1
|
Cruz NFSDA, Hudson JL, Sengillo JD, Shah SM, Lopez-Font F, Negron CI, Farah ME, Berrocal AM. Underlying Disease in Atypical Retinopathy of Prematurity. Am J Ophthalmol 2025; 274:67-75. [PMID: 40010420 DOI: 10.1016/j.ajo.2025.02.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 01/18/2025] [Accepted: 02/18/2025] [Indexed: 02/28/2025]
Abstract
BACKGROUND AND OBJECTIVE Retinopathy of prematurity (ROP), familial exudative vitreoretinopathy (FEVR), and telomere biology disorders (TBD) are classified as distinct diseases. However, emerging genetic research and evidence on multimodal imaging suggest a spectrum along which ROP may overlap with FEVR or TBD. DESIGN Retrospective case series. METHODS This was an institutional review board-approved, retrospective study. A literature review was performed, and medical records of all patients with phenotypic ROP evaluated by the pediatric retina service at Bascom Palmer Eye Institute from March 1, 2019 to July 30, 2023 were analyzed. RESULTS Eighteen patients with phenotypic and genetically confirmed FEVR or TBD were identified. Of these, the initial diagnosis was ROP with preterm gestational age (n = 11, 57.9%) or ROP at moderate to late preterm gestational age (n = 8, 42.1%). Final diagnosis for 15 patients (78.9%) was FEVR, and final diagnosis for 4 patients (21.1%) was TBD. The most common genetic variants in the FEVR group were identified in the genes LRP5 (n = 5, 33.3%) and FZD4 (n = 3, 20%), and in the TBD group, CTC1 (n = 3; 75%). The mean age at diagnosis was 5.7 years old (range 0.3-36.7 years). CONCLUSIONS The authors reinforce the classification of ROPER (ROP and FEVR) and introduce the term, ROPMERE (ROP and TBD), to classify these patients in a way that reflects their clinical presentation and underlying genetic diagnosis. Identification of this subset of patients will allow for sustained surveillance of infants with these diseases.
Collapse
Affiliation(s)
- Natasha F S DA Cruz
- Department of Ophthalmology (N.D.C., J.H., J.S., S.S., F.L.F., C.N., A.B.), Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, United States; Department of Ophthalmology (N.D.C., M.F.), Federal University of São Paulo, Escola Paulista de Medicina, São Paulo, Brazil; Department of Ophthalmology, Centro Ocular (N.D.C.), Belém, Brazil
| | - Julia L Hudson
- Department of Ophthalmology (N.D.C., J.H., J.S., S.S., F.L.F., C.N., A.B.), Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, United States
| | - Jesse D Sengillo
- Department of Ophthalmology (N.D.C., J.H., J.S., S.S., F.L.F., C.N., A.B.), Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, United States
| | - Serena M Shah
- Department of Ophthalmology (N.D.C., J.H., J.S., S.S., F.L.F., C.N., A.B.), Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, United States
| | - Francisco Lopez-Font
- Department of Ophthalmology (N.D.C., J.H., J.S., S.S., F.L.F., C.N., A.B.), Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, United States
| | - Catherin I Negron
- Department of Ophthalmology (N.D.C., J.H., J.S., S.S., F.L.F., C.N., A.B.), Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, United States
| | - Michel E Farah
- Department of Ophthalmology (N.D.C., M.F.), Federal University of São Paulo, Escola Paulista de Medicina, São Paulo, Brazil
| | - Audina M Berrocal
- Department of Ophthalmology (N.D.C., J.H., J.S., S.S., F.L.F., C.N., A.B.), Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, United States.
| |
Collapse
|
2
|
Ghoraba HH, Sears J, Traboulsi EI. Hereditary Vitreoretinopathies: Molecular Diagnosis, Clinical Presentation and Management. Clin Exp Ophthalmol 2025; 53:281-291. [PMID: 39837650 PMCID: PMC11962705 DOI: 10.1111/ceo.14494] [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: 10/21/2024] [Revised: 12/20/2024] [Accepted: 12/26/2024] [Indexed: 01/23/2025]
Abstract
Hereditary vitreoretinopathies (HVRs), also known as hereditary vitreoretinal degenerations comprise a heterogeneous group of inherited disorders of the retina and vitreous, collectively and variably characterised by vitreal abnormalities, such as fibrillary condensations, liquefaction or membranes, as well as peripheral retinal abnormalities, vascular changes in some, an increased risk of retinal detachment and early-onset cataract formation. The pathology often involves the vitreoretinal interface in some, while the major underlying abnormality is vascular in others. Recent advances in molecular diagnosis and identification of the responsible genes and have improved our understanding of the pathogenesis, risks and management of the HVRs. Clinically, HVRs can be classified according to the presence or absence of skeletal or other systemic abnormalities, retinal dysfunction or retinal vascular abnormalities [2]. There are some discrepancies in the literature regarding which diseases are included under the overarching term 'hereditary vitreoretinopathies'. Conditions such as Stickler syndrome, Wagner syndrome and familial exudative vitreoretinopathy are generally included, while others such as autosomal dominant neovascular inflammatory vitreoretinopathy (ADNIV) and autosomal dominant vitreoretinochoroidapathy (ADVIRC) may not. In this review, we will discuss some historical aspects, the molecular pathogenesis, clinical features and management of diseases and syndromes commonly considered as HVRs.
Collapse
|
3
|
DA Cruz NFS, Sengillo JD, Negron CI, Berrocal AM. Telomere Biology Disorders: Microvascular Abnormalities on Optical Coherence Tomography. Am J Ophthalmol 2025; 275:52-57. [PMID: 40139649 DOI: 10.1016/j.ajo.2025.03.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 03/15/2025] [Accepted: 03/17/2025] [Indexed: 03/29/2025]
Abstract
BACKGROUND AND OBJECTIVE Telomere biology disorders (TBDs) are inherited conditions caused by telomere dysfunction, impacting systemic and ocular health. We aim to explore the role of optical coherence tomography angiography (OCTA) in identifying retinal microvascular abnormalities in TBDs. DESIGN Retrospective case series. METHODS The electronic medical records of pediatric patients with TBD at a tertiary referral eye center were reviewed from January 2019 to July 2023. OCTA images were reviewed for anomalies of the retinal vasculature. RESULTS In total, 13 eyes of 7 patients were included in the study. All patients were genetically confirmed to have TBD. The most common genetic variants were CTC1 (5 patients; 71.4%), ACD (1 patient; 14.3%) and RTEL1 (1 patient; 14.3%). On OCTA, all 13 eyes showed some degree of macular microvascular abnormality in both the SVC and DVC. The most common microvascular abnormality seen in the SVC was blood vessels anastomosis (11; 84.6%), and in the DVC was decreased vessel density (9; 69.2%). CONCLUSIONS OCTA imaging reveals a high prevalence of microvascular abnormalities in patients with TBD, highlighting its potential role in assessing retinal vascular changes associated with the disease.
Collapse
Affiliation(s)
- Natasha F S DA Cruz
- From the Department of Ophthalmology (N.D.C., J.S., C.N., A.B.), Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, USA.; Department of Ophthalmology, Centro Ocular, Belém, Brazil
| | - Jesse D Sengillo
- From the Department of Ophthalmology (N.D.C., J.S., C.N., A.B.), Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Catherin I Negron
- From the Department of Ophthalmology (N.D.C., J.S., C.N., A.B.), Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Audina M Berrocal
- From the Department of Ophthalmology (N.D.C., J.S., C.N., A.B.), Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, USA..
| |
Collapse
|
4
|
da Cruz NFS, Sengillo JD, Shah SM, López-Font FJ, Negron CI, Berrocal AM. Telomere Biology Disorders: Clinical and Angiographic Findings. Ophthalmol Retina 2025; 9:272-277. [PMID: 39332705 DOI: 10.1016/j.oret.2024.09.011] [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: 05/22/2024] [Revised: 09/17/2024] [Accepted: 09/19/2024] [Indexed: 09/29/2024]
Abstract
PURPOSE To evaluate the retinal vasculature in pediatric patients with telomere biology disorders (TBDs). DESIGN Retrospective consecutive case series. SUBJECTS Pediatric patients with a diagnosis of TBD who underwent widefield fluorescein angiography (FA). METHODS Electronic medical records of pediatric patients with TBD at a tertiary referral eye center were reviewed from January 2019 to July 2023. Vascular phenotype was assessed by reviewing FA images. MAIN OUTCOME MEASURES Incomplete peripheral vascularization, aneurysmal dilatation, terminal arborization, anastomotic loops, capillary dropout, neovascularization, tortuosity, leakage from tractional membranes, and blockage from hemorrhage. RESULTS Fourteen eyes from 7 patients were included. All patients were genetically confirmed for TBD. The most common genetic variants were in CTC1 (5 patients; 71.4%), ACD (1 patient; 14.3%), and RTEL1 (1 patient; 14.3%). On FA, the most common findings were incomplete peripheral vascularization (14 eyes, 100%), aneurysmal dilatation (12 eyes, 85.7%), terminal arborization (12 eyes, 85.7%), anastomotic loops (12 eyes, 85.7%), capillary dropout (10 eyes, 71.4%), and neovascularization (9 eyes, 64.3%). Regarding treatment, laser photocoagulation (14 eyes, 100%), intravitreal bevacizumab injection (13 eyes, 92.6%), and subtenon's Kenalog (11 eyes, 78.6%) were utilized. All patients managed with laser photocoagulation and bevacizumab required multiple treatments. CONCLUSIONS Our study describes a spectrum of vascular changes evidenced by widefield FA in pediatric patients with genetically confirmed TBD. Although further research is warranted to fully understand the etiology of these subtle vascular anomalies, widefield FA should be conducted in patients with genetically confirmed or suspected TBD. FINANCIAL DISCLOSURE(S) Proprietary or commercial disclosure may be found in the Footnotes and Disclosures at the end of this article.
Collapse
Affiliation(s)
- Natasha F S da Cruz
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Jesse D Sengillo
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Serena M Shah
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Francisco J López-Font
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Catherin I Negron
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Audina M Berrocal
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida.
| |
Collapse
|
5
|
Guerrero-López R, Manguán-García C, Carrascoso-Rubio C, Lozano ML, Toldos-Torres M, García-Castro L, Sánchez-Dominguez R, Alberquilla O, Sánchez-Pérez I, Molina-Molina M, Bueren JA, Guenechea G, Perona R, Sastre L. Premature ageing of lung alveoli and bone marrow cells from Terc deficient mice with different telomere lengths. Sci Rep 2025; 15:6102. [PMID: 39971959 PMCID: PMC11840044 DOI: 10.1038/s41598-025-90246-2] [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: 11/05/2024] [Accepted: 02/11/2025] [Indexed: 02/21/2025] Open
Abstract
Telomeres are terminal protective chromosome structures. Genetic variants in genes coding for proteins required for telomere maintenance cause rare, life-threatening Telomere Biology Disorders (TBDs) such as dyskeratosis congenita, aplastic anemia or pulmonary fibrosis. The more frequently used mice strains have telomeres much longer than the human ones which question their use as in vivo models for TBDs. One mice model with shorter telomeres based on the CAST/EiJ mouse strain carrying a mutation in the Terc gene, coding for the telomerase RNA component, has been studied in comparison with C57BL/6J mice, carrying the same mutation and long telomeres. The possible alterations produced in lungs and the haematopoietic system, frequently affected in TBD patients, were determined at different ages of the mice. Homozygous mutant mice presented a very shortened life span, more notorious in the short-telomeres CAST/EiJ strain. The lungs of mutant mice presented a transitory increase in fibrosis and a significant decrease in the relative amount of the alveolar epithelial type 2 cells from six months of age. This decrease was larger in mutant homozygous animals but was also observed in heterozygous animals. On the contrary the expression of the senescence-related protein P21 increased from six months of age in mutant mice of both strains. The analysis of the haematopoietic system indicated a decrease in the number of megakaryocyte-erythroid progenitors in homozygous mutants and an increase in the clonogenic potential of bone marrow and LSK cells. Bone marrow cells from homozygous mutant animals presented decreasing in vitro expansion capacity. The alterations observed are compatible with precocious ageing of lung alveolar cells and the bone marrow cells that correlate with the alterations observed in TBD patients. The alterations seem to be more related to the genotype of the animals that to the basal telomere length of the strains although they are more pronounced in the short-telomere CAST/EiJ-derived strain than in C57BL/6J animals. Therefore, both animal models, at ages over 6-8 months, could represent valuable and convenient models for the study of TBDs and for the assay of new therapeutic products.
Collapse
Affiliation(s)
- Rosa Guerrero-López
- Instituto de Investigaciones Biomedicas Sols/Morreale, CSIC-UAM. Arturo Duperier, Madrid, 28029, Spain.
- Centro de Investigacion Biomedica en Red de Enfermedades Raras (CIBERER), Madrid, 28029, Spain.
| | - Cristina Manguán-García
- Instituto de Investigaciones Biomedicas Sols/Morreale, CSIC-UAM. Arturo Duperier, Madrid, 28029, Spain
- Centro de Investigacion Biomedica en Red de Enfermedades Raras (CIBERER), Madrid, 28029, Spain
| | - Carlos Carrascoso-Rubio
- Hematopoietic Innovative Therapies Division, Centro de Investigaciones Energeticas, Medioambientales y Tecnologicas (CIEMAT)) and Advanced Therapies Unit, Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD, UAM), Madrid, Spain
| | - M Luz Lozano
- Hematopoietic Innovative Therapies Division, Centro de Investigaciones Energeticas, Medioambientales y Tecnologicas (CIEMAT)) and Advanced Therapies Unit, Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD, UAM), Madrid, Spain
| | - Marta Toldos-Torres
- Instituto de Investigaciones Biomedicas Sols/Morreale, CSIC-UAM. Arturo Duperier, Madrid, 28029, Spain
| | - Laura García-Castro
- Instituto de Investigaciones Biomedicas Sols/Morreale, CSIC-UAM. Arturo Duperier, Madrid, 28029, Spain
| | - Rebeca Sánchez-Dominguez
- Centro de Investigacion Biomedica en Red de Enfermedades Raras (CIBERER), Madrid, 28029, Spain
- Hematopoietic Innovative Therapies Division, Centro de Investigaciones Energeticas, Medioambientales y Tecnologicas (CIEMAT)) and Advanced Therapies Unit, Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD, UAM), Madrid, Spain
| | - Omaira Alberquilla
- Centro de Investigacion Biomedica en Red de Enfermedades Raras (CIBERER), Madrid, 28029, Spain
- Hematopoietic Innovative Therapies Division, Centro de Investigaciones Energeticas, Medioambientales y Tecnologicas (CIEMAT)) and Advanced Therapies Unit, Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD, UAM), Madrid, Spain
| | - Isabel Sánchez-Pérez
- Instituto de Investigaciones Biomedicas Sols/Morreale, CSIC-UAM. Arturo Duperier, Madrid, 28029, Spain
- Centro de Investigacion Biomedica en Red de Enfermedades Raras (CIBERER), Madrid, 28029, Spain
| | - Maria Molina-Molina
- ILD Unit, Pneumatology Department, University Hospital of Bellvitge, IDIBELL. University of Barcelona, Barcelona, Spain
| | - Juan A Bueren
- Centro de Investigacion Biomedica en Red de Enfermedades Raras (CIBERER), Madrid, 28029, Spain
- Hematopoietic Innovative Therapies Division, Centro de Investigaciones Energeticas, Medioambientales y Tecnologicas (CIEMAT)) and Advanced Therapies Unit, Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD, UAM), Madrid, Spain
| | - Guillermo Guenechea
- Centro de Investigacion Biomedica en Red de Enfermedades Raras (CIBERER), Madrid, 28029, Spain.
- Hematopoietic Innovative Therapies Division, Centro de Investigaciones Energeticas, Medioambientales y Tecnologicas (CIEMAT)) and Advanced Therapies Unit, Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD, UAM), Madrid, Spain.
| | - Rosario Perona
- Centro de Investigacion Biomedica en Red de Enfermedades Raras (CIBERER), Madrid, 28029, Spain.
- Instituto de Salud Carlos III, Madrid, 28029, Spain.
| | - Leandro Sastre
- Instituto de Investigaciones Biomedicas Sols/Morreale, CSIC-UAM. Arturo Duperier, Madrid, 28029, Spain.
- Centro de Investigacion Biomedica en Red de Enfermedades Raras (CIBERER), Madrid, 28029, Spain.
| |
Collapse
|
6
|
Cazzola M, Malcovati L. Genome sequencing in the management of myelodysplastic syndromes and related disorders. Haematologica 2025; 110:312-329. [PMID: 39445412 PMCID: PMC11788631 DOI: 10.3324/haematol.2023.284947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Accepted: 07/18/2024] [Indexed: 10/25/2024] Open
Abstract
Myeloid neoplasms originate from the clonal proliferation of hematopoietic stem cells, which is driven by the acquisition of somatic genetic mutations. Within these disorders, myelodysplastic syndromes (MDS) are specifically characterized by morphological abnormalities (dysplasia) and impaired maturation of myeloid precursors (ineffective hematopoiesis), resulting in peripheral blood cytopenia. Several studies have advanced the field of MDS, with a few landmark papers leading to a paradigm shift, opening new avenues of research and enabling a molecular revolution. These seminal papers include the first description of the 5q- syndrome, the identification of somatic mutations of TET2 in myeloid neoplasms, the detection of common pathway mutations in the splicing machinery, and the discovery of clonal hematopoiesis. The somatic genomic landscape of MDS is now well defined. Genes that are recurrently mutated include epigenetic regulators, as well as genes of RNA splicing machinery, transcription regulation, DNA repair control, cohesin complex, and signal transduction. Furthermore, several disorders with a germline genetic predisposition to MDS have been identified, collectively accounting for up to 15% of all MDS cases. Genomic profiling can significantly improve the diagnostic approach to MDS, allowing the identification of distinct nosological entities such as SF3B1-mutant or TP53-mutant MDS. The Molecular International Prognostic Scoring System for MDS has already proven to be a valuable tool for individualized risk assessment and treatment decisions. In addition, the recently developed molecular taxonomy of MDS will likely facilitate the implementation of precision medicine approaches for these disorders. This will necessitate the establishment of specialized infrastructures within public health systems, involving close collaboration between healthcare institutions, academia, and the life-sciences industry.
Collapse
Affiliation(s)
- Mario Cazzola
- Department of Molecular Medicine, University of Pavia, and Department of Hematology Oncology, Fondazione IRCCS Policlinico San Matteo, Pavia.
| | - Luca Malcovati
- Department of Molecular Medicine, University of Pavia, and Department of Hematology Oncology, Fondazione IRCCS Policlinico San Matteo, Pavia
| |
Collapse
|
7
|
Ormerod MBEG, Ueland T, Aas M, Hjell G, Rødevand L, Sæther LS, Lunding SH, Johansen IT, Mlakar V, Andreou D, Ueland T, Lagerberg TV, Melle I, Djurovic S, Andreassen OA, Steen NE. Limited evidence of association between dysregulated immune marker levels and telomere length in severe mental disorders. Acta Neuropsychiatr 2025; 37:e4. [PMID: 39844366 DOI: 10.1017/neu.2024.62] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2025]
Abstract
OBJECTIVE Accelerated ageing indexed by telomere attrition is suggested in schizophrenia spectrum- (SCZ) and bipolar disorders (BD). While inflammation may promote telomere shortening, few studies have investigated the association between telomere length (TL) and markers of immune activation and inflammation in severe mental disorders. METHODS Leucocyte TL defined as telomere template/amount of single-copy gene template (T/S ratio), was determined in participants with SCZ (N = 301) or BD (N = 211) and a healthy control group (HC, N = 378). TL was analysed with linear regressions for associations with levels of 12 immune markers linked to SCZ or BD. Adjustments were made for a broad range of potential confounding variables. TL was measured by quantitative polymerase chain reaction (qPCR) and the immune markers were measured by enzyme immunoassays. RESULTS A positive association between levels of soluble tumour necrosis factor receptor 1A (sTNF-R1) and TL in SCZ (β = 0.191, p = 0.012) was observed. Plasma levels of the other immune markers were not significantly associated with TL in the BD, SCZ or HC groups. CONCLUSION There was limited evidence of association between immune markers and TL in SCZ and BD. The results provide little support for involvement of immune dysregulation, as reflected by current systemic markers, in telomere attrition-related accelerated ageing in severe mental disorders.
Collapse
Affiliation(s)
- Monica B E G Ormerod
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Thor Ueland
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Thrombosis Research Center (TREC), Division of Internal Medicine, University Hospital of North Norway, Tromsø, Norway
| | - Monica Aas
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, England, UK
- Department of Behavioural Sciences, OsloMet - Oslo Metropolitan University, Oslo, Norway
| | - Gabriela Hjell
- Department of Psychiatry, Ostfold Hospital, Graalum, Norway
| | - Linn Rødevand
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Linn Sofie Sæther
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- Department of Psychology, University of Oslo, Oslo, Norway
| | | | | | - Vid Mlakar
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Dimitrios Andreou
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet & Stockholm Health Care Services, Stockholm County Council, Stockholm, Sweden
- Division of Mental Health and Substance Abuse, Diakonhjemmet Hospital, Oslo, Norway
| | - Torill Ueland
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- Department of Psychology, University of Oslo, Oslo, Norway
| | - Trine V Lagerberg
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- Department of Psychology, University of Oslo, Oslo, Norway
| | - Ingrid Melle
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Srdjan Djurovic
- Department of Medical Genetics, Oslo University Hospital and University of Oslo, Oslo, Norway
- NORMENT, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Ole A Andreassen
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Nils Eiel Steen
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Division of Mental Health and Substance Abuse, Diakonhjemmet Hospital, Oslo, Norway
| |
Collapse
|
8
|
Savage SA, Bertuch AA. Different phenotypes with different endings-Telomere biology disorders and cancer predisposition with long telomeres. Br J Haematol 2025; 206:69-73. [PMID: 39462986 PMCID: PMC11739769 DOI: 10.1111/bjh.19851] [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: 08/21/2024] [Accepted: 10/11/2024] [Indexed: 10/29/2024]
Abstract
Rare germline pathogenic variants (GPVs) in genes essential in telomere length maintenance and function have been implicated in two broad classes of human disease. The telomere biology disorders (TBDs) are a spectrum of life-threatening conditions, including bone marrow failure, liver and lung disease, cancer and other complications caused by GPVs in telomere maintenance genes that result in short and/or dysfunctional telomeres and reduced cellular replicative capacity. In contrast, cancer predisposition with long telomeres (CPLT) is a disorder associated with elevated risk of a variety of cancers, primarily melanoma, thyroid cancer, sarcoma, glioma and lymphoproliferative neoplasms caused by GPVs in shelterin complex genes that lead to excessive telomere elongation and increased cellular replicative capacity. While telomeres are at the root of both disorders, the term TBD is used to convey the clinical phenotypes driven by critically short or otherwise dysfunctional telomeres and their biological consequences.
Collapse
Affiliation(s)
- Sharon A. Savage
- Clinical Genetics Branch, Division of Cancer Epidemiology and GeneticsNational Cancer InstituteBethesdaMarylandUSA
| | | |
Collapse
|
9
|
Duminy-Luppi D, Alcaide-Aldeano A, Planas-Cerezales L, Bermudo G, Vicens-Zygmunt V, Luburich P, Del Río-Carrero B, Llatjós R, Pijuan L, Escobar I, Rivas F, Montes-Worboys A, Gutiérrez-Rodríguez Y, Rodríguez-Plaza D, Padró-Miquel A, Esteve-Garcia A, Fernández-Varas B, Flores C, Fuentes M, Dorca J, Santos S, Perona R, Günther A, Shull J, Molina-Molina M. Diagnostic and prognostic implications of family history of fibrotic interstitial lung diseases. Respir Res 2024; 25:433. [PMID: 39695595 DOI: 10.1186/s12931-024-03063-y] [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: 06/09/2024] [Accepted: 12/04/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Patients with familial fibrotic interstitial lung disease (ILD) experience worse survival than patients with sporadic disease. Current guidelines do not consider family aggregation or genetic information in the diagnostic algorithm for idiopathic pulmonary fibrosis or other fibrotic ILDs. Better characterizing familial cases could help in diagnostic and treatment decision-making. METHODS This retrospective cohort study included 222 patients with fibrotic ILD (104 familial and 118 sporadic) from Bellvitge University Hospital. Clinical, radiological, pulmonary functional tests (PFT), and histological evaluations were performed at diagnosis and follow-up. Telomere shortening and disease-associated variants (DAVs) in telomerase-related genes were analysed in familial patients and sporadic patients with telomeric clinical signs. Primary outcomes were the presence of a UIP histological pattern and disease progression. RESULTS Patients with idiopathic pulmonary fibrosis (IPF) (52%), fibrotic hypersensitivity pneumonitis (23%), and other fibrotic ILDs (25%) were included. 42% of patients underwent lung biopsy. Patients with family aggregation were younger and less frequently associated comorbidities, male sex, and smoking history. However, usual interstitial pneumonia (UIP) was more frequent on pathology (p = 0.005; OR 3.37), especially in patients with indeterminate or non-UIP radiological patterns. Despite similar PFT results at diagnosis, familial patients were more likely to present with progressive disease (p = 0.001; OR 3.75). Carrying a DAV increased the risk of fibrotic progression in familial and sporadic patients (p = 0.029, OR 5.01). DISCUSSION Familial patients diagnosed with different fibrotic ILDs were more likely to exhibit a histological UIP pattern and disease progression than sporadic patients, independent of radiological findings and pulmonary function at diagnosis. CONCLUSION Considering the diagnostic likelihood of the histological UIP pattern and disease outcome, the presence of family aggregation would be useful in the decision making of multidisciplinary committees.
Collapse
Affiliation(s)
- D Duminy-Luppi
- University of Barcelona, School of Medicine. Respiratory Department, Bellvitge University Hospital, L'Hospitalet de Llobregat, Spain.
- Internal Medicine Department, ICMID, Hospital Clínic de Barcelona, Barcelona, Spain.
| | - A Alcaide-Aldeano
- University of Barcelona, School of Medicine. Respiratory Department, Bellvitge University Hospital, L'Hospitalet de Llobregat, Spain
- Cardiology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - L Planas-Cerezales
- Respiratory Department, Hospital of Viladecans, IDIBELL, University of Barcelona, Viladecans, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - G Bermudo
- Interstitial Lung Disease Unit, Respiratory Department, Bellvitge University Hospital, University of Barcelona, L'Hospitalet de Llobregat, Spain
- Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - V Vicens-Zygmunt
- Interstitial Lung Disease Unit, Respiratory Department, Bellvitge University Hospital, University of Barcelona, L'Hospitalet de Llobregat, Spain
- Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - P Luburich
- Radiology Department, Bellvitge University Hospital, IDIBELL, University of Barcelona, L'Hospitalet de Llobregat, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - B Del Río-Carrero
- Radiology Department, Bellvitge University Hospital, IDIBELL, University of Barcelona, L'Hospitalet de Llobregat, Spain
| | - R Llatjós
- Pathology Department, Bellvitge University, L'Hospitalet de Llobregat, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - L Pijuan
- Pathology Department, Bellvitge University, L'Hospitalet de Llobregat, Spain
| | - I Escobar
- Department of Thoracic Surgery, Bellvitge University Hospital, L'Hospitalet de Llobregat, Spain
| | - F Rivas
- Department of Thoracic Surgery, Bellvitge University Hospital, L'Hospitalet de Llobregat, Spain
| | - A Montes-Worboys
- Interstitial Lung Disease Unit, Respiratory Department, Bellvitge University Hospital, University of Barcelona, L'Hospitalet de Llobregat, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Y Gutiérrez-Rodríguez
- Interstitial Lung Disease Unit, Respiratory Department, Bellvitge University Hospital, University of Barcelona, L'Hospitalet de Llobregat, Spain
| | - D Rodríguez-Plaza
- Interstitial Lung Disease Unit, Respiratory Department, Bellvitge University Hospital, University of Barcelona, L'Hospitalet de Llobregat, Spain
| | - A Padró-Miquel
- Department of Clinical Genetics, Bellvitge University Hospital, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Spain
| | - A Esteve-Garcia
- Department of Clinical Genetics, Bellvitge University Hospital, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Spain
| | | | - C Flores
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
- Unidad de Investigación, del Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
- Área de Genómica, Instituto Tecnológico y de Energías Renovables, Santa Cruz de Tenerife, Spain
- Facultad de Ciencias de la Salud, Universidad Fernando de Pessoa Canarias, Las Palmas de Gran Canaria, Spain
| | - M Fuentes
- Interstitial Lung Disease Unit, Respiratory Department, Bellvitge University Hospital, University of Barcelona, L'Hospitalet de Llobregat, Spain
- Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Spain
| | - J Dorca
- University of Barcelona, School of Medicine. Respiratory Department, Bellvitge University Hospital, L'Hospitalet de Llobregat, Spain
| | - S Santos
- University of Barcelona, School of Medicine. Respiratory Department, Bellvitge University Hospital, L'Hospitalet de Llobregat, Spain
- Interstitial Lung Disease Unit, Respiratory Department, Bellvitge University Hospital, University of Barcelona, L'Hospitalet de Llobregat, Spain
- Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - R Perona
- Telomeropathies Lab, CSIC-IIB Alberto Sols, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
- Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - A Günther
- Justus Liebig University, Giessen, Germany
| | - J Shull
- Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Spain
| | - M Molina-Molina
- Interstitial Lung Disease Unit, Respiratory Department, Bellvitge University Hospital, University of Barcelona, L'Hospitalet de Llobregat, Spain.
- Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain.
| |
Collapse
|
10
|
Niewisch MR, Kim J, Giri N, Lunger JC, McReynolds LJ, Savage SA. Genotype and Associated Cancer Risk in Individuals With Telomere Biology Disorders. JAMA Netw Open 2024; 7:e2450111. [PMID: 39661387 PMCID: PMC11635530 DOI: 10.1001/jamanetworkopen.2024.50111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 10/18/2024] [Indexed: 12/12/2024] Open
Abstract
Importance Telomere biology disorders (TBDs) are inherited cancer-prone bone marrow failure syndromes with differences in morbidity and mortality based on mode of inheritance. Objective To quantify cancer risks in TBDs by genetic subgroups. Design, Setting, and Participants This longitudinal cohort study of TBDs assessed cancer occurrences from 2002 through 2022. Participants were individuals with a TBD-associated pathogenic germline variant recruited across institutions by self-referral. Data were collected and analyzed through June 30, 2022. Exposures The exposure was TBD genotypes, with subgroups defined by inheritance pattern (autosomal-dominant [AD-non-TINF2] vs autosomal-recessive/X-linked [AR/XLR] vs AD-TINF2). Main Outcomes and Measures The main outcome was cancer; secondary outcomes included death, or organ transplant. Cumulative cancer incidence was determined considering death or transplant as competing events. Observed:expected (O:E) ratios of cancer before and after any organ transplant were calculated using the National Cancer Institute's Surveillance, Epidemiology, and End Results Program. Results Among 230 individuals with TBD (135 [58.7%] male; median [range] age at last follow-up, 34.6 [1.4-82.2] years) included, the risk of cancer was 3-fold higher than the general population (O:E, 3.35 [95% CI, 2.32-4.68]). The highest risk was observed in individuals with AR/XLR (O:E, 19.16 [95% CI, 9.19-35.24]) with a significantly younger cancer onset than in individuals with AD-non-TINF2 (median [range] age, 36.7 [25.2-53.6] years vs 44.5 [32.2-67.5] years; P = .01). The risk of solid tumors was highest in individuals with AR/XLR (O:E = 23.97 [95% CI, 10.96-45.50]), predominantly head and neck squamous cell carcinomas (O:E, 276.00 [95% CI, 75.20-706.67]). Hematologic malignant neoplasm risk was highest in individuals with AD-non-TINF2 (O:E, 9.41 [95% CI, 4.30-17.86]). Solid tumor cumulative incidence increased to 12% for individuals with AR/XLR by age 45 years and to 13% for individuals with AD-non-TINF2 by age 70 years. The cumulative incidence of hematologic malignant neoplasms leveled off at 2% by age 30 years and 19% by age 70 years in individuals with AR/XLR and AD-non-TINF2, respectively. Individuals with AD-TINF2 showed the highest cumulative incidence for transplant or death (49% by age 15 years). Following transplant, individuals with AR/XLR (O:E, 136.11 [95% CI, 54.72-280.44) or AD-TINF2 (O:E, 81.07 [95% CI, 16.72-236.92]) had the highest cancer risk, predominantly young-onset head and neck squamous cell carcinomas (median [range] age, 32.2 [10.5-35.5] years). Conclusions and Relevance This cohort study of individuals with TBDs found an increased cancer risk compared with the general population, with the earliest age at onset for individuals with AR/XLR inheritance. Cancer risks increased after organ transplant across all subgroups. These differences in TBD-associated cancer risks by mode of inheritance suggest cancer screening could be tailored by genotype, but additional research is warranted.
Collapse
Affiliation(s)
- Marena R. Niewisch
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
- Department of Pediatric Hematology and Oncology, Medical School Hannover, Hannover, Germany
| | - Jung Kim
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Neelam Giri
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Judith C. Lunger
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Lisa J. McReynolds
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Sharon A. Savage
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
11
|
Liu Y, Calzone K, McReynolds LJ. Genetic predisposition to myelodysplastic syndrome: Genetic counseling and transplant implications. Semin Hematol 2024; 61:370-378. [PMID: 39443230 DOI: 10.1053/j.seminhematol.2024.09.003] [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: 05/31/2024] [Revised: 09/14/2024] [Accepted: 09/17/2024] [Indexed: 10/25/2024]
Abstract
The development of myelodysplastic syndromes (MDS) is influenced by various genetic predispositions. Several important genes contribute to disease susceptibility. This paper explores common genetic predisposition genes in MDS, including DDX41, CEBPA, and SAMD9/SAMD9L, which are linked to hereditary conditions presenting diagnostic and clinical challenges. It delves into hereditary conditions that affect platelet production and count, such as RUNX1, ETV6, and ANKRD26, detailing their clinical features and how they contribute to an increased risk of MDS. The discussion extends to additional genetic syndromes like GATA2 deficiency, telomere biology disorders, Fanconi anemia, and Li-Fraumeni syndrome, along with new findings on genes like ERG that offer new insights into disease etiology. The importance of genetic counseling in MDS is underscored, outlining its goals, methods for evaluating family history, risk assessment, and the ethical considerations involved. Furthermore, the role of hematopoietic cell transplantation in managing MDS, particularly in patients with germline syndromes, is reviewed, emphasizing the need for optimal donor selection and personalized treatment approaches. This comprehensive overview illustrates the critical role of genetic factors in MDS and highlights the need for continued research and tailored clinical practices to improve patient outcomes.
Collapse
Affiliation(s)
- Yi Liu
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD.
| | - Kathleen Calzone
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Lisa J McReynolds
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD
| |
Collapse
|
12
|
Roka K, Solomou E, Kattamis A, Stiakaki E. Telomere biology disorders: from dyskeratosis congenita and beyond. Postgrad Med J 2024; 100:879-889. [PMID: 39197110 DOI: 10.1093/postmj/qgae102] [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: 05/18/2024] [Revised: 07/05/2024] [Accepted: 07/31/2024] [Indexed: 08/30/2024]
Abstract
Defective telomerase function or telomere maintenance causes genomic instability. Alterations in telomere length and/or attrition are the primary features of rare diseases known as telomere biology disorders or telomeropathies. Recent advances in the molecular basis of these disorders and cutting-edge methods assessing telomere length have increased our understanding of this topic. Multiorgan manifestations and different phenotypes have been reported even in carriers within the same family. In this context, apart from dyskeratosis congenita, disorders formerly considered idiopathic (i.e. pulmonary fibrosis, liver cirrhosis) frequently correlate with underlying defective telomere maintenance mechanisms. Moreover, these patients are prone to developing specific cancer types and exhibit exceptional sensitivity and toxicity in standard chemotherapy regimens. The current review describes the diverse spectrum of clinical manifestations of telomere biology disorders in pediatric and adult patients, their correlation with pathogenic variants, and considerations during their management to increase awareness and improve a multidisciplinary approach.
Collapse
Affiliation(s)
- Kleoniki Roka
- Division of Pediatric Hematology-Oncology, First Department of Pediatrics, National and Kapodistrian University of Athens, Aghia Sophia Children's Hospital, Full Member of ERN GENTURIS and ERN EuroBloodnet, 8 Levadias Street, Goudi, Athens, 11527, Greece
| | - Elena Solomou
- Department of Internal Medicine, University of Patras Medical School, Rion, 26500, Greece
| | - Antonis Kattamis
- Division of Pediatric Hematology-Oncology, First Department of Pediatrics, National and Kapodistrian University of Athens, Aghia Sophia Children's Hospital, Full Member of ERN GENTURIS and ERN EuroBloodnet, 8 Levadias Street, Goudi, Athens, 11527, Greece
| | - Eftychia Stiakaki
- Department of Pediatric Hematology-Oncology & Autologous Hematopoietic Stem Cell Transplantation Unit, University Hospital of Heraklion & Laboratory of Blood Diseases and Childhood Cancer Biology, School of Medicine, University of Crete, Voutes, Heraklion, Crete, 71500, Greece
| |
Collapse
|
13
|
Guenechea G, Meza NW. The gray boundaries of aberrant shortening of the cellular timekeepers' edges. EMBO Mol Med 2024; 16:2276-2278. [PMID: 39198716 PMCID: PMC11473945 DOI: 10.1038/s44321-024-00122-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 08/05/2024] [Accepted: 08/08/2024] [Indexed: 09/01/2024] Open
Abstract
N. Meza and G. Guenechea discuss novel genetic variants identified in telomere biology disorder dyskeratosis congenita (DC) and DC-like patients, as reported by Tummala and colleagues in this issue of EMBO Mol Med .
Collapse
Affiliation(s)
- Guillermo Guenechea
- Centro de Investigaciones Energéticas Medioambientales y Tecnológicas and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIEMAT/CIBERER), Advanced Therapies Unit, Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD UAM), Madrid, Spain.
| | - Nestor W Meza
- Centro de Investigaciones Energéticas Medioambientales y Tecnológicas and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIEMAT/CIBERER), Advanced Therapies Unit, Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD UAM), Madrid, Spain
| |
Collapse
|
14
|
Wang CY, Chang SH, Hu CF, Hu YQ, Luo H, Liu L, Fan LL. ZCCHC8 p.P410A disrupts nucleocytoplasmic localization, promoting idiopathic pulmonary fibrosis and chronic obstructive pulmonary disease. Mol Med 2024; 30:144. [PMID: 39256642 PMCID: PMC11389302 DOI: 10.1186/s10020-024-00913-9] [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: 05/23/2024] [Accepted: 08/26/2024] [Indexed: 09/12/2024] Open
Abstract
BACKGROUND Idiopathic pulmonary fibrosis (IPF) is a special kind of chronic interstitial lung disease with insidious onset. Previous studies have revealed that mutations in ZCCHC8 may lead to IPF. The aim of this study is to explore the ZCCHC8 mutations in Chinese IPF patients. METHODS Here, we enrolled 124 patients with interstitial lung disease from 2017 to 2023 in our hospital. Whole exome sequencing and Sanger sequencing were employed to explore the genetic lesions of these patients. RESULTS Among these 124 patients, a novel mutation (NM_017612: c.1228 C > G/p.P410A) of Zinc Finger CCHC-Type Containing 8 (ZCCHC8)was identified in a family with IPF and chronic obstructive lung disease. As a component of the nuclear exosome-targeting complex that regulates the turnover of human telomerase RNA, ZCCHC8 mutations have been reported may lead to IPF in European population and American population. Functional study confirmed that the novel mutation can disrupt the nucleocytoplasmic localization of ZCCHC8, which further decreased the expression of DKC1 and RTEL1, and finally reduced the length of telomere and led to IPF and related disorders. CONCLUSIONS We may first report the ZCCHC8 mutation in Asian population with IPF. Our study broadens the mutation, phenotype, and population spectrum of ZCCHC8 deficiency.
Collapse
Affiliation(s)
- Chen-Yu Wang
- Department of Pulmonary and Critical Care Medicine, Research Unit of Respiratory Disease, Hunan Diagnosis and Treatment Center of Respiratory Disease, the Second Xiangya Hospital, Central South University, Changsha, 410011, China
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, 410013, China
| | - Si-Hua Chang
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, 410013, China
| | - Cheng-Feng Hu
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, 410013, China
| | - Yi-Qiao Hu
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, 410013, China
| | - Hong Luo
- Department of Pulmonary and Critical Care Medicine, Research Unit of Respiratory Disease, Hunan Diagnosis and Treatment Center of Respiratory Disease, the Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Lv Liu
- Department of Pulmonary and Critical Care Medicine, Research Unit of Respiratory Disease, Hunan Diagnosis and Treatment Center of Respiratory Disease, the Second Xiangya Hospital, Central South University, Changsha, 410011, China.
| | - Liang-Liang Fan
- Department of Pulmonary and Critical Care Medicine, Research Unit of Respiratory Disease, Hunan Diagnosis and Treatment Center of Respiratory Disease, the Second Xiangya Hospital, Central South University, Changsha, 410011, China.
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, 410013, China.
| |
Collapse
|
15
|
Coukos A, Saglietti C, Sempoux C, Haubitz M, Greuter T, Mittaz-Crettol L, Maurer F, Mdawar-Bailly E, Moradpour D, Alberio L, Good JM, Baerlocher GM, Fraga M. High prevalence of short telomeres in idiopathic porto-sinusoidal vascular disorder. Hepatol Commun 2024; 8:e0500. [PMID: 39037376 PMCID: PMC11265777 DOI: 10.1097/hc9.0000000000000500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 06/01/2024] [Indexed: 07/23/2024] Open
Abstract
BACKGROUND Telomeres prevent damage to coding DNA as end-nucleotides are lost during mitosis. Mutations in telomere maintenance genes cause excessive telomere shortening, a condition known as short telomere syndrome (STS). One hepatic manifestation documented in STS is porto-sinusoidal vascular disorder (PSVD). METHODS As the etiology of many cases of PSVD remains unknown, this study explored the extent to which short telomeres are present in patients with idiopathic PSVD. RESULTS This monocentric cross-sectional study included patients with histologically defined idiopathic PSVD. Telomere length in 6 peripheral blood leukocyte subpopulations was assessed using fluorescent in situ hybridization and flow cytometry. Variants of telomere-related genes were identified using high-throughput exome sequencing. In total, 22 patients were included, of whom 16 (73%) had short (9/22) or very short (7/22) telomeres according to age-adjusted reference ranges. Fourteen patients (64%) had clinically significant portal hypertension. Shorter telomeres were more frequent in males (p = 0.005) and patients with concomitant interstitial lung disease (p < 0.001), chronic kidney disease (p < 0.001), and erythrocyte macrocytosis (p = 0.007). Portal hypertension (p = 0.021), low serum albumin level (p < 0.001), low platelet count (p = 0.007), and hyperbilirubinemia (p = 0.053) were also associated with shorter telomeres. Variants in known STS-related genes were identified in 4 patients with VSTel and 1 with STel. CONCLUSIONS Short and very short telomeres were highly prevalent in patients with idiopathic PSVD, with 31% presenting with variants in telomere-related genes. Telomere biology may play an important role in vascular liver disease development. Clinicians should consider measuring telomeres in any patient presenting with PSVD.
Collapse
Affiliation(s)
- Alexander Coukos
- Divisions of Gastroenterology and Hepatology, Department of Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Chiara Saglietti
- Institute of Pathology, Department of Laboratory Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Christine Sempoux
- Institute of Pathology, Department of Laboratory Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Monika Haubitz
- Department of Biomedical Research, Laboratory for Hematopoiesis and Molecular Genetics, University of Bern, Bern, Switzerland
| | - Thomas Greuter
- Divisions of Gastroenterology and Hepatology, Department of Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Division of Gastroenterology and Hepatology, Department of Medicine, GZO-Zurich Regional Health Center, Wetzikon, Switzerland
| | - Laureane Mittaz-Crettol
- Genetic Medicine, Department of Laboratory Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Fabienne Maurer
- Genetic Medicine, Department of Laboratory Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Elise Mdawar-Bailly
- Divisions of Gastroenterology and Hepatology, Department of Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Darius Moradpour
- Divisions of Gastroenterology and Hepatology, Department of Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Lorenzo Alberio
- Department of Oncology, Hematology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Jean-Marc Good
- Genetic Medicine, Department of Laboratory Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Gabriela M. Baerlocher
- Department of Biomedical Research, Laboratory for Hematopoiesis and Molecular Genetics, University of Bern, Bern, Switzerland
| | - Montserrat Fraga
- Divisions of Gastroenterology and Hepatology, Department of Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
16
|
Küçüksolak M, Çoban HB, Bedir E. Optimization of biotransformation processes of Camarosporium laburnicola to improve production yields of potent telomerase activators. Microb Cell Fact 2024; 23:196. [PMID: 38987741 PMCID: PMC11234680 DOI: 10.1186/s12934-024-02468-0] [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: 05/09/2024] [Accepted: 06/30/2024] [Indexed: 07/12/2024] Open
Abstract
BACKGROUND Telomerase activators are promising agents for the healthy aging process and the treatment/prevention of short telomere-related and age-related diseases. The discovery of new telomerase activators and later optimizing their activities through chemical and biological transformations are crucial for the pharmaceutical sector. In our previous studies, several potent telomerase activators were discovered via fungal biotransformation, which in turn necessitated optimization of their production. It is practical to improve the production processes by implementing the design of experiment (DoE) strategy, leading to increased yield and productivity. In this study, we focused on optimizing biotransformation conditions utilizing Camarosporium laburnicola, a recently discovered filamentous fungus, to afford the target telomerase activators (E-CG-01, E-AG-01, and E-AG-02). RESULTS DoE approaches were used to optimize the microbial biotransformation processes of C. laburnicola. Nine parameters were screened by Plackett-Burman Design, and three significant parameters (biotransformation time, temperature, shaking speed) were optimized using Central Composite Design. After conducting validation experiments, we were able to further enhance the production yield of target metabolites through scale-up studies in shake flasks (55.3-fold for E-AG-01, 13-fold for E-AG-02, and 1.96-fold for E-CG-01). CONCLUSION Following a process optimization study using C. laburnicola, a significant increase was achieved in the production yields. Thus, the present study demonstrates a promising methodology to increase the production yield of potent telomerase activators. Furthermore, C. laburnicola is identified as a potential biocatalyst for further industrial utilization.
Collapse
Affiliation(s)
- Melis Küçüksolak
- Department of Bioengineering, Faculty of Engineering, İzmir Institute of Technology, Urla, İzmir, 35433, Turkey
| | - Hasan Buğra Çoban
- İzmir International Biomedicine and Genome Institute, Dokuz Eylül University, Balçova, İzmir, 35340, Turkey
| | - Erdal Bedir
- Department of Bioengineering, Faculty of Engineering, İzmir Institute of Technology, Urla, İzmir, 35433, Turkey.
| |
Collapse
|
17
|
Choudhary D, Lekshmon K, Singh C, Subramani VN, Singh Y, Mitra S, Sekar A, Malik M, Bhagat N, Shiva Kumar S, Taneja S, Gupta V, Ramachandran R, Singh S, Nada R, Kenwar D, Duseja AK, Yadav T, Malhotra P, Sharma A. Simultaneous Liver and Kidney Transplantation in a Patient With Telomere Biology Disorder: A Case Study. J Clin Exp Hepatol 2024; 14:101355. [PMID: 38389866 PMCID: PMC10879790 DOI: 10.1016/j.jceh.2024.101355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 01/27/2024] [Indexed: 02/24/2024] Open
Abstract
Organ transplantation is the primary therapy for organ failure caused by telomere biology disorder (TBD). We describe the first documented case of simultaneous liver and kidney transplantation (SLKTx) for TBD, although the diagnosis of TBD was reached only three months following SLKTx. The patient was born prematurely, displayed growth retardation, and developed chronic kidney and liver diseases. His pre-SLKTx autoimmune, metabolic, and viral assessments were negative, and persistent pancytopenia (bone marrow cellularity 70-80%) was attributed to renal disease-associated bone marrow changes. Following SLKTx, he was discharged with stable graft function on tacrolimus and prednisolone. Although mycophenolate mofetil was discontinued on the second postoperative day, his pancytopenia persisted. Despite extensive evaluations, including drug, immune, nutritional, and viral assessments, all results were negative. A bone marrow biopsy conducted three months post-transplant revealed significant hypocellularity (40-50%). Whole genome sequencing revealed a likely pathogenic variant of the TINF2 gene. The patient was subsequently treated with danazol. At the nine-month follow-up post-SLKTx, he exhibited stable graft function and improved cell counts while maintaining triple-drug immunosuppression. Given the lack of uniform diagnostic criteria for TBD, healthcare providers must be vigilant with patients presenting with multi-organ failure and persistent cytopenias. Effective pre-transplant screening for TBD can lead to timely diagnoses, better management, and improved post-transplant outcomes.
Collapse
Affiliation(s)
- Devprakash Choudhary
- Department of Renal Transplant Surgery, Post Graduate Institute of Medical Education & Research (PGIMER), Chandigarh, 160012, India
| | - K.S. Lekshmon
- Department of Haematology, Post Graduate Institute of Medical Education & Research (PGIMER), Chandigarh, 160012, India
| | - Charanpreet Singh
- Department of Haematology, Post Graduate Institute of Medical Education & Research (PGIMER), Chandigarh, 160012, India
| | - Vanji N. Subramani
- Department of Renal Transplant Surgery, Post Graduate Institute of Medical Education & Research (PGIMER), Chandigarh, 160012, India
| | - Yogendra Singh
- Department of Hepatology, Post Graduate Institute of Medical Education & Research (PGIMER), Chandigarh, 160012, India
| | - Suvradeep Mitra
- Department of Pathology, Post Graduate Institute of Medical Education & Research (PGIMER), Chandigarh, 160012, India
| | - Aravind Sekar
- Department of Pathology, Post Graduate Institute of Medical Education & Research (PGIMER), Chandigarh, 160012, India
| | - Manaswinee Malik
- Department of Haematology, Post Graduate Institute of Medical Education & Research (PGIMER), Chandigarh, 160012, India
| | - Naveen Bhagat
- Department of Hepatology, Post Graduate Institute of Medical Education & Research (PGIMER), Chandigarh, 160012, India
| | - S.P. Shiva Kumar
- Department of Renal Transplant Surgery, Post Graduate Institute of Medical Education & Research (PGIMER), Chandigarh, 160012, India
| | - Sunil Taneja
- Department of Hepatology, Post Graduate Institute of Medical Education & Research (PGIMER), Chandigarh, 160012, India
| | - Vikas Gupta
- Department of Surgical Gastroenterology, Post Graduate Institute of Medical Education & Research (PGIMER), Chandigarh, 160012, India
| | - Raja Ramachandran
- Department of Nephrology, Post Graduate Institute of Medical Education & Research (PGIMER), Chandigarh, 160012, India
| | - Sarbpreet Singh
- Department of Renal Transplant Surgery, Post Graduate Institute of Medical Education & Research (PGIMER), Chandigarh, 160012, India
| | - Ritambhra Nada
- Department of Pathology, Post Graduate Institute of Medical Education & Research (PGIMER), Chandigarh, 160012, India
| | - Deepesh Kenwar
- Department of Renal Transplant Surgery, Post Graduate Institute of Medical Education & Research (PGIMER), Chandigarh, 160012, India
| | - Ajay K. Duseja
- Department of Hepatology, Post Graduate Institute of Medical Education & Research (PGIMER), Chandigarh, 160012, India
| | - T.D. Yadav
- Department of Surgical Gastroenterology, Post Graduate Institute of Medical Education & Research (PGIMER), Chandigarh, 160012, India
| | - Pankaj Malhotra
- Department of Renal Transplant Surgery, Post Graduate Institute of Medical Education & Research (PGIMER), Chandigarh, 160012, India
| | - Ashish Sharma
- Department of Renal Transplant Surgery, Post Graduate Institute of Medical Education & Research (PGIMER), Chandigarh, 160012, India
| |
Collapse
|
18
|
Fernández-Varas B, Manguan-García C, Rodriguez-Centeno J, Mendoza-Lupiáñez L, Calatayud J, Perona R, Martín-Martínez M, Gutierrez-Rodriguez M, Benítez-Buelga C, Sastre L. Clinical mutations in the TERT and TERC genes coding for telomerase components induced oxidative stress, DNA damage at telomeres and cell apoptosis besides decreased telomerase activity. Hum Mol Genet 2024; 33:818-834. [PMID: 38641551 PMCID: PMC11031360 DOI: 10.1093/hmg/ddae015] [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: 10/02/2023] [Revised: 01/16/2024] [Accepted: 01/17/2024] [Indexed: 04/21/2024] Open
Abstract
Telomeres are nucleoprotein structures at the end of chromosomes that maintain their integrity. Mutations in genes coding for proteins involved in telomere protection and elongation produce diseases such as dyskeratosis congenita or idiopathic pulmonary fibrosis known as telomeropathies. These diseases are characterized by premature telomere shortening, increased DNA damage and oxidative stress. Genetic diagnosis of telomeropathy patients has identified mutations in the genes TERT and TERC coding for telomerase components but the functional consequences of many of these mutations still have to be experimentally demonstrated. The activity of twelve TERT and five TERC mutants, five of them identified in Spanish patients, has been analyzed. TERT and TERC mutants were expressed in VA-13 human cells that express low telomerase levels and the activity induced was analyzed. The production of reactive oxygen species, DNA oxidation and TRF2 association at telomeres, DNA damage response and cell apoptosis were determined. Most mutations presented decreased telomerase activity, as compared to wild-type TERT and TERC. In addition, the expression of several TERT and TERC mutants induced oxidative stress, DNA oxidation, DNA damage, decreased recruitment of the shelterin component TRF2 to telomeres and increased apoptosis. These observations might indicate that the increase in DNA damage and oxidative stress observed in cells from telomeropathy patients is dependent on their TERT or TERC mutations. Therefore, analysis of the effect of TERT and TERC mutations of unknown function on DNA damage and oxidative stress could be of great utility to determine the possible pathogenicity of these variants.
Collapse
Affiliation(s)
- Beatriz Fernández-Varas
- Instituto de Investigaciones Biomedicas Sols/Morreale CSIC/UAM, Arturo Duperier 4, 28029 Madrid, Spain
| | - Cristina Manguan-García
- Instituto de Investigaciones Biomedicas Sols/Morreale CSIC/UAM, Arturo Duperier 4, 28029 Madrid, Spain
- Centro de Investigacion Biomedica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III. C. Melchor Fernandez de Almagro, 3, 28029 Madrid, Spain
| | - Javier Rodriguez-Centeno
- Instituto de Investigaciones Biomedicas Sols/Morreale CSIC/UAM, Arturo Duperier 4, 28029 Madrid, Spain
| | - Lucía Mendoza-Lupiáñez
- Instituto de Investigaciones Biomedicas Sols/Morreale CSIC/UAM, Arturo Duperier 4, 28029 Madrid, Spain
| | - Joaquin Calatayud
- Departamento de Biología y Geología, Física y Química inorgánica. ESCET, Universidad Rey Juan Carlos, C/Tulipán s/n, Móstoles, C.P. 28933 Madrid, Spain
| | - Rosario Perona
- Centro de Investigacion Biomedica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III. C. Melchor Fernandez de Almagro, 3, 28029 Madrid, Spain
- Instituto de Salud Carlos III. Calle Monforte de Lemos 5, 28029 Madrid, Spain
| | | | | | - Carlos Benítez-Buelga
- Instituto de Investigaciones Biomedicas Sols/Morreale CSIC/UAM, Arturo Duperier 4, 28029 Madrid, Spain
| | - Leandro Sastre
- Instituto de Investigaciones Biomedicas Sols/Morreale CSIC/UAM, Arturo Duperier 4, 28029 Madrid, Spain
- Centro de Investigacion Biomedica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III. C. Melchor Fernandez de Almagro, 3, 28029 Madrid, Spain
| |
Collapse
|
19
|
Wang Q, Xu H, Yu W, Sun L, Zhao H, Shi X. Transformation of Severe Aplastic Anemia into Donor Cell Leukemia after Allogeneic Hematopoietic Stem Cell Transplantation: A Rare Case Report. AMERICAN JOURNAL OF CASE REPORTS 2024; 25:e943801. [PMID: 38632857 DOI: 10.12659/ajcr.943801] [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: 04/19/2024]
Abstract
BACKGROUND Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is an important treatment for severe aplastic anemia (SAA). It is known that SAA can evolve into malignant clonal diseases, such as acute myeloblastic leukemia (AML) or myelodysplastic syndrome. However, the transformation of SAA into AML after allo-HSCT is a rare phenomenon. Here, we report a case of SAA transformed into AML after patient received human leucocyte antigen (HLA)-matched sibling peripheral blood stem cell transplantation. CASE REPORT A 51-year-old female patient presented with petechiae and fatigue and received a diagnosis of idiopathic SAA. The immunosuppressive therapy combined with umbilical cord blood transplantation failed for this patient. Then, she received HLA-matched sibling allogeneic peripheral blood stem cell transplantation (allo-PBSCT). However, 445 days after allo-PBSCT, the patient had a diagnosis of AML by bone marrow puncture. Donor-recipient chimerism monitoring and cytogenetic analysis confirmed that the leukemia was donor cell origin. Notably, a new HOXA11 mutation was detected in the peripheral blood of the patient after transplantation by whole-exome sequencing, which was the same gene mutation detected in the donor. The patient received 1 cycle of induction chemotherapy with azacytidine and achieved complete remission. However, the leukemia relapsed after 2 cycles of consolidation chemotherapy. Unfortunately, the patient died of leukemia progression 575 days after allo-HSCT. CONCLUSIONS The mechanism of how normal donor hematopoietic cells transform to leukemia in the host remains unclear. Donor cell leukemia provides a unique opportunity to examine genetic variations in donors and hosts with regards to the progression to malignancy.
Collapse
Affiliation(s)
- Qianqian Wang
- Department of Hematology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China (mainland)
| | - Hong Xu
- Department of Hematology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China (mainland)
| | - Wei Yu
- Department of Hematology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China (mainland)
| | - Lingjie Sun
- Department of Hematology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China (mainland)
| | - Hongguo Zhao
- Department of Hematology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China (mainland)
| | - Xue Shi
- Department of Hematology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China (mainland)
| |
Collapse
|
20
|
Warsame F, Simonetto DA. Telomere Biology Disorder: A Focus on Gastrointestinal and Hepatic Manifestations. Curr Hematol Malig Rep 2024; 19:75-81. [PMID: 38372947 DOI: 10.1007/s11899-023-00723-6] [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: 12/29/2023] [Indexed: 02/20/2024]
Abstract
PURPOSE OF REVIEW Telomere biology disorders (TBD) encompass several illnesses caused by underlying mutations in telomere maintenance leading to premature telomere attrition and telomere dysfunction. These disorders have unique features but share common disease manifestations including pulmonary fibrosis, cirrhosis, and bone marrow failure. The goals of this article are to provide an overview of the gastrointestinal and hepatic manifestations of TBD, focusing on their pathophysiology, clinical disease states, and current management strategies. RECENT FINDINGS Telomere shortening has been observed in patients with chronic liver disease and is associated with a higher risk of progression to cirrhosis and portal hypertension. While the directionality of the association between telomere dysfunction and senescence on liver disease is not fully understood, research in TBD may provide clarity and could lead to future therapies for this increasingly prevalent disease. While treatment options remain limited in TBD-associated liver disease, recent studies point to the safety and efficacy of liver transplantation among patients with end-stage liver disease.
Collapse
Affiliation(s)
- Fatima Warsame
- Internal Medicine Residency, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Douglas A Simonetto
- Gastroenterology and Hepatology Fellowship Program, Division of Gastroenterology and Hepatology, Mayo Clinic, 200 1St SW, Rochester, MN, USA.
| |
Collapse
|
21
|
Paul S, McCourt PM, Le LTM, Ryu J, Czaja W, Bode AM, Contreras-Galindo R, Dong Z. Fyn-mediated phosphorylation of Menin disrupts telomere maintenance in stem cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.10.04.560876. [PMID: 37873235 PMCID: PMC10592958 DOI: 10.1101/2023.10.04.560876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Telomeres protect chromosome ends and determine the replication potential of dividing cells. The canonical telomere sequence TTAGGG is synthesized by telomerase holoenzyme, which maintains telomere length in proliferative stem cells. Although the core components of telomerase are well-defined, mechanisms of telomerase regulation are still under investigation. We report a novel role for the Src family kinase Fyn, which disrupts telomere maintenance in stem cells by phosphorylating the scaffold protein Menin. We found that Fyn knockdown prevented telomere erosion in human and mouse stem cells, validating the results with four telomere measurement techniques. We show that Fyn phosphorylates Menin at tyrosine 603 (Y603), which increases Menin's SUMO1 modification, C-terminal stability, and importantly, its association with the telomerase RNA component (TR). Using mass spectrometry, immunoprecipitation, and immunofluorescence experiments we found that SUMO1-Menin decreases TR's association with telomerase subunit Dyskerin, suggesting that Fyn's phosphorylation of Menin induces telomerase subunit mislocalization and may compromise telomerase function at telomeres. Importantly, we find that Fyn inhibition reduces accelerated telomere shortening in human iPSCs harboring mutations for dyskeratosis congenita.
Collapse
Affiliation(s)
- Souren Paul
- The Hormel Institute, University of Minnesota, 801 16th Avenue NE, Austin, MN 55912, USA
| | - Preston M. McCourt
- The Hormel Institute, University of Minnesota, 801 16th Avenue NE, Austin, MN 55912, USA
| | - Le Thi My Le
- The Hormel Institute, University of Minnesota, 801 16th Avenue NE, Austin, MN 55912, USA
| | - Joohyun Ryu
- The Hormel Institute, University of Minnesota, 801 16th Avenue NE, Austin, MN 55912, USA
- Department of Laboratory Medicine and Pathology, University of Minnesota School of Medicine, Minneapolis, MN, USA
| | - Wioletta Czaja
- The Hormel Institute, University of Minnesota, 801 16th Avenue NE, Austin, MN 55912, USA
- Department of Genetics, University of Alabama, Birmingham, AL 35294, USA
| | - Ann M. Bode
- The Hormel Institute, University of Minnesota, 801 16th Avenue NE, Austin, MN 55912, USA
| | - Rafael Contreras-Galindo
- The Hormel Institute, University of Minnesota, 801 16th Avenue NE, Austin, MN 55912, USA
- Department of Genetics, University of Alabama, Birmingham, AL 35294, USA
| | - Zigang Dong
- Department of Pathophysiology, School of Basic Medical Sciences, College of Medicine, Zhengzhou University, Henan, China 450001
| |
Collapse
|
22
|
Güllülü Ö, Mayer BE, Toplek FB. Linking Gene Fusions to Bone Marrow Failure and Malignant Transformation in Dyskeratosis Congenita. Int J Mol Sci 2024; 25:1606. [PMID: 38338888 PMCID: PMC10855549 DOI: 10.3390/ijms25031606] [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: 12/14/2023] [Revised: 01/26/2024] [Accepted: 01/26/2024] [Indexed: 02/12/2024] Open
Abstract
Dyskeratosis Congenita (DC) is a multisystem disorder intrinsically associated with telomere dysfunction, leading to bone marrow failure (BMF). Although the pathology of DC is largely driven by mutations in telomere-associated genes, the implications of gene fusions, which emerge due to telomere-induced genomic instability, remain unexplored. We meticulously analyzed gene fusions in RNA-Seq data from DC patients to provide deeper insights into DC's progression. The most significant DC-specific gene fusions were subsequently put through in silico assessments to ascertain biophysical and structural attributes, including charge patterning, inherent disorder, and propensity for self-association. Selected candidates were then analyzed using deep learning-powered structural predictions and molecular dynamics simulations to gauge their potential for forming higher-order oligomers. Our exploration revealed that genes participating in fusion events play crucial roles in upholding genomic stability, facilitating hematopoiesis, and suppressing tumors. Notably, our analysis spotlighted a particularly disordered polyampholyte fusion protein that exhibits robust higher-order oligomerization dynamics. To conclude, this research underscores the potential significance of several high-confidence gene fusions in the progression of BMF in DC, particularly through the dysregulation of genomic stability, hematopoiesis, and tumor suppression. Additionally, we propose that these fusion proteins might hold a detrimental role, specifically in inducing proteotoxicity-driven hematopoietic disruptions.
Collapse
Affiliation(s)
- Ömer Güllülü
- Department of Structural Biology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Benjamin E. Mayer
- Computational Biology & Simulation, Technische Universität Darmstadt, 64287 Darmstadt, Germany
| | - Fran Bačić Toplek
- Dipartimento di Bioscienze, Università degli Studi di Milano, 20133 Milano, Italy
| |
Collapse
|
23
|
Chini CCS, Cordeiro HS, Tran NLK, Chini EN. NAD metabolism: Role in senescence regulation and aging. Aging Cell 2024; 23:e13920. [PMID: 37424179 PMCID: PMC10776128 DOI: 10.1111/acel.13920] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 06/15/2023] [Accepted: 06/20/2023] [Indexed: 07/11/2023] Open
Abstract
The geroscience hypothesis proposes that addressing the biology of aging could directly prevent the onset or mitigate the severity of multiple chronic diseases. Understanding the interplay between key aspects of the biological hallmarks of aging is essential in delivering the promises of the geroscience hypothesis. Notably, the nucleotide nicotinamide adenine dinucleotide (NAD) interfaces with several biological hallmarks of aging, including cellular senescence, and changes in NAD metabolism have been shown to be involved in the aging process. The relationship between NAD metabolism and cellular senescence appears to be complex. On the one hand, the accumulation of DNA damage and mitochondrial dysfunction induced by low NAD+ can promote the development of senescence. On the other hand, the low NAD+ state that occurs during aging may inhibit SASP development as this secretory phenotype and the development of cellular senescence are both highly metabolically demanding. However, to date, the impact of NAD+ metabolism on the progression of the cellular senescence phenotype has not been fully characterized. Therefore, to explore the implications of NAD metabolism and NAD replacement therapies, it is essential to consider their interactions with other hallmarks of aging, including cellular senescence. We propose that a comprehensive understanding of the interplay between NAD boosting strategies and senolytic agents is necessary to advance the field.
Collapse
Affiliation(s)
- Claudia Christiano Silva Chini
- Metabolism and Molecular Nutrition Laboratory, Kogod Center on Aging, Department of Anesthesiology and Perioperative MedicineMayo Clinic College of MedicineRochesterMinnesotaUSA
- Metabolism and Molecular Nutrition Laboratory, Kogod Center on Aging, Department of Anesthesiology and Perioperative MedicineMayo Clinic College of MedicineJacksonvilleFloridaUSA
| | - Heidi Soares Cordeiro
- Metabolism and Molecular Nutrition Laboratory, Kogod Center on Aging, Department of Anesthesiology and Perioperative MedicineMayo Clinic College of MedicineRochesterMinnesotaUSA
- Metabolism and Molecular Nutrition Laboratory, Kogod Center on Aging, Department of Anesthesiology and Perioperative MedicineMayo Clinic College of MedicineJacksonvilleFloridaUSA
| | - Ngan Le Kim Tran
- Center for Clinical and Translational Science and Mayo Clinic Graduate School of Biomedical SciencesMayo ClinicJacksonvilleFloridaUSA
| | - Eduardo Nunes Chini
- Metabolism and Molecular Nutrition Laboratory, Kogod Center on Aging, Department of Anesthesiology and Perioperative MedicineMayo Clinic College of MedicineRochesterMinnesotaUSA
- Metabolism and Molecular Nutrition Laboratory, Kogod Center on Aging, Department of Anesthesiology and Perioperative MedicineMayo Clinic College of MedicineJacksonvilleFloridaUSA
| |
Collapse
|
24
|
Banaszak LG, Smith-Simmer K, Shoger K, Lovrien L, Malik A, Sandbo N, Sultan S, Guzy R, Lowery EM, Churpek JE. Implementation of a prospective screening strategy to identify adults with a telomere biology disorder among those undergoing lung transplant evaluation for interstitial lung disease. Respir Med 2023; 220:107464. [PMID: 37951311 DOI: 10.1016/j.rmed.2023.107464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 11/04/2023] [Accepted: 11/06/2023] [Indexed: 11/13/2023]
Abstract
INTRODUCTION Patients with interstitial lung disease (ILD) secondary to telomere biology disorders (TBD) experience increased morbidity after lung transplantation. Identifying patients with TBD may allow for personalized management to facilitate better outcomes. However, establishing a TBD diagnosis in adults is challenging. METHODS A TBD screening questionnaire was introduced prospectively into the lung transplant evaluation. Patients with ILD screening positive were referred for comprehensive TBD phenotyping and concurrent telomere length measurement and germline genetic testing. RESULTS Of 98 patients, 32 (33%) screened positive. Eight patients (8% of total; 25% of patients with a positive screen) met strict TBD diagnostic criteria, requiring either critically short lymphocyte telomeres (<1st percentile) (n = 4), a pathogenic variant in a TBD-associated gene (n = 1), or both (n = 3) along with a TBD clinical phenotype. Additional patients not meeting strict diagnostic criteria had histories consistent with TBD along with telomere lengths <10th percentile and/or rare variants in TBD-associated genes, highlighting a critical need to refine TBD diagnostic criteria for this patient population. CONCLUSION A TBD phenotype screening questionnaire in patients with ILD undergoing lung transplant evaluation has a diagnostic yield of 25%. Additional gene discovery, rare variant functional testing, and refined TBD diagnostic criteria are needed to realize the maximum benefit of testing for TBD in patients undergoing lung transplantation.
Collapse
Affiliation(s)
- Lauren G Banaszak
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, 53705, USA.
| | - Kelcy Smith-Simmer
- Oncology Genetics, University of Wisconsin Carbone Cancer Center, UW Health, Madison, WI, 53705, USA
| | - Kyle Shoger
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Lauren Lovrien
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Amy Malik
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Nathan Sandbo
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Samir Sultan
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Robert Guzy
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Erin M Lowery
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Jane E Churpek
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, 53705, USA.
| |
Collapse
|
25
|
Kanagal-Shamanna R, Schafernak KT, Calvo KR. Diagnostic work-up of hematological malignancies with underlying germline predisposition disorders (GPD). Semin Diagn Pathol 2023; 40:443-456. [PMID: 37977953 DOI: 10.1053/j.semdp.2023.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 11/07/2023] [Accepted: 11/09/2023] [Indexed: 11/19/2023]
Abstract
Hematological malignancies with underlying germline predisposition disorders have been recognized by the World Health Organization 5th edition and International Consensus Classification (ICC) classification systems. The list of genes and the associated phenotypes are expanding and involve both pediatric and adult populations. While the clinical presentation and underlying molecular pathogenesis are relatively well described, the knowledge regarding the bone marrow morphologic features, the landscape of somatic aberrations associated with progression to hematological malignancies is limited. These pose challenges in the diagnosis of low-grade myelodysplastic syndrome (MDS) to hematopathologists which carries direct implication for various aspects of clinical management of the patient, donor selection for transplantation, and family members. Here in, we provide a focused review on the diagnostic work-up of hematological malignancies with underlying germline predisposition disorders with emphasis on the spectrum of hematological malignancies associated with each entity, and characteristic bone marrow morphologic, somatic cytogenetic and molecular alterations at the time of diagnosis of hematological malignancies. We also review the key clinical, morphologic, and molecular features, that should initiate screening for these entities.
Collapse
Affiliation(s)
- Rashmi Kanagal-Shamanna
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Kristian T Schafernak
- Division of Pathology and Laboratory Medicine, Phoenix Children's Hospital, Phoenix, AZ, United States
| | - Katherine R Calvo
- Hematology Section, Department of Laboratory Medicine, National Institutes of Health Clinical Center, Bethesda, MD, United States.
| |
Collapse
|
26
|
Cuccuini W, Collonge-Rame MA, Auger N, Douet-Guilbert N, Coster L, Lafage-Pochitaloff M. Cytogenetics in the management of bone marrow failure syndromes: Guidelines from the Groupe Francophone de Cytogénétique Hématologique (GFCH). Curr Res Transl Med 2023; 71:103423. [PMID: 38016422 DOI: 10.1016/j.retram.2023.103423] [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: 06/30/2023] [Revised: 10/03/2023] [Accepted: 10/17/2023] [Indexed: 11/30/2023]
Abstract
Bone marrow failure syndromes are rare disorders characterized by bone marrow hypocellularity and resultant peripheral cytopenias. The most frequent form is acquired, so-called aplastic anemia or idiopathic aplastic anemia, an auto-immune disorder frequently associated with paroxysmal nocturnal hemoglobinuria, whereas inherited bone marrow failure syndromes are related to pathogenic germline variants. Among newly identified germline variants, GATA2 deficiency and SAMD9/9L syndromes have a special significance. Other germline variants impacting biological processes, such as DNA repair, telomere biology, and ribosome biogenesis, may cause major syndromes including Fanconi anemia, dyskeratosis congenita, Diamond-Blackfan anemia, and Shwachman-Diamond syndrome. Bone marrow failure syndromes are at risk of secondary progression towards myeloid neoplasms in the form of myelodysplastic neoplasms or acute myeloid leukemia. Acquired clonal cytogenetic abnormalities may be present before or at the onset of progression; some have prognostic value and/or represent somatic rescue mechanisms in inherited syndromes. On the other hand, the differential diagnosis between aplastic anemia and hypoplastic myelodysplastic neoplasm remains challenging. Here we discuss the value of cytogenetic abnormalities in bone marrow failure syndromes and propose recommendations for cytogenetic diagnosis and follow-up.
Collapse
Affiliation(s)
- Wendy Cuccuini
- Laboratoire d'Hématologie, Unité de Cytogénétique, Hôpital Saint-Louis, Assistance Publique Hôpitaux de Paris (APHP), 75475, Paris Cedex 10, France.
| | - Marie-Agnes Collonge-Rame
- Oncobiologie Génétique Bioinformatique UF Cytogénétique et Génétique Moléculaire, CHU de Besançon, Hôpital Minjoz, 25030, Besançon, France
| | - Nathalie Auger
- Laboratoire de Cytogénétique/Génétique des Tumeurs, Gustave Roussy, 94805, Villejuif, France
| | - Nathalie Douet-Guilbert
- Laboratoire de Génétique Chromosomique, CHU Brest, Hôpital Morvan, 29609, Brest Cedex, France
| | - Lucie Coster
- Laboratoire d'Hématologie, Secteur de Cytogénétique, Institut Universitaire de Cancérologie de Toulouse, CHU de Toulouse, 31059, Toulouse Cedex 9, France
| | - Marina Lafage-Pochitaloff
- Laboratoire de Cytogénétique Hématologique, CHU Timone, Assistance Publique Hôpitaux de Marseille (APHM), Aix Marseille Université, 13005, Marseille, France
| |
Collapse
|
27
|
Gachard N, Lafage-Pochitaloff M, Quessada J, Auger N, Collonge-Rame MA. Cytogenetics in the management of hematologic neoplasms with germline predisposition: guidelines from the Groupe Francophone de Cytogénétique Hématologique (GFCH). Curr Res Transl Med 2023; 71:103416. [PMID: 37865978 DOI: 10.1016/j.retram.2023.103416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 09/21/2023] [Accepted: 09/29/2023] [Indexed: 10/24/2023]
Abstract
The number of predisposing genes is continuously growing with the widespread availability of DNA sequencing, increasing the prevalence of hematologic malignancies with germline predisposition. Cytogenetic analyses provide an effective approach for the recognition of these malignancies with germline predisposition, which is critical for proper diagnosis, optimal treatment and genetic counseling. Based on the World Health Organization and the international consensus classifications as well as the European LeukemiaNet recommendations, this review first presents an advanced classification of neoplasms with germline predisposition focused on the acquired cytogenetic alterations during leukemogenesis. The various genetic rescue mechanisms and the progression to transformation are then explained. The review also outlines the specific constitutional and somatic cytogenetic aberrations indicative of germline predisposition disorders in B-acute lymphoblastic leukemia (ALL), T-ALL, bone marrow failure syndrome and myeloid neoplasms. An emphasis is made on monosomy 7 in the predisposition field, its frequency and diagnosis impact as well as its various circumstances of occurrence. Lastly, we propose cytogenetic technical recommendations and guidelines for clinical reporting of these specific aberrations.
Collapse
Affiliation(s)
- Nathalie Gachard
- Laboratoire d'hématologie, Centre de Biologie et de Recherche en Santé, CHU de Limoges, Limoges 87042, France; UMR CNRS 7276, INSERM U1262 Université de Limoges, Limoges 87025, France.
| | - Marina Lafage-Pochitaloff
- Laboratoire de Cytogénétique Hématologique, Département d'Hématologie, CHU Timone, APHM, Aix Marseille Université, Marseille 13005, France
| | - Julie Quessada
- Laboratoire de Cytogénétique Hématologique, Département d'Hématologie, CHU Timone, APHM, Aix Marseille Université, Marseille 13005, France
| | - Nathalie Auger
- Laboratoire de Cytogénétique -Génétique des Tumeurs - Gustave Roussy - 144 rue Edouard Vaillant, Villejuif 94805, France
| | - Marie-Agnès Collonge-Rame
- Oncobiologie Génétique Bioinformatique, UF Cytogénétique et Génétique Moléculaire, CHU de Besançon, Besançon 25030, France
| |
Collapse
|
28
|
Tichy ED, Lee JH, Li G, Estep KN, Brad Johnson F, Mourkioti F. Impacts of radiation exposure, hindlimb unloading, and recovery on murine skeletal muscle cell telomere length. NPJ Microgravity 2023; 9:76. [PMID: 37714858 PMCID: PMC10504369 DOI: 10.1038/s41526-023-00303-1] [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: 10/17/2022] [Accepted: 07/06/2023] [Indexed: 09/17/2023] Open
Abstract
Astronauts are exposed to harsh conditions, including cosmic radiation and microgravity. Spaceflight elongates human telomeres in peripheral blood, which shorten upon return to Earth and approach baseline levels during postflight recovery. Astronauts also encounter muscle atrophy, losing up to 20% loss of muscle mass on spaceflights. Telomere length changes in muscle cells of astronauts remain unexplored. This study investigates telomere alterations in grounded mice experiencing radiation exposure and muscle atrophy, via a hindlimb unloading spaceflight mimicking model. We find telomere lengthening is present in muscle stem cells and in myofiber nuclei, but not in muscle-resident endothelial cells. We further assessed telomere length in the model following hindlimb unloading recovery. We find that telomere length failed to return to baseline values. Our results suggest a role for telomeres in muscle acclimatization, which is relevant for the well-being of astronauts in space, and upon their return to Earth.
Collapse
Affiliation(s)
- Elisia D Tichy
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Ji-Hyung Lee
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Grant Li
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Katrina N Estep
- Department of Pathology and Laboratory Medicine, Institute on Aging, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - F Brad Johnson
- Department of Pathology and Laboratory Medicine, Institute on Aging, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Foteini Mourkioti
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Penn Institute for Regenerative Medicine, Musculoskeletal Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
29
|
Schreglmann SR, Goncalves T, Grant‐Peters M, Kia DA, Soreq L, Ryten M, Wood NW, Bhatia KP, Tomita K. Age-related telomere attrition in the human putamen. Aging Cell 2023; 22:e13861. [PMID: 37129365 PMCID: PMC10352551 DOI: 10.1111/acel.13861] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/10/2023] [Accepted: 04/18/2023] [Indexed: 05/03/2023] Open
Abstract
Age is a major risk factor for neurodegenerative diseases. Shortening of leucocyte telomeres with advancing age, arguably a measure of "biological" age, is a known phenomenon and epidemiologically correlated with age-related disease. The main mechanism of telomere shortening is cell division, rendering telomere length in post-mitotic cells presumably stable. Longitudinal measurement of human brain telomere length is not feasible, and cross-sectional cortical brain samples so far indicated no attrition with age. Hence, age-related changes in telomere length in the brain and the association between telomere length and neurodegenerative diseases remain unknown. Here, we demonstrate that mean telomere length in the putamen, a part of the basal ganglia, physiologically shortens with age, like leukocyte telomeres. This was achieved by using matched brain and leukocyte-rich spleen samples from 98 post-mortem healthy human donors. Using spleen telomeres as a reference, we further found that mean telomere length was brain region-specific, as telomeres in the putamen were significantly shorter than in the cerebellum. Expression analyses of genes involved in telomere length regulation and oxidative phosphorylation revealed that both region- and age-dependent expression pattern corresponded with region-dependent telomere length dynamics. Collectively, our results indicate that mean telomere length in the human putamen physiologically shortens with advancing age and that both local and temporal gene expression dynamics correlate with this, pointing at a potential mechanism for the selective, age-related vulnerability of the nigro-striatal network.
Collapse
Affiliation(s)
- Sebastian R. Schreglmann
- Queen Square Institute of NeurologyUniversity College LondonLondonUK
- Department of NeurologyUniversity Hospital WürzburgWürzburgGermany
| | - Tomas Goncalves
- Chromosome Maintenance Group, UCL Cancer InstituteUniversity College LondonLondonUK
- Centre for Genome Engineering and Maintenance, College of Health, Medicine and Life SciencesBrunel University LondonLondonUK
| | - Melissa Grant‐Peters
- Genetics and Genomic Medicine, Great Ormond Street Institute of Child HealthUniversity College LondonLondonUK
| | - Demis A. Kia
- Queen Square Institute of NeurologyUniversity College LondonLondonUK
| | - Lilach Soreq
- Queen Square Institute of NeurologyUniversity College LondonLondonUK
| | - Mina Ryten
- Genetics and Genomic Medicine, Great Ormond Street Institute of Child HealthUniversity College LondonLondonUK
- NIHR Great Ormond Street Hospital Biomedical Research CentreUniversity College LondonLondonUK
| | - Nicholas W. Wood
- Queen Square Institute of NeurologyUniversity College LondonLondonUK
| | - Kailash P. Bhatia
- Queen Square Institute of NeurologyUniversity College LondonLondonUK
| | - Kazunori Tomita
- Chromosome Maintenance Group, UCL Cancer InstituteUniversity College LondonLondonUK
- Centre for Genome Engineering and Maintenance, College of Health, Medicine and Life SciencesBrunel University LondonLondonUK
| |
Collapse
|
30
|
Roka K, Solomou EE, Kattamis A. Telomere biology: from disorders to hematological diseases. Front Oncol 2023; 13:1167848. [PMID: 37274248 PMCID: PMC10235513 DOI: 10.3389/fonc.2023.1167848] [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: 02/16/2023] [Accepted: 05/02/2023] [Indexed: 06/06/2023] Open
Abstract
Variations in the length of telomeres and pathogenic variants involved in telomere length maintenance have been correlated with several human diseases. Recent breakthroughs in telomere biology knowledge have contributed to the identification of illnesses named "telomeropathies" and revealed an association between telomere length and disease outcome. This review emphasizes the biology and physiology aspects of telomeres and describes prototype diseases in which telomeres are implicated in their pathophysiology. We also provide information on the role of telomeres in hematological diseases ranging from bone marrow failure syndromes to acute and chronic leukemias.
Collapse
Affiliation(s)
- Kleoniki Roka
- Division of Pediatric Hematology-Oncology, First Department of Pediatrics, National & Kapodistrian University of Athens, “Aghia Sophia” Children’s Hospital, Full Member of ERN GENTURIS, Athens, Greece
| | - Elena E. Solomou
- Department of Internal Medicine, University of Patras Medical School, Rion, Greece
| | - Antonis Kattamis
- Division of Pediatric Hematology-Oncology, First Department of Pediatrics, National & Kapodistrian University of Athens, “Aghia Sophia” Children’s Hospital, Full Member of ERN GENTURIS, Athens, Greece
| |
Collapse
|
31
|
Carlund O, Norberg A, Osterman P, Landfors M, Degerman S, Hultdin M. DNA methylation variations and epigenetic aging in telomere biology disorders. Sci Rep 2023; 13:7955. [PMID: 37193737 DOI: 10.1038/s41598-023-34922-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 05/10/2023] [Indexed: 05/18/2023] Open
Abstract
Telomere Biology Disorders (TBDs) are characterized by mutations in telomere-related genes leading to short telomeres and premature aging but with no strict correlation between telomere length and disease severity. Epigenetic alterations are also markers of aging and we aimed to evaluate whether DNA methylation (DNAm) could be part of the pathogenesis of TBDs. In blood from 35 TBD cases, genome-wide DNAm were analyzed and the cases were grouped based on relative telomere length (RTL): short (S), with RTL close to normal controls, and extremely short (ES). TBD cases had increased epigenetic age and DNAm alterations were most prominent in the ES-RTL group. Thus, the differentially methylated (DM) CpG sites could be markers of short telomeres but could also be one of the mechanisms contributing to disease phenotype since DNAm alterations were observed in symptomatic, but not asymptomatic, cases with S-RTL. Furthermore, two or more DM-CpGs were identified in four genes previously linked to TBD or telomere length (PRDM8, SMC4, VARS, and WNT6) and in three genes that were novel in telomere biology (MAS1L, NAV2, and TM4FS1). The DM-CpGs in these genes could be markers of aging in hematological cells, but they could also be of relevance for the progression of TBD.
Collapse
Affiliation(s)
- Olivia Carlund
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
| | - Anna Norberg
- Department of Medical Biosciences, Medical and Clinical Genetics, Umeå University, Umeå, Sweden
| | - Pia Osterman
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
| | - Mattias Landfors
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
| | - Sofie Degerman
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
- Department of Clinical Microbiology, Umeå University, Umeå, Sweden
| | - Magnus Hultdin
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden.
| |
Collapse
|
32
|
Tobai H, Endo M, Ishimura M, Moriya K, Yano J, Kanamori K, Sato N, Amanuma F, Maruyama H, Muramatsu H, Shibahara J, Narita M, Fumoto S, Peltier D, Ohga S. Neonatal intestinal obstruction in Hoyeraal-Hreidarsson syndrome with novel RTEL1 variants. Pediatr Blood Cancer 2023; 70:e30250. [PMID: 36776130 DOI: 10.1002/pbc.30250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 01/22/2023] [Accepted: 01/24/2023] [Indexed: 02/14/2023]
Affiliation(s)
- Hiromi Tobai
- Division of Pediatrics, Iwate Prefectural Iwai Hospital, Ichinoseki, Japan.,Department of Pediatrics, Graduate School of Medical Sciences, Kyusyu University, Fukuoka, Japan
| | - Mikiya Endo
- Department of Pediatrics, Iwate Medical University, School of Medicine, Morioka, Japan
| | - Masataka Ishimura
- Department of Pediatrics, Graduate School of Medical Sciences, Kyusyu University, Fukuoka, Japan
| | - Kunihiko Moriya
- Division of Pediatrics, Iwate Prefectural Iwai Hospital, Ichinoseki, Japan.,Department of Pediatrics, National Defense Medical College, Tokorozawa, Japan
| | - Jun Yano
- Division of Pediatrics, Iwate Prefectural Iwai Hospital, Ichinoseki, Japan
| | - Keita Kanamori
- Division of Pediatrics, Iwate Prefectural Iwai Hospital, Ichinoseki, Japan
| | - Norio Sato
- Division of Pediatrics, Iwate Prefectural Iwai Hospital, Ichinoseki, Japan
| | - Fumitaka Amanuma
- Division of Pediatrics, Iwate Prefectural Iwai Hospital, Ichinoseki, Japan
| | - Hidekazu Maruyama
- Division of Pediatrics, Iwate Prefectural Iwai Hospital, Ichinoseki, Japan
| | - Hideki Muramatsu
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Junji Shibahara
- Department of Pathology, Kyorin University School of Medicine, Mitaka, Japan
| | - Masami Narita
- Department of Pediatrics, Kyorin University School of Medicine, Mitaka, Japan
| | - Seiko Fumoto
- Department of Pediatrics, Kyorin University School of Medicine, Mitaka, Japan
| | - Daniel Peltier
- Pediatric Hematology and Oncology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Shouichi Ohga
- Department of Pediatrics, Graduate School of Medical Sciences, Kyusyu University, Fukuoka, Japan
| |
Collapse
|
33
|
Vulsteke JB, Smith V, Bonroy C, Derua R, Blockmans D, De Haes P, Vanderschueren S, Lenaerts JL, Claeys KG, Wuyts WA, Verschueren P, Vanhandsaeme G, Piette Y, De Langhe E, Bossuyt X. Identification of new telomere- and telomerase-associated autoantigens in systemic sclerosis. J Autoimmun 2023; 135:102988. [PMID: 36634459 DOI: 10.1016/j.jaut.2022.102988] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 12/16/2022] [Indexed: 01/12/2023]
Abstract
PURPOSE In up to 20% of patients with systemic sclerosis (SSc) no known autoantibody specificity can be identified. Recently discovered autoantigens, such as telomeric repeat binding factor 1 (TERF1), as well as established autoantigens, like RuvBL1/2, are associated with telomere and telomerase biology. We aimed to identify new telomere- and telomerase-associated autoantigens in patients with SSc without known autoantibody specificity. METHODS Unlabelled protein immunoprecipitation combined with gel-free liquid chromatography-tandem mass spectrometry (IP-MS) was performed with sera of 106 patients with SSc from two tertiary referral centres that had a nuclear pattern on HEp-2 indirect immunofluorescence without previously identified autoantibody. Telomere- or telomerase-associated proteins or protein complexes precipitated by individual sera were identified. Candidate autoantigens were confirmed through immunoprecipitation-western blot (IP-WB). A custom Luminex xMAP assay for 5 proteins was evaluated with sera from persons with SSc (n = 467), other systemic autoimmune rheumatic diseases (n = 923), non-rheumatic disease controls (n = 187) and healthy controls (n = 199). RESULTS Eight telomere- and telomerase-associated autoantigens were identified in a total of 11 index patients, including the THO complex (n = 3, all with interstitial lung disease and two with cardiac involvement), telomeric repeat-binding factor 2 (TERF2, n = 1), homeobox-containing protein 1 (HMBOX1, n = 2), regulator of chromosome condensation 1 (RCC1, n = 1), nucleolar and coiled-body phosphoprotein 1 (NOLC1, n = 1), dyskerin (DKC1, n = 1), probable 28S rRNA (cytosine(4447)-C(5))-methyltransferase (NOP2, n = 1) and nuclear valosin-containing protein-like (NVL, n = 2). A Luminex xMAP assay for THO complex subunit 1 (THOC1), TERF2, NOLC1, NOP2 and NVL revealed high reactivity in all index patients, but also in other patients with SSc and disease controls. However, the reactivity by xMAP assay in these other patients was not confirmed by IP-WB. CONCLUSION IP-MS revealed key telomere- and telomerase-associated proteins and protein complexes as autoantigens in patients with SSc.
Collapse
Affiliation(s)
- Jean-Baptiste Vulsteke
- KU Leuven, Department of Development and Regeneration, Skeletal Biology and Engineering Research Center, Leuven, Belgium; Rheumatology, University Hospitals Leuven, Leuven, Belgium
| | - Vanessa Smith
- Ghent University, Department of Internal Medicine, Ghent, Belgium; Unit for Molecular Immunology and Inflammation, VIB Inflammation Research Center (IRC), Ghent, Belgium; Rheumatology, Ghent University Hospital, Ghent, Belgium; European Reference Network on Rare and Complex Connective Tissue and Musculoskeletal Diseases (ERN ReCONNET), Belgium
| | - Carolien Bonroy
- Ghent University, Department of Diagnostic Sciences, Ghent, Belgium; Laboratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Rita Derua
- KU Leuven, Department of Cellular and Molecular Medicine, Laboratory of Protein Phosphorylation and Proteomics, Leuven, Belgium; KU Leuven, SyBioMa, Leuven, Belgium
| | - Daniel Blockmans
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Laboratory for Clinical Infectious and Inflammatory Disorders, Leuven, Belgium; General Internal Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Petra De Haes
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Leuven, Belgium; Dermatology, University Hospitals Leuven, Leuven, Belgium
| | - Steven Vanderschueren
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Laboratory for Clinical Infectious and Inflammatory Disorders, Leuven, Belgium; General Internal Medicine, University Hospitals Leuven, Leuven, Belgium; European Reference Network on Rare Immunodeficiency, Autoinflammatory and Autoimmune Diseases (ERN RITA), Belgium
| | - Jan L Lenaerts
- Rheumatology, University Hospitals Leuven, Leuven, Belgium
| | - Kristl G Claeys
- KU Leuven, Department of Neurosciences, Laboratory for Muscle Diseases and Neuropathies, Neurology, University Hospitals Leuven, Leuven, Belgium; European Reference Network on Rare Neuromuscular Diseases (ERN EURO-NMD), Belgium
| | - Wim A Wuyts
- KU Leuven, Department of Chronic Diseases and Metabolism, Laboratory of Respiratory Diseases and Thoracic Surgery, Unit for Interstitial Lung Diseases, Respiratory Medicine, University Hospitals Leuven, Leuven, Belgium; European Reference Network on Rare Respiratory Diseases (ERN LUNG), Belgium
| | - Patrick Verschueren
- KU Leuven, Department of Development and Regeneration, Skeletal Biology and Engineering Research Center, Leuven, Belgium; Rheumatology, University Hospitals Leuven, Leuven, Belgium
| | | | - Yves Piette
- Ghent University, Department of Internal Medicine, Ghent, Belgium; Rheumatology, Ghent University Hospital, Ghent, Belgium
| | - Ellen De Langhe
- KU Leuven, Department of Development and Regeneration, Skeletal Biology and Engineering Research Center, Leuven, Belgium; Rheumatology, University Hospitals Leuven, Leuven, Belgium; European Reference Network on Rare and Complex Connective Tissue and Musculoskeletal Diseases (ERN ReCONNET), Belgium; European Reference Network on Rare Immunodeficiency, Autoinflammatory and Autoimmune Diseases (ERN RITA), Belgium
| | - Xavier Bossuyt
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Clinical and Diagnostic Immunology, Leuven, Belgium; Laboratory Medicine, University Hospitals Leuven, Leuven, Belgium.
| |
Collapse
|
34
|
Atluri H, Gerstein YS, DiNardo CD. Approach Toward Germline Predisposition Syndromes in Patients with Hematologic Malignancies. Curr Hematol Malig Rep 2022; 17:275-285. [PMID: 36279069 DOI: 10.1007/s11899-022-00684-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/24/2022] [Indexed: 01/27/2023]
Abstract
PURPOSE OF REVIEW Hematologic malignancies were previously thought to be primarily sporadic cancers without germline predispositions. However, over the last two decades, with the widespread use of next generation sequencing (NGS), there have been several genes have been identified that carry a risk of inheriting hematologic malignancies. Identification of individuals with hereditary hematologic malignancies (HHM) involves a high index of suspicion and careful attention to family history, clinical features, and variant allele frequency on somatic NGS panels. RECENT FINDINGS Over the last several years, many genetic predisposition syndromes have been recognized to have unique features with both hematologic and non-hematologic co-morbidities. Multidisciplinary evaluation, including genetic counseling, is critical to optimizing diagnostic testing of individuals and at-risk family members. Prompt recognition of affected patients is imperative not only for personalized surveillance strategies but also for proper donor selection for those undergoing stem cell transplantation to avoid familial donors who also may share the same germline mutation. Herein, we describe our approach to recognizing patients suspected to carry a germline predisposition to hematologic malignancies and evaluation within a hereditary hematologic malignancies clinic (HHMC).
Collapse
Affiliation(s)
- Himachandana Atluri
- Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Yoheved S Gerstein
- Clinical Cancer Genetics Program, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Courtney D DiNardo
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| |
Collapse
|
35
|
Zafirovic S, Macvanin M, Stanimirovic J, Obradovic M, Radovanovic J, Melih I, Isenovic E. Association Between Telomere Length and Cardiovascular Risk: Pharmacological Treatments Affecting Telomeres and Telomerase Activity. Curr Vasc Pharmacol 2022; 20:465-474. [PMID: 35986545 DOI: 10.2174/1570161120666220819164240] [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: 02/18/2022] [Revised: 06/03/2022] [Accepted: 06/06/2022] [Indexed: 01/25/2023]
Abstract
Telomeres represent the ends of chromosomes, and they are composed of an extensive number of - TTAGGG nucleotide sequence repeats in humans. Telomeres prevent chromosome degradation, participate in stabilization, and regulate the DNA repair system. Inflammation and oxidative stress have been identified as important processes causing cardiovascular disease and accelerating telomere shortening rate. This review investigates the link between telomere length and pathological vascular conditions from experimental and human studies. Also, we discuss pharmacological treatments affecting telomeres and telomerase activity.
Collapse
Affiliation(s)
- Sonja Zafirovic
- Department of Radiobiology and Molecular Genetics, VINČA Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Mirjana Macvanin
- Department of Radiobiology and Molecular Genetics, VINČA Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Julijana Stanimirovic
- Department of Radiobiology and Molecular Genetics, VINČA Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Milan Obradovic
- Department of Radiobiology and Molecular Genetics, VINČA Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Jelena Radovanovic
- Department of Radiobiology and Molecular Genetics, VINČA Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Irena Melih
- Faculty of Stomatology, Pancevo, University Business Academy, 21000 Novi Sad, Serbia
| | - Esma Isenovic
- Department of Radiobiology and Molecular Genetics, VINČA Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
36
|
Ren HL, Zheng YC, He GQ, Gao J, Guo X. A Rare Heterozygous TINF2 Deletional Frameshift Mutation in a Chinese Pedigree With a Spectrum of TBDs Phenotypes. Front Genet 2022; 13:913133. [PMID: 35873475 PMCID: PMC9300939 DOI: 10.3389/fgene.2022.913133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 06/15/2022] [Indexed: 11/13/2022] Open
Abstract
Telomere biology disorders (TBDs) induced by TINF2 mutations manifest clinically with a spectrum of phenotypes, from silent carriers to a set of overlapping conditions. A rare TINF2 frameshift mutation (c.591delG) encoding a truncated mutant TIN2 protein (p.W198fs) was identified in a 6-years-and-3-month-old Chinese girl with neuroblastoma (NB) by next generation sequencing and confirmed by Sanger sequencing. To explore the possible implications of TINF2 mutations in TBDs development, the TINF2 mutant was transfected into the human embryonic kidney (HEK) 293T cells, and mRNA expression of the shelterin complex components as well as the cellular distribution of mutant TIN2 were examined. The TINF2 mutation was phenotypically associated with short stature in the proband, nail dystrophy and spotted hypopigmentation in her mother, and psoriasis in her older brother. I-TASSER modeling analysis revealed conformational changes of the mutant TIN2 protein and loss of pivotal domains downstream of the 198th amino acid. Additionally, mRNA expression of the shelterin components was downregulated, and TIN2 mutant protein expression was reduced in HEK293T cells transfected with mutant TINF2. Furthermore, instead of being restricted to the nucleus, the mutant TIN2 was identified in both the cytoplasm and the nucleus. The TINF2 gene mutation might impair the function of the shelterin complex and the telomere maintenance mechanisms, both of which are involved in the development of TBDs. TBDs have been associated with increased cancer risk. To the best of our knowledge, this is the first report of NB in patients with TBDs. The relationship between the TINF2 mutation and NB may need to further study.
Collapse
Affiliation(s)
- Hai-Long Ren
- Division of Spinal Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ying-Chun Zheng
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Guo-Qian He
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, China
| | - Ju Gao
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, China
- *Correspondence: Ju Gao, ; Xia Guo,
| | - Xia Guo
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, China
- *Correspondence: Ju Gao, ; Xia Guo,
| |
Collapse
|
37
|
Abbasi A, Chen C, Gandhi CK, Wu R, Pardo A, Selman M, Floros J. Single Nucleotide Polymorphisms (SNP) and SNP-SNP Interactions of the Surfactant Protein Genes Are Associated With Idiopathic Pulmonary Fibrosis in a Mexican Study Group; Comparison With Hypersensitivity Pneumonitis. Front Immunol 2022; 13:842745. [PMID: 35720392 PMCID: PMC9201215 DOI: 10.3389/fimmu.2022.842745] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 05/09/2022] [Indexed: 01/12/2023] Open
Abstract
Surfactant proteins (SPs) are important for normal lung function and innate immunity of the lungs and their genes have been identified with significant genetic variability. Changes in quantity or quality of SPs due to genetic mutations or natural genetic variability may alter their functions and contribute to the host susceptibility for particular diseases. Alternatively, SP single nucleotide polymorphisms (SNPs) can serve as markers to identify disease risk or response to therapies, as shown for other genes in a number of other studies. In the current study, we evaluated associations of SFTP SNPs with idiopathic pulmonary fibrosis (IPF) by studying novel computational models where the epistatic effects (dominant, additive, recessive) of SNP-SNP interactions could be evaluated, and then compared the results with a previously published hypersensitivity pneumonitis (HP) study where the same novel models were used. Mexican Hispanic patients (IPF=84 & HP=75) and 194 healthy control individuals were evaluated. The goal was to identify SP SNPs and SNP-SNP interactions that associate with IPF as well as SNPs and interactions that may be unique to each of these interstitial diseases or common between them. We observed: 1) in terms of IPF, i) three single SFTPA1 SNPs to associate with decreased IPF risk, ii) three SFTPA1 haplotypes to associate with increased IPF risk, and iii) a number of three-SNP interactions to associate with IPF susceptibility. 2) Comparison of IPF and HP, i) three SFTPA1 and one SFTPB SNP associated with decreased risk in IPF but increased risk in HP, and one SFTPA1 SNP associated with decreased risk in both IPF and HP, ii) a number of three-SNP interactions with the same or different effect pattern associated with IPF and/or HP susceptibility, iii) one of the three-SNP interactions that involved SNPs of SFTPA1, SFTPA2, and SFTPD, with the same effect pattern, was associated with a disease-specific outcome, a decreased and increased risk in HP and IPF, respectively. This is the first study that compares the SP gene variants in these two phenotypically similar diseases. Our findings indicate that SNPs of all SFTPs may play an important role in the genetic susceptibility to IPF and HP. Importantly, IPF and HP share some SP genetic variants, suggesting common pathophysiological mechanisms and pathways regarding surfactant biogenesis, but also some differences, highlighting the diverse underlying pathogenic mechanisms between an inflammatory-driven fibrosis (HP) and an epithelial-driven fibrosis (IPF). Alternatively, the significant SNPs identified here, along with SNPs of other genes, could serve as markers to distinguish these two devastating diseases.
Collapse
Affiliation(s)
- Ata Abbasi
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran.,Department of Pathology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Chixiang Chen
- Department of Public Health Science, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Chintan K Gandhi
- Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Rongling Wu
- Department of Public Health Science, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Annie Pardo
- Facultad de Ciencias, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Moises Selman
- Unidad de Investigación, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosio Villegas", Mexico City, Mexico
| | - Joanna Floros
- Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA, United States.,Department of Obstetrics & Gynecology, Pennsylvania State University College of Medicine, Hershey, PA, United States
| |
Collapse
|
38
|
Kim HY, Kim HJ, Kim SH. Genetics and genomics of bone marrow failure syndrome. Blood Res 2022; 57:86-92. [PMID: 35483932 PMCID: PMC9057661 DOI: 10.5045/br.2022.2022056] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 04/18/2022] [Accepted: 04/19/2022] [Indexed: 12/04/2022] Open
Abstract
Inherited bone marrow failure syndrome (IBMFS) is a group of clinically heterogeneous disorders characterized by significant hematological cytopenias of one or more hematopoietic cell lineages and is associated with an increased risk of cancer. The genetic etiology of IBMFS includes germline mutations impacting several key biological processes, such as DNA repair, telomere biology, and ribosome biogenesis, which may cause four major syndromes: Fanconi anemia, dyskeratosis congenita, Diamond-Blackfan anemia, and Shwachman-Diamond syndrome. Although the clinical features of some patients may be typical of a particular IBMFS, overlapping and atypical clinical manifestations and variable penetrance pose diagnostic challenges. Here, we review the clinical and genetic features of the major forms of IBMFS and discuss their molecular genetic diagnosis. Next-generation sequencing-based gene panel testing or whole exome sequencing will help elucidate the genetic causes and underlying mechanisms of this genetically heterogeneous group of diseases.
Collapse
Affiliation(s)
- Hyun-Young Kim
- Department of Laboratory Medicine and Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hee-Jin Kim
- Department of Laboratory Medicine and Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Sun-Hee Kim
- Department of Laboratory Medicine and Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
39
|
Vagher J, Gammon A, Kohlmann W, Jeter J. Non-Melanoma Skin Cancers and Other Cutaneous Manifestations in Bone Marrow Failure Syndromes and Rare DNA Repair Disorders. Front Oncol 2022; 12:837059. [PMID: 35359366 PMCID: PMC8960432 DOI: 10.3389/fonc.2022.837059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 02/17/2022] [Indexed: 11/17/2022] Open
Abstract
Although most non-melanoma skin cancers are felt to be sporadic in origin, these tumors do play a role in several cancer predisposition syndromes. The manifestations of skin cancers in these hereditary populations can include diagnosis at extremely early ages and/or multiple primary cancers, as well as tumors at less common sites. Awareness of baseline skin cancer risks for these individuals is important, particularly in the setting of treatments that may compromise the immune system and further increase risk of cutaneous malignancies. Additionally, diagnosis of these disorders and management of non-cutaneous manifestations of these diseases have profound implications for both the patient and their family. This review highlights the current literature on the diagnosis, features, and non-melanoma skin cancer risks associated with lesser-known cancer predisposition syndromes, including bone marrow failure disorders, genomic instability disorders, and base excision repair disorders.
Collapse
Affiliation(s)
- Jennie Vagher
- Family Cancer Assessment Clinic, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, United States
| | - Amanda Gammon
- Family Cancer Assessment Clinic, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, United States
| | - Wendy Kohlmann
- Family Cancer Assessment Clinic, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, United States
| | - Joanne Jeter
- Family Cancer Assessment Clinic, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, United States
| |
Collapse
|
40
|
Sharma R, Sahoo SS, Honda M, Granger SL, Goodings C, Sanchez L, Künstner A, Busch H, Beier F, Pruett-Miller SM, Valentine MB, Fernandez AG, Chang TC, Géli V, Churikov D, Hirschi S, Pastor VB, Boerries M, Lauten M, Kelaidi C, Cooper MA, Nicholas S, Rosenfeld JA, Polychronopoulou S, Kannengiesser C, Saintomé C, Niemeyer CM, Revy P, Wold MS, Spies M, Erlacher M, Coulon S, Wlodarski MW. Gain-of-function mutations in RPA1 cause a syndrome with short telomeres and somatic genetic rescue. Blood 2022; 139:1039-1051. [PMID: 34767620 PMCID: PMC8854676 DOI: 10.1182/blood.2021011980] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 10/15/2021] [Indexed: 11/20/2022] Open
Abstract
Human telomere biology disorders (TBD)/short telomere syndromes (STS) are heterogeneous disorders caused by inherited loss-of-function mutations in telomere-associated genes. Here, we identify 3 germline heterozygous missense variants in the RPA1 gene in 4 unrelated probands presenting with short telomeres and varying clinical features of TBD/STS, including bone marrow failure, myelodysplastic syndrome, T- and B-cell lymphopenia, pulmonary fibrosis, or skin manifestations. All variants cluster to DNA-binding domain A of RPA1 protein. RPA1 is a single-strand DNA-binding protein required for DNA replication and repair and involved in telomere maintenance. We showed that RPA1E240K and RPA1V227A proteins exhibit increased binding to single-strand and telomeric DNA, implying a gain in DNA-binding function, whereas RPA1T270A has binding properties similar to wild-type protein. To study the mutational effect in a cellular system, CRISPR/Cas9 was used to knock-in the RPA1E240K mutation into healthy inducible pluripotent stem cells. This resulted in severe telomere shortening and impaired hematopoietic differentiation. Furthermore, in patients with RPA1E240K, we discovered somatic genetic rescue in hematopoietic cells due to an acquired truncating cis RPA1 mutation or a uniparental isodisomy 17p with loss of mutant allele, coinciding with stabilized blood counts. Using single-cell sequencing, the 2 somatic genetic rescue events were proven to be independently acquired in hematopoietic stem cells. In summary, we describe the first human disease caused by germline RPA1 variants in individuals with TBD/STS.
Collapse
Affiliation(s)
- Richa Sharma
- Department of Hematology, St Jude Children's Research Hospital, Memphis, TN
| | - Sushree S Sahoo
- Department of Hematology, St Jude Children's Research Hospital, Memphis, TN
| | - Masayoshi Honda
- Department of Biochemistry and Molecular Biology, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - Sophie L Granger
- Department of Biochemistry and Molecular Biology, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - Charnise Goodings
- Department of Hematology, St Jude Children's Research Hospital, Memphis, TN
| | - Louis Sanchez
- Structure et Instabilité des Génomes, Muséum National d'Histoire Naturelle, Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 7196, INSERM Unité1154, Paris, France
| | - Axel Künstner
- Lübeck Institute of Experimental Dermatology and Institute of Cardiogenetics, University of Lübeck, Lübeck, Germany
- University Cancer Center Schleswig-Holstein, University Hospital of Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Hauke Busch
- Lübeck Institute of Experimental Dermatology and Institute of Cardiogenetics, University of Lübeck, Lübeck, Germany
- University Cancer Center Schleswig-Holstein, University Hospital of Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Fabian Beier
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | | | | | | | - Ti-Cheng Chang
- Center for Applied Bioinformatics, St Jude Children's Research Hospital, Memphis, TN
| | - Vincent Géli
- Marseille Cancer Research Centre, Unité1068 INSERM, UMR 7258 CNRS, Aix-Marseille University (UM 105), Institut Paoli-Calmettes, Equipe Labellisée par la Ligue Nationale contre le Cancer, Marseille, France
| | - Dmitri Churikov
- Marseille Cancer Research Centre, Unité1068 INSERM, UMR 7258 CNRS, Aix-Marseille University (UM 105), Institut Paoli-Calmettes, Equipe Labellisée par la Ligue Nationale contre le Cancer, Marseille, France
| | - Sandrine Hirschi
- Department of Respiratory Medicine and Rare Pulmonary Diseases, Strasbourg University Hospital, Strasbourg, France
| | - Victor B Pastor
- Center for Applied Bioinformatics, St Jude Children's Research Hospital, Memphis, TN
| | - Melanie Boerries
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK), Freiburg, Germany, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Melchior Lauten
- University Hospital Schleswig-Holstein, Department of Pediatrics, University of Lübeck, Lübeck, Germany
| | - Charikleia Kelaidi
- Department of Pediatric Hematology/Oncology, Aghia Sophia Children's Hospital, Athens, Greece
| | - Megan A Cooper
- Department of Pediatrics, Washington University School of Medicine, St Louis, MO
| | | | - Jill A Rosenfeld
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX
| | - Sophia Polychronopoulou
- Department of Pediatric Hematology/Oncology, Aghia Sophia Children's Hospital, Athens, Greece
| | - Caroline Kannengiesser
- Department of Genetics, Bichat Hospital, Assistance Publique-Hôpitaux de Paris, Paris University, INSERM U1152, Paris, France
| | - Carole Saintomé
- Structure et Instabilité des Génomes, Muséum National d'Histoire Naturelle, Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 7196, INSERM Unité1154, Paris, France
- Sorbonne Université, Education and Research Unit for Life Sciences (UFR 927), Paris, France
| | - Charlotte M Niemeyer
- German Cancer Consortium (DKTK), Freiburg, Germany, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, Medical Center, University of Freiburg, Freiburg, Germany; and
| | - Patrick Revy
- Université de Paris, Imagine Institute, Laboratory of Genome Dynamics in the Immune System, Laboratoire Labellisé Ligue, INSERM UMR 1163, Paris, France
| | - Marc S Wold
- Department of Biochemistry and Molecular Biology, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - Maria Spies
- Department of Biochemistry and Molecular Biology, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - Miriam Erlacher
- German Cancer Consortium (DKTK), Freiburg, Germany, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, Medical Center, University of Freiburg, Freiburg, Germany; and
| | - Stéphane Coulon
- Marseille Cancer Research Centre, Unité1068 INSERM, UMR 7258 CNRS, Aix-Marseille University (UM 105), Institut Paoli-Calmettes, Equipe Labellisée par la Ligue Nationale contre le Cancer, Marseille, France
| | - Marcin W Wlodarski
- Department of Hematology, St Jude Children's Research Hospital, Memphis, TN
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, Medical Center, University of Freiburg, Freiburg, Germany; and
| |
Collapse
|
41
|
Baarz BR, Rink L. Rebalancing the unbalanced aged immune system - A special focus on zinc. Ageing Res Rev 2022; 74:101541. [PMID: 34915196 DOI: 10.1016/j.arr.2021.101541] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 11/18/2021] [Accepted: 12/09/2021] [Indexed: 02/08/2023]
Abstract
Nowadays, aging is understood as a dynamic and multifaceted dysregulation process that spares almost no human organ or cell. The immune system being among the most affected, it has been shown predominantly that its integrity determines the tightrope walk between the difference of escaping or suffering from age-related diseases. Next to drug-based anti-aging strategies, micronutrient intervention may represent an emerging but less radical way to slow immune aging. While a sufficient supply of a variety of micronutrients is undeniably important, adequate intake of the trace element zinc appears to tower over others in terms of reaching old age. Inconveniently, zinc deficiency prevalence among the elderly is high, which in turn contributes to increased susceptibility to infection, decreased anti-tumor immunity as well as attenuated response to vaccination. Driven by this research, this review aims to provide a comprehensive and up-to-date overview of the various rebalancing capabilities of zinc in the unbalanced immune system of the elderly. This includes an in-depth and cell type-centered discussion on the role of zinc in immunosenescence and inflammaging. We further address upcoming translational aspects e.g. how zinc deficiency promotes the flourishing of certain pathogenic taxa of the gut microbiome and how zinc supply counteracts such alterations in a manner that may contribute to longevity. In the light of the ongoing COVID-19 pandemic, we also briefly review current knowledge on the interdependency between age, zinc status, and respiratory infections. Based on two concrete examples and considering the latest findings in the field we conclude our remarks by outlining tremendous parallels between suboptimal zinc status and accelerated aging on the one hand and an optimized zinc status and successful aging on the other hand.
Collapse
|
42
|
Utility of Telomerase Gene Mutation Testing in Patients with Idiopathic Pulmonary Fibrosis in Routine Practice. Cells 2022; 11:cells11030372. [PMID: 35159182 PMCID: PMC8834025 DOI: 10.3390/cells11030372] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 12/13/2021] [Accepted: 01/18/2022] [Indexed: 02/01/2023] Open
Abstract
Recent studies have suggested that causative variants in telomerase complex genes (TCGs) are present in around 10% of individuals with idiopathic pulmonary fibrosis (IPF) regardless of family history of the disease. However, the studies used a case-control rare variant enrichment study design which is not directly translatable to routine practice. To validate the prevalence results and to establish the individual level, routine clinical practice, and utility of those results we performed next generation sequencing of TCGs on a cohort of well-characterized consecutive individuals with IPF (diagnosis established according to ATS/ERS/JRS/ALAT guidelines). Of 27 IPF patients, three had a family history of idiopathic interstitial pneumonia (familial IPF) and 24 did not (sporadic IPF). Pathogenic/likely-pathogenic variants (according to American College of Medical Genetics criteria) in TCG were found in three individuals (11.1%) of the whole cohort; specifically, they were present in 2 out of 24 (8.3%) of the sporadic and in 1 out of 3 (33.3%) of the patients with familial IPF. Our results, which were established on an individual-patient level study design and in routine clinical practice (as opposed to the case-control study design), are roughly in line with the around 10% prevalence of causative TCG variants in patients with IPF.
Collapse
|
43
|
Guzonjić A, Sopić M, Ostanek B, Kotur-Stevuljević J. Telomere length as a biomarker of aging and diseases. ARHIV ZA FARMACIJU 2022. [DOI: 10.5937/arhfarm72-36376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
As research related to healthspan and lifespan has become a hot topic, the necessity for a reliable and practical biomarker of aging (BoA), which can provide information about mortality and morbidity risk, along with remaining life expectancy, has increased. The chromosome terminus non-coding protective structure that prevents genomic instability is called a telomere. The continual shortening of telomeres, which affects their structure as well as function, is a hallmark of agedness. The aforementioned process is a potential cause of age-related diseases (ARDs), leading to a bad prognosis and a low survival rate, which compromise health and longevity. Hence, studies scrutinizing the BoAs often include telomere length (TL) as a prospective candidate. The results of these studies suggest that TL measurement can only provide an approximate appraisal of the aging rate, and its implementation into clinical practice and routine use as a BoA has many limitations and challenges. Nevertheless, measuring TL while determining other biomarkers can be used to assess biological age. This review focuses on the importance of telomeres in health, senescence, and diseases, as well as on summarizing the results and conclusions of previous studies evaluating TL as a potential BoA.
Collapse
|
44
|
Guo X, Wang J, Zou W, Wei W, Guan X, Liu J. Exploring microenvironment strategies to delay mesenchymal stem cell senescence. Stem Cells Dev 2021; 31:38-52. [PMID: 34913751 DOI: 10.1089/scd.2021.0254] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have recently emerged as an important candidate for cell therapy and tissue regeneration. However, some limitations in translational research and therapies still exist, such as insufficient cell supply, inadequate differentiation potential, and decreased immune capacity, all of which result from replicative senescence during long-term in vitro culture. In vitro, stem cells lack a protective microenvironment owing to the absence of physical and biochemical cues compared with the in vivo niche, which provides dynamic physicochemical and biological cues. This difference results in accelerated aging after long-term in vitro culture. Therefore, it remains a great challenge to delay replicative senescence in culture. Constructing a microenvironment to delay replicative senescence of MSCs by maintaining their phenotypes, properties, and functions is a feasible strategy to solve this problem and has made measurable progress both in preclinical studies and clinical trials. Here, we review the current knowledge on the characteristics of senescent MSCs, explore the molecular mechanisms of MSCs senescence, describe the niche of MSCs, and discuss some current microenvironment strategies to delay MSCs replicative senescence that can broaden their range of therapeutic applications.
Collapse
Affiliation(s)
- Xunhui Guo
- First Affiliated Hospital of Dalian Medical University, 74710, Stem Cell Clinical Research Center, Dalian, China;
| | - Jiayi Wang
- First Affiliated Hospital of Dalian Medical University, 74710, Stem Cell Clinical Research Center, Dalian, Dalian, China;
| | - Wei Zou
- Liaoning Normal University, 66523, College of Life Sciences, Dalian, China;
| | - Wenjuan Wei
- First Affiliated Hospital of Dalian Medical University, 74710, Dalian, China, 116011;
| | - Xin Guan
- First Affiliated Hospital of Dalian Medical University, 74710, Dalian, China, 116011;
| | - Jing Liu
- First Affiliated Hospital of Dalian Medical University, 74710, Dalian, China, 116011;
| |
Collapse
|
45
|
Zhang K, Xu L, Cong YS. Telomere Dysfunction in Idiopathic Pulmonary Fibrosis. Front Med (Lausanne) 2021; 8:739810. [PMID: 34859008 PMCID: PMC8631932 DOI: 10.3389/fmed.2021.739810] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 10/05/2021] [Indexed: 12/14/2022] Open
Abstract
Idiopathic pulmonary fibrosis is an age-dependent progressive and fatal lung disease of unknown etiology, which is characterized by the excessive accumulation of extracellular matrix inside the interstitial layer of the lung parenchyma that leads to abnormal scar architecture and compromised lung function capacity. Recent genetic studies have attributed the pathological genes or genetic mutations associated with familial idiopathic pulmonary fibrosis (IPF) and sporadic IPF to telomere-related components, suggesting that telomere dysfunction is an important determinant of this disease. In this study, we summarized recent advances in our understanding of how telomere dysfunction drives IPF genesis. We highlighted the key role of alveolar stem cell dysfunction caused by telomere shortening or telomere uncapping, which bridged the gap between telomere abnormalities and fibrotic lung pathology. We emphasized that senescence-associated secretory phenotypes, innate immune cell infiltration, and/or inflammation downstream of lung stem cell dysfunction influenced the native microenvironment and local cell signals, including increased transforming growth factor-beta (TGF-β) signaling in the lung, to induce pro-fibrotic conditions. In addition, the failed regeneration of new alveoli due to alveolar stem cell dysfunction might expose lung cells to elevated mechanical tension, which could activate the TGF-β signaling loop to promote the fibrotic process, especially in a periphery-to-center pattern as seen in IPF patients. Understanding the telomere-related molecular and pathophysiological mechanisms of IPF would provide new insights into IPF etiology and therapeutic strategies for this fatal disease.
Collapse
Affiliation(s)
- Kexiong Zhang
- Key Laboratory of Aging and Cancer Biology of Zhejiang Province, School of Basic Medical Sciences, Hangzhou Normal University, Hangzhou, China
| | - Lu Xu
- Key Laboratory of Aging and Cancer Biology of Zhejiang Province, School of Basic Medical Sciences, Hangzhou Normal University, Hangzhou, China
| | - Yu-Sheng Cong
- Key Laboratory of Aging and Cancer Biology of Zhejiang Province, School of Basic Medical Sciences, Hangzhou Normal University, Hangzhou, China
| |
Collapse
|
46
|
Mackintosh JA, Pietsch M, Lutzky V, Enever D, Bancroft S, Apte SH, Tan M, Yerkovich ST, Dickinson JL, Pickett HA, Selvadurai H, Grainge C, Goh NS, Hopkins P, Glaspole I, Reynolds PN, Wrobel J, Jaffe A, Corte TJ, Chambers DC. TELO-SCOPE study: a randomised, double-blind, placebo-controlled, phase 2 trial of danazol for short telomere related pulmonary fibrosis. BMJ Open Respir Res 2021; 8:8/1/e001127. [PMID: 34857525 PMCID: PMC8640666 DOI: 10.1136/bmjresp-2021-001127] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 11/15/2021] [Indexed: 11/17/2022] Open
Abstract
Introduction Recent discoveries have identified shortened telomeres and related mutations in people with pulmonary fibrosis (PF). There is evidence to suggest that androgens, including danazol, may be effective in lengthening telomeres in peripheral blood cells. This study aims to assess the safety and efficacy of danazol in adults and children with PF associated with telomere shortening. Methods and analysis A multi-centre, double-blind, placebo-controlled, randomised trial of danazol will be conducted in subjects aged >5 years with PF associated with age-adjusted telomere length ≤10th centile measured by flow fluorescence in situ hybridisation; or in children, a diagnosis of dyskeratosis congenita. Adult participants will receive danazol 800 mg daily in two divided doses or identical placebo capsules orally for 12 months, in addition to standard of care (including pirfenidone or nintedanib). Paediatric participants will receive danazol 2 mg/kg/day orally in two divided doses or identical placebo for 6 months. If no side effects are encountered, the dose will be escalated to 4 mg/kg/day (maximum 800 mg daily) orally in two divided doses for a further 6 months. The primary outcome is change in absolute telomere length in base pairs, measured using the telomere shortest length assay (TeSLA), at 12 months in the intention to treat population. Ethics and dissemination Ethics approval has been granted in Australia by the Metro South Human Research Ethics Committee (HREC/2020/QMS/66385). The study will be conducted and reported according to Standard Protocol Items: Recommendations for Interventional Trials guidelines. Results will be published in peer-reviewed journals and presented at international and national conferences. Trial registration numbers NCT04638517; Australian New Zealand Clinical Trials Registry (ACTRN12620001363976p).
Collapse
Affiliation(s)
- John A Mackintosh
- Queensland Lung Transplant Service, Department of Thoracic Medicine, The Prince Charles Hospital, Chermside, Queensland, Australia
| | - Maria Pietsch
- Queensland Lung Transplant Service, Department of Thoracic Medicine, The Prince Charles Hospital, Chermside, Queensland, Australia
| | - Viviana Lutzky
- Queensland Lung Transplant Service, Department of Thoracic Medicine, The Prince Charles Hospital, Chermside, Queensland, Australia
| | - Debra Enever
- Queensland Lung Transplant Service, Department of Thoracic Medicine, The Prince Charles Hospital, Chermside, Queensland, Australia
| | - Sandra Bancroft
- Queensland Lung Transplant Service, Department of Thoracic Medicine, The Prince Charles Hospital, Chermside, Queensland, Australia
| | - Simon H Apte
- Queensland Lung Transplant Service, Department of Thoracic Medicine, The Prince Charles Hospital, Chermside, Queensland, Australia.,Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Maxine Tan
- Queensland Lung Transplant Service, Department of Thoracic Medicine, The Prince Charles Hospital, Chermside, Queensland, Australia
| | - Stephanie T Yerkovich
- Queensland Lung Transplant Service, Department of Thoracic Medicine, The Prince Charles Hospital, Chermside, Queensland, Australia.,Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Joanne L Dickinson
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Hilda A Pickett
- Children's Medical Research Institute, University of Sydney, Westmead, New South Wales, Australia
| | - Hiran Selvadurai
- Department of Respiratory Medicine, The Children's Hospital at Westmead, Sydney, New South Wales, Australia.,Discipline of Paediatrics and Child Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Christopher Grainge
- Department of Respiratory Medicine, John Hunter Hospital, Newcastle, New South Wales, Australia
| | - Nicole S Goh
- Respiratory and Sleep Medicine Department, Austin Health, Heidelberg, Victoria, Australia.,Institute for Breathing and Sleep, Melbourne, Victoria, Australia.,Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Victoria, Australia
| | - Peter Hopkins
- Queensland Lung Transplant Service, Department of Thoracic Medicine, The Prince Charles Hospital, Chermside, Queensland, Australia.,Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Ian Glaspole
- Department of Allergy and Respiratory Medicine, Alfred Hospital, Melbourne, Victoria, Australia
| | - Paul N Reynolds
- Department of Respiratory Medicine, Royal Adelaide Hospital, Adelaide, South Australia, Australia
| | - Jeremy Wrobel
- Advanced Lung Disease Unit, Fiona Stanley Hospital, Murdoch, Western Australia, Australia.,Department of Medicine, University of Notre Dame, Perth, Western Australia, Australia
| | - Adam Jaffe
- School of Women's and Children's Health, Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Tamera J Corte
- Respiratory Medicine, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia.,Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
| | - Daniel C Chambers
- Queensland Lung Transplant Service, Department of Thoracic Medicine, The Prince Charles Hospital, Chermside, Queensland, Australia.,Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|