1
|
Choi JC. Perinuclear organelle trauma at the nexus of cardiomyopathy pathogenesis arising from loss of function LMNA mutation. Nucleus 2025; 16:2449500. [PMID: 39789731 PMCID: PMC11730615 DOI: 10.1080/19491034.2024.2449500] [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: 10/31/2024] [Revised: 12/22/2024] [Accepted: 12/30/2024] [Indexed: 01/12/2025] Open
Abstract
Over the past 25 years, nuclear envelope (NE) perturbations have been reported in various experimental models with mutations in the LMNA gene. Although the hypothesis that NE perturbations from LMNA mutations are a fundamental feature of striated muscle damage has garnered wide acceptance, the molecular sequalae provoked by the NE damage and how they underlie disease pathogenesis such as cardiomyopathy (LMNA cardiomyopathy) remain poorly understood. We recently shed light on one such consequence, by employing a cardiomyocyte-specific Lmna deletion in vivo in the adult heart. We observed extensive NE perturbations prior to cardiac function deterioration with collateral damage in the perinuclear space. The Golgi is particularly affected, leading to cytoprotective stress responses that are likely disrupted by the progressive deterioration of the Golgi itself. In this review, we discuss the etiology of LMNA cardiomyopathy with perinuclear 'organelle trauma' as the nexus between NE damage and disease pathogenesis.
Collapse
Affiliation(s)
- Jason C. Choi
- Center for Translational Medicine, Department of Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
2
|
Nair A, Khanna J, Kler J, Ragesh R, Sengupta K. Nuclear envelope and chromatin choreography direct cellular differentiation. Nucleus 2025; 16:2449520. [PMID: 39943681 PMCID: PMC11834525 DOI: 10.1080/19491034.2024.2449520] [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: 10/01/2024] [Revised: 12/28/2024] [Accepted: 12/30/2024] [Indexed: 02/20/2025] Open
Abstract
The nuclear envelope plays an indispensable role in the spatiotemporal organization of chromatin and transcriptional regulation during the intricate process of cell differentiation. This review outlines the distinct regulatory networks between nuclear envelope proteins, transcription factors and epigenetic modifications in controlling the expression of cell lineage-specific genes during differentiation. Nuclear lamina with its associated nuclear envelope proteins organize heterochromatin via Lamina-Associated Domains (LADs), proximal to the nuclear periphery. Since nuclear lamina is mechanosensitive, we critically examine the impact of extracellular forces on differentiation outcomes. The nuclear envelope is spanned by nuclear pore complexes which, in addition to their central role in transport, are associated with chromatin organization. Furthermore, mutations in the nuclear envelope proteins disrupt differentiation, resulting in developmental disorders. Investigating the underlying nuclear envelope controlled regulatory mechanisms of chromatin remodelling during lineage commitment will accelerate our fundamental understanding of developmental biology and regenerative medicine.
Collapse
Affiliation(s)
- Anjitha Nair
- Chromosome Biology Lab (CBL), Indian Institute of Science Education and Research (IISER) Pune, Maharashtra, India
| | - Jayati Khanna
- Chromosome Biology Lab (CBL), Indian Institute of Science Education and Research (IISER) Pune, Maharashtra, India
| | - Jashan Kler
- Chromosome Biology Lab (CBL), Indian Institute of Science Education and Research (IISER) Pune, Maharashtra, India
| | - Rohith Ragesh
- Chromosome Biology Lab (CBL), Indian Institute of Science Education and Research (IISER) Pune, Maharashtra, India
| | - Kundan Sengupta
- Chromosome Biology Lab (CBL), Indian Institute of Science Education and Research (IISER) Pune, Maharashtra, India
| |
Collapse
|
3
|
Tukker M, Te Rijdt WP, Amin AS, Morris-Rosendahl DJ, Hirsch A, Ben-Haim Y, Houweling AC, Varnava A, Behr ER, Edwards M, Vanmaele A, Hajdarpasic A, von der Thusen J, Michels M, de Boer RA, van Slegtenhorst MA, Caliskan K. High incidence of malignant arrhythmias and heart failure in patients with RBM20-associated cardiomyopathy: A multicenter cohort study and review of the literature. Int J Cardiol 2025; 434:133350. [PMID: 40339755 DOI: 10.1016/j.ijcard.2025.133350] [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: 09/04/2024] [Revised: 03/22/2025] [Accepted: 04/30/2025] [Indexed: 05/10/2025]
Abstract
BACKGROUND Patients with RBM20 cardiomyopathy present with an aggressive phenotype, associated with premature malignant arrhythmias, sudden cardiac death, and progressive heart failure (HF). This study aimed to investigate genotype-phenotype correlations, clinical outcomes, and causes of death in patients with RBM20-associated cardiomyopathy and review the current literature. METHODS The cohort included patients with cardiomyopathy harboring pathogenic (P) or likely pathogenic (LP) RBM20 variants. For survival and regression analysis, a control group matched for sex, age, and presence of left ventricular dysfunction was included. Additionally, a comprehensive literature search was conducted. RESULTS Sixty-two patients (45 % male, 42 ± 15 years at presentation) were included. We found 11 truncating variants. Patients with truncating variants diagnosed with HF were older compared to patients with missense variants (mean age 62 ± 9 vs. 45 ± 14; p = 0.01). Over a median follow-up duration of 5.0 [1.0-10.5] years, 21 (34 %) patients reached the composite endpoint, with 19 (31 %) patients experiencing malignant ventricular arrhythmia (VA) (mean age 45 ± 15 years, 63 % males). Males exhibited higher risk for the composite endpoint (log-rank p = 0.02), particularly for VA (log-rank p = 0.007). The literature review analyzed 34 studies comprising 678 patients (53 % male). In these studies, 123 (24 %) patients experienced a VA, 58 (12 %) underwent a heart transplant or were treated with LVAD, and 52 (11 %) died. CONCLUSION This multicenter study highlights the severe phenotype associated with LP/P RBM20 variants, with a high incidence of VA, particularly in males. Additionally, this study presents 11 truncating variants mainly observed in older individuals.
Collapse
Affiliation(s)
- Martijn Tukker
- ErasmusMC, Cardiovascular Institute, Thoraxcenter, Department of Cardiology, Rotterdam, the Netherlands
| | | | - Ahmad S Amin
- Department of Clinical Cardiology, Heart Center, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
| | - Deborah J Morris-Rosendahl
- Clinical Genetics and Genomics Laboratory, Royal Brompton and Harefield NHS Foundation Trust, London, UK; Genomic Medicine, National Heart and Lung Institute, Imperial College London, London, UK
| | - Alexander Hirsch
- ErasmusMC, Cardiovascular Institute, Thoraxcenter, Department of Cardiology, Rotterdam, the Netherlands; Department of Radiology and Nuclear Medicine, Rotterdam, the Netherlands
| | - Yael Ben-Haim
- Department of Cardiology, St. George's University Hospitals NHS Foundation Trust, London, UK
| | - Arjan C Houweling
- Department of Human Genetics, Amsterdam University Medical Centre, Amsterdam, the Netherlands
| | - Amanda Varnava
- Hammersmith Hospital, Imperial NHS Healthcare Trust, London, UK
| | - Elijah R Behr
- Department of Cardiology, St. George's University Hospitals NHS Foundation Trust, London, UK
| | - Matthew Edwards
- Clinical Genetics and Genomics Laboratory, Royal Brompton and Harefield NHS Foundation Trust, London, UK
| | - Alexander Vanmaele
- ErasmusMC, Cardiovascular Institute, Thoraxcenter, Department of Cardiology, Rotterdam, the Netherlands
| | - Aida Hajdarpasic
- Department of Medical Genetics, Sarajevo Medical School, University Sarajevo School of Science and Technology, Sarajevo, Bosnia and Herzegovina
| | - Jan von der Thusen
- Department of Pathology, Erasmus MC University Medical Centre, Rotterdam, the Netherlands
| | - Michelle Michels
- ErasmusMC, Cardiovascular Institute, Thoraxcenter, Department of Cardiology, Rotterdam, the Netherlands
| | - Rudolf A de Boer
- ErasmusMC, Cardiovascular Institute, Thoraxcenter, Department of Cardiology, Rotterdam, the Netherlands
| | | | - Kadir Caliskan
- ErasmusMC, Cardiovascular Institute, Thoraxcenter, Department of Cardiology, Rotterdam, the Netherlands.
| |
Collapse
|
4
|
Bauer R, Parker C, Gorsic LK, Hayes MG, Kunselman AR, Legro RS, Welt CK, Urbanek M. Rare Variation in LMNA Underlies Polycystic Ovary Syndrome Pathogenesis in 2 Independent Cohorts. J Clin Endocrinol Metab 2025; 110:e2217-e2232. [PMID: 39484826 DOI: 10.1210/clinem/dgae761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 10/09/2024] [Accepted: 10/29/2024] [Indexed: 11/03/2024]
Abstract
CONTEXT Polycystic ovary syndrome (PCOS) is a common, heritable endocrinopathy that is a common cause of anovulatory infertility in reproductive age women. Variants in LMNA cause partial lipodystrophy, a syndrome with overlapping features to PCOS. OBJECTIVE We tested the hypothesis that rare variation in LMNA contributes to PCOS pathogenesis and selects a lipodystrophy-like subtype of PCOS. METHODS We sequenced LMNA by targeted sequencing a Discovery cohort of 811 PCOS patients and 164 healthy controls. We then analyzed LMNA from whole-exome sequencing of a Replication cohort of 718 PCOS patients and 281 healthy controls. We evaluated variation in the LMNA gene and hormone and lipid profiles of participants. RESULTS In the Discovery cohort, we identified 8 missense variants in 15/811 cases, and 1 variant in 1/172 reproductively healthy controls. There is strong evidence for association between the variants and PCOS compared to gnomAD non-Finnish European population controls (χ2 = 17, P = 3.7 × 10-5, OR = 2.9). In the Replication cohort, we identified 11 unique variants in 15/718 cases, and 1 variant in 281 reproductively healthy controls. Again, there is strong evidence for association with population controls (χ2 = 30.5, P = 3.4 × 10-8, OR = 4.0). In both the Discovery and Replication cohorts, variants in LMNA identify women with PCOS with high triglycerides and extreme insulin resistance. CONCLUSION Rare missense variation in LMNA is reproducibly associated with PCOS and identifies some individuals with lipodystrophy-like features. The overlap between this PCOS phenotype and genetic partial lipodystrophy syndromes warrants further investigation into additional lipodystrophy genes and their potential in PCOS etiology.
Collapse
Affiliation(s)
- Rosemary Bauer
- Division of Endocrinology, Metabolism, and Molecular Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Center for Reproductive Science, Northwestern University, Chicago, IL 60611, USA
| | - Chloe Parker
- Division of Endocrinology, Metabolism, and Molecular Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Lidija K Gorsic
- Division of Endocrinology, Metabolism, and Molecular Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Michael Geoffrey Hayes
- Division of Endocrinology, Metabolism, and Molecular Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Department of Anthropology, Northwestern University, Evanston, IL 60208, USA
| | - Allen R Kunselman
- Public Health Sciences, Pennsylvania State College of Medicine, Hershey, PA 17033, USA
| | - Richard S Legro
- Department of Obstetrics and Gynecology, Pennsylvania State College of Medicine, Hershey, PA 17033, USA
| | - Corrine K Welt
- Division of Endocrinology, Metabolism, and Diabetes, University of Utah, Salt Lake City, UT 84132, USA
| | - Margrit Urbanek
- Division of Endocrinology, Metabolism, and Molecular Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Center for Reproductive Science, Northwestern University, Chicago, IL 60611, USA
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| |
Collapse
|
5
|
Kort EJ, Sayed N, Liu C, Mondéjar-Parreño G, Forsberg J, Eugster E, Wu SM, Wu JC, Jovinge S. Olmesartan Restores LMNA Function in Haploinsufficient Cardiomyocytes. Circulation 2025; 151:1436-1448. [PMID: 40166828 PMCID: PMC12084018 DOI: 10.1161/circulationaha.121.058621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 02/17/2025] [Indexed: 04/02/2025]
Abstract
BACKGROUND Gene mutations are responsible for a sizeable proportion of cases of heart failure. However, the number of patients with any specific mutation is small. Repositioning of existing US Food and Drug Administration-approved compounds to target specific mutations is a promising approach to efficient identification of new therapies for these patients. METHODS The National Institutes of Health Library of Integrated Network-Based Cellular Signatures database was interrogated to identify US Food and Drug Administration-approved compounds that demonstrated the ability to reverse the transcriptional effects of LMNA knockdown. Top hits from this screening were validated in vitro with patient-specific induced pluripotent stem cell-derived cardiomyocytes combined with force measurement, gene expression profiling, electrophysiology, and protein expression analysis. RESULTS Several angiotensin receptor blockers were identified from our in silico screen. Of these, olmesartan significantly elevated the expression of sarcomeric genes and rate and force of contraction and ameliorated arrhythmogenic potential. In addition, olmesartan exhibited the ability to reduce phosphorylation of extracellular signal-regulated kinase 1 in LMNA-mutant induced pluripotent stem cell-derived cardiomyocytes. CONCLUSIONS In silico screening followed by in vitro validation with induced pluripotent stem cell-derived models can be an efficient approach to identifying repositionable therapies for monogenic cardiomyopathies.
Collapse
Affiliation(s)
- Eric J. Kort
- DeVos Cardiovascular Research Program, Fredrik Meijer Heart and Vascular Institute, Spectrum Health and Van Andel Institute, Grand Rapids, MI (E.J.K., J.F., E.E., S.J.)
- Department of Pediatrics and Human Development, Michigan State University, Grand Rapids (E.J.K.)
- Helen DeVos Children’s Hospital, Corewell Health, Grand Rapids, MI (E.J.K.)
| | - Nazish Sayed
- Cardiovascular Institute, Stanford University, CA (N.S., C.L., G.M.-P., S.M.W., J.C.W., S.J.)
- Division of Vascular Surgery, Department of Surgery, Stanford University, CA (N.S.)
| | - Chun Liu
- Cardiovascular Institute, Stanford University, CA (N.S., C.L., G.M.-P., S.M.W., J.C.W., S.J.)
| | - Gema Mondéjar-Parreño
- Cardiovascular Institute, Stanford University, CA (N.S., C.L., G.M.-P., S.M.W., J.C.W., S.J.)
| | - Jens Forsberg
- DeVos Cardiovascular Research Program, Fredrik Meijer Heart and Vascular Institute, Spectrum Health and Van Andel Institute, Grand Rapids, MI (E.J.K., J.F., E.E., S.J.)
| | - Emily Eugster
- DeVos Cardiovascular Research Program, Fredrik Meijer Heart and Vascular Institute, Spectrum Health and Van Andel Institute, Grand Rapids, MI (E.J.K., J.F., E.E., S.J.)
| | - Sean M. Wu
- Cardiovascular Institute, Stanford University, CA (N.S., C.L., G.M.-P., S.M.W., J.C.W., S.J.)
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University, CA (S.M.W., J.C.W.)
| | - Joseph C. Wu
- Cardiovascular Institute, Stanford University, CA (N.S., C.L., G.M.-P., S.M.W., J.C.W., S.J.)
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University, CA (S.M.W., J.C.W.)
- Department of Radiology, Stanford University, CA (J.C.W.)
| | - Stefan Jovinge
- DeVos Cardiovascular Research Program, Fredrik Meijer Heart and Vascular Institute, Spectrum Health and Van Andel Institute, Grand Rapids, MI (E.J.K., J.F., E.E., S.J.)
- Cardiovascular Institute, Stanford University, CA (N.S., C.L., G.M.-P., S.M.W., J.C.W., S.J.)
- Skåne University Hospital, Lund University, Lund, Sweden (S.J.)
| |
Collapse
|
6
|
Mahmoud AI, Sadek HA. Leveraging Transcriptional Readouts as a Platform for Drug Repurposing in Cardiomyopathy. Circulation 2025; 151:1449-1450. [PMID: 40388510 DOI: 10.1161/circulationaha.125.074556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/21/2025]
Affiliation(s)
- Ahmed I Mahmoud
- Center for Cardiovascular and Muscular Diseases, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA (A.I.M.)
| | - Hesham A Sadek
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain (H.A.S.)
- Division of Cardiology, University of Arizona College of Medicine, Tucson (H.A.S.)
- University of Arizona Sarver Heart Center, Tucson (H.A.S.)
| |
Collapse
|
7
|
Rouhi L. Cardiac phenotypes in LMNA mutations. Curr Opin Cardiol 2025; 40:131-138. [PMID: 39998502 PMCID: PMC11968229 DOI: 10.1097/hco.0000000000001209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/26/2025]
Abstract
PURPOSE OF REVIEW This review highlights the diverse cardiac manifestations of LMNA mutations, focusing on their underlying molecular mechanisms and clinical implications. As LMNA mutations are implicated in cardiomyopathies, such as dilated cardiomyopathy (DCM), arrhythmogenic cardiomyopathy (ARVC), and conduction system diseases, understanding these phenotypes is critical for advancing diagnosis and management strategies. RECENT FINDINGS Recent studies reveal that LMNA mutations disrupt nuclear envelope stability, activating the DNA damage response (DDR) and compromising chromatin organization and mechanotransduction. Mouse models have elucidated pathways linking LMNA dysfunction to fibrosis, arrhythmias, and myocardial remodeling. Emerging evidence demonstrates that fibroblasts play a crucial role in cardiac phenotypes. Advances in genetic screening have also underscored the importance of early identification and risk stratification, particularly for arrhythmias and sudden cardiac death. SUMMARY The diverse spectrum of LMNA-related cardiac phenotypes, from isolated conduction defects to severe DCM and ARVC, underscores the necessity of personalized care strategies. Bridging insights from molecular studies and clinical research paves the way for targeted therapies to slow disease progression and improve patient outcomes. Future efforts should prioritize translational research on molecular mechanisms with potential in mouse models, alongside a deeper exploration of genotype-phenotype correlations, to refine and implement effective therapeutic interventions.
Collapse
Affiliation(s)
- Leila Rouhi
- Center for Cardiovascular Genetics, Institute of Molecular Medicine and Department of Medicine, University of Texas Health Sciences Center at Houston, Houston, Texas, USA
| |
Collapse
|
8
|
Mohar NP, Langland CJ, Darr Z, Viles J, Moore SA, Darbro BW, Wallrath LL. A genetic variant in SMAD7 acts as a modifier of LMNA-associated muscular dystrophy, implicating SMAD signaling as a therapeutic target. SCIENCE ADVANCES 2025; 11:eads7903. [PMID: 40249815 PMCID: PMC12007578 DOI: 10.1126/sciadv.ads7903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 03/12/2025] [Indexed: 04/20/2025]
Abstract
Mutations in LMNA cause multiple types of muscular dystrophy (LMNA-MD). The symptoms of LMNA-MD are highly variable and sensitive to genetic background. To identify genetic contributions to this phenotypic variability, we performed whole-genome sequencing on four siblings possessing the same LMNA mutation with differing degrees of skeletal muscle disease severity. We identified a variant in SMAD7 that segregated with severe muscle disease. To functionally test the SMAD7 variant, we generated a Drosophila model possessing the LMNA mutation and the SMAD7 variant in the orthologous fly genes. The SMAD7 variant increased SMAD signaling and enhanced muscle defects caused by the mutant lamin. Conversely, overexpression of wild-type SMAD7 rescued muscle function. These findings were extended to humans by showing that SMAD signaling is increased in muscle biopsy tissue from individuals with LMNA-MD compared to age-matched controls. Collectively, our findings support SMAD7 as the first functionally tested genetic modifier for LMNA-MD and suggest components of the SMAD pathway as therapeutic targets.
Collapse
Affiliation(s)
- Nathaniel P. Mohar
- Interdisciplinary Graduate Program in Genetics, University of Iowa, Iowa City, IA 52242, USA
- Department of Biochemistry and Molecular Biology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Christopher J. Langland
- Department of Biochemistry and Molecular Biology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Zachary Darr
- Department of Biochemistry and Molecular Biology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Jill Viles
- Independent researcher, Gowrie, Iowa, USA
| | - Steven A. Moore
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Benjamin W. Darbro
- Interdisciplinary Graduate Program in Genetics, University of Iowa, Iowa City, IA 52242, USA
- Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Lori L. Wallrath
- Interdisciplinary Graduate Program in Genetics, University of Iowa, Iowa City, IA 52242, USA
- Department of Biochemistry and Molecular Biology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
9
|
Wang X, Lang Z, Yan Z, Xu J, Zhang J, Jiao L, Zhang H. Dilated cardiomyopathy: from genes and molecules to potential treatments. Mol Cell Biochem 2025:10.1007/s11010-025-05269-0. [PMID: 40155570 DOI: 10.1007/s11010-025-05269-0] [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: 01/17/2025] [Accepted: 03/22/2025] [Indexed: 04/01/2025]
Abstract
Dilated cardiomyopathy is a myocardial condition marked by the enlargement of the heart's ventricular chambers and the gradual decline in systolic function, frequently resulting in congestive heart failure. Dilated cardiomyopathy has obvious familial characteristics, and mutations in related pathogenic genes can account for about 50% of patients with dilated cardiomyopathy. The most common genes related to dilated cardiomyopathy include TTN, LMNA, MYH7, etc. With more and more research on these genes, it will undoubtedly provide more potential targets and therapeutic pathways for the treatment of dilated cardiomyopathy. In addition, myocardial inflammation, myocardial metabolism abnormalities and cardiomyocyte apoptosis all have an important impact on the pathogenesis of dilated cardiomyopathy. Approximately half of sudden deaths among children and adolescents, along with the majority of patients undergoing heart transplantation, stem from cardiomyopathy. Therefore, precise and prompt clinical diagnosis holds paramount importance. Currently, diagnosis primarily hinges on the patient's medical background and imaging tests, with the significance of genetic testing steadily gaining prominence. The primary treatment for dilated cardiomyopathy remains heart transplantation. However, the scarcity of donors and the risk of severe immune rejection underscore the pressing need for novel therapies. Presently, research is actively exploring preclinical treatments like stem cell therapy as potential solutions.
Collapse
Affiliation(s)
- Xiumei Wang
- Department of Anesthesiology and Operating Theater, The First Hospital of Lanzhou University, Lanzhou, 730000, The People's Republic of China
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, Gansu, The People's Republic of China
| | - Zekun Lang
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, Gansu, The People's Republic of China
| | - Zeyi Yan
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, Gansu, The People's Republic of China
| | - Jing Xu
- The Second Clinical Medical College of Lanzhou University, Lanzhou, 730000, Gansu, The People's Republic of China
| | - Jinyuan Zhang
- The Second Clinical Medical College of Lanzhou University, Lanzhou, 730000, Gansu, The People's Republic of China
| | - Lianhang Jiao
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, Gansu, The People's Republic of China
| | - Haijun Zhang
- Department of Anesthesiology and Operating Theater, The First Hospital of Lanzhou University, Lanzhou, 730000, The People's Republic of China.
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, Gansu, The People's Republic of China.
| |
Collapse
|
10
|
Bahl A, Seth S, Dhandapany PS, Mittal A, Chockalingam P, Ahamed H, Subramanian M, Nampoothiri S, Namboodiri N, Das S, Vaidya V, Anantharaman R, Khullar M, Rani DS, Thangaraj K, Naik N, Sivasubbu S, Roy D, Bang VH, Banerjee PS, Chandra Rath P, Sinha DP, Yadav R, Dastidar DG. Genetic testing of cardiomyopathies: Position statement of the Cardiological Society of India. Indian Heart J 2025:S0019-4832(25)00059-8. [PMID: 40157570 DOI: 10.1016/j.ihj.2025.03.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 06/22/2024] [Accepted: 03/25/2025] [Indexed: 04/01/2025] Open
Affiliation(s)
- Ajay Bahl
- Department of Cardiology, Postgraduate Institute of Medical Education and Research, Chandigarh, India.
| | - Sandeep Seth
- Department of Cardiology, All India Institute of Medical Sciences, New Delhi, India
| | | | - Anupam Mittal
- Department of Translational and Regenerative Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Priya Chockalingam
- Centre for Inherited Heart Diseases, Department of Cardiology, Kauvery Hospital, Chennai, India
| | - Hisham Ahamed
- Department of Cardiology, Amrita Institute of Medical Sciences and Research, Kochi, India
| | | | - Sheela Nampoothiri
- Department of Pediatric Genetics, Amrita Institute of Medical Sciences & Research Centre, Kochi, Kerala, India
| | - Narayanan Namboodiri
- Department of Cardiology, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram, India
| | - Soumi Das
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Vanya Vaidya
- Department of Cardiology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Rajaram Anantharaman
- Centre for Inherited Heart Diseases, Department of Cardiology, Kauvery Hospital, Chennai, India
| | - Madhu Khullar
- Department of Experimental Medicine and Biotechnology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Deepa Selvi Rani
- CSIR-Centre for Cellular and Molecular Biology, Hyderabad, India
| | | | - Nitish Naik
- Department of Cardiology, All India Institute of Medical Sciences, New Delhi, India
| | | | - Debabrata Roy
- Department of Cardiology, Rabindranath Tagore International Institute of Cardiac Sciences, Kolkata, India
| | | | - Partha Sarathi Banerjee
- Manipal Hospital, Kolkata, India; Formerly Department of Cardiology, Medical College, Kolkata, India
| | | | | | - Rakesh Yadav
- Department of Cardiology, All India Institute of Medical Sciences, New Delhi, India
| | | |
Collapse
|
11
|
Zheng Q, Liu Y, Guo M, Zhang X, Zhang Q, Yu XY, Lin Z. Discovery of therapeutic targets in cardiovascular diseases using high-throughput chromosome conformation capture (Hi-C). Front Genet 2025; 16:1515010. [PMID: 40182924 PMCID: PMC11966399 DOI: 10.3389/fgene.2025.1515010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 02/17/2025] [Indexed: 04/05/2025] Open
Abstract
Epigenetic changes have been associated with several cardiovascular diseases. In recent years, epigenetic inheritance based on spatial changes has gradually attracted attention. Alterations in three-dimensional chromatin structures have been shown to regulate gene expression and influence disease onset and progression. High-throughput Chromosome Conformation Capture (Hi-C) is a powerful method to detect spatial chromatin conformation changes. Since its development, Hi-C technology has been widely adopted for discovering novel therapeutic targets in cardiovascular research. In this review, we summarize key targets identified by Hi-C in cardiovascular diseases and discuss their potential implications for epigenetic therapy.
Collapse
Affiliation(s)
- Quan Zheng
- School of Pharmacy, Macau University of Science and Technology, Taipa, China
| | - Ying Liu
- School of Pharmacy, Macau University of Science and Technology, Taipa, China
- Department of Pharmacology, School of Pharmacy, Guangzhou Xinhua University, Guangzhou, China
| | - Minghao Guo
- School of Pharmacy, Macau University of Science and Technology, Taipa, China
| | - Xin Zhang
- School of Pharmacy, Macau University of Science and Technology, Taipa, China
| | - Qingbin Zhang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Xi-Yong Yu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Zhongxiao Lin
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
12
|
Lee TM, Ware SM, Kamsheh AM, Bhatnagar S, Absi M, Miller E, Purevjav E, Ryan KA, Towbin JA, Lipshultz SE. Genomics of pediatric cardiomyopathy. Pediatr Res 2025; 97:1381-1392. [PMID: 39922924 PMCID: PMC12106076 DOI: 10.1038/s41390-025-03819-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 11/21/2024] [Accepted: 11/27/2024] [Indexed: 02/10/2025]
Abstract
Cardiomyopathy in children is a leading cause of heart failure and cardiac transplantation. Disease-associated genetic variants play a significant role in the development of the different subtypes of disease. Genetic testing is increasingly being recognized as the standard of care for diagnosing this heterogeneous group of disorders, guiding management, providing prognostic information, and facilitating family-based risk stratification. The increase in clinical and research genetic testing within the field has led to new insights into this group of disorders. Mutations in genes encoding sarcomere, cytoskeletal, Z-disk, and sarcolemma proteins appear to play a major role in causing the overlapping clinical phenotypes called cardioskeletal myopathies through "final common pathway" links. For myocarditis, the high frequency of infectious exposures and wide spectrum of presentation suggest that genetic factors mediate the development and course of the disease, including genetic risk alleles, an association with cardiomyopathy, and undiagnosed arrhythmogenic cardiomyopathy. Finally, while we have made strides in elucidating the genetic architecture of pediatric cardiomyopathy, understanding the clinical implications of variants of uncertain significance remains a major issue. The need for continued genetic innovation in this field remains great, particularly as a basis to drive forward targeted precision medicine and gene therapy efforts. IMPACT: Cardiomyopathy and skeletal myopathy can occur in the same patient secondary to gene mutations that encode for sarcomeric or cytoskeletal proteins, which are expressed in both muscle groups, highlighting that there are common final pathways of disease. The heterogeneous presentation of myocarditis is likely secondary to a complex interaction of multiple environmental and genetic factors, suggesting a utility to genetic testing in pediatric patients with myocarditis, particularly those in higher risk groups. Given the high prevalence of variants of uncertain significance in genetic testing, better bioinformatic tools and pipelines are needed to resolve their clinical meaning.
Collapse
Affiliation(s)
- Teresa M Lee
- Department of Pediatrics, Columbia University Medical Center, New York, NY, USA
| | - Stephanie M Ware
- Departments of Pediatrics and Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Alicia M Kamsheh
- Department of Pediatrics, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Surbhi Bhatnagar
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Mohammed Absi
- Heart Institute, Division of Pediatric Cardiology, Le Bonheur Children's Hospital, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Elyse Miller
- Heart Institute, Division of Pediatric Cardiology, Le Bonheur Children's Hospital, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Enkhsaikhan Purevjav
- Heart Institute, Division of Pediatric Cardiology, Le Bonheur Children's Hospital, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Kaitlin A Ryan
- Heart Institute, Division of Pediatric Cardiology, Le Bonheur Children's Hospital, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Jeffrey A Towbin
- Heart Institute, Division of Pediatric Cardiology, Le Bonheur Children's Hospital, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Steven E Lipshultz
- Department of Pediatrics, University at Buffalo Jacobs School of Medicine and Biomedical Sciences, Clinical and Translational Research Center, Buffalo, NY, USA.
| |
Collapse
|
13
|
Parikh VN, Day SM, Lakdawala NK, Adler ED, Olivotto I, Seidman CE, Ho CY. Advances in the study and treatment of genetic cardiomyopathies. Cell 2025; 188:901-918. [PMID: 39983674 DOI: 10.1016/j.cell.2025.01.011] [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: 03/11/2024] [Revised: 10/21/2024] [Accepted: 01/07/2025] [Indexed: 02/23/2025]
Abstract
Cardiomyopathies are primary disorders of the heart muscle. Three key phenotypes have been defined, based on morphology and arrhythmia burden: hypertrophic cardiomyopathy (HCM), with thickened heart muscle and diastolic dysfunction; dilated cardiomyopathy (DCM), with left ventricular enlargement and systolic dysfunction; and arrhythmogenic cardiomyopathy (ACM), with right, left, or biventricular involvement and arrhythmias out of proportion to systolic dysfunction. Genetic discoveries of the molecular basis of disease are paving the way for greater precision in diagnosis and management and revealing mechanisms that account for distinguishing clinical features. This deeper understanding has propelled the development of new treatments for cardiomyopathies: disease-specific, mechanistically based medicines that counteract pathophysiology, and emergent gene therapies that aim to intercept disease progression and restore cardiac physiology. Together, these discoveries have advanced fundamental insights into cardiac biology and herald a new era for patients with cardiomyopathy.
Collapse
Affiliation(s)
- Victoria N Parikh
- Stanford Center for Inherited Cardiovascular Disease, Stanford School of Medicine, Stanford, CA, USA
| | - Sharlene M Day
- Division of Cardiovascular Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Neal K Lakdawala
- Cardiovascular Division, Brigham and Women's Hospital, Boston, MA, USA
| | - Eric D Adler
- Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla, San Diego, CA, USA
| | | | - Christine E Seidman
- Cardiovascular Division, Brigham and Women's Hospital, Boston, MA, USA; Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Carolyn Y Ho
- Cardiovascular Division, Brigham and Women's Hospital, Boston, MA, USA.
| |
Collapse
|
14
|
Hayes EA, Foreman K, Wright LK, Gajarski RJ, Hor K, Garg V, Cripe LH, Kertesz NJ, Nandi D. Extensive cardiac involvement in laminopathies diagnosed in pediatric-aged patients: A single-center study. Heart Rhythm 2025:S1547-5271(25)00104-3. [PMID: 39894135 DOI: 10.1016/j.hrthm.2025.01.034] [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: 11/04/2024] [Revised: 01/21/2025] [Accepted: 01/27/2025] [Indexed: 02/04/2025]
Abstract
BACKGROUND Pathogenic variations in lamin A/C (LMNA) result in a group of inherited conditions termed laminopathies. Cardiac manifestations of laminopathies include atrial and ventricular arrhythmias, atrioventricular conduction disorders, and cardiomyopathy, with or without skeletal muscle involvement. Because of rarity and previous cardiac characterization as adult onset, pediatric data are limited. OBJECTIVE This study sought to investigate the natural history of cardiac disease in pediatric patients with pathogenic LMNA variants. METHODS We identified patients ≤18 years with genetically confirmed pathogenic variants in LMNA observed at a single center between 2003 and 2024. Clinical phenotypes along with cardiac test results were retrospectively catalogued. RESULTS We identified 12 patients with pathogenic LMNA variant with a median age of 4.9 years at diagnosis (interquartile range, 3.7-10.7 years). Of the 12 patients, 9 (75%) were male and 10 (83%) had skeletal muscle involvement. Cardiac manifestations developed in 9 patients (75%) during a median follow-up of 9.5 years (interquartile range, 7.0-13.3 years). Nine patients (75%) had conduction abnormalities or arrhythmias (atrioventricular block, ventricular/atrial tachycardias), and 4 (33%) had cardiovascular implantable electronic devices placed. Two (17%) patients were diagnosed with cardiomyopathy, 1 (8%) requiring heart transplant. Two (17%) patients died during the study. CONCLUSION Cardiac involvement, specifically cardiomyopathy and progressive conduction system abnormalities, were common in pediatric patients with pathogenic LMNA variants. Early genetic diagnosis of laminopathies with frequent surveillance for arrhythmias and cardiac dysfunction is necessary for more timely initiation of advanced therapies to prevent adverse events. More comprehensive phenotype-genotype correlation is strongly needed to better understand early cardiac manifestations in laminopathies.
Collapse
Affiliation(s)
- Emily A Hayes
- The Heart Center, Nationwide Children's Hospital, Columbus, Ohio.
| | - Kaitlyn Foreman
- The Heart Center, Nationwide Children's Hospital, Columbus, Ohio; Pediatric Cardiology, Sunset Blvd, Los Angeles, CA
| | - Lydia K Wright
- The Heart Center, Nationwide Children's Hospital, Columbus, Ohio
| | | | - Kan Hor
- The Heart Center, Nationwide Children's Hospital, Columbus, Ohio
| | - Vidu Garg
- The Heart Center, Nationwide Children's Hospital, Columbus, Ohio; Department of Pediatrics, Center for Cardiovascular Research, Nationwide Children's Hospital, Columbus, Ohio; Department of Molecular Genetics, The Ohio State University, Columbus, Ohio
| | - Linda H Cripe
- The Heart Center, Nationwide Children's Hospital, Columbus, Ohio
| | - Naomi J Kertesz
- The Heart Center, Nationwide Children's Hospital, Columbus, Ohio
| | - Deipanjan Nandi
- The Heart Center, Nationwide Children's Hospital, Columbus, Ohio
| |
Collapse
|
15
|
Dai Y, Wang Y, Fan Y, Han B. Genotype-phenotype insights of pediatric dilated cardiomyopathy. Front Pediatr 2025; 13:1505830. [PMID: 39959410 PMCID: PMC11825472 DOI: 10.3389/fped.2025.1505830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 01/21/2025] [Indexed: 02/18/2025] Open
Abstract
Dilated cardiomyopathy (DCM) in children is a severe myocardial disease characterized by enlargement of the left ventricle or both ventricles with impaired contractile function. DCM can cause adverse consequences such as heart failure, sudden death, thromboembolism, and arrhythmias. This article reviews the latest advances in genotype and phenotype research in pediatric DCM. With the development of gene sequencing technologies, considerable progress has been made in genetic research on DCM. Research has shown that DCM exhibits notable genetic heterogeneity, with over 100 DCM-related genes identified to date, primarily involving functions such as calcium handling, the cytoskeleton, and ion channels. As human genomic variations are linked to phenotypes, DCM phenotypes are influenced by numerous genetic variations across the entire genome. Children with DCM display high genetic heterogeneity and are characterized by early onset, rapid disease progression, and poor prognosis. The genetic architecture of pediatric DCM markedly differs from that of adult DCM, necessitating analyses through clinical phenotyping, familial cosegregation studies, and functional validation. Clarifying the genotype-phenotype relationship can improve diagnostic accuracy, enhance prognosis, and guide follow-up treatment for genotype-positive and phenotype-negative patients identified through genetic testing, providing new insights for precision medicine. Future research should further explore novel pathogenic genes and mutations and strengthen genotype-phenotype correlation analyses to facilitate precise diagnosis and treatment of DCM in children.
Collapse
Affiliation(s)
| | | | - Youfei Fan
- Department of Pediatrics, Shandong Province Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Bo Han
- Department of Pediatrics, Shandong Province Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
16
|
Balakrishnan ID, Lakdawala NK. Contemporary Insights into LMNA Cardiomyopathy. Curr Cardiol Rep 2025; 27:40. [PMID: 39869235 DOI: 10.1007/s11886-025-02195-x] [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] [Accepted: 01/07/2025] [Indexed: 01/28/2025]
Abstract
PURPOSE OF REVIEW This review aims to explore how a diagnosis of LMNA-related cardiomyopathy (LMNA-CM) informs clinical management, focusing on the prevention and management of its complications, through practical clinical strategies. RECENT FINDINGS Longitudinal studies have enhanced our understanding of the natural history of LMNA-CM including its arrhythmic and non-arrhythmic complications. A LMNA specific ventricular arrhythmia risk prediction strategy has been integrated into clinical practice guidelines. Although less robust, observational studies are shaping gene-specific strategies for mitigating other complications including atrioventricular block, atrial fibrillation and cardiomyopathy, while novel therapies have been evaluated in clinical trials. LMNA-CM follows an aggressive yet generally stereotyped course. Early recognition of anticipated complications allows for more effective prevention and management in both symptomatic and asymptomatic patients.
Collapse
Affiliation(s)
- Iswaree D Balakrishnan
- Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Cardiology, National Heart Centre Singapore, Singapore, Singapore
| | - Neal K Lakdawala
- Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
17
|
Wang Z, Wu J, Lv Z, Liang P, Li Q, Li Y, Guo Y. LMNA-related cardiomyopathy: From molecular pathology to cardiac gene therapy. J Adv Res 2025:S2090-1232(25)00001-3. [PMID: 39827909 DOI: 10.1016/j.jare.2025.01.001] [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: 11/19/2024] [Revised: 12/29/2024] [Accepted: 01/01/2025] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND The genetic variants of LMNA cause an array of diseases that often affect the heart. LMNA-related cardiomyopathy exhibits high-penetrance and early-onset phenotypes that lead to late-stage heart failure or lethal arrhythmia. As a subtype of dilated cardiomyopathy and arrhythmogenic cardiomyopathy, LMNA-related cardiac dysfunction is resistant to existing cardiac therapeutic strategies, leaving a major unmet clinical need in cardiomyopathy management. AIM OF REVIEW Here we comprehensively summarize current knowledge about the genetic basis, disease models and pathological mechanisms of LMNA-related cardiomyopathy. Recent translational studies were highlighted to indicate new therapeutic modalities such as gene supplementation, gene silencing and genome editing therapy, which offer potential opportunities to overcome the difficulties in the development of specific drugs for this disease. KEY SCIENTIFIC CONCEPTS OF REVIEW LMNA-related cardiomyopathy involves many diverse disease mechanisms that preclude small-molecule drugs that target only a small fraction of the mechanisms. Agreeing to this notion, the first-in-human clinical trial for this disease recently reported futility. By contrast, gene therapy offers the new hope to directly intervene LMNA variants and demonstrates a tremendous potential for breakthrough therapy for this disease. Concepts in this review are also applicable to studies of other genetic diseases that lack effective therapeutics.
Collapse
Affiliation(s)
- Ze Wang
- School of Basic Medical Sciences, Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China
| | - Jiahao Wu
- Ministry of Education Key Laboratory of Birth Defects and Related Diseases of Women and Children, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Zhengyuan Lv
- School of Basic Medical Sciences, Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China
| | - Ping Liang
- Institute of Translational Medicine, Zhejiang University, Hangzhou 310029, China.
| | - Qirui Li
- Department of Cardiology, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China.
| | - Yifei Li
- Ministry of Education Key Laboratory of Birth Defects and Related Diseases of Women and Children, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China.
| | - Yuxuan Guo
- School of Basic Medical Sciences, Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China; Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China.
| |
Collapse
|
18
|
Wu S, Cai Y, Zhang L, Li X, Liu X, Zhou G, Luo H, Li R, Huo Y, Zhang Z, Chen S, Huang J, Shi J, Ding S, Sun Z, Zhou Z, Wang P, Wang G. Noncoding RNA Terc-53 and hyaluronan receptor Hmmr regulate aging in mice. Protein Cell 2025; 16:28-48. [PMID: 38721690 DOI: 10.1093/procel/pwae023] [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: 01/14/2024] [Accepted: 04/11/2024] [Indexed: 01/07/2025] Open
Abstract
One of the basic questions in the aging field is whether there is a fundamental difference between the aging of lower invertebrates and mammals. A major difference between the lower invertebrates and mammals is the abundancy of noncoding RNAs, most of which are not conserved. We have previously identified a noncoding RNA Terc-53 that is derived from the RNA component of telomerase Terc. To study its physiological functions, we generated two transgenic mouse models overexpressing the RNA in wild-type and early-aging Terc-/- backgrounds. Terc-53 mice showed age-related cognition decline and shortened life span, even though no developmental defects or physiological abnormality at an early age was observed, indicating its involvement in normal aging of mammals. Subsequent mechanistic study identified hyaluronan-mediated motility receptor (Hmmr) as the main effector of Terc-53. Terc-53 mediates the degradation of Hmmr, leading to an increase of inflammation in the affected tissues, accelerating organismal aging. adeno-associated virus delivered supplementation of Hmmr in the hippocampus reversed the cognition decline in Terc-53 transgenic mice. Neither Terc-53 nor Hmmr has homologs in C. elegans. Neither do arthropods express hyaluronan. These findings demonstrate the complexity of aging in mammals and open new paths for exploring noncoding RNA and Hmmr as means of treating age-related physical debilities and improving healthspan.
Collapse
Affiliation(s)
- Sipeng Wu
- State Key Laboratory for Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Yiqi Cai
- State Key Laboratory for Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Lixiao Zhang
- State Key Laboratory for Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Xiang Li
- State Key Laboratory for Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Xu Liu
- State Key Laboratory for Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Guangkeng Zhou
- State Key Laboratory for Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Hongdi Luo
- State Key Laboratory for Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Renjian Li
- State Key Laboratory for Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Yujia Huo
- State Key Laboratory for Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Zhirong Zhang
- State Key Laboratory for Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Siyi Chen
- State Key Laboratory for Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Jinliang Huang
- School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Jiahao Shi
- State Key Laboratory for Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Shanwei Ding
- State Key Laboratory for Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Zhe Sun
- State Key Laboratory for Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Zizhuo Zhou
- State Key Laboratory for Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Pengcheng Wang
- State Key Laboratory for Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Geng Wang
- State Key Laboratory for Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen 361102, China
| |
Collapse
|
19
|
Chen Y, Liang R, Li Y, Jiang L, Ma D, Luo Q, Song G. Chromatin accessibility: biological functions, molecular mechanisms and therapeutic application. Signal Transduct Target Ther 2024; 9:340. [PMID: 39627201 PMCID: PMC11615378 DOI: 10.1038/s41392-024-02030-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 08/04/2024] [Accepted: 10/17/2024] [Indexed: 12/06/2024] Open
Abstract
The dynamic regulation of chromatin accessibility is one of the prominent characteristics of eukaryotic genome. The inaccessible regions are mainly located in heterochromatin, which is multilevel compressed and access restricted. The remaining accessible loci are generally located in the euchromatin, which have less nucleosome occupancy and higher regulatory activity. The opening of chromatin is the most important prerequisite for DNA transcription, replication, and damage repair, which is regulated by genetic, epigenetic, environmental, and other factors, playing a vital role in multiple biological progresses. Currently, based on the susceptibility difference of occupied or free DNA to enzymatic cleavage, solubility, methylation, and transposition, there are many methods to detect chromatin accessibility both in bulk and single-cell level. Through combining with high-throughput sequencing, the genome-wide chromatin accessibility landscape of many tissues and cells types also have been constructed. The chromatin accessibility feature is distinct in different tissues and biological states. Research on the regulation network of chromatin accessibility is crucial for uncovering the secret of various biological processes. In this review, we comprehensively introduced the major functions and mechanisms of chromatin accessibility variation in different physiological and pathological processes, meanwhile, the targeted therapies based on chromatin dynamics regulation are also summarized.
Collapse
Affiliation(s)
- Yang Chen
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, PR China
| | - Rui Liang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, PR China
| | - Yong Li
- Hepatobiliary Pancreatic Surgery, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, PR China
| | - Lingli Jiang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, PR China
| | - Di Ma
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, PR China
| | - Qing Luo
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, PR China
| | - Guanbin Song
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, PR China.
| |
Collapse
|
20
|
Conte G. Increased pacemaker implantation and mortality rates in relatives of patients with early-onset sinus node dysfunction: can genetics explain all? Europace 2024; 26:euae289. [PMID: 39533833 PMCID: PMC11630514 DOI: 10.1093/europace/euae289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 11/10/2024] [Indexed: 11/16/2024] Open
Affiliation(s)
- Giulio Conte
- Division of Cardiology, Cardiocentro Ticino Institute, Ente Ospedaliero Cantonale, 6900 Lugano, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana (USI), 6900 Lugano, Switzerland
| |
Collapse
|
21
|
Zhao J, Zhang H, Pan C, He Q, Zheng K, Tang Y. Advances in research on the relationship between the LMNA gene and human diseases (Review). Mol Med Rep 2024; 30:236. [PMID: 39422026 PMCID: PMC11529173 DOI: 10.3892/mmr.2024.13358] [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: 06/18/2024] [Accepted: 08/29/2024] [Indexed: 10/19/2024] Open
Abstract
The LMNA gene, which is responsible for encoding lamin A/C proteins, is recognized as a primary constituent of the nuclear lamina. This protein serves crucial roles in various cellular physiological activities, including the maintenance of cellular structural stability, regulation of gene expression, mechanosensing and cellular motility. A significant association has been established between the LMNA gene and several major human diseases. Mutations, dysregulated expression of the LMNA gene, and improper processing of its encoded protein can result in a spectrum of pathological conditions. These diseases, collectively termed laminopathies, are directly attributed to LMNA gene dysfunction. The present review examines the recent advancements in research concerning the LMNA gene and its association with human diseases, while exploring its pathological roles. Particular emphasis is placed on the current status of LMNA gene research in the context of tumors. This includes an analysis of the abundance of LMNA alterations in cancer and its interplay with various signaling pathways. The aim of the present review was to provide novel perspectives for studying the development of LMNA‑related diseases and additional theoretical insights for basic and clinical translational research in this field.
Collapse
Affiliation(s)
- Jiumei Zhao
- Department of Laboratory, Chongqing Nanchuan District People's Hospital, Chongqing Medical University, Chongqing 408400, P.R. China
| | - Huijuan Zhang
- Forensic Science Centre, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Chenglong Pan
- Department of Pathology, The First Affiliated Hospital of Kunming Medical University, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Qian He
- School of Biomedical Engineering, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Kepu Zheng
- Forensic Science Centre, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Yu Tang
- Department of Pathology, The Third Affiliated Hospital of Kunming Medical University, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| |
Collapse
|
22
|
Garcia‐Pavia P, Lakdawala NK, Sinagra G, Ripoll‐Vera T, Afshar K, Priori SG, Ware JS, Owens A, Li H, Angeli FS, Elliott P, MacRae CA, Judge DP. Characterization and natural history of patients with LMNA-related dilated cardiomyopathy in the phase 3 REALM-DCM trial. ESC Heart Fail 2024; 11:4201-4208. [PMID: 39145700 PMCID: PMC11631308 DOI: 10.1002/ehf2.14955] [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: 10/20/2023] [Revised: 05/07/2024] [Accepted: 06/23/2024] [Indexed: 08/16/2024] Open
Abstract
AIMS LMNA-related dilated cardiomyopathy (DCM) is a rare disease with an incompletely defined phenotype. The phase 3 REALM-DCM trial evaluated a potential disease-modifying therapy for LMNA-related DCM but was terminated due to futility without safety concern. This study utilized pooled data from REALM-DCM to descriptively characterize the phenotype and progression of LMNA-related DCM in a contemporary cohort of patients using common heart failure (HF) measures. METHODS REALM-DCM enrolled patients with stable LMNA-related DCM, an implanted cardioverter defibrillator or cardiac resynchronization therapy defibrillator, and New York Heart Association (NYHA) Class II/III HF symptoms. RESULTS Between 2018 and 2022, 77 patients took part in REALM-DCM. The median patient age was 53 years (range: 23-72), and 57% were male. Overall, 88% of patients had a pathogenic or likely pathogenic LMNA variant, and 12% had a variant of uncertain significance with a concordant phenotype. Among patients with confirmed sequencing, 55% had a missense variant. Atrial fibrillation was present in 60% of patients; 79% of all patients had NYHA Class II and 21% had NYHA Class III HF symptoms at baseline. Median (range) left ventricular ejection fraction (LVEF), 6 min walk test (6MWT) distance, Kansas City Cardiomyopathy Questionnaire Overall Summary (KCCQ-OS) score and N-terminal pro-B-type natriuretic peptide (NT-proBNP) concentration at baseline were 42% (23-62), 403 m (173-481), 67 (18-97) and 866 pg/mL (57-5248), respectively. LVEF, 6MWT distance and KCCQ-OS score were numerically lower in patients who had NYHA Class III versus II symptoms at baseline (LVEF: 38% vs. 43%; 6MWT distance: 326 vs. 413 m; and KCCQ-OS score: 43 vs. 70), whereas NT-proBNP concentration was higher (1216 vs. 799 pg/mL). Median follow-up was 73 weeks (range: 0.4-218; 73 in NYHA Class II and 75 in NYHA Class III). Patients displayed variable change from baseline in 6MWT, KCCQ-OS and NT-proBNP values during follow-up. Overall, 25% of patients experienced ventricular tachycardia, and 8% had ventricular fibrillation. Ten (13%) patients met the composite endpoint of worsening HF (adjudicated HF-related hospitalization or urgent care visit) or all-cause death; six had NYHA Class II and four had NYHA Class III at baseline. All-cause mortality occurred in 6 (8%) patients; three had NYHA Class II and three had NYHA Class III symptoms at baseline. CONCLUSIONS Findings confirm the significant morbidity and mortality associated with LMNA-related DCM despite the standard of care management. Typical measures of HF, including 6MWT distance, KCCQ-OS score and NT-proBNP concentration, were variable but correlated with NYHA class. An unmet treatment need remains among patients with LMNA-related DCM. NCT03439514.
Collapse
Affiliation(s)
- Pablo Garcia‐Pavia
- Hospital Universitario Puerta de Hierro MajadahondaMadridSpain
- Instituto de Investigación Sanitaria Puerta de Hierro‐Segovia de Arana (IDIPHISA)MadridSpain
- Centro de Investigación en Red en Enfermedades Cardiovasculares (CIBERCV)MadridSpain
- Universidad Francisco de Vitoria (UFV)MadridSpain
- Centro Nacional de Investigaciones Cardiovasculares (CNIC)MadridSpain
| | - Neal K. Lakdawala
- Brigham and Women's HospitalBostonMassachusettsUSA
- Harvard Medical SchoolBostonMassachusettsUSA
| | - Gianfranco Sinagra
- Cardiothoracovascular Department, Azienda Sanitaria Universitaria Giuliano Isontina (ASUGI)University of TriesteTriesteItaly
| | - Tomas Ripoll‐Vera
- Hospital Universitario Son LlatzerMallorcaSpain
- Health Research Institute of the Balearic Islands (IdISBa)MallorcaSpain
- Department of MedicineUniversity of the Balearic IslandsMallorcaSpain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBERobn)Institute of Health Carlos IIIMadridSpain
| | - Kia Afshar
- Intermountain Heart InstituteSalt Lake CityUtahUSA
| | - Silvia G. Priori
- Centro Nacional de Investigaciones Cardiovasculares (CNIC)MadridSpain
- IRCCS Istituti Clinici Scientifici Maugeri SpA SB of PaviaPaviaItaly
- Department of Molecular MedicineUniversity of PaviaPaviaItaly
| | - James S. Ware
- National Heart & Lung Institute and MRC London Institute of Medical SciencesImperial College LondonLondonUK
- Royal Brompton & Harefield Hospitals, Guy's and St Thomas' NHS Foundation TrustLondonUK
| | - Anjali Owens
- University of Pennsylvania Heart and Vascular CenterPhiladelphiaPennsylvaniaUSA
| | - Huihua Li
- Pfizer Inc.CollegevillePennsylvaniaUSA
| | | | | | - Calum A. MacRae
- Brigham and Women's HospitalBostonMassachusettsUSA
- Harvard Medical SchoolBostonMassachusettsUSA
| | - Daniel P. Judge
- Cardiovascular GeneticsMedical University of South CarolinaCharlestonSouth CarolinaUSA
| |
Collapse
|
23
|
Gonçalves F, Duarte D, Reis F, Vaz A, Gomes A. Much More Than a Stroke: Emery-Dreifuss Muscular Dystrophy Type 2 Revealed by Ischemic Stroke. Cureus 2024; 16:e73815. [PMID: 39687831 PMCID: PMC11648976 DOI: 10.7759/cureus.73815] [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] [Accepted: 11/16/2024] [Indexed: 12/18/2024] Open
Abstract
Emery-Dreifuss muscular dystrophy type 2 (EDMD2) is a rare autosomal dominant neuromuscular disorder caused by LMNA gene mutations and characterized by progressive skeletal muscle weakness and significant cardiac involvement. We report the case of a 45-year-old woman who presented with sudden-onset, left-sided hemiparesis and dysarthria. Initial imaging was unremarkable, and symptoms transiently improved, suggesting a transient ischemic attack. However recurrent deficits led to the identification of right middle cerebral artery occlusion and new-onset atrial fibrillation. Mechanical thrombectomy was successfully performed. Subsequent cardiac evaluation revealed dilated cardiomyopathy with moderately depressed systolic function and segmental wall motion abnormalities, although coronary arteries were normal. Cardiac magnetic resonance imaging demonstrated myocardial fibrosis with late gadolinium enhancement in the subendocardial and mid-wall regions, suggestive of genetic cardiomyopathy. A neurological examination noted lordotic posture, waddling gait, positive Gowers' sign, generalized proximal muscle atrophy, and flaccid hyporeflexic tetraparesis. Electromyography confirmed a proximal myopathy. The patient reported limb weakness since her twenties. Family history was significant for similar neuromuscular and cardiac symptoms. Genetic testing identified a heterozygous LMNA missense variant (ClinVar ID: 804298), confirming the diagnosis of EDMD2. Despite the implantation of a cardiac resynchronization therapy defibrillator and optimal medical management, she experienced recurrent ventricular tachyarrhythmias, necessitating listing for heart transplantation. This case highlights the diagnostic challenges of EDMD2, particularly when the initial presentation is ischemic stroke, a rare manifestation of this genetic myopathy. It underscores the importance of a multidisciplinary approach for early diagnosis and management to improve the outcomes of this rare but impactful disorder.
Collapse
Affiliation(s)
- Francisco Gonçalves
- Department of Internal Medicine, Centro Hospitalar Tondela-Viseu, Viseu, PRT
| | - Daniela Duarte
- Department of Internal Medicine, Centro Hospitalar Tondela-Viseu, Viseu, PRT
| | - Filipa Reis
- Department of Internal Medicine, Centro Hospitalar Tondela-Viseu, Viseu, PRT
| | - Alexandra Vaz
- Department of Internal Medicine, Centro Hospitalar Tondela-Viseu, Viseu, PRT
| | - Ana Gomes
- Stroke Unit, Centro Hospitalar Tondela-Viseu, Viseu, PRT
| |
Collapse
|
24
|
Newman NA, Burke MA. Dilated Cardiomyopathy: A Genetic Journey from Past to Future. Int J Mol Sci 2024; 25:11460. [PMID: 39519012 PMCID: PMC11546582 DOI: 10.3390/ijms252111460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 10/21/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024] Open
Abstract
Dilated cardiomyopathy (DCM) is characterized by reduced systolic function and cardiac dilation. Cases without an identified secondary cause are classified as idiopathic dilated cardiomyopathy (IDC). Over the last 35 years, many cases of IDC have increasingly been recognized to be genetic in etiology with a core set of definitively causal genes in up to 40% of cases. While over 200 genes have been associated with DCM, the evidence supporting pathogenicity for most remains limited. Further, rapid advances in sequencing and bioinformatics have recently revealed a complex genetic spectrum ranging from monogenic to polygenic in DCM. These advances have also led to the discovery of causal and modifier genetic variants in secondary forms of DCM (e.g., alcohol-induced cardiomyopathy). Current guidelines recommend genetic counseling and screening, as well as endorsing a handful of genotype-specific therapies (e.g., device placement in LMNA cardiomyopathy). The future of genetics in DCM will likely involve polygenic risk scores, direct-to-consumer testing, and pharmacogenetics, requiring providers to have a thorough understanding of this rapidly developing field. Herein we outline three decades of genetics in DCM, summarize recent advances, and project possible future avenues for the field.
Collapse
Affiliation(s)
- Noah A. Newman
- Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Michael A. Burke
- Division of Cardiology, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
25
|
Wang X, Ma L, Lu D, Zhao G, Ren H, Lin Q, Jia M, Huang F, Wang S, Xu Z, Yang Z, Chu Y, Xu Z, Li W, Yu L, Jiang Q, Zhang C. Nuclear envelope budding inhibition slows down progerin-induced aging process. Proc Natl Acad Sci U S A 2024; 121:e2321378121. [PMID: 39352925 PMCID: PMC11474064 DOI: 10.1073/pnas.2321378121] [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: 12/11/2023] [Accepted: 08/08/2024] [Indexed: 10/04/2024] Open
Abstract
Progerin causes Hutchinson-Gilford progeria syndrome (HGPS), but how progerin accelerates aging is still an interesting question. Here, we provide evidence linking nuclear envelope (NE) budding and accelerated aging. Mechanistically, progerin disrupts nuclear lamina to induce NE budding in concert with lamin A/C, resulting in transport of chromatin into the cytoplasm where it is removed via autophagy, whereas emerin antagonizes this process. Primary cells from both HGPS patients and mouse models express progerin and display NE budding and chromatin loss, and ectopically expressing progerin in cells can mimic this process. More excitingly, we screen a NE budding inhibitor chaetocin by high-throughput screening, which can dramatically sequester progerin from the NE and prevent this NE budding through sustaining ERK1/2 activation. Chaetocin alleviates NE budding-induced chromatin loss and ameliorates HGPS defects in cells and mice and significantly extends lifespan of HGPS mice. Collectively, we propose that progerin-induced NE budding participates in the induction of progeria, highlight the roles of chaetocin and sustained ERK1/2 activation in anti-aging, and provide a distinct avenue for treating HGPS.
Collapse
Affiliation(s)
- Xiangyang Wang
- The Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, College of Life Sciences, Peking University, Beijing100871, China
- The Academy for Cell and Life Health, Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming650500, China
| | - Lin Ma
- Department of Dermatology, Beijing Children’s Hospital, Capital Medical University, National Center for Children's Health, Beijing100045, China
| | - Di Lu
- The State Key Laboratory of Membrane Biology, Tsinghua University-Peking University Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing100084, China
| | - Gan Zhao
- The Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, College of Life Sciences, Peking University, Beijing100871, China
| | - He Ren
- The Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, College of Life Sciences, Peking University, Beijing100871, China
| | - Qiaoyu Lin
- The Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, College of Life Sciences, Peking University, Beijing100871, China
| | - Mingkang Jia
- The Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, College of Life Sciences, Peking University, Beijing100871, China
| | - Fan Huang
- The Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, College of Life Sciences, Peking University, Beijing100871, China
| | - Shan Wang
- Department of Dermatology, Beijing Children’s Hospital, Capital Medical University, National Center for Children's Health, Beijing100045, China
| | - Zhe Xu
- Department of Dermatology, Beijing Children’s Hospital, Capital Medical University, National Center for Children's Health, Beijing100045, China
| | - Zhou Yang
- Department of Dermatology, Beijing Children’s Hospital, Capital Medical University, National Center for Children's Health, Beijing100045, China
| | - Yan Chu
- Department of Dermatology, Beijing Children’s Hospital, Capital Medical University, National Center for Children's Health, Beijing100045, China
| | - Zigang Xu
- Department of Dermatology, Beijing Children’s Hospital, Capital Medical University, National Center for Children's Health, Beijing100045, China
| | - Wei Li
- Genetics and Birth Defects Control Center, Beijing Children’s Hospital, Capital Medical University, National Center for Children's Health, Beijing100045, China
| | - Li Yu
- The State Key Laboratory of Membrane Biology, Tsinghua University-Peking University Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing100084, China
| | - Qing Jiang
- The Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, College of Life Sciences, Peking University, Beijing100871, China
| | - Chuanmao Zhang
- The Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, College of Life Sciences, Peking University, Beijing100871, China
- The Academy for Cell and Life Health, Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming650500, China
| |
Collapse
|
26
|
Van Gelder IC, Rienstra M, Bunting KV, Casado-Arroyo R, Caso V, Crijns HJGM, De Potter TJR, Dwight J, Guasti L, Hanke T, Jaarsma T, Lettino M, Løchen ML, Lumbers RT, Maesen B, Mølgaard I, Rosano GMC, Sanders P, Schnabel RB, Suwalski P, Svennberg E, Tamargo J, Tica O, Traykov V, Tzeis S, Kotecha D. 2024 ESC Guidelines for the management of atrial fibrillation developed in collaboration with the European Association for Cardio-Thoracic Surgery (EACTS). Eur Heart J 2024; 45:3314-3414. [PMID: 39210723 DOI: 10.1093/eurheartj/ehae176] [Citation(s) in RCA: 447] [Impact Index Per Article: 447.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/04/2024] Open
|
27
|
Iqbal MK, Ambreen A, Mujahid M, Zarlashat Y, Abid M, Yasin A, Ullah MN, Shahzad R, Harlina PW, Khan SU, Alissa M, Algopishi UB, Almubarak HA. Cardiomegaly: Navigating the uncharted territories of heart failure - A multimodal radiological journey through advanced imaging, pathophysiological landscapes, and innovative therapeutic frontiers. Curr Probl Cardiol 2024; 49:102748. [PMID: 39009253 DOI: 10.1016/j.cpcardiol.2024.102748] [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: 07/08/2024] [Accepted: 07/10/2024] [Indexed: 07/17/2024]
Abstract
Cardiomegaly is among the disorders categorized by a structural enlargement of the heart by any of the situations including pregnancy, resulting in damage to heart muscles and causing trouble in normal heart functioning. Cardiomegaly can be defined in terms of dilatation with an enlarged heart and decreased left or biventricular contraction. The genetic origin of cardiomegaly is becoming more evident due to extensive genomic research opening up new avenues to ensure the use of precision medicine. Cardiomegaly is usually assessed by using an array of radiological modalities, including computed tomography (CT) scans, chest X-rays, and MRIs. These imaging techniques have provided an important opportunity for the physiology and anatomy of the heart. This review aims to highlight the complexity of cardiomegaly, highlighting the contribution of both ecological and genetic variables to its progression. Moreover, we further highlight the worth of precise clinical diagnosis, which comprises blood biomarkers and electrocardiograms (EKG ECG), demonstrating the significance of distinguishing between numerous basic causes. Finally, the analysis highlights the extensive variation of treatment lines, such as lifestyle modifications, prescription drugs, surgery, and implantable devices, although highlighting the critical need for individualized and personalized care.
Collapse
Affiliation(s)
- Muhammad Khalid Iqbal
- Liaoning Provincial Key Laboratory of Cerebral Diseases, Department of Physiology, Dalian Medical University Liaoning Provence China; Department of Biochemistry, Government College University, Faisalabad, Pakistan
| | - Alia Ambreen
- Department of Biochemistry, Government College University, Faisalabad, Pakistan
| | - Muhammad Mujahid
- Department of Biochemistry, Government College University, Faisalabad, Pakistan
| | - Yusra Zarlashat
- Department of Biochemistry, Government College University, Faisalabad, Pakistan
| | - Muhammad Abid
- Academy of Integrative Medicine, Dalian Medical University, Dalian 116044, China
| | - Ayesha Yasin
- Department of Pathology and Forensic Medicine, Dalian Medical University Liaoning Provence, China
| | | | - Raheel Shahzad
- Research Center for Genetic Engineering, National Research and Innovation Agency (BRIN), KST-Cibinong, JI Raya Bogor KM46, Cibinong 16911, Indonesia
| | - Putri Widyanti Harlina
- Department of Food Industrial Technology, Faculty of Agro-Industrial Technology, Universitas Padjadjaran, 45363 Bandung, Indonesia
| | - Shahid Ullah Khan
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City and Southwest University, College of Agronomy and Biotechnology, Southwest University, Chongqing, 400715, China; Women Medical and Dental College, Khyber Medical University, Peshawar, KPK, 22020, Pakistan.
| | - Mohammed Alissa
- Department of Medical Laboratory, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | | | - Hassan Ali Almubarak
- Division of Radiology, Department of Medicine, College of Medicine and Surgery, King Khalid University, Abha, Saudi Arabia
| |
Collapse
|
28
|
Takase B, Ikeda T, Shimizu W, Abe H, Aiba T, Chinushi M, Koba S, Kusano K, Niwano S, Takahashi N, Takatsuki S, Tanno K, Watanabe E, Yoshioka K, Amino M, Fujino T, Iwasaki YK, Kohno R, Kinoshita T, Kurita Y, Masaki N, Murata H, Shinohara T, Yada H, Yodogawa K, Kimura T, Kurita T, Nogami A, Sumitomo N. JCS/JHRS 2022 Guideline on Diagnosis and Risk Assessment of Arrhythmia. Circ J 2024; 88:1509-1595. [PMID: 37690816 DOI: 10.1253/circj.cj-22-0827] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Affiliation(s)
| | - Takanori Ikeda
- Department of Cardiovascular Medicine, Toho University Faculty of Medicine
| | - Wataru Shimizu
- Department of Cardiovascular Medicine, Nippon Medical School
| | - Haruhiko Abe
- Department of Heart Rhythm Management, University of Occupational and Environmental Health, Japan
| | - Takeshi Aiba
- Department of Clinical Laboratory Medicine and Genetics, National Cerebral and Cardiovascular Center
| | - Masaomi Chinushi
- School of Health Sciences, Niigata University School of Medicine
| | - Shinji Koba
- Division of Cardiology, Department of Medicine, Showa University School of Medicine
| | - Kengo Kusano
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center
| | - Shinichi Niwano
- Department of Cardiovascular Medicine, Kitasato University School of Medicine
| | - Naohiko Takahashi
- Department of Cardiology and Clinical Examination, Faculty of Medicine, Oita University
| | - Seiji Takatsuki
- Department of Cardiology, Keio University School of Medicine
| | - Kaoru Tanno
- Cardiology Division, Cardiovascular Center, Showa University Koto-Toyosu Hospital
| | - Eiichi Watanabe
- Division of Cardiology, Department of Internal Medicine, Fujita Health University Bantane Hospital
| | | | - Mari Amino
- Department of Cardiology, Tokai University School of Medicine
| | - Tadashi Fujino
- Department of Cardiovascular Medicine, Toho University Faculty of Medicine
| | - Yu-Ki Iwasaki
- Department of Cardiovascular Medicine, Nippon Medical School
| | - Ritsuko Kohno
- Department of Heart Rhythm Management, University of Occupational and Environmental Health, Japan
| | - Toshio Kinoshita
- Department of Cardiovascular Medicine, Toho University Faculty of Medicine
| | - Yasuo Kurita
- Cardiovascular Center, International University of Health and Welfare, Mita Hospital
| | - Nobuyuki Masaki
- Department of Intensive Care Medicine, National Defense Medical College
| | | | - Tetsuji Shinohara
- Department of Cardiology and Clinical Examination, Faculty of Medicine, Oita University
| | - Hirotaka Yada
- Department of Cardiology, International University of Health and Welfare, Mita Hospital
| | - Kenji Yodogawa
- Department of Cardiovascular Medicine, Nippon Medical School
| | - Takeshi Kimura
- Cardiovascular Medicine, Kyoto University Graduate School of Medicine
| | | | - Akihiko Nogami
- Department of Cardiology, Faculty of Medicine, University of Tsukuba
| | - Naokata Sumitomo
- Department of Pediatric Cardiology, Saitama Medical University International Medical Center
| |
Collapse
|
29
|
Coelho-Rato LS, Parvanian S, Andrs Salajkova S, Medalia O, Eriksson JE. Intermediate filaments at a glance. J Cell Sci 2024; 137:jcs261386. [PMID: 39206824 DOI: 10.1242/jcs.261386] [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] [Indexed: 09/04/2024] Open
Abstract
Intermediate filaments (IFs) comprise a large family of versatile cytoskeletal proteins, divided into six subtypes with tissue-specific expression patterns. IFs have a wide repertoire of cellular functions, including providing structural support to cells, as well as active roles in mechanical support and signaling pathways. Consequently, defects in IFs are associated with more than 100 diseases. In this Cell Science at a Glance article, we discuss the established classes of IFs and their general features, their functions beyond structural support, and recent advances in the field. We also highlight their involvement in disease and potential use as clinical markers of pathological conditions. Finally, we provide our view on current knowledge gaps and the future directions of the IF field.
Collapse
Affiliation(s)
- Leila S Coelho-Rato
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520 Turku, Finland
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, 20520 Turku, Finland
| | - Sepideh Parvanian
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520 Turku, Finland
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, 20520 Turku, Finland
- Center for Systems Biology, Massachusetts General Hospital Research Institute and Harvard Medical School, Boston, MA 02114, USA
| | - Sarka Andrs Salajkova
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Ohad Medalia
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - John E Eriksson
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520 Turku, Finland
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, 20520 Turku, Finland
- Euro-Bioimaging ERIC, 20520 Turku, Finland
| |
Collapse
|
30
|
Wu XY, Lee YK, Lau YM, Au KW, Tse YL, Ng KM, Wong CK, Tse HF. The Pathogenic Mechanisms of and Novel Therapies for Lamin A/C-Related Dilated Cardiomyopathy Based on Patient-Specific Pluripotent Stem Cell Platforms and Animal Models. Pharmaceuticals (Basel) 2024; 17:1030. [PMID: 39204134 PMCID: PMC11357512 DOI: 10.3390/ph17081030] [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: 05/30/2024] [Revised: 07/12/2024] [Accepted: 07/16/2024] [Indexed: 09/03/2024] Open
Abstract
Variants (pathogenic) of the LMNA gene are a common cause of familial dilated cardiomyopathy (DCM), which is characterised by early-onset atrioventricular (AV) block, atrial fibrillation and ventricular tachyarrhythmias (VTs), and progressive heart failure. The unstable internal nuclear lamina observed in LMNA-related DCM is a consequence of the disassembly of lamins A and C. This suggests that LMNA variants produce truncated or alternative forms of protein that alter the nuclear structure and the signalling pathway related to cardiac muscle diseases. To date, the pathogenic mechanisms and phenotypes of LMNA-related DCM have been studied using different platforms, such as patient-specific induced pluripotent stem-cell-derived cardiomyocytes (iPSC-CMs) and transgenic mice. In this review, point variants in the LMNA gene that cause autosomal dominantly inherited forms of LMNA-related DCM are summarised. In addition, potential therapeutic targets based on preclinical studies of LMNA variants using transgenic mice and human iPSC-CMs are discussed. They include mitochondria deficiency, variants in nuclear deformation, chromatin remodelling, altered platelet-derived growth factor and ERK1/2-related pathways, and abnormal calcium handling.
Collapse
Affiliation(s)
- Xin-Yi Wu
- Cardiology Division, Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; (X.-Y.W.); (Y.-K.L.); (Y.-M.L.); (K.-W.A.); (Y.-L.T.); (K.-M.N.); (C.-K.W.)
| | - Yee-Ki Lee
- Cardiology Division, Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; (X.-Y.W.); (Y.-K.L.); (Y.-M.L.); (K.-W.A.); (Y.-L.T.); (K.-M.N.); (C.-K.W.)
| | - Yee-Man Lau
- Cardiology Division, Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; (X.-Y.W.); (Y.-K.L.); (Y.-M.L.); (K.-W.A.); (Y.-L.T.); (K.-M.N.); (C.-K.W.)
| | - Ka-Wing Au
- Cardiology Division, Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; (X.-Y.W.); (Y.-K.L.); (Y.-M.L.); (K.-W.A.); (Y.-L.T.); (K.-M.N.); (C.-K.W.)
| | - Yiu-Lam Tse
- Cardiology Division, Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; (X.-Y.W.); (Y.-K.L.); (Y.-M.L.); (K.-W.A.); (Y.-L.T.); (K.-M.N.); (C.-K.W.)
| | - Kwong-Man Ng
- Cardiology Division, Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; (X.-Y.W.); (Y.-K.L.); (Y.-M.L.); (K.-W.A.); (Y.-L.T.); (K.-M.N.); (C.-K.W.)
- Centre for Stem Cell Translational Biology, Hong Kong SAR, China
| | - Chun-Ka Wong
- Cardiology Division, Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; (X.-Y.W.); (Y.-K.L.); (Y.-M.L.); (K.-W.A.); (Y.-L.T.); (K.-M.N.); (C.-K.W.)
| | - Hung-Fat Tse
- Cardiology Division, Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; (X.-Y.W.); (Y.-K.L.); (Y.-M.L.); (K.-W.A.); (Y.-L.T.); (K.-M.N.); (C.-K.W.)
- Centre for Stem Cell Translational Biology, Hong Kong SAR, China
- Cardiac and Vascular Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
- Hong Kong-Guangdong Stem Cell and Regenerative Medicine Research Centre, The University of Hong Kong and Guangzhou Institutes of Biomedicine and Health, Hong Kong SAR, China
- Advanced Biomedical Instrumentation Centre, Hong Kong SAR, China
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong SAR, China
| |
Collapse
|
31
|
Agdamag AC, Nandar PP, Tang WHW. Advanced Heart Failure Therapies in Neuromuscular Diseases. CURRENT TREATMENT OPTIONS IN CARDIOVASCULAR MEDICINE 2024; 26:255-270. [PMID: 39777119 PMCID: PMC11706575 DOI: 10.1007/s11936-024-01046-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/13/2024] [Indexed: 01/11/2025]
Abstract
Purpose of Review The main objective of this review article is to discuss the prevalence, utilization, and outcomes associated with advanced heart failure therapies among patients with neuromuscular disorders. Recent Findings Neuromuscular disorders often have multisystem involvement with a high prevalence of cardiovascular pathology. With the improvement in management of respiratory related complications, heart failure is now the leading cause of mortality in this patient population. Advanced heart failure therapies with durable left ventricular assist devices and heart transplantation have proven to be feasible and safe treatment options in selected patients. Summary Management of neuromuscular disease involves multidisciplinary team involvement given the systemic nature of the disease. Early recognition and close monitoring of these patients will allow for timely initiation of advanced heart failure therapies that can lead to successful outcomes.
Collapse
Affiliation(s)
- Arianne Clare Agdamag
- Section of Advanced Heart Failure and Transplantation Medicine, Department of Cardiovascular Medicine, Miller Family Heart, Vascular and Thoracic Institute, Robert and Suzanne Tomsich, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Phoo Pwint Nandar
- Section of Advanced Heart Failure, Department of Medicine, MetroHealth Medical Center, Cleveland, OH 44109, USA
| | - W. H. Wilson Tang
- Section of Advanced Heart Failure and Transplantation Medicine, Department of Cardiovascular Medicine, Miller Family Heart, Vascular and Thoracic Institute, Robert and Suzanne Tomsich, Cleveland Clinic, Cleveland, OH 44195, USA
| |
Collapse
|
32
|
Takase B, Ikeda T, Shimizu W, Abe H, Aiba T, Chinushi M, Koba S, Kusano K, Niwano S, Takahashi N, Takatsuki S, Tanno K, Watanabe E, Yoshioka K, Amino M, Fujino T, Iwasaki Y, Kohno R, Kinoshita T, Kurita Y, Masaki N, Murata H, Shinohara T, Yada H, Yodogawa K, Kimura T, Kurita T, Nogami A, Sumitomo N, the Japanese Circulation Society and Japanese Heart Rhythm Society Joint Working Group. JCS/JHRS 2022 Guideline on Diagnosis and Risk Assessment of Arrhythmia. J Arrhythm 2024; 40:655-752. [PMID: 39139890 PMCID: PMC11317726 DOI: 10.1002/joa3.13052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 04/22/2024] [Indexed: 08/15/2024] Open
Affiliation(s)
| | - Takanori Ikeda
- Department of Cardiovascular MedicineToho University Faculty of Medicine
| | - Wataru Shimizu
- Department of Cardiovascular MedicineNippon Medical School
| | - Haruhiko Abe
- Department of Heart Rhythm ManagementUniversity of Occupational and Environmental HealthJapan
| | - Takeshi Aiba
- Department of Clinical Laboratory Medicine and GeneticsNational Cerebral and Cardiovascular Center
| | | | - Shinji Koba
- Division of Cardiology, Department of MedicineShowa University School of Medicine
| | - Kengo Kusano
- Department of Cardiovascular MedicineNational Cerebral and Cardiovascular Center
| | - Shinichi Niwano
- Department of Cardiovascular MedicineKitasato University School of Medicine
| | - Naohiko Takahashi
- Department of Cardiology and Clinical Examination, Faculty of MedicineOita University
| | | | - Kaoru Tanno
- Cardiovascular Center, Cardiology DivisionShowa University Koto‐Toyosu Hospital
| | - Eiichi Watanabe
- Division of Cardiology, Department of Internal MedicineFujita Health University Bantane Hospital
| | | | - Mari Amino
- Department of CardiologyTokai University School of Medicine
| | - Tadashi Fujino
- Department of Cardiovascular MedicineToho University Faculty of Medicine
| | - Yu‐ki Iwasaki
- Department of Cardiovascular MedicineNippon Medical School
| | - Ritsuko Kohno
- Department of Heart Rhythm ManagementUniversity of Occupational and Environmental HealthJapan
| | - Toshio Kinoshita
- Department of Cardiovascular MedicineToho University Faculty of Medicine
| | - Yasuo Kurita
- Cardiovascular Center, Mita HospitalInternational University of Health and Welfare
| | - Nobuyuki Masaki
- Department of Intensive Care MedicineNational Defense Medical College
| | | | - Tetsuji Shinohara
- Department of Cardiology and Clinical Examination, Faculty of MedicineOita University
| | - Hirotaka Yada
- Department of CardiologyInternational University of Health and Welfare Mita Hospital
| | - Kenji Yodogawa
- Department of Cardiovascular MedicineNippon Medical School
| | - Takeshi Kimura
- Cardiovascular MedicineKyoto University Graduate School of Medicine
| | | | - Akihiko Nogami
- Department of Cardiology, Faculty of MedicineUniversity of Tsukuba
| | - Naokata Sumitomo
- Department of Pediatric CardiologySaitama Medical University International Medical Center
| | | |
Collapse
|
33
|
En A, Bogireddi H, Thomas B, Stutzman AV, Ikegami S, LaForest B, Almakki O, Pytel P, Moskowitz IP, Ikegami K. Pervasive nuclear envelope ruptures precede ECM signaling and disease onset without activating cGAS-STING in Lamin-cardiomyopathy mice. Cell Rep 2024; 43:114284. [PMID: 38814785 PMCID: PMC11290591 DOI: 10.1016/j.celrep.2024.114284] [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: 10/03/2023] [Revised: 04/14/2024] [Accepted: 05/13/2024] [Indexed: 06/01/2024] Open
Abstract
Nuclear envelope (NE) ruptures are emerging observations in Lamin-related dilated cardiomyopathy, an adult-onset disease caused by loss-of-function mutations in Lamin A/C, a nuclear lamina component. Here, we test a prevailing hypothesis that NE ruptures trigger the pathological cGAS-STING cytosolic DNA-sensing pathway using a mouse model of Lamin cardiomyopathy. The reduction of Lamin A/C in cardio-myocyte of adult mice causes pervasive NE ruptures in cardiomyocytes, preceding inflammatory transcription, fibrosis, and fatal dilated cardiomyopathy. NE ruptures are followed by DNA damage accumulation without causing immediate cardiomyocyte death. However, cGAS-STING-dependent inflammatory signaling remains inactive. Deleting cGas or Sting does not rescue cardiomyopathy in the mouse model. The lack of cGAS-STING activation is likely due to the near absence of cGAS expression in adult cardiomyocytes at baseline. Instead, extracellular matrix (ECM) signaling is activated and predicted to initiate pro-inflammatory communication from Lamin-reduced cardiomyocytes to fibroblasts. Our work nominates ECM signaling, not cGAS-STING, as a potential inflammatory contributor in Lamin cardiomyopathy.
Collapse
Affiliation(s)
- Atsuki En
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Graduate School of Nanobioscience, Yokohama City University, Yokohama, Kanagawa 236-0027, Japan
| | - Hanumakumar Bogireddi
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Briana Thomas
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Alexis V Stutzman
- Department of Pediatrics, the University of Chicago, Chicago, IL 60637, USA
| | - Sachie Ikegami
- Department of Pediatrics, the University of Chicago, Chicago, IL 60637, USA
| | - Brigitte LaForest
- Department of Pediatrics, the University of Chicago, Chicago, IL 60637, USA
| | - Omar Almakki
- Department of Pediatrics, the University of Chicago, Chicago, IL 60637, USA
| | - Peter Pytel
- Department of Pathology, the University of Chicago, Chicago, IL 60637, USA
| | - Ivan P Moskowitz
- Department of Pediatrics, the University of Chicago, Chicago, IL 60637, USA; Department of Pathology, the University of Chicago, Chicago, IL 60637, USA; Department of Human Genetics, the University of Chicago, Chicago, IL 60637, USA
| | - Kohta Ikegami
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA.
| |
Collapse
|
34
|
Zuela-Sopilniak N, Morival J, Lammerding J. Multi-level transcriptomic analysis of LMNA -related dilated cardiomyopathy identifies disease-driving processes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.11.598511. [PMID: 38915720 PMCID: PMC11195185 DOI: 10.1101/2024.06.11.598511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
LMNA- related dilated cardiomyopathy ( LMNA -DCM) is one of the most severe forms of DCM. The incomplete understanding of the molecular disease mechanisms results in lacking treatment options, leading to high mortality amongst patients. Here, using an inducible, cardiomyocyte-specific lamin A/C depletion mouse model, we conducted a comprehensive transcriptomic study, combining both bulk and single nucleus RNA sequencing, and spanning LMNA -DCM disease progression, to identify potential disease drivers. Our refined analysis pipeline identified 496 genes already misregulated early in disease. The expression of these genes was largely driven by disease specific cardiomyocyte sub-populations and involved biological processes mediating cellular response to DNA damage, cytosolic pattern recognition, and innate immunity. Indeed, DNA damage in LMNA -DCM hearts was significantly increased early in disease and correlated with reduced cardiomyocyte lamin A levels. Activation of cytosolic pattern recognition in cardiomyocytes was independent of cGAS, which is rarely expressed in cardiomyocytes, but likely occurred downstream of other pattern recognition sensors such as IFI16. Altered gene expression in cardiac fibroblasts and immune cell infiltration further contributed to tissue-wide changes in gene expression. Our transcriptomic analysis further predicted significant alterations in cell-cell communication between cardiomyocytes, fibroblasts, and immune cells, mediated through early changes in the extracellular matrix (ECM) in the LMNA -DCM hearts. Taken together, our work suggests a model in which nuclear damage in cardiomyocytes leads to activation of DNA damage responses, cytosolic pattern recognition pathway, and other signaling pathways that activate inflammation, immune cell recruitment, and transcriptional changes in cardiac fibroblasts, which collectively drive LMNA -DCM pathogenesis.
Collapse
|
35
|
Anderson CL, Brown KA, North RJ, Walters JK, Kaska ST, Wolff MR, Kamp TJ, Ge Y, Eckhardt LL. Global Proteomic Analysis Reveals Alterations in Differentially Expressed Proteins between Cardiopathic Lamin A/C Mutations. J Proteome Res 2024; 23:1970-1982. [PMID: 38718259 PMCID: PMC11218822 DOI: 10.1021/acs.jproteome.3c00853] [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] [Indexed: 05/15/2024]
Abstract
Lamin A/C (LMNA) is an important component of nuclear lamina. Mutations cause arrhythmia, heart failure, and sudden cardiac death. While LMNA-associated cardiomyopathy typically has an aggressive course that responds poorly to conventional heart failure therapies, there is variability in severity and age of penetrance between and even within specific mutations, which is poorly understood at the cellular level. Further, this heterogeneity has not previously been captured to mimic the heterozygous state, nor have the hundreds of clinical LMNA mutations been represented. Herein, we have overexpressed cardiopathic LMNA variants in HEK cells and utilized state-of-the-art quantitative proteomics to compare the global proteomic profiles of (1) aggregating Q353 K alone, (2) Q353 K coexpressed with WT, (3) aggregating N195 K coexpressed with WT, and (4) nonaggregating E317 K coexpressed with WT to help capture some of the heterogeneity between mutations. We analyzed each data set to obtain the differentially expressed proteins (DEPs) and applied gene ontology (GO) and KEGG pathway analyses. We found a range of 162 to 324 DEPs from over 6000 total protein IDs with differences in GO terms, KEGG pathways, and DEPs important in cardiac function, further highlighting the complexity of cardiac laminopathies. Pathways disrupted by LMNA mutations were validated with redox, autophagy, and apoptosis functional assays in both HEK 293 cells and in induced pluripotent stem cell derived cardiomyocytes (iPSC-CMs) for LMNA N195 K. These proteomic profiles expand our repertoire for mutation-specific downstream cellular effects that may become useful as druggable targets for personalized medicine approach for cardiac laminopathies.
Collapse
Affiliation(s)
- Corey L. Anderson
- Department of Medicine, Division of Cardiovascular Medicine, University of Wisconsin-Madison, Madison, WI 53705
| | - Kyle A. Brown
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53705
| | - Ryan J. North
- Department of Medicine, Division of Cardiovascular Medicine, University of Wisconsin-Madison, Madison, WI 53705
| | - Janay K. Walters
- Department of Medicine, Division of Cardiovascular Medicine, University of Wisconsin-Madison, Madison, WI 53705
| | - Sara T. Kaska
- Department of Medicine, Division of Cardiovascular Medicine, University of Wisconsin-Madison, Madison, WI 53705
| | - Mathew R. Wolff
- Department of Medicine, Division of Cardiovascular Medicine, University of Wisconsin-Madison, Madison, WI 53705
| | - Timothy J. Kamp
- Department of Medicine, Division of Cardiovascular Medicine, University of Wisconsin-Madison, Madison, WI 53705
| | - Ying Ge
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53705
| | - Lee L. Eckhardt
- Department of Medicine, Division of Cardiovascular Medicine, University of Wisconsin-Madison, Madison, WI 53705
| |
Collapse
|
36
|
Shao Y, Liu C, Liao HK, Zhang R, Yuan B, Yang H, Li R, Zhu S, Fang X, Rodriguez Esteban C, Chen J, Izpisua Belmonte JC. In vivo rescue of genetic dilated cardiomyopathy by systemic delivery of nexilin. Genome Biol 2024; 25:135. [PMID: 38783323 PMCID: PMC11112773 DOI: 10.1186/s13059-024-03283-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 05/16/2024] [Indexed: 05/25/2024] Open
Abstract
BACKGROUND Dilated cardiomyopathy (DCM) is one of the most common causes of heart failure. Multiple identified mutations in nexilin (NEXN) have been suggested to be linked with severe DCM. However, the exact association between multiple mutations of Nexn and DCM remains unclear. Moreover, it is critical for the development of precise and effective therapeutics in treatments of DCM. RESULTS In our study, Nexn global knockout mice and mice carrying human equivalent G645del mutation are studied using functional gene rescue assays. AAV-mediated gene delivery is conducted through systemic intravenous injections at the neonatal stage. Heart tissues are analyzed by immunoblots, and functions are assessed by echocardiography. Here, we identify functional components of Nexilin and demonstrate that exogenous introduction could rescue the cardiac function and extend the lifespan of Nexn knockout mouse models. Similar therapeutic effects are also obtained in G645del mice, providing a promising intervention for future clinical therapeutics. CONCLUSIONS In summary, we demonstrated that a single injection of AAV-Nexn was capable to restore the functions of cardiomyocytes and extended the lifespan of Nexn knockout and G645del mice. Our study represented a long-term gene replacement therapy for DCM that potentially covers all forms of loss-of-function mutations in NEXN.
Collapse
Affiliation(s)
- Yanjiao Shao
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
- Altos Labs, San Diego, CA, 92121, USA
| | - Canzhao Liu
- Department of Cardiology, Translational Medicine Research Center, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, 510280, China
| | - Hsin-Kai Liao
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Ran Zhang
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Baolei Yuan
- Altos Labs, San Diego, CA, 92121, USA
- King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Kingdom of Saudi Arabia
| | - Hanyan Yang
- Department of Cardiology, Translational Medicine Research Center, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, 510280, China
| | - Ronghui Li
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
- Altos Labs, San Diego, CA, 92121, USA
| | - Siting Zhu
- Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Xi Fang
- Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Concepcion Rodriguez Esteban
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
- Altos Labs, San Diego, CA, 92121, USA
| | - Ju Chen
- Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA.
| | - Juan Carlos Izpisua Belmonte
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, 92037, USA.
- Altos Labs, San Diego, CA, 92121, USA.
| |
Collapse
|
37
|
Politano L. Is Cardiac Transplantation Still a Contraindication in Patients with Muscular Dystrophy-Related End-Stage Dilated Cardiomyopathy? A Systematic Review. Int J Mol Sci 2024; 25:5289. [PMID: 38791328 PMCID: PMC11121328 DOI: 10.3390/ijms25105289] [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: 03/08/2024] [Revised: 05/05/2024] [Accepted: 05/08/2024] [Indexed: 05/26/2024] Open
Abstract
Inherited muscular diseases (MDs) are genetic degenerative disorders typically caused by mutations in a single gene that affect striated muscle and result in progressive weakness and wasting in affected individuals. Cardiac muscle can also be involved with some variability that depends on the genetic basis of the MD (Muscular Dystrophy) phenotype. Heart involvement can manifest with two main clinical pictures: left ventricular systolic dysfunction with evolution towards dilated cardiomyopathy and refractory heart failure, or the presence of conduction system defects and serious life-threatening ventricular arrhythmias. The two pictures can coexist. In these cases, heart transplantation (HTx) is considered the most appropriate option in patients who are not responders to the optimized standard therapeutic protocols. However, cardiac transplant is still considered a relative contraindication in patients with inherited muscle disorders and end-stage cardiomyopathies. High operative risk related to muscle impairment and potential graft involvement secondary to the underlying myopathy have been the two main reasons implicated in the generalized reluctance to consider cardiac transplant as a viable option. We report an overview of cardiac involvement in MDs and its possible association with the underlying molecular defect, as well as a systematic review of HTx outcomes in patients with MD-related end-stage dilated cardiomyopathy, published so far in the literature.
Collapse
Affiliation(s)
- Luisa Politano
- Cardiomyology and Medical Genetics, University of Campania Luigi Vanvitelli, 80138 Naples, Italy
| |
Collapse
|
38
|
Chen C, Liu Y, Yang S, Chen M, Liao J. A bibliometric and visual analysis of research trends and hotspots of familial hypertrophic cardiomyopathy: A review. Medicine (Baltimore) 2024; 103:e37969. [PMID: 38701258 PMCID: PMC11062727 DOI: 10.1097/md.0000000000037969] [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: 10/30/2023] [Accepted: 03/29/2024] [Indexed: 05/05/2024] Open
Abstract
Familial hypertrophic cardiomyopathy (FHCM) is an inherited cardiac disease caused by mutations of sarcomere proteins and can be the underlining substrate for major cardiovascular events. Early identification and diagnosis of FHCM are essential to reduce sudden cardiac death. So, this paper summarized the current knowledge on FHCM, and displayed the analysis via bibliometrics method. The relevant literature on FHCM were screened searched via the Web of Science Core Collection database from 2012 to 2022. The literatures were was summarized and analyzed via the bibliometrics method analyzed via CiteSpace and VOSviewer according to topic categories, distribution of spatiotemporal omics and authors, as well as references. Since 2012, there are 909 research articles and reviews related to FHCM. The number of publication for the past 10 years have shown that the development of FHCM research has been steady, with the largest amount of literature in 2012. The most published papers were from the United States, followed by the United Kingdom and Italy. The University of London (63 papers) was the institution that published the most research articles, followed by Harvard University (45 papers) and University College London (45 papers). Keywords formed 3 clusters, focused on the pathogenesis of FHCM, the diagnosis of FHCM, FHCM complications, respectively. The bibliometric analysis and visualization techniques employed herein highlight key trends and focal points in the field, predominantly centered around FHCM's pathogenesis, diagnostic approaches, and its complications. These insights are instrumental in steering future research directions in this area.
Collapse
Affiliation(s)
- Cong Chen
- College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
- Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Yang Liu
- College of Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Songwei Yang
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
| | - Ming Chen
- College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Jing Liao
- College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
- Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
39
|
Marian AJ. Editorial: Gene-based renaming of human diseases. Curr Opin Cardiol 2024; 39:135-137. [PMID: 38567947 PMCID: PMC11421029 DOI: 10.1097/hco.0000000000001123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/05/2024]
Affiliation(s)
- Ali J Marian
- Center for Cardiovascular Genetics, Institute of Molecular Medicine, The University of Texas Health Science Center. Houston, Texas, USA
| |
Collapse
|
40
|
Cathcart B, Cheedipudi SM, Rouhi L, Zhao Z, Gurha P, Marian AJ. DNA double-stranded breaks, a hallmark of aging, defined at the nucleotide resolution, are increased and associated with transcription in the cardiac myocytes in LMNA-cardiomyopathy. Cardiovasc Res 2024:cvae063. [PMID: 38577741 DOI: 10.1093/cvr/cvae063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 03/19/2024] [Accepted: 03/20/2024] [Indexed: 04/06/2024] Open
Abstract
AIMS An intrinsic feature of gene transcription is the formation of DNA superhelices near the transcription bubble, which are resolved upon induction of transient double-stranded breaks (DSBs) by topoisomerases. Unrepaired DSBs are pathogenic as they lead to cell cycle arrest, senescence, inflammation, and organ dysfunction. We posit that DSBs would be more prevalent at the genomic sites that are associated with gene expression. The objectives were to identify and characterize genome-wide DSBs at the nucleotide resolution and determine the association of DSBs with transcription in cardiac myocytes. METHODS AND RESULTS We identified the genome-wide DSBs in ∼1 million cardiac myocytes per heart in three wild-type and three myocyte-specific LMNA-deficient (Myh6-Cre:LmnaF/F) mice by END-Sequencing. The prevalence of DSBs was 0.8% and 2.2% in the wild-type and Myh6-Cre:LmnaF/F myocytes, respectively. The END-Seq signals were enriched for 8 and 6764 DSBs in the wild-type and Myh6-Cre:LmnaF/F myocytes, respectively (q < 0.05). The DSBs were preferentially localized to the gene regions, transcription initiation sites, cardiac transcription factor motifs, and the G quadruplex forming structures. Because LMNA regulates transcription through the lamin-associated domains (LADs), we defined the LADs in cardiac myocytes by a Cleavage Under Targets & Release Using Nuclease (CUT&RUN) assay (N = 5). On average there were 818 LADs per myocyte. Constitutive LADs (cLADs), defined as LADs that were shared by at least three genomes (N = 2572), comprised about a third of the mouse cardiac myocyte genomes. Transcript levels of the protein-coding genes located at the cLADs (N = 3975) were ∼16-fold lower than those at the non-LAD regions (N = ∼17 778). The prevalence of DSBs was higher in the non-LAD as compared to the cLAD regions. Likewise, DSBs were more common in the loss-of-LAD regions, defined as the genomic regions in the Myh6-Cre:LmnaF/F that were juxtaposed to the LAD regions in the wild-type myocytes. CONCLUSION To our knowledge, this is the first identification of the DSBs, at the nucleotide resolution in the cardiovascular system. The prevalence of DSBs was higher in the genomic regions associated with transcription. Because transcription is pervasive, DSBs are expected to be common and pathogenic in various states and aging.
Collapse
Affiliation(s)
- Benjamin Cathcart
- Center for Cardiovascular Genetics, Institute of Molecular Medicine, The University of Texas Health Science Center, 6770 Bertner Street, Suite C900A, Houston, TX 77030, USA
| | - Sirisha M Cheedipudi
- Center for Cardiovascular Genetics, Institute of Molecular Medicine, The University of Texas Health Science Center, 6770 Bertner Street, Suite C900A, Houston, TX 77030, USA
| | - Leila Rouhi
- Center for Cardiovascular Genetics, Institute of Molecular Medicine, The University of Texas Health Science Center, 6770 Bertner Street, Suite C900A, Houston, TX 77030, USA
| | - Zhongming Zhao
- Center for Cardiovascular Genetics, Institute of Molecular Medicine, The University of Texas Health Science Center, 6770 Bertner Street, Suite C900A, Houston, TX 77030, USA
- Center for Precision Health, School of Biomedical Informatics and School of Public Health, UTHealth, Houston, TX 77030, USA
| | - Priyatansh Gurha
- Center for Cardiovascular Genetics, Institute of Molecular Medicine, The University of Texas Health Science Center, 6770 Bertner Street, Suite C900A, Houston, TX 77030, USA
| | - Ali J Marian
- Center for Cardiovascular Genetics, Institute of Molecular Medicine, The University of Texas Health Science Center, 6770 Bertner Street, Suite C900A, Houston, TX 77030, USA
| |
Collapse
|
41
|
Skriver SV, Krett B, Poulsen NS, Krag T, Walas HR, Christensen AH, Bundgaard H, Vissing J, Vissing CR. Skeletal Muscle Involvement in Patients With Truncations of Titin and Familial Dilated Cardiomyopathy. JACC. HEART FAILURE 2024; 12:740-753. [PMID: 37999665 DOI: 10.1016/j.jchf.2023.10.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 09/29/2023] [Accepted: 10/03/2023] [Indexed: 11/25/2023]
Abstract
BACKGROUND Genetic variants in titin (TTN) are associated with dilated cardiomyopathy (DCM) and skeletal myopathy. However, the skeletal muscle phenotype in individuals carrying heterozygous truncating TTN variants (TTNtv), the leading cause of DCM, is understudied. OBJECTIVES This study aimed to assess the skeletal muscle phenotype associated with TTNtv. METHODS Participants with TTNtv were included in a cross-sectional study. Skeletal muscle fat fraction was evaluated by magnetic resonance imaging (compared with healthy controls and controls with non-TTNtv DCM). Muscle strength was evaluated by dynamometry and muscle biopsy specimens were analyzed. RESULTS Twenty-five TTNtv participants (11 women, mean age 51 ± 15 years, left ventricular ejection fraction 45% ± 10%) were included (19 had DCM). Compared to healthy controls (n = 25), fat fraction was higher in calf (12.5% vs 9.9%, P = 0.013), thigh (12.2% vs 9.3%, P = 0.004), and paraspinal muscles (18.8% vs 13.9%, P = 0.008) of TTNtv participants. Linear mixed effects modelling found higher fat fractions in TTNtv participants compared to healthy controls (2.5%; 95% CI: 1.4-3.7; P < 0.001) and controls with non-TTNtv genetic DCM (n = 7) (1.5%; 95% CI: 0.2-2.8; P = 0.025). Muscle strength was within 1 SD of normal values. Biopsy specimens from 21 participants found myopathic features in 13 (62%), including central nuclei. Electron microscopy showed well-ordered Z-lines and T-tubuli but uneven and discontinuous M-lines and excessive glycogen depositions flanked by autophagosomes, lysosomes, and abnormal mitochondria with mitophagy. CONCLUSIONS Mild skeletal muscle involvement was prevalent in patients with TTNtv. The phenotype was characterized by an increased muscle fat fraction and excessive accumulation of glycogen, possibly due to reduced autophagic flux. These findings indicate an impact of TTNtv beyond the heart.
Collapse
Affiliation(s)
- Sofie Vinther Skriver
- Copenhagen Neuromuscular Center, Department of Neurology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Bjørg Krett
- Copenhagen Neuromuscular Center, Department of Neurology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Nanna Scharf Poulsen
- Copenhagen Neuromuscular Center, Department of Neurology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Thomas Krag
- Copenhagen Neuromuscular Center, Department of Neurology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Helle Rudkjær Walas
- Copenhagen Neuromuscular Center, Department of Neurology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Alex Hørby Christensen
- Department of Cardiology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark; Department of Cardiology, Copenhagen University Hospital, Herlev-Gentofte Hospital, Copenhagen, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Henning Bundgaard
- Department of Cardiology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - John Vissing
- Copenhagen Neuromuscular Center, Department of Neurology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | | |
Collapse
|
42
|
Tiwari V, Alam MJ, Bhatia M, Navya M, Banerjee SK. The structure and function of lamin A/C: Special focus on cardiomyopathy and therapeutic interventions. Life Sci 2024; 341:122489. [PMID: 38340979 DOI: 10.1016/j.lfs.2024.122489] [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: 11/11/2023] [Revised: 01/21/2024] [Accepted: 02/03/2024] [Indexed: 02/12/2024]
Abstract
Lamins are inner nuclear membrane proteins that belong to the intermediate filament family. Lamin A/C lie adjacent to the heterochromatin structure in polymer form, providing skeletal to the nucleus. Based on the localization, lamin A/C provides nuclear stability and cytoskeleton to the nucleus and modulates chromatin organization and gene expression. Besides being the structural protein making the inner nuclear membrane in polymer form, lamin A/C functions as a signalling molecule involved in gene expression as an enhancer inside the nucleus. Lamin A/C regulates various cellular pathways like autophagy and energy balance in the cytoplasm. Its expression is highly variable in differentiated tissues, higher in hard tissues like bone and muscle cells, and lower in soft tissues like the liver and brain. In muscle cells, including the heart, lamin A/C must be expressed in a balanced state. Lamin A/C mutation is linked with various diseases, such as muscular dystrophy, lipodystrophy, and cardiomyopathies. It has been observed that a good number of mutations in the LMNA gene impact cardiac activity and its function. Although several works have been published, there are still several unexplored areas left regarding the lamin A/C function and structure in the cardiovascular system and its pathological state. In this review, we focus on the structural organization, expression pattern, and function of lamin A/C, its interacting partners, and the pathophysiology associated with mutations in the lamin A/C gene, with special emphasis on cardiovascular diseases. With the recent finding on lamin A/C, we have summarized the possible therapeutic interventions to treat cardiovascular symptoms and reverse the molecular changes.
Collapse
Affiliation(s)
- Vikas Tiwari
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Guwahati 781101, Assam, India
| | - Md Jahangir Alam
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Guwahati 781101, Assam, India; Cell Biology and Physiology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Madhavi Bhatia
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Guwahati 781101, Assam, India
| | - Malladi Navya
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Guwahati 781101, Assam, India
| | - Sanjay K Banerjee
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Guwahati 781101, Assam, India.
| |
Collapse
|
43
|
Sengupta D, Sengupta K. Lamin A K97E leads to NF-κB-mediated dysfunction of inflammatory responses in dilated cardiomyopathy. Biol Cell 2024; 116:e2300094. [PMID: 38404031 DOI: 10.1111/boc.202300094] [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: 10/04/2023] [Revised: 12/07/2023] [Accepted: 01/04/2024] [Indexed: 02/27/2024]
Abstract
BACKGROUND INFORMATION Lamins are type V intermediate filament proteins underlying the inner nuclear membrane which provide structural rigidity to the nucleus, tether the chromosomes, maintain nuclear homeostasis, and remain dynamically associated with developmentally regulated regions of the genome. A large number of mutations particularly in the LMNA gene encoding lamin A/C results in a wide array of human diseases, collectively termed as laminopathies. Dilated Cardiomyopathy (DCM) is one such laminopathic cardiovascular disease which is associated with systolic dysfunction of left or both ventricles leading to cardiac arrhythmia which ultimately culminates into myocardial infarction. RESULTS In this work, we have unraveled the epigenetic landscape to address the regulation of gene expression in mouse myoblast cell line in the context of the missense mutation LMNA 289A CONCLUSIONS We report here for the first time that there is a significant downregulation of the NF-κB pathway, which has been implicated in cardio-protection elsewhere. SIGNIFICANCE This provides a new pathophysiological explanation that correlates an LMNA mutation and dilated cardiomyopathy.
Collapse
Affiliation(s)
- Duhita Sengupta
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, Kolkata, India
- Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Kaushik Sengupta
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, Kolkata, India
- Homi Bhabha National Institute (HBNI), Mumbai, India
| |
Collapse
|
44
|
Kataoka N, Imamura T, Nakamura M, Kinugawa K. A Clinical Diagnosis of Laminopathy without Systolic Dysfunction: When Does Nuclei Malformation Start? Intern Med 2024; 63:403-406. [PMID: 37316273 PMCID: PMC10901699 DOI: 10.2169/internalmedicine.1928-23] [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: 06/16/2023] Open
Abstract
Nuclear shape abnormalities in laminopathy are well known to occur in patients with cardiac systolic dysfunction. However, those in patients without systolic dysfunction are still unclear. We herein report a 42-year-old man who presented with advanced atrioventricular block without systolic dysfunction. Genetic testing identified a laminopathic mutation, c.497G>C, and an endocardial biopsy was performed. The hyperperfine structure on electron microscopy showed malformation of the nuclei, euchromatic nucleoplasm, and partial existence of heterochromatin clumps. Intrusion of heterochromatin into the nuclear fibrous lamina was observed. Cardiomyocyte nuclear shape abnormalities were observed before the progression of systolic dysfunction.
Collapse
Affiliation(s)
- Naoya Kataoka
- Second Department of Internal Medicine, University of Toyama, Japan
| | - Teruhiko Imamura
- Second Department of Internal Medicine, University of Toyama, Japan
| | - Makiko Nakamura
- Second Department of Internal Medicine, University of Toyama, Japan
| | | |
Collapse
|
45
|
Thompson SD, Barrett KL, Rugel CL, Redmond R, Rudofski A, Kurian J, Curtin JL, Dayanidhi S, Lavasani M. Sex-specific preservation of neuromuscular function and metabolism following systemic transplantation of multipotent adult stem cells in a murine model of progeria. GeroScience 2024; 46:1285-1302. [PMID: 37535205 PMCID: PMC10828301 DOI: 10.1007/s11357-023-00892-5] [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: 06/13/2023] [Accepted: 07/25/2023] [Indexed: 08/04/2023] Open
Abstract
Onset and rates of sarcopenia, a disease characterized by a loss of muscle mass and function with age, vary greatly between sexes. Currently, no clinical interventions successfully arrest age-related muscle impairments since the decline is frequently multifactorial. Previously, we found that systemic transplantation of our unique adult multipotent muscle-derived stem/progenitor cells (MDSPCs) isolated from young mice-but not old-extends the health-span in DNA damage mouse models of progeria, a disease of accelerated aging. Additionally, induced neovascularization in the muscles and brain-where no transplanted cells were detected-strongly suggests a systemic therapeutic mechanism, possibly activated through circulating secreted factors. Herein, we used ZMPSTE24-deficient mice, a lamin A defect progeria model, to investigate the ability of young MDSPCs to preserve neuromuscular tissue structure and function. We show that progeroid ZMPST24-deficient mice faithfully exhibit sarcopenia and age-related metabolic dysfunction. However, systemic transplantation of young MDSPCs into ZMPSTE24-deficient progeroid mice sustained healthy function and histopathology of muscular tissues throughout their 6-month life span in a sex-specific manner. Indeed, female-but not male-mice systemically transplanted with young MDSPCs demonstrated significant preservation of muscle endurance, muscle fiber size, mitochondrial respirometry, and neuromuscular junction morphometrics. These novel findings strongly suggest that young MDSPCs modulate the systemic environment of aged animals by secreted rejuvenating factors to maintain a healthy homeostasis in a sex-specific manner and that the female muscle microenvironment remains responsive to exogenous regenerative cues in older age. This work highlights the age- and sex-related differences in neuromuscular tissue degeneration and the future prospect of preserving health in older adults with systemic regenerative treatments.
Collapse
Affiliation(s)
- Seth D Thompson
- Shirley Ryan AbilityLab, 355 E. Erie St, Chicago, IL, 60611, USA.
- Department of Physical Medicine and Rehabilitation, Northwestern University, Chicago, IL, 60611, USA.
- Northwestern University Interdepartmental Neuroscience (NUIN) Graduate Program, Northwestern University, Chicago, IL, 60611, USA.
| | - Kelsey L Barrett
- Shirley Ryan AbilityLab, 355 E. Erie St, Chicago, IL, 60611, USA
| | - Chelsea L Rugel
- Shirley Ryan AbilityLab, 355 E. Erie St, Chicago, IL, 60611, USA
- Department of Physical Medicine and Rehabilitation, Northwestern University, Chicago, IL, 60611, USA
- Northwestern University Interdepartmental Neuroscience (NUIN) Graduate Program, Northwestern University, Chicago, IL, 60611, USA
| | - Robin Redmond
- Shirley Ryan AbilityLab, 355 E. Erie St, Chicago, IL, 60611, USA
| | - Alexia Rudofski
- Shirley Ryan AbilityLab, 355 E. Erie St, Chicago, IL, 60611, USA
| | - Jacob Kurian
- Department of Biomedical Engineering, Northwestern University, Chicago, IL, 60611, USA
| | - Jodi L Curtin
- Shirley Ryan AbilityLab, 355 E. Erie St, Chicago, IL, 60611, USA
| | - Sudarshan Dayanidhi
- Shirley Ryan AbilityLab, 355 E. Erie St, Chicago, IL, 60611, USA
- Department of Physical Medicine and Rehabilitation, Northwestern University, Chicago, IL, 60611, USA
| | - Mitra Lavasani
- Shirley Ryan AbilityLab, 355 E. Erie St, Chicago, IL, 60611, USA.
- Department of Physical Medicine and Rehabilitation, Northwestern University, Chicago, IL, 60611, USA.
- Northwestern University Interdepartmental Neuroscience (NUIN) Graduate Program, Northwestern University, Chicago, IL, 60611, USA.
| |
Collapse
|
46
|
Abstract
Dilated cardiomyopathy (DCM) is defined as dilation and/or reduced function of one or both ventricles and remains a common disease worldwide. An estimated 40% of cases of familial DCM have an identifiable genetic cause. Accordingly, there is a fast-growing interest in the field of molecular genetics as it pertains to DCM. Many gene mutations have been identified that contribute to phenotypically significant cardiomyopathy. DCM genes can affect a variety of cardiomyocyte functions, and particular genes whose function affects the cell-cell junction and cytoskeleton are associated with increased risk of arrhythmias and sudden cardiac death. Through advancements in next-generation sequencing and cardiac imaging, identification of genetic DCM has improved over the past couple decades, and precision medicine is now at the forefront of treatment for these patients and their families. In addition to standard treatment of heart failure and prevention of arrhythmias and sudden cardiac death, patients with genetic cardiomyopathy stand to benefit from gene mechanism-specific therapies.
Collapse
Affiliation(s)
- Ramone Eldemire
- Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA;
| | - Luisa Mestroni
- Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA;
- Cardiovascular Institute, University of Colorado, Aurora, Colorado, USA
| | - Matthew R G Taylor
- Cardiovascular Institute, University of Colorado, Aurora, Colorado, USA
- Adult Medical Genetics Program, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
47
|
Ebbinghaus H, Ueberham L, Husser-Bollmann D, Bollmann A, Paetsch I, Jahnke C, Laufs U, Dinov B. Case Report: Four cases of cardiac sarcoidosis in patients with inherited cardiomyopathy-a phenotypic overlap, co-existence of two rare cardiomyopathies or a second-hit disease. Front Cardiovasc Med 2023; 10:1328802. [PMID: 38173816 PMCID: PMC10763246 DOI: 10.3389/fcvm.2023.1328802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 12/04/2023] [Indexed: 01/05/2024] Open
Abstract
Cardiac sarcoidosis (CS), a rare condition characterized by non-caseating granulomas, can manifest with symptoms such as atrioventricular block and ventricular tachycardia (VT), as well as mimic inherited cardiomyopathies. A 48-year-old male presented with recurrent VT. The initial 18F-fluorodeoxyglucose positron emission tomography (18FDG-PET) scan showed uptake of the mediastinal lymph node. Cardiovascular magnetic resonance (CMR) demonstrated intramyocardial fibrosis. The follow-up 18FDG-PET scan revealed the presence of tracer uptake in the left ventricular (LV) septum, suggesting the likelihood of CS. Genetic testing identified a pathogenic LMNA variant. A 47-year-old female presented with complaints of palpitations and syncope. An Ajmaline provocation test confirmed Brugada syndrome (BrS). CMR revealed signs of cardiac inflammation. An endomyocardial biopsy (EMB) confirmed the diagnosis of cardiac sarcoidosis. Polymorphic VT was induced during an electrophysiological study, and an implantable cardioverter-defibrillator (ICD) was implanted. A 58-year-old woman presented with sustained VT with a prior diagnosis of hypertrophic cardiomyopathy (HCM). A genetic work-up identified the presence of a heterozygous MYBC3 variant of unknown significance (VUS). CMR revealed late gadolinium enhancement (LGE), while the 18FDG-PET scan demonstrated LV tracer uptake. The immunosuppressive therapy was adjusted, and no further VTs were observed. A 28-year-old male athlete with right ventricular dilatation and syncope experienced a cardiac arrest during training. Genetic testing identified a pathogenic mutation in PKP2. The autopsy has confirmed the presence of ACM and a distinctive extracardiac sarcoidosis. Cardiac sarcoidosis and inherited cardiomyopathies may interact in several different ways, altering the clinical presentation. Overlapping pathologies are frequently overlooked. Delayed or incomplete diagnosis risks inadequate treatment. Thus, genetic testing and endomyocardial biopsies should be recommended to obtain a clear diagnosis.
Collapse
Affiliation(s)
- Hans Ebbinghaus
- Department for Electrophysiology, Heart Center of Leipzig, Leipzig, Germany
| | - Laura Ueberham
- Klinik und Poliklinik für Kardiologie, Universitätsklinikum Leipzig, Leipzig,Germany
| | | | - Andreas Bollmann
- Department for Electrophysiology, Heart Center of Leipzig, Leipzig, Germany
| | - Ingo Paetsch
- Department for Electrophysiology, Heart Center of Leipzig, Leipzig, Germany
| | - Cosima Jahnke
- Department for Electrophysiology, Heart Center of Leipzig, Leipzig, Germany
| | - Ulrich Laufs
- Klinik und Poliklinik für Kardiologie, Universitätsklinikum Leipzig, Leipzig,Germany
| | - Borislav Dinov
- Department of Cardiology, Medical University of Giessen, Giessen, Germany
| |
Collapse
|
48
|
Rossi VA, Saguner AM. A silent interweaving: interatrial block and laminopathy. Eur Heart J Case Rep 2023; 7:ytad616. [PMID: 38130858 PMCID: PMC10733588 DOI: 10.1093/ehjcr/ytad616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2023]
Affiliation(s)
- Valentina A Rossi
- University Heart Centre, University Hospital of Zurich, Rämistrasse 100, Zurich 8091, Switzerland
| | - Ardan M Saguner
- University Heart Centre, University Hospital of Zurich, Rämistrasse 100, Zurich 8091, Switzerland
| |
Collapse
|
49
|
Wang S, Zhang Z, He J, Liu J, Guo X, Chu H, Xu H, Wang Y. Comprehensive review on gene mutations contributing to dilated cardiomyopathy. Front Cardiovasc Med 2023; 10:1296389. [PMID: 38107262 PMCID: PMC10722203 DOI: 10.3389/fcvm.2023.1296389] [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: 09/18/2023] [Accepted: 11/17/2023] [Indexed: 12/19/2023] Open
Abstract
Dilated cardiomyopathy (DCM) is one of the most common primary myocardial diseases. However, to this day, it remains an enigmatic cardiovascular disease (CVD) characterized by ventricular dilatation, which leads to myocardial contractile dysfunction. It is the most common cause of chronic congestive heart failure and the most frequent indication for heart transplantation in young individuals. Genetics and various other factors play significant roles in the progression of dilated cardiomyopathy, and variants in more than 50 genes have been associated with the disease. However, the etiology of a large number of cases remains elusive. Numerous studies have been conducted on the genetic causes of dilated cardiomyopathy. These genetic studies suggest that mutations in genes for fibronectin, cytoskeletal proteins, and myosin in cardiomyocytes play a key role in the development of DCM. In this review, we provide a comprehensive description of the genetic basis, mechanisms, and research advances in genes that have been strongly associated with DCM based on evidence-based medicine. We also emphasize the important role of gene sequencing in therapy for potential early diagnosis and improved clinical management of DCM.
Collapse
Affiliation(s)
- Shipeng Wang
- Department of Cardiovascular Medicine, The First Hospital of Jilin University, Changchun, China
| | - Zhiyu Zhang
- Department of Cardiovascular Medicine, The Second People's Hospital of Yibin, Yibin, China
| | - Jiahuan He
- Department of Cardiovascular Medicine, The First Hospital of Jilin University, Changchun, China
| | - Junqian Liu
- Department of Cardiovascular Medicine, The First Hospital of Jilin University, Changchun, China
| | - Xia Guo
- Department of Cardiovascular Medicine, The First Hospital of Jilin University, Changchun, China
| | - Haoxuan Chu
- Department of Cardiovascular Medicine, The First Hospital of Jilin University, Changchun, China
| | - Hanchi Xu
- Department of Cardiovascular Medicine, The First Hospital of Jilin University, Changchun, China
| | - Yushi Wang
- Department of Cardiovascular Medicine, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
50
|
Yang H, Gong K, Luo Y, Wang L, Tan Z, Yao Y, Xie L. Case report: A new de novo mutation of the Troponin T2 gene in a Chinese patient with dilated cardiomyopathy. Front Cardiovasc Med 2023; 10:1288328. [PMID: 38054088 PMCID: PMC10694197 DOI: 10.3389/fcvm.2023.1288328] [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: 09/04/2023] [Accepted: 11/01/2023] [Indexed: 12/07/2023] Open
Abstract
Dilated cardiomyopathy (DCM) is a cardiovascular disease characterized by persistent ventricular dilatation and systolic dysfunction. DCM has a variety of causes, including myocarditis; exposure to narcotics, alcohol, or other toxins; and metabolic or endocrine disorders. Genetic factors play a dominant role in 30%-40% of DCM cases. Here, we report a case of DCM with very severe heart failure. Because of the severity of heart failure, the patient underwent heart transplantation. We speculated that the patient's DCM might be due to a mutation; hence, we performed whole-exome sequencing of the patient and their parents, which showed a de novo heterozygous mutation (NM_001001431.2c.769G>A:p.E257K) in TNNT2, which was considered pathogenic according to the ACMG pathogenicity assessment. This finding expands the genetic map of DCM and TNNT2 and will be important for future studies on the genetic and disease relationships between DCM and TNNT2.
Collapse
Affiliation(s)
- Huan Yang
- Department of Pulmonary and Critical Care Medicine, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Ke Gong
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Central South University, Changsha, China
| | - Yong Luo
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Central South University, Changsha, China
| | - Lei Wang
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Central South University, Changsha, China
| | - Zhiping Tan
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Central South University, Changsha, China
| | - Yao Yao
- Department of Blood Transfusion, The Second Xiangya Hospital of Central South University, Central South University, Changsha, China
| | - Li Xie
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Central South University, Changsha, China
| |
Collapse
|