1
|
Rieder F, Nagy LE, Maher TM, Distler JHW, Kramann R, Hinz B, Prunotto M. Fibrosis: cross-organ biology and pathways to development of innovative drugs. Nat Rev Drug Discov 2025:10.1038/s41573-025-01158-9. [PMID: 40102636 DOI: 10.1038/s41573-025-01158-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/10/2025] [Indexed: 03/20/2025]
Abstract
Fibrosis is a pathophysiological mechanism involved in chronic and progressive diseases that results in excessive tissue scarring. Diseases associated with fibrosis include metabolic dysfunction-associated steatohepatitis (MASH), inflammatory bowel diseases (IBDs), chronic kidney disease (CKD), idiopathic pulmonary fibrosis (IPF) and systemic sclerosis (SSc), which are collectively responsible for substantial morbidity and mortality. Although a few drugs with direct antifibrotic activity are approved for pulmonary fibrosis and considerable progress has been made in the understanding of mechanisms of fibrosis, translation of this knowledge into effective therapies continues to be limited and challenging. With the aim of assisting developers of novel antifibrotic drugs, this Review integrates viewpoints of biologists and physician-scientists on core pathways involved in fibrosis across organs, as well as on specific characteristics and approaches to assess therapeutic interventions for fibrotic diseases of the lung, gut, kidney, skin and liver. This discussion is used as a basis to propose strategies to improve the translation of potential antifibrotic therapies.
Collapse
Affiliation(s)
- Florian Rieder
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.
- Department of Gastroenterology, Hepatology and Nutrition, Digestive Disease Institute, Cleveland Clinic, Cleveland, OH, USA.
- Program for Global Translational Inflammatory Bowel Diseases (GRID), Chicago, IL, USA.
| | - Laura E Nagy
- Department of Gastroenterology, Hepatology and Nutrition, Digestive Disease Institute, Cleveland Clinic, Cleveland, OH, USA
- Northern Ohio Alcohol Center, Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, OH, USA
| | - Toby M Maher
- Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- National Heart and Lung Institute, Imperial College, London, UK
| | - Jörg H W Distler
- Department of Rheumatology, University Hospital Düsseldorf, Heinrich-Heine-University, Düsseldorf, Germany
- Hiller Research Center, University Hospital Düsseldorf, Heinrich-Heine-University, Düsseldorf, Germany
| | - Rafael Kramann
- Department of Nephrology and Clinical Immunology, RWTH Aachen; Medical Faculty, Aachen, Germany
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus Medical Center, Rotterdam, Netherlands
| | - Boris Hinz
- Keenan Research Institute for Biomedical Science of the St Michael's Hospital, Toronto, Ontario, Canada
- Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Marco Prunotto
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland.
| |
Collapse
|
2
|
Fotook Kiaei SZ, Schwartz DA. Genetic underpinning of idiopathic pulmonary fibrosis: the role of mucin. Expert Rev Respir Med 2025:1-12. [PMID: 39912527 DOI: 10.1080/17476348.2025.2464035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 12/27/2024] [Accepted: 02/04/2025] [Indexed: 02/07/2025]
Abstract
INTRODUCTION Idiopathic pulmonary fibrosis (IPF) is a chronic lung disease characterized by progressive scarring and reduced survival. The development of IPF is influenced by rare and common genetic variants, cigarette smoking, aging, and environmental exposures. Among the two dozen genetic contributors, the MUC5B promoter variant (rs35705950) is the dominant risk factor, increasing the risk of both familial and sporadic IPF and accounting for nearly 50% of the genetic predisposition to the disease. AREAS COVERED This review provides an expert perspective on the genetic underpinnings of IPF rather than a systematic analysis, emphasizing key insights into its genetic basis. The articles referenced in this review were identified through targeted searches in PubMed, Scopus, and Web of Science for studies published between 2000 and 2023, prioritizing influential research on the genetic factors contributing to IPF. Search terms included 'idiopathic pulmonary fibrosis,' 'genetics,' 'MUC5B,' 'telomere dysfunction,' and 'surfactant proteins.' The selection of studies was guided by the authors' expertise, focusing on the most relevant publications. EXPERT OPINION The identification of genetic variants not only highlights the complexity of IPF but also offers potential for earlier diagnosis and personalized treatment strategies targeting specific genetic pathways, ultimately aiming to improve patient outcomes.
Collapse
Affiliation(s)
| | - David A Schwartz
- Department of Medicine, University of Colorado Denver, School of Medicine, Aurora, CO, USA
| |
Collapse
|
3
|
Adegunsoye A, Kropski JA, Behr J, Blackwell TS, Corte TJ, Cottin V, Glanville AR, Glassberg MK, Griese M, Hunninghake GM, Johannson KA, Keane MP, Kim JS, Kolb M, Maher TM, Oldham JM, Podolanczuk AJ, Rosas IO, Martinez FJ, Noth I, Schwartz DA. Genetics and Genomics of Pulmonary Fibrosis: Charting the Molecular Landscape and Shaping Precision Medicine. Am J Respir Crit Care Med 2024; 210:401-423. [PMID: 38573068 PMCID: PMC11351799 DOI: 10.1164/rccm.202401-0238so] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 04/04/2024] [Indexed: 04/05/2024] Open
Abstract
Recent genetic and genomic advancements have elucidated the complex etiology of idiopathic pulmonary fibrosis (IPF) and other progressive fibrotic interstitial lung diseases (ILDs), emphasizing the contribution of heritable factors. This state-of-the-art review synthesizes evidence on significant genetic contributors to pulmonary fibrosis (PF), including rare genetic variants and common SNPs. The MUC5B promoter variant is unusual, a common SNP that markedly elevates the risk of early and established PF. We address the utility of genetic variation in enhancing understanding of disease pathogenesis and clinical phenotypes, improving disease definitions, and informing prognosis and treatment response. Critical research gaps are highlighted, particularly the underrepresentation of non-European ancestries in PF genetic studies and the exploration of PF phenotypes beyond usual interstitial pneumonia/IPF. We discuss the role of telomere length, often critically short in PF, and its link to progression and mortality, underscoring the genetic complexity involving telomere biology genes (TERT, TERC) and others like SFTPC and MUC5B. In addition, we address the potential of gene-by-environment interactions to modulate disease manifestation, advocating for precision medicine in PF. Insights from gene expression profiling studies and multiomic analyses highlight the promise for understanding disease pathogenesis and offer new approaches to clinical care, therapeutic drug development, and biomarker discovery. Finally, we discuss the ethical, legal, and social implications of genomic research and therapies in PF, stressing the need for sound practices and informed clinical genetic discussions. Looking forward, we advocate for comprehensive genetic testing panels and polygenic risk scores to improve the management of PF and related ILDs across diverse populations.
Collapse
Affiliation(s)
- Ayodeji Adegunsoye
- Pulmonary/Critical Care, and
- Committee on Clinical Pharmacology and Pharmacogenomics, University of Chicago, Chicago, Illinois
| | - Jonathan A. Kropski
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee
- Department of Veterans Affairs Medical Center, Nashville, Tennessee
| | - Juergen Behr
- Department of Medicine V, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
- Comprehensive Pneumology Center Munich, member of the German Center for Lung Research (DZL), Munich, Germany
| | - Timothy S. Blackwell
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee
- Department of Veterans Affairs Medical Center, Nashville, Tennessee
| | - Tamera J. Corte
- Centre of Research Excellence in Pulmonary Fibrosis, Camperdown, New South Wales, Australia
- Department of Respiratory and Sleep Medicine, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
- University of Sydney, Sydney, New South Wales, Australia
| | - Vincent Cottin
- National Reference Center for Rare Pulmonary Diseases (OrphaLung), Louis Pradel Hospital, Hospices Civils de Lyon, ERN-LUNG (European Reference Network on Rare Respiratory Diseases), Lyon, France
- Claude Bernard University Lyon, Lyon, France
| | - Allan R. Glanville
- Lung Transplant Unit, St. Vincent’s Hospital Sydney, Sydney, New South Wales, Australia
| | - Marilyn K. Glassberg
- Department of Medicine, Loyola Chicago Stritch School of Medicine, Chicago, Illinois
| | - Matthias Griese
- Department of Pediatric Pneumology, Dr. von Hauner Children’s Hospital, Ludwig-Maximilians-University, German Center for Lung Research, Munich, Germany
| | - Gary M. Hunninghake
- Harvard Medical School, Boston, Massachusetts
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
| | | | - Michael P. Keane
- Department of Respiratory Medicine, St. Vincent’s University Hospital and School of Medicine, University College Dublin, Dublin, Ireland
| | - John S. Kim
- Department of Medicine, School of Medicine, and
| | - Martin Kolb
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Toby M. Maher
- Department of Medicine, Keck School of Medicine of University of Southern California, Los Angeles, California
- National Heart and Lung Institute, Imperial College, London, United Kingdom
| | - Justin M. Oldham
- Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, Michigan
| | | | | | - Fernando J. Martinez
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Weill Cornell Medicine, New York, New York; and
| | - Imre Noth
- Division of Pulmonary and Critical Care Medicine, University of Virginia, Charlottesville, Virginia
| | - David A. Schwartz
- Department of Medicine, School of Medicine, University of Colorado, Aurora, Colorado
| |
Collapse
|
4
|
Roodnat AW, Callaghan B, Doyle C, Vallabh NA, Atkinson SD, Willoughby CE. Genome-wide RNA sequencing of ocular fibroblasts from glaucomatous and normal eyes: Implications for glaucoma management. PLoS One 2024; 19:e0307227. [PMID: 38990974 PMCID: PMC11239048 DOI: 10.1371/journal.pone.0307227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 06/28/2024] [Indexed: 07/13/2024] Open
Abstract
Primary open angle glaucoma is a leading cause of visual impairment and blindness which is commonly treated with drugs or laser but may require surgery. Tenon's ocular fibroblasts are involved in wound-healing after glaucoma filtration surgery and may compromise a favourable outcome of glaucoma surgery by contributing to fibrosis. To investigate changes in gene expression and key pathways contributing to the glaucomatous state we performed genome-wide RNA sequencing. Human Tenon's ocular fibroblasts were cultured from normal and glaucomatous human donors undergoing eye surgery (n = 12). mRNA was extracted and RNA-Seq performed on the Illumina platform. Differentially expressed genes were identified using a bioinformatics pipeline consisting of FastQC, STAR, FeatureCounts and edgeR. Changes in biological functions and pathways were determined using Enrichr and clustered using Cytoscape. A total of 5817 genes were differentially expressed between Tenon's ocular fibroblasts from normal versus glaucomatous eyes. Enrichment analysis showed 787 significantly different biological functions and pathways which were clustered into 176 clusters. Tenon's ocular fibroblasts from glaucomatous eyes showed signs of fibrosis with fibroblast to myofibroblast transdifferentiation and associated changes in mitochondrial fission, remodeling of the extracellular matrix, proliferation, unfolded protein response, inflammation and apoptosis which may relate to the pathogenesis of glaucoma or the detrimental effects of topical glaucoma therapies. Altered gene expression in glaucomatous Tenon's ocular fibroblasts may contribute to an unfavourable outcome of glaucoma filtration surgery. This work presents a genome-wide transcriptome of glaucomatous versus normal Tenon's ocular fibroblasts which may identify genes or pathways of therapeutic value to improve surgical outcomes.
Collapse
Affiliation(s)
- Anton W. Roodnat
- Centre for Genomic Medicine, Biomedical Sciences Research Institute, Ulster University, Coleraine, Northern Ireland, United Kingdom
| | - Breedge Callaghan
- Centre for Genomic Medicine, Biomedical Sciences Research Institute, Ulster University, Coleraine, Northern Ireland, United Kingdom
| | - Chelsey Doyle
- Centre for Genomic Medicine, Biomedical Sciences Research Institute, Ulster University, Coleraine, Northern Ireland, United Kingdom
| | - Neeru A. Vallabh
- Department of Eye and Vision Science, Insitute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
- St. Paul’s Eye Unit, Liverpool University Hospital NHS Foundation Trust, Liverpool, United Kingdom
| | - Sarah D. Atkinson
- Centre for Genomic Medicine, Biomedical Sciences Research Institute, Ulster University, Coleraine, Northern Ireland, United Kingdom
| | - Colin E. Willoughby
- Centre for Genomic Medicine, Biomedical Sciences Research Institute, Ulster University, Coleraine, Northern Ireland, United Kingdom
| |
Collapse
|
5
|
Singh P, Guin D, Pattnaik B, Kukreti R. Mapping the genetic architecture of idiopathic pulmonary fibrosis: Meta-analysis and epidemiological evidence of case-control studies. Gene 2024; 895:147993. [PMID: 37977320 DOI: 10.1016/j.gene.2023.147993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 10/23/2023] [Accepted: 11/13/2023] [Indexed: 11/19/2023]
Abstract
BACKGROUND Idiopathic pulmonary fibrosis (IPF) is a rare and devastating fibrotic lung disorder with unknown etiology. Although it is believed that genetic component is an important risk factor for IPF, a comprehensive understanding of its genetic landscape is lacking. Hence, we aimed to highlight the susceptibility genes and pathways implicated in IPF pathogenesis through a two-staged systematic literature search of genetic association studies on IPF, followed by meta-analysis and pathway enrichment analysis. METHODS This study was performed based on PRISMA guidelines (PROSPERO, registration number: CRD42022297970). The first search was performed (using PubMed and Web of Science) retrieving a total of 5642 articles, of which 52 were eligible for inclusion in the first stage. The second search was performed (using PubMed, Web of Science and Scopus) for ten polymorphisms, identified from the first search, with 2 or more studies. Finally, seven polymorphisms, [rs35705950/MUC5B, rs2736100/TERT, rs2609255/FAM13A, rs2076295/DSP, rs12610495/DPP9, rs111521887/TOLLIP and rs1800470/TGF-β1] qualified for meta-analyses. The epidemiological credibility was evaluated using Venice criteria. RESULTS From the systematic review, 222 polymorphisms in 118 genes showed a significant association with IPF susceptibility. Meta-analyses findings revealed significant association of rs35705950/T [OR = 3.92(3.26-4.57)], rs2609255/G [OR = 1.50(1.18-1.82)], rs2076295/G [OR = 1.19(0.82-1.756)], rs12610495/G [OR = 1.28(1.12-1.44)], rs2736100/C [OR = 0.68(0.54-0.82), rs111521887/G [OR = 1.34(1.06-1.61)] and suggestive evidence for rs1800470/T [OR = 1.08(0.82-1.34)] with IPF susceptibility. Four polymorphisms- rs35705950/MUC5B, rs2736100/TERT, rs2076295/DSP and rs111521887/TOLLIP, exhibited substantial epidemiological evidence supporting their association with IPF risk. Gene ontology and pathway enrichment analysis performed on IPF risk-associated genes identified a critical role of genes in mucin production, immune response and inflammation, host defence, cell-cell adhesion and telomere maintenance. CONCLUSIONS Our findings present the most prominent IPF-associated genetic risk variants involved in alveolar epithelial injuries (MUC5B, TERT, FAM13A, DSP, DPP9) and epithelial-mesenchymal transition (TOLLIP, TGF-β1), providing genetic and biological insights into IPF pathogenesis. However, further experimental research and human studies with larger sample sizes, diverse ethnic representation, and rigorous design are warranted.
Collapse
Affiliation(s)
- Pooja Singh
- Academy of Scientific and Innovative Research (AcSIR), CSIR-HRDC, Ghaziabad, Uttar Pradesh, India; Genomics and Molecular Medicine Unit, CSIR-Institute of Genomics and Integrative Biology (IGIB), New Delhi, India
| | - Debleena Guin
- Department of Biotechnology, Delhi Technological University, Shahbad Daulatpur, Main Bawana Road, New Delhi, India; Genomics and Molecular Medicine Unit, CSIR-Institute of Genomics and Integrative Biology (IGIB), New Delhi, India
| | - Bijay Pattnaik
- Centre of Excellence for Translational Research in Asthma and Lung Diseases, CSIR-Institute of Genomics and Integrative Biology (IGIB), New Delhi, India; Department of Pulmonary, Critical Care and Sleep Medicine, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Ritushree Kukreti
- Academy of Scientific and Innovative Research (AcSIR), CSIR-HRDC, Ghaziabad, Uttar Pradesh, India; Genomics and Molecular Medicine Unit, CSIR-Institute of Genomics and Integrative Biology (IGIB), New Delhi, India.
| |
Collapse
|
6
|
Millá E, Ventura-Abreu N, Vendrell C, Muniesa MJ, Pazos M, Gasull X, Comes N. Differential Gene and Protein Expression of Conjunctival Bleb Hyperfibrosis in Early Failure of Glaucoma Surgery. Int J Mol Sci 2023; 24:11949. [PMID: 37569323 PMCID: PMC10418990 DOI: 10.3390/ijms241511949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 07/20/2023] [Indexed: 08/13/2023] Open
Abstract
The early failure of glaucoma surgery is mainly caused by over-fibrosis at the subconjunctival space, causing obliteration of the filtration bleb. Because fibrosis has a suspected basis of genetic predisposition, we have undertaken a prospective study to identify upregulated profibrotic genes in a population of glaucoma patients with signs of conjunctival fibrosis and early postoperative surgical failure. Clinical data of re-operated fibrosis patients, hyperfibrosis patients who re-operated more than once in a short time, and control patients with no fibrosis were recorded and analyzed at each follow-up visit. Conjunctival-Tenon surgical specimens were obtained intraoperatively to evaluate the local expression of a panel of genes potentially associated with fibrosis. In order to correlate gene expression signatures with protein levels, we quantified secreted proteins in primary cultures of fibroblasts from patients. Expression of VEGFA, CXCL8, MYC, and CDKN1A was induced in the conjunctiva of hyperfibrosis patients. VEGFA and IL8 protein levels were also increased in fibroblast supernatants. We propose that an increase in these proteins could be useful in detecting conjunctival fibrosis in glaucoma patients undergoing filtering surgery. Molecular markers could be crucial for early detection of patients at high risk of failure of filtration surgery, leading to more optimal and personalized treatments.
Collapse
Affiliation(s)
- Elena Millá
- Hospital Clínic de Barcelona, Institut Clinic d'Oftalmologia, ICOF, Sabino Arana nº1, 08028 Barcelona, Spain
- Institut Comtal d'Oftalmologia, Innova Ocular-ICO Barcelona, Via Augusta 48, 08006 Barcelona, Spain
| | | | - Cristina Vendrell
- Institut Comtal d'Oftalmologia, Innova Ocular-ICO Barcelona, Via Augusta 48, 08006 Barcelona, Spain
- Hospital de Viladecans, Avda. Gavà 38, 08840 Barcelona, Spain
| | - Maria Jesús Muniesa
- Hospital Clínic de Barcelona, Institut Clinic d'Oftalmologia, ICOF, Sabino Arana nº1, 08028 Barcelona, Spain
| | - Marta Pazos
- Hospital Clínic de Barcelona, Institut Clinic d'Oftalmologia, ICOF, Sabino Arana nº1, 08028 Barcelona, Spain
| | - Xavier Gasull
- Neurophysiology Laboratory, Department of Biomedicine, Medical School, University of Barcelona, Casanova 143, 08036 Barcelona, Spain
- Institute of Neurosciences, University of Barcelona, Edifici de Ponent, 2n vagó 3r pis, Passeig de la Vall d'Hebron 171, 08035 Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Rosselló 149, 08036 Barcelona, Spain
| | - Núria Comes
- Neurophysiology Laboratory, Department of Biomedicine, Medical School, University of Barcelona, Casanova 143, 08036 Barcelona, Spain
- Institute of Neurosciences, University of Barcelona, Edifici de Ponent, 2n vagó 3r pis, Passeig de la Vall d'Hebron 171, 08035 Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Rosselló 149, 08036 Barcelona, Spain
| |
Collapse
|
7
|
Kraven LM, Taylor AR, Molyneaux PL, Maher TM, McDonough JE, Mura M, Yang IV, Schwartz DA, Huang Y, Noth I, Ma SF, Yeo AJ, Fahy WA, Jenkins RG, Wain LV. Cluster analysis of transcriptomic datasets to identify endotypes of idiopathic pulmonary fibrosis. Thorax 2023; 78:551-558. [PMID: 35534152 PMCID: PMC9643664 DOI: 10.1136/thoraxjnl-2021-218563] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 04/14/2022] [Indexed: 11/04/2022]
Abstract
BACKGROUND Considerable clinical heterogeneity in idiopathic pulmonary fibrosis (IPF) suggests the existence of multiple disease endotypes. Identifying these endotypes would improve our understanding of the pathogenesis of IPF and could allow for a biomarker-driven personalised medicine approach. We aimed to identify clinically distinct groups of patients with IPF that could represent distinct disease endotypes. METHODS We co-normalised, pooled and clustered three publicly available blood transcriptomic datasets (total 220 IPF cases). We compared clinical traits across clusters and used gene enrichment analysis to identify biological pathways and processes that were over-represented among the genes that were differentially expressed across clusters. A gene-based classifier was developed and validated using three additional independent datasets (total 194 IPF cases). FINDINGS We identified three clusters of patients with IPF with statistically significant differences in lung function (p=0.009) and mortality (p=0.009) between groups. Gene enrichment analysis implicated mitochondrial homeostasis, apoptosis, cell cycle and innate and adaptive immunity in the pathogenesis underlying these groups. We developed and validated a 13-gene cluster classifier that predicted mortality in IPF (high-risk clusters vs low-risk cluster: HR 4.25, 95% CI 2.14 to 8.46, p=3.7×10-5). INTERPRETATION We have identified blood gene expression signatures capable of discerning groups of patients with IPF with significant differences in survival. These clusters could be representative of distinct pathophysiological states, which would support the theory of multiple endotypes of IPF. Although more work must be done to confirm the existence of these endotypes, our classifier could be a useful tool in patient stratification and outcome prediction in IPF.
Collapse
Affiliation(s)
- Luke M Kraven
- Department of Health Sciences, University of Leicester, Leicester, UK
- Research & Development, GlaxoSmithKline, Stevenage, UK
| | - Adam R Taylor
- Research & Development, GlaxoSmithKline, Stevenage, UK
| | - Philip L Molyneaux
- Guy's and St Thomas' NHS Foundation Trust, Royal Brompton and Harefield Hospitals, London, UK
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Toby M Maher
- Guy's and St Thomas' NHS Foundation Trust, Royal Brompton and Harefield Hospitals, London, UK
- National Heart and Lung Institute, Imperial College London, London, UK
- Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - John E McDonough
- Division of Pulmonary, Critical Care & Sleep Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Marco Mura
- Division of Respirology, Western University, London, Ontario, Canada
| | - Ivana V Yang
- Department of Medicine, University of Colorado, Denver, Colorado, USA
| | - David A Schwartz
- Department of Medicine, University of Colorado, Denver, Colorado, USA
| | - Yong Huang
- Division of Pulmonary & Critical Care Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Imre Noth
- Division of Pulmonary & Critical Care Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Shwu Fan Ma
- Division of Pulmonary & Critical Care Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Astrid J Yeo
- Research & Development, GlaxoSmithKline, Stevenage, UK
| | | | - R Gisli Jenkins
- National Heart and Lung Institute, Imperial College London, London, UK
- National Institute for Health Research Respiratory Clinical Research Facility, Royal Brompton Hospital, London, UK
| | - Louise V Wain
- Department of Health Sciences, University of Leicester, Leicester, UK
- National Institute for Health Research, Glenfield Hospital, Leicester, UK
| |
Collapse
|
8
|
Drake C, Wehr MM, Zobl W, Koschmann J, De Lucca D, Kühne BA, Hansen T, Knebel J, Ritter D, Boei J, Vrieling H, Bitsch A, Escher SE. Substantiate a read-across hypothesis by using transcriptome data-A case study on volatile diketones. FRONTIERS IN TOXICOLOGY 2023; 5:1155645. [PMID: 37206915 PMCID: PMC10188990 DOI: 10.3389/ftox.2023.1155645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 04/17/2023] [Indexed: 05/21/2023] Open
Abstract
This case study explores the applicability of transcriptome data to characterize a common mechanism of action within groups of short-chain aliphatic α-, β-, and γ-diketones. Human reference in vivo data indicate that the α-diketone diacetyl induces bronchiolitis obliterans in workers involved in the preparation of microwave popcorn. The other three α-diketones induced inflammatory responses in preclinical in vivo animal studies, whereas beta and gamma diketones in addition caused neuronal effects. We investigated early transcriptional responses in primary human bronchiolar (PBEC) cell cultures after 24 h and 72 h of air-liquid exposure. Differentially expressed genes (DEGs) were assessed based on transcriptome data generated with the EUToxRisk gene panel of Temp-O-Seq®. For each individual substance, genes were identified displaying a consistent differential expression across dose and exposure duration. The log fold change values of the DEG profiles indicate that α- and β-diketones are more active compared to γ-diketones. α-diketones in particular showed a highly concordant expression pattern, which may serve as a first indication of the shared mode of action. In order to gain a better mechanistic understanding, the resultant DEGs were submitted to a pathway analysis using ConsensusPathDB. The four α-diketones showed very similar results with regard to the number of activated and shared pathways. Overall, the number of signaling pathways decreased from α-to β-to γ-diketones. Additionally, we reconstructed networks of genes that interact with one another and are associated with different adverse outcomes such as fibrosis, inflammation or apoptosis using the TRANSPATH-database. Transcription factor enrichment and upstream analyses with the geneXplain platform revealed highly interacting gene products (called master regulators, MRs) per case study compound. The mapping of the resultant MRs on the reconstructed networks, visualized similar gene regulation with regard to fibrosis, inflammation and apoptosis. This analysis showed that transcriptome data can strengthen the similarity assessment of compounds, which is of particular importance, e.g., in read-across approaches. It is one important step towards grouping of compounds based on biological profiles.
Collapse
Affiliation(s)
- Christina Drake
- Fraunhofer Institute for Toxicology and Experimental Medicine, Chemical Safety and Toxicology, Hannover, Germany
- *Correspondence: Christina Drake,
| | - Matthias M. Wehr
- Fraunhofer Institute for Toxicology and Experimental Medicine, Chemical Safety and Toxicology, Hannover, Germany
| | - Walter Zobl
- Fraunhofer Institute for Toxicology and Experimental Medicine, Chemical Safety and Toxicology, Hannover, Germany
| | | | | | - Britta A. Kühne
- Fraunhofer Institute for Toxicology and Experimental Medicine, Chemical Safety and Toxicology, Hannover, Germany
| | - Tanja Hansen
- Fraunhofer Institute for Toxicology and Experimental Medicine, Chemical Safety and Toxicology, Hannover, Germany
| | - Jan Knebel
- Fraunhofer Institute for Toxicology and Experimental Medicine, Chemical Safety and Toxicology, Hannover, Germany
| | - Detlef Ritter
- Fraunhofer Institute for Toxicology and Experimental Medicine, Chemical Safety and Toxicology, Hannover, Germany
| | - Jan Boei
- Leiden University Medical Center, Leiden, Netherlands
| | | | - Annette Bitsch
- Fraunhofer Institute for Toxicology and Experimental Medicine, Chemical Safety and Toxicology, Hannover, Germany
| | - Sylvia E. Escher
- Fraunhofer Institute for Toxicology and Experimental Medicine, Chemical Safety and Toxicology, Hannover, Germany
| |
Collapse
|
9
|
Papiris SA, Kannengiesser C, Borie R, Kolilekas L, Kallieri M, Apollonatou V, Ba I, Nathan N, Bush A, Griese M, Dieude P, Crestani B, Manali ED. Genetics in Idiopathic Pulmonary Fibrosis: A Clinical Perspective. Diagnostics (Basel) 2022; 12:2928. [PMID: 36552935 PMCID: PMC9777433 DOI: 10.3390/diagnostics12122928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/17/2022] [Accepted: 11/18/2022] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Unraveling the genetic background in a significant proportion of patients with both sporadic and familial IPF provided new insights into the pathogenic pathways of pulmonary fibrosis. AIM The aim of the present study is to overview the clinical significance of genetics in IPF. PERSPECTIVE It is fascinating to realize the so-far underestimated but dynamically increasing impact that genetics has on aspects related to the pathophysiology, accurate and early diagnosis, and treatment and prevention of this devastating disease. Genetics in IPF have contributed as no other in unchaining the disease from the dogma of a "a sporadic entity of the elderly, limited to the lungs" and allowed all scientists, but mostly clinicians, all over the world to consider its many aspects and "faces" in all age groups, including its co-existence with several extra pulmonary conditions from cutaneous albinism to bone-marrow and liver failure. CONCLUSION By providing additional evidence for unsuspected characteristics such as immunodeficiency, impaired mucus, and surfactant and telomere maintenance that very often co-exist through the interaction of common and rare genetic variants in the same patient, genetics have created a generous and pluralistic yet unifying platform that could lead to the understanding of the injurious and pro-fibrotic effects of many seemingly unrelated extrinsic and intrinsic offending factors. The same platform constantly instructs us about our limitations as well as about the heritability, the knowledge and the wisdom that is still missing.
Collapse
Affiliation(s)
- Spyros A. Papiris
- 2nd Pulmonary Medicine Department, General University Hospital “Attikon”, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Caroline Kannengiesser
- Département de Génétique, APHP Hôpital Bichat, Université de Paris, 75018 Paris, France
- INSERM UMR 1152, Université de Paris, 75018 Paris, France
| | - Raphael Borie
- Service de Pneumologie A, INSERM UMR_1152, Centre de Référence des Maladies Pulmonaires Rares, FHU APOLLO, APHP Hôpital Bichat, Sorbonne Université, 75018 Paris, France
| | - Lykourgos Kolilekas
- 7th Pulmonary Department, Athens Chest Hospital “Sotiria”, 11527 Athens, Greece
| | - Maria Kallieri
- 2nd Pulmonary Medicine Department, General University Hospital “Attikon”, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Vasiliki Apollonatou
- 2nd Pulmonary Medicine Department, General University Hospital “Attikon”, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Ibrahima Ba
- Département de Génétique, APHP Hôpital Bichat, Université de Paris, 75018 Paris, France
| | - Nadia Nathan
- Peditric Pulmonology Department and Reference Centre for Rare Lung Diseases RespiRare, INSERM UMR_S933 Laboratory of Childhood Genetic Diseases, Armand Trousseau Hospital, Sorbonne University and APHP, 75012 Paris, France
| | - Andrew Bush
- Paediatrics and Paediatric Respirology, Imperial College, Imperial Centre for Paediatrics and Child Health, Royal Brompton Harefield NHS Foundation Trust, London SW3 6NP, UK
| | - Matthias Griese
- Department of Pediatric Pneumology, Dr von Hauner Children’s Hospital, Ludwig-Maximilians-University, German Center for Lung Research, 80337 Munich, Germany
| | - Philippe Dieude
- Department of Rheumatology, INSERM U1152, APHP Hôpital Bichat-Claude Bernard, Université de Paris, 75018 Paris, France
| | - Bruno Crestani
- Service de Pneumologie A, INSERM UMR_1152, Centre de Référence des Maladies Pulmonaires Rares, FHU APOLLO, APHP Hôpital Bichat, Sorbonne Université, 75018 Paris, France
| | - Effrosyni D. Manali
- 2nd Pulmonary Medicine Department, General University Hospital “Attikon”, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece
| |
Collapse
|
10
|
Hong X, Wang L, Zhang K, Liu J, Liu JP. Molecular Mechanisms of Alveolar Epithelial Stem Cell Senescence and Senescence-Associated Differentiation Disorders in Pulmonary Fibrosis. Cells 2022; 11:877. [PMID: 35269498 PMCID: PMC8909789 DOI: 10.3390/cells11050877] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/01/2022] [Accepted: 03/02/2022] [Indexed: 02/04/2023] Open
Abstract
Pulmonary senescence is accelerated by unresolved DNA damage response, underpinning susceptibility to pulmonary fibrosis. Recently it was reported that the SARS-Cov-2 viral infection induces acute pulmonary epithelial senescence followed by fibrosis, although the mechanism remains unclear. Here, we examine roles of alveolar epithelial stem cell senescence and senescence-associated differentiation disorders in pulmonary fibrosis, exploring the mechanisms mediating and preventing pulmonary fibrogenic crisis. Notably, the TGF-β signalling pathway mediates alveolar epithelial stem cell senescence by mechanisms involving suppression of the telomerase reverse transcriptase gene in pulmonary fibrosis. Alternatively, telomere uncapping caused by stress-induced telomeric shelterin protein TPP1 degradation mediates DNA damage response, pulmonary senescence and fibrosis. However, targeted intervention of cellular senescence disrupts pulmonary remodelling and fibrosis by clearing senescent cells using senolytics or preventing senescence using telomere dysfunction inhibitor (TELODIN). Studies indicate that the development of senescence-associated differentiation disorders is reprogrammable and reversible by inhibiting stem cell replicative senescence in pulmonary fibrosis, providing a framework for targeted intervention of the molecular mechanisms of alveolar stem cell senescence and pulmonary fibrosis. Abbreviations: DPS, developmental programmed senescence; IPF, idiopathic pulmonary fibrosis; OIS, oncogene-induced replicative senescence; SADD, senescence-associated differentiation disorder; SALI, senescence-associated low-grade inflammation; SIPS, stress-induced premature senescence; TERC, telomerase RNA component; TERT, telomerase reverse transcriptase; TIFs, telomere dysfunction-induced foci; TIS, therapy-induced senescence; VIS, virus-induced senescence.
Collapse
Affiliation(s)
- Xiaojing Hong
- Institute of Ageing Research, Hangzhou Normal University School of Medicine, Hangzhou 311121, China; (X.H.); (L.W.); (K.Z.); (J.L.)
| | - Lihui Wang
- Institute of Ageing Research, Hangzhou Normal University School of Medicine, Hangzhou 311121, China; (X.H.); (L.W.); (K.Z.); (J.L.)
| | - Kexiong Zhang
- Institute of Ageing Research, Hangzhou Normal University School of Medicine, Hangzhou 311121, China; (X.H.); (L.W.); (K.Z.); (J.L.)
| | - Jun Liu
- Institute of Ageing Research, Hangzhou Normal University School of Medicine, Hangzhou 311121, China; (X.H.); (L.W.); (K.Z.); (J.L.)
| | - Jun-Ping Liu
- Institute of Ageing Research, Hangzhou Normal University School of Medicine, Hangzhou 311121, China; (X.H.); (L.W.); (K.Z.); (J.L.)
- Department of Immunology and Pathology, Monash University Faculty of Medicine, Prahran, VIC 3181, Australia
- Hudson Institute of Medical Research, Monash University Department of Molecular and Translational Science, Clayton, VIC 3168, Australia
| |
Collapse
|
11
|
Moss BJ, Ryter SW, Rosas IO. Pathogenic Mechanisms Underlying Idiopathic Pulmonary Fibrosis. ANNUAL REVIEW OF PATHOLOGY 2022; 17:515-546. [PMID: 34813355 DOI: 10.1146/annurev-pathol-042320-030240] [Citation(s) in RCA: 344] [Impact Index Per Article: 114.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The pathogenesis of idiopathic pulmonary fibrosis (IPF) involves a complex interplay of cell types and signaling pathways. Recurrent alveolar epithelial cell (AEC) injury may occur in the context of predisposing factors (e.g., genetic, environmental, epigenetic, immunologic, and gerontologic), leading to metabolic dysfunction, senescence, aberrant epithelial cell activation, and dysregulated epithelial repair. The dysregulated epithelial cell interacts with mesenchymal, immune, and endothelial cells via multiple signaling mechanisms to trigger fibroblast and myofibroblast activation. Recent single-cell RNA sequencing studies of IPF lungs support the epithelial injury model. These studies have uncovered a novel type of AEC with characteristics of an aberrant basal cell, which may disrupt normal epithelial repair and propagate a profibrotic phenotype. Here, we review the pathogenesis of IPF in the context of novel bioinformatics tools as strategies to discover pathways of disease, cell-specific mechanisms, and cell-cell interactions that propagate the profibrotic niche.
Collapse
Affiliation(s)
- Benjamin J Moss
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Baylor College of Medicine, Houston, Texas 77030, USA; ,
| | - Stefan W Ryter
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY 10021, USA;
| | - Ivan O Rosas
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Baylor College of Medicine, Houston, Texas 77030, USA; ,
| |
Collapse
|
12
|
Michalski JE, Schwartz DA. Genetic Risk Factors for Idiopathic Pulmonary Fibrosis: Insights into Immunopathogenesis. J Inflamm Res 2021; 13:1305-1318. [PMID: 33447070 PMCID: PMC7801923 DOI: 10.2147/jir.s280958] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Accepted: 12/18/2020] [Indexed: 12/12/2022] Open
Abstract
Idiopathic pulmonary fibrosis is an etiologically complex interstitial lung disease characterized by progressive scarring of the lungs with a subsequent decline in lung function. While much of the pathogenesis of IPF still remains unclear, it is now understood that genetic variation accounts for at least one-third of the risk of developing the disease. The single-most validated and most significant risk factor, genetic or otherwise, is a gain-of-function promoter variant in the MUC5B gene. While the functional impact of these IPF risk variants at the cellular and tissue levels are areas of active investigation, there is a growing body of evidence that these genetic variants may influence disease pathogenesis through modulation of innate immune processes.
Collapse
Affiliation(s)
- Jacob E Michalski
- Department of Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| | - David A Schwartz
- Department of Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| |
Collapse
|
13
|
Jiang Y, Xiang C, Zhong F, Zhang Y, Wang L, Zhao Y, Wang J, Ding C, Jin L, He F, Wang H. Histone H3K27 methyltransferase EZH2 and demethylase JMJD3 regulate hepatic stellate cells activation and liver fibrosis. Am J Cancer Res 2021; 11:361-378. [PMID: 33391480 PMCID: PMC7681085 DOI: 10.7150/thno.46360] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 09/09/2020] [Indexed: 02/06/2023] Open
Abstract
Rationale: As the central hallmark of liver fibrosis, transdifferentiation of hepatic stellate cells (HSCs), the predominant contributor to fibrogenic hepatic myofibroblast responsible for extracellular matrix (ECM) deposition, is characterized with transcriptional and epigenetic remodeling. We aimed to characterize the roles of H3K27 methyltransferase EZH2 and demethylase JMJD3 and identify their effective pathways and novel target genes in HSCs activation and liver fibrosis. Methods: In primary HSCs, we analyzed effects of pharmacological inhibitions and genetic manipulations of EZH2 and JMJD3 on HSCs activation. In HSCs cell lines, we evaluated effects of EZH2 inhibition by DZNep on proliferation, cell cycling, senescence and apoptosis. In CCl4 and BDL murine models of liver fibrosis, we assessed in vivo effects of DZNep administration and Ezh2 silencing. We profiled rat primary HSCs transcriptomes with RNA-seq, screened the pathways and genes associated with DZNep treatment, analyzed EZH2 and JMJD3 regulation towards target genes by ChIP-qPCR. Results: EZH2 inhibition by DZNep resulted in retarded growth, lowered cell viability, cell cycle arrest in S and G2 phases, strengthened senescence, and enhanced apoptosis of HSCs, decreased hepatic collagen deposition and rescued the elevated serum ALT and AST activities of diseased mice, and downregulated cellular and hepatic expressions of H3K27me3, EZH2, α-SMA and COL1A. Ezh2 silencing by RNA interference in vitro and in vivo showed similar effects. JMJD3 inhibition by GSK-J4 and overexpression of wild-type but not mutant Jmjd3 enhanced or repressed HSCs activation respectively. EZH2 inhibition by DZNep transcriptionally inactivated TGF-β1 pathway, cell cycle pathways and vast ECM components in primary HSCs. EZH2 inhibition decreased H3K27me3 recruitment at target genes encoding TGF-β1 pseudoreceptor BAMBI, anti-inflammatory cytokine IL10 and cell cycle regulators CDKN1A, GADD45A and GADD45B, and increased their expressions, while Jmjd3 overexpression manifested alike effects. Conclusions: EZH2 and JMJD3 antagonistically modulate HSCs activation. The therapeutic effects of DZNep as epigenetic drug in liver fibrosis are associated with the regulation of EZH2 towards direct target genes encoding TGF-β1 pseudoreceptor BAMBI, anti-inflammatory cytokine IL10 and cell cycle regulators CDKN1A, GADD45A and GADD45B, which are also regulated by JMJD3. Our present study provides new mechanistic insight into the epigenetic modulation of EZH2 and JMJD3 in HSCs biology and hepatic fibrogenesis.
Collapse
|
14
|
p53: A Key Protein That Regulates Pulmonary Fibrosis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:6635794. [PMID: 33312337 PMCID: PMC7721501 DOI: 10.1155/2020/6635794] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 11/05/2020] [Accepted: 11/20/2020] [Indexed: 02/06/2023]
Abstract
Pulmonary fibrosis is a progressively aggravating lethal disease that is a serious public health concern. Although the incidence of this disease is increasing, there is a lack of effective therapies. In recent years, the pathogenesis of pulmonary fibrosis has become a research hotspot. p53 is a tumor suppressor gene with crucial roles in cell cycle, apoptosis, tumorigenesis, and malignant transformation. Previous studies on p53 have predominantly focused on its role in neoplastic disease. Following in-depth investigation, several studies have linked it to pulmonary fibrosis. This review covers the association between p53 and pulmonary fibrosis, with the aim of providing novel ideas to improve the clinical diagnosis, treatment, and prognosis of pulmonary fibrosis.
Collapse
|
15
|
Huidobro C, Martín-Vicente P, López-Martínez C, Alonso-López I, Amado-Rodríguez L, Crespo I, M Albaiceta G. Cellular and molecular features of senescence in acute lung injury. Mech Ageing Dev 2020; 193:111410. [PMID: 33249191 DOI: 10.1016/j.mad.2020.111410] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 11/20/2020] [Indexed: 12/14/2022]
Abstract
A wide range of insults can trigger acute injury in the lungs, which eventually may lead to respiratory failure and death of patients. Current treatment relies mainly on supportive measures and mechanical ventilation. Even so, survivors frequently develop important sequels that compromise quality of life. In the search for new approaches to prevent and treat acute lung injury, many investigations have focused on molecular and cellular pathways which could exert a pathogenic role in this disease. Herein, we review recent findings in the literature suggesting that cellular senescence could be involved in lung injury and discuss the potential use of senotherapies to prevent disease progression.
Collapse
Affiliation(s)
- Covadonga Huidobro
- Instituto de Investigación Sanitaria del Principado de Asturias, Avenida de Roma s/n, 33011, Oviedo, Spain.
| | - Paula Martín-Vicente
- Instituto de Investigación Sanitaria del Principado de Asturias, Avenida de Roma s/n, 33011, Oviedo, Spain; Centro de Investigación Biomédica en Red (CIBER)-Enfermedades Respiratorias, Calle de Melchor Fernández Almagro, 3, 28029, Madrid, Spain; Instituto Universitario de Oncología del Principado de Asturias, Calle Fernando Bongera s/n, 33006, Oviedo, Spain
| | - Cecilia López-Martínez
- Instituto de Investigación Sanitaria del Principado de Asturias, Avenida de Roma s/n, 33011, Oviedo, Spain; Centro de Investigación Biomédica en Red (CIBER)-Enfermedades Respiratorias, Calle de Melchor Fernández Almagro, 3, 28029, Madrid, Spain; Instituto Universitario de Oncología del Principado de Asturias, Calle Fernando Bongera s/n, 33006, Oviedo, Spain
| | - Inés Alonso-López
- Instituto de Investigación Sanitaria del Principado de Asturias, Avenida de Roma s/n, 33011, Oviedo, Spain; Centro de Investigación Biomédica en Red (CIBER)-Enfermedades Respiratorias, Calle de Melchor Fernández Almagro, 3, 28029, Madrid, Spain; Instituto Universitario de Oncología del Principado de Asturias, Calle Fernando Bongera s/n, 33006, Oviedo, Spain
| | - Laura Amado-Rodríguez
- Instituto de Investigación Sanitaria del Principado de Asturias, Avenida de Roma s/n, 33011, Oviedo, Spain; Unidad de Cuidados Intensivos Cardiológicos, Hospital Universitario Central de Asturias, Oviedo, Avenida de Roma s/n, 33011, Oviedo, Spain
| | - Irene Crespo
- Departamento de Biología Funcional. Universidad de Oviedo, C/ Julián Clavería s/n, 33006, Oviedo, Spain
| | - Guillermo M Albaiceta
- Instituto de Investigación Sanitaria del Principado de Asturias, Avenida de Roma s/n, 33011, Oviedo, Spain; Centro de Investigación Biomédica en Red (CIBER)-Enfermedades Respiratorias, Calle de Melchor Fernández Almagro, 3, 28029, Madrid, Spain; Instituto Universitario de Oncología del Principado de Asturias, Calle Fernando Bongera s/n, 33006, Oviedo, Spain; Unidad de Cuidados Intensivos Cardiológicos, Hospital Universitario Central de Asturias, Oviedo, Avenida de Roma s/n, 33011, Oviedo, Spain; Departamento de Biología Funcional. Universidad de Oviedo, C/ Julián Clavería s/n, 33006, Oviedo, Spain
| |
Collapse
|
16
|
Dagneaux L, Owen AR, Bettencourt JW, Barlow JD, Amadio PC, Kocher JP, Morrey ME, Sanchez-Sotelo J, Berry DJ, van Wijnen AJ, Abdel MP. Human Fibrosis: Is There Evidence for a Genetic Predisposition in Musculoskeletal Tissues? J Arthroplasty 2020; 35:3343-3352. [PMID: 32593486 PMCID: PMC7842876 DOI: 10.1016/j.arth.2020.05.070] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 05/26/2020] [Accepted: 05/28/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Pathologic fibrosis is characterized by dysregulation of gene expression with excessive extracellular matrix production. The genetic basis for solid organ fibrosis is well described in the literature. However, there is a paucity of evidence for similar processes in the musculoskeletal (MSK) system. The purpose of this review is to provide an overview of existing evidence of genetic predisposition to pathologic fibrosis in the cardiac, pulmonary, and MSK systems, and to describe common genetic variants associated with these processes. METHODS A comprehensive search of several databases from 2000 to 2019 was conducted using relevant keywords in the English language. Genes reported as involved in idiopathic fibrotic processes in the heart, lung, hand, shoulder, and knee were recorded by 2 independent authors. RESULTS Among 2373 eligible studies, 52 studies investigated genetic predisposition in terms of variant analysis with the following organ system distribution: 36 pulmonary studies (69%), 15 hand studies (29%), and 1 knee study (2%). Twenty-two percent of gene variants identified were associated with both pulmonary and MSK fibrosis (ie, ADAM, HLA, CARD, EIF, TGF, WNT, and ZNF genes). Genetic variants known to be involved in the MSK tissue development or contractility properties in muscle were identified in the pulmonary fibrosis. CONCLUSION Despite shared genetic variations in both the lung and hand, there remains limited information about genetic variants associated with fibrosis in other MSK regions. This finding establishes the necessity of further studies to elucidate the genetic determinants involved in the knee, shoulder, and other joint fibrotic pathways. LEVEL OF EVIDENCE Level III.
Collapse
Affiliation(s)
- Louis Dagneaux
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN
| | - Aaron R. Owen
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN
| | | | | | - Peter C. Amadio
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN
| | - Jean P. Kocher
- Department of Bioinformatics, Mayo Clinic, Rochester, MN
| | - Mark E. Morrey
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN
| | | | - Daniel J. Berry
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN
| | | | - Matthew P. Abdel
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN,Reprint requests: Matthew P. Abdel, MD, Department of Orthopedic Surgery, Mayo Clinic, 200 First Street S.W., Rochester, MN 55905
| |
Collapse
|
17
|
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
|
18
|
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
|
19
|
Ballester B, Milara J, Cortijo J. Idiopathic Pulmonary Fibrosis and Lung Cancer: Mechanisms and Molecular Targets. Int J Mol Sci 2019; 20:ijms20030593. [PMID: 30704051 PMCID: PMC6387034 DOI: 10.3390/ijms20030593] [Citation(s) in RCA: 202] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 01/18/2019] [Accepted: 01/28/2019] [Indexed: 12/18/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is the most common idiopathic interstitial pulmonary disease with a median survival of 2–4 years after diagnosis. A significant number of IPF patients have risk factors, such as a history of smoking or concomitant emphysema, both of which can predispose the patient to lung cancer (LC) (mostly non-small cell lung cancer (NSCLC)). In fact, IPF itself increases the risk of LC development by 7% to 20%. In this regard, there are multiple common genetic, molecular, and cellular processes that connect lung fibrosis with LC, such as myofibroblast/mesenchymal transition, myofibroblast activation and uncontrolled proliferation, endoplasmic reticulum stress, alterations of growth factors expression, oxidative stress, and large genetic and epigenetic variations that can predispose the patient to develop IPF and LC. The current approved IPF therapies, pirfenidone and nintedanib, are also active in LC. In fact, nintedanib is approved as a second line treatment in NSCLC, and pirfenidone has shown anti-neoplastic effects in preclinical studies. In this review, we focus on the current knowledge on the mechanisms implicated in the development of LC in patients with IPF as well as in current IPF and LC-IPF candidate therapies based on novel molecular advances.
Collapse
Affiliation(s)
- Beatriz Ballester
- Department of Pharmacology, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain.
- CIBERES, Health Institute Carlos III, 28029 Valencia, Spain.
| | - Javier Milara
- CIBERES, Health Institute Carlos III, 28029 Valencia, Spain.
- Pharmacy Unit, University Clinic Hospital of Valencia, 46010 Valencia, Spain.
- Institute of Health Research-INCLIVA, 46010 Valencia, Spain.
| | - Julio Cortijo
- Department of Pharmacology, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain.
- CIBERES, Health Institute Carlos III, 28029 Valencia, Spain.
- Research and teaching Unit, University General Hospital Consortium, 46014 Valencia, Spain.
| |
Collapse
|
20
|
Hwang JA, Kim D, Chun SM, Bae S, Song JS, Kim MY, Koo HJ, Song JW, Kim WS, Lee JC, Kim HR, Choi CM, Jang SJ. Genomic profiles of lung cancer associated with idiopathic pulmonary fibrosis. J Pathol 2018; 244:25-35. [PMID: 28862766 DOI: 10.1002/path.4978] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 08/22/2017] [Accepted: 08/25/2017] [Indexed: 12/22/2022]
Abstract
Little is known about the pathogenesis or molecular profiles of idiopathic pulmonary fibrosis-associated lung cancer (IPF-LC). This study was performed to investigate the genomic profiles of IPF-LC and to explore the possibility of defining potential therapeutic targets in IPF-LC. We assessed genomic profiles of IPF-LC by using targeted exome sequencing (OncoPanel version 2) in 35 matched tumour/normal pairs surgically resected between 2004 and 2014. Germline and somatic variant calling was performed with GATK HaplotypeCaller and MuTect with GATK SomaticIndelocator, respectively. Copy number analysis was conducted with CNVkit, with focal events determined by Genomic Identification of Significant Targets in Cancer 2.0, and pathway analysis (KEGG) with DAVID. Germline mutations in TERT (rs2736100, n = 33) and CDKN1A (rs2395655, n = 27) associated with idiopathic pulmonary fibrosis risk were detected in most samples. A total of 410 somatic mutations were identified, with an average of 11.7 per tumour, including 69 synonymous, 177 missense, 17 nonsense, 1 nonstop and 11 splice-site mutations, and 135 small coding indels. Spectra of the somatic mutations revealed predominant C > T transitions despite an extensive smoking history in most patients, suggesting a potential association between APOBEC-related mutagenesis and the development of IPF-LC. TP53 (22/35, 62.9%) and BRAF (6/35, 17.1%) were found to be significantly mutated in IPF-LC. Recurrent focal amplifications in three chromosomal loci (3q26.33, 7q31.2, and 12q14.3) and 9p21.3 deletion were identified, and genes associated with the JAK-STAT signalling pathway were significantly amplified in IPF-LC (P = 0.012). This study demonstrates that IPF-LC is genetically characterized by the presence of somatic mutations reflecting a variety of environmental exposures on the background of specific germline mutations, and is associated with potentially targetable alterations such as BRAF mutations. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Ji An Hwang
- Department of Pulmonary and Critical Care Medicine, Asan Medical Centre, University of Ulsan College of Medicine, Seoul, Korea.,Department of Biomedical Sciences, University of Ulsan College of Medicine, Asan Medical Centre, Seoul, Korea
| | - Deokhoon Kim
- Asan Institute for Life Sciences, University of Ulsan College of medicine, Asan Medical Centre, Seoul, Korea.,Department of Pathology, Asan Medical Centre, University of Ulsan College of Medicine, Seoul, Korea
| | - Sung-Min Chun
- Department of Pathology, Asan Medical Centre, University of Ulsan College of Medicine, Seoul, Korea
| | - SooHyun Bae
- Department of Pulmonary and Critical Care Medicine, Asan Medical Centre, University of Ulsan College of Medicine, Seoul, Korea
| | - Joon Seon Song
- Department of Pathology, Asan Medical Centre, University of Ulsan College of Medicine, Seoul, Korea
| | - Mi Young Kim
- Department of Radiology and Research Institute of Radiology, Asan Medical Centre, University of Ulsan College of Medicine, Seoul, Korea
| | - Hyun Jung Koo
- Department of Radiology and Research Institute of Radiology, Asan Medical Centre, University of Ulsan College of Medicine, Seoul, Korea
| | - Jin Woo Song
- Department of Pulmonary and Critical Care Medicine, Asan Medical Centre, University of Ulsan College of Medicine, Seoul, Korea
| | - Woo Sung Kim
- Department of Pulmonary and Critical Care Medicine, Asan Medical Centre, University of Ulsan College of Medicine, Seoul, Korea
| | - Jae Cheol Lee
- Department of Oncology, Asan Medical Centre, University of Ulsan College of Medicine, Seoul, Korea
| | - Hyeong Ryul Kim
- Department of Thoracic Surgery, Asan Medical Centre, University of Ulsan College of Medicine, Seoul, Korea
| | - Chang-Min Choi
- Department of Pulmonary and Critical Care Medicine, Asan Medical Centre, University of Ulsan College of Medicine, Seoul, Korea.,Department of Oncology, Asan Medical Centre, University of Ulsan College of Medicine, Seoul, Korea
| | - Se Jin Jang
- Department of Pathology, Asan Medical Centre, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
21
|
Abstract
PURPOSE OF REVIEW Interstitial lung disease (ILD) is comprised of a heterogeneous group of disorders with highly variable natural histories and response to therapies. Pharmacogenetics focuses on the variability in drug response because of the presence of genetic factors that influence drug metabolism or disease activity. In this article, we review relevant drug-specific and disease-specific polymorphisms that may influence therapeutic response, and then highlight a recently identified drug-gene interaction in patients with idiopathic pulmonary fibrosis (IPF). RECENT FINDINGS The emergence of high-throughput genomic technology has allowed for identification of gene polymorphisms associated with susceptibility to specific disease states, including IPF and several connective tissue diseases known to cause ILD. IPF risk loci span a diverse group of genes, while most associated with connective tissue disease are critical to immune signaling. A recent pharmacogenetic analysis of patients enrolled in an IPF clinical trial identified a variant within TOLLIP to be associated with differential response to N-acetylcysteine therapy. SUMMARY Though few pharmacogenetic investigations have been conducted in patients with ILD to date, ample opportunities for pharmacogenetic exploration exist in this patient population. Such exploration will advance our understanding of specific ILDs and help usher in an era of personalized medicine.
Collapse
|
22
|
Analysis of Microarray-Identified Genes and MicroRNAs Associated with Idiopathic Pulmonary Fibrosis. Mediators Inflamm 2017; 2017:1804240. [PMID: 28588348 PMCID: PMC5446886 DOI: 10.1155/2017/1804240] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 03/13/2017] [Accepted: 03/23/2017] [Indexed: 12/12/2022] Open
Abstract
The aim of this study was to identify potential microRNAs and genes associated with idiopathic pulmonary fibrosis (IPF) through web-available microarrays. The microRNA microarray dataset GSE32538 and the mRNA datasets GSE32537, GSE53845, and GSE10667 were downloaded from the Gene Expression Omnibus (GEO) database. Differentially expressed miRNAs (DE-miRNAs)/genes (DEGs) were screened with GEO2R, and their associations with IPF were analyzed by comprehensive bioinformatic analyses. A total of 45 DE-microRNAs were identified between IPF and control tissues, whereas 67 common DEGs were determined to exhibit the same expression trends in all three microarrays. Furthermore, functional analysis indicated that microRNAs in cancer and ECM-receptor interaction were the most significant pathways and were enriched by the 45 DE-miRNAs and 67 common DEGs. Finally, we predicted potential microRNA-target interactions between 17 DE-miRNAs and 17 DEGs by using at least three online programs. A microRNA-mediated regulatory network among the DE-miRNAs and DEGs was constructed that might shed new light on potential biomarkers for the prediction of IPF progression.
Collapse
|
23
|
Spagnolo P, Cottin V. Genetics of idiopathic pulmonary fibrosis: from mechanistic pathways to personalised medicine. J Med Genet 2016; 54:93-99. [DOI: 10.1136/jmedgenet-2016-103973] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Accepted: 11/28/2016] [Indexed: 01/07/2023]
|
24
|
Kishore A, Žižková V, Kocourková L, Petrkova J, Bouros E, Nunes H, Loštáková V, Müller-Quernheim J, Zissel G, Kolek V, Bouros D, Valeyre D, Petrek M. Association Study for 26 Candidate Loci in Idiopathic Pulmonary Fibrosis Patients from Four European Populations. Front Immunol 2016; 7:274. [PMID: 27462317 PMCID: PMC4939450 DOI: 10.3389/fimmu.2016.00274] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 06/29/2016] [Indexed: 11/18/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) affects lung parenchyma with progressing fibrosis. In this study, we aimed to replicate MUC5B rs35705950 variants and determine new plausible candidate variants for IPF among four different European populations. We genotyped 26 IPF candidate loci in 165 IPF patients from four European countries, such as Czech Republic (n = 41), Germany (n = 33), Greece (n = 40), France (n = 51), and performed association study comparing observed variant distribution with that obtained in a genetically similar Czech healthy control population (n = 96) described in our earlier data report. A highly significant association for a promoter variant (rs35705950) of mucin encoding MUC5B gene was observed in all IPF populations, individually and combined [odds ratio (95% confidence interval); p-value as 5.23 (8.94–3.06); 1.80 × 10−11]. Another non-coding variant, rs7934606 in MUC2 was significant among German patients [2.85 (5.05–1.60); 4.03 × 10−4] and combined European IPF cases [2.18 (3.16–1.50); 3.73 × 10−5]. The network analysis for these variants indicated gene–gene and gene–phenotype interactions in IPF and lung biology. With replication of MUC5B rs35705950 previously reported in U.S. populations of European descent and indicating other plausible polymorphic variants relevant for IPF, we provide additional reference information for future extended functional and population studies aimed, ideally with inclusion of clinical parameters, at identification of IPF genetic markers.
Collapse
Affiliation(s)
- Amit Kishore
- Laboratory of Immunogenomics, Department of Pathological Physiology, Faculty of Medicine and Dentistry, Palacký University , Olomouc , Czech Republic
| | - Veronika Žižková
- Laboratory of Immunogenomics, Department of Pathological Physiology, Faculty of Medicine and Dentistry, Palacký University , Olomouc , Czech Republic
| | - Lenka Kocourková
- Laboratory of Immunogenomics, Department of Pathological Physiology, Faculty of Medicine and Dentistry, Palacký University , Olomouc , Czech Republic
| | - Jana Petrkova
- Laboratory of Immunogenomics, Department of Pathological Physiology, Faculty of Medicine and Dentistry, Palacký University , Olomouc , Czech Republic
| | - Evangelos Bouros
- Laboratory of Pharmacology, University Hospital Alexandroupolis, Democritus University of Thrace , Athens , Greece
| | - Hilario Nunes
- Université Paris 13, COMUE Sorbonne Paris Cité, Bobigny , Paris , France
| | - Vladimíra Loštáková
- Department of Respiratory Medicine, Faculty of Medicine and Dentistry, Palacký University , Olomouc , Czech Republic
| | - Joachim Müller-Quernheim
- Department of Pneumology, Center for Medicine, Medical Center, University of Freiburg , Freiburg , Germany
| | - Gernot Zissel
- Department of Pneumology, Center for Medicine, Medical Center, University of Freiburg , Freiburg , Germany
| | - Vitezslav Kolek
- Department of Respiratory Medicine, Faculty of Medicine and Dentistry, Palacký University , Olomouc , Czech Republic
| | - Demosthenes Bouros
- Academic Department of Pneumonology, Hospital for Diseases of the Chest 'Sotiria', Medical School, University of Athens , Athens , Greece
| | - Dominique Valeyre
- Université Paris 13, COMUE Sorbonne Paris Cité, Bobigny , Paris , France
| | - Martin Petrek
- Laboratory of Immunogenomics, Department of Pathological Physiology, Faculty of Medicine and Dentistry, Palacký University, Olomouc, Czech Republic; Faculty of Medicine and Dentistry, Institute of Molecular and Translational Medicine, Palacký University and Faculty Hospital, Olomouc, Czech Republic
| |
Collapse
|
25
|
Donati B, Valenti L. Telomeres, NAFLD and Chronic Liver Disease. Int J Mol Sci 2016; 17:383. [PMID: 26999107 PMCID: PMC4813240 DOI: 10.3390/ijms17030383] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 03/09/2016] [Accepted: 03/10/2016] [Indexed: 12/14/2022] Open
Abstract
Telomeres consist of repeat DNA sequences located at the terminal portion of chromosomes that shorten during mitosis, protecting the tips of chromosomes. During chronic degenerative conditions associated with high cell replication rate, progressive telomere attrition is accentuated, favoring senescence and genomic instability. Several lines of evidence suggest that this process is involved in liver disease progression: (a) telomere shortening and alterations in the expression of proteins protecting the telomere are associated with cirrhosis and hepatocellular carcinoma; (b) advanced liver damage is a feature of a spectrum of genetic diseases impairing telomere function, and inactivating germline mutations in the telomerase complex (including human Telomerase Reverse Transcriptase (hTERT) and human Telomerase RNA Component (hTERC)) are enriched in cirrhotic patients independently of the etiology; and (c) experimental models suggest that telomerase protects from liver fibrosis progression. Conversely, reactivation of telomerase occurs during hepatocarcinogenesis, allowing the immortalization of the neoplastic clone. The role of telomere attrition may be particularly relevant in the progression of nonalcoholic fatty liver, an emerging cause of advanced liver disease. Modulation of telomerase or shelterins may be exploited to prevent liver disease progression, and to define specific treatments for different stages of liver disease.
Collapse
Affiliation(s)
- Benedetta Donati
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Policlinico Milano, 20122 Milano, Italy.
| | - Luca Valenti
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Policlinico Milano, 20122 Milano, Italy.
| |
Collapse
|
26
|
de Pablo A, Bogoi R, Bejarano I, Toro C, Valencia E, Moreno V, Martín-Carbonero L, Gómez-Hernando C, Rodés B. Short Communication: p21/CDKN1A Expression Shows Broad Interindividual Diversity in a Subset of HIV-1 Elite Controllers. AIDS Res Hum Retroviruses 2016; 32:232-6. [PMID: 26537458 DOI: 10.1089/aid.2015.0137] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
The p21/CDKN1A protein has been described in vitro as well as in a small subset of patients as a restriction factor for HIV infection. We evaluated p21/CDKN1A mRNA expression on CD4(+) T cells from HIV-infected individuals with two outcomes (18 elite controllers and 28 viremic progressors). Our results show broad interindividual variation in this factor, which is unrelated to the patient's phenotype. Considering the gene's genetic surroundings in chromosome 6, such as HLA genotype and single nucleotide polymorphisms (SNPs), there was a positive association with carrying HLA-B2705 alleles and the rs733590 SNP. Thus, this natural variation of p21/CDKN1A alone does not appear to be a prognostic indicator of effective viral control in vivo and other factors must be considered.
Collapse
Affiliation(s)
- Alicia de Pablo
- Foundation for Biomedical Research, University Hospital La Paz, Madrid, Spain
- IdiPAZ, Madrid, Spain
| | - Roberta Bogoi
- Foundation for Biomedical Research, University Hospital La Paz, Madrid, Spain
- IdiPAZ, Madrid, Spain
| | - Irene Bejarano
- Foundation for Biomedical Research, Hospital Carlos III, Madrid, Spain
| | - Carlos Toro
- Microbiology Department, University Hospital La Paz - Carlos III, Madrid, Spain
| | - Eulalia Valencia
- Infectious Diseases Department, University Hospital La Paz - Carlos III, Madrid, Spain
| | - Victoria Moreno
- Infectious Diseases Department, University Hospital La Paz - Carlos III, Madrid, Spain
| | - Luz Martín-Carbonero
- Infectious Diseases Department, University Hospital La Paz - Carlos III, Madrid, Spain
| | | | - Berta Rodés
- Foundation for Biomedical Research, University Hospital La Paz, Madrid, Spain
- IdiPAZ, Madrid, Spain
| |
Collapse
|
27
|
Understanding Idiopathic Interstitial Pneumonia: A Gene-Based Review of Stressed Lungs. BIOMED RESEARCH INTERNATIONAL 2015; 2015:304186. [PMID: 26539479 PMCID: PMC4619788 DOI: 10.1155/2015/304186] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 08/26/2015] [Indexed: 12/17/2022]
Abstract
Pulmonary fibrosis is the main cause of severe morbidity and mortality in idiopathic interstitial pneumonias (IIP). In the past years, there has been major progress in the discovery of genetic factors that contribute to disease. Genes with highly penetrant mutations or strongly predisposing common risk alleles have been identified in familial and sporadic IIP. This review summarizes genes harbouring causative rare mutations and replicated common predisposing alleles. To date, rare mutations in nine different genes and five risk alleles fulfil this criterion. Mutated genes represent three genes involved in surfactant homeostasis and six genes involved in telomere maintenance. We summarize gene function, gene expressing cells, and pathological consequences of genetic alterations associated with disease. Consequences of the genetic alteration include dysfunctional surfactant processing, ER stress, immune dysregulation, and maintenance of telomere length. Biological evidence shows that these processes point towards a central role for alveolar epithelial type II cell dysfunction. However, tabulation also shows that function and consequence of most common risk alleles are not known. Most importantly, the predisposition of the MUC5B risk allele to disease is not understood. We propose a mechanism whereby MUC5B decreases surface tension lowering capacity of alveolar surfactant at areas with maximal mechanical stress.
Collapse
|
28
|
Kishore A, Žižková V, Kocourková L, Petřek M. A Dataset of 26 Candidate Gene and Pro-Inflammatory Cytokine Variants for Association Studies in Idiopathic Pulmonary Fibrosis: Frequency Distribution in Normal Czech Population. Front Immunol 2015; 6:476. [PMID: 26441981 PMCID: PMC4585032 DOI: 10.3389/fimmu.2015.00476] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 09/02/2015] [Indexed: 12/31/2022] Open
Affiliation(s)
- Amit Kishore
- Department of Pathological Physiology, Laboratory of Immunogenomics, Faculty of Medicine and Dentistry, Palacký University , Olomouc , Czech Republic
| | - Veronika Žižková
- Department of Pathological Physiology, Laboratory of Immunogenomics, Faculty of Medicine and Dentistry, Palacký University , Olomouc , Czech Republic
| | - Lenka Kocourková
- Department of Pathological Physiology, Laboratory of Immunogenomics, Faculty of Medicine and Dentistry, Palacký University , Olomouc , Czech Republic
| | - Martin Petřek
- Department of Pathological Physiology, Laboratory of Immunogenomics, Faculty of Medicine and Dentistry, Palacký University , Olomouc , Czech Republic ; Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University , Olomouc , Czech Republic
| |
Collapse
|
29
|
Kropski JA, Blackwell TS, Loyd JE. The genetic basis of idiopathic pulmonary fibrosis. Eur Respir J 2015; 45:1717-27. [PMID: 25837031 DOI: 10.1183/09031936.00163814] [Citation(s) in RCA: 140] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Accepted: 03/17/2015] [Indexed: 02/06/2023]
Abstract
Throughout the past decade, there have been substantial advances in understanding the pathogenesis of idiopathic pulmonary fibrosis (IPF). Recently, several large genome-wide association and linkage studies have identified common genetic variants in more than a dozen loci that appear to contribute to IPF risk. In addition, family-based studies have led to the identification of rare genetic variants in genes related to surfactant function and telomere biology, and mechanistic studies suggest pathophysiological derangements associated with these rare genetic variants are also found in sporadic cases of IPF. Current evidence suggests that rather than existing as distinct syndromes, sporadic and familial cases of IPF (familial interstitial pneumonia) probably reflect a continuum of genetic risk. Rapidly evolving bioinformatic and molecular biology techniques, combined with next-generation sequencing technologies, hold great promise for developing a comprehensive, integrated approach to defining the fundamental molecular mechanisms that underlie IPF pathogenesis.
Collapse
Affiliation(s)
- Jonathan A Kropski
- Division of Allergy, Pulmonary and Critical Care Medicine, Dept of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Timothy S Blackwell
- Division of Allergy, Pulmonary and Critical Care Medicine, Dept of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA Dept of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA Dept of Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN, USA Department of Veterans Affairs Medical Center, Nashville, TN, USA
| | - James E Loyd
- Division of Allergy, Pulmonary and Critical Care Medicine, Dept of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| |
Collapse
|
30
|
Abstract
The occurrence of pulmonary fibrosis in numerous individuals from the same family suggests a genetic cause for the disease. During the last 10 years, mutations involving proteins from the telomerase complex and from the surfactant system have been identified in association with pulmonary fibrosis. Mutations of TERT, the coding gene for the telomerase reverse transcriptase, are the most frequently identified mutations and are present in 15% of cases of familial pulmonary fibrosis. Other mutations (TERC, surfactant proteins genes) are only rarely evidenced in adults. Patients with mutations involving the telomerase complex may present with pulmonary fibrosis, hematologic, cutaneous or liver diseases. Other genetic variations associated with pulmonary fibrosis such as a polymorphism in the promoter of MUC5B or a polymorphism in TERT have been recently described, and could be considered to be part of a polygenic transmission. Evidence for mutations associated with the development of pulmonary fibrosis raises numerous clinical questions from establishing a diagnosis, providing counselling to deciding on therapy, and requires specific studies. From a pathophysiological point of view, the function of the genes highlights the central role of alveolar epithelium and aging in fibrogenesis.
Collapse
|
31
|
Abstract
PURPOSE OF REVIEW Recent genetic findings have identified new targets of investigation in the field of pulmonary fibrosis and have the potential to change clinical care. RECENT FINDINGS These findings implicate alterations in host defense, cell-to-cell adhesion, and aging and senescence in the pathophysiology of pulmonary fibrosis. At least one common genetic variant strongly associated with pulmonary fibrosis appears to have prognostic implications for patients. SUMMARY The inherited risk for pulmonary fibrosis is substantial, and recent data suggest that genetic risk for familial and sporadic forms of the disease are similar. Further characterizing this genetic risk will influence clinical practice in terms of categorization, diagnosis, and screening of individuals for this disease.
Collapse
Affiliation(s)
- Susan K. Mathai
- University of Colorado Denver, Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, Aurora, Colorado
| | - David A. Schwartz
- University of Colorado Denver, Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, Aurora, Colorado
- National Jewish Health, Denver, Colorado
| | - Laura A. Warg
- University of Colorado Denver, Medical Scientist Training Program, Aurora, Colorado
| |
Collapse
|
32
|
Spagnolo P, Rossi G, Cavazza A. Pathogenesis of idiopathic pulmonary fibrosis and its clinical implications. Expert Rev Clin Immunol 2014; 10:1005-17. [PMID: 24953006 DOI: 10.1586/1744666x.2014.917050] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Idiopathic pulmonary fibrosis (IPF) is the most common and lethal form of idiopathic interstitial pneumonia. The disease is thought to arise following an aberrant reparative response to recurrent alveolar epithelial cell injury leading to progressive loss of function. The median survival time is 3-5 years from diagnosis. Cigarette smoking, exposure to organic and inorganic dust and genetic factors have been shown to increase the risk of disease, although the cause of IPF remains elusive and its pathogenesis incompletely understood. In the last decade, several clinical trials evaluating novel therapies for IPF have been conducted but the results have been mostly disappointing. Conversely, compounds that target anti-fibrotic and growth factor pathways have been proven effective in slowing functional decline and disease progression. These promising results notwithstanding, truly effective therapeutic strategies will likely require combinations of drugs in order to target the multitude of pathways involved in disease pathogenesis.
Collapse
Affiliation(s)
- Paolo Spagnolo
- Center for Rare Lung Diseases, Respiratory Disease Unit, University Hospital of Modena, Via del Pozzo 71, 41124 Modena, Italy
| | | | | |
Collapse
|
33
|
Spagnolo P, Grunewald J, du Bois RM. Genetic determinants of pulmonary fibrosis: evolving concepts. THE LANCET RESPIRATORY MEDICINE 2014; 2:416-28. [DOI: 10.1016/s2213-2600(14)70047-5] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
34
|
Aravinthan A, Mells G, Allison M, Leathart J, Kotronen A, Yki-Jarvinen H, Daly AK, Day CP, Anstee QM, Alexander G. Gene polymorphisms of cellular senescence marker p21 and disease progression in non-alcohol-related fatty liver disease. Cell Cycle 2014; 13:1489-94. [PMID: 24626178 PMCID: PMC4050146 DOI: 10.4161/cc.28471] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Revised: 02/16/2014] [Accepted: 03/07/2014] [Indexed: 02/08/2023] Open
Abstract
Non-alcohol-related fatty liver disease (NAFLD) encompasses a wide spectrum, ranging from steatosis alone to steatohepatitis and fibrosis. Presence of steatohepatitis and fibrosis are key hallmarks of disease progression. Previous studies have demonstrated an association between hepatocyte p21 expression and fibrosis stage in NAFLD. The aim of this study is to investigate the association between the variants of CDKN1A, which encodes p21, and disease progression in NAFLD. To this end, the relation between CDKN1A polymorphism and liver fibrosis was studied in 2 cohorts of biopsy-proven NAFLD patients from UK (n = 323) and Finland (n = 123). Genotyping was performed using DNA isolated from lymphocytes collected at the time of liver biopsy. The findings of the UK cohort were tested in the Finnish cohort. Both the UK and Finnish cohorts were significantly different from each other in basic demographics. In the UK cohort, rs762623, of the 6 SNPs across CDKN1A tested, was significantly associated with disease progression in NAFLD. This association was confirmed in the Finnish cohort. Despite the influence on fibrosis development, SNPs across CDKN1A did not affect the progression of liver fibrosis. In conclusion, CDKN1A variant rs762623 is associated with the development but not the propagation of progressive liver disease in NAFLD.
Collapse
Affiliation(s)
| | - George Mells
- Department of Medicine; University of Cambridge; Cambridge, UK
| | - Michael Allison
- Department of Medicine; University of Cambridge; Cambridge, UK
| | - Julian Leathart
- Institute of Cellular Medicine; Newcastle University; Newcastle upon Tyne, UK
| | - Anna Kotronen
- Department of Medicine; University of Helsinki; Helsinki, Finland
| | | | - Ann K Daly
- Institute of Cellular Medicine; Newcastle University; Newcastle upon Tyne, UK
| | - Christopher P Day
- Institute of Cellular Medicine; Newcastle University; Newcastle upon Tyne, UK
| | - Quentin M Anstee
- Institute of Cellular Medicine; Newcastle University; Newcastle upon Tyne, UK
| | | |
Collapse
|
35
|
Zhang R, Zhao Y, Chu M, Mehta A, Wei Y, Liu Y, Xun P, Bai J, Yu H, Su L, Zhang H, Hu Z, Shen H, Chen F, Christiani DC. A large scale gene-centric association study of lung function in newly-hired female cotton textile workers with endotoxin exposure. PLoS One 2013; 8:e59035. [PMID: 23527081 PMCID: PMC3602449 DOI: 10.1371/journal.pone.0059035] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Accepted: 02/08/2013] [Indexed: 02/02/2023] Open
Abstract
Background Occupational exposure to endotoxin is associated with decrements in pulmonary function, but how much variation in this association is explained by genetic variants is not well understood. Objective We aimed to identify single nucleotide polymorphisms (SNPs) that are associated with the rate of forced expiratory volume in one second (FEV1) decline by a large scale genetic association study in newly-hired healthy young female cotton textile workers. Methods DNA samples were genotyped using the Illumina Human CVD BeadChip. Change rate in FEV1 was modeled as a function of each SNP genotype in linear regression model with covariate adjustment. We controlled the type 1 error in study-wide level by permutation method. The false discovery rate (FDR) and the family-wise error rate (FWER) were set to be 0.10 and 0.15 respectively. Results Two SNPs were found to be significant (P<6.29×10−5), including rs1910047 (P = 3.07×10−5, FDR = 0.0778) and rs9469089 (P = 6.19×10−5, FDR = 0.0967), as well as other eight suggestive (P<5×10−4) associated SNPs. Gene-gene and gene-environment interactions were also observed, such as rs1910047 and rs1049970 (P = 0.0418, FDR = 0.0895); rs9469089 and age (P = 0.0161, FDR = 0.0264). Genetic risk score analysis showed that the more risk loci the subjects carried, the larger the rate of FEV1 decline occurred (Ptrend = 3.01×10−18). However, the association was different among age subgroups (P = 7.11×10−6) and endotoxin subgroups (P = 1.08×10−2). Functional network analysis illustrates potential biological connections of all interacted genes. Conclusions Genetic variants together with environmental factors interact to affect the rate of FEV1 decline in cotton textile workers.
Collapse
Affiliation(s)
- Ruyang Zhang
- Department of Epidemiology and Biostatistics and Ministry of Education (MOE) Key Lab for Modern Toxicology, School of Public Health, Nanjing Medical University, Nanjing, China
- Section of Clinical Epidemiology, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Cancer Center, Nanjing Medical University, Nanjing, China
| | - Yang Zhao
- Department of Epidemiology and Biostatistics and Ministry of Education (MOE) Key Lab for Modern Toxicology, School of Public Health, Nanjing Medical University, Nanjing, China
- Department of Environmental Health, Harvard School of Public Health, Boston, Massachusetts, United States of America
| | - Minjie Chu
- Department of Epidemiology and Biostatistics and Ministry of Education (MOE) Key Lab for Modern Toxicology, School of Public Health, Nanjing Medical University, Nanjing, China
- Section of Clinical Epidemiology, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Cancer Center, Nanjing Medical University, Nanjing, China
| | - Amar Mehta
- Department of Environmental Health, Harvard School of Public Health, Boston, Massachusetts, United States of America
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Yongyue Wei
- Department of Epidemiology and Biostatistics and Ministry of Education (MOE) Key Lab for Modern Toxicology, School of Public Health, Nanjing Medical University, Nanjing, China
- Department of Environmental Health, Harvard School of Public Health, Boston, Massachusetts, United States of America
| | - Yao Liu
- Department of Epidemiology and Biostatistics and Ministry of Education (MOE) Key Lab for Modern Toxicology, School of Public Health, Nanjing Medical University, Nanjing, China
- Section of Clinical Epidemiology, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Cancer Center, Nanjing Medical University, Nanjing, China
| | - Pengcheng Xun
- Department of Epidemiology and Biostatistics, School of Public Health, Indiana University Bloomington, Bloomington, Indiana, United States of America
| | - Jianling Bai
- Department of Epidemiology and Biostatistics and Ministry of Education (MOE) Key Lab for Modern Toxicology, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Hao Yu
- Department of Epidemiology and Biostatistics and Ministry of Education (MOE) Key Lab for Modern Toxicology, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Li Su
- Department of Environmental Health, Harvard School of Public Health, Boston, Massachusetts, United States of America
| | - Hongxi Zhang
- Putuo District Peoples Hospital, Shanghai Second Medical University, Shanghai, China
| | - Zhibin Hu
- Department of Epidemiology and Biostatistics and Ministry of Education (MOE) Key Lab for Modern Toxicology, School of Public Health, Nanjing Medical University, Nanjing, China
- Section of Clinical Epidemiology, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Cancer Center, Nanjing Medical University, Nanjing, China
| | - Hongbing Shen
- Department of Epidemiology and Biostatistics and Ministry of Education (MOE) Key Lab for Modern Toxicology, School of Public Health, Nanjing Medical University, Nanjing, China
- Section of Clinical Epidemiology, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Cancer Center, Nanjing Medical University, Nanjing, China
| | - Feng Chen
- Department of Epidemiology and Biostatistics and Ministry of Education (MOE) Key Lab for Modern Toxicology, School of Public Health, Nanjing Medical University, Nanjing, China
- * E-mail: (FC); (DCC)
| | - David C. Christiani
- Department of Environmental Health, Harvard School of Public Health, Boston, Massachusetts, United States of America
- Pulmonary and Critical Care Unit, Department of Medicine, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail: (FC); (DCC)
| |
Collapse
|
36
|
Familial forms of nonspecific interstitial pneumonia/idiopathic pulmonary fibrosis: clinical course and genetic background. Curr Opin Pulm Med 2012; 18:455-61. [PMID: 22781209 DOI: 10.1097/mcp.0b013e328356b15c] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
PURPOSE OF REVIEW Familial pulmonary fibrosis has long been recognized and suggests that pulmonary fibrosis may have a genetic origin in some cases with an autosomal dominant transmission. RECENT FINDINGS Mutations in the telomerase complex and in the surfactant pathways have been discovered in the last decade. Almost 20% of the cases of familial pulmonary fibrosis are related to known functional mutations in one of these systems. A polymorphism in the promoter of the MUC5B gene has been associated with both sporadic and familial forms of idiopathic pulmonary fibrosis; however, the impact of this association remains to be determined. SUMMARY These genes point to alveolar epithelium injury and repair as a major component of the fibrotic process.
Collapse
|
37
|
Abstract
There is clear evidence that environmental exposures and genetic predisposition contribute to the pathogenesis of idiopathic pulmonary fibrosis (IPF). Cigarette smoking increases the risk of developing IPF several-fold, as do other exposures such as metal-fume and wood-dust exposure. Occupations that increase the risk of IPF are agricultural work, hairdressing, and stone polishing, supporting the role of environmental exposure in disease pathogenesis. Genetic predisposition to IPF is evident from its familial aggregation and the fact that pulmonary fibrosis develops in several rare genetic disorders. Mutations in surfactant proteins lead to pulmonary fibrosis and are associated with endoplasmic reticulum stress in alveolar type II epithelial cells. Mutations in telomerase have been found in several families with IPF, and shortened telomeres are found in sporadic cases of IPF. A common variant in mucin 5B predisposes to both familial and sporadic IPF and is present in the majority of cases, indicating sporadic IPF occurs in those with genetic predisposition.
Collapse
Affiliation(s)
- Mark P Steele
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, USA.
| | | |
Collapse
|