1
|
Ren AJ, Wei C, Liu YJ, Liu M, Wang P, Fan J, Wang K, Zhang S, Qin Z, Ren QX, Zheng Y, Chen YX, Xie Z, Gao L, Zhu Y, Zhang Y, Yang HT, Zhang WJ. ZBTB20 Regulates SERCA2a Activity and Myocardial Contractility Through Phospholamban. Circ Res 2024; 134:252-265. [PMID: 38166470 DOI: 10.1161/circresaha.123.323798] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 12/20/2023] [Indexed: 01/04/2024]
Abstract
BACKGROUND Intracellular Ca2+ cycling determines myocardial contraction and relaxation in response to physiological demands. SERCA2a (sarcoplasmic/endoplasmic reticulum Ca2+-ATPase 2a) is responsible for the sequestration of cytosolic Ca2+ into intracellular stores during cardiac relaxation, and its activity is reversibly inhibited by PLN (phospholamban). However, the regulatory hierarchy of SERCA2a activity remains unclear. METHODS Cardiomyocyte-specific ZBTB20 knockout mice were generated by crossing ZBTB20flox mice with Myh6-Cre mice. Echocardiography, blood pressure measurements, Langendorff perfusion, histological analysis and immunohistochemistry, quantitative reverse transcription-PCR, Western blot analysis, electrophysiological measurements, and chromatin immunoprecipitation assay were performed to clarify the phenotype and elucidate the molecular mechanisms. RESULTS Specific ablation of ZBTB20 in cardiomyocyte led to a significant increase in basal myocardial contractile parameters both in vivo and in vitro, accompanied by an impairment in cardiac reserve and exercise capacity. Moreover, the cardiomyocytes lacking ZBTB20 showed an increase in sarcoplasmic reticular Ca2+ content and exhibited a remarkable enhancement in both SERCA2a activity and electrically stimulated contraction. Mechanistically, PLN expression was dramatically reduced in cardiomyocytes at the mRNA and protein levels by ZBTB20 deletion or silencing, and PLN overexpression could largely restore the basal contractility in ZBTB20-deficient cardiomyocytes. CONCLUSIONS These data point to ZBTB20 as a fine-tuning modulator of PLN expression and SERCA2a activity, thereby offering new perspective on the regulation of basal contractility in the mammalian heart.
Collapse
Affiliation(s)
- An-Jing Ren
- Department of Pathophysiology, Naval Medical University, Shanghai, China (A.-J.R., C.W., M.L., P.W., K.W., Z.Q., Q.-X.R., Y.-X.C., W.J.Z.)
- Experimental Teaching Center, College of Basic Medical Sciences, Naval Medical University, Shanghai, China (A.-J.R., J.F.)
| | - Chunchun Wei
- Department of Pathophysiology, Naval Medical University, Shanghai, China (A.-J.R., C.W., M.L., P.W., K.W., Z.Q., Q.-X.R., Y.-X.C., W.J.Z.)
| | - Ya-Jin Liu
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Tianjin Institute of Endocrinology and Chu Hsien-I Memorial Hospital, Tianjin Medical University Tianjin, China (Y.-J.L., Y. Zhu, W.J.Z.)
| | - Mengna Liu
- Department of Pathophysiology, Naval Medical University, Shanghai, China (A.-J.R., C.W., M.L., P.W., K.W., Z.Q., Q.-X.R., Y.-X.C., W.J.Z.)
| | - Ping Wang
- Department of Pathophysiology, Naval Medical University, Shanghai, China (A.-J.R., C.W., M.L., P.W., K.W., Z.Q., Q.-X.R., Y.-X.C., W.J.Z.)
| | - Juan Fan
- Experimental Teaching Center, College of Basic Medical Sciences, Naval Medical University, Shanghai, China (A.-J.R., J.F.)
| | - Kai Wang
- Department of Pathophysiology, Naval Medical University, Shanghai, China (A.-J.R., C.W., M.L., P.W., K.W., Z.Q., Q.-X.R., Y.-X.C., W.J.Z.)
| | - Sha Zhang
- Department of Cardiovascular Diseases, Changhai Hospital, Naval Medical University, Shanghai, China (S.Z.)
| | - Zhenbang Qin
- Department of Pathophysiology, Naval Medical University, Shanghai, China (A.-J.R., C.W., M.L., P.W., K.W., Z.Q., Q.-X.R., Y.-X.C., W.J.Z.)
| | - Qiu-Xiao Ren
- Department of Pathophysiology, Naval Medical University, Shanghai, China (A.-J.R., C.W., M.L., P.W., K.W., Z.Q., Q.-X.R., Y.-X.C., W.J.Z.)
| | - Yanjun Zheng
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Laboratory of Molecular Cardiology, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, China (Y. Zheng, H.-T.Y.)
| | - Yu-Xia Chen
- Department of Pathophysiology, Naval Medical University, Shanghai, China (A.-J.R., C.W., M.L., P.W., K.W., Z.Q., Q.-X.R., Y.-X.C., W.J.Z.)
| | - Zhifang Xie
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Institute of Early Life Health, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, China (Z.X.)
| | - Ling Gao
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, China (L.G.)
| | - Yi Zhu
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Tianjin Institute of Endocrinology and Chu Hsien-I Memorial Hospital, Tianjin Medical University Tianjin, China (Y.-J.L., Y. Zhu, W.J.Z.)
| | - Youyi Zhang
- Institute of Vascular Medicine, National Key Laboratory of Cardiovascular Homeostasis and Remodeling, Peking University Third Hospital, Beijing, China (Y. Zhang)
| | - Huang-Tian Yang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Laboratory of Molecular Cardiology, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, China (Y. Zheng, H.-T.Y.)
| | - Weiping J Zhang
- Department of Pathophysiology, Naval Medical University, Shanghai, China (A.-J.R., C.W., M.L., P.W., K.W., Z.Q., Q.-X.R., Y.-X.C., W.J.Z.)
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Tianjin Institute of Endocrinology and Chu Hsien-I Memorial Hospital, Tianjin Medical University Tianjin, China (Y.-J.L., Y. Zhu, W.J.Z.)
| |
Collapse
|
2
|
Vora N, Patel P, Gajjar A, Ladani P, Konat A, Bhanderi D, Gadam S, Prajjwal P, Sharma K, Arunachalam SP. Gene therapy for heart failure: A novel treatment for the age old disease. Dis Mon 2024; 70:101636. [PMID: 37734966 DOI: 10.1016/j.disamonth.2023.101636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/23/2023]
Abstract
Across the globe, cardiovascular disease (CVD) is the leading cause of mortality. According to reports, around 6.2 million people in the United states have heart failure. Current standards of care for heart failure can delay but not prevent progression of disease. Gene therapy is one of the novel treatment modalities that promises to fill this limitation in the current standard of care for Heart Failure. In this paper we performed an extensive search of the literature on various advances made in gene therapy for heart failure till date. We review the delivery methods, targets, current applications, trials, limitations and feasibility of gene therapy for heart failure. Various methods have been employed till date for administering gene therapies including but not limited to arterial and venous infusion, direct myocardial injection and pericardial injection. Various strategies such as AC6 expression, S100A1 protein upregulation, VEGF-B and SDF-1 gene therapy have shown promise in recent preclinical trials. Furthermore, few studies even show that stimulation of cardiomyocyte proliferation such as through cyclin A2 overexpression is a realistic avenue. However, a considerable number of obstacles need to be overcome for gene therapy to be part of standard treatment of care such as definitive choice of gene, gene delivery systems and a suitable method for preclinical trials and clinical trials on patients. Considering the challenges and taking into account the recent advances in gene therapy research, there are encouraging signs to indicate gene therapy for heart failure to be a promising treatment modality for the future. However, the time and feasibility of this option remains in a situation of balance.
Collapse
Affiliation(s)
- Neel Vora
- B. J. Medical College, Ahmedabad, India
| | - Parth Patel
- Pramukhswami Medical College, Karamsad, India
| | | | | | - Ashwati Konat
- University School of Sciences, Gujarat University, Ahmedabad, India
| | | | | | | | - Kamal Sharma
- U. N. Mehta Institute of Cardiology and Research Centre, Ahmedabad, India.
| | | |
Collapse
|
3
|
Mattiazzi A, Kranias EG. Unleashing the Power of Genetics: PLN Ablation, Phospholambanopathies and Evolving Challenges. Circ Res 2024; 134:138-142. [PMID: 38236951 DOI: 10.1161/circresaha.123.323053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2024]
Affiliation(s)
- Alicia Mattiazzi
- Centro de Investigaciones Cardiovasculares, Centro Cientifico Tecnologico-La Plata CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas), Facultad de Ciencias Médicas, Universidad Nacional de La Plata, Argentina (A.M.)
| | - Evangelia G Kranias
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, OH (E.G.K.)
| |
Collapse
|
4
|
Fan X, Yang G, Duru F, Grilli M, Akin I, Zhou X, Saguner AM, Ei-Battrawy I. Arrhythmogenic Cardiomyopathy: from Preclinical Models to Genotype-phenotype Correlation and Pathophysiology. Stem Cell Rev Rep 2023; 19:2683-2708. [PMID: 37731079 PMCID: PMC10661732 DOI: 10.1007/s12015-023-10615-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/23/2023] [Indexed: 09/22/2023]
Abstract
Arrhythmogenic cardiomyopathy (ACM) is a hereditary myocardial disease characterized by the replacement of the ventricular myocardium with fibrous fatty deposits. ACM is usually inherited in an autosomal dominant pattern with variable penetrance and expressivity, which is mainly related to ventricular tachyarrhythmia and sudden cardiac death (SCD). Importantly, significant progress has been made in determining the genetic background of ACM due to the development of new techniques for genetic analysis. The exact molecular pathomechanism of ACM, however, is not completely clear and the genotype-phenotype correlations have not been fully elucidated, which are useful to predict the prognosis and treatment of ACM patients. Different gene-targeted and transgenic animal models, human-induced pluripotent stem cell-derived cardiomyocyte (hiPSC-CM) models, and heterologous expression systems have been developed. Here, this review aims to summarize preclinical ACM models and platforms promoting our understanding of the pathogenesis of ACM and assess their value in elucidating the ACM genotype-phenotype relationship.
Collapse
Affiliation(s)
- Xuehui Fan
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention of Cardiovascular Diseases, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, China
- Cardiology, Angiology, Haemostaseology, and Medical Intensive Care, Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
- European Center for AngioScience (ECAS), German Center for Cardiovascular Research (DZHK) Partner Site Heidelberg/ Mannheim, and Centre for Cardiovascular Acute Medicine Mannheim (ZKAM), Medical Centre Mannheim, Heidelberg University, Partner Site, Heidelberg-Mannheim, Germany
| | - Guoqiang Yang
- Cardiology, Angiology, Haemostaseology, and Medical Intensive Care, Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
- Department of Acupuncture and Rehabilitation, the Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
- Research Unit of Molecular Imaging Probes, Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Firat Duru
- Department of Cardiology, University Heart Centre, University Hospital Zurich, Zurich, Switzerland
| | - Maurizio Grilli
- Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany
| | - Ibrahim Akin
- Cardiology, Angiology, Haemostaseology, and Medical Intensive Care, Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
- European Center for AngioScience (ECAS), German Center for Cardiovascular Research (DZHK) Partner Site Heidelberg/ Mannheim, and Centre for Cardiovascular Acute Medicine Mannheim (ZKAM), Medical Centre Mannheim, Heidelberg University, Partner Site, Heidelberg-Mannheim, Germany
| | - Xiaobo Zhou
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention of Cardiovascular Diseases, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, China.
- Cardiology, Angiology, Haemostaseology, and Medical Intensive Care, Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany.
- European Center for AngioScience (ECAS), German Center for Cardiovascular Research (DZHK) Partner Site Heidelberg/ Mannheim, and Centre for Cardiovascular Acute Medicine Mannheim (ZKAM), Medical Centre Mannheim, Heidelberg University, Partner Site, Heidelberg-Mannheim, Germany.
- First Department of Medicine, University Medical Centre Mannheim, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany.
| | - Ardan Muammer Saguner
- Department of Cardiology, University Heart Centre, University Hospital Zurich, Zurich, Switzerland
| | - Ibrahim Ei-Battrawy
- European Center for AngioScience (ECAS), German Center for Cardiovascular Research (DZHK) Partner Site Heidelberg/ Mannheim, and Centre for Cardiovascular Acute Medicine Mannheim (ZKAM), Medical Centre Mannheim, Heidelberg University, Partner Site, Heidelberg-Mannheim, Germany.
- Department of Cardiology and Angiology, Ruhr University, Bochum, Germany; Institute of Physiology, Department of Cellular and Translational Physiology and Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr- University Bochum, Bochum, Germany.
| |
Collapse
|
5
|
Masenga SK, Kirabo A. Salt and Gut Microbiota in Heart Failure. Curr Hypertens Rep 2023; 25:173-184. [PMID: 37219766 DOI: 10.1007/s11906-023-01245-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/09/2023] [Indexed: 05/24/2023]
Abstract
PURPOSE OF REVIEW The role and underlying mechanisms mediated by dietary salt in modulating the gut microbiota and contributing to heart failure (HF) are not clear. This review summarizes the mechanisms of dietary salt and the gut-heart axis in HF. RECENT FINDINGS The gut microbiota has been implicated in several cardiovascular diseases (CVDs) including HF. Dietary factors including high consumption of salt play a role in influencing the gut microbiota, resulting in dysbiosis. An imbalance of microbial species due to a reduction in microbial diversity with accompanying immune cell activation has been implicated in the pathogenesis of HF via several mechanisms. The gut microbiota and gut-associated metabolites contribute to HF by reducing gut microbiota biodiversity and activating several signaling pathways. High dietary salt modulates the gut microbiota composition and exacerbate or induce HF by increasing the expression of the epithelial sodium/hydrogen exchanger isoform 3 in the gut, cardiac expression of beta myosin heavy chain, activation of the myocyte enhancer factor/nuclear factor of activated T cell, and salt-inducible kinase 1. These mechanisms explain the resulting structural and functional derangements in patients with HF.
Collapse
Affiliation(s)
- Sepiso K Masenga
- HAND Research Group, School of Medicine and Health Sciences, Mulungushi University, Livingstone Campus, Zambia
- Department of Medicine, Vanderbilt University Medical Center, Room 536 Robinson Research Building, Nashville, TN, 37232-6602, USA
| | - Annet Kirabo
- Department of Medicine, Vanderbilt University Medical Center, Room 536 Robinson Research Building, Nashville, TN, 37232-6602, USA.
| |
Collapse
|
6
|
Hou J, Liang WY, Xiong S, Long P, Yue T, Wen X, Wang T, Deng H. Identification of hub genes and potential ceRNA networks of diabetic cardiomyopathy. Sci Rep 2023; 13:10258. [PMID: 37355664 PMCID: PMC10290640 DOI: 10.1038/s41598-023-37378-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 06/21/2023] [Indexed: 06/26/2023] Open
Abstract
Diabetic cardiomyopathy (DCM), a common complication of diabetes, is defined as ventricular dysfunction in the absence of underlying heart disease. Noncoding RNAs (ncRNAs), including long noncoding RNAs (lncRNAs) and microRNAs (miRNAs), play a crucial role in the development of DCM. Weighted Gene Co-Expression Network Analysis (WGCNA) was used to identify key modules in DCM-related pathways. DCM-related miRNA-mRNA network and DCM-related ceRNA network were constructed by miRNA-seq to identify hub genes in these modules. We identified five hub genes that are associated with the onset of DCM, including Troponin C1 (Tnnc1), Phospholamban (Pln), Fatty acid binding proteins 3 (Fabp3), Popeye domain containing 2 (Popdc2), and Tripartite Motif-containing Protein 63 (Trim63). miRNAs that target the hub genes were mainly involved in TGF-β and Wnt signaling pathways. GO BP enrichment analysis found these miRNAs were involved in the signaling of TGF-β and glucose homeostasis. Q-PCR results found the gene expressions of Pln, Fabp3, Trim63, Tnnc1, and Popdc2 were significantly increased in DCM. Our study identified five hub genes (Tnnc1, Pln, Fabp3, Popdc2, Trim63) whose associated ceRNA networks are responsible for the onset of DCM.
Collapse
Affiliation(s)
- Jun Hou
- Department of Cardiology, The Third People's Hospital of Chengdu/Affiliated Hospital of Southwest Jiaotong University, Chengdu, Sichuan, China
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, China
| | - Wan Yi Liang
- Department of Microbiology and Immunology, Faculty of Science, University of British Columbia, Vancouver, BC, Canada
| | - Shiqiang Xiong
- Department of Cardiology, The Third People's Hospital of Chengdu/Affiliated Hospital of Southwest Jiaotong University, Chengdu, Sichuan, China
| | - Pan Long
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, China
| | - Tian Yue
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, China
| | - Xudong Wen
- Department of Gastroenterology and Hepatology, Chengdu First People's Hospital, Chengdu, Sichuan, China
| | - Tianchen Wang
- Alfred E. Mann Department of Biomedical Engineering, University of South California, Los Angeles, CA, USA
| | - Haoyu Deng
- Department of Medicine, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada.
- Centre for Heart and Lung Innovation, St. Paul's Hospital, University of British Columbia, 1081 Burrard Street, Vancouver, BC, V6Z 1Y6, Canada.
| |
Collapse
|
7
|
Rustad MD, Roopnarine O, Cornea RL, Thomas DD. Interaction of DWORF with SERCA and PLB as determined by EPR spectroscopy. Biochem Biophys Res Commun 2023; 645:97-102. [PMID: 36682333 PMCID: PMC9951557 DOI: 10.1016/j.bbrc.2023.01.041] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 01/13/2023] [Indexed: 01/15/2023]
Abstract
Insufficient sarco/endoplasmic reticulum calcium ATPase (SERCA) activity significantly contributes to heart failure, which is a leading cause of death worldwide. A characteristic pathology of cardiac disease is the slow and incomplete Ca2+ removal from the myocyte cytoplasm in diastole, which is primarily driven by SERCA, the integral transmembrane Ca2+ pump. Phospholamban (PLB) allosterically inhibits SERCA by reducing its apparent Ca2+ affinity. Recently, the 34-codon novel dwarf open reading frame (DWORF) micropeptide has been identified as a muscle-specific SERCA effector, capable of reversing the inhibitory effects of PLB and independently activating SERCA in the absence of PLB. However, the structural basis for these functions has not yet been determined in a system of defined molecular components. We have used electron paramagnetic resonance (EPR) spectroscopy to investigate the protein-protein interactions of DWORF, co-reconstituted in proteoliposomes with SERCA and spin-labeled PLB. We analyzed the change of PLB rotational mobility in response to varying DWORF concentration, to quantify competitive binding of DWORF and PLB. We determined that DWORF competes with PLB for binding to SERCA at low [Ca2+], although the measured affinity of DWORF for SERCA is an order of magnitude weaker than that of PLB for SERCA, indicating cooperativity. The sensitivity of EPR to structural dynamics, using stereospecifically attached spin labels, allows us to obtain new information needed to refine the molecular model for regulation of SERCA activity, as needed for development of novel therapeutic remedies against cardiac pathologies.
Collapse
Affiliation(s)
- Mark D Rustad
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, 55455, USA; School of Physics and Astronomy, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Osha Roopnarine
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Razvan L Cornea
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, 55455, USA
| | - David D Thomas
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, 55455, USA.
| |
Collapse
|
8
|
Sarco/endoplasmic reticulum calcium ATPase activity is unchanged despite increased myofilament calcium sensitivity in Zucker type 2 diabetic fatty rat heart. Sci Rep 2022; 12:16904. [PMID: 36207382 PMCID: PMC9546843 DOI: 10.1038/s41598-022-20520-0] [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] [Received: 05/19/2022] [Accepted: 09/14/2022] [Indexed: 11/11/2022] Open
Abstract
Systolic and diastolic dysfunction in diabetes have frequently been associated with abnormal calcium (Ca2+) regulation. However, there is emerging evidence that Ca2+ mishandling alone is insufficient to fully explain diabetic heart dysfunction, with focus shifting to the properties of the myofilament proteins. Our aim was to examine the effects of diabetes on myofilament Ca2+ sensitivity and Ca2+ handling in left ventricular tissues isolated from the same type 2 diabetic rat hearts. We measured the force-pCa relationship in skinned left ventricular cardiomyocytes isolated from 20-week-old type 2 diabetic and non-diabetic rats. Myofilament Ca2+ sensitivity was greater in the diabetic relative to non-diabetic cardiomyocytes, and this corresponded with lower phosphorylation of cardiac troponin I (cTnI) at ser23/24 in the diabetic left ventricular tissues. Protein expression of sarco/endoplasmic reticulum Ca2+-ATPase (SERCA), phosphorylation of phospholamban (PLB) at Ser16, and SERCA/PLB ratio were lower in the diabetic left ventricular tissues. However, the maximum SERCA Ca2+ uptake rate was not different between the diabetic and non-diabetic myocardium. Our data suggest that impaired contractility in the diabetic heart is not caused by SERCA Ca2+ mishandling. This study highlights the important role of the cardiac myofilament and provides new insight on the pathophysiology of diabetic heart dysfunction.
Collapse
|
9
|
Val‐Blasco A, Gil‐Fernández M, Rueda A, Pereira L, Delgado C, Smani T, Ruiz Hurtado G, Fernández‐Velasco M. Ca 2+ mishandling in heart failure: Potential targets. Acta Physiol (Oxf) 2021; 232:e13691. [PMID: 34022101 DOI: 10.1111/apha.13691] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 05/17/2021] [Accepted: 05/19/2021] [Indexed: 12/14/2022]
Abstract
Ca2+ mishandling is a common feature in several cardiovascular diseases such as heart failure (HF). In many cases, impairment of key players in intracellular Ca2+ homeostasis has been identified as the underlying mechanism of cardiac dysfunction and cardiac arrhythmias associated with HF. In this review, we summarize primary novel findings related to Ca2+ mishandling in HF progression. HF research has increasingly focused on the identification of new targets and the contribution of their role in Ca2+ handling to the progression of the disease. Recent research studies have identified potential targets in three major emerging areas implicated in regulation of Ca2+ handling: the innate immune system, bone metabolism factors and post-translational modification of key proteins involved in regulation of Ca2+ handling. Here, we describe their possible contributions to the progression of HF.
Collapse
Affiliation(s)
| | | | - Angélica Rueda
- Department of Biochemistry Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV‐IPN) México City Mexico
| | - Laetitia Pereira
- INSERM UMR‐S 1180 Laboratory of Ca Signaling and Cardiovascular Physiopathology University Paris‐Saclay Châtenay‐Malabry France
| | - Carmen Delgado
- Instituto de Investigaciones Biomédicas Alberto Sols Madrid Spain
- Department of Metabolism and Cell Signalling Biomedical Research Institute "Alberto Sols" CSIC‐UAM Madrid Spain
| | - Tarik Smani
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV) Madrid Spain
- Department of Medical Physiology and Biophysics University of Seville Seville Spain
- Group of Cardiovascular Pathophysiology Institute of Biomedicine of Seville University Hospital of Virgen del Rocío, University of Seville, CSIC Seville Spain
| | - Gema Ruiz Hurtado
- Cardiorenal Translational Laboratory Institute of Research i+12 University Hospital 12 de Octubre Madrid Spain
- CIBER‐CV University Hospita1 12 de Octubre Madrid Spain
| | - Maria Fernández‐Velasco
- La Paz University Hospital Health Research Institute IdiPAZ Madrid Spain
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV) Madrid Spain
| |
Collapse
|
10
|
Cuello F, Knaust AE, Saleem U, Loos M, Raabe J, Mosqueira D, Laufer S, Schweizer M, van der Kraak P, Flenner F, Ulmer BM, Braren I, Yin X, Theofilatos K, Ruiz‐Orera J, Patone G, Klampe B, Schulze T, Piasecki A, Pinto Y, Vink A, Hübner N, Harding S, Mayr M, Denning C, Eschenhagen T, Hansen A. Impairment of the ER/mitochondria compartment in human cardiomyocytes with PLN p.Arg14del mutation. EMBO Mol Med 2021; 13:e13074. [PMID: 33998164 PMCID: PMC8185541 DOI: 10.15252/emmm.202013074] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 03/31/2021] [Accepted: 04/07/2021] [Indexed: 12/28/2022] Open
Abstract
The phospholamban (PLN) p.Arg14del mutation causes dilated cardiomyopathy, with the molecular disease mechanisms incompletely understood. Patient dermal fibroblasts were reprogrammed to hiPSC, isogenic controls were established by CRISPR/Cas9, and cardiomyocytes were differentiated. Mutant cardiomyocytes revealed significantly prolonged Ca2+ transient decay time, Ca2+ -load dependent irregular beating pattern, and lower force. Proteomic analysis revealed less endoplasmic reticulum (ER) and ribosomal and mitochondrial proteins. Electron microscopy showed dilation of the ER and large lipid droplets in close association with mitochondria. Follow-up experiments confirmed impairment of the ER/mitochondria compartment. PLN p.Arg14del end-stage heart failure samples revealed perinuclear aggregates positive for ER marker proteins and oxidative stress in comparison with ischemic heart failure and non-failing donor heart samples. Transduction of PLN p.Arg14del EHTs with the Ca2+ -binding proteins GCaMP6f or parvalbumin improved the disease phenotype. This study identified impairment of the ER/mitochondria compartment without SR dysfunction as a novel disease mechanism underlying PLN p.Arg14del cardiomyopathy. The pathology was improved by Ca2+ -scavenging, suggesting impaired local Ca2+ cycling as an important disease culprit.
Collapse
|
11
|
Raad N, Bittihn P, Cacheux M, Jeong D, Ilkan Z, Ceholski D, Kohlbrenner E, Zhang L, Cai CL, Kranias EG, Hajjar RJ, Stillitano F, Akar FG. Arrhythmia Mechanism and Dynamics in a Humanized Mouse Model of Inherited Cardiomyopathy Caused by Phospholamban R14del Mutation. Circulation 2021; 144:441-454. [PMID: 34024116 DOI: 10.1161/circulationaha.119.043502] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Arginine (Arg) 14 deletion (R14del) in the calcium regulatory protein phospholamban (hPLNR14del) has been identified as a disease-causing mutation in patients with an inherited cardiomyopathy. Mechanisms underlying the early arrhythmogenic phenotype that predisposes carriers of this mutation to sudden death with no apparent structural remodeling remain unclear. METHODS To address this, we performed high spatiotemporal resolution optical mapping of intact hearts from adult knock-in mice harboring the human PLNWT (wildtype [WT], n=12) or the heterozygous human PLNR14del mutation (R14del, n=12) before and after ex vivo challenge with isoproterenol and rapid pacing. RESULTS Adverse electrophysiological remodeling was evident in the absence of significant structural or hemodynamic changes. R14del hearts exhibited increased arrhythmia susceptibility compared with wildtype. Underlying this susceptibility was preferential right ventricular action potential prolongation that was unresponsive to β-adrenergic stimulation. A steep repolarization gradient at the left ventricular/right ventricular interface provided the substrate for interventricular activation delays and ultimately local conduction block during rapid pacing. This was followed by the initiation of macroreentrant circuits supporting the onset of ventricular tachycardia. Once sustained, these circuits evolved into high-frequency rotors, which in their majority were pinned to the right ventricle. These rotors exhibited unique spatiotemporal dynamics that promoted their increased stability in R14del compared with wildtype hearts. CONCLUSIONS Our findings highlight the crucial role of primary electric remodeling caused by the hPLNR14del mutation. These inherently arrhythmogenic features form the substrate for adrenergic-mediated VT at early stages of PLNR14del induced cardiomyopathy.
Collapse
Affiliation(s)
- Nour Raad
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (N.R., M.C., D.J., Z.I., D.C., E.K., F.S., F.G.A.).,Poliklinik für Innere Medizin I, Rechts der Isar Hospital, Technical University Munich, Germany (N.R.).,German Center for Cardiovascular Research, Munich Heart Alliance (N.R.)
| | - Philip Bittihn
- BioCircuits Institute, University of California, San Diego (P.B.).,Max Planck Institute for Dynamics and Self-Organization, Göttingen, Germany (P.B.)
| | - Marine Cacheux
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (N.R., M.C., D.J., Z.I., D.C., E.K., F.S., F.G.A.)
| | - Dongtak Jeong
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (N.R., M.C., D.J., Z.I., D.C., E.K., F.S., F.G.A.)
| | - Zeki Ilkan
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (N.R., M.C., D.J., Z.I., D.C., E.K., F.S., F.G.A.)
| | - Delaine Ceholski
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (N.R., M.C., D.J., Z.I., D.C., E.K., F.S., F.G.A.)
| | - Erik Kohlbrenner
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (N.R., M.C., D.J., Z.I., D.C., E.K., F.S., F.G.A.)
| | - Lu Zhang
- Indiana University School of Medicine, Indianapolis (L.Z., C.C.)
| | - Chen-Leng Cai
- Indiana University School of Medicine, Indianapolis (L.Z., C.C.)
| | | | - Roger J Hajjar
- Phospholamban Foundation, Middenmeer, The Netherlands (R.J.H.)
| | - Francesca Stillitano
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (N.R., M.C., D.J., Z.I., D.C., E.K., F.S., F.G.A.)
| | - Fadi G Akar
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (N.R., M.C., D.J., Z.I., D.C., E.K., F.S., F.G.A.).,School of Medicine (F.G.A.), Yale University, New Haven, CT.,Department of Biomedical Engineering (F.G.A.). Yale University, New Haven, CT
| |
Collapse
|
12
|
Patel Y, Joseph J. Sodium Intake and Heart Failure. Int J Mol Sci 2020; 21:ijms21249474. [PMID: 33322108 PMCID: PMC7763082 DOI: 10.3390/ijms21249474] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 12/05/2020] [Accepted: 12/10/2020] [Indexed: 12/27/2022] Open
Abstract
Sodium is an essential mineral and nutrient used in dietary practices across the world and is important to maintain proper blood volume and blood pressure. A high sodium diet is associated with increased expression of β—myosin heavy chain, decreased expression of α/β—myosin heavy chain, increased myocyte enhancer factor 2/nuclear factor of activated T cell transcriptional activity, and increased salt-inducible kinase 1 expression, which leads to alteration in myocardial mechanical performance. A high sodium diet is also associated with alterations in various proteins responsible for calcium homeostasis and myocardial contractility. Excessive sodium intake is associated with the development of a variety of comorbidities including hypertension, chronic kidney disease, stroke, and cardiovascular diseases. While the American College of Cardiology/American Heart Association/Heart Failure Society of America guidelines recommend limiting sodium intake to both prevent and manage heart failure, the evidence behind such recommendations is unclear. Our review article highlights evidence and underlying mechanisms favoring and contradicting limiting sodium intake in heart failure.
Collapse
Affiliation(s)
- Yash Patel
- Lifespan Cardiovascular Institute, Warren Alpert Medical School at Brown University, Providence, RI 02914, USA;
| | - Jacob Joseph
- Department of Medicine, Veterans Affairs Boston Healthcare System, Boston, MA 02132, USA
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Correspondence: ; Tel.: +1-857-203-6841
| |
Collapse
|
13
|
Zhang XH, Morad M. Ca 2+ signaling of human pluripotent stem cells-derived cardiomyocytes as compared to adult mammalian cardiomyocytes. Cell Calcium 2020; 90:102244. [PMID: 32585508 PMCID: PMC7483365 DOI: 10.1016/j.ceca.2020.102244] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 06/10/2020] [Accepted: 06/11/2020] [Indexed: 12/23/2022]
Abstract
Human induced pluripotent stem cells derived cardiomyocytes (hiPSC-CMs) have been extensively used for in vitro modeling of human cardiovascular disease, drug screening and pharmacotherapy, but little rigorous studies have been reported on their biophysical or Ca2+ signaling properties. There is also considerable concern as to the level of their maturity and whether they can serve as reliable models for adult human cardiac myocytes. Ultrastructural difference such as lack of t-tubular network, their polygonal shapes, disorganized sarcomeric myofilament, and their rhythmic automaticity, among others, have been cited as evidence for immaturity of hiPSC-CMs. In this review, we will deal with Ca2+ signaling, its regulation, and its stage of maturity as compared to the mammalian adult cardiomyocytes. We shall summarize the data on functional aspects of Ca2+signaling and its parameters that include: L-type calcium channel (Cav1.2), ICa-induced Ca2+release, CICR, and its parameters, cardiac Na/Ca exchanger (NCX1), the ryanodine receptors (RyR2), sarco-reticular Ca2+pump, SERCA2a/PLB, and the contribution of mitochondrial Ca2+ to hiPSC-CMs excitation-contraction (EC)-coupling as compared with adult mammalian cardiomyocytes. The comparative studies suggest that qualitatively hiPSC-CMs have similar Ca2+signaling properties as those of adult cardiomyocytes, but quantitative differences do exist. This review, we hope, will allow the readers to judge for themselves to what extent Ca2+signaling of hiPSC-CMs represents the adult form of this signaling pathway, and whether these cells can be used as good models of human cardiomyocytes.
Collapse
Affiliation(s)
- Xiao-Hua Zhang
- Cardiac Signaling Center of University of South Carolina, Medical University of South Carolina, Clemson University, Charleston SC, United States
| | - Martin Morad
- Cardiac Signaling Center of University of South Carolina, Medical University of South Carolina, Clemson University, Charleston SC, United States.
| |
Collapse
|
14
|
Hamstra SI, Whitley KC, Baranowski RW, Kurgan N, Braun JL, Messner HN, Fajardo VA. The role of phospholamban and GSK3 in regulating rodent cardiac SERCA function. Am J Physiol Cell Physiol 2020; 319:C694-C699. [PMID: 32755452 DOI: 10.1152/ajpcell.00318.2020] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cardiac contractile function is largely mediated by the regulation of Ca2+ cycling throughout the lifespan. The sarco(endo)plasmic reticulum Ca2+ ATPase (SERCA) pump is paramount to cardiac Ca2+ regulation, and it is well established that SERCA dysfunction pathologically contributes to cardiomyopathy and heart failure. Phospholamban (PLN) is a well-known inhibitor of the SERCA pump and its regulation of SERCA2a-the predominant cardiac SERCA isoform-contributes significantly to proper cardiac function. Glycogen synthase kinase 3 (GSK3) is a serine/threonine kinase involved in several metabolic pathways, and we and others have shown that it regulates SERCA function. In this mini-review, we highlight the underlying mechanisms behind GSK3's regulation of SERCA function specifically discussing changes in SERCA2a and PLN expression and its potential protection against oxidative stress. Ultimately, these recent findings that we discuss could have clinical implications in the treatment and prevention of cardiomyopathies and heart failure.
Collapse
Affiliation(s)
- Sophie I Hamstra
- Department of Kinesiology, Brock University, St. Catharines, Ontario, Canada.,Centre for Bone and Muscle Health, Brock University, St. Catharines, Ontario, Canada
| | - Kennedy C Whitley
- Department of Kinesiology, Brock University, St. Catharines, Ontario, Canada.,Centre for Bone and Muscle Health, Brock University, St. Catharines, Ontario, Canada
| | - Ryan W Baranowski
- Department of Kinesiology, Brock University, St. Catharines, Ontario, Canada.,Centre for Bone and Muscle Health, Brock University, St. Catharines, Ontario, Canada
| | - Nigel Kurgan
- Department of Kinesiology, Brock University, St. Catharines, Ontario, Canada.,Centre for Bone and Muscle Health, Brock University, St. Catharines, Ontario, Canada
| | - Jessica L Braun
- Department of Kinesiology, Brock University, St. Catharines, Ontario, Canada.,Centre for Bone and Muscle Health, Brock University, St. Catharines, Ontario, Canada
| | - Holt N Messner
- Department of Kinesiology, Brock University, St. Catharines, Ontario, Canada.,Centre for Bone and Muscle Health, Brock University, St. Catharines, Ontario, Canada
| | - Val A Fajardo
- Department of Kinesiology, Brock University, St. Catharines, Ontario, Canada.,Centre for Bone and Muscle Health, Brock University, St. Catharines, Ontario, Canada
| |
Collapse
|
15
|
Hamstra SI, Kurgan N, Baranowski RW, Qiu L, Watson CJF, Messner HN, MacPherson REK, MacNeil AJ, Roy BD, Fajardo VA. Low-dose lithium feeding increases the SERCA2a-to-phospholamban ratio, improving SERCA function in murine left ventricles. Exp Physiol 2020; 105:666-675. [PMID: 32087034 DOI: 10.1113/ep088061] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 02/21/2020] [Indexed: 12/24/2022]
Abstract
NEW FINDINGS What is the central question of this study? Inhibition of glycogen synthase kinase-3 (GSK3) has been shown to improve cardiac SERCA2a function. Lithium can inhibit GSK3, but therapeutic doses used in treating bipolar disorder can have toxic effects. It has not been determined whether subtherapeutic doses of lithium can improve cardiac SERCA function. What is the main finding and its importance? Using left ventricles from wild-type mice, we found that subtherapeutic lithium feeding for 6 weeks decreased GSK3 activity and increased cardiac SERCA function compared with control-fed mice. These findings warrant the investigation of low-dose lithium feeding in preclinical models of cardiomyopathy and heart failure to determine the therapeutic benefit of GSK3 inhibition. ABSTRACT The sarco(endo)plasmic reticulum Ca2+ -ATPase (SERCA) pump is responsible for regulating calcium (Ca2+ ) within myocytes, with SERCA2a being the dominant isoform in cardiomyocytes. Its inhibitor, phospholamban (PLN), acts by decreasing the affinity of SERCA for Ca2+ . Changes in the SERCA2a:PLN ratio can cause Ca2+ dysregulation often seen in patients with dilated cardiomyopathy and heart failure. The enzyme glycogen synthase kinase-3 (GSK3) is known to downregulate SERCA function by decreasing the SERCA2a:PLN ratio. In this study, we sought to determine whether feeding mice low-dose lithium, a natural GSK3 inhibitor, would improve left ventricular SERCA function by altering the SERCA2a:PLN ratio. To this end, male wild-type C57BL/6J mice were fed low-dose lithium via drinking water (10 mg kg-1 day-1 LiCl for 6 weeks) and left ventricles were harvested. GSK3 activity was significantly reduced in LiCl-fed versus control-fed mice. The apparent affinity of SERCA for Ca2+ was also increased (pCa50 ; control, 6.09 ± 0.03 versus LiCl, 6.26 ± 0.04, P < 0.0001) along with a 2.0-fold increase in SERCA2a:PLN ratio in LiCl-fed versus control-fed mice. These findings suggest that low-dose lithium supplementation can improve SERCA function by increasing the SERCA2a:PLN ratio. Future studies in murine preclinical models will determine whether GSK3 inhibition via low-dose lithium could be a potential therapeutic strategy for dilated cardiomyopathy and heart failure.
Collapse
Affiliation(s)
- Sophie I Hamstra
- Department of Kinesiology, Brock University, St. Catharines, ON, L2S 3A1, Canada.,Centre for Bone and Muscle Health, Brock University, St. Catharines, ON, L2S 3A1, Canada
| | - Nigel Kurgan
- Department of Kinesiology, Brock University, St. Catharines, ON, L2S 3A1, Canada.,Centre for Bone and Muscle Health, Brock University, St. Catharines, ON, L2S 3A1, Canada
| | - Ryan W Baranowski
- Department of Kinesiology, Brock University, St. Catharines, ON, L2S 3A1, Canada.,Centre for Bone and Muscle Health, Brock University, St. Catharines, ON, L2S 3A1, Canada
| | - Liqun Qiu
- Department of Chemistry, Brock University, St. Catharines, ON, L2S 3A1, Canada
| | - Colton J F Watson
- Department of Health Sciences, Brock University, St. Catharines, ON, L2S 3A1, Canada
| | - Holt N Messner
- Department of Biological Sciences, Brock University, St. Catharines, ON, L2S 3A1, Canada
| | | | - Adam J MacNeil
- Department of Health Sciences, Brock University, St. Catharines, ON, L2S 3A1, Canada
| | - Brian D Roy
- Department of Kinesiology, Brock University, St. Catharines, ON, L2S 3A1, Canada.,Centre for Bone and Muscle Health, Brock University, St. Catharines, ON, L2S 3A1, Canada
| | - Val A Fajardo
- Department of Kinesiology, Brock University, St. Catharines, ON, L2S 3A1, Canada.,Centre for Bone and Muscle Health, Brock University, St. Catharines, ON, L2S 3A1, Canada
| |
Collapse
|
16
|
Law ML, Cohen H, Martin AA, Angulski ABB, Metzger JM. Dysregulation of Calcium Handling in Duchenne Muscular Dystrophy-Associated Dilated Cardiomyopathy: Mechanisms and Experimental Therapeutic Strategies. J Clin Med 2020; 9:jcm9020520. [PMID: 32075145 PMCID: PMC7074327 DOI: 10.3390/jcm9020520] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 02/06/2020] [Indexed: 02/07/2023] Open
Abstract
: Duchenne muscular dystrophy (DMD) is an X-linked recessive disease resulting in the loss of dystrophin, a key cytoskeletal protein in the dystrophin-glycoprotein complex. Dystrophin connects the extracellular matrix with the cytoskeleton and stabilizes the sarcolemma. Cardiomyopathy is prominent in adolescents and young adults with DMD, manifesting as dilated cardiomyopathy (DCM) in the later stages of disease. Sarcolemmal instability, leading to calcium mishandling and overload in the cardiac myocyte, is a key mechanistic contributor to muscle cell death, fibrosis, and diminished cardiac contractile function in DMD patients. Current therapies for DMD cardiomyopathy can slow disease progression, but they do not directly target aberrant calcium handling and calcium overload. Experimental therapeutic targets that address calcium mishandling and overload include membrane stabilization, inhibition of stretch-activated channels, ryanodine receptor stabilization, and augmentation of calcium cycling via modulation of the Serca2a/phospholamban (PLN) complex or cytosolic calcium buffering. This paper addresses what is known about the mechanistic basis of calcium mishandling in DCM, with a focus on DMD cardiomyopathy. Additionally, we discuss currently utilized therapies for DMD cardiomyopathy, and review experimental therapeutic strategies targeting the calcium handling defects in DCM and DMD cardiomyopathy.
Collapse
Affiliation(s)
- Michelle L. Law
- Department of Family and Consumer Sciences, Robbins College of Health and Human Sciences, Baylor University, Waco, TX 76706, USA;
| | - Houda Cohen
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN 55455, USA; (H.C.); (A.A.M.); (A.B.B.A.)
| | - Ashley A. Martin
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN 55455, USA; (H.C.); (A.A.M.); (A.B.B.A.)
| | - Addeli Bez Batti Angulski
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN 55455, USA; (H.C.); (A.A.M.); (A.B.B.A.)
| | - Joseph M. Metzger
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN 55455, USA; (H.C.); (A.A.M.); (A.B.B.A.)
- Correspondence: ; Tel.: +1-612-625-5902; Fax: +1-612-625-5149
| |
Collapse
|
17
|
da Silva Vasconcelos E, Kalinin AL, Cipriano RC, Dos Santos Beserra S, Lopes AG, da Costa Leite CA, Monteiro DA. Effects of feeding and digestion on myocardial contractility and expression of calcium-handling proteins in Burmese pythons (Python molurus). Comp Biochem Physiol B Biochem Mol Biol 2019; 240:110371. [PMID: 31676333 DOI: 10.1016/j.cbpb.2019.110371] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 10/07/2019] [Accepted: 10/14/2019] [Indexed: 12/20/2022]
Abstract
Pythons are important models of studies on postprandial metabolism because their physiological responses are exacerbated when digesting large prey. Prior studies of these animals have shown hypertrophy of the cardiac tissue 2 to 3 days after feeding, coinciding with the peak of the specific dynamic action (SDA), but the consequences of this remodeling in myocardial contractility have not been studied, which is the purpose of this work. Specimens of Python molurus were divided into two groups: a Digesting group (2 days after feeding, at the peak of SDA), and a Fasting group (28 days after feeding). When compared to the Fasting group, the Digesting group showed higher relative ventricular mass and calcium-handling protein expression such as sarcoplasmic reticulum Ca2+-ATPase (SERCA), phospholamban (PLB), and the Na+/Ca2+ exchanger (NCX). Digesting pythons also exhibited significant increases in the cardiac contraction force (Fc), rates of force development and relaxation, and cardiac pumping capacity. Therefore, the higher SERCA, PLB and NCX expression levels increased cytosolic Ca2+ transient amplitude, improving myofilament force. These changes are crucial to maintain cardiac output and a relatively high and continuous blood flow required by metabolic expenditure that occurs in postprandial animals.
Collapse
Affiliation(s)
- Eliton da Silva Vasconcelos
- Department of Physiological Sciences, Federal University of São Carlos (UFSCar), São Carlos, São Paulo, Brazil
| | - Ana Lúcia Kalinin
- Department of Physiological Sciences, Federal University of São Carlos (UFSCar), São Carlos, São Paulo, Brazil
| | - Rafael Correa Cipriano
- Department of Physiological Sciences, Federal University of São Carlos (UFSCar), São Carlos, São Paulo, Brazil
| | | | - André Guelli Lopes
- Department of Physiological Sciences, Federal University of São Carlos (UFSCar), São Carlos, São Paulo, Brazil
| | | | - Diana Amaral Monteiro
- Department of Physiological Sciences, Federal University of São Carlos (UFSCar), São Carlos, São Paulo, Brazil.
| |
Collapse
|
18
|
Affiliation(s)
- Evangelia G Kranias
- From the Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, OH (E.G.K.)
| | - Pieter A Doevendans
- Netherlands Heart Institute, Utrecht (P.A.D.).,UMC Utrecht Department of Cardiology, The Netherlands; Central Military Hospital Utrecht, The Netherlands (P.A.D.)
| | - Pieter C Glijnis
- Phospholamban Foundation, Wieringerwerf, The Netherlands (P.C.G.)
| | - Roger J Hajjar
- Department of Cardiology, Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York (R.J.H.)
| |
Collapse
|
19
|
Newly Discovered Micropeptide Regulators of SERCA Form Oligomers but Bind to the Pump as Monomers. J Mol Biol 2019; 431:4429-4443. [PMID: 31449798 DOI: 10.1016/j.jmb.2019.07.037] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 07/26/2019] [Accepted: 07/31/2019] [Indexed: 12/22/2022]
Abstract
The recently-discovered single-span transmembrane proteins endoregulin (ELN), dwarf open reading frame (DWORF), myoregulin (MLN), and another-regulin (ALN) are reported to bind to the SERCA calcium pump in a manner similar to that of known regulators of SERCA activity, phospholamban (PLB) and sarcolipin (SLN). To determine how micropeptide assembly into oligomers affects the availability of the micropeptide to bind to SERCA in a regulatory complex, we used co-immunoprecipitation and fluorescence resonance energy transfer (FRET) to quantify micropeptide oligomerization and SERCA-binding. Micropeptides formed avid homo-oligomers with high-order stoichiometry (n > 2 protomers per homo-oligomer), but it was the monomeric form of all micropeptides that interacted with SERCA. In view of these two alternative binding interactions, we evaluated the possibility that oligomerization occurs at the expense of SERCA-binding. However, even the most avidly oligomeric micropeptide species still showed robust FRET with SERCA, and there was a surprising positive correlation between oligomerization affinity and SERCA-binding. This comparison of micropeptide family members suggests that the same structural determinants that support oligomerization are also important for binding to SERCA. Moreover, the unique oligomerization/SERCA-binding profile of DWORF is in harmony with its distinct role as a PLB-competing SERCA activator, in contrast to the inhibitory function of the other SERCA-binding micropeptides.
Collapse
|
20
|
Gamu D, Juracic ES, Hall KJ, Tupling AR. The sarcoplasmic reticulum and SERCA: a nexus for muscular adaptive thermogenesis. Appl Physiol Nutr Metab 2019; 45:1-10. [PMID: 31116956 DOI: 10.1139/apnm-2019-0067] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
We are currently facing an "obesity epidemic" worldwide. Promoting inefficient metabolism in muscle represents a potential treatment for obesity and its complications. Sarco(endo)plasmic reticulum (SR) Ca2+-ATPase (SERCA) pumps in muscle are responsible for maintaining low cytosolic Ca2+ concentration through the ATP-dependent pumping of Ca2+ from the cytosol into the SR lumen. SERCA activity has the potential to be a critical regulator of body mass and adiposity given that it is estimated to contribute upwards of 20% of daily energy expenditure. More interestingly, this fraction can be modified physiologically in the face of stressors, such as ambient temperature and diet, through its physical interaction with several regulators known to inhibit Ca2+ uptake and muscle function. In this review, we discuss advances in our understanding of Ca2+-cycling thermogenesis within skeletal muscle, focusing on SERCA and its protein regulators, which were thought previously to only modulate muscular contractility. Novelty ATP consumption by SERCA pumps comprises a large proportion of resting energy expenditure in muscle and is dynamically regulated through interactions with small SERCA regulatory proteins. SERCA efficiency correlates significantly with resting metabolism, such that individuals with a higher resting metabolic rate have less energetically efficient SERCA Ca2+ pumping in muscle (i.e., lower coupling ratio). Futile Ca2+ cycling is a versatile heat generating mechanism utilized by both skeletal muscle and beige fat.
Collapse
Affiliation(s)
- Daniel Gamu
- Department of Kinesiology, University of Waterloo, Waterloo, ON N2L 3G1, Canada.,Department of Kinesiology, University of Waterloo, Waterloo, ON N2L 3G1, Canada
| | - Emma Sara Juracic
- Department of Kinesiology, University of Waterloo, Waterloo, ON N2L 3G1, Canada.,Department of Kinesiology, University of Waterloo, Waterloo, ON N2L 3G1, Canada
| | - Karlee J Hall
- Department of Kinesiology, University of Waterloo, Waterloo, ON N2L 3G1, Canada.,Department of Kinesiology, University of Waterloo, Waterloo, ON N2L 3G1, Canada
| | - A Russell Tupling
- Department of Kinesiology, University of Waterloo, Waterloo, ON N2L 3G1, Canada.,Department of Kinesiology, University of Waterloo, Waterloo, ON N2L 3G1, Canada
| |
Collapse
|
21
|
Niemeyer J, Mentrup T, Heidasch R, Müller SA, Biswas U, Meyer R, Papadopoulou AA, Dederer V, Haug-Kröper M, Adamski V, Lüllmann-Rauch R, Bergmann M, Mayerhofer A, Saftig P, Wennemuth G, Jessberger R, Fluhrer R, Lichtenthaler SF, Lemberg MK, Schröder B. The intramembrane protease SPPL2c promotes male germ cell development by cleaving phospholamban. EMBO Rep 2019; 20:e46449. [PMID: 30733280 PMCID: PMC6399600 DOI: 10.15252/embr.201846449] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 12/21/2018] [Accepted: 12/21/2018] [Indexed: 11/09/2022] Open
Abstract
Signal peptide peptidase (SPP) and the four homologous SPP-like (SPPL) proteases constitute a family of intramembrane aspartyl proteases with selectivity for type II-oriented transmembrane segments. Here, we analyse the physiological function of the orphan protease SPPL2c, previously considered to represent a non-expressed pseudogene. We demonstrate proteolytic activity of SPPL2c towards selected tail-anchored proteins. Despite shared ER localisation, SPPL2c and SPP exhibit distinct, though partially overlapping substrate spectra and inhibitory profiles, and are organised in different high molecular weight complexes. Interestingly, SPPL2c is specifically expressed in murine and human testis where it is primarily localised in spermatids. In mice, SPPL2c deficiency leads to a partial loss of elongated spermatids and reduced motility of mature spermatozoa, but preserved fertility. However, matings of male and female SPPL2c-/- mice exhibit reduced litter sizes. Using proteomics we identify the sarco/endoplasmic reticulum Ca2+-ATPase (SERCA2)-regulating protein phospholamban (PLN) as a physiological SPPL2c substrate. Accumulation of PLN correlates with a decrease in intracellular Ca2+ levels in elongated spermatids that likely contribute to the compromised male germ cell differentiation and function of SPPL2c-/- mice.
Collapse
Affiliation(s)
- Johannes Niemeyer
- Biochemical Institute, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Torben Mentrup
- Biochemical Institute, Christian-Albrechts-University of Kiel, Kiel, Germany
- Institute of Physiological Chemistry, Technische Universität Dresden, Dresden, Germany
| | - Ronny Heidasch
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), DKFZ-ZMBH Allianz, Heidelberg, Germany
| | - Stephan A Müller
- DZNE - German Center for Neurodegenerative Diseases, Munich, Germany
- Neuroproteomics, School of Medicine, Klinikum rechts der Isar and Institute for Advanced Study, Technical University of Munich, Munich, Germany
| | - Uddipta Biswas
- Institute of Physiological Chemistry, Technische Universität Dresden, Dresden, Germany
| | - Rieke Meyer
- Biochemical Institute, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Alkmini A Papadopoulou
- Institute for Metabolic Biochemistry, Biomedical Center (BMC) München, Ludwig Maximilians University of Munich, Munich, Germany
| | - Verena Dederer
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), DKFZ-ZMBH Allianz, Heidelberg, Germany
| | - Martina Haug-Kröper
- Institute for Metabolic Biochemistry, Biomedical Center (BMC) München, Ludwig Maximilians University of Munich, Munich, Germany
| | - Vivian Adamski
- Biochemical Institute, Christian-Albrechts-University of Kiel, Kiel, Germany
| | | | - Martin Bergmann
- Institute of Veterinary Anatomy, Justus Liebig University of Gießen, Gießen, Germany
| | - Artur Mayerhofer
- Cell Biology, Anatomy III, Biomedical Center (BMC) München, Ludwig Maximilians University of Munich, Munich, Germany
| | - Paul Saftig
- Biochemical Institute, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Gunther Wennemuth
- Institute of Anatomy, University Hospital, Duisburg-Essen University, Essen, Germany
| | - Rolf Jessberger
- Institute of Physiological Chemistry, Technische Universität Dresden, Dresden, Germany
| | - Regina Fluhrer
- DZNE - German Center for Neurodegenerative Diseases, Munich, Germany
- Institute for Metabolic Biochemistry, Biomedical Center (BMC) München, Ludwig Maximilians University of Munich, Munich, Germany
| | - Stefan F Lichtenthaler
- DZNE - German Center for Neurodegenerative Diseases, Munich, Germany
- Neuroproteomics, School of Medicine, Klinikum rechts der Isar and Institute for Advanced Study, Technical University of Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Marius K Lemberg
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), DKFZ-ZMBH Allianz, Heidelberg, Germany
| | - Bernd Schröder
- Biochemical Institute, Christian-Albrechts-University of Kiel, Kiel, Germany
- Institute of Physiological Chemistry, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
22
|
Affiliation(s)
- Evangelia G Kranias
- From the Department of Pharmacology and Cell Biophysics, University of Cincinnati College of Medicine, OH (E.G.K.); and Cardiovascular Research Center, Department of Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY (R.J.H.).
| | - Roger J Hajjar
- From the Department of Pharmacology and Cell Biophysics, University of Cincinnati College of Medicine, OH (E.G.K.); and Cardiovascular Research Center, Department of Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY (R.J.H.)
| |
Collapse
|
23
|
Groban L, Tran QK, Ferrario CM, Sun X, Cheng CP, Kitzman DW, Wang H, Lindsey SH. Female Heart Health: Is GPER the Missing Link? Front Endocrinol (Lausanne) 2019; 10:919. [PMID: 31993020 PMCID: PMC6970950 DOI: 10.3389/fendo.2019.00919] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Accepted: 12/17/2019] [Indexed: 12/20/2022] Open
Abstract
The G Protein-Coupled Estrogen Receptor (GPER) is a novel membrane-bound receptor that mediates non-genomic actions of the primary female sex hormone 17β-estradiol. Studies over the past two decades have elucidated the beneficial actions of this receptor in a number of cardiometabolic diseases. This review will focus specifically on the cardiac actions of GPER, since this receptor is expressed in cardiomyocytes as well as other cells within the heart and most likely contributes to estrogen-induced cardioprotection. Studies outlining the impact of GPER on diastolic function, mitochondrial function, left ventricular stiffness, calcium dynamics, cardiac inflammation, and aortic distensibility are discussed. In addition, recent data using genetic mouse models with global or cardiomyocyte-specific GPER gene deletion are highlighted. Since estrogen loss due to menopause in combination with chronological aging contributes to unique aspects of cardiac dysfunction in women, this receptor may provide novel therapeutic effects. While clinical studies are still required to fully understand the potential for pharmacological targeting of this receptor in postmenopausal women, this review will summarize the evidence gathered thus far on its likely beneficial effects.
Collapse
Affiliation(s)
- Leanne Groban
- Department of Anesthesiology, Wake Forest School of Medicine, Winston Salem, NC, United States
- Department of Internal Medicine-Molecular Medicine, Wake Forest School of Medicine, Winston Salem, NC, United States
- *Correspondence: Leanne Groban
| | - Quang-Kim Tran
- Department of Physiology & Pharmacology, Des Moines University College of Osteopathic Medicine, Des Moines, IA, United States
| | - Carlos M. Ferrario
- Department of Surgery, Wake Forest School of Medicine, Winston Salem, NC, United States
- Department of Physiology-Pharmacology, Wake Forest School of Medicine, Winston Salem, NC, United States
| | - Xuming Sun
- Department of Anesthesiology, Wake Forest School of Medicine, Winston Salem, NC, United States
| | - Che Ping Cheng
- Department of Internal Medicine, Cardiovascular Medicine Section, Wake Forest School of Medicine, Winston Salem, NC, United States
| | - Dalane W. Kitzman
- Department of Internal Medicine, Cardiovascular Medicine Section, Wake Forest School of Medicine, Winston Salem, NC, United States
| | - Hao Wang
- Department of Anesthesiology, Wake Forest School of Medicine, Winston Salem, NC, United States
- Department of Internal Medicine-Molecular Medicine, Wake Forest School of Medicine, Winston Salem, NC, United States
| | - Sarah H. Lindsey
- Department of Pharmacology, Tulane University, New Orleans, LA, United States
| |
Collapse
|
24
|
Makarewich CA, Munir AZ, Schiattarella GG, Bezprozvannaya S, Raguimova ON, Cho EE, Vidal AH, Robia SL, Bassel-Duby R, Olson EN. The DWORF micropeptide enhances contractility and prevents heart failure in a mouse model of dilated cardiomyopathy. eLife 2018; 7:e38319. [PMID: 30299255 PMCID: PMC6202051 DOI: 10.7554/elife.38319] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2018] [Accepted: 09/26/2018] [Indexed: 01/01/2023] Open
Abstract
Calcium (Ca2+) dysregulation is a hallmark of heart failure and is characterized by impaired Ca2+ sequestration into the sarcoplasmic reticulum (SR) by the SR-Ca2+-ATPase (SERCA). We recently discovered a micropeptide named DWORF (DWarf Open Reading Frame) that enhances SERCA activity by displacing phospholamban (PLN), a potent SERCA inhibitor. Here we show that DWORF has a higher apparent binding affinity for SERCA than PLN and that DWORF overexpression mitigates the contractile dysfunction associated with PLN overexpression, substantiating its role as a potent activator of SERCA. Additionally, using a well-characterized mouse model of dilated cardiomyopathy (DCM) due to genetic deletion of the muscle-specific LIM domain protein (MLP), we show that DWORF overexpression restores cardiac function and prevents the pathological remodeling and Ca2+ dysregulation classically exhibited by MLP knockout mice. Our results establish DWORF as a potent activator of SERCA within the heart and as an attractive candidate for a heart failure therapeutic.
Collapse
Affiliation(s)
- Catherine A Makarewich
- Department of Molecular Biology and Hamon Center for Regenerative Science and MedicineUniversity of Texas Southwestern Medical CenterDallasUnited States
| | - Amir Z Munir
- Department of Molecular Biology and Hamon Center for Regenerative Science and MedicineUniversity of Texas Southwestern Medical CenterDallasUnited States
| | - Gabriele G Schiattarella
- Department of Internal MedicineUniversity of Texas Southwestern Medical CenterDallasUnited States
| | - Svetlana Bezprozvannaya
- Department of Molecular Biology and Hamon Center for Regenerative Science and MedicineUniversity of Texas Southwestern Medical CenterDallasUnited States
| | - Olga N Raguimova
- Department of Cell and Molecular PhysiologyLoyola University ChicagoMaywoodUnited States
| | - Ellen E Cho
- Department of Cell and Molecular PhysiologyLoyola University ChicagoMaywoodUnited States
| | - Alexander H Vidal
- Department of Molecular Biology and Hamon Center for Regenerative Science and MedicineUniversity of Texas Southwestern Medical CenterDallasUnited States
| | - Seth L Robia
- Department of Cell and Molecular PhysiologyLoyola University ChicagoMaywoodUnited States
| | - Rhonda Bassel-Duby
- Department of Molecular Biology and Hamon Center for Regenerative Science and MedicineUniversity of Texas Southwestern Medical CenterDallasUnited States
| | - Eric N Olson
- Department of Molecular Biology and Hamon Center for Regenerative Science and MedicineUniversity of Texas Southwestern Medical CenterDallasUnited States
| |
Collapse
|
25
|
Chen M, Xu D, Wu AZ, Kranias E, Lin SF, Chen PS, Chen Z. Phospholamban regulates nuclear Ca 2+ stores and inositol 1,4,5-trisphosphate mediated nuclear Ca 2+ cycling in cardiomyocytes. J Mol Cell Cardiol 2018; 123:185-197. [PMID: 30261161 DOI: 10.1016/j.yjmcc.2018.09.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 09/04/2018] [Accepted: 09/21/2018] [Indexed: 01/15/2023]
Abstract
AIMS Phospholamban (PLB) is the key regulator of the cardiac Ca2+ pump (SERCA2a)-mediated sarcoplasmic reticulum Ca2+ stores. We recently reported that PLB is highly concentrated in the nuclear envelope (NE) from where it can modulate perinuclear Ca2+ handling of the cardiomyocytes (CMs). Since inositol 1,4,5-trisphosphate (IP3) receptor (IP3R) mediates nuclear Ca2+ release, we examined whether the nuclear pool of PLB regulates IP3-induced nuclear Ca2+ handling. METHODS AND RESULTS Fluo-4 based confocal Ca2+ imaging was performed to measure Ca2+ dynamics across both nucleus and cytosol in saponin-permeabilized CMs isolated from wild-type (WT) or PLB-knockout (PLB-KO) mice. At diastolic intracellular Ca2+ ([Ca2+]i = 100 nM), the Fab fragment of the monoclonal PLB antibody (anti-PLB Fab) facilitated the formation and increased the length of spontaneous Ca2+ waves (SCWs) originating from the nuclear region in CMs from WT but not from PLB-KO mice. We next examined nuclear Ca2+ activities at basal condition and after sequential addition of IP3, anti-PLB Fab, and the IP3R inhibitor 2-aminoethoxydiphenyl borate (2-APB) at a series of [Ca2+]i. In WT mice, at 10 nM [Ca2+]i where ryanodine receptor (RyR2) based spontaneous Ca2+ sparks rarely occurred, IP3 increased fluorescence amplitude (F/F0) of overall nuclear region to 1.19 ± 0.02. Subsequent addition of anti-PLB Fab significantly decreased F/F0 to 1.09 ± 0.02. At 50 nM [Ca2+]i, anti-PLB Fab not only decreased the overall nuclear F/F0 previously elevated by IP3, but also increased the amplitude and duration of spark-like nuclear Ca2+ release events. These nuclear Ca2+ releases were blocked by 2-APB. At 100 nM [Ca2+]i, IP3 induced short SCWs originating from nucleus. Anti-PLB Fab transformed those short waves into long SCWs with propagation from the nucleus into the cytosol. In contrast, neither nuclear nor cytosolic Ca2+ dynamics was affected by anti-PLB Fab in CMs from PLB-KO mice in all these conditions. Furthermore, in WT CMs pretreated with RyR2 blocker tetracaine, IP3 and anti-PLB Fab still increased the magnitude of nuclear Ca2+ release but failed to regenerate SCWs. Finally, anti-PLB Fab increased low Ca2+ affinity mag-fluo 4 fluorescence intensity in the lumen of NE of nuclei isolated from WT but not in PLB-KO mice. CONCLUSION PLB regulates nuclear Ca2+ handling. By increasing Ca2+ uptake into lumen of the NE and perhaps other perinuclear membranes, the acute reversal of PLB inhibition decreases global Ca2+ concentration at rest in the nucleoplasm, and increases Ca2+ release into the nucleus, through mechanisms involving IP3R and RyR2 in the vicinity.
Collapse
Affiliation(s)
- Mu Chen
- Krannert Institute of Cardiology, Indiana University, Indianapolis, IN, USA; Department of Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Dongzhu Xu
- Krannert Institute of Cardiology, Indiana University, Indianapolis, IN, USA; Cardiovascular Division, Institute of Clinical Medicine, Faculty of Medicine, University of Tsukuba, Japan
| | - Adonis Z Wu
- Krannert Institute of Cardiology, Indiana University, Indianapolis, IN, USA
| | - Evangelia Kranias
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Shien-Fong Lin
- Krannert Institute of Cardiology, Indiana University, Indianapolis, IN, USA; Institute of Biomedical Engineering, College of Electrical and Computer Engineering, National Chiao Tung University, Hsin-Chu, Taiwan
| | - Peng-Sheng Chen
- Krannert Institute of Cardiology, Indiana University, Indianapolis, IN, USA
| | - Zhenhui Chen
- Krannert Institute of Cardiology, Indiana University, Indianapolis, IN, USA.
| |
Collapse
|
26
|
Hammoudi N, Jeong D, Singh R, Farhat A, Komajda M, Mayoux E, Hajjar R, Lebeche D. Empagliflozin Improves Left Ventricular Diastolic Dysfunction in a Genetic Model of Type 2 Diabetes. Cardiovasc Drugs Ther 2018. [PMID: 28643218 DOI: 10.1007/s10557-017-6734-1] [Citation(s) in RCA: 108] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
PURPOSE Cardiovascular (CV) diseases in type 2 diabetes (T2DM) represent an enormous burden with high mortality and morbidity. Sodium-glucose cotransporter 2 (SGLT2) inhibitors have recently emerged as a new antidiabetic class that improves glucose control, as well as body weight and blood pressure with no increased risk of hypoglycemia. The first CV outcome study terminated with empagliflozin, a specific SGLT2 inhibitor, has shown a reduction in CV mortality and in heart failure hospitalization, suggesting a beneficial impact on cardiac function which remains to be demonstrated. This study was designed to examine the chronic effect of empagliflozin on left ventricular (LV) systolic and diastolic functions in a genetic model of T2DM, ob/ob mice. METHODS AND RESULTS Cardiac phenotype was characterized by echocardiography, in vivo hemodynamics, histology, and molecular profiling. Our results demonstrate that empagliflozin significantly lowered HbA1c and slightly reduced body weight compared to vehicle treatment with no obvious changes in insulin levels. Empagliflozin also improved LV maximum pressure and in vivo indices of diastolic function. While systolic function was grossly not affected in both groups at steady state, response to dobutamine stimulation was significantly improved in the empagliflozin-treated group, suggesting amelioration of contractile reserve. This was paralleled by an increase in phospholamban (PLN) phosphorylation and increased SERCA2a/PLN ratio, indicative of enhanced SERCA2a function, further supporting improved cardiac relaxation and diastolic function. In addition, empagliflozin reconciled diabetes-associated increase in MAPKs and dysregulated phosphorylation of IRS1 and Akt, leading to improvement in myocardial insulin sensitivity and glucose utilization. CONCLUSION The data show that chronic treatment with empagliflozin improves diastolic function, preserves calcium handling and growth signaling pathways and attenuates myocardial insulin resistance in ob/ob mice, findings suggestive of a potential clinical utility for empagliflozin in the treatment of diastolic dysfunction.
Collapse
Affiliation(s)
- Nadjib Hammoudi
- Cardiovascular Research Institute, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.,Sorbonne Universités, UPMC University Paris 06, Institut de Cardiologie (AP-HP), Centre Hospitalier Universitaire Pitié-Salpêtrière, Institute of Cardiometabolism and Nutrition (ICAN), INSERM UMRS 1166, Paris, France
| | - Dongtak Jeong
- Cardiovascular Research Institute, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Rajvir Singh
- Cardiovascular Research Institute, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Ahmed Farhat
- Cardiovascular Research Institute, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.,Graduate School of Biological Sciences, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Michel Komajda
- Sorbonne Universités, UPMC University Paris 06, Institut de Cardiologie (AP-HP), Centre Hospitalier Universitaire Pitié-Salpêtrière, Institute of Cardiometabolism and Nutrition (ICAN), INSERM UMRS 1166, Paris, France
| | - Eric Mayoux
- Boehringer Ingelheim Pharma GmbH & Co. KG, Cardio-metabolic Diseases, Binger Straße 173, 55216, Ingelheim am Rhein, Germany
| | - Roger Hajjar
- Cardiovascular Research Institute, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Djamel Lebeche
- Cardiovascular Research Institute, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA. .,Graduate School of Biological Sciences, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA. .,Diabetes, Obesity and Metabolism Institute, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
27
|
Berger RCM, Benetti A, Girardi ACC, Forechi L, de Oliveira RM, Vassallo PF, Mill JG. Influence of Long-Term Salt Diets on Cardiac Ca2+ Handling and Contractility Proteins in Hypertensive Rats. Am J Hypertens 2018. [PMID: 29518186 DOI: 10.1093/ajh/hpy023] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND High sodium intake contributes to the pathogenesis of hypertension and adversely affects cardiac function. Conversely, sodium reduction is associated with a blood pressure decrease and improved cardiovascular function. However, the mechanisms that underlie the cardiac effects induced by salt intake in hypertension have not been fully elucidated. Ca2+ handling is critical for efficient myocardial function; thus, we aimed to investigate the long-term effects of diets with different salt contents on cardiac function and Ca2+ handling proteins in spontaneously hypertensive rats (SHRs). METHODS Cardiac function was evaluated by catheterization. Ca2+ handling and contractile proteins were evaluated by immunoblotting in hearts from SHRs fed for 6 months with diets containing high (HS, 3%), low (LS, 0.03%), or normal salt content (NS, 0.3%). Diets were introduced immediately after weaning. Tail cuff pletismography was assessed at the 3rd and 7th months of follow-up. RESULTS Compared to the NS group, the HS group exhibited worsened hypertension, increased cardiac expression of β-myosin heavy chain (MHC), a decreased α/β-MHC ratio and reduced expression of both phospholamban (PLB) and Na+/Ca2+ exchanger (NCX). LS intake attenuated the blood pressure increase and left ventricle hypertrophy, slightly decreased the cardiac contractility and relaxation index, and increased the α/β-MHC ratio. These effects were accompanied by increased cardiac PLB expression and decreased Ca2+ L-type channel and NCX expression. CONCLUSIONS These findings indicate that the modulation of Ca2+ handling may be one of the molecular mechanisms underlying the effect of salt intake on myocardial function in hypertension.
Collapse
Affiliation(s)
| | - Acaris Benetti
- Heart Institute (InCor), University of São Paulo Medical School, São Paulo, São Paulo, Brazil
| | | | - Ludimila Forechi
- Department of Physiological Sciences, Federal University of Espirito Santo, Vitória, Espirito Santo, Brazil
| | | | - Paula Frizera Vassallo
- Department of Physiological Sciences, Federal University of Espirito Santo, Vitória, Espirito Santo, Brazil
| | - José Geraldo Mill
- Department of Physiological Sciences, Federal University of Espirito Santo, Vitória, Espirito Santo, Brazil
| |
Collapse
|
28
|
|
29
|
Ribeiro Júnior RF, Ronconi KS, Jesus ICG, Almeida PWM, Forechi L, Vassallo DV, Guatimosim S, Stefanon I, Fernandes AA. Testosterone deficiency prevents left ventricular contractility dysfunction after myocardial infarction. Mol Cell Endocrinol 2018; 460:14-23. [PMID: 28606867 DOI: 10.1016/j.mce.2017.06.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Revised: 05/17/2017] [Accepted: 06/08/2017] [Indexed: 11/30/2022]
Abstract
Testosterone may affect myocardial contractility since its deficiency decreases the contraction and relaxation of the heart. Meanwhile, testosterone replacement therapy has raised concerns because it may worsen cardiac dysfunction and remodeling after myocardial infarction (MI). In this study, we evaluate cardiac contractility 60 days after MI in rats with suppressed testosterone. Male Wistar rats underwent bilateral orchidectomy one week before the ligation of the anterior descending left coronary artery. The animals were divided into orchidectomized (OCT); MI; orchidectomized + MI (OCT + MI); orchidectomized + MI + testosterone (OCT + MI + T) and control (Sham) groups. Eight weeks after MI, papillary muscle contractility was analyzed under increasing calcium (0.62, 1.25, 2.5 and 3.75 mM) and isoproterenol (10-8 to 10-2 M) concentrations. Ventricular myocytes were isolated for intracellular calcium measurements and assessment of Ca2+ handling proteins. Contractility was preserved in the orchidectomized animals after myocardial infarction and was reduced when testosterone was replaced (Ca2+ 3.75 mM: Sham: 608 ± 70 (n = 11); OCT: 590 ± 37 (n = 16); MI: 311 ± 33* (n = 9); OCT + MI: 594 ± 76 (n = 7); OCT + MI + T: 433 ± 38* (n=4), g/g *p < 0.05 vs Sham). Orchidectomy also increased the Ca2+ transient amplitude of the ventricular myocytes and SERCA-2a protein expression levels. PLB phosphorylation levels at Thr17 were not different in the orchidectomized animals compared to the Sham animals but were reduced after testosterone replacement. CAMKII phosphorylation and protein nitrosylation increased in the orchidectomized animals. Our results support the view that testosterone deficiency prevents MI contractility dysfunction by altering the key proteins involved in Ca2+ handling.
Collapse
Affiliation(s)
- R F Ribeiro Júnior
- Department of Physiological Sciences, Universidade Federal do Espírito Santo, Vitória, ES, Brazil.
| | - K S Ronconi
- Department of Physiological Sciences, Universidade Federal do Espírito Santo, Vitória, ES, Brazil
| | - I C G Jesus
- Department of Physiology and Biophysics, Universidade Federal de Minas Gerais, Minas Gerais, MG, Brazil
| | - P W M Almeida
- Department of Physiology and Biophysics, Universidade Federal de Minas Gerais, Minas Gerais, MG, Brazil
| | - L Forechi
- Department of Physiological Sciences, Universidade Federal do Espírito Santo, Vitória, ES, Brazil
| | - D V Vassallo
- Department of Physiological Sciences, Universidade Federal do Espírito Santo, Vitória, ES, Brazil
| | - S Guatimosim
- Department of Physiology and Biophysics, Universidade Federal de Minas Gerais, Minas Gerais, MG, Brazil
| | - I Stefanon
- Department of Physiological Sciences, Universidade Federal do Espírito Santo, Vitória, ES, Brazil
| | - A A Fernandes
- Department of Physiological Sciences, Universidade Federal do Espírito Santo, Vitória, ES, Brazil
| |
Collapse
|
30
|
Kraev A. Insertional Mutagenesis Confounds the Mechanism of the Morbid Phenotype of a PLN R9C Transgenic Mouse Line. J Card Fail 2018; 24:115-125. [PMID: 29325795 DOI: 10.1016/j.cardfail.2017.12.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 12/18/2017] [Accepted: 12/21/2017] [Indexed: 12/29/2022]
Abstract
BACKGROUND A mouse line with heterozygous transgenic expression of phospholamban carrying a substitution of cysteine for arginine 9 (TgPLNR9C) under the control of α-myosin heavy chain (αMHC) promoter features dilated cardiomyopathy, heart failure, and premature death. METHODS AND RESULTS Determination of transgene chromosomal localization by conventional methods shows that in this line the transgenic array of 13 PLNR9C expression cassettes, arranged in a head-to-tail tandem orientation, have integrated into the bidirectional promoter of the αMHC (Myh6) gene and the gene for the regulatory noncoding RNA Myheart (Mhrt), both of which are known to be involved in cardiac development and pathology. Expression of the noncoding RNA Mhrt in TgPLNR9C mice exhibits profound deregulation, despite the presence of the second, intact allele. CONCLUSIONS The TgPLNR9C mouse strain is, in the best case, a functionally ambiguous phenocopy of the human PLNR9C heterozygote, because a similar constellation of genetically programmed events can not occur in a patient. Publications featuring "cardiac-specific overexpression" are focused on the phenotype and typically forgo the definition of the transgene integration site or transgene temporal expression profile, so caution should be exercised in attributing clinical relevance to pathologic phenomena observed in αMHC-driven transgenes.
Collapse
Affiliation(s)
- Alexander Kraev
- University of Toronto, 27 King's College Circle, Toronto, Ontario M5S 1A1, Canada.
| |
Collapse
|
31
|
Bidwell PA, Haghighi K, Kranias EG. The antiapoptotic protein HAX-1 mediates half of phospholamban's inhibitory activity on calcium cycling and contractility in the heart. J Biol Chem 2017; 293:359-367. [PMID: 29150445 DOI: 10.1074/jbc.ra117.000128] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 11/17/2017] [Indexed: 12/25/2022] Open
Abstract
The antiapoptotic protein HAX-1 (HS-associated protein X-1) localizes to sarcoplasmic reticulum (SR) in the heart and interacts with the small membrane protein phospholamban (PLN), inhibiting the cardiac sarco/endoplasmic reticulum calcium ATPase 2a (SERCA2a) in the regulation of overall calcium handling and heart muscle contractility. However, because global HAX-1 deletion causes early lethality, how much endogenous HAX-1 contributes to PLN's inhibitory activity on calcium cycling is unknown. We therefore generated a cardiac-specific and inducible knock-out mouse model. HAX-1 ablation in the adult heart significantly increased contractile parameters and calcium kinetics, associated with increased SR calcium load. These changes occurred without any changes in the protein expression of SERCA2a, PLN, and ryanodine receptor or in the PLN phosphorylation status. The enhanced calcium cycling in the HAX-1-depleted heart was mediated through increases in the calcium affinity of SERCA2a and reduced PLN-SERCA2a binding. Comparison of the HAX-1 deletion-induced stimulatory effects with those elicited by PLN ablation indicated that HAX-1 mediates ∼50% of the PLN-associated inhibitory effects in the heart. Stimulation with the inotropic and lusitropic agent isoproterenol eliminated the differences among wild-type, HAX-1-deficient, and PLN-deficient hearts, and maximally stimulated contractile and calcium kinetic parameters were similar among these three groups. Furthermore, PLN overexpression in the HAX-1-null cardiomyocytes did not elicit any inhibitory effects, indicating that HAX-1 may limit PLN activity. These findings suggest that HAX-1 is a major mediator of PLN's inhibitory activity and a critical gatekeeper of SR calcium cycling and contractility in the heart.
Collapse
Affiliation(s)
- Philip A Bidwell
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio 45267
| | - Kobra Haghighi
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio 45267
| | - Evangelia G Kranias
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio 45267; Department of Molecular Biology, Center of Basic Research, Biomedical Research Foundation, Academy of Athens, 115 27 Athens, Greece.
| |
Collapse
|
32
|
Rissoli RZ, Vasconcelos EDS, Rantin FT, Kalinin AL. Effects of exercise training on excitation-contraction coupling, calcium dynamics and protein expression in the heart of the Neotropical fish Brycon amazonicus. Comp Biochem Physiol A Mol Integr Physiol 2017; 214:85-93. [PMID: 28966144 DOI: 10.1016/j.cbpa.2017.09.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 09/21/2017] [Accepted: 09/21/2017] [Indexed: 10/18/2022]
Abstract
Matrinxã (Brycon amazonicus) is a great swimming performance teleost fish from the Amazon basin. However, the possible cardiac adaptations of this ability are still unknown. Therefore, the aim of the present work was to investigate the effects of prolonged exercise (EX group - 60days under 0.4BL·s-1) on ventricular contractility by (i) in-vitro analysis of contractility comparing the relative roles of sodium/calcium exchanger (NCX) and sarcoplasmic reticulum (SR) in the excitation-contraction (E-C) coupling and (ii) molecular analysis of NCX, sarcoplasmic reticulum Ca2+ ATPase (SERCA2) and phospholamban (PLB) expression and quantification. The exercise training significantly improved twitch tension, cardiac pumping capacity and the contraction rate when compared to controls (CT). Inhibition of the NCX function, replacing Na+ by Li+ in the physiological solutions, diminished cardiac contractility in the EX group, reduced all analyzed parameters under both high and low stimulation frequencies. The SR blockage, using 10μM ryanodine, caused ~50% tension reduction in CT at most analyzed frequencies while in EX, reductions (34-54%) were only found at higher frequencies. SR inhibition also decreased contraction and relaxation rates in both groups. Additionally, higher post-rest contraction values were recorded for EX, indicating an increase in SR Ca2+ loading. Higher NCX and PLB expression rates and lower SERCA2 rates were found in EX. Our data indicate that matrinxã presents a modulation in E-C coupling after exercise-training, enhancing the SR function under higher frequencies. This was the first study to functionally analyze the effects of swimming-induced exercise on fish cardiac E-C coupling.
Collapse
Affiliation(s)
- Rafael Zanelli Rissoli
- Department of Physiological Sciences, Federal University of São Carlos, São Carlos, SP, Brazil
| | | | - Francisco Tadeu Rantin
- Department of Physiological Sciences, Federal University of São Carlos, São Carlos, SP, Brazil
| | - Ana Lúcia Kalinin
- Department of Physiological Sciences, Federal University of São Carlos, São Carlos, SP, Brazil.
| |
Collapse
|
33
|
Rajtik T, Goncalvesova E, Varga ZV, Leszek P, Kusmierczyk M, Hulman M, Kyselovic J, Ferdinandy P, Adameova A. Posttranslational modifications of calcium/calmodulin-dependent protein kinase IIδ and its downstream signaling in human failing hearts. Am J Transl Res 2017; 9:3573-3585. [PMID: 28861149 PMCID: PMC5575172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 07/16/2017] [Indexed: 06/07/2023]
Abstract
BACKGROUND In human failing hearts (HF) of different origin (coronary artery disease-CAD, dilated-DCM, restrictive and hypertrophic cardiomyopathy-OTHER), we investigated the active forms of Ca2+/calmodulin-dependent protein kinase IIδ (p-Thr287-CaMKIIδ, oxMet281/282-CaMKIIδ) and their role in phenotypes of the disease. METHODS AND RESULTS Although basic diagnostic and clinical markers indicating the attenuated cardiac contractility and remodeling were comparable in HF groups, CaMKIIδ-mediated axis was different. P-Thr287-CaMKIIδ was unaltered in CAD group, whereas it was upregulated in non-ischemic cardiomyopathic groups. No correlation between the upregulated p-Thr287-CaMKIIδ and QT interval prolongation was detected. Unlike in DCM, oxMet281/282-CaMKIIδ did not differ among HF groups. Independently of CaMKIIδ phosphorylation/oxidation, activation of its downstreams-phospholamban and cardiac myosin binding protein-C was significantly downregulated supporting both diminished cardiac lusitropy and inotropy in all hearts. Content of sarcoplasmic reticulum Ca2+-ATPase 2a in all HF was unchanged. Protein phosphatase1β was upregulated in CAD and DCM only, while 2A did not differ among groups. CONCLUSION This is the first demonstration that the posttranslational activation of CaMKIIδ differs in HF depending on etiology. Lower levels of downstream molecular targets of CaMKIIδ do not correlate with either activation of CaMKIIδ or the expression of major protein phosphatases in the HF. Thus, it is unlikely that these mechanisms exclusively underlie failing of the heart.
Collapse
Affiliation(s)
- Tomas Rajtik
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Comenius UniversityBratislava, Slovak Republic
| | - Eva Goncalvesova
- Department of Heart Failure & Transplantation, The National Institute of Cardiovascular DiseasesBratislava, Slovak Republic
| | - Zoltan V Varga
- Department of Pharmacology & Pharmacotherapy, Semmelweis UniversityBudapest, Hungary
| | | | | | - Michal Hulman
- Clinic of Heart Surgery, The National Institute of Cardiovascular DiseasesBratislava, Slovak Republic
| | - Jan Kyselovic
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Comenius UniversityBratislava, Slovak Republic
| | - Peter Ferdinandy
- Department of Pharmacology & Pharmacotherapy, Semmelweis UniversityBudapest, Hungary
| | - Adriana Adameova
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Comenius UniversityBratislava, Slovak Republic
| |
Collapse
|
34
|
Savi M, Bocchi L, Mena P, Dall'Asta M, Crozier A, Brighenti F, Stilli D, Del Rio D. In vivo administration of urolithin A and B prevents the occurrence of cardiac dysfunction in streptozotocin-induced diabetic rats. Cardiovasc Diabetol 2017; 16:80. [PMID: 28683791 PMCID: PMC5501434 DOI: 10.1186/s12933-017-0561-3] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 06/15/2017] [Indexed: 12/13/2022] Open
Abstract
Background Emerging evidence suggests that specific (poly)phenols may constitute new preventative strategies to counteract cell oxidative stress and myocardial tissue inflammation, which have a key role in the patho-physiology of diabetic cardiomyopathy. In a rat model of early diabetes, we evaluated whether in vivo administration of urolithin A (UA) or urolithin B (UB), the main gut microbiota phenolic metabolites of ellagitannin-rich foods, can reduce diabetes-induced microenvironmental changes in myocardial tissue, preventing cardiac functional impairment. Methods Adult Wistar rats with streptozotocin-induced type-1 diabetes (n = 29) were studied in comparison with 10 control animals. Diabetic rats were either untreated (n = 9) or subjected to daily i.p. injection of UA (n = 10) or UB (n = 10). After 3 weeks of hyperglycaemia, hemodynamics, cardiomyocyte contractile properties and calcium transients were measured to assess cardiac performance. The myocardial expression of the pro-inflammatory cytokine fractalkine and proteins involved in calcium dynamics (sarcoplasmic reticulum calcium ATPase, phospholamban and phosphorylated phospholamban) were evaluated by immunoblotting. Plasma, urine and tissue distribution of UA, UB and their phase II metabolites were determined. Results In vivo urolithin treatment reduced by approximately 30% the myocardial expression of the pro-inflammatory cytokine fractalkine, preventing the early inflammatory response of cardiac cells to hyperglycaemia. The improvement in myocardial microenvironment had a functional counterpart, as documented by the increase in the maximal rate of ventricular pressure rise compared to diabetic group (+18% and +31% in UA and UB treated rats, respectively), and the parallel reduction in the isovolumic contraction time (−12%). In line with hemodynamic data, both urolithins induced a recovery of cardiomyocyte contractility and calcium dynamics, leading to a higher re-lengthening rate (+21%, on average), lower re-lengthening times (−56%), and a more efficient cytosolic calcium clearing (−32% in tau values). UB treatment also increased the velocity of shortening (+27%). Urolithin metabolites accumulated in the myocardium, with a higher concentration of UB and UB-sulphate, potentially explaining the slightly higher efficacy of UB administration. Conclusions In vivo urolithin administration may be able to prevent the initial inflammatory response of myocardial tissue to hyperglycaemia and the negative impact of the altered diabetic milieu on cardiac performance.
Collapse
Affiliation(s)
- Monia Savi
- Department of Food and Drugs, University of Parma, Parco Area delle Scienze 27/A, 43124, Parma, Italy.,Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parco Area delle Scienze 11/A, 43124, Parma, Italy
| | - Leonardo Bocchi
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parco Area delle Scienze 11/A, 43124, Parma, Italy
| | - Pedro Mena
- Department of Food and Drugs, University of Parma, Parco Area delle Scienze 27/A, 43124, Parma, Italy
| | - Margherita Dall'Asta
- Department of Food and Drugs, University of Parma, Parco Area delle Scienze 27/A, 43124, Parma, Italy
| | - Alan Crozier
- Department of Nutrition, University of California, 3143 Meyer Hall One Shields Avenue, Davis, CA, 95616-5270, USA
| | - Furio Brighenti
- Department of Food and Drugs, University of Parma, Parco Area delle Scienze 27/A, 43124, Parma, Italy
| | - Donatella Stilli
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parco Area delle Scienze 11/A, 43124, Parma, Italy.
| | - Daniele Del Rio
- Department of Food and Drugs, University of Parma, Parco Area delle Scienze 27/A, 43124, Parma, Italy.
| |
Collapse
|
35
|
Grinshpon M, Bondarenko VE. Simulation of the effects of moderate stimulation/inhibition of the β1-adrenergic signaling system and its components in mouse ventricular myocytes. Am J Physiol Cell Physiol 2016; 310:C844-56. [DOI: 10.1152/ajpcell.00002.2016] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 03/02/2016] [Indexed: 01/08/2023]
Abstract
The β1-adrenergic signaling system is one of the most important protein signaling systems in cardiac cells. It regulates cardiac action potential duration, intracellular Ca2+concentration ([Ca2+]i) transients, and contraction force. In this paper, a comprehensive experimentally based mathematical model of the β1-adrenergic signaling system for mouse ventricular myocytes is explored to simulate the effects of moderate stimulations of β1-adrenergic receptors (β1-ARs) on the action potential, Ca2+and Na+dynamics, as well as the effects of inhibition of protein kinase A (PKA) and phosphodiesterase of type 4 (PDE4). Simulation results show that the action potential prolongations reach saturating values at relatively small concentrations of isoproterenol (∼0.01 μM), while the [Ca2+]itransient amplitude saturates at significantly larger concentrations (∼0.1–1.0 μM). The differences in the response of Ca2+and Na+fluxes to moderate stimulation of β1-ARs are also observed. Sensitivity analysis of the mathematical model is performed and the model limitations are discussed. The investigated model reproduces most of the experimentally observed effects of moderate stimulation of β1-ARs, PKA, and PDE4 inhibition on the L-type Ca2+current, [Ca2+]itransients, and the sarcoplasmic reticulum Ca2+load and makes testable predictions for the action potential duration and [Ca2+]itransients as functions of isoproterenol concentration.
Collapse
Affiliation(s)
- Mark Grinshpon
- Department of Mathematics and Statistics, Georgia State University, Atlanta, Georgia; and
| | - Vladimir E. Bondarenko
- Department of Mathematics and Statistics, Georgia State University, Atlanta, Georgia; and
- Neuroscience Institute, Georgia State University, Atlanta, Georgia
| |
Collapse
|
36
|
dos Santos RL, da Silva FB, Ribeiro RF, Stefanon I. Sex hormones in the cardiovascular system. Horm Mol Biol Clin Investig 2015; 18:89-103. [PMID: 25390005 DOI: 10.1515/hmbci-2013-0048] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2013] [Accepted: 04/29/2014] [Indexed: 12/24/2022]
Abstract
Gender-associated differences in the development of cardiovascular diseases have been described in humans and animals. These differences could explain the low incidence of cardiovascular disease in women in the reproductive period, such as stroke, hypertension, and atherosclerosis. The cardiovascular protection observed in females has been attributed to the beneficial effects of estrogen on endothelial function. Besides estrogen, sex hormones are able to modulate blood pressure by acting on important systems as cardiovascular, renal, and neural. They can have complementary or antagonistic actions. For example, testosterone can raise blood pressure by stimulating the renin-angiotensin-aldosterone system, whereas estrogen alone or combined with progesterone has been associated with decreased blood pressure. The effects of testosterone in the development of cardiovascular disease are contradictory. Although some researchers suggest a positive effect, others indicate negative actions of testosterone. Estrogens physiologically stimulate the release of endothelium-derived vasodilator factors and inhibit the renin-angiotensin system. Although the cardioprotective effects of estrogen are widely appreciated, little is known about the effects of progesterone, which is commonly used in hormone replacement therapy. Progesterone has both vasodilatory and vasoconstrictive effects in the vasculature, depending on the location of the vessel and the level of exposure. Nevertheless, the mechanisms through which sex hormones modulate blood pressure have not been fully elucidated. Therefore, the characterization of those could lead to a better understanding of hypertension in women and men and perhaps to improved forms of therapy.
Collapse
|
37
|
Dalpiaz PLM, Lamas AZ, Caliman IF, Ribeiro RF, Abreu GR, Moyses MR, Andrade TU, Gouvea SA, Alves MF, Carmona AK, Bissoli NS. Sex Hormones Promote Opposite Effects on ACE and ACE2 Activity, Hypertrophy and Cardiac Contractility in Spontaneously Hypertensive Rats. PLoS One 2015; 10:e0127515. [PMID: 26010093 PMCID: PMC4444272 DOI: 10.1371/journal.pone.0127515] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Accepted: 04/16/2015] [Indexed: 11/25/2022] Open
Abstract
Background There is growing interest in sex differences and RAS components. However, whether gender influences cardiac angiotensin I-converting enzyme (ACE) and angiotensin-converting enzyme 2 (ACE2) activity is still unknown. In the present work, we determined the relationship between ACE and ACE2 activity, left ventricular function and gender in spontaneously hypertensive rats (SHRs). Methodology / Principal Findings Twelve-week-old female (F) and male (M) SHRs were divided into 2 experimental groups (n = 7 in each group): sham (S) and gonadectomized (G). Fifty days after gonadectomy, we measured positive and negative first derivatives (dP/dt maximum left ventricle (LV) and dP/dt minimum LV, respectively), hypertrophy (morphometric analysis) and ACE and ACE2 catalytic activity (fluorimetrically). Expression of calcium handling proteins was measured by western blot. Male rats exhibited higher cardiac ACE and ACE2 activity as well as hypertrophy compared to female rats. Orchiectomy decreased the activity of these enzymes and hypertrophy, while ovariectomy increased hypertrophy and ACE2, but did not change ACE activity. For cardiac function, the male sham group had a lower +dP/dt than the female sham group. After gonadectomy, the +dP/dt increased in males and reduced in females. The male sham group had a lower -dP/dt than the female group. After gonadectomy, the -dP/dt increased in the male and decreased in the female groups when compared to the sham group. No difference was observed among the groups in SERCA2a protein expression. Gonadectomy increased protein expression of PLB (phospholamban) and the PLB to SERCA2a ratio in female rats, but did not change in male rats. Conclusion Ovariectomy leads to increased cardiac hypertrophy, ACE2 activity, PLB expression and PLB to SERCA2a ratio, and worsening of hemodynamic variables, whereas in males the removal of testosterone has the opposite effects on RAS components.
Collapse
Affiliation(s)
- P. L. M. Dalpiaz
- Department of Physiological Sciences, Federal University of Espirito Santo, Vitória, Espirito Santo, Brazil
- * E-mail:
| | - A. Z. Lamas
- Department of Physiological Sciences, Federal University of Espirito Santo, Vitória, Espirito Santo, Brazil
| | - I. F. Caliman
- Department of Physiological Sciences, Federal University of Espirito Santo, Vitória, Espirito Santo, Brazil
| | - R. F. Ribeiro
- Department of Physiological Sciences, Federal University of Espirito Santo, Vitória, Espirito Santo, Brazil
| | - G. R. Abreu
- Department of Physiological Sciences, Federal University of Espirito Santo, Vitória, Espirito Santo, Brazil
| | - M. R. Moyses
- Department of Physiological Sciences, Federal University of Espirito Santo, Vitória, Espirito Santo, Brazil
| | - T. U. Andrade
- Department of Pharmacy, University Vila Velha, Vila Velha, Espirito Santo, Brazil
| | - S. A. Gouvea
- Nucleus of Biotechnology, Federal University of Espirito Santo, Vitória, Espirito Santo, Brazil
| | - M. F. Alves
- Department of Biophysics, Escola Paulista de Medicina, Federal University of São Paulo, São Paulo, Brazil
| | - A. K. Carmona
- Department of Biophysics, Escola Paulista de Medicina, Federal University of São Paulo, São Paulo, Brazil
| | - N. S. Bissoli
- Department of Physiological Sciences, Federal University of Espirito Santo, Vitória, Espirito Santo, Brazil
| |
Collapse
|
38
|
Sprenger JU, Perera RK, Steinbrecher JH, Lehnart SE, Maier LS, Hasenfuss G, Nikolaev VO. In vivo model with targeted cAMP biosensor reveals changes in receptor-microdomain communication in cardiac disease. Nat Commun 2015; 6:6965. [PMID: 25917898 DOI: 10.1038/ncomms7965] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 03/18/2015] [Indexed: 11/09/2022] Open
Abstract
3',5'-cyclic adenosine monophosphate (cAMP) is an ubiquitous second messenger that regulates physiological functions by acting in distinct subcellular microdomains. Although several targeted cAMP biosensors are developed and used in single cells, it is unclear whether such biosensors can be successfully applied in vivo, especially in the context of disease. Here, we describe a transgenic mouse model expressing a targeted cAMP sensor and analyse microdomain-specific second messenger dynamics in the vicinity of the sarcoplasmic/endoplasmic reticulum calcium ATPase (SERCA). We demonstrate the biocompatibility of this targeted sensor and its potential for real-time monitoring of compartmentalized cAMP signalling in adult cardiomyocytes isolated from a healthy mouse heart and from an in vivo cardiac disease model. In particular, we uncover the existence of a phosphodiesterase-dependent receptor-microdomain communication, which is affected in hypertrophy, resulting in reduced β-adrenergic receptor-cAMP signalling to SERCA.
Collapse
Affiliation(s)
- Julia U Sprenger
- 1] Emmy Noether Group of the DFG, European Heart Research Institute Göttingen, University Medical Center Göttingen, D-37075 Göttingen, Germany [2] Department of Cardiology and Pulmonology, Heart Research Center Göttingen, University Medical Center Göttingen, Georg August University, D-37075 Göttingen, Germany [3] Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, D-20246 Hamburg, Germany
| | - Ruwan K Perera
- 1] Emmy Noether Group of the DFG, European Heart Research Institute Göttingen, University Medical Center Göttingen, D-37075 Göttingen, Germany [2] Department of Cardiology and Pulmonology, Heart Research Center Göttingen, University Medical Center Göttingen, Georg August University, D-37075 Göttingen, Germany [3] Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, D-20246 Hamburg, Germany
| | - Julia H Steinbrecher
- Department of Cardiology and Pulmonology, Heart Research Center Göttingen, University Medical Center Göttingen, Georg August University, D-37075 Göttingen, Germany
| | - Stephan E Lehnart
- 1] Department of Cardiology and Pulmonology, Heart Research Center Göttingen, University Medical Center Göttingen, Georg August University, D-37075 Göttingen, Germany [2] German Center for Cardiovascular Research (DZHK), D-93053 Regensburg, Germany
| | - Lars S Maier
- Department of Internal Medicine II, University Hospital Regensburg, D-93053 Regensburg, Germany
| | - Gerd Hasenfuss
- 1] Department of Cardiology and Pulmonology, Heart Research Center Göttingen, University Medical Center Göttingen, Georg August University, D-37075 Göttingen, Germany [2] German Center for Cardiovascular Research (DZHK), D-93053 Regensburg, Germany
| | - Viacheslav O Nikolaev
- 1] Emmy Noether Group of the DFG, European Heart Research Institute Göttingen, University Medical Center Göttingen, D-37075 Göttingen, Germany [2] Department of Cardiology and Pulmonology, Heart Research Center Göttingen, University Medical Center Göttingen, Georg August University, D-37075 Göttingen, Germany [3] Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, D-20246 Hamburg, Germany [4] German Center for Cardiovascular Research (DZHK), D-93053 Regensburg, Germany
| |
Collapse
|
39
|
Liu GS, Morales A, Vafiadaki E, Lam CK, Cai WF, Haghighi K, Adly G, Hershberger RE, Kranias EG. A novel human R25C-phospholamban mutation is associated with super-inhibition of calcium cycling and ventricular arrhythmia. Cardiovasc Res 2015; 107:164-74. [PMID: 25852082 DOI: 10.1093/cvr/cvv127] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 03/20/2015] [Indexed: 12/26/2022] Open
Abstract
AIMS Depressed sarcoplasmic reticulum (SR) Ca(2+) cycling, a universal characteristic of human and experimental heart failure, may be associated with genetic alterations in key Ca(2+)-handling proteins. In this study, we identified a novel PLN mutation (R25C) in dilated cardiomyopathy (DCM) and investigated its functional significance in cardiomyocyte Ca(2+)-handling and contractility. METHODS AND RESULTS Exome sequencing identified a C73T substitution in the coding region of PLN in a family with DCM. The four heterozygous family members had implantable cardiac defibrillators, and three developed prominent ventricular arrhythmias. Overexpression of R25C-PLN in adult rat cardiomyocytes significantly suppressed the Ca(2+) affinity of SR Ca(2+)-ATPase (SERCA2a), resulting in decreased SR Ca(2+) content, Ca(2+) transients, and impaired contractile function, compared with WT-PLN. These inhibitory effects were associated with enhanced interaction of R25C-PLN with SERCA2, which was prevented by PKA phosphorylation. Accordingly, isoproterenol stimulation relieved the depressive effects of R25C-PLN in cardiomyocytes. However, R25C-PLN also elicited increases in the frequency of Ca(2+) sparks and waves as well as stress-induced aftercontractions. This was accompanied by increased Ca(2+)/calmodulin-dependent protein kinase II activity and hyper-phosphorylation of RyR2 at serine 2814. CONCLUSION The findings demonstrate that human R25C-PLN is associated with super-inhibition of SERCA2a and Ca(2+) transport as well as increased SR Ca(2+) leak, promoting arrhythmogenesis under stress conditions. This is the first mechanistic evidence that increased PLN inhibition may impact both SR Ca(2+) uptake and Ca(2+) release activities and suggests that the human R25C-PLN may be a prognostic factor for increased ventricular arrhythmia risk in DCM carriers.
Collapse
Affiliation(s)
- Guan-Sheng Liu
- Department of Pharmacology and Cell Biophysics, University of Cincinnati College of Medicine, PO Box 670575, 231 Albert Sabin Way, Cincinnati, OH, USA
| | - Ana Morales
- Division of Human Genetics, Ohio State University College of Medicine, Columbus, OH, USA Dorothy M. Davis Heart and Lung Research Institute, Ohio State University College of Medicine, Columbus, OH 45267-0575, USA
| | - Elizabeth Vafiadaki
- Molecular Biology Division, Biomedical Research Foundation, Academy of Athens, Greece
| | - Chi Keung Lam
- Department of Pharmacology and Cell Biophysics, University of Cincinnati College of Medicine, PO Box 670575, 231 Albert Sabin Way, Cincinnati, OH, USA
| | - Wen-Feng Cai
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Kobra Haghighi
- Department of Pharmacology and Cell Biophysics, University of Cincinnati College of Medicine, PO Box 670575, 231 Albert Sabin Way, Cincinnati, OH, USA
| | - George Adly
- Department of Pharmacology and Cell Biophysics, University of Cincinnati College of Medicine, PO Box 670575, 231 Albert Sabin Way, Cincinnati, OH, USA
| | - Ray E Hershberger
- Division of Human Genetics, Ohio State University College of Medicine, Columbus, OH, USA Dorothy M. Davis Heart and Lung Research Institute, Ohio State University College of Medicine, Columbus, OH 45267-0575, USA Division of Cardiovascular Medicine, Department of Internal Medicine, Ohio State University College of Medicine, Columbus, OH, USA
| | - Evangelia G Kranias
- Department of Pharmacology and Cell Biophysics, University of Cincinnati College of Medicine, PO Box 670575, 231 Albert Sabin Way, Cincinnati, OH, USA Molecular Biology Division, Biomedical Research Foundation, Academy of Athens, Greece
| |
Collapse
|
40
|
Shanmugam M, Li D, Gao S, Fefelova N, Shah V, Voit A, Pachon R, Yehia G, Xie LH, Babu GJ. Cardiac specific expression of threonine 5 to alanine mutant sarcolipin results in structural remodeling and diastolic dysfunction. PLoS One 2015; 10:e0115822. [PMID: 25671318 PMCID: PMC4324845 DOI: 10.1371/journal.pone.0115822] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 12/02/2014] [Indexed: 12/23/2022] Open
Abstract
The functional importance of threonine 5 (T5) in modulating the activity of sarcolipin (SLN), a key regulator of sarco/endoplasmic reticulum (SR) Ca2+ ATPase (SERCA) pump was studied using a transgenic mouse model with cardiac specific expression of threonine 5 to alanine mutant SLN (SLNT5A). In these transgenic mice, the SLNT5A protein replaces the endogenous SLN in atria, while maintaining the total SLN content. The cardiac specific expression of SLNT5A results in severe cardiac structural remodeling accompanied by bi-atrial enlargement. Biochemical analyses reveal a selective downregulation of SR Ca2+ handling proteins and a reduced SR Ca2+ uptake both in atria and in the ventricles. Optical mapping analysis shows slower action potential propagation in the transgenic mice atria. Doppler echocardiography and hemodynamic measurements demonstrate a reduced atrial contractility and an impaired diastolic function. Together, these findings suggest that threonine 5 plays an important role in modulating SLN function in the heart. Furthermore, our studies suggest that alteration in SLN function can cause abnormal Ca2+ handling and subsequent cardiac remodeling and dysfunction.
Collapse
Affiliation(s)
- Mayilvahanan Shanmugam
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, New Jersey, United States of America
| | - Dan Li
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, New Jersey, United States of America
| | - Shumin Gao
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, New Jersey, United States of America
| | - Nadezhda Fefelova
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, New Jersey, United States of America
| | - Vikas Shah
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, New Jersey, United States of America
| | - Antanina Voit
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, New Jersey, United States of America
| | - Ronald Pachon
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, New Jersey, United States of America
| | - Ghassan Yehia
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, New Jersey, United States of America
| | - Lai-Hua Xie
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, New Jersey, United States of America
| | - Gopal J. Babu
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, New Jersey, United States of America
- * E-mail:
| |
Collapse
|
41
|
Kao DP, Lowes BD, Gilbert EM, Minobe W, Epperson LE, Meyer LK, Ferguson DA, Volkman AK, Zolty R, Borg CD, Quaife RA, Bristow MR. Therapeutic Molecular Phenotype of β-Blocker-Associated Reverse-Remodeling in Nonischemic Dilated Cardiomyopathy. ACTA ACUST UNITED AC 2015; 8:270-83. [PMID: 25637602 DOI: 10.1161/circgenetics.114.000767] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Accepted: 01/14/2015] [Indexed: 01/01/2023]
Abstract
BACKGROUND When β-blockers produce reverse-remodeling in idiopathic dilated cardiomyopathy, they partially reverse changes in fetal-adult/contractile protein, natriuretic peptide, SR-Ca(2+)-ATPase gene program constituents. The objective of the current study was to further test the hypothesis that reverse-remodeling is associated with favorable changes in myocardial gene expression by measuring additional contractile, signaling, and metabolic genes that exhibit a fetal/adult expression predominance, are thyroid hormone-responsive, and are regulated by β1-adrenergic receptor signaling. A secondary objective was to identify which of these putative regulatory networks is most closely associated with observed changes. METHODS AND RESULTS Forty-seven patients with idiopathic dilated cardiomyopathy (left ventricular ejection fraction, 0.24±0.09) were randomized to the adrenergic-receptor blockers metoprolol (β1-selective), metoprolol+doxazosin (β1/α1), or carvedilol (β1/β2/α1). Serial radionuclide ventriculography and endomyocardial biopsies were performed at baseline, 3, and 12 months. Expression of 50 mRNA gene products was measured by quantitative polymerase chain reaction. Thirty-one patients achieved left ventricular ejection fraction reverse-remodeling response defined as improvement by ≥0.08 at 12 months or by ≥0.05 at 3 months (Δ left ventricular ejection fraction, 0.21±0.10). Changes in gene expression in responders versus nonresponders were decreases in NPPA and NPPB and increases in MYH6, ATP2A2, PLN, RYR2, ADRA1A, ADRB1, MYL3, PDFKM, PDHX, and CPT1B. All except PDHX involved increase in adult or decrease in fetal cardiac genes, but 100% were concordant with changes predicted by inhibition of β1-adrenergic signaling. CONCLUSIONS In addition to known gene expression changes, additional calcium-handling, sarcomeric, adrenergic signaling, and metabolic genes were associated with reverse-remodeling. The pattern suggests a fetal-adult paradigm but may be because of reversal of gene expression controlled by a β1-adrenergic receptor gene network. CLINICAL TRIAL REGISTRATION URL: www.clinicaltrials.gov. Unique Identifier: NCT01798992.
Collapse
Affiliation(s)
- David P Kao
- From the Division of Cardiology, Department of Medicine, University of Colorado School of Medicine, Aurora (D.P.K., W.M., L.E.E., L.K.M., D.A.F., R.A.Q., M.R.B.); Division of Cardiology, Department of Medicine, University of Nebraska Medical Center, Omaha (B.D.L.); Division of Cardiology, Department of Medicine, University of Utah, Salt Lake City (E.M.G., A.K.V.); Division of Cardiology, Department of Medicine, Albert Einstein College of Medicine, Bronx, NY (R.Z.); and Heart Clinic of Arkansas, Little Rock (C.D.B.)
| | - Brian D Lowes
- From the Division of Cardiology, Department of Medicine, University of Colorado School of Medicine, Aurora (D.P.K., W.M., L.E.E., L.K.M., D.A.F., R.A.Q., M.R.B.); Division of Cardiology, Department of Medicine, University of Nebraska Medical Center, Omaha (B.D.L.); Division of Cardiology, Department of Medicine, University of Utah, Salt Lake City (E.M.G., A.K.V.); Division of Cardiology, Department of Medicine, Albert Einstein College of Medicine, Bronx, NY (R.Z.); and Heart Clinic of Arkansas, Little Rock (C.D.B.)
| | - Edward M Gilbert
- From the Division of Cardiology, Department of Medicine, University of Colorado School of Medicine, Aurora (D.P.K., W.M., L.E.E., L.K.M., D.A.F., R.A.Q., M.R.B.); Division of Cardiology, Department of Medicine, University of Nebraska Medical Center, Omaha (B.D.L.); Division of Cardiology, Department of Medicine, University of Utah, Salt Lake City (E.M.G., A.K.V.); Division of Cardiology, Department of Medicine, Albert Einstein College of Medicine, Bronx, NY (R.Z.); and Heart Clinic of Arkansas, Little Rock (C.D.B.)
| | - Wayne Minobe
- From the Division of Cardiology, Department of Medicine, University of Colorado School of Medicine, Aurora (D.P.K., W.M., L.E.E., L.K.M., D.A.F., R.A.Q., M.R.B.); Division of Cardiology, Department of Medicine, University of Nebraska Medical Center, Omaha (B.D.L.); Division of Cardiology, Department of Medicine, University of Utah, Salt Lake City (E.M.G., A.K.V.); Division of Cardiology, Department of Medicine, Albert Einstein College of Medicine, Bronx, NY (R.Z.); and Heart Clinic of Arkansas, Little Rock (C.D.B.)
| | - L Elaine Epperson
- From the Division of Cardiology, Department of Medicine, University of Colorado School of Medicine, Aurora (D.P.K., W.M., L.E.E., L.K.M., D.A.F., R.A.Q., M.R.B.); Division of Cardiology, Department of Medicine, University of Nebraska Medical Center, Omaha (B.D.L.); Division of Cardiology, Department of Medicine, University of Utah, Salt Lake City (E.M.G., A.K.V.); Division of Cardiology, Department of Medicine, Albert Einstein College of Medicine, Bronx, NY (R.Z.); and Heart Clinic of Arkansas, Little Rock (C.D.B.)
| | - Leslie K Meyer
- From the Division of Cardiology, Department of Medicine, University of Colorado School of Medicine, Aurora (D.P.K., W.M., L.E.E., L.K.M., D.A.F., R.A.Q., M.R.B.); Division of Cardiology, Department of Medicine, University of Nebraska Medical Center, Omaha (B.D.L.); Division of Cardiology, Department of Medicine, University of Utah, Salt Lake City (E.M.G., A.K.V.); Division of Cardiology, Department of Medicine, Albert Einstein College of Medicine, Bronx, NY (R.Z.); and Heart Clinic of Arkansas, Little Rock (C.D.B.)
| | - Debra A Ferguson
- From the Division of Cardiology, Department of Medicine, University of Colorado School of Medicine, Aurora (D.P.K., W.M., L.E.E., L.K.M., D.A.F., R.A.Q., M.R.B.); Division of Cardiology, Department of Medicine, University of Nebraska Medical Center, Omaha (B.D.L.); Division of Cardiology, Department of Medicine, University of Utah, Salt Lake City (E.M.G., A.K.V.); Division of Cardiology, Department of Medicine, Albert Einstein College of Medicine, Bronx, NY (R.Z.); and Heart Clinic of Arkansas, Little Rock (C.D.B.)
| | - Ann Kirkpatrick Volkman
- From the Division of Cardiology, Department of Medicine, University of Colorado School of Medicine, Aurora (D.P.K., W.M., L.E.E., L.K.M., D.A.F., R.A.Q., M.R.B.); Division of Cardiology, Department of Medicine, University of Nebraska Medical Center, Omaha (B.D.L.); Division of Cardiology, Department of Medicine, University of Utah, Salt Lake City (E.M.G., A.K.V.); Division of Cardiology, Department of Medicine, Albert Einstein College of Medicine, Bronx, NY (R.Z.); and Heart Clinic of Arkansas, Little Rock (C.D.B.)
| | - Ronald Zolty
- From the Division of Cardiology, Department of Medicine, University of Colorado School of Medicine, Aurora (D.P.K., W.M., L.E.E., L.K.M., D.A.F., R.A.Q., M.R.B.); Division of Cardiology, Department of Medicine, University of Nebraska Medical Center, Omaha (B.D.L.); Division of Cardiology, Department of Medicine, University of Utah, Salt Lake City (E.M.G., A.K.V.); Division of Cardiology, Department of Medicine, Albert Einstein College of Medicine, Bronx, NY (R.Z.); and Heart Clinic of Arkansas, Little Rock (C.D.B.)
| | - C Douglas Borg
- From the Division of Cardiology, Department of Medicine, University of Colorado School of Medicine, Aurora (D.P.K., W.M., L.E.E., L.K.M., D.A.F., R.A.Q., M.R.B.); Division of Cardiology, Department of Medicine, University of Nebraska Medical Center, Omaha (B.D.L.); Division of Cardiology, Department of Medicine, University of Utah, Salt Lake City (E.M.G., A.K.V.); Division of Cardiology, Department of Medicine, Albert Einstein College of Medicine, Bronx, NY (R.Z.); and Heart Clinic of Arkansas, Little Rock (C.D.B.)
| | - Robert A Quaife
- From the Division of Cardiology, Department of Medicine, University of Colorado School of Medicine, Aurora (D.P.K., W.M., L.E.E., L.K.M., D.A.F., R.A.Q., M.R.B.); Division of Cardiology, Department of Medicine, University of Nebraska Medical Center, Omaha (B.D.L.); Division of Cardiology, Department of Medicine, University of Utah, Salt Lake City (E.M.G., A.K.V.); Division of Cardiology, Department of Medicine, Albert Einstein College of Medicine, Bronx, NY (R.Z.); and Heart Clinic of Arkansas, Little Rock (C.D.B.)
| | - Michael R Bristow
- From the Division of Cardiology, Department of Medicine, University of Colorado School of Medicine, Aurora (D.P.K., W.M., L.E.E., L.K.M., D.A.F., R.A.Q., M.R.B.); Division of Cardiology, Department of Medicine, University of Nebraska Medical Center, Omaha (B.D.L.); Division of Cardiology, Department of Medicine, University of Utah, Salt Lake City (E.M.G., A.K.V.); Division of Cardiology, Department of Medicine, Albert Einstein College of Medicine, Bronx, NY (R.Z.); and Heart Clinic of Arkansas, Little Rock (C.D.B.).
| |
Collapse
|
42
|
Haghighi K, Bidwell P, Kranias EG. Phospholamban interactome in cardiac contractility and survival: A new vision of an old friend. J Mol Cell Cardiol 2014; 77:160-7. [PMID: 25451386 PMCID: PMC4312245 DOI: 10.1016/j.yjmcc.2014.10.005] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Revised: 10/06/2014] [Accepted: 10/09/2014] [Indexed: 01/10/2023]
Abstract
Depressed sarcoplasmic reticulum (SR) calcium cycling, reflecting impaired SR Ca-transport and Ca-release, is a key and universal characteristic of human and experimental heart failure. These SR processes are regulated by multimeric protein complexes, including protein kinases and phosphatases as well as their anchoring and regulatory subunits that fine-tune Ca-handling in specific SR sub-compartments. SR Ca-transport is mediated by the SR Ca-ATPase (SERCA2a) and its regulatory phosphoprotein, phospholamban (PLN). Dephosphorylated PLN is an inhibitor of SERCA2a and phosphorylation by protein kinase A (PKA) or calcium-calmodulin-dependent protein kinases (CAMKII) relieves these inhibitory effects. Recent studies identified additional regulatory proteins, associated with PLN, that control SR Ca-transport. These include the inhibitor-1 (I-1) of protein phosphatase 1 (PP1), the small heat shock protein 20 (Hsp20) and the HS-1 associated protein X-1 (HAX1). In addition, the intra-luminal histidine-rich calcium binding protein (HRC) has been shown to interact with both SERCA2a and triadin. Notably, there is physical and direct interaction between these protein players, mediating a fine-cross talk between SR Ca-uptake, storage and release. Importantly, regulation of SR Ca-cycling by the PLN/SERCA interactome does not only impact cardiomyocyte contractility, but also survival and remodeling. Indeed, naturally occurring variants in these Ca-cycling genes modulate their activity and interactions with other protein partners, resulting in depressed contractility and accelerated remodeling. These genetic variants may serve as potential prognostic or diagnostic markers in cardiac pathophysiology.
Collapse
Affiliation(s)
- Kobra Haghighi
- Department of Pharmacology and Cell Biophysics, University of Cincinnati, College of Medicine, Cincinnati, OH 45267, USA
| | - Philip Bidwell
- Department of Pharmacology and Cell Biophysics, University of Cincinnati, College of Medicine, Cincinnati, OH 45267, USA
| | - Evangelia G Kranias
- Department of Pharmacology and Cell Biophysics, University of Cincinnati, College of Medicine, Cincinnati, OH 45267, USA.
| |
Collapse
|
43
|
Locatelli J, de Assis LVM, Isoldi MC. Calcium handling proteins: structure, function, and modulation by exercise. Heart Fail Rev 2014; 19:207-25. [PMID: 23436107 DOI: 10.1007/s10741-013-9373-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Heart failure is a serious public health issue with a growing prevalence, and it is related with the aging of the population. Hypertension is identified as the main precursor of left ventricular hypertrophy and therefore can lead to diastolic dysfunction and heart failure. Scientific studies have confirmed the beneficial effects of the physical exercise by reducing the blood pressure and improving the functional status of the heart in hypertension. Several proteins are involved in the mobilization of calcium during the coupling excitation-contraction process in the heart among those are sarcoplasmic reticulum Ca(2+)-ATPase, phospholamban, calsequestrin, sodium-calcium exchanger, L-type calcium's channel, and ryanodine receptors. Our goal is to address the beneficial effects of exercise on the calcium handling proteins in a heart with hypertension.
Collapse
Affiliation(s)
- Jamille Locatelli
- Institute of Exact and Biological Sciences, Federal University of Ouro Preto, Ouro Prêto, Brazil
| | | | | |
Collapse
|
44
|
Sinnecker D, Dirschinger RJ, Barthel P, Müller A, Morley-Davies A, Hapfelmeier A, Dommasch M, Huster KM, Hasenfuss G, Laugwitz KL, Malik M, Schmidt G. Postextrasystolic blood pressure potentiation predicts poor outcome of cardiac patients. J Am Heart Assoc 2014; 3:e000857. [PMID: 24895163 PMCID: PMC4309081 DOI: 10.1161/jaha.114.000857] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background Postextrasystolic blood pressure potentiation (PESP), the pulse wave augmentation after an extrasystolic beat, is typically enhanced in heart failure (HF) patients. This study prospectively tested the association of PESP and mortality in cardiac patients. Methods and Results Consecutive patients (n=941; mean age, 61 years; 19% female) presenting with acute myocardial infarction were enrolled between May 2000 and March 2005 and followed up until August 2010. The main study outcome was 5‐year all‐cause mortality. Patients underwent noninvasive 30‐minute recordings of ECG and continuous blood pressure. PESP presence was based on the ratio between the first postectopic pulse wave amplitude and the mean of the subsequent 9 pulse wave amplitudes. A ratio above 1 was prospectively defined as PESP present. Ventricular premature complexes (VPCs) suitable for PESP quantification were present in recordings of 220 patients. PESP was present in 62 of these patients. Patients without suitable VPCs were classified as PESP absent. During the follow‐up, 72 patients died. Among the 220 patients in whom PESP was measurable, 27 died. Under univariable analysis, PESP was a significant predictor of death (P<0.001) as were GRACE score (P<0.001), left ventricular ejection fraction (LVEF) (P<0.001), and the number of recorded VPCs (P<0.001). Under multivariable analysis, PESP (P<0.001), GRACE score (P<0.001), and LVEF (P=0.001) were independently associated with outcome. The combination of PESP presence and LVEF ≤35% identified a subgroup of patients with a particularly high mortality of 46.7%. Separate validation reproduced the finding in an unrelated population of 146 HF patients. Conclusions PESP, which likely reflects abnormalities of myocardial calcium cycling, predicts the mortality risk in postinfarction patients. Clinical Trial Registration URL: ClinicalTrials.gov. Unique identifier: NCT00196274.
Collapse
Affiliation(s)
- Daniel Sinnecker
- Medizinische Klinik und Deutsches Herzzentrum, München der Technischen Universität München, Munich, Germany (D.S., R.J.D., P.B., A., M.D., K.M.H., K.L.L., G.S.)
| | - Ralf J Dirschinger
- Medizinische Klinik und Deutsches Herzzentrum, München der Technischen Universität München, Munich, Germany (D.S., R.J.D., P.B., A., M.D., K.M.H., K.L.L., G.S.)
| | - Petra Barthel
- Medizinische Klinik und Deutsches Herzzentrum, München der Technischen Universität München, Munich, Germany (D.S., R.J.D., P.B., A., M.D., K.M.H., K.L.L., G.S.)
| | - Alexander Müller
- Medizinische Klinik und Deutsches Herzzentrum, München der Technischen Universität München, Munich, Germany (D.S., R.J.D., P.B., A., M.D., K.M.H., K.L.L., G.S.)
| | - Adrian Morley-Davies
- Department of Cardiology, University Hospital of North Staffordshire, NHS Trust, City General Hospital, Staffordshire, UK (A.M.D.)
| | - Alexander Hapfelmeier
- Institut für Medizinische Statistik und Epidemiologie der Technischen, Universität München, Munich, Germany (A.H.)
| | - Michael Dommasch
- Medizinische Klinik und Deutsches Herzzentrum, München der Technischen Universität München, Munich, Germany (D.S., R.J.D., P.B., A., M.D., K.M.H., K.L.L., G.S.)
| | - Katharina M Huster
- Medizinische Klinik und Deutsches Herzzentrum, München der Technischen Universität München, Munich, Germany (D.S., R.J.D., P.B., A., M.D., K.M.H., K.L.L., G.S.)
| | - Gerd Hasenfuss
- Department of Cardiology and Pneumology, Georg-August-University Göttingen, Göttingen, Germany (G.H.)
| | - Karl-Ludwig Laugwitz
- Medizinische Klinik und Deutsches Herzzentrum, München der Technischen Universität München, Munich, Germany (D.S., R.J.D., P.B., A., M.D., K.M.H., K.L.L., G.S.) DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany (K.L.L., G.S.)
| | - Marek Malik
- St. Paul's Cardiac Electrophysiology, University of London and Imperial College, London, UK (M.M.)
| | - Georg Schmidt
- Medizinische Klinik und Deutsches Herzzentrum, München der Technischen Universität München, Munich, Germany (D.S., R.J.D., P.B., A., M.D., K.M.H., K.L.L., G.S.) DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany (K.L.L., G.S.)
| |
Collapse
|
45
|
Simon JN, Duglan D, Casadei B, Carnicer R. Nitric oxide synthase regulation of cardiac excitation-contraction coupling in health and disease. J Mol Cell Cardiol 2014; 73:80-91. [PMID: 24631761 DOI: 10.1016/j.yjmcc.2014.03.004] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Revised: 03/04/2014] [Accepted: 03/05/2014] [Indexed: 02/07/2023]
Abstract
Significant advances in our understanding of the ability of nitric oxide synthases (NOS) to modulate cardiac function have provided key insights into the role NOS play in the regulation of excitation-contraction (EC) coupling in health and disease. Through both cGMP-dependent and cGMP-independent (e.g. S-nitrosylation) mechanisms, NOS have the ability to alter intracellular Ca(2+) handling and the myofilament response to Ca(2+), thereby impacting the systolic and diastolic performance of the myocardium. Findings from experiments using nitric oxide (NO) donors and NOS inhibition or gene deletion clearly implicate dysfunctional NOS as a critical contributor to many cardiovascular disease states. However, studies to date have only partially addressed NOS isoform-specific effects and, more importantly, how subcellular localization of NOS influences ion channels involved in myocardial EC coupling and excitability. In this review, we focus on the contribution of each NOS isoform to cardiac dysfunction and on the role of uncoupled NOS activity in common cardiac disease states, including heart failure, diabetic cardiomyopathy, ischemia/reperfusion injury and atrial fibrillation. We also review evidence that clearly indicates the importance of NO in cardioprotection. This article is part of a Special Issue entitled "Redox Signalling in the Cardiovascular System".
Collapse
Affiliation(s)
- Jillian N Simon
- Radcliffe Department of Medicine, Division of Cardiovascular Medicine, University of Oxford, Oxford, UK
| | - Drew Duglan
- Radcliffe Department of Medicine, Division of Cardiovascular Medicine, University of Oxford, Oxford, UK
| | - Barbara Casadei
- Radcliffe Department of Medicine, Division of Cardiovascular Medicine, University of Oxford, Oxford, UK
| | - Ricardo Carnicer
- Radcliffe Department of Medicine, Division of Cardiovascular Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
46
|
Mattiazzi A, Kranias EG. The role of CaMKII regulation of phospholamban activity in heart disease. Front Pharmacol 2014; 5:5. [PMID: 24550830 PMCID: PMC3913884 DOI: 10.3389/fphar.2014.00005] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Accepted: 01/07/2014] [Indexed: 01/06/2023] Open
Abstract
Phospholamban (PLN) is a phosphoprotein in cardiac sarcoplasmic reticulum (SR) that is a reversible regulator of the Ca2+-ATPase (SERCA2a) activity and cardiac contractility. Dephosphorylated PLN inhibits SERCA2a and PLN phosphorylation, at either Ser16 by PKA or Thr17 by Ca2+-calmodulin-dependent protein kinase (CaMKII), reverses this inhibition. Through this mechanism, PLN is a key modulator of SR Ca2+ uptake, Ca2+ load, contractility, and relaxation. PLN phosphorylation is also the main determinant of β1-adrenergic responses in the heart. Although phosphorylation of Thr17 by CaMKII contributes to this effect, its role is subordinate to the PKA-dependent increase in cytosolic Ca2+, necessary to activate CaMKII. Furthermore, the effects of PLN and its phosphorylation on cardiac function are subject to additional regulation by its interacting partners, the anti-apoptotic HAX-1 protein and Gm or the anchoring unit of protein phosphatase 1. Regulation of PLN activity by this multimeric complex becomes even more important in pathological conditions, characterized by aberrant Ca2+-cycling. In this scenario, CaMKII-dependent PLN phosphorylation has been associated with protective effects in both acidosis and ischemia/reperfusion. However, the beneficial effects of increasing SR Ca2+ uptake through PLN phosphorylation may be lost or even become deleterious, when these occur in association with alterations in SR Ca2+ leak. Moreover, a major characteristic in human and experimental heart failure (HF) is depressed SR Ca2+ uptake, associated with decreased SERCA2a levels and dephosphorylation of PLN, leading to decreased SR Ca2+ load and impaired contractility. Thus, the strategy of altering SERCA2a and/or PLN levels or activity to restore perturbed SR Ca2+ uptake is a potential therapeutic tool for HF treatment. We will review here the role of CaMKII-dependent phosphorylation of PLN at Thr17 on cardiac function under physiological and pathological conditions.
Collapse
Affiliation(s)
- Alicia Mattiazzi
- Facultad de Medicina, Centro de Investigaciones Cardiovasculares, Conicet La Plata-Universidad Nacional de La Plata La Plata, Argentina
| | - Evangelia G Kranias
- Department of Pharmacology and Cell Biophysics, College of Medicine, University of Cincinnati Cincinnati, OH, USA
| |
Collapse
|
47
|
Abstract
Heart failure is one of the leading causes of sudden death in developed countries. While current therapies are mostly aimed at mitigating associated symptoms, novel therapies targeting the subcellular mechanisms underlying heart failure are emerging. Failing hearts are characterized by reduced contractile properties caused by impaired Ca(2+) cycling between the sarcoplasm and sarcoplasmic reticulum (SR). Sarcoplasmic/ endoplasmic reticulum Ca(2+)ATPase 2a (SERCA2a) mediates Ca(2+) reuptake into the SR in cardiomyocytes. Of note, the expression level and/or activity of SERCA2a, translating to the quantity of SR Ca(2+) uptake, are significantly reduced in failing hearts. Normalization of the SERCA2a expression level by gene delivery has been shown to restore hampered cardiac functions and ameliorate associated symptoms in pre-clinical as well as clinical studies. SERCA2a activity can be regulated at multiple levels of a signaling cascade comprised of phospholamban, protein phosphatase 1, inhibitor-1, and PKCα. SERCA2 activity is also regulated by post-translational modifications including SUMOylation and acetylation. In this review, we will highlight the molecular mechanisms underlying the regulation of SERCA2a activity and the potential therapeutic modalities for the treatment of heart failure.
Collapse
Affiliation(s)
- Woo Jin Park
- Global Research Laboratory and College of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 500-712, Korea.
| | | |
Collapse
|
48
|
Abstract
Ca(2+)-ATPases (pumps) are key to the regulation of Ca(2+) in eukaryotic cells: nine are known today, belonging to three multigene families. The three endo(sarco)plasmic reticulum (SERCA) and the four plasma membrane (PMCA) pumps have been known for decades, the two Secretory Pathway Ca(2+) ATPase (SPCA) pumps have only become known recently. The number of pump isoforms is further increased by alternative splicing processes. The three pump types share the basic features of the catalytic mechanism, but differ in a number of properties related to tissue distribution, regulation, and role in the cellular homeostasis of Ca(2+). The molecular understanding of the function of all pumps has received great impetus from the solution of the three-dimensional (3D) structure of one of them, the SERCA pump. This landmark structural advance has been accompanied by the emergence and rapid expansion of the area of pump malfunction. Most of the pump defects described so far are genetic and produce subtler, often tissue and isoform specific, disturbances that affect individual components of the Ca(2+)-controlling and/or processing machinery, compellingly indicating a specialized role for each Ca(2+) pump type and/or isoform.
Collapse
Affiliation(s)
- Marisa Brini
- Department of Comparative Biomedicine and Food Science, University of Padova, Legnaro Padova, Italy.
| | | | | | | |
Collapse
|
49
|
Tuncay E, Okatan EN, Vassort G, Turan B. ß-blocker timolol prevents arrhythmogenic Ca²⁺ release and normalizes Ca²⁺ and Zn²⁺ dyshomeostasis in hyperglycemic rat heart. PLoS One 2013; 8:e71014. [PMID: 23923043 PMCID: PMC3726605 DOI: 10.1371/journal.pone.0071014] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Accepted: 06/25/2013] [Indexed: 01/08/2023] Open
Abstract
Defective cardiac mechanical activity in diabetes results from alterations in intracellular Ca(2+) handling, in part, due to increased oxidative stress. Beta-blockers demonstrate marked beneficial effects in heart dysfunction with scavenging free radicals and/or acting as an antioxidant. The aim of this study was to address how β-blocker timolol-treatment of diabetic rats exerts cardioprotection. Timolol-treatment (12-week), one-week following diabetes induction, prevented diabetes-induced depressed left ventricular basal contractile activity, prolonged cellular electrical activity, and attenuated the increase in isolated-cardiomyocyte size without hyperglycemic effect. Both in vivo and in vitro timolol-treatment of diabetic cardiomyocytes prevented the altered kinetic parameters of Ca(2+) transients and reduced Ca(2+) loading of sarcoplasmic reticulum (SR), basal intracellular free Ca(2+) and Zn(2+) ([Ca(2+)]i and [Zn(2+)]i), and spatio-temporal properties of the Ca(2+) sparks, significantly. Timolol also antagonized hyperphosphorylation of cardiac ryanodine receptor (RyR2), and significantly restored depleted protein levels of both RyR2 and calstabin2. Western blot analysis demonstrated that timolol-treatment also significantly normalized depressed levels of some [Ca(2+)]i-handling regulators, such as Na(+)/Ca(2+) exchanger (NCX) and phospho-phospholamban (pPLN) to PLN ratio. Incubation of diabetic cardiomyocytes with 4-mM glutathione exerted similar beneficial effects on RyR2-macromolecular complex and basal levels of both [Ca(2+)]i and [Zn(2+)]i, increased intracellular Zn(2+) hyperphosphorylated RyR2 in a concentration-dependent manner. Timolol also led to a balanced oxidant/antioxidant level in both heart and circulation and prevented altered cellular redox state of the heart. We thus report, for the first time, that the preventing effect of timolol, directly targeting heart, seems to be associated with a normalization of macromolecular complex of RyR2 and some Ca(2+) handling regulators, and prevention of Ca(2+) leak, and thereby normalization of both [Ca(2+)]i and [Zn(2+)]i homeostasis in diabetic rat heart, at least in part by controlling the cellular redox status of hyperglycemic cardiomyocytes.
Collapse
Affiliation(s)
- Erkan Tuncay
- Department of Biophysics, Faculty of Medicine, Ankara University, Ankara, Turkey
| | - Esma N. Okatan
- Department of Biophysics, Faculty of Medicine, Ankara University, Ankara, Turkey
| | - Guy Vassort
- INSERM U-1046, CHU Arnaud de Villeneuve, Montpellier, France
| | - Belma Turan
- Department of Biophysics, Faculty of Medicine, Ankara University, Ankara, Turkey
- * E-mail:
| |
Collapse
|
50
|
Ribeiro RF, Potratz FF, Pavan BMM, Forechi L, Lima FLM, Fiorim J, Fernandes AA, Vassallo DV, Stefanon I. Carvedilol prevents ovariectomy-induced myocardial contractile dysfunction in female rat. PLoS One 2013; 8:e53226. [PMID: 23308166 PMCID: PMC3538779 DOI: 10.1371/journal.pone.0053226] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2012] [Accepted: 11/26/2012] [Indexed: 11/18/2022] Open
Abstract
Carvedilol has beneficial effects on cardiac function in patients with heart failure but its effect on ovariectomy-induced myocardial contractile dysfunction remains unclear. Estrogen deficiency induces myocardial contractile dysfunction and increases cardiovascular disease risk in postmenopausal women. Our aim was to investigate whether carvedilol, a beta receptor blocker, would prevent ovariectomy-induced myocardial contractile dysfunction. Female rats (8 weeks old) that underwent bilateral ovariectomy were randomly assigned to receive daily treatment with carvedilol (OVX+CAR, 20 mg/kg), placebo (OVX) and SHAM for 58 days. Left ventricle papillary muscle was mounted for isometric tension recordings. The inotropic response to Ca2+ (0.62 to 3.75 mM) and isoproterenol (Iso 10−8 to 10−2 M) were assessed. Expression of calcium handling proteins was measured by western blot analysis. Carvedilol treatment in the OVX animals: prevented weight gain and slight hypertrophy, restored the reduced positive inotropic responses to Ca2+ and isoproterenol, prevented the reduction in SERCA2a expression, abolished the increase in superoxide anion production, normalized the increase in p22phox expression, and decreased serum angiotensin converting enzyme (ACE) activity. This study demonstrated that myocardial contractile dysfunction and SERCA2a down regulation were prevented by carvedilol treatment. Superoxide anion production and NADPH oxidase seem to be involved in this response.
Collapse
|