1
|
Nehme RD, Sinno L, Shouman W, Ziade JA, Ammar LA, Amin G, Booz GW, Zouein FA. Cardiac Channelopathies: Clinical Diagnosis and Promising Therapeutics. J Am Heart Assoc 2025; 14:e040072. [PMID: 40281647 DOI: 10.1161/jaha.124.040072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/29/2025]
Abstract
Cardiac channelopathies, also known as primary electrical heart diseases, are inherited genetic abnormalities of cardiomyocyte electrical behavior. Notable for their absence of structural heart diseases, they include a diverse group of diseases such as long QT syndrome, short QT syndrome, Brugada syndrome, early repolarization syndrome, catecholaminergic polymorphic ventricular tachycardia, and idiopathic ventricular fibrillation, and carry the risk of malignant arrhythmias leading to sudden cardiac death. The genetic and molecular foundations of these diseases are diverse and complex, with evolving research highlighting the multifactorial nature of their pathophysiology and the intricate interplay of various genes in the manifestation of arrhythmias. While advances in diagnostic techniques, such as genetic testing and electrophysiological studies, have improved the identification and management of these conditions, the relationship between specific genetic mutations and sudden cardiac death remains incompletely understood. This review provides an overview of the molecular and genetic mechanisms underlying those inherited arrhythmias, exploring both well-established and emerging data. Additionally, it discusses current diagnostic approaches and management strategies, aiming to enhance the understanding of these conditions and contribute to better sudden cardiac death prevention.
Collapse
Affiliation(s)
- Ryan Dib Nehme
- Department of Pharmacology and Toxicology American University of Beirut Faculty of Medicine Beirut Lebanon
- The Cardiovascular, Renal, and Metabolic Diseases Research Center of Excellence American University of Beirut Medical Center Beirut Lebanon
| | - Lilas Sinno
- Department of Pharmacology and Toxicology American University of Beirut Faculty of Medicine Beirut Lebanon
- The Cardiovascular, Renal, and Metabolic Diseases Research Center of Excellence American University of Beirut Medical Center Beirut Lebanon
| | - Wael Shouman
- Department of Pharmacology and Toxicology American University of Beirut Faculty of Medicine Beirut Lebanon
- The Cardiovascular, Renal, and Metabolic Diseases Research Center of Excellence American University of Beirut Medical Center Beirut Lebanon
| | - Joanna A Ziade
- Department of Pharmacology and Toxicology American University of Beirut Faculty of Medicine Beirut Lebanon
- The Cardiovascular, Renal, and Metabolic Diseases Research Center of Excellence American University of Beirut Medical Center Beirut Lebanon
| | - Lama A Ammar
- Department of Pharmacology and Toxicology American University of Beirut Faculty of Medicine Beirut Lebanon
| | - Ghadir Amin
- The Cardiovascular, Renal, and Metabolic Diseases Research Center of Excellence American University of Beirut Medical Center Beirut Lebanon
- Department of Pharmacology and Toxicology, School of Medicine University of Mississippi Medical Center Jackson MS USA
| | - George W Booz
- Department of Pharmacology and Toxicology, School of Medicine University of Mississippi Medical Center Jackson MS USA
| | - Fouad A Zouein
- Department of Pharmacology and Toxicology American University of Beirut Faculty of Medicine Beirut Lebanon
- The Cardiovascular, Renal, and Metabolic Diseases Research Center of Excellence American University of Beirut Medical Center Beirut Lebanon
- Department of Pharmacology and Toxicology, School of Medicine University of Mississippi Medical Center Jackson MS USA
| |
Collapse
|
2
|
Komissarova SM, Chakova NN, Niyazova SS, Dolmatovich TV, Troyanova-Shchutskaia TA, Rineiska NM. [Catecholaminergic Polymorphic Ventricular Tachycardia Caused by a Homozygous Pathogenic Variant in Calsequestrin 2 Gene]. KARDIOLOGIIA 2025; 65:57-64. [PMID: 40331653 DOI: 10.18087/cardio.2025.4.n2877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Accepted: 03/21/2025] [Indexed: 05/08/2025]
Abstract
The article presents a clinical case of a 19-year-old patient with catecholaminergic polymorphic ventricular tachycardia caused by the pathogenic homozygous variant p.Ile193Asnfs*17 (rs397516643) in the CASQ2 gene, the early manifestations of which were recurrent syncope during emotional stress, supraventricular and polymorphic ventricular arrhythmias in the absence of structural changes in the heart. The article showed the evolution of heart rhythm disorders during the observation period. The authors discussed the issues of risk stratification for sudden cardiac death and the strategy for its prevention in this pathology.
Collapse
Affiliation(s)
- S M Komissarova
- Republican Scientific and Practical Center "Cardiology", Minsk
| | - N N Chakova
- Institute of Genetics and Cytology of the Belarus National Academy of Sciences, Minsk
| | - S S Niyazova
- Institute of Genetics and Cytology of the Belarus National Academy of Sciences, Minsk
| | - T V Dolmatovich
- Institute of Genetics and Cytology of the Belarus National Academy of Sciences, Minsk
| | | | - N M Rineiska
- Republican Scientific and Practical Center "Cardiology", Minsk
| |
Collapse
|
3
|
Siu A, Tandanu E, Ma B, Osas EE, Liu H, Liu T, Chou OHI, Huang H, Tse G. Precision medicine in catecholaminergic polymorphic ventricular tachycardia: Recent advances toward personalized care. Ann Pediatr Cardiol 2023; 16:431-446. [PMID: 38817258 PMCID: PMC11135882 DOI: 10.4103/apc.apc_96_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 12/12/2023] [Accepted: 01/14/2024] [Indexed: 06/01/2024] Open
Abstract
Catecholaminergic polymorphic ventricular tachycardia (CPVT) is a rare inherited cardiac ion channelopathy where the initial disease presentation is during childhood or adolescent stages, leading to increased risks of sudden cardiac death. Despite advances in medical science and technology, several gaps remain in the understanding of the molecular mechanisms, risk prediction, and therapeutic management of patients with CPVT. Recent studies have identified and validated seven sets of genes responsible for various CPVT phenotypes, including RyR2, CASQ-2, TRDN, CALM1, 2, and 3, and TECRL, providing novel insights into the molecular mechanisms. However, more data on atypical CPVT genotypes are required to investigate the underlying mechanisms further. The complexities of the underlying genetics contribute to challenges in risk stratification as well as the uncertainty surrounding nongenetic modifiers. Therapeutically, although medical management involving beta-blockers and flecainide, or insertion of an implantable cardioverter defibrillator remains the mainstay of treatment, animal and stem cell studies on gene therapy for CPVT have shown promising results. However, its clinical applicability remains unclear. Current gene therapy studies have primarily focused on the RyR2 and CASQ-2 variants, which constitute 75% of all CPVT cases. Alternative approaches that target a broader population, such as CaMKII inhibition, could be more feasible for clinical implementation. Together, this review provides an update on recent research on CPVT, highlighting the need for further investigation of the molecular mechanisms, risk stratification, and therapeutic management of this potentially lethal condition.
Collapse
Affiliation(s)
- Anthony Siu
- Cardiac Electrophysiology Unit, Cardiovascular Analytics Group, Powerhealth Research Institute, Hong Kong, China
- GKT School of Medical Education, King’s College London, London, United Kingdom
| | - Edelyne Tandanu
- GKT School of Medical Education, King’s College London, London, United Kingdom
| | - Brian Ma
- Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China
| | | | - Haipeng Liu
- Research Centre for Intelligent Healthcare, Coventry University, Coventry, United Kingdom
| | - Tong Liu
- Department of Cardiology, Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin, China
| | - Oscar Hou In Chou
- Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China
| | - Helen Huang
- University of Medicine and Health Science, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Gary Tse
- Department of Cardiology, Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin, China
- Kent and Medway Medical School, University of Kent, Canterbury, United Kingdom
- School of Nursing and Health Studies, Hong Kong Metropolitan University, Hong Kong, China
| |
Collapse
|
4
|
Cahill T, Chan S, Overton IM, Hardiman G. Transcriptome Profiling Reveals Enhanced Mitochondrial Activity as a Cold Adaptive Strategy to Hypothermia in Zebrafish Muscle. Cells 2023; 12:1366. [PMID: 37408201 PMCID: PMC10216211 DOI: 10.3390/cells12101366] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 05/01/2023] [Accepted: 05/07/2023] [Indexed: 07/07/2023] Open
Abstract
The utilisation of synthetic torpor for interplanetary travel once seemed farfetched. However, mounting evidence points to torpor-induced protective benefits from the main hazards of space travel, namely, exposure to radiation and microgravity. To determine the radio-protective effects of an induced torpor-like state we exploited the ectothermic nature of the Danio rerio (zebrafish) in reducing their body temperatures to replicate the hypothermic states seen during natural torpor. We also administered melatonin as a sedative to reduce physical activity. Zebrafish were then exposed to low-dose radiation (0.3 Gy) to simulate radiation exposure on long-term space missions. Transcriptomic analysis found that radiation exposure led to an upregulation of inflammatory and immune signatures and a differentiation and regeneration phenotype driven by STAT3 and MYOD1 transcription factors. In addition, DNA repair processes were downregulated in the muscle two days' post-irradiation. The effects of hypothermia led to an increase in mitochondrial translation including genes involved in oxidative phosphorylation and a downregulation of extracellular matrix and developmental genes. Upon radiation exposure, increases in endoplasmic reticulum stress genes were observed in a torpor+radiation group with downregulation of immune-related and ECM genes. Exposing hypothermic zebrafish to radiation also resulted in a downregulation of ECM and developmental genes however, immune/inflammatory related pathways were downregulated in contrast to that observed in the radiation only group. A cross-species comparison was performed with the muscle of hibernating Ursus arctos horribilis (brown bear) to define shared mechanisms of cold tolerance. Shared responses show an upregulation of protein translation and metabolism of amino acids, as well as a hypoxia response with the shared downregulation of glycolysis, ECM, and developmental genes.
Collapse
Affiliation(s)
- Thomas Cahill
- School of Biological Sciences, Institute for Global Food Security, Queen’s University Belfast, Belfast BT9 5DL, UK;
| | - Sherine Chan
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, SC 29425, USA;
- JLABS at the Children’s National Research and Innovation Campus, Washington, DC 20012, USA
| | - Ian M. Overton
- Patrick G Johnston Centre for Cancer Research, Queen’s University Belfast, Belfast BT9 7AE, UK;
| | - Gary Hardiman
- School of Biological Sciences, Institute for Global Food Security, Queen’s University Belfast, Belfast BT9 5DL, UK;
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, SC 29425, USA;
| |
Collapse
|
5
|
Kakkat S, Pramanik P, Singh S, Singh AP, Sarkar C, Chakroborty D. Cardiovascular Complications in Patients with Prostate Cancer: Potential Molecular Connections. Int J Mol Sci 2023; 24:ijms24086984. [PMID: 37108147 PMCID: PMC10138415 DOI: 10.3390/ijms24086984] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/21/2023] [Accepted: 03/30/2023] [Indexed: 04/29/2023] Open
Abstract
Cardiovascular diseases (CVDs) and complications are often seen in patients with prostate cancer (PCa) and affect their clinical management. Despite acceptable safety profiles and patient compliance, androgen deprivation therapy (ADT), the mainstay of PCa treatment and chemotherapy, has increased cardiovascular risks and metabolic syndromes in patients. A growing body of evidence also suggests that patients with pre-existing cardiovascular conditions show an increased incidence of PCa and present with fatal forms of the disease. Therefore, it is possible that a molecular link exists between the two diseases, which has not yet been unraveled. This article provides insight into the connection between PCa and CVDs. In this context, we present our findings linking PCa progression with patients' cardiovascular health by performing a comprehensive gene expression study, gene set enrichment (GSEA) and biological pathway analysis using publicly available data extracted from patients with advanced metastatic PCa. We also discuss the common androgen deprivation strategies and CVDs most frequently reported in PCa patients and present evidence from various clinical trials that suggest that therapy induces CVD in PCa patients.
Collapse
Affiliation(s)
- Sooraj Kakkat
- Department of Pathology, University of South Alabama, Mobile, AL 36617, USA
- Cancer Biology Program, Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA
| | - Paramahansa Pramanik
- Department of Mathematics and Statistics, University of South Alabama, Mobile, AL 36688, USA
| | - Seema Singh
- Department of Pathology, University of South Alabama, Mobile, AL 36617, USA
- Cancer Biology Program, Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, AL 36688, USA
| | - Ajay Pratap Singh
- Department of Pathology, University of South Alabama, Mobile, AL 36617, USA
- Cancer Biology Program, Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, AL 36688, USA
| | - Chandrani Sarkar
- Department of Pathology, University of South Alabama, Mobile, AL 36617, USA
- Cancer Biology Program, Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, AL 36688, USA
| | - Debanjan Chakroborty
- Department of Pathology, University of South Alabama, Mobile, AL 36617, USA
- Cancer Biology Program, Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, AL 36688, USA
| |
Collapse
|
6
|
Paudel R, Jafri MS, Ullah A. Pacing Dynamics Determines the Arrhythmogenic Mechanism of the CPVT2-Causing CASQ2 G112+5X Mutation in a Guinea Pig Ventricular Myocyte Computational Model. Genes (Basel) 2022; 14:23. [PMID: 36672764 PMCID: PMC9858930 DOI: 10.3390/genes14010023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 12/05/2022] [Accepted: 12/20/2022] [Indexed: 12/25/2022] Open
Abstract
Calsequestrin Type 2 (CASQ2) is a high-capacity, low-affinity, Ca2+-binding protein expressed in the sarcoplasmic reticulum (SR) of the cardiac myocyte. Mutations in CASQ2 have been linked to the arrhythmia catecholaminergic polymorphic ventricular tachycardia (CPVT2) that occurs with acute emotional stress or exercise can result in sudden cardiac death (SCD). CASQ2G112+5X is a 16 bp (339-354) deletion CASQ2 mutation that prevents the protein expression due to premature stop codon. Understanding the subcellular mechanisms of CPVT2 is experimentally challenging because the occurrence of arrhythmia is rare. To obtain an insight into the characteristics of this rare disease, simulation studies using a local control stochastic computational model of the Guinea pig ventricular myocyte investigated how the mutant CASQ2s may be responsible for the development of an arrhythmogenic episode under the condition of β-adrenergic stimulation or in the slowing of heart rate afterward once β-adrenergic stimulation ceases. Adjustment of the computational model parameters based upon recent experiments explore the functional changes caused by the CASQ2 mutation. In the simulation studies under rapid pacing (6 Hz), electromechanically concordant cellular alternans appeared under β-adrenergic stimulation in the CPVT mutant but not in the wild-type nor in the non-β-stimulated mutant. Similarly, the simulations of accelerating pacing from slow to rapid and back to the slow pacing did not display alternans but did generate early afterdepolarizations (EADs) during the period of second slow pacing subsequent acceleration of rapid pacing.
Collapse
Affiliation(s)
- Roshan Paudel
- School of Systems Biology, George Mason University, Fairfax, VA 22030, USA
- School of Computer, Mathematical, and Natural Sciences, Morgan State University, Baltimore, MD 21251, USA
| | - Mohsin Saleet Jafri
- School of Systems Biology, George Mason University, Fairfax, VA 22030, USA
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD 20201, USA
| | - Aman Ullah
- School of Systems Biology, George Mason University, Fairfax, VA 22030, USA
| |
Collapse
|
7
|
Charafeddine F, Assaf N, Ismail A, Bulbul Z. Novel Trans-2,3-enoyl-CoA reductase-like Variant Associated with Catecholaminergic Polymorphic Ventricular Tachycardia Type 3. HeartRhythm Case Rep 2022; 9:171-177. [PMID: 36970382 PMCID: PMC10030308 DOI: 10.1016/j.hrcr.2022.12.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
|
8
|
Abstract
Flecainide, a cardiac class 1C blocker of the surface membrane sodium channel (NaV1.5), has also been reported to reduce cardiac ryanodine receptor (RyR2)-mediated sarcoplasmic reticulum (SR) Ca2+ release. It has been introduced as a clinical antiarrhythmic agent for catecholaminergic polymorphic ventricular tachycardia (CPVT), a condition most commonly associated with gain-of-function RyR2 mutations. Current debate concerns both cellular mechanisms of its antiarrhythmic action and molecular mechanisms of its RyR2 actions. At the cellular level, it targets NaV1.5, RyR2, Na+/Ca2+ exchange (NCX), and additional proteins involved in excitation-contraction (EC) coupling and potentially contribute to the CPVT phenotype. This Viewpoint primarily addresses the various direct molecular actions of flecainide on isolated RyR2 channels in artificial lipid bilayers. Such studies demonstrate different, multifarious, flecainide binding sites on RyR2, with voltage-dependent binding in the channel pore or voltage-independent binding at distant peripheral sites. In contrast to its single NaV1.5 pore binding site, flecainide may bind to at least four separate inhibitory sites on RyR2 and one activation site. None of these binding sites have been specifically located in the linear RyR2 sequence or high-resolution structure. Furthermore, it is not clear which of the inhibitory sites contribute to flecainide's reduction of spontaneous Ca2+ release in cellular studies. A confounding observation is that flecainide binding to voltage-dependent inhibition sites reduces cation fluxes in a direction opposite to physiological Ca2+ flow from SR lumen to cytosol. This may suggest that, rather than directly blocking Ca2+ efflux, flecainide can reduce Ca2+ efflux by blocking counter currents through the pore which otherwise limit SR membrane potential change during systolic Ca2+ efflux. In summary, the antiarrhythmic effects of flecainide in CPVT seem to involve multiple components of EC coupling and multiple actions on RyR2. Their clarification may identify novel specific drug targets and facilitate flecainide's clinical utilization in CPVT.
Collapse
Affiliation(s)
| | - Christopher L.-H. Huang
- Department of Biochemistry, University of Cambridge, Cambridge, UK
- Physiological Laboratory, University of Cambridge, Cambridge, UK
| | - James A. Fraser
- Physiological Laboratory, University of Cambridge, Cambridge, UK
| | - Angela F. Dulhunty
- Eccles Institute of Neuroscience, John Curtin School of Medical Research, The Australian National University, Acton, Australia
| |
Collapse
|
9
|
Wang M, Tu X. The Genetics and Epigenetics of Ventricular Arrhythmias in Patients Without Structural Heart Disease. Front Cardiovasc Med 2022; 9:891399. [PMID: 35783865 PMCID: PMC9240357 DOI: 10.3389/fcvm.2022.891399] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 05/25/2022] [Indexed: 12/19/2022] Open
Abstract
Ventricular arrhythmia without structural heart disease is an arrhythmic disorder that occurs in structurally normal heart and no transient or reversible arrhythmia factors, such as electrolyte disorders and myocardial ischemia. Ventricular arrhythmias without structural heart disease can be induced by multiple factors, including genetics and environment, which involve different genetic and epigenetic regulation. Familial genetic analysis reveals that cardiac ion-channel disorder and dysfunctional calcium handling are two major causes of this type of heart disease. Genome-wide association studies have identified some genetic susceptibility loci associated with ventricular tachycardia and ventricular fibrillation, yet relatively few loci associated with no structural heart disease. The effects of epigenetics on the ventricular arrhythmias susceptibility genes, involving non-coding RNAs, DNA methylation and other regulatory mechanisms, are gradually being revealed. This article aims to review the knowledge of ventricular arrhythmia without structural heart disease in genetics, and summarizes the current state of epigenetic regulation.
Collapse
|
10
|
Therapeutic Approaches of Ryanodine Receptor-Associated Heart Diseases. Int J Mol Sci 2022; 23:ijms23084435. [PMID: 35457253 PMCID: PMC9031589 DOI: 10.3390/ijms23084435] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 04/11/2022] [Accepted: 04/14/2022] [Indexed: 01/08/2023] Open
Abstract
Cardiac diseases are the leading causes of death, with a growing number of cases worldwide, posing a challenge for both healthcare and research. Therefore, the most relevant aim of cardiac research is to unravel the molecular pathomechanisms and identify new therapeutic targets. Cardiac ryanodine receptor (RyR2), the Ca2+ release channel of the sarcoplasmic reticulum, is believed to be a good therapeutic target in a group of certain heart diseases, collectively called cardiac ryanopathies. Ryanopathies are associated with the impaired function of the RyR, leading to heart diseases such as congestive heart failure (CHF), catecholaminergic polymorphic ventricular tachycardia (CPVT), arrhythmogenic right ventricular dysplasia type 2 (ARVD2), and calcium release deficiency syndrome (CRDS). The aim of the current review is to provide a short insight into the pathological mechanisms of ryanopathies and discuss the pharmacological approaches targeting RyR2.
Collapse
|
11
|
Kim JH, Lee E, Yun J, Ryu HS, Kim HK, Ju YW, Kim K, Kim J, Moon H. Calsequestrin 2 overexpression in breast cancer increases tumorigenesis and metastasis by modulating the tumor microenvironment. Mol Oncol 2022; 16:466-484. [PMID: 34743414 PMCID: PMC8763655 DOI: 10.1002/1878-0261.13136] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 10/05/2021] [Accepted: 11/04/2021] [Indexed: 01/16/2023] Open
Abstract
The spatial tumor shape is determined by the complex interactions between tumor cells and their microenvironment. Here, we investigated the role of a newly identified breast cancer-related gene, calsequestrin 2 (CASQ2), in tumor-microenvironment interactions during tumor growth and metastasis. We analyzed gene expression and three-dimensional tumor shape data from the breast cancer dataset of The Cancer Genome Atlas (TCGA) and identified CASQ2 as a potential regulator of tumor-microenvironment interaction. In TCGA breast cancer cases containing information of three-dimensional tumor shapes, CASQ2 mRNA showed the highest correlation with the spatial tumor shapes. Furthermore, we investigated the expression pattern of CASQ2 in human breast cancer tissues. CASQ2 was not detected in breast cancer cell lines in vitro but was induced in the xenograft tumors and human breast cancer tissues. To evaluate the role of CASQ2, we established CASQ2-overexpressing breast cancer cell lines for in vitro and in vivo experiments. CASQ2 overexpression in breast cancer cells resulted in a more aggressive phenotype and altered epithelial-mesenchymal transition (EMT) markers in vitro. CASQ2 overexpression induced cancer-associated fibroblast characteristics along with increased hypoxia-inducible factor 1α (HIF1α) expression in stromal fibroblasts. CASQ2 overexpression accelerated tumorigenesis, induced collagen structure remodeling, and increased distant metastasis in vivo. CASQ2 conferred more metaplastic features to triple-negative breast cancer cells. Our data suggest that CASQ2 is a key regulator of breast cancer tumorigenesis and metastasis by modulating diverse aspects of tumor-microenvironment interactions.
Collapse
Affiliation(s)
- Ju Hee Kim
- Biomedical Research InstituteSeoul National University HospitalSouth Korea
| | - Eun‐Shin Lee
- Biomedical Research InstituteSeoul National University HospitalSouth Korea
- Department of PathologySeoul National University School of MedicineSouth Korea
| | - Jihui Yun
- Genomic Medicine InstituteMedical Research CenterSeoul National UniversityKorea
- Department of Biomedical SciencesSeoul National University College of MedicineKorea
| | - Han Suk Ryu
- Department of PathologySeoul National University HospitalSouth Korea
| | - Hong Kyu Kim
- Department of SurgerySeoul National University HospitalKorea
| | - Young Wook Ju
- Department of SurgerySeoul National University HospitalKorea
| | - Kwangsoo Kim
- Division of Clinical BioinformaticsSeoul National University HospitalKorea
| | - Jong‐Il Kim
- Genomic Medicine InstituteMedical Research CenterSeoul National UniversityKorea
- Department of Biomedical SciencesSeoul National University College of MedicineKorea
- Cancer Research InstituteSeoul National UniversityKorea
- Department of Biochemistry and Molecular BiologySeoul National University College of MedicineKorea
| | - Hyeong‐Gon Moon
- Department of SurgerySeoul National University HospitalKorea
- Cancer Research InstituteSeoul National UniversityKorea
- Department of SurgerySeoul National University College of MedicineSouth Korea
| |
Collapse
|
12
|
Li S, Jia Z, Zhang Z, Li Y, Yan M, Yu T. Association Study of Genetic Variants in Calcium Signaling-Related Genes With Cardiovascular Diseases. Front Cell Dev Biol 2021; 9:642141. [PMID: 34912794 PMCID: PMC8666440 DOI: 10.3389/fcell.2021.642141] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 10/01/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Calcium ions (Ca2+) play an essential role in excitation-contraction coupling in the heart. The association between cardiovascular diseases (CVDs) and genetic polymorphisms in key regulators of Ca2+ homeostasis is well established but still inadequately understood. Methods: The associations of 11,274 genetic variants located in nine calcium signaling-related genes with 118 diseases of the circulatory system were explored using a large sample from the United Kingdom Biobank (N = 308,366). The clinical outcomes in electronic health records were mapped to the phecode system. Survival analyses were employed to study the role of variants in CVDs incidence and mortality. Phenome-wide association studies (PheWAS) were performed to investigate the effect of variants on cardiovascular risk factors. Results: The reported association between rs1801253 in β1-adrenergic receptor (ADRB1) and hypertension was successfully replicated, and we additionally found the blood pressure-lowering G allele of this variant was associated with a delayed onset of hypertension and a decreased level of apolipoprotein A. The association of rs4484922 in calsequestrin 2 (CASQ2) with atrial fibrillation/flutter was identified, and this variant also displayed nominal evidence of association with QRS duration and carotid intima-medial thickness. Moreover, our results indicated suggestive associations of rs79613429 in ryanodine receptor 2 (RYR2) with precordial pain. Conclusion: Multiple novel associations established in our study highlight genetic testing as a useful method for CVDs diagnosis and prevention.
Collapse
Affiliation(s)
- Sen Li
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | | | | | | | | | | |
Collapse
|
13
|
Woll KA, Van Petegem F. Calcium Release Channels: Structure and Function of IP3 Receptors and Ryanodine Receptors. Physiol Rev 2021; 102:209-268. [PMID: 34280054 DOI: 10.1152/physrev.00033.2020] [Citation(s) in RCA: 132] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Ca2+-release channels are giant membrane proteins that control the release of Ca2+ from the endoplasmic and sarcoplasmic reticulum. The two members, ryanodine receptors (RyRs) and inositol-1,4,5-trisphosphate Receptors (IP3Rs), are evolutionarily related and are both activated by cytosolic Ca2+. They share a common architecture, but RyRs have evolved additional modules in the cytosolic region. Their massive size allows for the regulation by tens of proteins and small molecules, which can affect the opening and closing of the channels. In addition to Ca2+, other major triggers include IP3 for the IP3Rs, and depolarization of the plasma membrane for a particular RyR subtype. Their size has made them popular targets for study via electron microscopic methods, with current structures culminating near 3Å. The available structures have provided many new mechanistic insights int the binding of auxiliary proteins and small molecules, how these can regulate channel opening, and the mechanisms of disease-associated mutations. They also help scrutinize previously proposed binding sites, as some of these are now incompatible with the structures. Many questions remain around the structural effects of post-translational modifications, additional binding partners, and the higher-order complexes these channels can make in situ. This review summarizes our current knowledge about the structures of Ca2+-release channels and how this informs on their function.
Collapse
Affiliation(s)
- Kellie A Woll
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| | - Filip Van Petegem
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
14
|
Arrhythmia Mechanisms in Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes. J Cardiovasc Pharmacol 2020; 77:300-316. [PMID: 33323698 DOI: 10.1097/fjc.0000000000000972] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 12/08/2020] [Indexed: 12/30/2022]
Abstract
ABSTRACT Despite major efforts by clinicians and researchers, cardiac arrhythmia remains a leading cause of morbidity and mortality in the world. Experimental work has relied on combining high-throughput strategies with standard molecular and electrophysiological studies, which are, to a great extent, based on the use of animal models. Because this poses major challenges for translation, the progress in the development of novel antiarrhythmic agents and clinical care has been mostly disappointing. Recently, the advent of human induced pluripotent stem cell-derived cardiomyocytes has opened new avenues for both basic cardiac research and drug discovery; now, there is an unlimited source of cardiomyocytes of human origin, both from healthy individuals and patients with cardiac diseases. Understanding arrhythmic mechanisms is one of the main use cases of human induced pluripotent stem cell-derived cardiomyocytes, in addition to pharmacological cardiotoxicity and efficacy testing, in vitro disease modeling, developing patient-specific models and personalized drugs, and regenerative medicine. Here, we review the advances that the human induced pluripotent stem cell-derived-based modeling systems have brought so far regarding the understanding of both arrhythmogenic triggers and substrates, while also briefly speculating about the possibilities in the future.
Collapse
|
15
|
Wang Q, Paskevicius T, Filbert A, Qin W, Kim HJ, Chen XZ, Tang J, Dacks JB, Agellon LB, Michalak M. Phylogenetic and biochemical analysis of calsequestrin structure and association of its variants with cardiac disorders. Sci Rep 2020; 10:18115. [PMID: 33093545 PMCID: PMC7582152 DOI: 10.1038/s41598-020-75097-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 10/08/2020] [Indexed: 12/16/2022] Open
Abstract
Calsequestrin is among the most abundant proteins in muscle sarcoplasmic reticulum and displays a high capacity but a low affinity for Ca2+ binding. In mammals, calsequestrin is encoded by two genes, CASQ1 and CASQ2, which are expressed almost exclusively in skeletal and cardiac muscles, respectively. Phylogenetic analysis indicates that calsequestrin is an ancient gene in metazoans, and that the duplication of the ancestral calsequestrin gene took place after the emergence of the lancelet. CASQ2 gene variants associated with catecholaminergic polymorphic ventricular tachycardia (CPVT) in humans are positively correlated with a high degree of evolutionary conservation across all calsequestrin homologues. The mutations are distributed in diverse locations of the calsequestrin protein and impart functional diversity but remarkably manifest in a similar phenotype in humans.
Collapse
Affiliation(s)
- Qian Wang
- Department of Biochemistry, University of Alberta, Edmonton, Alberta, T6G 2H7, Canada
| | - Tautvydas Paskevicius
- Department of Biochemistry, University of Alberta, Edmonton, Alberta, T6G 2H7, Canada
| | - Alexander Filbert
- Division of Infectious Disease, Department of Medicine, University of Alberta, Edmonton, AB, T6G 2G3, Canada
| | - Wenying Qin
- Institute of Biomedical and Pharmaceutical Sciences, Key Laboratory of Fermentation Engineering, Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, Hubei, China
| | - Hyeong Jin Kim
- Department of Biochemistry, University of Alberta, Edmonton, Alberta, T6G 2H7, Canada
| | - Xing-Zhen Chen
- Institute of Biomedical and Pharmaceutical Sciences, Key Laboratory of Fermentation Engineering, Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, Hubei, China.,Department of Physiology, University of Alberta, Edmonton, Alberta, T6G 2H7, Canada
| | - Jingfeng Tang
- Institute of Biomedical and Pharmaceutical Sciences, Key Laboratory of Fermentation Engineering, Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, Hubei, China
| | - Joel B Dacks
- Division of Infectious Disease, Department of Medicine, University of Alberta, Edmonton, AB, T6G 2G3, Canada.
| | - Luis B Agellon
- School of Dietetics and Human Nutrition, McGill University, Ste. Anne de Bellevue, Quebec, H9X 3V9, Canada.
| | - Marek Michalak
- Department of Biochemistry, University of Alberta, Edmonton, Alberta, T6G 2H7, Canada. .,Institute of Biomedical and Pharmaceutical Sciences, Key Laboratory of Fermentation Engineering, Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, Hubei, China.
| |
Collapse
|
16
|
Molecular adaptation to calsequestrin 2 (CASQ2) point mutations leading to catecholaminergic polymorphic ventricular tachycardia (CPVT): comparative analysis of R33Q and D307H mutants. J Muscle Res Cell Motil 2020; 41:251-258. [PMID: 32902830 PMCID: PMC7666291 DOI: 10.1007/s10974-020-09587-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 08/29/2020] [Indexed: 12/16/2022]
Abstract
Homozygous calsequestrin 2 (CASQ2) point mutations leads to catecholaminergic polymorphic ventricular tachycardia: a common pathogenetic feature appears to be the drastic reduction of mutant CASQ2 in spite of normal transcription. Comparative biochemical analysis of R33Q and D307H knock in mutant mice identifies different pathogenetic mechanisms for CASQ2 degradation and different molecular adaptive mechanisms. In particular, each CASQ2 point mutation evokes specific adaptive cellular and molecular processes in each of the four adaptive pathways investigated. Thus, similar clinical phenotypes and identical cellular mechanism for cardiac arrhythmia might imply different molecular adaptive mechanisms.
Collapse
|
17
|
Saadeh K, Achercouk Z, Fazmin IT, Nantha Kumar N, Salvage SC, Edling CE, Huang CLH, Jeevaratnam K. Protein expression profiles in murine ventricles modeling catecholaminergic polymorphic ventricular tachycardia: effects of genotype and sex. Ann N Y Acad Sci 2020; 1478:63-74. [PMID: 32713021 DOI: 10.1111/nyas.14426] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 05/27/2020] [Accepted: 06/15/2020] [Indexed: 12/12/2022]
Abstract
Catecholaminergic polymorphic ventricular tachycardia (CPVT) is associated with mutations in the cardiac ryanodine receptor (RyR2). These result in stress-induced ventricular arrhythmic episodes, with clinical symptoms and prognosis reported more severe in male than female patients. Murine homozygotic RyR2-P2328S (RyR2S/S ) hearts replicate the proarrhythmic CPVT phenotype of abnormal sarcoplasmic reticular Ca2+ leak and disrupted Ca2+ homeostasis. In addition, RyR2S/S hearts show decreased myocardial action potential conduction velocities (CV), all features implicated in arrhythmic trigger and substrate. The present studies explored for independent and interacting effects of RyR2S/S genotype and sex on expression levels of molecular determinants of Ca2+ homeostasis (CASQ2, FKBP12, SERCA2a, NCX1, and CaV 1.2) and CV (NaV 1.5, Connexin (Cx)-43, phosphorylated-Cx43, and TGF-β1) in mice. Expression levels of Ca2+ homeostasis proteins were not altered, hence implicating abnormal RyR2 function alone in disrupted cytosolic Ca2+ homeostasis. Furthermore, altered NaV 1.5, phosphorylated Cx43, and TGF-β1 expression were not implicated in the development of slowed CV. By contrast, decreased Cx43 expression correlated with slowed CV, in female, but not male, RyR2S/S mice. The CV changes may reflect acute actions of the increased cytosolic Ca2+ on NaV 1.5 and Cx43 function.
Collapse
Affiliation(s)
- Khalil Saadeh
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom.,School of Clinical Medicine, Addenbrooke's Hospital, University of Cambridge, Cambridge, United Kingdom
| | - Zakaria Achercouk
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
| | - Ibrahim T Fazmin
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom.,School of Clinical Medicine, Addenbrooke's Hospital, University of Cambridge, Cambridge, United Kingdom
| | - Nakulan Nantha Kumar
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom.,Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Samantha C Salvage
- Physiological Laboratory, University of Cambridge, Cambridge, United Kingdom.,Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - Charlotte E Edling
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
| | - Christopher L-H Huang
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom.,Physiological Laboratory, University of Cambridge, Cambridge, United Kingdom.,Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - Kamalan Jeevaratnam
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom.,Physiological Laboratory, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
18
|
Wang Q, Michalak M. Calsequestrin. Structure, function, and evolution. Cell Calcium 2020; 90:102242. [PMID: 32574906 DOI: 10.1016/j.ceca.2020.102242] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 06/05/2020] [Accepted: 06/06/2020] [Indexed: 12/25/2022]
Abstract
Calsequestrin is the major Ca2+ binding protein in the sarcoplasmic reticulum (SR), serves as the main Ca2+ storage and buffering protein and is an important regulator of Ca2+ release channels in both skeletal and cardiac muscle. It is anchored at the junctional SR membrane through interactions with membrane proteins and undergoes reversible polymerization with increasing Ca2+ concentration. Calsequestrin provides high local Ca2+ at the junctional SR and communicates changes in luminal Ca2+ concentration to Ca2+ release channels, thus it is an essential component of excitation-contraction coupling. Recent studies reveal new insights on calsequestrin trafficking, Ca2+ binding, protein evolution, protein-protein interactions, stress responses and the molecular basis of related human muscle disease, including catecholaminergic polymorphic ventricular tachycardia (CPVT). Here we provide a comprehensive overview of calsequestrin, with recent advances in structure, diverse functions, phylogenetic analysis, and its role in muscle physiology, stress responses and human pathology.
Collapse
Affiliation(s)
- Qian Wang
- Department of Biochemistry, University of Alberta, Edmonton, AB, T6H 2S7, Canada
| | - Marek Michalak
- Department of Biochemistry, University of Alberta, Edmonton, AB, T6H 2S7, Canada.
| |
Collapse
|
19
|
Kohli U, Aziz Z, Beaser AD, Nayak HM. Ventricular arrhythmia suppression with ivabradine in a patient with catecholaminergic polymorphic ventricular tachycardia refractory to nadolol, flecainide, and sympathectomy. PACING AND CLINICAL ELECTROPHYSIOLOGY: PACE 2020; 43:527-533. [DOI: 10.1111/pace.13913] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 03/26/2020] [Accepted: 03/29/2020] [Indexed: 11/28/2022]
Affiliation(s)
- Utkarsh Kohli
- Division of Pediatric Cardiology, Department of PediatricsComer Children's Hospital and the Pritzker School of Medicine of the University of Chicago Chicago Illinois
| | - Zaid Aziz
- Center for Arrhythmia Care, Heart and Vascular CenterPritzker School of Medicine of the University of Chicago Chicago Illinois
| | - Andrew D. Beaser
- Center for Arrhythmia Care, Heart and Vascular CenterPritzker School of Medicine of the University of Chicago Chicago Illinois
| | - Hemal M. Nayak
- Center for Arrhythmia Care, Heart and Vascular CenterPritzker School of Medicine of the University of Chicago Chicago Illinois
| |
Collapse
|
20
|
Kistamás K, Veress R, Horváth B, Bányász T, Nánási PP, Eisner DA. Calcium Handling Defects and Cardiac Arrhythmia Syndromes. Front Pharmacol 2020; 11:72. [PMID: 32161540 PMCID: PMC7052815 DOI: 10.3389/fphar.2020.00072] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 01/24/2020] [Indexed: 12/13/2022] Open
Abstract
Calcium ions (Ca2+) play a major role in the cardiac excitation-contraction coupling. Intracellular Ca2+ concentration increases during systole and falls in diastole thereby determining cardiac contraction and relaxation. Normal cardiac function also requires perfect organization of the ion currents at the cellular level to drive action potentials and to maintain action potential propagation and electrical homogeneity at the tissue level. Any imbalance in Ca2+ homeostasis of a cardiac myocyte can lead to electrical disturbances. This review aims to discuss cardiac physiology and pathophysiology from the elementary membrane processes that can cause the electrical instability of the ventricular myocytes through intracellular Ca2+ handling maladies to inherited and acquired arrhythmias. Finally, the paper will discuss the current therapeutic approaches targeting cardiac arrhythmias.
Collapse
Affiliation(s)
- Kornél Kistamás
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,Division of Cardiovascular Sciences, School of Medical Sciences, University of Manchester, Manchester, United Kingdom
| | - Roland Veress
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Balázs Horváth
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Tamás Bányász
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Péter P Nánási
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,Department of Dental Physiology, Faculty of Dentistry, University of Debrecen, Debrecen, Hungary
| | - David A Eisner
- Division of Cardiovascular Sciences, School of Medical Sciences, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
21
|
Wang WA, Agellon LB, Michalak M. Organellar Calcium Handling in the Cellular Reticular Network. Cold Spring Harb Perspect Biol 2019; 11:cshperspect.a038265. [PMID: 31358518 DOI: 10.1101/cshperspect.a038265] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Ca2+ is an important intracellular messenger affecting diverse cellular processes. In eukaryotic cells, Ca2+ is handled by a myriad of Ca2+-binding proteins found in organelles that are organized into the cellular reticular network (CRN). The network is comprised of the endoplasmic reticulum, Golgi apparatus, lysosomes, membranous components of the endocytic and exocytic pathways, peroxisomes, and the nuclear envelope. Membrane contact sites between the different components of the CRN enable the rapid movement of Ca2+, and communication of Ca2+ status, within the network. Ca2+-handling proteins that reside in the CRN facilitate Ca2+ sensing, buffering, and cellular signaling to coordinate the many processes that operate within the cell.
Collapse
Affiliation(s)
- Wen-An Wang
- Department of Biochemistry, University of Alberta, Edmonton, Alberta T6G 2S7, Canada
| | - Luis B Agellon
- School of Human Nutrition, McGill University, Ste. Anne de Bellevue, Quebec H9X 3V9, Canada
| | - Marek Michalak
- Department of Biochemistry, University of Alberta, Edmonton, Alberta T6G 2S7, Canada
| |
Collapse
|
22
|
Pathophysiology of Calcium Mediated Ventricular Arrhythmias and Novel Therapeutic Options with Focus on Gene Therapy. Int J Mol Sci 2019; 20:ijms20215304. [PMID: 31653119 PMCID: PMC6862059 DOI: 10.3390/ijms20215304] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Revised: 10/16/2019] [Accepted: 10/21/2019] [Indexed: 12/19/2022] Open
Abstract
Cardiac arrhythmias constitute a major health problem with a huge impact on mortality rates and health care costs. Despite ongoing research efforts, the understanding of the molecular mechanisms and processes responsible for arrhythmogenesis remains incomplete. Given the crucial role of Ca2+-handling in action potential generation and cardiac contraction, Ca2+ channels and Ca2+ handling proteins represent promising targets for suppression of ventricular arrhythmias. Accordingly, we report the different roles of Ca2+-handling in the development of congenital as well as acquired ventricular arrhythmia syndromes. We highlight the therapeutic potential of gene therapy as a novel and innovative approach for future arrhythmia therapy. Furthermore, we discuss various promising cellular and mitochondrial targets for therapeutic gene transfer currently under investigation.
Collapse
|
23
|
Hwang HS, Baldo MP, Rodriguez JP, Faggioni M, Knollmann BC. Efficacy of Flecainide in Catecholaminergic Polymorphic Ventricular Tachycardia Is Mutation-Independent but Reduced by Calcium Overload. Front Physiol 2019; 10:992. [PMID: 31456692 PMCID: PMC6701460 DOI: 10.3389/fphys.2019.00992] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 07/18/2019] [Indexed: 11/22/2022] Open
Abstract
Background The dual Na+ and cardiac Ca2+-release channel inhibitor, Flecainide (FLEC) is effective in patients with catecholaminergic polymorphic ventricular tachycardia (CPVT), a disease caused by mutations in cardiac Ca2+-release channels (RyR2), calsequestrin (Casq2), or calmodulin. FLEC suppresses spontaneous Ca2+ waves in Casq2-knockout (Casq2−/−) cardiomyocytes, a CPVT model. However, a report failed to find FLEC efficacy against Ca2+ waves in another CPVT model, RyR2-R4496C heterozygous mice (RyR2R4496C+/−), raising the possibility that FLEC efficacy may be mutation dependent. Objective To address this controversy, we compared FLEC in Casq2−/− and RyR2R4496C+/− cardiomyocytes and mice under identical conditions. Methods After 30 min exposure to FLEC (6 μM) or vehicle (VEH), spontaneous Ca2+ waves were quantified during a 40 s pause after 1 Hz pacing train in the presence of isoproterenol (ISO, 1 μM). FLEC efficacy was also tested in vivo using a low dose (LOW: 3 mg/kg ISO + 60 mg/kg caffeine) or a high dose catecholamine challenge (HIGH: 3 mg/kg ISO + 120 mg/kg caffeine). Results In cardiomyocytes, FLEC efficacy was dependent on extracellular [Ca2+]. At 2 mM [Ca2+], only Casq2−/− myocytes exhibited Ca2+ waves, which were strongly suppressed by FLEC. At 3 mM [Ca2+] both groups exhibited Ca2+ waves that were suppressed by FLEC. At 4 mM [Ca2+], FLEC no longer suppressed Ca2+ waves in both groups. Analogous to the results in myocytes, RyR2R4496C+/− mice (n = 12) had significantly lower arrhythmia scores than Casq2−/− mice (n = 9), but the pattern of FLEC efficacy was similar in both groups (i.e., reduced FLEC efficacy after HIGH dose catecholamine challenge). Conclusion FLEC inhibits Ca2+ waves in RyR2R4496C+/− cardiomyocytes, indicating that RyR2 channel block by FLEC is not mutation-specific. However, FLEC efficacy is reduced by Ca2+ overload in vitro or by high dose catecholamine challenge in vivo, which could explain conflicting literature reports.
Collapse
Affiliation(s)
- Hyun Seok Hwang
- Department of Nutrition, Food and Exercise Sciences, Florida State University, Tallahassee, FL, United States.,Division of Clinical Pharmacology, Oates Institute for Experimental Therapeutics, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Marcelo P Baldo
- Division of Clinical Pharmacology, Oates Institute for Experimental Therapeutics, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Jose Pindado Rodriguez
- Department of Nutrition, Food and Exercise Sciences, Florida State University, Tallahassee, FL, United States
| | - Michela Faggioni
- Division of Clinical Pharmacology, Oates Institute for Experimental Therapeutics, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Bjorn C Knollmann
- Division of Clinical Pharmacology, Oates Institute for Experimental Therapeutics, Vanderbilt University School of Medicine, Nashville, TN, United States
| |
Collapse
|
24
|
Flores DJ, Duong T, Brandenberger LO, Mitra A, Shirali A, Johnson JC, Springer D, Noguchi A, Yu ZX, Ebert SN, Ludwig A, Knollmann BC, Levin MD, Pfeifer K. Conditional ablation and conditional rescue models for Casq2 elucidate the role of development and of cell-type specific expression of Casq2 in the CPVT2 phenotype. Hum Mol Genet 2019; 27:1533-1544. [PMID: 29452352 DOI: 10.1093/hmg/ddy060] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 02/12/2018] [Indexed: 01/30/2023] Open
Abstract
Cardiac calsequestrin (Casq2) associates with the ryanodine receptor 2 channel in the junctional sarcoplasmic reticulum to regulate Ca2+ release into the cytoplasm. Patients carrying mutations in CASQ2 display low resting heart rates under basal conditions and stress-induced polymorphic ventricular tachycardia (CPVT). In this study, we generate and characterize novel conditional deletion and conditional rescue mouse models to test the influence of developmental programs on the heart rate and CPVT phenotypes. We also compare the requirements for Casq2 function in the cardiac conduction system (CCS) and in working cardiomyocytes. Our study shows that the CPVT phenotype is dependent upon concurrent loss of Casq2 function in both the CCS and in working cardiomyocytes. Accordingly, restoration of Casq2 in only the CCS prevents CPVT. In addition, occurrence of CPVT is independent of the developmental history of Casq2-deficiency. In contrast, resting heart rate depends upon Casq2 gene activity only in the CCS and upon developmental history. Finally, our data support a model where low basal heart rate is a significant risk factor for CPVT.
Collapse
Affiliation(s)
- Daniel J Flores
- Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - ThuyVy Duong
- Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Luke O Brandenberger
- Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Apratim Mitra
- Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Aditya Shirali
- Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - John C Johnson
- Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Danielle Springer
- Murine Phenotyping Core, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Audrey Noguchi
- Murine Phenotyping Core, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Zu-Xi Yu
- Pathology Core, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Steven N Ebert
- Division of Metabolic and Cardiovascular Sciences, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Andreas Ludwig
- Institut fuer Experimentelle und Klinische Pharmakologie und Toxikologie, Friedrich-Alexander-Universitaet Erlangen-Nuernberg, Erlangen 91054, Germany
| | - Bjorn C Knollmann
- Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Mark D Levin
- Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Karl Pfeifer
- Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
25
|
Abstract
Genetic testing has an increasingly important role in the diagnosis and management of cardiac disorders, where it confirms the diagnosis, aids prognostication and risk stratification and guides treatment. A genetic diagnosis in the proband also enables clarification of the risk for family members by cascade testing. Genetics in cardiac disorders is complex where epigenetic and environmental factors might come into interplay. Incomplete penetrance and variable expressivity is also common. Genetic results in cardiac conditions are mostly probabilistic and should be interpreted with all available clinical information. With this complexity in cardiac genetics, testing is only indicated in patients with a strong suspicion of an inheritable cardiac disorder after a full clinical evaluation. In this review we discuss the genetics underlying the major cardiomyopathies and channelopathies, and the practical aspects of diagnosing these conditions in the laboratory.
Collapse
|
26
|
|
27
|
Hamilton S, Terentyev D. Proarrhythmic Remodeling of Calcium Homeostasis in Cardiac Disease; Implications for Diabetes and Obesity. Front Physiol 2018. [PMID: 30425651 DOI: 10.3389/fphys.2018.01517, 10.3389/fpls.2018.01517] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
A rapid growth in the incidence of diabetes and obesity has transpired to a major heath issue and economic burden in the postindustrial world, with more than 29 million patients affected in the United States alone. Cardiovascular defects have been established as the leading cause of mortality and morbidity of diabetic patients. Over the last decade, significant progress has been made in delineating mechanisms responsible for the diminished cardiac contractile function and enhanced propensity for malignant cardiac arrhythmias characteristic of diabetic disease. Rhythmic cardiac contractility relies upon the precise interplay between several cellular Ca2+ transport protein complexes including plasmalemmal L-type Ca2+ channels (LTCC), Na+-Ca2+ exchanger (NCX1), Sarco/endoplasmic Reticulum (SR) Ca2+-ATPase (SERCa2a) and ryanodine receptors (RyR2s), the SR Ca2+ release channels. Here we provide an overview of changes in Ca2+ homeostasis in diabetic ventricular myocytes and discuss the therapeutic potential of targeting Ca2+ handling proteins in the prevention of diabetes-associated cardiomyopathy and arrhythmogenesis.
Collapse
Affiliation(s)
- Shanna Hamilton
- Department of Medicine, The Warren Alpert Medical School of Brown University, Providence, RI, United States.,Cardiovascular Research Center, Rhode Island Hospital, Providence, RI, United States
| | - Dmitry Terentyev
- Department of Medicine, The Warren Alpert Medical School of Brown University, Providence, RI, United States.,Cardiovascular Research Center, Rhode Island Hospital, Providence, RI, United States
| |
Collapse
|
28
|
Hamilton S, Terentyev D. Proarrhythmic Remodeling of Calcium Homeostasis in Cardiac Disease; Implications for Diabetes and Obesity. Front Physiol 2018; 9:1517. [PMID: 30425651 PMCID: PMC6218530 DOI: 10.3389/fphys.2018.01517] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 10/09/2018] [Indexed: 12/28/2022] Open
Abstract
A rapid growth in the incidence of diabetes and obesity has transpired to a major heath issue and economic burden in the postindustrial world, with more than 29 million patients affected in the United States alone. Cardiovascular defects have been established as the leading cause of mortality and morbidity of diabetic patients. Over the last decade, significant progress has been made in delineating mechanisms responsible for the diminished cardiac contractile function and enhanced propensity for malignant cardiac arrhythmias characteristic of diabetic disease. Rhythmic cardiac contractility relies upon the precise interplay between several cellular Ca2+ transport protein complexes including plasmalemmal L-type Ca2+ channels (LTCC), Na+-Ca2+ exchanger (NCX1), Sarco/endoplasmic Reticulum (SR) Ca2+-ATPase (SERCa2a) and ryanodine receptors (RyR2s), the SR Ca2+ release channels. Here we provide an overview of changes in Ca2+ homeostasis in diabetic ventricular myocytes and discuss the therapeutic potential of targeting Ca2+ handling proteins in the prevention of diabetes-associated cardiomyopathy and arrhythmogenesis.
Collapse
Affiliation(s)
- Shanna Hamilton
- Department of Medicine, The Warren Alpert Medical School of Brown University, Providence, RI, United States.,Cardiovascular Research Center, Rhode Island Hospital, Providence, RI, United States
| | - Dmitry Terentyev
- Department of Medicine, The Warren Alpert Medical School of Brown University, Providence, RI, United States.,Cardiovascular Research Center, Rhode Island Hospital, Providence, RI, United States
| |
Collapse
|
29
|
Hamilton S, Terentyev D. Proarrhythmic Remodeling of Calcium Homeostasis in Cardiac Disease; Implications for Diabetes and Obesity. Front Physiol 2018; 9:1517. [PMID: 30425651 PMCID: PMC6218530 DOI: 10.3389/fphys.2018.01517,+10.3389/fpls.2018.01517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/06/2022] Open
Abstract
A rapid growth in the incidence of diabetes and obesity has transpired to a major heath issue and economic burden in the postindustrial world, with more than 29 million patients affected in the United States alone. Cardiovascular defects have been established as the leading cause of mortality and morbidity of diabetic patients. Over the last decade, significant progress has been made in delineating mechanisms responsible for the diminished cardiac contractile function and enhanced propensity for malignant cardiac arrhythmias characteristic of diabetic disease. Rhythmic cardiac contractility relies upon the precise interplay between several cellular Ca2+ transport protein complexes including plasmalemmal L-type Ca2+ channels (LTCC), Na+-Ca2+ exchanger (NCX1), Sarco/endoplasmic Reticulum (SR) Ca2+-ATPase (SERCa2a) and ryanodine receptors (RyR2s), the SR Ca2+ release channels. Here we provide an overview of changes in Ca2+ homeostasis in diabetic ventricular myocytes and discuss the therapeutic potential of targeting Ca2+ handling proteins in the prevention of diabetes-associated cardiomyopathy and arrhythmogenesis.
Collapse
Affiliation(s)
- Shanna Hamilton
- Department of Medicine, The Warren Alpert Medical School of Brown University, Providence, RI, United States,Cardiovascular Research Center, Rhode Island Hospital, Providence, RI, United States
| | - Dmitry Terentyev
- Department of Medicine, The Warren Alpert Medical School of Brown University, Providence, RI, United States,Cardiovascular Research Center, Rhode Island Hospital, Providence, RI, United States,*Correspondence: Dmitry Terentyev,
| |
Collapse
|
30
|
Jones DC, Gong JQX, Sobie EA. A privileged role for neuronal Na + channels in regulating ventricular [Ca 2+] and arrhythmias. J Gen Physiol 2018; 150:901-905. [PMID: 29899058 PMCID: PMC6028496 DOI: 10.1085/jgp.201812120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Jones et al. provide commentary on the intricate crosstalk between ion transporters that goes awry in long QT arrhythmia.
Collapse
Affiliation(s)
- DeAnalisa C Jones
- Department of Pharmacological Sciences, Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Jingqi Q X Gong
- Department of Pharmacological Sciences, Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Eric A Sobie
- Department of Pharmacological Sciences, Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| |
Collapse
|
31
|
Josephs K, Patel K, Janson CM, Montagna C, McDonald TV. Compound heterozygous CASQ2 mutations and long-term course of catecholaminergic polymorphic ventricular tachycardia. Mol Genet Genomic Med 2017; 5:788-794. [PMID: 29178653 PMCID: PMC5702571 DOI: 10.1002/mgg3.323] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 07/07/2017] [Accepted: 07/10/2017] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Catecholaminergic polymorphic ventricular tachycardia (CPVT) is a potentially lethal inherited cardiac disorder characterized by episodic ventricular tachycardia during adrenergic stimulation. It is associated with significant morbidity and mortality. Knowledge of the underlying genetic cause, pathogenesis, and the natural history of the disease remains incomplete. Approximately 50% of CPVT cases are caused by dominant mutations in the cardiac ryanodine receptor (RYR2) gene, <5% of cases are accounted for by recessive mutations in cardiac calsequestrin (CASQ2) or Triadin (TRDN). METHODS We report a family with two CASQ2 gene mutations. A research-based next-generation sequencing (NGS) initiative was used in a patient with a severe CPVT phenotype and her clinically unaffected son. Reverse transcription polymerase chain reaction (RT-PCR) from platelet RNA was used to assess the consequences of predicted splice variants. RESULTS NGS revealed that the proband carried a novel c.199C>T (p.Gln67*) mutation and a previously reported splice site mutation c.532+1G>A in CASQ2. Her son is a heterozygous carrier of the c.199C>T (p.Gln67*) mutation alone and the proband was compound heterozygous at CASQ2. RNA analysis demonstrated that the splice site mutation results in the retention of intron 3 with no full-length CASQ2 mRNA. CONCLUSION This study describes a novel CPVT genotype and further characterizes the effect of a previously reported CASQ2 splice site mutation. The long-term follow-up of 23 years since first symptom provides additional insight into the natural history of CASQ2-associated CPVT.
Collapse
Affiliation(s)
- Katherine Josephs
- Department of GeneticsAlbert Einstein College of MedicineBronxNew York
| | - Kunjan Patel
- Department of GeneticsAlbert Einstein College of MedicineBronxNew York
| | - Christopher M. Janson
- Department of Pediatrics (Cardiology)Albert Einstein College of Medicine & Montefiore Medical CenterBronxNew York
| | - Cristina Montagna
- Department of GeneticsAlbert Einstein College of MedicineBronxNew York
| | - Thomas V. McDonald
- Department of Medicine (Cardiology)Albert Einstein College of Medicine & Montefiore Medical CenterBronxNew York
- Department of Molecular PharmacologyAlbert Einstein College of MedicineBronxNew York
| |
Collapse
|
32
|
Barone V, Del Re V, Gamberucci A, Polverino V, Galli L, Rossi D, Costanzi E, Toniolo L, Berti G, Malandrini A, Ricci G, Siciliano G, Vattemi G, Tomelleri G, Pierantozzi E, Spinozzi S, Volpi N, Fulceri R, Battistutta R, Reggiani C, Sorrentino V. Identification and characterization of three novel mutations in the CASQ1 gene in four patients with tubular aggregate myopathy. Hum Mutat 2017; 38:1761-1773. [PMID: 28895244 DOI: 10.1002/humu.23338] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2017] [Revised: 08/14/2017] [Accepted: 09/04/2017] [Indexed: 12/22/2022]
Abstract
Here, we report the identification of three novel missense mutations in the calsequestrin-1 (CASQ1) gene in four patients with tubular aggregate myopathy. These CASQ1 mutations affect conserved amino acids in position 44 (p.(Asp44Asn)), 103 (p.(Gly103Asp)), and 385 (p.(Ile385Thr)). Functional studies, based on turbidity and dynamic light scattering measurements at increasing Ca2+ concentrations, showed a reduced Ca2+ -dependent aggregation for the CASQ1 protein containing p.Asp44Asn and p.Gly103Asp mutations and a slight increase in Ca2+ -dependent aggregation for the p.Ile385Thr. Accordingly, limited trypsin proteolysis assay showed that p.Asp44Asn and p.Gly103Asp were more susceptible to trypsin cleavage in the presence of Ca2+ in comparison with WT and p.Ile385Thr. Analysis of single muscle fibers of a patient carrying the p.Gly103Asp mutation showed a significant reduction in response to caffeine stimulation, compared with normal control fibers. Expression of CASQ1 mutations in eukaryotic cells revealed a reduced ability of all these CASQ1 mutants to store Ca2+ and a reduced inhibitory effect of p.Ile385Thr and p.Asp44Asn on store operated Ca2+ entry. These results widen the spectrum of skeletal muscle diseases associated with CASQ1 and indicate that these mutations affect properties critical for correct Ca2+ handling in skeletal muscle fibers.
Collapse
Affiliation(s)
- Virginia Barone
- Department of Molecular and Developmental Medicine, Molecular Medicine Section, University of Siena, Siena, Italy
| | - Valeria Del Re
- Department of Molecular and Developmental Medicine, Molecular Medicine Section, University of Siena, Siena, Italy
| | - Alessandra Gamberucci
- Department of Molecular and Developmental Medicine, Molecular Medicine Section, University of Siena, Siena, Italy
| | - Valentina Polverino
- Department of Molecular and Developmental Medicine, Molecular Medicine Section, University of Siena, Siena, Italy
| | - Lucia Galli
- Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | - Daniela Rossi
- Department of Molecular and Developmental Medicine, Molecular Medicine Section, University of Siena, Siena, Italy.,Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | - Elisa Costanzi
- Department of Chemical Sciences, University of Padova, Padova, Italy
| | - Luana Toniolo
- Department of Biomedical Sciences, University of Padova, Padova, Italy.,CNR, Institute of Neuroscience, Padova, Italy
| | - Gianna Berti
- Department of Medical, Surgical and Neurological Sciences, University of Siena, Siena, Italy
| | - Alessandro Malandrini
- Department of Medical, Surgical and Neurological Sciences, University of Siena, Siena, Italy
| | - Giulia Ricci
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Gabriele Siciliano
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Gaetano Vattemi
- Department of Neurological Neurosciences, Biomedicine and Movement Sciences, Section of Clinical Neurology, University of Verona, Verona, Italy
| | - Giuliano Tomelleri
- Department of Neurological Neurosciences, Biomedicine and Movement Sciences, Section of Clinical Neurology, University of Verona, Verona, Italy
| | - Enrico Pierantozzi
- Department of Molecular and Developmental Medicine, Molecular Medicine Section, University of Siena, Siena, Italy
| | - Simone Spinozzi
- Department of Molecular and Developmental Medicine, Molecular Medicine Section, University of Siena, Siena, Italy
| | - Nila Volpi
- Department of Medical, Surgical and Neurological Sciences, University of Siena, Siena, Italy
| | - Rosella Fulceri
- Department of Molecular and Developmental Medicine, Molecular Medicine Section, University of Siena, Siena, Italy
| | | | - Carlo Reggiani
- Department of Biomedical Sciences, University of Padova, Padova, Italy.,CNR, Institute of Neuroscience, Padova, Italy
| | - Vincenzo Sorrentino
- Department of Molecular and Developmental Medicine, Molecular Medicine Section, University of Siena, Siena, Italy.,Azienda Ospedaliera Universitaria Senese, Siena, Italy
| |
Collapse
|
33
|
Walweel K, Oo YW, Laver DR. The emerging role of calmodulin regulation of RyR2 in controlling heart rhythm, the progression of heart failure and the antiarrhythmic action of dantrolene. Clin Exp Pharmacol Physiol 2017; 44:135-142. [PMID: 27626620 DOI: 10.1111/1440-1681.12669] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Revised: 08/27/2016] [Accepted: 09/09/2016] [Indexed: 11/28/2022]
Abstract
Cardiac output and rhythm depend on the release and the take-up of calcium from the sarcoplasmic reticulum (SR). Excessive diastolic calcium leak from the SR due to dysfunctional calcium release channels (RyR2) contributes to the formation of delayed after-depolarizations, which underlie the fatal arrhythmias that occur in heart failure and inherited syndromes. Calmodulin (CaM) is a calcium-binding protein that regulates target proteins and acts as a calcium sensor. CaM is comprised of two calcium-binding EF-hand domains and a flexible linker. CaM is an accessory protein that partially inhibits RyR2 channel activity. CaM is critical for normal cardiac function, and altered CaM binding and efficacy may contribute to defects in SR calcium release. The present paper reviews CaM binding to RyR2 and how it regulates RyR2 channel activity. It then goes on to review how mutations in the CaM amino acid sequence give rise to inherited syndromes such as Catecholaminergic Polymorphic Ventricular Tachychardia (CPVT) and long QT syndrome (LQTS). In addition, the role of reduced CaM binding to RyR2 that results from RyR2 phosphorylation or from oxidation of either RyR2 or CaM contributes to the progression of heart failure is reviewed. Finally, this manuscript reviews recent evidence that CaM binding to RyR2 is required for the inhibitory action of a pharmaceutical agent (dantrolene) on RyR2. Dantrolene is a clinically used muscle relaxant that has recently been found to exert antiarrhythmic effects against SR Ca2+ overload arrhythmias.
Collapse
Affiliation(s)
- Kafa Walweel
- School of Biomedical Sciences and Pharmacy, University of Newcastle and Hunter Medical Research Institute, Callaghan, NSW, 2308, Australia
| | - Ye Win Oo
- School of Biomedical Sciences and Pharmacy, University of Newcastle and Hunter Medical Research Institute, Callaghan, NSW, 2308, Australia
| | - Derek R Laver
- School of Biomedical Sciences and Pharmacy, University of Newcastle and Hunter Medical Research Institute, Callaghan, NSW, 2308, Australia
| |
Collapse
|
34
|
Abstract
There has been a significant progress in our understanding of the molecular mechanisms by which calcium (Ca2+) ions mediate various types of cardiac arrhythmias. A growing list of inherited gene defects can cause potentially lethal cardiac arrhythmia syndromes, including catecholaminergic polymorphic ventricular tachycardia, congenital long QT syndrome, and hypertrophic cardiomyopathy. In addition, acquired deficits of multiple Ca2+-handling proteins can contribute to the pathogenesis of arrhythmias in patients with various types of heart disease. In this review article, we will first review the key role of Ca2+ in normal cardiac function-in particular, excitation-contraction coupling and normal electric rhythms. The functional involvement of Ca2+ in distinct arrhythmia mechanisms will be discussed, followed by various inherited arrhythmia syndromes caused by mutations in Ca2+-handling proteins. Finally, we will discuss how changes in the expression of regulation of Ca2+ channels and transporters can cause acquired arrhythmias, and how these mechanisms might be targeted for therapeutic purposes.
Collapse
Affiliation(s)
- Andrew P Landstrom
- From the Section of Cardiology, Department of Pediatrics (A.P.L.), Cardiovascular Research Institute (A.P.L., X.H.T.W.), and Departments of Molecular Physiology and Biophysics, Medicine (Cardiology), Center for Space Medicine (X.H.T.W.), Baylor College of Medicine, Houston, TX; and Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (D.D.)
| | - Dobromir Dobrev
- From the Section of Cardiology, Department of Pediatrics (A.P.L.), Cardiovascular Research Institute (A.P.L., X.H.T.W.), and Departments of Molecular Physiology and Biophysics, Medicine (Cardiology), Center for Space Medicine (X.H.T.W.), Baylor College of Medicine, Houston, TX; and Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (D.D.)
| | - Xander H T Wehrens
- From the Section of Cardiology, Department of Pediatrics (A.P.L.), Cardiovascular Research Institute (A.P.L., X.H.T.W.), and Departments of Molecular Physiology and Biophysics, Medicine (Cardiology), Center for Space Medicine (X.H.T.W.), Baylor College of Medicine, Houston, TX; and Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (D.D.).
| |
Collapse
|
35
|
Reddish FN, Miller CL, Gorkhali R, Yang JJ. Calcium Dynamics Mediated by the Endoplasmic/Sarcoplasmic Reticulum and Related Diseases. Int J Mol Sci 2017; 18:E1024. [PMID: 28489021 PMCID: PMC5454937 DOI: 10.3390/ijms18051024] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2017] [Revised: 04/28/2017] [Accepted: 05/01/2017] [Indexed: 12/17/2022] Open
Abstract
The flow of intracellular calcium (Ca2+) is critical for the activation and regulation of important biological events that are required in living organisms. As the major Ca2+ repositories inside the cell, the endoplasmic reticulum (ER) and the sarcoplasmic reticulum (SR) of muscle cells are central in maintaining and amplifying the intracellular Ca2+ signal. The morphology of these organelles, along with the distribution of key calcium-binding proteins (CaBPs), regulatory proteins, pumps, and receptors fundamentally impact the local and global differences in Ca2+ release kinetics. In this review, we will discuss the structural and morphological differences between the ER and SR and how they influence localized Ca2+ release, related diseases, and the need for targeted genetically encoded calcium indicators (GECIs) to study these events.
Collapse
Affiliation(s)
- Florence N Reddish
- Department of Chemistry, Center for Diagnostics and Therapeutics (CDT), Georgia State University, Atlanta, GA 30303, USA.
| | - Cassandra L Miller
- Department of Chemistry, Center for Diagnostics and Therapeutics (CDT), Georgia State University, Atlanta, GA 30303, USA.
| | - Rakshya Gorkhali
- Department of Chemistry, Center for Diagnostics and Therapeutics (CDT), Georgia State University, Atlanta, GA 30303, USA.
| | - Jenny J Yang
- Department of Chemistry, Center for Diagnostics and Therapeutics (CDT), Georgia State University, Atlanta, GA 30303, USA.
| |
Collapse
|
36
|
Characterization of Post-Translational Modifications to Calsequestrins of Cardiac and Skeletal Muscle. Int J Mol Sci 2016; 17:ijms17091539. [PMID: 27649144 PMCID: PMC5037814 DOI: 10.3390/ijms17091539] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 09/05/2016] [Accepted: 09/08/2016] [Indexed: 11/26/2022] Open
Abstract
Calsequestrin is glycosylated and phosphorylated during its transit to its final destination in the junctional sarcoplasmic reticulum. To determine the significance and universal profile of these post-translational modifications to mammalian calsequestrin, we characterized, via mass spectrometry, the glycosylation and phosphorylation of skeletal muscle calsequestrin from cattle (B. taurus), lab mice (M. musculus) and lab rats (R. norvegicus) and cardiac muscle calsequestrin from cattle, lab rats and humans. On average, glycosylation of skeletal calsequestrin consisted of two N-acetylglucosamines and one mannose (GlcNAc2Man1), while cardiac calsequestrin had five additional mannoses (GlcNAc2Man6). Skeletal calsequestrin was not phosphorylated, while the C-terminal tails of cardiac calsequestrin contained between zero to two phosphoryls, indicating that phosphorylation of cardiac calsequestrin may be heterogeneous in vivo. Static light scattering experiments showed that the Ca2+-dependent polymerization capabilities of native bovine skeletal calsequestrin are enhanced, relative to the non-glycosylated, recombinant isoform, which our crystallographic studies suggest may be due to glycosylation providing a dynamic “guiderail”-like scaffold for calsequestrin polymerization. Glycosylation likely increases a polymerization/depolymerization response to changing Ca2+ concentrations, and proper glycosylation, in turn, guarantees both effective Ca2+ storage/buffering of the sarcoplasmic reticulum and localization of calsequestrin (Casq) at its target site.
Collapse
|
37
|
Rajagopalan A, Pollanen MS. Sudden death during struggle in the setting of heterozygosity for a mutation in calsequesterin 2. Forensic Sci Med Pathol 2015; 12:86-9. [PMID: 26671417 DOI: 10.1007/s12024-015-9733-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/24/2015] [Indexed: 01/02/2023]
Abstract
Instances of sudden and unexpected death while in police custody remain complex and controversial cases in forensic pathology, and provide unique diagnostic challenges. In general, the circumstances of these cases have resulted in two major theories to account for these deaths: "excited delirium syndrome", and positional (restraint) asphyxia. However, some cases that are not easily explained by one of these theories may be best explained by a theory from another emergent area in forensic pathology, non-structural genetic heart disease. We present one such case, a sudden arrhythmic death during struggle/restraint. A 45 year old man with developmental delay was walking outdoors as part of his daily routine. He was misidentified as a criminal suspect by police officers, who attempted to take him into custody. He resisted this arrest violently. He was taken to the ground, and restrained in a face-down position. Both police and civilian witness state that he was pushing his chest off the ground with his arms, when he suddenly collapsed and died. The interaction with police lasted approximately 3 min. There was no prior excited delirium. At autopsy, minor external blunt force injuries were observed. The heart showed mild cardiomegaly with concentric left ventricular hypertrophy, and sub-occlusive coronary atherosclerosis. Toxicological testing was negative for common drugs, including cocaine and its metabolites. Post-mortem molecular testing demonstrated this man to be heterozygous for a catecholaminergic polymorphic ventricular tachycardia (CPVT) associated mutation (Phe189Leu) in the calsequestrin 2 (CASQ2) gene. This mutation was classified as a class I mutation (deleterious), that may cause disease in a heterozygous state. The cause of death was given as cardiac arrhythmia precipitated by struggle/restraint in a man with CPVT. This case illustrates the difficulty assigning a scientific cause of death in rare and controversial cases, and the value of the molecular autopsy in identifying disease causing mutations.
Collapse
Affiliation(s)
- Ashwyn Rajagopalan
- Department of Laboratory Medicine and Pathobiology, Ontario Forensic Pathology Service and Centre for Forensic Science and Medicine, University of Toronto, Toronto, ON, Canada.
| | - Michael S Pollanen
- Department of Laboratory Medicine and Pathobiology, Ontario Forensic Pathology Service and Centre for Forensic Science and Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
38
|
Ning F, Luo L, Ahmad S, Valli H, Jeevaratnam K, Wang T, Guzadhur L, Yang D, Fraser JA, Huang CLH, Ma A, Salvage SC. The RyR2-P2328S mutation downregulates Nav1.5 producing arrhythmic substrate in murine ventricles. Pflugers Arch 2015; 468:655-65. [PMID: 26545784 PMCID: PMC4792352 DOI: 10.1007/s00424-015-1750-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2015] [Revised: 09/25/2015] [Accepted: 10/19/2015] [Indexed: 01/05/2023]
Abstract
Catecholaminergic polymorphic ventricular tachycardia (CPVT) predisposes to ventricular arrhythmia due to altered Ca2+ homeostasis and can arise from ryanodine receptor (RyR2) mutations including RyR2-P2328S. Previous reports established that homozygotic murine RyR2-P2328S (RyR2S/S) hearts show an atrial arrhythmic phenotype associated with reduced action potential (AP) conduction velocity and sodium channel (Nav1.5) expression. We now relate ventricular arrhythmogenicity and slowed AP conduction in RyR2S/S hearts to connexin-43 (Cx43) and Nav1.5 expression and Na+ current (INa). Stimulation protocols applying extrasystolic S2 stimulation following 8 Hz S1 pacing at progressively decremented S1S2 intervals confirmed an arrhythmic tendency despite unchanged ventricular effective refractory periods (VERPs) in Langendorff-perfused RyR2S/S hearts. Dynamic pacing imposing S1 stimuli then demonstrated that progressive reductions of basic cycle lengths (BCLs) produced greater reductions in conduction velocity at equivalent BCLs and diastolic intervals in RyR2S/S than WT, but comparable changes in AP durations (APD90) and their alternans. Western blot analyses demonstrated that Cx43 protein expression in whole ventricles was similar, but Nav1.5 expression in both whole tissue and membrane fractions were significantly reduced in RyR2S/S compared to wild-type (WT). Loose patch-clamp studies similarly demonstrated reduced INa in RyR2S/S ventricles. We thus attribute arrhythmogenesis in RyR2S/S ventricles resulting from arrhythmic substrate produced by reduced conduction velocity to downregulated Nav1.5 reducing INa, despite normal determinants of repolarization and passive conduction. The measured changes were quantitatively compatible with earlier predictions of linear relationships between conduction velocity and the peak INa of the AP but nonlinear relationships between peak INa and maximum Na+ permeability.
Collapse
Affiliation(s)
- Feifei Ning
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi'an Jiaotong University, No. 277 West Yanta Road, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Ling Luo
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi'an Jiaotong University, No. 277 West Yanta Road, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Shiraz Ahmad
- Physiological Laboratory, University of Cambridge, Cambridge, CB2 3EG, UK
| | - Haseeb Valli
- Physiological Laboratory, University of Cambridge, Cambridge, CB2 3EG, UK
| | - Kamalan Jeevaratnam
- Faculty of Health and Medical Science, Duke of Kent Building, University of Surrey, Guildford, GU2 7TE, UK
- Perdana University-Royal College of Surgeons Ireland, 43400 Serdang, Selangor, Darul Ehsan, Malaysia
| | - Tingzhong Wang
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi'an Jiaotong University, No. 277 West Yanta Road, Xi'an, Shaanxi, 710061, People's Republic of China
- Key Laboratory of Molecular Cardiology, Shaanxi Province, People's Republic of China
- Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an, People's Republic of China
| | - Laila Guzadhur
- Department of Biochemistry, University of Cambridge, Cambridge, CB2 1QW, UK
- Niche Science and Technology, Falstaff House, Bardolph Road, Richmond, UK
| | - Dandan Yang
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi'an Jiaotong University, No. 277 West Yanta Road, Xi'an, Shaanxi, 710061, People's Republic of China
| | - James A Fraser
- Physiological Laboratory, University of Cambridge, Cambridge, CB2 3EG, UK
| | - Christopher L-H Huang
- Physiological Laboratory, University of Cambridge, Cambridge, CB2 3EG, UK
- Department of Biochemistry, University of Cambridge, Cambridge, CB2 1QW, UK
| | - Aiqun Ma
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi'an Jiaotong University, No. 277 West Yanta Road, Xi'an, Shaanxi, 710061, People's Republic of China.
- Key Laboratory of Molecular Cardiology, Shaanxi Province, People's Republic of China.
- Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an, People's Republic of China.
| | - Samantha C Salvage
- Physiological Laboratory, University of Cambridge, Cambridge, CB2 3EG, UK.
| |
Collapse
|
39
|
Lewis KM, Ronish LA, Ríos E, Kang C. Characterization of Two Human Skeletal Calsequestrin Mutants Implicated in Malignant Hyperthermia and Vacuolar Aggregate Myopathy. J Biol Chem 2015; 290:28665-74. [PMID: 26416891 DOI: 10.1074/jbc.m115.686261] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Indexed: 12/14/2022] Open
Abstract
Calsequestrin 1 is the principal Ca(2+) storage protein of the sarcoplasmic reticulum of skeletal muscle. Its inheritable D244G mutation causes a myopathy with vacuolar aggregates, whereas its M87T "variant" is weakly associated with malignant hyperthermia. We characterized the consequences of these mutations with studies of the human proteins in vitro. Equilibrium dialysis and turbidity measurements showed that D244G and, to a lesser extent, M87T partially lose Ca(2+) binding exhibited by wild type calsequestrin 1 at high Ca(2+) concentrations. D244G aggregates abruptly and abnormally, a property that fully explains the protein inclusions that characterize its phenotype. D244G crystallized in low Ca(2+) concentrations lacks two Ca(2+) ions normally present in wild type that weakens the hydrophobic core of Domain II. D244G crystallized in high Ca(2+) concentrations regains its missing ions and Domain II order but shows a novel dimeric interaction. The M87T mutation causes a major shift of the α-helix bearing the mutated residue, significantly weakening the back-to-back interface essential for tetramerization. D244G exhibited the more severe structural and biophysical property changes, which matches the different pathophysiological impacts of these mutations.
Collapse
Affiliation(s)
- Kevin M Lewis
- From the Department of Chemistry, Washington State University, Pullman, Washington 99164-4630
| | - Leslie A Ronish
- School of Molecular Biosciences, Washington State University, Pullman, Washington 99164-4660, and
| | - Eduardo Ríos
- Department of Molecular Biophysics and Physiology, Rush University, Chicago, Illinois 60612
| | - ChulHee Kang
- From the Department of Chemistry, Washington State University, Pullman, Washington 99164-4630, School of Molecular Biosciences, Washington State University, Pullman, Washington 99164-4660, and
| |
Collapse
|
40
|
Eschenhagen T, Mummery C, Knollmann BC. Modelling sarcomeric cardiomyopathies in the dish: from human heart samples to iPSC cardiomyocytes. Cardiovasc Res 2015; 105:424-38. [PMID: 25618410 PMCID: PMC4349163 DOI: 10.1093/cvr/cvv017] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
One of the obstacles to a better understanding of the pathogenesis of human cardiomyopathies has been poor availability of heart-tissue samples at early stages of disease development. This has possibly changed by the advent of patient-derived induced pluripotent stem cell (hiPSC) from which cardiomyocytes can be derived in vitro. The main promise of hiPSC technology is that by capturing the effects of thousands of individual gene variants, the phenotype of differentiated derivatives of these cells will provide more information on a particular disease than simple genotyping. This article summarizes what is known about the ‘human cardiomyopathy or heart failure phenotype in vitro’, which constitutes the reference for modelling sarcomeric cardiomyopathies in hiPSC-derived cardiomyocytes. The current techniques for hiPSC generation and cardiac myocyte differentiation are briefly reviewed and the few published reports of hiPSC models of sarcomeric cardiomyopathies described. A discussion of promises and challenges of hiPSC-modelling of sarcomeric cardiomyopathies and individualized approaches is followed by a number of questions that, in the view of the authors, need to be answered before the true potential of this technology can be evaluated.
Collapse
Affiliation(s)
- Thomas Eschenhagen
- Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Martinistr. 52, 20246 Hamburg, Germany DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck
| | - Christine Mummery
- Department of Anatomy and Embryology, Leiden University Medical Centre, Einthovenweg 20, 2333ZC Leiden, The Netherlands
| | - Bjorn C Knollmann
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University School of Medicine, 2215 Garland Ave, Nashville, TN 37232, USA
| |
Collapse
|
41
|
Zanella F, Lyon RC, Sheikh F. Modeling heart disease in a dish: from somatic cells to disease-relevant cardiomyocytes. Trends Cardiovasc Med 2013; 24:32-44. [PMID: 24054750 DOI: 10.1016/j.tcm.2013.06.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Revised: 06/07/2013] [Accepted: 06/10/2013] [Indexed: 01/02/2023]
Abstract
A scientific milestone that has tremendously impacted the cardiac research field has been the discovery and establishment of human-induced pluripotent stem cells (hiPSC). Key to this discovery has been uncovering a viable path in generating human patient and disease-specific cardiac cells to dynamically model and study human cardiac diseases in an in vitro setting. Recent studies have demonstrated that hiPSC-derived cardiomyocytes can be used to model and recapitulate various known disease features in hearts of patient donors harboring genetic-based cardiac diseases. Experimental drugs have also been tested in this setting and shown to alleviate disease phenotypes in hiPSC-derived cardiomyocytes, further paving the way for therapeutic interventions for cardiac disease. Here, we review state-of-the-art methods to generate high-quality hiPSC and differentiate them towards cardiomyocytes as well as the full range of genetic-based cardiac diseases, which have been modeled using hiPSC. We also provide future perspectives on exploiting the potential of hiPSC to compliment existing studies and gain new insights into the mechanisms underlying cardiac disease.
Collapse
Affiliation(s)
- Fabian Zanella
- Department of Medicine (Cardiology Division), University of California-San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Robert C Lyon
- Department of Medicine (Cardiology Division), University of California-San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Farah Sheikh
- Department of Medicine (Cardiology Division), University of California-San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA.
| |
Collapse
|
42
|
Knollmann BC. Induced pluripotent stem cell-derived cardiomyocytes: boutique science or valuable arrhythmia model? Circ Res 2013; 112:969-76; discussion 976. [PMID: 23569106 DOI: 10.1161/circresaha.112.300567] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
This article reviews the strengths and limitations of induced pluripotent stem cell-derived cardiomyocytes (iPSC-CM) as models of cardiac arrhythmias. Specifically, the article attempts to answer the following questions: Which clinical arrhythmias can be modeled by iPSC-CM? How well can iPSC-CM model adult ventricular myocytes? What are the strengths and limitations of published iPSC-CM arrhythmia models? What new mechanistic insight has been gained? What is the evidence that would support using iPSC-CM to personalize antiarrhythmic drug therapy? The review also discusses the pros and cons of using the iPSC-CM technology for modeling specific genetic arrhythmia disorders, such as long QT syndrome, Brugada Syndrome, or Catecholaminergic Polymorphic Ventricular Tachycardia.
Collapse
Affiliation(s)
- Björn C Knollmann
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical School, Nashville, TN 37232-0575, USA.
| |
Collapse
|