1
|
Hespe S, Gray B, Puranik R, Peters S, Sweeting J, Ingles J. The role of genetic testing in management and prognosis of individuals with inherited cardiomyopathies. Trends Cardiovasc Med 2025; 35:34-44. [PMID: 39004295 DOI: 10.1016/j.tcm.2024.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 06/23/2024] [Accepted: 06/24/2024] [Indexed: 07/16/2024]
Abstract
Inherited cardiomyopathies are a heterogeneous group of heart muscle conditions where disease classification has traditionally been based on clinical characteristics. However, this does not always align with genotype. While there are well described challenges of genetic testing, understanding the role of genotype in patient management is increasingly required. We take a gene-by-gene approach, reviewing current evidence for the role of genetic testing in guiding prognosis and management of individuals with inherited cardiomyopathies. In particular, focusing on causal variants in genes definitively associated with arrhythmogenic cardiomyopathy, dilated cardiomyopathy, and hypertrophic cardiomyopathy. This review identifies genotype-specific disease sub-groups with strong evidence supporting the use of genetics in clinical management and highlights that at present, the spectrum of clinical utility is not reflected in current guidelines. Of 13 guideline or expert consensus statements for management of cardiomyopathies, there are seven gene-specific therapeutic recommendations that have been published from four documents. Understanding how genotype influences phenotype provides evidence for the role of genetic testing for prognostic and therapeutic purposes, moving us closer to precision-medicine based care.
Collapse
Affiliation(s)
- Sophie Hespe
- Genomics and Inherited Disease Program, Garvan Institute of Medical Research, and UNSW Sydney, Sydney, Australia; Faculty of Medicine and Health, The University of Sydney, Australia
| | - Belinda Gray
- Faculty of Medicine and Health, The University of Sydney, Australia; Department of Cardiology, Royal Prince Alfred Hospital, Sydney, Australia
| | - Rajesh Puranik
- Faculty of Medicine and Health, The University of Sydney, Australia; Department of Cardiology, Royal Prince Alfred Hospital, Sydney, Australia
| | - Stacey Peters
- Department of Cardiology and Genomic Medicine, Royal Melbourne Hospital, Melbourne, Australia; Department of Medicine, University of Melbourne, Melbourne, Australia
| | - Joanna Sweeting
- Genomics and Inherited Disease Program, Garvan Institute of Medical Research, and UNSW Sydney, Sydney, Australia
| | - Jodie Ingles
- Genomics and Inherited Disease Program, Garvan Institute of Medical Research, and UNSW Sydney, Sydney, Australia; Faculty of Medicine and Health, The University of Sydney, Australia; Department of Cardiology, Royal Prince Alfred Hospital, Sydney, Australia.
| |
Collapse
|
2
|
Guo J, Jiang H, Schuftan D, Moreno JD, Ramahdita G, Aryan L, Bhagavan D, Silva J, Huebsch N. Substrate mechanics unveil early structural and functional pathology in iPSC micro-tissue models of hypertrophic cardiomyopathy. iScience 2024; 27:109954. [PMID: 38827401 PMCID: PMC11141149 DOI: 10.1016/j.isci.2024.109954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 03/22/2024] [Accepted: 05/08/2024] [Indexed: 06/04/2024] Open
Abstract
Hypertension is a major cause of morbidity and mortality in patients with hypertrophic cardiomyopathy (HCM), suggesting a potential role for mechanics in HCM pathogenesis. Here, we developed an in vitro physiological model to investigate how mechanics acts together with HCM-linked myosin binding protein C (MYBPC3) mutations to trigger disease. Micro-heart muscles (μHM) were engineered from induced pluripotent stem cell (iPSC)-derived cardiomyocytes bearing MYBPC3+/- mutations and challenged to contract against substrates of different elasticity. μHMs that worked against substrates with stiffness at or exceeding the stiffness of healthy adult heart muscle exhibited several hallmarks of HCM, including cellular hypertrophy, impaired contractile energetics, and maladaptive calcium handling. Remarkably, we discovered changes in troponin C and T localization in MYBPC3+/- μHM that were entirely absent in 2D culture. Pharmacologic studies suggested that excessive Ca2+ intake through membrane-embedded channels underlie the observed electrophysiological abnormalities. These results illustrate the power of physiologically relevant engineered tissue models to study inherited disease with iPSC technology.
Collapse
Affiliation(s)
- Jingxuan Guo
- Department of Mechanical Engineering and Material Science, Washington University in Saint Louis, Saint Louis, MO 63130, USA
| | - Huanzhu Jiang
- Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, MO 63130, USA
| | - David Schuftan
- Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, MO 63130, USA
| | - Jonathan D. Moreno
- Division of Cardiology, Department of Medicine, Washington University in Saint Louis, Saint Louis, MO 63130, USA
| | - Ghiska Ramahdita
- Department of Mechanical Engineering and Material Science, Washington University in Saint Louis, Saint Louis, MO 63130, USA
- NSF Science and Technology Center for Engineering Mechanobiology, Washington University in Saint Louis, Saint Louis, MO 63130, USA
| | - Lavanya Aryan
- Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, MO 63130, USA
| | - Druv Bhagavan
- Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, MO 63130, USA
| | - Jonathan Silva
- Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, MO 63130, USA
| | - Nathaniel Huebsch
- Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, MO 63130, USA
- NSF Science and Technology Center for Engineering Mechanobiology, Washington University in Saint Louis, Saint Louis, MO 63130, USA
- Center for Cardiovascular Research, Center for Regenerative Medicine, Center for Investigation of Membrane Excitability Diseases, Washington University in Saint Louis, Saint Louis, MO 63130, USA
| |
Collapse
|
3
|
Mikšiūnas R, Labeit S, Bironaite D. Class I and II Histone Deacetylase Inhibitors as Therapeutic Modulators of Dilated Cardiac Tissue-Derived Mesenchymal Stem/Stromal Cells. Int J Mol Sci 2024; 25:6758. [PMID: 38928463 PMCID: PMC11203858 DOI: 10.3390/ijms25126758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 06/14/2024] [Accepted: 06/15/2024] [Indexed: 06/28/2024] Open
Abstract
The prevalence of dilated cardiomyopathy (DCM) is increasing globally, highlighting the need for innovative therapeutic approaches to prevent its onset. In this study, we examined the energetic and epigenetic distinctions between dilated and non-dilated human myocardium-derived mesenchymal stem/stromal cells (hmMSCs) and assessed the effects of class I and II HDAC inhibitors (HDACi) on these cells and their cardiomyogenic differentiation. Cells were isolated from myocardium biopsies using explant outgrowth methods. Mitochondrial and histone deacetylase activities, ATP levels, cardiac transcription factors, and structural proteins were assessed using flow cytometry, PCR, chemiluminescence, Western blotting, and immunohistochemistry. The data suggest that the tested HDAC inhibitors improved acetylation and enhanced the energetic status of both types of cells, with significant effects observed in dilated myocardium-derived hmMSCs. Additionally, the HDAC inhibitors activated the cardiac transcription factors Nkx2-5, HOPX, GATA4, and Mef2C, and upregulated structural proteins such as cardiac troponin T and alpha cardiac actin at both the protein and gene levels. In conclusion, our findings suggest that HDACi may serve as potential modulators of the energetic status and cardiomyogenic differentiation of human heart hmMSCs. This avenue of exploration could broaden the search for novel therapeutic interventions for dilated cardiomyopathy, ultimately leading to improvements in heart function.
Collapse
Affiliation(s)
- Rokas Mikšiūnas
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Santariškių 5, LT-08406 Vilnius, Lithuania;
| | | | - Daiva Bironaite
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Santariškių 5, LT-08406 Vilnius, Lithuania;
| |
Collapse
|
4
|
Xu M, Bermea KC, Ayati M, Kim HB, Yang X, Medina A, Fu Z, Heravi A, Zhang X, Na CH, Everett AD, Gabrielson K, Foster DB, Paolocci N, Murphy AM, Ramirez-Correa GA. Alteration in tyrosine phosphorylation of cardiac proteome and EGFR pathway contribute to hypertrophic cardiomyopathy. Commun Biol 2022; 5:1251. [PMID: 36380187 PMCID: PMC9666710 DOI: 10.1038/s42003-022-04021-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 09/22/2022] [Indexed: 11/16/2022] Open
Abstract
Alterations of serine/threonine phosphorylation of the cardiac proteome are a hallmark of heart failure. However, the contribution of tyrosine phosphorylation (pTyr) to the pathogenesis of cardiac hypertrophy remains unclear. We use global mapping to discover and quantify site-specific pTyr in two cardiac hypertrophic mouse models, i.e., cardiac overexpression of ErbB2 (TgErbB2) and α myosin heavy chain R403Q (R403Q-αMyHC Tg), compared to control hearts. From this, there are significant phosphoproteomic alterations in TgErbB2 mice in right ventricular cardiomyopathy, hypertrophic cardiomyopathy (HCM), and dilated cardiomyopathy (DCM) pathways. On the other hand, R403Q-αMyHC Tg mice indicated that the EGFR1 pathway is central for cardiac hypertrophy, along with angiopoietin, ErbB, growth hormone, and chemokine signaling pathways activation. Surprisingly, most myofilament proteins have downregulation of pTyr rather than upregulation. Kinase-substrate enrichment analysis (KSEA) shows a marked downregulation of MAPK pathway activity downstream of k-Ras in TgErbB2 mice and activation of EGFR, focal adhesion, PDGFR, and actin cytoskeleton pathways. In vivo ErbB2 inhibition by AG-825 decreases cardiomyocyte disarray. Serine/threonine and tyrosine phosphoproteome confirm the above-described pathways and the effectiveness of AG-825 Treatment. Thus, altered pTyr may play a regulatory role in cardiac hypertrophic models.
Collapse
Affiliation(s)
- Mingguo Xu
- grid.21107.350000 0001 2171 9311Department of Pediatrics/Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD USA ,Department of Pediatrics, The Third People’s Hospital of Longgang District, Shenzhen, 518115 China
| | - Kevin C. Bermea
- grid.21107.350000 0001 2171 9311Department of Pediatrics/Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Marzieh Ayati
- grid.449717.80000 0004 5374 269XDeparment of Computer Science/College of Engineering and Computer Science, University of Texas Rio Grande Valley School of Medicine, Edinburgh, Texas USA
| | - Han Byeol Kim
- grid.21107.350000 0001 2171 9311Department of Neurology/Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Xiaomei Yang
- grid.27255.370000 0004 1761 1174Department of Anesthesiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Ji’nan, China
| | - Andres Medina
- Department of Molecular Science/UT Health Rio Grande Valley, McAllen, TX USA
| | - Zongming Fu
- grid.21107.350000 0001 2171 9311Department of Pediatrics/Division of Hematology, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Amir Heravi
- grid.21107.350000 0001 2171 9311Department of Pediatrics/Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Xinyu Zhang
- grid.27255.370000 0004 1761 1174Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Ji’nan, China
| | - Chan Hyun Na
- grid.21107.350000 0001 2171 9311Department of Neurology/Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD USA ,grid.21107.350000 0001 2171 9311Department of Biological Chemistry/McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Allen D. Everett
- grid.21107.350000 0001 2171 9311Department of Pediatrics/Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Kathleen Gabrielson
- grid.21107.350000 0001 2171 9311Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - D. Brian Foster
- grid.21107.350000 0001 2171 9311Department of Medicine/Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Nazareno Paolocci
- grid.21107.350000 0001 2171 9311Department of Medicine/Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD USA ,grid.5608.b0000 0004 1757 3470Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Anne M. Murphy
- grid.21107.350000 0001 2171 9311Department of Pediatrics/Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Genaro A. Ramirez-Correa
- grid.21107.350000 0001 2171 9311Department of Pediatrics/Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD USA ,Department of Molecular Science/UT Health Rio Grande Valley, McAllen, TX USA
| |
Collapse
|
5
|
Genetic Restrictive Cardiomyopathy: Causes and Consequences-An Integrative Approach. Int J Mol Sci 2021; 22:ijms22020558. [PMID: 33429969 PMCID: PMC7827163 DOI: 10.3390/ijms22020558] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 01/05/2021] [Accepted: 01/06/2021] [Indexed: 12/12/2022] Open
Abstract
The sarcomere as the smallest contractile unit is prone to alterations in its functional, structural and associated proteins. Sarcomeric dysfunction leads to heart failure or cardiomyopathies like hypertrophic (HCM) or restrictive cardiomyopathy (RCM) etc. Genetic based RCM, a very rare but severe disease with a high mortality rate, might be induced by mutations in genes of non-sarcomeric, sarcomeric and sarcomere associated proteins. In this review, we discuss the functional effects in correlation to the phenotype and present an integrated model for the development of genetic RCM.
Collapse
|
6
|
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
|
7
|
Histone Deacetylase Inhibitor Suberoylanilide Hydroxamic Acid Improves Energetic Status and Cardiomyogenic Differentiation of Human Dilated Myocardium-Derived Primary Mesenchymal Cells. Int J Mol Sci 2020; 21:ijms21144845. [PMID: 32650632 PMCID: PMC7402340 DOI: 10.3390/ijms21144845] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 07/03/2020] [Accepted: 07/05/2020] [Indexed: 02/08/2023] Open
Abstract
Background. In this study the effect of histone deacetylase (HDAC) inhibitor suberoylanilide hydroxamic acid (SAHA) on the energetic status and cardiomyogenic differentiation of human healthy and dilated myocardium-derived mesenchymal stromal cells (hmMSC) have been investigated. Methods. The hmMSC were isolated from the healthy and dilated post-operation heart biopsies by explant outgrowth method. Cell proliferation, HDAC activity, mitochondrial membrane potential, and level of adenosine triphosphate (ATP) were evaluated. The effect of SAHA on mitochondrial parameters has been investigated also by Seahorse XF analyzer and cardiomyogenic differentiation was confirmed by the expression of transcription factor NK2 Homeobox 5 (Nkx2.5), cardiac troponin T and alpha cardiac actin at gene and protein levels. Results. Dilated myocardium-derived hmMSC had almost 1.5 folds higher HDAC activity compared to the healthy cells and significantly lower mitochondrial membrane potential and ATP level. HDAC class I and II inhibitor SAHA improved energetic status of mitochondria in dilated myocardium-isolated hmMSC and increased expression of cardiac specific proteins during 14 days of exposure of cells to SAHA. Conclusions. HDAC inhibitor SAHA can be a promising therapeutic for dilated cardiomyopathy (DCM). Dilated hmMSC exposed to SAHA improved energetic status and, subsequently, cardiomyogenic differentiation. Data suggest that human dilated myocardium-derived MSC still have cardio tissue regenerative potential, which might be stimulated by HDAC inhibitors.
Collapse
|
8
|
Nayak A, Amrute-Nayak M. SUMO system - a key regulator in sarcomere organization. FEBS J 2020; 287:2176-2190. [PMID: 32096922 DOI: 10.1111/febs.15263] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 02/07/2020] [Accepted: 02/24/2020] [Indexed: 01/14/2023]
Abstract
Skeletal muscles constitute roughly 40% of human body mass. Muscles are specialized tissues that generate force to drive movements through ATP-driven cyclic interactions between the protein filaments, namely actin and myosin filaments. The filaments are organized in an intricate structure called the 'sarcomere', which is a fundamental contractile unit of striated skeletal and cardiac muscle, hosting a fine assembly of macromolecular protein complexes. The micrometer-sized sarcomere units are arranged in a reiterated array within myofibrils of muscle cells. The precise spatial organization of sarcomere is tightly controlled by several molecular mechanisms, indispensable for its force-generating function. Disorganized sarcomeres, either due to erroneous molecular signaling or due to mutations in the sarcomeric proteins, lead to human diseases such as cardiomyopathies and muscle atrophic conditions prevalent in cachexia. Protein post-translational modifications (PTMs) of the sarcomeric proteins serve a critical role in sarcomere formation (sarcomerogenesis), as well as in the steady-state maintenance of sarcomeres. PTMs such as phosphorylation, acetylation, ubiquitination, and SUMOylation provide cells with a swift and reversible means to adapt to an altered molecular and therefore cellular environment. Over the past years, SUMOylation has emerged as a crucial modification with implications for different aspects of cell function, including organizing higher-order protein assemblies. In this review, we highlight the fundamentals of the small ubiquitin-like modifiers (SUMO) pathway and its link specifically to the mechanisms of sarcomere assembly. Furthermore, we discuss recent studies connecting the SUMO pathway-modulated protein homeostasis with sarcomere organization and muscle-related pathologies.
Collapse
Affiliation(s)
- Arnab Nayak
- Institute of Molecular and Cell Physiology, Hannover Medical School, Hannover, Germany
| | - Mamta Amrute-Nayak
- Institute of Molecular and Cell Physiology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
9
|
The Pathogenic Role of Very Low Density Lipoprotein on Atrial Remodeling in the Metabolic Syndrome. Int J Mol Sci 2020; 21:ijms21030891. [PMID: 32019138 PMCID: PMC7037013 DOI: 10.3390/ijms21030891] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 01/24/2020] [Accepted: 01/28/2020] [Indexed: 12/16/2022] Open
Abstract
Atrial fibrillation (AF) is the most common persistent arrhythmia, and can lead to systemic thromboembolism and heart failure. Aging and metabolic syndrome (MetS) are major risks for AF. One of the most important manifestations of MetS is dyslipidemia, but its correlation with AF is ambiguous in clinical observational studies. Although there is a paradoxical relationship between fasting cholesterol and AF incidence, the benefit from lipid lowering therapy in reduction of AF is significant. Here, we reviewed the health burden from AF and MetS, the association between two disease entities, and the metabolism of triglyceride, which is elevated in MetS. We also reviewed scientific evidence for the mechanistic links between very low density lipoproteins (VLDL), which primarily carry circulatory triglyceride, to atrial cardiomyopathy and development of AF. The effects of VLDL to atria suggesting pathogenic to atrial cardiomyopathy and AF include excess lipid accumulation, direct cytotoxicity, abbreviated action potentials, disturbed calcium regulation, delayed conduction velocities, modulated gap junctions, and sarcomere protein derangements. The electrical remodeling and structural changes in concert promote development of atrial cardiomyopathy in MetS and ultimately lead to vulnerability to AF. As VLDL plays a major role in lipid metabolism after meals (rather than fasting state), further human studies that focus on the effects/correlation of postprandial lipids to atrial remodeling are required to determine whether VLDL-targeted therapy can reduce MetS-related AF. On the basis of our scientific evidence, we propose a pivotal role of VLDL in MetS-related atrial cardiomyopathy and vulnerability to AF.
Collapse
|
10
|
Eschenhagen T, Carrier L. Cardiomyopathy phenotypes in human-induced pluripotent stem cell-derived cardiomyocytes-a systematic review. Pflugers Arch 2018; 471:755-768. [PMID: 30324321 PMCID: PMC6475632 DOI: 10.1007/s00424-018-2214-0] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 09/19/2018] [Accepted: 10/02/2018] [Indexed: 12/12/2022]
Abstract
Human-induced pluripotent stem cells (hiPSC) can be differentiated to cardiomyocytes at high efficiency and are increasingly used to study cardiac disease in a human context. This review evaluated 38 studies on hypertrophic (HCM) and dilated cardiomyopathy (DCM) of different genetic causes asking to which extent published data allow the definition of an in vitro HCM/DCM hiPSC-CM phenotype. The data are put in context with the prevailing hypotheses on HCM/DCM dysfunction and pathophysiology. Relatively consistent findings in HCM not reported in DCM were larger cell size (156 ± 85%, n = 15), more nuclear localization of nuclear factor of activated T cells (NFAT; 175 ± 65%, n = 3), and higher β-myosin heavy chain gene expression levels (500 ± 547%, n = 8) than respective controls. Conversely, DCM lines showed consistently less force development than controls (47 ± 23%, n = 9), while HCM forces scattered without clear trend. Both HCM and DCM lines often showed sarcomere disorganization, higher NPPA/NPPB expression levels, and arrhythmic beating behaviour. The data have to be taken with the caveat that reporting frequencies of the various parameters (e.g. cell size, NFAT expression) differ widely between HCM and DCM lines, in which data scatter is large and that only 9/38 studies used isogenic controls. Taken together, the current data provide interesting suggestions for disease-specific phenotypes in HCM/DCM hiPSC-CM but indicate that the field is still in its early days. Systematic, quantitative comparisons and robust, high content assays are warranted to advance the field.
Collapse
Affiliation(s)
- Thomas Eschenhagen
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany. .,Partner Site Hamburg/Kiel/Lübeck, DZHK (German Centre for Cardiovascular Research), Hamburg, Germany.
| | - Lucie Carrier
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany. .,Partner Site Hamburg/Kiel/Lübeck, DZHK (German Centre for Cardiovascular Research), Hamburg, Germany.
| |
Collapse
|
11
|
Zhang H, Yu Z, He J, Hua B, Zhang G. Identification of the molecular mechanisms underlying dilated cardiomyopathy via bioinformatic analysis of gene expression profiles. Exp Ther Med 2016; 13:273-279. [PMID: 28123501 PMCID: PMC5245080 DOI: 10.3892/etm.2016.3953] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 04/21/2016] [Indexed: 12/29/2022] Open
Abstract
In the present study, gene expression profiles of patients with dilated cardiomyopathy (DCM) were re-analyzed with bioinformatics tools to investigate the molecular mechanisms underlying DCM. Gene expression dataset GSE3585 was downloaded from Gene Expression Omnibus, which included seven heart biopsy samples obtained from patients with DCM and five healthy controls. Differential analysis was performed using a Limma package in R to screen for differentially expressed genes (DEGs). Functional enrichment analysis was subsequently conducted for DEGs using the Database for Annotation, Visualization and Integration Discovery. A protein-protein interaction (PPI) network was constructed using information from Search Tool for the Retrieval of Interacting Genes software. A total of 89 DEGs were identified in the patients with DCM, including 67 upregulated and 22 downregulated genes. Functional enrichment analysis demonstrated that the downregulated genes predominantly encoded chromosomal proteins and transport-related proteins, which were significantly associated with the biological processes of ‘nucleosome assembly’, ‘chromatin assembly’, ‘protein-DNA complex assembly’, ‘nucleosome organization’ and ‘DNA packaging’ (H1 histone family member 0, histone cluster 1 H1c, histone cluster 1 H2bd and H2A histone family member Z). The upregulated genes detected in the present study encoded secreted proteins or phosphotransferase, which were associated with biological processes including ‘cell adhesion’ [connective tissue growth factor (CTGF)], ‘skeletal system development’ [CTGF and insulin-like growth factor binding protein 3 (IGFBP3)], ‘muscle organ development’ (SMAD7) and ‘regulation of cell migration’ [SMAD7, IGFBP3 and insulin receptor (INSR)]. Notably, signal transducer and activator of transcription 3, SMAD7, INSR, CTGF, exportin 1, IGFBP3 and phosphatidylinositol-4,5-bisphosphate 3-kinase, catalytic subunit alpha were hub nodes with the higher degree in the PPI network. Therefore, the results of the present study suggested that DEGs may alter the biological processes of ‘nucleosome formation’, ‘cell adhesion’, ‘skeletal system development’, ‘muscle organ development’ and ‘regulation of cell migration’ in the development of DCM.
Collapse
Affiliation(s)
- Hu Zhang
- Department of Cardiaovascular Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Zhuo Yu
- Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Jianchao He
- Department of Cardiaovascular Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Baotong Hua
- Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Guiming Zhang
- Department of Cardiaovascular Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| |
Collapse
|
12
|
Matsa E, Burridge PW, Yu KH, Ahrens JH, Termglinchan V, Wu H, Liu C, Shukla P, Sayed N, Churko JM, Shao N, Woo NA, Chao AS, Gold JD, Karakikes I, Snyder MP, Wu JC. Transcriptome Profiling of Patient-Specific Human iPSC-Cardiomyocytes Predicts Individual Drug Safety and Efficacy Responses In Vitro. Cell Stem Cell 2016; 19:311-25. [PMID: 27545504 PMCID: PMC5087997 DOI: 10.1016/j.stem.2016.07.006] [Citation(s) in RCA: 113] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Revised: 04/20/2016] [Accepted: 07/15/2016] [Indexed: 01/24/2023]
Abstract
Understanding individual susceptibility to drug-induced cardiotoxicity is key to improving patient safety and preventing drug attrition. Human induced pluripotent stem cells (hiPSCs) enable the study of pharmacological and toxicological responses in patient-specific cardiomyocytes (CMs) and may serve as preclinical platforms for precision medicine. Transcriptome profiling in hiPSC-CMs from seven individuals lacking known cardiovascular disease-associated mutations and in three isogenic human heart tissue and hiPSC-CM pairs showed greater inter-patient variation than intra-patient variation, verifying that reprogramming and differentiation preserve patient-specific gene expression, particularly in metabolic and stress-response genes. Transcriptome-based toxicology analysis predicted and risk-stratified patient-specific susceptibility to cardiotoxicity, and functional assays in hiPSC-CMs using tacrolimus and rosiglitazone, drugs targeting pathways predicted to produce cardiotoxicity, validated inter-patient differential responses. CRISPR/Cas9-mediated pathway correction prevented drug-induced cardiotoxicity. Our data suggest that hiPSC-CMs can be used in vitro to predict and validate patient-specific drug safety and efficacy, potentially enabling future clinical approaches to precision medicine.
Collapse
Affiliation(s)
- Elena Matsa
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Departments of Medicine and Radiology, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | - Paul W Burridge
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Departments of Medicine and Radiology, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Kun-Hsing Yu
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA; Biomedical Informatics Training Program, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - John H Ahrens
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Vittavat Termglinchan
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Departments of Medicine and Radiology, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Haodi Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Departments of Medicine and Radiology, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Chun Liu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Departments of Medicine and Radiology, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Praveen Shukla
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Departments of Medicine and Radiology, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Nazish Sayed
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Departments of Medicine and Radiology, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jared M Churko
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Departments of Medicine and Radiology, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Ningyi Shao
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Departments of Medicine and Radiology, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Nicole A Woo
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Alexander S Chao
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Joseph D Gold
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Ioannis Karakikes
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Departments of Medicine and Radiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Michael P Snyder
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Departments of Medicine and Radiology, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
13
|
Abstract
Cardiomyopathy is a rare disorder of the heart muscle, affecting 1.13 cases per 100,000 children, from birth to 18 years of age. Cardiomyopathy is the leading cause of heart transplantation in children over the age of 1. The Pediatric Cardiomyopathy Registry funded in 1994 by the National Heart, Lung, and Blood Institute was established to examine the epidemiology of the disease in children below 18 years of age. More than 3500 children across the United States and Canada have been enrolled in the Pediatric Cardiomyopathy Registry, which has followed-up these patients until death, heart transplantation, or loss to follow-up. The Pediatric Cardiomyopathy Registry has provided the most in-depth illustration of this disease regarding its aetiology, clinical course, associated risk factors, and patient outcomes. Data from the registry have helped in guiding the clinical management of cardiomyopathy in children under 18 years of age; however, questions still remain regarding the most clinically effective diagnostic and treatment approaches for these patients. Future directions of the registry include the use of next-generation whole-exome sequencing and cardiac biomarkers to identify aetiology-specific treatments and improve diagnostic strategies. This article provides a brief synopsis of the work carried out by the Pediatric Cardiomyopathy Registry since its inception, including the current knowledge on the aetiologies, outcomes, and treatments of cardiomyopathy in children.
Collapse
|
14
|
Barefield D, Kumar M, Gorham J, Seidman JG, Seidman CE, de Tombe PP, Sadayappan S. Haploinsufficiency of MYBPC3 exacerbates the development of hypertrophic cardiomyopathy in heterozygous mice. J Mol Cell Cardiol 2015; 79:234-43. [PMID: 25463273 PMCID: PMC4642280 DOI: 10.1016/j.yjmcc.2014.11.018] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Revised: 11/10/2014] [Accepted: 11/17/2014] [Indexed: 01/01/2023]
Abstract
Mutations in MYBPC3, the gene encoding cardiac myosin binding protein-C (cMyBP-C), account for ~40% of hypertrophic cardiomyopathy (HCM) cases. Most pathological MYBPC3 mutations encode truncated protein products not found in tissue. Reduced protein levels occur in symptomatic heterozygous human HCM carriers, suggesting haploinsufficiency as an underlying mechanism of disease. However, we do not know if reduced cMyBP-C content results from, or initiates the development of HCM. In previous studies, heterozygous (HET) mice with a MYBPC3 C'-terminal truncation mutation and normal cMyBP-C levels show altered contractile function prior to any overt hypertrophy. Therefore, this study aimed to test whether haploinsufficiency occurs, with decreased cMyBP-C content, following cardiac stress and whether the functional impairment in HET MYBPC3 hearts leads to worsened disease progression. To address these questions, transverse aortic constriction (TAC) was performed on three-month-old wild-type (WT) and HET MYBPC3-truncation mutant mice and then characterized at 4 and 12weeks post-surgery. HET-TAC mice showed increased hypertrophy and reduced ejection fraction compared to WT-TAC mice. At 4weeks post-surgery, HET myofilaments showed significantly reduced cMyBP-C content. Functionally, HET-TAC cardiomyocytes showed impaired force generation, higher Ca(2+) sensitivity, and blunted length-dependent increase in force generation. RNA sequencing revealed several differentially regulated genes between HET and WT groups, including regulators of remodeling and hypertrophic response. Collectively, these results demonstrate that haploinsufficiency occurs in HET MYBPC3 mutant carriers following stress, causing, in turn, reduced cMyBP-C content and exacerbating the development of dysfunction at myofilament and whole-heart levels.
Collapse
Affiliation(s)
- David Barefield
- Department of Cell and Molecular Physiology, Health Sciences Division, Loyola University Chicago, Maywood, IL, USA
| | - Mohit Kumar
- Department of Cell and Molecular Physiology, Health Sciences Division, Loyola University Chicago, Maywood, IL, USA
| | - Joshua Gorham
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | | | | | - Pieter P de Tombe
- Department of Cell and Molecular Physiology, Health Sciences Division, Loyola University Chicago, Maywood, IL, USA
| | - Sakthivel Sadayappan
- Department of Cell and Molecular Physiology, Health Sciences Division, Loyola University Chicago, Maywood, IL, USA.
| |
Collapse
|
15
|
Strande JL. Haploinsufficiency MYBPC3 mutations: another stress induced cardiomyopathy? Let's take a look! J Mol Cell Cardiol 2014; 79:284-6. [PMID: 25524041 DOI: 10.1016/j.yjmcc.2014.12.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Revised: 12/08/2014] [Accepted: 12/09/2014] [Indexed: 01/18/2023]
Affiliation(s)
- Jennifer L Strande
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA; Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA; Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, USA.
| |
Collapse
|
16
|
Ramirez-Correa GA, Frazier AH, Zhu G, Zhang P, Rappold T, Kooij V, Bedja D, Snyder GA, Lugo-Fagundo NS, Hariharan R, Li Y, Shen X, Gao WD, Cingolani OH, Takimoto E, Foster DB, Murphy AM. Cardiac troponin I Pro82Ser variant induces diastolic dysfunction, blunts β-adrenergic response, and impairs myofilament cooperativity. J Appl Physiol (1985) 2014; 118:212-23. [PMID: 25324519 DOI: 10.1152/japplphysiol.00463.2014] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Troponin I (TnI) variant Pro82Ser (cTnIP82S) was initially considered a disease-causing mutation; however, later studies suggested the contrary. We tested the hypothesis of whether a causal link exists between cTnIP82S and cardiac structural and functional remodeling, such as during aging or chronic pressure overload. A cardiac-specific transgenic (Tg) mouse model of cTnIP82S was created to test this hypothesis. During aging, Tg cTnIP82S displayed diastolic dysfunction, characterized by longer isovolumetric relaxation time, and impaired ejection and relaxation time. In young, Tg mice in vivo pressure-volume loops and intact trabecular preparations revealed normal cardiac contractility at baseline. However, upon β-adrenergic stimulation, a blunted contractile reserve and no hastening in left ventricle relaxation were evident in vivo, whereas, in isolated muscles, Ca(2+) transient amplitude isoproterenol dose-response was blunted. In addition, when exposed to chronic pressure overload, Tg mice show exacerbated hypertrophy and decreased contractility compared with age-matched non-Tg littermates. At the molecular level, this mutation significantly impairs myofilament cooperative activation. Importantly, this occurs in the absence of alterations in TnI or myosin-binding protein C phosphorylation. The cTnIP82S variant occurs near a region of interactions with troponin T; therefore, structural changes in this region could explain its meaningful effects on myofilament cooperativity. Our data indicate that cTnIP82S mutation modifies age-dependent diastolic dysfunction and impairs overall contractility after β-adrenergic stimulation or chronic pressure overload. Thus cTnIP82S variant should be regarded as a disease-modifying factor for dysfunction and adverse remodeling with aging and chronic pressure overload.
Collapse
Affiliation(s)
- Genaro A Ramirez-Correa
- Department of Pediatrics and Division of Pediatric Cardiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Aisha H Frazier
- Department of Pediatrics and Division of Pediatric Cardiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Guangshuo Zhu
- Department of Medicine and Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Pingbo Zhang
- The Hopkins Bayview Proteomics Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Thomas Rappold
- The Hopkins Bayview Proteomics Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Viola Kooij
- The Hopkins Bayview Proteomics Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Djahida Bedja
- Department of Comparative Medicine and Comparative Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Greg A Snyder
- Institute of Human Virology, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Nahyr S Lugo-Fagundo
- Department of Pediatrics and Division of Pediatric Cardiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Raena Hariharan
- Department of Pediatrics and Division of Pediatric Cardiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Yuejin Li
- Department of Pediatrics and Division of Pediatric Cardiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Xiaoxu Shen
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland; and
| | - Wei Dong Gao
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland; and
| | - Oscar H Cingolani
- Department of Medicine and Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Eiki Takimoto
- Department of Medicine and Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - D Brian Foster
- Department of Medicine and Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Anne M Murphy
- Department of Pediatrics and Division of Pediatric Cardiology, Johns Hopkins University School of Medicine, Baltimore, Maryland;
| |
Collapse
|
17
|
Ramirez-Correa GA, Martinez-Ferrando MI, Zhang P, Murphy AM. Targeted proteomics of myofilament phosphorylation and other protein posttranslational modifications. Proteomics Clin Appl 2014; 8:543-53. [DOI: 10.1002/prca.201400034] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Revised: 05/29/2014] [Accepted: 06/24/2014] [Indexed: 12/26/2022]
Affiliation(s)
- Genaro A. Ramirez-Correa
- Department of Pediatrics/Division of Cardiology; Johns Hopkins University School of Medicine; Baltimore MD USA
| | | | - Pingbo Zhang
- The Hopkins Bayview Proteomics Center; Johns Hopkins University School of Medicine; Baltimore MD USA
| | - Anne M. Murphy
- Department of Pediatrics/Division of Cardiology; Johns Hopkins University School of Medicine; Baltimore MD USA
| |
Collapse
|
18
|
Garcia-Pavia P, Cobo-Marcos M, Guzzo-Merello G, Gomez-Bueno M, Bornstein B, Lara-Pezzi E, Segovia J, Alonso-Pulpon L. Genetics in dilated cardiomyopathy. Biomark Med 2014; 7:517-33. [PMID: 23905888 DOI: 10.2217/bmm.13.77] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Discoveries made during the last 20 years have revealed a genetic origin in many cases of dilated cardiomyopathy (DCM). Currently, over 40 genes have been associated with the disease. Mutations in DCM-causing genes induce the condition through a variety of different pathological pathways with complex and not completely understood mechanisms. Genes that encode for sarcomeric, cytoskeletal, nuclear membrane, dystrophin-associated glycoprotein complex and desmosomal proteins are the principal genes involved. In this review we discuss the most frequent DCM-causing genes. We propose a classification in which DCM genes are considered as being major or minor genes according to their mutation frequency and the available supporting evidence. The main phenotypic characteristics associated with each gene are discussed.
Collapse
Affiliation(s)
- Pablo Garcia-Pavia
- Heart Failure & Cardiomyopathy Unit, Department of Cardiology, Hospital Universitario Puerta de Hierro, Madrid, Spain.
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Ryan TD, Gupta A, Gupta D, Goldenberg P, Taylor MD, Lorts A, Jefferies JL. Dilated cardiomyopathy in a 32-year-old woman with Russell-Silver syndrome. Cardiovasc Pathol 2013; 23:21-7. [PMID: 24075556 DOI: 10.1016/j.carpath.2013.08.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Revised: 08/08/2013] [Accepted: 08/14/2013] [Indexed: 10/26/2022] Open
Abstract
INTRODUCTION Russell-Silver Syndrome (RSS) is a genetically determined condition characterized by severe intrauterine and postnatal growth retardation; relative macrocephaly; a small, triangular face; and fifth-finger clinodactyly. The etiology of RSS involves epigenetic regulation through either uniparental disomy or genomic imprinting via DNA methylation. There has been no documented association between RSS and cardiomyopathy. METHODS We present an original case of a 32-year-old woman with RSS with dilated a cardiomyopathy who on cardiac biopsy showed occasional hypertrophic and atrophic myocytes with no evidence of inflammation, abnormal sarcomeres and disintegration of the Z bands on ultrastructural analysis, abnormal desmin, and normal C9 immunoreactivity. CONCLUSION This case represents the first reported association between RSS and cardiomyopathy. Given the complex mechanisms of disease etiology in RSS, this novel case provides insights into the mechanism of progressive dilated cardiomyopathy in an older individual with RSS.
Collapse
Affiliation(s)
- Thomas D Ryan
- The Heart Institute, Cincinnati Children's Hospital, Cincinnati, OH 45229, USA
| | | | | | | | | | | | | |
Collapse
|
20
|
Legge CH, López A, Hanna P, Côté E, Hare E, Martinson SA. Histological characterization of dilated cardiomyopathy in the juvenile toy Manchester terrier. Vet Pathol 2013; 50:1043-52. [PMID: 23456967 DOI: 10.1177/0300985813480509] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Dilated cardiomyopathy (DCM), the most common form of cardiomyopathy in the dog, most often occurs in certain breeds. The objective of this study was to describe a rapidly progressive form of DCM that has been recently recognized in juvenile Toy Manchester Terrier dogs (TMTs). The clinical history and gross findings were reviewed in a group of 14 TMTs, and histologic sections of heart were examined in 12 of those 14 TMTs with DCM. Histochemical and histomorphometric analyses were employed to compare the heart in TMTs affected by DCM with that of control dogs. TMTs ranged in age from 10 to 58.3 weeks, with males and females being equally affected. Affected TMT hearts contained foci of degeneration and loss of myofibers with fibrosis and mild lymphoplasmacytic infiltrates. Less prominent features included foci of acute myofiber degeneration and necrosis with or without intralesional mineralization and mild to moderate suppurative and lymphoplasmacytic infiltrates. Morphometric quantification demonstrated that the right ventricle was more severely affected (P ≤ .05) than the left ventricle with variable involvement of the interventricular septum. Immunohistochemistry for canine parvovirus was negative in all heart samples. However, the absence of parvoviral antigen does not rule out a possible viral or autoimmune cause. The presence of these myocardial lesions among closely related dogs suggests a genetic contribution to this disease process in the TMT.
Collapse
Affiliation(s)
- C H Legge
- Department of Pathology and Microbiology, University of Prince Edward Island, 550 University Avenue, Charlottetown, Prince Edward Island, Canada C1A 4P3
| | | | | | | | | | | |
Collapse
|
21
|
Kurosaka S, Leu NA, Pavlov I, Han X, Ribeiro PAB, Xu T, Bunte R, Saha S, Wang J, Cornachione A, Mai W, Yates JR, Rassier DE, Kashina A. Arginylation regulates myofibrils to maintain heart function and prevent dilated cardiomyopathy. J Mol Cell Cardiol 2012; 53:333-41. [PMID: 22626847 DOI: 10.1016/j.yjmcc.2012.05.007] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2012] [Revised: 04/25/2012] [Accepted: 05/12/2012] [Indexed: 01/25/2023]
Abstract
Protein arginylation mediated by arginyltransferase (ATE1) is essential for heart formation during embryogenesis, however its cell-autonomous role in cardiomyocytes and the differentiated heart muscle has never been investigated. To address this question, we generated cardiac muscle-specific Ate1 knockout mice, in which Ate1 deletion was driven by α-myosin heavy chain promoter (αMHC-Ate1 mouse). These mice were initially viable, but developed severe cardiac contractility defects, dilated cardiomyopathy, and thrombosis over time, resulting in high rates of lethality after 6months of age. These symptoms were accompanied by severe ultrastructural defects in cardiac myofibrils, seen in the newborns and far preceding the onset of cardiomyopathy, suggesting that these defects were primary and likely underlay the development of the future heart defects. Several major sarcomeric proteins were arginylated in vivo. Moreover, Ate1 deletion in the hearts resulted in a significant reduction of active and passive myofibril forces, suggesting that arginylation is critical for both myofibril structural integrity and contractility. Thus, arginylation is essential for maintaining the heart function by regulation of the major myofibril proteins and myofibril forces, and its absence in the heart muscle leads to progressive heart failure through cardiomyocyte-specific defects.
Collapse
Affiliation(s)
- Satoshi Kurosaka
- Department of Animal Biology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia 19104, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Erdal C, Karakülah G, Fermancı E, Kunter I, Silistreli E, Canda T, Erdal E, Hepaguslar H. Early biventricular molecular responses to an acute myocardial infarction. Int J Med Sci 2012; 9:74-82. [PMID: 22211093 PMCID: PMC3245415 DOI: 10.7150/ijms.9.74] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2011] [Accepted: 11/16/2011] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND Acute myocardial infarction (AMI) remains as one of the most common lethal diseases in the world and therefore it is necessary to understand its effect on molecular basis. Genome-wide microarray analysis provides us to predict potential biomarkers and signaling pathways for this purpose. OBJECTIVES The aim of this study is to understand the molecular basis of the immediate right ventricular cellular response to left ventricular AMI. MATERIAL AND METHODS A rat model of left anterior descending coronary artery ligation was used to assess the effect of left ventricular AMI on both the right ventricle as a remote zone and the left ventricle as an ischemic/infarct zone. Microarray technology was applied to detect the gene expression. Gene Ontology and KEGG pathways analysis were done to identify effected pathways and related genes. RESULTS We found that immune response, cell chemotaxis, inflammation, cytoskeleton organization are significantly deregulated in ischemic zone as early response within 30 min. Unexpectedly, there were several affected signaling pathways such as cell chemotaxis, regulation of endothelial cell proliferation, and regulation of caveolea regulation of anti-apoptosis, regulation of cytoskeleton organization and cell adhesion on the remote zone in the right ventricle. CONCLUSION This data demonstrates that there is an immediate molecular response in both ventricles after an AMI. Although the ischemia did not histologically involve the right ventricle; there is a clear molecular response to the infarct in the left ventricle. This provides us new insights to understand molecular mechanisms behind AMI and to find more effective drug targets.
Collapse
Affiliation(s)
- Cenk Erdal
- Department of Cardiovascular Surgery, Dokuz Eylül University, Faculty of Medicine, 35340 İnciraltı İzmir, Turkey.
| | | | | | | | | | | | | | | |
Collapse
|