1
|
Carabetta N, Siracusa C, Leo I, Panuccio G, Strangio A, Sabatino J, Torella D, De Rosa S. Cardiomyopathies: The Role of Non-Coding RNAs. Noncoding RNA 2024; 10:53. [PMID: 39449507 PMCID: PMC11503404 DOI: 10.3390/ncrna10060053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 10/16/2024] [Accepted: 10/21/2024] [Indexed: 10/26/2024] Open
Abstract
Cardiomyopathies are the structural and functional disorders of the myocardium. Etiopathogenesis is complex and involves an interplay of genetic, environmental, and lifestyle factors eventually leading to myocardial abnormalities. It is known that non-coding (Nc) RNAs, including micro (mi)-RNAs and long non-coding (lnc) RNAs, play a crucial role in regulating gene expression. Several studies have explored the role of miRNAs in the development of various pathologies, including heart diseases. In this review, we analyzed various patterns of ncRNAs expressed in the most common cardiomyopathies: dilated cardiomyopathy, hypertrophic cardiomyopathy and arrhythmogenic cardiomyopathy. Understanding the role of different ncRNAs implicated in cardiomyopathic processes may contribute to the identification of potential therapeutic targets and novel risk stratification models based on gene expression. The analysis of ncRNAs may also be helpful to unveil the molecular mechanisms subtended to these diseases.
Collapse
Affiliation(s)
- Nicole Carabetta
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy; (N.C.); (C.S.)
| | - Chiara Siracusa
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy; (N.C.); (C.S.)
| | - Isabella Leo
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy; (I.L.); (G.P.); (A.S.); (J.S.); (D.T.)
| | - Giuseppe Panuccio
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy; (I.L.); (G.P.); (A.S.); (J.S.); (D.T.)
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité Berlin, 12200 Berlin, Germany
| | - Antonio Strangio
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy; (I.L.); (G.P.); (A.S.); (J.S.); (D.T.)
| | - Jolanda Sabatino
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy; (I.L.); (G.P.); (A.S.); (J.S.); (D.T.)
| | - Daniele Torella
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy; (I.L.); (G.P.); (A.S.); (J.S.); (D.T.)
| | - Salvatore De Rosa
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy; (N.C.); (C.S.)
| |
Collapse
|
2
|
Orgil BO, Chintanaphol M, Alberson NR, Letourneau L, Martinez HR, Towbin JA, Purevjav E. Animal Models for Mechanical Circulatory Support: A Research Review. Rev Cardiovasc Med 2024; 25:351. [PMID: 39484122 PMCID: PMC11522838 DOI: 10.31083/j.rcm2510351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 04/25/2024] [Accepted: 06/19/2024] [Indexed: 11/03/2024] Open
Abstract
Heart failure is a clinical syndrome that has become a leading public health problem worldwide. Globally, nearly 64 million individuals are currently affected by heart failure, causing considerable medical, financial, and social challenges. One therapeutic option for patients with advanced heart failure is mechanical circulatory support (MCS) which is widely used for short-term or long-term management. MCS with various ventricular assist devices (VADs) has gained traction in end-stage heart failure treatment as a bridge-to-recovery, -decision, -transplant or -destination therapy. Due to limitations in studying VADs in humans, animal studies have substantially contributed to the development and advancement of MCS devices. Large animals have provided an avenue for developing and testing new VADs and improving surgical strategies for VAD implantation and for evaluating the effects and complications of MCS on hemodynamics and organ function. VAD modeling by utilizing rodents and small animals has been successfully implemented for investigating molecular mechanisms of cardiac unloading after the implantation of MCS. This review will cover the animal research that has resulted in significant advances in the development of MCS devices and the therapeutic care of advanced heart failure.
Collapse
Affiliation(s)
- Buyan-Ochir Orgil
- The Heart Institute, Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN 38103, USA
- Children’s Foundation Research Institute, Le Bonheur Children’s Hospital, Memphis, TN 38103, USA
| | - Michelle Chintanaphol
- College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Neely R. Alberson
- The Heart Institute, Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN 38103, USA
- Children’s Foundation Research Institute, Le Bonheur Children’s Hospital, Memphis, TN 38103, USA
| | | | - Hugo R. Martinez
- The Heart Institute, Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN 38103, USA
- Children’s Foundation Research Institute, Le Bonheur Children’s Hospital, Memphis, TN 38103, USA
| | - Jeffrey A. Towbin
- The Heart Institute, Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN 38103, USA
- Children’s Foundation Research Institute, Le Bonheur Children’s Hospital, Memphis, TN 38103, USA
- Pediatric Cardiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Enkhsaikhan Purevjav
- The Heart Institute, Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN 38103, USA
- Children’s Foundation Research Institute, Le Bonheur Children’s Hospital, Memphis, TN 38103, USA
| |
Collapse
|
3
|
Wang X, Baksh SS, Pratt RE, Dzau VJ, Hodgkinson CP. Modifying miRs for effective reprogramming of fibroblasts to cardiomyocytes. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102160. [PMID: 38495845 PMCID: PMC10943962 DOI: 10.1016/j.omtn.2024.102160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 02/22/2024] [Indexed: 03/19/2024]
Abstract
Reprogramming scar fibroblasts into cardiomyocytes has been proposed to reverse the damage associated with myocardial infarction. However, the limited improvement in cardiac function calls for enhanced strategies. We reported enhanced efficacy of our miR reprogramming cocktail miR combo (miR-1, miR-133a, miR-208a, and miR-499) via RNA-sensing receptor stimulation. We hypothesized that we could combine RNA-sensing receptor activation with fibroblast reprogramming by chemically modifying miR combo. To test the hypothesis, miR combo was modified to enhance interaction with the RNA-sensing receptor Rig1 via the addition of a 5'-triphosphate (5'ppp) group. Importantly, when compared with unmodified miR combo, 5'ppp-modified miR combo markedly improved reprogramming efficacy in vitro. Enhanced reprogramming efficacy correlated with a type-I interferon immune response with strong and selective secretion of interferon β (IFNβ). Antibody blocking studies and media replacement experiments indicated that 5'ppp-miR combo utilized IFNβ to enhance fibroblast reprogramming efficacy. In conclusion, miRs can acquire powerful additional roles through chemical modification that potentially increases their clinical applications.
Collapse
Affiliation(s)
- Xinghua Wang
- Mandel Center for Hypertension and Atherosclerosis, and the Duke Cardiovascular Research Center, Duke University Medical Center, Durham, NC 27710, USA
| | - Syeda S. Baksh
- Mandel Center for Hypertension and Atherosclerosis, and the Duke Cardiovascular Research Center, Duke University Medical Center, Durham, NC 27710, USA
| | - Richard E. Pratt
- Mandel Center for Hypertension and Atherosclerosis, and the Duke Cardiovascular Research Center, Duke University Medical Center, Durham, NC 27710, USA
| | - Victor J. Dzau
- Mandel Center for Hypertension and Atherosclerosis, and the Duke Cardiovascular Research Center, Duke University Medical Center, Durham, NC 27710, USA
| | - Conrad P. Hodgkinson
- Mandel Center for Hypertension and Atherosclerosis, and the Duke Cardiovascular Research Center, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
4
|
Delaunay M, Paterek A, Gautschi I, Scherler G, Diviani D. AKAP2-anchored extracellular signal-regulated kinase 1 (ERK1) regulates cardiac myofibroblast migration. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119674. [PMID: 38242328 DOI: 10.1016/j.bbamcr.2024.119674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 12/22/2023] [Accepted: 01/10/2024] [Indexed: 01/21/2024]
Abstract
Cardiac fibrosis is a major cause of dysfunctions and arrhythmias in failing hearts. At the cellular level fibrosis is mediated by cardiac myofibroblasts, which display an increased migratory capacity and secrete large amounts of extracellular matrix. These properties allow myofibroblasts to invade, remodel and stiffen the myocardium and eventually alter cardiac function. While the enhanced ability of cardiac myofibroblasts to migrate has been proposed to contribute to the initiation of the fibrotic process, the molecular mechanisms controlling their motile function have been poorly defined. In this context, our current findings indicate that A-kinase anchoring protein 2 (AKAP2) associates with actin at the leading edge of migrating cardiac myofibroblasts. Proteomic analysis of the AKAP2 interactome revealed that this anchoring protein assembles a signaling complex composed of the extracellular regulated kinase 1 (ERK1) and its upstream activator Grb2 that mediates the activation of ERK in cardiac myofibroblasts. Silencing AKAP2 expression results in a significant reduction in the phosphorylation of ERK1 and its downstream effector WAVE2, a protein involved in actin polymerization, and impairs the ability of cardiac myofibroblasts to migrate. Importantly, disruption of the interaction between AKAP2 and F-actin using cell-permeant competitor peptides, inhibits the activation of the ERK-WAVE2 signaling axis, resulting in a reduction of the translocation of Arp2 to the leading-edge membrane and in inhibition of cardiac myofibroblast migration. Collectively, these findings suggest that AKAP2 functions as an F-actin bound molecular scaffold mediating the activation of an ERK1-dependent promigratory transduction pathway in cardiac myofibroblasts.
Collapse
Affiliation(s)
- Marion Delaunay
- Department of Biomedical Sciences, Faculty of Biology et Medicine, University of Lausanne, 1011 Lausanne, Switzerland
| | - Aleksandra Paterek
- Department of Biomedical Sciences, Faculty of Biology et Medicine, University of Lausanne, 1011 Lausanne, Switzerland
| | - Ivan Gautschi
- Department of Biomedical Sciences, Faculty of Biology et Medicine, University of Lausanne, 1011 Lausanne, Switzerland
| | - Greta Scherler
- Department of Biomedical Sciences, Faculty of Biology et Medicine, University of Lausanne, 1011 Lausanne, Switzerland
| | - Dario Diviani
- Department of Biomedical Sciences, Faculty of Biology et Medicine, University of Lausanne, 1011 Lausanne, Switzerland.
| |
Collapse
|
5
|
Orgil BO, Purevjav E. Molecular Pathways and Animal Models of Cardiomyopathies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1441:991-1019. [PMID: 38884766 DOI: 10.1007/978-3-031-44087-8_64] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
Cardiomyopathies are a heterogeneous group of disorders of the heart muscle that ultimately result in congestive heart failure. Rapid progress in genetics, molecular and cellular biology with breakthrough innovative genetic-engineering techniques, such as next-generation sequencing and multiomics platforms, stem cell reprogramming, as well as novel groundbreaking gene-editing systems over the past 25 years has greatly improved the understanding of pathogenic signaling pathways in inherited cardiomyopathies. This chapter will focus on intracellular and intercellular molecular signaling pathways that are activated by a genetic insult in cardiomyocytes to maintain tissue and organ level regulation and resultant cardiac remodeling in certain forms of cardiomyopathies. In addition, animal models of different clinical forms of human cardiomyopathies with their summaries of triggered key molecules and signaling pathways will be described.
Collapse
Affiliation(s)
- Buyan-Ochir Orgil
- Department of Pediatrics, The Heart Institute, Division of Cardiology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Enkhsaikhan Purevjav
- Department of Pediatrics, The Heart Institute, Division of Cardiology, University of Tennessee Health Science Center, Memphis, TN, USA.
| |
Collapse
|
6
|
Marcoux E, Sosnowski D, Ninni S, Mackasey M, Cadrin-Tourigny J, Roberts JD, Olesen MS, Fatkin D, Nattel S. Genetic Atrial Cardiomyopathies: Common Features, Specific Differences, and Broader Relevance to Understanding Atrial Cardiomyopathy. Circ Arrhythm Electrophysiol 2023; 16:675-698. [PMID: 38018478 DOI: 10.1161/circep.123.003750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2023]
Abstract
Atrial cardiomyopathy is a condition that causes electrical and contractile dysfunction of the atria, often along with structural and functional changes. Atrial cardiomyopathy most commonly occurs in conjunction with ventricular dysfunction, in which case it is difficult to discern the atrial features that are secondary to ventricular dysfunction from those that arise as a result of primary atrial abnormalities. Isolated atrial cardiomyopathy (atrial-selective cardiomyopathy [ASCM], with minimal or no ventricular function disturbance) is relatively uncommon and has most frequently been reported in association with deleterious rare genetic variants. The genes involved can affect proteins responsible for various biological functions, not necessarily limited to the heart but also involving extracardiac tissues. Atrial enlargement and atrial fibrillation are common complications of ASCM and are often the predominant clinical features. Despite progress in identifying disease-causing rare variants, an overarching understanding and approach to the molecular pathogenesis, phenotypic spectrum, and treatment of genetic ASCM is still lacking. In this review, we aim to analyze the literature relevant to genetic ASCM to understand the key features of this rather rare condition, as well as to identify distinct characteristics of ASCM and its arrhythmic complications that are related to specific genotypes. We outline the insights that have been gained using basic research models of genetic ASCM in vitro and in vivo and correlate these with patient outcomes. Finally, we provide suggestions for the future investigation of patients with genetic ASCM and improvements to basic scientific models and systems. Overall, a better understanding of the genetic underpinnings of ASCM will not only provide a better understanding of this condition but also promises to clarify our appreciation of the more commonly occurring forms of atrial cardiomyopathy associated with ventricular dysfunction.
Collapse
Affiliation(s)
- Edouard Marcoux
- Research Center, Montreal Heart Institute, Université de Montréal. (E.M., D.S., S. Ninni, M.M., S. Nattel)
- Faculty of Pharmacy, Université de Montréal. (E.M.)
| | - Deanna Sosnowski
- Research Center, Montreal Heart Institute, Université de Montréal. (E.M., D.S., S. Ninni, M.M., S. Nattel)
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada (D.S., M.M., S. Nattel)
| | - Sandro Ninni
- Research Center, Montreal Heart Institute, Université de Montréal. (E.M., D.S., S. Ninni, M.M., S. Nattel)
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, France (S. Ninni)
| | - Martin Mackasey
- Research Center, Montreal Heart Institute, Université de Montréal. (E.M., D.S., S. Ninni, M.M., S. Nattel)
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada (D.S., M.M., S. Nattel)
| | - Julia Cadrin-Tourigny
- Cardiovascular Genetics Center, Montreal Heart Institute, Faculty of Medicine, Université de Montréal. (J.C.-T.)
| | - Jason D Roberts
- Population Health Research Institute, McMaster University and Hamilton Health Sciences, Canada (J.D.R.)
| | - Morten Salling Olesen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark (M.S.O.)
| | - Diane Fatkin
- Victor Chang Cardiac Research Institute, Darlinghurst (D.F.)
- School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Kensington (D.F.)
- Department of Cardiology, St Vincent's Hospital, Darlinghurst, NSW, Australia (D.F.)
| | - Stanley Nattel
- Research Center, Montreal Heart Institute, Université de Montréal. (E.M., D.S., S. Ninni, M.M., S. Nattel)
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal. (S. Nattel.)
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada (D.S., M.M., S. Nattel)
- Institute of Pharmacology. West German Heart and Vascular Center, University Duisburg-Essen, Germany (S. Nattel)
- IHU LYRIC & Fondation Bordeaux Université de Bordeaux, France (S. Nattel)
| |
Collapse
|
7
|
Castaldi B, Padalino M, Sirico D, Guariento A, Vida V, Di Salvo G. Hybrid approach for end-stage heart failure treatment in a 6-month-old baby. Cardiol Young 2023; 33:1738-1740. [PMID: 36927615 DOI: 10.1017/s104795112300046x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/18/2023]
Abstract
A 6-month-old baby girl, with a history of 2 months of intensive care management and two episodes of cardiac arrest, was transferred from another European country to initiate the "Giessen approach" for end-stage heart failure in children. At the admission, left ventricular ejection fraction was 20%. Severe mitral valve regurgitation and severe left atrial dilatation were present. Right ventricular function was preserved, and tricuspid valve regurgitation was mild. As a result, the patient underwent surgical pulmonary artery banding. Additionally, unloading of the left atrium was achieved by implanting an 8 mm atrial flow regulator device through a hybrid per-atrial approach. Two months after the procedure, the patient was progressively weaned from the inotropes and transferred to the ward.
Collapse
Affiliation(s)
- Biagio Castaldi
- Department of Women's and Children's Health, University of Padua, Italy
| | - Massimo Padalino
- Department of Cardiac Thoracic and Vascular Sciences and Public Health, University of Padua, Italy
| | - Domenico Sirico
- Department of Women's and Children's Health, University of Padua, Italy
| | - Alvise Guariento
- Department of Cardiac Thoracic and Vascular Sciences and Public Health, University of Padua, Italy
| | - Vladimiro Vida
- Department of Cardiac Thoracic and Vascular Sciences and Public Health, University of Padua, Italy
| | - Giovanni Di Salvo
- Department of Women's and Children's Health, University of Padua, Italy
| |
Collapse
|
8
|
Srivastava S, Gajwani P, Jousma J, Miyamoto H, Kwon Y, Jana A, Toth PT, Yan G, Ong SG, Rehman J. Nuclear translocation of mitochondrial dehydrogenases as an adaptive cardioprotective mechanism. Nat Commun 2023; 14:4360. [PMID: 37468519 DOI: 10.1038/s41467-023-40084-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 07/12/2023] [Indexed: 07/21/2023] Open
Abstract
Chemotherapy-induced cardiac damage remains a leading cause of death amongst cancer survivors. Anthracycline-induced cardiotoxicity is mediated by severe mitochondrial injury, but little is known about the mechanisms by which cardiomyocytes adaptively respond to the injury. We observed the translocation of selected mitochondrial tricarboxylic acid (TCA) cycle dehydrogenases to the nucleus as an adaptive stress response to anthracycline-cardiotoxicity in human induced pluripotent stem cell-derived cardiomyocytes and in vivo. The expression of nuclear-targeted mitochondrial dehydrogenases shifts the nuclear metabolic milieu to maintain their function both in vitro and in vivo. This protective effect is mediated by two parallel pathways: metabolite-induced chromatin accessibility and AMP-kinase (AMPK) signaling. The extent of chemotherapy-induced cardiac damage thus reflects a balance between mitochondrial injury and the protective response initiated by the nuclear pool of mitochondrial dehydrogenases. Our study identifies nuclear translocation of mitochondrial dehydrogenases as an endogenous adaptive mechanism that can be leveraged to attenuate cardiomyocyte injury.
Collapse
Affiliation(s)
- Shubhi Srivastava
- Department of Biochemistry and Molecular Genetics, The University of Illinois College of Medicine, Chicago, IL, USA
| | - Priyanka Gajwani
- Department of Biochemistry and Molecular Genetics, The University of Illinois College of Medicine, Chicago, IL, USA
| | - Jordan Jousma
- Department of Pharmacology and Regenerative Medicine, The University of Illinois College of Medicine, Chicago, IL, USA
| | - Hiroe Miyamoto
- Department of Pharmacology and Regenerative Medicine, The University of Illinois College of Medicine, Chicago, IL, USA
| | - Youjeong Kwon
- Department of Pharmacology and Regenerative Medicine, The University of Illinois College of Medicine, Chicago, IL, USA
| | - Arundhati Jana
- Division of Cardiology, Department of Medicine, The University of Illinois College of Medicine, Chicago, IL, USA
| | - Peter T Toth
- Department of Pharmacology and Regenerative Medicine, The University of Illinois College of Medicine, Chicago, IL, USA
- Research Resources Center, University of Illinois, Chicago, IL, USA
| | - Gege Yan
- Department of Pharmacology and Regenerative Medicine, The University of Illinois College of Medicine, Chicago, IL, USA
- Division of Cardiology, Department of Medicine, The University of Illinois College of Medicine, Chicago, IL, USA
| | - Sang-Ging Ong
- Department of Pharmacology and Regenerative Medicine, The University of Illinois College of Medicine, Chicago, IL, USA.
- Division of Cardiology, Department of Medicine, The University of Illinois College of Medicine, Chicago, IL, USA.
| | - Jalees Rehman
- Department of Biochemistry and Molecular Genetics, The University of Illinois College of Medicine, Chicago, IL, USA.
- Department of Pharmacology and Regenerative Medicine, The University of Illinois College of Medicine, Chicago, IL, USA.
- Division of Cardiology, Department of Medicine, The University of Illinois College of Medicine, Chicago, IL, USA.
- University of Illinois Cancer Center, Chicago, IL, USA.
| |
Collapse
|
9
|
Hu X, Kong J, Niu T, Chen L, Yang J. Single coronary artery presenting dilated cardiomyopathy and hyperlipidemia with the SCN5A and APOA5 gene mutation: A case report and review of the literature. Front Cardiovasc Med 2023; 10:1113886. [PMID: 37288251 PMCID: PMC10242075 DOI: 10.3389/fcvm.2023.1113886] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 03/23/2023] [Indexed: 06/09/2023] Open
Abstract
We present a 55-year-old man with chest tightness and dyspnoea after activity lasting for 2 months who was diagnosed with single coronary artery (SCA) and presented with dilated cardiomyopathy (DCM) with the c.1858C > T mutation in the SCN5A gene. The computed tomography coronary angiogram (CTCA) showed congenital absence of the right coronary artery (RCA), and the right heart was nourished by the left coronary artery branch with no apparent stenosis. Transthoracic echocardiography (TTE) revealed enlargement of the left heart and cardiomyopathy. Cardiac magnetic resonance imaging (CMR) revealed DCM. Genetic testing showed that the c.1858C > T variant of the SCN5A gene could lead to Brugada syndrome and DCM. SCA is a rare congenital anomaly of the coronary anatomy, and this case reported as SCA accompanied by DCM is even rarer. We present a rare case of a 55-year-old man with DCM with the c.1858C > T (p. Arg620Cys)/c.1008G > A (p.(Pro336=) variant of the SCN5A gene, congenital absence of RCA, and c.990_993delAACA (p. Asp332Valfs*5) variant of the APOA5 gene. To our knowledge, this is the first report of DCM combined with the SCN5A gene mutation in SCA after searching the PubMed, CNKI and Wanfang databases.
Collapse
Affiliation(s)
- Xiaoxia Hu
- Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Jing Kong
- Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Tingting Niu
- Department of Medical Technology, Jinan Vocational College of Nursing, Jinan, Shandong, China
| | - Liang Chen
- Department of Emergency Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Jingjing Yang
- Department of Emergency Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, China
| |
Collapse
|
10
|
Cheng X, Jian D, Xing J, Liu C, Liu Y, Cui C, Li Z, Wang S, Li R, Ma X, Wang Y, Gu X, Ge Z, Tang H, Liu L. Circulating cardiac MicroRNAs safeguard against dilated cardiomyopathy. Clin Transl Med 2023; 13:e1258. [PMID: 37138538 PMCID: PMC10157268 DOI: 10.1002/ctm2.1258] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 03/28/2023] [Accepted: 04/25/2023] [Indexed: 05/05/2023] Open
Abstract
BACKGROUND Cardiac-resident or -enriched microRNAs (miRNAs) could be released into the bloodstream becoming circulating cardiac miRNAs, which are increasingly recognized as non-invasive and accessible biomarkers of multiple heart diseases. However, dilated cardiomyopathy (DCM)-associated circulating miRNAs (DACMs) and their roles in DCM pathogenesis remain largely unexplored. METHODS Two human cohorts, consisting of healthy individuals and DCM patients, were enrolled for serum miRNA sequencing (10 vs. 10) and quantitative polymerase chain reaction validation (46 vs. 54), respectively. Rigorous screening strategy was enacted to define DACMs and their potentials for diagnosis. DCM mouse model, different sources of cardiomyocytes, adeno-associated virus 9 (AAV9), gene knockout, RNAscope miRNA in situ hybridization, mRFP-GFP-LC3B reporter, echocardiography and transmission electron microscopy were adopted for mechanistic explorations. RESULTS Serum miRNA sequencing revealed a unique expression pattern for DCM circulating miRNAs. DACMs miR-26a-5p, miR-30c-5p, miR-126-5p and miR-126-3p were found to be depleted in DCM circulation as well as heart tissues. Their expressions in circulation and heart tissues were proven to be correlated significantly, and a combination of these miRNAs was suggested potential values for DCM diagnosis. FOXO3, a predicted common target, was experimentally demonstrated to be co-repressed within cardiomyocytes by these DACMs except miR-26a-5p. Delivery of a combination of miR-30c-5p, miR-126-5p and miR-126-3p into the murine myocardium via AAV9 carrying an expression cassette driven by cTnT promoter, or cardiac-specific knockout of FOXO3 (Myh6-CreERT2 , FOXO3 flox+/+ ) dramatically attenuated cardiac apoptosis and autophagy involved in DCM progression. Moreover, competitively disrupting the interplay between DACMs and FOXO3 mRNA by specifically introducing their interacting regions into murine myocardium crippled the cardioprotection of DACMs against DCM. CONCLUSIONS Circulating cardiac miRNA-FOXO3 axis plays a pivotal role in safeguarding against myocardial apoptosis and excessive autophagy in DCM development, which may provide serological cues for DCM non-invasive diagnosis and shed light on DCM pathogenesis and therapeutic targets.
Collapse
Affiliation(s)
- Xiaolei Cheng
- National Health Commission Key Laboratory of Cardiovascular Regenerative MedicineHeart Center of Henan Provincial People's HospitalCentral China Fuwai Hospital of Zhengzhou UniversityFuwai Central China Cardiovascular Hospital and Central China Branch of National Center for Cardiovascular DiseasesZhengzhouChina
- Department of AnesthesiologyAffiliated Drum Tower Hospital of Medical School of Nanjing UniversityNanjingChina
| | - Dongdong Jian
- National Health Commission Key Laboratory of Cardiovascular Regenerative MedicineHeart Center of Henan Provincial People's HospitalCentral China Fuwai Hospital of Zhengzhou UniversityFuwai Central China Cardiovascular Hospital and Central China Branch of National Center for Cardiovascular DiseasesZhengzhouChina
- Department of Biochemistry and Molecular BiologyBeijing Key Laboratory of Protein Posttranslational Modifications and Cell FunctionSchool of Basic Medical SciencesPeking University Health Science CenterBeijingChina
| | - Junyue Xing
- National Health Commission Key Laboratory of Cardiovascular Regenerative MedicineHeart Center of Henan Provincial People's HospitalCentral China Fuwai Hospital of Zhengzhou UniversityFuwai Central China Cardiovascular Hospital and Central China Branch of National Center for Cardiovascular DiseasesZhengzhouChina
- Henan Key Laboratory of Chronic Disease ManagementDepartment of Health Management CenterHenan Provincial People's HospitalDepartment of Health Management Center of Central China Fuwai HospitalCentral China Fuwai Hospital of Zhengzhou UniversityZhengzhouChina
| | - Cihang Liu
- Department of AnesthesiologyAffiliated Drum Tower Hospital of Medical School of Nanjing UniversityNanjingChina
- Department of Biochemistry and Molecular BiologyBeijing Key Laboratory of Protein Posttranslational Modifications and Cell FunctionSchool of Basic Medical SciencesPeking University Health Science CenterBeijingChina
| | - Yong Liu
- National Health Commission Key Laboratory of Cardiovascular Regenerative MedicineHeart Center of Henan Provincial People's HospitalCentral China Fuwai Hospital of Zhengzhou UniversityFuwai Central China Cardiovascular Hospital and Central China Branch of National Center for Cardiovascular DiseasesZhengzhouChina
- Department of PhysiologyShanxi Medical UniversityTaiyuanChina
| | - Cunying Cui
- National Health Commission Key Laboratory of Cardiovascular Regenerative MedicineHeart Center of Henan Provincial People's HospitalCentral China Fuwai Hospital of Zhengzhou UniversityFuwai Central China Cardiovascular Hospital and Central China Branch of National Center for Cardiovascular DiseasesZhengzhouChina
| | - Zhen Li
- National Health Commission Key Laboratory of Cardiovascular Regenerative MedicineHeart Center of Henan Provincial People's HospitalCentral China Fuwai Hospital of Zhengzhou UniversityFuwai Central China Cardiovascular Hospital and Central China Branch of National Center for Cardiovascular DiseasesZhengzhouChina
| | - Shixing Wang
- National Health Commission Key Laboratory of Cardiovascular Regenerative MedicineHeart Center of Henan Provincial People's HospitalCentral China Fuwai Hospital of Zhengzhou UniversityFuwai Central China Cardiovascular Hospital and Central China Branch of National Center for Cardiovascular DiseasesZhengzhouChina
| | - Ran Li
- National Health Commission Key Laboratory of Cardiovascular Regenerative MedicineHeart Center of Henan Provincial People's HospitalCentral China Fuwai Hospital of Zhengzhou UniversityFuwai Central China Cardiovascular Hospital and Central China Branch of National Center for Cardiovascular DiseasesZhengzhouChina
| | - Xiaohan Ma
- National Health Commission Key Laboratory of Cardiovascular Regenerative MedicineHeart Center of Henan Provincial People's HospitalCentral China Fuwai Hospital of Zhengzhou UniversityFuwai Central China Cardiovascular Hospital and Central China Branch of National Center for Cardiovascular DiseasesZhengzhouChina
| | - Yingying Wang
- National Health Commission Key Laboratory of Cardiovascular Regenerative MedicineHeart Center of Henan Provincial People's HospitalCentral China Fuwai Hospital of Zhengzhou UniversityFuwai Central China Cardiovascular Hospital and Central China Branch of National Center for Cardiovascular DiseasesZhengzhouChina
| | - Xiaoping Gu
- Department of AnesthesiologyAffiliated Drum Tower Hospital of Medical School of Nanjing UniversityNanjingChina
| | - Zhenwei Ge
- National Health Commission Key Laboratory of Cardiovascular Regenerative MedicineHeart Center of Henan Provincial People's HospitalCentral China Fuwai Hospital of Zhengzhou UniversityFuwai Central China Cardiovascular Hospital and Central China Branch of National Center for Cardiovascular DiseasesZhengzhouChina
| | - Hao Tang
- National Health Commission Key Laboratory of Cardiovascular Regenerative MedicineHeart Center of Henan Provincial People's HospitalCentral China Fuwai Hospital of Zhengzhou UniversityFuwai Central China Cardiovascular Hospital and Central China Branch of National Center for Cardiovascular DiseasesZhengzhouChina
- Henan Key Laboratory of Chronic Disease ManagementDepartment of Health Management CenterHenan Provincial People's HospitalDepartment of Health Management Center of Central China Fuwai HospitalCentral China Fuwai Hospital of Zhengzhou UniversityZhengzhouChina
| | - Lin Liu
- National Health Commission Key Laboratory of Cardiovascular Regenerative MedicineHeart Center of Henan Provincial People's HospitalCentral China Fuwai Hospital of Zhengzhou UniversityFuwai Central China Cardiovascular Hospital and Central China Branch of National Center for Cardiovascular DiseasesZhengzhouChina
| |
Collapse
|
11
|
Pagano M, Fumagalli C, Girolami F, Passantino S, Gozzini A, Brambilla A, Spinelli V, Morrone A, Procopio E, Pochiero F, Donati MA, Olivotto I, Favilli S. Clinical profile and outcome of cardiomyopathies in infants and children seen at a tertiary centre. Int J Cardiol 2023; 371:516-522. [PMID: 36130621 DOI: 10.1016/j.ijcard.2022.09.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 07/12/2022] [Accepted: 09/15/2022] [Indexed: 01/05/2023]
Abstract
INTRODUCTION Due to their rare prevalence and marked heterogeneity, pediatric cardiomyopathies (CMPs) are little known and scarcely reported. We report the etiology, clinical profile and outcome of a consecutive cohort of children diagnosed with CMP and followed at Meyer Children's Hospital over a decade. PATIENTS AND METHODS We retrospectively reviewed patients consecutively referred from May 2008 to May 2019 for pediatric onset CMP (<18 years). Heart disease caused by arrhythmic disorders, toxic agents, rheumatic conditions and maternal disease were excluded. RESULTS We enrolled 110 patients (65 males), diagnosed at a median age of 27 [4-134] months; 35% had an infant onset (<1 year of age). A positive family history was more often associated with childhood-onset (38.8%). Hypertrophic cardiomyopathy (HCM; 48 patients) was the most frequent phenotype, followed by dilated cardiomyopathy (DCM; 35 patients). While metabolic and idiopathic etiologies were preponderant in infants, metabolic and sarcomeric diseases were most frequent in the childhood-onset group. Major adverse cardiac events (MACE) occurred in 31.8% of patients, including hospitalization for acute heart failure in 25.5% of patients, most commonly due to DCM. Overall, the most severe outcomes were documented in patients with metabolic diseases. CONCLUSIONS In a consecutive cohort of pediatric patients with CMP, those with infantile onset and with a metabolic etiology had the worst prognosis. Overall, MACE occurred in 41% of the entire population, most commonly associated with DCM, inborn errors of metabolism and genetic syndromes. Systematic NGS genetic testing was critical for etiological diagnosis and management.
Collapse
Affiliation(s)
- M Pagano
- Cardiology Unit, Meyer Children's Hospital, Florence, Italy.
| | - C Fumagalli
- Cardiomyopathies Unit, Careggi University Hospital (AOUC), Florence, Italy
| | - F Girolami
- Cardiology Unit, Meyer Children's Hospital, Florence, Italy
| | - S Passantino
- Cardiology Unit, Meyer Children's Hospital, Florence, Italy
| | - A Gozzini
- Cardiology Unit, Meyer Children's Hospital, Florence, Italy
| | - A Brambilla
- Cardiology Unit, Meyer Children's Hospital, Florence, Italy
| | - V Spinelli
- Cardiology Unit, Meyer Children's Hospital, Florence, Italy
| | - A Morrone
- Molecular and Cell Biology Laboratory of Neurometabolic Diseases, Meyer Children's Hospital, Neuroscience Department, Florence, Italy; Department of NEUROFARBA, University of Florence, Florence, Italy
| | - E Procopio
- Metabolic and Muscular Unit, Meyer Children's Hospital, Neuroscience Department, Florence, Italy
| | - F Pochiero
- Metabolic and Muscular Unit, Meyer Children's Hospital, Neuroscience Department, Florence, Italy
| | - M A Donati
- Metabolic and Muscular Unit, Meyer Children's Hospital, Neuroscience Department, Florence, Italy
| | - I Olivotto
- Cardiology Unit, Meyer Children's Hospital, Florence, Italy; Cardiomyopathies Unit, Careggi University Hospital (AOUC), Florence, Italy; Department of Experimental and Clinical Medicine, University of Florence, Italy
| | - S Favilli
- Cardiology Unit, Meyer Children's Hospital, Florence, Italy
| |
Collapse
|
12
|
Spurrell CH, Barozzi I, Kosicki M, Mannion BJ, Blow MJ, Fukuda-Yuzawa Y, Slaven N, Afzal SY, Akiyama JA, Afzal V, Tran S, Plajzer-Frick I, Novak CS, Kato M, Lee EA, Garvin TH, Pham QT, Kronshage AN, Lisgo S, Bristow J, Cappola TP, Morley MP, Margulies KB, Pennacchio LA, Dickel DE, Visel A. Genome-wide fetalization of enhancer architecture in heart disease. Cell Rep 2022; 40:111400. [PMID: 36130500 PMCID: PMC9534044 DOI: 10.1016/j.celrep.2022.111400] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 06/10/2022] [Accepted: 09/01/2022] [Indexed: 11/22/2022] Open
Abstract
Heart disease is associated with re-expression of key transcription factors normally active only during prenatal development of the heart. However, the impact of this reactivation on the regulatory landscape in heart disease is unclear. Here, we use RNA-seq and ChIP-seq targeting a histone modification associated with active transcriptional enhancers to generate genome-wide enhancer maps from left ventricle tissue from up to 26 healthy controls, 18 individuals with idiopathic dilated cardiomyopathy (DCM), and five fetal hearts. Healthy individuals have a highly reproducible epigenomic landscape, consisting of more than 33,000 predicted heart enhancers. In contrast, we observe reproducible disease-associated changes in activity at 6,850 predicted heart enhancers. Combined analysis of adult and fetal samples reveals that the heart disease epigenome and transcriptome both acquire fetal-like characteristics, with 3,400 individual enhancers sharing fetal regulatory properties. We also provide a comprehensive data resource (http://heart.lbl.gov) for the mechanistic exploration of DCM etiology.
Collapse
Affiliation(s)
- Cailyn H Spurrell
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Iros Barozzi
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Michael Kosicki
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Brandon J Mannion
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Matthew J Blow
- U.S. Department of Energy Joint Genome Institute, Walnut Creek, CA 94598, USA
| | - Yoko Fukuda-Yuzawa
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Neil Slaven
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Sarah Y Afzal
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Jennifer A Akiyama
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Veena Afzal
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Stella Tran
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Ingrid Plajzer-Frick
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Catherine S Novak
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Momoe Kato
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Elizabeth A Lee
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Tyler H Garvin
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Quan T Pham
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Anne N Kronshage
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Steven Lisgo
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - James Bristow
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Thomas P Cappola
- Cardiovascular Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Michael P Morley
- Cardiovascular Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Kenneth B Margulies
- Cardiovascular Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Len A Pennacchio
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA; U.S. Department of Energy Joint Genome Institute, Walnut Creek, CA 94598, USA; Comparative Biochemistry Program, University of California, Berkeley, Berkeley, CA 94720, USA.
| | - Diane E Dickel
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Axel Visel
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA; U.S. Department of Energy Joint Genome Institute, Walnut Creek, CA 94598, USA; School of Natural Sciences, University of California, Merced, Merced, CA 95343, USA.
| |
Collapse
|
13
|
Li H, Wu F, Huang G, Wu D, Wang T, Wang X, Wang K, Feng Y, Xu A. Cardiomyocytes induced from hiPSCs by well-defined compounds have therapeutic potential in heart failure by secreting PDGF-BB. Signal Transduct Target Ther 2022; 7:253. [PMID: 35902567 PMCID: PMC9334380 DOI: 10.1038/s41392-022-01045-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Revised: 04/27/2022] [Accepted: 05/30/2022] [Indexed: 01/14/2023] Open
Abstract
Recent studies have suggested that transplant of hiPS-CMs is a promising approach for treating heart failure. However, the optimally clinical benefits have been hampered by the immature nature of the hiPS-CMs, and the hiPS-CMs-secreted proteins contributing to the repair of cardiomyocytes remain largely unidentified. Here, we established a saponin+ compound optimally induced system to generate hiPS-CMs with stable functional attributes in vitro and transplanted in heart failure mice. Our study showed enhanced therapeutic effects of optimally induced hiPS-CMs by attenuating cardiac remodeling and dysfunction, these beneficial effects were concomitant with reduced cardiomyocytes death and increased angiogenesis. Moreover, the optimally induced hiPS-CMs could gathering to the injured heart and secret an abundant PDGF-BB. The reparative effect of the optimally induced hiPS-CMs in the hypoxia-injured HCMs was mimicked by PDGF-BB but inhibited by PDGF-BB neutralizing antibody, which was accompanied by the changed expression of p-PI3K and p-Akt proteins. It is highly possible that the PI3K/Akt pathway is regulated by the PDGF-BB secreted from the compound induced hiPS-CMs to achieve a longer lasting myocardial repair effect compared with the standard induced hiPS-CMs. Taken together, our data strongly implicate that the compound induced hiPS-CMs promote the recovery of injured hearts via paracrine action. In this process, the paracrine factor PDGF-BB derived from the compound induced hiPS-CMs reduces isoproterenol-induced adverse cardiac remodeling, which is associated with improved cardiac function, and these effects are mediated by the PI3K/Akt pathway, suggesting that the optimally induced hiPS-CMs may serve as a new promising cell therapy for clinical applications.
Collapse
Affiliation(s)
- Hongmei Li
- School of Life Science, Beijing University of Chinese Medicine, Beijing, China.,Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Fenfang Wu
- Shenzhen Hospital, Beijing University of Chinese Medicine, Shenzhen, China
| | - Guangrui Huang
- School of Life Science, Beijing University of Chinese Medicine, Beijing, China
| | - Di Wu
- College of Life Sciences, Sun Yat-Sen University, Guangzhou, China
| | - Ting Wang
- School of Life Science, Beijing University of Chinese Medicine, Beijing, China
| | - Xiashuang Wang
- College of Life Sciences, Sun Yat-Sen University, Guangzhou, China
| | - Kai Wang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yuyin Feng
- School of Life Science, Beijing University of Chinese Medicine, Beijing, China
| | - Anlong Xu
- School of Life Science, Beijing University of Chinese Medicine, Beijing, China. .,College of Life Sciences, Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|
14
|
Martínez-Falguera D, Iborra-Egea O, Gálvez-Montón C. iPSC Therapy for Myocardial Infarction in Large Animal Models: Land of Hope and Dreams. Biomedicines 2021; 9:1836. [PMID: 34944652 PMCID: PMC8698445 DOI: 10.3390/biomedicines9121836] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 11/30/2021] [Accepted: 12/01/2021] [Indexed: 02/07/2023] Open
Abstract
Myocardial infarction is the main driver of heart failure due to ischemia and subsequent cell death, and cell-based strategies have emerged as promising therapeutic methods to replace dead tissue in cardiovascular diseases. Research in this field has been dramatically advanced by the development of laboratory-induced pluripotent stem cells (iPSCs) that harbor the capability to become any cell type. Like other experimental strategies, stem cell therapy must meet multiple requirements before reaching the clinical trial phase, and in vivo models are indispensable for ensuring the safety of such novel therapies. Specifically, translational studies in large animal models are necessary to fully evaluate the therapeutic potential of this approach; to empirically determine the optimal combination of cell types, supplementary factors, and delivery methods to maximize efficacy; and to stringently assess safety. In the present review, we summarize the main strategies employed to generate iPSCs and differentiate them into cardiomyocytes in large animal species; the most critical differences between using small versus large animal models for cardiovascular studies; and the strategies that have been pursued regarding implanted cells' stage of differentiation, origin, and technical application.
Collapse
Affiliation(s)
- Daina Martínez-Falguera
- Faculty of Medicine, University of Barcelona (UB), 08036 Barcelona, Spain;
- ICREC Research Program, Germans Trias i Pujol Health Research Institute, Can Ruti Campus, 08916 Badalona, Spain;
- Heart Institute (iCor), Germans Trias i Pujol University Hospital, 08916 Badalona, Spain
| | - Oriol Iborra-Egea
- ICREC Research Program, Germans Trias i Pujol Health Research Institute, Can Ruti Campus, 08916 Badalona, Spain;
- Heart Institute (iCor), Germans Trias i Pujol University Hospital, 08916 Badalona, Spain
| | - Carolina Gálvez-Montón
- ICREC Research Program, Germans Trias i Pujol Health Research Institute, Can Ruti Campus, 08916 Badalona, Spain;
- Heart Institute (iCor), Germans Trias i Pujol University Hospital, 08916 Badalona, Spain
- CIBERCV, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Institut d’Investigació Biomèdica de Bellvitge-IDIBELL, L’Hospitalet de Llobregat, 08908 Barcelona, Spain
| |
Collapse
|
15
|
Abstract
Barth syndrome is a rare and potentially fatal X-linked disease characterized by cardiomyopathy, skeletal muscle weakness, growth delays, and cyclic neutropenia. Patients with Barth syndrome are prone to high risk of mortality in infancy and the development of cardiomyopathy with severe weakening of the immune system. Elamipretide is a water-soluble, aromatic-cationic, mitochondria-targeting tetrapeptide that readily penetrates and transiently localizes to the inner mitochondrial membrane. Therapy with elamipretide facilitates cell health by improving energy production and inhibiting excessive formation of reactive oxygen species, thus alleviating oxidative stress. Elamipretide crosses the outer membrane of the mitochondrion and becomes associated with cardiolipin, a constituent phospholipid of the inner membrane. Elamipretide improves mitochondrial bioenergetics and morphology rapidly in induced pluripotent stem cells from patients with Barth syndrome and other genetically related diseases characterized by pediatric cardiomyopathy. Data with elamipretide across multiple models of disease are especially promising, with results from several studies supporting the use of elamipretide as potential therapy for patients with Barth syndrome, particularly where there is a confirmed diagnosis of cardiomyopathy. This review highlights the challenges and opportunities presented in treating Barth syndrome cardiomyopathy patients with elamipretide and addresses evidence supporting the durability of effect of elamipretide as a therapeutic agent for Barth syndrome, especially its likely durable effects on progression of cardiomyopathy following the cessation of drug treatment and the capability of elamipretide to structurally reverse remodel the failing left ventricle at the global, cellular, and molecular level in a gradual manner through specific targeting of the mitochondrial inner membrane.
Collapse
|
16
|
Akhtar MM, Lorenzini M, Pavlou M, Ochoa JP, O’Mahony C, Restrepo-Cordoba MA, Segura-Rodriguez D, Bermúdez-Jiménez F, Molina P, Cuenca S, Ader F, Larrañaga-Moreira JM, Sabater-Molina M, Garcia-Alvarez MI, Arantzamendi LG, Truszkowska G, Ortiz-Genga M, Ruiz IS, Nielsen SK, Rasmussen TB, Robles Mezcua A, Alvarez-Rubio J, Eiskjaer H, Gautel M, Garcia-Pinilla JM, Ripoll-Vera T, Mogensen J, Limeres Freire J, Rodríguez-Palomares JF, Peña-Peña ML, Rangel-Sousa D, Palomino-Doza J, Arana Achaga X, Bilinska Z, Zamarreño Golvano E, Climent V, Peñalver MN, Barriales-Villa R, Charron P, Yotti R, Zorio E, Jiménez-Jáimez J, Garcia-Pavia P, Elliott PM. Association of Left Ventricular Systolic Dysfunction Among Carriers of Truncating Variants in Filamin C With Frequent Ventricular Arrhythmia and End-stage Heart Failure. JAMA Cardiol 2021; 6:891-901. [PMID: 33978673 PMCID: PMC8117057 DOI: 10.1001/jamacardio.2021.1106] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 02/22/2021] [Indexed: 12/28/2022]
Abstract
Importance Truncating variants in the gene encoding filamin C (FLNCtv) are associated with arrhythmogenic and dilated cardiomyopathies with a reportedly high risk of ventricular arrhythmia. Objective To determine the frequency of and risk factors associated with adverse events among FLNCtv carriers compared with individuals carrying TTN truncating variants (TTNtv). Design, Setting, and Participants This cohort study recruited 167 consecutive FLNCtv carriers and a control cohort of 244 patients with TTNtv matched for left ventricular ejection fraction (LVEF) from 19 European cardiomyopathy referral units between 1990 and 2018. Data analyses were conducted between June and October, 2020. Main Outcomes and Measures The primary end point was a composite of malignant ventricular arrhythmia (MVA) (sudden cardiac death, aborted sudden cardiac death, appropriate implantable cardioverter-defibrillator shock, and sustained ventricular tachycardia) and end-stage heart failure (heart transplant or mortality associated with end-stage heart failure). The secondary end point comprised MVA events only. Results In total, 167 patients with FLNCtv were studied (55 probands [33%]; 89 men [53%]; mean [SD] age at baseline evaluation, 43 [18] years). For a median follow-up of 20 months (interquartile range, 7-60 months), 29 patients (17.4%) reached the primary end point (19 patients with MVA and 10 patients with end-stage heart failure). Eight (44%) arrhythmic events occurred among individuals with baseline mild to moderate left ventricular systolic dysfunction (LVSD) (LVEF = 36%-49%). Univariable risk factors associated with the primary end point included proband status, LVEF decrement per 10%, ventricular ectopy (≥500 in 24 hours) and myocardial fibrosis detected on cardiac magnetic resonance imaging. The LVEF decrement (hazard ratio [HR] per 10%, 1.83 [95% CI, 1.30-2.57]; P < .001) and proband status (HR, 3.18 [95% CI, 1.12-9.04]; P = .03) remained independent risk factors on multivariable analysis (excluding myocardial fibrosis and ventricular ectopy owing to case censoring). There was no difference in freedom from MVA between FLNCtv carriers with mild to moderate or severe (LVEF ≤35%) LVSD (HR, 1.29 [95% CI, 0.45-3.72]; P = .64). Carriers of FLNCtv with impaired LVEF at baseline evaluation (n = 69) had reduced freedom from MVA compared with 244 TTNtv carriers with similar baseline LVEF (for mild to moderate LVSD: HR, 16.41 [95% CI, 3.45-78.11]; P < .001; for severe LVSD: HR, 2.47 [95% CI, 1.04-5.87]; P = .03). Conclusions and Relevance The high frequency of MVA among patients with FLNCtv with mild to moderate LVSD suggests that higher LVEF values than those currently recommended should be considered for prophylactic implantable cardioverter-defibrillator therapy in FLNCtv carriers.
Collapse
MESH Headings
- Adult
- Cardiomyopathy, Dilated/genetics
- Cardiomyopathy, Dilated/mortality
- Cardiomyopathy, Dilated/physiopathology
- Cardiomyopathy, Dilated/therapy
- Codon, Nonsense
- Connectin/genetics
- Death, Sudden, Cardiac/prevention & control
- Defibrillators, Implantable
- Female
- Filamins/genetics
- Heart Failure/genetics
- Heart Failure/mortality
- Heart Failure/physiopathology
- Heart Failure/therapy
- Heart Transplantation/statistics & numerical data
- Humans
- Male
- Middle Aged
- Mutation
- Stroke Volume
- Tachycardia, Ventricular/epidemiology
- Tachycardia, Ventricular/genetics
- Tachycardia, Ventricular/physiopathology
- Ventricular Dysfunction, Left/genetics
- Ventricular Dysfunction, Left/physiopathology
Collapse
Affiliation(s)
- Mohammed Majid Akhtar
- Department of Inherited Cardiovascular Diseases, Bart’s Heart Centre St Bartholomew’s Hospital, London, United Kingdom
- Institute of Cardiovascular Science, University College London, London, United Kingdom
| | - Massimiliano Lorenzini
- Department of Inherited Cardiovascular Diseases, Bart’s Heart Centre St Bartholomew’s Hospital, London, United Kingdom
| | - Menelaos Pavlou
- Department of Statistical Science, University College London, London, United Kingdom
| | | | - Constantinos O’Mahony
- Department of Inherited Cardiovascular Diseases, Bart’s Heart Centre St Bartholomew’s Hospital, London, United Kingdom
- Institute of Cardiovascular Science, University College London, London, United Kingdom
| | - Maria Alejandra Restrepo-Cordoba
- Department of Cardiology, Universidad Francisco de Vitoria, Pozuelo de Alarcón, Spain
- Heart Failure and Inherited Cardiac Diseases Unit, Department of Cardiology, Hospital Universitario Puerta de Hierro, Madrid, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart (ERN GUARDHEART)
| | | | | | - Pilar Molina
- Pathology Department, Institute of Legal Medicine and Forensic Sciences of Valencia and Faculty of Medicine of the Universitat de València, CAFAMUSME Research Group, IIS La Fe, Valencia, Spain
| | - Sofia Cuenca
- Hospital General Universitario Gregorio Marañon, Madrid, Spain
- Instituto de Investigación Sanitarias Gregorio Marañón, Spain
| | - Flavie Ader
- APHP, UF Cardiogénétique et Myogénétique, Service de Biochimie Métabolique, Hôpitaux Universitaires de la Pitié- Salpêtrière- Charles Foix, 47-83 Bd de l’Hôpital, Paris, France
- Sorbonne Universités, UPMC Univ. Paris 06, INSERM, UMR_S 1166 and ICAN Institute for Cardiometabolism and Nutrition, Paris, France
| | - Jose M. Larrañaga-Moreira
- Unidad de Cardiopatías Familiares, Instituto de Investigación Biomédica de A Coruña (INIBIC), A Coruña, Spain
- Complexo Hospitalario Universitario de A Coruña, Servizo Galego de Saúde (SERGAS), A Coruña, Spain
- Department of Cardiology, Universidade da Coruña, A Coruña, Spain
- Centro de Investigación Biomédica en Red (CIBERCV), Madrid, Spain
| | - Maria Sabater-Molina
- Inherited Cardiac Disease Unit, Hospital Universitario Virgen Arrixaca, Murcia, Spain
- Universidad de Murcia, Murcia, Spain
| | - Maria I. Garcia-Alvarez
- Cardiology Department, University General Hospital of Alicante, Alicante, Spain
- Institute of Health and Biomedical Research (ISABIAL), Alicante, Spain
| | | | - Grazyna Truszkowska
- Molecular Biology Laboratory, Department of Medical Biology, The Cardinal Stefan Wyszynski Institute of Cardiology, Warsaw, Poland
| | | | - Itziar Solla Ruiz
- Cardiology Specialist in Heart Failure and Inherited Cardiac Diseases, Department of Cardiology, Hospital Universitario Donostia, Spain
| | | | | | - Ainhoa Robles Mezcua
- Heart Failure and Familial Heart Diseases Unit, Cardiology Department, Hospital Universitario Virgen de la Victoria, CIBER-CV, IBIMA, Malaga, Spain
| | - Jorge Alvarez-Rubio
- Inherited Cardiovascular Diseases Unit, Son Llatzer University Hospital & IdISBa, Palma de Mallorca, Spain
| | - Hans Eiskjaer
- Department of Cardiology, Aarhus University Hospital, Hjertesygdomme, Aarhus, Denmark
| | - Mathias Gautel
- Randall Institute, King’s College London, London, United Kingdom
| | - José M. Garcia-Pinilla
- Heart Failure and Familial Heart Diseases Unit, Cardiology Department, Hospital Universitario Virgen de la Victoria, CIBER-CV, IBIMA, Malaga, Spain
| | - Tomas Ripoll-Vera
- Inherited Cardiovascular Diseases Unit, Son Llatzer University Hospital & IdISBa, Palma de Mallorca, Spain
| | - Jens Mogensen
- Department of Cardiology, Odense University Hospital, Odense, Denmark
| | - Javier Limeres Freire
- Department of Cardiology, Vall d’ Hebron Institut de Recerca (VHIR), Hospital Universitari Vall d’ Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Jose F. Rodríguez-Palomares
- Department of Cardiology, Vall d’ Hebron Institut de Recerca (VHIR), Hospital Universitari Vall d’ Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Maria Luisa Peña-Peña
- Heart Failure and Heart Transplantation Unit, Virgen del Rocio University Hospital, Sevilla, Spain
| | - Diego Rangel-Sousa
- Heart Failure and Heart Transplantation Unit, Virgen del Rocio University Hospital, Sevilla, Spain
| | - Julian Palomino-Doza
- Hereditary Cardiopathies Unit, Hospital Universitario 12 de Octubre, Madrid, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- Instituto de Investigación 12 de Octubre i+12, Madrid, Spain
| | - Xabier Arana Achaga
- Cardiology Specialist in Heart Failure and Inherited Cardiac Diseases, Department of Cardiology, Hospital Universitario Donostia, Spain
| | - Zofia Bilinska
- Unit for Screening Studies in Inherited Cardiovascular Diseases, The Cardinal Stefan Wyszynski Institute of Cardiology, Warsaw, Poland
| | | | - Vincent Climent
- Cardiology Department, University General Hospital of Alicante, Alicante, Spain
- Institute of Health and Biomedical Research (ISABIAL), Alicante, Spain
| | | | - Roberto Barriales-Villa
- Unidad de Cardiopatías Familiares, Instituto de Investigación Biomédica de A Coruña (INIBIC), A Coruña, Spain
- Complexo Hospitalario Universitario de A Coruña, Servizo Galego de Saúde (SERGAS), A Coruña, Spain
- Department of Cardiology, Universidade da Coruña, A Coruña, Spain
- Centro de Investigación Biomédica en Red (CIBERCV), Madrid, Spain
| | - Philippe Charron
- Sorbonne Universités, UPMC Univ. Paris 06, INSERM, UMR_S 1166 and ICAN Institute for Cardiometabolism and Nutrition, Paris, France
- APHP, Centre de Référence pour les Maladies Cardiaques Héréditaires, Département de Génétique, Hôpital Pitié-Salpêtrière, Paris, France
| | - Raquel Yotti
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- Hospital General Universitario Gregorio Marañon, Madrid, Spain
- Instituto de Investigación Sanitarias Gregorio Marañón, Spain
| | - Esther Zorio
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- Cardiology Department at Hospital Universitario y Politécnico La Fe and Research Group on Inherited Heart Diseases, Sudden Death and Mechanisms of Disease (CaFaMuSMe) from the Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| | - Juan Jiménez-Jáimez
- Cardiology Department, Hospital Universitario Virgen de las Nieves, Granada, Spain
| | - Pablo Garcia-Pavia
- Department of Cardiology, Universidad Francisco de Vitoria, Pozuelo de Alarcón, Spain
- Heart Failure and Inherited Cardiac Diseases Unit, Department of Cardiology, Hospital Universitario Puerta de Hierro, Madrid, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart (ERN GUARDHEART)
| | - Perry M. Elliott
- Department of Inherited Cardiovascular Diseases, Bart’s Heart Centre St Bartholomew’s Hospital, London, United Kingdom
- Institute of Cardiovascular Science, University College London, London, United Kingdom
| |
Collapse
|
17
|
Giri P, Mukhopadhyay A, Gupta M, Mohapatra B. Dilated cardiomyopathy: a new insight into the rare but common cause of heart failure. Heart Fail Rev 2021; 27:431-454. [PMID: 34245424 DOI: 10.1007/s10741-021-10125-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/25/2021] [Indexed: 12/26/2022]
Abstract
Heart failure is a global health burden responsible for high morbidity and mortality with a prevalence of greater than 60 million individuals worldwide. One of the major causes of heart failure is dilated cardiomyopathy (DCM), characterized by associated systolic dysfunction. During the last few decades, there have been remarkable advances in our understanding about the genetics of dilated cardiomyopathy. The genetic causes were initially thought to be associated with mutations in genes encoding proteins that are localized to cytoskeleton and sarcomere only; however, with the advancement in mechanistic understanding, the roles of ion channels, Z-disc, mitochondria, nuclear proteins, cardiac transcription factors (e.g., NKX-2.5, TBX20, GATA4), and the factors involved in calcium homeostasis have also been identified and found to be implicated in both familial and sporadic DCM cases. During past few years, next-generation sequencing (NGS) has been established as a diagnostic tool for genetic analysis and it has added significantly to the existing candidate gene list for DCM. The animal models have also provided novel insights to develop a better treatment strategy based on phenotype-genotype correlation, epigenetic and phenomic profiling. Most of the DCM biomarkers that are used in routine genetic and clinical testing are structural proteins, but during the last few years, the role of mi-RNA has also emerged as a biomarker due to their accessibility through noninvasive methods. Our increasing genetic knowledge can improve the clinical management of DCM by bringing clinicians and geneticists on one platform, thereby influencing the individualized clinical decision making and leading to precision medicine.
Collapse
Affiliation(s)
- Prerna Giri
- Cytogenetics Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Uttar Pradesh, Varanasi-5, India
| | - Amrita Mukhopadhyay
- Cytogenetics Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Uttar Pradesh, Varanasi-5, India
| | - Mohini Gupta
- Cytogenetics Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Uttar Pradesh, Varanasi-5, India
| | - Bhagyalaxmi Mohapatra
- Cytogenetics Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Uttar Pradesh, Varanasi-5, India.
| |
Collapse
|
18
|
Rohde S, Muslem R, Kaya E, Dalinghaus M, van Waning JI, Majoor-Krakauer D, Towbin J, Caliskan K. State-of-the art review: Noncompaction cardiomyopathy in pediatric patients. Heart Fail Rev 2021; 27:15-28. [PMID: 33715140 PMCID: PMC8739285 DOI: 10.1007/s10741-021-10089-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/15/2021] [Indexed: 12/13/2022]
Abstract
Noncompaction cardiomyopathy (NCCM) is a disease characterized by hypertrabeculation, commonly hypothesized due to an arrest in compaction during fetal development. In 2006, NCCM was classified as a distinct form of cardiomyopathy (CMP) by the American Heart Association. NCCM in childhood is more frequently familial than when diagnosed in adulthood and is associated with other congenital heart diseases (CHDs), other genetic CMPs, and neuromuscular diseases (NMDs). It is yet a rare cardiac diseased with an estimated incidence of 0.12 per 100.000 in children up to 10 years of age. Diagnosing NCCM can be challenging due to non-uniform diagnostic criteria, unawareness, presumed other CMPs, and presence of CHD. Therefore, the incidence of NCCM in children might be an underestimation. Nonetheless, NCCM is the third most common cardiomyopathy in childhood and is associated with heart failure, arrhythmias, and/or thromboembolic events. This state-of-the-art review provides an overview on pediatric NCCM. In addition, we discuss the natural history, epidemiology, genetics, clinical presentation, outcome, and therapeutic options of NCCM in pediatric patients, including fetuses, neonates, infants, and children. Furthermore, we provide a simple classification of different forms of the disease. Finally, the differences between the pediatric population and the adult population are described.
Collapse
Affiliation(s)
- Sofie Rohde
- Thoraxcenter, Department of Cardiology, Erasmus University Medical Center, Room RG 431, 3015 GD, Rotterdam, The Netherlands
| | - Rahatullah Muslem
- Thoraxcenter, Department of Cardiology, Erasmus University Medical Center, Room RG 431, 3015 GD, Rotterdam, The Netherlands
| | - Emrah Kaya
- Thoraxcenter, Department of Cardiology, Erasmus University Medical Center, Room RG 431, 3015 GD, Rotterdam, The Netherlands
| | - Michel Dalinghaus
- Division of Pediatric Cardiology, Sophia Children's Hospital, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Jaap I van Waning
- Department of Clinical Genetics, Erasmus University Medical Center, Rotterdam, The Netherlands
| | | | - Jeffery Towbin
- The Heart Institute, Le Bonheur Children's Hospital, Memphis, TN, USA
| | - Kadir Caliskan
- Thoraxcenter, Department of Cardiology, Erasmus University Medical Center, Room RG 431, 3015 GD, Rotterdam, The Netherlands.
| |
Collapse
|
19
|
Abstract
Barth syndrome (BTHS) is a rare, X-linked recessive, infantile-onset debilitating disorder characterized by early-onset cardiomyopathy, skeletal muscle myopathy, growth delay, and neutropenia, with a worldwide incidence of 1/300,000-400,000 live births. The high mortality rate throughout infancy in BTHS patients is related primarily to progressive cardiomyopathy and a weakened immune system. BTHS is caused by defects in the TAZ gene that encodes tafazzin, a transacylase responsible for the remodeling and maturation of the mitochondrial phospholipid cardiolipin (CL), which is critical to normal mitochondrial structure and function (i.e., ATP generation). A deficiency in tafazzin results in up to a 95% reduction in levels of structurally mature CL. Because the heart is the most metabolically active organ in the body, with the highest mitochondrial content of any tissue, mitochondrial dysfunction plays a key role in the development of heart failure in patients with BTHS. Changes in mitochondrial oxidative phosphorylation reduce the ability of mitochondria to meet the ATP demands of the human heart as well as skeletal muscle, namely ATP synthesis does not match the rate of ATP consumption. The presence of several cardiomyopathic phenotypes have been described in BTHS, including dilated cardiomyopathy, left ventricular noncompaction, either alone or in conjunction with other cardiomyopathic phenotypes, endocardial fibroelastosis, hypertrophic cardiomyopathy, and an apical form of hypertrophic cardiomyopathy, among others, all of which can be directly attributed to the lack of CL synthesis, remodeling, and maturation with subsequent mitochondrial dysfunction. Several mechanisms by which these cardiomyopathic phenotypes exist have been proposed, thereby identifying potential targets for treatment. Dysfunction of the sarcoplasmic reticulum Ca2+-ATPase pump and inflammation potentially triggered by circulating mitochondrial components have been identified. Currently, treatment modalities are aimed at addressing symptomatology of HF in BTHS, but do not address the underlying pathology. One novel therapeutic approach includes elamipretide, which crosses the mitochondrial outer membrane to localize to the inner membrane where it associates with cardiolipin to enhance ATP synthesis in several organs, including the heart. Encouraging clinical results of the use of elamipretide in treating patients with BTHS support the potential use of this drug for management of this rare disease.
Collapse
Affiliation(s)
- Hani N Sabbah
- Department of Medicine, Division of Cardiovascular Medicine, Henry Ford Hospital, Henry Ford Health System, 2799 West Grand Boulevard, Detroit, MI, 48202, USA.
| |
Collapse
|
20
|
Ovics P, Regev D, Baskin P, Davidor M, Shemer Y, Neeman S, Ben-Haim Y, Binah O. Drug Development and the Use of Induced Pluripotent Stem Cell-Derived Cardiomyocytes for Disease Modeling and Drug Toxicity Screening. Int J Mol Sci 2020; 21:E7320. [PMID: 33023024 PMCID: PMC7582587 DOI: 10.3390/ijms21197320] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 09/23/2020] [Accepted: 09/27/2020] [Indexed: 12/19/2022] Open
Abstract
: Over the years, numerous groups have employed human induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) as a superb human-compatible model for investigating the function and dysfunction of cardiomyocytes, drug screening and toxicity, disease modeling and for the development of novel drugs for heart diseases. In this review, we discuss the broad use of iPSC-CMs for drug development and disease modeling, in two related themes. In the first theme-drug development, adverse drug reactions, mechanisms of cardiotoxicity and the need for efficient drug screening protocols-we discuss the critical need to screen old and new drugs, the process of drug development, marketing and Adverse Drug reactions (ADRs), drug-induced cardiotoxicity, safety screening during drug development, drug development and patient-specific effect and different mechanisms of ADRs. In the second theme-using iPSC-CMs for disease modeling and developing novel drugs for heart diseases-we discuss the rationale for using iPSC-CMs and modeling acquired and inherited heart diseases with iPSC-CMs.
Collapse
Affiliation(s)
- Paz Ovics
- Department of Physiology, Biophysics and Systems Biology, The Rappaport Institute, Ruth & Bruce Rappaport Faculty of Medicine, Technion, Haifa 31096, Israel; (P.O.); (D.R.); (P.B.); (M.D.); (Y.S.); (S.N.)
| | - Danielle Regev
- Department of Physiology, Biophysics and Systems Biology, The Rappaport Institute, Ruth & Bruce Rappaport Faculty of Medicine, Technion, Haifa 31096, Israel; (P.O.); (D.R.); (P.B.); (M.D.); (Y.S.); (S.N.)
| | - Polina Baskin
- Department of Physiology, Biophysics and Systems Biology, The Rappaport Institute, Ruth & Bruce Rappaport Faculty of Medicine, Technion, Haifa 31096, Israel; (P.O.); (D.R.); (P.B.); (M.D.); (Y.S.); (S.N.)
| | - Mor Davidor
- Department of Physiology, Biophysics and Systems Biology, The Rappaport Institute, Ruth & Bruce Rappaport Faculty of Medicine, Technion, Haifa 31096, Israel; (P.O.); (D.R.); (P.B.); (M.D.); (Y.S.); (S.N.)
| | - Yuval Shemer
- Department of Physiology, Biophysics and Systems Biology, The Rappaport Institute, Ruth & Bruce Rappaport Faculty of Medicine, Technion, Haifa 31096, Israel; (P.O.); (D.R.); (P.B.); (M.D.); (Y.S.); (S.N.)
| | - Shunit Neeman
- Department of Physiology, Biophysics and Systems Biology, The Rappaport Institute, Ruth & Bruce Rappaport Faculty of Medicine, Technion, Haifa 31096, Israel; (P.O.); (D.R.); (P.B.); (M.D.); (Y.S.); (S.N.)
| | - Yael Ben-Haim
- Institute of Molecular and Clinical Sciences, St. George’s University of London, London SW17 0RE, UK;
- Cardiology Clinical Academic Group, St. George’s University Hospitals NHS Foundation Trust, London SW17 0QT, UK
| | - Ofer Binah
- Department of Physiology, Biophysics and Systems Biology, The Rappaport Institute, Ruth & Bruce Rappaport Faculty of Medicine, Technion, Haifa 31096, Israel; (P.O.); (D.R.); (P.B.); (M.D.); (Y.S.); (S.N.)
| |
Collapse
|
21
|
Akhtar MM, Lorenzini M, Cicerchia M, Ochoa JP, Hey TM, Sabater Molina M, Restrepo-Cordoba MA, Dal Ferro M, Stolfo D, Johnson R, Larrañaga-Moreira JM, Robles-Mezcua A, Rodriguez-Palomares JF, Casas G, Peña-Peña ML, Lopes LR, Gallego-Delgado M, Franaszczyk M, Laucey G, Rangel-Sousa D, Basurte M, Palomino-Doza J, Villacorta E, Bilinska Z, Limeres Freire J, Garcia Pinilla JM, Barriales-Villa R, Fatkin D, Sinagra G, Garcia-Pavia P, Gimeno JR, Mogensen J, Monserrat L, Elliott PM. Clinical Phenotypes and Prognosis of Dilated Cardiomyopathy Caused by Truncating Variants in the TTN Gene. Circ Heart Fail 2020; 13:e006832. [PMID: 32964742 DOI: 10.1161/circheartfailure.119.006832] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Truncating variants in the TTN gene (TTNtv) are the commonest cause of heritable dilated cardiomyopathy. This study aimed to study the phenotypes and outcomes of TTNtv carriers. METHODS Five hundred thirty-seven individuals (61% men; 317 probands) with TTNtv were recruited in 14 centers (372 [69%] with baseline left ventricular systolic dysfunction [LVSD]). Baseline and longitudinal clinical data were obtained. The primary end point was a composite of malignant ventricular arrhythmia and end-stage heart failure. The secondary end point was left ventricular reverse remodeling (left ventricular ejection fraction increase by ≥10% or normalization to ≥50%). RESULTS Median follow-up was 49 (18-105) months. Men developed LVSD more frequently and earlier than women (45±14 versus 49±16 years, respectively; P=0.04). By final evaluation, 31%, 45%, and 56% had atrial fibrillation, frequent ventricular ectopy, and nonsustained ventricular tachycardia, respectively. Seventy-six (14.2%) individuals reached the primary end point (52 [68%] end-stage heart failure events, 24 [32%] malignant ventricular arrhythmia events). Malignant ventricular arrhythmia end points most commonly occurred in patients with severe LVSD. Male sex (hazard ratio, 1.89 [95% CI, 1.04-3.44]; P=0.04) and left ventricular ejection fraction (per 10% decrement from left ventricular ejection fraction, 50%; hazard ratio, 1.63 [95% CI, 1.30-2.04]; P<0.001) were independent predictors of the primary end point. Two hundred seven of 300 (69%) patients with LVSD had evidence of left ventricular reverse remodeling. In a subgroup of 29 of 74 (39%) patients with initial left ventricular reverse remodeling, there was a subsequent left ventricular ejection fraction decrement. TTNtv location was not associated with statistically significant differences in baseline clinical characteristics, left ventricular reverse remodeling, or outcomes on multivariable analysis (P=0.07). CONCLUSIONS TTNtv is characterized by frequent arrhythmia, but malignant ventricular arrhythmias are most commonly associated with severe LVSD. Male sex and LVSD are independent predictors of outcomes. Mutation location does not impact clinical phenotype or outcomes.
Collapse
Affiliation(s)
- Mohammed Majid Akhtar
- Department of Inherited Cardiovascular Diseases, Bart's Heart Centre, St. Bartholomew's Hospital, London, United Kingdom (M.M.A., M.L., L.R.L., P.M.E.).,Institute of Cardiovascular Science, University College London, United Kingdom (M.M.A., M.L., L.R.L., P.M.E.)
| | - Massimiliano Lorenzini
- Department of Inherited Cardiovascular Diseases, Bart's Heart Centre, St. Bartholomew's Hospital, London, United Kingdom (M.M.A., M.L., L.R.L., P.M.E.).,Institute of Cardiovascular Science, University College London, United Kingdom (M.M.A., M.L., L.R.L., P.M.E.)
| | - Marcos Cicerchia
- Health in Code S.L. Scientific Department, A Coruña, Spain (M.C., J.P.O., L.M.).,Instituto de Investigación Biomédica de A Coruña, Complexo Hospitalario Universitario de A Coruña, Sergas, Universidade da Coruña, Spain (M.C., J.P.O., L.M.)
| | - Juan Pablo Ochoa
- Health in Code S.L. Scientific Department, A Coruña, Spain (M.C., J.P.O., L.M.).,Instituto de Investigación Biomédica de A Coruña, Complexo Hospitalario Universitario de A Coruña, Sergas, Universidade da Coruña, Spain (M.C., J.P.O., L.M.)
| | - Thomas Morris Hey
- Department of Cardiology, Odense University Hospital, Denmark (T.M.H., J.M.).,Odense Patient Data Explorative Network, University of Southern Denmark (T.M.H., J.M.)
| | - Maria Sabater Molina
- Inherited Cardiac Disease Unit, Hospital Universitario Virgen Arrixaca, Murcia, Spain (M.S.M., J.R.G.)
| | - Maria Alejandra Restrepo-Cordoba
- Heart Failure and Inherited Cardiac Diseases Unit, Department of Cardiology, Hospital Universitario Puerta de Hierro, Centro de Investigación en Red en Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain (M.A.R.-C., P.G.-P.).,Universidad Francisco de Vitoria, Pozuelo de Alarcon, Spain (M.A.R.-C., P.G.-P.)
| | - Matteo Dal Ferro
- Cardiovascular Department, Azienda Sanitaria Universitaria Integrata of Trieste, Trieste Hospital, Italy (M.D.F., D.S., G.S.)
| | - Davide Stolfo
- Cardiovascular Department, Azienda Sanitaria Universitaria Integrata of Trieste, Trieste Hospital, Italy (M.D.F., D.S., G.S.)
| | - Renee Johnson
- Molecular Cardiology and Biophysics Division (R.J.), Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
| | - José M Larrañaga-Moreira
- Unidad de Cardiopatías Familiares/Cardiology Service, CIBERCV, Instituto de Investigación Biomédica de A Coruña, Complexo Hospitalario Universitario de A Coruña, Sergas, Universidade da Coruña, Spain (J.M.L.-M., R.B.-V.)
| | - Ainhoa Robles-Mezcua
- Heart Failure and Familial Heart Diseases Unit, Cardiology Department, Hospital Universitario Virgen de la Victoria, CIBERCV, IBIMA, Malaga, Spain (A.R.-M., J.M.G.P.)
| | - Jose F Rodriguez-Palomares
- Department of Cardiology, Vall d' Hebron Institut de Recerca, Hospital Universitari Vall d' Hebron, Universitat Autònoma de Barcelona, Spain (J.F.R.-P., G.C., J.L.F.)
| | - Guillem Casas
- Department of Cardiology, Vall d' Hebron Institut de Recerca, Hospital Universitari Vall d' Hebron, Universitat Autònoma de Barcelona, Spain (J.F.R.-P., G.C., J.L.F.)
| | - Maria Luisa Peña-Peña
- Heart Failure and Heart Transplantation Unit, Virgen del Rocio University Hospital, Sevilla, Spain (M.L.P.-P., D.R.-S.)
| | - Luis Rocha Lopes
- Department of Inherited Cardiovascular Diseases, Bart's Heart Centre, St. Bartholomew's Hospital, London, United Kingdom (M.M.A., M.L., L.R.L., P.M.E.).,Institute of Cardiovascular Science, University College London, United Kingdom (M.M.A., M.L., L.R.L., P.M.E.)
| | - Maria Gallego-Delgado
- Inherited Cardiovascular Disease Unit, Cardiology Department, Instituto de Investigación Biomédica de Salamanca, Complejo Asistencial Universitario de Salamanca, Spain (M.G.-D., E.V.)
| | - Maria Franaszczyk
- Department of Medical Biology (M.F.), Cardinal Stefan Wyszynski Institute of Cardiology, Warsaw, Poland
| | - Gemma Laucey
- Complejo Hospitalario de Navarra, Pamplona, Spain (G.L., M.B.)
| | - Diego Rangel-Sousa
- Heart Failure and Heart Transplantation Unit, Virgen del Rocio University Hospital, Sevilla, Spain (M.L.P.-P., D.R.-S.)
| | - Mayte Basurte
- Complejo Hospitalario de Navarra, Pamplona, Spain (G.L., M.B.)
| | - Julian Palomino-Doza
- Inherited Cardiac Disease Unit, Instituto de investigación I+12, Hospital Universitario 12 de Octubre, Madrid, Spain (J.P.-D.).,Centro de Investigación Biomedica en Red en Enfermedades Cardiovasculares, CIBERCV, Madrid, Spain (J.P.-D.)
| | - Eduardo Villacorta
- Inherited Cardiovascular Disease Unit, Cardiology Department, Instituto de Investigación Biomédica de Salamanca, Complejo Asistencial Universitario de Salamanca, Spain (M.G.-D., E.V.)
| | - Zofia Bilinska
- Unit for Screening Studies in Inherited Cardiovascular Diseases (Z.B.), Cardinal Stefan Wyszynski Institute of Cardiology, Warsaw, Poland
| | - Javier Limeres Freire
- Department of Cardiology, Vall d' Hebron Institut de Recerca, Hospital Universitari Vall d' Hebron, Universitat Autònoma de Barcelona, Spain (J.F.R.-P., G.C., J.L.F.)
| | - José M Garcia Pinilla
- Heart Failure and Familial Heart Diseases Unit, Cardiology Department, Hospital Universitario Virgen de la Victoria, CIBERCV, IBIMA, Malaga, Spain (A.R.-M., J.M.G.P.)
| | - Roberto Barriales-Villa
- Unidad de Cardiopatías Familiares/Cardiology Service, CIBERCV, Instituto de Investigación Biomédica de A Coruña, Complexo Hospitalario Universitario de A Coruña, Sergas, Universidade da Coruña, Spain (J.M.L.-M., R.B.-V.)
| | - Diane Fatkin
- Molecular Cardiology and Biophysics Division (D.F.), Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia.,St. Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Kensington, NSW, Australia (D.F.).,Cardiology Department, St. Vincent's Hospital, Darlinghurst, NSW, Australia (D.F.)
| | - Gianfranco Sinagra
- Cardiovascular Department, Azienda Sanitaria Universitaria Integrata of Trieste, Trieste Hospital, Italy (M.D.F., D.S., G.S.)
| | - Pablo Garcia-Pavia
- Heart Failure and Inherited Cardiac Diseases Unit, Department of Cardiology, Hospital Universitario Puerta de Hierro, Centro de Investigación en Red en Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain (M.A.R.-C., P.G.-P.).,Universidad Francisco de Vitoria, Pozuelo de Alarcon, Spain (M.A.R.-C., P.G.-P.)
| | - Juan R Gimeno
- Inherited Cardiac Disease Unit, Hospital Universitario Virgen Arrixaca, Murcia, Spain (M.S.M., J.R.G.)
| | - Jens Mogensen
- Department of Cardiology, Odense University Hospital, Denmark (T.M.H., J.M.).,Odense Patient Data Explorative Network, University of Southern Denmark (T.M.H., J.M.)
| | - Lorenzo Monserrat
- Health in Code S.L. Scientific Department, A Coruña, Spain (M.C., J.P.O., L.M.).,Instituto de Investigación Biomédica de A Coruña, Complexo Hospitalario Universitario de A Coruña, Sergas, Universidade da Coruña, Spain (M.C., J.P.O., L.M.)
| | - Perry M Elliott
- Department of Inherited Cardiovascular Diseases, Bart's Heart Centre, St. Bartholomew's Hospital, London, United Kingdom (M.M.A., M.L., L.R.L., P.M.E.).,Institute of Cardiovascular Science, University College London, United Kingdom (M.M.A., M.L., L.R.L., P.M.E.)
| |
Collapse
|
22
|
Gi WT, Haas J, Sedaghat-Hamedani F, Kayvanpour E, Tappu R, Lehmann DH, Shirvani Samani O, Wisdom M, Keller A, Katus HA, Meder B. Epigenetic Regulation of Alternative mRNA Splicing in Dilated Cardiomyopathy. J Clin Med 2020; 9:jcm9051499. [PMID: 32429430 PMCID: PMC7291244 DOI: 10.3390/jcm9051499] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 05/04/2020] [Accepted: 05/12/2020] [Indexed: 12/19/2022] Open
Abstract
In recent years, the genetic architecture of dilated cardiomyopathy (DCM) has been more thoroughly elucidated. However, there is still insufficient knowledge on the modifiers and regulatory principles that lead to the failure of myocardial function. The current study investigates the association of epigenome-wide DNA methylation and alternative splicing, both of which are important regulatory principles in DCM. We analyzed screening and replication cohorts of cases and controls and identified distinct transcriptomic patterns in the myocardium that differ significantly, and we identified a strong association of intronic DNA methylation and flanking exons usage (p < 2 × 10-16). By combining differential exon usage (DEU) and differential methylation regions (DMR), we found a significant change of regulation in important sarcomeric and other DCM-associated pathways. Interestingly, inverse regulation of Titin antisense non-coding RNA transcript splicing and DNA methylation of a locus reciprocal to TTN substantiate these findings and indicate an additional role for non-protein-coding transcripts. In summary, this study highlights for the first time the close interrelationship between genetic imprinting by DNA methylation and the transport of this epigenetic information towards the dynamic mRNA splicing landscape. This expands our knowledge of the genome-environment interaction in DCM besides simple gene expression regulation.
Collapse
Affiliation(s)
- Weng-Tein Gi
- Institute for Cardiomyopathies Heidelberg (ICH), Heart Center Heidelberg, University of Heidelberg, 69121 Heidelberg, Germany; (W.-T.G.); (J.H.); (F.S.-H.); (E.K.); (R.T.); (D.H.L.); (O.S.S.); (M.W.); (H.A.K.)
- DZHK (German Center for Cardiovascular Research), 69121 Heidelberg, Germany
- Department of Medicine III, University of Heidelberg, INF 410, 69120 Heidelberg, Germany
| | - Jan Haas
- Institute for Cardiomyopathies Heidelberg (ICH), Heart Center Heidelberg, University of Heidelberg, 69121 Heidelberg, Germany; (W.-T.G.); (J.H.); (F.S.-H.); (E.K.); (R.T.); (D.H.L.); (O.S.S.); (M.W.); (H.A.K.)
- DZHK (German Center for Cardiovascular Research), 69121 Heidelberg, Germany
- Department of Medicine III, University of Heidelberg, INF 410, 69120 Heidelberg, Germany
| | - Farbod Sedaghat-Hamedani
- Institute for Cardiomyopathies Heidelberg (ICH), Heart Center Heidelberg, University of Heidelberg, 69121 Heidelberg, Germany; (W.-T.G.); (J.H.); (F.S.-H.); (E.K.); (R.T.); (D.H.L.); (O.S.S.); (M.W.); (H.A.K.)
- DZHK (German Center for Cardiovascular Research), 69121 Heidelberg, Germany
- Department of Medicine III, University of Heidelberg, INF 410, 69120 Heidelberg, Germany
| | - Elham Kayvanpour
- Institute for Cardiomyopathies Heidelberg (ICH), Heart Center Heidelberg, University of Heidelberg, 69121 Heidelberg, Germany; (W.-T.G.); (J.H.); (F.S.-H.); (E.K.); (R.T.); (D.H.L.); (O.S.S.); (M.W.); (H.A.K.)
- DZHK (German Center for Cardiovascular Research), 69121 Heidelberg, Germany
- Department of Medicine III, University of Heidelberg, INF 410, 69120 Heidelberg, Germany
| | - Rewati Tappu
- Institute for Cardiomyopathies Heidelberg (ICH), Heart Center Heidelberg, University of Heidelberg, 69121 Heidelberg, Germany; (W.-T.G.); (J.H.); (F.S.-H.); (E.K.); (R.T.); (D.H.L.); (O.S.S.); (M.W.); (H.A.K.)
- DZHK (German Center for Cardiovascular Research), 69121 Heidelberg, Germany
- Department of Medicine III, University of Heidelberg, INF 410, 69120 Heidelberg, Germany
| | - David Hermann Lehmann
- Institute for Cardiomyopathies Heidelberg (ICH), Heart Center Heidelberg, University of Heidelberg, 69121 Heidelberg, Germany; (W.-T.G.); (J.H.); (F.S.-H.); (E.K.); (R.T.); (D.H.L.); (O.S.S.); (M.W.); (H.A.K.)
- Department of Medicine III, University of Heidelberg, INF 410, 69120 Heidelberg, Germany
| | - Omid Shirvani Samani
- Institute for Cardiomyopathies Heidelberg (ICH), Heart Center Heidelberg, University of Heidelberg, 69121 Heidelberg, Germany; (W.-T.G.); (J.H.); (F.S.-H.); (E.K.); (R.T.); (D.H.L.); (O.S.S.); (M.W.); (H.A.K.)
- DZHK (German Center for Cardiovascular Research), 69121 Heidelberg, Germany
- Department of Medicine III, University of Heidelberg, INF 410, 69120 Heidelberg, Germany
| | - Michael Wisdom
- Institute for Cardiomyopathies Heidelberg (ICH), Heart Center Heidelberg, University of Heidelberg, 69121 Heidelberg, Germany; (W.-T.G.); (J.H.); (F.S.-H.); (E.K.); (R.T.); (D.H.L.); (O.S.S.); (M.W.); (H.A.K.)
- DZHK (German Center for Cardiovascular Research), 69121 Heidelberg, Germany
- Department of Medicine III, University of Heidelberg, INF 410, 69120 Heidelberg, Germany
| | - Andreas Keller
- Department of Clinical Bioinformatics, Medical Faculty, Saarland University, 66123 Saarbrücken, Germany;
| | - Hugo A. Katus
- Institute for Cardiomyopathies Heidelberg (ICH), Heart Center Heidelberg, University of Heidelberg, 69121 Heidelberg, Germany; (W.-T.G.); (J.H.); (F.S.-H.); (E.K.); (R.T.); (D.H.L.); (O.S.S.); (M.W.); (H.A.K.)
- DZHK (German Center for Cardiovascular Research), 69121 Heidelberg, Germany
- Department of Medicine III, University of Heidelberg, INF 410, 69120 Heidelberg, Germany
| | - Benjamin Meder
- Institute for Cardiomyopathies Heidelberg (ICH), Heart Center Heidelberg, University of Heidelberg, 69121 Heidelberg, Germany; (W.-T.G.); (J.H.); (F.S.-H.); (E.K.); (R.T.); (D.H.L.); (O.S.S.); (M.W.); (H.A.K.)
- DZHK (German Center for Cardiovascular Research), 69121 Heidelberg, Germany
- Department of Medicine III, University of Heidelberg, INF 410, 69120 Heidelberg, Germany
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
- Correspondence:
| |
Collapse
|
23
|
Shaboodien G, Spracklen TF, Kamuli S, Ndibangwi P, Van Niekerk C, Ntusi NAB. Genetics of inherited cardiomyopathies in Africa. Cardiovasc Diagn Ther 2020; 10:262-278. [PMID: 32420109 DOI: 10.21037/cdt.2019.10.03] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In sub-Saharan Africa (SSA), the burden of noncommunicable diseases (NCDs) is rising disproportionately in comparison to the rest of the world, affecting urban, semi-urban and rural dwellers alike. NCDs are predicted to surpass infections like human immunodeficiency virus, tuberculosis and malaria as the leading cause of mortality in SSA over the next decade. Heart failure (HF) is the dominant form of cardiovascular disease (CVD), and a leading cause of NCD in SSA. The main causes of HF in SSA are hypertension, cardiomyopathies, rheumatic heart disease, pericardial disease, and to a lesser extent, coronary heart disease. Of these, the cardiomyopathies deserve greater attention because of the relatively poor understanding of mechanisms of disease, poor outcomes and the disproportionate impact they have on young, economically active individuals. Morphofunctionally, cardiomyopathies are classified as dilated, hypertrophic, restrictive and arrhythmogenic; regardless of classification, at least half of these are inherited forms of CVD. In this review, we summarise all studies that have investigated the incidence of cardiomyopathy across Africa, with a focus on the inherited cardiomyopathies. We also review data on the molecular genetic underpinnings of cardiomyopathy in Africa, where there is a striking lack of studies reporting on the genetics of cardiomyopathy. We highlight the impact that genetic testing, through candidate gene screening, association studies and next generation sequencing technologies such as whole exome sequencing and targeted resequencing has had on the understanding of cardiomyopathy in Africa. Finally, we emphasise the need for future studies to fill large gaps in our knowledge in relation to the genetics of inherited cardiomyopathies in Africa.
Collapse
Affiliation(s)
- Gasnat Shaboodien
- Cardiovascular Genetics Laboratory, Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.,Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Timothy F Spracklen
- Cardiovascular Genetics Laboratory, Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.,Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Stephen Kamuli
- Cardiovascular Genetics Laboratory, Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.,Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Polycarp Ndibangwi
- Cardiovascular Genetics Laboratory, Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.,Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Carla Van Niekerk
- Cardiovascular Genetics Laboratory, Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.,Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Ntobeko A B Ntusi
- Cardiovascular Genetics Laboratory, Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.,Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.,Cape Universities Body Imaging Centre, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
24
|
Zhou T, Wang J, Xu J, Zheng C, Niu Y, Wang C, Xu F, Yuan L, Zhao X, Liang L, Xu P. A Smart Fluorescent Probe for NO Detection and Application in Myocardial Fibrosis Imaging. Anal Chem 2020; 92:5064-5072. [DOI: 10.1021/acs.analchem.9b05435] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
25
|
Wang DF, Lyu JL, Fang J, Chen J, Chen WW, Huang JQ, Xia SD, Jin JM, Dong FH, Cheng HQ, Xu YK, Guo XG. Impact of LDB3 gene polymorphisms on clinical presentation and implantable cardioverter defibrillator (ICD) implantation in Chinese patients with idiopathic dilated cardiomyopathy. J Zhejiang Univ Sci B 2020; 20:766-775. [PMID: 31379146 DOI: 10.1631/jzus.b1900017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
OBJECTIVE Mutations in LIM domain binding 3 (LDB3) gene cause idiopathic dilated cardiomyopathy (IDCM), a structural heart disease with a complicated genetic background. However, the association of polymorphisms in the LDB3 gene with susceptibility to IDCM in Chinese populations remains unexplored as dose the impact on clinical presentation. METHODS We sequenced all exons and the adjacent part of introns of the LDB3 gene in 159 Chinese Han IDCM patients and 247 healthy controls. Then we detected the distribution of polymorphisms in the LDB3 gene in all participants and assessed their associations with risk of IDCM. Additionally, we conducted a stratified genotype-phenotype correlation analysis. RESULTS The A allele of rs4468255 was significantly associated with IDCM (P<0.01). The rs4468255, rs11812601, rs56165849, and rs3740346 were also associated with diastolic blood pressure (DBP) and left ventricular ejection fraction (LVEF) (P<0.05). Notably, a higher frequency of rs4468255 polymorphism was observed in implantable cardioverter defibrillator (ICD) recipients under a recessive model (P<0.01), whereas the significant association disappeared after adjusting for potential confounders. However, in the dominant model, notable correlations could only be observed after adjusting for multi parameters. CONCLUSIONS The rs4468255 was significantly correlated with IDCM of Chinese Han population. A allele of rs4468255 is higher in IDCM patients with ICD implantation, suggesting the influence of genetic background in the generation of this response. In addition, rs11812601, rs56165849, and rs3740346 in LDB3 show association with brain natriuretic peptide, DBP, and LVEF levels in patients with IDCM but did not show any association with IDCM susceptibility.
Collapse
Affiliation(s)
- Dong-Fei Wang
- Department of Cardiology, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Jia-Lan Lyu
- Department of Cardiology, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Juan Fang
- Department of Cardiology, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Jian Chen
- Department of Cardiology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Wan-Wan Chen
- Department of Cardiology, Pujiang Branch of the First Affiliated Hospital, School of Medicine, Zhejiang University, Jinhua 322200, China
| | - Jia-Qi Huang
- Department of Pathology and Pathophysiology, School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Shu-Dong Xia
- Department of Cardiology, the Forth Affiliated Hospital, School of Medicine, Zhejiang University, Yiwu 322000, China
| | - Jian-Mei Jin
- Department of Cardiology, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Fang-Hong Dong
- Department of Cardiology, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Hong-Qiang Cheng
- Department of Pathology and Pathophysiology, School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Ying-Ke Xu
- Key Laboratory of Biomedical Engineering of Ministry of Education, Department of Biomedical Engineering, Zhejiang University, Hangzhou 310027, China
| | - Xiao-Gang Guo
- Department of Cardiology, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| |
Collapse
|
26
|
Zhang XL, Xie J, Lan RF, Kang LN, Wang L, Xu W, Xu B. Genetic Basis and Genotype-Phenotype Correlations in Han Chinese Patients with Idiopathic Dilated Cardiomyopathy. Sci Rep 2020; 10:2226. [PMID: 32041989 PMCID: PMC7010767 DOI: 10.1038/s41598-020-58984-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 01/22/2020] [Indexed: 12/30/2022] Open
Abstract
Dilated cardiomyopathy (DCM) is one of the leading causes of heart failure. A large proportion of genetic cause remains unexplained, especially in idiopathic DCM. We performed target next-generation sequencing of 102 genes which were known causes or candidate genes for cardiomyopathies and channelpathies in 118 prospectively recruited Han Chinese patients with idiopathic DCM. 41 of the 118 patients carried 40 pathogenic or likely pathogenic variants, providing a molecular diagnosis in 34.7% of patients. 32 of these variants were novel. TTN truncating variants were predominant, with a frequency of 31.0%, followed by variants of LMNA (14.3%), RBM20 (4.8%), and NEXN (4.8%). These 4 genes accounted for over half variants identified. No significant difference in clinical characteristics or rates of reaching the composite end point (cardiac transplantation and death from cardiac causes) between pathogenic or likely pathogenic variant carriers and noncarriers (hazard ratio 1.11, 95% CI: 0.41 to 3.00), or between patients with TTN truncating variants or without (hazard ratio 0.49, 95% CI: 0.36 to 6.10). In our prospective study, we first determined the overall genetic profiles and genotype-phenotype correlations in Han Chinese idiopathic DCM patients, which could provide insight for genetic diagnosis of DCM in this population.
Collapse
Affiliation(s)
- Xin-Lin Zhang
- Department of Cardiology, Affiliated Drum Tower Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Jun Xie
- Department of Cardiology, Affiliated Drum Tower Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Rong-Fang Lan
- Department of Cardiology, Affiliated Drum Tower Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Li-Na Kang
- Department of Cardiology, Affiliated Drum Tower Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Lian Wang
- Department of Cardiology, Affiliated Drum Tower Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Wei Xu
- Department of Cardiology, Affiliated Drum Tower Hospital, Nanjing University School of Medicine, Nanjing, China.
| | - Biao Xu
- Department of Cardiology, Affiliated Drum Tower Hospital, Nanjing University School of Medicine, Nanjing, China.
| |
Collapse
|
27
|
|
28
|
Wang L, Yu P, Zhou B, Song J, Li Z, Zhang M, Guo G, Wang Y, Chen X, Han L, Hu S. Single-cell reconstruction of the adult human heart during heart failure and recovery reveals the cellular landscape underlying cardiac function. Nat Cell Biol 2020; 22:108-119. [PMID: 31915373 DOI: 10.1038/s41556-019-0446-7] [Citation(s) in RCA: 255] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 11/28/2019] [Indexed: 01/04/2023]
Abstract
Owing to the prevalence and high mortality rates of cardiac diseases, a more detailed characterization of the human heart is necessary; however, this has been largely impeded by the cellular diversity of cardiac tissue and limited access to samples. Here, we show transcriptome profiling of 21,422 single cells-including cardiomyocytes (CMs) and non-CMs (NCMs)-from normal, failed and partially recovered (left ventricular assist device treatment) adult human hearts. Comparative analysis of atrial and ventricular cells revealed pronounced inter- and intracompartmental CM heterogeneity as well as compartment-specific utilization of NCM cell types as major cell-communication hubs. Systematic analysis of cellular compositions and cell-cell interaction networks showed that CM contractility and metabolism are the most prominent aspects that are correlated with changes in heart function. We also uncovered active engagement of NCMs in regulating the behaviour of CMs, exemplified by ACKR1+-endothelial cells, injection of which preserved cardiac function after injury. Beyond serving as a rich resource, our study provides insights into cell-type-targeted intervention of heart diseases.
Collapse
Affiliation(s)
- Li Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Peng Yu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Bingying Zhou
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiangping Song
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zheng Li
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Mingzhi Zhang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Guangran Guo
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yin Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiao Chen
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Leng Han
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX, USA
| | - Shengshou Hu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
29
|
The Role of Cyclic AMP Signaling in Cardiac Fibrosis. Cells 2019; 9:cells9010069. [PMID: 31888098 PMCID: PMC7016856 DOI: 10.3390/cells9010069] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 12/23/2019] [Accepted: 12/24/2019] [Indexed: 12/18/2022] Open
Abstract
Myocardial stress and injury invariably promote remodeling of the cardiac tissue, which is associated with cardiomyocyte death and development of fibrosis. The fibrotic process is initially triggered by the differentiation of resident cardiac fibroblasts into myofibroblasts. These activated fibroblasts display increased proliferative capacity and secrete large amounts of extracellular matrix. Uncontrolled myofibroblast activation can thus promote heart stiffness, cardiac dysfunction, arrhythmias, and progression to heart failure. Despite the well-established role of myofibroblasts in mediating cardiac disease, our current knowledge on how signaling pathways promoting fibrosis are regulated and coordinated in this cell type is largely incomplete. In this respect, cyclic adenosine monophosphate (cAMP) signaling acts as a major modulator of fibrotic responses activated in fibroblasts of injured or stressed hearts. In particular, accumulating evidence now suggests that upstream cAMP modulators including G protein-coupled receptors, adenylyl cyclases (ACs), and phosphodiesterases (PDEs); downstream cAMP effectors such as protein kinase A (PKA) and the guanine nucleotide exchange factor Epac; and cAMP signaling organizers such as A-kinase anchoring proteins (AKAPs) modulate a variety of fundamental cellular processes involved in myocardial fibrosis including myofibroblast differentiation, proliferation, collagen secretion, and invasiveness. The current review will discuss recent advances highlighting the role of cAMP and AKAP-mediated signaling in regulating pathophysiological responses controlling cardiac fibrosis.
Collapse
|
30
|
Ichida F. Left ventricular noncompaction - Risk stratification and genetic consideration. J Cardiol 2019; 75:1-9. [PMID: 31629663 DOI: 10.1016/j.jjcc.2019.09.011] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 09/18/2019] [Indexed: 11/30/2022]
Abstract
Left ventricular noncompaction (LVNC) is a cardiomyopathy characterized by two layered structures composed of prominent trabecular meshwork and deep intertrabecular recesses. LVNC was thought to be rare; however, heightened awareness has resulted in an increased detection of the morphological features of LVNC in routine clinical practice especially in the adult population. Although LVNC was classified as an independent primary cardiomyopathy of genetic origin by the American Heart Association in 2006, its definition, diagnostic criteria and clinical implications are still being debated. Clinical manifestations are highly variable, even in the same family, ranging from no symptoms to disabling congestive heart failure, life-threatening arrhythmias, systemic thromboemboli, and sudden cardiac death. Among phenotypic subtypes of LVNC, children with isolated LVNC with normal cardiac function had the best outcomes: children with LVNC and dilated cardiomyopathy had the worst outcomes. Myocardial dysfunction or ventricular arrhythmias are predictors of mortality in adults with LVNC. LVNC, like other forms of inherited cardiomyopathy, is genetically heterogeneous and can be inherited as an autosomal dominant or X-linked recessive disorder. It has been linked to mutations in many genes, including ZASP, TAZ/G4.5, and those encoding sarcomeric, Z-disc, cytoskeleton proteins, and mitochondria. Disturbance of the NOTCH signaling pathway has been reported to be part of genetic pathway for LVNC as well. Although there are an increasing number of reports, genotype-phenotype correlations have been challenging and investigations are ongoing. Patients with mutations are more likely to have major adverse cardiovascular events, further, LV systolic dysfunction in mutation carriers makes them at high risk for cardiac events. Treatments focus on improvement in cardiac function and reduction of mechanical stress in patients with systolic dysfunction and on treatment of arrhythmia and implantation of an automatic implantable cardioverter-defibrillator for prevention of sudden death. Given that 20-40% of cases may be familial, family screening is recommended.
Collapse
Affiliation(s)
- Fukiko Ichida
- Department of Pediatrics, International University of Health and Welfare, Sanno Hospital, 8-10-16, Akasaka, Minato-ku, Tokyo 107-0052, Japan.
| |
Collapse
|
31
|
Nakamura Y, Miyagawa S, Yoshida S, Sasawatari S, Toyofuku T, Toda K, Sawa Y. Natural killer cells impede the engraftment of cardiomyocytes derived from induced pluripotent stem cells in syngeneic mouse model. Sci Rep 2019; 9:10840. [PMID: 31346220 PMCID: PMC6658523 DOI: 10.1038/s41598-019-47134-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 07/11/2019] [Indexed: 02/06/2023] Open
Abstract
Transplantation of cardiomyocytes derived from induced pluripotent stem cell (iPSC-CMs) is a promising approach for increasing functional CMs during end-stage heart failure. Although major histocompatibility complex (MHC) class I matching between donor cells and recipient could reduce acquired immune rejection, innate immune responses may have negative effects on transplanted iPSC-CMs. Here, we demonstrated that natural killer cells (NKCs) infiltrated in iPSC-CM transplants even in a syngeneic mouse model. The depletion of NKCs using an anti-NKC antibody rescued transplanted iPSC-CMs, suggesting that iPSC-CMs activated NKC-mediated innate immunity. Surprisingly, iPSC-CMs lost inhibitory MHCs but not activating ligands for NKCs. Re-expression of MHC class I induced by IFN-γ as well as suppression of activating ligands by an antibody rescued the transplanted iPSC-CMs. Thus, NKCs impede the engraftment of transplanted iPSC-CMs because of lost MHC class I, and our results provide a basis for an approach to improve iPSC-CM engraftment.
Collapse
Affiliation(s)
- Yuki Nakamura
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Shigeru Miyagawa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Shohei Yoshida
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Shigemi Sasawatari
- Department of Immunology and Regenerative Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Toshihiko Toyofuku
- Department of Immunology and Regenerative Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Koichi Toda
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Yoshiki Sawa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
32
|
Kagemoto T, Oyama K, Yamane M, Tsukamoto S, Kobirumaki-Shimozawa F, Li A, Dos Remedios C, Fukuda N, Ishiwata S. Sarcomeric Auto-Oscillations in Single Myofibrils From the Heart of Patients With Dilated Cardiomyopathy. Circ Heart Fail 2019; 11:e004333. [PMID: 29980594 DOI: 10.1161/circheartfailure.117.004333] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 05/31/2018] [Indexed: 01/30/2023]
Abstract
BACKGROUND Left ventricular wall motion is depressed in patients with dilated cardiomyopathy (DCM). However, whether or not the depressed left ventricular wall motion is caused by impairment of sarcomere dynamics remains to be fully clarified. METHODS AND RESULTS We analyzed the mechanical properties of single sarcomere dynamics during sarcomeric auto-oscillations (calcium spontaneous oscillatory contractions [Ca-SPOC]) that occurred at partial activation under the isometric condition in myofibrils from donor hearts and from patients with severe DCM (New York Heart Association classification III-IV). Ca-SPOC reproducibly occurred in the presence of 1 μmol/L free Ca2+ in both nonfailing and DCM myofibrils, and sarcomeres exhibited a saw-tooth waveform along single myofibrils composed of quick lengthening and slow shortening. The period of Ca-SPOC was longer in DCM myofibrils than in nonfailing myofibrils, in association with prolonged shortening time. Lengthening time was similar in both groups. Then, we performed Tn (troponin) exchange in myofibrils with a DCM-causing homozygous mutation (K36Q) in cTnI (cardiac TnI). On exchange with the Tn complex from healthy porcine ventricles, period, shortening time, and shortening velocity in cTnI-K36Q myofibrils became similar to those in Tn-reconstituted nonfailing myofibrils. Protein kinase A abbreviated period in both Tn-reconstituted nonfailing and cTnI-K36Q myofibrils, demonstrating acceleration of cross-bridge kinetics. CONCLUSIONS Sarcomere dynamics was found to be depressed under loaded conditions in DCM myofibrils because of impairment of thick-thin filament sliding. Thus, microscopic analysis of Ca-SPOC in human cardiac myofibrils is beneficial to systematically unveil the kinetic properties of single sarcomeres in various types of heart disease.
Collapse
Affiliation(s)
- Tatsuya Kagemoto
- Department of Physics, Faculty of Science and Engineering, Waseda University, Tokyo, Japan (T.K., M.Y., S.I.)
| | - Kotaro Oyama
- Department of Cell Physiology, The Jikei University School of Medicine, Tokyo, Japan (K.O., S.T., F.K.-S., N.F.)
| | - Mitsunori Yamane
- Department of Physics, Faculty of Science and Engineering, Waseda University, Tokyo, Japan (T.K., M.Y., S.I.)
| | - Seiichi Tsukamoto
- Department of Cell Physiology, The Jikei University School of Medicine, Tokyo, Japan (K.O., S.T., F.K.-S., N.F.)
| | - Fuyu Kobirumaki-Shimozawa
- Department of Cell Physiology, The Jikei University School of Medicine, Tokyo, Japan (K.O., S.T., F.K.-S., N.F.)
| | - Amy Li
- School of Medical Sciences, Bosch Institute, The University of Sydney, Australia (A.L., C.D.R.)
| | - Cristobal Dos Remedios
- School of Medical Sciences, Bosch Institute, The University of Sydney, Australia (A.L., C.D.R.)
| | - Norio Fukuda
- Department of Cell Physiology, The Jikei University School of Medicine, Tokyo, Japan (K.O., S.T., F.K.-S., N.F.).
| | - Shin'ichi Ishiwata
- Department of Physics, Faculty of Science and Engineering, Waseda University, Tokyo, Japan (T.K., M.Y., S.I.).
| |
Collapse
|
33
|
Lu D, Wang J, Li J, Guan F, Zhang X, Dong W, Liu N, Gao S, Zhang L. Meox1 accelerates myocardial hypertrophic decompensation through Gata4. Cardiovasc Res 2019; 114:300-311. [PMID: 29155983 DOI: 10.1093/cvr/cvx222] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 11/15/2017] [Indexed: 12/22/2022] Open
Abstract
Aims Pathological hypertrophy is the result of gene network regulation, which ultimately leads to adverse cardiac remodelling and heart failure (HF) and is accompanied by the reactivation of a 'foetal gene programme'. The Mesenchyme homeobox 1 (Meox1) gene is one of the foetal programme genes. Meox1 may play a role in embryonic development, but its regulation of pathological hypertrophy is not known. Therefore, this study investigated the effect of Meox1 on pathological hypertrophy, including familial and pressure overload-induced hypertrophy, and its potential mechanism of action. Methods and results Meox1 expression was markedly down-regulated in the wild-type adult mouse heart with age, and expression was up-regulated in heart tissues from familial dilated cardiomyopathy (FDCM) mice of the cTnTR141W strain, familial hypertrophic cardiomyopathy (FHCM) mice of the cTnTR92Q strain, pressure overload-induced HF mice, and hypertrophic cardiomyopathy (HCM) patients. Echocardiography, histopathology, and hypertrophic molecular markers consistently demonstrated that Meox1 overexpression exacerbated the phenotypes in FHCM and in mice with thoracic aorta constriction (TAC), and that Meox1 knockdown improved the pathological changes. Gata4 was identified as a potential downstream target of Meox1 using digital gene expression (DGE) profiling, real-time PCR, and bioinformatics analysis. Promoter activity data and chromatin immunoprecipitation (ChIP) and Gata4 knockdown analyses indicated that Meox1 acted via activation of Gata4 transcription. Conclusion Meox1 accelerated decompensation via the downstream target Gata4, at least in part directly. Meox1 and other foetal programme genes form a highly interconnected network, which offers multiple therapeutic entry points to dampen the aberrant expression of foetal genes and pathological hypertrophy.
Collapse
Affiliation(s)
- Dan Lu
- Key Laboratory of Human Disease Comparative Medicine, NHFPC, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences & Comparative Medical Center, Peking Union Medical College, Building 5, Panjiayuan Nanli, Chaoyang District, Beijing 100021, China
| | - Jizheng Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishilu, Beijing 100037, China
| | - Jing Li
- Key Laboratory of Human Disease Comparative Medicine, NHFPC, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences & Comparative Medical Center, Peking Union Medical College, Building 5, Panjiayuan Nanli, Chaoyang District, Beijing 100021, China
| | - Feifei Guan
- Key Laboratory of Human Disease Comparative Medicine, NHFPC, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences & Comparative Medical Center, Peking Union Medical College, Building 5, Panjiayuan Nanli, Chaoyang District, Beijing 100021, China
| | - Xu Zhang
- Key Laboratory of Human Disease Comparative Medicine, NHFPC, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences & Comparative Medical Center, Peking Union Medical College, Building 5, Panjiayuan Nanli, Chaoyang District, Beijing 100021, China
| | - Wei Dong
- Key Laboratory of Human Disease Comparative Medicine, NHFPC, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences & Comparative Medical Center, Peking Union Medical College, Building 5, Panjiayuan Nanli, Chaoyang District, Beijing 100021, China
| | - Ning Liu
- Key Laboratory of Human Disease Comparative Medicine, NHFPC, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences & Comparative Medical Center, Peking Union Medical College, Building 5, Panjiayuan Nanli, Chaoyang District, Beijing 100021, China
| | - Shan Gao
- Key Laboratory of Human Disease Comparative Medicine, NHFPC, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences & Comparative Medical Center, Peking Union Medical College, Building 5, Panjiayuan Nanli, Chaoyang District, Beijing 100021, China
| | - Lianfeng Zhang
- Key Laboratory of Human Disease Comparative Medicine, NHFPC, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences & Comparative Medical Center, Peking Union Medical College, Building 5, Panjiayuan Nanli, Chaoyang District, Beijing 100021, China
| |
Collapse
|
34
|
Bauer A, Esmaeili A, deRosa R, Voelkel NF, Schranz D. Restrictive atrial communication in right and left heart failure. Transl Pediatr 2019; 8:133-139. [PMID: 31161080 PMCID: PMC6514282 DOI: 10.21037/tp.2019.04.03] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Heart failure (HF) is usually defined by the dominantly affected heart chamber; therefore, termed right or left HF (RHF or LHF). Pulmonologists understand RHF as a complex syndrome characterized by insufficient delivery of blood from the right ventricle associated with elevated systemic venous pressure at rest or exercise. Cardiologists specify LHF by its clinical functional class and the relation to a reduced (HFrEF), preserved (HFpEF) or mid-range ejection fraction (HFmrEF). Pediatric cardiologist, dealing also with patients with a failing single ventricle, define HF as a condition of insufficient systemic oxygen delivery (DO2). Certainly, pediatricians do not think of the right and left heart, or even a single ventricle as an isolated, independently acting entity. Because of the importance of cardiac interactions, the creation of a restrictive atrial communication aims at a palliative approach with the goal to diminish the congestive consequences of a dysfunctional ventricle; further to serve as a pop-off valve in order to prevent syncope and cardiovascular collapse. This review covers the background, the particular indications, the techniques and preliminary results achieved following the creation of a restrictive atrial septum defect (rASD) in different pathophysiological settings. Based on the institutional experience, percutaneous trans-catheter perforation of the atrial septum, followed by gradual balloon dilatation can be performed at any age and location worldwide. Medical institutions in low resource countries can make use of such palliating procedures in the setting of right as well as LHF independent of their pharmacological facilities.
Collapse
Affiliation(s)
- Anna Bauer
- Pediatric Heart Center, Justus-Liebig University, Giessen, Germany
| | - Anoosh Esmaeili
- Department of Pediatric Cardiology, Johann-Wolfgang Goethe University, Frankfurt, Germany
| | - Roberta deRosa
- Cardiovascular Department, University of Salerno, Salerno, Italy
| | | | - Dietmar Schranz
- Pediatric Heart Center, Justus-Liebig University, Giessen, Germany
| |
Collapse
|
35
|
Akinrinade O, Heliö T, Lekanne Deprez RH, Jongbloed JDH, Boven LG, van den Berg MP, Pinto YM, Alastalo TP, Myllykangas S, Spaendonck-Zwarts KV, van Tintelen JP, van der Zwaag PA, Koskenvuo J. Relevance of Titin Missense and Non-Frameshifting Insertions/Deletions Variants in Dilated Cardiomyopathy. Sci Rep 2019; 9:4093. [PMID: 30858397 PMCID: PMC6412046 DOI: 10.1038/s41598-019-39911-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 02/04/2019] [Indexed: 12/30/2022] Open
Abstract
Recent advancements in next generation sequencing (NGS) technology have led to the identification of the giant sarcomere gene, titin (TTN), as a major human disease gene. Truncating variants of TTN (TTNtv) especially in the A-band region account for 20% of dilated cardiomyopathy (DCM) cases. Much attention has been focused on assessment and interpretation of TTNtv in human disease; however, missense and non-frameshifting insertions/deletions (NFS-INDELs) are difficult to assess and interpret in clinical diagnostic workflow. Targeted sequencing covering all exons of TTN was performed on a cohort of 530 primary DCM patients from three cardiogenetic centres across Europe. Using stringent bioinformatic filtering, twenty-nine and two rare TTN missense and NFS-INDELs variants predicted deleterious were identified in 6.98% and 0.38% of DCM patients, respectively. However, when compared with those identified in the largest available reference population database, no significant enrichment of such variants was identified in DCM patients. Moreover, DCM patients and reference individuals had comparable frequencies of splice-region missense variants with predicted splicing alteration. DCM patients and reference populations had comparable frequencies of rare predicted deleterious TTN missense variants including splice-region missense variants suggesting that these variants are not independently causative for DCM. Hence, these variants should be classified as likely benign in the clinical diagnostic workflow, although a modifier effect cannot be excluded at this stage.
Collapse
Affiliation(s)
- Oyediran Akinrinade
- Children's Hospital, Institute of Clinical Medicine, Helsinki University Central Hospital, University of Helsinki, Helsinki, Finland.
- Institute of Biomedicine, University of Helsinki, Helsinki, Finland.
| | - Tiina Heliö
- Heart and Lung Centre, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Ronald H Lekanne Deprez
- Department of Clinical Genetics, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - Jan D H Jongbloed
- University of Groningen, University Medical Centre Groningen, Department of Genetics, Groningen, The Netherlands
| | - Ludolf G Boven
- University of Groningen, University Medical Centre Groningen, Department of Genetics, Groningen, The Netherlands
| | - Maarten P van den Berg
- Department of Cardiology, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Yigal M Pinto
- Department of Cardiology, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - Tero-Pekka Alastalo
- Children's Hospital, Institute of Clinical Medicine, Helsinki University Central Hospital, University of Helsinki, Helsinki, Finland
- Blueprint Genetics, Helsinki, Finland
| | - Samuel Myllykangas
- Institute of Biomedicine, University of Helsinki, Helsinki, Finland
- Blueprint Genetics, Helsinki, Finland
| | - Karin van Spaendonck-Zwarts
- Department of Clinical Genetics, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - J Peter van Tintelen
- Department of Clinical Genetics, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
- Durrer Centre for Cardiovascular Research, Netherlands Heart Institute, Utrecht, The Netherlands
| | - Paul A van der Zwaag
- University of Groningen, University Medical Centre Groningen, Department of Genetics, Groningen, The Netherlands
| | | |
Collapse
|
36
|
Transcatheter left atrial decompression in patients with dilated cardiomyopathy: bridging to cardiac transplantation or recovery. Cardiol Young 2019; 29:355-362. [PMID: 30806346 DOI: 10.1017/s1047951118002433] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Left atrial congestion results from backward failure in dilated cardiomyopathy. We aimed to evaluate feasibility and efficacy of percutaneous atrioseptostomy to create a restrictive atrial septum defect in management of dilated cardiomyopathy.Methods and resultsFrom June 2009 to December 2016, 27 interventions comprised left atria decompressions in 22 dilated cardiomyopathy patients; 9 females; age: 24 days to 36.9 years; weight: 3-50 kg; NYHA-/Ross class IV (n=16). Mean left ventricular ejection fraction was 21.5±9.7% and brain natriuretic peptide was 2291±1992 pg/ml. Dilated cardiomyopathy was classified as chronic (n=9); acute (n=1) myocarditis; idiopathic (n=5); left ventricular non-compaction (n=4); mitochondriopathy, pacemaker induced, and arrhythmogenic (n=3). Atrioseptostomy was concomitantly performed with myocardial biopsies 6.5 days (±11.7) after admission (n=11). Trans-septal puncture was used in 18 patients; foramen ovale dilatation was done in four patients. Mean balloon size was 11 mm (range 7-14 mm); total procedure time was 133±38 minutes. No procedural complications were observed. Mean left atrial pressure decreased from 15.8±6.8 to 12.2±4.8 mmHg (p=0.005), left/right atrial pressure gradient from 9.6±5.6 to 5±3.5 mmHg; brain natriuretic peptide (n=18) decreased from 1968±1606 to 830±1083 pg/ml (p=0.01). One patient unsuitable for heart transplantation died at home despite additionally performed pulmonary artery banding and three further left atrial decompressions; five patients were bridged to transplantation, two died afterwards. Functional recovery occurred in the remaining 14 patients and in six after additional pulmonary artery banding. No patient required assist device. CONCLUSIONS Percutaneous left atrial decompression is an age-independent, effective palliation treating patients with dilated cardiomyopathy.
Collapse
|
37
|
The Effects of Exercise Training Intensity on the Expression of C/EBPβ and CITED4 in Rats with Myocardial Infarction. Asian J Sports Med 2018. [DOI: 10.5812/asjsm.59300] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
|
38
|
Proteomic signature of circulating extracellular vesicles in dilated cardiomyopathy. J Transl Med 2018; 98:1291-1299. [PMID: 29540862 DOI: 10.1038/s41374-018-0044-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 01/29/2018] [Accepted: 01/30/2018] [Indexed: 11/08/2022] Open
Abstract
Dilated cardiomyopathy (DCM) remains a major cause of heart failure and carries a poor prognosis despite important advances in recent years. Better disease characterization using novel molecular techniques is needed to refine its progression. This study explored the proteomic signature of plasma-derived extracellular vesicles (EVs) obtained from DCM patients and healthy controls using size-exclusion chromatography (SEC). EV-enriched fractions were analyzed by liquid chromatography-mass spectrometry (LC-MS/MS). Raw data obtained from LC-MS/MS were analyzed against the Uniprot human database using MaxQuant software. Additional analyses using Perseus software were based on the Intensity-Based Absolute Quantification (iBAQ) values from MaxQuant analyses. A total of 90.07 ± 21 proteins (227 different proteins) in the DCM group and 96.52 ± 17.91 proteins (183 different proteins) in the control group were identified. A total of 176 proteins (74.6%) were shared by controls and DCM patients, whereas 51 proteins were exclusive for the DCM group and 7 proteins were exclusive for the control group. Fibrinogen (α, β and γ chain), serotransferrin, α-1-antitrypsin, and a variety of apolipoprotein family members (C-I, C-III, D, H or β-2-glycoprotein, and J or clusterin) were clustered in SEC-EVs derived from DCM patients relative to controls (p < 0.05). Regarding Gene Ontology analysis, response to stress and protein activation-related proteins were enriched in DCM-EVs compared with controls. Thus, the present study reports the distinct proteomic signature of circulating DCM-EVs compared with control-EVs. Furthermore, we confirm that SEC obtains highly purified EV fractions from peripheral blood samples for subsequent use in determining disease-specific proteomic signatures.
Collapse
|
39
|
Cassalett-Bustillo G. Falla cardíaca en pacientes pediátricos. Fisiopatología y tratamiento. Parte II. REVISTA COLOMBIANA DE CARDIOLOGÍA 2018. [DOI: 10.1016/j.rccar.2018.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
40
|
Benitez‐Amaro A, Samouillan V, Jorge E, Dandurand J, Nasarre L, de Gonzalo‐Calvo D, Bornachea O, Amoros‐Figueras G, Lacabanne C, Vilades D, Leta R, Carreras F, Gallardo A, Lerma E, Cinca J, Guerra JM, Llorente‐Cortés V. Identification of new biophysical markers for pathological ventricular remodelling in tachycardia-induced dilated cardiomyopathy. J Cell Mol Med 2018; 22:4197-4208. [PMID: 29921039 PMCID: PMC6111813 DOI: 10.1111/jcmm.13699] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 04/17/2018] [Indexed: 11/28/2022] Open
Abstract
Our aim was to identify biophysical biomarkers of ventricular remodelling in tachycardia-induced dilated cardiomyopathy (DCM). Our study includes healthy controls (N = 7) and DCM pigs (N = 10). Molecular analysis showed global myocardial metabolic abnormalities, some of them related to myocardial hibernation in failing hearts, supporting the translationality of our model to study cardiac remodelling in dilated cardiomyopathy. Histological analysis showed unorganized and agglomerated collagen accumulation in the dilated ventricles and a higher percentage of fibrosis in the right (RV) than in the left (LV) ventricle (P = .016). The Fourier Transform Infrared Spectroscopy (FTIR) 1st and 2nd indicators, which are markers of the myofiber/collagen ratio, were reduced in dilated hearts, with the 1st indicator reduced by 45% and 53% in the RV and LV, respectively, and the 2nd indicator reduced by 25% in the RV. The 3rd FTIR indicator, a marker of the carbohydrate/lipid ratio, was up-regulated in the right and left dilated ventricles but to a greater extent in the RV (2.60-fold vs 1.61-fold, P = .049). Differential scanning calorimetry (DSC) showed a depression of the freezable water melting point in DCM ventricles - indicating structural changes in the tissue architecture - and lower protein stability. Our results suggest that the 1st, 2nd and 3rd FTIR indicators are useful markers of cardiac remodelling. Moreover, the 2nd and 3rd FITR indicators, which are altered to a greater extent in the right ventricle, are associated with greater fibrosis.
Collapse
Affiliation(s)
- Aleyda Benitez‐Amaro
- Group of Lipids and Cardiovascular PathologyICCC ProgramBiomedical Research Institute Sant Pau (IIB Sant Pau)Hospital de la Santa Creu i Sant PauBarcelonaSpain
- Institute of Biomedical Research of Barcelona (IIBB)Spanish National Research Council (CSIC)BarcelonaSpain
| | - Valerie Samouillan
- CIRIMATUniversité de ToulouseUniversité Paul Sabatier, Physique des PolymèresToulouseFrance
| | - Esther Jorge
- CIBERCVBarcelonaSpain
- Department of CardiologyHospital de la Santa Creu i Sant PauBiomedical Research Institute Sant Pau (IIB Sant Pau)Universitat Autonoma de BarcelonaBarcelonaSpain
| | - Jany Dandurand
- CIRIMATUniversité de ToulouseUniversité Paul Sabatier, Physique des PolymèresToulouseFrance
| | - Laura Nasarre
- Group of Lipids and Cardiovascular PathologyICCC ProgramBiomedical Research Institute Sant Pau (IIB Sant Pau)Hospital de la Santa Creu i Sant PauBarcelonaSpain
| | - David de Gonzalo‐Calvo
- Group of Lipids and Cardiovascular PathologyICCC ProgramBiomedical Research Institute Sant Pau (IIB Sant Pau)Hospital de la Santa Creu i Sant PauBarcelonaSpain
- Institute of Biomedical Research of Barcelona (IIBB)Spanish National Research Council (CSIC)BarcelonaSpain
- CIBERCVBarcelonaSpain
| | - Olga Bornachea
- Group of Lipids and Cardiovascular PathologyICCC ProgramBiomedical Research Institute Sant Pau (IIB Sant Pau)Hospital de la Santa Creu i Sant PauBarcelonaSpain
- Institute of Biomedical Research of Barcelona (IIBB)Spanish National Research Council (CSIC)BarcelonaSpain
| | - Gerard Amoros‐Figueras
- CIBERCVBarcelonaSpain
- Department of CardiologyHospital de la Santa Creu i Sant PauBiomedical Research Institute Sant Pau (IIB Sant Pau)Universitat Autonoma de BarcelonaBarcelonaSpain
| | - Colette Lacabanne
- CIRIMATUniversité de ToulouseUniversité Paul Sabatier, Physique des PolymèresToulouseFrance
| | - David Vilades
- Department of CardiologyHospital de la Santa Creu i Sant PauBiomedical Research Institute Sant Pau (IIB Sant Pau)Universitat Autonoma de BarcelonaBarcelonaSpain
| | - Ruben Leta
- Department of CardiologyHospital de la Santa Creu i Sant PauBiomedical Research Institute Sant Pau (IIB Sant Pau)Universitat Autonoma de BarcelonaBarcelonaSpain
| | - Francesc Carreras
- CIBERCVBarcelonaSpain
- Department of CardiologyHospital de la Santa Creu i Sant PauBiomedical Research Institute Sant Pau (IIB Sant Pau)Universitat Autonoma de BarcelonaBarcelonaSpain
| | - Alberto Gallardo
- Department of PathologyHospital de la Santa Creu i Sant PauBarcelonaSpain
| | - Enrique Lerma
- Department of PathologyHospital de la Santa Creu i Sant PauBarcelonaSpain
| | - Juan Cinca
- CIBERCVBarcelonaSpain
- Department of CardiologyHospital de la Santa Creu i Sant PauBiomedical Research Institute Sant Pau (IIB Sant Pau)Universitat Autonoma de BarcelonaBarcelonaSpain
| | - Jose M. Guerra
- CIBERCVBarcelonaSpain
- Department of CardiologyHospital de la Santa Creu i Sant PauBiomedical Research Institute Sant Pau (IIB Sant Pau)Universitat Autonoma de BarcelonaBarcelonaSpain
| | - Vicenta Llorente‐Cortés
- Group of Lipids and Cardiovascular PathologyICCC ProgramBiomedical Research Institute Sant Pau (IIB Sant Pau)Hospital de la Santa Creu i Sant PauBarcelonaSpain
- Institute of Biomedical Research of Barcelona (IIBB)Spanish National Research Council (CSIC)BarcelonaSpain
- CIBERCVBarcelonaSpain
| |
Collapse
|
41
|
Translating emerging molecular genetic insights into clinical practice in inherited cardiomyopathies. J Mol Med (Berl) 2018; 96:993-1024. [PMID: 30128729 DOI: 10.1007/s00109-018-1685-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 07/22/2018] [Accepted: 08/08/2018] [Indexed: 12/19/2022]
Abstract
Cardiomyopathies are primarily genetic disorders of the myocardium associated with higher risk of life-threatening cardiac arrhythmias, heart failure, and sudden cardiac death. The evolving knowledge in genomic medicine during the last decade has reshaped our understanding of cardiomyopathies as diseases of multifactorial nature and complex pathophysiology. Genetic testing in cardiomyopathies has subsequently grown from primarily a research tool into an essential clinical evaluation piece with important clinical implications for patients and their families. The purpose of this review is to provide with a contemporary insight into the implications of genetic testing in diagnosis, therapy, and prognosis of patients with inherited cardiomyopathies. Here, we summarize the contemporary knowledge on genotype-phenotype correlations in inherited cardiomyopathies and highlight the recent significant achievements in the field of translational cardiovascular genetics.
Collapse
|
42
|
Vikhorev PG, Vikhoreva NN. Cardiomyopathies and Related Changes in Contractility of Human Heart Muscle. Int J Mol Sci 2018; 19:ijms19082234. [PMID: 30065175 PMCID: PMC6121228 DOI: 10.3390/ijms19082234] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Revised: 07/22/2018] [Accepted: 07/27/2018] [Indexed: 02/07/2023] Open
Abstract
About half of hypertrophic and dilated cardiomyopathies cases have been recognized as genetic diseases with mutations in sarcomeric proteins. The sarcomeric proteins are involved in cardiomyocyte contractility and its regulation, and play a structural role. Mutations in non-sarcomeric proteins may induce changes in cell signaling pathways that modify contractile response of heart muscle. These facts strongly suggest that contractile dysfunction plays a central role in initiation and progression of cardiomyopathies. In fact, abnormalities in contractile mechanics of myofibrils have been discovered. However, it has not been revealed how these mutations increase risk for cardiomyopathy and cause the disease. Much research has been done and still much is being done to understand how the mechanism works. Here, we review the facts of cardiac myofilament contractility in patients with cardiomyopathy and heart failure.
Collapse
Affiliation(s)
- Petr G Vikhorev
- National Heart and Lung Institute, Imperial College London, London W12 0NN, UK.
| | - Natalia N Vikhoreva
- Heart Science Centre, Magdi Yacoub Institute, Harefield Hospital, London UB9 6JH, UK.
| |
Collapse
|
43
|
Leifheit-Nestler M, Grabner A, Hermann L, Richter B, Schmitz K, Fischer DC, Yanucil C, Faul C, Haffner D. Vitamin D treatment attenuates cardiac FGF23/FGFR4 signaling and hypertrophy in uremic rats. Nephrol Dial Transplant 2018; 32:1493-1503. [PMID: 28339837 DOI: 10.1093/ndt/gfw454] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 12/06/2016] [Indexed: 11/13/2022] Open
Abstract
Background Vitamin D deficiency and excess of circulating fibroblast growth factor 23 (FGF23) contribute to cardiovascular mortality in patients with chronic kidney disease (CKD). FGF23 activates FGF receptor 4 and (FGFR4) calcineurin/nuclear factor of activated T cells (NFAT) signaling in cardiac myocytes, thereby causing left ventricular hypertrophy (LVH). Here, we determined if 1,25-dihydroxyvitamin D (calcitriol) inhibits FGF23-induced cardiac signaling and LVH. Methods 5/6 nephrectomized (5/6 Nx) rats were treated with different doses of calcitriol for 4 or 10 weeks and cardiac expression of FGF23/FGFR4 and activation of calcineurin/NFAT as well as LVH were analyzed. FGFR4 activation and hypertrophic cell growth were studied in cultured cardiac myocytes that were co-treated with FGF23 and calcitriol. Results In 5/6Nx rats with LVH, we detected elevated FGF23 expression in bone and myocardium, increased cardiac expression of FGFR4 and elevated cardiac activation of calcineurin/NFAT signaling. Cardiac expression levels of FGF23 and FGFR4 significantly correlated with the presence of LVH in uremic rats. Treatment with calcitriol reduced LVH as well as cardiac FGFR4 expression and calcineurin/NFAT activation. Bone and cardiac FGF23 expression were further stimulated by calcitriol in a dose-dependent manner, but levels of intact cardiac FGF23 protein were suppressed by high-dose calcitriol. In cultured cardiac myocytes, co-treatment with calcitriol blocked FGF23-induced activation of FGFR4 and hypertrophic cell growth. Conclusions Our data suggest that in CKD, cardioprotective effects of calcitriol stem from its inhibitory actions on the cardiac FGF23/FGFR4 system, and based on their counterbalancing effects on cardiac myocytes, high FGF23 and low calcitriol synergistically contribute to cardiac hypertrophy.
Collapse
Affiliation(s)
- Maren Leifheit-Nestler
- Department of Pediatric Kidney, Liver and Metabolic Diseases, Hannover Medical School, Hannover, Germany
| | - Alexander Grabner
- Department of Medicine, Katz Family Drug Discovery Center and Division of Nephrology and Hypertension, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Laura Hermann
- Department of Pediatric Kidney, Liver and Metabolic Diseases, Hannover Medical School, Hannover, Germany
| | - Beatrice Richter
- Department of Pediatric Kidney, Liver and Metabolic Diseases, Hannover Medical School, Hannover, Germany
| | - Karin Schmitz
- Department of Pediatrics, University Hospital Rostock, Rostock, Germany
| | | | - Christopher Yanucil
- Department of Medicine, Katz Family Drug Discovery Center and Division of Nephrology and Hypertension, University of Miami Miller School of Medicine, Miami, FL, USA.,Department of Cell Biology and Anatomy, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA
| | - Christian Faul
- Department of Medicine, Katz Family Drug Discovery Center and Division of Nephrology and Hypertension, University of Miami Miller School of Medicine, Miami, FL, USA.,Department of Cell Biology and Anatomy, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA
| | - Dieter Haffner
- Department of Pediatric Kidney, Liver and Metabolic Diseases, Hannover Medical School, Hannover, Germany
| |
Collapse
|
44
|
Sárközy M, Kahán Z, Csont T. A myriad of roles of miR-25 in health and disease. Oncotarget 2018; 9:21580-21612. [PMID: 29765562 PMCID: PMC5940376 DOI: 10.18632/oncotarget.24662] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 01/30/2018] [Indexed: 02/06/2023] Open
Abstract
Small non-coding RNAs including microRNAs (miRNAs) have been recently recognized as important regulators of gene expression. MicroRNAs play myriads of roles in physiological processes as well as in the pathogenesis of a number of diseases by translational repression or mRNA destabilization of numerous target genes. The miR-106b-25 cluster is highly conserved in vertebrates and consists of three members including miR-106b, miR-93 and miR-25. MiR-106b and miR-93 share the same seed sequences; however, miR-25 has only a similar seed sequence resulting in different predicted target mRNAs. In this review, we specifically focus on the role of miR-25 in healthy and diseased conditions. Many of miR-25 target mRNAs are involved in biological processes such as cell proliferation, differentiation, and migration, apoptosis, oxidative stress, inflammation, calcium handling, etc. Therefore, it is no surprise that miR-25 has been reported as a key regulator of common cancerous and non-cancerous diseases. MiR-25 plays an important role in the pathogenesis of acute myocardial infarction, left ventricular hypertrophy, heart failure, diabetes mellitus, diabetic nephropathy, tubulointerstitial nephropathy, asthma bronchiale, cerebral ischemia/reperfusion injury, neurodegenerative diseases, schizophrenia, multiple sclerosis, etc. MiR-25 is also a well-described oncogenic miRNA playing a crucial role in the development of many tumor types including brain tumors, lung, breast, ovarian, prostate, thyroid, oesophageal, gastric, colorectal, hepatocellular cancers, etc. In this review, our aim is to discuss the translational therapeutic role of miR-25 in common diseased conditions based on relevant basic research and clinical studies.
Collapse
Affiliation(s)
- Márta Sárközy
- Department of Biochemistry, Faculty of Medicine, University of Szeged, H-6720 Szeged, Hungary
| | - Zsuzsanna Kahán
- Department of Oncotherapy, Faculty of Medicine, University of Szeged, H-6720 Szeged, Hungary
| | - Tamás Csont
- Department of Biochemistry, Faculty of Medicine, University of Szeged, H-6720 Szeged, Hungary
| |
Collapse
|
45
|
Bauer A, Khalil M, Lüdemann M, Bauer J, Esmaeili A, De-Rosa R, Voelkel NF, Akintuerk H, Schranz D. Creation of a restrictive atrial communication in heart failure with preserved and mid-range ejection fraction: effective palliation of left atrial hypertension and pulmonary congestion. Clin Res Cardiol 2018; 107:845-857. [DOI: 10.1007/s00392-018-1255-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 04/12/2018] [Indexed: 11/28/2022]
|
46
|
Shi X, Zhang Y, Li B, Peng M, Yuan Y, Wang X, Li X, Yu D, Li Y, Qin D. NOTCH4 is a possible novel susceptibility gene for dilated cardiomyopathy in the Chinese population: A case-control study. J Clin Lab Anal 2018; 32:e22436. [PMID: 29577422 DOI: 10.1002/jcla.22436] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Accepted: 02/27/2018] [Indexed: 11/07/2022] Open
Abstract
BACKGROUND The incidence of dilated cardiomyopathy (DCM) has increased in recent years, and many studies have sought to further improve the general understanding of this condition. Previous studies have demonstrated that some single nucleotide polymorphisms (SNPs) associated with systemic lupus erythematosus also affect susceptibility to DCM, suggesting that immune-related diseases may share similar genetic susceptibility. Recent large-scale and genome-wide association studies have identified NCR3, NOTCH4, CYP1A2, ITGA1, OPRM1, ST8SIA2, and LINC00704 as genetic risk factors associated with cardiac manifestations of neonatal lupus. Here, we aimed to determine whether these SNPs conferred susceptibility to DCM in the Chinese Han population. METHODS We investigated the relationship between these polymorphisms and DCM risk in 273 patients with DCM and 548 healthy controls. Genotyping was performed using MassArray iPLEX system. RESULTS Logistic regression analysis indicated that the T allele of rs3134942 in NOTCH4 gene increased the risk of DCM by 61% compared with the G allele (Pa = 6.57 × 10-3 ). The SNP rs3134942 was also significantly associated with increased DCM risk in the additive (Pa = 6.57 × 10-3 ) and dominant models (Pa = 1.01 × 10-2 ). Additionally, rs2472299 in CYP1A2 gene showed suggestive association with reduced risk of DCM in the dominant model (Pa = 4.24 × 10-2 ) and was correlated with smoking status in patients with DCM (Pa = 1.56 × 10-2 ). CONCLUSIONS Our findings suggested that rs3134942 in NOTCH4 may be involved in DCM risk. Further, studies in larger and ethnically diverse populations are required to confirm the results reported in this study.
Collapse
Affiliation(s)
- Xiaoqing Shi
- Department of Clinical Laboratory, Key Laboratory of Laboratory Medicine of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yang Zhang
- Center of Laboratory Medicine, National Center for Cardiovascular Diseases & Fuwai Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, , Beijing, China
| | - Bingjie Li
- Department of Clinical Laboratory, Key Laboratory of Laboratory Medicine of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Mengle Peng
- Department of Clinical Laboratory, Key Laboratory of Laboratory Medicine of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yingying Yuan
- Department of Clinical Laboratory, Key Laboratory of Laboratory Medicine of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Ximing Wang
- Center of Laboratory Medicine, National Center for Cardiovascular Diseases & Fuwai Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, , Beijing, China
| | - Xinqiang Li
- Center of Laboratory Medicine, National Center for Cardiovascular Diseases & Fuwai Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, , Beijing, China
| | - Dongze Yu
- Center of Laboratory Medicine, National Center for Cardiovascular Diseases & Fuwai Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, , Beijing, China
| | - Yongzhe Li
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Peking Union Medical College, Beijing, China
| | - Dongchun Qin
- Department of Clinical Laboratory, Key Laboratory of Laboratory Medicine of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
47
|
Diviani D, Osman H, Reggi E. A-Kinase Anchoring Protein-Lbc: A Molecular Scaffold Involved in Cardiac Protection. J Cardiovasc Dev Dis 2018; 5:E12. [PMID: 29419761 PMCID: PMC5872360 DOI: 10.3390/jcdd5010012] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 02/02/2018] [Accepted: 02/06/2018] [Indexed: 12/13/2022] Open
Abstract
Heart failure is a lethal disease that can develop after myocardial infarction, hypertension, or anticancer therapy. In the damaged heart, loss of function is mainly due to cardiomyocyte death and associated cardiac remodeling and fibrosis. In this context, A-kinase anchoring proteins (AKAPs) constitute a family of scaffolding proteins that facilitate the spatiotemporal activation of the cyclic adenosine monophosphate (AMP)-dependent protein kinase (PKA) and other transduction enzymes involved in cardiac remodeling. AKAP-Lbc, a cardiac enriched anchoring protein, has been shown to act as a key coordinator of the activity of signaling pathways involved in cardiac protection and remodeling. This review will summarize and discuss recent advances highlighting the role of the AKAP-Lbc signalosome in orchestrating adaptive responses in the stressed heart.
Collapse
Affiliation(s)
- Dario Diviani
- Département de Pharmacologie et de Toxicologie, Faculté de Biologie et de Médecine, Lausanne 1005, Switzerland.
| | - Halima Osman
- Département de Pharmacologie et de Toxicologie, Faculté de Biologie et de Médecine, Lausanne 1005, Switzerland.
| | - Erica Reggi
- Département de Pharmacologie et de Toxicologie, Faculté de Biologie et de Médecine, Lausanne 1005, Switzerland.
| |
Collapse
|
48
|
Field LJ, Shou W, Markham L. 2017 Riley Heart Center Symposium on Cardiac Development: Development and Repair of the Ventricular Wall. Pediatr Cardiol 2018; 39:1067-1068. [PMID: 30066104 PMCID: PMC6096844 DOI: 10.1007/s00246-018-1942-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Loren J. Field
- The Riley Heart Center and the Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN USA
| | - Weinian Shou
- The Riley Heart Center and the Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN USA
| | - Larry Markham
- The Riley Heart Center and the Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN USA
| |
Collapse
|
49
|
Bryan CE, Bossart GD, Christopher SJ, Davis WC, Kilpatrick LE, McFee WE, O'Brien TX. Selenium protein identification and profiling by mass spectrometry: A tool to assess progression of cardiomyopathy in a whale model. J Trace Elem Med Biol 2017; 44:40-49. [PMID: 28965599 PMCID: PMC5657608 DOI: 10.1016/j.jtemb.2017.05.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 05/18/2017] [Accepted: 05/19/2017] [Indexed: 11/19/2022]
Abstract
Non-ischemic cardiomyopathy is a leading cause of congestive heart failure and sudden cardiac death in humans and in some cases the etiology of cardiomyopathy can include the downstream effects of an essential element deficiency. Of all mammal species, pygmy sperm whales (Kogia breviceps) present the greatest known prevalence of cardiomyopathy with more than half of examined individuals indicating the presence of cardiomyopathy from gross and histo-pathology. Several factors such as genetics, infectious agents, contaminants, biotoxins, and inappropriate dietary intake (vitamins, selenium, mercury, and pro-oxidants), may contribute to the development of idiopathic cardiomyopathy in K. breviceps. Due to the important role Se can play in antioxidant biochemistry and protein formation, Se protein presence and relative abundance were explored in cardiomyopathy related cases. Selenium proteins were separated and detected by multi-dimension liquid chromatography inductively coupled plasma mass spectrometry (LC-ICP-MS), Se protein identification was performed by liquid chromatography electrospray tandem mass spectrometry (LC-ESI-MS/MS), and Se protein profiles were examined in liver (n=30) and heart tissue (n=5) by SEC/UV/ICP-MS detection. Data collected on selenium proteins was evaluated in the context of individual animal trace element concentration, life history, and histological information. Selenium containing protein peak profiles varied in presence and intensity between animals with no pathological findings of cardiomyopathy and animals exhibiting evidence of cardiomyopathy. In particular, one class of proteins, metallothioneins, was found to be associated with Se and was in greater abundance in animals with cardiomyopathy than those with no pathological findings. Profiling Se species with SEC/ICP-MS proved to be a useful tool to identify Se protein pattern differences between heart disease stages in K. breviceps and an approach similar to this may be applied to other species to study Se protein associations with cardiomyopathy.
Collapse
Affiliation(s)
- Colleen E Bryan
- Chemical Sciences Division, National Institute of Standards and Technology, Hollings Marine Laboratory, 331 Fort Johnson Road, Charleston, SC 29412, USA.
| | | | - Steven J Christopher
- Chemical Sciences Division, National Institute of Standards and Technology, Hollings Marine Laboratory, 331 Fort Johnson Road, Charleston, SC 29412, USA
| | - W Clay Davis
- Chemical Sciences Division, National Institute of Standards and Technology, Hollings Marine Laboratory, 331 Fort Johnson Road, Charleston, SC 29412, USA
| | - Lisa E Kilpatrick
- Biomolecular Measurement Division, National Institute of Standards and Technology, 100 Bureau Drive, Gaithersburg, MD 20899, USA
| | - Wayne E McFee
- Center for Coastal Environmental Health and Biomolecular Research, National Ocean Service, National Oceanic and Atmospheric Administration, 219 Fort Johnson Road, Charleston, SC 29412, USA
| | - Terrence X O'Brien
- Department of Medicine, Division of Cardiology, Medical University of South Carolina, Carolina 25 Courtenay Dr. ART 7063 and the Office of Research and Development, Ralph H. Johnson Veterans Administration Medical Center, Charleston, SC 29425, USA
| |
Collapse
|
50
|
Nishiuchi S, Makiyama T, Aiba T, Nakajima K, Hirose S, Kohjitani H, Yamamoto Y, Harita T, Hayano M, Wuriyanghai Y, Chen J, Sasaki K, Yagihara N, Ishikawa T, Onoue K, Murakoshi N, Watanabe I, Ohkubo K, Watanabe H, Ohno S, Doi T, Shizuta S, Minamino T, Saito Y, Oginosawa Y, Nogami A, Aonuma K, Kusano K, Makita N, Shimizu W, Horie M, Kimura T. Gene-Based Risk Stratification for Cardiac Disorders in
LMNA
Mutation Carriers. ACTA ACUST UNITED AC 2017; 10:CIRCGENETICS.116.001603. [DOI: 10.1161/circgenetics.116.001603] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 09/25/2017] [Indexed: 01/01/2023]
Abstract
Background—
Mutations in
LMNA
(
lamin A/C
), which encodes lamin A and C, typically cause age-dependent cardiac phenotypes, including dilated cardiomyopathy, cardiac conduction disturbance, atrial fibrillation, and malignant ventricular arrhythmias. Although the type of
LMNA
mutations have been reported to be associated with susceptibility to malignant ventricular arrhythmias, the gene-based risk stratification for cardiac complications remains unexplored.
Methods and Results—
The multicenter cohort included 77
LMNA
mutation carriers from 45 families; cardiac disorders were retrospectively analyzed. The mean age of patients when they underwent genetic testing was 45±17, and they were followed for a median 49 months. Of the 77 carriers, 71 (92%) were phenotypically affected and showed cardiac conduction disturbance (81%), low left ventricular ejection fraction (<50%; 45%), atrial arrhythmias (58%), and malignant ventricular arrhythmias (26%). During the follow-up period, 9 (12%) died, either from end-stage heart failure (n=7) or suddenly (n=2). Genetic analysis showed truncation mutations in 58 patients from 31 families and missense mutations in 19 patients from 14 families. The onset of cardiac disorders indicated that subjects with truncation mutations had an earlier occurrence of cardiac conduction disturbance and low left ventricular ejection fraction, than those with missense mutations. In addition, the truncation mutation was found to be a risk factor for the early onset of cardiac conduction disturbance and the occurrence of atrial arrhythmias and low left ventricular ejection fraction, as estimated using multivariable analyses.
Conclusions—
The truncation mutations were associated with manifestation of cardiac phenotypes in
LMNA
-related cardiomyopathy, suggesting that genetic analysis might be useful for diagnosis and risk stratification.
Collapse
|