1
|
Vouloagkas I, Agbariah A, Zegkos T, Gossios TD, Tziomalos G, Parcharidou D, Didagelos M, Kamperidis V, Ziakas A, Efthimiadis GK. The many faces of SCN5A pathogenic variants: from channelopathy to cardiomyopathy. Heart Fail Rev 2024:10.1007/s10741-024-10459-x. [PMID: 39465469 DOI: 10.1007/s10741-024-10459-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/18/2024] [Indexed: 10/29/2024]
Abstract
The SCN5A gene encodes the alpha subunit of the cardiac sodium channel, which plays a fundamental role in the generation and propagation of the action potential in the heart muscle. During the past years our knowledge concerning the function of the cardiac sodium channel and the diseases caused by mutations of the SCN5A gene has grown. Although initially SCN5A pathogenic variants were mainly associated with channelopathies, increasing recent evidence suggests an association with structural heart disease in the form of heart muscle disease. The pathways leading to a cardiomyopathic phenotype remain unclear and require further elucidation. The aim of the present review is to provide a concise summary regarding the mechanisms through which SCN5A pathogenic variants result in heart disease, focusing in cardiomyopathy, highlighting along the way the complex role of the SCN5A gene at the intersection of cardiac excitability and contraction networks.
Collapse
Affiliation(s)
- Ioannis Vouloagkas
- Department of Medicine, Cantonal Hospital of Winterthur, Winterthur, Switzerland
| | - Andrea Agbariah
- Department of Cardiology, Università Degli Studi Di Verona, Verona, Italy
| | - Thomas Zegkos
- Cardiomyopathies Laboratory, 1st Aristotle University of Thessaloniki Cardiology Department, AHEPA University Hospital, Thessaloniki, Greece
| | - Thomas D Gossios
- Cardiomyopathies Laboratory, 1st Aristotle University of Thessaloniki Cardiology Department, AHEPA University Hospital, Thessaloniki, Greece.
| | - Georgios Tziomalos
- Cardiomyopathies Laboratory, 1st Aristotle University of Thessaloniki Cardiology Department, AHEPA University Hospital, Thessaloniki, Greece
| | - Despoina Parcharidou
- Cardiomyopathies Laboratory, 1st Aristotle University of Thessaloniki Cardiology Department, AHEPA University Hospital, Thessaloniki, Greece
| | - Matthaios Didagelos
- 1st Aristotle University of Thessaloniki Cardiology Department, AHEPA University Hospital, Thessaloniki, Greece
| | - Vasileios Kamperidis
- 1st Aristotle University of Thessaloniki Cardiology Department, AHEPA University Hospital, Thessaloniki, Greece
| | - Antonios Ziakas
- 1st Aristotle University of Thessaloniki Cardiology Department, AHEPA University Hospital, Thessaloniki, Greece
| | - Georgios K Efthimiadis
- Cardiomyopathies Laboratory, 1st Aristotle University of Thessaloniki Cardiology Department, AHEPA University Hospital, Thessaloniki, Greece
| |
Collapse
|
2
|
Foo B, Amedei H, Kaur S, Jaawan S, Boshnakovska A, Gall T, de Boer RA, Silljé HHW, Urlaub H, Rehling P, Lenz C, Lehnart SE. Unbiased complexome profiling and global proteomics analysis reveals mitochondrial impairment and potential changes at the intercalated disk in presymptomatic R14Δ/+ mice hearts. PLoS One 2024; 19:e0311203. [PMID: 39446877 PMCID: PMC11501035 DOI: 10.1371/journal.pone.0311203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 09/15/2024] [Indexed: 10/26/2024] Open
Abstract
Phospholamban (PLN) is a sarco-endoplasmic reticulum (SER) membrane protein that regulates cardiac contraction/relaxation by reversibly inhibiting the SERCA2a Ca2+-reuptake pump. The R14Δ-PLN mutation causes severe cardiomyopathy that is resistant to conventional treatment. Protein complexes and higher-order supercomplexes such as intercalated disk components and Ca+2-cycling domains underlie many critical cardiac functions, a subset of which may be disrupted by R14Δ-PLN. Complexome profiling (CP) is a proteomics workflow for systematic analysis of high molecular weight (MW) protein complexes and supercomplexes. We hypothesize that R14Δ-PLN may alter a subset of these assemblies, and apply CP workflows to explore these changes in presymptomatic R14Δ/+ mice hearts. Ventricular tissues from presymptomatic 28wk-old WT and R14Δ/+ mice were homogenized under non-denaturing conditions, fractionated by size-exclusion chromatography (SEC) with a linear MW-range exceeding 5 MDa, and subjected to quantitative data-independent acquisition mass spectrometry (DIA-MS) analysis. Unfortunately, current workflows for the systematic analysis of CP data proved ill-suited for use in cardiac samples. Most rely upon curated protein complex databases to provide ground-truth for analysis; however, these are derived primarily from cancerous or immortalized cell lines and, consequently, cell-type specific complexes (including cardiac-specific machinery potentially affected in R14Δ-PLN hearts) are poorly covered. We thus developed PERCOM: a novel CP data-analysis strategy that does not rely upon these databases and can, furthermore, be implemented on widely available spreadsheet software. Applying PERCOM to our CP dataset resulted in the identification of 296 proteins with disrupted elution profiles. Hits were significantly enriched for mitochondrial and intercalated disk (ICD) supercomplex components. Changes to mitochondrial supercomplexes were associated with reduced expression of mitochondrial proteins and maximal oxygen consumption rate. The observed alterations to mitochondrial and ICD supercomplexes were replicated in a second cohort of "juvenile" 9wk-old mice. These early-stage changes to key cardiac machinery may contribute to R14Δ-PLN pathogenesis.
Collapse
Affiliation(s)
- Brian Foo
- Department of Cardiology and Pneumology, Heart Research Center Göttingen, Cellular Biophysics and Translational Cardiology Section, University Medical Center Göttingen, Göttingen, Germany
- Cluster of Excellence “Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells” (MBExC), University of Göttingen, Göttingen, Germany
| | - Hugo Amedei
- Department of Clinical Chemistry, University Medical Center Göttingen, Göttingen, Germany
| | - Surmeet Kaur
- Department of Clinical Chemistry, University Medical Center Göttingen, Göttingen, Germany
| | - Samir Jaawan
- Department of Cardiology and Pneumology, Heart Research Center Göttingen, Cellular Biophysics and Translational Cardiology Section, University Medical Center Göttingen, Göttingen, Germany
- Cluster of Excellence “Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells” (MBExC), University of Göttingen, Göttingen, Germany
| | - Angela Boshnakovska
- Department of Cellular Biochemistry, University Medical Center Göttingen, Göttingen, Germany
| | - Tanja Gall
- Department of Cellular Biochemistry, University Medical Center Göttingen, Göttingen, Germany
| | - Rudolf A. de Boer
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
- Department of Cardiology, Erasmus MC, Thorax Center, Cardiovascular Institute, Rotterdam, the Netherlands
| | - Herman H. W. Silljé
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Henning Urlaub
- Cluster of Excellence “Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells” (MBExC), University of Göttingen, Göttingen, Germany
- Department of Clinical Chemistry, University Medical Center Göttingen, Göttingen, Germany
- Bioanalytical Mass Spectrometry Group, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Peter Rehling
- Department of Cellular Biochemistry, University Medical Center Göttingen, Göttingen, Germany
| | - Christof Lenz
- Cluster of Excellence “Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells” (MBExC), University of Göttingen, Göttingen, Germany
- Department of Clinical Chemistry, University Medical Center Göttingen, Göttingen, Germany
- Bioanalytical Mass Spectrometry Group, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Stephan E. Lehnart
- Department of Cardiology and Pneumology, Heart Research Center Göttingen, Cellular Biophysics and Translational Cardiology Section, University Medical Center Göttingen, Göttingen, Germany
- Cluster of Excellence “Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells” (MBExC), University of Göttingen, Göttingen, Germany
| |
Collapse
|
3
|
Sommerfeld LC, Holmes AP, Yu TY, O'Shea C, Kavanagh DM, Pike JM, Wright T, Syeda F, Aljehani A, Kew T, Cardoso VR, Kabir SN, Hepburn C, Menon PR, Broadway-Stringer S, O'Reilly M, Witten A, Fortmueller L, Lutz S, Kulle A, Gkoutos GV, Pavlovic D, Arlt W, Lavery GG, Steeds R, Gehmlich K, Stoll M, Kirchhof P, Fabritz L. Reduced plakoglobin increases the risk of sodium current defects and atrial conduction abnormalities in response to androgenic anabolic steroid abuse. J Physiol 2024; 602:4409-4436. [PMID: 38345865 DOI: 10.1113/jp284597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 01/16/2024] [Indexed: 03/07/2024] Open
Abstract
Androgenic anabolic steroids (AAS) are commonly abused by young men. Male sex and increased AAS levels are associated with earlier and more severe manifestation of common cardiac conditions, such as atrial fibrillation, and rare ones, such as arrhythmogenic right ventricular cardiomyopathy (ARVC). Clinical observations suggest a potential atrial involvement in ARVC. Arrhythmogenic right ventricular cardiomyopathy is caused by desmosomal gene defects, including reduced plakoglobin expression. Here, we analysed clinical records from 146 ARVC patients to identify that ARVC is more common in males than females. Patients with ARVC also had an increased incidence of atrial arrhythmias and P wave changes. To study desmosomal vulnerability and the effects of AAS on the atria, young adult male mice, heterozygously deficient for plakoglobin (Plako+/-), and wild type (WT) littermates were chronically exposed to 5α-dihydrotestosterone (DHT) or placebo. The DHT increased atrial expression of pro-hypertrophic, fibrotic and inflammatory transcripts. In mice with reduced plakoglobin, DHT exaggerated P wave abnormalities, atrial conduction slowing, sodium current depletion, action potential amplitude reduction and the fall in action potential depolarization rate. Super-resolution microscopy revealed a decrease in NaV1.5 membrane clustering in Plako+/- atrial cardiomyocytes after DHT exposure. In summary, AAS combined with plakoglobin deficiency cause pathological atrial electrical remodelling in young male hearts. Male sex is likely to increase the risk of atrial arrhythmia, particularly in those with desmosomal gene variants. This risk is likely to be exaggerated further by AAS use. KEY POINTS: Androgenic male sex hormones, such as testosterone, might increase the risk of atrial fibrillation in patients with arrhythmogenic right ventricular cardiomyopathy (ARVC), which is often caused by desmosomal gene defects (e.g. reduced plakoglobin expression). In this study, we observed a significantly higher proportion of males who had ARVC compared with females, and atrial arrhythmias and P wave changes represented a common observation in advanced ARVC stages. In mice with reduced plakoglobin expression, chronic administration of 5α-dihydrotestosterone led to P wave abnormalities, atrial conduction slowing, sodium current depletion and a decrease in membrane-localized NaV1.5 clusters. 5α-Dihydrotestosterone, therefore, represents a stimulus aggravating the pro-arrhythmic phenotype in carriers of desmosomal mutations and can affect atrial electrical function.
Collapse
Affiliation(s)
- Laura C Sommerfeld
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
- University Center of Cardiovascular Science, University Heart and Vascular Center, UKE Hamburg, Hamburg, Germany
- German Center for Cardiovascular Research (DZHK), Standort Hamburg/Kiel/Lübeck, Germany
| | - Andrew P Holmes
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
- School of Biomedical Sciences, Institute of Clinical Sciences, University of Birmingham, Birmingham, UK
| | - Ting Y Yu
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
- Research and Training Centre in Physical Sciences for Health, Birmingham, UK
| | - Christopher O'Shea
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
- Research and Training Centre in Physical Sciences for Health, Birmingham, UK
| | - Deirdre M Kavanagh
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
- Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham, Birmingham, UK
| | - Jeremy M Pike
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
- Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham, Birmingham, UK
| | - Thomas Wright
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
| | - Fahima Syeda
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
| | - Areej Aljehani
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
| | - Tania Kew
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
| | - Victor R Cardoso
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - S Nashitha Kabir
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
| | - Claire Hepburn
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
| | - Priyanka R Menon
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
| | | | - Molly O'Reilly
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
| | - Anika Witten
- Genetic Epidemiology, Institute for Human Genetics, University of Münster, Münster, Germany
- Core Facility Genomics of the Medical Faculty, University of Münster, Münster, Germany
| | - Lisa Fortmueller
- University Center of Cardiovascular Science, University Heart and Vascular Center, UKE Hamburg, Hamburg, Germany
- German Center for Cardiovascular Research (DZHK), Standort Hamburg/Kiel/Lübeck, Germany
- Genetic Epidemiology, Institute for Human Genetics, University of Münster, Münster, Germany
| | - Susanne Lutz
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Göttingen, Germany
| | - Alexandra Kulle
- Division of Paediatric Endocrinology and Diabetes, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Georgios V Gkoutos
- University Center of Cardiovascular Science, University Heart and Vascular Center, UKE Hamburg, Hamburg, Germany
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
- Institute of Translational Medicine, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
- MRC Health Data Research UK (HDR), Midlands Site, UK
| | - Davor Pavlovic
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
| | - Wiebke Arlt
- Institute of Metabolism and Systems Research (IMSR), University of Birmingham, Birmingham, UK
- Centre for Endocrinology, Diabetes and Metabolism (CEDAM), Birmingham Health Partners, Birmingham, UK
- Medical Research Council London Institute of Medical Sciences, London UK & Institute of Clinical Sciences, Faculty of Medicine, Imperial College, London, UK
| | - Gareth G Lavery
- Institute of Metabolism and Systems Research (IMSR), University of Birmingham, Birmingham, UK
- Centre for Endocrinology, Diabetes and Metabolism (CEDAM), Birmingham Health Partners, Birmingham, UK
| | - Richard Steeds
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
- Department of Cardiology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Katja Gehmlich
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
| | - Monika Stoll
- Genetic Epidemiology, Institute for Human Genetics, University of Münster, Münster, Germany
- Core Facility Genomics of the Medical Faculty, University of Münster, Münster, Germany
- Cardiovascular Research Institute Maastricht, Department of Biochemistry, Maastricht University, Maastricht, The Netherlands
| | - Paulus Kirchhof
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
- German Center for Cardiovascular Research (DZHK), Standort Hamburg/Kiel/Lübeck, Germany
- Department of Cardiology, University Heart & Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Larissa Fabritz
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
- University Center of Cardiovascular Science, University Heart and Vascular Center, UKE Hamburg, Hamburg, Germany
- German Center for Cardiovascular Research (DZHK), Standort Hamburg/Kiel/Lübeck, Germany
- Department of Cardiology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
- Department of Cardiology, University Heart & Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
4
|
Nguyen Tat T, Lien NTK, Luu Sy H, Ta Van T, Dang Viet D, Nguyen Thi H, Tung NV, Thanh LT, Xuan NT, Hoang NH. Identifying the Pathogenic Variants in Heart Genes in Vietnamese Sudden Unexplained Death Victims by Next-Generation Sequencing. Diagnostics (Basel) 2024; 14:1876. [PMID: 39272661 PMCID: PMC11394071 DOI: 10.3390/diagnostics14171876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 08/16/2024] [Accepted: 08/17/2024] [Indexed: 09/15/2024] Open
Abstract
In forensics, one-third of sudden deaths remain unexplained after a forensic autopsy. A majority of these sudden unexplained deaths (SUDs) are considered to be caused by inherited cardiovascular diseases. In this study, we investigated 40 young SUD cases (<40 years), with non-diagnostic structural cardiac abnormalities, using Targeted NGS (next-generation sequencing) for 167 genes previously associated with inherited cardiomyopathies and channelopathies. Fifteen cases identified 17 variants on related genes including the following: AKAP9, CSRP3, GSN, HTRA1, KCNA5, LAMA4, MYBPC3, MYH6, MYLK, RYR2, SCN5A, SCN10A, SLC4A3, TNNI3, TNNI3K, and TNNT2. Of these, eight variants were novel, and nine variants were reported in the ClinVar database. Five were determined to be pathogenic and four were not evaluated. The novel and unevaluated variants were predicted by using in silico tools, which revealed that four novel variants (c.5187_5188dup, p.Arg1730llefsTer4 in the AKAP9 gene; c.1454A>T, p.Lys485Met in the MYH6 gene; c.2535+1G>A in the SLC4A3 gene; and c.10498G>T, p.Asp3500Tyr in the RYR2 gene) were pathogenic and three variants (c.292C>G, p.Arg98Gly in the TNNI3 gene; c.683C>A, p.Pro228His in the KCN5A gene; and c.2275G>A, p.Glu759Lys in the MYBPC3 gene) still need to be further verified experimentally. The results of our study contributed to the general understanding of the causes of SUDs. They provided a scientific basis for screening the risk of sudden death in family members of victims. They also suggested that the Targeted NGS method may be used to identify the pathogenic variants in SUD victims.
Collapse
Affiliation(s)
- Tho Nguyen Tat
- Department of Forensic Medicine, Hanoi Medical University, 1 Ton That Tung Str., Dongda, Hanoi 100000, Vietnam
| | - Nguyen Thi Kim Lien
- Institute of Genome Research, Vietnam Academy of Science and Technology, 18-Hoang Quoc Viet Str., Caugiay, Hanoi 100000, Vietnam
| | - Hung Luu Sy
- Department of Forensic Medicine, Hanoi Medical University, 1 Ton That Tung Str., Dongda, Hanoi 100000, Vietnam
| | - To Ta Van
- Department of Pathology, National Cancer Hospital, 43 Quan Su Str., Hoan Kiem, Hanoi 100000, Vietnam
| | - Duc Dang Viet
- Cardiovascular Intensive Care Unit, Heart Institute, 108 Military Central Hospital, 1B Tran Hung Dao Str., Hai Ba Trung, Hanoi 100000, Vietnam
| | - Hoa Nguyen Thi
- Faculty of Biotechnology, Graduate University of Science and Technology, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet Str., Caugiay, Hanoi 100000, Vietnam
| | - Nguyen Van Tung
- Institute of Genome Research, Vietnam Academy of Science and Technology, 18-Hoang Quoc Viet Str., Caugiay, Hanoi 100000, Vietnam
- Faculty of Biotechnology, Graduate University of Science and Technology, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet Str., Caugiay, Hanoi 100000, Vietnam
| | - Le Tat Thanh
- Institute of Genome Research, Vietnam Academy of Science and Technology, 18-Hoang Quoc Viet Str., Caugiay, Hanoi 100000, Vietnam
| | - Nguyen Thi Xuan
- Institute of Genome Research, Vietnam Academy of Science and Technology, 18-Hoang Quoc Viet Str., Caugiay, Hanoi 100000, Vietnam
| | - Nguyen Huy Hoang
- Institute of Genome Research, Vietnam Academy of Science and Technology, 18-Hoang Quoc Viet Str., Caugiay, Hanoi 100000, Vietnam
- Faculty of Biotechnology, Graduate University of Science and Technology, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet Str., Caugiay, Hanoi 100000, Vietnam
| |
Collapse
|
5
|
Vanaja IP, Scalco A, Ronfini M, Bona AD, Olianti C, Rizzo S, Chelko SP, Corrado D, Sacconi L, Basso C, Mongillo M, Zaglia T. Cardiac sympathetic neurons are additional cells affected in genetically determined arrhythmogenic cardiomyopathy. J Physiol 2024. [PMID: 39141822 DOI: 10.1113/jp286845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 07/05/2024] [Indexed: 08/16/2024] Open
Abstract
Arrhythmogenic cardiomyopathy (AC) is a familial cardiac disease, mainly caused by mutations in desmosomal genes, which accounts for most cases of stress-related arrhythmic sudden death, in young and athletes. AC hearts display fibro-fatty lesions that generate the arrhythmic substrate and cause contractile dysfunction. A correlation between physical/emotional stresses and arrhythmias supports the involvement of sympathetic neurons (SNs) in the disease, but this has not been confirmed previously. Here, we combined molecular, in vitro and ex vivo analyses to determine the role of AC-linked DSG2 downregulation on SN biology and assess cardiac sympathetic innervation in desmoglein-2 mutant (Dsg2mut/mut) mice. Molecular assays showed that SNs express DSG2, implying that DSG2-mutation carriers would harbour the mutant protein in SNs. Confocal immunofluorescence of heart sections and 3-D reconstruction of SN network in clarified heart blocks revealed significant changes in the physiologialc SN topology, with massive hyperinnervation of the intact subepicardial layers and heterogeneous distribution of neurons in fibrotic areas. Cardiac SNs isolated from Dsg2mut/mut neonatal mice, prior to the establishment of cardiac innervation, show alterations in axonal sprouting, process development and distribution of varicosities. Consistently, virus-assisted DSG2 downregulation replicated, in PC12-derived SNs, the phenotypic alterations displayed by Dsg2mut/mut primary neurons, corroborating that AC-linked Dsg2 variants may affect SNs. Our results reveal that altered sympathetic innervation is an unrecognized feature of AC hearts, which may result from the combination of cell-autonomous and context-dependent factors implicated in myocardial remodelling. Our results favour the concept that AC is a disease of multiple cell types also hitting cardiac SNs. KEY POINTS: Arrhythmogenic cardiomyopathy is a genetically determined cardiac disease, which accounts for most cases of stress-related arrhythmic sudden death. Arrhythmogenic cardiomyopathy linked to mutations in desmoglein-2 (DSG2) is frequent and leads to a left-dominant form of the disease. Arrhythmogenic cardiomyopathy has been approached thus far as a disease of cardiomyocytes, but we here unveil that DSG2 is expressed, in addition to cardiomyocytes, by cardiac and extracardiac sympathetic neurons, although not organized into desmosomes. AC-linked DSG2 downregulation primarily affect sympathetic neurons, resulting in the significant increase in cardiac innervation density, accompanied by alterations in sympathetic neuron distribution. Our data supports the notion that AC develops with the contribution of several 'desmosomal protein-carrying' cell types and systems.
Collapse
Affiliation(s)
- Induja Perumal Vanaja
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
- Veneto Institute of Molecular Medicine (VIMM), Padova, Italy
| | - Arianna Scalco
- Veneto Institute of Molecular Medicine (VIMM), Padova, Italy
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Marco Ronfini
- Veneto Institute of Molecular Medicine (VIMM), Padova, Italy
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Anna Di Bona
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
- Veneto Institute of Molecular Medicine (VIMM), Padova, Italy
| | - Camilla Olianti
- Institute of Clinical Physiology (IFC), National Research Council, Florence, Florence, Italy
| | - Stefania Rizzo
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Stephen P Chelko
- Department of Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
- Department of Biomedical Sciences, Florida State University, College of Medicine, Tallahassee, FL, USA
| | - Domenico Corrado
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Leonardo Sacconi
- Institute of Clinical Physiology (IFC), National Research Council, Florence, Florence, Italy
- Institute for Experimental Cardiovascular Medicine, University Heart Center and Medical Faculty, University of Freiburg, Freiburg, Germany
| | - Cristina Basso
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Marco Mongillo
- Veneto Institute of Molecular Medicine (VIMM), Padova, Italy
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Tania Zaglia
- Veneto Institute of Molecular Medicine (VIMM), Padova, Italy
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| |
Collapse
|
6
|
Risato G, Brañas Casas R, Cason M, Bueno Marinas M, Pinci S, De Gaspari M, Visentin S, Rizzo S, Thiene G, Basso C, Pilichou K, Tiso N, Celeghin R. In Vivo Approaches to Understand Arrhythmogenic Cardiomyopathy: Perspectives on Animal Models. Cells 2024; 13:1264. [PMID: 39120296 PMCID: PMC11311808 DOI: 10.3390/cells13151264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 07/23/2024] [Accepted: 07/24/2024] [Indexed: 08/10/2024] Open
Abstract
Arrhythmogenic cardiomyopathy (AC) is a hereditary cardiac disorder characterized by the gradual replacement of cardiomyocytes with fibrous and adipose tissue, leading to ventricular wall thinning, chamber dilation, arrhythmias, and sudden cardiac death. Despite advances in treatment, disease management remains challenging. Animal models, particularly mice and zebrafish, have become invaluable tools for understanding AC's pathophysiology and testing potential therapies. Mice models, although useful for scientific research, cannot fully replicate the complexity of the human AC. However, they have provided valuable insights into gene involvement, signalling pathways, and disease progression. Zebrafish offer a promising alternative to mammalian models, despite the phylogenetic distance, due to their economic and genetic advantages. By combining animal models with in vitro studies, researchers can comprehensively understand AC, paving the way for more effective treatments and interventions for patients and improving their quality of life and prognosis.
Collapse
Affiliation(s)
- Giovanni Risato
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, I-35128 Padua, Italy; (G.R.); (M.C.); (M.B.M.); (S.P.); (M.D.G.); (S.R.); (G.T.); (C.B.); (K.P.); (R.C.)
- Department of Biology, University of Padua, I-35131 Padua, Italy;
- Department of Women’s and Children’s Health, University of Padua, I-35128 Padua, Italy;
| | | | - Marco Cason
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, I-35128 Padua, Italy; (G.R.); (M.C.); (M.B.M.); (S.P.); (M.D.G.); (S.R.); (G.T.); (C.B.); (K.P.); (R.C.)
| | - Maria Bueno Marinas
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, I-35128 Padua, Italy; (G.R.); (M.C.); (M.B.M.); (S.P.); (M.D.G.); (S.R.); (G.T.); (C.B.); (K.P.); (R.C.)
| | - Serena Pinci
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, I-35128 Padua, Italy; (G.R.); (M.C.); (M.B.M.); (S.P.); (M.D.G.); (S.R.); (G.T.); (C.B.); (K.P.); (R.C.)
| | - Monica De Gaspari
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, I-35128 Padua, Italy; (G.R.); (M.C.); (M.B.M.); (S.P.); (M.D.G.); (S.R.); (G.T.); (C.B.); (K.P.); (R.C.)
| | - Silvia Visentin
- Department of Women’s and Children’s Health, University of Padua, I-35128 Padua, Italy;
| | - Stefania Rizzo
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, I-35128 Padua, Italy; (G.R.); (M.C.); (M.B.M.); (S.P.); (M.D.G.); (S.R.); (G.T.); (C.B.); (K.P.); (R.C.)
| | - Gaetano Thiene
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, I-35128 Padua, Italy; (G.R.); (M.C.); (M.B.M.); (S.P.); (M.D.G.); (S.R.); (G.T.); (C.B.); (K.P.); (R.C.)
| | - Cristina Basso
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, I-35128 Padua, Italy; (G.R.); (M.C.); (M.B.M.); (S.P.); (M.D.G.); (S.R.); (G.T.); (C.B.); (K.P.); (R.C.)
| | - Kalliopi Pilichou
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, I-35128 Padua, Italy; (G.R.); (M.C.); (M.B.M.); (S.P.); (M.D.G.); (S.R.); (G.T.); (C.B.); (K.P.); (R.C.)
| | - Natascia Tiso
- Department of Biology, University of Padua, I-35131 Padua, Italy;
| | - Rudy Celeghin
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, I-35128 Padua, Italy; (G.R.); (M.C.); (M.B.M.); (S.P.); (M.D.G.); (S.R.); (G.T.); (C.B.); (K.P.); (R.C.)
| |
Collapse
|
7
|
Vencato S, Romanato C, Rampazzo A, Calore M. Animal Models and Molecular Pathogenesis of Arrhythmogenic Cardiomyopathy Associated with Pathogenic Variants in Intercalated Disc Genes. Int J Mol Sci 2024; 25:6208. [PMID: 38892395 PMCID: PMC11172742 DOI: 10.3390/ijms25116208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 05/28/2024] [Accepted: 06/01/2024] [Indexed: 06/21/2024] Open
Abstract
Arrhythmogenic cardiomyopathy (ACM) is a rare genetic cardiac disease characterized by the progressive substitution of myocardium with fibro-fatty tissue. Clinically, ACM shows wide variability among patients; symptoms can include syncope and ventricular tachycardia but also sudden death, with the latter often being its sole manifestation. Approximately half of ACM patients have been found with variations in one or more genes encoding cardiac intercalated discs proteins; the most involved genes are plakophilin 2 (PKP2), desmoglein 2 (DSG2), and desmoplakin (DSP). Cardiac intercalated discs provide mechanical and electro-metabolic coupling among cardiomyocytes. Mechanical communication is guaranteed by the interaction of proteins of desmosomes and adheren junctions in the so-called area composita, whereas electro-metabolic coupling between adjacent cardiac cells depends on gap junctions. Although ACM has been first described almost thirty years ago, the pathogenic mechanism(s) leading to its development are still only partially known. Several studies with different animal models point to the involvement of the Wnt/β-catenin signaling in combination with the Hippo pathway. Here, we present an overview about the existing murine models of ACM harboring variants in intercalated disc components with a particular focus on the underlying pathogenic mechanisms. Prospectively, mechanistic insights into the disease pathogenesis will lead to the development of effective targeted therapies for ACM.
Collapse
Affiliation(s)
- Sara Vencato
- Department of Biology, University of Padova, Via U. Bassi 58/B, 35121 Padova, Italy; (S.V.); (C.R.); (A.R.)
| | - Chiara Romanato
- Department of Biology, University of Padova, Via U. Bassi 58/B, 35121 Padova, Italy; (S.V.); (C.R.); (A.R.)
| | - Alessandra Rampazzo
- Department of Biology, University of Padova, Via U. Bassi 58/B, 35121 Padova, Italy; (S.V.); (C.R.); (A.R.)
| | - Martina Calore
- Department of Biology, University of Padova, Via U. Bassi 58/B, 35121 Padova, Italy; (S.V.); (C.R.); (A.R.)
- Department of Molecular Genetics, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6211 LK Maastricht, The Netherlands
| |
Collapse
|
8
|
Stevens TL, Coles S, Sturm AC, Hoover CA, Borzok MA, Mohler PJ, El Refaey M. Molecular Pathways and Animal Models of Arrhythmias. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1441:1057-1090. [PMID: 38884769 DOI: 10.1007/978-3-031-44087-8_67] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
Arrhythmias account for over 300,000 annual deaths in the United States, and approximately half of all deaths are associated with heart disease. Mechanisms underlying arrhythmia risk are complex; however, work in humans and animal models over the past 25 years has identified a host of molecular pathways linked with both arrhythmia substrates and triggers. This chapter will focus on select arrhythmia pathways solved by linking human clinical and genetic data with animal models.
Collapse
Affiliation(s)
- Tyler L Stevens
- The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Sara Coles
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Amy C Sturm
- Genomic Medicine Institute, 23andMe, Sunnyvale, CA, USA
| | - Catherine A Hoover
- Department of Biochemistry, Chemistry, Engineering and Physics, Commonwealth University of Pennsylvania, Mansfield, PA, USA
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, USA
| | - Maegen A Borzok
- Department of Biochemistry, Chemistry, Engineering and Physics, Commonwealth University of Pennsylvania, Mansfield, PA, USA
| | - Peter J Mohler
- The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- Department of Internal Medicine, Division of Cardiovascular Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Mona El Refaey
- The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
- Department of Surgery, Division of Cardiac Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
| |
Collapse
|
9
|
Struckman HL, Moise N, King DR, Soltisz A, Buxton A, Dunlap I, Chen Z, Radwański PB, Weinberg SH, Veeraraghavan R. Unraveling Impacts of Chamber-Specific Differences in Intercalated Disc Ultrastructure and Molecular Organization on Cardiac Conduction. JACC Clin Electrophysiol 2023; 9:2425-2443. [PMID: 37498248 PMCID: PMC11102000 DOI: 10.1016/j.jacep.2023.05.042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 05/26/2023] [Accepted: 05/30/2023] [Indexed: 07/28/2023]
Abstract
BACKGROUND Propagation of action potentials through the heart coordinates the heartbeat. Thus, intercalated discs, specialized cell-cell contact sites that provide electrical and mechanical coupling between cardiomyocytes, are an important target for study. Impaired propagation leads to arrhythmias in many pathologies, where intercalated disc remodeling is a common finding, hence the importance and urgency of understanding propagation dependence on intercalated disc structure. Conventional modeling approaches cannot predict changes in propagation elicited by perturbations that alter intercalated disc ultrastructure or molecular organization, because of lack of quantitative structural data at subcellular through nano scales. OBJECTIVES This study sought to quantify intercalated disc structure at these spatial scales in the healthy adult mouse heart and relate them to chamber-specific properties of propagation as a precursor to understanding the effects of pathological intercalated disc remodeling. METHODS Using super-resolution light microscopy, electron microscopy, and computational image analysis, we provide here the first ever systematic, multiscale quantification of intercalated disc ultrastructure and molecular organization. RESULTS By incorporating these data into a rule-based model of cardiac tissue with realistic intercalated disc structure, and comparing model predictions of electrical propagation with experimental measures of conduction velocity, we reveal that atrial intercalated discs can support faster conduction than their ventricular counterparts, which is normally masked by interchamber differences in myocyte geometry. Further, we identify key ultrastructural and molecular organization features underpinning the ability of atrial intercalated discs to support faster conduction. CONCLUSIONS These data provide the first stepping stone to elucidating chamber-specific effects of pathological intercalated disc remodeling, as occurs in many arrhythmic diseases.
Collapse
Affiliation(s)
- Heather L Struckman
- Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, Ohio, USA; The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Nicolae Moise
- The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - D Ryan King
- The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Andrew Soltisz
- Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, Ohio, USA; The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Andrew Buxton
- Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, Ohio, USA
| | - Izabella Dunlap
- The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Zhenhui Chen
- Krannert Cardiovascular Research Center, Department of Medicine, Indiana University, Indianapolis, Indiana, USA
| | - Przemysław B Radwański
- The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA; Division of Outcomes and Translational Sciences, College of Pharmacy, The Ohio State University, Columbus, Ohio, USA
| | - Seth H Weinberg
- Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, Ohio, USA; The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Rengasayee Veeraraghavan
- Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, Ohio, USA; The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA.
| |
Collapse
|
10
|
Fan X, Yang G, Duru F, Grilli M, Akin I, Zhou X, Saguner AM, Ei-Battrawy I. Arrhythmogenic Cardiomyopathy: from Preclinical Models to Genotype-phenotype Correlation and Pathophysiology. Stem Cell Rev Rep 2023; 19:2683-2708. [PMID: 37731079 PMCID: PMC10661732 DOI: 10.1007/s12015-023-10615-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/23/2023] [Indexed: 09/22/2023]
Abstract
Arrhythmogenic cardiomyopathy (ACM) is a hereditary myocardial disease characterized by the replacement of the ventricular myocardium with fibrous fatty deposits. ACM is usually inherited in an autosomal dominant pattern with variable penetrance and expressivity, which is mainly related to ventricular tachyarrhythmia and sudden cardiac death (SCD). Importantly, significant progress has been made in determining the genetic background of ACM due to the development of new techniques for genetic analysis. The exact molecular pathomechanism of ACM, however, is not completely clear and the genotype-phenotype correlations have not been fully elucidated, which are useful to predict the prognosis and treatment of ACM patients. Different gene-targeted and transgenic animal models, human-induced pluripotent stem cell-derived cardiomyocyte (hiPSC-CM) models, and heterologous expression systems have been developed. Here, this review aims to summarize preclinical ACM models and platforms promoting our understanding of the pathogenesis of ACM and assess their value in elucidating the ACM genotype-phenotype relationship.
Collapse
Affiliation(s)
- Xuehui Fan
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention of Cardiovascular Diseases, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, China
- Cardiology, Angiology, Haemostaseology, and Medical Intensive Care, Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
- European Center for AngioScience (ECAS), German Center for Cardiovascular Research (DZHK) Partner Site Heidelberg/ Mannheim, and Centre for Cardiovascular Acute Medicine Mannheim (ZKAM), Medical Centre Mannheim, Heidelberg University, Partner Site, Heidelberg-Mannheim, Germany
| | - Guoqiang Yang
- Cardiology, Angiology, Haemostaseology, and Medical Intensive Care, Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
- Department of Acupuncture and Rehabilitation, the Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
- Research Unit of Molecular Imaging Probes, Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Firat Duru
- Department of Cardiology, University Heart Centre, University Hospital Zurich, Zurich, Switzerland
| | - Maurizio Grilli
- Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany
| | - Ibrahim Akin
- Cardiology, Angiology, Haemostaseology, and Medical Intensive Care, Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
- European Center for AngioScience (ECAS), German Center for Cardiovascular Research (DZHK) Partner Site Heidelberg/ Mannheim, and Centre for Cardiovascular Acute Medicine Mannheim (ZKAM), Medical Centre Mannheim, Heidelberg University, Partner Site, Heidelberg-Mannheim, Germany
| | - Xiaobo Zhou
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention of Cardiovascular Diseases, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, China.
- Cardiology, Angiology, Haemostaseology, and Medical Intensive Care, Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany.
- European Center for AngioScience (ECAS), German Center for Cardiovascular Research (DZHK) Partner Site Heidelberg/ Mannheim, and Centre for Cardiovascular Acute Medicine Mannheim (ZKAM), Medical Centre Mannheim, Heidelberg University, Partner Site, Heidelberg-Mannheim, Germany.
- First Department of Medicine, University Medical Centre Mannheim, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany.
| | - Ardan Muammer Saguner
- Department of Cardiology, University Heart Centre, University Hospital Zurich, Zurich, Switzerland
| | - Ibrahim Ei-Battrawy
- European Center for AngioScience (ECAS), German Center for Cardiovascular Research (DZHK) Partner Site Heidelberg/ Mannheim, and Centre for Cardiovascular Acute Medicine Mannheim (ZKAM), Medical Centre Mannheim, Heidelberg University, Partner Site, Heidelberg-Mannheim, Germany.
- Department of Cardiology and Angiology, Ruhr University, Bochum, Germany; Institute of Physiology, Department of Cellular and Translational Physiology and Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr- University Bochum, Bochum, Germany.
| |
Collapse
|
11
|
Zhang B, Wu Y, Yang X, Xiang Y, Yang B. Molecular insight into arrhythmogenic cardiomyopathy caused by DSG2 mutations. Biomed Pharmacother 2023; 167:115448. [PMID: 37696084 DOI: 10.1016/j.biopha.2023.115448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 09/01/2023] [Accepted: 09/04/2023] [Indexed: 09/13/2023] Open
Abstract
Mutant desmoglein 2 (DSG2) is the second most common pathogenic gene in arrhythmogenic cardiomyopathy (ACM), accounting for approximately 10% of ACM cases. In addition to common clinical and pathological features, ACM caused by mutant DSG2 has specific characteristics, manifesting as left ventricle involvement and a high risk of heart failure. Pathological studies have shown extensive cardiomyocyte necrosis, infiltration of immune cells, and fibrofatty replacement in both ventricles, as well as abnormal desmosome structures in the hearts of humans and mice with mutant DSG2-related ACM. Although desmosome dysfunction is a common pathway in the pathogenesis of mutant DSG2-related ACM, the mechanisms underlying this dysfunction vary among mutations. Desmosome dysfunction induces cardiomyocyte injury, plakoglobin dislocation, and gap junction dysfunction, all of which contribute to the initiation and progression of ACM. Additionally, dysregulated inflammation, overactivation of transforming growth factor-beta-1 signaling and endoplasmic reticulum stress, and cardiac metabolic dysfunction contribute to the pathogenesis of ACM caused by mutant DSG2. These features demonstrate that patients with mutant DSG2-related ACM should be managed individually and precisely based on the genotype and phenotype. Further studies are needed to investigate the underlying mechanisms and to identify novel therapies to reverse or attenuate the progression of ACM caused by mutant DSG2.
Collapse
Affiliation(s)
- Baowei Zhang
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Pudong, Shanghai 200120, PR China
| | - Yizhang Wu
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Pudong, Shanghai 200120, PR China
| | - Xingbo Yang
- Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, 150 Jimo Road, Pudong, Shanghai 200120, PR China
| | - Yaozu Xiang
- Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, 150 Jimo Road, Pudong, Shanghai 200120, PR China.
| | - Bing Yang
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Pudong, Shanghai 200120, PR China.
| |
Collapse
|
12
|
Ng R, Gokhan I, Stankey P, Akar FG, Campbell SG. Chronic diastolic stretch unmasks conduction defects in an in vitro model of arrhythmogenic cardiomyopathy. Am J Physiol Heart Circ Physiol 2023; 325:H1373-H1385. [PMID: 37830983 PMCID: PMC10977872 DOI: 10.1152/ajpheart.00709.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 10/05/2023] [Accepted: 10/06/2023] [Indexed: 10/14/2023]
Abstract
We seek to elucidate the precise nature of mechanical loading that precipitates conduction deficits in a concealed-phase model of arrhythmogenic cardiomyopathy (ACM). ACM is a progressive disorder often resulting from mutations in desmosomal proteins. Exercise has been shown to worsen disease progression and unmask arrhythmia vulnerability, yet the underlying pathomechanisms may depend on the type and intensity of exercise. Because exercise causes myriad changes to multiple inter-dependent hemodynamic parameters, it is difficult to isolate its effects to specific changes in mechanical load. Here, we use engineered heart tissues (EHTs) with iPSC-derived cardiomyocytes expressing R451G desmoplakin, an ACM-linked mutation, which results in a functionally null model of desmoplakin (DSP). We also use a novel bioreactor to independently perturb tissue strain at different time points during the cardiac cycle. We culture EHTs under three strain regimes: normal physiological shortening; increased diastolic stretch, simulating high preload; and isometric culture, simulating high afterload. DSPR451G EHTs that have been cultured isometrically undergo adaptation, with no change in action potential parameters, conduction velocity, or contractile function, a phenotype confirmed by global proteomic analysis. However, when DSPR451G EHTs are subjected to increased diastolic stretch, they exhibit concomitant reductions in conduction velocity and the expression of connexin-43. These effects are rescued by inhibition of both lysosome activity and ERK signaling. Our results indicate that the response of DSPR451G EHTs to mechanical stimuli depends on the strain and the timing of the applied stimulus, with increased diastolic stretch unmasking conduction deficits in a concealed-phase model of ACM.
Collapse
Affiliation(s)
- Ronald Ng
- Yale University, New Haven, United States
| | | | | | - Fadi G Akar
- Cardiovascular Medicine and Biomedical Engineering, Yale University, New Haven, CT, United States
| | - Stuart G Campbell
- Division of Cardiology, Department of Internal Medicine, Yale University, New Haven, CT, United States
| |
Collapse
|
13
|
Uchechukwu CF, Anyaduba UL, Udekwu CC, Orababa OQ, Kade AE. Desmoglein-2 and COVID-19 complications: insights into its role as a biomarker, pathogenesis and clinical implications. J Gen Virol 2023; 104. [PMID: 37815458 DOI: 10.1099/jgv.0.001902] [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] [Indexed: 10/11/2023] Open
Abstract
Desmoglein-2 (DSG2) has emerged as a potential biomarker for coronavirus disease 2019 (COVID-19) complications, particularly cardiac and cardiovascular involvement. The expression of DSG2 in lung tissues has been detected at elevated levels, and circulating DSG2 levels correlate with COVID-19 severity. DSG2 may contribute to myocardial injury, cardiac dysfunction and vascular endothelial dysfunction in COVID-19. Monitoring DSG2 levels could aid in risk stratification, early detection and prognostication of COVID-19 complications. However, further research is required to validate DSG2 as a biomarker. Such research will aim to elucidate its precise role in pathogenesis, establishing standardized assays for its measurement and possibly identifying therapeutic targets.
Collapse
Affiliation(s)
- Chidiebere F Uchechukwu
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
- Warwick Medical School, University of Warwick, Coventry, UK
- School of Life Sciences, University of Warwick, Coventry, UK
- Michael Okpara University of Agriculture, Umudike, Nigeria
| | | | | | | | | |
Collapse
|
14
|
Chua CJ, Morrissette-McAlmon J, Tung L, Boheler KR. Understanding Arrhythmogenic Cardiomyopathy: Advances through the Use of Human Pluripotent Stem Cell Models. Genes (Basel) 2023; 14:1864. [PMID: 37895213 PMCID: PMC10606441 DOI: 10.3390/genes14101864] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 09/11/2023] [Accepted: 09/16/2023] [Indexed: 10/29/2023] Open
Abstract
Cardiomyopathies (CMPs) represent a significant healthcare burden and are a major cause of heart failure leading to premature death. Several CMPs are now recognized to have a strong genetic basis, including arrhythmogenic cardiomyopathy (ACM), which predisposes patients to arrhythmic episodes. Variants in one of the five genes (PKP2, JUP, DSC2, DSG2, and DSP) encoding proteins of the desmosome are known to cause a subset of ACM, which we classify as desmosome-related ACM (dACM). Phenotypically, this disease may lead to sudden cardiac death in young athletes and, during late stages, is often accompanied by myocardial fibrofatty infiltrates. While the pathogenicity of the desmosome genes has been well established through animal studies and limited supplies of primary human cells, these systems have drawbacks that limit their utility and relevance to understanding human disease. Human induced pluripotent stem cells (hiPSCs) have emerged as a powerful tool for modeling ACM in vitro that can overcome these challenges, as they represent a reproducible and scalable source of cardiomyocytes (CMs) that recapitulate patient phenotypes. In this review, we provide an overview of dACM, summarize findings in other model systems linking desmosome proteins with this disease, and provide an up-to-date summary of the work that has been conducted in hiPSC-cardiomyocyte (hiPSC-CM) models of dACM. In the context of the hiPSC-CM model system, we highlight novel findings that have contributed to our understanding of disease and enumerate the limitations, prospects, and directions for research to consider towards future progress.
Collapse
Affiliation(s)
- Christianne J. Chua
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (C.J.C.); (J.M.-M.); (L.T.)
| | - Justin Morrissette-McAlmon
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (C.J.C.); (J.M.-M.); (L.T.)
| | - Leslie Tung
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (C.J.C.); (J.M.-M.); (L.T.)
| | - Kenneth R. Boheler
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (C.J.C.); (J.M.-M.); (L.T.)
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
15
|
Vielmuth F, Radeva MY, Yeruva S, Sigmund AM, Waschke J. cAMP: A master regulator of cadherin-mediated binding in endothelium, epithelium and myocardium. Acta Physiol (Oxf) 2023; 238:e14006. [PMID: 37243909 DOI: 10.1111/apha.14006] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 05/05/2023] [Accepted: 05/22/2023] [Indexed: 05/29/2023]
Abstract
Regulation of cadherin-mediated cell adhesion is crucial not only for maintaining tissue integrity and barrier function in the endothelium and epithelium but also for electromechanical coupling within the myocardium. Therefore, loss of cadherin-mediated adhesion causes various disorders, including vascular inflammation and desmosome-related diseases such as the autoimmune blistering skin dermatosis pemphigus and arrhythmogenic cardiomyopathy. Mechanisms regulating cadherin-mediated binding contribute to the pathogenesis of diseases and may also be used as therapeutic targets. Over the last 30 years, cyclic adenosine 3',5'-monophosphate (cAMP) has emerged as one of the master regulators of cell adhesion in endothelium and, more recently, also in epithelial cells as well as in cardiomyocytes. A broad spectrum of experimental models from vascular physiology and cell biology applied by different generations of researchers provided evidence that not only cadherins of endothelial adherens junctions (AJ) but also desmosomal contacts in keratinocytes and the cardiomyocyte intercalated discs are central targets in this scenario. The molecular mechanisms involve protein kinase A- and exchange protein directly activated by cAMP-mediated regulation of Rho family GTPases and S665 phosphorylation of the AJ and desmosome adaptor protein plakoglobin. In line with this, phosphodiesterase 4 inhibitors such as apremilast have been proposed as a therapeutic strategy to stabilize cadherin-mediated adhesion in pemphigus and may also be effective to treat other disorders where cadherin-mediated binding is compromised.
Collapse
Affiliation(s)
- Franziska Vielmuth
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Mariya Y Radeva
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Sunil Yeruva
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Anna M Sigmund
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Jens Waschke
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, LMU Munich, Munich, Germany
| |
Collapse
|
16
|
Ward KE, Steadman L, Karim AR, Reynolds GM, Pugh M, Chua W, Faustini SE, Veenith T, Thwaites RS, Openshaw PJM, Drayson MT, Shields AM, Cunningham AF, Wraith DC, Richter AG. SARS-CoV-2 infection is associated with anti-desmoglein 2 autoantibody detection. Clin Exp Immunol 2023; 213:243-251. [PMID: 37095599 PMCID: PMC10651225 DOI: 10.1093/cei/uxad046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 03/08/2023] [Accepted: 04/24/2023] [Indexed: 04/26/2023] Open
Abstract
Post-acute cardiac sequelae, following SARS-CoV-2 infection, are well recognized as complications of COVID-19. We have previously shown the persistence of autoantibodies against antigens in skin, muscle, and heart in individuals following severe COVID-19; the most common staining on skin tissue displayed an inter-cellular cement pattern consistent with antibodies against desmosomal proteins. Desmosomes play a critical role in maintaining the structural integrity of tissues. For this reason, we analyzed desmosomal protein levels and the presence of anti-desmoglein (DSG) 1, 2, and 3 antibodies in acute and convalescent sera from patients with COVID-19 of differing clinical severity. We find increased levels of DSG2 protein in sera from acute COVID-19 patients. Furthermore, we find that DSG2 autoantibody levels are increased significantly in convalescent sera following severe COVID-19 but not in hospitalized patients recovering from influenza infection or healthy controls. Levels of autoantibody in sera from patients with severe COVID-19 were comparable to levels in patients with non-COVID-19-associated cardiac disease, potentially identifying DSG2 autoantibodies as a novel biomarker for cardiac damage. To determine if there was any association between severe COVID-19 and DSG2, we stained post-mortem cardiac tissue from patients who died from COVID-19 infection. This confirmed DSG2 protein within the intercalated discs and disruption of the intercalated disc between cardiomyocytes in patients who died from COVID-19. Our results reveal the potential for DSG2 protein and autoimmunity to DSG2 to contribute to unexpected pathologies associated with COVID-19 infection.
Collapse
Affiliation(s)
- Kerensa E Ward
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, West Midlands, UK
| | - Lora Steadman
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, West Midlands, UK
| | - Abid R Karim
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, West Midlands, UK
| | - Gary M Reynolds
- Centre for Liver and Gastrointestinal Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, West Midlands, UK
| | - Matthew Pugh
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, West Midlands, UK
| | - Winnie Chua
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, West Midlands, UK
| | - Sian E Faustini
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, West Midlands, UK
| | - Tonny Veenith
- Department of Critical Care, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Ryan S Thwaites
- National Heart and Lung Institute, Imperial College London, London, UK
| | | | - Mark T Drayson
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, West Midlands, UK
| | - Adrian M Shields
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, West Midlands, UK
- Department of Clinical Immunology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Adam F Cunningham
- Institute of Microbiology and Infection, University of Birmingham, Birmingham, UK
| | - David C Wraith
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, West Midlands, UK
| | - Alex G Richter
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, West Midlands, UK
- Department of Clinical Immunology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| |
Collapse
|
17
|
Thiene G, Basso C, Pilichou K, Bueno Marinas M. Desmosomal Arrhythmogenic Cardiomyopathy: The Story Telling of a Genetically Determined Heart Muscle Disease. Biomedicines 2023; 11:2018. [PMID: 37509658 PMCID: PMC10377062 DOI: 10.3390/biomedicines11072018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 06/28/2023] [Accepted: 07/10/2023] [Indexed: 07/30/2023] Open
Abstract
The history of arrhythmogenic cardiomyopathy (AC) as a genetically determined desmosomal disease started since the original discovery by Lancisi in a four-generation family, published in 1728. Contemporary history at the University of Padua started with Dalla Volta, who haemodynamically investigated patients with "auricularization" of the right ventricle, and with Nava, who confirmed familiarity. The contemporary knowledge advances consisted of (a) AC as a heart muscle disease with peculiar electrical instability of the right ventricle; (b) the finding of pathological substrates, in keeping with a myocardial dystrophy; (c) the inclusion of AC in the cardiomyopathies classification; (d) AC as the main cause of sudden death in athletes; (e) the discovery of the culprit genes coding proteins of the intercalated disc (desmosome); (f) progression in clinical diagnosis with specific ECG abnormalities, angiocardiography, endomyocardial biopsy, 2D echocardiography, electron anatomic mapping and cardiac magnetic resonance; (g) the discovery of left ventricular AC; (h) prevention of SCD with the invention and application of the lifesaving implantable cardioverter defibrillator and external defibrillator scattered in public places and playgrounds as well as the ineligibility for competitive sport activity for AC patients; (i) genetic screening of the proband family to unmask asymptomatic carriers. Nondesmosomal ACs, with a phenotype overlapping desmosomal AC, are also treated, including genetics: Transmembrane protein 43, SCN5A, Desmin, Phospholamban, Lamin A/C, Filamin C, Cadherin 2, Tight junction protein 1.
Collapse
Affiliation(s)
- Gaetano Thiene
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, Medical School, University of Padua, 35121 Padova, Italy
| | - Cristina Basso
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, Medical School, University of Padua, 35121 Padova, Italy
| | - Kalliopi Pilichou
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, Medical School, University of Padua, 35121 Padova, Italy
| | - Maria Bueno Marinas
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, Medical School, University of Padua, 35121 Padova, Italy
| |
Collapse
|
18
|
Nielsen MS, van Opbergen CJM, van Veen TAB, Delmar M. The intercalated disc: a unique organelle for electromechanical synchrony in cardiomyocytes. Physiol Rev 2023; 103:2271-2319. [PMID: 36731030 PMCID: PMC10191137 DOI: 10.1152/physrev.00021.2022] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 01/24/2023] [Accepted: 01/30/2023] [Indexed: 02/04/2023] Open
Abstract
The intercalated disc (ID) is a highly specialized structure that connects cardiomyocytes via mechanical and electrical junctions. Although described in some detail by light microscopy in the 19th century, it was in 1966 that electron microscopy images showed that the ID represented apposing cell borders and provided detailed insight into the complex ID nanostructure. Since then, much has been learned about the ID and its molecular composition, and it has become evident that a large number of proteins, not all of them involved in direct cell-to-cell coupling via mechanical or gap junctions, reside at the ID. Furthermore, an increasing number of functional interactions between ID components are emerging, leading to the concept that the ID is not the sum of isolated molecular silos but an interacting molecular complex, an "organelle" where components work in concert to bring about electrical and mechanical synchrony. The aim of the present review is to give a short historical account of the ID's discovery and an updated overview of its composition and organization, followed by a discussion of the physiological implications of the ID architecture and the local intermolecular interactions. The latter will focus on both the importance of normal conduction of cardiac action potentials as well as the impact on the pathophysiology of arrhythmias.
Collapse
Affiliation(s)
- Morten S Nielsen
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Chantal J M van Opbergen
- The Leon Charney Division of Cardiology, New York University Grossmann School of Medicine, New York, New York, United States
| | - Toon A B van Veen
- Department of Medical Physiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Mario Delmar
- The Leon Charney Division of Cardiology, New York University Grossmann School of Medicine, New York, New York, United States
| |
Collapse
|
19
|
Remme CA. SCN5A channelopathy: arrhythmia, cardiomyopathy, epilepsy and beyond. Philos Trans R Soc Lond B Biol Sci 2023; 378:20220164. [PMID: 37122208 PMCID: PMC10150216 DOI: 10.1098/rstb.2022.0164] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Accepted: 12/31/2022] [Indexed: 05/02/2023] Open
Abstract
Influx of sodium ions through voltage-gated sodium channels in cardiomyocytes is essential for proper electrical conduction within the heart. Both acquired conditions associated with sodium channel dysfunction (myocardial ischaemia, heart failure) as well as inherited disorders secondary to mutations in the gene SCN5A encoding for the cardiac sodium channel Nav1.5 are associated with life-threatening arrhythmias. Research in the last decade has uncovered the complex nature of Nav1.5 distribution, function, in particular within distinct subcellular subdomains of cardiomyocytes. Nav1.5-based channels furthermore display previously unrecognized non-electrogenic actions and may impact on cardiac structural integrity, leading to cardiomyopathy. Moreover, SCN5A and Nav1.5 are expressed in cell types other than cardiomyocytes as well as various extracardiac tissues, where their functional role in, e.g. epilepsy, gastrointestinal motility, cancer and the innate immune response is increasingly investigated and recognized. This review provides an overview of these novel insights and how they deepen our mechanistic knowledge on SCN5A channelopathies and Nav1.5 (dys)function. This article is part of the theme issue 'The heartbeat: its molecular basis and physiological mechanisms'.
Collapse
Affiliation(s)
- Carol Ann Remme
- Department of Experimental Cardiology, Heart Centre, Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Amsterdam UMC location AMC, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
20
|
Miles C, Boukens BJ, Scrocco C, Wilde AA, Nademanee K, Haissaguerre M, Coronel R, Behr ER. Subepicardial Cardiomyopathy: A Disease Underlying J-Wave Syndromes and Idiopathic Ventricular Fibrillation. Circulation 2023; 147:1622-1633. [PMID: 37216437 PMCID: PMC11073566 DOI: 10.1161/circulationaha.122.061924] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 03/27/2023] [Indexed: 05/24/2023]
Abstract
Brugada syndrome (BrS), early repolarization syndrome (ERS), and idiopathic ventricular fibrillation (iVF) have long been considered primary electrical disorders associated with malignant ventricular arrhythmia and sudden cardiac death. However, recent studies have revealed the presence of subtle microstructural abnormalities of the extracellular matrix in some cases of BrS, ERS, and iVF, particularly within right ventricular subepicardial myocardium. Substrate-based ablation within this region has been shown to ameliorate the electrocardiographic phenotype and to reduce arrhythmia frequency in BrS. Patients with ERS and iVF may also exhibit low-voltage and fractionated electrograms in the ventricular subepicardial myocardium, which can be treated with ablation. A significant proportion of patients with BrS and ERS, as well as some iVF survivors, harbor pathogenic variants in the voltage-gated sodium channel gene, SCN5A, but the majority of genetic susceptibility of these disorders is likely to be polygenic. Here, we postulate that BrS, ERS, and iVF may form part of a spectrum of subtle subepicardial cardiomyopathy. We propose that impaired sodium current, along with genetic and environmental susceptibility, precipitates a reduction in epicardial conduction reserve, facilitating current-to-load mismatch at sites of structural discontinuity, giving rise to electrocardiographic changes and the arrhythmogenic substrate.
Collapse
Affiliation(s)
- Chris Miles
- Cardiovascular Clinical Academic Group, St. George’s University Hospitals’ NHS Foundation Trust and Molecular and Clinical Sciences Institute, St. George’s, University of London, UK (C.M., C.S., E.R.B.)
| | - Bastiaan J. Boukens
- Department of Medical Biology, University of Amsterdam, the Netherlands (B.J.B.)
- University of Maastricht, Cardiovascular Research Institute Maastricht, Maastricht University Medical Center, the Netherlands (B.J.B.)
| | - Chiara Scrocco
- Cardiovascular Clinical Academic Group, St. George’s University Hospitals’ NHS Foundation Trust and Molecular and Clinical Sciences Institute, St. George’s, University of London, UK (C.M., C.S., E.R.B.)
| | - Arthur A.M. Wilde
- Amsterdam UMC, University of Amsterdam, Department of Cardiology, the Netherlands (A.A.M.W.)
- Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, the Netherlands (A.A.M.W.)
- European Reference Network for rare, low-prevalence, and complex diseases of the heart: ERN GUARD-Heart (A.A.M.W., M.H.)
| | - Koonlawee Nademanee
- Center of Excellence in Arrhythmia Research Chulalongkorn University, Department of Medicine, Chulalongkorn University, Thailand (K.N.)
- Pacific Rim Electrophysiology Research Institute, Bumrungrad Hospital, Bangkok, Thailand (K.N.)
| | - Michel Haissaguerre
- European Reference Network for rare, low-prevalence, and complex diseases of the heart: ERN GUARD-Heart (A.A.M.W., M.H.)
- Institut Hospitalo-Universitaire Liryc, Electrophysiology and Heart Modeling Institute, Pessac, France (M.H.)
- Department of Electrophysiology and Cardiac Stimulation, Centre Hospitalier Universitaire de Bordeaux, France (M.H.)
| | - Ruben Coronel
- Department of Experimental Cardiology, Amsterdam University Medical Centers, Cardiovascular Science, the Netherlands (R.C.)
| | - Elijah R. Behr
- Cardiovascular Clinical Academic Group, St. George’s University Hospitals’ NHS Foundation Trust and Molecular and Clinical Sciences Institute, St. George’s, University of London, UK (C.M., C.S., E.R.B.)
- Mayo Clinic Healthcare, London, UK (E.R.B.)
| |
Collapse
|
21
|
Reisqs JB, Moreau A, Sleiman Y, Boutjdir M, Richard S, Chevalier P. Arrhythmogenic cardiomyopathy as a myogenic disease: highlights from cardiomyocytes derived from human induced pluripotent stem cells. Front Physiol 2023; 14:1191965. [PMID: 37250123 PMCID: PMC10210147 DOI: 10.3389/fphys.2023.1191965] [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: 03/22/2023] [Accepted: 05/02/2023] [Indexed: 05/31/2023] Open
Abstract
Arrhythmogenic cardiomyopathy (ACM) is an inherited cardiomyopathy characterized by the replacement of myocardium by fibro-fatty infiltration and cardiomyocyte loss. ACM predisposes to a high risk for ventricular arrhythmias. ACM has initially been defined as a desmosomal disease because most of the known variants causing the disease concern genes encoding desmosomal proteins. Studying this pathology is complex, in particular because human samples are rare and, when available, reflect the most advanced stages of the disease. Usual cellular and animal models cannot reproduce all the hallmarks of human pathology. In the last decade, human-induced pluripotent stem cells (hiPSC) have been proposed as an innovative human cellular model. The differentiation of hiPSCs into cardiomyocytes (hiPSC-CM) is now well-controlled and widely used in many laboratories. This hiPSC-CM model recapitulates critical features of the pathology and enables a cardiomyocyte-centered comprehensive approach to the disease and the screening of anti-arrhythmic drugs (AAD) prescribed sometimes empirically to the patient. In this regard, this model provides unique opportunities to explore and develop new therapeutic approaches. The use of hiPSC-CMs will undoubtedly help the development of precision medicine to better cure patients suffering from ACM. This review aims to summarize the recent advances allowing the use of hiPSCs in the ACM context.
Collapse
Affiliation(s)
- J. B. Reisqs
- Cardiovascular Research Program, VA New York Harbor Healthcare System, Brooklyn, NY, United States
| | - A. Moreau
- Université de Montpellier, Institut National de la Santé et de la Recherche Médicale, Centre National de la Recherche Scientifique, PhyMedExp, Montpellier, France
| | - Y. Sleiman
- Cardiovascular Research Program, VA New York Harbor Healthcare System, Brooklyn, NY, United States
| | - M. Boutjdir
- Cardiovascular Research Program, VA New York Harbor Healthcare System, Brooklyn, NY, United States
- Department of Medicine, Cell Biology and Pharmacology, State University of New York Downstate Health Sciences University, NY, United States
- Department of Medicine, New York University School of Medicine, NY, United States
| | - S. Richard
- Université de Montpellier, Institut National de la Santé et de la Recherche Médicale, Centre National de la Recherche Scientifique, PhyMedExp, Montpellier, France
| | - P. Chevalier
- Neuromyogene Institute, Claude Bernard University, Lyon 1, Villeurbanne, France
- Service de Rythmologie, Hospices Civils de Lyon, Lyon, France
| |
Collapse
|
22
|
Peretto G, Sommariva E, Di Resta C, Rabino M, Villatore A, Lazzeroni D, Sala S, Pompilio G, Cooper LT. Myocardial Inflammation as a Manifestation of Genetic Cardiomyopathies: From Bedside to the Bench. Biomolecules 2023; 13:646. [PMID: 37189393 PMCID: PMC10136351 DOI: 10.3390/biom13040646] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 03/24/2023] [Accepted: 03/24/2023] [Indexed: 05/17/2023] Open
Abstract
Over recent years, preclinical and clinical evidence has implicated myocardial inflammation (M-Infl) in the pathophysiology and phenotypes of traditionally genetic cardiomyopathies. M-Infl resembling myocarditis on imaging and histology occurs frequently as a clinical manifestation of classically genetic cardiac diseases, including dilated and arrhythmogenic cardiomyopathy. The emerging role of M-Infl in disease pathophysiology is leading to the identification of druggable targets for molecular treatment of the inflammatory process and a new paradigm in the field of cardiomyopathies. Cardiomyopathies constitute a leading cause of heart failure and arrhythmic sudden death in the young population. The aim of this review is to present, from bedside to bench, the current state of the art about the genetic basis of M-Infl in nonischemic cardiomyopathies of the dilated and arrhythmogenic spectrum in order to prompt future research towards the identification of novel mechanisms and treatment targets, with the ultimate goal of lowering disease morbidity and mortality.
Collapse
Affiliation(s)
- Giovanni Peretto
- Department of Cardiac Electrophysiology and Arrhythmology, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
- School of Medicine, Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Elena Sommariva
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino IRCCS, 20139 Milan, Italy
| | - Chiara Di Resta
- School of Medicine, Vita-Salute San Raffaele University, 20132 Milan, Italy
- Genomic Unit for the Diagnosis of Human Pathologies, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Martina Rabino
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino IRCCS, 20139 Milan, Italy
| | - Andrea Villatore
- Department of Cardiac Electrophysiology and Arrhythmology, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | | | - Simone Sala
- Department of Cardiac Electrophysiology and Arrhythmology, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Giulio Pompilio
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino IRCCS, 20139 Milan, Italy
- Department of Biomedical, Surgical and Dental Sciences, Università degli Studi di Milano, 20122 Milan, Italy
| | - Leslie T. Cooper
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL 32224, USA
| |
Collapse
|
23
|
Struckman HL, Moise N, King DR, Soltisz A, Buxton A, Dunlap I, Chen Z, Radwański PB, Weinberg SH, Veeraraghavan R. Unraveling Chamber-specific Differences in Intercalated Disc Ultrastructure and Molecular Organization and Their Impact on Cardiac Conduction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.13.528369. [PMID: 36824727 PMCID: PMC9949041 DOI: 10.1101/2023.02.13.528369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
During each heartbeat, the propagation of action potentials through the heart coordinates the contraction of billions of individual cardiomyocytes and is thus, a critical life process. Unsurprisingly, intercalated discs, which are cell-cell contact sites specialized to provide electrical and mechanical coupling between adjacent cardiomyocytes, have been the focus of much investigation. Slowed or disrupted propagation leads to potentially life-threatening arrhythmias in a wide range of pathologies, where intercalated disc remodeling is a common finding. Hence, the importance and urgency of understanding intercalated disc structure and its influence on action potential propagation. Surprisingly, however, conventional modeling approaches cannot predict changes in propagation elicited by perturbations that alter intercalated disc ultrastructure or molecular organization, owing to lack of quantitative structural data at subcellular through nano scales. In order to address this critical gap in knowledge, we sought to quantify intercalated disc structure at these finer spatial scales in the healthy adult mouse heart and relate them to function in a chamber-specific manner as a precursor to understanding the impacts of pathological intercalated disc remodeling. Using super-resolution light microscopy, electron microscopy, and computational image analysis, we provide here the first ever systematic, multiscale quantification of intercalated disc ultrastructure and molecular organization. By incorporating these data into a rule-based model of cardiac tissue with realistic intercalated disc structure, and comparing model predictions of electrical propagation with experimental measures of conduction velocity, we reveal that atrial intercalated discs can support faster conduction than their ventricular counterparts, which is normally masked by inter-chamber differences in myocyte geometry. Further, we identify key ultrastructural and molecular organization features underpinning the ability of atrial intercalated discs to support faster conduction. These data provide the first stepping stone to elucidating chamber-specific impacts of pathological intercalated disc remodeling, as occurs in many arrhythmic diseases.
Collapse
|
24
|
Marchal GA, Remme CA. Subcellular diversity of Nav1.5 in cardiomyocytes: distinct functions, mechanisms and targets. J Physiol 2023; 601:941-960. [PMID: 36469003 DOI: 10.1113/jp283086] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 11/24/2022] [Indexed: 12/11/2022] Open
Abstract
In cardiomyocytes, the rapid depolarisation of the membrane potential is mediated by the α-subunit of the cardiac voltage-gated Na+ channel (NaV 1.5), encoded by the gene SCN5A. This ion channel allows positively charged Na+ ions to enter the cardiomyocyte, resulting in the fast upstroke of the action potential and is therefore crucial for cardiac excitability and electrical propagation. This essential role is underscored by the fact that dysfunctional NaV 1.5 is associated with high risk for arrhythmias and sudden cardiac death. However, development of therapeutic interventions regulating NaV 1.5 has been limited due to the complexity of NaV 1.5 structure and function and its diverse roles within the cardiomyocyte. In particular, research from the last decade has provided us with increased knowledge on the subcellular distribution of NaV 1.5 as well as the proteins which it interacts with in distinct cardiomyocyte microdomains. We here review these insights, detailing the potential role of NaV 1.5 within subcellular domains as well as its dysfunction in the setting of arrhythmia disorders. We furthermore provide an overview of current knowledge on the pathways involved in (microdomain-specific) trafficking of NaV 1.5, and their potential as novel targets. Unravelling the complexity of NaV 1.5 (dys)function may ultimately facilitate the development of therapeutic strategies aimed at preventing lethal arrhythmias. This is not only of importance for pathophysiological conditions where sodium current is specifically decreased within certain subcellular regions, such as in arrhythmogenic cardiomyopathy and Duchenne muscular dystrophy, but also for other acquired and inherited disorders associated with NaV 1.5.
Collapse
Affiliation(s)
- Gerard A Marchal
- Department of Experimental Cardiology, Heart Centre, Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands.,National Institute of Optics, National Research Council (CNR-INO), Sesto Fiorentino, Florence, Italy
| | - Carol Ann Remme
- Department of Experimental Cardiology, Heart Centre, Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
25
|
de Boer M, Te Lintel Hekkert M, Chang J, van Thiel BS, Martens L, Bos MM, de Kleijnen MGJ, Ridwan Y, Octavia Y, van Deel ED, Blonden LA, Brandt RMC, Barnhoorn S, Bautista-Niño PK, Krabbendam-Peters I, Wolswinkel R, Arshi B, Ghanbari M, Kupatt C, de Windt LJ, Danser AHJ, van der Pluijm I, Remme CA, Stoll M, Pothof J, Roks AJM, Kavousi M, Essers J, van der Velden J, Hoeijmakers JHJ, Duncker DJ. DNA repair in cardiomyocytes is critical for maintaining cardiac function in mice. Aging Cell 2023; 22:e13768. [PMID: 36756698 PMCID: PMC10014058 DOI: 10.1111/acel.13768] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 11/30/2022] [Accepted: 12/19/2022] [Indexed: 02/10/2023] Open
Abstract
Heart failure has reached epidemic proportions in a progressively ageing population. The molecular mechanisms underlying heart failure remain elusive, but evidence indicates that DNA damage is enhanced in failing hearts. Here, we tested the hypothesis that endogenous DNA repair in cardiomyocytes is critical for maintaining normal cardiac function, so that perturbed repair of spontaneous DNA damage drives early onset of heart failure. To increase the burden of spontaneous DNA damage, we knocked out the DNA repair endonucleases xeroderma pigmentosum complementation group G (XPG) and excision repair cross-complementation group 1 (ERCC1), either systemically or cardiomyocyte-restricted, and studied the effects on cardiac function and structure. Loss of DNA repair permitted normal heart development but subsequently caused progressive deterioration of cardiac function, resulting in overt congestive heart failure and premature death within 6 months. Cardiac biopsies revealed increased oxidative stress associated with increased fibrosis and apoptosis. Moreover, gene set enrichment analysis showed enrichment of pathways associated with impaired DNA repair and apoptosis, and identified TP53 as one of the top active upstream transcription regulators. In support of the observed cardiac phenotype in mutant mice, several genetic variants in the ERCC1 and XPG gene in human GWAS data were found to be associated with cardiac remodelling and dysfunction. In conclusion, unrepaired spontaneous DNA damage in differentiated cardiomyocytes drives early onset of cardiac failure. These observations implicate DNA damage as a potential novel therapeutic target and highlight systemic and cardiomyocyte-restricted DNA repair-deficient mouse mutants as bona fide models of heart failure.
Collapse
Affiliation(s)
- Martine de Boer
- Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus MC, Rotterdam, The Netherlands
| | - Maaike Te Lintel Hekkert
- Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus MC, Rotterdam, The Netherlands
| | - Jiang Chang
- Department of Molecular Genetics, Erasmus MC, Rotterdam, The Netherlands
| | - Bibi S van Thiel
- Department of Molecular Genetics, Erasmus MC, Rotterdam, The Netherlands.,Department of Vascular Surgery, Erasmus MC, Rotterdam, The Netherlands.,Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Leonie Martens
- Department of Genetic Epidemiology, Institute of Human Genetics, University Hospital Münster, Münster, Germany
| | - Maxime M Bos
- Department of Epidemiology, Erasmus MC, Rotterdam, The Netherlands
| | - Marion G J de Kleijnen
- Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus MC, Rotterdam, The Netherlands
| | - Yanto Ridwan
- Department of Molecular Genetics, Erasmus MC, Rotterdam, The Netherlands.,Department of Radiotherapy, Erasmus MC, Rotterdam, The Netherlands
| | - Yanti Octavia
- Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus MC, Rotterdam, The Netherlands
| | - Elza D van Deel
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Lau A Blonden
- Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus MC, Rotterdam, The Netherlands
| | - Renata M C Brandt
- Department of Molecular Genetics, Erasmus MC, Rotterdam, The Netherlands
| | - Sander Barnhoorn
- Department of Molecular Genetics, Erasmus MC, Rotterdam, The Netherlands
| | - Paula K Bautista-Niño
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands.,Centro de Investigaciones, Fundación Cardiovascular de Colombia- FCV, Bucaramanga, Colombia
| | - Ilona Krabbendam-Peters
- Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus MC, Rotterdam, The Netherlands
| | - Rianne Wolswinkel
- Department of Clinical and Experimental Cardiology, Heart Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Banafsheh Arshi
- Department of Epidemiology, Erasmus MC, Rotterdam, The Netherlands
| | - Mohsen Ghanbari
- Department of Epidemiology, Erasmus MC, Rotterdam, The Netherlands
| | - Christian Kupatt
- I. Medizinische Klinik und Poliklinik, University Clinic Rechts der Isar, Technical University of Munich, Munich, Germany.,DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany.,Walter-Brendel-Centre for Experimental Medicine, Ludwig Maximilian University of Munich, Munich, Germany
| | - Leon J de Windt
- Department of Molecular Genetics, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands.,Faculty of Science and Engineering, Maastricht University, Maastricht, The Netherlands
| | - A H Jan Danser
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Ingrid van der Pluijm
- Department of Molecular Genetics, Erasmus MC, Rotterdam, The Netherlands.,Department of Vascular Surgery, Erasmus MC, Rotterdam, The Netherlands
| | - Carol Ann Remme
- Department of Clinical and Experimental Cardiology, Heart Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Monika Stoll
- Department of Genetic Epidemiology, Institute of Human Genetics, University Hospital Münster, Münster, Germany.,Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands
| | - Joris Pothof
- Department of Molecular Genetics, Erasmus MC, Rotterdam, The Netherlands
| | - Anton J M Roks
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Maryam Kavousi
- Department of Epidemiology, Erasmus MC, Rotterdam, The Netherlands
| | - Jeroen Essers
- Department of Molecular Genetics, Erasmus MC, Rotterdam, The Netherlands.,Department of Vascular Surgery, Erasmus MC, Rotterdam, The Netherlands.,Department of Radiotherapy, Erasmus MC, Rotterdam, The Netherlands
| | - Jolanda van der Velden
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.,Netherlands Heart Institute, Utrecht, The Netherlands
| | - Jan H J Hoeijmakers
- Department of Molecular Genetics, Erasmus MC, Rotterdam, The Netherlands.,CECAD Forschungszentrum, Universität zu Köln, Köln, Germany.,Princess Máxima Center for Pediatric Oncology, Genome Instability and Nutrition, ONCODE Institute, Utrecht, The Netherlands
| | - Dirk J Duncker
- Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus MC, Rotterdam, The Netherlands
| |
Collapse
|
26
|
Schinner C, Xu L, Franz H, Zimmermann A, Wanuske MT, Rathod M, Hanns P, Geier F, Pelczar P, Liang Y, Lorenz V, Stüdle C, Maly PI, Kauferstein S, Beckmann BM, Sheikh F, Kuster GM, Spindler V. Defective Desmosomal Adhesion Causes Arrhythmogenic Cardiomyopathy by Involving an Integrin-αVβ6/TGF-β Signaling Cascade. Circulation 2022; 146:1610-1626. [PMID: 36268721 PMCID: PMC9674449 DOI: 10.1161/circulationaha.121.057329] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
BACKGROUND Arrhythmogenic cardiomyopathy (ACM) is characterized by progressive loss of cardiomyocytes with fibrofatty tissue replacement, systolic dysfunction, and life-threatening arrhythmias. A substantial proportion of ACM is caused by mutations in genes of the desmosomal cell-cell adhesion complex, but the underlying mechanisms are not well understood. In the current study, we investigated the relevance of defective desmosomal adhesion for ACM development and progression. METHODS We mutated the binding site of DSG2 (desmoglein-2), a crucial desmosomal adhesion molecule in cardiomyocytes. This DSG2-W2A mutation abrogates the tryptophan swap, a central interaction mechanism of DSG2 on the basis of structural data. Impaired adhesive function of DSG2-W2A was confirmed by cell-cell dissociation assays and force spectroscopy measurements by atomic force microscopy. The DSG2-W2A knock-in mouse model was analyzed by echocardiography, ECG, and histologic and biomolecular techniques including RNA sequencing and transmission electron and superresolution microscopy. The results were compared with ACM patient samples, and their relevance was confirmed in vivo and in cardiac slice cultures by inhibitor studies applying the small molecule EMD527040 or an inhibitory integrin-αVβ6 antibody. RESULTS The DSG2-W2A mutation impaired binding on molecular level and compromised intercellular adhesive function. Mice bearing this mutation develop a severe cardiac phenotype recalling the characteristics of ACM, including cardiac fibrosis, impaired systolic function, and arrhythmia. A comparison of the transcriptome of mutant mice with ACM patient data suggested deregulated integrin-αVβ6 and subsequent transforming growth factor-β signaling as driver of cardiac fibrosis. Blocking integrin-αVβ6 led to reduced expression of profibrotic markers and reduced fibrosis formation in mutant animals in vivo. CONCLUSIONS We show that disruption of desmosomal adhesion is sufficient to induce a phenotype that fulfils the clinical criteria to establish the diagnosis of ACM, confirming the dysfunctional adhesion hypothesis. Deregulation of integrin-αVβ6 and transforming growth factor-β signaling was identified as a central step toward fibrosis. A pilot in vivo drug test revealed this pathway as a promising target to ameliorate fibrosis. This highlights the value of this model to discern mechanisms of cardiac fibrosis and to identify and test novel treatment options for ACM.
Collapse
Affiliation(s)
- Camilla Schinner
- Department of Biomedicine, Section Anatomy (C. Schinner, H.F., A.Z., M.-T.W., M.R., P.H., C. Stüdle, P.I.M., V.S.), University of Basel, Switzerland
| | - Lifen Xu
- Department of Biomedicine, University Hospital Basel and University of Basel, Switzerland (L.X., V.L., G.M.K.)
| | - Henriette Franz
- Department of Biomedicine, Section Anatomy (C. Schinner, H.F., A.Z., M.-T.W., M.R., P.H., C. Stüdle, P.I.M., V.S.), University of Basel, Switzerland
| | - Aude Zimmermann
- Department of Biomedicine, Section Anatomy (C. Schinner, H.F., A.Z., M.-T.W., M.R., P.H., C. Stüdle, P.I.M., V.S.), University of Basel, Switzerland
| | - Marie-Therès Wanuske
- Department of Biomedicine, Section Anatomy (C. Schinner, H.F., A.Z., M.-T.W., M.R., P.H., C. Stüdle, P.I.M., V.S.), University of Basel, Switzerland
| | - Maitreyi Rathod
- Department of Biomedicine, Section Anatomy (C. Schinner, H.F., A.Z., M.-T.W., M.R., P.H., C. Stüdle, P.I.M., V.S.), University of Basel, Switzerland
| | - Pauline Hanns
- Department of Biomedicine, Section Anatomy (C. Schinner, H.F., A.Z., M.-T.W., M.R., P.H., C. Stüdle, P.I.M., V.S.), University of Basel, Switzerland
| | - Florian Geier
- Department of Biomedicine, Bioinformatics Core Facility (F.G.), University Hospital Basel, Switzerland
- Swiss Institute of Bioinformatics, Basel, Switzerland (F.G.)
| | - Pawel Pelczar
- Center for Transgenic Models (P.P.), University of Basel, Switzerland
| | - Yan Liang
- Department of Medicine, University of California San Diego (Y.L., F.S.)
| | - Vera Lorenz
- Department of Biomedicine, University Hospital Basel and University of Basel, Switzerland (L.X., V.L., G.M.K.)
| | - Chiara Stüdle
- Department of Biomedicine, Section Anatomy (C. Schinner, H.F., A.Z., M.-T.W., M.R., P.H., C. Stüdle, P.I.M., V.S.), University of Basel, Switzerland
| | - Piotr I. Maly
- Department of Biomedicine, Section Anatomy (C. Schinner, H.F., A.Z., M.-T.W., M.R., P.H., C. Stüdle, P.I.M., V.S.), University of Basel, Switzerland
| | - Silke Kauferstein
- Department of Legal Medicine, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany (S.K., B.M.B.)
| | - Britt M. Beckmann
- Department of Legal Medicine, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany (S.K., B.M.B.)
- Department of Medicine I, University Hospital, LMU Munich, Germany (B.M.B.)
| | - Farah Sheikh
- Department of Medicine, University of California San Diego (Y.L., F.S.)
| | - Gabriela M. Kuster
- Department of Biomedicine, University Hospital Basel and University of Basel, Switzerland (L.X., V.L., G.M.K.)
- Division of Cardiology (G.M.K.), University Hospital Basel, Switzerland
| | - Volker Spindler
- Department of Biomedicine, Section Anatomy (C. Schinner, H.F., A.Z., M.-T.W., M.R., P.H., C. Stüdle, P.I.M., V.S.), University of Basel, Switzerland
| |
Collapse
|
27
|
Laurita KR, Vasireddi SK, Mackall JA. Elucidating arrhythmogenic right ventricular cardiomyopathy with stem cells. Birth Defects Res 2022; 114:948-958. [PMID: 35396927 PMCID: PMC9790231 DOI: 10.1002/bdr2.2010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 03/17/2022] [Accepted: 03/23/2022] [Indexed: 12/31/2022]
Abstract
Human stems cells have sparked many novel strategies for treating heart disease and for elucidating their underlying mechanisms. For example, arrhythmogenic right ventricular cardiomyopathy (ARVC) is an inherited heart muscle disorder that is associated with fatal arrhythmias often occurring in healthy young adults. Fibro-fatty infiltrate, a clinical hallmark, progresses with the disease and can develop across both ventricles. Pathogenic variants in genes have been identified, with most being responsible for encoding cardiac desmosome proteins that reside at myocyte boundaries that are critical for cell-to-cell coupling. Despite some understanding of the molecular signaling mechanisms associated with ARVC mutations, their relationship with arrhythmogenesis is complex and not well understood for a monogenetic disorder. This review article focuses on arrhythmia mechanisms in ARVC based on clinical and animal studies and their relationship with disease causing variants. We also discuss the ways in which stem cells can be leveraged to improve our understanding of the role cardiac myocytes, nonmyocytes, metabolic signals, and inflammatory mediators play in an early onset disease such as ARVC.
Collapse
Affiliation(s)
- Kenneth R. Laurita
- Heart and Vascular Research CenterMetroHealth Campus, Case Western Reserve UniversityClevelandOhioUSA
| | - Sunil K. Vasireddi
- Stanford Cardiovascular Institute, Department of MedicineStanford UniversityCaliforniaUSA
| | - Judith A. Mackall
- Harrington Heart and Vascular InstituteUniversity Hospitals Cleveland Medical Center, Case Western Reserve UniversityClevelandOhioUSA
| |
Collapse
|
28
|
Zink M, Seewald A, Rohrbach M, Brodehl A, Liedtke D, Williams T, Childs SJ, Gerull B. Altered Expression of TMEM43 Causes Abnormal Cardiac Structure and Function in Zebrafish. Int J Mol Sci 2022; 23:9530. [PMID: 36076925 PMCID: PMC9455580 DOI: 10.3390/ijms23179530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 08/17/2022] [Accepted: 08/19/2022] [Indexed: 11/21/2022] Open
Abstract
Arrhythmogenic cardiomyopathy (ACM) is an inherited heart muscle disease caused by heterozygous missense mutations within the gene encoding for the nuclear envelope protein transmembrane protein 43 (TMEM43). The disease is characterized by myocyte loss and fibro-fatty replacement, leading to life-threatening ventricular arrhythmias and sudden cardiac death. However, the role of TMEM43 in the pathogenesis of ACM remains poorly understood. In this study, we generated cardiomyocyte-restricted transgenic zebrafish lines that overexpress eGFP-linked full-length human wild-type (WT) TMEM43 and two genetic variants (c.1073C>T, p.S358L; c.332C>T, p.P111L) using the Tol2-system. Overexpression of WT and p.P111L-mutant TMEM43 was associated with transcriptional activation of the mTOR pathway and ribosome biogenesis, and resulted in enlarged hearts with cardiomyocyte hypertrophy. Intriguingly, mutant p.S358L TMEM43 was found to be unstable and partially redistributed into the cytoplasm in embryonic and adult hearts. Moreover, both TMEM43 variants displayed cardiac morphological defects at juvenile stages and ultrastructural changes within the myocardium, accompanied by dysregulated gene expression profiles in adulthood. Finally, CRISPR/Cas9 mutants demonstrated an age-dependent cardiac phenotype characterized by heart enlargement in adulthood. In conclusion, our findings suggest ultrastructural remodeling and transcriptomic alterations underlying the development of structural and functional cardiac defects in TMEM43-associated cardiomyopathy.
Collapse
Affiliation(s)
- Miriam Zink
- Comprehensive Heart Failure Center, Department of Internal Medicine I, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Anne Seewald
- Comprehensive Heart Failure Center, Department of Internal Medicine I, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Mareike Rohrbach
- Comprehensive Heart Failure Center, Department of Internal Medicine I, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Andreas Brodehl
- Erich and Hanna Klessmann Institute, Heart and Diabetes Center NRW, University Hospital of the Ruhr-University Bochum, 32545 Bad Oeynhausen, Germany
| | - Daniel Liedtke
- Institute for Human Genetics, Biocenter, Julius-Maximilians-University Würzburg, 97074 Würzburg, Germany
| | - Tatjana Williams
- Comprehensive Heart Failure Center, Department of Internal Medicine I, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Sarah J. Childs
- Department of Biochemistry and Molecular Biology, Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Brenda Gerull
- Comprehensive Heart Failure Center, Department of Internal Medicine I, University Hospital Würzburg, 97080 Würzburg, Germany
| |
Collapse
|
29
|
Abstract
Brugada syndrome is a heritable channelopathy characterized by a peculiar electrocardiogram (ECG) pattern and increased risk of cardiac arrhythmias and sudden death. The arrhythmias originate because of an imbalance between the repolarizing and depolarizing currents that modulate the cardiac action potential. Even if an overt structural cardiomyopathy is not typical of Brugada syndrome, fibrosis and structural changes in the right ventricle contribute to a conduction slowing, which ultimately facilitates ventricular arrhythmias. Currently, Mendelian autosomal dominant transmission is detected in less than 25% of all clinical confirmed cases. Although 23 genes have been associated with the condition, only SCN5A, encoding the cardiac sodium channel, is considered clinically actionable and disease causing. The limited monogenic inheritance has pointed toward new perspectives on the possible complex genetic architecture of the disease, involving polygenic inheritance and a polygenic risk score that can influence penetrance and risk stratification. Expected final online publication date for the Annual Review of Genomics and Human Genetics, Volume 23 is October 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Marina Cerrone
- Leon H. Charney Division of Cardiology, Grossman School of Medicine, New York University, New York, NY, USA;
| | - Sarah Costa
- Department of Internal Medicine, Kantonsspital Baden, Baden, Switzerland
| | - Mario Delmar
- Leon H. Charney Division of Cardiology, Grossman School of Medicine, New York University, New York, NY, USA;
| |
Collapse
|
30
|
Zhao M, Han M, Liang L, Song Q, Li X, Du Y, Hu D, Cheng Y, Wang QK, Ke T. Mog1 deficiency promotes cardiac contractile dysfunction and isoproterenol-induced arrhythmias associated with cardiac fibrosis and Cx43 remodeling. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166429. [PMID: 35533905 DOI: 10.1016/j.bbadis.2022.166429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 04/21/2022] [Accepted: 05/02/2022] [Indexed: 10/18/2022]
Abstract
Our earlier studies identified MOG1 as a Nav1.5-binding protein that promotes Nav1.5 intracellular trafficking to plasma membranes. Genetic studies have identified MOG1 variants responsible for cardiac arrhythmias. However, the physiological functions of MOG1 in vivo remain incompletely characterized. In this study, we generated Mog1 knockout (Mog1-/-) mice. Mog1-/- mice did not develop spontaneous arrhythmias at the baseline, but exhibited a prolongation of QRS duration. Mog1-/- mice treated with isoproterenol (ISO), but not with flecainide, exhibited an increased risk of arrhythmias and even sudden death. Mog1-/- mice had normal cardiac morphology, however, LV systolic dysfunction was identified and associated with an increase in ventricular fibrosis. Whole-cell patch-clamping and Western blotting analysis clearly demonstrated the normal cardiac expression and function of Nav1.5 in Mog1-/- mice. Further RNA-seq and iTRAQ analysis identified critical pathways and genes, including extracellular matrix (Mmp2), gap junction (Gja1), and mitochondrial components that were dysregulated in Mog1-/- mice. RT-qPCR, Western blotting, and immunofluorescence assays revealed reduced cardiac expression of Gja1 in Mog1-/- mice. Dye transfer assays confirmed impairment of gap-junction function; Cx43 gap-junction enhancer ZP123 decreased arrhythmia inducibility in ISO-treated Mog1-/- mice. Transmission electron microscopy analysis revealed abnormal sarcomere ultrastructure and altered mitochondrial morphology in Mog1-/- mice. Mitochondrial dynamics was found to be disturbed, and associated with a trend toward increased mitochondrial fusion in Mog1-/- mice. Meanwhile, the level of ATP supply was increased in the hearts of Mog1-/- mice. These results indicate that MOG1 plays an important role in cardiac electrophysiology and cardiac contractile function.
Collapse
Affiliation(s)
- Miao Zhao
- Center for Human Genome Research, The Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Cardio-X Institute, Huazhong University of Science and Technology, Wuhan, 430074, Hubei Province, PR China
| | - Meng Han
- Hubei Provincial Key Laboratory of Occurrence and Intervention of Kidney Diseases, Medical College, Hubei Polytechnic University, Huangshi, Hubei Province, PR China
| | - Lina Liang
- Center for Human Genome Research, The Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Cardio-X Institute, Huazhong University of Science and Technology, Wuhan, 430074, Hubei Province, PR China
| | - Qixue Song
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 518055 Shenzhen, PR China
| | - Xia Li
- Center for Human Genome Research, The Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Cardio-X Institute, Huazhong University of Science and Technology, Wuhan, 430074, Hubei Province, PR China
| | - Yimei Du
- Research Center of Ion Channelopathy, Institute of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, PR China
| | - Dongping Hu
- Center for Human Genome Research, The Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Cardio-X Institute, Huazhong University of Science and Technology, Wuhan, 430074, Hubei Province, PR China
| | - Yu Cheng
- Center for Human Genome Research, The Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Cardio-X Institute, Huazhong University of Science and Technology, Wuhan, 430074, Hubei Province, PR China
| | - Qing K Wang
- Center for Human Genome Research, The Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Cardio-X Institute, Huazhong University of Science and Technology, Wuhan, 430074, Hubei Province, PR China.
| | - Tie Ke
- Center for Human Genome Research, The Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Cardio-X Institute, Huazhong University of Science and Technology, Wuhan, 430074, Hubei Province, PR China.
| |
Collapse
|
31
|
Costa S, Koch K, Gasperetti A, Akdis D, Brunckhorst C, Fu G, Tanner FC, Ruschitzka F, Duru F, Saguner AM. Changes in Exercise Capacity and Ventricular Function in Arrhythmogenic Right Ventricular Cardiomyopathy: The Impact of Sports Restriction during Follow-Up. J Clin Med 2022; 11:1150. [PMID: 35268241 PMCID: PMC8911196 DOI: 10.3390/jcm11051150] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 02/11/2022] [Accepted: 02/12/2022] [Indexed: 02/07/2023] Open
Abstract
(1) Background: Physical exercise has been suggested to promote disease progression in patients with arrhythmogenic right ventricular cardiomyopathy (ARVC). We aimed to investigate the exercise performance and ventricular function of ARVC patients during follow-up, while taking into account their adherence to exercise restriction recommendations. (2) Methods: This retrospective study included 49 patients (33 male, 67%) who had an exercise test at baseline and after 4.2 ± 1.6 years. Of the 49 ARVC patients, 27 (55%) were athletes, while 22 (45%) were non-athletes. Of the athletes, 12 (44%) continued intensive sports activity (non-adherent), while 15 (56%) stopped intensive physical activity upon recommendation (adherent). The maximum workload in Watts (W), percentage of the target workload (W%), and double product (DP) factor were measured for all patients. (3) Results: The non-adherent cohort had a significant decrease in physical performance (W at baseline vs. follow-up, p = 0.012; W% at baseline vs. follow-up, p = 0.025; DP-factor at baseline vs. follow-up, p = 0.012) over time. Left ventricular (LV) function (LV ejection fraction at baseline vs. follow-up, p = 0.082) showed a decreasing trend in the non-adherent cohort, while the performance of the adherent cohort remained at a similar level. (4) Conclusions: If intensive sports activities are not discontinued, exercise capacity and left ventricular function of athletes with ARVC deteriorates during follow-up. All patients with ARVC need to strictly adhere to the recommendation to cease intense sports activity in order to halt disease progression.
Collapse
Affiliation(s)
- Sarah Costa
- Department of Cardiology, University Heart Center Zurich, CH-8091 Zurich, Switzerland; (S.C.); (K.K.); (A.G.); (D.A.); (C.B.); (G.F.); (F.C.T.); (F.R.); (F.D.)
| | - Kristina Koch
- Department of Cardiology, University Heart Center Zurich, CH-8091 Zurich, Switzerland; (S.C.); (K.K.); (A.G.); (D.A.); (C.B.); (G.F.); (F.C.T.); (F.R.); (F.D.)
| | - Alessio Gasperetti
- Department of Cardiology, University Heart Center Zurich, CH-8091 Zurich, Switzerland; (S.C.); (K.K.); (A.G.); (D.A.); (C.B.); (G.F.); (F.C.T.); (F.R.); (F.D.)
| | - Deniz Akdis
- Department of Cardiology, University Heart Center Zurich, CH-8091 Zurich, Switzerland; (S.C.); (K.K.); (A.G.); (D.A.); (C.B.); (G.F.); (F.C.T.); (F.R.); (F.D.)
| | - Corinna Brunckhorst
- Department of Cardiology, University Heart Center Zurich, CH-8091 Zurich, Switzerland; (S.C.); (K.K.); (A.G.); (D.A.); (C.B.); (G.F.); (F.C.T.); (F.R.); (F.D.)
| | - Guan Fu
- Department of Cardiology, University Heart Center Zurich, CH-8091 Zurich, Switzerland; (S.C.); (K.K.); (A.G.); (D.A.); (C.B.); (G.F.); (F.C.T.); (F.R.); (F.D.)
| | - Felix C. Tanner
- Department of Cardiology, University Heart Center Zurich, CH-8091 Zurich, Switzerland; (S.C.); (K.K.); (A.G.); (D.A.); (C.B.); (G.F.); (F.C.T.); (F.R.); (F.D.)
| | - Frank Ruschitzka
- Department of Cardiology, University Heart Center Zurich, CH-8091 Zurich, Switzerland; (S.C.); (K.K.); (A.G.); (D.A.); (C.B.); (G.F.); (F.C.T.); (F.R.); (F.D.)
| | - Firat Duru
- Department of Cardiology, University Heart Center Zurich, CH-8091 Zurich, Switzerland; (S.C.); (K.K.); (A.G.); (D.A.); (C.B.); (G.F.); (F.C.T.); (F.R.); (F.D.)
- Center for Integrative Human Physiology (ZIHP), University of Zurich, CH-8057 Zurich, Switzerland
| | - Ardan M. Saguner
- Department of Cardiology, University Heart Center Zurich, CH-8091 Zurich, Switzerland; (S.C.); (K.K.); (A.G.); (D.A.); (C.B.); (G.F.); (F.C.T.); (F.R.); (F.D.)
| |
Collapse
|
32
|
Daimi H, Lozano-Velasco E, Aranega A, Franco D. Genomic and Non-Genomic Regulatory Mechanisms of the Cardiac Sodium Channel in Cardiac Arrhythmias. Int J Mol Sci 2022; 23:1381. [PMID: 35163304 PMCID: PMC8835759 DOI: 10.3390/ijms23031381] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 12/30/2021] [Accepted: 01/06/2022] [Indexed: 12/19/2022] Open
Abstract
Nav1.5 is the predominant cardiac sodium channel subtype, encoded by the SCN5A gene, which is involved in the initiation and conduction of action potentials throughout the heart. Along its biosynthesis process, Nav1.5 undergoes strict genomic and non-genomic regulatory and quality control steps that allow only newly synthesized channels to reach their final membrane destination and carry out their electrophysiological role. These regulatory pathways are ensured by distinct interacting proteins that accompany the nascent Nav1.5 protein along with different subcellular organelles. Defects on a large number of these pathways have a tremendous impact on Nav1.5 functionality and are thus intimately linked to cardiac arrhythmias. In the present review, we provide current state-of-the-art information on the molecular events that regulate SCN5A/Nav1.5 and the cardiac channelopathies associated with defects in these pathways.
Collapse
Affiliation(s)
- Houria Daimi
- Biochemistry and Molecular Biology Laboratory, Faculty of Pharmacy, University of Monastir, Monastir 5000, Tunisia
| | - Estefanía Lozano-Velasco
- Department of Experimental Biology, University of Jaen, 23071 Jaen, Spain; (E.L.-V.); (A.A.); (D.F.)
- Medina Foundation, Technology Park of Health Sciences, Av. del Conocimiento, 34, 18016 Granada, Spain
| | - Amelia Aranega
- Department of Experimental Biology, University of Jaen, 23071 Jaen, Spain; (E.L.-V.); (A.A.); (D.F.)
- Medina Foundation, Technology Park of Health Sciences, Av. del Conocimiento, 34, 18016 Granada, Spain
| | - Diego Franco
- Department of Experimental Biology, University of Jaen, 23071 Jaen, Spain; (E.L.-V.); (A.A.); (D.F.)
- Medina Foundation, Technology Park of Health Sciences, Av. del Conocimiento, 34, 18016 Granada, Spain
| |
Collapse
|
33
|
Pathology of sudden death, cardiac arrhythmias, and conduction system. Cardiovasc Pathol 2022. [DOI: 10.1016/b978-0-12-822224-9.00007-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
34
|
Pitsch M, Kant S, Mytzka C, Leube RE, Krusche CA. Autophagy and Endoplasmic Reticulum Stress during Onset and Progression of Arrhythmogenic Cardiomyopathy. Cells 2021; 11:96. [PMID: 35011658 PMCID: PMC8750195 DOI: 10.3390/cells11010096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/20/2021] [Accepted: 12/23/2021] [Indexed: 11/16/2022] Open
Abstract
Arrhythmogenic cardiomyopathy (AC) is a heritable, potentially lethal disease without a causal therapy. AC is characterized by focal cardiomyocyte death followed by inflammation and progressive formation of connective tissue. The pathomechanisms leading to structural disease onset and progression, however, are not fully elucidated. Recent studies revealed that dysregulation of autophagy and endoplasmic/sarcoplasmic reticulum (ER/SR) stress plays an important role in cardiac pathophysiology. We therefore examined the temporal and spatial expression patterns of autophagy and ER/SR stress indicators in murine AC models by qRT-PCR, immunohistochemistry, in situ hybridization and electron microscopy. Cardiomyocytes overexpressing the autophagy markers LC3 and SQSTM1/p62 and containing prominent autophagic vacuoles were detected next to regions of inflammation and fibrosis during onset and chronic disease progression. mRNAs of the ER stress markers Chop and sXbp1 were elevated in both ventricles at disease onset. During chronic disease progression Chop mRNA was upregulated in right ventricles. In addition, reduced Ryr2 mRNA expression together with often drastically enlarged ER/SR cisternae further indicated SR dysfunction during this disease phase. Our observations support the hypothesis that locally altered autophagy and enhanced ER/SR stress play a role in AC pathogenesis both at the onset and during chronic progression.
Collapse
Affiliation(s)
| | | | | | - Rudolf E. Leube
- Institute of Molecular and Cellular Anatomy, RWTH Aachen University, Wendlingweg 2, 52074 Aachen, Germany; (M.P.); (S.K.); (C.M.)
| | - Claudia A. Krusche
- Institute of Molecular and Cellular Anatomy, RWTH Aachen University, Wendlingweg 2, 52074 Aachen, Germany; (M.P.); (S.K.); (C.M.)
| |
Collapse
|
35
|
Moazzen H, Venger K, Kant S, Leube RE, Krusche CA. Desmoglein 2 regulates cardiogenesis by restricting hematopoiesis in the developing murine heart. Sci Rep 2021; 11:21687. [PMID: 34737300 PMCID: PMC8569146 DOI: 10.1038/s41598-021-00996-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 10/21/2021] [Indexed: 02/05/2023] Open
Abstract
Cardiac morphogenesis relies on intricate intercellular signaling. Altered signaling impacts cardiac function and is detrimental to embryonic survival. Here we report an unexpected regulatory role of the desmosomal cell adhesion molecule desmoglein 2 (Dsg2) on murine heart development. A large percentage of Dsg2-mutant embryos develop pericardial hemorrhage. Lethal myocardial rupture is occasionally observed, which is not associated with loss of cardiomyocyte contact but with expansion of abnormal, non-myocyte cell clusters within the myocardial wall. Two types of abnormal cell clusters can be distinguished: Type A clusters involve endocard-associated, round-shaped CD31+ cells, which proliferate and invade the myocardium. They acquire Runx1- and CD44-positivity indicating a shift towards a hematopoietic phenotype. Type B clusters expand subepicardially and next to type A clusters. They consist primarily of Ter119+ erythroid cells with interspersed Runx1+/CD44+ cells suggesting that they originate from type A cell clusters. The observed pericardial hemorrhage is caused by migration of erythrocytes from type B clusters through the epicardium and rupture of the altered cardiac wall. Finally, evidence is presented that structural defects of Dsg2-depleted cardiomyocytes are primary to the observed pathogenesis. We propose that cardiomyocyte-driven paracrine signaling, which likely involves Notch1, directs subsequent trans-differentiation of endo- and epicardial cells. Together, our observations uncover a hitherto unknown regulatory role of Dsg2 in cardiogenesis.
Collapse
Affiliation(s)
- Hoda Moazzen
- Institute of Molecular and Cellular Anatomy, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Kateryna Venger
- Institute of Molecular and Cellular Anatomy, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Sebastian Kant
- Institute of Molecular and Cellular Anatomy, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Rudolf E Leube
- Institute of Molecular and Cellular Anatomy, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany.
| | - Claudia A Krusche
- Institute of Molecular and Cellular Anatomy, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany.
| |
Collapse
|
36
|
Zhang J, Liang Y, Bradford WH, Sheikh F. Desmosomes: emerging pathways and non-canonical functions in cardiac arrhythmias and disease. Biophys Rev 2021; 13:697-706. [PMID: 34765046 PMCID: PMC8555023 DOI: 10.1007/s12551-021-00829-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 08/12/2021] [Indexed: 12/14/2022] Open
Abstract
Desmosomes are critical adhesion structures in cardiomyocytes, with mutation/loss linked to the heritable cardiac disease, arrhythmogenic right ventricular cardiomyopathy (ARVC). Early studies revealed the ability of desmosomal protein loss to trigger ARVC disease features including structural remodeling, arrhythmias, and inflammation; however, the precise mechanisms contributing to diverse disease presentations are not fully understood. Recent mechanistic studies demonstrated the protein degradation component CSN6 is a resident cardiac desmosomal protein which selectively restricts cardiomyocyte desmosomal degradation and disease. This suggests defects in protein degradation can trigger the structural remodeling underlying ARVC. Additionally, a subset of ARVC-related mutations show enhanced vulnerability to calpain-mediated degradation, further supporting the relevance of these mechanisms in disease. Desmosomal gene mutations/loss has been shown to impact arrhythmogenic pathways in the absence of structural disease within ARVC patients and model systems. Studies have shown the involvement of connexins, calcium handling machinery, and sodium channels as early drivers of arrhythmias, suggesting these may be distinct pathways regulating electrical function from the desmosome. Emerging evidence has suggested inflammation may be an early mechanism in disease pathogenesis, as clinical reports have shown an overlap between myocarditis and ARVC. Recent studies focus on the association between desmosomal mutations/loss and inflammatory processes including autoantibodies and signaling pathways as a way to understand the involvement of inflammation in ARVC pathogenesis. A specific focus will be to dissect ongoing fields of investigation to highlight diverse pathogenic pathways associated with desmosomal mutations/loss.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093 USA
| | - Yan Liang
- Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093 USA
| | - William H. Bradford
- Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093 USA
| | - Farah Sheikh
- Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093 USA
| |
Collapse
|
37
|
Ng KE, Delaney PJ, Thenet D, Murtough S, Webb CM, Zaman N, Tsisanova E, Mastroianni G, Walker SLM, Westaby JD, Pennington DJ, Pink R, Kelsell DP, Tinker A. Early inflammation precedes cardiac fibrosis and heart failure in desmoglein 2 murine model of arrhythmogenic cardiomyopathy. Cell Tissue Res 2021; 386:79-98. [PMID: 34236518 PMCID: PMC8526453 DOI: 10.1007/s00441-021-03488-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 06/18/2021] [Indexed: 12/19/2022]
Abstract
The study of a desmoglein 2 murine model of arrhythmogenic cardiomyopathy revealed cardiac inflammation as a key early event leading to fibrosis. Arrhythmogenic cardiomyopathy (AC) is an inherited heart muscle disorder leading to ventricular arrhythmias and heart failure due to abnormalities in the cardiac desmosome. We examined how loss of desmoglein 2 (Dsg2) in the young murine heart leads to development of AC. Apoptosis was an early cellular phenotype, and RNA sequencing analysis revealed early activation of inflammatory-associated pathways in Dsg2-null (Dsg2-/-) hearts at postnatal day 14 (2 weeks) that were absent in the fibrotic heart of adult mice (10 weeks). This included upregulation of iRhom2/ADAM17 and its associated pro-inflammatory cytokines and receptors such as TNFα, IL6R and IL-6. Furthermore, genes linked to specific macrophage populations were also upregulated. This suggests cardiomyocyte stress triggers an early immune response to clear apoptotic cells allowing tissue remodelling later on in the fibrotic heart. Our analysis at the early disease stage suggests cardiac inflammation is an important response and may be one of the mechanisms responsible for AC disease progression.
Collapse
Affiliation(s)
- K E Ng
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK.,Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, E1 2AT, UK
| | - P J Delaney
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, E1 2AT, UK
| | - D Thenet
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, E1 2AT, UK
| | - S Murtough
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, E1 2AT, UK
| | - C M Webb
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, E1 2AT, UK
| | - N Zaman
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, E1 2AT, UK
| | - E Tsisanova
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - G Mastroianni
- School of Biological and Chemical Sciences, Queen Mary University of London, Mile End Road, London, E1 4NS, UK
| | - S L M Walker
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - J D Westaby
- CRY Dept. of Cardiovascular Pathology, Cardiology Clinical Academic Group, Molecular and Clinical Sciences Research Institute, St. George's University of London, Jenner WingCranmer Terrace, London, SW17 0RE, UK
| | - D J Pennington
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, E1 2AT, UK
| | - R Pink
- Department of Biological and Medical Sciences, Faculty of Health and Life Sciences, Oxford Brookes University, Headington Campus, Oxford, OX3 0BP, UK
| | - D P Kelsell
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, E1 2AT, UK.
| | - A Tinker
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK.
| |
Collapse
|
38
|
Gerull B, Brodehl A. Insights Into Genetics and Pathophysiology of Arrhythmogenic Cardiomyopathy. Curr Heart Fail Rep 2021; 18:378-390. [PMID: 34478111 PMCID: PMC8616880 DOI: 10.1007/s11897-021-00532-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/09/2021] [Indexed: 02/07/2023]
Abstract
Purpose of Review Arrhythmogenic cardiomyopathy (ACM) is a genetic disease characterized by life-threatening ventricular arrhythmias and sudden cardiac death (SCD) in apparently healthy young adults. Mutations in genes encoding for cellular junctions can be found in about half of the patients. However, disease onset and severity, risk of arrhythmias, and outcome are highly variable and drug-targeted treatment is currently unavailable. Recent Findings This review focuses on advances in clinical risk stratification, genetic etiology, and pathophysiological concepts. The desmosome is the central part of the disease, but other intercalated disc and associated structural proteins not only broaden the genetic spectrum but also provide novel molecular and cellular insights into the pathogenesis of ACM. Signaling pathways and the role of inflammation will be discussed and targets for novel therapeutic approaches outlined. Summary Genetic discoveries and experimental-driven preclinical research contributed significantly to the understanding of ACM towards mutation- and pathway-specific personalized medicine.
Collapse
Affiliation(s)
- Brenda Gerull
- Comprehensive Heart Failure Center (CHFC), Department of Medicine I, University Clinic Würzburg, Am Schwarzenberg 15, 97078, Würzburg, Germany.
| | - Andreas Brodehl
- Heart and Diabetes Center NRW, Erich and Hanna Klessmann Institute, University Hospital of the Ruhr-University Bochum, Georgstrasse 11, 32545, Bad Oeynhausen, Germany
| |
Collapse
|
39
|
Rivaud MR, Delmar M, Remme CA. Heritable arrhythmia syndromes associated with abnormal cardiac sodium channel function: ionic and non-ionic mechanisms. Cardiovasc Res 2021; 116:1557-1570. [PMID: 32251506 PMCID: PMC7341171 DOI: 10.1093/cvr/cvaa082] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 03/01/2020] [Accepted: 04/01/2020] [Indexed: 12/19/2022] Open
Abstract
The cardiac sodium channel NaV1.5, encoded by the SCN5A gene, is responsible for the fast upstroke of the action potential. Mutations in SCN5A may cause sodium channel dysfunction by decreasing peak sodium current, which slows conduction and facilitates reentry-based arrhythmias, and by enhancing late sodium current, which prolongs the action potential and sets the stage for early afterdepolarization and arrhythmias. Yet, some NaV1.5-related disorders, in particular structural abnormalities, cannot be directly or solely explained on the basis of defective NaV1.5 expression or biophysics. An emerging concept that may explain the large disease spectrum associated with SCN5A mutations centres around the multifunctionality of the NaV1.5 complex. In this alternative view, alterations in NaV1.5 affect processes that are independent of its canonical ion-conducting role. We here propose a novel classification of NaV1.5 (dys)function, categorized into (i) direct ionic effects of sodium influx through NaV1.5 on membrane potential and consequent action potential generation, (ii) indirect ionic effects of sodium influx on intracellular homeostasis and signalling, and (iii) non-ionic effects of NaV1.5, independent of sodium influx, through interactions with macromolecular complexes within the different microdomains of the cardiomyocyte. These indirect ionic and non-ionic processes may, acting alone or in concert, contribute significantly to arrhythmogenesis. Hence, further exploration of these multifunctional effects of NaV1.5 is essential for the development of novel preventive and therapeutic strategies.
Collapse
Affiliation(s)
- Mathilde R Rivaud
- Department of Clinical and Experimental Cardiology, Heart Center, Amsterdam UMC (location AMC), University of Amsterdam, Amsterdam Cardiovascular Sciences, Meigberdreef 15, 1105AZ Amsterdam, The Netherlands
| | - Mario Delmar
- The Leon H. Charney Division of Cardiology, New York University School of Medicine, 435 E 30th St, NSB 707, New York, NY 10016, USA
| | - Carol Ann Remme
- Department of Clinical and Experimental Cardiology, Heart Center, Amsterdam UMC (location AMC), University of Amsterdam, Amsterdam Cardiovascular Sciences, Meigberdreef 15, 1105AZ Amsterdam, The Netherlands
| |
Collapse
|
40
|
van der Voorn SM, Te Riele ASJM, Basso C, Calkins H, Remme CA, van Veen TAB. Arrhythmogenic cardiomyopathy: pathogenesis, pro-arrhythmic remodelling, and novel approaches for risk stratification and therapy. Cardiovasc Res 2021; 116:1571-1584. [PMID: 32246823 PMCID: PMC7526754 DOI: 10.1093/cvr/cvaa084] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 03/10/2020] [Accepted: 03/30/2020] [Indexed: 02/07/2023] Open
Abstract
Arrhythmogenic cardiomyopathy (ACM) is a life-threatening cardiac disease caused by mutations in genes predominantly encoding for desmosomal proteins that lead to alterations in the molecular composition of the intercalated disc. ACM is characterized by progressive replacement of cardiomyocytes by fibrofatty tissue, ventricular dilatation, cardiac dysfunction, and heart failure but mostly dominated by the occurrence of life-threatening arrhythmias and sudden cardiac death (SCD). As SCD appears mostly in apparently healthy young individuals, there is a demand for better risk stratification of suspected ACM mutation carriers. Moreover, disease severity, progression, and outcome are highly variable in patients with ACM. In this review, we discuss the aetiology of ACM with a focus on pro-arrhythmic disease mechanisms in the early concealed phase of the disease. We summarize potential new biomarkers which might be useful for risk stratification and prediction of disease course. Finally, we explore novel therapeutic strategies to prevent arrhythmias and SCD in the early stages of ACM.
Collapse
Affiliation(s)
- Stephanie M van der Voorn
- Division of Heart and Lungs, Department of Medical Physiology, University Medical Center Utrecht, PO Box 85060, Utrecht 3508 AB, The Netherlands
| | - Anneline S J M Te Riele
- Division of Heart and Lungs, Department of Cardiology, University Medical Center Utrecht, PO Box 85060, Utrecht 3508 AB, The Netherlands
| | - Cristina Basso
- Cardiovascular Pathology Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padua Medical School, Via A. Gabelli, 61 35121 Padova, Italy
| | - Hugh Calkins
- Johns Hopkins Hospital, Sheikh Zayed Tower 7125R, Baltimore, MD 21287, USA
| | - Carol Ann Remme
- Department of Clinical and Experimental Cardiology, Heart Centre, Amsterdam UMC, Location Academic Medical Center, University of Amsterdam, Amsterdam 1105AZ, The Netherlands
| | - Toon A B van Veen
- Division of Heart and Lungs, Department of Medical Physiology, University Medical Center Utrecht, PO Box 85060, Utrecht 3508 AB, The Netherlands
| |
Collapse
|
41
|
Odening KE, Gomez AM, Dobrev D, Fabritz L, Heinzel FR, Mangoni ME, Molina CE, Sacconi L, Smith G, Stengl M, Thomas D, Zaza A, Remme CA, Heijman J. ESC working group on cardiac cellular electrophysiology position paper: relevance, opportunities, and limitations of experimental models for cardiac electrophysiology research. Europace 2021; 23:1795-1814. [PMID: 34313298 DOI: 10.1093/europace/euab142] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 05/19/2021] [Indexed: 12/19/2022] Open
Abstract
Cardiac arrhythmias are a major cause of death and disability. A large number of experimental cell and animal models have been developed to study arrhythmogenic diseases. These models have provided important insights into the underlying arrhythmia mechanisms and translational options for their therapeutic management. This position paper from the ESC Working Group on Cardiac Cellular Electrophysiology provides an overview of (i) currently available in vitro, ex vivo, and in vivo electrophysiological research methodologies, (ii) the most commonly used experimental (cellular and animal) models for cardiac arrhythmias including relevant species differences, (iii) the use of human cardiac tissue, induced pluripotent stem cell (hiPSC)-derived and in silico models to study cardiac arrhythmias, and (iv) the availability, relevance, limitations, and opportunities of these cellular and animal models to recapitulate specific acquired and inherited arrhythmogenic diseases, including atrial fibrillation, heart failure, cardiomyopathy, myocarditis, sinus node, and conduction disorders and channelopathies. By promoting a better understanding of these models and their limitations, this position paper aims to improve the quality of basic research in cardiac electrophysiology, with the ultimate goal to facilitate the clinical translation and application of basic electrophysiological research findings on arrhythmia mechanisms and therapies.
Collapse
Affiliation(s)
- Katja E Odening
- Translational Cardiology, Department of Cardiology, Inselspital, Bern University Hospital, Bern, Switzerland.,Institute of Physiology, University of Bern, Bern, Switzerland
| | - Ana-Maria Gomez
- Signaling and cardiovascular pathophysiology-UMR-S 1180, Inserm, Université Paris-Saclay, 92296 Châtenay-Malabry, France
| | - Dobromir Dobrev
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany
| | - Larissa Fabritz
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK.,Department of Cardiology, University Hospital Birmingham NHS Trust, Birmingham, UK
| | - Frank R Heinzel
- Department of Internal Medicine and Cardiology, Charité - Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site, Berlin, Germany
| | - Matteo E Mangoni
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Cristina E Molina
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,German Centre for Cardiovascular Research (DZHK), Partner Site, Hamburg/Kiel/Lübeck, Germany
| | - Leonardo Sacconi
- National Institute of Optics and European Laboratory for Non Linear Spectroscopy, Italy.,Institute for Experimental Cardiovascular Medicine, University Freiburg, Germany
| | - Godfrey Smith
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, UK
| | - Milan Stengl
- Department of Physiology, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| | - Dierk Thomas
- Department of Cardiology, University Hospital Heidelberg, Heidelberg, Germany; Heidelberg Center for Heart Rhythm Disorders (HCR), University Hospital Heidelberg, Heidelberg, Germany.,German Centre for Cardiovascular Research (DZHK), Partner Site, Heidelberg/Mannheim, Germany
| | - Antonio Zaza
- Department of Biotechnology and Bioscience, University of Milano-Bicocca, Milano, Italy
| | - Carol Ann Remme
- Department of Experimental Cardiology, Amsterdam UMC, location AMC, Amsterdam, The Netherlands
| | - Jordi Heijman
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
42
|
Marchal GA, Jouni M, Chiang DY, Pérez-Hernández M, Podliesna S, Yu N, Casini S, Potet F, Veerman CC, Klerk M, Lodder EM, Mengarelli I, Guan K, Vanoye CG, Rothenberg E, Charpentier F, Redon R, George AL, Verkerk AO, Bezzina CR, MacRae CA, Burridge PW, Delmar M, Galjart N, Portero V, Remme CA. Targeting the Microtubule EB1-CLASP2 Complex Modulates Na V1.5 at Intercalated Discs. Circ Res 2021; 129:349-365. [PMID: 34092082 PMCID: PMC8298292 DOI: 10.1161/circresaha.120.318643] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Gerard A Marchal
- Department of Experimental Cardiology, Amsterdam UMC - location AMC, The Netherlands (G.A.M., S.P., S.C., C.C.V., E.M.L., I.M., A.O.V., C.R.B., V.P., C.A.R.)
| | - Mariam Jouni
- Department of Pharmacology, University Feinberg School of Medicine, Chicago, IL (M.J., F.P., C.G.V., A.L.G., P.W.B.)
| | - David Y Chiang
- Brigham and Women's Hospital and Harvard Medical School, Boston, MA (D.Y.C., C.A.M.)
| | | | - Svitlana Podliesna
- Department of Experimental Cardiology, Amsterdam UMC - location AMC, The Netherlands (G.A.M., S.P., S.C., C.C.V., E.M.L., I.M., A.O.V., C.R.B., V.P., C.A.R.)
| | - Nuo Yu
- Department of Cell Biology, Erasmus Medical Centre Rotterdam, The Netherlands (N.Y., N.G.)
| | - Simona Casini
- Department of Experimental Cardiology, Amsterdam UMC - location AMC, The Netherlands (G.A.M., S.P., S.C., C.C.V., E.M.L., I.M., A.O.V., C.R.B., V.P., C.A.R.)
| | - Franck Potet
- Department of Pharmacology, University Feinberg School of Medicine, Chicago, IL (M.J., F.P., C.G.V., A.L.G., P.W.B.)
| | - Christiaan C Veerman
- Department of Experimental Cardiology, Amsterdam UMC - location AMC, The Netherlands (G.A.M., S.P., S.C., C.C.V., E.M.L., I.M., A.O.V., C.R.B., V.P., C.A.R.)
| | - Mischa Klerk
- Department of Medical Biology, Amsterdam UMC - location AMC, The Netherlands (M.K., A.O.V.)
| | - Elisabeth M Lodder
- Department of Experimental Cardiology, Amsterdam UMC - location AMC, The Netherlands (G.A.M., S.P., S.C., C.C.V., E.M.L., I.M., A.O.V., C.R.B., V.P., C.A.R.)
| | - Isabella Mengarelli
- Department of Experimental Cardiology, Amsterdam UMC - location AMC, The Netherlands (G.A.M., S.P., S.C., C.C.V., E.M.L., I.M., A.O.V., C.R.B., V.P., C.A.R.)
| | - Kaomei Guan
- Institute of Pharmacology and Toxicology, Technische Universität Dresden, Germany (K.G.)
| | - Carlos G Vanoye
- Department of Pharmacology, University Feinberg School of Medicine, Chicago, IL (M.J., F.P., C.G.V., A.L.G., P.W.B.)
| | - Eli Rothenberg
- Department of Biochemistry and Pharmacology (E.R.), NYU School of Medicine
| | - Flavien Charpentier
- Université de Nantes, CNRS, INSERM, l'institut du Thorax, Nantes, France (F.C., R.R., V.P.)
| | - Richard Redon
- Université de Nantes, CNRS, INSERM, l'institut du Thorax, Nantes, France (F.C., R.R., V.P.)
| | - Alfred L George
- Department of Pharmacology, University Feinberg School of Medicine, Chicago, IL (M.J., F.P., C.G.V., A.L.G., P.W.B.)
| | - Arie O Verkerk
- Department of Experimental Cardiology, Amsterdam UMC - location AMC, The Netherlands (G.A.M., S.P., S.C., C.C.V., E.M.L., I.M., A.O.V., C.R.B., V.P., C.A.R.)
- Department of Medical Biology, Amsterdam UMC - location AMC, The Netherlands (M.K., A.O.V.)
| | - Connie R Bezzina
- Department of Experimental Cardiology, Amsterdam UMC - location AMC, The Netherlands (G.A.M., S.P., S.C., C.C.V., E.M.L., I.M., A.O.V., C.R.B., V.P., C.A.R.)
| | - Calum A MacRae
- Brigham and Women's Hospital and Harvard Medical School, Boston, MA (D.Y.C., C.A.M.)
| | - Paul W Burridge
- Department of Pharmacology, University Feinberg School of Medicine, Chicago, IL (M.J., F.P., C.G.V., A.L.G., P.W.B.)
| | - Mario Delmar
- Division of Cardiology (M.P.-H., M.D.), NYU School of Medicine
| | - Niels Galjart
- Department of Cell Biology, Erasmus Medical Centre Rotterdam, The Netherlands (N.Y., N.G.)
| | - Vincent Portero
- Department of Experimental Cardiology, Amsterdam UMC - location AMC, The Netherlands (G.A.M., S.P., S.C., C.C.V., E.M.L., I.M., A.O.V., C.R.B., V.P., C.A.R.)
- Université de Nantes, CNRS, INSERM, l'institut du Thorax, Nantes, France (F.C., R.R., V.P.)
| | - Carol Ann Remme
- Department of Experimental Cardiology, Amsterdam UMC - location AMC, The Netherlands (G.A.M., S.P., S.C., C.C.V., E.M.L., I.M., A.O.V., C.R.B., V.P., C.A.R.)
| |
Collapse
|
43
|
Hawthorne RN, Blazeski A, Lowenthal J, Kannan S, Teuben R, DiSilvestre D, Morrissette-McAlmon J, Saffitz JE, Boheler KR, James CA, Chelko SP, Tomaselli G, Tung L. Altered Electrical, Biomolecular, and Immunologic Phenotypes in a Novel Patient-Derived Stem Cell Model of Desmoglein-2 Mutant ARVC. J Clin Med 2021; 10:jcm10143061. [PMID: 34300226 PMCID: PMC8306340 DOI: 10.3390/jcm10143061] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 07/05/2021] [Accepted: 07/08/2021] [Indexed: 12/27/2022] Open
Abstract
Arrhythmogenic right ventricular cardiomyopathy (ARVC) is a progressive heart condition which causes fibro-fatty myocardial scarring, ventricular arrhythmias, and sudden cardiac death. Most cases of ARVC can be linked to pathogenic mutations in the cardiac desmosome, but the pathophysiology is not well understood, particularly in early phases when arrhythmias can develop prior to structural changes. Here, we created a novel human induced pluripotent stem cell-derived cardiomyocyte (hiPSC-CM) model of ARVC from a patient with a c.2358delA variant in desmoglein-2 (DSG2). These DSG2-mutant (DSG2Mut) hiPSC-CMs were compared against two wildtype hiPSC-CM lines via immunostaining, RT-qPCR, Western blot, RNA-Seq, cytokine expression and optical mapping. Mutant cells expressed reduced DSG2 mRNA and had altered localization of desmoglein-2 protein alongside thinner, more disorganized myofibrils. No major changes in other desmosomal proteins were noted. There was increased pro-inflammatory cytokine expression that may be linked to canonical and non-canonical NFκB signaling. Action potentials in DSG2Mut CMs were shorter with increased upstroke heterogeneity, while time-to-peak calcium and calcium decay rate were reduced. These were accompanied by changes in ion channel and calcium handling gene expression. Lastly, suppressing DSG2 in control lines via siRNA allowed partial recapitulation of electrical anomalies noted in DSG2Mut cells. In conclusion, the aberrant cytoskeletal organization, cytokine expression, and electrophysiology found DSG2Mut hiPSC-CMs could underlie early mechanisms of disease manifestation in ARVC patients.
Collapse
Affiliation(s)
- Robert N. Hawthorne
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA; (R.N.H.); (A.B.); (J.L.); (S.K.); (R.T.); (J.M.-M.); (K.R.B.)
- Medical Scientist Training Program, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Adriana Blazeski
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA; (R.N.H.); (A.B.); (J.L.); (S.K.); (R.T.); (J.M.-M.); (K.R.B.)
| | - Justin Lowenthal
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA; (R.N.H.); (A.B.); (J.L.); (S.K.); (R.T.); (J.M.-M.); (K.R.B.)
- Medical Scientist Training Program, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Suraj Kannan
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA; (R.N.H.); (A.B.); (J.L.); (S.K.); (R.T.); (J.M.-M.); (K.R.B.)
- Medical Scientist Training Program, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Roald Teuben
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA; (R.N.H.); (A.B.); (J.L.); (S.K.); (R.T.); (J.M.-M.); (K.R.B.)
| | - Deborah DiSilvestre
- Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA; (D.D.); (C.A.J.)
| | - Justin Morrissette-McAlmon
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA; (R.N.H.); (A.B.); (J.L.); (S.K.); (R.T.); (J.M.-M.); (K.R.B.)
| | - Jeffrey E. Saffitz
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA;
| | - Kenneth R. Boheler
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA; (R.N.H.); (A.B.); (J.L.); (S.K.); (R.T.); (J.M.-M.); (K.R.B.)
- Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA; (D.D.); (C.A.J.)
| | - Cynthia A. James
- Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA; (D.D.); (C.A.J.)
| | - Stephen P. Chelko
- Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA; (D.D.); (C.A.J.)
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, FL 32306, USA
- Correspondence: (S.P.C.); (G.T.); (L.T.); Tel.: +1-850-644-2215 (S.P.C.); +1-718-430-2801 (G.T.); +1-410-955-9603 (L.T.)
| | - Gordon Tomaselli
- Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA; (D.D.); (C.A.J.)
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Correspondence: (S.P.C.); (G.T.); (L.T.); Tel.: +1-850-644-2215 (S.P.C.); +1-718-430-2801 (G.T.); +1-410-955-9603 (L.T.)
| | - Leslie Tung
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA; (R.N.H.); (A.B.); (J.L.); (S.K.); (R.T.); (J.M.-M.); (K.R.B.)
- Correspondence: (S.P.C.); (G.T.); (L.T.); Tel.: +1-850-644-2215 (S.P.C.); +1-718-430-2801 (G.T.); +1-410-955-9603 (L.T.)
| |
Collapse
|
44
|
Vite A, Gandjbakhch E, Hery T, Fressart V, Gary F, Simon F, Varnous S, Hidden Lucet F, Charron P, Villard E. Desmoglein-2 mutations in propeptide cleavage-site causes arrhythmogenic right ventricular cardiomyopathy/dysplasia by impairing extracellular 1-dependent desmosomal interactions upon cellular stress. Europace 2021; 22:320-329. [PMID: 31845994 DOI: 10.1093/europace/euz329] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 11/18/2019] [Indexed: 02/04/2023] Open
Abstract
AIMS Desmoglein-2 (DSG2) mutations, which encode a heart-specific cadherin crucial for desmosomal adhesion, are frequent in arrhythmogenic right ventricular cardiomyopathy/dysplasia (ARVC/D). DSG2 mutations have been associated with higher risk of biventricular involvement. Among DSG2 mutations, mutations of the inhibitory propeptide consensus cleavage-site (Arg-X-Arg/Lys-Arg), are particularly frequent. In the present work, we explored the functional consequences of DSG2 propeptide cleavage site mutations p.Arg49His, p.Arg46Trp, and p.Arg46Gln on localization, adhesive properties, and desmosome incorporation of DSG2. METHODS AND RESULTS We studied the expression of mutant-DSG2 in human heart and in epithelial and cardiac cellular models expressing wild-type or mutant (p.Arg49His, p.Arg46Trp, and p.Arg46Gln) proDSG2-GFP fusion proteins. The consequences of the p.Arg46Trp mutation on DSG2 adhesiveness were studied by surface plasmon resonance. Incorporation of mutant p.Arg46Trp DSG2 into desmosomes was studied under low-calcium culture conditions and cyclic mechanical stress. We demonstrated in human heart and cellular models that all three mutations prevented N-terminal propeptide cleavage, but did not modify intercellular junction targeting. Surface plasmon resonance experiments showed a propeptide-dependent loss of interaction between the cadherin N-terminal extracellular 1 (EC1) domains. Additionally, proDSG2 mutant proteins were abnormally incorporated into desmosomes under low-calcium culture conditions or following mechanical stress. This was accompanied by an epidermal growth factor receptor-dependent internalization of proDSG2, suggesting increased turnover of unprocessed proDSG2. CONCLUSION Our results strongly suggest weakened desmosomal adhesiveness due to abnormal incorporation of uncleaved mutant proDSG2 in cellular stress conditions. These results provide new insights into desmosomal cadherin regulation and ARVC/D pathophysiology, in particular, the potential role of mechanical stress on desmosomal dysfunction.
Collapse
Affiliation(s)
- Alexia Vite
- Sorbonne Université, Faculté de médecine Pitié-Salpêtrière, INSERM U1166, IHU ICAN, F-75013 Paris, France
| | - Estelle Gandjbakhch
- Sorbonne Université, Faculté de médecine Pitié-Salpêtrière, INSERM U1166, IHU ICAN, F-75013 Paris, France.,Département de Cardiologie, AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Paris, France.,Centre de référence des maladies cardiaques héréditaires, AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Paris, France
| | - Tiphaine Hery
- Sorbonne Université, Faculté de médecine Pitié-Salpêtrière, INSERM U1166, IHU ICAN, F-75013 Paris, France
| | - Veronique Fressart
- Service de Biochimie Métabolique, AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Unité de Cardiogénétique et Myogénétique, Paris, France
| | - Francoise Gary
- Sorbonne Université, Faculté de médecine Pitié-Salpêtrière, INSERM U1166, IHU ICAN, F-75013 Paris, France
| | - Francoise Simon
- Service de Biochimie Métabolique, AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Unité de Cardiogénétique et Myogénétique, Paris, France
| | - Shaida Varnous
- Département de Chirurgie Cardio-thoracique, AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Paris, France
| | | | - Philippe Charron
- Sorbonne Université, Faculté de médecine Pitié-Salpêtrière, INSERM U1166, IHU ICAN, F-75013 Paris, France.,Département de Cardiologie, AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Paris, France.,Centre de référence des maladies cardiaques héréditaires, AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Paris, France.,Département de Génétique, AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Paris, France
| | - Eric Villard
- Sorbonne Université, Faculté de médecine Pitié-Salpêtrière, INSERM U1166, IHU ICAN, F-75013 Paris, France
| |
Collapse
|
45
|
Zhu Y, Shamblin I, Rodriguez E, Salzer GE, Araysi L, Margolies KA, Halade GV, Litovsky SH, Pogwizd S, Gray M, Huke S. Progressive cardiac arrhythmias and ECG abnormalities in the Huntington's disease BACHD mouse model. Hum Mol Genet 2021; 29:369-381. [PMID: 31816043 DOI: 10.1093/hmg/ddz295] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 11/11/2019] [Accepted: 12/05/2019] [Indexed: 02/03/2023] Open
Abstract
Huntington's disease (HD) is a dominantly inherited neurodegenerative disease. There is accumulating evidence that HD patients have increased prevalence of conduction abnormalities and compromised sinoatrial node function which could lead to increased risk for arrhythmia. We used mutant Huntingtin (mHTT) expressing bacterial artificial chromosome Huntington's disease mice to determine if they exhibit electrocardiogram (ECG) abnormalities involving cardiac conduction that are known to increase risk of sudden arrhythmic death in humans. We obtained surface ECGs and analyzed arrhythmia susceptibility; we observed prolonged QRS duration, increases in PVCs as well as PACs. Abnormal histological and structural changes that could lead to cardiac conduction system dysfunction were seen. Finally, we observed decreases in desmosomal proteins, plakophilin-2 and desmoglein-2, which have been reported to cause cardiac arrhythmias and reduced conduction. Our study indicates that mHTT could cause progressive cardiac conduction system pathology that could increase the susceptibility to arrhythmias and sudden cardiac death in HD patients.
Collapse
Affiliation(s)
- Yujie Zhu
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Isaac Shamblin
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Efrain Rodriguez
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Grace E Salzer
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Lita Araysi
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Katherine A Margolies
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Ganesh V Halade
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Silvio H Litovsky
- Department of Pathology, Division of Anatomic Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Steven Pogwizd
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Michelle Gray
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Sabine Huke
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
46
|
Cha MJ, Seo JW, Kim HJ, Kim MK, Yoon HS, Jo SW, Oh S, Chang JH. Early Changes in Rat Heart After High-Dose Irradiation: Implications for Antiarrhythmic Effects of Cardiac Radioablation. J Am Heart Assoc 2021; 10:e019072. [PMID: 33660526 PMCID: PMC8174197 DOI: 10.1161/jaha.120.019072] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Background Noninvasive cardiac radioablation is employed to treat ventricular arrhythmia. However, myocardial changes leading to early‐period antiarrhythmic effects induced by high‐dose irradiation are unknown. This study investigated dose‐responsive histologic, ultrastructural, and functional changes within 1 month after irradiation in rat heart. Methods and Results Whole hearts of wild‐type Lewis rats (N=95) were irradiated with single fraction 20, 25, 30, 40, or 50 Gy and explanted at 1 day or 1, 2, 3, or 4 weeks’ postirradiation. Microscopic pathologic changes of cardiac structures by light microscope with immunohistopathologic staining, ultrastructure by electron microscopy, and functional evaluation by ECG and echocardiography were studied. Despite high‐dose irradiation, no myocardial necrosis and apoptosis were observed. Intercalated discs were widened and disrupted, forming uneven and twisted junctions between adjacent myocytes. Diffuse vacuolization peaked at 3 weeks, suggesting irradiation dose‐responsiveness, which was correlated with interstitial and intracellular edema. CD68 immunostaining accompanying vacuolization suggested mononuclear cell infiltration. These changes were prominent in working myocardium but not cardiac conduction tissue. Intracardiac conduction represented by PR and QTc intervals on ECG was delayed compared with baseline measurements. ST segment was initially depressed and gradually elevated. Ventricular chamber dimensions and function remained intact without pericardial effusion. Conclusions Mononuclear cell–related intracellular and extracellular edema with diffuse vacuolization and intercalated disc widening were observed within 1 month after high‐dose irradiation. ECG indicated intracardiac conduction delay with prominent ST‐segment changes. These observations suggest that early antiarrhythmic effects after cardiac radioablation result from conduction disturbances and membrane potential alterations without necrosis.
Collapse
Affiliation(s)
- Myung-Jin Cha
- Division of Cardiology Department of Internal Medicine Seoul National University Hospital Seoul South Korea
| | - Jeong-Wook Seo
- Departments of Pathology Seoul National University Hospital Seoul South Korea
| | - Hak Jae Kim
- Department of Radiation Oncology Seoul National University College of Medicine Seoul Korea.,Department of Radiation Oncology Seoul National University Hospital Seoul South Korea.,Cancer Research InstituteSeoul National University College of Medicine Seoul Korea
| | - Moo-Kang Kim
- Division of Cardiology Department of Internal Medicine Seoul National University Hospital Seoul South Korea
| | - Hye-Sun Yoon
- Division of Cardiology Department of Internal Medicine Seoul National University Hospital Seoul South Korea
| | - Seong Won Jo
- Seoul National University College of Medicine Seoul Korea
| | - Seil Oh
- Division of Cardiology Department of Internal Medicine Seoul National University Hospital Seoul South Korea
| | - Ji Hyun Chang
- Department of Radiation Oncology Seoul National University College of Medicine Seoul Korea.,Department of Radiation Oncology Seoul National University Hospital Seoul South Korea
| |
Collapse
|
47
|
Dong C, Wang Y, Ma A, Wang T. Life Cycle of the Cardiac Voltage-Gated Sodium Channel Na V1.5. Front Physiol 2020; 11:609733. [PMID: 33391024 PMCID: PMC7773603 DOI: 10.3389/fphys.2020.609733] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 11/12/2020] [Indexed: 12/13/2022] Open
Abstract
Cardiac voltage-gated sodium channel NaV1.5, encoded by SCN5A, is crucial for the upstroke of action potential and excitation of cardiomyocytes. NaV1.5 undergoes complex processes before it reaches the target membrane microdomains and performs normal functions. A variety of protein partners are needed to achieve the balance between SCN5A transcription and mRNA decay, endoplasmic reticulum retention and export, Golgi apparatus retention and export, selective anchoring and degradation, activation, and inactivation of sodium currents. Subtle alterations can impair NaV1.5 in terms of expression or function, eventually leading to NaV1.5-associated diseases such as lethal arrhythmias and cardiomyopathy.
Collapse
Affiliation(s)
- Caijuan Dong
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Ya Wang
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Aiqun Ma
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,Key Laboratory of Molecular Cardiology, Shaanxi Province, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education, Xi'an, China
| | - Tingzhong Wang
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,Key Laboratory of Molecular Cardiology, Shaanxi Province, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education, Xi'an, China
| |
Collapse
|
48
|
Beffagna G, Sommariva E, Bellin M. Mechanotransduction and Adrenergic Stimulation in Arrhythmogenic Cardiomyopathy: An Overview of in vitro and in vivo Models. Front Physiol 2020; 11:568535. [PMID: 33281612 PMCID: PMC7689294 DOI: 10.3389/fphys.2020.568535] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 10/19/2020] [Indexed: 01/09/2023] Open
Abstract
Arrhythmogenic Cardiomyopathy (AC) is a rare inherited heart disease, manifesting with progressive myocardium degeneration and dysfunction, and life-threatening arrhythmic events that lead to sudden cardiac death. Despite genetic determinants, most of AC patients admitted to hospital are athletes or very physically active people, implying the existence of other disease-causing factors. It is recognized that AC phenotypes are enhanced and triggered by strenuous physical activity, while excessive mechanical stretch and load, and repetitive adrenergic stimulation are mechanisms influencing disease penetrance. Different approaches have been undertaken to recapitulate and study both mechanotransduction and adrenergic signaling in AC, including the use of in vitro cellular and tissue models, and the development of in vivo models (particularly rodents but more recently also zebrafish). However, it remains challenging to reproduce mechanical load stimuli and physical activity in laboratory experimental settings. Thus, more work to drive the innovation of advanced AC models is needed to recapitulate these subtle physiological influences. Here, we review the state-of-the-art in this field both in clinical and laboratory-based modeling scenarios. Specific attention will be focused on highlighting gaps in the knowledge and how they may be resolved by utilizing novel research methodology.
Collapse
Affiliation(s)
- Giorgia Beffagna
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, Padua, Italy.,Department of Biology, University of Padua, Padua, Italy
| | - Elena Sommariva
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino IRCCS, Milan, Italy
| | - Milena Bellin
- Department of Biology, University of Padua, Padua, Italy.,Veneto Institute of Molecular Medicine, Padua, Italy.,Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
49
|
Established and Emerging Mechanisms in the Pathogenesis of Arrhythmogenic Cardiomyopathy: A Multifaceted Disease. Int J Mol Sci 2020; 21:ijms21176320. [PMID: 32878278 PMCID: PMC7503882 DOI: 10.3390/ijms21176320] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 08/28/2020] [Accepted: 08/29/2020] [Indexed: 12/13/2022] Open
Abstract
Arrhythmogenic cardiomyopathy (ACM) is a heritable myocardial disease that manifests with cardiac arrhythmias, syncope, sudden cardiac death, and heart failure in the advanced stages. The pathological hallmark of ACM is a gradual replacement of the myocardium by fibroadiposis, which typically starts from the epicardium. Molecular genetic studies have identified causal mutations predominantly in genes encoding for desmosomal proteins; however, non-desmosomal causal mutations have also been described, including genes coding for nuclear proteins, cytoskeleton componentsand proteins involved in excitation-contraction coupling. Despite the poor prognosis, currently available treatments can only partially control symptoms and to date there is no effective therapy for ACM. Inhibition of the canonical Wnt/β-catenin pathway and activation of the Hippo and the TGF-β pathways have been implicated in the pathogenesis of ACM. Yet, our understanding of the molecular mechanisms involved in the development of the disease and the cell source of fibroadiposis remains incomplete. Elucidation of the pathogenesis of the disease could facilitate targeted approaches for treatment. In this manuscript we will provide a comprehensive review of the proposed molecular and cellular mechanisms of the pathogenesis of ACM, including the emerging evidence on abnormal calcium homeostasis and inflammatory/autoimmune response. Moreover, we will propose novel hypothesis about the role of epicardial cells and paracrine factors in the development of the phenotype. Finally, we will discuss potential innovative therapeutic approaches based on the growing knowledge in the field.
Collapse
|
50
|
Yeung C, Enriquez A, Suarez-Fuster L, Baranchuk A. Atrial fibrillation in patients with inherited cardiomyopathies. Europace 2020; 21:22-32. [PMID: 29684120 DOI: 10.1093/europace/euy064] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 03/13/2018] [Indexed: 12/19/2022] Open
Abstract
Atrial fibrillation (AF) often complicates the course of inherited cardiomyopathies and, in some cases, may be the presenting feature. Each inherited cardiomyopathy has its own peculiar pathogenetic characteristics that can contribute to the development and maintenance of AF. Atrial fibrillation may occur as a consequence of disease-specific defects, non-specific cardiac chamber changes secondary to the primary illness, or a combination thereof. The presence of AF can denote a turning point in the progression of the disease, promoting clinical deterioration and increasing morbidity and mortality. Furthermore, the management of AF can be particularly challenging in patients with inherited cardiomyopathies. In this article, we review the current information on the prevalence, pathophysiology, risk factors, and treatment of AF in three different inherited cardiomyopathies: hypertrophic cardiomyopathy, arrhythmogenic right ventricular dysplasia/cardiomyopathy, familial dilated cardiomyopathy, and left ventricular non-compaction cardiomyopathy.
Collapse
Affiliation(s)
- Cynthia Yeung
- Kingston General Hospital, Queen's University, Kingston, ON, Canada
| | - Andres Enriquez
- Kingston General Hospital, Queen's University, Kingston, ON, Canada
| | | | - Adrian Baranchuk
- Kingston General Hospital, Queen's University, Kingston, ON, Canada
| |
Collapse
|