1
|
Chakraborty P, Po SS, Yabluchanskiy A, Dasari TW. Protein kinase A: A potential marker of sympathovagal imbalance in heart failure. Life Sci 2023; 331:122069. [PMID: 37666387 DOI: 10.1016/j.lfs.2023.122069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 08/23/2023] [Accepted: 09/01/2023] [Indexed: 09/06/2023]
Abstract
Mitigation of cardiac autonomic dysregulation by neuromodulation technologies is emerging as a new therapeutic modality of heart failure (HF). This recent progress has necessitated the identification of a biomarker for the quantification of sympathovagal balance, the potential target of 'neuromodulation' strategies. The currently available autonomic nervous system assessment parameters do not truly reflect the sympathovagal balance of the ventricle. Protein kinase A (PKA) is an intracellular enzyme that plays a major role in the pathophysiology of functional and structural ventricular remodeling in HF. Interestingly, sympathetic and parasympathetic activations exert reciprocal influence on the activity of PKA. The current review attempts to evaluate the potential concept and feasibility of using in vitro assessment of PKA activity as a marker of sympathovagal balance in HF.
Collapse
Affiliation(s)
- Praloy Chakraborty
- Cardiovascular Section, Department of Internal Medicine, Heart Rhythm Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Sunny S Po
- Cardiovascular Section, Department of Internal Medicine, Heart Rhythm Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Andriy Yabluchanskiy
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Tarun W Dasari
- Cardiovascular Section, Department of Internal Medicine, Heart Rhythm Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| |
Collapse
|
2
|
Yang BY, Coult J, Blackwood J, Kwok H, Rajah A, Goldenberg I, Sotoodehenia N, Harris JR, Kudenchuk PJ, Rea TD. Title: Age, sex, and survival following ventricular fibrillation cardiac arrest: a mechanistic evaluation of the ECG waveform: Short title: Age, sex, and survival via VF waveform in OHCA. Resuscitation 2023:109891. [PMID: 37390958 DOI: 10.1016/j.resuscitation.2023.109891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 06/20/2023] [Accepted: 06/21/2023] [Indexed: 07/02/2023]
Abstract
BACKGROUND Studies of outcome differences by sex in out-of-hospital cardiac arrest (OHCA) have produced mixed results that may depend on age, a potential surrogate for menopausal status. OBJECTIVE We used quantitative measures of ventricular fibrillation (VF) waveforms - indicators of the myocardium's physiology - to assess whether survival differences according to sex and age group may be mediated via a biologic mechanism. METHODS We conducted a cohort study of VF-OHCA in a metropolitan EMS system. We used multivariable logistic regression to assess the association of survival to hospital discharge with sex and age group (<55, ≥55 years). We determined the proportion of outcome difference mediated by VF waveform measures: VitalityScore and amplitude spectrum area (AMSA). RESULTS Among 1526 VF-OHCA patients, the average age was 62 years, and 29% were female. Overall, younger women were more likely to survive than younger men (survival 67% vs 54%, p=0.02), while survival among older women and older men did not differ (40% vs 44%, p=0.3). Adjusting for Utstein characteristics, women <55 compared to men <55 had greater odds of survival to hospital discharge (OR=1.93, 95% CI 1.23-3.09), an association not observed between the ≥55 groups. Waveform measures were more favorable among women and mediated some of the beneficial association between female sex and survival among those <55 years: 47% for VitalityScore and 25% for AMSA. CONCLUSIONS Women <55 years were more likely to survive than men <55 years following VF-OHCA. The biologic mechanism represented by VF waveform mediated some, though not all, of the outcome difference.
Collapse
Affiliation(s)
- Betty Y Yang
- Department of Emergency Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, United States
| | - Jason Coult
- Department of Medicine, Division of General Medicine, University of Washington, Seattle, WA, United States
| | - Jennifer Blackwood
- Emergency Medical Services Division of Public Health - Seattle & King County, Seattle, WA, United States
| | - Heemun Kwok
- Department of Emergency Medicine, University of Washington. Postal address: 326 9th Avenue, Seattle, WA, 98104, United States
| | - Anjali Rajah
- Department of Medicine, Division of General Medicine, University of Washington, Seattle, WA, United States
| | - Ilan Goldenberg
- Clinical Cardiovascular Research Center, Division of Cardiology, Department of Medicine, University of Rochester Medical Center, Rochester, New York, United States
| | - Nona Sotoodehenia
- Department of Medicine, Division of Cardiology, University of Washington, Seattle, Washington, United States
| | - Jeffrey R Harris
- Department of Health Systems and Population Health, University of Washington School of Public Health, Seattle, WA, United States
| | - Peter J Kudenchuk
- Emergency Medical Services Division of Public Health - Seattle & King County, Seattle, WA, United States; Department of Medicine, Division of Cardiology, University of Washington, Seattle, Washington, United States
| | - Thomas D Rea
- Department of Medicine, Division of General Medicine, University of Washington, Seattle, WA, United States; Emergency Medical Services Division of Public Health - Seattle & King County, Seattle, WA, United States
| |
Collapse
|
3
|
Kumar M, Haghighi K, Koch S, Rubinstein J, Stillitano F, Hajjar RJ, Kranias EG, Sadayappan S. Myofilament Alterations Associated with Human R14del-Phospholamban Cardiomyopathy. Int J Mol Sci 2023; 24:2675. [PMID: 36768995 PMCID: PMC9917359 DOI: 10.3390/ijms24032675] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/23/2023] [Accepted: 01/27/2023] [Indexed: 02/01/2023] Open
Abstract
Phospholamban (PLN) is a major regulator of cardiac contractility, and human mutations in this gene give rise to inherited cardiomyopathies. The deletion of Arginine 14 is the most-prevalent cardiomyopathy-related mutation, and it has been linked to arrhythmogenesis and early death. Studies in PLN-humanized mutant mice indicated an increased propensity to arrhythmias, but the underlying cellular mechanisms associated with R14del-PLN cardiac dysfunction in the absence of any apparent structural remodeling remain unclear. The present study addressed the specific role of myofilaments in the setting of R14del-PLN and the long-term effects of R14del-PLN in the heart. Maximal force was depressed in skinned cardiomyocytes from both left and right ventricles, but this effect was more pronounced in the right ventricle of R14del-PLN mice. In addition, the Ca2+ sensitivity of myofilaments was increased in both ventricles of mutant mice. However, the depressive effects of R14del-PLN on contractile parameters could be reversed with the positive inotropic drug omecamtiv mecarbil, a myosin activator. At 12 months of age, corresponding to the mean symptomatic age of R14del-PLN patients, contractile parameters and Ca2+ transients were significantly depressed in the right ventricular R14del-PLN cardiomyocytes. Echocardiography did not reveal any alterations in cardiac function or remodeling, although histological and electron microscopy analyses indicated subtle alterations in mutant hearts. These findings suggest that both aberrant myocyte calcium cycling and aberrant contractility remain specific to the right ventricle in the long term. In addition, altered myofilament activity is an early characteristic of R14del-PLN mutant hearts and the positive inotropic drug omecamtiv mecarbil may be beneficial in treating R14del-PLN cardiomyopathy.
Collapse
Affiliation(s)
- Mohit Kumar
- Department of Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Kobra Haghighi
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Sheryl Koch
- Department of Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Jack Rubinstein
- Department of Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Francesca Stillitano
- Division Heart and Lung, Department of Cardiology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Roger J. Hajjar
- Phospholamban Heart Foundation, Postbus 66, 1775 ZH Middenmeer, The Netherlands
| | - Evangelia G. Kranias
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Sakthivel Sadayappan
- Department of Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| |
Collapse
|
4
|
Henderson MJ, Trychta KA, Yang SM, Bäck S, Yasgar A, Wires ES, Danchik C, Yan X, Yano H, Shi L, Wu KJ, Wang AQ, Tao D, Zahoránszky-Kőhalmi G, Hu X, Xu X, Maloney D, Zakharov AV, Rai G, Urano F, Airavaara M, Gavrilova O, Jadhav A, Wang Y, Simeonov A, Harvey BK. A target-agnostic screen identifies approved drugs to stabilize the endoplasmic reticulum-resident proteome. Cell Rep 2021; 35:109040. [PMID: 33910017 DOI: 10.1016/j.celrep.2021.109040] [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: 09/01/2020] [Revised: 02/12/2021] [Accepted: 04/06/2021] [Indexed: 12/24/2022] Open
Abstract
Endoplasmic reticulum (ER) dysregulation is associated with pathologies including neurodegenerative, muscular, and diabetic conditions. Depletion of ER calcium can lead to the loss of resident proteins in a process termed exodosis. To identify compounds that attenuate the redistribution of ER proteins under pathological conditions, we performed a quantitative high-throughput screen using the Gaussia luciferase (GLuc)-secreted ER calcium modulated protein (SERCaMP) assay, which monitors secretion of ER-resident proteins triggered by calcium depletion. We identify several clinically used drugs, including bromocriptine, and further characterize them using assays to measure effects on ER calcium, ER stress, and ER exodosis. Bromocriptine elicits protective effects in cell-based models of exodosis as well as in vivo models of stroke and diabetes. Bromocriptine analogs with reduced dopamine receptor activity retain similar efficacy in stabilizing the ER proteome, indicating a non-canonical mechanism of action. This study describes a strategic approach to identify small-molecule drugs capable of improving ER proteostasis in human disease conditions.
Collapse
Affiliation(s)
- Mark J Henderson
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA.
| | - Kathleen A Trychta
- National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD 21224, USA
| | - Shyh-Ming Yang
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| | - Susanne Bäck
- National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD 21224, USA
| | - Adam Yasgar
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| | - Emily S Wires
- National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD 21224, USA
| | - Carina Danchik
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| | - Xiaokang Yan
- National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD 21224, USA
| | - Hideaki Yano
- National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD 21224, USA
| | - Lei Shi
- National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD 21224, USA
| | - Kuo-Jen Wu
- Center for Neuropsychiatric Research, National Health Research Institutes, Zhunan 35053, Taiwan
| | - Amy Q Wang
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| | - Dingyin Tao
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| | - Gergely Zahoránszky-Kőhalmi
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| | - Xin Hu
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| | - Xin Xu
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| | - David Maloney
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| | - Alexey V Zakharov
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| | - Ganesha Rai
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| | - Fumihiko Urano
- Department of Medicine, Division of Endocrinology, Metabolism, and Lipid Research, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Mikko Airavaara
- Neuroscience Center, HiLIFE & Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Oksana Gavrilova
- National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD 20892, USA
| | - Ajit Jadhav
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| | - Yun Wang
- Center for Neuropsychiatric Research, National Health Research Institutes, Zhunan 35053, Taiwan
| | - Anton Simeonov
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| | - Brandon K Harvey
- National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD 21224, USA.
| |
Collapse
|
5
|
Liu Y, Chen J, Fontes SK, Bautista EN, Cheng Z. Physiological And Pathological Roles Of Protein Kinase A In The Heart. Cardiovasc Res 2021; 118:386-398. [PMID: 33483740 DOI: 10.1093/cvr/cvab008] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 11/30/2020] [Accepted: 01/08/2021] [Indexed: 12/21/2022] Open
Abstract
Protein kinase A (PKA) is a central regulator of cardiac performance and morphology. Myocardial PKA activation is induced by a variety of hormones, neurotransmitters and stress signals, most notably catecholamines secreted by the sympathetic nervous system. Catecholamines bind β-adrenergic receptors to stimulate cAMP-dependent PKA activation in cardiomyocytes. Elevated PKA activity enhances Ca2+ cycling and increases cardiac muscle contractility. Dynamic control of PKA is essential for cardiac homeostasis, as dysregulation of PKA signaling is associated with a broad range of heart diseases. Specifically, abnormal PKA activation or inactivation contributes to the pathogenesis of myocardial ischemia, hypertrophy, heart failure, as well as diabetic, takotsubo, or anthracycline cardiomyopathies. PKA may also determine sex-dependent differences in contractile function and heart disease predisposition. Here, we describe the recent advances regarding the roles of PKA in cardiac physiology and pathology, highlighting previous study limitations and future research directions. Moreover, we discuss the therapeutic strategies and molecular mechanisms associated with cardiac PKA biology. In summary, PKA could serve as a promising drug target for cardioprotection. Depending on disease types and mechanisms, therapeutic intervention may require either inhibition or activation of PKA. Therefore, specific PKA inhibitors or activators may represent valuable drug candidates for the treatment of heart diseases.
Collapse
Affiliation(s)
- Yuening Liu
- Department of Pharmaceutical Sciences, Washington State University, PBS 423, 412 E. Spokane Falls Blvd, ., Spokane, WA, 99202-2131, USA
| | - Jingrui Chen
- Department of Pharmaceutical Sciences, Washington State University, PBS 423, 412 E. Spokane Falls Blvd, ., Spokane, WA, 99202-2131, USA
| | - Shayne K Fontes
- Department of Pharmaceutical Sciences, Washington State University, PBS 423, 412 E. Spokane Falls Blvd, ., Spokane, WA, 99202-2131, USA
| | - Erika N Bautista
- Department of Pharmaceutical Sciences, Washington State University, PBS 423, 412 E. Spokane Falls Blvd, ., Spokane, WA, 99202-2131, USA
| | - Zhaokang Cheng
- Department of Pharmaceutical Sciences, Washington State University, PBS 423, 412 E. Spokane Falls Blvd, ., Spokane, WA, 99202-2131, USA
| |
Collapse
|
6
|
Florea V, Rieger AC, Natsumeda M, Tompkins BA, Banerjee MN, Schulman IH, Premer C, Khan A, Valasaki K, Heidecker B, Mantero A, Balkan W, Mitrani RD, Hare JM. The impact of patient sex on the response to intramyocardial mesenchymal stem cell administration in patients with non-ischaemic dilated cardiomyopathy. Cardiovasc Res 2020; 116:2131-2141. [PMID: 32053144 PMCID: PMC7584465 DOI: 10.1093/cvr/cvaa004] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 10/31/2019] [Accepted: 02/05/2020] [Indexed: 12/11/2022] Open
Abstract
AIMS Sex differences impact the occurrence, presentation, prognosis, and response to therapy in heart disease. Particularly, the phenotypic presentation of patients with non-ischaemic dilated cardiomyopathy (NIDCM) differs between men and women. However, whether the response to mesenchymal stem cell (MSC) therapy is influenced by sex remains unknown. We hypothesize that males and females with NIDCM respond similarly to MSC therapy. METHODS AND RESULTS Male (n = 24) and female (n = 10) patients from the POSEIDON-DCM trial who received MSCs via transendocardial injections were evaluated over 12 months. Endothelial function was measured at baseline and 3 months post-transendocardial stem cell injection (TESI). At baseline, ejection fraction (EF) was lower (P = 0.004) and end-diastolic volume (EDV; P = 0.0002) and end-systolic volume (ESV; P = 0.0002) were higher in males vs. females. In contrast, baseline demographic characteristics, Minnesota Living with Heart Failure Questionnaire (MLHFQ), and 6-min walk test (6MWT) were similar between groups. EF improved in males by 6.2 units (P = 0.04) and in females by 8.6 units (P = 0.04; males vs. females, P = 0.57). EDV and ESV were unchanged over time. The MLHFQ score, New York Heart Association (NYHA) class, endothelial progenitor cell-colony forming units, and serum tumour necrosis factor alpha improved similarly in both groups. CONCLUSION Despite major differences in phenotypic presentation of NIDCM in males and females, this study is the first of its kind to demonstrate that MSC therapy improves a variety of parameters in NIDCM irrespective of patient sex. These findings have important clinical and pathophysiologic implications regarding the impact of sex on responses to cell-based therapy for NIDCM.
Collapse
Affiliation(s)
- Victoria Florea
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Biomedical Research Building - 9th Floor 1501 NW 10th Ave, Miami, FL 33136, USA
| | - Angela C Rieger
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Biomedical Research Building - 9th Floor 1501 NW 10th Ave, Miami, FL 33136, USA
| | - Makoto Natsumeda
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Biomedical Research Building - 9th Floor 1501 NW 10th Ave, Miami, FL 33136, USA
| | - Bryon A Tompkins
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Biomedical Research Building - 9th Floor 1501 NW 10th Ave, Miami, FL 33136, USA
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Monisha N Banerjee
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Biomedical Research Building - 9th Floor 1501 NW 10th Ave, Miami, FL 33136, USA
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Ivonne H Schulman
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Biomedical Research Building - 9th Floor 1501 NW 10th Ave, Miami, FL 33136, USA
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Courtney Premer
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Biomedical Research Building - 9th Floor 1501 NW 10th Ave, Miami, FL 33136, USA
| | - Aisha Khan
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Biomedical Research Building - 9th Floor 1501 NW 10th Ave, Miami, FL 33136, USA
| | - Krystalenia Valasaki
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Biomedical Research Building - 9th Floor 1501 NW 10th Ave, Miami, FL 33136, USA
| | - Bettina Heidecker
- Department of Cardiology, Charite Berlin University of Medicine, Berlin, Germany
| | - Alejandro Mantero
- Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Wayne Balkan
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Biomedical Research Building - 9th Floor 1501 NW 10th Ave, Miami, FL 33136, USA
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Raul D Mitrani
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Biomedical Research Building - 9th Floor 1501 NW 10th Ave, Miami, FL 33136, USA
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Joshua M Hare
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Biomedical Research Building - 9th Floor 1501 NW 10th Ave, Miami, FL 33136, USA
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
7
|
Stølen TO, Høydal MA, Ahmed MS, Jørgensen K, Garten K, Hortigon-Vinagre MP, Zamora V, Scrimgeour NR, Berre AMO, Nes BM, Skogvoll E, Johnsen AB, Moreira JBN, McMullen JR, Attramadal H, Smith GL, Ellingsen Ø, Wisløff U. Exercise training reveals micro-RNAs associated with improved cardiac function and electrophysiology in rats with heart failure after myocardial infarction. J Mol Cell Cardiol 2020; 148:106-119. [PMID: 32918915 DOI: 10.1016/j.yjmcc.2020.08.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 08/20/2020] [Accepted: 08/25/2020] [Indexed: 02/09/2023]
Abstract
AIMS Endurance training improves aerobic fitness and cardiac function in individuals with heart failure. However, the underlying mechanisms are not well characterized. Exercise training could therefore act as a tool to discover novel targets for heart failure treatment. We aimed to associate changes in Ca2+ handling and electrophysiology with micro-RNA (miRNA) profile in exercise trained heart failure rats to establish which miRNAs induce heart failure-like effects in Ca2+ handling and electrophysiology. METHODS AND RESULTS Post-myocardial infarction (MI) heart failure was induced in Sprague Dawley rats. Rats with MI were randomized to sedentary control (sed), moderate (mod)- or high-intensity (high) endurance training for 8 weeks. Exercise training improved cardiac function, Ca2+ handling and electrophysiology including reduced susceptibility to arrhythmia in an exercise intensity-dependent manner where high intensity gave a larger effect. Fifty-five miRNAs were significantly regulated (up or down) in MI-sed, of which 18 and 3 were changed towards Sham-sed in MI-high and MI-mod, respectively. Thereafter we experimentally altered expression of these "exercise-miRNAs" individually in human induced pluripotent stem cell-derived cardiomyocytes (hIPSC-CM) in the same direction as they were changed in MI. Of the "exercise-miRNAs", miR-214-3p prolonged AP duration, whereas miR-140 and miR-208a shortened AP duration. miR-497-5p prolonged Ca2+ release whereas miR-214-3p and miR-31a-5p prolonged Ca2+ decay. CONCLUSION Using exercise training as a tool, we discovered that miR-214-3p, miR-497-5p, miR-31a-5p contribute to heart-failure like behaviour in Ca2+ handling and electrophysiology and could be potential treatment targets.
Collapse
Affiliation(s)
- Tomas O Stølen
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health, Norwegian University of Science and Technology, Trondheim, Norway; Department of Cardiology, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway; Department of Cardiothoracic Surgery, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway.
| | - Morten A Høydal
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health, Norwegian University of Science and Technology, Trondheim, Norway; Department of Cardiology, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway; Department of Cardiothoracic Surgery, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Muhammad Shakil Ahmed
- Institute for Surgical Research, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Kari Jørgensen
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health, Norwegian University of Science and Technology, Trondheim, Norway
| | - Karin Garten
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health, Norwegian University of Science and Technology, Trondheim, Norway
| | - Maria P Hortigon-Vinagre
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Science, University of Glasgow 126 University Place, Glasgow G12 8TA, United Kingdom
| | - Victor Zamora
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Science, University of Glasgow 126 University Place, Glasgow G12 8TA, United Kingdom
| | - Nathan R Scrimgeour
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health, Norwegian University of Science and Technology, Trondheim, Norway
| | - Anne Marie Ormbostad Berre
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health, Norwegian University of Science and Technology, Trondheim, Norway
| | - Bjarne M Nes
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health, Norwegian University of Science and Technology, Trondheim, Norway; Department of Cardiology, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Eirik Skogvoll
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health, Norwegian University of Science and Technology, Trondheim, Norway; Department of Anesthesia and Intensive Care Medicine, St. Olav University Hospital, Trondheim, Norway
| | - Anne Berit Johnsen
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health, Norwegian University of Science and Technology, Trondheim, Norway
| | - Jose B N Moreira
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health, Norwegian University of Science and Technology, Trondheim, Norway
| | - Julie R McMullen
- Cardiac Hypertrophy Laboratory, Baker Heart & Diabetes Institute, 75 Commercial Road, Melbourne, VIC, Australia
| | - Håvard Attramadal
- Institute for Surgical Research, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Godfrey L Smith
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health, Norwegian University of Science and Technology, Trondheim, Norway; Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Science, University of Glasgow 126 University Place, Glasgow G12 8TA, United Kingdom
| | - Øyvind Ellingsen
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health, Norwegian University of Science and Technology, Trondheim, Norway; Department of Cardiology, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Ulrik Wisløff
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health, Norwegian University of Science and Technology, Trondheim, Norway; School of Human Movement & Nutrition Sciences, University of Queensland, Australia
| |
Collapse
|
8
|
Association with SERCA2a directs phospholamban trafficking to sarcoplasmic reticulum from a nuclear envelope pool. J Mol Cell Cardiol 2020; 143:107-119. [DOI: 10.1016/j.yjmcc.2020.04.025] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 04/08/2020] [Accepted: 04/23/2020] [Indexed: 11/22/2022]
|
9
|
Zhao J, Lei Y, Yang Y, Gao H, Gai Z, Li X. Metoprolol alleviates arginine vasopressin-induced cardiomyocyte hypertrophy by upregulating the AKT1-SERCA2 cascade in H9C2 cells. Cell Biosci 2020; 10:72. [PMID: 32489586 PMCID: PMC7247229 DOI: 10.1186/s13578-020-00434-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 05/16/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Arginine vasopressin (AVP) is elevated in patients with heart failure, and the increase in the AVP concentration in plasma is positively correlated with disease severity and mortality. Metoprolol (Met) is a beta blocker that is widely used in the clinic to treat pathological cardiac hypertrophy and to improve heart function. However, the specific mechanism by which Met alleviates AVP-induced pathological cardiac hypertrophy is still unknown. Our current study aimed to evaluate the inhibitory effects of Met on AVP-induced cardiomyocyte hypertrophy and the underlying mechanisms. METHODS AVP alone or AVP plus Met was added to the wild type or AKT1-overexpressing rat cardiac H9C2 cell line. The cell surface areas and ANP/BNP/β-MHC expressions were used to evaluate the levels of hypertrophy. Western bolting was used to analyze AKT1/P-AKT1, AKT2/P-AKT2, total AKT, SERCA2, and Phospholamban (PLN) expression. Fluo3-AM was used to measure the intracellular Ca2+ stores. RESULTS In the current study, we found that AKT1 but not AKT2 mediated the pathogenesis of AVP-induced cardiomyocyte hypertrophy. Sustained stimulation (48 h) with AVP led to hypertrophy in the H9C2 rat cardiomyocytes, resulting in the downregulation of AKT1 (0.48 fold compared to control) and SERCA2 (0.62 fold), the upregulation of PLN (1.32 fold), and the increase in the cytoplasmic calcium concentration (1.52 fold). In addition, AKT1 overexpression increased the expression of SERCA2 (1.34 fold) and decreased the expression of PLN (0.48 fold) in the H9C2 cells. Moreover, we found that Met could attenuate the AVP-induced changes in AKT1, SERCA2 and PLN expression and decreased the cytoplasmic calcium concentration in the H9C2 cells. CONCLUSIONS Our results demonstrated that the AKT1-SERCA2 cascade served as an important regulatory pathway in AVP-induced pathological cardiac hypertrophy.
Collapse
Affiliation(s)
- Jieqiong Zhao
- Department of Cardiology, Tangdu Hospital, Air Force Medical University, Xi’an, 710038 Shaanxi People’s Republic of China
| | - Yonghong Lei
- Department of Plastic Surgery, General Hospital of Chinese PLA, Beijing, 100853 People’s Republic of China
| | - Yanping Yang
- Department of Cardiology, Tangdu Hospital, Air Force Medical University, Xi’an, 710038 Shaanxi People’s Republic of China
| | - Haibo Gao
- Department of Cardiology, Tangdu Hospital, Air Force Medical University, Xi’an, 710038 Shaanxi People’s Republic of China
| | - Zhongchao Gai
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi’an, 710021 Shaanxi People’s Republic of China
| | - Xue Li
- Department of Cardiology, Tangdu Hospital, Air Force Medical University, Xi’an, 710038 Shaanxi People’s Republic of China
| |
Collapse
|
10
|
Jiao L, Machuki JO, Wu Q, Shi M, Fu L, Adekunle AO, Tao X, Xu C, Hu X, Yin Z, Sun H. Estrogen and calcium handling proteins: new discoveries and mechanisms in cardiovascular diseases. Am J Physiol Heart Circ Physiol 2020; 318:H820-H829. [PMID: 32083972 DOI: 10.1152/ajpheart.00734.2019] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Estrogen deficiency is considered to be an important factor leading to cardiovascular diseases (CVDs). Indeed, the prevalence of CVDs in postmenopausal women exceeds that of premenopausal women and men of the same age. Recent research findings provide evidence that estrogen plays a pivotal role in the regulation of calcium homeostasis and therefore fine-tunes normal cardiomyocyte contraction and relaxation processes. Disruption of calcium homeostasis is closely associated with the pathological mechanism of CVDs. Thus, this paper maps out and summarizes the effects and mechanisms of estrogen on calcium handling proteins in cardiac myocytes, including L-type Ca2+ channel, the sarcoplasmic reticulum Ca2+ release channel named ryanodine receptor, sarco(endo)plasmic reticulum Ca2+-ATPase, and sodium-calcium exchanger. In so doing, we provide theoretical and experimental evidence for the successful design of estrogen-based prevention and treatment therapies for CVDs.
Collapse
Affiliation(s)
- Lijuan Jiao
- Department of Physiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | | | - Qi Wu
- Department of Physiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Mingjin Shi
- Department of Physiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Lu Fu
- Department of Physiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | | | - Xi Tao
- Department of Physiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Chenxi Xu
- Department of Physiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xide Hu
- Department of Physiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Zeyuan Yin
- Department of Physiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Hong Sun
- Department of Physiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| |
Collapse
|
11
|
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: 55] [Impact Index Per Article: 11.0] [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
|
12
|
Mesquita TRR, Miguel-dos-Santos R, Jesus ICGD, de Almeida GKM, Fernandes VA, Gomes AAL, Guatimosim S, Martins-Silva L, Ferreira AJ, Capettini LDSA, Pesquero JL, Lauton-Santos S. Ablation of B1- and B2-kinin receptors causes cardiac dysfunction through redox-nitroso unbalance. Life Sci 2019; 228:121-127. [DOI: 10.1016/j.lfs.2019.04.062] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 04/24/2019] [Accepted: 04/26/2019] [Indexed: 01/03/2023]
|
13
|
Zhang Y, Wang WE, Zhang X, Li Y, Chen B, Liu C, Ai X, Zhang X, Tian Y, Zhang C, Tang M, Szeto C, Hua X, Xie M, Zeng C, Wu Y, Zhou L, Zhu W, Yu D, Houser SR, Chen X. Cardiomyocyte PKA Ablation Enhances Basal Contractility While Eliminates Cardiac β-Adrenergic Response Without Adverse Effects on the Heart. Circ Res 2019; 124:1760-1777. [PMID: 30982412 DOI: 10.1161/circresaha.118.313417] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
RATIONALE PKA (Protein Kinase A) is a major mediator of β-AR (β-adrenergic) regulation of cardiac function, but other mediators have also been suggested. Reduced PKA basal activity and activation are linked to cardiac diseases. However, how complete loss of PKA activity impacts on cardiac physiology and if it causes cardiac dysfunction have never been determined. OBJECTIVES We set to determine how the heart adapts to the loss of cardiomyocyte PKA activity and if it elicits cardiac abnormalities. METHODS AND RESULTS (1) Cardiac PKA activity was almost completely inhibited by expressing a PKA inhibitor peptide in cardiomyocytes (cPKAi) in mice; (2) cPKAi reduced basal phosphorylation of 2 myofilament proteins (TnI [troponin I] and cardiac myosin binding protein C), and one longitudinal SR (sarcoplasmic reticulum) protein (PLB [phospholamban]) but not of the sarcolemmal proteins (Cav1.2 α1c and PLM [phospholemman]), dyadic protein RyR2, and nuclear protein CREB (cAMP response element binding protein) at their PKA phosphorylation sites; (3) cPKAi increased the expression of CaMKII (Ca2+/calmodulin-dependent kinase II), the Cav1.2 β subunits and current, but decreased CaMKII phosphorylation and CaMKII-mediated phosphorylation of PLB and RyR2; (4) These changes resulted in significantly enhanced myofilament Ca2+ sensitivity, prolonged contraction, slowed relaxation but increased myocyte Ca2+ transient and contraction amplitudes; (5) Isoproterenol-induced PKA and CaMKII activation and their phosphorylation of proteins were prevented by cPKAi; (6) cPKAi abolished the increases of heart rate, and cardiac and myocyte contractility by a β-AR agonist (isoproterenol), showing an important role of PKA and a minimal role of PKA-independent β-AR signaling in acute cardiac regulation; (7) cPKAi mice have partial exercise capability probably by enhancing vascular constriction and ventricular filling during β-AR stimulation; and (8) cPKAi mice did not show any cardiac functional or structural abnormalities during the 1-year study period. CONCLUSIONS PKA activity suppression induces a unique Ca2+ handling phenotype, eliminates β-AR regulation of heart rates and cardiac contractility but does not cause cardiac abnormalities.
Collapse
Affiliation(s)
- Ying Zhang
- Cardiology, Daping Hospital, Third Military Medical University, Chongqing (Y.Z., W.E.W., C. Zeng, L.Z.)
- Endocrinology and Metabolism, The First Affiliated Hospital of Zhengzhou University (Y.Z.)
- Department of Physiology and Cardiovascular Research Center (Y.Z., W.E.W., X.Z., Y.L., C.L., X.A., X.Z., C.Z., M.T., C.S., S.R.H., X.C.), Temple University School of Medicine, Philadelphia, PA
| | - Wei Eric Wang
- Cardiology, Daping Hospital, Third Military Medical University, Chongqing (Y.Z., W.E.W., C. Zeng, L.Z.)
- Department of Physiology and Cardiovascular Research Center (Y.Z., W.E.W., X.Z., Y.L., C.L., X.A., X.Z., C.Z., M.T., C.S., S.R.H., X.C.), Temple University School of Medicine, Philadelphia, PA
| | - Xiaoying Zhang
- Department of Physiology and Cardiovascular Research Center (Y.Z., W.E.W., X.Z., Y.L., C.L., X.A., X.Z., C.Z., M.T., C.S., S.R.H., X.C.), Temple University School of Medicine, Philadelphia, PA
| | - Ying Li
- Department of Physiology and Cardiovascular Research Center (Y.Z., W.E.W., X.Z., Y.L., C.L., X.A., X.Z., C.Z., M.T., C.S., S.R.H., X.C.), Temple University School of Medicine, Philadelphia, PA
- The General Hospital of The PLA Rocket Force, Beijing, China (Y.L.)
| | - Biyi Chen
- Division of Cardiovascular Medicine, Department of Internal Medicine, François M. Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine (B.C.)
| | - Chong Liu
- Department of Physiology and Cardiovascular Research Center (Y.Z., W.E.W., X.Z., Y.L., C.L., X.A., X.Z., C.Z., M.T., C.S., S.R.H., X.C.), Temple University School of Medicine, Philadelphia, PA
- Pharmacology, Second Military Medical University, Shanghai (C.L.)
| | - Xiaojie Ai
- Department of Physiology and Cardiovascular Research Center (Y.Z., W.E.W., X.Z., Y.L., C.L., X.A., X.Z., C.Z., M.T., C.S., S.R.H., X.C.), Temple University School of Medicine, Philadelphia, PA
- School of Agriculture and Biology, Shanghai Key Laboratory of Veterinary Biotechnology, Shanghai Jiao Tong University (X.A.)
| | - Xiaoxiao Zhang
- Department of Physiology and Cardiovascular Research Center (Y.Z., W.E.W., X.Z., Y.L., C.L., X.A., X.Z., C.Z., M.T., C.S., S.R.H., X.C.), Temple University School of Medicine, Philadelphia, PA
- Department of Ultrasound, Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan (X.Z., M.X.)
| | - Ying Tian
- Department of Pharmacology, Center for Translational Medicine (Y.T., W.Z.), Temple University School of Medicine, Philadelphia, PA
| | - Chen Zhang
- Cardiology, Daping Hospital, Third Military Medical University, Chongqing (Y.Z., W.E.W., C. Zeng, L.Z.)
- Department of Physiology and Cardiovascular Research Center (Y.Z., W.E.W., X.Z., Y.L., C.L., X.A., X.Z., C.Z., M.T., C.S., S.R.H., X.C.), Temple University School of Medicine, Philadelphia, PA
| | - Mingxin Tang
- Department of Physiology and Cardiovascular Research Center (Y.Z., W.E.W., X.Z., Y.L., C.L., X.A., X.Z., C.Z., M.T., C.S., S.R.H., X.C.), Temple University School of Medicine, Philadelphia, PA
| | - Christopher Szeto
- Department of Physiology and Cardiovascular Research Center (Y.Z., W.E.W., X.Z., Y.L., C.L., X.A., X.Z., C.Z., M.T., C.S., S.R.H., X.C.), Temple University School of Medicine, Philadelphia, PA
| | - Xiang Hua
- Fox Chase Cancer Center, Philadelphia, PA (X.H.)
| | - Mingxin Xie
- Department of Ultrasound, Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan (X.Z., M.X.)
| | | | - Yingjie Wu
- Institute of Genome Engineered Animal Models for Human Diseases, National Center of Genetically Engineered Animal Models for International Research, Dalian Medical University, Liaoning (Y.W.)
| | - Lin Zhou
- Cardiology, Daping Hospital, Third Military Medical University, Chongqing (Y.Z., W.E.W., C. Zeng, L.Z.)
| | - Weizhong Zhu
- Department of Pharmacology, Center for Translational Medicine (Y.T., W.Z.), Temple University School of Medicine, Philadelphia, PA
- Pharmacology, School of Pharmacy, Nantong University, Jiangsu (W.Z.)
| | - Daohai Yu
- Clinical Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA (D.Y.)
| | - Steven R Houser
- Department of Physiology and Cardiovascular Research Center (Y.Z., W.E.W., X.Z., Y.L., C.L., X.A., X.Z., C.Z., M.T., C.S., S.R.H., X.C.), Temple University School of Medicine, Philadelphia, PA
| | - Xiongwen Chen
- Department of Physiology and Cardiovascular Research Center (Y.Z., W.E.W., X.Z., Y.L., C.L., X.A., X.Z., C.Z., M.T., C.S., S.R.H., X.C.), Temple University School of Medicine, Philadelphia, PA
| |
Collapse
|
14
|
Saad NS, Elnakish MT, Ahmed AAE, Janssen PML. Protein Kinase A as a Promising Target for Heart Failure Drug Development. Arch Med Res 2018; 49:530-537. [PMID: 30642654 PMCID: PMC6451668 DOI: 10.1016/j.arcmed.2018.12.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 12/13/2018] [Indexed: 12/24/2022]
Abstract
Heart failure (HF) is a clinical syndrome characterized by impaired ability of the heart to fill or eject blood. HF is rather prevalent and it represents the foremost reason of hospitalization in the United States. The costs linked to HF overrun those of all other causes of disabilities, and death in the United States and all over the developed as well as the developing countries which amplify the supreme significance of its prevention. Protein kinase (PK) A plays multiple roles in heart functions including, contraction, metabolism, ion fluxes, and gene transcription. Altered PKA activity is likely to cause the progression to cardiomyopathy and HF. Thus, this review is intended to focus on the roles of PKA and PKA-mediated signal transduction in the healthy heart as well as during the development of HF. Furthermore, the impact of cardiac PKA inhibition/activation will be highlighted to identify PKA as a potential target for the HF drug development.
Collapse
Affiliation(s)
- Nancy S Saad
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, USA; Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Helwan University, Cairo, Egypt
| | - Mohammad T Elnakish
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, USA; Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Helwan University, Cairo, Egypt
| | - Amany A E Ahmed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Helwan University, Cairo, Egypt
| | - Paul M L Janssen
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, USA; Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
15
|
Mollanoori H, Naderi N, Amin A, Hassani B, Shahraki H, Teimourian S. A novel human T17N-phospholamban variation in idiopathic dilated cardiomyopathy. GENE REPORTS 2018. [DOI: 10.1016/j.genrep.2018.06.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
16
|
Kajimoto K, Minami Y, Sato N, Otsubo S, Kasanuki H. Gender Differences in Left Ventricular Ejection Fraction and Outcomes Among Patients Hospitalized for Acute Decompensated Heart Failure. Am J Cardiol 2017; 119:1623-1630. [PMID: 28325570 DOI: 10.1016/j.amjcard.2017.02.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 02/08/2017] [Accepted: 02/08/2017] [Indexed: 11/19/2022]
Abstract
In patients with acute decompensated heart failure (HF), the association of gender and left ventricular ejection fraction (LVEF) with clinical outcomes has not been fully investigated. The aim of this study was to evaluate gender differences in LVEF and adverse outcomes across the full spectrum of LVEF in patients hospitalized for acute decompensated HF. Of the 4,842 patients enrolled in the Acute Decompensated Heart Failure Syndromes registry, 4,231 patients (2,461 men and 1,770 women) discharged alive after hospitalization for acute decompensated HF were investigated to assess the association of gender and LVEF with the primary end point (all-cause death and readmission for HF). Men or women were divided into 5 groups based on the LVEF at hospital discharge (<30%, 30% to <40%, 40% to <50%, 50% to <60%, and ≥60%). The median follow-up period after discharge was 523 (384 to 791) days. The frequency of the primary end point did not differ between men and women (36.5% vs 38.1%, p = 0.291). After adjustment for multiple comorbidities, male patients with an LVEF <30%, 30% to <40%, 40% to <50%, or 50% to <60% had a significantly higher risk of the primary end point than those with an LVEF ≥60%, indicating an inverse association between LVEF and adverse outcomes. In contrast, the adjusted risk of the primary end point was similar for all 5 LVEF groups of female patients. In conclusions, the association between LVEF and outcomes differs markedly between men and women hospitalized for acute decompensated HF, although event-free survival is similar for both genders.
Collapse
Affiliation(s)
| | - Yuichiro Minami
- Department of Cardiology, Tokyo Women's Medical University, Tokyo, Japan
| | - Naoki Sato
- Department of Internal Medicine, Cardiology, and Intensive Care Unit, Nippon Medical School Musashi-Kosugi Hospital, Kanagawa, Japan
| | - Shigeru Otsubo
- Department of Blood Purification, Tohto Sangenjaya Clinic, Tokyo, Japan
| | - Hiroshi Kasanuki
- Faculty of Science and Engineering, Department of Bioscience and Biotechnology, Waseda University, Tokyo, Japan
| |
Collapse
|
17
|
Structure-Function Relationship of the SERCA Pump and Its Regulation by Phospholamban and Sarcolipin. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 981:77-119. [DOI: 10.1007/978-3-319-55858-5_5] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
18
|
Li L, Hou X, Xu R, Liu C, Tu M. Research review on the pharmacological effects of astragaloside IV. Fundam Clin Pharmacol 2016; 31:17-36. [PMID: 27567103 DOI: 10.1111/fcp.12232] [Citation(s) in RCA: 240] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 07/09/2016] [Accepted: 08/26/2016] [Indexed: 12/11/2022]
Abstract
Astragalus membranaceus Bunge has been used to treat numerous diseases for thousands of years. As the main active substance of Astragalus membranaceus Bunge, astragaloside IV (AS-IV) also demonstrates the potent protective effect on focal cerebral ischemia/reperfusion, cardiovascular disease, pulmonary disease, liver fibrosis, and diabetic nephropathy. Based on studies published during the past several decades, the current state of AS-IV research and the pharmacological effects are detailed, elucidated, and summarized. This review systematically summarizes the pharmacological effects, metabolism mechanism, and the toxicity of AS-IV. AS-IV has multiple pharmacologic effects, including anti-inflammatory, antifibrotic, antioxidative stress, anti-asthma, antidiabetes, immunoregulation, and cardioprotective effect via numerous signaling pathways. According to the existing studies and clinical practices, AS-IV possesses potential for broad application in many diseases.
Collapse
Affiliation(s)
- Lei Li
- College of Animal Science, Anhui Science and Technology University, Chuzhou, China
| | - Xiaojiao Hou
- Engineering Research Center of Chinese Traditional Veterinary Medicine, Beijing, China
| | - Rongfang Xu
- Engineering Research Center of Chinese Traditional Veterinary Medicine, Beijing, China
| | - Chang Liu
- College of Animal Science, Anhui Science and Technology University, Chuzhou, China
| | - Menbayaer Tu
- Engineering Research Center of Chinese Traditional Veterinary Medicine, Beijing, China
| |
Collapse
|
19
|
Gong H, Li Y, Wang L, Lv Q, Wang X. Short-term effects of β2-AR blocker ICI 118,551 on sarcoplasmic reticulum SERCA2a and cardiac function of rats with heart failure. Exp Ther Med 2016; 12:1355-1362. [PMID: 27602067 PMCID: PMC4998176 DOI: 10.3892/etm.2016.3450] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 06/13/2016] [Indexed: 01/15/2023] Open
Abstract
The study was conducted to examine the effects of ICI 118,551 on the systolic function of cardiac muscle cells of rats in heart failure and determine the molecular mechanism of selective β2-adrenergic receptor (β2-AR) antagonist on these cells. The chronic heart failure model for rats was prepared through abdominal aortic constriction and separate cardiac muscle cells using the collagenase digestion method. The rats were then divided into Sham, HF and HF+ICI 50 nM goups and cultivated for 48 h. β2-AR, Gi/Gs and sarcoplasmic reticulum Ca2+-ATPase (SERCA2a) protein expression levels in the cardiac muscle cells were evaluated by western blotting and changes in the systolic function of cardiac muscle cells based on the boundary detection system of contraction dynamics for individual cells was measured. The results showed that compared with the Sham group, the survival rate, percentage of basic contraction and maximum contraction amplitude percentage of cardiac muscle cells with heart failure decreased, Gi protein expression increased while Gs and SERCA2a protein expression decreased. Compared with the HF group, the maximum contraction amplitude percentage of cardiac muscle cells in group HF+ICI 50 nM decreased, the Gi protein expression level increased while the SERCA2a protein expression level decreased. Following the stimulation of Ca2+ and ISO, the maximum contraction amplitude percentage of cardiac muscle cells in the HF+ICI 50 nM group was lower than that in group HF. This indicated that ICI 118,551 has negative inotropic effects on cardiac muscle cells with heart failure, which may be related to Gi protein. Systolic function of cardiac muscle cells with heart failure can therefore be reduced by increasing Gi protein expression and lowering SERCA2a protein expression.
Collapse
Affiliation(s)
- Haibin Gong
- Department of Cardiology, Xuzhou Central Hospital, Xuzhou Cardiovascular Disease Institute, Xuzhou, Jiangsu 221009, P.R. China
| | - Yanfei Li
- Department of Cardiology, Xuzhou Central Hospital, Xuzhou Cardiovascular Disease Institute, Xuzhou, Jiangsu 221009, P.R. China
| | - Lei Wang
- Department of Cardiology, Xuzhou Central Hospital, Xuzhou Cardiovascular Disease Institute, Xuzhou, Jiangsu 221009, P.R. China
| | - Qian Lv
- Department of Cardiology, Xuzhou Central Hospital, Xuzhou Cardiovascular Disease Institute, Xuzhou, Jiangsu 221009, P.R. China
| | - Xiuli Wang
- Department of Cardiology, Xuzhou Central Hospital, Xuzhou Cardiovascular Disease Institute, Xuzhou, Jiangsu 221009, P.R. China
| |
Collapse
|
20
|
Schwertz DW, Beck JM, Kowalski JM, Ross JD. Sex Differences in the Response of Rat Heart Ventricle to Calcium. Biol Res Nurs 2016; 5:286-98. [PMID: 15068658 DOI: 10.1177/1099800403262615] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Calcium (Ca2+ ) is a key mediator of myocardial function. Calcium regulates contraction, and disruption of myocellular Ca2+ handling plays a role in cardiac pathologies such as arrhythmias and heart failure. This investigation examines sex differences in sensitivity of the contractile proteins to Ca2+ and myofibrillar Ca2+ delivery in the ventricular myocardium. Sensitivity of contractile proteins to Ca2+ was measured in weight-matched male and female Sprague-Dawley rats using the skinned ventricular papillary muscle fiber and Ca2+ -stimulated Mg2+ -dependent adenosine triphosphatase (ATPase) activity methodologies. Calcium delivery was examined by measuring the contractile response to a range of extracellular Ca2+ concentrations in isolated ventricular myocytes, papillary muscle, and the isolated perfused whole heart. Findings from studies in the whole heart suggest that at a fixed preload, the male left ventricle generates more pressure than a female ventricle over a range of extracellular Ca2+ concentrations. In contrast, results from myocyte and papillary muscle studies suggest that females require less extracellular Ca2+ to elicit a similar contractile response. Results obtained from the 2 methods used to determine sex differences in Ca2+ sensitivity were equivocal. Further studies are required to elucidate sex differences in myocardial Ca2+ handling and the reasons for disparate results in different heart muscle preparations. The results of these studies will lead to the design of sex-optimized therapeutic interventions for cardiac disease.
Collapse
Affiliation(s)
- Dorie W Schwertz
- College of Nursing M/C 802, University of Illinois, at Chicago, 845 South Damen Avenue, Chicago, IL 60612, USA.
| | | | | | | |
Collapse
|
21
|
Nascimento AMD, Lima EMD, Brasil GA, Caliman IF, Silva JFD, Lemos VS, Andrade TUD, Bissoli NS. Serca2a and Na+/Ca2+ exchanger are involved in left ventricular function following cardiac remodelling of female rats treated with anabolic androgenic steroid. Toxicol Appl Pharmacol 2016; 301:22-30. [DOI: 10.1016/j.taap.2016.04.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Revised: 03/16/2016] [Accepted: 04/01/2016] [Indexed: 11/16/2022]
|
22
|
Shuai XX, Meng YD, Lu YX, Su GH, Tao XF, Han J, Xu SD, Luo P. Relaxin-2 improves diastolic function of pressure-overloaded rats via phospholamban by activating Akt. Int J Cardiol 2016; 218:305-311. [PMID: 27240156 DOI: 10.1016/j.ijcard.2016.05.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Revised: 04/18/2016] [Accepted: 05/12/2016] [Indexed: 02/05/2023]
Abstract
BACKGROUND Relaxin is a peptide hormone which has been demonstrated to be safe and has a therapeutic effect on acute heart failure in clinic trials. However, its effect on diastolic function is still unknown. The aims of the study were to determine whether relaxin could improve the diastolic function in pressure-overloaded rat model and to analyze potential mechanisms. METHODS AND RESULTS In the present study, a pressure-overloaded rat model induced by transaortic constriction (TAC) was established. Four weeks after TAC, echocardiography was performed and then all the rat models were randomly divided into 3 groups: models without intramyocardial injection (TAC), with intramyocardial injection of empty adenoviral vector (TAC+GFP) and adenoviral vector overexpression relaxin-2 gene (TAC+RLN2). A sham group was also included. Twelve days after intramyocardial injection, echocardiography and hemodynamics were carried out to evaluate diastolic function in sham, TAC, TAC+GFP and TAC+RLN2 groups. Then hearts were harvested for subsequent examinations. The results indicated that relaxin-2 had ameliorated diastolic function in the pressure-overloaded rats. Compared with the TAC and TAC+GFP groups, the relaxin-2 gene transfer increased phosphorylation of Akt at both the Ser473 and Thr308 sites. Meanwhile, it increased the Ser16 and Thr17- phosphorylation levels of phospholamban (PLB). Furthermore, SERCA2 activity was enhanced in the TAC+RLN2 group more than in the TAC and TAC+GFP groups. CONCLUSIONS These results demonstrated that relaxin-2 gene therapy improved diastolic function in pressure-overloaded rats. The potential mechanism may be that relaxin-2 gene transfer enhances SERCA2 activity in hearts by increasing phospholamban phosphorylation through nuclear-targeted Akt phosphorylation.
Collapse
Affiliation(s)
- Xin-Xin Shuai
- Department of Cardiology, Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yi-di Meng
- Department of Cardiology, Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yong-Xin Lu
- Department of Cardiology, Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Guan-Hua Su
- Department of Cardiology, Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xiao-Fang Tao
- Department of Cardiology, Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jun Han
- Department of Cardiology, Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430022, China
| | - Su-Dan Xu
- Department of Cardiology, Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430022, China
| | - Ping Luo
- Department of Cardiology, Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
23
|
Sato M, Yamanaka H, Iwasaki M, Miyata Y, Kamibayashi T, Fujino Y, Hayashi Y. Altered Phosphatidylinositol 3-Kinase and Calcium Signaling in Cardiac Dysfunction After Brain Death in Rats. Ann Thorac Surg 2016; 102:556-63. [PMID: 27130251 DOI: 10.1016/j.athoracsur.2016.02.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Revised: 01/02/2016] [Accepted: 02/08/2016] [Indexed: 11/28/2022]
Abstract
BACKGROUND Phosphatidylinositol 3-kinase is involved in myocardial function, including contractility. To date, myocardial regulation by phosphatidylinositol 3-kinase after brain death has not been investigated. The present study using a brain death model was designed to examine the role of phosphatidylinositol 3-kinase in myocardial function after brain death. METHODS After anesthesia with sevoflurane, a Fogarty catheter was placed intracranially for induction of brain death. A conductance catheter was inserted into the left ventricle for measurement of myocardial function. Rats were assigned to the following groups: one group undergoing sham operation (with catheter placement but no brain death introduction); one group receiving saline before brain death; and one group receiving wortmannin, an inhibitor of phosphatidylinositol 3-kinase, before brain death. Various measurements, including mean blood pressure, heart rate, maximal rate of rise of left ventricular pressure, and ejection fraction, were obtained every 30 minutes for 6 hours after brain death. The phosphorylation status of Akt and phospholamban was determined 360 minutes after brain death. RESULTS After induction of brain death, rats showed significant decreases in blood pressure, maximal rate of rise of left ventricular pressure, and ejection fraction. Inhibition of phosphatidylinositol 3-kinase using wortmannin significantly improved these measurements, resulting in increased survival. Western blot analysis demonstrated that brain death increased Akt phosphorylation and decreased phospholamban phosphorylation; these effects were abolished by wortmannin. CONCLUSIONS Inhibition of phosphatidylinositol 3-kinase prevented myocardial dysfunction after brain death in association with inhibition of the decrease in phosphorylation of myocardial phospholamban, characteristic of brain death.
Collapse
Affiliation(s)
- Masanori Sato
- Department of Anesthesiology, Osaka University Medical School, Osaka, Japan
| | - Hiroo Yamanaka
- Department of Anesthesia, Kansai Rosai Hospital, Osaka, Japan
| | - Mitsuo Iwasaki
- Department of Anesthesiology, Osaka University Medical School, Osaka, Japan
| | - Yuka Miyata
- Anesthesiology Service, Sakurabashi-Watanabe Hospital, Osaka, Japan
| | | | - Yuji Fujino
- Department of Anesthesiology, Osaka University Medical School, Osaka, Japan
| | - Yukio Hayashi
- Department of Anesthesiology, Osaka University Medical School, Osaka, Japan; Anesthesiology Service, Sakurabashi-Watanabe Hospital, Osaka, Japan.
| |
Collapse
|
24
|
Providência R, Marijon E, Lambiase PD, Bouzeman A, Defaye P, Klug D, Amet D, Perier MC, Gras D, Algalarrondo V, Deharo JC, Leclercq C, Fauchier L, Babuty D, Bordachar P, Sadoul N, Piot O, Boveda S. Primary Prevention Implantable Cardioverter Defibrillator (ICD) Therapy in Women-Data From a Multicenter French Registry. J Am Heart Assoc 2016; 5:e002756. [PMID: 26873687 PMCID: PMC4802475 DOI: 10.1161/jaha.115.002756] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 12/21/2015] [Indexed: 11/24/2022]
Abstract
BACKGROUND There are limited data describing sex specificities regarding implantable cardioverter defibrillators (ICDs) in the real-world European setting. METHODS AND RESULTS Using a large multicenter cohort of consecutive patients referred for ICD implantation for primary prevention (2002-2012), in ischemic and nonischemic cardiomyopathy, we examined the sex differences in subjects' characteristics and outcomes. Of 5539 patients, only 837 (15.1%) were women and 53.8% received cardiac resynchronization therapy. Compared to men, women presented a significantly higher proportion of nonischemic cardiomyopathy (60.2% versus 36.2%, P<0.001), wider QRS complex width (QRS >120 ms: 74.6% versus 68.5%, P=0.003), higher New York Heart Association functional class (≥III in 54.2%♀ versus 47.8%♂, P=0.014), and lower prevalence of atrial fibrillation (18.7% versus 24.9%, P<0.001). During a 16 786 patient-years follow-up, overall, fewer appropriate therapies were observed in women (hazard ratio=0.59, 95% CI 0.45-0.76; P<0.001). By contrast, no sex-specific interaction was observed for inappropriate shocks (odds ratio ♀=0.84, 95% CI 0.50-1.39, P=0.492), early complications (odds ratio=1.00, 95% CI 0.75-1.32, P=0.992), and all-cause mortality (hazard ratio=0.87 95% CI 0.66-1.15, P=0.324). Analysis of sex-by- cardiac resynchronization therapy interaction shows than female cardiac resynchronization therapy recipients experienced fewer appropriate therapies than men (hazard ratio=0.62, 95% CI 0.50-0.77; P<0.001) and lower mortality (hazard ratio=0.68, 95% CI 0.47-0.97; P=0.034). CONCLUSIONS In our real-life registry, women account for the minority of ICD recipients and presented with a different clinical profile. Whereas female cardiac resynchronization therapy recipients had a lower incidence of appropriate ICD therapies and all-cause death than their male counterparts, the observed rates of inappropriate shocks and early complications in all ICD recipients were comparable. CLINICAL TRIAL REGISTRATION URL: https://www.clinicaltrials.gov/. Unique identifier: NCT01992458.
Collapse
Affiliation(s)
- Rui Providência
- Clinique Pasteur, Toulouse, France Barts Heart Centre, Barts Health NHS Trust, London, United Kingdom
| | - Eloi Marijon
- European Georges Pompidou Hospital and Paris Descartes University, Paris, France
| | - Pier D Lambiase
- Barts Heart Centre, Barts Health NHS Trust, London, United Kingdom
| | | | | | | | - Denis Amet
- European Georges Pompidou Hospital and Paris Descartes University, Paris, France
| | | | - Daniel Gras
- Nouvelles Cliniques Nantaises, Nantes, France
| | | | | | | | | | | | | | | | - Olivier Piot
- Centre Cardiologique du Nord, Saint Denis, France
| | | |
Collapse
|
25
|
Haghighi K, Pritchard TJ, Liu GS, Singh VP, Bidwell P, Lam CK, Vafiadaki E, Das P, Ma J, Kunduri S, Sanoudou D, Florea S, Vanderbilt E, Wang HS, Rubinstein J, Hajjar RJ, Kranias EG. Human G109E-inhibitor-1 impairs cardiac function and promotes arrhythmias. J Mol Cell Cardiol 2015; 89:349-59. [PMID: 26455482 PMCID: PMC4689614 DOI: 10.1016/j.yjmcc.2015.10.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Revised: 09/18/2015] [Accepted: 10/04/2015] [Indexed: 01/09/2023]
Abstract
A hallmark of human and experimental heart failure is deficient sarcoplasmic reticulum (SR) Ca-uptake reflecting impaired contractile function. This is at least partially attributed to dephosphorylation of phospholamban by increased protein phosphatase 1 (PP1) activity. Indeed inhibition of PP1 by transgenic overexpression or gene-transfer of constitutively active inhibitor-1 improved Ca-cycling, preserved function and decreased fibrosis in small and large animal models of heart failure, suggesting that inhibitor-1 may represent a potential therapeutic target. We recently identified a novel human polymorphism (G109E) in the inhibitor-1 gene with a frequency of 7% in either normal or heart failure patients. Transgenic mice, harboring cardiac-specific expression of G109E inhibitor-1, exhibited decreases in contractility, Ca-kinetics and SR Ca-load. These depressive effects were relieved by isoproterenol stimulation. Furthermore, stress conditions (2Hz +/- Iso) induced increases in Ca-sparks, Ca-waves (60% of G109E versus 20% in wild types) and after-contractions (76% of G109E versus 23% of wild types) in mutant cardiomyocytes. Similar findings were obtained by acute expression of the G109E variant in adult cardiomyocytes in the absence or presence of endogenous inhibitor-1. The underlying mechanisms included reduced binding of mutant inhibitor-1 to PP1, increased PP1 activity, and dephosphorylation of phospholamban at Ser16 and Thr17. However, phosphorylation of the ryanodine receptor at Ser2808 was not altered while phosphorylation at Ser2814 was increased, consistent with increased activation of Ca/calmodulin-dependent protein kinase II (CaMKII), promoting aberrant SR Ca-release. Parallel in vivo studies revealed that mutant mice developed ventricular ectopy and complex ventricular arrhythmias (including bigeminy, trigeminy and ventricular tachycardia), when challenged with isoproterenol. Inhibition of CaMKII activity by KN-93 prevented the increased propensity to arrhythmias. These findings suggest that the human G109E inhibitor-1 variant impairs SR Ca-cycling and promotes arrhythmogenesis under stress conditions, which may present an additional insult in the compromised function of heart failure carriers.
Collapse
Affiliation(s)
- Kobra Haghighi
- Department of Pharmacology and Cell Biophysics, University of Cincinnati, College of Medicine, Cincinnati, OH 45267, United States
| | - Tracy J Pritchard
- Department of Pharmacology and Cell Biophysics, University of Cincinnati, College of Medicine, Cincinnati, OH 45267, United States
| | - Guan-Sheng Liu
- Department of Pharmacology and Cell Biophysics, University of Cincinnati, College of Medicine, Cincinnati, OH 45267, United States
| | - Vivek P Singh
- Department of Pharmacology and Cell Biophysics, University of Cincinnati, College of Medicine, Cincinnati, OH 45267, United States
| | - Philip Bidwell
- Department of Pharmacology and Cell Biophysics, University of Cincinnati, College of Medicine, Cincinnati, OH 45267, United States
| | - Chi Keung Lam
- Department of Pharmacology and Cell Biophysics, University of Cincinnati, College of Medicine, Cincinnati, OH 45267, United States
| | - Elizabeth Vafiadaki
- Molecular Biology Division, Biomedical Research Foundation, Academy of Athens, Greece
| | - Parthib Das
- Department of Pharmacology and Cell Biophysics, University of Cincinnati, College of Medicine, Cincinnati, OH 45267, United States
| | - Jianyong Ma
- Department of Pharmacology and Cell Biophysics, University of Cincinnati, College of Medicine, Cincinnati, OH 45267, United States
| | - Swati Kunduri
- Department of Pharmacology and Cell Biophysics, University of Cincinnati, College of Medicine, Cincinnati, OH 45267, United States
| | - Despina Sanoudou
- Molecular Biology Division, Biomedical Research Foundation, Academy of Athens, Greece; 4th Department of Internal Medicine, Medical School, University of Athens and Attikon Hospital, Greece
| | - Stela Florea
- Department of Pharmacology and Cell Biophysics, University of Cincinnati, College of Medicine, Cincinnati, OH 45267, United States
| | - Erica Vanderbilt
- Department of Pharmacology and Cell Biophysics, University of Cincinnati, College of Medicine, Cincinnati, OH 45267, United States
| | - Hong-Shang Wang
- Department of Pharmacology and Cell Biophysics, University of Cincinnati, College of Medicine, Cincinnati, OH 45267, United States
| | - Jack Rubinstein
- Division of Cardiology, Internal Medicine, University of Cincinnati College of Medicine, 231 Albert Sabin Way, Cincinnati, OH 45267, United States
| | - Roger J Hajjar
- Cardiovascular Research Center, Ichan School of Medicine at Mount Sinai, One Gustave Levy Place, New York, NY 10029, United States
| | - Evangelia G Kranias
- Department of Pharmacology and Cell Biophysics, University of Cincinnati, College of Medicine, Cincinnati, OH 45267, United States; Molecular Biology Division, Biomedical Research Foundation, Academy of Athens, Greece.
| |
Collapse
|
26
|
TOMPKINS CHRISTINEM, KUTYIFA VALENTINA, ARSHAD AYSHA, MCNITT SCOTT, POLONSKY BRONISLAVA, WANG PAULJ, MOSS ARTHURJ, ZAREBA WOJCIECH. Sex Differences in Device Therapies for Ventricular Arrhythmias or Death in the Multicenter Automatic Defibrillator Implantation Trial With Cardiac Resynchronization Therapy (MADIT-CRT) Trial. J Cardiovasc Electrophysiol 2015; 26:862-871. [DOI: 10.1111/jce.12701] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Revised: 04/24/2015] [Accepted: 04/27/2015] [Indexed: 11/26/2022]
Affiliation(s)
| | - VALENTINA KUTYIFA
- Heart Research Follow-Up Program; University of Rochester Medical Center; Rochester New York USA
| | - AYSHA ARSHAD
- Valley Health System; Columbia University; New York USA
| | - SCOTT MCNITT
- Heart Research Follow-Up Program; University of Rochester Medical Center; Rochester New York USA
| | - BRONISLAVA POLONSKY
- Heart Research Follow-Up Program; University of Rochester Medical Center; Rochester New York USA
| | - PAUL J. WANG
- Stanford University of Medicine; Palo Alto California USA
| | - ARTHUR J. MOSS
- Heart Research Follow-Up Program; University of Rochester Medical Center; Rochester New York USA
| | - WOJCIECH ZAREBA
- Heart Research Follow-Up Program; University of Rochester Medical Center; Rochester New York USA
| |
Collapse
|
27
|
Mehta NK, Abraham WT, Maytin M. ICD and CRT use in ischemic heart disease in women. Curr Atheroscler Rep 2015; 17:512. [PMID: 25921310 DOI: 10.1007/s11883-015-0512-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Although the role of implantable cardioverter defibrillator (ICD) and cardiac resynchronization therapy (CRT) in improving outcomes in ischemic cardiomyopathy (ICM) has been described, the data regarding gender-based survival outcomes are limited. There is a higher preponderance of non-ischemic cardiomyopathy (NICM) in women, and most of the ICM literature is derived from sub-study analysis. This review summarizes the current body of literature on prognosis, pathophysiology, and the present clinical practice for device implantation in women with ICM.
Collapse
Affiliation(s)
- Nishaki Kiran Mehta
- Division of Cardiovascular Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, 43220, USA,
| | | | | |
Collapse
|
28
|
Truszkowska GT, Bilińska ZT, Kosińska J, Śleszycka J, Rydzanicz M, Sobieszczańska-Małek M, Franaszczyk M, Bilińska M, Stawiński P, Michalak E, Małek ŁA, Chmielewski P, Foss-Nieradko B, Machnicki MM, Stokłosa T, Ponińska J, Szumowski Ł, Grzybowski J, Piwoński J, Drygas W, Zieliński T, Płoski R. A study in Polish patients with cardiomyopathy emphasizes pathogenicity of phospholamban (PLN) mutations at amino acid position 9 and low penetrance of heterozygous null PLN mutations. BMC MEDICAL GENETICS 2015; 16:21. [PMID: 25928149 PMCID: PMC4421997 DOI: 10.1186/s12881-015-0167-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Accepted: 03/23/2015] [Indexed: 12/16/2022]
Abstract
Background In humans mutations in the PLN gene, encoding phospholamban - a regulator of sarcoplasmic reticulum calcium ATPase (SERCA), cause cardiomyopathy with prevalence depending on the population. Our purpose was to identify PLN mutations in Polish cardiomyopathy patients. Methods We studied 161 unrelated subjects referred for genetic testing for cardiomyopathies: 135 with dilated cardiomyopathy, 22 with hypertrophic cardiomyopathy and 4 with other cardiomyopathies. In 23 subjects multiple genes were sequenced by next generation sequencing and in all subjects PLN exons were analyzed by Sanger sequencing. Control group included 200 healthy subjects matched with patients for ethnicity, sex and age. Large deletions/insertions were screened by real time polymerase chain reaction. Results We detected three different heterozygous mutations in the PLN gene: a novel null c.9_10insA:(p.Val4Serfs*15) variant and two missense variants: c.25C > T:(p.Arg9Cys) and c.26G > T:(p.Arg9Leu). The (p.Val4Serfs*15) variant occurred in the patient with Wolff-Parkinson-White syndrome in whom the diagnosis of cardiomyopathy was not confirmed and his mother who had concentric left ventricular remodeling but normal left ventricular mass and function. We did not detect large deletions/insertions in PLN in cohort studied. Conclusions In Poland, similar to most populations, PLN mutations rarely cause cardiomyopathy. The 9thPLN residue is apparently a mutation hot spot whereas a single dose of c.9_10insA, and likely other null PLN mutations, cause the disease only with low penetrance or are not pathogenic. Electronic supplementary material The online version of this article (doi:10.1186/s12881-015-0167-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Grażyna T Truszkowska
- Laboratory of Molecular Biology, Institute of Cardiology, ul. Alpejska 42, 04-628, Warszawa, Poland.
| | - Zofia T Bilińska
- Unit for Screening Studies in Inherited Cardiovascular Diseases, Institute of Cardiology, ul. Alpejska 42, 04-628, Warszawa, Poland.
| | - Joanna Kosińska
- Department of Medical Genetics, Warsaw Medical University, ul. Pawińskiego 3C, 02-106, Warszawa, Poland.
| | - Justyna Śleszycka
- Department of Cardiomyopathies, Institute of Cardiology, ul. Alpejska 42, 04-628, Warszawa, Poland.
| | - Małgorzata Rydzanicz
- Department of Medical Genetics, Warsaw Medical University, ul. Pawińskiego 3C, 02-106, Warszawa, Poland.
| | | | - Maria Franaszczyk
- Laboratory of Molecular Biology, Institute of Cardiology, ul. Alpejska 42, 04-628, Warszawa, Poland.
| | - Maria Bilińska
- Department of Arrhythmia, Institute of Cardiology, ul. Alpejska 42, 04-628, Warszawa, Poland.
| | - Piotr Stawiński
- Department of Immunology, Center for Biostructure Research, Medical University of Warsaw, Warszawa, Poland.
| | - Ewa Michalak
- Unit for Screening Studies in Inherited Cardiovascular Diseases, Institute of Cardiology, ul. Alpejska 42, 04-628, Warszawa, Poland.
| | - Łukasz A Małek
- Department of Interventional Cardiology and Angiology, Institute of Cardiology, ul. Alpejska 42, 04-628, Warszawa, Poland.
| | - Przemysław Chmielewski
- Unit for Screening Studies in Inherited Cardiovascular Diseases, Institute of Cardiology, ul. Alpejska 42, 04-628, Warszawa, Poland.
| | - Bogna Foss-Nieradko
- Unit for Screening Studies in Inherited Cardiovascular Diseases, Institute of Cardiology, ul. Alpejska 42, 04-628, Warszawa, Poland.
| | - Marcin M Machnicki
- Department of Immunology, Center for Biostructure Research, Medical University of Warsaw, Warszawa, Poland.
| | - Tomasz Stokłosa
- Department of Immunology, Center for Biostructure Research, Medical University of Warsaw, Warszawa, Poland.
| | - Joanna Ponińska
- Laboratory of Molecular Biology, Institute of Cardiology, ul. Alpejska 42, 04-628, Warszawa, Poland.
| | - Łukasz Szumowski
- Department of Arrhythmia, Institute of Cardiology, ul. Alpejska 42, 04-628, Warszawa, Poland.
| | - Jacek Grzybowski
- Department of Cardiomyopathies, Institute of Cardiology, ul. Alpejska 42, 04-628, Warszawa, Poland.
| | - Jerzy Piwoński
- Department of Epidemiology, Cardiovascular Diseases Prevention and Promotion of Health, Institute of Cardiology, ul. Niemodlińska 33, 04-635, Warszawa, Poland.
| | - Wojciech Drygas
- Department of Epidemiology, Cardiovascular Diseases Prevention and Promotion of Health, Institute of Cardiology, ul. Niemodlińska 33, 04-635, Warszawa, Poland.
| | - Tomasz Zieliński
- Department of Heart Failure and Transplantology, Institute of Cardiology, ul. Alpejska 42, 04-628, Warszawa, Poland.
| | - Rafał Płoski
- Department of Medical Genetics, Warsaw Medical University, ul. Pawińskiego 3C, 02-106, Warszawa, Poland.
| |
Collapse
|
29
|
Nonaka M, Morimoto S, Murayama T, Kurebayashi N, Li L, Wang YY, Arioka M, Yoshihara T, Takahashi-Yanaga F, Sasaguri T. Stage-dependent benefits and risks of pimobendan in mice with genetic dilated cardiomyopathy and progressive heart failure. Br J Pharmacol 2015; 172:2369-82. [PMID: 25560565 DOI: 10.1111/bph.13062] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Revised: 12/16/2014] [Accepted: 12/18/2014] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND AND PURPOSE The Ca(2+) sensitizer pimobendan is a unique inotropic agent that improves cardiac contractility with less of an increase in oxygen consumption and potentially fewer adverse effects on myocardial remodelling and arrhythmia, compared with traditional inotropes. However, clinical trials report contradictory effects of pimobendan in patients with heart failure (HF). We provide mechanistic experimental evidence of the efficacy of pimobendan using a novel mouse model of progressive HF. EXPERIMENTAL APPROACH A knock-in mouse model of human genetic dilated cardiomyopathy, which shows a clear transition from compensatory to end-stage HF at a fixed time during growth, was used to evaluate the efficacy of pimobendan and explore the underlying molecular and cellular mechanisms. KEY RESULTS Pimobendan prevented myocardial remodelling in compensated HF and significantly extended life span in both compensated and end-stage HF, but dose-dependently increased sudden death in end-stage HF. In cardiomyocytes isolated from end-stage HF mice, pimobendan induced triggered activity probably because of early or delayed afterdepolarizations. The L-type Ca(2+) channel blocker verapamil decreased the incidence of triggered activity, suggesting that this was from over-elevated cytoplasmic Ca(2+) through increased Ca(2+) entry by PDE3 inhibition under diminished sarcoplasmic reticulum Ca(2+) reuptake and increased Ca(2+) leakage from sarcoplasmic reticulum in end-stage HF. CONCLUSIONS AND IMPLICATIONS Pimobendan was beneficial regardless of HF stage, but increased sudden cardiac death in end-stage HF with extensive remodelling of Ca(2+) handling. Reduction of cytoplasmic Ca(2+) elevated by PDE3 inhibition might decrease this risk of sudden cardiac death.
Collapse
Affiliation(s)
- Miki Nonaka
- Department of Clinical Pharmacology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Walweel K, Laver DR. Mechanisms of SR calcium release in healthy and failing human hearts. Biophys Rev 2015; 7:33-41. [PMID: 28509976 PMCID: PMC5425750 DOI: 10.1007/s12551-014-0152-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Accepted: 11/25/2014] [Indexed: 01/08/2023] Open
Abstract
Normal heart contraction and rhythm relies on the proper flow of calcium ions (Ca2+) into cardiac cells and between their intracellular organelles, and any disruption can lead to arrhythmia and sudden cardiac death. Electrical excitation of the surface membrane activates voltage-dependent L-type Ca2+ channels to open and allow Ca2+ to enter the cytoplasm. The subsequent increase in cytoplasmic Ca2+ concentration activates calcium release channels (RyR2) located at specialised Ca2+ release sites in the sarcoplasmic reticulum (SR), which serves as an intracellular Ca2+ store. Animal models have provided valuable insights into how intracellular Ca2+ transport mechanisms are altered in human heart failure. The aim of this review is to examine how Ca2+ release sites are remodelled in heart failure and how this affects intracellular Ca2+ transport with an emphasis on Ca2+ release mechanisms in the SR. Current knowledge on how heart failure alters the regulation of RyR2 by Ca2+ and Mg2+ and how these mechanisms control the activity of RyR2 in the confines of the Ca2+ release sites is reviewed.
Collapse
Affiliation(s)
- K Walweel
- School of Biomedical Sciences and Pharmacy, University of Newcastle and Hunter Medical Research Institute, Callaghan, NSW, 2308, Australia
| | - D R Laver
- School of Biomedical Sciences and Pharmacy, University of Newcastle and Hunter Medical Research Institute, Callaghan, NSW, 2308, Australia.
| |
Collapse
|
31
|
RUWALD MARTINH, MOSS ARTHURJ, ZAREBA WOJCIECH, JONS CHRISTIAN, RUWALD ANNECHRISTINE, MCNITT SCOTT, POLONSKY BRONISLAVA, KUTYIFA VALENTINA. Circadian Distribution of Ventricular Tachyarrhythmias and Association with Mortality in the MADIT-CRT Trial. J Cardiovasc Electrophysiol 2015; 26:291-9. [DOI: 10.1111/jce.12592] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Revised: 10/23/2014] [Accepted: 10/30/2014] [Indexed: 11/28/2022]
Affiliation(s)
- MARTIN H. RUWALD
- Heart Research Follow-up Program; University of Rochester Medical Center; Rochester New York USA
- Department of Cardiology; Gentofte Hospital; Hellerup Denmark
| | - ARTHUR J. MOSS
- Heart Research Follow-up Program; University of Rochester Medical Center; Rochester New York USA
| | - WOJCIECH ZAREBA
- Heart Research Follow-up Program; University of Rochester Medical Center; Rochester New York USA
| | - CHRISTIAN JONS
- Department of Cardiology; Gentofte Hospital; Hellerup Denmark
| | - ANNE-CHRISTINE RUWALD
- Heart Research Follow-up Program; University of Rochester Medical Center; Rochester New York USA
- Department of Cardiology; Gentofte Hospital; Hellerup Denmark
| | - SCOTT MCNITT
- Heart Research Follow-up Program; University of Rochester Medical Center; Rochester New York USA
| | - BRONISLAVA POLONSKY
- Heart Research Follow-up Program; University of Rochester Medical Center; Rochester New York USA
| | - VALENTINA KUTYIFA
- Heart Research Follow-up Program; University of Rochester Medical Center; Rochester New York USA
| |
Collapse
|
32
|
Nonaka M, Morimoto S. Experimental models of inherited cardiomyopathy and its therapeutics. World J Cardiol 2014; 6:1245-1251. [PMID: 25548614 PMCID: PMC4278159 DOI: 10.4330/wjc.v6.i12.1245] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Revised: 07/08/2014] [Accepted: 10/16/2014] [Indexed: 02/06/2023] Open
Abstract
Cardiomyopathy is a disease of myocardium categorized into three major forms, hypertrophic (HCM), dilated (DCM) and restrictive cardiomyopathy (RCM), which has recently been demonstrated to be a monogenic disease due to mutations in various proteins expressed in cardiomyocytes. Mutations in HCM and RCM typically increase the myofilament sensitivity to cytoplasmic Ca2+, leading to systolic hyperfunction and diastolic dysfunction. In contrast, mutations in DCM typically decrease the myofilament sensitivity to cytoplasmic Ca2+ and/or force generation/transmission, leading to systolic dysfunction. Creation of genetically-manipulated transgenic and knock-in animals expressing mutant proteins exogenously and endogenously, respectively, in their hearts provides valuable animal models to discover the molecular and cellular mechanisms for pathogenesis and promising therapeutic strategy in vivo. Recently, cardiomyocytes have been differentiated from patient’s induced pluripotent stem cells as a model of inherited cardiomyopathies in vitro. In this review, we provide overview of experimental models of cardiomyopathies with a focus on revealed molecular and cellular pathogenic mechanisms and potential therapeutics.
Collapse
|
33
|
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: 6.7] [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
|
34
|
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: 1.3] [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
|
35
|
Ablorh NAD, Dong X, James ZM, Xiong Q, Zhang J, Thomas DD, Karim CB. Synthetic phosphopeptides enable quantitation of the content and function of the four phosphorylation states of phospholamban in cardiac muscle. J Biol Chem 2014; 289:29397-405. [PMID: 25190804 DOI: 10.1074/jbc.m114.556621] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We have studied the differential effects of phospholamban (PLB) phosphorylation states on the activity of the sarcoplasmic reticulum Ca-ATPase (SERCA). It has been shown that unphosphorylated PLB (U-PLB) inhibits SERCA and that phosphorylation of PLB at Ser-16 or Thr-17 relieves this inhibition in cardiac sarcoplasmic reticulum. However, the levels of the four phosphorylation states of PLB (U-PLB, P16-PLB, P17-PLB, and doubly phosphorylated 2P-PLB) have not been measured quantitatively in cardiac tissue, and their functional effects on SERCA have not been determined directly. We have solved both problems through the chemical synthesis of all four PLB species. We first used the synthetic PLB as standards for a quantitative immunoblot assay, to determine the concentrations of all four PLB phosphorylation states in pig cardiac tissue, with and without left ventricular hypertrophy (LVH) induced by aortic banding. In both LVH and sham hearts, all phosphorylation states were significantly populated, but LVH hearts showed a significant decrease in U-PLB, with a corresponding increase in the ratio of total phosphorylated PLB to U-PLB. To determine directly the functional effects of each PLB species, we co-reconstituted each of the synthetic peptides in phospholipid membranes with SERCA and measured calcium-dependent ATPase activity. SERCA inhibition was maximally relieved by P16-PLB (the most highly populated PLB state in cardiac tissue homogenates), followed by 2P-PLB, then P17-PLB. These results show that each PLB phosphorylation state uniquely alters Ca(2+) homeostasis, with important implications for cardiac health, disease, and therapy.
Collapse
Affiliation(s)
| | - Xiaoqiong Dong
- From the Departments of Biochemistry, Molecular Biology and Biophysics and
| | - Zachary M James
- From the Departments of Biochemistry, Molecular Biology and Biophysics and
| | - Qiang Xiong
- Medicine, University of Minnesota, Minneapolis, Minnesota 55455
| | - Jianyi Zhang
- Medicine, University of Minnesota, Minneapolis, Minnesota 55455
| | - David D Thomas
- From the Departments of Biochemistry, Molecular Biology and Biophysics and
| | - Christine B Karim
- From the Departments of Biochemistry, Molecular Biology and Biophysics and
| |
Collapse
|
36
|
Bennett MK, Sweet WE, Baicker-McKee S, Looney E, Karohl K, Mountis M, Tang WHW, Starling RC, Moravec CS. S100A1 in human heart failure: lack of recovery following left ventricular assist device support. Circ Heart Fail 2014; 7:612-8. [PMID: 24842913 PMCID: PMC4102621 DOI: 10.1161/circheartfailure.113.000849] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND We hypothesized that S100A1 is regulated during human hypertrophy and heart failure and that it may be implicated in remodeling after left ventricular assist device. S100A1 is decreased in animal and human heart failure, and restoration produces functional recovery in animal models and in failing human myocytes. With the potential for gene therapy, it is important to carefully explore human cardiac S100A1 regulation and its role in remodeling. METHODS AND RESULTS We measured S100A1, the sarcoplasmic endoplasmic reticulum Ca(2+)ATPase, phospholamban, and ryanodine receptor proteins, as well as β-adrenergic receptor density in nonfailing, hypertrophied (left ventricular hypertrophy), failing, and failing left ventricular assist device-supported hearts. We determined functional consequences of protein alterations in isolated contracting muscles from the same hearts. S100A1, sarcoplasmic endoplasmic reticulum Ca(2+)ATPase and phospholamban were normal in left ventricular hypertrophy, but decreased in failing hearts, while ryanodine receptor was unchanged in either group. Baseline muscle contraction was not altered in left ventricular hypertrophy or failing hearts. β-Adrenergic receptor and inotropic response were decreased in failing hearts. In failing left ventricular assist device-supported hearts, S100A1 and sarcoplasmic endoplasmic reticulum Ca(2+)ATPase showed no recovery, while phospholamban, β-adrenergic receptor, and the inotropic response fully recovered. CONCLUSIONS S100A1 and sarcoplasmic endoplasmic reticulum Ca(2+)ATPase, both key Ca(2+)-regulatory proteins, are decreased in human heart failure, and these changes are not reversed after left ventricular assist device. The clinical significance of these findings for cardiac recovery remains to be addressed.
Collapse
Affiliation(s)
- Mosi K Bennett
- From the Kaufman Center for Heart Failure, Department of Cardiovascular Medicine, Cleveland Clinic, OH
| | - Wendy E Sweet
- From the Kaufman Center for Heart Failure, Department of Cardiovascular Medicine, Cleveland Clinic, OH
| | - Sara Baicker-McKee
- From the Kaufman Center for Heart Failure, Department of Cardiovascular Medicine, Cleveland Clinic, OH
| | - Elizabeth Looney
- From the Kaufman Center for Heart Failure, Department of Cardiovascular Medicine, Cleveland Clinic, OH
| | - Kristen Karohl
- From the Kaufman Center for Heart Failure, Department of Cardiovascular Medicine, Cleveland Clinic, OH
| | - Maria Mountis
- From the Kaufman Center for Heart Failure, Department of Cardiovascular Medicine, Cleveland Clinic, OH
| | - W H Wilson Tang
- From the Kaufman Center for Heart Failure, Department of Cardiovascular Medicine, Cleveland Clinic, OH
| | - Randall C Starling
- From the Kaufman Center for Heart Failure, Department of Cardiovascular Medicine, Cleveland Clinic, OH
| | - Christine S Moravec
- From the Kaufman Center for Heart Failure, Department of Cardiovascular Medicine, Cleveland Clinic, OH.
| |
Collapse
|
37
|
Pritchard TJ, Kawase Y, Haghighi K, Anjak A, Cai W, Jiang M, Nicolaou P, Pylar G, Karakikes I, Rapti K, Rubinstein J, Hajjar RJ, Kranias EG. Active inhibitor-1 maintains protein hyper-phosphorylation in aging hearts and halts remodeling in failing hearts. PLoS One 2013; 8:e80717. [PMID: 24312496 PMCID: PMC3846572 DOI: 10.1371/journal.pone.0080717] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Accepted: 10/16/2013] [Indexed: 01/14/2023] Open
Abstract
Impaired sarcoplasmic reticulum calcium cycling and depressed contractility are key characteristics in heart failure. Defects in sarcoplasmic reticulum function are characterized by decreased SERCA2a Ca-transport that is partially attributable to dephosphorylation of its regulator phospholamban by increased protein phosphatase 1 activity. Inhibition of protein phosphatase 1 through activation of its endogenous inhibitor-1 has been shown to enhance cardiac Ca-handling and contractility as well as protect from pathological stress remodeling in young mice. In this study, we assessed the long-term effects of inducible expression of constitutively active inhibitor-1 in the adult heart and followed function and remodeling through the aging process, up to 20 months. Mice with inhibitor-1 had normal survival and similar function to WTs. There was no overt remodeling as evidenced by measures of left ventricular end-systolic and diastolic diameters and posterior wall dimensions, heart weight to tibia length ratio, and histology. Higher phosphorylation of phospholamban at both Ser16 and Thr17 was maintained in aged hearts with active inhibitor-1, potentially offsetting the effects of elevated Ser2815-phosphorylation in ryanodine receptor, as there were no increases in arrhythmias under stress conditions in 20-month old mice. Furthermore, long-term expression of active inhibitor-1 via recombinant adeno-associated virus type 9 gene transfer in rats with pressure-overload induced heart failure improved function and prevented remodeling, associated with increased phosphorylation of phospholamban at Ser16 and Thr17. Thus, chronic inhibition of protein phosphatase 1, through increases in active inhibitor-1, does not accelerate age-related cardiomyopathy and gene transfer of this molecule in vivo improves function and halts remodeling in the long term.
Collapse
Affiliation(s)
- Tracy J. Pritchard
- Department of Pharmacology and Cell Biophysics, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Yoshiaki Kawase
- Cardiovascular Research Center, Ichan School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Kobra Haghighi
- Department of Pharmacology and Cell Biophysics, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Ahmad Anjak
- Department of Internal Medicine, Division of Cardiovascular Diseases, University of Cincinnati, Cincinnati, Ohio, United States of America
| | - Wenfeng Cai
- Department of Pharmacology and Cell Biophysics, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Min Jiang
- Department of Internal Medicine, Division of Cardiovascular Diseases, University of Cincinnati, Cincinnati, Ohio, United States of America
| | - Persoulla Nicolaou
- Department of Pharmacology and Cell Biophysics, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - George Pylar
- Department of Pharmacology and Cell Biophysics, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Ioannis Karakikes
- Cardiovascular Research Center, Ichan School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Kleopatra Rapti
- Cardiovascular Research Center, Ichan School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Jack Rubinstein
- Department of Internal Medicine, Division of Cardiovascular Diseases, University of Cincinnati, Cincinnati, Ohio, United States of America
| | - Roger J. Hajjar
- Cardiovascular Research Center, Ichan School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Evangelia G. Kranias
- Department of Pharmacology and Cell Biophysics, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
- * E-mail:
| |
Collapse
|
38
|
Coppini R, Ferrantini C, Mazzoni L, Sartiani L, Olivotto I, Poggesi C, Cerbai E, Mugelli A. Regulation of intracellular Na(+) in health and disease: pathophysiological mechanisms and implications for treatment. Glob Cardiol Sci Pract 2013; 2013:222-42. [PMID: 24689024 PMCID: PMC3963757 DOI: 10.5339/gcsp.2013.30] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Accepted: 09/01/2013] [Indexed: 12/19/2022] Open
Abstract
Transmembrane sodium (Na+) fluxes and intracellular sodium homeostasis are central players in the physiology of the cardiac myocyte, since they are crucial for both cell excitability and for the regulation of the intracellular calcium concentration. Furthermore, Na+ fluxes across the membrane of mitochondria affect the concentration of protons and calcium in the matrix, regulating mitochondrial function. In this review we first analyze the main molecular determinants of sodium fluxes across the sarcolemma and the mitochondrial membrane and describe their role in the physiology of the healthy myocyte. In particular we focus on the interplay between intracellular Ca2+ and Na+. A large part of the review is dedicated to discuss the changes of Na+ fluxes and intracellular Na+ concentration([Na+]i) occurring in cardiac disease; we specifically focus on heart failure and hypertrophic cardiomyopathy, where increased intracellular [Na+]i is an established determinant of myocardial dysfunction. We review experimental evidence attributing the increase of [Na+]i to either decreased Na+ efflux (e.g. via the Na+/K+ pump) or increased Na+ influx into the myocyte (e.g. via Na+ channels). In particular, we focus on the role of the “late sodium current” (INaL), a sustained component of the fast Na+ current of cardiac myocytes, which is abnormally enhanced in cardiac diseases and contributes to both electrical and contractile dysfunction. We analyze the pathophysiological role of INaL enhancement in heart failure and hypertrophic cardiomyopathy and the consequences of its pharmacological modulation, highlighting the clinical implications. The central role of Na+ fluxes and intracellular Na+ physiology and pathophysiology of cardiac myocytes has been highlighted by a large number of recent works. The possibility of modulating Na+ inward fluxes and [Na+]i with specific INaL inhibitors, such as ranolazine, has made Na+a novel suitable target for cardiac therapy, potentially capable of addressing arrhythmogenesis and diastolic dysfunction in severe conditions such as heart failure and hypertrophic cardiomyopathy.
Collapse
Affiliation(s)
- Raffaele Coppini
- Department NeuroFarBa, Division of Pharmacology, University of Florence, Italy
| | - Cecilia Ferrantini
- Department of Clinical and Experimental Medicine, division of Physiology, University of Florence, Italy
| | - Luca Mazzoni
- Department NeuroFarBa, Division of Pharmacology, University of Florence, Italy
| | - Laura Sartiani
- Department NeuroFarBa, Division of Pharmacology, University of Florence, Italy
| | - Iacopo Olivotto
- Referral Center for Cardiomyopathies, Careggi University Hospital, Florence, Italy
| | - Corrado Poggesi
- Department of Clinical and Experimental Medicine, division of Physiology, University of Florence, Italy
| | - Elisabetta Cerbai
- Department NeuroFarBa, Division of Pharmacology, University of Florence, Italy
| | - Alessandro Mugelli
- Department NeuroFarBa, Division of Pharmacology, University of Florence, Italy
| |
Collapse
|
39
|
Kwon DH, Hachamovitch R, Adeniyi A, Nutter B, Popovic ZB, Wilkoff BL, Desai MY, Flamm SD, Marwick T. Myocardial scar burden predicts survival benefit with implantable cardioverter defibrillator implantation in patients with severe ischaemic cardiomyopathy: influence of gender. Heart 2013; 100:206-13. [PMID: 24186562 PMCID: PMC3913110 DOI: 10.1136/heartjnl-2013-304261] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Objective We sought to assess the impact of myocardial scar burden (MSB) on the association between implantable cardioverter defibrillator (ICD) implantation and mortality in patients with ischaemic cardiomyopathy (ICM) and left ventricular EF ≤40%. In addition, we sought to determine the impact of gender on survival benefit with ICD implantation. Design Retrospective observational study. Setting Single US tertiary care centre. Patients Consecutive patients with significant ICM who underwent delayed hyperenhancement-MRI between 2002 and 2006. Interventions ICD implantation. Main outcome measures All-cause mortality and cardiac transplantation. Results Follow-up of 450 consecutive patients, over a mean of 5.8 years, identified 186 deaths. Cox proportional hazard modelling was used to evaluate associations among MSB, gender and ICD with respect to all-cause death as the primary endpoint. ICDs were implanted in 163 (36%) patients. On multivariable analysis, Scar% (χ2 28.21, p<0.001), Gender (χ2 12.39, p=0.015) and ICD (χ2 9.57, p=0.022) were independent predictors of mortality after adjusting for multiple parameters. An interaction between MSB×ICD (χ2 9.47, p=0.009) demonstrated significant differential survival with ICD based on MSB severity. Additionally, Scar%×ICD×Gender (χ2 6.18, p=0.048) suggested that men with larger MSB had significant survival benefit with ICD, but men with smaller MSB derived limited benefit with ICD implantation. However, the inverse relationship was found in women. Conclusions MSB is a powerful independent predictor of mortality in patients with and without ICD implantation. In addition, MSB may predict gender-based significant differences in survival benefit from ICDs in patients with severe ICM.
Collapse
Affiliation(s)
- Deborah H Kwon
- Heart and Vascular Institute, Cleveland Clinic, , Cleveland, Ohio, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Sivakumaran V, Stanley BA, Tocchetti CG, Ballin JD, Caceres V, Zhou L, Keceli G, Rainer PP, Lee DI, Huke S, Ziolo MT, Kranias EG, Toscano JP, Wilson GM, O'Rourke B, Kass DA, Mahaney JE, Paolocci N. HNO enhances SERCA2a activity and cardiomyocyte function by promoting redox-dependent phospholamban oligomerization. Antioxid Redox Signal 2013; 19:1185-97. [PMID: 23919584 PMCID: PMC3785857 DOI: 10.1089/ars.2012.5057] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
AIMS Nitroxyl (HNO) interacts with thiols to act as a redox-sensitive modulator of protein function. It enhances sarcoplasmic reticular Ca(2+) uptake and myofilament Ca(2+) sensitivity, improving cardiac contractility. This activity has led to clinical testing of HNO donors for heart failure. Here we tested whether HNO alters the inhibitory interaction between phospholamban (PLN) and the sarcoplasmic reticulum Ca(2+)-ATPase (SERCA2a) in a redox-dependent manner, improving Ca(2+) handling in isolated myocytes/hearts. RESULTS Ventriculocytes, sarcoplasmic reticulum (SR) vesicles, and whole hearts were isolated from control (wildtype [WT]) or PLN knockout (pln(-/-)) mice. Compared to WT, pln(-/-) myocytes displayed enhanced resting sarcomere shortening, peak Ca(2+) transient, and blunted β-adrenergic responsiveness. HNO stimulated shortening, relaxation, and Ca(2+) transient in WT cardiomyocytes, and evoked positive inotropy/lusitropy in intact hearts. These changes were markedly blunted in pln(-/-) cells/hearts. HNO enhanced SR Ca(2+) uptake in WT but not pln(-/-) SR-vesicles. Spectroscopic studies in insect cell microsomes expressing SERCA2a±PLN showed that HNO increased Ca(2+)-dependent SERCA2a conformational flexibility but only when PLN was present. In cardiomyocytes, HNO achieved this effect by stabilizing PLN in an oligomeric disulfide bond-dependent configuration, decreasing the amount of free inhibitory monomeric PLN available. INNOVATION HNO-dependent redox changes in myocyte PLN oligomerization relieve PLN inhibition of SERCA2a. CONCLUSIONS PLN plays a central role in HNO-induced enhancement of SERCA2a activity, leading to increased inotropy/lusitropy in intact myocytes and hearts. PLN remains physically associated with SERCA2a; however, less monomeric PLN is available resulting in decreased inhibition of the enzyme. These findings offer new avenues to improve Ca(2+) handling in failing hearts.
Collapse
Affiliation(s)
- Vidhya Sivakumaran
- 1 Division of Cardiology, Johns Hopkins Medical Institutions , Baltimore, Maryland
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Myofilament Ca2+ desensitization mediates positive lusitropic effect of neuronal nitric oxide synthase in left ventricular myocytes from murine hypertensive heart. J Mol Cell Cardiol 2013; 60:107-15. [DOI: 10.1016/j.yjmcc.2013.04.017] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2012] [Revised: 04/13/2013] [Accepted: 04/15/2013] [Indexed: 11/23/2022]
|
42
|
van der Heijden AC, Thijssen J, Borleffs CJW, van Rees JB, Höke U, van der Velde ET, van Erven L, Schalij MJ. Gender-specific differences in clinical outcome of primary prevention implantable cardioverter defibrillator recipients. Heart 2013; 99:1244-9. [PMID: 23723448 DOI: 10.1136/heartjnl-2013-304013] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
OBJECTIVE To assess differences in clinical outcome of implantable cardioverter-defibrillator (ICD) treatment in men and women. DESIGN Prospective cohort study. SETTING University Medical Center. PATIENTS 1946 primary prevention ICD recipients (1528 (79%) men and 418 (21%) women). Patients with congenital heart disease were excluded for this analysis. MAIN OUTCOME MEASURES All-cause mortality, ICD therapy (antitachycardia pacing and shock) and ICD shock. RESULTS During a median follow-up of 3.3 years (25th-75th percentile 1.4-5.4), 387 (25%) men and 76 (18%) women died. The estimated 5-year cumulative incidence for all-cause mortality was 20% (95% CI 18% to 23%) for men and 14% (95% CI 9% to 19%) for women (log rank p<0.01). After adjustment for potential confounding covariates all-cause mortality was lower in women (HR 0.65; 95% CI 0.49 to 0.84; p<0.01). The 5-year cumulative incidence for appropriate therapy in men was 24% (95% CI 21% to 28%) as compared with 20% (95% CI 14% to 26%) in women (log rank p=0.07). After adjustment, a non-significant trend remained (HR 0.82; 95% CI 0.64 to 1.06; p=0.13). CONCLUSIONS In clinical practice, 21% of primary prevention ICD recipients are women. Women have lower mortality and tend to experience less appropriate ICD therapy as compared with their male peers.
Collapse
|
43
|
Witayavanitkul N, Woranush W, Bupha-Intr T, Wattanapermpool J. Testosterone regulates cardiac contractile activation by modulating SERCA but not NCX activity. Am J Physiol Heart Circ Physiol 2013. [DOI: 10.1152/ajpheart.00555.2012] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Alterations in intracellular Ca2+ transients of cardiomyocytes in orchidectomized (ORX) rats could be a cause of cardiac dysfunction in the hypogonadal condition. To investigate the role of male sex hormones in intracellular Ca2+ homeostasis during relaxation, Ca2+-handling activities by sarco(endo)plasmic reticulum Ca2+-ATPase (SERCA) and the Na+/Ca2+ exchanger (NCX) were evaluated in the ventricular muscle of 10-wk-old ORX rats with and without testosterone supplementation (2.5 mg/kg testosterone propionate, 2 times/wk). ORX induced a 50% decrease in contraction force accompanied by a prolonged time to achieve 50% relaxation ( T50) in isolated intact ventricular trabeculae, which was partially corrected by testosterone administration. Maximum active tension was also suppressed in ORX rats without changes in myofilament Ca2+ sensitivity and passive stiffness of the heart. Using a sarcoplasmic reticulum-enriched membrane preparation, the maximum thapsigargin-sensitive SERCA activity of the ORX rat was 27% lower with an increased Ca2+ sensitivity, which was prevented by testosterone treatment. However, neither changes in SERCA content nor its modulating components, sarcolipin and heat shock protein 20, were detected in the ORX rat, but there was a significant decrease in the phosphorylated Thr17 form of phospholamban. Despite a lower level of NCX protein in the heart of ORX rats, prolonged T50 disappeared after an incubation with thapsigargin (10 μM), implying a lack of effect of male sex hormone deficiency on NCX function. These findings indicate that male sex hormones can regulate cardiac relaxation by acting mainly through SERCA. However, a detailed mechanism of SERCA modulation under male sex hormone deficiency status remains to be explored.
Collapse
Affiliation(s)
| | - Warunya Woranush
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Tepmanas Bupha-Intr
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | | |
Collapse
|
44
|
Abstract
Although cardiac arrhythmia had long been considered a predominantly male syndrome, it is now clear that arrhythmia is also a primary cause of mortality in women. Notably, the manifestation of specific arrhythmia syndromes appears to be gender specific. In particular, female sex is an independent risk factor for development of torsade de pointes (TdP) arrhythmias not only in congenital long QT syndromes but also in acquired long QT syndromes which occur as adverse effects of existing drugs. Males, on the other hand, are more likely to develop Brugada syndrome. Recent clinical and experimental studies suggest that these differences may stem from intrinsic sex differences in cardiac tissue. These include fundamental electrical differences resulting from variable ion channel expression and diverse sex hormonal regulation via long-term genomic and acute nongenomic pathways, and sex differences in drug responses and metabolisms. Undoubtedly, determining the effect of gender on cardiac function will be difficult and require sophisticated methodologies. However, gender differences underlying predilection to distinct arrhythmia syndromes must be revealed so that new therapeutic strategies that take gender into account can be applied to at-risk patients.
Collapse
Affiliation(s)
- Junko Kurokawa
- Department of Bio-Informational Pharmacology, Tokyo Medical and Dental University, Tokyo, Japan.
| | | | | | | |
Collapse
|
45
|
Rho RW, Patton KK, Poole JE, Cleland JG, Shadman R, Anand I, Maggioni AP, Carson PE, Swedberg K, Levy WC. Important differences in mode of death between men and women with heart failure who would qualify for a primary prevention implantable cardioverter-defibrillator. Circulation 2012; 126:2402-7. [PMID: 23072904 DOI: 10.1161/circulationaha.111.069245] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Whether sex differences in implantable cardioverter-defibrillator (ICD) benefit exist remains unanswered. We evaluated sex differences in mode of death among a large cohort of ambulatory heart failure patients who meet criteria for a primary prevention ICD. METHODS AND RESULTS Patients from 5 trials or registries were included if they met American College of Cardiology/American Heart Association/Heart Rhythm Society guideline criteria for implantation of a primary prevention ICD. We investigated the potential sex differences in total deaths and total deaths by mode of death. The relationship between the estimated total mortality and mode of death by percentage of total mortality was also analyzed by sex. The Seattle Heart Failure Model was used to estimate total mortality in this analysis. A total of 8337 patients (1685 [20%] women) met inclusion criteria. One-year mortality was 10.8±0.3%. In women, the age-adjusted all-cause mortality was 24% lower (hazard ratio [HR], 0.76; confidence interval [CI], 0.68-0.85; P<0.0001), the risk of sudden death was 31% lower (HR, 0.69; CI, 0.58-0.83; P<0.0001), but no significant difference in pump failure death was observed. Throughout a range of total mortality risk, women had a 20% lower all-cause mortality (HR, 0.80; CI, 0.71-0.89; P<0.001) and 29% fewer deaths that were sudden (HR, 0.71; CI, 0.59-0.86;P<0.001) compared with men. CONCLUSIONS Women with heart failure have a lower mortality than men, and fewer of those deaths are sudden throughout a spectrum of all-cause mortality risk. These data provide a plausible reason for and thus support the possibility that sex differences in ICD benefit may exist.
Collapse
Affiliation(s)
- Robert W Rho
- Division of Cardiology, University of Washington, Seattle, WA 98195, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Ling LH, Khammy O, Byrne M, Amirahmadi F, Foster A, Li G, Zhang L, dos Remedios C, Chen C, Kaye DM. Irregular rhythm adversely influences calcium handling in ventricular myocardium: implications for the interaction between heart failure and atrial fibrillation. Circ Heart Fail 2012; 5:786-93. [PMID: 23014130 DOI: 10.1161/circheartfailure.112.968321] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
BACKGROUND Despite adequate rate control, the combination of atrial fibrillation with heart failure (HF) has been shown, in a number of studies, to hasten HF progression. In this context, we aimed to test the hypothesis that an irregular ventricular rhythm causes an alteration in ventricular cardiomyocyte excitation-contraction coupling which contributes to the progression of HF. METHODS AND RESULTS We investigated the effects of electrical field stimulation (average frequency 2 Hz) in an irregular versus regular drive train pattern on the expression of calcium-handling genes and proteins in rat ventricular myocytes. The effect of rhythm on intracellular calcium transients was examined using Fura-2AM fluorescence spectroscopy. In conjunction, calcium-handling protein expression was examined in left ventricular samples obtained from end-stage HF patients, in patients with either persistent atrial fibrillation or sinus rhythm. Compared with regularly paced ventricular cardiomyocytes, in cells paced irregularly for 24 hours, there was a significant reduction in the expression of sarcoplasmic reticulum calcium (Ca(2+)) ATPase together with reduced serine-16 phosphorylation of phospholamban. These findings were accompanied by a 59% reduction (P<0.01) in the peak Ca2+ transient in irregulary paced myocytes compared with those with regular pacing. Consistent with these observations, we observed a 54% (P<0.05) decrease in sarcoplasmic reticulum Ca(2+)ATPase protein expression and an 85% (P<0.01) reduction in the extent of phosphorylation of phospholamban in the left ventricular myocardium of HF patients in atrial fibrillation compared with those in sinus rhythm. CONCLUSIONS Together, these data demonstrate that ventricular rhythmicity contributes significantly to excitation-contraction coupling by altering the expression and activity of key calcium-handling proteins. These data suggest that control of rhythm may be of benefit in patients with HF.
Collapse
Affiliation(s)
- Liang-han Ling
- Heart Failure Research Group, Baker IDI Heart and Diabetes Institute, Melbourne, Australia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Li L, Morimoto S, Take S, Zhan DY, Du CK, Wang YY, Fan XL, Yoshihara T, Takahashi-Yanaga F, Katafuchi T, Sasaguri T. Role of brain serotonin dysfunction in the pathophysiology of congestive heart failure. J Mol Cell Cardiol 2012; 53:760-7. [PMID: 22921782 DOI: 10.1016/j.yjmcc.2012.08.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2012] [Revised: 08/01/2012] [Accepted: 08/10/2012] [Indexed: 12/17/2022]
Abstract
Inherited or non-inherited dilated cardiomyopathy (DCM) patients develop varied disease phenotypes leading to death after developing congestive heart failure (HF) or sudden death with mild or no overt HF symptoms, suggesting that environmental and/or genetic factors may modify the disease phenotype of DCM. In this study, we sought to explore unknown genetic factors affecting the disease phenotype of monogenic inherited human DCM. Knock-in mice bearing a sarcomeric protein mutation that causes DCM were created on different genetic backgrounds; BALB/c and C57Bl/6. DCM mice on the BALB/c background showed cardiac enlargement and systolic dysfunction and developed congestive HF before died. In contrast, DCM mice on the C57Bl/6 background developed no overt HF symptoms and died suddenly, although they showed considerable cardiac enlargement and systolic dysfunction. BALB/c mice have brain serotonin dysfunction due to a single nucleotide polymorphism (SNP) in tryptophan hydroxylase 2 (TPH2). Brain serotonin dysfunction plays a critical role in depression and anxiety and BALB/c mice exhibit depression- and anxiety-related behaviors. Since depression is common and associated with poor prognosis in HF patients, we examined therapeutic effects of anti-depression drug paroxetine and anti-anxiety drug buspirone that could improve the brain serotonin function in mice. Both drugs reduced cardiac enlargement and improved systolic dysfunction and symptoms of severe congestive HF in DCM mice on the BALB/c background. These results strongly suggest that genetic backgrounds involving brain serotonin dysfunction, such as TPH2 gene SNP, may play an important role in the development of congestive HF in DCM.
Collapse
Affiliation(s)
- Lei Li
- Department of Clinical Pharmacology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Abstract
Heart disease remains the leading cause of death and disability in the Western world. Current therapies aim at treating the symptoms rather than the subcellular mechanisms, underlying the etiology and pathological remodeling in heart failure. A universal characteristic, contributing to the decreased contractile performance in human and experimental failing hearts, is impaired calcium sequestration into the sarcoplasmic reticulum (SR). SR calcium uptake is mediated by a Ca(2+)-ATPase (SERCA2), whose activity is reversibly regulated by phospholamban (PLN). Dephosphorylated PLN is an inhibitor of SERCA and phosphorylation of PLN relieves this inhibition. However, the initial simple view of a PLN/SERCA regulatory complex has been modified by our recent identification of SUMO, S100 and the histidine-rich Ca-binding protein as regulators of SERCA activity. In addition, PLN activity is regulated by 2 phosphoproteins, the inhibitor-1 of protein phosphatase 1 and the small heat shock protein 20, which affect the overall SERCA-mediated Ca-transport. This review will highlight the regulatory mechanisms of cardiac contractility by the multimeric SERCA/PLN-ensemble and the potential for new therapeutic avenues targeting this complex by using small molecules and gene transfer methods.
Collapse
Affiliation(s)
- Evangelia G Kranias
- Department of Pharmacology and Cell Biophysics, College of Medicine, University of Cincinnati, 231 Albert Sabin Way, Cincinnati, OH 45267-0575, USA.
| | | |
Collapse
|
49
|
Chen Y, Zhao J, Du J, Xu G, Tang C, Geng B. Hydrogen sulfide regulates cardiac sarcoplasmic reticulum Ca(2+) uptake via K(ATP) channel and PI3K/Akt pathway. Life Sci 2012; 91:271-8. [PMID: 22884808 DOI: 10.1016/j.lfs.2012.07.026] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2012] [Revised: 07/05/2012] [Accepted: 07/13/2012] [Indexed: 12/18/2022]
Abstract
AIMS To investigate the effects of hydrogen sulfide (H(2)S) on calcium uptake activity of the rat cardiac sarcoplasmic reticulum (SR) and possible signaling. MAIN METHODS Crude SR was isolated after treatment with H(2)S, then SR Ca(2+) uptake and SR Ca(2+)-ATPase (SERCA) activity was measured by the isotopic tracer method. The possible roles of the K(ATP) channel and PI3K/Akt and SR-membrane protein phospholamban (PLB) pathway were analyzed by specific blockers, and target protein activation was assayed by measuring protein phosphorylation. KEY FINDINGS Exogenous H(2)S lowered Ca(2+) uptake into the SR time or concentration dependently, which was associated with decreased SERCA activity. Inhibiting endogenous H(2)S production by DL-propargylglycine increased SR Ca(2+) uptake and SERCA activity. H(2)S inhibition of PLB phosphorylation was through SERCA activity and was reversed by two PI3K inhibitors, wortmannin and LY294002. Glibenclamide (a K(ATP) channel blocker) blocked the inhibitory effects of H(2)S on PLB and Akt phosphorylation. Pinacidil (a K(ATP) channel opener) reduced the phosphorylation of PLB and reversed the effects of DL-propargylglycine. H(2)S preconditioning increased PLB phosphorylation but did not affect SERCA activity. SIGNIFICANCE Endogenous H(2)S transiently and reversibly inhibits SR Ca(2+) uptake in rat heart SR because of downregulated SERCA activity associated with PLB phosphorylation by the PI3K/Akt or K(ATP) channel. The transient negative regulation of SR Ca(2+) uptake and the L-type Ca(2+) channel contributes to Ca(2+) cycle homeostasis, which might be an important molecular mechanism in ischemic diseases.
Collapse
Affiliation(s)
- Yu Chen
- Department of Physiology and Pathophysiology, School of Basic Medical Science, Peking University, PR China
| | | | | | | | | | | |
Collapse
|
50
|
Abstract
Electrical storm (ES), characterized by recurrent ventricular tachycardia/fibrillation, is a serious condition, adversely affecting prognosis in patients with implantable cardioverter/defibrillators. Electrical storm patients often die of progressive heart failure, but the underlying molecular basis is poorly understood. We have recently created an animal model of ES that features repetitive implantable cardioverter/defibrillator firing for recurrent ventricular fibrillation and found that ES events cause striking activation of Ca(2+)/calmodulin-dependent protein kinase II and prominent alteration of Ca(2+)-handling protein phosphorylation, possibly explaining mechanical dysfunction and arrhythmia promotion that characterize ES. Here, the pathophysiology and potential therapeutic strategies for ES, based on experimental and clinical studies by us and others, are described.
Collapse
|