Columbia University, New York, New York; VU University Medical Center, Amsterdam, the Netherlands; Nephrology Associates, Newark, Delaware; Krysiewicza Children's Hospital, Poznań, Poland; Poznań University of Medical Sciences and Center for Medical Genetics GENESIS, Poznań, Poland; IRCCS Giannina Gaslini Children's Hospital, Genova, Italy; IRCCS San Raffaele Scientific Institute, Milan, Italy; New York University School of Medicine, New York, New York; University of Texas Southwestern Medical Center, Dallas, Texas; and French Institute of Health and Medical Research (INSERM) U1163, Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, and Necker Hospital, Paris, France
| | - Yifu Li
- From Columbia University, New York, New York; VU University Medical Center, Amsterdam, the Netherlands; Nephrology Associates, Newark, Delaware; Krysiewicza Children's Hospital, Poznań, Poland; Poznań University of Medical Sciences and Center for Medical Genetics GENESIS, Poznań, Poland; IRCCS Giannina Gaslini Children's Hospital, Genova, Italy; IRCCS San Raffaele Scientific Institute, Milan, Italy; New York University School of Medicine, New York, New York; University of Texas Southwestern Medical Center, Dallas, Texas; and French Institute of Health and Medical Research (INSERM) U1163, Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, and Necker Hospital, Paris, France
| | - David A Fasel
- From Columbia University, New York, New York; VU University Medical Center, Amsterdam, the Netherlands; Nephrology Associates, Newark, Delaware; Krysiewicza Children's Hospital, Poznań, Poland; Poznań University of Medical Sciences and Center for Medical Genetics GENESIS, Poznań, Poland; IRCCS Giannina Gaslini Children's Hospital, Genova, Italy; IRCCS San Raffaele Scientific Institute, Milan, Italy; New York University School of Medicine, New York, New York; University of Texas Southwestern Medical Center, Dallas, Texas; and French Institute of Health and Medical Research (INSERM) U1163, Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, and Necker Hospital, Paris, France
| | - Emily Groopman
- From Columbia University, New York, New York; VU University Medical Center, Amsterdam, the Netherlands; Nephrology Associates, Newark, Delaware; Krysiewicza Children's Hospital, Poznań, Poland; Poznań University of Medical Sciences and Center for Medical Genetics GENESIS, Poznań, Poland; IRCCS Giannina Gaslini Children's Hospital, Genova, Italy; IRCCS San Raffaele Scientific Institute, Milan, Italy; New York University School of Medicine, New York, New York; University of Texas Southwestern Medical Center, Dallas, Texas; and French Institute of Health and Medical Research (INSERM) U1163, Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, and Necker Hospital, Paris, France
| | - Vaidehi Jobanputra
- From Columbia University, New York, New York; VU University Medical Center, Amsterdam, the Netherlands; Nephrology Associates, Newark, Delaware; Krysiewicza Children's Hospital, Poznań, Poland; Poznań University of Medical Sciences and Center for Medical Genetics GENESIS, Poznań, Poland; IRCCS Giannina Gaslini Children's Hospital, Genova, Italy; IRCCS San Raffaele Scientific Institute, Milan, Italy; New York University School of Medicine, New York, New York; University of Texas Southwestern Medical Center, Dallas, Texas; and French Institute of Health and Medical Research (INSERM) U1163, Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, and Necker Hospital, Paris, France
| | - Hila Rasouly
- From Columbia University, New York, New York; VU University Medical Center, Amsterdam, the Netherlands; Nephrology Associates, Newark, Delaware; Krysiewicza Children's Hospital, Poznań, Poland; Poznań University of Medical Sciences and Center for Medical Genetics GENESIS, Poznań, Poland; IRCCS Giannina Gaslini Children's Hospital, Genova, Italy; IRCCS San Raffaele Scientific Institute, Milan, Italy; New York University School of Medicine, New York, New York; University of Texas Southwestern Medical Center, Dallas, Texas; and French Institute of Health and Medical Research (INSERM) U1163, Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, and Necker Hospital, Paris, France
| | - Adele Mitrotti
- From Columbia University, New York, New York; VU University Medical Center, Amsterdam, the Netherlands; Nephrology Associates, Newark, Delaware; Krysiewicza Children's Hospital, Poznań, Poland; Poznań University of Medical Sciences and Center for Medical Genetics GENESIS, Poznań, Poland; IRCCS Giannina Gaslini Children's Hospital, Genova, Italy; IRCCS San Raffaele Scientific Institute, Milan, Italy; New York University School of Medicine, New York, New York; University of Texas Southwestern Medical Center, Dallas, Texas; and French Institute of Health and Medical Research (INSERM) U1163, Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, and Necker Hospital, Paris, France
| | - Rik Westland
- From Columbia University, New York, New York; VU University Medical Center, Amsterdam, the Netherlands; Nephrology Associates, Newark, Delaware; Krysiewicza Children's Hospital, Poznań, Poland; Poznań University of Medical Sciences and Center for Medical Genetics GENESIS, Poznań, Poland; IRCCS Giannina Gaslini Children's Hospital, Genova, Italy; IRCCS San Raffaele Scientific Institute, Milan, Italy; New York University School of Medicine, New York, New York; University of Texas Southwestern Medical Center, Dallas, Texas; and French Institute of Health and Medical Research (INSERM) U1163, Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, and Necker Hospital, Paris, France
| | - Miguel Verbitsky
- From Columbia University, New York, New York; VU University Medical Center, Amsterdam, the Netherlands; Nephrology Associates, Newark, Delaware; Krysiewicza Children's Hospital, Poznań, Poland; Poznań University of Medical Sciences and Center for Medical Genetics GENESIS, Poznań, Poland; IRCCS Giannina Gaslini Children's Hospital, Genova, Italy; IRCCS San Raffaele Scientific Institute, Milan, Italy; New York University School of Medicine, New York, New York; University of Texas Southwestern Medical Center, Dallas, Texas; and French Institute of Health and Medical Research (INSERM) U1163, Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, and Necker Hospital, Paris, France
| | - Jordan Nestor
- From Columbia University, New York, New York; VU University Medical Center, Amsterdam, the Netherlands; Nephrology Associates, Newark, Delaware; Krysiewicza Children's Hospital, Poznań, Poland; Poznań University of Medical Sciences and Center for Medical Genetics GENESIS, Poznań, Poland; IRCCS Giannina Gaslini Children's Hospital, Genova, Italy; IRCCS San Raffaele Scientific Institute, Milan, Italy; New York University School of Medicine, New York, New York; University of Texas Southwestern Medical Center, Dallas, Texas; and French Institute of Health and Medical Research (INSERM) U1163, Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, and Necker Hospital, Paris, France
| | - Lindsey M Slater
- From Columbia University, New York, New York; VU University Medical Center, Amsterdam, the Netherlands; Nephrology Associates, Newark, Delaware; Krysiewicza Children's Hospital, Poznań, Poland; Poznań University of Medical Sciences and Center for Medical Genetics GENESIS, Poznań, Poland; IRCCS Giannina Gaslini Children's Hospital, Genova, Italy; IRCCS San Raffaele Scientific Institute, Milan, Italy; New York University School of Medicine, New York, New York; University of Texas Southwestern Medical Center, Dallas, Texas; and French Institute of Health and Medical Research (INSERM) U1163, Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, and Necker Hospital, Paris, France
| | - Vivette D'Agati
- From Columbia University, New York, New York; VU University Medical Center, Amsterdam, the Netherlands; Nephrology Associates, Newark, Delaware; Krysiewicza Children's Hospital, Poznań, Poland; Poznań University of Medical Sciences and Center for Medical Genetics GENESIS, Poznań, Poland; IRCCS Giannina Gaslini Children's Hospital, Genova, Italy; IRCCS San Raffaele Scientific Institute, Milan, Italy; New York University School of Medicine, New York, New York; University of Texas Southwestern Medical Center, Dallas, Texas; and French Institute of Health and Medical Research (INSERM) U1163, Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, and Necker Hospital, Paris, France
| | - Marcin Zaniew
- From Columbia University, New York, New York; VU University Medical Center, Amsterdam, the Netherlands; Nephrology Associates, Newark, Delaware; Krysiewicza Children's Hospital, Poznań, Poland; Poznań University of Medical Sciences and Center for Medical Genetics GENESIS, Poznań, Poland; IRCCS Giannina Gaslini Children's Hospital, Genova, Italy; IRCCS San Raffaele Scientific Institute, Milan, Italy; New York University School of Medicine, New York, New York; University of Texas Southwestern Medical Center, Dallas, Texas; and French Institute of Health and Medical Research (INSERM) U1163, Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, and Necker Hospital, Paris, France
| | - Anna Materna-Kiryluk
- From Columbia University, New York, New York; VU University Medical Center, Amsterdam, the Netherlands; Nephrology Associates, Newark, Delaware; Krysiewicza Children's Hospital, Poznań, Poland; Poznań University of Medical Sciences and Center for Medical Genetics GENESIS, Poznań, Poland; IRCCS Giannina Gaslini Children's Hospital, Genova, Italy; IRCCS San Raffaele Scientific Institute, Milan, Italy; New York University School of Medicine, New York, New York; University of Texas Southwestern Medical Center, Dallas, Texas; and French Institute of Health and Medical Research (INSERM) U1163, Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, and Necker Hospital, Paris, France
| | - Francesca Lugani
- From Columbia University, New York, New York; VU University Medical Center, Amsterdam, the Netherlands; Nephrology Associates, Newark, Delaware; Krysiewicza Children's Hospital, Poznań, Poland; Poznań University of Medical Sciences and Center for Medical Genetics GENESIS, Poznań, Poland; IRCCS Giannina Gaslini Children's Hospital, Genova, Italy; IRCCS San Raffaele Scientific Institute, Milan, Italy; New York University School of Medicine, New York, New York; University of Texas Southwestern Medical Center, Dallas, Texas; and French Institute of Health and Medical Research (INSERM) U1163, Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, and Necker Hospital, Paris, France
| | - Gianluca Caridi
- From Columbia University, New York, New York; VU University Medical Center, Amsterdam, the Netherlands; Nephrology Associates, Newark, Delaware; Krysiewicza Children's Hospital, Poznań, Poland; Poznań University of Medical Sciences and Center for Medical Genetics GENESIS, Poznań, Poland; IRCCS Giannina Gaslini Children's Hospital, Genova, Italy; IRCCS San Raffaele Scientific Institute, Milan, Italy; New York University School of Medicine, New York, New York; University of Texas Southwestern Medical Center, Dallas, Texas; and French Institute of Health and Medical Research (INSERM) U1163, Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, and Necker Hospital, Paris, France
| | - Luca Rampoldi
- From Columbia University, New York, New York; VU University Medical Center, Amsterdam, the Netherlands; Nephrology Associates, Newark, Delaware; Krysiewicza Children's Hospital, Poznań, Poland; Poznań University of Medical Sciences and Center for Medical Genetics GENESIS, Poznań, Poland; IRCCS Giannina Gaslini Children's Hospital, Genova, Italy; IRCCS San Raffaele Scientific Institute, Milan, Italy; New York University School of Medicine, New York, New York; University of Texas Southwestern Medical Center, Dallas, Texas; and French Institute of Health and Medical Research (INSERM) U1163, Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, and Necker Hospital, Paris, France
| | - Aditya Mattoo
- From Columbia University, New York, New York; VU University Medical Center, Amsterdam, the Netherlands; Nephrology Associates, Newark, Delaware; Krysiewicza Children's Hospital, Poznań, Poland; Poznań University of Medical Sciences and Center for Medical Genetics GENESIS, Poznań, Poland; IRCCS Giannina Gaslini Children's Hospital, Genova, Italy; IRCCS San Raffaele Scientific Institute, Milan, Italy; New York University School of Medicine, New York, New York; University of Texas Southwestern Medical Center, Dallas, Texas; and French Institute of Health and Medical Research (INSERM) U1163, Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, and Necker Hospital, Paris, France
| | - Chad A Newton
- From Columbia University, New York, New York; VU University Medical Center, Amsterdam, the Netherlands; Nephrology Associates, Newark, Delaware; Krysiewicza Children's Hospital, Poznań, Poland; Poznań University of Medical Sciences and Center for Medical Genetics GENESIS, Poznań, Poland; IRCCS Giannina Gaslini Children's Hospital, Genova, Italy; IRCCS San Raffaele Scientific Institute, Milan, Italy; New York University School of Medicine, New York, New York; University of Texas Southwestern Medical Center, Dallas, Texas; and French Institute of Health and Medical Research (INSERM) U1163, Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, and Necker Hospital, Paris, France
| | - Maya K Rao
- From Columbia University, New York, New York; VU University Medical Center, Amsterdam, the Netherlands; Nephrology Associates, Newark, Delaware; Krysiewicza Children's Hospital, Poznań, Poland; Poznań University of Medical Sciences and Center for Medical Genetics GENESIS, Poznań, Poland; IRCCS Giannina Gaslini Children's Hospital, Genova, Italy; IRCCS San Raffaele Scientific Institute, Milan, Italy; New York University School of Medicine, New York, New York; University of Texas Southwestern Medical Center, Dallas, Texas; and French Institute of Health and Medical Research (INSERM) U1163, Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, and Necker Hospital, Paris, France
| | - Jai Radhakrishnan
- From Columbia University, New York, New York; VU University Medical Center, Amsterdam, the Netherlands; Nephrology Associates, Newark, Delaware; Krysiewicza Children's Hospital, Poznań, Poland; Poznań University of Medical Sciences and Center for Medical Genetics GENESIS, Poznań, Poland; IRCCS Giannina Gaslini Children's Hospital, Genova, Italy; IRCCS San Raffaele Scientific Institute, Milan, Italy; New York University School of Medicine, New York, New York; University of Texas Southwestern Medical Center, Dallas, Texas; and French Institute of Health and Medical Research (INSERM) U1163, Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, and Necker Hospital, Paris, France
| | - Wooin Ahn
- From Columbia University, New York, New York; VU University Medical Center, Amsterdam, the Netherlands; Nephrology Associates, Newark, Delaware; Krysiewicza Children's Hospital, Poznań, Poland; Poznań University of Medical Sciences and Center for Medical Genetics GENESIS, Poznań, Poland; IRCCS Giannina Gaslini Children's Hospital, Genova, Italy; IRCCS San Raffaele Scientific Institute, Milan, Italy; New York University School of Medicine, New York, New York; University of Texas Southwestern Medical Center, Dallas, Texas; and French Institute of Health and Medical Research (INSERM) U1163, Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, and Necker Hospital, Paris, France
| | - Pietro A Canetta
- From Columbia University, New York, New York; VU University Medical Center, Amsterdam, the Netherlands; Nephrology Associates, Newark, Delaware; Krysiewicza Children's Hospital, Poznań, Poland; Poznań University of Medical Sciences and Center for Medical Genetics GENESIS, Poznań, Poland; IRCCS Giannina Gaslini Children's Hospital, Genova, Italy; IRCCS San Raffaele Scientific Institute, Milan, Italy; New York University School of Medicine, New York, New York; University of Texas Southwestern Medical Center, Dallas, Texas; and French Institute of Health and Medical Research (INSERM) U1163, Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, and Necker Hospital, Paris, France
| | - Andrew S Bomback
- From Columbia University, New York, New York; VU University Medical Center, Amsterdam, the Netherlands; Nephrology Associates, Newark, Delaware; Krysiewicza Children's Hospital, Poznań, Poland; Poznań University of Medical Sciences and Center for Medical Genetics GENESIS, Poznań, Poland; IRCCS Giannina Gaslini Children's Hospital, Genova, Italy; IRCCS San Raffaele Scientific Institute, Milan, Italy; New York University School of Medicine, New York, New York; University of Texas Southwestern Medical Center, Dallas, Texas; and French Institute of Health and Medical Research (INSERM) U1163, Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, and Necker Hospital, Paris, France
| | - Gerald B Appel
- From Columbia University, New York, New York; VU University Medical Center, Amsterdam, the Netherlands; Nephrology Associates, Newark, Delaware; Krysiewicza Children's Hospital, Poznań, Poland; Poznań University of Medical Sciences and Center for Medical Genetics GENESIS, Poznań, Poland; IRCCS Giannina Gaslini Children's Hospital, Genova, Italy; IRCCS San Raffaele Scientific Institute, Milan, Italy; New York University School of Medicine, New York, New York; University of Texas Southwestern Medical Center, Dallas, Texas; and French Institute of Health and Medical Research (INSERM) U1163, Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, and Necker Hospital, Paris, France
| | - Corinne Antignac
- From Columbia University, New York, New York; VU University Medical Center, Amsterdam, the Netherlands; Nephrology Associates, Newark, Delaware; Krysiewicza Children's Hospital, Poznań, Poland; Poznań University of Medical Sciences and Center for Medical Genetics GENESIS, Poznań, Poland; IRCCS Giannina Gaslini Children's Hospital, Genova, Italy; IRCCS San Raffaele Scientific Institute, Milan, Italy; New York University School of Medicine, New York, New York; University of Texas Southwestern Medical Center, Dallas, Texas; and French Institute of Health and Medical Research (INSERM) U1163, Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, and Necker Hospital, Paris, France
| | - Glen S Markowitz
- From Columbia University, New York, New York; VU University Medical Center, Amsterdam, the Netherlands; Nephrology Associates, Newark, Delaware; Krysiewicza Children's Hospital, Poznań, Poland; Poznań University of Medical Sciences and Center for Medical Genetics GENESIS, Poznań, Poland; IRCCS Giannina Gaslini Children's Hospital, Genova, Italy; IRCCS San Raffaele Scientific Institute, Milan, Italy; New York University School of Medicine, New York, New York; University of Texas Southwestern Medical Center, Dallas, Texas; and French Institute of Health and Medical Research (INSERM) U1163, Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, and Necker Hospital, Paris, France
| | - Christine K Garcia
- From Columbia University, New York, New York; VU University Medical Center, Amsterdam, the Netherlands; Nephrology Associates, Newark, Delaware; Krysiewicza Children's Hospital, Poznań, Poland; Poznań University of Medical Sciences and Center for Medical Genetics GENESIS, Poznań, Poland; IRCCS Giannina Gaslini Children's Hospital, Genova, Italy; IRCCS San Raffaele Scientific Institute, Milan, Italy; New York University School of Medicine, New York, New York; University of Texas Southwestern Medical Center, Dallas, Texas; and French Institute of Health and Medical Research (INSERM) U1163, Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, and Necker Hospital, Paris, France
| | - Krzysztof Kiryluk
- From Columbia University, New York, New York; VU University Medical Center, Amsterdam, the Netherlands; Nephrology Associates, Newark, Delaware; Krysiewicza Children's Hospital, Poznań, Poland; Poznań University of Medical Sciences and Center for Medical Genetics GENESIS, Poznań, Poland; IRCCS Giannina Gaslini Children's Hospital, Genova, Italy; IRCCS San Raffaele Scientific Institute, Milan, Italy; New York University School of Medicine, New York, New York; University of Texas Southwestern Medical Center, Dallas, Texas; and French Institute of Health and Medical Research (INSERM) U1163, Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, and Necker Hospital, Paris, France
| | - Simone Sanna-Cherchi
- From Columbia University, New York, New York; VU University Medical Center, Amsterdam, the Netherlands; Nephrology Associates, Newark, Delaware; Krysiewicza Children's Hospital, Poznań, Poland; Poznań University of Medical Sciences and Center for Medical Genetics GENESIS, Poznań, Poland; IRCCS Giannina Gaslini Children's Hospital, Genova, Italy; IRCCS San Raffaele Scientific Institute, Milan, Italy; New York University School of Medicine, New York, New York; University of Texas Southwestern Medical Center, Dallas, Texas; and French Institute of Health and Medical Research (INSERM) U1163, Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, and Necker Hospital, Paris, France
| | - Ali G Gharavi
- From Columbia University, New York, New York; VU University Medical Center, Amsterdam, the Netherlands; Nephrology Associates, Newark, Delaware; Krysiewicza Children's Hospital, Poznań, Poland; Poznań University of Medical Sciences and Center for Medical Genetics GENESIS, Poznań, Poland; IRCCS Giannina Gaslini Children's Hospital, Genova, Italy; IRCCS San Raffaele Scientific Institute, Milan, Italy; New York University School of Medicine, New York, New York; University of Texas Southwestern Medical Center, Dallas, Texas; and French Institute of Health and Medical Research (INSERM) U1163, Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, and Necker Hospital, Paris, France
| |
Collapse
|
243
|
Abstract
Genetic investigations of fibrotic diseases, including those of late onset, often yield unanticipated insights into disease pathogenesis. This Review focuses on pathways underlying lung fibrosis that are generalizable to other organs. Herein, we discuss genetic variants subdivided into those that shorten telomeres, activate the DNA damage response, change resident protein expression or function, or affect organelle activity. Genetic studies provide a window into the downstream cascade of maladaptive responses and pathways that lead to tissue fibrosis. In addition, these studies reveal interactions between genetic variants, environmental factors, and age that influence the phenotypic spectrum of disease. The discovery of forces counterbalancing inherited risk alleles identifies potential therapeutic targets, thus providing hope for future prevention or reversal of fibrosis.
Collapse
|
244
|
Molina-Molina M, Planas-Cerezales L, Perona R. Acortamiento de los telómeros en fibrosis pulmonar idiopática. Arch Bronconeumol 2018; 54:3-4. [DOI: 10.1016/j.arbres.2017.07.026] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 07/26/2017] [Accepted: 07/28/2017] [Indexed: 11/25/2022]
|
245
|
Borie R, Kannengiesser C, Debray MP, Crestani B. The Genetic Diagnosis of Interstitial Lung Disease: A Need for an International Consensus. Am J Respir Crit Care Med 2017; 195:1538-1539. [PMID: 28569587 DOI: 10.1164/rccm.201611-2209le] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Raphael Borie
- 1 Hôpital Bichat Paris, France Université Paris Diderot Paris, France and.,2 INSERM U1152 Paris, France
| | | | | | - Bruno Crestani
- 1 Hôpital Bichat Paris, France Université Paris Diderot Paris, France and.,2 INSERM U1152 Paris, France
| |
Collapse
|
246
|
Borie R, Kannengiesser C, Sicre de Fontbrune F, Boutboul D, Tabeze L, Brunet-Possenti F, Lainey E, Debray MP, Cazes A, Crestani B. Pneumocystosis revealing immunodeficiency secondary to TERC mutation. Eur Respir J 2017; 50:50/5/1701443. [PMID: 29167304 DOI: 10.1183/13993003.01443-2017] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 09/01/2017] [Indexed: 12/22/2022]
Affiliation(s)
- Raphael Borie
- APHP, Hôpital Bichat, Service de Pneumologie A, Centre de référence des maladies pulmonaires rares, Paris, France .,INSERM, Unité 1152, Université Paris Diderot, Paris, France
| | | | - Flore Sicre de Fontbrune
- Service d'Hématologie greffe, Centre de référence maladie rare aplasie médullaire, APHP, Hôpital St Louis, Paris, France
| | - David Boutboul
- INSERM UMR 1163, Laboratory of Genome Dynamics in the Immune System, Paris Descartes - Sorbonne Paris Cité University, Imagine Institute, Paris, France
| | - Laure Tabeze
- APHP, Hôpital Bichat, Service de Pneumologie A, Centre de référence des maladies pulmonaires rares, Paris, France
| | | | - Elodie Lainey
- APHP, Hopital Robert Debré, Laboratoire d'hématologie, Paris, France
| | | | - Aurélie Cazes
- INSERM, Unité 1152, Université Paris Diderot, Paris, France.,APHP, Hôpital Bichat, Laboratoire d'anatomopathologie, Paris, France
| | - Bruno Crestani
- APHP, Hôpital Bichat, Service de Pneumologie A, Centre de référence des maladies pulmonaires rares, Paris, France.,INSERM, Unité 1152, Université Paris Diderot, Paris, France
| |
Collapse
|
247
|
Adegunsoye A, Oldham JM, Husain AN, Chen L, Hsu S, Montner S, Chung JH, Vij R, Noth I, Strek ME. Autoimmune Hypothyroidism As a Predictor of Mortality in Chronic Hypersensitivity Pneumonitis. Front Med (Lausanne) 2017; 4:170. [PMID: 29085824 PMCID: PMC5650730 DOI: 10.3389/fmed.2017.00170] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 09/26/2017] [Indexed: 11/13/2022] Open
Abstract
Background Chronic hypersensitivity pneumonitis (CHP) is a fibrotic parenchymal lung disease that occurs when inhalation of environmental antigens leads to immune dysregulation. Autoimmune features have recently been identified as potentially important among patients with CHP. However, the relationship between hypothyroidism (HT) and CHP is unknown. In this study, we investigate the prevalence and impact of HT among patients with CHP. Methods We conducted a retrospective, case–control analysis. We identified 121 patients at the University of Chicago Interstitial Lung Disease Center with a multidisciplinary diagnosis of CHP. These patients were matched 3:1 according to age, sex, and race to 363 control subjects with asthma from 2006 to 2015. We analyzed demographics, clinical characteristics, and survival between both groups and assessed the relationship of HT with CHP. Survival analysis was performed using Cox proportional hazards modeling. Results Patients with CHP had higher prevalence of HT (25.6%, n = 31) compared to controls (10.7%, n = 39; OR, 2.86; 95% CI, 1.62–4.99; P < 0.0001). Compared to CHP alone, patients with CHP/HT were more likely to be female (80.6 vs 51.1%, P = 0.004), have increased incidence of autoimmune disease (19.4 vs 3.3%, P = 0.009), antinuclear antibody seropositivity (80.6 vs 57.0%, P = 0.019), and higher TSH levels (4.0 vs 1.9 mIU/L, P < 0.0001). HT was a significant independent predictor of mortality among CHP patients with seropositive ANA (HR, 3.39; 95% CI, 1.31–8.80; P = 0.012). Conclusion HT is common in patients with CHP and may carry prognostic significance in patients with features of autoimmunity. Further research exploring common pathogenic pathways between autoimmune HT and CHP may illuminate the association of HT with survival.
Collapse
Affiliation(s)
- Ayodeji Adegunsoye
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, University of Chicago, Chicago, IL, United States
| | - Justin M Oldham
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, Davis, Davis, CA, United States
| | - Aliya N Husain
- Department of Pathology, University of Chicago, Chicago, IL, United States
| | - Lena Chen
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, University of Chicago, Chicago, IL, United States
| | - Scully Hsu
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, University of Chicago, Chicago, IL, United States
| | - Steven Montner
- Department of Radiology, University of Chicago, Chicago, IL, United States
| | - Jonathan H Chung
- Department of Radiology, University of Chicago, Chicago, IL, United States
| | - Rekha Vij
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, University of Chicago, Chicago, IL, United States
| | - Imre Noth
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, University of Chicago, Chicago, IL, United States
| | - Mary E Strek
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, University of Chicago, Chicago, IL, United States
| |
Collapse
|
248
|
Cottin V, Crestani B, Cadranel J, Cordier JF, Marchand-Adam S, Prévot G, Wallaert B, Bergot E, Camus P, Dalphin JC, Dromer C, Gomez E, Israel-Biet D, Jouneau S, Kessler R, Marquette CH, Reynaud-Gaubert M, Aguilaniu B, Bonnet D, Carré P, Danel C, Faivre JB, Ferretti G, Just N, Lebargy F, Philippe B, Terrioux P, Thivolet-Béjui F, Trumbic B, Valeyre D. French practical guidelines for the diagnosis and management of idiopathic pulmonary fibrosis – 2017 update. Full-length version. Rev Mal Respir 2017; 34:900-968. [DOI: 10.1016/j.rmr.2017.07.017] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
249
|
Cottin V, Crestani B, Cadranel J, Cordier JF, Marchand-Adam S, Prévot G, Wallaert B, Bergot E, Camus P, Dalphin JC, Dromer C, Gomez E, Israel-Biet D, Jouneau S, Kessler R, Marquette CH, Reynaud-Gaubert M, Aguilaniu B, Bonnet D, Carré P, Danel C, Faivre JB, Ferretti G, Just N, Lebargy F, Philippe B, Terrioux P, Thivolet-Béjui F, Trumbic B, Valeyre D. [French practical guidelines for the diagnosis and management of idiopathic pulmonary fibrosis. 2017 update. Full-length update]. Rev Mal Respir 2017:S0761-8425(17)30209-7. [PMID: 28943227 DOI: 10.1016/j.rmr.2017.07.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- V Cottin
- Centre national de référence des maladies pulmonaires rares, pneumologie, hôpital Louis-Pradel, hospices civils de Lyon, université Claude-Bernard-Lyon 1, Lyon, France.
| | - B Crestani
- Service de pneumologie A, centre de compétences pour les maladies pulmonaires rares, CHU Bichat, université Paris Diderot, Paris, France
| | - J Cadranel
- Service de pneumologie et oncologie thoracique, centre de compétences pour les maladies pulmonaires rares, hôpital Tenon, université Pierre-et-Marie-Curie, Paris 6, GH-HUEP, Assistance publique-Hôpitaux de Paris, Paris, France
| | - J-F Cordier
- Centre national de référence des maladies pulmonaires rares, pneumologie, hôpital Louis-Pradel, hospices civils de Lyon, université Claude-Bernard-Lyon 1, Lyon, France
| | - S Marchand-Adam
- Service de pneumologie, centre de compétences pour les maladies pulmonaires rares, CHU de Tours, Tours, France
| | - G Prévot
- Service de pneumologie, centre de compétences pour les maladies pulmonaires rares, CHU Larrey, Toulouse, France
| | - B Wallaert
- Service de pneumologie et immuno-allergologie, centre de compétences pour les maladies pulmonaires rares, hôpital Calmette, CHRU de Lille, Lille, France
| | - E Bergot
- Service de pneumologie et oncologie thoracique, centre de compétences pour les maladies pulmonaires rares, CHU de Caen, Caen, France
| | - P Camus
- Service de pneumologie et oncologie thoracique, centre de compétences pour les maladies pulmonaires rares, CHU Dijon-Bourgogne, Dijon, France
| | - J-C Dalphin
- Service de pneumologie, allergologie et oncologie thoracique, centre de compétences pour les maladies pulmonaires rares, hôpital Jean-Minjoz, CHRU de Besançon, Besançon, France
| | - C Dromer
- Service de pneumologie, centre de compétences pour les maladies pulmonaires rares, hôpital Haut-Lévèque, CHU de Bordeaux, Bordeaux, France
| | - E Gomez
- Département de pneumologie, centre de compétences pour les maladies pulmonaires rares, CHU de Nancy, Vandœuvre-lès-Nancy, France
| | - D Israel-Biet
- Service de pneumologie, centre de compétences pour les maladies pulmonaires rares, hôpital européen Georges-Pompidou, université Paris-Descartes, Paris, France
| | - S Jouneau
- Service de pneumologie, centre de compétences pour les maladies pulmonaires rares, CHU de Rennes, IRSET UMR 1085, université de Rennes 1, Rennes, France
| | - R Kessler
- Service de pneumologie, centre de compétences pour les maladies pulmonaires rares, hôpital civil, CHU de Strasbourg, Strasbourg, France
| | - C-H Marquette
- Service de pneumologie, centre de compétences pour les maladies pulmonaires rares, CHU de Nice, FHU Oncoage, université Côte d'Azur, France
| | - M Reynaud-Gaubert
- Service de pneumologie, centre de compétence des maladies pulmonaires rares, CHU Nord, Marseille, France
| | | | - D Bonnet
- Service de pneumologie, centre hospitalier de la Côte-Basque, Bayonne, France
| | - P Carré
- Service de pneumologie, centre hospitalier, Carcassonne, France
| | - C Danel
- Département de pathologie, hôpital Bichat-Claude-Bernard, université Paris Diderot, Assistance publique-Hôpitaux de Paris, Paris 7, Paris, France
| | - J-B Faivre
- Service d'imagerie thoracique, hôpital Calmette, CHRU de Lille, Lille, France
| | - G Ferretti
- Clinique universitaire de radiologie et imagerie médicale, CHU Grenoble-Alpes, Grenoble, France
| | - N Just
- Service de pneumologie, centre hospitalier Victor-Provo, Roubaix, France
| | - F Lebargy
- Service des maladies respiratoires, CHU Maison-Blanche, Reims, France
| | - B Philippe
- Service de pneumologie, centre hospitalier René-Dubos, Pontoise, France
| | - P Terrioux
- Service de pneumologie, centre hospitalier de Meaux, Meaux, France
| | - F Thivolet-Béjui
- Service d'anatomie et cytologie pathologiques, hôpital Louis-Pradel, Lyon, France
| | | | - D Valeyre
- Service de pneumologie, centre de compétences pour les maladies pulmonaires rares, hôpital Avicenne, CHU Paris-Seine-Saint-Denis, Bobigny, France
| |
Collapse
|
250
|
French practical guidelines for the diagnosis and management of idiopathic pulmonary fibrosis - 2017 update. Short-length version. Rev Mal Respir 2017; 34:852-899. [PMID: 28939154 DOI: 10.1016/j.rmr.2017.07.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|