1
|
Ma W, Wright DL, Parra O, Shah ND, Black CC, Baker ML, Khan WA. Identification of novel TTN gene variant in a patient exhibiting severe dilated cardiomyopathy co-occurring with acute fibrinoid organizing pneumonia. Am J Clin Pathol 2025; 163:102-108. [PMID: 39180762 DOI: 10.1093/ajcp/aqae100] [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: 01/05/2024] [Accepted: 07/08/2024] [Indexed: 08/26/2024] Open
Abstract
OBJECTIVES Dilated cardiomyopathy (DCM) is often hereditary, with 20% to 40% of nonischemic cases showing familial linkage, yet genetic testing is underused. This report describes an unreported pathogenic nonsense variant in the Titin (TTN) gene (NM_001267550.2:c.92603G>A) in a 24-year-old man with severe DCM and acute fibrinoid organizing pneumonia, highlighting a unique cardiopulmonary pathology. METHODS We conducted detailed gross, histopathologic, immunophenotypic, and exome-based DNA sequencing analysis in the workup of this case. We also included the patient's clinical and radiologic findings in our study. RESULTS With rapid clinical deterioration and complex comorbidities, including substance abuse and psychiatric conditions, which precluded transplantation, the patient's cardiac function progressively worsened. Autopsy findings included extreme cardiomegaly, biventricular hypertrophy, and acute and chronic pericarditis. Significant pulmonary pathology consistent with acute fibrinoid organizing pneumonia was also noted. Molecular testing confirmed a deleterious maternally inherited TTN variant that was absent in the sibling of the proband and the extant medical literature, highlighting its rarity and significance. CONCLUSIONS This case contributes to the ongoing body of work on the impact of TTN variants on DCM. It suggests a potential link between genetic variants and complex cardiac injury patterns, emphasizing the need for further investigation into the interplay between cardiomyopathy and pulmonary pathology.
Collapse
Affiliation(s)
- Weijie Ma
- Department of Pathology and Laboratory Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, NH, US
- Geisel School of Medicine at Dartmouth, Hanover, NH, US
| | - Dana L Wright
- Department of Pathology and Laboratory Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, NH, US
- Geisel School of Medicine at Dartmouth, Hanover, NH, US
| | - Ourania Parra
- Department of Pathology and Laboratory Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, NH, US
- Geisel School of Medicine at Dartmouth, Hanover, NH, US
| | - Nidhi D Shah
- Department of Pathology and Laboratory Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, NH, US
- Geisel School of Medicine at Dartmouth, Hanover, NH, US
| | - Candice C Black
- Department of Pathology and Laboratory Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, NH, US
- Geisel School of Medicine at Dartmouth, Hanover, NH, US
| | - Michael L Baker
- Department of Pathology and Laboratory Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, NH, US
- Geisel School of Medicine at Dartmouth, Hanover, NH, US
| | - Wahab A Khan
- Department of Pathology and Laboratory Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, NH, US
- Geisel School of Medicine at Dartmouth, Hanover, NH, US
| |
Collapse
|
2
|
Wei F, Rui H, Bian R, Liu S. The causal relationship between circulating inflammatory proteins and heart failure: A two-sample Mendelian randomization study. Medicine (Baltimore) 2025; 104:e41115. [PMID: 40184136 PMCID: PMC11709175 DOI: 10.1097/md.0000000000041115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 11/25/2024] [Accepted: 12/11/2024] [Indexed: 04/05/2025] Open
Abstract
This study aims to explore the causal associations of 91 circulating inflammatory proteins with ischemic cardiomyopathy heart failure (ICM), dilated cardiomyopathy heart failure (DCM), and hypertrophic cardiomyopathy heart failure (HCM) to provide new ideas for the study of relevant heart failure mechanisms, adjunctive diagnosis and differentiation, and the clinical application of relevant drug targets. An analysis of the causal relationship between circulating inflammatory proteins and heart failure was conducted via inverse-variance weighted, weighted median estimator (WME), weighted mode (WM), and Mendelian randomization-Egger regression with Mendelian randomization. A Mendelian randomization analysis of 91 circulating inflammatory proteins revealed that natural killer cell receptor 2B4 levels, CXCL-6, fibroblast growth factor 5 levels, and interleukin-10 levels had positive causal relationships with ICM, whereas CX3CL-1, C-X-C motif chemokine 9 levels, interleukin-10 levels, leukemia inhibitory factor receptor levels, and signaling lymphocytic activation molecule levels had negative causal relationships; C-C motif chemokine 20 levels, C-X-C motif chemokine 5 levels, C-X-C motif chemokine 9 levels, fibroblast growth factor 5 levels, and oncostatin-M levels were positively correlated with DCM, whereas eukaryotic translation initiation factor 4E-binding protein 1 levels and Fms-related tyrosine kinase 3 ligand levels were negatively associated with DCM; and the CD40L receptor, Fms-related tyrosine kinase 3 ligand levels, hepatocyte growth factor levels, and sulfotransferase 1A1 levels were negatively associated with HCM. In this study, 9 of the 91 circulating inflammatory proteins were causally related to the ICM (4 positive, 5 negative), 7 were causally related to the DCM (5 positive, 2 negative), and 4 were causally related to the HCM (all negative). This study provides a theoretical foundation for the study of the relevant mechanisms of heart failure, clinical diagnosis, and treatment, as well as potential drug candidates closely related to heart failure.
Collapse
Affiliation(s)
- Fangxiang Wei
- The Second Clinical Medical College of Henan University of Chinese Medicine, Zhengzhou, China
| | - Haomiao Rui
- Henan Province Hospital of TCM (The Second Affiliated Hospital of Henan University of Chinese Medicine), Zhengzhou, China
| | - Rutao Bian
- Zhengzhou Traditional Chinese Medicine Hospital, Zhengzhou, China
| | - Shunyu Liu
- The Second Clinical Medical College of Henan University of Chinese Medicine, Zhengzhou, China
| |
Collapse
|
3
|
McNamee L, Schoch K, Huang A, Lee H, Wang LK, Smith EC, Lark RK, Buckley AF, Jobanputra V, Nelson SF, Shashi V. Reanalysis of RNA sequencing data ends diagnostic odyssey and expands the phenotypic spectrum of congenital titinopathy. Am J Med Genet A 2024; 194:e63798. [PMID: 38924341 PMCID: PMC11934095 DOI: 10.1002/ajmg.a.63798] [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: 03/29/2024] [Revised: 06/06/2024] [Accepted: 06/12/2024] [Indexed: 06/28/2024]
Abstract
Although next-generation sequencing has enabled diagnoses for many patients with Mendelian disorders, the majority remain undiagnosed. Here, we present a sibling pair who were clinically diagnosed with Escobar syndrome, however targeted gene testing was negative. Exome sequencing (ES), and later genome sequencing (GS), revealed compound heterozygous TTN variants in both siblings, a maternally inherited frameshift variant [(NM_133378.4):c.36812del; p.(Asp12271Valfs*10)], and a paternally inherited missense variant [(NM_133378.4):c.12322G > A; p.(Asp4108Asn)]. This result was considered nondiagnostic due to poor clinical fit and limited pathogenicity evidence for the missense variant of uncertain significance (VUS). Following initial nondiagnostic RNA sequencing (RNAseq) on muscle and further pursuit of other variants detected on the ES/GS, a reanalysis of noncanonical splice sites in the muscle transcriptome identified an out-of-frame exon retraction in TTN, near the known VUS. Interim literature included reports of patients with similar TTN variants who had phenotypic concordance with the siblings, and a diagnosis of a congenital titinopathy was given 4 years after the TTN variants had been initially reported. This report highlights the value of reanalysis of RNAseq with a different approach, expands the phenotypic spectrum of congenital titinopathy and also illustrates how a perceived phenotypic mismatch, and failure to consider known variants, can result in a prolongation of the diagnostic journey.
Collapse
Affiliation(s)
- Lucy McNamee
- UNC-Greensboro Genetic Counseling Program, Greensboro, North Carolina, USA
| | - Kelly Schoch
- Division of Medical Genetics, Department of Pediatrics, Duke University School of Medicine, Durham, North Carolina, USA
| | - Alden Huang
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, California, USA
- Institute for Precision Health, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, California, USA
| | - Hane Lee
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, California, USA
- Department of Human Genetics, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, California, USA
| | - Lee-kai Wang
- Institute for Precision Health, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, California, USA
| | - Edward C. Smith
- Division of Pediatric Neurology, Department of Pediatrics, Duke University School of Medicine, Durham, North Carolina, USA
| | - Robert K. Lark
- Department of Orthopedics, Duke University Medical Center, Durham, North Carolina, USA
| | - Anne F. Buckley
- Department of Pathology, Duke University Medical Center, Durham, North Carolina, USA
| | - Vaidehi Jobanputra
- Department of Pathology and Cell Biology, Columbia University, New York, New York, USA
- New York Genome Center, New York, New York, USA
| | - Stanley F. Nelson
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, California, USA
- Institute for Precision Health, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, California, USA
- Department of Human Genetics, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, California, USA
- Department of Pediatrics, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, California, USA
| | - Vandana Shashi
- Division of Medical Genetics, Department of Pediatrics, Duke University School of Medicine, Durham, North Carolina, USA
| | | |
Collapse
|
4
|
Wang BX. Investigating Inherited Heart Diseases Using Human Induced Pluripotent Stem Cell-Based Models. Life (Basel) 2024; 14:1370. [PMID: 39598169 PMCID: PMC11595871 DOI: 10.3390/life14111370] [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: 09/26/2024] [Revised: 10/22/2024] [Accepted: 10/23/2024] [Indexed: 11/29/2024] Open
Abstract
Inherited heart diseases (IHDs) are caused by genetic mutations that disrupt the physiological structure and function of the heart. Understanding the mechanisms behind these diseases is crucial for developing personalised interventions in cardiovascular medicine. Development of induced pluripotent stem cells, which can then be differentiated to any nucleated adult cell type, has enabled the creation of personalised single-cell and multicellular models, providing unprecedented insights into the pathophysiology of IHDs. This review provides a comprehensive overview of recent advancements in human iPSC models used to dissect the molecular and genetic underpinnings of common IHDs. We examine multicellular models and tissue engineering approaches, such as cardiac organoids, engineered heart tissue, and multicellular co-culture systems, which simulate complex intercellular interactions within heart tissue. Recent advancements in stem cell models offer a more physiologically relevant platform to study disease mechanisms, enabling researchers to observe cellular interactions, study disease progression, and identify therapeutic strategies. By leveraging these innovative models, we can gain deeper insights into the molecular and cellular mechanisms underlying IHDs, ultimately paving the way for more effective diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Brian Xiangzhi Wang
- Department of Cardiology, Jersey General Hospital, Gloucester Street, St. Helier JE1 3QS, Jersey, UK
| |
Collapse
|
5
|
Jolfayi AG, Kohansal E, Ghasemi S, Naderi N, Hesami M, MozafaryBazargany M, Moghadam MH, Fazelifar AF, Maleki M, Kalayinia S. Exploring TTN variants as genetic insights into cardiomyopathy pathogenesis and potential emerging clues to molecular mechanisms in cardiomyopathies. Sci Rep 2024; 14:5313. [PMID: 38438525 PMCID: PMC10912352 DOI: 10.1038/s41598-024-56154-7] [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: 11/22/2023] [Accepted: 03/01/2024] [Indexed: 03/06/2024] Open
Abstract
The giant protein titin (TTN) is a sarcomeric protein that forms the myofibrillar backbone for the components of the contractile machinery which plays a crucial role in muscle disorders and cardiomyopathies. Diagnosing TTN pathogenic variants has important implications for patient management and genetic counseling. Genetic testing for TTN variants can help identify individuals at risk for developing cardiomyopathies, allowing for early intervention and personalized treatment strategies. Furthermore, identifying TTN variants can inform prognosis and guide therapeutic decisions. Deciphering the intricate genotype-phenotype correlations between TTN variants and their pathologic traits in cardiomyopathies is imperative for gene-based diagnosis, risk assessment, and personalized clinical management. With the increasing use of next-generation sequencing (NGS), a high number of variants in the TTN gene have been detected in patients with cardiomyopathies. However, not all TTN variants detected in cardiomyopathy cohorts can be assumed to be disease-causing. The interpretation of TTN variants remains challenging due to high background population variation. This narrative review aimed to comprehensively summarize current evidence on TTN variants identified in published cardiomyopathy studies and determine which specific variants are likely pathogenic contributors to cardiomyopathy development.
Collapse
Affiliation(s)
- Amir Ghaffari Jolfayi
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Erfan Kohansal
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Serwa Ghasemi
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Niloofar Naderi
- Cardiogenetic Research Center, Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mahshid Hesami
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | | | - Maryam Hosseini Moghadam
- Cardiogenetic Research Center, Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Amir Farjam Fazelifar
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Majid Maleki
- Cardiogenetic Research Center, Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Samira Kalayinia
- Cardiogenetic Research Center, Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
6
|
ElSaygh J, Zaher A, Peterson SJ, Parikh MA, Frishman WH. Titin: The Missing Link in Cardiac Physiology. Cardiol Rev 2024:00045415-990000000-00209. [PMID: 38334419 DOI: 10.1097/crd.0000000000000656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/10/2024]
Abstract
Titin, an extraordinary protein known for its colossal size and multifaceted roles, is a cornerstone in the structural and functional dynamics of striated muscle tissues, including the heart and skeletal muscles. Its sheer enormity, with a molecular weight exceeding 3000 kDa, is paralleled only by the immense influence it exerts on muscle physiology. This review will delve into the remarkable structural organization of Titin and the genetics of this molecule, including the common mutations resulting in various cardiomyopathies. We will delve deeper into its role in dilated cardiomyopathy, familial restrictive cardiomyopathy, hypertrophic cardiomyopathy, and left ventricular noncompaction cardiomyopathy. This review culminates by discussing the prospects of therapeutic strategies targeting Titin. While these interventions remain primarily theoretical, the possibilities are intriguing. Patients with Titin truncation mutations present unique challenges, but innovative approaches like gene therapy or preemptive treatments with drugs such as angiotensin-converting enzyme inhibitors or beta-blockers offer hope. This multi-pronged approach highlights the significance of understanding Titin's multifaceted role and its potential as a target for future therapeutic interventions.
Collapse
Affiliation(s)
- Jude ElSaygh
- From the Department of Medicine, New York Presbyterian Brooklyn Methodist Hospital, Brooklyn, NY
| | - Anas Zaher
- From the Department of Medicine, New York Presbyterian Brooklyn Methodist Hospital, Brooklyn, NY
| | - Stephen J Peterson
- From the Department of Medicine, New York Presbyterian Brooklyn Methodist Hospital, Brooklyn, NY
- Weill Department of Medicine, Weill Cornell Medicine, NY
| | - Manish A Parikh
- From the Department of Medicine, New York Presbyterian Brooklyn Methodist Hospital, Brooklyn, NY
- Weill Department of Medicine, Weill Cornell Medicine, NY
| | | |
Collapse
|
7
|
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
|
8
|
Lian H, Song S, Chen W, Shi A, Jiang H, Hu S. Genetic characterization of dilated cardiomyopathy patients undergoing heart transplantation in the Chinese population by whole-exome sequencing. J Transl Med 2023; 21:476. [PMID: 37461109 PMCID: PMC10351148 DOI: 10.1186/s12967-023-04282-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 06/17/2023] [Indexed: 07/20/2023] Open
Abstract
BACKGROUND Dilated cardiomyopathy (DCM) is one of the most frequent causes of heart failure and heart transplantation (HTx). The genetic basis of DCM among patients undergoing HTx remains to be further studied. This study aimed to characterize the genetic basis of DCM HTx in the Chinese population. METHODS In total, 208 unrelated DCM patients who underwent HTx at Fuwai Hospital between June 2004 and June 2017 were included in this study. Whole-exome sequencing (WES) was performed for all patients. Gene burden analysis, variant classification, and genotype-phenotype correlation analysis were subsequently performed. RESULTS After completing the bioinformatics analysis, gene burden analysis suggested that titin (TTN), filamin C (FLNC) and lamin A/C (LMNA) were significantly enriched with rare protein-altering variants. The frequencies of TTN and FLNC truncating variants in our cohort were 18.8% and 8.7%, respectively. Among the 165 rare variants in high evidence DCM-related genes, 27 (16.4%) and 59 (35.8%) were interpreted as pathogenic (P) and likely pathogenic (LP), respectively. In addition, 41 (47.7%) and 16 (18.6%) of these 86 P/LP variants are located in TTN and FLNC, respectively. The FLNC group contained more patients with NYHA class IV than the P/LP-negative group (FLNC, 16/18 vs. P/LP-negative, 81/123, P = 0.049). CONCLUSIONS Based on WES, we provided a primary genetic spectrum of DCM patients undergoing HTx in the Chinese population. TTN and FLNC harbour the most P/LP variants. FLNC truncation may lead to severe clinical symptoms in DCM patients.
Collapse
Affiliation(s)
- Hong Lian
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Center, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Shen Song
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Wenzheng Chen
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Anteng Shi
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Haobin Jiang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Shengshou Hu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China.
| |
Collapse
|
9
|
Koslow M, Mondaca-Ruff D, Xu X. Transcriptome studies of inherited dilated cardiomyopathies. Mamm Genome 2023; 34:312-322. [PMID: 36749382 PMCID: PMC10426000 DOI: 10.1007/s00335-023-09978-z] [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: 07/25/2022] [Accepted: 01/16/2023] [Indexed: 02/08/2023]
Abstract
Dilated cardiomyopathy (DCM) is a group of heart muscle diseases that often lead to heart failure, with more than 50 causative genes have being linked to DCM. The heterogenous nature of the inherited DCMs suggest the need of precision medicine. Consistent with this emerging concept, transcriptome studies in human patients with DCM indicated distinct molecular signature for DCMs of different genetic etiology. To facilitate this line of research, we reviewed the status of transcriptome studies of inherited DCMs by focusing on three predominant DCM causative genes, TTN, LMNA, and BAG3. Besides studies in human patients, we summarized transcriptomic analysis of these inherited DCMs in a variety of model systems ranging from iPSCs to rodents and zebrafish. We concluded that the RNA-seq technology is a powerful genomic tool that has already led to the discovery of new modifying genes, signaling pathways, and related therapeutic avenues. We also pointed out that both temporal (different pathological stages) and spatial (different cell types) information need to be considered for future transcriptome studies. While an important bottle neck is the low throughput in experimentally testing differentially expressed genes, new technologies in efficient animal models such as zebrafish starts to be developed. It is anticipated that the RNA-seq technology will continue to uncover both unique and common pathological events, aiding the development of precision medicine for inherited DCMs.
Collapse
Affiliation(s)
- Matthew Koslow
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
- Department of Cardiovascular Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - David Mondaca-Ruff
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
- Department of Cardiovascular Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Xiaolei Xu
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA.
- Department of Cardiovascular Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| |
Collapse
|
10
|
Verdonschot JA, Wang P, Derks KW, Adriaens ME, Stroeks SL, Henkens MT, Raafs AG, Sikking M, de Koning B, van den Wijngaard A, Krapels IP, Nabben M, Brunner HG, Heymans SR. Clustering of Cardiac Transcriptome Profiles Reveals Unique: Subgroups of Dilated Cardiomyopathy Patients. JACC Basic Transl Sci 2023; 8:406-418. [PMID: 37138803 PMCID: PMC10149655 DOI: 10.1016/j.jacbts.2022.10.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 10/26/2022] [Accepted: 10/26/2022] [Indexed: 02/04/2023]
Abstract
Dilated cardiomyopathy is a heterogeneous disease characterized by multiple genetic and environmental etiologies. The majority of patients are treated the same despite these differences. The cardiac transcriptome provides information on the patient's pathophysiology, which allows targeted therapy. Using clustering techniques on data from the genotype, phenotype, and cardiac transcriptome of patients with early- and end-stage dilated cardiomyopathy, more homogeneous patient subgroups are identified based on shared underlying pathophysiology. Distinct patient subgroups are identified based on differences in protein quality control, cardiac metabolism, cardiomyocyte function, and inflammatory pathways. The identified pathways have the potential to guide future treatment and individualize patient care.
Collapse
Affiliation(s)
- Job A.J. Verdonschot
- Department of Clinical Genetics, Maastricht University Medical Center, Maastricht, the Netherlands
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
| | - Ping Wang
- Department of Clinical Genetics, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Kasper W.J. Derks
- Department of Clinical Genetics, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Michiel E. Adriaens
- Maastricht Centre for Systems Biology, Maastricht University, Maastricht, the Netherlands
| | - Sophie L.V.M. Stroeks
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
- Department of Cardiology, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Michiel T.H.M. Henkens
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
- Department of Cardiology, Maastricht University Medical Center, Maastricht, the Netherlands
- Netherlands Heart Institute (NLHI), Utrecht, the Netherlands
| | - Anne G. Raafs
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
- Department of Cardiology, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Maurits Sikking
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
- Department of Cardiology, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Bart de Koning
- Department of Clinical Genetics, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Arthur van den Wijngaard
- Department of Clinical Genetics, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Ingrid P.C. Krapels
- Department of Clinical Genetics, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Miranda Nabben
- Department of Clinical Genetics, Maastricht University Medical Center, Maastricht, the Netherlands
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
| | - Han G. Brunner
- Department of Clinical Genetics, Maastricht University Medical Center, Maastricht, the Netherlands
- GROW Institute for Developmental Biology and Cancer, Maastricht University, Maastricht, the Netherlands
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Stephane R.B. Heymans
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
- Centre for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Belgium
| |
Collapse
|
11
|
Shi HY, Xie MS, Guo YH, Yang CX, Gu JN, Qiao Q, Di RM, Qiu XB, Xu YJ, Yang YQ. VEZF1 loss-of-function mutation underlying familial dilated cardiomyopathy. Eur J Med Genet 2023; 66:104705. [PMID: 36657711 DOI: 10.1016/j.ejmg.2023.104705] [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: 07/30/2021] [Revised: 11/17/2022] [Accepted: 01/15/2023] [Indexed: 01/18/2023]
Abstract
Dilated cardiomyopathy (DCM), characteristic of left ventricular or biventricular dilation with systolic dysfunction, is the most common form of cardiomyopathy, and a leading cause of heart failure and sudden cardiac death. Aggregating evidence highlights the underlying genetic basis of DCM, and mutations in over 100 genes have been causally linked to DCM. Nevertheless, due to pronounced genetic heterogeneity, the genetic defects underpinning DCM in most cases remain obscure. Hence, this study was sought to identify novel genetic determinants of DCM. In this investigation, whole-exome sequencing and bioinformatics analyses were conducted in a family suffering from DCM, and a novel heterozygous mutation in the VEZF1 gene (coding for a zinc finger-containing transcription factor critical for cardiovascular development and structural remodeling), NM_007146.3: c.490A > T; p.(Lys164*), was identified. The nonsense mutation was validated by Sanger sequencing and segregated with autosome-dominant DCM in the family with complete penetrance. The mutation was neither detected in another cohort of 200 unrelated DCM patients nor observed in 400 unrelated healthy individuals nor retrieved in the Single Nucleotide Polymorphism database, the Human Gene Mutation Database and the Genome Aggregation Database. Biological analyses by utilizing a dual-luciferase reporter assay system revealed that the mutant VEZF1 protein failed to transactivate the promoters of MYH7 and ET1, two genes that have been associated with DCM. The findings indicate VEZF1 as a new gene responsible for DCM, which provides novel insight into the molecular pathogenesis of DCM, implying potential implications for personalized precisive medical management of the patients affected with DCM.
Collapse
Affiliation(s)
- Hong-Yu Shi
- Department of Cardiology, Zhongshan Hospital Wusong Branch, Fudan University, Shanghai, China
| | - Meng-Shi Xie
- Department of Cardiology, Zhongshan Hospital Wusong Branch, Fudan University, Shanghai, China
| | - Yu-Han Guo
- Department of Cardiology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Chen-Xi Yang
- Department of Cardiology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Jia-Ning Gu
- Department of Cardiology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Qi Qiao
- Department of Cardiology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Ruo-Min Di
- Department of Cardiology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Xing-Biao Qiu
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Ying-Jia Xu
- Department of Cardiology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China.
| | - Yi-Qing Yang
- Department of Cardiology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China; Department of Cardiovascular Research Laboratory, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China; Department of Central Laboratory, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
12
|
The Involvement of Krüppel-like Factors in Cardiovascular Diseases. Life (Basel) 2023; 13:life13020420. [PMID: 36836777 PMCID: PMC9962890 DOI: 10.3390/life13020420] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 01/16/2023] [Accepted: 01/29/2023] [Indexed: 02/05/2023] Open
Abstract
Krüppel-like factors (KLFs) are a set of DNA-binding proteins belonging to a family of zinc-finger transcription factors, which have been associated with many biological processes related to the activation or repression of genes, inducing cell growth, differentiation, and death, and the development and maintenance of tissues. In response to metabolic alterations caused by disease and stress, the heart will undergo cardiac remodeling, leading to cardiovascular diseases (CVDs). KLFs are among the transcriptional factors that take control of many physiological and, in this case, pathophysiological processes of CVD. KLFs seem to be associated with congenital heart disease-linked syndromes, malformations because of autosomal diseases, mutations that relate to protein instability, and/or loss of functions such as atheroprotective activities. Ischemic damage also relates to KLF dysregulation because of the differentiation of cardiac myofibroblasts or a modified fatty acid oxidation related to the formation of a dilated cardiomyopathy, myocardial infarctions, left ventricular hypertrophy, and diabetic cardiomyopathies. In this review, we describe the importance of KLFs in cardiovascular diseases such as atherosclerosis, myocardial infarction, left ventricle hypertrophy, stroke, diabetic cardiomyopathy, and congenital heart diseases. We further discuss microRNAs that have been involved in certain regulatory loops of KLFs as they may act as critical in CVDs.
Collapse
|
13
|
Kulikova OV, Myasnikov RP, Meshkov AN, Mershina EA, Kiseleva AV, Sotnikova EA, Kudryavtseva MM, Kharlap MS, Divashuk MG, Zharikova AA, Angarsky RK, Koretsky SN, Filatova DА, Sinitsyn VE, Drapkina OM. RBM20 nucleotide sequence variant in a family with a dilated phenotype of left ventricular non-compaction. КАРДИОВАСКУЛЯРНАЯ ТЕРАПИЯ И ПРОФИЛАКТИКА 2023. [DOI: 10.15829/1728-8800-2022-3470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Aim. To demonstrate two generations of a family with a progressive course of left ventricular non-compaction (LVNC) and the presence of a RBM20 gene variant.Material and methods. Based on the multicenter registry of patients with LVNC, a family with LVNC with a dilated phenotype was selected at the National Medical Research Center for Therapy and Preventive Medicine. Next generation sequencing was performed on a Nextseq 550 systen (Illumina, USA). For clinical interpretation, nucleotide sequence variants in the genes associated with LVNC development were selected according to the available literature data, with frequencies <0,5% in the gnomAD database. The identified variants were verified using Sanger sequencing on an Applied Biosystem 3500 Genetic Analyzer (Thermo Fisher Scientific, USA).Results. The article presents the results of clinical, paraclinical and molecular genetic studies of two generations of a family diagnosed with LVNC with a dilated phenotype and the progression of isolated LVNC to a dilated type. As a result of a molecular genetic study, all family members with the LVNC were found to have a likely pathogenic variant in the RBM20 NP_001127835.2:p.Pro638Leu (rs267607003) gene. RBM20 is a key splicing regulator that controls the processing of several important transcripts predominantly expressed in striated muscle, especially cardiac tissue. RBM20 gene variants can lead to disruption of splicing at several points and, as a result, to cardiomyopathy progression. Most known pathogenic RBM20 variants are associated with dilated cardiomyopathy; however, a number of studies have found RBM20 gene variants in patients with LVNC. The segregation of nucleotide sequence variant with symptoms in two generations testifies in favor of the association of the detected variant with LVNC development.Conclusion. Currently, the boundaries of the cardiomyopathy genetics are expanding. Pathogenic and likely pathogenic RBM20 gene variants are associated primarily with a dilated phenotype and a high risk of sudden cardiac death. The article presents the results of a survey of two generations of a family with LVNC and progressive myocardial remodeling.
Collapse
Affiliation(s)
- O. V. Kulikova
- National Medical Research Center for Therapy and Preventive Medicine
| | - R. P. Myasnikov
- National Medical Research Center for Therapy and Preventive Medicine
| | - A. N. Meshkov
- National Medical Research Center for Therapy and Preventive Medicine
| | - E. A. Mershina
- Medical Research and Educational Center, Lomonosov Moscow State University
| | - A. V. Kiseleva
- National Medical Research Center for Therapy and Preventive Medicine
| | - E. A. Sotnikova
- National Medical Research Center for Therapy and Preventive Medicine
| | | | - M. S. Kharlap
- National Medical Research Center for Therapy and Preventive Medicine
| | - M. G. Divashuk
- National Medical Research Center for Therapy and Preventive Medicine; Kurchatov Center for Genome Research, All-Russia Research Institute of Agricultural Biotechnology
| | - A. A. Zharikova
- National Medical Research Center for Therapy and Preventive Medicine; Lomonosov Moscow State University
| | - R. K. Angarsky
- National Medical Research Center for Therapy and Preventive Medicine
| | - S. N. Koretsky
- National Medical Research Center for Therapy and Preventive Medicine
| | - D. А. Filatova
- Medical Research and Educational Center, Lomonosov Moscow State University
| | - V. E. Sinitsyn
- Medical Research and Educational Center, Lomonosov Moscow State University
| | - O. M. Drapkina
- National Medical Research Center for Therapy and Preventive Medicine
| |
Collapse
|
14
|
Guo Y, Wang J, Guo X, Gao R, Yang C, Li L, Sun Y, Qiu X, Xu Y, Yang Y. KLF13 Loss‐of‐Function Mutations Underlying Familial Dilated Cardiomyopathy. J Am Heart Assoc 2022; 11:e027578. [DOI: 10.1161/jaha.122.027578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Background
Dilated cardiomyopathy (DCM), characterized by progressive left ventricular enlargement and systolic dysfunction, is the most common type of cardiomyopathy and a leading cause of heart failure and cardiac death. Accumulating evidence underscores the critical role of genetic defects in the pathogenesis of DCM, and >250 genes have been implicated in DCM to date. However, DCM is of substantial genetic heterogeneity, and the genetic basis underpinning DCM remains elusive in most cases.
Methods and Results
By genome‐wide scan with microsatellite markers and genetic linkage analysis in a 4‐generation family inflicted with autosomal‐dominant DCM, a new locus for DCM was mapped on chromosome 15q13.1–q13.3, a 4.77‐cM (≈3.43 Mbp) interval between markers D15S1019 and D15S1010, with the largest 2‐point logarithm of odds score of 5.1175 for the marker D15S165 at recombination fraction (θ)=0.00. Whole‐exome sequencing analyses revealed that within the mapping chromosomal region, only the mutation in the
KLF13
gene, c.430G>T (p.E144X), cosegregated with DCM in the family. In addition, sequencing analyses of
KLF13
in another cohort of 266 unrelated patients with DCM and their available family members unveiled 2 new mutations, c.580G>T (p.E194X) and c.595T>C (p.C199R), which cosegregated with DCM in 2 families, respectively. The 3 mutations were absent from 418 healthy subjects. Functional assays demonstrated that the 3 mutants had no transactivation on the target genes
ACTC1
and
MYH7
(2 genes causally linked to DCM), alone or together with GATA4 (another gene contributing to DCM), and a diminished ability to bind the promoters of
ACTC1
and
MYH7
. Add, the E144X‐mutant KLF13 showed a defect in intracellular distribution.
Conclusions
This investigation indicates
KLF13
as a new gene predisposing to DCM, which adds novel insight to the molecular pathogenesis underlying DCM, implying potential implications for prenatal prevention and precision treatment of DCM in a subset of patients.
Collapse
Affiliation(s)
- Yu‐Han Guo
- Department of Cardiology, Shanghai Fifth People’s Hospital Fudan University Shanghai China
| | - Jun Wang
- Department of Cardiology, Shanghai Jing’an District Central Hospital Fudan University Shanghai China
| | - Xiao‐Juan Guo
- Department of Cardiology, Shanghai Fifth People’s Hospital Fudan University Shanghai China
| | - Ri‐Feng Gao
- Department of Cardiology, Shanghai Fifth People’s Hospital Fudan University Shanghai China
| | - Chen‐Xi Yang
- Department of Cardiology, Shanghai Fifth People’s Hospital Fudan University Shanghai China
| | - Li Li
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital Tongji University School of Medicine Shanghai China
- Institute of Medical Genetics Tongji University Shanghai China
| | - Yu‐Min Sun
- Department of Cardiology, Shanghai Jing’an District Central Hospital Fudan University Shanghai China
| | - Xing‐Biao Qiu
- Department of Cardiology, Shanghai Chest Hospital Shanghai Jiao Tong University Shanghai China
| | - Ying‐Jia Xu
- Department of Cardiology, Shanghai Fifth People’s Hospital Fudan University Shanghai China
| | - Yi‐Qing Yang
- Department of Cardiology, Shanghai Fifth People’s Hospital Fudan University Shanghai China
- Cardiovascular Research Laboratory and Central Laboratory, Shanghai Fifth People’s Hospital Fudan University Shanghai China
| |
Collapse
|
15
|
Mazzaccara C, Lombardi R, Mirra B, Barretta F, Esposito MV, Uomo F, Caiazza M, Monda E, Losi MA, Limongelli G, D’Argenio V, Frisso G. Next-Generation Sequencing Gene Panels in Inheritable Cardiomyopathies and Channelopathies: Prevalence of Pathogenic Variants and Variants of Unknown Significance in Uncommon Genes. Biomolecules 2022; 12:1417. [PMID: 36291626 PMCID: PMC9599286 DOI: 10.3390/biom12101417] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/27/2022] [Accepted: 09/30/2022] [Indexed: 08/17/2023] Open
Abstract
The diffusion of next-generation sequencing (NGS)-based approaches allows for the identification of pathogenic mutations of cardiomyopathies and channelopathies in more than 200 different genes. Since genes considered uncommon for a clinical phenotype are also now included in molecular testing, the detection rate of disease-causing variants has increased. Here, we report the prevalence of genetic variants detected by using a NGS custom panel in a cohort of 133 patients with inherited cardiomyopathies (n = 77) or channelopathies (n = 56). We identified 82 variants, of which 50 (61%) were identified in genes without a strong or definitive evidence of disease association according to the NIH-funded Clinical Genome Resource (ClinGen; "uncommon genes"). Among these, 35 (70%) were variants of unknown significance (VUSs), 13 (26%) were pathogenic (P) or likely pathogenic (LP) mutations, and 2 (4%) benign (B) or likely benign (LB) variants according to American College of Medical Genetics (ACMG) classifications. These data reinforce the need for the screening of uncommon genes in order to increase the diagnostic sensitivity of the genetic testing of inherited cardiomyopathies and channelopathies by allowing for the identification of mutations in genes that are not usually explored due to a currently poor association with the clinical phenotype.
Collapse
Affiliation(s)
- Cristina Mazzaccara
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, 80131 Napoli, Italy
- CEINGE Biotecnologie Avanzate, 80145 Napoli, Italy
| | - Raffaella Lombardi
- Department of Advanced Biomedical Sciences, University of Naples Federico II, 80131 Napoli, Italy
- Department of Medicine, Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Bruno Mirra
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, 80131 Napoli, Italy
- CEINGE Biotecnologie Avanzate, 80145 Napoli, Italy
| | - Ferdinando Barretta
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, 80131 Napoli, Italy
- CEINGE Biotecnologie Avanzate, 80145 Napoli, Italy
| | | | - Fabiana Uomo
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, 80131 Napoli, Italy
- CEINGE Biotecnologie Avanzate, 80145 Napoli, Italy
| | - Martina Caiazza
- Monaldi Hospital, AO Colli, 80131 Napoli, Italy
- Department of Translational Medical Sciences, University of Campania ‘Luigi Vanvitelli’, 81100 Caserta, Italy
| | - Emanuele Monda
- Monaldi Hospital, AO Colli, 80131 Napoli, Italy
- Department of Translational Medical Sciences, University of Campania ‘Luigi Vanvitelli’, 81100 Caserta, Italy
| | - Maria Angela Losi
- Department of Advanced Biomedical Sciences, University of Naples Federico II, 80131 Napoli, Italy
| | - Giuseppe Limongelli
- Monaldi Hospital, AO Colli, 80131 Napoli, Italy
- Department of Translational Medical Sciences, University of Campania ‘Luigi Vanvitelli’, 81100 Caserta, Italy
| | - Valeria D’Argenio
- CEINGE Biotecnologie Avanzate, 80145 Napoli, Italy
- Department of Human Sciences and Quality of Life Promotion, San Raffaele Open University, 00166 Roma, Italy
| | - Giulia Frisso
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, 80131 Napoli, Italy
- CEINGE Biotecnologie Avanzate, 80145 Napoli, Italy
| |
Collapse
|
16
|
Yang Y, Fu Z, Zhu W, Hu H, Wang J. Application of optical tweezers in cardiovascular research: More than just a measuring tool. Front Bioeng Biotechnol 2022; 10:947918. [PMID: 36147537 PMCID: PMC9486066 DOI: 10.3389/fbioe.2022.947918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 08/12/2022] [Indexed: 12/04/2022] Open
Abstract
Recent advances in the field of optical tweezer technology have shown intriguing potential for applications in cardiovascular medicine, bringing this laboratory nanomechanical instrument into the spotlight of translational medicine. This article summarizes cardiovascular system findings generated using optical tweezers, including not only rigorous nanomechanical measurements but also multifunctional manipulation of biologically active molecules such as myosin and actin, of cells such as red blood cells and cardiomyocytes, of subcellular organelles, and of microvessels in vivo. The implications of these findings in the diagnosis and treatment of diseases, as well as potential perspectives that could also benefit from this tool, are also discussed.
Collapse
Affiliation(s)
- Yi Yang
- Department of Cardiology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, China
| | - Zhenhai Fu
- Quantum Sensing Center, Zhejiang Lab, Hangzhou, China
| | - Wei Zhu
- Department of Cardiology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, China
- *Correspondence: Wei Zhu, ; Huizhu Hu, ; Jian’an Wang,
| | - Huizhu Hu
- Quantum Sensing Center, Zhejiang Lab, Hangzhou, China
- State Key Laboratory of Modern Optical Instrumentation, College of Optical Science and Engineering, Zhejiang University, Hangzhou, China
- *Correspondence: Wei Zhu, ; Huizhu Hu, ; Jian’an Wang,
| | - Jian’an Wang
- Department of Cardiology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, China
- *Correspondence: Wei Zhu, ; Huizhu Hu, ; Jian’an Wang,
| |
Collapse
|
17
|
Shah RA, Asatryan B, Sharaf Dabbagh G, Aung N, Khanji MY, Lopes LR, van Duijvenboden S, Holmes A, Muser D, Landstrom AP, Lee AM, Arora P, Semsarian C, Somers VK, Owens AT, Munroe PB, Petersen SE, Chahal CAA. Frequency, Penetrance, and Variable Expressivity of Dilated Cardiomyopathy-Associated Putative Pathogenic Gene Variants in UK Biobank Participants. Circulation 2022; 146:110-124. [PMID: 35708014 PMCID: PMC9375305 DOI: 10.1161/circulationaha.121.058143] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
BACKGROUND There is a paucity of data regarding the phenotype of dilated cardiomyopathy (DCM) gene variants in the general population. We aimed to determine the frequency and penetrance of DCM-associated putative pathogenic gene variants in a general adult population, with a focus on the expression of clinical and subclinical phenotype, including structural, functional, and arrhythmic disease features. METHODS UK Biobank participants who had undergone whole exome sequencing, ECG, and cardiovascular magnetic resonance imaging were selected for study. Three variant-calling strategies (1 primary and 2 secondary) were used to identify participants with putative pathogenic variants in 44 DCM genes. The observed phenotype was graded DCM (clinical or cardiovascular magnetic resonance diagnosis); early DCM features, including arrhythmia or conduction disease, isolated ventricular dilation, and hypokinetic nondilated cardiomyopathy; or phenotype-negative. RESULTS Among 18 665 individuals included in the study, 1463 (7.8%) possessed ≥1 putative pathogenic variant in 44 DCM genes by the main variant calling strategy. A clinical diagnosis of DCM was present in 0.34% and early DCM features in 5.7% of individuals with putative pathogenic variants. ECG and cardiovascular magnetic resonance analysis revealed evidence of subclinical DCM in an additional 1.6% and early DCM features in an additional 15.9% of individuals with putative pathogenic variants. Arrhythmias or conduction disease (15.2%) were the most common early DCM features, followed by hypokinetic nondilated cardiomyopathy (4%). The combined clinical/subclinical penetrance was ≤30% with all 3 variant filtering strategies. Clinical DCM was slightly more prevalent among participants with putative pathogenic variants in definitive/strong evidence genes as compared with those with variants in moderate/limited evidence genes. CONCLUSIONS In the UK Biobank, ≈1 of 6 of adults with putative pathogenic variants in DCM genes exhibited early DCM features potentially associated with DCM genotype, most commonly manifesting with arrhythmias in the absence of substantial ventricular dilation or dysfunction.
Collapse
Affiliation(s)
- Ravi A Shah
- Imperial College Healthcare NHS Trust, London, United Kingdom (R.A.S.)
| | - Babken Asatryan
- Department of Cardiology, Inselspital, Bern University Hospital, University of Bern, Switzerland (B.A.)
| | - Ghaith Sharaf Dabbagh
- Center for Inherited Cardiovascular Diseases, WellSpan Health, Lancaster, PA (G.S.D., C.A.A.C.).,University of Michigan, Division of Cardiovascular Medicine, Ann Arbor (G.S.D.)
| | - Nay Aung
- Barts Heart Centre, St Bartholomew's Hospital, Barts Health NHS Trust, London, United Kingdom (N.A., M.Y.K., L.R.L., A.M.L., S.E.P., C.A.A.C.).,NIHR Barts Biomedical Research Centre, William Harvey Research Institute, Queen Mary University of London, United Kingdom (N.A., M.Y.K., S.v.D., A.M.L., P.B.M., S.E.P.)
| | - Mohammed Y Khanji
- Barts Heart Centre, St Bartholomew's Hospital, Barts Health NHS Trust, London, United Kingdom (N.A., M.Y.K., L.R.L., A.M.L., S.E.P., C.A.A.C.).,NIHR Barts Biomedical Research Centre, William Harvey Research Institute, Queen Mary University of London, United Kingdom (N.A., M.Y.K., S.v.D., A.M.L., P.B.M., S.E.P.)
| | - Luis R Lopes
- Centre for Heart Muscle Disease, Institute of Cardiovascular Science, University College London, United Kingdom (L.R.L.)
| | - Stefan van Duijvenboden
- NIHR Barts Biomedical Research Centre, William Harvey Research Institute, Queen Mary University of London, United Kingdom (N.A., M.Y.K., S.v.D., A.M.L., P.B.M., S.E.P.)
| | | | - Daniele Muser
- Cardiac Electrophysiology, Cardiovascular Division, Hospital of the University of Pennsylvania, Philadelphia (D.M., C.A.A.C.)
| | - Andrew P Landstrom
- Departments of Pediatrics, Division of Cardiology, and Cell Biology, Duke University School of Medicine, Durham, NC (A.P.L.)
| | - Aaron Mark Lee
- NIHR Barts Biomedical Research Centre, William Harvey Research Institute, Queen Mary University of London, United Kingdom (N.A., M.Y.K., S.v.D., A.M.L., P.B.M., S.E.P.)
| | - Pankaj Arora
- Division of Cardiovascular Disease, University of Alabama at Birmingham (P.A.)
| | - Christopher Semsarian
- Agnes Ginges Centre for Molecular Cardiology at Centenary Institute (C.S.), The University of Sydney, New South Wales, Australia.,Sydney Medical School Faculty of Medicine and Health (C.S.), The University of Sydney, New South Wales, Australia.,Department of Cardiology, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia (C.S.)
| | - Virend K Somers
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN (V.K.S., C.A.A.C.)
| | - Anjali T Owens
- Center for Inherited Cardiovascular Disease, Cardiovascular Division, University of Pennsylvania Perelman School of Medicine, Philadelphia (A.T.O.)
| | - Patricia B Munroe
- NIHR Barts Biomedical Research Centre, William Harvey Research Institute, Queen Mary University of London, United Kingdom (N.A., M.Y.K., S.v.D., A.M.L., P.B.M., S.E.P.)
| | - Steffen E Petersen
- NIHR Barts Biomedical Research Centre, William Harvey Research Institute, Queen Mary University of London, United Kingdom (N.A., M.Y.K., S.v.D., A.M.L., P.B.M., S.E.P.)
| | - C Anwar A Chahal
- Center for Inherited Cardiovascular Diseases, WellSpan Health, Lancaster, PA (G.S.D., C.A.A.C.).,Cardiac Electrophysiology, Cardiovascular Division, Hospital of the University of Pennsylvania, Philadelphia (D.M., C.A.A.C.).,Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN (V.K.S., C.A.A.C.)
| | | |
Collapse
|
18
|
Exploring the Potential of Symmetric Exon Deletion to Treat Non-Ischemic Dilated Cardiomyopathy by Removing Frameshift Mutations in TTN. Genes (Basel) 2022; 13:genes13061093. [PMID: 35741855 PMCID: PMC9222585 DOI: 10.3390/genes13061093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 05/30/2022] [Accepted: 06/10/2022] [Indexed: 11/17/2022] Open
Abstract
Non-ischemic dilated cardiomyopathy (DCM) is one of the most frequent pathologies requiring cardiac transplants. Even though the etiology of this disease is complex, frameshift mutations in the giant sarcomeric protein Titin could explain up to 25% of the familial and 18% of the sporadic cases of DCM. Many studies have shown the potential of genome editing using CRISPR/Cas9 to correct truncating mutations in sarcomeric proteins and have established the grounds for myoediting. However, these therapies are still in an immature state, with only few studies showing an efficient treatment of cardiac diseases. This publication hypothesizes that the Titin (TTN)-specific gene structure allows the application of myoediting approaches in a broad range of locations to reframe TTNtvvariants and to treat DCM patients. Additionally, to pave the way for the generation of efficient myoediting approaches for DCM, we screened and selected promising target locations in TTN. We conceptually explored the deletion of symmetric exons as a therapeutic approach to restore TTN’s reading frame in cases of frameshift mutations. We identified a set of 94 potential candidate exons of TTN that we consider particularly suitable for this therapeutic deletion. With this study, we aim to contribute to the development of new therapies to efficiently treat titinopathies and other diseases caused by mutations in genes encoding proteins with modular structures, e.g., Obscurin.
Collapse
|
19
|
Panwar D, Singh KG, Mathur S, Prasad B, Joshi A, Lal V, Thatai A. Heterozygous missense variant in the TTN gene causing Tibial muscular dystrophy. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2022. [DOI: 10.1186/s43042-022-00284-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Tibial muscular dystrophy (TMD), tardive, is a dominantly inherited mild degenerative disorder of anterior tibial muscles. Mutations of Titin (TTN) have been reported in patients with different phenotypes such as skeletal muscular abnormalities or complex overlapping disorders of muscles. Titin (TTN) is a large 363 exon gene that encodes an abundant protein (the longest polypeptide known in nature) expressed in the heart and skeletal muscles.
Methods
DNA from peripheral blood sample was extracted, whole exome sequencing (WES) was performed, and a neuromuscular disorders related gene-filtering strategy was used to analyse the disease-causing mutations. Further, sanger sequencing was applied to confirm the variant.
Results
A novel missense variant (c.41529G > C;p.Arg13843Ser) of TTN gene was identified in a patient with lower limb weakness, occasional tongue fasciculation and mild scoliosis. This variant leads to a substitution of arginine with serine, causing structural changes in titin protein that is responsible for the TMD disease.
Conclusion
The novel variant detected has widened the genetic spectrum of TTN-associated diseases, further functional studies will aid in establishing the clinical diagnosis.
Collapse
|
20
|
Parker LE, Landstrom AP. The clinical utility of pediatric cardiomyopathy genetic testing: From diagnosis to a precision medicine-based approach to care. PROGRESS IN PEDIATRIC CARDIOLOGY 2021; 62. [PMID: 34776723 DOI: 10.1016/j.ppedcard.2021.101413] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Background Pediatric-onset cardiomyopathies are rare yet cause significant morbidity and mortality in affected children. Genetic testing has a major role in the clinical evaluation of pediatric-onset cardiomyopathies, and identification of a variant in an associated gene can be used to confirm the clinical diagnosis and exclude syndromic causes that may warrant different treatment strategies. Further, risk-predictive testing of first-degree relatives can assess who is at-risk of disease and requires continued clinical follow-up. Aim of Review In this review, we seek to describe the current role of genetic testing in the clinical diagnosis and management of patients and families with the five major cardiomyopathies. Further, we highlight the ongoing development of precision-based approaches to diagnosis, prognosis, and treatment. Key Scientific Concepts of Review Emerging application of genotype-phenotype correlations opens the door for genetics to guide a precision medicine-based approach to prognosis and potentially for therapies. Despite advances in our understanding of the genetic etiology of cardiomyopathy and increased accessibility of clinical genetic testing, not all pediatric cardiomyopathy patients have a clear genetic explanation for their disease. Expanded genomic studies are needed to understand the cause of disease in these patients, improve variant classification and genotype-driven prognostic predictions, and ultimately develop truly disease preventing treatment.
Collapse
Affiliation(s)
- Lauren E Parker
- Department of Pediatrics, Division of Cardiology, Duke University School of Medicine, Durham, NC, United States
| | - Andrew P Landstrom
- Department of Pediatrics, Division of Cardiology, Duke University School of Medicine, Durham, NC, United States.,Department of Cell Biology, Duke University School of Medicine, Durham, NC, United States
| |
Collapse
|
21
|
Nguyen TV, Tran Vu MT, Do TNP, Tran THN, Do TH, Nguyen TMH, Tran Huynh BN, Le LA, Nguyen Pham NT, Nguyen TDA, Nguyen TMN, Le NHP, Pham Nguyen V, Ho Huynh TD. Genetic Determinants and Genotype-Phenotype Correlations in Vietnamese Patients With Dilated Cardiomyopathy. Circ J 2021; 85:1469-1478. [PMID: 34011823 DOI: 10.1253/circj.cj-21-0077] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND Dilated cardiomyopathy (DCM) is an important cause of heart failure and cardiac transplantation. This study determined the prevalence of DCM-associated genes and evaluated the genotype-phenotype correlation in Vietnamese patients. METHODS AND RESULTS This study analyzed 58 genes from 230 patients. The study cohort consisted of 64.3% men; age at diagnosis 47.9±13.7 years; familial (10.9%) and sporadic DCM (82.2%). The diagnostic yield was 23.5%, 44.0% in familial and 19.6% in sporadic DCM.TTNtruncating variants (TTNtv) were predominant (46.4%), followed byTPM1,DSP,LMNA,MYBPC3,MYH6,MYH7,DES,TNNT2,ACTC1,ACTN2,BAG3,DMD,FKTN,PLN,TBX5,RBM20,TCAP(2-6%). Familial DCM, genotype-positive andTTNtv-positive patients were younger than those with genotype-negative and sporadic DCM. Genotype-positive patients displayed a decreased systolic blood pressure and left ventricular wall thickness compared to genotype-negative patients. Genotype-positive patients, particularly those withTTNtv, had a family history of DCM, higher left atrial volume index and body mass index, and lower right ventricle-fractional area change than genotype-negative patients. Genotype-positive patients reached the combined outcomes more frequently and at a younger age than genotype-negative patients. Major cardiac events occurred more frequently in patients positive with genes other thanTTNtv. CONCLUSIONS The study findings provided an overview of Vietnamese DCM patients' genetic profile and suggested that management of environmental factors may be beneficial for DCM patients.
Collapse
Affiliation(s)
- Thuy Vy Nguyen
- Department of Genetics, Faculty of Biology and Biotechnology, University of Science, VNUHCM [Vietnam National University, Ho Chi Minh City]
| | | | | | | | | | | | | | | | | | | | - Thi My Nuong Nguyen
- Department of Genetics, Faculty of Biology and Biotechnology, University of Science, VNUHCM [Vietnam National University, Ho Chi Minh City]
| | - Ngoc Hong Phuong Le
- Research Center for Genetics and Reproductive Health, School of Medicine, VNUHCM [Vietnam National University, Ho Chi Minh City]
| | | | - Thuy Duong Ho Huynh
- Department of Genetics, Faculty of Biology and Biotechnology, University of Science, VNUHCM [Vietnam National University, Ho Chi Minh City]
- Research Center for Genetics and Reproductive Health, School of Medicine, VNUHCM [Vietnam National University, Ho Chi Minh City]
- KTEST Science Company
| |
Collapse
|
22
|
The Sarcomeric Spring Protein Titin: Biophysical Properties, Molecular Mechanisms, and Genetic Mutations Associated with Heart Failure and Cardiomyopathy. Curr Cardiol Rep 2021; 23:121. [PMID: 34269900 DOI: 10.1007/s11886-021-01550-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/07/2021] [Indexed: 01/06/2023]
Abstract
PURPOSE OF REVIEW The giant protein titin forms the "elastic" filament of the sarcomere, essential for the mechanical compliance of the heart muscle. Titin serves a biological spring, and therefore structural modifications of titin affect function of the myocardium and are associated with heart failure and cardiomyopathy. RECENT FINDINGS In this review, we discuss the current understanding of titin's biophysical properties and how modifications contribute to cardiac function and heart failure. In addition, we review the most recent data on the clinical impact and phenotype heterogeneity of TTN truncating variants, including diseases involving striated muscles, and prospects for future therapies. Because of the giant structure of the titin protein and the complexity of its function, titin's role in health and disease is not yet completely understood. Future research efforts need to focus on novel therapeutic approaches able to modulate titin transcriptional and post-translational modification.
Collapse
|
23
|
Giri P, Mukhopadhyay A, Gupta M, Mohapatra B. Dilated cardiomyopathy: a new insight into the rare but common cause of heart failure. Heart Fail Rev 2021; 27:431-454. [PMID: 34245424 DOI: 10.1007/s10741-021-10125-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/25/2021] [Indexed: 12/26/2022]
Abstract
Heart failure is a global health burden responsible for high morbidity and mortality with a prevalence of greater than 60 million individuals worldwide. One of the major causes of heart failure is dilated cardiomyopathy (DCM), characterized by associated systolic dysfunction. During the last few decades, there have been remarkable advances in our understanding about the genetics of dilated cardiomyopathy. The genetic causes were initially thought to be associated with mutations in genes encoding proteins that are localized to cytoskeleton and sarcomere only; however, with the advancement in mechanistic understanding, the roles of ion channels, Z-disc, mitochondria, nuclear proteins, cardiac transcription factors (e.g., NKX-2.5, TBX20, GATA4), and the factors involved in calcium homeostasis have also been identified and found to be implicated in both familial and sporadic DCM cases. During past few years, next-generation sequencing (NGS) has been established as a diagnostic tool for genetic analysis and it has added significantly to the existing candidate gene list for DCM. The animal models have also provided novel insights to develop a better treatment strategy based on phenotype-genotype correlation, epigenetic and phenomic profiling. Most of the DCM biomarkers that are used in routine genetic and clinical testing are structural proteins, but during the last few years, the role of mi-RNA has also emerged as a biomarker due to their accessibility through noninvasive methods. Our increasing genetic knowledge can improve the clinical management of DCM by bringing clinicians and geneticists on one platform, thereby influencing the individualized clinical decision making and leading to precision medicine.
Collapse
Affiliation(s)
- Prerna Giri
- Cytogenetics Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Uttar Pradesh, Varanasi-5, India
| | - Amrita Mukhopadhyay
- Cytogenetics Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Uttar Pradesh, Varanasi-5, India
| | - Mohini Gupta
- Cytogenetics Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Uttar Pradesh, Varanasi-5, India
| | - Bhagyalaxmi Mohapatra
- Cytogenetics Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Uttar Pradesh, Varanasi-5, India.
| |
Collapse
|
24
|
Genetic variants associated with inherited cardiovascular disorders among 13,131 asymptomatic older adults of European descent. NPJ Genom Med 2021; 6:51. [PMID: 34135346 PMCID: PMC8209162 DOI: 10.1038/s41525-021-00211-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 05/20/2021] [Indexed: 12/21/2022] Open
Abstract
Genetic testing is used to optimise the management of inherited cardiovascular disorders that can cause sudden cardiac death. Yet more genotype–phenotype correlation studies from populations not ascertained on clinical symptoms or family history of disease are required to improve understanding of gene penetrance. We performed targeted sequencing of 25 genes used routinely in clinical genetic testing for inherited cardiovascular disorders in a population of 13,131 asymptomatic older individuals (mean age 75 years) enrolled in the ASPREE trial. Participants had no prior history of cardiovascular disease events, dementia or physical disability at enrolment. Variants were classified following ACMG/AMP standards. Sudden and rapid cardiac deaths were clinically adjudicated as ASPREE trial endpoints, and assessed during mean 4.7 years of follow-up. In total, 119 participants had pathogenic/deleterious variants in one of the 25 genes analysed (carrier rate of 1 in 110 or 0.9%). Participants carried variants associated with hypertrophic cardiomyopathy (N = 24), dilated cardiomyopathy (N = 29), arrhythmogenic right-ventricular cardiomyopathy (N = 22), catecholaminergic polymorphic ventricular tachycardia (N = 4), aortopathies (N = 1), and long-QT syndrome (N = 39). Among 119 carriers, two died from presumed sudden/rapid cardiac deaths during follow-up (1.7%); both with pathogenic variants in long-QT syndrome genes (KCNQ1, SCN5A). Among non-carriers, the rate of sudden/rapid cardiac deaths was significantly lower (0.08%, 11/12936, p < 0.001). Variants associated with inherited cardiovascular disorders are found in asymptomatic individuals aged 70 years and older without a history of cardiovascular disease.
Collapse
|
25
|
The Combined Human Genotype of Truncating TTN and RBM20 Mutations Is Associated with Severe and Early Onset of Dilated Cardiomyopathy. Genes (Basel) 2021; 12:genes12060883. [PMID: 34201072 PMCID: PMC8228627 DOI: 10.3390/genes12060883] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/01/2021] [Accepted: 06/05/2021] [Indexed: 12/30/2022] Open
Abstract
A major cause of heart failure is cardiomyopathies, with dilated cardiomyopathy (DCM) as the most common form. Over 40 genes are linked to DCM, among them TTN and RBM20. Next Generation Sequencing in clinical DCM cohorts revealed truncating variants in TTN (TTNtv), accounting for up to 25% of familial DCM cases. Mutations in the cardiac splicing factor RNA binding motif protein 20 (RBM20) are also known to be associated with severe cardiomyopathies. TTN is one of the major RBM20 splicing targets. Most of the pathogenic RBM20 mutations are localized in the highly conserved arginine serine rich domain (RS), leading to a cytoplasmic mislocalization of mutant RBM20. Here, we present a patient with an early onset DCM carrying a combination of (likely) pathogenic TTN and RBM20 mutations. We show that the splicing of RBM20 target genes is affected in the mutation carrier. Furthermore, we reveal RBM20 haploinsufficiency presumably caused by the frameshift mutation in RBM20.
Collapse
|
26
|
Magavern EF, Kaski JC, Turner RM, Janmohamed A, Borry P, Pirmohamed M. The Interface of Therapeutics and Genomics in Cardiovascular Medicine. Cardiovasc Drugs Ther 2021; 35:663-676. [PMID: 33528719 PMCID: PMC7851637 DOI: 10.1007/s10557-021-07149-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/19/2021] [Indexed: 01/31/2023]
Abstract
Pharmacogenomics has a burgeoning role in cardiovascular medicine, from warfarin dosing to antiplatelet choice, with recent developments in sequencing bringing the promise of personalised medicine ever closer to the bedside. Further scientific evidence, real-world clinical trials, and economic modelling are needed to fully realise this potential. Additionally, tools such as polygenic risk scores, and results from Mendelian randomisation analyses, are only in the early stages of clinical translation and merit further investigation. Genetically targeted rational drug design has a strong evidence base and, due to the nature of genetic data, academia, direct-to-consumer companies, healthcare systems, and industry may meet in an unprecedented manner. Data sharing navigation may prove problematic. The present manuscript addresses these issues and concludes a need for further guidance to be provided to prescribers by professional bodies to aid in the consideration of such complexities and guide translation of scientific knowledge to personalised clinical action, thereby striving to improve patient care. Additionally, technologic infrastructure equipped to handle such large complex data must be adapted to pharmacogenomics and made user friendly for prescribers and patients alike.
Collapse
Affiliation(s)
- E F Magavern
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- Department of Clinical Pharmacology, Cardiovascular Medicine, Barts Health NHS Trust, London, UK
| | - J C Kaski
- Molecular and Clinical Sciences Research Institute, St George's, University of London, Cranmer Terrace, London, SW17 0RE, UK.
| | - R M Turner
- The Wolfson Centre for Personalised Medicine, Institute of Systems, Molecular and Integrative Biology (ISMIB), University of Liverpool, Liverpool, UK
- Liverpool University Hospitals NHS Foundation Trust, Liverpool, UK
| | - A Janmohamed
- Department of Clinical Pharmacology, St George's, University of London, London, UK
| | - P Borry
- Center for Biomedical Ethics and Law, Department of Public Health and Primary Care, KU Leuven, Leuven, Belgium
- Leuven Institute for Human Genetics and Society, Leuven, Belgium
| | - M Pirmohamed
- The Wolfson Centre for Personalised Medicine, Institute of Systems, Molecular and Integrative Biology (ISMIB), University of Liverpool, Liverpool, UK
- Liverpool University Hospitals NHS Foundation Trust, Liverpool, UK
- Liverpool Health Partners, Liverpool, UK
| |
Collapse
|
27
|
Hershberger RE, Cowan J, Jordan E, Kinnamon DD. The Complex and Diverse Genetic Architecture of Dilated Cardiomyopathy. Circ Res 2021; 128:1514-1532. [PMID: 33983834 DOI: 10.1161/circresaha.121.318157] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Our insight into the diverse and complex nature of dilated cardiomyopathy (DCM) genetic architecture continues to evolve rapidly. The foundations of DCM genetics rest on marked locus and allelic heterogeneity. While DCM exhibits a Mendelian, monogenic architecture in some families, preliminary data from our studies and others suggests that at least 20% to 30% of DCM may have an oligogenic basis, meaning that multiple rare variants from different, unlinked loci, determine the DCM phenotype. It is also likely that low-frequency and common genetic variation contribute to DCM complexity, but neither has been examined within a rare variant context. Other types of genetic variation are also likely relevant for DCM, along with gene-by-environment interaction, now established for alcohol- and chemotherapy-related DCM. Collectively, this suggests that the genetic architecture of DCM is broader in scope and more complex than previously understood. All of this elevates the impact of DCM genetics research, as greater insight into the causes of DCM can lead to interventions to mitigate or even prevent it and thus avoid the morbid and mortal scourge of human heart failure.
Collapse
Affiliation(s)
- Ray E Hershberger
- Divisions of Cardiovascular Medicine (R.E.H.), The Ohio State University Wexner Medical Center, Columbus.,Human Genetics (R.E.H., J.C., E.J., D.D.K.), The Ohio State University Wexner Medical Center, Columbus.,Department of Internal Medicine and the Davis Heart and Lung Research Institute (R.E.H., J.C., E.J., D.D.K.), The Ohio State University Wexner Medical Center, Columbus
| | - Jason Cowan
- Human Genetics (R.E.H., J.C., E.J., D.D.K.), The Ohio State University Wexner Medical Center, Columbus.,Department of Internal Medicine and the Davis Heart and Lung Research Institute (R.E.H., J.C., E.J., D.D.K.), The Ohio State University Wexner Medical Center, Columbus
| | - Elizabeth Jordan
- Human Genetics (R.E.H., J.C., E.J., D.D.K.), The Ohio State University Wexner Medical Center, Columbus.,Department of Internal Medicine and the Davis Heart and Lung Research Institute (R.E.H., J.C., E.J., D.D.K.), The Ohio State University Wexner Medical Center, Columbus
| | - Daniel D Kinnamon
- Human Genetics (R.E.H., J.C., E.J., D.D.K.), The Ohio State University Wexner Medical Center, Columbus.,Department of Internal Medicine and the Davis Heart and Lung Research Institute (R.E.H., J.C., E.J., D.D.K.), The Ohio State University Wexner Medical Center, Columbus
| |
Collapse
|
28
|
Xiao L, Li C, Sun Y, Chen Y, Wei H, Hu D, Yu T, Li X, Jin L, Shi L, Marian AJ, Wang DW. Clinical Significance of Variants in the TTN Gene in a Large Cohort of Patients With Sporadic Dilated Cardiomyopathy. Front Cardiovasc Med 2021; 8:657689. [PMID: 33996946 PMCID: PMC8120103 DOI: 10.3389/fcvm.2021.657689] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Accepted: 03/05/2021] [Indexed: 12/11/2022] Open
Abstract
Background: Mutations in the TTN gene are the most common causes of dilated cardiomyopathy (DCM). The clinical significance of TTN gene variants remains inadequately understood. Methods: Whole-exome sequencing and phenotypic characterisation were performed, and patients were followed up for a median of 44 months. Results: We analyzed the association of the TTN variants with the clinical outcomes in a prospective study of 1,041 patients with sporadic DCM. TTN truncating variants (tTTN) were detected in 120 (11.5%) patients as compared with 2.4/10,000 East Asian populations in the Genome Aggregation Database (GnomAD; p < 0.0001). Pathogenic TTN missense variants were also enriched in DCM as compared with the GnomAD populations (27.6 vs. 5.9%, p < 0.0001). DCM patients with tTTN had a lower left ventricular ejection fraction (28.89 ± 8.72 vs. 31.81 ± 9.97, p = 0.002) and a lower frequency of the left bundle branch block (3.3 vs. 11.3%, p = 0.011) than those without or with mutations in other known causal genes (OCG). However, tTTN were not associated with the composite primary endpoint of cardiac death and heart transplantation during the follow-up period [adjusted hazard ratio (HR): 0.912; 95% confidence interval: 0.464–1.793; p = 0.790]. There was also no sex-dependent effect. Concomitant tTTN and pathogenic variants in OCG were present in only eight DCM patients and did not affect the outcome. Conclusion: The phenotype of DCM caused by tTTN, major causes of sporadic DCM, is not distinctly different from those caused by other causal genes for DCM.
Collapse
Affiliation(s)
- Lei Xiao
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, China
| | - Chenze Li
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, China
| | - Yang Sun
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, China
| | - Yanghui Chen
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, China
| | - Haoran Wei
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, China
| | - Dong Hu
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, China
| | - Ting Yu
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, China
| | - Xianqing Li
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, China
| | - Li Jin
- Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Leming Shi
- Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Ali J Marian
- Center for Cardiovascular Genetics, Houston, TX, United States
| | - Dao Wen Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, China.,Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| |
Collapse
|
29
|
Discovery of TITIN Gene Truncating Variant Mutations and 5-Year Outcomes in Patients With Nonischemic Dilated Cardiomyopathy. Am J Cardiol 2020; 137:97-102. [PMID: 32998006 DOI: 10.1016/j.amjcard.2020.09.026] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 09/11/2020] [Accepted: 09/15/2020] [Indexed: 01/07/2023]
Abstract
Genetic factors play an important role in nonischemic dilated cardiomyopathy (NIDC). However, prime opportunities remain for genetic discovery and prognostic understanding. TITIN gene truncating variant mutations (TTNtv) are of interest because of their frequent appearance in NIDC series. We sought to discover known and novel TTNtv mutations in a NIDC cohort and assess 5-year outcomes. Patients with NIDC entered into the INSPIRE Registry with ≥3 years of follow-up were studied. Whole exome sequencing (WES) was performed using an Illumina Novaseq platform. Genetic analysis used Sentieon software and the GRCh38 human reference genome. Variant calls were annotated with ClinVar. Five-year outcomes were determined by functional assessment and ejection fraction (EF) as recovered (EF ≥50%), persistent (EF 21% to 49%), or progressive (left ventricular assist device, transplant, heart failure [HF] or arrhythmic death, or EF ≤20%). The study comprised 229 NIDC patients (age = 50 ± 15 years, 58% men). TTNtv's were discovered in 27 patients with 22 unique mutations; (7 known, 15 novel). TTNtv+ patients more frequently presented with severe NIDC (EF ≤20%) (p = 0.032). By 5-year, outcomes were worse in TTNtv+ patients (p = 0.027), and patients less often recovered (11% vs. 30%). Prognosis was similar with known and novel mutations. Nongenetic (e.g., environmental) cocausal risk factors for HF were frequently present, and these factors frequently appeared to act in concert with genetic variants to precipitate clinical HF. In conclusion, our study expands the library of likely pathogenic TTN mutations and increases our understanding of their clinical impact in association with other HF risk factors.
Collapse
|
30
|
Qiao Q, Zhao CM, Yang CX, Gu JN, Guo YH, Zhang M, Li RG, Qiu XB, Xu YJ, Yang YQ. Detection and functional characterization of a novel MEF2A variation responsible for familial dilated cardiomyopathy. Clin Chem Lab Med 2020; 59:955-963. [PMID: 33554560 DOI: 10.1515/cclm-2020-1318] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Accepted: 11/25/2020] [Indexed: 12/17/2022]
Abstract
OBJECTIVES Dilated cardiomyopathy (DCM) represents the most frequent form of cardiomyopathy, leading to heart failure, cardiac arrhythmias and death. Accumulating evidence convincingly demonstrates the crucial role of genetic defects in the pathogenesis of DCM, and over 100 culprit genes have been implicated with DCM. However, DCM is of substantial genetic heterogeneity, and the genetic determinants underpinning DCM remain largely elusive. METHODS Whole-exome sequencing and bioinformatical analyses were implemented in a consanguineous Chinese family with DCM. A total of 380 clinically annotated control individuals and 166 more DCM index cases then underwent Sanger sequencing analysis for the identified genetic variation. The functional characteristics of the variant were delineated by utilizing a dual-luciferase assay system. RESULTS A heterozygous variation in the MEF2A gene (encoding myocyte enhancer factor 2A, a transcription factor pivotal for embryonic cardiogenesis and postnatal cardiac adaptation), NM_001365204.1: c.718G>T; p. (Gly240*), was identified, and verified by Sanger sequencing to segregate with autosome-dominant DCM in the family with complete penetrance. The nonsense variation was neither detected in 760 control chromosomes nor found in 166 more DCM probands. Functional analyses revealed that the variant lost transactivation on the validated target genes MYH6 and FHL2, both causally linked to DCM. Furthermore, the variation nullified the synergistic activation between MEF2A and GATA4, another key transcription factor involved in DCM. CONCLUSIONS The findings firstly indicate that MEF2A loss-of-function variation predisposes to DCM in humans, providing novel insight into the molecular mechanisms of DCM and suggesting potential implications for genetic testing and prognostic evaluation of DCM patients.
Collapse
Affiliation(s)
- Qi Qiao
- Department of Cardiology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, P.R. China
| | - Cui-Mei Zhao
- Department of Cardiology, Tongji Hospital, Tongji University School of Medicine, Shanghai, P.R. China
| | - Chen-Xi Yang
- Department of Cardiology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, P.R. China
| | - Jia-Ning Gu
- Department of Cardiology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, P.R. China
| | - Yu-Han Guo
- Department of Cardiology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, P.R. China
| | - Min Zhang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Ruo-Gu Li
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Xing-Biao Qiu
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Ying-Jia Xu
- Department of Cardiology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, P.R. China
| | - Yi-Qing Yang
- Department of Cardiology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, P.R. China.,Cardiovascular Research Laboratory, Shanghai Fifth People's Hospital, Fudan University, Shanghai, P.R. China.,Center Laboratory, Shanghai Fifth People's Hospital, Fudan University, Shanghai, P.R. China
| |
Collapse
|
31
|
Vissing CR, Rasmussen TB, Dybro AM, Olesen MS, Pedersen LN, Jensen M, Bundgaard H, Christensen AH. Dilated cardiomyopathy caused by truncating titin variants: long-term outcomes, arrhythmias, response to treatment and sex differences. J Med Genet 2020; 58:832-841. [PMID: 33106378 DOI: 10.1136/jmedgenet-2020-107178] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 08/24/2020] [Accepted: 09/11/2020] [Indexed: 11/04/2022]
Abstract
BACKGROUND Truncating variants in titin (TTNtv) are the most common cause of dilated cardiomyopathy (DCM). We evaluated the genotype-phenotype correlation in TTNtv-DCM, with a special focus on long-term outcomes, arrhythmias, response to treatment and sex-related presentation. METHODS Data on patient characteristics and outcomes were collected retrospectively from electronic health records of patients genotyped at two Danish heart transplantation centres. RESULTS We included 115 patients (66% men). At diagnosis of DCM, mean age was 46±13 years and left ventricular ejection fraction (LVEF) was 28%±13%. During a median follow-up of 7.9 years, 26% reached a composite outcome of left ventricular assist device implantation, heart transplantation or death. In 20% an arrhythmia preceded the DCM diagnosis. In total, 43% had atrial fibrillation (AF) and 23% had ventricular arrhythmias. Long-term left ventricular reverse remodelling (LVRR; LVEF increase ≥10% points or normalisation) was achieved in 58% and occurred more frequently in women (72% vs 51%, p=0.042).In multivariable proportional hazards analyses, occurrence of LVRR was a strong independent negative predictor of the composite outcome (HR: 0.05 (95% CI 0.02 to 0.14); p<0.001). Female sex independently predicted lower rates of ventricular arrhythmias (HR: 0.33 (95% CI 0.11 to 0.99); p=0.05), while the location of the TTNtv was not associated with cardiovascular outcomes. CONCLUSION DCM caused by TTNtv presented in midlife and was associated with a high burden of AF and ventricular arrhythmias, which often preceded DCM diagnosis. Furthermore, LVRR occurred in a high proportion of patients and was a strong negative predictor of the composite outcome. Female sex was positively associated with occurrence of LVRR and longer event-free survival.
Collapse
Affiliation(s)
- Christoffer Rasmus Vissing
- The Capital Region's Unit for Inherited Cardiac Diseases, Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | | | - Anne Mette Dybro
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
| | - Morten Salling Olesen
- Laboratory of Molecular Cardiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark.,Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Morten Jensen
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
| | - Henning Bundgaard
- The Capital Region's Unit for Inherited Cardiac Diseases, Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Alex Hørby Christensen
- The Capital Region's Unit for Inherited Cardiac Diseases, Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark.,Department of Cardiology, Herlev-Gentofte Hospital, Copenhagen University Hospital, Copenhagen, Denmark
| |
Collapse
|
32
|
Understanding the role of titin in dilated cardiomyopathy. Int J Cardiol 2020; 316:186-187. [DOI: 10.1016/j.ijcard.2020.06.064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 06/29/2020] [Indexed: 11/16/2022]
|
33
|
Barretta F, Mirra B, Monda E, Caiazza M, Lombardo B, Tinto N, Scudiero O, Frisso G, Mazzaccara C. The Hidden Fragility in the Heart of the Athletes: A Review of Genetic Biomarkers. Int J Mol Sci 2020; 21:E6682. [PMID: 32932687 PMCID: PMC7555257 DOI: 10.3390/ijms21186682] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 09/02/2020] [Accepted: 09/08/2020] [Indexed: 12/16/2022] Open
Abstract
Sudden cardiac death (SCD) is a devastating event which can also affect people in apparent good health, such as young athletes. It is known that intense and continuous exercise along with a genetic background that predisposes a person to the risk of fatal arrhythmias is a trigger for SCD. Therefore, knowledge of the athlete's genetic conditions underlying the onset of SCD must be extended, in order to develop new effective prevention and/or therapeutic strategies. Arrhythmic features occur across a broad spectrum of cardiac diseases, sometimes presenting with overlapping phenotypes. The genetic basis of arrhythmogenic disorders has been greatly highlighted in the last 30 years, and has shown marked heterogeneity. The advent of next-generation sequencing has constantly updated our understanding of the genetic basis of arrhythmogenic diseases and is laying the foundation for precision medicine. With the exception of a few clinical cases involving a single athlete showing a highly suspected phenotype for the presence of a heart disease, there are few studies to date that analysed the applicability of genetic testing on cohorts of athletes. This evidence shows that genetic testing can contribute to the diagnosis of up to 13% of athletes; however, the presence of clinical markers is essential. This review aims to provide a reference collection on current knowledge of the genetic basis of sudden cardiac death in athletes and to review updated evidence on the effectiveness of genetic testing in early identification of athletes at risk for SCD.
Collapse
Affiliation(s)
- Ferdinando Barretta
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (F.B.); (B.M.); (B.L.); (N.T.); (O.S.); (C.M.)
- CEINGE Advanced Biotechnologies, 80131 Naples, Italy
| | - Bruno Mirra
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (F.B.); (B.M.); (B.L.); (N.T.); (O.S.); (C.M.)
- CEINGE Advanced Biotechnologies, 80131 Naples, Italy
| | - Emanuele Monda
- Department of Translational Medical Sciences, University of Campania ‘Luigi Vanvitelli’, Monaldi Hospital, 80131 Naples, Italy; (E.M.); (M.C.)
| | - Martina Caiazza
- Department of Translational Medical Sciences, University of Campania ‘Luigi Vanvitelli’, Monaldi Hospital, 80131 Naples, Italy; (E.M.); (M.C.)
| | - Barbara Lombardo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (F.B.); (B.M.); (B.L.); (N.T.); (O.S.); (C.M.)
- CEINGE Advanced Biotechnologies, 80131 Naples, Italy
| | - Nadia Tinto
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (F.B.); (B.M.); (B.L.); (N.T.); (O.S.); (C.M.)
- CEINGE Advanced Biotechnologies, 80131 Naples, Italy
| | - Olga Scudiero
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (F.B.); (B.M.); (B.L.); (N.T.); (O.S.); (C.M.)
- CEINGE Advanced Biotechnologies, 80131 Naples, Italy
| | - Giulia Frisso
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (F.B.); (B.M.); (B.L.); (N.T.); (O.S.); (C.M.)
- CEINGE Advanced Biotechnologies, 80131 Naples, Italy
| | - Cristina Mazzaccara
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (F.B.); (B.M.); (B.L.); (N.T.); (O.S.); (C.M.)
- CEINGE Advanced Biotechnologies, 80131 Naples, Italy
| |
Collapse
|
34
|
Modifications of Titin Contribute to the Progression of Cardiomyopathy and Represent a Therapeutic Target for Treatment of Heart Failure. J Clin Med 2020; 9:jcm9092770. [PMID: 32859027 PMCID: PMC7564493 DOI: 10.3390/jcm9092770] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 08/22/2020] [Accepted: 08/24/2020] [Indexed: 12/20/2022] Open
Abstract
Titin is the largest human protein and an essential component of the cardiac sarcomere. With multiple immunoglobulin(Ig)-like domains that serve as molecular springs, titin contributes significantly to the passive tension, systolic function, and diastolic function of the heart. Mutations leading to early termination of titin are the most common genetic cause of dilated cardiomyopathy. Modifications of titin, which change protein length, and relative stiffness affect resting tension of the ventricle and are associated with acquired forms of heart failure. Transcriptional and post-translational changes that increase titin’s length and extensibility, making the sarcomere longer and softer, are associated with systolic dysfunction and left ventricular dilation. Modifications of titin that decrease its length and extensibility, making the sarcomere shorter and stiffer, are associated with diastolic dysfunction in animal models. There has been significant progress in understanding the mechanisms by which titin is modified. As molecular pathways that modify titin’s mechanical properties are elucidated, they represent therapeutic targets for treatment of both systolic and diastolic dysfunction. In this article, we review titin’s contribution to normal cardiac physiology, the pathophysiology of titin truncation variations leading to dilated cardiomyopathy, and transcriptional and post-translational modifications of titin. Emphasis is on how modification of titin can be utilized as a therapeutic target for treatment of heart failure.
Collapse
|
35
|
Lombardo B, Izzo V, Terracciano D, Ranieri A, Mazzaccara C, Fimiani F, Cesaro A, Gentile L, Leggiero E, Pero R, Izzo B, D'Alicandro AC, Ercolini D, D'Alicandro G, Frisso G, Pastore L, Calabrò P, Scudiero O. Laboratory medicine: health evaluation in elite athletes. Clin Chem Lab Med 2020; 57:1450-1473. [PMID: 30835249 DOI: 10.1515/cclm-2018-1107] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 01/24/2019] [Indexed: 02/06/2023]
Abstract
The need to evaluate the health status of an athlete represents a crucial aim in preventive and protective sports science in order to identify the best diagnostic strategy to improve performance and reduce risks related to physical exercise. In the present review we aim to define the main biochemical and haematological markers that vary significantly during and after sports training to identify risk factors, at competitive and professional levels and to highlight the set up of a specific parameter's panel for elite athletes. Moreover, we also intend to consider additional biomarkers, still under investigation, which could further contribute to laboratory sports medicine and provide reliable data that can be used by athlete's competent staff in order to establish personal attitudes and prevent sports injuries.
Collapse
Affiliation(s)
- Barbara Lombardo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Naples, Italy.,CEINGE Advanced Biotechnologies, Naples, Italy
| | - Viviana Izzo
- Department of Medicine, Surgery and Dentistry, University of Salerno, Salerno, Italy
| | - Daniela Terracciano
- Department of Translational Medical Sciences, University of Naples "Federico II", Naples, Italy
| | - Annaluisa Ranieri
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Naples, Italy.,CEINGE Advanced Biotechnologies, Naples, Italy
| | - Cristina Mazzaccara
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Naples, Italy.,CEINGE Advanced Biotechnologies, Naples, Italy
| | - Fabio Fimiani
- Division of Cardiology, Department of Cardio-Thoracic and Respiratory Sciences, University of Campania 'Luigi Vanvitelli', Naples, Italy
| | - Arturo Cesaro
- Division of Cardiology, Department of Cardio-Thoracic and Respiratory Sciences, University of Campania 'Luigi Vanvitelli', Naples, Italy
| | | | | | - Raffaela Pero
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Naples, Italy.,Task Force on Microbiome Studies, University of Naples "Federico II", Naples, Italy
| | - Barbara Izzo
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, Italy
| | | | - Danilo Ercolini
- Task Force on Microbiome Studies, University of Naples "Federico II", Naples, Italy.,Division of Microbiology, Department of Agricultural Sciences, University of Naples "Federico II", Naples, Italy
| | - Giovanni D'Alicandro
- Department of Neuroscience and Rehabilitation, Center of Sports Medicine and Disability, AORN, Santobono-Pausillipon, Naples, Italy
| | - Giulia Frisso
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Naples, Italy.,CEINGE Advanced Biotechnologies, Naples, Italy
| | - Lucio Pastore
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Naples, Italy.,CEINGE Advanced Biotechnologies, Naples, Italy.,Task Force on Microbiome Studies, University of Naples "Federico II", Naples, Italy
| | - Paolo Calabrò
- Division of Cardiology, Department of Cardio-Thoracic and Respiratory Sciences, University of Campania 'Luigi Vanvitelli', Naples, Italy
| | - Olga Scudiero
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Naples, Italy.,CEINGE Advanced Biotechnologies, Naples, Italy.,Task Force on Microbiome Studies, University of Naples "Federico II", Naples, Italy
| |
Collapse
|
36
|
Di RM, Yang CX, Zhao CM, Yuan F, Qiao Q, Gu JN, Li XM, Xu YJ, Yang YQ. Identification and functional characterization of KLF5 as a novel disease gene responsible for familial dilated cardiomyopathy. Eur J Med Genet 2020; 63:103827. [DOI: 10.1016/j.ejmg.2019.103827] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 10/25/2019] [Accepted: 12/14/2019] [Indexed: 02/08/2023]
|
37
|
Tharp CA, Haywood ME, Sbaizero O, Taylor MRG, Mestroni L. The Giant Protein Titin's Role in Cardiomyopathy: Genetic, Transcriptional, and Post-translational Modifications of TTN and Their Contribution to Cardiac Disease. Front Physiol 2019; 10:1436. [PMID: 31849696 PMCID: PMC6892752 DOI: 10.3389/fphys.2019.01436] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 11/07/2019] [Indexed: 02/06/2023] Open
Abstract
Dilated cardiomyopathy (DCM) is a leading cause of heart failure, sudden cardiac death and heart transplant. DCM is inherited in approximately 50% of cases, in which the most frequent genetic defects are truncation variants of the titin gene (TTNtv). TTN encodes titin, which is the largest protein in the body and is an essential component of the sarcomere. Titin serves as a biological spring, spanning half of the sarcomere and connecting the Z-disk to the M-line, with scaffold and signaling functions. Truncations of titin are believed to lead to either haploinsufficiency and loss-of-function, or to a “poison peptide” effect. However, other titin mechanisms are postulated to influence cardiac function including post-translational modifications, in particular changes in titin phosphorylation that alters the stiffness of the protein, and diversity of alternative splicing that generates different titin isoforms. In this article, we review the role of TTN mutations in development of DCM, how differential expression of titin isoforms relate to DCM pathophysiology, and discuss how post-translational modifications of titin can affect cardiomyocyte function. Current research efforts aim to elucidate the contribution of titin to myofibril assembly, stability, and signal transduction, and how mutant titin leads to cardiac dysfunction and human disease. Future research will need to translate this knowledge toward novel therapeutic approaches that can modulate titin transcriptional and post-translational defects to treat DCM and heart failure.
Collapse
Affiliation(s)
- Charles A Tharp
- Adult Medical Genetics Program and Cardiovascular Institute, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Mary E Haywood
- Adult Medical Genetics Program and Cardiovascular Institute, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Orfeo Sbaizero
- Department of Engineering and Architecture, University of Trieste, Trieste, Italy
| | - Matthew R G Taylor
- Adult Medical Genetics Program and Cardiovascular Institute, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Luisa Mestroni
- Adult Medical Genetics Program and Cardiovascular Institute, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
38
|
An Overview of Circular RNAs and Their Implications in Myotonic Dystrophy. Int J Mol Sci 2019; 20:ijms20184385. [PMID: 31500099 PMCID: PMC6769675 DOI: 10.3390/ijms20184385] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Revised: 09/02/2019] [Accepted: 09/03/2019] [Indexed: 02/07/2023] Open
Abstract
Circular RNAs (circRNAs) are a class of single-stranded covalently closed RNA rings. Biogenesis of circRNAs, which may occur co-transcriptionally and post-transcriptionally via a back-splicing mechanism, requires the presence of complementary and/or inverted repeat sequences in introns flanking back-spliced exons and is facilitated by RNA-binding proteins. CircRNAs are abundant across eukaryotes; however, their biological functions remain largely speculative. Recently, they have been emerging as new members of a gene regulatory network and contributing factors in various human diseases including cancer, neurological, muscular and cardiovascular disorders. In this review, we present an overview of the current knowledge about circRNAs biogenesis and their aberrant expression in various human disorders. In particular, we focus on the latest discovery of circRNAs global upregulation in myotonic dystrophy type 1 (DM1) skeletal muscles and the role these prospective biomarkers might have for prognosis and therapeutic response in DM1.
Collapse
|
39
|
Verdonschot JAJ, Hazebroek MR, Wang P, Sanders-van Wijk S, Merken JJ, Adriaansen YA, van den Wijngaard A, Krapels IPC, Brunner-La Rocca HP, Brunner HG, Heymans SRB. Clinical Phenotype and Genotype Associations With Improvement in Left Ventricular Function in Dilated Cardiomyopathy. Circ Heart Fail 2019; 11:e005220. [PMID: 30571196 DOI: 10.1161/circheartfailure.118.005220] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
BACKGROUND Improvement of left ventricular function (also called left ventricular reverse remodeling [LVRR]) is an important treatment goal in patients with dilated cardiomyopathy (DCM) and hypokinetic non-DCM (HNDC) and is prognostically favorable. We tested whether genetic DCM mutations impact LVRR independent from clinical parameters. METHODS AND RESULTS Patients with DCM and hypokinetic non-DCM (n=346; mean left ventricular ejection fraction, 30%) underwent genotyping for 47 DCM-associated genes in addition to extensive phenotyping. LVRR was defined as improvement of left ventricular ejection fraction >50% or ≥10% absolute increase, with cardiac dimensions (left ventricular end diastolic diameter) ≤33 mm/m2 or ≥10% relative decrease. LVRR occurred in 180 (52%) patients after a median follow-up of 12-month optimal medical treatment. Low baseline left ventricular ejection fraction, a hypokinetic non-DCM phenotype, high systolic blood pressure, absence of a family history of DCM, female sex, absence of atrioventricular block, and treatment with β-blockers were all independent positive clinical predictors of LVRR. With the exception of TTN, genetic mutations were strongly associated with a lower rate of LVRR (odds ratio, 0.19 [0.09-0.42]; P<0.0001). TTN and LMNA were independently associated with LVRR (odds ratio, 2.49 [1.09-6.20]; P=0.038 and 0.11 [0.01-0.99]; P=0.049, respectively). Adding mutation status significantly improved discrimination (C statistics) and reclassification (integrated discrimination improvement/net reclassification index) of the clinical model predicting LVRR. Furthermore, the risk for heart failure hospitalization and cardiovascular death is lower in the LVRR patients on the long term (hazard ratio, 0.47 [0.24-0.91]; P=0.009 and 0.18 [0.04-0.82]; P=0.007, respectively), and LVRR is an independent predictor for event-free survival. CONCLUSIONS The genetic substrate is associated with the clinical course and long-term prognosis of patients with DCM/hypokinetic non-DCM.
Collapse
Affiliation(s)
- Job A J Verdonschot
- Department of Cardiology, Maastricht University Medical Centre, the Netherlands (J.A.J.V., M.R.H., S.S.-v.W., J.J.M., H.-P.B.-L.R., S.R.B.H.).,Department of Clinical Genetics, Maastricht University Medical Centre, the Netherlands (J.A.J.V., P.W., Y.A.A., A.v.d.W., I.P.C.K., H.G.B.)
| | - Mark R Hazebroek
- Department of Cardiology, Maastricht University Medical Centre, the Netherlands (J.A.J.V., M.R.H., S.S.-v.W., J.J.M., H.-P.B.-L.R., S.R.B.H.)
| | - Ping Wang
- Department of Clinical Genetics, Maastricht University Medical Centre, the Netherlands (J.A.J.V., P.W., Y.A.A., A.v.d.W., I.P.C.K., H.G.B.)
| | - Sandra Sanders-van Wijk
- Department of Cardiology, Maastricht University Medical Centre, the Netherlands (J.A.J.V., M.R.H., S.S.-v.W., J.J.M., H.-P.B.-L.R., S.R.B.H.)
| | - Jort J Merken
- Department of Cardiology, Maastricht University Medical Centre, the Netherlands (J.A.J.V., M.R.H., S.S.-v.W., J.J.M., H.-P.B.-L.R., S.R.B.H.)
| | - Yvonne A Adriaansen
- Department of Clinical Genetics, Maastricht University Medical Centre, the Netherlands (J.A.J.V., P.W., Y.A.A., A.v.d.W., I.P.C.K., H.G.B.)
| | - Arthur van den Wijngaard
- Department of Clinical Genetics, Maastricht University Medical Centre, the Netherlands (J.A.J.V., P.W., Y.A.A., A.v.d.W., I.P.C.K., H.G.B.)
| | - Ingrid P C Krapels
- Department of Clinical Genetics, Maastricht University Medical Centre, the Netherlands (J.A.J.V., P.W., Y.A.A., A.v.d.W., I.P.C.K., H.G.B.)
| | - Hans-Peter Brunner-La Rocca
- Department of Cardiology, Maastricht University Medical Centre, the Netherlands (J.A.J.V., M.R.H., S.S.-v.W., J.J.M., H.-P.B.-L.R., S.R.B.H.)
| | - Han G Brunner
- Department of Clinical Genetics, Maastricht University Medical Centre, the Netherlands (J.A.J.V., P.W., Y.A.A., A.v.d.W., I.P.C.K., H.G.B.).,Department of Human Genetics, Donders Center for Neuroscience, Radboudumc, Nijmegen, the Netherlands (H.G.B.)
| | - Stephane R B Heymans
- Department of Cardiology, Maastricht University Medical Centre, the Netherlands (J.A.J.V., M.R.H., S.S.-v.W., J.J.M., H.-P.B.-L.R., S.R.B.H.).,Department of Cardiovascular Research, University of Leuven, Belgium (S.R.B.H.).,Netherlands Heart Institute, Utrecht (S.R.B.H.)
| |
Collapse
|
40
|
Tabish AM, Arif M, Song T, Elbeck Z, Becker RC, Knöll R, Sadayappan S. Association of intronic DNA methylation and hydroxymethylation alterations in the epigenetic etiology of dilated cardiomyopathy. Am J Physiol Heart Circ Physiol 2019; 317:H168-H180. [PMID: 31026178 PMCID: PMC6692731 DOI: 10.1152/ajpheart.00758.2018] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 04/15/2019] [Accepted: 04/22/2019] [Indexed: 01/03/2023]
Abstract
In this study, we investigated the role of DNA methylation [5-methylcytosine (5mC)] and 5-hydroxymethylcytosine (5hmC), epigenetic modifications that regulate gene activity, in dilated cardiomyopathy (DCM). A MYBPC3 mutant mouse model of DCM was compared with wild type and used to profile genomic 5mC and 5hmC changes by Chip-seq, and gene expression levels were analyzed by RNA-seq. Both 5mC-altered genes (957) and 5hmC-altered genes (2,022) were identified in DCM hearts. Diverse gene ontology and KEGG pathways were enriched for DCM phenotypes, such as inflammation, tissue fibrosis, cell death, cardiac remodeling, cardiomyocyte growth, and differentiation, as well as sarcomere structure. Hierarchical clustering of mapped genes affected by 5mC and 5hmC clearly differentiated DCM from wild-type phenotype. Based on these data, we propose that genomewide 5mC and 5hmC contents may play a major role in DCM pathogenesis. NEW & NOTEWORTHY Our data demonstrate that development of dilated cardiomyopathy in mice is associated with significant epigenetic changes, specifically in intronic regions, which, when combined with gene expression profiling data, highlight key signaling pathways involved in pathological cardiac remodeling and heart contractile dysfunction.
Collapse
Affiliation(s)
- Ali M Tabish
- Integrated Cardio-Metabolic Centre, Karolinska Institutet , Stockholm , Sweden
| | - Mohammed Arif
- Heart, Lung, Vascular Institute, Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati , Cincinnati, Ohio
| | - Taejeong Song
- Heart, Lung, Vascular Institute, Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati , Cincinnati, Ohio
| | - Zaher Elbeck
- Integrated Cardio-Metabolic Centre, Karolinska Institutet , Stockholm , Sweden
| | - Richard C Becker
- Heart, Lung, Vascular Institute, Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati , Cincinnati, Ohio
| | - Ralph Knöll
- Integrated Cardio-Metabolic Centre, Karolinska Institutet , Stockholm , Sweden
- Cardiovascular and Metabolic Disease Innovative Medicines and Early Development Unit, AstraZeneca R&D, Gothenburg , Sweden
| | - Sakthivel Sadayappan
- Heart, Lung, Vascular Institute, Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati , Cincinnati, Ohio
| |
Collapse
|
41
|
Müller D, Hagenah D, Biswanath S, Coffee M, Kampmann A, Zweigerdt R, Heisterkamp A, Kalies SMK. Femtosecond laser-based nanosurgery reveals the endogenous regeneration of single Z-discs including physiological consequences for cardiomyocytes. Sci Rep 2019; 9:3625. [PMID: 30842507 PMCID: PMC6403391 DOI: 10.1038/s41598-019-40308-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 02/13/2019] [Indexed: 11/24/2022] Open
Abstract
A highly organized cytoskeleton architecture is the basis for continuous and controlled contraction in cardiomyocytes (CMs). Abnormalities in cytoskeletal elements, like the Z-disc, are linked to several diseases. It is challenging to reveal the mechanisms of CM failure, endogenous repair, or mechanical homeostasis on the scale of single cytoskeletal elements. Here, we used a femtosecond (fs) laser to ablate single Z-discs in human pluripotent stem cells (hPSC) -derived CMs (hPSC-CM) and neonatal rat CMs. We show, that CM viability was unaffected by the loss of a single Z-disc. Furthermore, more than 40% of neonatal rat and 68% of hPSC-CMs recovered the Z-disc loss within 24 h. Significant differences to control cells, after the Z-disc loss, in terms of cell perimeter, x- and y-expansion and calcium homeostasis were not found. Only 14 days in vitro old hPSC-CMs reacted with a significant decrease in cell area, x- and y-expansion 24 h past nanosurgery. This demonstrates that CMs can compensate the loss of a single Z-disc and recover a regular sarcomeric pattern during spontaneous contraction. It also highlights the significant potential of fs laser-based nanosurgery to physically micro manipulate CMs to investigate cytoskeletal functions and organization of single elements.
Collapse
Affiliation(s)
- Dominik Müller
- Institute of Quantum Optics, Leibniz University Hannover, Hannover, Germany. .,REBIRTH-Cluster of Excellence, Hannover Medical School, Hannover, Germany. .,Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), Hannover, Germany.
| | - Dorian Hagenah
- Institute of Quantum Optics, Leibniz University Hannover, Hannover, Germany.,REBIRTH-Cluster of Excellence, Hannover Medical School, Hannover, Germany.,Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), Hannover, Germany
| | - Santoshi Biswanath
- REBIRTH-Cluster of Excellence, Hannover Medical School, Hannover, Germany.,Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiac, Thoracic, Transplantation and Vascular Surgery (HTTG), Hannover Medical, School, Hannover, Germany
| | - Michelle Coffee
- REBIRTH-Cluster of Excellence, Hannover Medical School, Hannover, Germany.,Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiac, Thoracic, Transplantation and Vascular Surgery (HTTG), Hannover Medical, School, Hannover, Germany
| | - Andreas Kampmann
- Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), Hannover, Germany.,Clinic for Cranio-Maxillo-Facial Surgery, Hannover Medical School, Hannover, Germany
| | - Robert Zweigerdt
- REBIRTH-Cluster of Excellence, Hannover Medical School, Hannover, Germany.,Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiac, Thoracic, Transplantation and Vascular Surgery (HTTG), Hannover Medical, School, Hannover, Germany
| | - Alexander Heisterkamp
- Institute of Quantum Optics, Leibniz University Hannover, Hannover, Germany.,REBIRTH-Cluster of Excellence, Hannover Medical School, Hannover, Germany.,Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), Hannover, Germany
| | - Stefan M K Kalies
- Institute of Quantum Optics, Leibniz University Hannover, Hannover, Germany.,REBIRTH-Cluster of Excellence, Hannover Medical School, Hannover, Germany.,Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), Hannover, Germany
| |
Collapse
|
42
|
van der Velden J, Stienen GJM. Cardiac Disorders and Pathophysiology of Sarcomeric Proteins. Physiol Rev 2019; 99:381-426. [PMID: 30379622 DOI: 10.1152/physrev.00040.2017] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The sarcomeric proteins represent the structural building blocks of heart muscle, which are essential for contraction and relaxation. During recent years, it has become evident that posttranslational modifications of sarcomeric proteins, in particular phosphorylation, tune cardiac pump function at rest and during exercise. This delicate, orchestrated interaction is also influenced by mutations, predominantly in sarcomeric proteins, which cause hypertrophic or dilated cardiomyopathy. In this review, we follow a bottom-up approach starting from a description of the basic components of cardiac muscle at the molecular level up to the various forms of cardiac disorders at the organ level. An overview is given of sarcomere changes in acquired and inherited forms of cardiac disease and the underlying disease mechanisms with particular reference to human tissue. A distinction will be made between the primary defect and maladaptive/adaptive secondary changes. Techniques used to unravel functional consequences of disease-induced protein changes are described, and an overview of current and future treatments targeted at sarcomeric proteins is given. The current evidence presented suggests that sarcomeres not only form the basis of cardiac muscle function but also represent a therapeutic target to combat cardiac disease.
Collapse
Affiliation(s)
- Jolanda van der Velden
- Amsterdam UMC, Vrije Universiteit Amsterdam, Physiology, Amsterdam Cardiovascular Sciences, Amsterdam , The Netherlands ; and Department of Physiology, Kilimanjaro Christian Medical University College, Moshi, Tanzania
| | - Ger J M Stienen
- Amsterdam UMC, Vrije Universiteit Amsterdam, Physiology, Amsterdam Cardiovascular Sciences, Amsterdam , The Netherlands ; and Department of Physiology, Kilimanjaro Christian Medical University College, Moshi, Tanzania
| |
Collapse
|
43
|
Mazzaccara C, Limongelli G, Petretta M, Vastarella R, Pacileo G, Bonaduce D, Salvatore F, Frisso G. A common polymorphism in the SCN5A gene is associated with dilated cardiomyopathy. J Cardiovasc Med (Hagerstown) 2019; 19:344-350. [PMID: 29782370 PMCID: PMC6012048 DOI: 10.2459/jcm.0000000000000670] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Aims SCN5A is a disease-causing gene associated with familial dilated cardiomyopathy (FDC). We examined the possible association between a common polymorphism in the SCN5A gene (c.1673A>G-p.H558R; rs1805124) and the risk of dilated cardiomyopathy (DCM) occurrence. Methods We genotyped 185 DCM cases (familial DCM, idiopathic DCM and postischemic DCM) and 251 controls for the p.H558R polymorphism in the SCN5A gene, to test the association of the molecular epidemiology of the individuals with the presence/absence of various types of DCM. Results Our results showed that the rs1805124 polymorphism was significantly associated with DCM, and the association was more significant in patients with FDC; furthermore, in these individuals, the less frequent GG genotype was associated with a 7.39-fold increased risk of disease [95% confidence interval (95% CI) = 2.88–18.96; P < 0.0001] compared with the AA genotype. Moreover, logistic regression analysis showed that GG carriers had a higher risk of DCM than AA + AG carriers (odds ratio = 5.45, 95% CI = 2.23–13.35; P < 0.001). No association was observed between the rs1805124 and DCM risk in postischemic DCM patients. Conclusion Our study demonstrates an association between familial DCM and the rs1805124 polymorphism in the SCN5A gene, which may unravel additional genetic predisposition to the development of a multifactorial disease as DCM.
Collapse
Affiliation(s)
- Cristina Mazzaccara
- CEINGE-Biotecnologie Avanzate s.c.a r.l.,Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli 'Federico II'
| | - Giuseppe Limongelli
- Dipartimento di Scienze Cardiotoraciche e Respiratorie, Università degli Studi della Campania 'Luigi Vanvitelli', A.O. Monaldi, Azienda dei Colli
| | - Mario Petretta
- Dipartimento di Scienze Mediche Traslazionali, Università di Napoli 'Federico II'
| | - Rossella Vastarella
- Dipartimento di Scienze Cardiotoraciche e Respiratorie, Università degli Studi della Campania 'Luigi Vanvitelli', A.O. Monaldi, Azienda dei Colli
| | - Giuseppe Pacileo
- Dipartimento di Scienze Cardiotoraciche e Respiratorie, Università degli Studi della Campania 'Luigi Vanvitelli', A.O. Monaldi, Azienda dei Colli
| | - Domenico Bonaduce
- Dipartimento di Scienze Mediche Traslazionali, Università di Napoli 'Federico II'
| | - Francesco Salvatore
- CEINGE-Biotecnologie Avanzate s.c.a r.l.,IRCCS-Fondazione SDN, Napoli, Italy
| | - Giulia Frisso
- CEINGE-Biotecnologie Avanzate s.c.a r.l.,Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli 'Federico II'
| |
Collapse
|
44
|
Knyazeva A, Krutikov A, Golovkin A, Mishanin A, Pavlov G, Smolina N, Hushkina A, Sejersen T, Sjoberg G, Galagudza M, Kostareva A. Time- and Ventricular-Specific Expression Profiles of Genes Encoding Z-Disk Proteins in Pressure Overload Model of Left Ventricular Hypertrophy. Front Genet 2019; 9:684. [PMID: 30666270 PMCID: PMC6330284 DOI: 10.3389/fgene.2018.00684] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 12/07/2018] [Indexed: 01/20/2023] Open
Abstract
Mechanotransduction is an essential mechanism of transforming external mechanical stimulus to biochemical response. In cardiomyocytes mechanotransduction plays an important role in contraction, stretch sensing and homeostasis regulation. One of the major mechanosensitive area in cardiomyocytes, the Z-disk, consists of numbers of structural and signaling proteins, that may undergo conformational or gene expression changes under pathological stress conditions. In present study we examined a rat model of pressure overload cardiac hypertrophy validated by echocardiographic and histopathological examinations. We revealed, that during hypertrophy progression expression of several genes encoding Z-disk proteins (Actn2, Ldb3, Cmya5, Nebl) is different at early and late points of cardiac remodeling. Moreover, expression patterns of several genes are opposite in myocardium of overloaded left ventricle and hemodynamically unaffected right ventricle, and expression profiles in interventricular septum are more similar to right ventricle. Additionally, we revealed inconsistencies between mRNA and protein level changes of Actn2, one of the major structural Z-disk element. All these findings point out, that investigated Z-disk proteins participate in pathological stress adaptation through undergoing the gene expression changes, and suggest the novel important role of hypertrophic response modulation during different stages of cardiac remodeling.
Collapse
Affiliation(s)
| | | | - Alexey Golovkin
- Almazov National Medical Research Centre, Saint Petersburg, Russia
| | | | - Georgii Pavlov
- Saint Petersburg State Academy of Veterinary Medicine, Saint Petersburg, Russia
| | - Natalia Smolina
- Almazov National Medical Research Centre, Saint Petersburg, Russia.,Information Technologies and Programming Faculty, ITMO University, Saint Petersburg, Russia
| | | | - Thomas Sejersen
- Department of Women's and Children's Health, Center for Molecular Medicine, Karolinska Institute, Stockholm, Sweden
| | - Gunnar Sjoberg
- Department of Women's and Children's Health, Center for Molecular Medicine, Karolinska Institute, Stockholm, Sweden
| | | | - Anna Kostareva
- Almazov National Medical Research Centre, Saint Petersburg, Russia.,Information Technologies and Programming Faculty, ITMO University, Saint Petersburg, Russia
| |
Collapse
|
45
|
Interpretation of Genomic Sequencing Results in Healthy and Ill Newborns: Results from the BabySeq Project. Am J Hum Genet 2019; 104:76-93. [PMID: 30609409 DOI: 10.1016/j.ajhg.2018.11.016] [Citation(s) in RCA: 183] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 11/23/2018] [Indexed: 12/27/2022] Open
Abstract
Genomic sequencing provides many opportunities in newborn clinical care, but the challenges of interpreting and reporting newborn genomic sequencing (nGS) results need to be addressed for its broader and effective application. The BabySeq Project is a pilot randomized clinical trial that explores the medical, behavioral, and economic impacts of nGS in well newborns and those admitted to a neonatal intensive care unit (NICU). Here we present childhood-onset and actionable adult-onset disease risk, carrier status, and pharmacogenomics findings from nGS of 159 newborns in the BabySeq Project. nGS revealed a risk of childhood-onset disease in 15/159 (9.4%) newborns; none of the disease risks were anticipated based on the infants' known clinical or family histories. nGS also revealed actionable adult-onset disease risk in 3/85 (3.5%) newborns whose parents consented to receive this information. Carrier status for recessive diseases and pharmacogenomics variants were reported in 88% and 5% of newborns, respectively. Additional indication-based analyses were performed in 29/32 (91%) NICU newborns and 6/127 (5%) healthy newborns who later had presentations that prompted a diagnostic analysis. No variants that sufficiently explained the reason for the indications were identified; however, suspicious but uncertain results were reported in five newborns. Testing parental samples contributed to the interpretation and reporting of results in 13/159 (8%) newborns. Our results suggest that nGS can effectively detect risk and carrier status for a wide range of disorders that are not detectable by current newborn screening assays or predicted based on the infant's known clinical or family history, and the interpretation of results can substantially benefit from parental testing.
Collapse
|
46
|
Abstract
Genetic testing has an increasingly important role in the diagnosis and management of cardiac disorders, where it confirms the diagnosis, aids prognostication and risk stratification and guides treatment. A genetic diagnosis in the proband also enables clarification of the risk for family members by cascade testing. Genetics in cardiac disorders is complex where epigenetic and environmental factors might come into interplay. Incomplete penetrance and variable expressivity is also common. Genetic results in cardiac conditions are mostly probabilistic and should be interpreted with all available clinical information. With this complexity in cardiac genetics, testing is only indicated in patients with a strong suspicion of an inheritable cardiac disorder after a full clinical evaluation. In this review we discuss the genetics underlying the major cardiomyopathies and channelopathies, and the practical aspects of diagnosing these conditions in the laboratory.
Collapse
|
47
|
Identification and Functional Characterization of an ISL1 Mutation Predisposing to Dilated Cardiomyopathy. J Cardiovasc Transl Res 2018; 12:257-267. [DOI: 10.1007/s12265-018-9851-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Accepted: 12/03/2018] [Indexed: 02/06/2023]
|
48
|
|
49
|
Takotsubo as Initial Manifestation of Non-Myopathic Cardiomyopathy Due to the Titin Variant c.1489G > T. MEDICINES 2018; 5:medicines5030080. [PMID: 30061524 PMCID: PMC6164847 DOI: 10.3390/medicines5030080] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 06/20/2018] [Accepted: 07/13/2018] [Indexed: 11/19/2022]
Abstract
Background: Whether patients with subclinical cardiomyopathy (CMP) are more prone to experience Takotsubo syndrome (TTS) than patients without CMP, is unknown. We present a patient with TTS as the initial manifestation of a hitherto unrecognized genetic CMP. Method: case report. Results: At age 55 after the unexpected death of her father, a now 61-year-old female had developed precordial pressure. Work-up revealed moderately reduced systolic function, dyskinesia of the interventricular septum, and indications for a TTS. Coronary angiography was normal but ventriculography showed TTS. Cardiac MRI confirmed reduced systolic function and TTS. TTS resolved without treatment and sequelae. At age 57 atrial fibrillation was recorded. After deterioration of systolic function at age 59 dilated CMP was diagnosed. Despite application of levosimendan, sacubitril, valsartan, and ivabradine, complete remission could not be achieved. Upon genetic work-up by means of a gene panel, the heterozygous mutation c.1489G > T (p. E497X) in exon 9 of the titin gene was detected and made responsible for the phenotype. Neurological work-up precluded involvement of the skeletal muscles. The further course was complicated by ventricular arrhythmias, requiring implantation of an implantable cardioverter defibrillator (ICD). Conclusions: previously subclinical CMP may initially manifest as TTS. Since patients with titin CMP are at risk of developing ventricular arrhythmias and thus to experience sudden cardiac death, appropriate anti-arrhythmic therapy needs to be established.
Collapse
|
50
|
Stöhr EJ, Takayama H, Ferrari G. Stretch your heart-but not too far: The role of titin mutations in dilated cardiomyopathy. J Thorac Cardiovasc Surg 2018; 156:209-214. [PMID: 29685583 PMCID: PMC6724204 DOI: 10.1016/j.jtcvs.2017.10.160] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 09/29/2017] [Accepted: 10/24/2017] [Indexed: 12/23/2022]
Affiliation(s)
- Eric J Stöhr
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY; School of Health Sciences, Cardiff Metropolitan University, Cardiff, United Kingdom.
| | - Hiroo Takayama
- Division of Cardiothoracic and Vascular Surgery, Department of Surgery, Columbia University Irving Medical Center, New York, NY
| | - Giovanni Ferrari
- Division of Cardiothoracic and Vascular Surgery, Department of Surgery, Columbia University Irving Medical Center, New York, NY
| |
Collapse
|