1
|
Hong Y, Lee JM, Lee C, Na D, Jung J, Ahn A, Yoo JW, Lee JW, Chung NG, Kim M, Kim Y. Telomere Length and Genetic Variations in Acquired Pediatric Aplastic Anemia: A Flow-FISH Study in Korean Patients. Diagnostics (Basel) 2025; 15:931. [PMID: 40218281 PMCID: PMC11988933 DOI: 10.3390/diagnostics15070931] [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: 03/12/2025] [Accepted: 03/31/2025] [Indexed: 04/14/2025] Open
Abstract
Background: Aplastic anemia (AA) is a rare bone marrow failure syndrome characterized by notably short telomere length, which is associated with treatment responses. In this study, we measured telomere lengths in Korean pediatric AA patients using flow-fluorescence in situ hybridization (Flow-FISH) and explored their shortening in relation to disease characteristics, genetic conditions and patient outcomes. Methods: We analyzed peripheral blood samples from 75 AA patients and 101 healthy controls. Telomere lengths were measured using Flow-FISH, and relative telomere length (RTL) and delta RTL assessments were conducted. Genetic evaluations included karyotyping, chromosome breakage tests and clinical exome sequencing (CES) to identify inherited bone marrow failure syndrome (IBMFS)-associated genetic variants. Results: Telomere lengths in AA patients were significantly lower than those of age-adjusted healthy controls. Patients receiving immunosuppressive therapy tended to have long telomeres, as indicated by high delta RTL values. Patients with genetic abnormalities, including karyotype abnormalities (n = 2) and genetic variants (n = 11) such as carrier genes of IBMFS or variants of unclear significance, showed significantly short telomere lengths. Conclusions: This study reinforces the importance of telomere length as a biomarker in acquired AA. Utilizing Flow-FISH, we were able to accurately measure telomere lengths and establish confidence in this method as an appropriate laboratory test. We found significant reduction in telomere lengths in AA patients, and importantly, longer telomeres were correlated with better outcomes in immunosuppressive therapy. Additionally, our genetic analysis underscored the relevance of variants in IBMFS-associated genes to the pathophysiology of short telomeres.
Collapse
Affiliation(s)
- Yuna Hong
- Catholic Genetic Laboratory Center, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (Y.H.); (J.-M.L.); (C.L.); (D.N.); (Y.K.)
- Department of Medical Sciences, Graduate School of The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Jong-Mi Lee
- Catholic Genetic Laboratory Center, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (Y.H.); (J.-M.L.); (C.L.); (D.N.); (Y.K.)
- Department of Laboratory Medicine, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea;
| | - Chaeyeon Lee
- Catholic Genetic Laboratory Center, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (Y.H.); (J.-M.L.); (C.L.); (D.N.); (Y.K.)
- Department of Medical Sciences, Graduate School of The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Duyeon Na
- Catholic Genetic Laboratory Center, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (Y.H.); (J.-M.L.); (C.L.); (D.N.); (Y.K.)
- Department of Medical Sciences, Graduate School of The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Jin Jung
- Department of Laboratory Medicine, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea;
| | - Ari Ahn
- Department of Laboratory Medicine, Incheon St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea;
| | - Jae Won Yoo
- Department of Pediatrics, Catholic Hematology Hospital, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (J.W.Y.); (J.W.L.)
| | - Jae Wook Lee
- Department of Pediatrics, Catholic Hematology Hospital, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (J.W.Y.); (J.W.L.)
| | - Nack-Gyun Chung
- Department of Pediatrics, Catholic Hematology Hospital, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (J.W.Y.); (J.W.L.)
| | - Myungshin Kim
- Catholic Genetic Laboratory Center, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (Y.H.); (J.-M.L.); (C.L.); (D.N.); (Y.K.)
- Department of Laboratory Medicine, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea;
| | - Yonggoo Kim
- Catholic Genetic Laboratory Center, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (Y.H.); (J.-M.L.); (C.L.); (D.N.); (Y.K.)
- Department of Laboratory Medicine, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea;
| |
Collapse
|
2
|
Shah PD, Armanios M. Viewpoint: Pre- and post-lung transplant considerations for patients with ultra-short telomere length. Eur Respir J 2025; 65:2401545. [PMID: 39884762 PMCID: PMC11883148 DOI: 10.1183/13993003.01545-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Accepted: 01/09/2025] [Indexed: 02/01/2025]
Abstract
Lung transplantation remains the only life-extending procedure for patients with idiopathic pulmonary fibrosis (IPF) and related progressive interstitial lung disease (ILD). Discoveries from recent decades have shown that mutations in telomerase and other telomere maintenance genes are their most common inherited risk factor, identifiable in up to 30–35% of families with pulmonary fibrosis [1]. Mutations in nine telomerase and telomere maintenance genes are confirmed to predispose to adult-onset pulmonary fibrosis by co-segregation in large families and functional studies (table 1) [2–13]. They compromise telomerase abundance, recruitment and function [1, 14]. Patients with ultra-short telomere length develop recurrent complications after lung transplantation; therefore, pre-transplant assessment and individualised post-transplant management may improve outcome in carefully defined high risk patient subsets https://bit.ly/3WvfLC1
Collapse
Affiliation(s)
- Pali D Shah
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mary Armanios
- Departments of Oncology, Genetic Medicine and Pathology, Telomere Center at Johns Hopkins, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
3
|
Southern BD, Gadre SK. Telomeropathies in Interstitial Lung Disease and Lung Transplant Recipients. J Clin Med 2025; 14:1496. [PMID: 40095034 PMCID: PMC11900913 DOI: 10.3390/jcm14051496] [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: 12/07/2024] [Revised: 01/23/2025] [Accepted: 02/09/2025] [Indexed: 03/19/2025] Open
Abstract
Telomeropathies, or telomere biology disorders (TBDs), are syndromes that can cause a number of medical conditions, including interstitial lung disease (ILD), bone marrow failure, liver fibrosis, and other diseases. They occur due to genetic mutations to the telomerase complex enzymes that result in premature shortening of telomeres, the caps on the ends of cellular DNA that protect chromosome length during cell division, leading to early cell senescence and death. Idiopathic pulmonary fibrosis (IPF) is the most common manifestation of the telomere biology disorders, although it has been described in other interstitial lung diseases as well, such as rheumatoid arthritis-associated ILD and chronic hypersensitivity pneumonitis. Telomere-related mutations can be inherited or can occur sporadically. Identifying these patients and offering genetic counseling is important because telomerapathies have been associated with poorer outcomes including death, lung transplantation, hospitalization, and FVC decline. Additionally, treatment with immunosuppressants has been shown to be associated with worse outcomes. Currently, there is no specific treatment for TBD except to transplant the organ that is failing, although there are a number of promising treatment strategies currently under investigation. Shortened telomere length is routinely discovered in patients undergoing lung transplantation for IPF. Testing to detect early TBD in patients with suggestive signs or symptoms can allow for more comprehensive treatment and multidisciplinary care pre- and post-transplant. Patients with TBD undergoing lung transplantation have been reported to have both pulmonary and extrapulmonary complications at a higher frequency than other lung transplant recipients, such as graft-specific complications, increased infections, and complications related to immunosuppressive therapy.
Collapse
Affiliation(s)
- Brian D. Southern
- Integrated Hospital-Care Institute, Department of Pulmonary Medicine, Cleveland Clinic, Cleveland, OH 44195, USA;
| | | |
Collapse
|
4
|
Pennington KM, Simonetto D, Taner T, Mangaonkar AA. Pulmonary, Hepatic, and Allogeneic Hematopoietic Stem Cell Transplantation in Patients with Telomere Biology Disorders. Curr Hematol Malig Rep 2024; 19:293-299. [PMID: 38315384 DOI: 10.1007/s11899-024-00724-z] [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] [Accepted: 01/17/2024] [Indexed: 02/07/2024]
Abstract
PURPOSE OF THE REVIEW This study aimed to summarize evidence and provide consensus-based guidelines for management of transplantation in patients with telomere biology disorders (TBD). Specifically, this review focuses on clinical management of lung, liver, and bone marrow transplantation in TBD patients. RECENT FINDINGS TBD patients have specific unique biological vulnerabilities such as T cell immunodeficiency, susceptibility to infections, hypersensitivity to chemotherapy and radiation, and cytopenias. Furthermore, multiple organ involvement at diagnosis makes clinical management especially challenging due to higher degree of organ damage, and stress-induced telomeric crisis. Sequential and combined organ transplants, development of novel radiation and alkylator-free conditioning regimen, and use of novel drugs for graft-versus-host disease prophylaxis are some of the recent updates in the field. Multidisciplinary management is essential to optimize transplant outcomes in patients with TBD. In this review, we provide consensus-based transplant management guidelines for clinical management of transplant in TBD.
Collapse
Affiliation(s)
| | - Douglas Simonetto
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, USA.
| | - Timucin Taner
- Departments of Surgery and Immunology, Mayo Clinic, Rochester, USA.
| | | |
Collapse
|
5
|
Borie R, Ba I, Debray MP, Kannengiesser C, Crestani B. Syndromic genetic causes of pulmonary fibrosis. Curr Opin Pulm Med 2024; 30:473-483. [PMID: 38896087 DOI: 10.1097/mcp.0000000000001088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
PURPOSE OF REVIEW The identification of extra-pulmonary symptoms plays a crucial role in diagnosing interstitial lung disease (ILD). These symptoms not only indicate autoimmune diseases but also hint at potential genetic disorders, suggesting a potential overlap between genetic and autoimmune origins. RECENT FINDINGS Genetic factors contributing to ILD are predominantly associated with telomere (TRG) and surfactant-related genes. While surfactant-related gene mutations typically manifest with pulmonary involvement alone, TRG mutations were initially linked to syndromic forms of pulmonary fibrosis, known as telomeropathies, which may involve hematological and hepatic manifestations with variable penetrance. Recognizing extra-pulmonary signs indicative of telomeropathy should prompt the analysis of TRG mutations, the most common genetic cause of familial pulmonary fibrosis. Additionally, various genetic diseases causing ILD, such as alveolar proteinosis, alveolar hemorrhage, or unclassifiable pulmonary fibrosis, often present as part of syndromes that include hepatic, hematological, or skin disorders. SUMMARY This review explores the main genetic conditions identified over the past two decades.
Collapse
Affiliation(s)
- Raphaël Borie
- Service de Pneumologie A Hôpital Bichat, APHP, Paris, France, Université Paris Cité, Inserm, PHERE, Université Paris Cité
| | | | | | | | - Bruno Crestani
- Service de Pneumologie A Hôpital Bichat, APHP, Paris, France, Université Paris Cité, Inserm, PHERE, Université Paris Cité
| |
Collapse
|
6
|
Hinchie AM, Sanford SL, Loughridge KE, Sutton RM, Parikh AH, Gil Silva AA, Sullivan DI, Chun-On P, Morrell MR, McDyer JF, Opresko PL, Alder JK. A persistent variant telomere sequence in a human pedigree. Nat Commun 2024; 15:4681. [PMID: 38824190 PMCID: PMC11144197 DOI: 10.1038/s41467-024-49072-9] [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: 01/20/2024] [Accepted: 05/22/2024] [Indexed: 06/03/2024] Open
Abstract
The telomere sequence, TTAGGG, is conserved across all vertebrates and plays an essential role in suppressing the DNA damage response by binding a set of proteins termed shelterin. Changes in the telomere sequence impair shelterin binding, initiate a DNA damage response, and are toxic to cells. Here we identify a family with a variant in the telomere template sequence of telomerase, the enzyme responsible for telomere elongation, that led to a non-canonical telomere sequence. The variant is inherited across at least one generation and one family member reports no significant medical concerns despite ~9% of their telomeres converting to the novel sequence. The variant template disrupts telomerase repeat addition processivity and decreased the binding of the telomere-binding protein POT1. Despite these disruptions, the sequence is readily incorporated into cellular chromosomes. Incorporation of a variant sequence prevents POT1-mediated inhibition of telomerase suggesting that incorporation of a variant sequence may influence telomere addition. These findings demonstrate that telomeres can tolerate substantial degeneracy while remaining functional and provide insights as to how incorporation of a non-canonical telomere sequence might alter telomere length dynamics.
Collapse
Affiliation(s)
- Angela M Hinchie
- Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Samantha L Sanford
- Environmental and Occupational Health Department, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
- University of Pittsburgh Medical Center, Hillman Cancer Center, Pittsburgh, PA, USA
| | - Kelly E Loughridge
- Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Rachel M Sutton
- Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Anishka H Parikh
- Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Agustin A Gil Silva
- Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Daniel I Sullivan
- Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Pattra Chun-On
- Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Matthew R Morrell
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - John F McDyer
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Patricia L Opresko
- Environmental and Occupational Health Department, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
- University of Pittsburgh Medical Center, Hillman Cancer Center, Pittsburgh, PA, USA
- Pharmacology and Chemical Biology Department, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jonathan K Alder
- Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease, University of Pittsburgh, Pittsburgh, PA, USA.
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
7
|
Lasho T, Patnaik MM. Adaptive and Maladaptive Clonal Hematopoiesis in Telomere Biology Disorders. Curr Hematol Malig Rep 2024; 19:35-44. [PMID: 38095828 DOI: 10.1007/s11899-023-00719-2] [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] [Accepted: 11/24/2023] [Indexed: 01/30/2024]
Abstract
PURPOSE OF REVIEW Telomere biology disorders (TBDs) are germline-inherited conditions characterized by reduction in telomerase function, accelerated shortening of telomeres, predisposition to organ-failure syndromes, and increased risk of neoplasms, especially myeloid malignancies. In normal cells, critically short telomeres trigger apoptosis and/or cellular senescence. However, the evolutionary mechanism by which TBD-related telomerase-deficient cells can overcome this fitness constraint remains elusive. RECENT FINDINGS Preliminary data suggests the existence of adaptive somatic mosaic states characterized by variants in TBD-related genes and maladaptive somatic mosaic states that attempt to overcome hematopoietic fitness constraints by alternative methods leading to clonal hematopoiesis. TBDs are both rare and highly heterogeneous in presentation, and the association of TBD with malignant transformation is unclear. Understanding the clonal complexity and mechanisms behind TBD-associated molecular signatures that lead to somatic adaptation in the setting of defective hematopoiesis will help inform therapy and treatment for this set of diseases.
Collapse
Affiliation(s)
- Terra Lasho
- Division of Hematology, Mayo Clinic Rochester, 200 First Street SW, Rochester, MN, 55905, USA
| | - Mrinal M Patnaik
- Division of Hematology, Mayo Clinic Rochester, 200 First Street SW, Rochester, MN, 55905, USA.
| |
Collapse
|
8
|
Ghanim GE, Sekne Z, Balch S, van Roon AMM, Nguyen THD. 2.7 Å cryo-EM structure of human telomerase H/ACA ribonucleoprotein. Nat Commun 2024; 15:746. [PMID: 38272871 PMCID: PMC10811338 DOI: 10.1038/s41467-024-45002-x] [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: 06/27/2023] [Accepted: 01/03/2024] [Indexed: 01/27/2024] Open
Abstract
Telomerase is a ribonucleoprotein (RNP) enzyme that extends telomeric repeats at eukaryotic chromosome ends to counterbalance telomere loss caused by incomplete genome replication. Human telomerase is comprised of two distinct functional lobes tethered by telomerase RNA (hTR): a catalytic core, responsible for DNA extension; and a Hinge and ACA (H/ACA) box RNP, responsible for telomerase biogenesis. H/ACA RNPs also have a general role in pseudouridylation of spliceosomal and ribosomal RNAs, which is critical for the biogenesis of the spliceosome and ribosome. Much of our structural understanding of eukaryotic H/ACA RNPs comes from structures of the human telomerase H/ACA RNP. Here we report a 2.7 Å cryo-electron microscopy structure of the telomerase H/ACA RNP. The significant improvement in resolution over previous 3.3 Å to 8.2 Å structures allows us to uncover new molecular interactions within the H/ACA RNP. Many disease mutations are mapped to these interaction sites. The structure also reveals unprecedented insights into a region critical for pseudouridylation in canonical H/ACA RNPs. Together, our work advances understanding of telomerase-related disease mutations and the mechanism of pseudouridylation by eukaryotic H/ACA RNPs.
Collapse
Affiliation(s)
| | - Zala Sekne
- MRC Laboratory of Molecular Biology, Cambridge, CB2 0QH, UK
| | | | | | | |
Collapse
|
9
|
Al-Mutairy EA, Al Qattan S, Khalid M, Al-Enazi AA, Al-Saif MM, Imtiaz F, Ramzan K, Raveendran V, Alaiya A, Meyer BF, Atamas SP, Collison KS, Khabar KS, Hasday JD, Al-Mohanna F. Wild-type S100A3 and S100A13 restore calcium homeostasis and mitigate mitochondrial dysregulation in pulmonary fibrosis patient-derived cells. Front Cell Dev Biol 2023; 11:1282868. [PMID: 38099297 PMCID: PMC10720433 DOI: 10.3389/fcell.2023.1282868] [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: 08/25/2023] [Accepted: 11/20/2023] [Indexed: 12/17/2023] Open
Abstract
Patients with digenic S100A3 and S100A13 mutations exhibited an atypical and progressive interstitial pulmonary fibrosis, with impaired intracellular calcium homeostasis and mitochondrial dysfunction. Here we provide direct evidence of a causative effect of the mutation on receptor mediated calcium signaling and calcium store responses in control cells transfected with mutant S100A3 and mutant S100A13. We demonstrate that the mutations lead to increased mitochondrial mass and hyperpolarization, both of which were reversed by transfecting patient-derived cells with the wild type S100A3 and S100A13, or extracellular treatment with the recombinant proteins. In addition, we demonstrate increased secretion of inflammatory mediators in patient-derived cells and in control cells transfected with the mutant-encoding constructs. These findings indicate that treatment of patients' cells with recombinant S100A3 and S100A13 proteins is sufficient to normalize most of cellular responses, and may therefore suggest the use of these recombinant proteins in the treatment of this devastating disease.
Collapse
Affiliation(s)
- Eid A. Al-Mutairy
- Department of Cell Biology, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
- Department of Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Somaya Al Qattan
- Department of Cell Biology, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Mohammed Khalid
- Department of Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Azizah A. Al-Enazi
- Department of Cell Biology, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Maher M. Al-Saif
- BioMolecular Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Faiqa Imtiaz
- Clinical Genomics, Center of Genomic Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Khushnooda Ramzan
- Clinical Genomics, Center of Genomic Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Vineesh Raveendran
- Department of Cell Biology, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Ayodele Alaiya
- Stem Cell Therapy Program, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Brian F. Meyer
- Clinical Genomics, Center of Genomic Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Sergei P. Atamas
- University of Maryland School of Medicine, Baltimore, MD, United States
- Baltimore VA Medical Center, Baltimore, MD, United States
| | - Kate S. Collison
- Department of Cell Biology, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Khalid S. Khabar
- BioMolecular Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Jeffrey D. Hasday
- University of Maryland School of Medicine, Baltimore, MD, United States
- Baltimore VA Medical Center, Baltimore, MD, United States
| | - Futwan Al-Mohanna
- Department of Cell Biology, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| |
Collapse
|
10
|
Liao P, Yan B, Wang C, Lei P. Telomeres: Dysfunction, Maintenance, Aging and Cancer. Aging Dis 2023; 15:2595-2631. [PMID: 38270117 PMCID: PMC11567242 DOI: 10.14336/ad.2023.1128] [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: 10/17/2023] [Accepted: 11/28/2023] [Indexed: 01/26/2024] Open
Abstract
Aging has emerged at the forefront of scientific research due to the growing social and economic costs associated with the growing aging global population. The defining features of aging involve a variety of molecular processes and cellular systems, which are interconnected and collaboratively contribute to the aging process. Herein, we analyze how telomere dysfunction potentially amplifies or accelerates the molecular and biochemical mechanisms underpinning each feature of aging and contributes to the emergence of age-associated illnesses, including cancer and neurodegeneration, via the perspective of telomere biology. Furthermore, the recently identified novel mechanistic actions for telomere maintenance offer a fresh viewpoint and approach to the management of telomeres and associated disorders. Telomeres and the defining features of aging are intimately related, which has implications for therapeutic and preventive approaches to slow aging and reduce the prevalence of age-related disorders.
Collapse
Affiliation(s)
- Pan Liao
- The School of Medicine, Nankai University, Tianjin, China.
- Haihe Laboratory of Cell Ecosystem, Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin, China.
- Tianjin Geriatrics Institute, Tianjin Medical University General Hospital, Tianjin, China.
| | - Bo Yan
- Haihe Laboratory of Cell Ecosystem, Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin, China.
- Tianjin Geriatrics Institute, Tianjin Medical University General Hospital, Tianjin, China.
| | - Conglin Wang
- Haihe Laboratory of Cell Ecosystem, Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin, China.
- Tianjin Geriatrics Institute, Tianjin Medical University General Hospital, Tianjin, China.
| | - Ping Lei
- The School of Medicine, Nankai University, Tianjin, China.
- Haihe Laboratory of Cell Ecosystem, Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin, China.
- Tianjin Geriatrics Institute, Tianjin Medical University General Hospital, Tianjin, China.
| |
Collapse
|
11
|
Borie R, Kannengiesser C, Antoniou K, Bonella F, Crestani B, Fabre A, Froidure A, Galvin L, Griese M, Grutters JC, Molina-Molina M, Poletti V, Prasse A, Renzoni E, van der Smagt J, van Moorsel CHM. European Respiratory Society statement on familial pulmonary fibrosis. Eur Respir J 2023; 61:13993003.01383-2022. [PMID: 36549714 DOI: 10.1183/13993003.01383-2022] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 10/26/2022] [Indexed: 12/24/2022]
Abstract
Genetic predisposition to pulmonary fibrosis has been confirmed by the discovery of several gene mutations that cause pulmonary fibrosis. Although genetic sequencing of familial pulmonary fibrosis (FPF) cases is embedded in routine clinical practice in several countries, many centres have yet to incorporate genetic sequencing within interstitial lung disease (ILD) services and proper international consensus has not yet been established. An international and multidisciplinary expert Task Force (pulmonologists, geneticists, paediatrician, pathologist, genetic counsellor, patient representative and librarian) reviewed the literature between 1945 and 2022, and reached consensus for all of the following questions: 1) Which patients may benefit from genetic sequencing and clinical counselling? 2) What is known of the natural history of FPF? 3) Which genes are usually tested? 4) What is the evidence for telomere length measurement? 5) What is the role of common genetic variants (polymorphisms) in the diagnostic workup? 6) What are the optimal treatment options for FPF? 7) Which family members are eligible for genetic sequencing? 8) Which clinical screening and follow-up parameters may be considered in family members? Through a robust review of the literature, the Task Force offers a statement on genetic sequencing, clinical management and screening of patients with FPF and their relatives. This proposal may serve as a basis for a prospective evaluation and future international recommendations.
Collapse
Affiliation(s)
- Raphael Borie
- Université Paris Cité, Inserm, PHERE, Hôpital Bichat, AP-HP, Service de Pneumologie A, Centre Constitutif du Centre de Référence des Maladies Pulmonaires Rares, FHU APOLLO, Paris, France
| | | | - Katerina Antoniou
- Laboratory of Molecular and Cellular Pneumonology, Department of Respiratory Medicine, School of Medicine, University of Crete, Heraklion, Greece
| | - Francesco Bonella
- Center for Interstitial and Rare Lung Diseases, Pneumology Department, Ruhrlandklinik, University Hospital, University of Essen, European Reference Network (ERN)-LUNG, ILD Core Network, Essen, Germany
| | - Bruno Crestani
- Université Paris Cité, Inserm, PHERE, Hôpital Bichat, AP-HP, Service de Pneumologie A, Centre Constitutif du Centre de Référence des Maladies Pulmonaires Rares, FHU APOLLO, Paris, France
| | - Aurélie Fabre
- Department of Histopathology, St Vincent's University Hospital and UCD School of Medicine, University College Dublin, Dublin, Ireland
| | - Antoine Froidure
- Pulmonology Department, Cliniques Universitaires Saint-Luc and Institut de Recherche Expérimentale et Clinique, UCLouvain, Brussels, Belgium
| | - Liam Galvin
- European Pulmonary Fibrosis Federation, Blackrock, Ireland
| | - Matthias Griese
- Dr von Haunersches Kinderspital, University of Munich, German Center for Lung Research (DZL), Munich, Germany
| | - Jan C Grutters
- ILD Center of Excellence, St Antonius Hospital, Nieuwegein, The Netherlands
- Division of Heart and Lungs, UMC Utrecht, Utrecht, The Netherlands
| | - Maria Molina-Molina
- Interstitial Lung Disease Unit, Respiratory Department, University Hospital of Bellvitge, IDIBELL, Hospitalet de Llobregat (Barcelona), CIBERES, Barcelona, Spain
| | - Venerino Poletti
- Department of Diseases of the Thorax, Ospedale GB Morgagni, Forlì, Italy
- Department of Experimental, Diagnostics and Speciality Medicine, University of Bologna, Bologna, Italy
| | - Antje Prasse
- Department of Pulmonology, Hannover Medical School, German Center for Lung Research (DZL), BREATH, Hannover, Germany
- Fraunhofer ITEM, Hannover, Germany
| | - Elisabetta Renzoni
- Interstitial Lung Disease Unit, Royal Brompton and Harefield Clinical Group, Guy's and St Thomas' NHS Foundation Trust, London, UK
- Margaret Turner Warwick Centre for Fibrosing Lung Disease, National Heart and Lung Institute, Imperial College London, London, UK
| | - Jasper van der Smagt
- Division of Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | | |
Collapse
|
12
|
Groen K, van der Vis JJ, van Batenburg AA, Kazemier KM, Grutters JC, van Moorsel CHM. Genetic Variant Overlap Analysis Identifies Established and Putative Genes Involved in Pulmonary Fibrosis. Int J Mol Sci 2023; 24:ijms24032790. [PMID: 36769106 PMCID: PMC9917193 DOI: 10.3390/ijms24032790] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/23/2023] [Accepted: 01/28/2023] [Indexed: 02/05/2023] Open
Abstract
In only around 40% of families with pulmonary fibrosis (PF) a suspected genetic cause can be found. Genetic overlap analysis of Whole Exome Sequencing (WES) data may be a powerful tool to discover new shared variants in novel genes for PF. As a proof of principle, we first selected unrelated PF patients for whom a genetic variant was detected (n = 125) in established PF genes and searched for overlapping variants. Second, we performed WES (n = 149) and identified novel potentially deleterious variants shared by at least two unrelated PF patients. These variants were genotyped in validation cohorts (n = 2748). In 125 unrelated patients, a potentially deleterious variant was detected in known PF genes of which 15 variants in six genes overlapped, involving 51 patients. Overlap analysis of WES data identified two novel variants of interest: TOM1L2 c.421T > C p.(Y141H) and TDP1c.1373dupG p.(S459fs*5), neither gene had been related to pulmonary fibrosis before. Both proteins were present in the alveolar epithelium. No apparent characteristics of telomere disease were observed. This study underlines the potential of searching for overlapping rare potentially deleterious variants to identify disease-associated variants and genes. A previously unreported variant was found in two putative new PF genes, but further research is needed to determine causality.
Collapse
Affiliation(s)
- Karlijn Groen
- Department of Pulmonology, St. Antonius ILD Center of Excellence, St. Antonius Hospital, 3435 CM Nieuwegein, The Netherlands
| | - Joanne J. van der Vis
- Department of Pulmonology, St. Antonius ILD Center of Excellence, St. Antonius Hospital, 3435 CM Nieuwegein, The Netherlands
- Department of Clinical Chemistry, St. Antonius ILD Center of Excellence, St. Antonius Hospital, 3435 CM Nieuwegein, The Netherlands
| | - Aernoud A. van Batenburg
- Department of Pulmonology, St. Antonius ILD Center of Excellence, St. Antonius Hospital, 3435 CM Nieuwegein, The Netherlands
| | - Karin M. Kazemier
- Center of Translational Immunology, University Medical Center Utrecht, 3508 GA Utrecht, The Netherlands
- Division of Hearts and Lungs, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Jan C. Grutters
- Department of Pulmonology, St. Antonius ILD Center of Excellence, St. Antonius Hospital, 3435 CM Nieuwegein, The Netherlands
- Division of Hearts and Lungs, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Coline H. M. van Moorsel
- Department of Pulmonology, St. Antonius ILD Center of Excellence, St. Antonius Hospital, 3435 CM Nieuwegein, The Netherlands
- Correspondence:
| |
Collapse
|
13
|
Revy P, Kannengiesser C, Bertuch AA. Genetics of human telomere biology disorders. Nat Rev Genet 2023; 24:86-108. [PMID: 36151328 DOI: 10.1038/s41576-022-00527-z] [Citation(s) in RCA: 68] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/11/2022] [Indexed: 01/24/2023]
Abstract
Telomeres are specialized nucleoprotein structures at the ends of linear chromosomes that prevent the activation of DNA damage response and repair pathways. Numerous factors localize at telomeres to regulate their length, structure and function, to avert replicative senescence or genome instability and cell death. In humans, Mendelian defects in several of these factors can result in abnormally short or dysfunctional telomeres, causing a group of rare heterogeneous premature-ageing diseases, termed telomeropathies, short-telomere syndromes or telomere biology disorders (TBDs). Here, we review the TBD-causing genes identified so far and describe their main functions associated with telomere biology. We present molecular aspects of TBDs, including genetic anticipation, phenocopy, incomplete penetrance and somatic genetic rescue, which underlie the complexity of these diseases. We also discuss the implications of phenotypic and genetic features of TBDs on fundamental aspects related to human telomere biology, ageing and cancer, as well as on diagnostic, therapeutic and clinical approaches.
Collapse
Affiliation(s)
- Patrick Revy
- INSERM UMR 1163, Laboratory of Genome Dynamics in the Immune System, Equipe Labellisée Ligue Nationale contre le Cancer, Paris, France.
- Université Paris Cité, Imagine Institute, Paris, France.
| | - Caroline Kannengiesser
- APHP Service de Génétique, Hôpital Bichat, Paris, France
- Inserm U1152, Université Paris Cité, Paris, France
| | - Alison A Bertuch
- Departments of Paediatrics and Molecular & Human Genetics, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
14
|
Ding D, Gao R, Xue Q, Luan R, Yang J. Genomic Fingerprint Associated with Familial Idiopathic Pulmonary Fibrosis: A Review. Int J Med Sci 2023; 20:329-345. [PMID: 36860670 PMCID: PMC9969503 DOI: 10.7150/ijms.80358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 01/12/2023] [Indexed: 02/04/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a severe interstitial lung disease; although the recent introduction of two anti-fibrosis drugs, pirfenidone and Nidanib, have resulted in a significant reduction in lung function decline, IPF is still not curable. Approximately 2-20% of patients with IPF have a family history of the disease, which is considered the strongest risk factor for idiopathic interstitial pneumonia. However, the genetic predispositions of familial IPF (f-IPF), a particular type of IPF, remain largely unknown. Genetics affect the susceptibility and progression of f-IPF. Genomic markers are increasingly being recognized for their contribution to disease prognosis and drug therapy outcomes. Existing data suggest that genomics may help identify individuals at risk for f-IPF, accurately classify patients, elucidate key pathways involved in disease pathogenesis, and ultimately develop more effective targeted therapies. Since several genetic variants associated with the disease have been found in f-IPF, this review systematically summarizes the latest progress in the gene spectrum of the f-IPF population and the underlying mechanisms of f-IPF. The genetic susceptibility variation related to the disease phenotype is also illustrated. This review aims to improve the understanding of the IPF pathogenesis and facilitate his early detection.
Collapse
Affiliation(s)
- Dongyan Ding
- Department of Respiratory Medicine, The Second Hospital of Jilin University, Changchun, China
| | - Rong Gao
- Department of Respiratory Medicine, The Second Hospital of Jilin University, Changchun, China
| | - Qianfei Xue
- Hospital of Jilin University, Changchun, China
| | - Rumei Luan
- Department of Respiratory Medicine, The Second Hospital of Jilin University, Changchun, China
| | - Junling Yang
- Department of Respiratory Medicine, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
15
|
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
|
16
|
Abstract
Telomere biology was first studied in maize, ciliates, yeast, and mice, and in recent decades, it has informed understanding of common disease mechanisms with broad implications for patient care. Short telomere syndromes are the most prevalent premature aging disorders, with prominent phenotypes affecting the lung and hematopoietic system. Less understood are a newly recognized group of cancer-prone syndromes that are associated with mutations that lengthen telomeres. A large body of new data from Mendelian genetics and epidemiology now provides an opportunity to reconsider paradigms related to the role of telomeres in human aging and cancer, and in some cases, the findings diverge from what was interpreted from model systems. For example, short telomeres have been considered potent drivers of genome instability, but age-associated solid tumors are rare in individuals with short telomere syndromes, and T cell immunodeficiency explains their spectrum. More commonly, short telomeres promote clonal hematopoiesis, including somatic reversion, providing a new leukemogenesis paradigm that is independent of genome instability. Long telomeres, on the other hand, which extend the cellular life span in vitro, are now appreciated to be the most common shared germline risk factor for cancer in population studies. Through this contemporary lens, I revisit here the role of telomeres in human aging, focusing on how short and long telomeres drive cancer evolution but through distinct mechanisms.
Collapse
Affiliation(s)
- Mary Armanios
- Departments of Oncology, Genetic Medicine, Pathology, and Molecular Biology and Genetics; Telomere Center at Johns Hopkins; and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA;
| |
Collapse
|
17
|
Guérin C, Crestani B, Dupin C, Kawano-Dourado L, Ba I, Kannengiesser C, Borie R. [Telomeres and lung]. Rev Mal Respir 2022; 39:595-606. [PMID: 35715316 DOI: 10.1016/j.rmr.2022.03.011] [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: 03/12/2021] [Accepted: 02/26/2022] [Indexed: 10/18/2022]
Abstract
Genetic studies of familial forms of interstitial lung disease (ILD) have led to the discovery of telomere-related gene (TRG) mutations (TERT, TERC, RTEL1, PARN, DKC1, TINF2, NAF1, NOP10, NHP2, ACD, ZCCH8) in approximately 30% of familial ILD forms. ILD patients with TRG mutation are also subject to extra-pulmonary (immune-hematological, hepatic and/or mucosal-cutaneous) manifestations. TRG mutations may be associated not only with idiopathic pulmonary fibrosis (IPF), but also with non-IPF ILDs, including idiopathic and secondary ILDs, such as hypersensitivity pneumonitis (HP). The presence of TRG mutation may also be associated with an accelerated decline of forced vital capacity (FVC) or poorer prognosis after lung transplantation, notwithstanding which, usual ILD treatments may be proposed. Lastly, patients and their relatives are called upon to reduce their exposure to environmental lung toxicity, and are likely to derive benefit from specific genetic counseling and pre-symptomatic genetic testing.
Collapse
Affiliation(s)
- C Guérin
- Service de Pneumologie A, Centre de compétences maladies pulmonaires rares, AP-HP, Hôpital Bichat, Paris, France..
| | - B Crestani
- Service de Pneumologie A, Centre de compétences maladies pulmonaires rares, AP-HP, Hôpital Bichat, Paris, France.; INSERM, Unité 1152; Université Paris Diderot, Paris, France
| | - C Dupin
- Service de Pneumologie A, Centre de compétences maladies pulmonaires rares, AP-HP, Hôpital Bichat, Paris, France.; INSERM, Unité 1152; Université Paris Diderot, Paris, France
| | - L Kawano-Dourado
- INSERM, Unité 1152; Université Paris Diderot, Paris, France.; HCor Research Institute, Hôpital de Caracao, Sao Paulo, Brésil.; Département de Pneumologie, InCor, Université de Sao Paulo, Sao Paulo, Brésil
| | - I Ba
- INSERM, Unité 1152; Université Paris Diderot, Paris, France.; Département de Génétique, AP-HP, Hôpital Bichat, Paris, France
| | - C Kannengiesser
- INSERM, Unité 1152; Université Paris Diderot, Paris, France.; Département de Génétique, AP-HP, Hôpital Bichat, Paris, France
| | - R Borie
- Service de Pneumologie A, Centre de compétences maladies pulmonaires rares, AP-HP, Hôpital Bichat, Paris, France.; INSERM, Unité 1152; Université Paris Diderot, Paris, France
| |
Collapse
|
18
|
Abstract
Parenchymal lung disease is the fourth leading cause of death in the United States; among the top causes, it continues on the rise. Telomeres and telomerase have historically been linked to cellular processes related to aging and cancer, but surprisingly, in the recent decade genetic discoveries have linked the most apparent manifestations of telomere and telomerase dysfunction in humans to the etiology of lung disease: both idiopathic pulmonary fibrosis (IPF) and emphysema. The short telomere defect is pervasive in a subset of IPF patients, and human IPF is the phenotype most intimately tied to germline defects in telomere maintenance. One-third of families with pulmonary fibrosis carry germline mutations in telomerase or other telomere maintenance genes, and one-half of patients with apparently sporadic IPF have short telomere length. Beyond explaining genetic susceptibility, short telomere length uncovers clinically relevant syndromic extrapulmonary disease, including a T-cell immunodeficiency and a propensity to myeloid malignancies. Recognition of this subset of patients who share a unifying molecular defect has provided a precision medicine paradigm wherein the telomere-mediated lung disease diagnosis provides more prognostic value than histopathology or multidisciplinary evaluation. Here, we critically evaluate this progress, emphasizing how the genetic findings put forth a new pathogenesis paradigm of age-related lung disease that links telomere abnormalities to alveolar stem senescence, remodeling, and defective gas exchange.
Collapse
Affiliation(s)
- Jonathan K. Alder
- Division of Pulmonary and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh PA, United States
| | - Mary Armanios
- Departments of Oncology and Genetic Medicine, Telomere Center at Johns Hopkins, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
19
|
Functional interaction between compound heterozygous TERT mutations causes severe telomere biology disorder. Blood Adv 2022; 6:3779-3791. [PMID: 35477117 DOI: 10.1182/bloodadvances.2022007029] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 04/07/2022] [Indexed: 11/20/2022] Open
Abstract
Telomere biology disorders (TBDs) are a spectrum of multisystem inherited disorders characterized by bone marrow failure, resulting from mutations in genes encoding telomerase or other proteins involved in maintaining telomere length and integrity. Pathogenicity of variants in these genes can be hard to evaluate, since TBD mutations show highly variable penetrance and genetic anticipation due to inheritance of shorter telomeres with each generation. Thus, detailed functional analysis of newly identified variants is often essential. Here we describe a patient with compound heterozygous variants in the TERT gene, which encodes the catalytic subunit of telomerase, hTERT; this patient has the extremely severe Hoyeraal-Hreidarsson form of TBD, although his heterozygous parents are clinically unaffected. Molecular dynamic modeling and detailed biochemical analyses demonstrate that 1 allele (L557P) affects association of hTERT with its cognate RNA component hTR, while the other (K1050E) affects the binding of telomerase to its DNA substrate and enzyme processivity. Unexpectedly, the data demonstrate a functional interaction between the proteins encoded by the 2 alleles, with WT hTERT able to rescue the effect of K1050E on processivity, whereas L557P hTERT cannot. These data contribute to the mechanistic understanding of telomerase, indicating that RNA binding in 1 hTERT molecule affects the processivity of telomere addition by the other molecule. This work emphasizes the importance of functional characterization of TERT variants to reach a definitive molecular diagnosis for TBD patients, and in particular it illustrates the importance of analyzing the effects of compound heterozygous variants in combination to reveal interallelic effects.
Collapse
|
20
|
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
|
21
|
Zhang D, Newton CA. Familial Pulmonary Fibrosis: Genetic Features and Clinical Implications. Chest 2021; 160:1764-1773. [PMID: 34186035 PMCID: PMC8628177 DOI: 10.1016/j.chest.2021.06.037] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 06/21/2021] [Accepted: 06/22/2021] [Indexed: 11/24/2022] Open
Abstract
Pulmonary fibrosis comprises a wide range of fibrotic lung diseases with unknown pathogenesis and poor prognosis. Familial pulmonary fibrosis (FPF) represents a unique subgroup of patients in which at least one other relative is also affected. Patients with FPF exhibit a wide range of pulmonary fibrosis phenotypes, although idiopathic pulmonary fibrosis is the most common subtype. Despite variable disease manifestations, patients with FPF experience worse survival compared with their counterparts with the sporadic disease form. Therefore, ascertaining a positive family history not only provides prognostic value but should also raise suspicion for the inheritance of an underlying causative genetic variant within kindreds. By focusing on FPF kindreds, rare variants within surfactant metabolism and telomere maintenance genes have been discovered. However, such genetic variation is not solely restricted to FPF, as similar rare variants are found in patients with seemingly sporadic pulmonary fibrosis, further supporting the idea of genetic susceptibility underlying pulmonary fibrosis as a whole. Researchers are beginning to show how the presence of rare variants may inform clinical management, such as informing predisposition risk for yet unaffected relatives as well as informing prognosis and therapeutic strategy for those already affected. Despite these advances, rare variants in surfactant and telomere-related genes only explain the genetic basis in about one-quarter of FPF kindreds. Therefore, research is needed to identify the missing genetic contributors of pulmonary fibrosis, which would not only improve our understanding of disease pathobiology but may offer additional opportunities to improve the health of patients.
Collapse
Affiliation(s)
- David Zhang
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Columbia University Irving Medical Center, New York, NY
| | - Chad A Newton
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Texas Southwestern Medical Center, Dallas, TX.
| |
Collapse
|
22
|
Borie R, Renzoni E. Pulmonary fibrosis associated with telomere-related gene mutations: A complex inheritance. Respirology 2021; 26:1098-1100. [PMID: 34617352 DOI: 10.1111/resp.14168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 09/29/2021] [Indexed: 10/20/2022]
Affiliation(s)
- Raphael Borie
- Service de Pneumologie, Centre constitutif du centre de référence, des Maladies Pulmonaires Rares, Hôpital Bichat, APHP, INSERM, Université Paris Diderot, Paris, France
| | - Elisabetta Renzoni
- Interstitial Lung Disease Unit, Royal Brompton and Harefield Clinical Group, Guy's and St Thomas' NHS Foundation Trust, London, UK.,Margaret Turner Warwick Centre for Fibrosing Lung Diseases, NHLI, Imperial College, London, UK
| |
Collapse
|
23
|
van der Vis JJ, van der Smagt JJ, van Batenburg AA, Goldschmeding R, van Es HW, Grutters JC, van Moorsel CHM. Pulmonary fibrosis in non-mutation carriers of families with short telomere syndrome gene mutations. Respirology 2021; 26:1160-1170. [PMID: 34580961 DOI: 10.1111/resp.14145] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 07/16/2021] [Accepted: 08/23/2021] [Indexed: 11/29/2022]
Abstract
BACKGROUND AND OBJECTIVE Diagnostic and predictive genetic testing for disease cause and risk estimation is common in many countries. For genetic diseases, predictive test results are commonly straightforward: presence of the mutation involves increased risk for disease and absence of the mutation involves no inherit risk for disease. Germline mutations in telomere-related genes (TRGs) can lead to telomere shortening and are associated with short telomere syndrome (STS). Telomere length is heritable, and in families with STS due to a TRG mutation, progeny with and without the TRG mutation is known to have shorter than average telomeres. We hypothesize that progeny of TRG mutation carriers who did not inherit the TRG mutation may still develop pulmonary fibrosis. METHODS A genetic screen of 99 unrelated families with familial pulmonary fibrosis revealed five patients with features of pulmonary fibrosis but without carrying the familial disease-causing TRG mutation. RESULTS Features of STS were present in each family, including short telomeres in blood and tissue of the non-mutation carrying patients. Additional genetic, clinical or environmental risk factors for pulmonary fibrosis were present in each non-mutation carrying patient. CONCLUSION Our study shows that non-mutation carrying first-degree relatives in families with STS are at increased risk for pulmonary fibrosis. Disease development may be triggered by inherited short telomeres and additional risk factors for disease. This observation has profound consequences for genetic counselling. Unlike any other genetic syndrome, absence of the mutation does not imply absence of disease risk. Therefore, clinical follow-up is still urged for non-mutation carrying first-degree family members.
Collapse
Affiliation(s)
- Joanne J van der Vis
- ILD Center of Excellence, Department of Pulmonology, St Antonius Hospital, Nieuwegein, The Netherlands
| | - Jasper J van der Smagt
- ILD Center of Excellence, Department of Clinical Chemistry, St Antonius Hospital, Nieuwegein, The Netherlands
| | | | - Roel Goldschmeding
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - H Wouter van Es
- ILD Center of Excellence, Department of Radiology, St Antonius Hospital, Nieuwegein, The Netherlands
| | - Jan C Grutters
- Division of Heart and Lungs, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Coline H M van Moorsel
- Division of Heart and Lungs, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
24
|
Planas-Cerezales L, Arias-Salgado EG, Berastegui C, Montes-Worboys A, González-Montelongo R, Lorenzo-Salazar JM, Vicens-Zygmunt V, Garcia-Moyano M, Dorca J, Flores C, Perona R, Román A, Molina-Molina M. Lung Transplant Improves Survival and Quality of Life Regardless of Telomere Dysfunction. Front Med (Lausanne) 2021; 8:695919. [PMID: 34395476 PMCID: PMC8362799 DOI: 10.3389/fmed.2021.695919] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 05/27/2021] [Indexed: 12/30/2022] Open
Abstract
Introduction: Fibrotic interstitial lung diseases (ILDs) are the first indication for lung transplantation (LT). Telomere dysfunction has been associated with poor post-transplant outcomes. The aim of the study was to evaluate the morbi-mortality and quality of life in fibrotic ILDs after lung transplant depending on telomere biology. Methods: Fibrotic ILD patients that underwent lung transplant were allocated to two arms; with or without telomere dysfunction at diagnosis based on the telomere length and telomerase related gene mutations revealed by whole-exome sequencing. Post-transplant evaluation included: (1) short and long-term mortality and complications and (2) quality of life. Results: Fifty-five percent of patients that underwent LT carried rare coding mutations in telomerase-related genes. Patients with telomere shortening more frequently needed extracorporeal circulation and presented a higher rate of early post-transplant hematological complications, longer stay in the intensive care unit (ICU), and a higher number of long-term hospital admissions. However, post-transplant 1-year survival was higher than 80% regardless of telomere dysfunction, with improvement in the quality of life and oxygen therapy withdrawal. Conclusions: Post-transplant morbidity is higher in patients with telomere dysfunction and differs according to elapsed time from transplantation. However, lung transplant improves survival and quality of life and the associated complications are manageable.
Collapse
Affiliation(s)
- Lurdes Planas-Cerezales
- ILD Multidisciplinary Unit, Hospital Universitari Bellvitge, IDIBELL, Universitat de Barcelona, Hospitalet de Llobregat, Spain
| | - Elena G Arias-Salgado
- Biomedical Research Institute CSIC/UAM, IdIPAZ, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain
| | - Cristina Berastegui
- Respiratory Department, Institute of Research, Hospital Universitari Vall d'Hebrón, Universitat Autónoma de Barcelona, Barcelona, Spain
| | - Ana Montes-Worboys
- ILD Multidisciplinary Unit, Hospital Universitari Bellvitge, IDIBELL, Universitat de Barcelona, Hospitalet de Llobregat, Spain
| | | | - José M Lorenzo-Salazar
- Genomics Division, Instituto Tecnológico y de Energías Renovables, Santa Cruz de Tenerife, Spain
| | - Vanesa Vicens-Zygmunt
- ILD Multidisciplinary Unit, Hospital Universitari Bellvitge, IDIBELL, Universitat de Barcelona, Hospitalet de Llobregat, Spain
| | | | - Jordi Dorca
- ILD Multidisciplinary Unit, Hospital Universitari Bellvitge, IDIBELL, Universitat de Barcelona, Hospitalet de Llobregat, Spain
| | - Carlos Flores
- Genomics Division, Instituto Tecnológico y de Energías Renovables, Santa Cruz de Tenerife, Spain.,Research Unit, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain.,Instituto de Tecnologías Biomédicas, Universidad de La Laguna, Santa Cruz de Tenerife, Spain.,Centro Investigación Biomédica en Red de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
| | - Rosario Perona
- Biomedical Research Institute CSIC/UAM, IdIPAZ, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain
| | - Antonio Román
- Respiratory Department, Institute of Research, Hospital Universitari Vall d'Hebrón, Universitat Autónoma de Barcelona, Barcelona, Spain
| | - María Molina-Molina
- ILD Multidisciplinary Unit, Hospital Universitari Bellvitge, IDIBELL, Universitat de Barcelona, Hospitalet de Llobregat, Spain.,Centro Investigación Biomédica en Red de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
25
|
Ptasinski V, Stegmayr J, Belvisi MG, Wagner DE, Murray LA. Targeting Alveolar Repair in Idiopathic Pulmonary Fibrosis. Am J Respir Cell Mol Biol 2021; 65:347-365. [PMID: 34129811 PMCID: PMC8525210 DOI: 10.1165/rcmb.2020-0476tr] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Idiopathic pulmonary fibrosis is a fatal interstitial lung disease with limited therapeutic options. Current evidence suggests that IPF may be initiated by repeated epithelial injury in the distal lung followed by abnormal wound healing responses which occur due to intrinsic and extrinsic factors. Mechanisms contributing to chronic damage of the alveolar epithelium in IPF include dysregulated cellular processes such as apoptosis, senescence, abnormal activation of developmental pathways, aging, as well as genetic mutations. Therefore, targeting the regenerative capacity of the lung epithelium is an attractive approach in the development of novel therapies for IPF. Endogenous lung regeneration is a complex process involving coordinated cross-talk between multiple cell types and re-establishment of a normal extracellular matrix environment. This review will describe the current knowledge of reparative epithelial progenitor cells in the alveolar region of the lung and discuss potential novel therapeutic approaches for IPF focusing on endogenous alveolar repair. This article is open access and distributed under the terms of the Creative Commons Attribution Non-Commercial No Derivatives License 4.0 (http://creativecommons.org/licenses/by-nc-nd/4.0/).
Collapse
Affiliation(s)
- Victoria Ptasinski
- Lund University Faculty of Medicine, 59568, Lund, Sweden.,AstraZeneca R&D Gothenburg, 128698, Goteborg, Sweden
| | - John Stegmayr
- Lunds University Faculty of Medicine, 59568, Lund, Sweden
| | - Maria G Belvisi
- Imperial College London, 4615, London, United Kingdom of Great Britain and Northern Ireland
| | - Darcy E Wagner
- Lunds Universitet, 5193, Experimental Medical Sciences, Lund, Sweden
| | - Lynne A Murray
- AstraZeneca PLC, 4625, Cambridge, United Kingdom of Great Britain and Northern Ireland;
| |
Collapse
|
26
|
van Batenburg AA, Kazemier KM, van Oosterhout MFM, van der Vis JJ, Grutters JC, Goldschmeding R, van Moorsel CHM. Telomere shortening and DNA damage in culprit cells of different types of progressive fibrosing interstitial lung disease. ERJ Open Res 2021; 7:00691-2020. [PMID: 34084786 PMCID: PMC8165375 DOI: 10.1183/23120541.00691-2020] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 12/14/2020] [Indexed: 12/11/2022] Open
Abstract
Pulmonary fibrosis is strongly associated with telomere shortening and increased DNA damage. Key cells in the pathogenesis involve alveolar type 2 (AT2) cells, club cells and myofibroblasts; however, to what extent these cells are affected by telomere shortening and DNA damage is not yet known. We sought to determine the degree of, and correlation between, telomere shortening and DNA damage in different cell types involved in the pathogenesis of progressive fibrosing interstitial lung disease. Telomere length and DNA damage were quantified, using combined fluorescence in situ hybridisation and immunofluorescence staining techniques, in AT2 cells, club cells and myofibroblasts of controls and patients with pulmonary fibrosis and a telomerase reverse transcriptase mutation (TERT-PF), idiopathic pulmonary fibrosis (IPF) and fibrotic hypersensitivity pneumonitis (fHP). In IPF and TERT-PF lungs, AT2 cells contained shorter telomeres and expressed higher DNA damage signals than club cells and myofibroblasts. In fHP lungs, club cells contained highly elevated levels of DNA damage, while telomeres were not obviously short. In vitro, we found significantly shorter telomeres and higher DNA damage levels only in AT2 surrogate cell lines treated with telomerase inhibitor BIBR1532. Our study demonstrated that in IPF and TERT-PF lungs, telomere shortening and accumulation of DNA damage primarily affects AT2 cells, further supporting the importance of AT2 cells in these diseases, while in fHP the particularly high telomere-independent DNA damage signals in club cells underscores its bronchiolocentric pathogenesis. These findings suggest that cell type-specific telomere shortening and DNA damage may help to discriminate between different drivers of fibrogenesis.
Collapse
Affiliation(s)
- Aernoud A van Batenburg
- Dept of Pulmonology, St Antonius ILD Center of Excellence, St Antonius Hospital, Nieuwegein, The Netherlands
| | - Karin M Kazemier
- Center of Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands.,Division of Heart and Lungs, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Matthijs F M van Oosterhout
- Dept of Pathology, Pathology DNA, St Antonius ILD Center of Excellence, St Antonius Hospital, Nieuwegein, The Netherlands
| | - Joanne J van der Vis
- Dept of Pulmonology, St Antonius ILD Center of Excellence, St Antonius Hospital, Nieuwegein, The Netherlands.,Dept of Clinical Chemistry, St Antonius ILD Center of Excellence, St Antonius Hospital, Nieuwegein, The Netherlands
| | - Jan C Grutters
- Dept of Pulmonology, St Antonius ILD Center of Excellence, St Antonius Hospital, Nieuwegein, The Netherlands.,Division of Heart and Lungs, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Roel Goldschmeding
- Dept of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Coline H M van Moorsel
- Dept of Pulmonology, St Antonius ILD Center of Excellence, St Antonius Hospital, Nieuwegein, The Netherlands.,Division of Heart and Lungs, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
27
|
Cocconcelli E, Tonelli R, Abbati G, Marchioni A, Castaniere I, Pelizzaro F, Russo FP, Vegetti A, Balestro E, Pietrangelo A, Richeldi L, Luppi F, Spagnolo P, Clini E, Cerri S. Subclinical liver fibrosis in patients with idiopathic pulmonary fibrosis. Intern Emerg Med 2021; 16:349-357. [PMID: 32451930 DOI: 10.1007/s11739-020-02376-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Accepted: 05/12/2020] [Indexed: 01/29/2023]
Abstract
Data on the presence of subclinical fibrosis across multiple organs in patients with idiopathic lung fibrosis (IPF) are lacking. Our study aimed at investigating through hepatic transient elastography (HTE) the prevalence and clinical impact of subclinical liver fibrosis in a cohort of patients with IPF. Patients referred to the Centre for Rare Lung Disease of the University Hospital of Modena (Italy) from March 2012 to February 2013 with established diagnosis of IPF and without a documented history of liver diseases were consecutively enrolled and underwent HTE. Based on hepatic stiffness status as assessed through METAVIR score patients were categorized as "with liver fibrosis" (corresponding to a METAVIR score of F1-F4) and "without liver fibrosis" (METAVIR F0). Potential predictors of liver fibrosis were investigated through logistic regression model among clinical and serological variables. The overall survival (OS) was assessed according to liver fibrosis and multivariate Cox regression analysis was used to identify independent predictors. In 13 out of 37 patients (35%) with IPF, a certain degree of liver fibrosis was documented. No correlation was found between liver stiffness and clinical-functional parameters. OS was lower in patients 'with liver fibrosis' than in patients 'without liver fibrosis' (median months 33 [23-55] vs. 63 [26-94], p = 0.038). Patients 'with liver fibrosis' presented a higher risk of death at seven years as compared to patients 'without liver fibrosis' (HR = 2.6, 95% CI [1.003-6.7], p = 0.049). Higher level of AST to platelet ratio index (APRI) was an independent predictor of survival (HR = 4.52 95% CI [1.3-15.6], p = 0.02). In our cohort, more than one-third of IPF patients had concomitant subclinical liver fibrosis that negatively affected OS. These preliminary claims further investigation aimed at clarifying the mechanisms beyond multiorgan fibrosis and its clinical implication in patients with IPF.
Collapse
Affiliation(s)
- Elisabetta Cocconcelli
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University Hospital of Padova, Padua, Italy
| | - Roberto Tonelli
- Department of Medical and Surgical Sciences, University Hospital of Modena, Respiratory Diseases Unit and Centre for Rare Lung Diseases, University of Modena Reggio Emilia, Modena, Italy
- Clinical and Experimental Medicine PhD Program, University of Modena Reggio Emilia, Modena, Italy
| | - Gianluca Abbati
- Department of Medical and Surgical Sciences, University Hospital of Modena, Internal Medicine Unit, University of Modena Reggio Emilia, Modena, Italy
| | - Alessandro Marchioni
- Department of Medical and Surgical Sciences, University Hospital of Modena, Respiratory Diseases Unit and Centre for Rare Lung Diseases, University of Modena Reggio Emilia, Modena, Italy
| | - Ivana Castaniere
- Department of Medical and Surgical Sciences, University Hospital of Modena, Respiratory Diseases Unit and Centre for Rare Lung Diseases, University of Modena Reggio Emilia, Modena, Italy
- Clinical and Experimental Medicine PhD Program, University of Modena Reggio Emilia, Modena, Italy
| | - Filippo Pelizzaro
- Department of Surgical, Oncological and Gastroenterological Sciences, Unit of Gastroenterology, University of Padova, Padua, Italy
| | - Francesco Paolo Russo
- Department of Surgical, Oncological and Gastroenterological Sciences, Unit of Gastroenterology, University of Padova, Padua, Italy
| | - Alberto Vegetti
- Department of Medical and Surgical Sciences, University Hospital of Modena, Internal Medicine Unit, University of Modena Reggio Emilia, Modena, Italy
| | - Elisabetta Balestro
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University Hospital of Padova, Padua, Italy
| | - Antonello Pietrangelo
- Department of Medical and Surgical Sciences, University Hospital of Modena, Internal Medicine Unit, University of Modena Reggio Emilia, Modena, Italy
| | - Luca Richeldi
- University Cattolica del Sacro Cuore, Respiratory Diseases Unit, University Hospital Agostino Gemelli, Rome, Italy
| | - Fabrizio Luppi
- Respiratory Unit, S. Gerardo Hospital, University of Milano Bicocca, Monza, Italy
| | - Paolo Spagnolo
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University Hospital of Padova, Padua, Italy
| | - Enrico Clini
- Department of Medical and Surgical Sciences, University Hospital of Modena, Respiratory Diseases Unit and Centre for Rare Lung Diseases, University of Modena Reggio Emilia, Modena, Italy.
| | - Stefania Cerri
- Department of Medical and Surgical Sciences, University Hospital of Modena, Respiratory Diseases Unit and Centre for Rare Lung Diseases, University of Modena Reggio Emilia, Modena, Italy
| |
Collapse
|
28
|
Pardo A, Selman M. The Interplay of the Genetic Architecture, Aging, and Environmental Factors in the Pathogenesis of Idiopathic Pulmonary Fibrosis. Am J Respir Cell Mol Biol 2021; 64:163-172. [PMID: 32946290 DOI: 10.1165/rcmb.2020-0373ps] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic fibrosing lung disease of indeterminate etiology and limited therapeutic options. The initiation, development, and progression of IPF are influenced by genetic predisposition, aging, and host and environmental factors, but the magnitude of the contribution of each of them and the sequence of the pathogenic events are uncertain. Current evidence indicates that accumulated environmental exposures in a genetically predisposed individual, usually over 60 years of age, leads to phenotypic and functional alterations of the lung epithelium. Aberrant activation of epithelial cells results, through a complex release of numerous mediators, in the local expansion of peculiar subsets of aggressive fibroblasts and myofibroblasts, which are crucial effector cells of fibrotic remodeling and loss of the normal lung architecture and function. Progressive increase of the mechanical stiffness activates cell-autonomous and matrix-dependent processes contributing to the perpetuation of the fibrotic response. This Perspective provides an integral overview of the major risk factors underpinning the pathogenesis of IPF, including gene variants, aging alterations, environmental factors, host risk factors, and epigenetic reprogramming.
Collapse
Affiliation(s)
- Annie Pardo
- Facultad de Ciencias, Universidad Nacional Autónoma de México, México City, Mexico; and
| | - Moisés Selman
- Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas," México City, Mexico
| |
Collapse
|
29
|
Cutting CC, Bowman WS, Dao N, Pugashetti JV, Garcia CK, Oldham JM, Newton CA. Family History of Pulmonary Fibrosis Predicts Worse Survival in Patients With Interstitial Lung Disease. Chest 2021; 159:1913-1921. [PMID: 33484728 DOI: 10.1016/j.chest.2021.01.026] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 01/13/2021] [Accepted: 01/14/2021] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND A number of genetic markers linked to familial pulmonary fibrosis predict differential survival in interstitial lung disease (ILD) patients. Although genetic testing is not performed routinely for ILD, family history commonly is obtained and may inform outcome risk. RESEARCH QUESTION Does survival vary between patients with and without self-reported familial pulmonary fibrosis? METHODS Family history was acquired systematically for consecutive ILD patients who consented to clinical registry enrollment at the University of Texas Southwestern and the University of California at Davis. Patients were stratified by idiopathic pulmonary fibrosis (IPF) and non-IPF ILD diagnosis and were substratified by presence or absence of familial pulmonary fibrosis, defined as one or more additional affected family members. Transplant-free survival was compared using multilevel, mixed-effects Cox proportional hazards regression. RESULTS Of the 1,262 patients included, 534 (42%) had IPF ILD and 728 (58%) had non-IPF ILD. Of those with non-IPF ILD, 18.5% had connective tissue disease, 15.6% had chronic hypersensitivity pneumonitis, and 23.5% had unclassifiable ILD. Familial pulmonary fibrosis was reported in 134 IPF ILD patients (25.1%) and 90 non-IPF ILD patients (12.4%). Those with familial IPF showed an 80% increased risk of death or transplantation compared with those with sporadic IPF (hazard ratio [HR], 1.8; 95% CI, 1.37-2.37; P < .001), whereas those with familial non-IPF ILD showed a twofold increased risk compared with their counterparts with sporadic disease (HR, 2.08; 95% CI, 1.46-2.96; P < .001). Outcome risk among those with familial non-IPF ILD was no different than for those with sporadic IPF ILD (HR, 1.27; 95% CI, 0.89-1.84; P = .19). INTERPRETATION Patient-reported familial pulmonary fibrosis is predictive of reduced transplant-free survival in IPF and non-IPF ILD patients. Because survival among patients with familial non-IPF ILD approximates that of sporadic IPF ILD, early intervention should be considered for such patients. Until clinical genetic testing is widely available and provides actionable results, family history should be ascertained and considered in risk stratification.
Collapse
Affiliation(s)
- Claire C Cutting
- Department of Internal Medicine, University of California at Davis, Sacramento, CA.
| | - Willis S Bowman
- Division of Pulmonary, Critical Care and Sleep Medicine, University of California at Davis, Sacramento, CA
| | - Nam Dao
- Department of Internal Medicine, University of California at Davis, Sacramento, CA
| | - Janelle Vu Pugashetti
- Division of Pulmonary, Critical Care and Sleep Medicine, University of California at Davis, Sacramento, CA
| | - Christine Kim Garcia
- Medicine within the Department of Medicine at Columbia University, College of Physicians and Surgeons, New York, NY
| | - Justin M Oldham
- Division of Pulmonary, Critical Care and Sleep Medicine, University of California at Davis, Sacramento, CA
| | - Chad A Newton
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX
| |
Collapse
|
30
|
FBW7 Mediates Senescence and Pulmonary Fibrosis through Telomere Uncapping. Cell Metab 2020; 32:860-877.e9. [PMID: 33086033 DOI: 10.1016/j.cmet.2020.10.004] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 07/28/2020] [Accepted: 10/02/2020] [Indexed: 12/19/2022]
Abstract
Tissue stem cells undergo premature senescence under stress, promoting age-related diseases; however, the associated mechanisms remain unclear. Here, we report that in response to radiation, oxidative stress, or bleomycin, the E3 ubiquitin ligase FBW7 mediates cell senescence and tissue fibrosis through telomere uncapping. FBW7 binding to telomere protection protein 1 (TPP1) facilitates TPP1 multisite polyubiquitination and accelerates degradation, triggering telomere uncapping and DNA damage response. Overexpressing TPP1 or inhibiting FBW7 by genetic ablation, epigenetic interference, or peptidomimetic telomere dysfunction inhibitor (TELODIN) reduces telomere uncapping and shortening, expanding the pulmonary alveolar AEC2 stem cell population in mice. TELODIN, synthesized from the seventh β strand blade of FBW7 WD40 propeller domain, increases TPP1 stability, lung respiratory function, and resistance to senescence and fibrosis in animals chronically exposed to environmental stress. Our findings elucidate a pivotal mechanism underlying stress-induced pulmonary epithelial stem cell senescence and fibrosis, providing a framework for aging-related disorder interventions.
Collapse
|
31
|
Strikoudis A, Cieślak A, Loffredo L, Chen YW, Patel N, Saqi A, Lederer DJ, Snoeck HW. Modeling of Fibrotic Lung Disease Using 3D Organoids Derived from Human Pluripotent Stem Cells. Cell Rep 2020; 27:3709-3723.e5. [PMID: 31216486 DOI: 10.1016/j.celrep.2019.05.077] [Citation(s) in RCA: 149] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 03/27/2019] [Accepted: 05/20/2019] [Indexed: 02/07/2023] Open
Abstract
The pathogenesis of idiopathic pulmonary fibrosis (IPF), an intractable interstitial lung disease, is unclear. Recessive mutations in some genes implicated in Hermansky-Pudlak syndrome (HPS) cause HPS-associated interstitial pneumonia (HPSIP), a clinical entity that is similar to IPF. We previously reported that HPS1-/- embryonic stem cell-derived 3D lung organoids showed fibrotic changes. Here, we show that the introduction of all HPS mutations associated with HPSIP promotes fibrotic changes in lung organoids, while the deletion of HPS8, which is not associated with HPSIP, does not. Genome-wide expression analysis revealed the upregulation of interleukin-11 (IL-11) in epithelial cells from HPS mutant fibrotic organoids. IL-11 was detected predominantly in type 2 alveolar epithelial cells in end-stage IPF, but was expressed more broadly in HPSIP. Finally, IL-11 induced fibrosis in WT organoids, while its deletion prevented fibrosis in HPS4-/- organoids, suggesting IL-11 as a therapeutic target. hPSC-derived 3D lung organoids are, therefore, a valuable resource to model fibrotic lung disease.
Collapse
Affiliation(s)
- Alexandros Strikoudis
- Columbia Center for Human Development, Columbia University Medical Center, New York, NY 10032, USA; Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA; Division of Pulmonary Medicine, Allergy, and Critical Care, Columbia University Medical Center, New York, NY 10032, USA
| | - Anna Cieślak
- Columbia Center for Human Development, Columbia University Medical Center, New York, NY 10032, USA; Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA; Division of Pulmonary Medicine, Allergy, and Critical Care, Columbia University Medical Center, New York, NY 10032, USA
| | - Lucas Loffredo
- Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY 10032, USA
| | - Ya-Wen Chen
- Columbia Center for Human Development, Columbia University Medical Center, New York, NY 10032, USA; Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA; Division of Pulmonary Medicine, Allergy, and Critical Care, Columbia University Medical Center, New York, NY 10032, USA
| | - Nina Patel
- Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA
| | - Anjali Saqi
- Department of Pathology & Cell Biology, Columbia University Medical Center, New York, NY 10032, USA
| | - David J Lederer
- Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA
| | - Hans-Willem Snoeck
- Columbia Center for Human Development, Columbia University Medical Center, New York, NY 10032, USA; Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA; Division of Pulmonary Medicine, Allergy, and Critical Care, Columbia University Medical Center, New York, NY 10032, USA; Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY 10032, USA.
| |
Collapse
|
32
|
van der Vis JJ, van der Smagt JJ, Hennekam FA, Grutters JC, van Moorsel CH. Pulmonary Fibrosis and a TERT Founder Mutation With a Latency Period of 300 Years. Chest 2020; 158:612-619. [DOI: 10.1016/j.chest.2020.03.069] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 02/10/2020] [Accepted: 03/04/2020] [Indexed: 10/24/2022] Open
|
33
|
Borie R, Kannengiesser C, Dupin C, Debray MP, Cazes A, Crestani B. Impact of genetic factors on fibrosing interstitial lung diseases. Incidence and clinical presentation in adults. Presse Med 2020; 49:104024. [PMID: 32437840 DOI: 10.1016/j.lpm.2020.104024] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 02/11/2019] [Indexed: 12/20/2022] Open
Abstract
At least 10% of patients with pulmonary fibrosis, whether idiopathic or secondary, present heritable pulmonary fibrosis suspected on familial aggregation of pulmonary fibrosis, specific syndromes or early age of diagnosis. Approximately 30% of those patients have an identified mutation mostly in telomere related genes (TRG) more rarely in surfactant homeostasis or other genes. TRG mutation may be associated with hematological and hepatic diseases that may worsen after lung transplantation requiring a specific care and adapted immunosuppression. Surfactant genes mutations are usually associated with ground-glass opacities and cysts on CT scan and may improve with steroids, hydroxychloroquine or azithromycin. Moreover relatives should benefit from a genetic analysis associated with a clinical evaluation according to the gene involved. Genetics of pulmonary fibrosis raise specific problems from diagnosis, therapy or genetic counseling varying from one gene to another.
Collapse
Affiliation(s)
- Raphael Borie
- Unité 1152, Inserm, DHU FIRE, service de pneumologie A, centre de référence des maladies pulmonaires rares, université Paris Diderot, hôpital Bichat, AP-HP, 75013 Paris, France.
| | - Caroline Kannengiesser
- Unité 1152, Inserm, laboratoire de génétique, université Paris Diderot, hôpital Bichat, AP-HP, 75013 Paris, France
| | - Clairelyne Dupin
- Unité 1152, Inserm, DHU FIRE, service de pneumologie A, centre de référence des maladies pulmonaires rares, université Paris Diderot, hôpital Bichat, AP-HP, 75013 Paris, France
| | - Marie-Pierre Debray
- Unité 1152, Inserm, service de radiologie, hôpital Bichat, AP-HP, 75018 Paris, France
| | - Aurélie Cazes
- Inserm, unité 1152, service d'antomopathologie, université Paris Diderot, hôpital Bichat, AP-HP, 75018 Paris, France
| | - Bruno Crestani
- Unité 1152, Inserm, DHU FIRE, service de pneumologie A, centre de référence des maladies pulmonaires rares, université Paris Diderot, hôpital Bichat, AP-HP, 75013 Paris, France
| |
Collapse
|
34
|
Yuan B, Wen X, Li L, Li Y, Li C, Li B, Yuan W, Cui L. NAF1 rs4691896 Is Significantly Associated with Coal Workers' Pneumoconiosis in a Chinese Han Population: A Case-Control Study. Med Sci Monit 2020; 26:e918709. [PMID: 32333749 PMCID: PMC7197226 DOI: 10.12659/msm.918709] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Background Previous studies have demonstrated the important role of genetic predisposition in coal workers’ pneumoconiosis (CWP) in addition to environmental factors. The pathogenesis of pulmonary fibrosis disease is related to telomere activity. We performed this study to assess the association between genetic variants of telomere-related genes and the risk of CWP. Material/Methods We enrolled 652 CWP Chinese Han patients and 648 dust-exposed controls in this case-control design study, genotyping 8 single-nucleotide polymorphisms (SNPs) including TERT (rs2736100), TERC (rs10936599 and rs12696304), and NAF1 (rs7675998, rs3822304, rs12331717, rs936562 and rs4691896) using the Sequenom MassARRAY system. Results We identified a significant allele association between NAF1 rs4691896 and CWP by comparing patients with controls (22.0% vs. 13.0%, odds ratio [OR]: 1.89, 95% confidence interval [CI]: 1.54–2.33, Pc=1.14×10−8). The genotype frequency of rs4691896 differed significantly between the patients and controls (Pc=1.49×10−8). In addition, rs4691896 was correlated with CWP in an additive genetic model (OR: 1.96, 95% CI: 1.58–2.44, Pc=8.96×10−9) and a dominant model (OR: 2.15, 95% CI: 1.70–2.73, Pc=2.39×10−9). Conclusions Our study for the first time demonstrates an association between a telomere-related gene (NAF1) and CWP in a Chinese Han population, and provides valuable insight to further understand the possible pathogenetic mechanism of fibrosis in CWP.
Collapse
Affiliation(s)
- Baojun Yuan
- Department of Clinical Laboratory, Kai Luan General Hospital, Tangshan, Hebei, China (mainland)
| | - Xiaoting Wen
- Department of Clinical Laboratory, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China (mainland)
| | - Liubing Li
- Department of Clinical Laboratory, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China (mainland)
| | - Yongzhe Li
- Department of Clinical Laboratory, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China (mainland)
| | - Chao Li
- Department of Clinical Laboratory, Kai Luan General Hospital, Tangshan, Hebei, China (mainland)
| | - Baolin Li
- Department of Clinical Laboratory, Kai Luan General Hospital, Tangshan, Hebei, China (mainland)
| | - Wei Yuan
- Department of Rheumatology and Clinical Immunology, Kai Luan General Hospital, Tangshan, Hebei, China (mainland)
| | - Liufu Cui
- Department of Rheumatology and Clinical Immunology, Kai Luan General Hospital, Tangshan, Hebei, China (mainland)
| |
Collapse
|
35
|
Abstract
The interstitial lung diseases (ILDs) are a group of progressive disorders characterized by chronic inflammation and/or fibrosis in the lung. While some ILDs can be linked to specific environmental causes (i.e., asbestosis, silicosis), in many individuals, no culprit exposure can be identified; these patients are deemed to have "idiopathic interstitial pneumonia" (IIP). Family history is now recognized as the strongest risk factor for IIP, and IIP cases that run in families comprise a syndrome termed "familial interstitial pneumonia" (FIP). Mutations in more than 10 different genes have been implicated as responsible for disease in FIP families. Diverse ILD clinical phenotypes can be seen within a family, and available evidence suggests underlying genetic risk is the primary determinant of disease outcomes. Together, these FIP studies have provided unique insights into the pathobiology of ILDs, and brought focus on the unique issues that arise in the care of patients with FIP.
Collapse
Affiliation(s)
- 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
- U.S. Department of Veterans Affairs Medical Center, Nashville, Tennessee
| |
Collapse
|
36
|
van Batenburg AA, Kazemier KM, van Oosterhout MFM, van der Vis JJ, van Es HW, Grutters JC, Goldschmeding R, van Moorsel CHM. From organ to cell: Multi-level telomere length assessment in patients with idiopathic pulmonary fibrosis. PLoS One 2020; 15:e0226785. [PMID: 31910222 PMCID: PMC6946122 DOI: 10.1371/journal.pone.0226785] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 12/03/2019] [Indexed: 12/15/2022] Open
Abstract
Rationale A subset of patients with idiopathic pulmonary fibrosis (IPF) contains short leukocyte telomeres or telomere related mutations. We previously showed that alveolar type 2 cells have short telomeres in fibrotic lesions. Our objectives were to better understand how telomere shortening associates with fibrosis in IPF lung and identify a subset of patients with telomere-related disease. Methods Average telomere length was determined in multiple organs, basal and apical lung, and diagnostic and end-stage fibrotic lung biopsies. Alveolar type 2 cells telomere length was determined in different areas of IPF lungs. Results In IPF but not in controls, telomere length in lung was shorter than in other organs, providing rationale to focus on telomere length in lung. Telomere length did not correlate with age and no difference in telomere length was found between diagnostic and explant lung or between basal and apical lung, irrespective of the presence of a radiological apicobasal gradient or fibrosis. Fifteen out of 28 IPF patients had average lung telomere length in the range of patients with a telomerase (TERT) mutation, and formed the IPFshort group. Only in this IPFshort and TERT group telomeres of alveolar type 2 cells were extremely short in fibrotic areas. Additionally, whole exome sequencing of IPF patients revealed two genetic variations in RTEL1 and one in PARN in the IPFshort group. Conclusions Average lung tissue telomere shortening does not associated with fibrotic patterns in IPF, however, approximately half of IPF patients show excessive lung telomere shortening that is associated with pulmonary fibrosis driven by telomere attrition.
Collapse
Affiliation(s)
- Aernoud A. van Batenburg
- Department of Pulmonology, St Antonius ILD Center of Excellence, St Antonius Hospital, Nieuwegein, the Netherlands
| | - Karin M. Kazemier
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
- Department of Respiratory Medicine, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Matthijs F. M. van Oosterhout
- Pathology–DNA, Department of Pathology, St Antonius ILD Center of Excellence St Antonius Hospital, Nieuwegein, The Netherlands
| | - Joanne J. van der Vis
- Department of Pulmonology, St Antonius ILD Center of Excellence, St Antonius Hospital, Nieuwegein, the Netherlands
- Department of Clinical Chemistry, St Antonius ILD Center of Excellence, St Antonius Hospital, Nieuwegein, the Netherlands
| | - Hendrik W. van Es
- Department of Radiology, St Antonius ILD Center of Excellence, St Antonius Hospital, Nieuwegein, the Netherlands
| | - Jan C. Grutters
- Department of Pulmonology, St Antonius ILD Center of Excellence, St Antonius Hospital, Nieuwegein, the Netherlands
- Division of Heart and Lungs, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Roel Goldschmeding
- Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Coline H. M. van Moorsel
- Department of Pulmonology, St Antonius ILD Center of Excellence, St Antonius Hospital, Nieuwegein, the Netherlands
- Division of Heart and Lungs, University Medical Center Utrecht, Utrecht, the Netherlands
- * E-mail:
| |
Collapse
|
37
|
Combining conservation and species-specific differences to determine how human telomerase binds telomeres. Proc Natl Acad Sci U S A 2019; 116:26505-26515. [PMID: 31822618 DOI: 10.1073/pnas.1911912116] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Telomerase catalyzes telomeric DNA synthesis at chromosome ends to allow for continued cell division. The telomeric protein TPP1 is essential for enhancing the processivity of telomerase and recruiting the enzyme to telomeres. The telomerase interaction surface on human TPP1 has been mapped to 2 regions of the N-terminal oligosaccharide/oligonucleotide-binding (OB) domain, namely the TPP1 glutamate (E) and leucine (L)-rich (TEL) patch and the N terminus of TPP1-oligosaccharide/oligonucleotide-binding (NOB) region. To map the telomerase side of the interface, we exploited the predicted structural similarities for human and Tetrahymena thermophila telomerase as well as the species specificity of human and mouse telomerase for their cognate TPP1 partners. We show that swapping in the telomerase essential N-terminal (TEN) and insertions in fingers domain (IFD)-TRAP regions of the human telomerase catalytic protein subunit TERT into the mouse TERT backbone is sufficient to bias the species specificity toward human TPP1. Employing a structural homology-based mutagenesis screen focused on surface residues of the TEN and IFD regions, we identified TERT residues that are critical for contacting TPP1 but dispensable for other aspects of telomerase structure or function. We present a functionally validated structural model for how human telomerase engages TPP1 at telomeres, setting the stage for a high-resolution structure of this interface.
Collapse
|
38
|
Zalzman M, Meltzer WA, Portney BA, Brown RA, Gupta A. The Role of Ubiquitination and SUMOylation in Telomere Biology. Curr Issues Mol Biol 2019; 35:85-98. [PMID: 31422934 DOI: 10.21775/cimb.035.085] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Telomeres are a unique structure of DNA repeats covered by proteins at the ends of the chromosomes that protect the coding regions of the genome and function as a biological clock. They require a tight regulation of the factors covering and protecting their structure, as they are shortened with each cell division to limit the ability of cells to replicate uncontrollably. Additionally, they protect the chromosome ends from DNA damage responses and thereby, prevent genomic instability. Telomere dysfunction can lead to chromosomal abnormalities and cancer. Therefore, dysregulation of any of the factors that regulate the integrity of the telomeres will have implications to chromosomal stability, replicative lifespan and may lead to cell transformation. This review will cover the main factors participating in the normal function of the telomeres and how these are regulated by the ubiquitin and SUMO systems. Accumulating evidence indicate that the ubiquitin and SUMO pathways are significant regulators of the shelterin complex and other chromatin modifiers, which are important for telomere structure integrity. Furthermore, the crosstalk between these two pathways has been reported in telomeric DNA repair. A better understanding of the factors contributing to telomere biology, and how they are regulated, is important for the design of new strategies for cancer therapies and regenerative medicine.
Collapse
Affiliation(s)
- Michal Zalzman
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - W Alex Meltzer
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Benjamin A Portney
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Robert A Brown
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Aditi Gupta
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
39
|
Alnafakh RAA, Adishesh M, Button L, Saretzki G, Hapangama DK. Telomerase and Telomeres in Endometrial Cancer. Front Oncol 2019; 9:344. [PMID: 31157162 PMCID: PMC6533802 DOI: 10.3389/fonc.2019.00344] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 04/15/2019] [Indexed: 12/11/2022] Open
Abstract
Telomeres at the termini of human chromosomes are shortened with each round of cell division due to the “end replication problem” as well as oxidative stress. During carcinogenesis, cells acquire or retain mechanisms to maintain telomeres to avoid initiation of cellular senescence or apoptosis and halting cell division by critically short telomeres. The unique reverse transcriptase enzyme complex, telomerase, catalyzes the maintenance of telomeres but most human somatic cells do not have sufficient telomerase activity to prevent telomere shortening. Tissues with high and prolonged replicative potential demonstrate adequate cellular telomerase activity to prevent telomere erosion, and high telomerase activity appears to be a critical feature of most (80–90%) epithelial cancers, including endometrial cancer. Endometrial cancers regress in response to progesterone which is frequently used to treat advanced endometrial cancer. Endometrial telomerase is inhibited by progestogens and deciphering telomere and telomerase biology in endometrial cancer is therefore important, as targeting telomerase (a downstream target of progestogens) in endometrial cancer may provide novel and more effective therapeutic avenues. This review aims to examine the available evidence for the role and importance of telomere and telomerase biology in endometrial cancer.
Collapse
Affiliation(s)
- Rafah A A Alnafakh
- Liverpool Women's Hospital NHS Foundation Trust, Liverpool, United Kingdom.,Department of Women's and Children's Health, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Meera Adishesh
- Liverpool Women's Hospital NHS Foundation Trust, Liverpool, United Kingdom.,Department of Women's and Children's Health, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Lucy Button
- Liverpool Women's Hospital NHS Foundation Trust, Liverpool, United Kingdom.,Department of Women's and Children's Health, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Gabriele Saretzki
- The Ageing Biology Centre and Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Dharani K Hapangama
- Liverpool Women's Hospital NHS Foundation Trust, Liverpool, United Kingdom.,Department of Women's and Children's Health, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
40
|
Telomere DNA G-quadruplex folding within actively extending human telomerase. Proc Natl Acad Sci U S A 2019; 116:9350-9359. [PMID: 31019071 DOI: 10.1073/pnas.1814777116] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Telomerase reverse transcribes short guanine (G)-rich DNA repeat sequences from its internal RNA template to maintain telomere length. G-rich telomere DNA repeats readily fold into G-quadruplex (GQ) structures in vitro, and the presence of GQ-prone sequences throughout the genome introduces challenges to replication in vivo. Using a combination of ensemble and single-molecule telomerase assays, we discovered that GQ folding of the nascent DNA product during processive addition of multiple telomere repeats modulates the kinetics of telomerase catalysis and dissociation. Telomerase reactions performed with telomere DNA primers of varying sequence or using GQ-stabilizing K+ versus GQ-destabilizing Li+ salts yielded changes in DNA product profiles consistent with formation of GQ structures within the telomerase-DNA complex. Addition of the telomerase processivity factor POT1-TPP1 altered the DNA product profile, but was not sufficient to recover full activity in the presence of Li+ cations. This result suggests GQ folding synergizes with POT1-TPP1 to support telomerase function. Single-molecule Förster resonance energy transfer experiments reveal complex DNA structural dynamics during real-time catalysis in the presence of K+ but not Li+, supporting the notion of nascent product folding within the active telomerase complex. To explain the observed distributions of telomere products, we globally fit telomerase time-series data to a kinetic model that converges to a set of rate constants describing each successive telomere repeat addition cycle. Our results highlight the potential influence of the intrinsic folding properties of telomere DNA during telomerase catalysis, and provide a detailed characterization of GQ modulation of polymerase function.
Collapse
|
41
|
Abstract
PURPOSE OF REVIEW Genomic mutations in telomere-related genes have been recognized as a cause of familial forms of idiopathic pulmonary fibrosis (IPF). However, it has become increasingly clear that telomere syndromes and telomere shortening are associated with various types of pulmonary disease. Additionally, it was found that also single nucleotide polymorphisms (SNPs) in telomere-related genes are risk factors for the development of pulmonary disease. This review focuses on recent updates on pulmonary phenotypes associated with genetic variation in telomere-related genes. RECENT FINDINGS Genomic mutations in seven telomere-related genes cause pulmonary disease. Pulmonary phenotypes associated with these mutations range from many forms of pulmonary fibrosis to emphysema and pulmonary vascular disease. Telomere-related mutations account for up to 10% of sporadic IPF, 25% of familial IPF, 10% of connective-tissue disease-associated interstitial lung disease, and 1% of COPD. Mixed disease forms have also been found. Furthermore, SNPs in TERT, TERC, OBFC1, and RTEL1, as well as short telomere length, have been associated with several pulmonary diseases. Treatment of pulmonary disease caused by telomere-related gene variation is currently based on disease diagnosis and not on the underlying cause. SUMMARY Pulmonary phenotypes found in carriers of telomere-related gene mutations and SNPs are primarily pulmonary fibrosis, sometimes emphysema and rarely pulmonary vascular disease. Genotype-phenotype relations are weak, suggesting that environmental factors and genetic background of patients determine disease phenotypes to a large degree. A disease model is presented wherever genomic variation in telomere-related genes cause specific pulmonary disease phenotypes whenever triggered by environmental exposure, comorbidity, or unknown factors.
Collapse
|
42
|
|
43
|
Planas-Cerezales L, Arias-Salgado EG, Buendia-Roldán I, Montes-Worboys A, López CE, Vicens-Zygmunt V, Hernaiz PL, Sanuy RL, Leiro-Fernandez V, Vilarnau EB, Llinás ES, Sargatal JD, Abellón RP, Selman M, Molina-Molina M. Predictive factors and prognostic effect of telomere shortening in pulmonary fibrosis. Respirology 2018; 24:146-153. [PMID: 30320420 DOI: 10.1111/resp.13423] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Revised: 09/11/2018] [Accepted: 09/12/2018] [Indexed: 01/04/2023]
Abstract
BACKGROUND AND OBJECTIVE The abnormal shortening of telomeres is a mechanism linking ageing to idiopathic pulmonary fibrosis (IPF) that could be useful in the clinical setting. The objective of this study was to identify the IPF patients with higher risk for telomere shortening and to investigate the outcome implications. METHODS Consecutive Spanish patients were included at diagnosis and followed up for 3 years. DNA blood samples from a Mexican cohort were used to validate the results found in Spanish sporadic IPF. Prior to treatment, telomere length was measured through quantitative polymerase chain reaction (qPCR) and Southern blot. Outcome was assessed according to mortality or need for lung transplantation. A multivariate regression logistic model was used for statistical analysis. RESULTS Family aggregation, age of <60 years and the presence of non-specific immunological or haematological abnormalities were associated with a higher probability of telomere shortening. Overall, 66.6% of patients younger than 60 years with telomere shortening died or required lung transplantation, independent of functional impairment at diagnosis. By contrast, in patients older than 60 years with telomere shortening, the negative impact of telomere shortening in outcome was not significant. CONCLUSION Our data indicate that young sporadic IPF patients (<60 years) with some non-specific immunological or haematological abnormalities had higher risk of telomere shortening, and furthermore, they presented a poorer prognosis.
Collapse
Affiliation(s)
- Lurdes Planas-Cerezales
- Centro de Investigación en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain.,Unidad Funcional de Intersticio Pulmonar, Servicio Neumología, Hospital Universitario de Bellvitge, Instituto de Investigación Biomédica de Bellvitge (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
| | - Elena G Arias-Salgado
- Advanced Medical Projects, Madrid, Spain.,Instituto de Investigaciones Biomédicas, Consejo Superior de Investigaciones Científicas (CSIC)/Universidad Autónoma de Madrid (UAM), Centro de Investigación en Red de Enfermedades Raras (CIBERER), Madrid, Spain
| | - Ivette Buendia-Roldán
- Instituto Nacional de Enfermedades Respiratorias, "Ismael Cosío Villegas", México City, Mexico
| | - Ana Montes-Worboys
- Centro de Investigación en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain.,Unidad Funcional de Intersticio Pulmonar, Servicio Neumología, Hospital Universitario de Bellvitge, Instituto de Investigación Biomédica de Bellvitge (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
| | - Cristina Esquinas López
- Centro de Investigación en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain.,Servicio de Neumología, Hospital Vall d'Hebron, Barcelona, Spain
| | - Vanesa Vicens-Zygmunt
- Unidad Funcional de Intersticio Pulmonar, Servicio Neumología, Hospital Universitario de Bellvitge, Instituto de Investigación Biomédica de Bellvitge (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
| | - Patricio Luburich Hernaiz
- Unidad Funcional de Intersticio Pulmonar. Servicio Radiodiagnóstico, Hospital Universitario de Bellvitge, Instituto de Investigación Biomédica de Bellvitge (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
| | - Roger Llatjós Sanuy
- Unidad Funcional de Intersticio Pulmonar, Servicio de Anatomía Patológica, Hospital Universitario de Bellvitge, Instituto de Investigación Biomédica de Bellvitge (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
| | - Virginia Leiro-Fernandez
- Servicio de Neumología, Complexo Hospitalario Universitario de Vigo (CHUVI), Vigo, Spain.,Grupo de Investigación en Respiratorio, Instituto de Investigación Biomédica de Vigo, Vigo, Spain
| | - Eva Balcells Vilarnau
- Centro de Investigación en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain.,Servicio de Neumología, Hospital del Mar, Instituto Hospital del Mar de Investigaciones Médicas (IMIM), Universidad Pompeu Fabra (UPF), Barcelona, Spain
| | - Ernest Sala Llinás
- Centro de Investigación en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain.,Servicio de Neumología, Hospital Son Espases, Instituto de Investigación Sanitaria Islas Baleares (IdISBa), Palma de Mallorca, Spain
| | - Jordi Dorca Sargatal
- Centro de Investigación en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain.,Unidad Funcional de Intersticio Pulmonar, Servicio Neumología, Hospital Universitario de Bellvitge, Instituto de Investigación Biomédica de Bellvitge (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
| | - Rosario Perona Abellón
- Instituto de Investigaciones Biomédicas, Consejo Superior de Investigaciones Científicas (CSIC)/Universidad Autónoma de Madrid (UAM), Centro de Investigación en Red de Enfermedades Raras (CIBERER), Madrid, Spain
| | - Moisés Selman
- Instituto Nacional de Enfermedades Respiratorias, "Ismael Cosío Villegas", México City, Mexico
| | - Maria Molina-Molina
- Centro de Investigación en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain.,Unidad Funcional de Intersticio Pulmonar, Servicio Neumología, Hospital Universitario de Bellvitge, Instituto de Investigación Biomédica de Bellvitge (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
| |
Collapse
|
44
|
Deshpande AP, Collins K. Mechanisms of template handling and pseudoknot folding in human telomerase and their manipulation to expand the sequence repertoire of processive repeat synthesis. Nucleic Acids Res 2018; 46:7886-7901. [PMID: 29986069 PMCID: PMC6125678 DOI: 10.1093/nar/gky601] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Accepted: 06/26/2018] [Indexed: 01/12/2023] Open
Abstract
Telomerase adds telomeric repeats to chromosome ends by processive copying of a template within the telomerase RNA bound to telomerase reverse transcriptase. Telomerase RNAs have single-stranded regions that separate the template from a 5' stem and 3' pseudoknot, and mammals gained additional stem P2a.1 separating the template from the pseudoknot. Using human telomerase, we show that the length of template 3'-flanking single-stranded RNA is a determinant of repeat addition processivity whereas template 5'-flanking single-stranded RNA and P2a.1 are critical for activity but not processivity. In comparison, requirements for the template sequence itself are confounding: different substitutions of the same position have strikingly different consequences, from improved processivity and activity to complete inactivation. We discovered that some altered-template sequences stabilize an alternative RNA conformation that precludes the pseudoknot by base-pairing of one pseudoknot strand to the template 3' end. Using mutations to reduce over-stability of the alternative conformation, we restore high activity and processivity to otherwise inactive altered-template telomerase ribonucleoproteins. In cells, over-stabilization or destabilization of the alternative state severely inhibited biogenesis of active telomerase. Our findings delineate roles for human telomerase RNA template-flanking regions, establish a biologically relevant pseudoknot-alternative RNA conformation, and expand the repertoire of human telomerase repeat synthesis.
Collapse
Affiliation(s)
- Aishwarya P Deshpande
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720, USA
| | - Kathleen Collins
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720, USA
| |
Collapse
|
45
|
Mangaonkar AA, Patnaik MM. Short Telomere Syndromes in Clinical Practice: Bridging Bench and Bedside. Mayo Clin Proc 2018; 93:904-916. [PMID: 29804726 PMCID: PMC6035054 DOI: 10.1016/j.mayocp.2018.03.020] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 03/19/2018] [Indexed: 10/16/2022]
Abstract
Short telomere syndromes (STSs) are accelerated aging syndromes often caused by inheritable gene mutations resulting in decreased telomere lengths. Consequently, organ systems with increased cell turnover, such as the skin, bone marrow, lungs, and gastrointestinal tract, are commonly affected. Owing to diverse clinical presentations, STSs pose a diagnostic challenge, with bone marrow failure and idiopathic pulmonary fibrosis being frequent manifestations, occurring in association with gene mutations involving DKC1 (for expansion of gene symbols, use search tool at www.genenames.org), TERT, TERC, and others. Inherited STSs demonstrate genetic anticipation, occurring at an earlier age with more severe manifestations in the affected progeny. Telomere lengths can be assessed in peripheral blood granulocytes and lymphocytes using a sensitive technique called flow cytometry-fluorescence in situ hybridization, and mutational analysis can be performed using next-generation sequencing assays. In approximately 40% of patients with shortened telomere lengths, gene mutations cannot be identified due to the fact that all STS-associated genes have not yet been defined or due to alternative mechanisms of telomere shortening. Danazol, an anabolic steroid, has been associated with hematologic responses in patients with STSs and associated bone marrow failure; however, its reported ability to increase telomerase activity and reduce telomere attrition needs further elucidation. Organ transplant is reserved for patients with end-organ failure and is associated with substantial morbidity and mortality. Herein, we summarize the clinical and laboratory characteristics of STSs and offer a stepwise approach to diagnose and manage complications in affected patients.
Collapse
Affiliation(s)
| | - Mrinal M Patnaik
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN.
| |
Collapse
|
46
|
Affiliation(s)
- Mary Armanios
- Department of Oncology, McKusick-Nathans Institute of Genetic Medicine, Telomere Center, Johns Hopkins University School of Medicine, Baltimore, MD.
| |
Collapse
|
47
|
Analysis of protein-altering variants in telomerase genes and their association with MUC5B common variant status in patients with idiopathic pulmonary fibrosis: a candidate gene sequencing study. THE LANCET RESPIRATORY MEDICINE 2018; 6:603-614. [PMID: 29891356 DOI: 10.1016/s2213-2600(18)30135-8] [Citation(s) in RCA: 129] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 03/14/2018] [Accepted: 03/16/2018] [Indexed: 12/22/2022]
Abstract
BACKGROUND Idiopathic pulmonary fibrosis (IPF) risk has a strong genetic component. Studies have implicated variations at several loci, including TERT, surfactant genes, and a single nucleotide polymorphism at chr11p15 (rs35705950) in the intergenic region between TOLLIP and MUC5B. Patients with IPF who have risk alleles at rs35705950 have longer survival from the time of IPF diagnosis than do patients homozygous for the non-risk allele, whereas patients with shorter telomeres have shorter survival times. We aimed to assess whether rare protein-altering variants in genes regulating telomere length are enriched in patients with IPF homozygous for the non-risk alleles at rs35705950. METHODS Between Nov 1, 2014, and Nov 1, 2016, we assessed blood samples from patients aged 40 years or older and of European ancestry with sporadic IPF from three international phase 3 clinical trials (INSPIRE, CAPACITY, ASCEND), one phase 2 study (RIFF), and US-based observational studies (Vanderbilt Clinical Interstitial Lung Disease Registry and the UCSF Interstitial Lung Disease Clinic registry cohorts) at the Broad Institute (Cambridge, MA, USA) and Human Longevity (San Diego, CA, USA). We also assessed blood samples from non-IPF controls in several clinical trials. We did whole-genome sequencing to assess telomere length and identify rare protein-altering variants, stratified by rs35705950 genotype. We also assessed rare functional variation in TERT exons and compared telomere length and disease progression across genotypes. FINDINGS We assessed samples from 1510 patients with IPF and 1874 non-IPF controls. 30 (3%) of 1046 patients with an rs35705950 risk allele had a rare protein-altering variant in TERT compared with 34 (7%) of 464 non-risk allele carriers (odds ratio 0·40 [95% CI 0·24-0·66], p=0·00039). Subsequent analyses identified enrichment of rare protein-altering variants in PARN and RTEL1, and rare variation in TERC in patients with IPF compared with controls. We expanded our study population to provide a more accurate estimation of rare variant frequency in these four loci, and to calculate telomere length. The proportion of patients with at least one rare variant in TERT, PARN, TERC, or RTEL1 was higher in patients with IPF than in controls (149 [9%] of 1739 patients vs 205 [2%] of 8645 controls, p=2·44 × 10-8). Patients with IPF who had a variant in any of the four identified telomerase component genes had telomeres that were 3·69-16·10% shorter than patients without a variant in any of the four genes and had an earlier mean age of disease onset than patients without one or more variants (65·1 years [SD 7·8] vs 67·1 years [7·9], p=0·004). In the placebo arms of clinical trials, shorter telomeres were significantly associated with faster disease progression (1·7% predicted forced vital capacity per kb per year, p=0·002). Pirfenidone had treatment benefit regardless of telomere length (p=4·24 × 10-8 for telomere length lower than the median, p=0·0044 for telomere length greater than the median). INTERPRETATION Rare protein-altering variants in TERT, PARN, TERC, and RTEL1 are enriched in patients with IPF compared with controls, and, in the case of TERT, particularly in individuals without a risk allele at the rs35705950 locus. This suggests that multiple genetic factors contribute to sporadic IPF, which might implicate distinct mechanisms of pathogenesis and disease progression. FUNDING Genentech, National Institutes of Health, Francis Family Foundation, Pulmonary Fibrosis Foundation, Nina Ireland Program for Lung Health, US Department of Veterans Affairs.
Collapse
|
48
|
Merck SJ, Armanios M. Shall we call them "telomere-mediated"? Renaming the idiopathic after the cause is found. Eur Respir J 2018; 48:1556-1558. [PMID: 27903687 DOI: 10.1183/13993003.02115-2016] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 10/28/2016] [Indexed: 01/10/2023]
Affiliation(s)
- Samantha J Merck
- Dept of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mary Armanios
- Dept of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA .,McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
49
|
|
50
|
Abstract
Telomere length (TL) predicts the onset of cellular senescence in vitro but the diagnostic utility of TL measurement in clinical settings is not fully known. We tested the value of TL measurement by flow cytometry and FISH (flowFISH) in patients with mutations in telomerase and telomere maintenance genes. TL had a discrete and reproducible normal range with definable upper and lower boundaries. While TL above the 50th age-adjusted percentile had a 100% negative predictive value for clinically relevant mutations, the lower threshold in mutation carriers was age-dependent, and adult mutation carriers often overlapped with the lowest decile of controls. The extent of telomere shortening correlated with the age at diagnosis as well as the short telomere syndrome phenotype. Extremely short TL caused bone marrow failure and immunodeficiency in children and young adults, while milder defects manifested as pulmonary fibrosis-emphysema in adults. We prospectively examined whether TL altered treatment decisions for newly diagnosed idiopathic bone marrow failure patients and found abnormally short TL enriched for patients with mutations in some inherited bone marrow failure genes, such as RUNX1, in addition to telomerase and telomere maintenance genes. The result was actionable, altering the choice of treatment regimen and/or hematopoietic stem cell donor in one-fourth of the cases (9 of 38, 24%). We conclude that TL measurement by flowFISH, when used for targeted clinical indications and in limited settings, can influence treatment decisions in ways that improve outcome.
Collapse
|