1
|
Hajdarpašić A, Tukker M, Rijdt WT, Mohamedhoesein S, Meijers WC, Caliskan K. Epigenetics of cardiomyopathies: the next frontier. Heart Fail Rev 2025; 30:257-270. [PMID: 39586986 DOI: 10.1007/s10741-024-10460-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/18/2024] [Indexed: 11/27/2024]
Abstract
Cardiomyopathies (CMP) are a diverse group of myocardial diseases that cause structural, functional, and pathological changes to the heart. Alterations at the molecular level associated with the clinical phenotype and progression of CMPs cannot be solely explained by the genetic mutations, even in inherited cardiomyopathies. Epigenetics and environmental factors are likely to significantly modify the clinical manifestations of CMPs, resulting in variable clinical expression and different age-related penetrance. This review examines the role of dysfunctional DNA methylation, histone modifications, chromatin remodelling, and noncoding RNAs in the development and exacerbation of CMPs, highlighting their potential as diagnostic markers and therapeutic targets, including the use of histone deacetylase inhibitors. Additionally, it explores how environmental exposures can influence epigenetic changes and potentially be used for preventive strategies and personalized care in CMP patients. Monozygotic twin studies and intergenerational studies are discussed as valuable tools for understanding the interplay between genetics, epigenetics, and environmental factors. Lastly, this review addresses current challenges and future perspectives, such as the need for greater specificity in epigenetic therapies, minimizing off-target effects, and investigating sex differences in CMP research and treatment.
Collapse
Affiliation(s)
- Aida Hajdarpašić
- Department of Medical Biology and Genetics, Sarajevo Medical School, University Sarajevo School of Science and Technology, Hrasnička Cesta 3a, 71210, Sarajevo, Ilidža, Bosnia and Herzegovina.
| | - Martijn Tukker
- Thoraxcenter, Department of Cardiology, Cardiovascular Institute. Erasmus MC - University Medical Center Rotterdam, Office RG-431Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - Wouter Te Rijdt
- Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Sharida Mohamedhoesein
- Thoraxcenter, Department of Cardiology, Cardiovascular Institute. Erasmus MC - University Medical Center Rotterdam, Office RG-431Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - Wouter C Meijers
- Thoraxcenter, Department of Cardiology, Cardiovascular Institute. Erasmus MC - University Medical Center Rotterdam, Office RG-431Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - Kadir Caliskan
- Thoraxcenter, Department of Cardiology, Cardiovascular Institute. Erasmus MC - University Medical Center Rotterdam, Office RG-431Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands.
| |
Collapse
|
2
|
Peñarroya A, Lorca R, Rodríguez Reguero JJ, Gómez J, Avanzas P, Tejedor JR, Fernandez AF, Fraga MF. Epigenetic Study of Cohort of Monozygotic Twins With Hypertrophic Cardiomyopathy Due to MYBPC3 (Cardiac Myosin-Binding Protein C). J Am Heart Assoc 2024; 13:e035777. [PMID: 39470061 PMCID: PMC11935665 DOI: 10.1161/jaha.124.035777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 08/12/2024] [Indexed: 10/30/2024]
Abstract
BACKGROUND Hypertrophic cardiomyopathy is an autosomal dominant cardiac disease. The mechanisms that determine its variable expressivity are poorly understood. Epigenetics could play a crucial role in bridging the gap between genotype and phenotype by orchestrating the interplay between the environment and the genome regulation. In this study we aimed to establish a possible correlation between the peripheral blood DNA methylation patterns and left ventricular hypertrophy severity in patients with hypertrophic cardiomyopathy, evaluating the potential impact of lifestyle variables and providing a biological context to the observed changes. METHODS AND RESULTS Methylation data were obtained from peripheral blood samples (Infinium MethylationEPIC BeadChip arrays). We employed multiple pair-matched models to extract genomic positions whose methylation correlates with the degree of left ventricular hypertrophy in 3 monozygotic twin pairs carrying the same founder pathogenic variant (MYBPC3 p.Gly263Ter). This model enables the isolation of the environmental influence, beyond age, on DNA methylation changes by removing the genetic background. Our results revealed a more anxious personality among more severely affected individuals. We identified 56 differentially methylated positions that exhibited moderate, proportional changes in methylation associated with left ventricular hypertrophy. These differentially methylated positions were enriched in regions regulated by repressor histone marks and tended to cluster at genes involved in left ventricular hypertrophy development, such as HOXA5, TRPC3, UCN3, or PLSCR2, suggesting that changes in peripheral blood may reflect myocardial alterations. CONCLUSIONS We present a unique pair-matched model, based on 3 monozygotic twin pairs carrying the same founder pathogenic variant and different phenotypes. This study provides further evidence of the pivotal role of epigenetics in hypertrophic cardiomyopathy variable expressivity.
Collapse
Affiliation(s)
- Alfonso Peñarroya
- Nanomaterials and Nanotechnology Research Center (CINN)Spanish National Research Council (CSIC)El EntregoAsturiasSpain
- Health Research Institute of the Principality of Asturias (ISPA)OviedoAsturiasSpain
| | - Rebeca Lorca
- Health Research Institute of the Principality of Asturias (ISPA)OviedoAsturiasSpain
- Unidad de Cardiopatías Familiares, Área del Corazón y Departamento de Genética MolecularHospital Universitario Central AsturiasOviedoSpain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORs)MadridSpain
- Departamento de Biología FuncionalUniversidad de OviedoOviedoSpain
| | - José Julián Rodríguez Reguero
- Health Research Institute of the Principality of Asturias (ISPA)OviedoAsturiasSpain
- Unidad de Cardiopatías Familiares, Área del Corazón y Departamento de Genética MolecularHospital Universitario Central AsturiasOviedoSpain
| | - Juan Gómez
- Health Research Institute of the Principality of Asturias (ISPA)OviedoAsturiasSpain
- Unidad de Cardiopatías Familiares, Área del Corazón y Departamento de Genética MolecularHospital Universitario Central AsturiasOviedoSpain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORs)MadridSpain
| | - Pablo Avanzas
- Health Research Institute of the Principality of Asturias (ISPA)OviedoAsturiasSpain
- Unidad de Cardiopatías Familiares, Área del Corazón y Departamento de Genética MolecularHospital Universitario Central AsturiasOviedoSpain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORs)MadridSpain
- Departamento de MedicinaUniversidad de OviedoOviedoSpain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV)OviedoSpain
| | - Juan Ramon Tejedor
- Nanomaterials and Nanotechnology Research Center (CINN)Spanish National Research Council (CSIC)El EntregoAsturiasSpain
- Health Research Institute of the Principality of Asturias (ISPA)OviedoAsturiasSpain
- Spanish Biomedical Research Network in Rare Diseases (CIBERER)MadridSpain
- Institute of Oncology of Asturias (IUOPA), University of OviedoOviedoAsturiasSpain
| | - Agustín F. Fernandez
- Nanomaterials and Nanotechnology Research Center (CINN)Spanish National Research Council (CSIC)El EntregoAsturiasSpain
- Health Research Institute of the Principality of Asturias (ISPA)OviedoAsturiasSpain
- Spanish Biomedical Research Network in Rare Diseases (CIBERER)MadridSpain
- Institute of Oncology of Asturias (IUOPA), University of OviedoOviedoAsturiasSpain
| | - Mario F. Fraga
- Nanomaterials and Nanotechnology Research Center (CINN)Spanish National Research Council (CSIC)El EntregoAsturiasSpain
- Health Research Institute of the Principality of Asturias (ISPA)OviedoAsturiasSpain
- Spanish Biomedical Research Network in Rare Diseases (CIBERER)MadridSpain
- Institute of Oncology of Asturias (IUOPA), University of OviedoOviedoAsturiasSpain
| |
Collapse
|
3
|
Wu KJ, Chen Q, Leung CH, Sun N, Gao F, Chen Z. Recent discoveries of the role of histone modifications and related inhibitors in pathological cardiac hypertrophy. Drug Discov Today 2024; 29:103878. [PMID: 38211819 DOI: 10.1016/j.drudis.2024.103878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 12/19/2023] [Accepted: 01/05/2024] [Indexed: 01/13/2024]
Abstract
Pathological cardiac hypertrophy is a common response of the heart to various pathological stimuli. In recent years, various histone modifications, including acetylation, methylation, phosphorylation and ubiquitination, have been identified to have crucial roles in regulating chromatin remodeling and cardiac hypertrophy. Novel drugs targeting these epigenetic changes have emerged as potential treatments for pathological cardiac hypertrophy. In this review, we provide a comprehensive summary of the roles of histone modifications in regulating the development of pathological cardiac hypertrophy, and discuss potential therapeutic targets that could be utilized for its treatment.
Collapse
Affiliation(s)
- Ke-Jia Wu
- Wuxi School of Medicine, Jiangnan University, Jiangsu 214082, PR China
| | - Qi Chen
- Wuxi School of Medicine, Jiangnan University, Jiangsu 214082, PR China
| | - Chung-Hang Leung
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa 999078, Macau; Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Taipa 999078, Macau; Macao Centre for Research and Development in Chinese Medicine, University of Macau, Taipa 999078, Macau; MoE Frontiers Science Centre for Precision Oncology, University of Macau, Taipa 999078, Macau.
| | - Ning Sun
- Wuxi School of Medicine, Jiangnan University, Jiangsu 214082, PR China.
| | - Fei Gao
- Department of Cardiology, Beijing An Zhen Hospital, Capital Medical University, Chaoyang District, Beijing 100029, PR China.
| | - Zhaoyang Chen
- Department of Cardiology, Heart Center of Fujian Province, Fujian Medical University Union Hospital, 29 Xin-Quan Road, Fuzhou, Fujian 350001, PR China.
| |
Collapse
|
4
|
Elias J. T-Wave Microalternans in Hypertrophic Cardiomyopathy: The Complexity of an Inherited Cardiac Condition with Multiple Phenotypic Expressions. Arq Bras Cardiol 2023; 120:e20230615. [PMID: 37909504 PMCID: PMC10586819 DOI: 10.36660/abc.20230615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 09/06/2023] [Accepted: 09/06/2023] [Indexed: 11/03/2023] Open
Affiliation(s)
- Jorge Elias
- Vitória Apart HospitalSerraESBrasilVitória Apart Hospital – Serviço de Eletrofisiologia, Serra, ES – Brasil
- Hospital Universitário Cassiano Antonio MoraesUfesVitóriaESBrasilHospital Universitário Cassiano Antonio Moraes (Hucam) – Ufes, Vitória, ES – Brasil
| |
Collapse
|
5
|
Laura Francés J, Pagiatakis C, Di Mauro V, Climent M. Therapeutic Potential of EVs: Targeting Cardiovascular Diseases. Biomedicines 2023; 11:1907. [PMID: 37509546 PMCID: PMC10377624 DOI: 10.3390/biomedicines11071907] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 06/25/2023] [Accepted: 06/29/2023] [Indexed: 07/30/2023] Open
Abstract
Due to their different biological functions, extracellular vesicles (EVs) have great potential from a therapeutic point of view. They are released by all cell types, carrying and delivering different kinds of biologically functional cargo. Under pathological events, cells can increase their secretion of EVs and can release different amounts of cargo, thus making EVs great biomarkers as indicators of pathological progression. Moreover, EVs are also known to be able to transport and deliver cargo to different recipient cells, having an important role in cellular communication. Interestingly, EVs have recently been explored as biological alternatives for the delivery of therapeutics, being considered natural drug delivery carriers. Because cardiovascular disorders (CVDs) are the leading cause of death worldwide, in this review, we will discuss the up-to-date knowledge regarding the biophysical properties and biological components of EVs, focusing on myocardial infarction, diabetic cardiomyopathy, and sepsis-induced cardiomyopathy, three very different types of CVDs.
Collapse
Affiliation(s)
| | - Christina Pagiatakis
- IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy
- Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy
| | - Vittoria Di Mauro
- IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy
- Veneto Institute of Molecular Medicine, Via Orus 2, 35129 Padova, Italy
- Department of Pathology and Laboratory Medicine, Cardiovascular Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
| | | |
Collapse
|
6
|
Segreti A, Piccirillo F, Crispino SP, Cocchia F, Martucciello A, Calabrese V, Gurrieri F, Grigioni F. Simultaneous presence of Brugada and overgrowth syndromes. Monaldi Arch Chest Dis 2023; 94. [PMID: 37114354 DOI: 10.4081/monaldi.2023.2521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 04/12/2023] [Indexed: 04/29/2023] Open
Abstract
In the present article, we describe the case of a 21-year-old male presenting to the emergency department following a syncopal episode. Physical examination revealed a distinctive facial appearance in the context of an overgrowth syndrome. Also, an ajmaline test was performed because of the evidence of an incomplete right bundle branch block with ST-T segment elevation in the right precordial derivations, revealing a type-1 Brugada electrocardiographic pattern. Considering the high cardiovascular risk phenotype, the patient underwent subcutaneous cardiac defibrillator implantation. The subsequent comprehensive genomic testing analysis led to the diagnosis of a variant of uncertain significance of the nuclear receptor binding SET domain protein 1 (NSD1) gene and a heterozygous mutation of the calsequestrin 2 (CASQ2) gene. NSD1 gene alterations are usually responsible for the Sotos syndrome, characterized by distinctive facial appearance, learning disability, and overgrowth, in addition to cardiac anomalies ranging from single self-limiting alterations to more severe, complex cardiac abnormalities. On the contrary, a compound heterozygous or homozygous alteration of the CASQ2 gene is usually associated with catecholaminergic polymorphic ventricular tachycardia; however, the significance of a merely heterozygous alteration in the CASQ2 gene, as in the present case report, is not yet clear. In conclusion, to the best of our knowledge, this is the first description of the coexisting presence of Brugada and overgrowth syndromes in a single patient.
Collapse
Affiliation(s)
- Andrea Segreti
- Unit of Cardiovascular Science, Campus Bio-Medico University Hospital Foundation, Rome; Department of Movement, Human and Health Sciences, University of Rome "Foro Italico", Rome.
| | - Francesco Piccirillo
- Unit of Cardiovascular Science, Campus Bio-Medico University Hospital Foundation, Rome.
| | | | - Francesca Cocchia
- Unit of Cardiovascular Science, Campus Bio-Medico University Hospital Foundation, Rome.
| | - Arianna Martucciello
- Unit of Cardiovascular Science, Campus Bio-Medico University Hospital Foundation, Rome.
| | - Vito Calabrese
- Unit of Cardiovascular Science, Campus Bio-Medico University Hospital Foundation, Rome.
| | - Fiorella Gurrieri
- Research Unit of Medical Genetics, Department of Medicine and Surgery, Campus Bio-Medico University of Rome; Operative Research Unit of Medical Genetics, Campus Bio-Medico University Hospital Foundation, Rome.
| | - Francesco Grigioni
- Unit of Cardiovascular Science, Campus Bio-Medico University Hospital Foundation, Rome.
| |
Collapse
|
7
|
Singh T, Kaur P, Singh P, Singh S, Munshi A. Differential molecular mechanistic behavior of HDACs in cancer progression. MEDICAL ONCOLOGY (NORTHWOOD, LONDON, ENGLAND) 2022; 39:171. [PMID: 35972597 DOI: 10.1007/s12032-022-01770-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 06/10/2022] [Indexed: 12/13/2022]
Abstract
Genetic aberration including mutation in oncogenes and tumor suppressor genes transforms normal cells into tumor cells. Epigenetic modifications work concertedly with genetic factors in controlling cancer development. Histone acetyltransferases (HATs), histone deacetylases (HDACs), DNA methyltransferases (DNMTs) and chromatin structure modifier are prospective epigenetic regulators. Specifically, HDACs are histone modifiers regulating the expression of genes implicated in cell survival, growth, apoptosis, and metabolism. The majority of HDACs are highly upregulated in cancer, whereas some have a varied function and expression in cancer progression. Distinct HDACs have a positive and negative role in controlling cancer progression. HDACs are also significantly involved in tumor cells acquiring metastatic and angiogenic potential in order to withstand the anti-tumor microenvironment. HDACs' role in modulating metabolic genes has also been associated with tumor development and survival. This review highlights and discusses the molecular mechanisms of HDACs by which they regulate cell survival, apoptosis, metastasis, invasion, stemness potential, angiogenesis, and epithelial to mesenchymal transitions (EMT) in tumor cells. HDACs are the potential target for anti-cancer drug development and various inhibitors have been developed and FDA approved for a variety of cancers. The primary HDAC inhibitors with proven anti-cancer efficacy have also been highlighted in this review.
Collapse
Affiliation(s)
- Tashvinder Singh
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda, 151401, India
| | - Prabhsimran Kaur
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda, 151401, India
| | | | - Sandeep Singh
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda, 151401, India.
| | - Anjana Munshi
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda, 151401, India.
| |
Collapse
|
8
|
Volani C, Pagliaro A, Rainer J, Paglia G, Porro B, Stadiotti I, Foco L, Cogliati E, Paolin A, Lagrasta C, Frati C, Corradini E, Falco A, Matzinger T, Picard A, Ermon B, Piazza S, De Bortoli M, Tondo C, Philippe R, Medici A, Lavdas AA, Blumer MJF, Pompilio G, Sommariva E, Pramstaller PP, Troppmair J, Meraviglia V, Rossini A. GCN5 contributes to intracellular lipid accumulation in human primary cardiac stromal cells from patients affected by Arrhythmogenic cardiomyopathy. J Cell Mol Med 2022; 26:3687-3701. [PMID: 35712781 PMCID: PMC9258704 DOI: 10.1111/jcmm.17396] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 03/20/2022] [Accepted: 04/28/2022] [Indexed: 11/29/2022] Open
Abstract
Arrhythmogenic cardiomyopathy (ACM) is a genetic disease associated with sudden cardiac death and cardiac fibro‐fatty replacement. Over the last years, several works have demonstrated that different epigenetic enzymes can affect not only gene expression changes in cardiac diseases but also cellular metabolism. Specifically, the histone acetyltransferase GCN5 is known to facilitate adipogenesis and modulate cardiac metabolism in heart failure. Our group previously demonstrated that human primary cardiac stromal cells (CStCs) contribute to adipogenesis in the ACM pathology. Thus, this study aims to evaluate the role of GCN5 in ACM intracellular lipid accumulation. To do so, CStCs were obtained from right ventricle biopsies of ACM patients and from samples of healthy cadaveric donors (CTR). GCN5 expression was increased both in ex vivo and in vitro ACM samples compared to CTR. When GCN5 expression was silenced or pharmacologically inhibited by the administration of MB‐3, we observed a reduction in lipid accumulation and a mitigation of reactive oxygen species (ROS) production in ACM CStCs. In agreement, transcriptome analysis revealed that the presence of MB‐3 modified the expression of pathways related to cellular redox balance. Altogether, our findings suggest that GCN5 inhibition reduces fat accumulation in ACM CStCs, partially by modulating intracellular redox balance pathways.
Collapse
Affiliation(s)
- Chiara Volani
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, Bolzano, Italy.,The Cell Physiology MiLab, Department of Biosciences, Università degli Studi di Milano, Milano, Italy
| | - Alessandra Pagliaro
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, Bolzano, Italy
| | - Johannes Rainer
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, Bolzano, Italy
| | - Giuseppe Paglia
- School of Medicine and Surgery, Università degli Studi di Milano-Bicocca, Vedano al Lambro, MB, Italy
| | - Benedetta Porro
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino IRCCS, Milano, Italy
| | - Ilaria Stadiotti
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino IRCCS, Milano, Italy
| | - Luisa Foco
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, Bolzano, Italy
| | | | - Adolfo Paolin
- Fondazione Banca dei Tessuti di Treviso, Treviso, Italy
| | - Costanza Lagrasta
- Department of Medicine and Surgery, Università degli Studi di Parma, Parma, Italy
| | - Caterina Frati
- Department of Medicine and Surgery, Università degli Studi di Parma, Parma, Italy
| | - Emilia Corradini
- Department of Medicine and Surgery, Università degli Studi di Parma, Parma, Italy
| | - Angela Falco
- Department of Medicine and Surgery, Università degli Studi di Parma, Parma, Italy
| | - Theresa Matzinger
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, Bolzano, Italy
| | - Anne Picard
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, Bolzano, Italy
| | - Benedetta Ermon
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, Bolzano, Italy
| | - Silvano Piazza
- Department of Cellular, Computational and Integrative Biology - CIBIO, Università degli Studi di Trento, Povo, TN, Italy.,Computational Biology, International Centre for Genetic Engineering and Biotechnology, ICGEB, Trieste, Italy
| | - Marzia De Bortoli
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, Bolzano, Italy
| | - Claudio Tondo
- Heart Rhythm Center, Centro Cardiologico Monzino IRCCS, Milano, Italy.,Department of Biomedical, Surgical and Dental Sciences, Università degli Studi di Milano, Milano, Italy.,Department of Clinical Electrophysiology&Cardiac Pacing, Università degli Studi di Milano, Milano, Italy
| | - Réginald Philippe
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, Bolzano, Italy
| | - Andrea Medici
- Daniel Swarovski Research Laboratory, Department of Visceral, Transplant and Thoracic Surgery, Medical University Innsbruck, Innsbruck, Austria
| | - Alexandros A Lavdas
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, Bolzano, Italy
| | - Michael J F Blumer
- Department of Anatomy, Histology and Embryology, Institute of Clinical and Functional Anatomy, Medical University Innsbruck, Innsbruck, Austria
| | - Giulio Pompilio
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino IRCCS, Milano, Italy.,Heart Rhythm Center, Centro Cardiologico Monzino IRCCS, Milano, Italy
| | - Elena Sommariva
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino IRCCS, Milano, Italy
| | - Peter P Pramstaller
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, Bolzano, Italy
| | - Jakob Troppmair
- Daniel Swarovski Research Laboratory, Department of Visceral, Transplant and Thoracic Surgery, Medical University Innsbruck, Innsbruck, Austria
| | - Viviana Meraviglia
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, Bolzano, Italy
| | - Alessandra Rossini
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, Bolzano, Italy
| |
Collapse
|
9
|
Yan F, Chen Z, Cui W. H3K9me2 regulation of BDNF expression via G9a partakes in the progression of heart failure. BMC Cardiovasc Disord 2022; 22:182. [PMID: 35439934 PMCID: PMC9020036 DOI: 10.1186/s12872-022-02621-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 04/11/2022] [Indexed: 11/23/2022] Open
Abstract
Background Heart disease is a major cause of mortality in developed countries. The associated pathology is mainly characterized by the loss of cardiomyocytes that contributes to heart failure (HF). This study aims to investigate the mechanism of euchromatic histone lysine methyltransferase 2 (EHMT2, also term G9a) in HF in rats. Methods Differentially expressed mRNAs in HF were screened using GEO database. Sera from subjects with or without HF were collected, and PCR was performed to detect the G9a expression. G9a was downregulated in cardiomyocytes exposed to oxygen–glucose deprivation (OGD), followed by CCK8, flow cytometry, colorimetric method, and western blot assays. Established HF rats were delivered with lentiviral vectors carrying sh-G9a, and TTC staining, HE staining, TUNEL, ELISA, and western blot were performed. The regulation of G9a on the downstream target BDNF was investigated by RT-qPCR, Western blot, and ChIP-qPCR. Finally, rescue experiments were carried out to substantiate the effect of G9a on cardiomyocyte apoptosis and injury via the BDNF/TrkB axis. Results G9a was overexpressed, whereas BDNF was downregulated in HF. Knockdown of G9a inhibited apoptosis and injury in OGD-treated cardiomyocytes and attenuated the extent of HF and myocardial injury in rats. Silencing of G9a promoted BDNF transcription by repressing H3K9me2 modification of the BDNF promoter. Further depletion of BDNF partially reversed the effect of sh-G9a in alleviating cardiomyocyte apoptosis and injury by inhibiting the TrkB signaling pathway. Conclusion G9a inhibits BDNF expression through H3K9me2 modification, thereby impairing the TrkB signaling pathway and exacerbating the development of HF. Supplementary Information The online version contains supplementary material available at 10.1186/s12872-022-02621-w.
Collapse
Affiliation(s)
- Fang Yan
- Department of Cardiac Surgery, Hebei Medical University, Shijiazhuang, 050011, Hebei, People's Republic of China.,Department of Cardiac Surgery, The Second Hospital of Hebei Medical University, No. 215, Heping West Road, Xinhua District, Shijiazhuang, 050000, Hebei, People's Republic of China
| | - Ziying Chen
- Department of Cardiac Surgery, The Second Hospital of Hebei Medical University, No. 215, Heping West Road, Xinhua District, Shijiazhuang, 050000, Hebei, People's Republic of China.
| | - Wei Cui
- Department of Cardiology, The Second Hospital of Hebei Medical University, No. 215, Heping West Road, Xinhua District, Shijiazhuang, 050000, Hebei, People's Republic of China.
| |
Collapse
|
10
|
Nguyen MB, Mital S, Mertens L, Jeewa A, Friedberg MK, Aguet J, Adler A, Lam CZ, Dragulescu A, Rakowski H, Villemain O. Pediatric Hypertrophic Cardiomyopathy: Exploring the Genotype-Phenotype Association. J Am Heart Assoc 2022; 11:e024220. [PMID: 35179047 PMCID: PMC9075072 DOI: 10.1161/jaha.121.024220] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 01/11/2022] [Indexed: 11/19/2022]
Abstract
Pediatric hypertrophic cardiomyopathy (HCM) is the most common form of cardiomyopathy in children and a leading cause of sudden cardiac death. Yet, the association between genotype variation, phenotype expression, and adverse events in pediatric HCM has not been fully elucidated. Although the literature on this topic is evolving in adult HCM, the evidence in children is lacking. Solidifying our understanding of this relationship could improve risk stratification as well as improve our comprehension of the underlying pathophysiological characteristics of pediatric HCM. In this state-of-the-art review, we examine the current literature on genetic variations in HCM and their association with outcomes in children, discuss the current approaches to identifying cardiovascular phenotypes in pediatric HCM, and explore possible avenues that could improve sudden cardiac death risk assessment.
Collapse
Affiliation(s)
- Minh B. Nguyen
- Division of CardiologyLabatt Family Heart CentreHospital for Sick ChildrenUniversity of TorontoOntarioCanada
| | - Seema Mital
- Division of CardiologyLabatt Family Heart CentreHospital for Sick ChildrenUniversity of TorontoOntarioCanada
| | - Luc Mertens
- Division of CardiologyLabatt Family Heart CentreHospital for Sick ChildrenUniversity of TorontoOntarioCanada
| | - Aamir Jeewa
- Division of CardiologyLabatt Family Heart CentreHospital for Sick ChildrenUniversity of TorontoOntarioCanada
| | - Mark K. Friedberg
- Division of CardiologyLabatt Family Heart CentreHospital for Sick ChildrenUniversity of TorontoOntarioCanada
| | - Julien Aguet
- Department of Diagnostic ImagingHospital for Sick ChildrenUniversity of TorontoOntarioCanada
| | - Arnon Adler
- Division of CardiologyPeter Munk Cardiac CentreToronto General HospitalUniversity of TorontoOntarioCanada
| | - Christopher Z. Lam
- Division of CardiologyLabatt Family Heart CentreHospital for Sick ChildrenUniversity of TorontoOntarioCanada
| | - Andreea Dragulescu
- Division of CardiologyLabatt Family Heart CentreHospital for Sick ChildrenUniversity of TorontoOntarioCanada
| | - Harry Rakowski
- Division of CardiologyPeter Munk Cardiac CentreToronto General HospitalUniversity of TorontoOntarioCanada
| | - Olivier Villemain
- Division of CardiologyLabatt Family Heart CentreHospital for Sick ChildrenUniversity of TorontoOntarioCanada
| |
Collapse
|
11
|
Wang Z, Xia Q, Su W, Cao M, Sun Y, Zhang M, Chen W, Jiang T. Exploring the Communal Pathogenesis, Ferroptosis Mechanism, and Potential Therapeutic Targets of Dilated Cardiomyopathy and Hypertrophic Cardiomyopathy via a Microarray Data Analysis. Front Cardiovasc Med 2022; 9:824756. [PMID: 35282347 PMCID: PMC8907834 DOI: 10.3389/fcvm.2022.824756] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 01/20/2022] [Indexed: 12/13/2022] Open
Abstract
Background Cardiomyopathies are a heterogeneous group of heart diseases that can gradually cause severe heart failure. In particular, dilated cardiomyopathy (DCM) and hypertrophic cardiomyopathy (HCM) are the two main types of cardiomyopathies, yet the independent and communal biological mechanisms of both remain far from elucidated. Meanwhile, ferroptosis is a non-apoptotic form of cell death that has been proven to be associated with cardiomyopathies, but the concrete nature of the interaction remains unclear. Hence, this study explored the pathogenesis and ferroptosis mechanism of HCM and DCM via a bioinformatics analysis. Methods Six datasets were downloaded from the Gene Expression Omnibus (GEO) database based on the study inclusion/exclusion criteria. After screening the differentially expressed genes (DEGs) and hub genes of HCM and DCM, subsequent analyses, including functional annotation, co-expression, validation, and transcription factors (TF)–mRNA–microRNA (miRNA) regulatory network construction, were performed. In addition, ferroptosis-related DEGs were also identified and verified in HCM and DCM. Results We found 171 independent DEGs of HCM mainly enriched in the regulation of ERK1 and ERK2 cascade, while 171 independent DEGs of DCM were significantly involved in cell adhesion. Meanwhile, 32 communal DEGs (26 upregulated genes and 6 downregulated genes) and 3 hub genes [periostin (POSTN), insulin-like growth factor-binding protein-5 (IGFBP5), and fibromodulin (FMOD)] were determined to be shared between HCM and DCM and the functional annotation of these genes highlighted the important position of growth hormone in HCM and DCM. Moreover, we identified activating transcription factor 3 (ATF3), lysophosphatidylcholine acyltransferase 3 (LPCAT3), and solute carrier family 1 member 5 (SLC1A5) as ferroptosis-related genes in HCM and STAT3 as a ferroptosis-related gene in DCM. Conclusion The identified independent and communal DEGs contribute to uncover a potentially distinct and common mechanism of HCM and DCM and ferroptosis-related genes could provide us with a novel direction for exploration. In addition, 3 hub genes could be potential biomarkers or therapeutic targets in patients with cardiomyopathy.
Collapse
Affiliation(s)
- Zuoxiang Wang
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Department of Medicine, Soochow University, Suzhou, China
| | - Qingyue Xia
- Department of Dermatology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wenxing Su
- Department of Plastic and Burn Surgery, The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, China
| | - Mingqiang Cao
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yunjuan Sun
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Mingyang Zhang
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Department of Medicine, Soochow University, Suzhou, China
| | - Weixiang Chen
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, China
- *Correspondence: Weixiang Chen
| | - Tingbo Jiang
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Tingbo Jiang
| |
Collapse
|
12
|
Liu S, Chong W. Roles of LncRNAs in Regulating Mitochondrial Dysfunction in Septic Cardiomyopathy. Front Immunol 2021; 12:802085. [PMID: 34899764 PMCID: PMC8652231 DOI: 10.3389/fimmu.2021.802085] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 11/10/2021] [Indexed: 01/20/2023] Open
Abstract
Sepsis is an abnormal systemic inflammatory response of the host immune system to infection and can lead to fatal multiorgan dysfunction syndrome. Epidemiological studies have shown that approximately 10-70% of sepsis cases can lead to septic cardiomyopathy. Since the pathogenesis of septic cardiomyopathy is not clear, it is difficult for medical doctors to treat the disease. Therefore, finding effective interventions to prevent and reduce myocardial damage in septic cardiomyopathy is clinically significant. Epigenetics is the study of stable genetic phenotype inheritance that does not involve changing gene sequences. Epigenetic inheritance is affected by both gene and environmental regulation. Epigenetic studies focus on the modification and influence of chromatin structure, mainly including chromatin remodelling, DNA methylation, histone modification and noncoding RNA (ncRNA)-related mechanisms. Recently, long ncRNA (lncRNA)-related mechanisms have been the focus of epigenetic studies. LncRNAs are expected to become important targets to prevent, diagnose and treat human diseases. As the energy metabolism centre of cells, mitochondria are important targets in septic cardiomyopathy. Intervention measures to prevent and treat mitochondrial damage are of great significance for improving the prognosis of septic cardiomyopathy. LncRNAs play important roles in life activities. Recently, studies have focused on the involvement of lncRNAs in regulating mitochondrial dysfunction. However, few studies have revealed the involvement of lncRNAs in regulating mitochondrial dysfunction in septic cardiomyopathy. In this article, we briefly review recent research in this area.
Collapse
Affiliation(s)
- Shuang Liu
- Department of Emergency, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Wei Chong
- Department of Emergency, The First Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|