1
|
Wang P, Ahmed MS, Nguyen NUN, Menendez-Montes I, Hsu CC, Farag AB, Thet S, Lam NT, Wansapura JP, Crossley E, Ma N, Zhao SR, Zhang T, Morimoto S, Singh R, Elhelaly W, Tassin TC, Cardoso AC, Williams NS, Pointer HL, Elliott DA, McNamara JW, Watt KI, Porrello ER, Sadayappan S, Sadek HA. An FDA-approved drug structurally and phenotypically corrects the K210del mutation in genetic cardiomyopathy models. J Clin Invest 2025; 135:e174081. [PMID: 39959972 PMCID: PMC11827848 DOI: 10.1172/jci174081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 11/19/2024] [Indexed: 02/20/2025] Open
Abstract
Dilated cardiomyopathy (DCM) due to genetic disorders results in decreased myocardial contractility, leading to high morbidity and mortality rates. There are several therapeutic challenges in treating DCM, including poor understanding of the underlying mechanism of impaired myocardial contractility and the difficulty of developing targeted therapies to reverse mutation-specific pathologies. In this report, we focused on K210del, a DCM-causing mutation, due to 3-nucleotide deletion of sarcomeric troponin T (TnnT), resulting in loss of Lysine210. We resolved the crystal structure of the troponin complex carrying the K210del mutation. K210del induced an allosteric shift in the troponin complex resulting in distortion of activation Ca2+-binding domain of troponin C (TnnC) at S69, resulting in calcium discoordination. Next, we adopted a structure-based drug repurposing approach to identify bisphosphonate risedronate as a potential structural corrector for the mutant troponin complex. Cocrystallization of risedronate with the mutant troponin complex restored the normal configuration of S69 and calcium coordination. Risedronate normalized force generation in K210del patient-induced pluripotent stem cell-derived (iPSC-derived) cardiomyocytes and improved calcium sensitivity in skinned papillary muscles isolated from K210del mice. Systemic administration of risedronate to K210del mice normalized left ventricular ejection fraction. Collectively, these results identify the structural basis for decreased calcium sensitivity in K210del and highlight structural and phenotypic correction as a potential therapeutic strategy in genetic cardiomyopathies.
Collapse
Affiliation(s)
- Ping Wang
- Division of Cardiology, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Mahmoud Salama Ahmed
- Division of Cardiology, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, Amarillo, Texas, USA
| | - Ngoc Uyen Nhi Nguyen
- Division of Cardiology, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Ivan Menendez-Montes
- Division of Cardiology, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Ching-Cheng Hsu
- Division of Cardiology, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Ayman B. Farag
- Division of Cardiology, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Suwannee Thet
- Division of Cardiology, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Nicholas T. Lam
- Division of Cardiology, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | | | - Eric Crossley
- Department of Pharmacology, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Ning Ma
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California, USA
| | - Shane Rui Zhao
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California, USA
| | - Tiejun Zhang
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California, USA
| | - Sachio Morimoto
- School of Health Sciences Fukuoka, International University of Health and Welfare, Okawa, Japan
| | - Rohit Singh
- Heart, Lung and Vascular Institute, Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati, Cincinnati, Ohio, USA
| | - Waleed Elhelaly
- Division of Cardiology, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Tara C. Tassin
- Division of Cardiology, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Alisson C. Cardoso
- Biosciences National Laboratory, Brazilian Center for Research in Energy and Materials, Campinas, Brazil
| | - Noelle S. Williams
- Department of Pharmacology, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Hayley L. Pointer
- Novo Nordisk Foundation Center for Stem Cell Medicine, Murdoch Children’s Research Institute, Royal Children’s Hospital, Parkville, Victoria, Australia
| | - David A. Elliott
- Novo Nordisk Foundation Center for Stem Cell Medicine, Murdoch Children’s Research Institute, Royal Children’s Hospital, Parkville, Victoria, Australia
- Department of Paediatrics, and
| | - James W. McNamara
- Novo Nordisk Foundation Center for Stem Cell Medicine, Murdoch Children’s Research Institute, Royal Children’s Hospital, Parkville, Victoria, Australia
- Department of Anatomy and Physiology, University of Melbourne, Parkville, Victoria, Australia
| | - Kevin I. Watt
- Novo Nordisk Foundation Center for Stem Cell Medicine, Murdoch Children’s Research Institute, Royal Children’s Hospital, Parkville, Victoria, Australia
- Department of Anatomy and Physiology, University of Melbourne, Parkville, Victoria, Australia
| | - Enzo R. Porrello
- Novo Nordisk Foundation Center for Stem Cell Medicine, Murdoch Children’s Research Institute, Royal Children’s Hospital, Parkville, Victoria, Australia
- Department of Paediatrics, and
- Department of Anatomy and Physiology, University of Melbourne, Parkville, Victoria, Australia
| | - Sakthivel Sadayappan
- Heart, Lung and Vascular Institute, Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati, Cincinnati, Ohio, USA
- Department of Cellular & Molecular Medicine, The University of Arizona College of Medicine, Tucson, Arizona, USA
| | - Hesham A. Sadek
- Division of Cardiology, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Division of Cardiology, The University of Arizona College of Medicine, Tucson, Arizona, USA
- The University of Arizona Sarver Heart Center, Tucson, Arizona, USA
| |
Collapse
|
2
|
Kiessling M, Djalinac N, Voglhuber J, Ljubojevic-Holzer S. Nuclear Calcium in Cardiac (Patho)Physiology: Small Compartment, Big Impact. Biomedicines 2023; 11:biomedicines11030960. [PMID: 36979939 PMCID: PMC10046765 DOI: 10.3390/biomedicines11030960] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 03/17/2023] [Indexed: 03/30/2023] Open
Abstract
The nucleus of a cardiomyocyte has been increasingly recognized as a morphologically distinct and partially independent calcium (Ca2+) signaling microdomain, with its own Ca2+-regulatory mechanisms and important effects on cardiac gene expression. In this review, we (1) provide a comprehensive overview of the current state of research on the dynamics and regulation of nuclear Ca2+ signaling in cardiomyocytes, (2) address the role of nuclear Ca2+ in the development and progression of cardiac pathologies, such as heart failure and atrial fibrillation, and (3) discuss novel aspects of experimental methods to investigate nuclear Ca2+ handling and its downstream effects in the heart. Finally, we highlight current challenges and limitations and recommend future directions for addressing key open questions.
Collapse
Affiliation(s)
- Mara Kiessling
- Department of Cardiology, Medical University of Graz, 8036 Graz, Austria
| | - Nataša Djalinac
- Department of Biology, University of Padua, 35122 Padova, Italy
| | - Julia Voglhuber
- Department of Cardiology, Medical University of Graz, 8036 Graz, Austria
- BioTechMed Graz, 8010 Graz, Austria
| | - Senka Ljubojevic-Holzer
- Department of Cardiology, Medical University of Graz, 8036 Graz, Austria
- BioTechMed Graz, 8010 Graz, Austria
- Gottfried Schatz Research Center, Division of Molecular Biology and Biochemistry, Medical University of Graz, 8010 Graz, Austria
| |
Collapse
|
3
|
Kitaoka H, Tsutsui H, Kubo T, Ide T, Chikamori T, Fukuda K, Fujino N, Higo T, Isobe M, Kamiya C, Kato S, Kihara Y, Kinugawa K, Kinugawa S, Kogaki S, Komuro I, Hagiwara N, Ono M, Maekawa Y, Makita S, Matsui Y, Matsushima S, Sakata Y, Sawa Y, Shimizu W, Teraoka K, Tsuchihashi-Makaya M, Ishibashi-Ueda H, Watanabe M, Yoshimura M, Fukusima A, Hida S, Hikoso S, Imamura T, Ishida H, Kawai M, Kitagawa T, Kohno T, Kurisu S, Nagata Y, Nakamura M, Morita H, Takano H, Shiga T, Takei Y, Yuasa S, Yamamoto T, Watanabe T, Akasaka T, Doi Y, Kimura T, Kitakaze M, Kosuge M, Takayama M, Tomoike H. JCS/JHFS 2018 Guideline on the Diagnosis and Treatment of Cardiomyopathies. Circ J 2021; 85:1590-1689. [PMID: 34305070 DOI: 10.1253/circj.cj-20-0910] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Hiroaki Kitaoka
- Department of Cardiology and Geriatrics, Kochi Medical School, Kochi University
| | | | - Toru Kubo
- Department of Cardiology and Geriatrics, Kochi Medical School, Kochi University
| | - Tomomi Ide
- Department of Cardiovascular Medicine, Kyushu University
| | | | - Keiichi Fukuda
- Department of Cardiology, Keio University School of Medicine
| | - Noboru Fujino
- Department of Cardiovascular and Internal Medicine, Kanazawa University, Graduate School of Medical Science
| | - Taiki Higo
- Department of Cardiovascular Medicine, Kyushu University Graduate School of Medical Sciences
| | | | - Chizuko Kamiya
- Department of Perinatology and Gynecology, National Cerebral and Cardiovascular Center
| | - Seiya Kato
- Division of Pathology, Saiseikai Fukuoka General Hospital
| | | | | | | | - Shigetoyo Kogaki
- Department of Pediatrics and Neonatology, Osaka General Medical Center
| | - Issei Komuro
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo
| | | | - Minoru Ono
- Department of Cardiac Surgery, The University of Tokyo Hospital
| | - Yuichiro Maekawa
- Division of Cardiology, Internal Medicine III, Hamamatsu University School of Medicine
| | - Shigeru Makita
- Department of Cardiac Rehabilitation, Saitama International Medical Center, Saitama Medical University
| | - Yoshiro Matsui
- Department of Cardiac Surgery, Hanaoka Seishu Memorial Hospital
| | | | - Yasushi Sakata
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine
| | - Yoshiki Sawa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine
| | - Wataru Shimizu
- Department of Cardiovascular Medicine, Nippon Medical School
| | | | | | | | - Masafumi Watanabe
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University Faculty of Medicine
| | - Michihiro Yoshimura
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine
| | | | - Satoshi Hida
- Department of Cardiovascular Medicine, Tokyo Medical University
| | - Shungo Hikoso
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine
| | | | | | - Makoto Kawai
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine
| | - Toshiro Kitagawa
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences
| | - Takashi Kohno
- Department of Cardiovascular Medicine, Kyorin University School of Medicine
| | - Satoshi Kurisu
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences
| | - Yoji Nagata
- Division of Cardiology, Fukui CardioVascular Center
| | - Makiko Nakamura
- Second Department of Internal Medicine, University of Toyama
| | - Hiroyuki Morita
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo
| | - Hitoshi Takano
- Department of Cardiovascular Medicine, Nippon Medical School Hospital
| | - Tsuyoshi Shiga
- Department of Clinical Pharmacology and Therapeutics, The Jikei University School of Medicine
| | | | - Shinsuke Yuasa
- Department of Cardiology, Keio University School of Medicine
| | - Teppei Yamamoto
- Department of Cardiovascular Medicine, Nippon Medical School
| | - Tetsu Watanabe
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University Faculty of Medicine
| | - Takashi Akasaka
- Department of Cardiovascular Medicine, Wakayama Medical University
| | | | - Takeshi Kimura
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine
| | | | - Masami Kosuge
- Division of Cardiology, Yokohama City University Medical Center
| | | | | | | |
Collapse
|
4
|
Chen YJ, Chien CS, Chiang CE, Chen CH, Cheng HM. From Genetic Mutations to Molecular Basis of Heart Failure Treatment: An Overview of the Mechanism and Implication of the Novel Modulators for Cardiac Myosin. Int J Mol Sci 2021; 22:6617. [PMID: 34205587 PMCID: PMC8234187 DOI: 10.3390/ijms22126617] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 06/11/2021] [Accepted: 06/17/2021] [Indexed: 02/06/2023] Open
Abstract
Heart failure (HF) is a syndrome encompassing several important etiologies that lead to the imbalance between oxygen demand and supply. Despite the usage of guideline-directed medical therapy for HF has shown better outcomes, novel therapeutic strategies are desirable, especially for patients with preserved or mildly reduced left ventricular ejection fraction. In this regard, understanding the molecular basis for cardiomyopathies is expected to fill in the knowledge gap and generate new therapies to improve prognosis for HF. This review discusses an evolutionary mechanism designed to regulate cardiac contraction and relaxation through the most often genetically determined cardiomyopathies associated with HF. In addition, both the myosin inhibitor and myosin activator are promising new treatments for cardiomyopathies. A comprehensive review from genetic mutations to the molecular basis of direct sarcomere modulators will help shed light on future studies for a better characterization of HF etiologies and potential therapeutic targets.
Collapse
Affiliation(s)
- Yu-Jen Chen
- Department of Internal Medicine, Division of Cardiovascular Medicine, Wan Fang Hospital, Taipei Medical University, Taipei 116081, Taiwan;
- Department of Internal Medicine, Division of Cardiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110301, Taiwan
- Institute of Public Health, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
| | - Chian-Shiu Chien
- Innovative Cellular Therapy Center, Department of Medical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan;
| | - Chern-En Chiang
- General Clinical Research Center, Taipei Veterans General Hospital, Taipei 112201, Taiwan;
- Department of Medicine, National Yang Ming Chiao Tung University College of Medicine, Taipei 112304, Taiwan
| | - Chen-Huan Chen
- Department of Medical Education, Taipei Veterans General Hospital, Taipei 112201, Taiwan;
- College of Medicine, National Yang Ming Chiao Tung University, Taipei 112201, Taiwan
| | - Hao-Min Cheng
- Institute of Public Health, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
- Department of Medicine, National Yang Ming Chiao Tung University College of Medicine, Taipei 112304, Taiwan
- Center for Evidence-Based Medicine, Taipei Veterans General Hospital, Taipei 112201, Taiwan
| |
Collapse
|
5
|
Li B, Guo Y, Zhan Y, Zhou X, Li Y, Zhao C, Sun N, Xu C, Liang Q. Cardiac Overexpression of XIN Prevents Dilated Cardiomyopathy Caused by TNNT2 ΔK210 Mutation. Front Cell Dev Biol 2021; 9:691749. [PMID: 34222259 PMCID: PMC8247596 DOI: 10.3389/fcell.2021.691749] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 05/25/2021] [Indexed: 11/13/2022] Open
Abstract
TNNT2 mutation is associated with a range of cardiac diseases, including dilated cardiomyopathy (DCM). However, the mechanisms underlying the development of DCM and heart failure remain incompletely understood. In the present study, we found the expression of cardiac XIN protein was reduced in TNNT2-ΔK210 hESCs-derived cardiomyocytes and mouse heart tissues. We further investigated whether XIN protects against TNNT2 mutation-induced DCM. Overexpression of the repeat-containing isoform XINB decreased the percentage of myofilaments disorganization and increased cell contractility of TNNT2-ΔK210 cardiomyocytes. Moreover, overexpression of XINB by heart-specific delivery via AAV9 ameliorates DCM remodeling caused by TNNT2-ΔK210 mutation in mice, revealed by partially reversed cardiac dilation, systolic dysfunction and heart fibrosis. These results suggest that deficiency of XIN may play a critical role in the development of DCM. Consequently, our findings may provide a new mechanistic insight and represent a therapeutic target for the treatment of idiopathic DCM.
Collapse
Affiliation(s)
- Bin Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
- Shanghai Institute of Precision Medicine, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yifan Guo
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yongkun Zhan
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Xinyan Zhou
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yongbo Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Chao Zhao
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Ning Sun
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Chen Xu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Qianqian Liang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
6
|
Sarcomeric Gene Variants and Their Role with Left Ventricular Dysfunction in Background of Coronary Artery Disease. Biomolecules 2020; 10:biom10030442. [PMID: 32178433 PMCID: PMC7175236 DOI: 10.3390/biom10030442] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Accepted: 03/11/2020] [Indexed: 12/18/2022] Open
Abstract
: Cardiovascular diseases are one of the leading causes of death in developing countries, generally originating as coronary artery disease (CAD) or hypertension. In later stages, many CAD patients develop left ventricle dysfunction (LVD). Left ventricular ejection fraction (LVEF) is the most prevalent prognostic factor in CAD patients. LVD is a complex multifactorial condition in which the left ventricle of the heart becomes functionally impaired. Various genetic studies have correlated LVD with dilated cardiomyopathy (DCM). In recent years, enormous progress has been made in identifying the genetic causes of cardiac diseases, which has further led to a greater understanding of molecular mechanisms underlying each disease. This progress has increased the probability of establishing a specific genetic diagnosis, and thus providing new opportunities for practitioners, patients, and families to utilize this genetic information. A large number of mutations in sarcomeric genes have been discovered in cardiomyopathies. In this review, we will explore the role of the sarcomeric genes in LVD in CAD patients, which is a major cause of cardiac failure and results in heart failure.
Collapse
|
7
|
Kresin N, Stücker S, Krämer E, Flenner F, Mearini G, Münch J, Patten M, Redwood C, Carrier L, Friedrich FW. Analysis of Contractile Function of Permeabilized Human Hypertrophic Cardiomyopathy Multicellular Heart Tissue. Front Physiol 2019; 10:239. [PMID: 30984009 PMCID: PMC6447666 DOI: 10.3389/fphys.2019.00239] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 02/22/2019] [Indexed: 01/08/2023] Open
Affiliation(s)
- Nico Kresin
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Sabrina Stücker
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Elisabeth Krämer
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Frederik Flenner
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Giulia Mearini
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Julia Münch
- University Heart Center Hamburg, Hamburg, Germany
| | | | - Charles Redwood
- Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Lucie Carrier
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Felix W Friedrich
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| |
Collapse
|
8
|
Tikunova SB, Cuesta A, Price M, Li MX, Belevych N, Biesiadecki BJ, Reiser PJ, Hwang PM, Davis JP. 3-Chlorodiphenylamine activates cardiac troponin by a mechanism distinct from bepridil or TFP. J Gen Physiol 2018; 151:9-17. [PMID: 30442775 PMCID: PMC6314390 DOI: 10.1085/jgp.201812131] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 10/02/2018] [Indexed: 01/14/2023] Open
Abstract
Cardiac troponin activators could be beneficial in systolic heart failure. Tikunova et al. demonstrate that, unlike previously known calcium sensitizers, the small molecule 3-chlorodiphenylamine does not activate isolated cardiac troponin C but instead activates the intact troponin complex. Despite extensive efforts spanning multiple decades, the development of highly effective Ca2+ sensitizers for the heart remains an elusive goal. Existing Ca2+ sensitizers have other targets in addition to cardiac troponin (cTn), which can lead to adverse side effects, such as hypotension or arrhythmias. Thus, there is a need to design Ca2+-sensitizing drugs with higher affinity and selectivity for cTn. Previously, we determined that many compounds based on diphenylamine (DPA) were able to bind to a cTnC–cTnI chimera with moderate affinity (Kd ∼10–120 µM). Of these compounds, 3-chlorodiphenylamine (3-Cl-DPA) bound most tightly (Kd of 10 µM). Here, we investigate 3-Cl-DPA further and find that it increases the Ca2+ sensitivity of force development in skinned cardiac muscle. Using NMR, we show that, like the known Ca2+ sensitizers, trifluoperazine (TFP) and bepridil, 3-Cl-DPA is able to bind to the isolated N-terminal domain (N-domain) of cTnC (Kd of 6 µM). However, while the bulky molecules of TFP and bepridil stabilize the open state of the N-domain of cTnC, the small and flexible 3-Cl-DPA molecule is able to bind without stabilizing this open state. Thus, unlike TFP, which drastically slows the rate of Ca2+ dissociation from the N-domain of isolated cTnC in a dose-dependent manner, 3-Cl-DPA has no effect on the rate of Ca2+ dissociation. On the other hand, the affinity of 3-Cl-DPA for a cTnC–TnI chimera is at least an order of magnitude higher than that of TFP or bepridil, likely because 3-Cl-DPA is less disruptive of cTnI binding to cTnC. Therefore, 3-Cl-DPA has a bigger effect on the rate of Ca2+ dissociation from the entire cTn complex than TFP and bepridil. Our data suggest that 3-Cl-DPA activates the cTn complex via a unique mechanism and could be a suitable scaffold for the development of novel treatments for systolic heart failure.
Collapse
Affiliation(s)
- Svetlana B Tikunova
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH
| | - Andres Cuesta
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH
| | - Morgan Price
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH
| | - Monica X Li
- Departments of Medicine and Biochemistry, University of Alberta, Edmonton, AB, Canada
| | - Natalya Belevych
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH
| | | | - Peter J Reiser
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH
| | - Peter M Hwang
- Departments of Medicine and Biochemistry, University of Alberta, Edmonton, AB, Canada
| | - Jonathan P Davis
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH
| |
Collapse
|
9
|
Tikunova S, Belevych N, Doan K, Reiser PJ. Desensitizing mouse cardiac troponin C to calcium converts slow muscle towards a fast muscle phenotype. J Physiol 2018; 596:4651-4663. [PMID: 29992562 PMCID: PMC6166084 DOI: 10.1113/jp276296] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 06/27/2018] [Indexed: 12/11/2022] Open
Abstract
KEY POINTS The Ca2+ -desensitizing D73N mutation in slow skeletal/cardiac troponin C caused dilatated cardiomyopathy in mice, but the consequences of this mutation in skeletal muscle were not known. The D73N mutation led to a rightward shift in the force versus pCa (-log [Ca]) relationship in slow-twitch mouse fibres. The D73N mutation led to a rightward shift in the force-stimulation frequency relationship and reduced fatigue resistance of mouse soleus muscle. The D73N mutation led to reduced cross-sectional area of slow-twitch fibres in mouse soleus muscle without affecting fibre type composition of the muscle. The D73N mutation resulted in significantly shorter times to peak force and to relaxation during isometric twitches and tetani in mouse soleus muscle. The D73N mutation led to major changes in physiological properties of mouse soleus muscle, converting slow muscle toward a fast muscle phenotype. ABSTRACT The missense mutation, D73N, in mouse cardiac troponin C has a profound impact on cardiac function, mediated by a decreased myofilament Ca2+ sensitivity. Mammalian cardiac muscle and slow skeletal muscle normally share expression of the same troponin C isoform. Therefore, the objective of this study was to determine the consequences of the D73N mutation in skeletal muscle, as a potential mechanism that contributes to the morbidity associated with heart failure or other conditions in which Ca2+ sensitivity might be altered. Effects of the D73N mutation on physiological properties of mouse soleus muscle, in which slow-twitch fibres are prevalent, were examined. The mutation resulted in a rightward shift of the force-stimulation frequency relationship, and significantly faster kinetics of isometric twitches and tetani in isolated soleus muscle. Furthermore, soleus muscles from D73N mice underwent a significantly greater reduction in force during a fatigue test. The mutation significantly reduced slow fibre mean cross-sectional area without affecting soleus fibre type composition. The effects of the mutation on Ca2+ sensitivity of force development in soleus skinned slow and fast fibres were also examined. As expected, the D73N mutation did not affect the Ca2+ sensitivity of force development in fast fibres but resulted in substantially decreased Ca2+ sensitivity in slow fibres. The results demonstrate that a point mutation in a single constituent of myofilaments (slow/cardiac troponin C) led to major changes in physiological properties of skeletal muscle and converted slow muscle toward a fast muscle phenotype with reduced fatigue resistance and Ca2+ sensitivity of force generation.
Collapse
Affiliation(s)
- Svetlana Tikunova
- Department of Physiology and Cell BiologyCollege of MedicineColumbusOH 43210USA
| | - Natalya Belevych
- Division of Biosciences, College of DentistryOhio State UniversityColumbusOH 43210USA
| | - Kelly Doan
- Division of Biosciences, College of DentistryOhio State UniversityColumbusOH 43210USA
| | - Peter J. Reiser
- Division of Biosciences, College of DentistryOhio State UniversityColumbusOH 43210USA
| |
Collapse
|
10
|
Hong L, Lai H, Fang Y, Tao Y, Qiu Y. Silencing CTGF/CCN2 inactivates the MAPK signaling pathway to alleviate myocardial fibrosis and left ventricular hypertrophy in rats with dilated cardiomyopathy. J Cell Biochem 2018; 119:9519-9531. [PMID: 30129221 DOI: 10.1002/jcb.27268] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Accepted: 06/26/2018] [Indexed: 01/04/2023]
Affiliation(s)
- Lang Hong
- 2nd Department of Cardiology Jiangxi Provincial People’s Hospital Nanchang China
| | - Heng‐Li Lai
- 2nd Department of Cardiology Jiangxi Provincial People’s Hospital Nanchang China
| | - Yan Fang
- 2nd Department of Cardiology Jiangxi Provincial People’s Hospital Nanchang China
| | - Yu Tao
- 2nd Department of Cardiology Jiangxi Provincial People’s Hospital Nanchang China
| | - Yun Qiu
- 2nd Department of Cardiology Jiangxi Provincial People’s Hospital Nanchang China
| |
Collapse
|
11
|
Incidence, Clinical Characteristics, and Long-term Outcome of the Dilated Phase of Hypertrophic Cardiomyopathy. Keio J Med 2018; 68:87-94. [DOI: 10.2302/kjm.2018-0004-oa] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
12
|
Johnston JR, Chase PB, Pinto JR. Troponin through the looking-glass: emerging roles beyond regulation of striated muscle contraction. Oncotarget 2017; 9:1461-1482. [PMID: 29416706 PMCID: PMC5787451 DOI: 10.18632/oncotarget.22879] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 11/20/2017] [Indexed: 01/03/2023] Open
Abstract
Troponin is a heterotrimeric Ca2+-binding protein that has a well-established role in regulating striated muscle contraction. However, mounting evidence points to novel cellular functions of troponin, with profound implications in cancer, cardiomyopathy pathogenesis and skeletal muscle aging. Here, we highlight the non-canonical roles and aberrant expression patterns of troponin beyond the sarcomeric milieu. Utilizing bioinformatics tools and online databases, we also provide pathway, subcellular localization, and protein-protein/DNA interaction analyses that support a role for troponin in multiple subcellular compartments. This emerging knowledge challenges the conventional view of troponin as a sarcomere-specific protein exclusively involved in muscle contraction and may transform the way we think about sarcomeric proteins, particularly in the context of human disease and aging.
Collapse
Affiliation(s)
- Jamie R Johnston
- Department of Biomedical Sciences, The Florida State University College of Medicine, Tallahassee, FL, 32306-4300, USA
| | - P Bryant Chase
- Department of Biological Science, The Florida State University, Tallahassee, FL, 32306-4370, USA
| | - Jose Renato Pinto
- Department of Biomedical Sciences, The Florida State University College of Medicine, Tallahassee, FL, 32306-4300, USA
| |
Collapse
|
13
|
Phenotypic expression of a novel desmin gene mutation: hypertrophic cardiomyopathy followed by systemic myopathy. J Hum Genet 2017; 63:249-254. [DOI: 10.1038/s10038-017-0383-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 10/07/2017] [Accepted: 10/15/2017] [Indexed: 11/08/2022]
|
14
|
Stücker S, Kresin N, Carrier L, Friedrich FW. Nebivolol Desensitizes Myofilaments of a Hypertrophic Cardiomyopathy Mouse Model. Front Physiol 2017; 8:558. [PMID: 28824454 PMCID: PMC5539082 DOI: 10.3389/fphys.2017.00558] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 07/17/2017] [Indexed: 01/19/2023] Open
Abstract
Background: Hypertrophic cardiomyopathy (HCM) patients often present with diastolic dysfunction and a normal to supranormal systolic function. To counteract this hypercontractility, guideline therapies advocate treatment with beta-adrenoceptor and Ca2+ channel blockers. One well established pathomechanism for the hypercontractile phenotype frequently observed in HCM patients and several HCM mouse models is an increased myofilament Ca2+ sensitivity. Nebivolol, a commonly used beta-adrenoceptor antagonist, has been reported to lower maximal force development and myofilament Ca2+ sensitivity in rabbit and human heart tissues. The aim of this study was to evaluate the effect of nebivolol in cardiac muscle strips of an established HCM Mybpc3 mouse model. Furthermore, we investigated actions of nebivolol and epigallocatechin-gallate, which has been shown to desensitize myofilaments for Ca2+ in mouse and human HCM models, in cardiac strips of HCM patients with a mutation in the most frequently mutated HCM gene MYBPC3. Methods and Results: Nebivolol effects were tested on contractile parameters and force-Ca2+ relationship of skinned ventricular muscle strips isolated from Mybpc3-targeted knock-in (KI), wild-type (WT) mice and cardiac strips of three HCM patients with MYBPC3 mutations. At baseline, KI strips showed no difference in maximal force development compared to WT mouse heart strips. Neither 1 nor 10 μM nebivolol had an effect on maximal force development in both genotypes. 10 μM nebivolol induced myofilament Ca2+ desensitization in WT strips and to a greater extent in KI strips. Neither 1 nor 10 μM nebivolol had an effect on Ca2+ sensitivity in cardiac muscle strips of three HCM patients with MYBPC3 mutations, whereas epigallocatechin-gallate induced a right shift in the force-Ca2+ curve. Conclusion: Nebivolol induced a myofilament Ca2+ desensitization in both WT and KI strips, which was more pronounced in KI muscle strips. In human cardiac muscle strips of three HCM patients nebivolol had no effect on myofilament Ca2+ sensitivity.
Collapse
Affiliation(s)
- Sabrina Stücker
- Department of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-EppendorfHamburg, Germany.,German Centre for Cardiovascular Research (DZHK)Hamburg, Germany
| | - Nico Kresin
- Department of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-EppendorfHamburg, Germany.,German Centre for Cardiovascular Research (DZHK)Hamburg, Germany
| | - Lucie Carrier
- Department of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-EppendorfHamburg, Germany.,German Centre for Cardiovascular Research (DZHK)Hamburg, Germany
| | - Felix W Friedrich
- Department of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-EppendorfHamburg, Germany.,German Centre for Cardiovascular Research (DZHK)Hamburg, Germany
| |
Collapse
|
15
|
Na I, Kong MJ, Straight S, Pinto JR, Uversky VN. Troponins, intrinsic disorder, and cardiomyopathy. Biol Chem 2017; 397:731-51. [PMID: 27074551 DOI: 10.1515/hsz-2015-0303] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 04/05/2016] [Indexed: 11/15/2022]
Abstract
Cardiac troponin is a dynamic complex of troponin C, troponin I, and troponin T (TnC, TnI, and TnT, respectively) found in the myocyte thin filament where it plays an essential role in cardiac muscle contraction. Mutations in troponin subunits are found in inherited cardiomyopathies, such as hypertrophic cardiomyopathy (HCM) and dilated cardiomyopathy (DCM). The highly dynamic nature of human cardiac troponin and presence of numerous flexible linkers in its subunits suggest that understanding of structural and functional properties of this important complex can benefit from the consideration of the protein intrinsic disorder phenomenon. We show here that mutations causing decrease in the disorder score in TnI and TnT are significantly more abundant in HCM and DCM than mutations leading to the increase in the disorder score. Identification and annotation of intrinsically disordered regions in each of the troponin subunits conducted in this study can help in better understanding of the roles of intrinsic disorder in regulation of interactomes and posttranslational modifications of these proteins. These observations suggest that disease-causing mutations leading to a decrease in the local flexibility of troponins can trigger a whole plethora of functional changes in the heart.
Collapse
|
16
|
Friedrich FW, Flenner F, Nasib M, Eschenhagen T, Carrier L. Epigallocatechin-3-Gallate Accelerates Relaxation and Ca 2+ Transient Decay and Desensitizes Myofilaments in Healthy and Mybpc3-Targeted Knock-in Cardiomyopathic Mice. Front Physiol 2016; 7:607. [PMID: 27994558 PMCID: PMC5136558 DOI: 10.3389/fphys.2016.00607] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 11/22/2016] [Indexed: 11/13/2022] Open
Abstract
Background: Hypertrophic cardiomyopathy (HCM) is the most common inherited cardiac muscle disease with left ventricular hypertrophy, interstitial fibrosis and diastolic dysfunction. Increased myofilament Ca2+ sensitivity could be the underlying cause of diastolic dysfunction. Epigallocatechin-3-gallate (EGCg), a catechin found in green tea, has been reported to decrease myofilament Ca2+ sensitivity in HCM models with troponin mutations. However, whether this is also the case for HCM-associated thick filament mutations is not known. Therefore, we evaluated whether EGCg affects the behavior of cardiomyocytes and myofilaments of an HCM mouse model carrying a gene mutation in cardiac myosin-binding protein C and exhibiting both increased myofilament Ca2+ sensitivity and diastolic dysfunction. Methods and Results: Acute effects of EGCg were tested on fractional sarcomere shortening and Ca2+ transients in intact ventricular myocytes and on force-Ca2+ relationship of skinned ventricular muscle strips isolated from Mybpc3-targeted knock-in (KI) and wild-type (WT) mice. Fractional sarcomere shortening and Ca2+ transients were analyzed at 37°C under 1-Hz pacing in the absence or presence of EGCg (1.8 μM). At baseline and in the absence of Fura-2, KI cardiomyocytes displayed lower diastolic sarcomere length, higher fractional sarcomere shortening, longer time to peak shortening and time to 50% relengthening than WT cardiomyocytes. In WT and KI neither diastolic sarcomere length nor fractional sarcomere shortening were influenced by EGCg treatment, but relaxation time was reduced, to a greater extent in KI cells. EGCg shortened time to peak Ca2+ and Ca2+ transient decay in Fura-2-loaded WT and KI cardiomyocytes. EGCg did not influence phosphorylation of phospholamban. In skinned cardiac muscle strips, EGCg (30 μM) decreased Ca2+ sensitivity in both groups. Conclusion: EGCg hastened relaxation and Ca2+ transient decay to a larger extent in KI than in WT cardiomyocytes. This effect could be partially explained by myofilament Ca2+ desensitization.
Collapse
Affiliation(s)
- Felix W Friedrich
- Cardiovascular Research Center, Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-EppendorfHamburg, Germany; German Centre for Cardiovascular Research (DZHK)Hamburg, Germany
| | - Frederik Flenner
- Cardiovascular Research Center, Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-EppendorfHamburg, Germany; German Centre for Cardiovascular Research (DZHK)Hamburg, Germany
| | - Mahtab Nasib
- Cardiovascular Research Center, Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-EppendorfHamburg, Germany; German Centre for Cardiovascular Research (DZHK)Hamburg, Germany
| | - Thomas Eschenhagen
- Cardiovascular Research Center, Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-EppendorfHamburg, Germany; German Centre for Cardiovascular Research (DZHK)Hamburg, Germany
| | - Lucie Carrier
- Cardiovascular Research Center, Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-EppendorfHamburg, Germany; German Centre for Cardiovascular Research (DZHK)Hamburg, Germany
| |
Collapse
|
17
|
Fucikova A, Lenco J, Tambor V, Rehulkova H, Pudil R, Stulik J. Plasma concentration of fibronectin is decreased in patients with hypertrophic cardiomyopathy. Clin Chim Acta 2016; 463:62-66. [DOI: 10.1016/j.cca.2016.09.024] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 09/11/2016] [Accepted: 09/28/2016] [Indexed: 11/30/2022]
|
18
|
Häggström J, Luis Fuentes V, Wess G. Screening for hypertrophic cardiomyopathy in cats. J Vet Cardiol 2016; 17 Suppl 1:S134-49. [PMID: 26776573 DOI: 10.1016/j.jvc.2015.07.003] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Revised: 06/27/2015] [Accepted: 07/01/2015] [Indexed: 11/17/2022]
Abstract
Hypertrophic cardiomyopathy (HCM) is the most common heart disease in cats, and it can lead to increased morbidity and mortality. Cats are often screened for HCM because of the presence of a heart murmur, but screening for breeding purposes has also become common. These cats are usually purebred cats of breeding age, and generally do not present with severe disease or with any clinical signs. This type of screening is particularly challenging because mild disease may be difficult to differentiate from a normal phenotype, and the margin for error is small, with potentially major consequences for the breeder. This article reviews HCM screening methods, with particular emphasis on echocardiography.
Collapse
Affiliation(s)
- Jens Häggström
- Department of Clinical Sciences, Faculty of Veterinary Medicine and Animal Science, Swedish University of Agricultural Sciences, Box 7054, Uppsala, Sweden.
| | - Virginia Luis Fuentes
- The Royal Veterinary College, Department of Clinical Science and Services, Hawkshead Lane, Hatfield AL9 7TA, United Kingdom
| | - Gerhard Wess
- Clinic of Small Animal Medicine, LMU University, Veterinaerstr. 13, 80539 Munich, Germany
| |
Collapse
|
19
|
Koh W, Wong C, Tang WHW. Genetic Predispositions to Heart Failure. CURRENT CARDIOVASCULAR RISK REPORTS 2016. [DOI: 10.1007/s12170-016-0525-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
20
|
Kumar S, Mishra A, Srivastava A, Bhatt M, Garg N, Agarwal SK, Pande S, Mittal B. Role of common sarcomeric gene polymorphisms in genetic susceptibility to left ventricular dysfunction. J Genet 2016; 95:263-272. [PMID: 27350668 DOI: 10.1007/s12041-016-0623-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Mutations in sarcomeric genes are common genetic cause of cardiomyopathies. An intronic 25-bp deletion in cardiac myosin binding protein C (MYBPC3) at 3' region is associated with dilated and hypertrophic cardiomyopathies in Southeast Asia. However, the frequency of sarcomeric gene polymorphisms and associated clinical presentation have not been established with left ventricular dysfunction (LVD). Therefore, the aim of the present study was to explore the association of MYBPC3 25-bp deletion, titin (TTN) 18 bp I/D, troponin T type 2 (TNNT2) 5 bp I/D and myospryn K2906N polymorphisms with LVD. This study includes 988 consecutive patients with angiographically confirmed coronary artery disease (CAD) and 300 healthy controls. Among the 988 CAD patients, 253 with reduced left ventricle ejection fraction (LVEF≤45%) were categorized as LVD. MYBPC3 25-bp deletion, TTN 18 bp I/D and TNNT2 5 bp I/D polymorphisms were determined by direct polymerase chain reaction method, while myospryn K2906N polymorphism by TaqMan assay. Our results showed that MYBPC3 25-bp deletion polymorphism was significantly associated with elevated risk of LVD (LVEF <45) (healthy controls versus LVD: OR=3.85, P <0.001; and nonLVD versus LVD: OR=1.65, P = 0.035), while TTN 18 bp I/D, TNNT2 5 bp I/D and myospryn K2906N polymorphisms did not show any significant association with LVD. The results also showed that MYBPC3 25-bp deletion polymorphism was significantly associated with other parameters of LV remodelling, i.e. LV dimensions (LV end diastole dimension, LVEDD: P = 0.037 and LV end systolic dimension, LVESD: P = 0.032). Our data suggests that MYBPC3 25-bp deletion may play significant role in conferring LVD as well as CAD risk in north Indian population.
Collapse
Affiliation(s)
- Surendra Kumar
- Department of Genetics, Sanjay Gandhi Postgraduate Institute of Medical Sciences (SGPGIMS), Lucknow 226 014, India.
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Scheid LM, Mosqueira M, Hein S, Kossack M, Juergensen L, Mueller M, Meder B, Fink RH, Katus HA, Hassel D. Essential light chain S195 phosphorylation is required for cardiac adaptation under physical stress. Cardiovasc Res 2016; 111:44-55. [DOI: 10.1093/cvr/cvw066] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 03/18/2016] [Indexed: 01/10/2023] Open
|
22
|
Evaluation of the Mayo Clinic Phenotype-Based Genotype Predictor Score in Patients with Clinically Diagnosed Hypertrophic Cardiomyopathy. J Cardiovasc Transl Res 2016; 9:153-61. [PMID: 26914223 DOI: 10.1007/s12265-016-9681-5] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 02/14/2016] [Indexed: 02/08/2023]
Abstract
Genetic testing for hypertrophic cardiomyopathy (HCM) can provide an important clinical marker for disease outcome and family screening. This study set out to validate our recently developed phenotype-based HCM genotype predictor score. Patients clinically diagnosed with HCM and evaluated by genetic counselors comprised the study cohort. Genotype score was derived based on clinical and echocardiographic variables. Total score was correlated with the yield of genetic testing. Of 564 HCM patients, 198 sought genetic testing (35 %; 55 % male; mean age at diagnosis, 50 ± 20 years). Of these, 101 patients (51 %) were genotype positive for a HCM-associated genetic mutation (55 % male; mean age at diagnosis, 42 ± 18 years). Cochran-Armitage analysis showed similar, statistically significant trends of increased yields for higher genotype scores for both the original and study cohort. Validated by the current study, this scoring system provides an easy-to-use, clinical tool to aid in determining the likelihood of a positive HCM genetic test.
Collapse
|
23
|
|
24
|
Linking Genes to Cardiovascular Diseases: Gene Action and Gene-Environment Interactions. J Cardiovasc Transl Res 2015; 8:506-27. [PMID: 26545598 DOI: 10.1007/s12265-015-9658-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 10/08/2015] [Indexed: 01/22/2023]
Abstract
A unique myocardial characteristic is its ability to grow/remodel in order to adapt; this is determined partly by genes and partly by the environment and the milieu intérieur. In the "post-genomic" era, a need is emerging to elucidate the physiologic functions of myocardial genes, as well as potential adaptive and maladaptive modulations induced by environmental/epigenetic factors. Genome sequencing and analysis advances have become exponential lately, with escalation of our knowledge concerning sometimes controversial genetic underpinnings of cardiovascular diseases. Current technologies can identify candidate genes variously involved in diverse normal/abnormal morphomechanical phenotypes, and offer insights into multiple genetic factors implicated in complex cardiovascular syndromes. The expression profiles of thousands of genes are regularly ascertained under diverse conditions. Global analyses of gene expression levels are useful for cataloging genes and correlated phenotypes, and for elucidating the role of genes in maladies. Comparative expression of gene networks coupled to complex disorders can contribute insights as to how "modifier genes" influence the expressed phenotypes. Increasingly, a more comprehensive and detailed systematic understanding of genetic abnormalities underlying, for example, various genetic cardiomyopathies is emerging. Implementing genomic findings in cardiology practice may well lead directly to better diagnosing and therapeutics. There is currently evolving a strong appreciation for the value of studying gene anomalies, and doing so in a non-disjointed, cohesive manner. However, it is challenging for many-practitioners and investigators-to comprehend, interpret, and utilize the clinically increasingly accessible and affordable cardiovascular genomics studies. This survey addresses the need for fundamental understanding in this vital area.
Collapse
|
25
|
Macadangdang J, Guan X, Smith AS, Lucero R, Czerniecki S, Childers MK, Mack DL, Kim DH. Nanopatterned Human iPSC-based Model of a Dystrophin-Null Cardiomyopathic Phenotype. Cell Mol Bioeng 2015; 8:320-332. [PMID: 26366230 PMCID: PMC4564135 DOI: 10.1007/s12195-015-0413-8] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2015] [Accepted: 08/05/2015] [Indexed: 11/30/2022] Open
Abstract
Human induced pluripotent stem cell derived cardiomyocytes (hiPSC-CMs) offer unprecedented opportunities to study inherited heart conditions in vitro, but are phenotypically immature, limiting their ability to effectively model adult-onset diseases. Cardiomyopathy is becoming the leading cause of death in patients with Duchenne muscular dystrophy (DMD), but the pathogenesis of this disease phenotype is not fully understood. Therefore, we aimed to test whether biomimetic nanotopography could further stratify the disease phenotype of DMD hiPSC-CMs to create more translationally relevant cardiomyocytes for disease modeling applications. We found that anisotropic nanotopography was necessary to distinguish structural differences between normal and DMD hiPSC-CMs, as these differences were masked on conventional flat substrates. DMD hiPSC-CMs exhibited a diminished structural and functional response to the underlying nanotopography compared to normal cardiomyocytes at both the macroscopic and subcellular levels. This blunted response may be due to a lower level of actin cytoskeleton turnover as measured by fluorescence recovery after photobleaching. Taken together these data suggest that DMD hiPSC-CMs are less adaptable to changes in their extracellular environment, and highlight the utility of nanotopographic substrates for effectively stratifying normal and structural cardiac disease phenotypes in vitro.
Collapse
Affiliation(s)
- Jesse Macadangdang
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Institute of Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| | - Xuan Guan
- Department of Physiology and Pharmacology, School of Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, USA
- Department of Rehabilitation Medicine, University of Washington, Seattle, WA, USA
- Institute of Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| | - Alec S.T. Smith
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Rachel Lucero
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Stefan Czerniecki
- Department of Rehabilitation Medicine, University of Washington, Seattle, WA, USA
- Institute of Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| | - Martin K. Childers
- Department of Rehabilitation Medicine, University of Washington, Seattle, WA, USA
- Institute of Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| | - David L. Mack
- Department of Rehabilitation Medicine, University of Washington, Seattle, WA, USA
- Institute of Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| | - Deok-Ho Kim
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Institute of Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| |
Collapse
|
26
|
McConnell BK, Singh S, Fan Q, Hernandez A, Portillo JP, Reiser PJ, Tikunova SB. Knock-in mice harboring a Ca(2+) desensitizing mutation in cardiac troponin C develop early onset dilated cardiomyopathy. Front Physiol 2015; 6:242. [PMID: 26379556 PMCID: PMC4550777 DOI: 10.3389/fphys.2015.00242] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 08/11/2015] [Indexed: 12/05/2022] Open
Abstract
The physiological consequences of aberrant Ca(2+) binding and exchange with cardiac myofilaments are not clearly understood. In order to examine the effect of decreasing Ca(2+) sensitivity of cTnC on cardiac function, we generated knock-in mice carrying a D73N mutation (not known to be associated with heart disease in human patients) in cTnC. The D73N mutation was engineered into the regulatory N-domain of cTnC in order to reduce Ca(2+) sensitivity of reconstituted thin filaments by increasing the rate of Ca(2+) dissociation. In addition, the D73N mutation drastically blunted the extent of Ca(2+) desensitization of reconstituted thin filaments induced by cTnI pseudo-phosphorylation. Compared to wild-type mice, heterozygous knock-in mice carrying the D73N mutation exhibited a substantially decreased Ca(2+) sensitivity of force development in skinned ventricular trabeculae. Kaplan-Meier survival analysis revealed that median survival time for knock-in mice was 12 weeks. Echocardiographic analysis revealed that knock-in mice exhibited increased left ventricular dimensions with thinner walls. Echocardiographic analysis also revealed that measures of systolic function, such as ejection fraction (EF) and fractional shortening (FS), were dramatically reduced in knock-in mice. In addition, knock-in mice displayed electrophysiological abnormalities, namely prolonged QRS and QT intervals. Furthermore, ventricular myocytes isolated from knock-in mice did not respond to β-adrenergic stimulation. Thus, knock-in mice developed pathological features similar to those observed in human patients with dilated cardiomyopathy (DCM). In conclusion, our results suggest that decreasing Ca(2+) sensitivity of the regulatory N-domain of cTnC is sufficient to trigger the development of DCM.
Collapse
Affiliation(s)
- Bradley K. McConnell
- Department of Pharmacological and Pharmaceutical Sciences, University of HoustonHouston, TX, USA
| | - Sonal Singh
- Department of Pharmacological and Pharmaceutical Sciences, University of HoustonHouston, TX, USA
| | - Qiying Fan
- Department of Pharmacological and Pharmaceutical Sciences, University of HoustonHouston, TX, USA
| | - Adriana Hernandez
- Department of Pharmacological and Pharmaceutical Sciences, University of HoustonHouston, TX, USA
| | - Jesus P. Portillo
- Department of Pharmacological and Pharmaceutical Sciences, University of HoustonHouston, TX, USA
| | - Peter J. Reiser
- Division of Biosciences, College of Dentistry, The Ohio State UniversityColumbus, OH, USA
| | - Svetlana B. Tikunova
- Department of Pharmacological and Pharmaceutical Sciences, University of HoustonHouston, TX, USA
| |
Collapse
|
27
|
Cardiovascular Disease Modeling Using Patient-Specific Induced Pluripotent Stem Cells. Int J Mol Sci 2015; 16:18894-922. [PMID: 26274955 PMCID: PMC4581278 DOI: 10.3390/ijms160818894] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Revised: 08/01/2015] [Accepted: 08/03/2015] [Indexed: 12/20/2022] Open
Abstract
The generation of induced pluripotent stem cells (iPSCs) has opened up a new scientific frontier in medicine. This technology has made it possible to obtain pluripotent stem cells from individuals with genetic disorders. Because iPSCs carry the identical genetic anomalies related to those disorders, iPSCs are an ideal platform for medical research. The pathophysiological cellular phenotypes of genetically heritable heart diseases such as arrhythmias and cardiomyopathies, have been modeled on cell culture dishes using disease-specific iPSC-derived cardiomyocytes. These model systems can potentially provide new insights into disease mechanisms and drug discoveries. This review focuses on recent progress in cardiovascular disease modeling using iPSCs, and discusses problems and future perspectives concerning their use.
Collapse
|
28
|
Screening of sarcomere gene mutations in young athletes with abnormal findings in electrocardiography: identification of a MYH7 mutation and MYBPC3 mutations. J Hum Genet 2015; 60:641-5. [PMID: 26178432 DOI: 10.1038/jhg.2015.81] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Revised: 05/15/2015] [Accepted: 06/15/2015] [Indexed: 11/09/2022]
Abstract
There is an overlap between the physiological cardiac remodeling associated with training in athletes, the so-called athlete's heart, and mild forms of hypertrophic cardiomyopathy (HCM), the most common hereditary cardiac disease. HCM is often accompanied by unfavorable outcomes including a sudden cardiac death in the adolescents. Because one of the initial signs of HCM is abnormality in electrocardiogram (ECG), athletes may need to monitor for ECG findings to prevent any unfavorable outcomes. HCM is caused by mutations in genes for sarcomere proteins, but there is no report on the systematic screening of gene mutations in athletes. One hundred and two genetically unrelated young Japanese athletes with abnormal ECG findings were the subjects for the analysis of four sarcomere genes, MYH7, MYBPC3, TNNT2 and TNNI3. We found that 5 out of 102 (4.9%) athletes carried mutations: a heterozygous MYH7 Glu935Lys mutation, a heterozygous MYBPC3 Arg160Trp mutation and another heterozygous MYBPC3 Thr1046Met mutation, all of which had been reported as HCM-associated mutations, in 1, 2 and 2 subjects, respectively. This is the first study of systematic screening of sarcomere gene mutations in a cohort of athletes with abnormal ECG, demonstrating the presence of sarcomere gene mutations in the athlete's heart.
Collapse
|
29
|
Kimura A. Molecular genetics and pathogenesis of cardiomyopathy. J Hum Genet 2015; 61:41-50. [PMID: 26178429 DOI: 10.1038/jhg.2015.83] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 06/15/2015] [Indexed: 12/19/2022]
Abstract
Cardiomyopathy is defined as a disease of functional impairment in the cardiac muscle and its etiology includes both extrinsic and intrinsic factors. Cardiomyopathy caused by the intrinsic factors is called as primary cardiomyopathy of which two major clinical phenotypes are hypertrophic cardiomyopathy (HCM) and dilated cardiomyopathy (DCM). Genetic approaches have revealed the disease genes for hereditary primary cardiomyopathy and functional studies have demonstrated that characteristic functional alterations induced by the disease-associated mutations are closely related to the clinical types, such that increased and decreased Ca(2+) sensitivities of muscle contraction are associated with HCM and DCM, respectively. In addition, recent studies have suggested that mutations in the Z-disc components found in HCM and DCM may result in increased and decreased stiffness of sarcomere, respectively. Moreover, functional analysis of mutations in the other components of cardiac muscle have suggested that the altered response to metabolic stresses is associated with cardiomyopathy, further indicating the heterogeneity in the etiology and pathogenesis of cardiomyopathy.
Collapse
Affiliation(s)
- Akinori Kimura
- Department of Molecular Pathogenesis, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| |
Collapse
|
30
|
Aksel T, Choe Yu E, Sutton S, Ruppel KM, Spudich JA. Ensemble force changes that result from human cardiac myosin mutations and a small-molecule effector. Cell Rep 2015; 11:910-920. [PMID: 25937279 PMCID: PMC4431957 DOI: 10.1016/j.celrep.2015.04.006] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Revised: 03/19/2015] [Accepted: 04/01/2015] [Indexed: 11/24/2022] Open
Abstract
Cardiomyopathies due to mutations in human β-cardiac myosin are a significant cause of heart failure, sudden death, and arrhythmia. To understand the underlying molecular basis of changes in the contractile system's force production due to such mutations and search for potential drugs that restore force generation, an in vitro assay is necessary to evaluate cardiac myosin's ensemble force using purified proteins. Here, we characterize the ensemble force of human α- and β-cardiac myosin isoforms and those of β-cardiac myosins carrying left ventricular non-compaction (M531R) and dilated cardiomyopathy (S532P) mutations using a utrophin-based loaded in vitro motility assay and new filament-tracking software. Our results show that human α- and β-cardiac myosin, as well as the mutants, show opposite mechanical and enzymatic phenotypes with respect to each other. We also show that omecamtiv mecarbil, a previously discovered cardiac-specific myosin activator, increases β-cardiac myosin force generation.
Collapse
Affiliation(s)
- Tural Aksel
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Elizabeth Choe Yu
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA 94305, USA; Cancer Biology Program, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Shirley Sutton
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Kathleen M Ruppel
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Pediatrics (Cardiology), Stanford University School of Medicine, Stanford, CA 94305, USA.
| | - James A Spudich
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
31
|
A novel de novo mutation of β-cardiac myosin heavy chain gene found in a twelve-year-old boy with hypertrophic cardiomyopathy. J Genet 2015; 93:557-60. [PMID: 25189259 DOI: 10.1007/s12041-014-0414-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
32
|
Jasnic-Savovic J, Nestorovic A, Savic S, Karasek S, Vitulo N, Valle G, Faulkner G, Radojkovic D, Kojic S. Profiling of skeletal muscle Ankrd2 protein in human cardiac tissue and neonatal rat cardiomyocytes. Histochem Cell Biol 2015; 143:583-97. [PMID: 25585647 DOI: 10.1007/s00418-015-1307-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/26/2014] [Indexed: 10/24/2022]
Abstract
Muscle-specific mechanosensors Ankrd2/Arpp (ankyrin repeat protein 2) and Ankrd1/CARP (cardiac ankyrin repeat protein) have an important role in transcriptional regulation, myofibrillar assembly, cardiogenesis and myogenesis. In skeletal muscle myofibrils, Ankrd2 has a structural role as a component of a titin associated stretch-sensing complex, while in the nucleus it exerts regulatory function as transcriptional co-factor. It is also involved in myogenic differentiation and coordination of myoblast proliferation. Although expressed in the heart, the role of Ankrd2 in the cardiac muscle is completely unknown. Recently, we have shown that hypertrophic and dilated cardiomyopathy pathways are altered upon Ankrd2 silencing suggesting the importance of this protein in cardiac tissue. Here we provide the underlying basis for the functional investigation of Ankrd2 in the heart. We confirmed reduced Ankrd2 expression levels in human heart in comparison with Ankrd1 using RNAseq and Western blot. For the first time we demonstrated that, apart from the sarcomere and nucleus, both proteins are localized to the intercalated disks of human cardiomyocytes. We further tested the expression and localization of endogenous Ankrd2 in rat neonatal cardiomyocytes, a well-established model for studying cardiac-specific proteins. Ankrd2 was found to be expressed in both the cytoplasm and nucleus, independently from maturation status of cardiomyocytes. In contrast to Ankrd1, it is not responsive to the cardiotoxic drug Doxorubicin, suggesting that different mechanisms govern their expression in cardiac cells.
Collapse
Affiliation(s)
- Jovana Jasnic-Savovic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, PO Box 23, 11010, Belgrade, Serbia
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Friedrich FW, Reischmann S, Schwalm A, Unger A, Ramanujam D, Münch J, Müller OJ, Hengstenberg C, Galve E, Charron P, Linke WA, Engelhardt S, Patten M, Richard P, van der Velden J, Eschenhagen T, Isnard R, Carrier L. FHL2 expression and variants in hypertrophic cardiomyopathy. Basic Res Cardiol 2014; 109:451. [PMID: 25358972 PMCID: PMC4215105 DOI: 10.1007/s00395-014-0451-8] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Revised: 10/04/2014] [Accepted: 10/22/2014] [Indexed: 11/28/2022]
Abstract
Based on evidence that FHL2 (four and a half LIM domains protein 2) negatively regulates cardiac hypertrophy we tested whether FHL2 altered expression or variants could be associated with hypertrophic cardiomyopathy (HCM). HCM is a myocardial disease characterized by left ventricular hypertrophy, diastolic dysfunction and increased interstitial fibrosis and is mainly caused by mutations in genes coding for sarcomeric proteins. FHL2 mRNA level, FHL2 protein level and I-band-binding density were lower in HCM patients than control individuals. Screening of 121 HCM patients without mutations in established disease genes identified 2 novel (T171M, V187L) and 4 known (R177Q, N226N, D268D, P273P) FHL2 variants in unrelated HCM families. We assessed the structural and functional consequences of the nonsynonymous substitutions after adeno-associated viral-mediated gene transfer in cardiac myocytes and in 3D-engineered heart tissue (EHT). Overexpression of FHL2 wild type or nonsynonymous substitutions in cardiac myocytes markedly down-regulated α-skeletal actin and partially blunted hypertrophy induced by phenylephrine or endothelin-1. After gene transfer in EHTs, force and velocity of both contraction and relaxation were higher with T171M and V187L FHL2 variants than wild type under basal conditions. Finally, chronic phenylephrine stimulation depressed EHT function in all groups, but to a lower extent in T171M-transduced EHTs. These data suggest that (1) FHL2 is down-regulated in HCM, (2) both FHL2 wild type and variants partially protected phenylephrine- or endothelin-1-induced hypertrophy in cardiac myocytes, and (3) FHL2 T171M and V187L nonsynonymous variants induced altered EHT contractility. These findings provide evidence that the 2 novel FHL2 variants could increase cardiac function in HCM.
Collapse
Affiliation(s)
- Felix W. Friedrich
- Department of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Silke Reischmann
- Department of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Aileen Schwalm
- Department of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Andreas Unger
- Department of Cardiovascular Physiology, Ruhr University Bochum, Bochum, Germany
| | - Deepak Ramanujam
- Institute of Pharmacology and Toxicology, Technical University Munich, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich, Munich, Germany
| | - Julia Münch
- University Heart Center Hamburg, Hamburg, Germany
| | - Oliver J. Müller
- Department of Cardiology, Internal Medicine III, University Hospital Heidelberg, Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, Germany
| | - Christian Hengstenberg
- Present Address: German Heart Centre of the Technical University Munich, Munich, Germany
- Klinik und Poliklinik für Innere Medizin II, Universitätsklinikum Regensburg, Regensburg, Germany
| | - Enrique Galve
- Unitat d’Insuficiència Cardiaca, Servei de Cardiologia, Hospital Vall d’Hebron, Barcelona, Spain
| | - Philippe Charron
- Inserm, U956, Paris, France
- ICAN Institute, UPMC Univ Paris 06, Paris, France
| | - Wolfgang A. Linke
- Department of Cardiovascular Physiology, Ruhr University Bochum, Bochum, Germany
| | - Stefan Engelhardt
- Institute of Pharmacology and Toxicology, Technical University Munich, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich, Munich, Germany
| | | | - Pascale Richard
- Inserm, U956, Paris, France
- ICAN Institute, UPMC Univ Paris 06, Paris, France
- Groupe Hospitalier Pitié-Salpêtrière, AP-HP Centre de référence des maladies cardiaques héréditaires, Paris, France
- Groupe Hospitalier Pitié-Salpêtrière, AP-HP,UF Cardiogénétique et Myogénétique, Paris, France
| | - Jolanda van der Velden
- Laboratory for Physiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands
| | - Thomas Eschenhagen
- Department of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Richard Isnard
- Inserm, U956, Paris, France
- ICAN Institute, UPMC Univ Paris 06, Paris, France
- Groupe Hospitalier Pitié-Salpêtrière, AP-HP Centre de référence des maladies cardiaques héréditaires, Paris, France
| | - Lucie Carrier
- Department of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany
| |
Collapse
|
34
|
Girolami F, Iascone M, Tomberli B, Bardi S, Benelli M, Marseglia G, Pescucci C, Pezzoli L, Sana ME, Basso C, Marziliano N, Merlini PA, Fornaro A, Cecchi F, Torricelli F, Olivotto I. Novel α-actinin 2 variant associated with familial hypertrophic cardiomyopathy and juvenile atrial arrhythmias: a massively parallel sequencing study. ACTA ACUST UNITED AC 2014; 7:741-50. [PMID: 25173926 DOI: 10.1161/circgenetics.113.000486] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Next-generation sequencing might be particularly advantageous in genetically heterogeneous conditions, such as hypertrophic cardiomyopathy (HCM), in which a considerable proportion of patients remain undiagnosed after Sanger. In this study, we present an Italian family with atypical HCM in which a novel disease-causing variant in α-actinin 2 (ACTN2) was identified by next-generation sequencing. METHODS AND RESULTS A large family spanning 4 generations was examined, exhibiting an autosomal dominant cardiomyopathic trait comprising a variable spectrum of (1) midapical HCM with restrictive evolution with marked biatrial dilatation, (2) early-onset atrial fibrillation and atrioventricular block, and (3) left ventricular noncompaction. In the proband, 48 disease genes for HCM, selected on the basis of published reports, were analyzed by targeted resequencing with a customized enrichment system. After bioinformatics analysis, 4 likely pathogenic variants were identified: TTN c.21977G>A (p.Arg7326Gln); TTN c.8749A>C (p.Thr2917Pro); ACTN2 c.683T>C (p.Met228Thr); and OBSCN c.13475T>G (p.Leu4492Arg). The novel variant ACTN2 c.683T>C (p.Met228Thr), located in the actin-binding domain, proved to be the only mutation fully cosegregating with the cardiomyopathic trait in 18 additional family members (of whom 11 clinically affected). ACTN2 c.683T>C (p.Met228Thr) was absent in 570 alleles of healthy controls and in 1000 Genomes Project and was labeled as Damaging by in silico analysis using polymorphism phenotyping v2, as Deleterious by sorts intolerant from tolerant, and as Disease-Causing by Mutation Taster. CONCLUSIONS A targeted next-generation sequencing approach allowed the identification of a novel ACTN2 variant associated with midapical HCM and juvenile onset of atrial fibrillation, emphasizing the potential of such approach in HCM diagnostic screening.
Collapse
Affiliation(s)
- Francesca Girolami
- From the Genetic Diagnostic Unit, Careggi University Hospital, Florence, Italy (F.G., S.B., M.B., G.M., C.P., F.T.); USSD Laboratorio Genetica Medica, Ospedali Riuniti, Bergamo, Italy (M.I., L.P., M.E.S.); Division of Cardiology, Department of Cardiological Thoracic and Vascular Sciences, University of Padua, Padua, Italy (C.B.); Azienda Ospedaliera Ospedale Niguarda Cà Granda, IV Division of Cardiology, Milan, Italy (N.M.); Division of Cardiology, Azienda Ospedaliera Universitaria di Parma, Parma, Italy (N.M.); Referral Center for Myocardial Diseases, Careggi University Hospital, Florence, Italy (B.T., A.F., I.O.); and Department of Clinical and Experimental Medicine (P.A.M.), University of Florence, Florence, Italy (F.C.).
| | - Maria Iascone
- From the Genetic Diagnostic Unit, Careggi University Hospital, Florence, Italy (F.G., S.B., M.B., G.M., C.P., F.T.); USSD Laboratorio Genetica Medica, Ospedali Riuniti, Bergamo, Italy (M.I., L.P., M.E.S.); Division of Cardiology, Department of Cardiological Thoracic and Vascular Sciences, University of Padua, Padua, Italy (C.B.); Azienda Ospedaliera Ospedale Niguarda Cà Granda, IV Division of Cardiology, Milan, Italy (N.M.); Division of Cardiology, Azienda Ospedaliera Universitaria di Parma, Parma, Italy (N.M.); Referral Center for Myocardial Diseases, Careggi University Hospital, Florence, Italy (B.T., A.F., I.O.); and Department of Clinical and Experimental Medicine (P.A.M.), University of Florence, Florence, Italy (F.C.)
| | - Benedetta Tomberli
- From the Genetic Diagnostic Unit, Careggi University Hospital, Florence, Italy (F.G., S.B., M.B., G.M., C.P., F.T.); USSD Laboratorio Genetica Medica, Ospedali Riuniti, Bergamo, Italy (M.I., L.P., M.E.S.); Division of Cardiology, Department of Cardiological Thoracic and Vascular Sciences, University of Padua, Padua, Italy (C.B.); Azienda Ospedaliera Ospedale Niguarda Cà Granda, IV Division of Cardiology, Milan, Italy (N.M.); Division of Cardiology, Azienda Ospedaliera Universitaria di Parma, Parma, Italy (N.M.); Referral Center for Myocardial Diseases, Careggi University Hospital, Florence, Italy (B.T., A.F., I.O.); and Department of Clinical and Experimental Medicine (P.A.M.), University of Florence, Florence, Italy (F.C.)
| | - Sara Bardi
- From the Genetic Diagnostic Unit, Careggi University Hospital, Florence, Italy (F.G., S.B., M.B., G.M., C.P., F.T.); USSD Laboratorio Genetica Medica, Ospedali Riuniti, Bergamo, Italy (M.I., L.P., M.E.S.); Division of Cardiology, Department of Cardiological Thoracic and Vascular Sciences, University of Padua, Padua, Italy (C.B.); Azienda Ospedaliera Ospedale Niguarda Cà Granda, IV Division of Cardiology, Milan, Italy (N.M.); Division of Cardiology, Azienda Ospedaliera Universitaria di Parma, Parma, Italy (N.M.); Referral Center for Myocardial Diseases, Careggi University Hospital, Florence, Italy (B.T., A.F., I.O.); and Department of Clinical and Experimental Medicine (P.A.M.), University of Florence, Florence, Italy (F.C.)
| | - Matteo Benelli
- From the Genetic Diagnostic Unit, Careggi University Hospital, Florence, Italy (F.G., S.B., M.B., G.M., C.P., F.T.); USSD Laboratorio Genetica Medica, Ospedali Riuniti, Bergamo, Italy (M.I., L.P., M.E.S.); Division of Cardiology, Department of Cardiological Thoracic and Vascular Sciences, University of Padua, Padua, Italy (C.B.); Azienda Ospedaliera Ospedale Niguarda Cà Granda, IV Division of Cardiology, Milan, Italy (N.M.); Division of Cardiology, Azienda Ospedaliera Universitaria di Parma, Parma, Italy (N.M.); Referral Center for Myocardial Diseases, Careggi University Hospital, Florence, Italy (B.T., A.F., I.O.); and Department of Clinical and Experimental Medicine (P.A.M.), University of Florence, Florence, Italy (F.C.)
| | - Giuseppina Marseglia
- From the Genetic Diagnostic Unit, Careggi University Hospital, Florence, Italy (F.G., S.B., M.B., G.M., C.P., F.T.); USSD Laboratorio Genetica Medica, Ospedali Riuniti, Bergamo, Italy (M.I., L.P., M.E.S.); Division of Cardiology, Department of Cardiological Thoracic and Vascular Sciences, University of Padua, Padua, Italy (C.B.); Azienda Ospedaliera Ospedale Niguarda Cà Granda, IV Division of Cardiology, Milan, Italy (N.M.); Division of Cardiology, Azienda Ospedaliera Universitaria di Parma, Parma, Italy (N.M.); Referral Center for Myocardial Diseases, Careggi University Hospital, Florence, Italy (B.T., A.F., I.O.); and Department of Clinical and Experimental Medicine (P.A.M.), University of Florence, Florence, Italy (F.C.)
| | - Chiara Pescucci
- From the Genetic Diagnostic Unit, Careggi University Hospital, Florence, Italy (F.G., S.B., M.B., G.M., C.P., F.T.); USSD Laboratorio Genetica Medica, Ospedali Riuniti, Bergamo, Italy (M.I., L.P., M.E.S.); Division of Cardiology, Department of Cardiological Thoracic and Vascular Sciences, University of Padua, Padua, Italy (C.B.); Azienda Ospedaliera Ospedale Niguarda Cà Granda, IV Division of Cardiology, Milan, Italy (N.M.); Division of Cardiology, Azienda Ospedaliera Universitaria di Parma, Parma, Italy (N.M.); Referral Center for Myocardial Diseases, Careggi University Hospital, Florence, Italy (B.T., A.F., I.O.); and Department of Clinical and Experimental Medicine (P.A.M.), University of Florence, Florence, Italy (F.C.)
| | - Laura Pezzoli
- From the Genetic Diagnostic Unit, Careggi University Hospital, Florence, Italy (F.G., S.B., M.B., G.M., C.P., F.T.); USSD Laboratorio Genetica Medica, Ospedali Riuniti, Bergamo, Italy (M.I., L.P., M.E.S.); Division of Cardiology, Department of Cardiological Thoracic and Vascular Sciences, University of Padua, Padua, Italy (C.B.); Azienda Ospedaliera Ospedale Niguarda Cà Granda, IV Division of Cardiology, Milan, Italy (N.M.); Division of Cardiology, Azienda Ospedaliera Universitaria di Parma, Parma, Italy (N.M.); Referral Center for Myocardial Diseases, Careggi University Hospital, Florence, Italy (B.T., A.F., I.O.); and Department of Clinical and Experimental Medicine (P.A.M.), University of Florence, Florence, Italy (F.C.)
| | - Maria Elena Sana
- From the Genetic Diagnostic Unit, Careggi University Hospital, Florence, Italy (F.G., S.B., M.B., G.M., C.P., F.T.); USSD Laboratorio Genetica Medica, Ospedali Riuniti, Bergamo, Italy (M.I., L.P., M.E.S.); Division of Cardiology, Department of Cardiological Thoracic and Vascular Sciences, University of Padua, Padua, Italy (C.B.); Azienda Ospedaliera Ospedale Niguarda Cà Granda, IV Division of Cardiology, Milan, Italy (N.M.); Division of Cardiology, Azienda Ospedaliera Universitaria di Parma, Parma, Italy (N.M.); Referral Center for Myocardial Diseases, Careggi University Hospital, Florence, Italy (B.T., A.F., I.O.); and Department of Clinical and Experimental Medicine (P.A.M.), University of Florence, Florence, Italy (F.C.)
| | - Cristina Basso
- From the Genetic Diagnostic Unit, Careggi University Hospital, Florence, Italy (F.G., S.B., M.B., G.M., C.P., F.T.); USSD Laboratorio Genetica Medica, Ospedali Riuniti, Bergamo, Italy (M.I., L.P., M.E.S.); Division of Cardiology, Department of Cardiological Thoracic and Vascular Sciences, University of Padua, Padua, Italy (C.B.); Azienda Ospedaliera Ospedale Niguarda Cà Granda, IV Division of Cardiology, Milan, Italy (N.M.); Division of Cardiology, Azienda Ospedaliera Universitaria di Parma, Parma, Italy (N.M.); Referral Center for Myocardial Diseases, Careggi University Hospital, Florence, Italy (B.T., A.F., I.O.); and Department of Clinical and Experimental Medicine (P.A.M.), University of Florence, Florence, Italy (F.C.)
| | - Nicola Marziliano
- From the Genetic Diagnostic Unit, Careggi University Hospital, Florence, Italy (F.G., S.B., M.B., G.M., C.P., F.T.); USSD Laboratorio Genetica Medica, Ospedali Riuniti, Bergamo, Italy (M.I., L.P., M.E.S.); Division of Cardiology, Department of Cardiological Thoracic and Vascular Sciences, University of Padua, Padua, Italy (C.B.); Azienda Ospedaliera Ospedale Niguarda Cà Granda, IV Division of Cardiology, Milan, Italy (N.M.); Division of Cardiology, Azienda Ospedaliera Universitaria di Parma, Parma, Italy (N.M.); Referral Center for Myocardial Diseases, Careggi University Hospital, Florence, Italy (B.T., A.F., I.O.); and Department of Clinical and Experimental Medicine (P.A.M.), University of Florence, Florence, Italy (F.C.)
| | - Piera Angelica Merlini
- From the Genetic Diagnostic Unit, Careggi University Hospital, Florence, Italy (F.G., S.B., M.B., G.M., C.P., F.T.); USSD Laboratorio Genetica Medica, Ospedali Riuniti, Bergamo, Italy (M.I., L.P., M.E.S.); Division of Cardiology, Department of Cardiological Thoracic and Vascular Sciences, University of Padua, Padua, Italy (C.B.); Azienda Ospedaliera Ospedale Niguarda Cà Granda, IV Division of Cardiology, Milan, Italy (N.M.); Division of Cardiology, Azienda Ospedaliera Universitaria di Parma, Parma, Italy (N.M.); Referral Center for Myocardial Diseases, Careggi University Hospital, Florence, Italy (B.T., A.F., I.O.); and Department of Clinical and Experimental Medicine (P.A.M.), University of Florence, Florence, Italy (F.C.)
| | - Alessandra Fornaro
- From the Genetic Diagnostic Unit, Careggi University Hospital, Florence, Italy (F.G., S.B., M.B., G.M., C.P., F.T.); USSD Laboratorio Genetica Medica, Ospedali Riuniti, Bergamo, Italy (M.I., L.P., M.E.S.); Division of Cardiology, Department of Cardiological Thoracic and Vascular Sciences, University of Padua, Padua, Italy (C.B.); Azienda Ospedaliera Ospedale Niguarda Cà Granda, IV Division of Cardiology, Milan, Italy (N.M.); Division of Cardiology, Azienda Ospedaliera Universitaria di Parma, Parma, Italy (N.M.); Referral Center for Myocardial Diseases, Careggi University Hospital, Florence, Italy (B.T., A.F., I.O.); and Department of Clinical and Experimental Medicine (P.A.M.), University of Florence, Florence, Italy (F.C.)
| | - Franco Cecchi
- From the Genetic Diagnostic Unit, Careggi University Hospital, Florence, Italy (F.G., S.B., M.B., G.M., C.P., F.T.); USSD Laboratorio Genetica Medica, Ospedali Riuniti, Bergamo, Italy (M.I., L.P., M.E.S.); Division of Cardiology, Department of Cardiological Thoracic and Vascular Sciences, University of Padua, Padua, Italy (C.B.); Azienda Ospedaliera Ospedale Niguarda Cà Granda, IV Division of Cardiology, Milan, Italy (N.M.); Division of Cardiology, Azienda Ospedaliera Universitaria di Parma, Parma, Italy (N.M.); Referral Center for Myocardial Diseases, Careggi University Hospital, Florence, Italy (B.T., A.F., I.O.); and Department of Clinical and Experimental Medicine (P.A.M.), University of Florence, Florence, Italy (F.C.)
| | - Francesca Torricelli
- From the Genetic Diagnostic Unit, Careggi University Hospital, Florence, Italy (F.G., S.B., M.B., G.M., C.P., F.T.); USSD Laboratorio Genetica Medica, Ospedali Riuniti, Bergamo, Italy (M.I., L.P., M.E.S.); Division of Cardiology, Department of Cardiological Thoracic and Vascular Sciences, University of Padua, Padua, Italy (C.B.); Azienda Ospedaliera Ospedale Niguarda Cà Granda, IV Division of Cardiology, Milan, Italy (N.M.); Division of Cardiology, Azienda Ospedaliera Universitaria di Parma, Parma, Italy (N.M.); Referral Center for Myocardial Diseases, Careggi University Hospital, Florence, Italy (B.T., A.F., I.O.); and Department of Clinical and Experimental Medicine (P.A.M.), University of Florence, Florence, Italy (F.C.)
| | - Iacopo Olivotto
- From the Genetic Diagnostic Unit, Careggi University Hospital, Florence, Italy (F.G., S.B., M.B., G.M., C.P., F.T.); USSD Laboratorio Genetica Medica, Ospedali Riuniti, Bergamo, Italy (M.I., L.P., M.E.S.); Division of Cardiology, Department of Cardiological Thoracic and Vascular Sciences, University of Padua, Padua, Italy (C.B.); Azienda Ospedaliera Ospedale Niguarda Cà Granda, IV Division of Cardiology, Milan, Italy (N.M.); Division of Cardiology, Azienda Ospedaliera Universitaria di Parma, Parma, Italy (N.M.); Referral Center for Myocardial Diseases, Careggi University Hospital, Florence, Italy (B.T., A.F., I.O.); and Department of Clinical and Experimental Medicine (P.A.M.), University of Florence, Florence, Italy (F.C.)
| |
Collapse
|
35
|
Arimura T, Takeya R, Ishikawa T, Yamano T, Matsuo A, Tatsumi T, Nomura T, Sumimoto H, Kimura A. Dilated cardiomyopathy-associated FHOD3 variant impairs the ability to induce activation of transcription factor serum response factor. Circ J 2014; 77:2990-6. [PMID: 24088304 DOI: 10.1253/circj.cj-13-0255] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND Dilated cardiomyopathy (DCM) is characterized by a dilated left ventricular cavity with systolic dysfunction manifested by heart failure. It has been revealed that mutations in genes for cytoskeleton or sarcomere proteins cause DCM. However, the disease-causing mutations can be found only in far less than half of patients with a family history, indicating that there should be other disease genes for DCM. Formin homology 2 domain containing 3 (FHOD3) is a sarcomeric protein expressed in the heart that plays an essential role in sarcomere organization during myofibrillogenesis. The purpose of this study was to explore a possible novel disease gene for DCM. METHODS AND RESULTS We analyzed 48 Japanese familial DCM patients for mutations in FHOD3, and a missense variant, Tyr1249Asn, which was predicted to modify the 3D structure and damage protein function, was found in a case with adult-onset DCM. Functional studies revealed that the DCM-associated mutation significantly reduced the ability to induce actin dynamics-dependent activation of serum response factor, although no remarkable change in the cellular localization was induced in neonatal rat cardiomyocytes transfected with a mutant construct of FHOD3. CONCLUSIONS The DCM-associated FHOD3 variant may cause DCM by interfering with actin filament assembly.
Collapse
Affiliation(s)
- Takuro Arimura
- Department of Molecular Pathogenesis, Medical Research Institute, Tokyo Medical and Dental University
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Kirk JA, Holewinski RJ, Kooij V, Agnetti G, Tunin RS, Witayavanitkul N, de Tombe PP, Gao WD, Van Eyk J, Kass DA. Cardiac resynchronization sensitizes the sarcomere to calcium by reactivating GSK-3β. J Clin Invest 2014; 124:129-38. [PMID: 24292707 DOI: 10.1172/jci69253] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2013] [Accepted: 09/19/2013] [Indexed: 01/10/2023] Open
Abstract
Cardiac resynchronization therapy (CRT), the application of biventricular stimulation to correct discoordinate contraction, is the only heart failure treatment that enhances acute and chronic systolic function, increases cardiac work, and reduces mortality. Resting myocyte function also increases after CRT despite only modest improvement in calcium transients, suggesting that CRT may enhance myofilament calcium responsiveness. To test this hypothesis, we examined adult dogs subjected to tachypacing-induced heart failure for 6 weeks, concurrent with ventricular dyssynchrony (HF(dys)) or CRT. Myofilament force-calcium relationships were measured in skinned trabeculae and/or myocytes. Compared with control, maximal calcium-activated force and calcium sensitivity declined globally in HF(dys); however, CRT restored both. Phosphatase PP1 induced calcium desensitization in control and CRT-treated cells, while HF(dys) cells were unaffected, implying that CRT enhances myofilament phosphorylation. Proteomics revealed phosphorylation sites on Z-disk and M-band proteins, which were predicted to be targets of glycogen synthase kinase-3β (GSK-3β). We found that GSK-3β was deactivated in HF(dys) and reactivated by CRT. Mass spectrometry of myofilament proteins from HF(dys) animals incubated with GSK-3β confirmed GSK-3β–dependent phosphorylation at many of the same sites observed with CRT. GSK-3β restored calcium sensitivity in HF(dys), but did not affect control or CRT cells. These data indicate that CRT improves calcium responsiveness of myofilaments following HF(dys) through GSK-3β reactivation, identifying a therapeutic approach to enhancing contractile function
Collapse
|
37
|
Xiao X, Moreno-Moral A, Rotival M, Bottolo L, Petretto E. Multi-tissue analysis of co-expression networks by higher-order generalized singular value decomposition identifies functionally coherent transcriptional modules. PLoS Genet 2014; 10:e1004006. [PMID: 24391511 PMCID: PMC3879165 DOI: 10.1371/journal.pgen.1004006] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2013] [Accepted: 10/22/2013] [Indexed: 12/27/2022] Open
Abstract
Recent high-throughput efforts such as ENCODE have generated a large body of genome-scale transcriptional data in multiple conditions (e.g., cell-types and disease states). Leveraging these data is especially important for network-based approaches to human disease, for instance to identify coherent transcriptional modules (subnetworks) that can inform functional disease mechanisms and pathological pathways. Yet, genome-scale network analysis across conditions is significantly hampered by the paucity of robust and computationally-efficient methods. Building on the Higher-Order Generalized Singular Value Decomposition, we introduce a new algorithmic approach for efficient, parameter-free and reproducible identification of network-modules simultaneously across multiple conditions. Our method can accommodate weighted (and unweighted) networks of any size and can similarly use co-expression or raw gene expression input data, without hinging upon the definition and stability of the correlation used to assess gene co-expression. In simulation studies, we demonstrated distinctive advantages of our method over existing methods, which was able to recover accurately both common and condition-specific network-modules without entailing ad-hoc input parameters as required by other approaches. We applied our method to genome-scale and multi-tissue transcriptomic datasets from rats (microarray-based) and humans (mRNA-sequencing-based) and identified several common and tissue-specific subnetworks with functional significance, which were not detected by other methods. In humans we recapitulated the crosstalk between cell-cycle progression and cell-extracellular matrix interactions processes in ventricular zones during neocortex expansion and further, we uncovered pathways related to development of later cognitive functions in the cortical plate of the developing brain which were previously unappreciated. Analyses of seven rat tissues identified a multi-tissue subnetwork of co-expressed heat shock protein (Hsp) and cardiomyopathy genes (Bag3, Cryab, Kras, Emd, Plec), which was significantly replicated using separate failing heart and liver gene expression datasets in humans, thus revealing a conserved functional role for Hsp genes in cardiovascular disease.
Collapse
Affiliation(s)
- Xiaolin Xiao
- Medical Research Council (MRC) Clinical Sciences Centre, Faculty of Medicine, Imperial College, London, United Kingdom
| | - Aida Moreno-Moral
- Medical Research Council (MRC) Clinical Sciences Centre, Faculty of Medicine, Imperial College, London, United Kingdom
| | - Maxime Rotival
- Medical Research Council (MRC) Clinical Sciences Centre, Faculty of Medicine, Imperial College, London, United Kingdom
| | - Leonardo Bottolo
- Department of Mathematics, Imperial College, London, United Kingdom
| | - Enrico Petretto
- Medical Research Council (MRC) Clinical Sciences Centre, Faculty of Medicine, Imperial College, London, United Kingdom
- * E-mail:
| |
Collapse
|
38
|
Gosselin-Badaroudine P, Moreau A, Chahine M. Nav 1.5 mutations linked to dilated cardiomyopathy phenotypes: Is the gating pore current the missing link? Channels (Austin) 2013; 8:90-94. [PMID: 24300601 PMCID: PMC4048347 DOI: 10.4161/chan.27179] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Accepted: 11/12/2013] [Indexed: 12/19/2022] Open
Abstract
Nav 1.5 dysfunctions are commonly linked to rhythms disturbances that include type 3 long QT syndrome (LQT3), Brugada syndrome (BrS), sick sinus syndrome (SSS) and conduction defects. Recently, this channel protein has been also linked to structural heart diseases such as dilated cardiomyopathy (DCM).
Collapse
Affiliation(s)
| | - Adrien Moreau
- Centre de recherche; Institut universitaire en santé mentale de Québec; Quebec City, QC Canada
| | - Mohamed Chahine
- Centre de recherche; Institut universitaire en santé mentale de Québec; Quebec City, QC Canada
- Department of Medicine; Université Laval; Quebec City, QC Canada
| |
Collapse
|
39
|
Scholten A, Preisinger C, Corradini E, Bourgonje VJ, Hennrich ML, van Veen TAB, Swaminathan PD, Joiner ML, Vos MA, Anderson ME, Heck AJR. Phosphoproteomics study based on in vivo inhibition reveals sites of calmodulin-dependent protein kinase II regulation in the heart. J Am Heart Assoc 2013; 2:e000318. [PMID: 23926118 PMCID: PMC3828808 DOI: 10.1161/jaha.113.000318] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
BACKGROUND The multifunctional Ca(2+)- and calmodulin-dependent protein kinase II (CaMKII) is a crucial mediator of cardiac physiology and pathology. Increased expression and activation of CaMKII has been linked to elevated risk for arrhythmic events and is a hallmark of human heart failure. A useful approach to determining CaMKII's role therein is large-scale analysis of phosphorylation events by mass spectrometry. However, current large-scale phosphoproteomics approaches have proved inadequate for high-fidelity identification of kinase-specific roles. The purpose of this study was to develop a phosphoproteomics approach to specifically identify CaMKII's downstream effects in cardiac tissue. METHODS AND RESULTS To identify putative downstream CaMKII targets in cardiac tissue, animals with myocardial-delimited expression of the specific peptide inhibitor of CaMKII (AC3-I) or an inactive control (AC3-C) were compared using quantitative phosphoproteomics. The hearts were isolated after isoproterenol injection to induce CaMKII activation downstream of β-adrenergic receptor agonist stimulation. Enriched phosphopeptides from AC3-I and AC3-C mice were differentially quantified using stable isotope dimethyl labeling, strong cation exchange chromatography and high-resolution LC-MS/MS. Phosphorylation levels of several hundred sites could be profiled, including 39 phosphoproteins noticeably affected by AC3-I-mediated CaMKII inhibition. CONCLUSIONS Our data set included known CaMKII substrates, as well as several new candidate proteins involved in functions not previously implicated in CaMKII signaling.
Collapse
Affiliation(s)
- Arjen Scholten
- Biomolecular Mass Spectrometry & Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, The Netherlands
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
|
41
|
Mook ORF, Haagmans MA, Soucy JF, van de Meerakker JBA, Baas F, Jakobs ME, Hofman N, Christiaans I, Lekanne Deprez RH, Mannens MMAM. Targeted sequence capture and GS-FLX Titanium sequencing of 23 hypertrophic and dilated cardiomyopathy genes: implementation into diagnostics. J Med Genet 2013; 50:614-26. [PMID: 23785128 PMCID: PMC3756457 DOI: 10.1136/jmedgenet-2012-101231] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
BACKGROUND Genetic evaluation of cardiomyopathies poses a challenge. Multiple genes are involved but no clear genotype-phenotype correlations have been found so far. In the past, genetic evaluation for hypertrophic (HCM) and dilated (DCM) cardiomyopathies was performed by sequential screening of a very limited number of genes. Recent developments in sequencing have increased the throughput, enabling simultaneous screening of multiple genes for multiple patients in a single sequencing run. OBJECTIVE Development and implementation of a next generation sequencing (NGS) based genetic test as replacement for Sanger sequencing. METHODS AND RESULTS In order to increase the number of genes that can be screened in a shorter time period, we enriched all exons of 23 of the most relevant HCM and DCM related genes using on-array multiplexed sequence capture followed by massively parallel pyrosequencing on the GS-FLX Titanium. After optimisation of array based sequence capture it was feasible to reliably detect a large panel of known and unknown variants in HCM and DCM patients, whereby the unknown variants could be confirmed by Sanger sequencing. CONCLUSIONS The rate of detection of (pathogenic) variants in both HCM and DCM patients was increased due to a larger number of genes studied. Array based target enrichment followed by NGS showed the same accuracy as Sanger sequencing. Therefore, NGS is ready for implementation in a diagnostic setting.
Collapse
Affiliation(s)
- Olaf R F Mook
- Department of Clinical Genetics, Academic Medical Center, Amsterdam, The Netherlands.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Wilson AJ, Schoenauer R, Ehler E, Agarkova I, Bennett PM. Cardiomyocyte growth and sarcomerogenesis at the intercalated disc. Cell Mol Life Sci 2013; 71:165-81. [PMID: 23708682 PMCID: PMC3889684 DOI: 10.1007/s00018-013-1374-5] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Revised: 04/27/2013] [Accepted: 05/13/2013] [Indexed: 12/02/2022]
Abstract
Cardiomyocytes grow during heart maturation or disease-related cardiac remodeling. We present evidence that the intercalated disc (ID) is integral to both longitudinal and lateral growth: increases in width are accommodated by lateral extension of the plicate tread regions and increases in length by sarcomere insertion within the ID. At the margin between myofibril and the folded membrane of the ID lies a transitional junction through which the thin filaments from the last sarcomere run to the ID membrane and it has been suggested that this junction acts as a proto Z-disc for sarcomere addition. In support of this hypothesis, we have investigated the ultrastructure of the ID in mouse hearts from control and dilated cardiomyopathy (DCM) models, the MLP-null and a cardiac-specific β-catenin mutant, cΔex3, as well as in human left ventricle from normal and DCM samples. We find that the ID amplitude can vary tenfold from 0.2 μm up to a maximum of ~2 μm allowing gradual expansion during heart growth. At the greatest amplitude, equivalent to a sarcomere length, A-bands and thick filaments are found within the ID membrane loops together with a Z-disc, which develops at the transitional junction position. Here, also, the tops of the membrane folds, which are rich in αII spectrin, become enlarged and associated with junctional sarcoplasmic reticulum. Systematically larger ID amplitudes are found in DCM samples. Other morphological differences between mouse DCM and normal hearts suggest that sarcomere inclusion is compromised in the diseased hearts.
Collapse
Affiliation(s)
- Amanda J Wilson
- Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London, SE1 1UL, UK,
| | | | | | | | | |
Collapse
|
43
|
Impact of ANKRD1 mutations associated with hypertrophic cardiomyopathy on contraction parameters of engineered heart tissue. Basic Res Cardiol 2013; 108:349. [PMID: 23572067 DOI: 10.1007/s00395-013-0349-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2012] [Revised: 03/11/2013] [Accepted: 03/26/2013] [Indexed: 12/25/2022]
Abstract
Hypertrophic cardiomyopathy (HCM) is a myocardial disease associated with mutations in sarcomeric genes. Three mutations were found in ANKRD1, encoding ankyrin repeat domain 1 (ANKRD1), a transcriptional co-factor located in the sarcomere. In the present study, we investigated whether expression of HCM-associated ANKRD1 mutations affects contraction parameters after gene transfer in engineered heart tissues (EHTs). EHTs were generated from neonatal rat heart cells and were transduced with adeno-associated virus encoding GFP or myc-tagged wild-type (WT) or mutant (P52A, T123M, or I280V) ANKRD1. Contraction parameters were analyzed from day 8 to day 16 of culture, and evaluated in the absence or presence of the proteasome inhibitor epoxomicin for 24 h. Under standard conditions, only WT- and T123M-ANKRD1 were correctly incorporated in the sarcomere. T123M-ANKRD1-transduced EHTs exhibited higher force and velocities of contraction and relaxation than WT- P52A- and I280V-ANKRD1 were highly unstable, not incorporated into the sarcomere, and did not induce contractile alterations. After epoxomicin treatment, P52A and I280V were both stabilized and incorporated into the sarcomere. I280V-transduced EHTs showed prolonged relaxation. These data suggest different impacts of ANKRD1 mutations on cardiomyocyte function: gain-of-function for T123M mutation under all conditions and dominant-negative effect for the I280V mutation which may come into play only when the proteasome is impaired.
Collapse
|
44
|
Andreasen C, Nielsen JB, Refsgaard L, Holst AG, Christensen AH, Andreasen L, Sajadieh A, Haunsø S, Svendsen JH, Olesen MS. New population-based exome data are questioning the pathogenicity of previously cardiomyopathy-associated genetic variants. Eur J Hum Genet 2013; 21:918-28. [PMID: 23299917 PMCID: PMC3746259 DOI: 10.1038/ejhg.2012.283] [Citation(s) in RCA: 161] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2012] [Revised: 09/04/2012] [Accepted: 11/23/2012] [Indexed: 11/09/2022] Open
Abstract
Cardiomyopathies are a heterogeneous group of diseases with various etiologies. We focused on three genetically determined cardiomyopathies: hypertrophic (HCM), dilated (DCM), and arrhythmogenic right ventricular cardiomyopathy (ARVC). Eighty-four genes have so far been associated with these cardiomyopathies, but the disease-causing effect of reported variants is often dubious. In order to identify possible false-positive variants, we investigated the prevalence of previously reported cardiomyopathy-associated variants in recently published exome data. We searched for reported missense and nonsense variants in the NHLBI-Go Exome Sequencing Project (ESP) containing exome data from 6500 individuals. In ESP, we identified 94 variants out of 687 (14%) variants previously associated with HCM, 58 out of 337 (17%) variants associated with DCM, and 38 variants out of 209 (18%) associated with ARVC. These findings correspond to a genotype prevalence of 1:4 for HCM, 1:6 for DCM, and 1:5 for ARVC. PolyPhen-2 predictions were conducted on all previously published cardiomyopathy-associated missense variants. We found significant overrepresentation of variants predicted as being benign among those present in ESP compared with the ones not present. In order to validate our findings, seven variants associated with cardiomyopathy were genotyped in a control population and this revealed frequencies comparable with the ones found in ESP. In conclusion, we identified genotype prevalences up to more than one thousand times higher than expected from the phenotype prevalences in the general population (HCM 1:500, DCM 1:2500, and ARVC 1:5000) and our data suggest that a high number of these variants are not monogenic causes of cardiomyopathy.
Collapse
Affiliation(s)
- Charlotte Andreasen
- The Danish National Research Foundation Centre for Cardiac Arrhythmia, Copenhagen, Denmark
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Kekenes-Huskey PM, Lindert S, McCammon JA. Molecular basis of calcium-sensitizing and desensitizing mutations of the human cardiac troponin C regulatory domain: a multi-scale simulation study. PLoS Comput Biol 2012; 8:e1002777. [PMID: 23209387 PMCID: PMC3510055 DOI: 10.1371/journal.pcbi.1002777] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2012] [Accepted: 09/28/2012] [Indexed: 11/18/2022] Open
Abstract
Troponin C (TnC) is implicated in the initiation of myocyte contraction via binding of cytosolic Ca²⁺ and subsequent recognition of the Troponin I switch peptide. Mutations of the cardiac TnC N-terminal regulatory domain have been shown to alter both calcium binding and myofilament force generation. We have performed molecular dynamics simulations of engineered TnC variants that increase or decrease Ca²⁺ sensitivity, in order to understand the structural basis of their impact on TnC function. We will use the distinction for mutants that are associated with increased Ca²⁺ affinity and for those mutants with reduced affinity. Our studies demonstrate that for GOF mutants V44Q and L48Q, the structure of the physiologically-active site II Ca²⁺ binding site in the Ca²⁺-free (apo) state closely resembled the Ca²⁺-bound (holo) state. In contrast, site II is very labile for LOF mutants E40A and V79Q in the apo form and bears little resemblance with the holo conformation. We hypothesize that these phenomena contribute to the increased association rate, k(on), for the GOF mutants relative to LOF. Furthermore, we observe significant positive and negative positional correlations between helices in the GOF holo mutants that are not found in the LOF mutants. We anticipate these correlations may contribute either directly to Ca²⁺ affinity or indirectly through TnI association. Our observations based on the structure and dynamics of mutant TnC provide rationale for binding trends observed in GOF and LOF mutants and will guide the development of inotropic drugs that target TnC.
Collapse
Affiliation(s)
- Peter Michael Kekenes-Huskey
- Department of Pharmacology, Center for Theoretical Biological Physics, National Computational Biomedical Resource and Howard Hughes Medical Institute, University of California San Diego, La Jolla, California, United States of America.
| | | | | |
Collapse
|
46
|
Faita F, Vecoli C, Foffa I, Andreassi MG. Next generation sequencing in cardiovascular diseases. World J Cardiol 2012; 4:288-95. [PMID: 23110245 PMCID: PMC3482622 DOI: 10.4330/wjc.v4.i10.288] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2012] [Revised: 09/08/2012] [Accepted: 09/15/2012] [Indexed: 02/06/2023] Open
Abstract
In the last few years, the advent of next generation sequencing (NGS) has revolutionized the approach to genetic studies, making whole-genome sequencing a possible way of obtaining global genomic information. NGS has very recently been shown to be successful in identifying novel causative mutations of rare or common Mendelian disorders. At the present time, it is expected that NGS will be increasingly important in the study of inherited and complex cardiovascular diseases (CVDs). However, the NGS approach to the genetics of CVDs represents a territory which has not been widely investigated. The identification of rare and frequent genetic variants can be very important in clinical practice to detect pathogenic mutations or to establish a profile of risk for the development of pathology. The purpose of this paper is to discuss the recent application of NGS in the study of several CVDs such as inherited cardiomyopathies, channelopathies, coronary artery disease and aortic aneurysm. We also discuss the future utility and challenges related to NGS in studying the genetic basis of CVDs in order to improve diagnosis, prevention, and treatment.
Collapse
Affiliation(s)
- Francesca Faita
- Francesca Faita, Cecilia Vecoli, Ilenia Foffa, Maria Grazia Andreassi, CNR, Institute of Clinical Physiology, 54100 Massa, Italy
| | | | | | | |
Collapse
|
47
|
Otsuka H, Arimura T, Abe T, Kawai H, Aizawa Y, Kubo T, Kitaoka H, Nakamura H, Nakamura K, Okamoto H, Ichida F, Ayusawa M, Nunoda S, Isobe M, Matsuzaki M, Doi YL, Fukuda K, Sasaoka T, Izumi T, Ashizawa N, Kimura A. Prevalence and Distribution of Sarcomeric Gene Mutations in Japanese Patients With Familial Hypertrophic Cardiomyopathy. Circ J 2012; 76:453-461. [DOI: 10.1253/circj.cj-11-0876] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Affiliation(s)
- Haruna Otsuka
- Department of Molecular Pathogenesis, Medical Research Institute, Tokyo Medical and Dental University
| | - Takuro Arimura
- Department of Molecular Pathogenesis, Medical Research Institute, Tokyo Medical and Dental University
| | - Tadaaki Abe
- Department of Pediatric Cardiovascular Medicine, National Cerebral and Cardiovascular Center
| | - Hiroya Kawai
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine
| | | | - Toru Kubo
- Department of Medicine and Geriatrics, Kochi Medical School
| | | | - Hiroshi Nakamura
- Department of Community Health and Medicine, Yamaguchi University School of Medicine
| | - Kazufumi Nakamura
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences
| | - Hiroshi Okamoto
- Department of Cardiovascular Medicine, Hokkaido Medical Center
| | - Fukiko Ichida
- Department of Pediatrics, Toyama University School of Medicine
| | - Mamoru Ayusawa
- Department of Pediatrics and Child Health, Nihon University School of Medicine
| | - Shinichi Nunoda
- Cardiovascular Division, Tokyo Women's Medical University Medical Center East
| | - Mitsuaki Isobe
- Department of Cardiovascular Medicine, Graduate School of Medical and Dental Science, Tokyo Medical and Dental University
| | - Masunori Matsuzaki
- Department of Cardiovascular Medicine, Yamaguchi University School of Medicine
| | | | - Keiichi Fukuda
- Department of Cardiology, Keio University School of Medicine
| | - Taishi Sasaoka
- Department of Cardiovascular Medicine, Kitasato University School of Medicine
| | - Toru Izumi
- Department of Cardiovascular Medicine, Kitasato University School of Medicine
| | - Naoto Ashizawa
- Department of Cardiovascular Medicine, Nagasaki University Graduate School of Biomedical Sciences
| | - Akinori Kimura
- Department of Molecular Pathogenesis, Medical Research Institute, Tokyo Medical and Dental University
| |
Collapse
|
48
|
Kojic S, Radojkovic D, Faulkner G. Muscle ankyrin repeat proteins: their role in striated muscle function in health and disease. Crit Rev Clin Lab Sci 2011; 48:269-94. [DOI: 10.3109/10408363.2011.643857] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
49
|
Benian GM, Epstein HF. Caenorhabditis elegans muscle: a genetic and molecular model for protein interactions in the heart. Circ Res 2011; 109:1082-95. [PMID: 21998299 DOI: 10.1161/circresaha.110.237685] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The nematode Caenorhabditis elegans has become established as a major experimental organism with applications to many biomedical research areas. The body wall muscle cells are a useful model for the study of human cardiomyocytes and their homologous structures and proteins. The ability to readily identify mutations affecting these proteins and structures in C elegans and to be able to rigorously characterize their genotypes and phenotypes at the cellular and molecular levels permits mechanistic studies of the responsible interactions relevant to the inherited human cardiomyopathies. Future work in C elegans muscle holds great promise in uncovering new mechanisms in the pathogenesis of these cardiac disorders.
Collapse
Affiliation(s)
- Guy M Benian
- Department of Pathology, Emory University, Atlanta, GA 30322, USA.
| | | |
Collapse
|
50
|
Niwano S. Multicenter study of the prevalence and distribution of sarcometric gene mutations in familial hypertrophic cardiomyopathy: – a milestone for genetic diagnosis in the Japanese population –. Circ J 2011; 76:303-4. [PMID: 22122969 DOI: 10.1253/circj.cj-11-1260] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|