1
|
Alibhai FJ, Li RK. Rejuvenation of the Aging Heart: Molecular Determinants and Applications. Can J Cardiol 2024; 40:1394-1411. [PMID: 38460612 DOI: 10.1016/j.cjca.2024.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 02/20/2024] [Accepted: 03/04/2024] [Indexed: 03/11/2024] Open
Abstract
In Canada and worldwide, the elderly population (ie, individuals > 65 years of age) is increasing disproportionately relative to the total population. This is expected to have a substantial impact on the health care system, as increased aged is associated with a greater incidence of chronic noncommunicable diseases. Within the elderly population, cardiovascular disease is a leading cause of death, therefore developing therapies that can prevent or slow disease progression in this group is highly desirable. Historically, aging research has focused on the development of anti-aging therapies that are implemented early in life and slow the age-dependent decline in cell and organ function. However, accumulating evidence supports that late-in-life therapies can also benefit the aged cardiovascular system by limiting age-dependent functional decline. Moreover, recent studies have demonstrated that rejuvenation (ie, reverting cellular function to that of a younger phenotype) of the already aged cardiovascular system is possible, opening new avenues to develop therapies for older individuals. In this review, we first provide an overview of the functional changes that occur in the cardiomyocyte with aging and how this contributes to the age-dependent decline in heart function. We then discuss the various anti-aging and rejuvenation strategies that have been pursued to improve the function of the aged cardiomyocyte, with a focus on therapies implemented late in life. These strategies include 1) established systemic approaches (caloric restriction, exercise), 2) pharmacologic approaches (mTOR, AMPK, SIRT1, and autophagy-targeting molecules), and 3) emerging rejuvenation approaches (partial reprogramming, parabiosis/modulation of circulating factors, targeting endogenous stem cell populations, and senotherapeutics). Collectively, these studies demonstrate the exciting potential and limitations of current rejuvenation strategies and highlight future areas of investigation that will contribute to the development of rejuvenation therapies for the aged heart.
Collapse
Affiliation(s)
- Faisal J Alibhai
- Toronto General Research Hospital Institute, University Health Network, Toronto, Ontario, Canada
| | - Ren-Ke Li
- Toronto General Research Hospital Institute, University Health Network, Toronto, Ontario, Canada; Department of Surgery, Division of Cardiovascular Surgery, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
2
|
Hastings MH, Castro C, Freeman R, Abdul Kadir A, Lerchenmüller C, Li H, Rhee J, Roh JD, Roh K, Singh AP, Wu C, Xia P, Zhou Q, Xiao J, Rosenzweig A. Intrinsic and Extrinsic Contributors to the Cardiac Benefits of Exercise. JACC Basic Transl Sci 2024; 9:535-552. [PMID: 38680954 PMCID: PMC11055208 DOI: 10.1016/j.jacbts.2023.07.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 07/06/2023] [Accepted: 07/20/2023] [Indexed: 05/01/2024]
Abstract
Among its many cardiovascular benefits, exercise training improves heart function and protects the heart against age-related decline, pathological stress, and injury. Here, we focus on cardiac benefits with an emphasis on more recent updates to our understanding. While the cardiomyocyte continues to play a central role as both a target and effector of exercise's benefits, there is a growing recognition of the important roles of other, noncardiomyocyte lineages and pathways, including some that lie outside the heart itself. We review what is known about mediators of exercise's benefits-both those intrinsic to the heart (at the level of cardiomyocytes, fibroblasts, or vascular cells) and those that are systemic (including metabolism, inflammation, the microbiome, and aging)-highlighting what is known about the molecular mechanisms responsible.
Collapse
Affiliation(s)
- Margaret H. Hastings
- Institute for Heart and Brain Health, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Claire Castro
- Cardiovascular Research Center, Division of Cardiology, Corrigan Minehan Heart Center, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Rebecca Freeman
- Cardiovascular Research Center, Division of Cardiology, Corrigan Minehan Heart Center, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Azrul Abdul Kadir
- Cardiovascular Research Center, Division of Cardiology, Corrigan Minehan Heart Center, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Carolin Lerchenmüller
- Department of Cardiology, University Hospital Heidelberg, German Center for Cardiovascular Research (DZHK), Partner Site Heidelberg/Mannheim, Heidelberg, Germany
| | - Haobo Li
- Cardiovascular Research Center, Division of Cardiology, Corrigan Minehan Heart Center, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - James Rhee
- Cardiovascular Research Center, Division of Cardiology, Corrigan Minehan Heart Center, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Anesthesiology and Critical Care, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jason D. Roh
- Cardiovascular Research Center, Division of Cardiology, Corrigan Minehan Heart Center, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Kangsan Roh
- Cardiovascular Research Center, Division of Cardiology, Corrigan Minehan Heart Center, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Anesthesiology and Critical Care, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Anand P. Singh
- Institute for Heart and Brain Health, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Chao Wu
- Institute for Heart and Brain Health, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Peng Xia
- Cardiovascular Research Center, Division of Cardiology, Corrigan Minehan Heart Center, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Qiulian Zhou
- Institute for Heart and Brain Health, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Junjie Xiao
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, Shanghai, China
| | - Anthony Rosenzweig
- Institute for Heart and Brain Health, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| |
Collapse
|
3
|
Kho C. Targeting calcium regulators as therapy for heart failure: focus on the sarcoplasmic reticulum Ca-ATPase pump. Front Cardiovasc Med 2023; 10:1185261. [PMID: 37534277 PMCID: PMC10392702 DOI: 10.3389/fcvm.2023.1185261] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 07/06/2023] [Indexed: 08/04/2023] Open
Abstract
Impaired myocardial Ca2+ cycling is a critical contributor to the development of heart failure (HF), causing changes in the contractile function and structure remodeling of the heart. Within cardiomyocytes, the regulation of sarcoplasmic reticulum (SR) Ca2+ storage and release is largely dependent on Ca2+ handling proteins, such as the SR Ca2+ ATPase (SERCA2a) pump. During the relaxation phase of the cardiac cycle (diastole), SERCA2a plays a critical role in transporting cytosolic Ca2+ back to the SR, which helps to restore both cytosolic Ca2+ levels to their resting state and SR Ca2+ content for the next contraction. However, decreased SERCA2a expression and/or pump activity are key features in HF. As a result, there is a growing interest in developing therapeutic approaches to target SERCA2a. This review provides an overview of the regulatory mechanisms of the SERCA2a pump and explores potential strategies for SERCA2a-targeted therapy, which are being investigated in both preclinical and clinical studies.
Collapse
Affiliation(s)
- Changwon Kho
- Division of Applied Medicine, School of Korean Medicine, Pusan National University, Yangsan, Republic of Korea
| |
Collapse
|
4
|
Bencurova M, Lysikova T, Leskova Majdova K, Kaplan P, Racay P, Lehotsky J, Tatarkova Z. Age-Dependent Changes in Calcium Regulation after Myocardial Ischemia-Reperfusion Injury. Biomedicines 2023; 11:biomedicines11041193. [PMID: 37189811 DOI: 10.3390/biomedicines11041193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 04/11/2023] [Accepted: 04/15/2023] [Indexed: 05/17/2023] Open
Abstract
During aging, heart structure and function gradually deteriorate, which subsequently increases susceptibility to ischemia-reperfusion (IR). Maintenance of Ca2+ homeostasis is critical for cardiac contractility. We used Langendorff's model to monitor the susceptibility of aging (6-, 15-, and 24-month-old) hearts to IR, with a specific focus on Ca2+-handling proteins. IR, but not aging itself, triggered left ventricular changes when the maximum rate of pressure development decreased in 24-month-olds, and the maximum rate of relaxation was most affected in 6-month-old hearts. Aging caused a deprivation of Ca2+-ATPase (SERCA2a), Na+/Ca2+ exchanger, mitochondrial Ca2+ uniporter, and ryanodine receptor contents. IR-induced damage to ryanodine receptor stimulates Ca2+ leakage in 6-month-old hearts and elevated phospholamban (PLN)-to-SERCA2a ratio can slow down Ca2+ reuptake seen at 2-5 μM Ca2+. Total and monomeric PLN mirrored the response of overexpressed SERCA2a after IR in 24-month-old hearts, resulting in stable Ca2+-ATPase activity. Upregulated PLN accelerated inhibition of Ca2+-ATPase activity at low free Ca2+ in 15-month-old after IR, and reduced SERCA2a content subsequently impairs the Ca2+-sequestering capacity. In conclusion, our study suggests that aging is associated with a significant decrease in the abundance and function of Ca2+-handling proteins. However, the IR-induced damage was not increased during aging.
Collapse
Affiliation(s)
- Maria Bencurova
- Department of Medical Biochemistry, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Terezia Lysikova
- Department of Medical Biochemistry, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Katarina Leskova Majdova
- Department of Medical Biochemistry, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Peter Kaplan
- Department of Medical Biochemistry, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Peter Racay
- Department of Medical Biochemistry, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Jan Lehotsky
- Department of Medical Biochemistry, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Zuzana Tatarkova
- Department of Medical Biochemistry, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 03601 Martin, Slovakia
| |
Collapse
|
5
|
Ribeiro ASF, Zerolo BE, López-Espuela F, Sánchez R, Fernandes VS. Cardiac System during the Aging Process. Aging Dis 2023:AD.2023.0115. [PMID: 37163425 PMCID: PMC10389818 DOI: 10.14336/ad.2023.0115] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 01/15/2023] [Indexed: 05/12/2023] Open
Abstract
The aging process is accompanied by a continuous decline of the cardiac system, disrupting the homeostatic regulation of cells, organs, and systems. Aging increases the prevalence of cardiovascular diseases, thus heart failure and mortality. Understanding the cardiac aging process is of pivotal importance once it allows us to design strategies to prevent age-related cardiac events and increasing the quality of live in the elderly. In this review we provide an overview of the cardiac aging process focus on the following topics: cardiac structural and functional modifications; cellular mechanisms of cardiac dysfunction in the aging; genetics and epigenetics in the development of cardiac diseases; and aging heart and response to the exercise.
Collapse
Affiliation(s)
| | - Blanca Egea Zerolo
- Escuela de Enfermería y Fisioterapia San Juan de Dios. Universidad Pontificia Comillas, Madrid, Spain
| | - Fidel López-Espuela
- Metabolic Bone Diseases Research Group, Nursing and Occupational Therapy College, University of Extremadura, Caceres, Spain
| | - Raúl Sánchez
- Unidad de Cardiopatías Congénitas, Hospital Universitario La Paz, Madrid, Spain
| | - Vítor S Fernandes
- Departamento de Fisiología, Facultad de Farmacia, Universidad Complutense de Madrid, Madrid, Spain
| |
Collapse
|
6
|
Mercurio V, Ambrosio G, Correale M, Dini FL, Ghio S, Nodari S, Palazzuoli A, Ruocco G, Pedrinelli R, Mercuro G, Filardi PP, Indolfi C, Agostoni P, Tocchetti CG, Paolillo S. Innovations in medical therapy of heart failure with reduced ejection fraction. J Cardiovasc Med (Hagerstown) 2022; 24:e47-e54. [PMID: 36729606 DOI: 10.2459/jcm.0000000000001413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Heart failure with reduced ejection fraction (HFrEF) is a pathological condition still characterized by high rates of mortality and disease exacerbation frequently leading to hospitalization, thus there is a continuous need for pharmacological treatments impacting on disease stability and long-term prognosis. Moreover, the phenotype of heart failure patients is continuously changing over time, and the development of new heart failure drugs is crucial to promote a personalized and targeted approach. In recent years, several therapeutic innovations have emerged in the landscape of acute and chronic HFrEF, largely changing and improving our approach to the disease. Various studies on new drugs and experimental therapeutic approaches are ongoing. The present review discusses the latest data on both recently approved drugs and developing therapeutic targets, in order to provide a critical overview for an informed and optimal approach to such a complex disease.
Collapse
Affiliation(s)
- Valentina Mercurio
- Department of Translational Medical Sciences, Federico II University, Naples.,Interdepartmental Center of Clinical and Translational Sciences (CIRCET), Federico II University
| | | | | | - Frank L Dini
- Cardiac, Thoracic and Vascular Department, University of Pisa, Pisa
| | - Stefano Ghio
- Division of Cardiology, Fondazione IRCCS Policlinico S.Matteo, Pavia
| | - Savina Nodari
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia
| | - Alberto Palazzuoli
- Cardiovascular Disease Unit, Department of Internal Medicine, University of Siena, Siena
| | - Gaetano Ruocco
- Cardiology Unit, Riuniti of Valdichiana Hospitals, USL Sud Est Toscana, Montepulciano
| | - Roberto Pedrinelli
- Dipartimento di Patologia Chirurgica, Medica, Molecolare e dell'Area Critica, Università di Pisa
| | - Giuseppe Mercuro
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari
| | - Pasquale Perrone Filardi
- Department of Advanced Biomedical Sciences, Federico II University, Naples.,Mediterranea Cardiocentro, Naples
| | - Ciro Indolfi
- Cardiology Unit, University Magna Graecia of Catanzaro, Catanza
| | - Piergiuseppe Agostoni
- Centro Cardiologico Monzino, IRCCS.,Department of Clinical Sciences and Community Health, Cardiovascular Section, University of Milan, Milan
| | - Carlo G Tocchetti
- Department of Translational Medical Sciences, Federico II University, Naples.,Interdepartmental Center of Clinical and Translational Sciences (CIRCET), Federico II University.,Interdepartmental Hypertension Research Center (CIRIAPA).,Center for Basic and Clinical Immunology Research (CISI), Federico II University, Naples, Italy
| | - Stefania Paolillo
- Department of Advanced Biomedical Sciences, Federico II University, Naples.,Mediterranea Cardiocentro, Naples
| |
Collapse
|
7
|
Oknińska M, Mączewski M, Mackiewicz U. Ventricular arrhythmias in acute myocardial ischaemia-Focus on the ageing and sex. Ageing Res Rev 2022; 81:101722. [PMID: 36038114 DOI: 10.1016/j.arr.2022.101722] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 08/17/2022] [Accepted: 08/20/2022] [Indexed: 01/31/2023]
Abstract
Annually, approximately 17 million people die from cardiovascular diseases worldwide, half of them suddenly. The most common direct cause of sudden cardiac death is ventricular arrhythmia triggered by an acute coronary syndrome (ACS). The study summarizes the knowledge of the mechanisms of arrhythmia onset during ACS in humans and in animal models and factors that may influence the susceptibility to life-threatening arrhythmias during ACS with particular focus on the age and sex. The real impact of age and sex on the arrhythmic susceptibility within the setting of acute ischaemia is masked by the fact that ACSs result from coronary artery disease appearing with age much earlier among men than among women. However, results of researches show that in ageing process changes with potential pro-arrhythmic significance, such as increased fibrosis, cardiomyocyte hypertrophy, decrease number of gap junction channels, disturbances of the intracellular Ca2+ signalling or changes in electrophysiological parameters, occur independently of the development of cardiovascular diseases and are more severe in male individuals. A review of the literature also indicates a marked paucity of research in this area in female and elderly individuals. Greater awareness of sex differences in the aging process could help in the development of personalized prevention methods targeting potential pro-arrhythmic factors in patients of both sexes to reduce mortality during the acute phase of myocardial infarction. This is especially important in an era of aging populations in which women will predominate due to their longer lifespan.
Collapse
Affiliation(s)
- Marta Oknińska
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, Marymoncka 99/103, 01-813 Warsaw, Poland
| | - Michał Mączewski
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, Marymoncka 99/103, 01-813 Warsaw, Poland
| | - Urszula Mackiewicz
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, Marymoncka 99/103, 01-813 Warsaw, Poland.
| |
Collapse
|
8
|
Lu Q, Pan B, Bai H, Zhao W, Liu L, Li G, Liu R, Lv T, Huang X, Li X, Tian J. Intranuclear cardiac troponin I plays a functional role in regulating Atp2a2 expression in cardiomyocytes. Genes Dis 2022; 9:1689-1700. [PMID: 36157491 PMCID: PMC9485201 DOI: 10.1016/j.gendis.2021.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 03/11/2021] [Accepted: 04/15/2021] [Indexed: 11/21/2022] Open
Abstract
In the past studies, it is shown that cardiac troponin I (cTnI, encoded by TNNI3), as a cytoplasmic protein, is an inhibitory subunit in troponin complex, and involves in cardiomyocyte diastolic regulation. Here, we assessed a novel role of cTnI as a nucleoprotein. Firstly, the nuclear translocation of cTnI was found in mouse, human fetuses and rat heart tissues. In addition, there were differences in percentage of intranuclear cTnI in different conditions. Based on weighted gene co-expression network analyses (WGCNA) and verification in cell experiments, a strong expression correlation was found between TNNI3 and Atp2a2, which encodes sarco-endoplasmic reticulum Ca2+ ATPase isoform 2a (SERCA2a), and involves in ATP hydrolysis and Ca2+ transient. TNNI3 gain and loss caused Atpa2a2 increase/decrease in a dose-dependent manner both in mRNA and protein levels, in vivo and in vitro. By using ChIP-sequence we demonstrated specific binding DNA sequences of cTnI were enriched in ATP2a2 promoter −239∼–889 region and the specific binding sequence motif of cTnI was analyzed by software as "CCAT", which has been reported to be required for YY1 binding to the promoter region of YY1-related genes. Moreover, it was further verified that pcDNA3.1 (−)-TNNI3 could express cTnI proteins and increase the promoter activity of Atp2a2 through luciferase report assay. In the end, we evaluated beat frequencies, total ATP contents, Ca2+ transients in TNNI3-siRNA myocardial cells. These findings indicated, for the first time, cTnI may regulate Atp2a2 in cardiomyocytes as a co-regulatory factor and participate in the regulation of intracellular Ca ions.
Collapse
Affiliation(s)
- Qian Lu
- Department of Pediatric Cardiology, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, PR China.,Chongqing Key Laboratory of Pediatrics, Chongqing 400014, PR China.,Department of Biomedical Science, Charlie E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Bo Pan
- Department of Pediatric Cardiology, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, PR China.,Chongqing Key Laboratory of Pediatrics, Chongqing 400014, PR China
| | - Haobo Bai
- Department of Orthopedic, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China
| | - Weian Zhao
- Department of Pediatric Cardiology, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, PR China.,Chongqing Key Laboratory of Pediatrics, Chongqing 400014, PR China
| | - Lingjuan Liu
- Department of Pediatric Cardiology, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, PR China.,Chongqing Key Laboratory of Pediatrics, Chongqing 400014, PR China
| | - Gu Li
- Department of Pediatric Cardiology, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, PR China.,Chongqing Key Laboratory of Pediatrics, Chongqing 400014, PR China
| | - Ruimin Liu
- Department of Pediatric Cardiology, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, PR China.,Chongqing Key Laboratory of Pediatrics, Chongqing 400014, PR China
| | - Tiewei Lv
- Department of Pediatric Cardiology, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, PR China.,Chongqing Key Laboratory of Pediatrics, Chongqing 400014, PR China
| | - Xupei Huang
- Department of Biomedical Science, Charlie E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Xi Li
- Biology Science Institutes of Chongqing Medical University, Chongqing 400016, PR China
| | - Jie Tian
- Department of Pediatric Cardiology, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, PR China.,Chongqing Key Laboratory of Pediatrics, Chongqing 400014, PR China
| |
Collapse
|
9
|
Lackner I, Weber B, Pressmar J, Odwarka A, Lam C, Haffner-Luntzer M, Marcucio R, Miclau T, Kalbitz M. Cardiac alterations following experimental hip fracture - inflammaging as independent risk factor. Front Immunol 2022; 13:895888. [PMID: 36131923 PMCID: PMC9484325 DOI: 10.3389/fimmu.2022.895888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 08/08/2022] [Indexed: 12/04/2022] Open
Abstract
Background Cardiac injuries following trauma are associated with a worse clinical outcome. So-called trauma-induced secondary cardiac injuries have been recently described after experimental long bone fracture even in absence of direct heart damage. With the progressive aging of our society, the number of elderly trauma victims rises and therefore the incidence of hip fractures increases. Hip fractures were previously shown to be associated with adverse cardiac events in elderly individuals, which have mainly been attributed to pre-conditioned cardiac diseases. The aim of the present study was to investigate the effect of hip fractures on the heart in healthy young and middle-aged mice. Materials and Methods Young (12-week-old) and middle-aged (52-week-old) female C57BL/6 mice either received an intramedullary stabilized proximal femur fracture or sham treatment. The observation time points included 6 and 24 h. Systemic levels of pro-inflammatory mediators as well as local inflammation and alterations in myocardial structure, metabolism and calcium homeostasis in left ventricular tissue was analyzed following hip fracture by multiplex analysis, RT-qPCR and immunohistochemistry. Results After hip fracture young and middle-aged mice showed increased systemic IL-6 and KC levels, which were significantly elevated in the middle-aged animals. Furthermore, the middle-aged mice showed enhanced myocardial expression of HMGB1, TLR2/4, TNF, IL1β and NLRP3 as well as considerable alterations in the myocardial expression of glucose- and fatty acid transporters (HFABP, GLUT4), calcium homeostasis proteins (SERCA) and cardiac structure proteins (desmin, troponin I) compared to the young animals following hip fracture. Conclusion Young and middle-aged mice showed local myocardial alterations, which might predispose for the development of secondary cardiac injury following hip fracture. Age and the age-associated phenomenon of ‘inflammaging’ seemed to be an independent risk factor aggravating and accelerating cardiac alterations following hip fracture.
Collapse
Affiliation(s)
- Ina Lackner
- Department of Trauma and Orthopedic Surgery, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
- Department of Traumatology, Hand, Plastic, and Reconstructive Surgery, University Medical Center Ulm, Ulm, Germany
| | - Birte Weber
- Department of Traumatology, Hand, Plastic, and Reconstructive Surgery, University Medical Center Ulm, Ulm, Germany
- Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, Zuckerberg San Francisco General Hospital, University of California, San Francisco, San Francisco, CA, United States
- Department of Trauma, Hand and Reconstructive Surgery, Goethe University of Frankfurt, Frankfurt, Germany
| | - Jochen Pressmar
- Department of Trauma and Orthopedic Surgery, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
- Department of Traumatology, Hand, Plastic, and Reconstructive Surgery, University Medical Center Ulm, Ulm, Germany
| | - Anna Odwarka
- Department of Traumatology, Hand, Plastic, and Reconstructive Surgery, University Medical Center Ulm, Ulm, Germany
| | - Charles Lam
- Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, Zuckerberg San Francisco General Hospital, University of California, San Francisco, San Francisco, CA, United States
| | - Melanie Haffner-Luntzer
- Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, Zuckerberg San Francisco General Hospital, University of California, San Francisco, San Francisco, CA, United States
- Institute of Orthopaedic Research and Biomechanics, University Medical Center Ulm, Ulm, Germany
| | - Ralph Marcucio
- Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, Zuckerberg San Francisco General Hospital, University of California, San Francisco, San Francisco, CA, United States
| | - Theodore Miclau
- Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, Zuckerberg San Francisco General Hospital, University of California, San Francisco, San Francisco, CA, United States
| | - Miriam Kalbitz
- Department of Trauma and Orthopedic Surgery, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
- Department of Traumatology, Hand, Plastic, and Reconstructive Surgery, University Medical Center Ulm, Ulm, Germany
- Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, Zuckerberg San Francisco General Hospital, University of California, San Francisco, San Francisco, CA, United States
- *Correspondence: Miriam Kalbitz,
| |
Collapse
|
10
|
Iakovou E, Kourti M. A Comprehensive Overview of the Complex Role of Oxidative Stress in Aging, The Contributing Environmental Stressors and Emerging Antioxidant Therapeutic Interventions. Front Aging Neurosci 2022; 14:827900. [PMID: 35769600 PMCID: PMC9234325 DOI: 10.3389/fnagi.2022.827900] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 05/10/2022] [Indexed: 12/15/2022] Open
Abstract
Introduction Aging is a normal, inevitable, irreversible, and progressive process which is driven by internal and external factors. Oxidative stress, that is the imbalance between prooxidant and antioxidant molecules favoring the first, plays a key role in the pathophysiology of aging and comprises one of the molecular mechanisms underlying age-related diseases. However, the oxidative stress theory of aging has not been successfully proven in all animal models studying lifespan, meaning that altering oxidative stress/antioxidant defense systems did not always lead to a prolonged lifespan, as expected. On the other hand, animal models of age-related pathological phenotypes showed a well-correlated relationship with the levels of prooxidant molecules. Therefore, it seems that oxidative stress plays a more complicated role than the one once believed and this role might be affected by the environment of each organism. Environmental factors such as UV radiation, air pollution, and an unbalanced diet, have also been implicated in the pathophysiology of aging and seem to initiate this process more rapidly and even at younger ages. Aim The purpose of this review is to elucidate the role of oxidative stress in the physiology of aging and the effect of certain environmental factors in initiating and sustaining this process. Understanding the pathophysiology of aging will contribute to the development of strategies to postpone this phenomenon. In addition, recent studies investigating ways to alter the antioxidant defense mechanisms in order to prevent aging will be presented. Conclusions Careful exposure to harmful environmental factors and the use of antioxidant supplements could potentially affect the biological processes driving aging and slow down the development of age-related diseases. Maybe a prolonged lifespan could not be achieved by this strategy alone, but a longer healthspan could also be a favorable target.
Collapse
Affiliation(s)
- Evripides Iakovou
- Department of Life Sciences, European University Cyprus, Nicosia, Cyprus
| | - Malamati Kourti
- Department of Life Sciences, European University Cyprus, Nicosia, Cyprus
- Angiogenesis and Cancer Drug Discovery Group, Basic and Translational Cancer Research Center, Department of Life Sciences, European University Cyprus, Nicosia, Cyprus
- *Correspondence: Malamati Kourti
| |
Collapse
|
11
|
Das BB. Therapeutic Approaches in Heart Failure with Preserved Ejection Fraction (HFpEF) in Children: Present and Future. Paediatr Drugs 2022; 24:235-246. [PMID: 35501560 DOI: 10.1007/s40272-022-00508-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/07/2022] [Indexed: 12/29/2022]
Abstract
For a long time, pediatric heart failure (HF) with preserved systolic function (HFpEF) has been noted in patients with cardiomyopathies and congenital heart disease. HFpEF is infrequently reported in children and instead of using the HFpEF terminology the HF symptoms are attributed to diastolic dysfunction. Identifying HFpEF in children is challenging because of heterogeneous etiologies and unknown pathophysiological mechanisms. Advances in echocardiography and cardiac magnetic resonance imaging techniques have further increased our understanding of HFpEF in children. However, the literature does not describe the incidence, etiology, clinical features, and treatment of HFpEF in children. At present, treatment of HFpEF in children is extrapolated from clinical trials in adults. There are significant differences between pediatric and adult HF with reduced ejection fraction, supported by a lack of adequate response to adult HF therapies. Evidence-based clinical trials in children are still not available because of the difficulty of conducting trials with a limited number of pediatric patients with HF. The treatment of HFpEF in children is based upon the clinician's experience, and the majority of children receive off-level medications. There are significant differences between pediatric and adult HFpEF pharmacotherapies in many areas, including side-effect profiles, underlying pathophysiologies, the β-receptor physiology, and pharmacokinetics and pharmacodynamics. This review describes the present and future treatments for children with HFpEF compared with adults. This review also highlights the need to urgently test new therapies in children with HFpEF to demonstrate the safety and efficacy of drugs and devices with proven benefits in adults.
Collapse
Affiliation(s)
- Bibhuti B Das
- Department of Pediatrics, Division of Cardiology, University of Mississippi Medical Center, 2500 N State St., Jackson, MS, 39216, USA.
| |
Collapse
|
12
|
Sanchis-Gomar F, Lavie CJ, Marín J, Perez-Quilis C, Eijsvogels TMH, O'Keefe JH, Perez MV, Blair SN. Exercise Effects On Cardiovascular Disease: From Basic Aspects To Clinical Evidence. Cardiovasc Res 2021; 118:2253-2266. [PMID: 34478520 DOI: 10.1093/cvr/cvab272] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 08/31/2021] [Indexed: 12/16/2022] Open
Abstract
Cardiovascular (CV) disease (CVD) remains the leading cause of major morbidity and CVD- and all-cause mortality in most of the world. It is now clear that regular physical activity (PA) and exercise training (ET) induces a wide range of direct and indirect physiologic adaptations and pleiotropic benefits for human general and CV health. Generally, higher levels of PA, ET, and cardiorespiratory fitness (CRF) are correlated with reduced risk of CVD, including myocardial infarction, CVD-related death, and all-cause mortality. Although exact details regarding the ideal doses of ET, including resistance and, especially, aerobic ET, as well as the potential adverse effects of extreme levels of ET, continue to be investigated, there is no question that most of the world's population have insufficient levels of PA/ET, and many also have lower than ideal levels of CRF. Therefore, assessment and promotion of PA, ET, and efforts to improve levels of CRF should be integrated into all health professionals' practices worldwide. In this state-of-the-art review, we discuss the exercise effects on many areas related to CVD, from basic aspects to clinical practice.
Collapse
Affiliation(s)
- Fabian Sanchis-Gomar
- Department of Physiology, Faculty of Medicine, University of Valencia and INCLIVA Biomedical Research Institute, Valencia, Spain.,Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Carl J Lavie
- John Ochsner Heart and Vascular Institute, Ochsner Clinical School, The University of Queensland School of Medicine, New Orleans, LA, USA
| | - Jorge Marín
- Growth, Exercise, Nutrition and Development Group, Faculty of Health and Sport Sciences, University of Zaragoza, Zaragoza, Spain
| | - Carme Perez-Quilis
- Department of Physiology, Faculty of Medicine, University of Valencia and INCLIVA Biomedical Research Institute, Valencia, Spain
| | - Thijs M H Eijsvogels
- Radboud Institute for Health Science, Department of Physiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - James H O'Keefe
- St. Luke's Mid America Heart Institute, University of Missouri-Kansas City, Kansas City, Missouri, USA
| | - Marco V Perez
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Steven N Blair
- Department of Exercise Sciences, University of South Carolina, Columbia, USA
| |
Collapse
|
13
|
Oknińska M, Paterek A, Bierła J, Czarnowska E, Mączewski M, Mackiewicz U. Effect of age and sex on the incidence of ventricular arrhythmia in a rat model of acute ischemia. Biomed Pharmacother 2021; 142:111983. [PMID: 34392089 DOI: 10.1016/j.biopha.2021.111983] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 07/23/2021] [Accepted: 07/26/2021] [Indexed: 10/20/2022] Open
Abstract
BACKGROUND The impact of sex and age on the arrhythmic susceptibility within the setting of acute ischemia is masked by the fact that acute coronary events result from coronary artery disease appearing with age much earlier among men than among women. METHODS AND RESULTS LAD ligation or sham operations were performed in rats of both sexes at the age 3 and 24 months. An ECG was recorded continuously for 6 h after the operation. The number of early and late premature ventricular beats (PVBs), episodes of ventricular tachycardia (VT) and fibrillation (VF), heart rate, QRS, QT and Tpeak-Tend duration were analysed. Epicardial action potentials were recorded in vivo, Ca2+ signaling was evaluated in isolated cardiomyocytes, fibrosis and connexin-43 expression and localization were measured in the septum. PVBs, VT and VF episodes are much more common in older males than in young males and females independently from their age. Fibrosis with varying intensity in different muscle layers, hypertrophy of cardiomyocytes, reduced number of gap junctions and their appearance on the lateral myocyte membrane, QT prolongation, increase transmural dispersion of repolarisation and a decreased function of SERCA2a may increase the propensity to arrhythmia within the setting of acute ischemia. CONCLUSION We show that the male sex, especially in case of older individuals is a strong predictor of increased arrhythmic susceptibility within the acute ischemia setting regardless of its impact on the occurrence of cardiovascular diseases. A personalized sex-dependent prevention treatment is needed to reduce the mortality in acute phases of myocardial infarction.
Collapse
Affiliation(s)
- Marta Oknińska
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, Marymoncka 99/103, 01-813 Warsaw, Poland
| | - Aleksandra Paterek
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, Marymoncka 99/103, 01-813 Warsaw, Poland
| | - Joanna Bierła
- Department of Pathology, The Children's Memorial Health Institute, Aleja Dzieci Polskich 20, 04-730 Warsaw, Poland
| | - Elżbieta Czarnowska
- Department of Pathology, The Children's Memorial Health Institute, Aleja Dzieci Polskich 20, 04-730 Warsaw, Poland
| | - Michał Mączewski
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, Marymoncka 99/103, 01-813 Warsaw, Poland
| | - Urszula Mackiewicz
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, Marymoncka 99/103, 01-813 Warsaw, Poland.
| |
Collapse
|
14
|
Dridi H, Kushnir A, Zalk R, Yuan Q, Melville Z, Marks AR. Intracellular calcium leak in heart failure and atrial fibrillation: a unifying mechanism and therapeutic target. Nat Rev Cardiol 2020; 17:732-747. [PMID: 32555383 PMCID: PMC8362847 DOI: 10.1038/s41569-020-0394-8] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/06/2020] [Indexed: 12/14/2022]
Abstract
Ca2+ is a fundamental second messenger in all cell types and is required for numerous essential cellular functions, including cardiac and skeletal muscle contraction. The intracellular concentration of free Ca2+ ([Ca2+]) is regulated primarily by ion channels, pumps (ATPases), exchangers and Ca2+-binding proteins. Defective regulation of [Ca2+] is found in a diverse spectrum of pathological states that affect all the major organs. In the heart, abnormalities in the regulation of cytosolic and mitochondrial [Ca2+] occur in heart failure (HF) and atrial fibrillation (AF), two common forms of heart disease and leading contributors to morbidity and mortality. In this Review, we focus on the mechanisms that regulate ryanodine receptor 2 (RYR2), the major sarcoplasmic reticulum (SR) Ca2+-release channel in the heart, how RYR2 becomes dysfunctional in HF and AF, and its potential as a therapeutic target. Inherited RYR2 mutations and/or stress-induced phosphorylation and oxidation of the protein destabilize the closed state of the channel, resulting in a pathological diastolic Ca2+ leak from the SR that both triggers arrhythmias and impairs contractility. On the basis of our increased understanding of SR Ca2+ leak as a shared Ca2+-dependent pathological mechanism in HF and AF, a new class of drugs developed in our laboratory, known as rycals, which stabilize RYR2 channels and prevent Ca2+ leak from the SR, are undergoing investigation in clinical trials.
Collapse
Affiliation(s)
- Haikel Dridi
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Alexander Kushnir
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Ran Zalk
- The National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Qi Yuan
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Zephan Melville
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Andrew R Marks
- Department of Physiology and Cellular Biophysics, Clyde and Helen Wu Center for Molecular Cardiology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA.
| |
Collapse
|
15
|
Moreira JBN, Wohlwend M, Wisløff U. Exercise and cardiac health: physiological and molecular insights. Nat Metab 2020; 2:829-839. [PMID: 32807982 DOI: 10.1038/s42255-020-0262-1] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 07/13/2020] [Indexed: 12/19/2022]
Abstract
The cardiac benefits of exercise have been recognized for centuries. Studies have undisputedly shown that regular exercise is beneficial for the cardiovascular system in young, old, healthy and diseased populations. For these reasons, physical activity has been recommended worldwide for cardiovascular disease prevention and treatment. Although the benefits of exercise are clear, understanding of the molecular triggers that orchestrate these effects remains incomplete and has been a topic of intense research in recent years. Here, we provide a comprehensive review of the cardiac effects of physical activity, beginning with a brief history of exercise in cardiovascular medicine and then discussing seminal work on the physiological effects of exercise in healthy, diseased and aged hearts. Later, we revisit pioneering work on the molecular mechanisms underlying the cardiac benefits of exercise, and we conclude with our view on the translational potential of this knowledge as a powerful platform for cardiovascular disease drug discovery.
Collapse
Affiliation(s)
- Jose B N Moreira
- Cardiac Exercise Research Group at the Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Martin Wohlwend
- Cardiac Exercise Research Group at the Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Ulrik Wisløff
- Cardiac Exercise Research Group at the Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway.
- School of Human Movement & Nutrition Sciences, University of Queensland, Brisbane, Queensland, Australia.
| |
Collapse
|
16
|
Tracy E, Rowe G, LeBlanc AJ. Cardiac tissue remodeling in healthy aging: the road to pathology. Am J Physiol Cell Physiol 2020; 319:C166-C182. [PMID: 32432929 DOI: 10.1152/ajpcell.00021.2020] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
This review aims to highlight the normal physiological remodeling that occurs in healthy aging hearts, including changes that occur in contractility, conduction, valve function, large and small coronary vessels, and the extracellular matrix. These "normal" age-related changes serve as the foundation that supports decreased plasticity and limited ability for tissue remodeling during pathophysiological states such as myocardial ischemia and heart failure. This review will identify populations at greater risk for poor tissue remodeling in advanced age along with present and future therapeutic strategies that may ameliorate dysfunctional tissue remodeling in aging hearts.
Collapse
Affiliation(s)
- Evan Tracy
- Department of Physiology, Cardiovascular Innovation Institute, University of Louisville, Louisville, Kentucky
| | - Gabrielle Rowe
- Department of Physiology, Cardiovascular Innovation Institute, University of Louisville, Louisville, Kentucky
| | - Amanda J LeBlanc
- Department of Physiology, Cardiovascular Innovation Institute, University of Louisville, Louisville, Kentucky
| |
Collapse
|
17
|
Dong M, Yang Z, Fang H, Xiang J, Xu C, Zhou Y, Wu Q, Liu J. Aging Attenuates Cardiac Contractility and Affects Therapeutic Consequences for Myocardial Infarction. Aging Dis 2020; 11:365-376. [PMID: 32257547 PMCID: PMC7069457 DOI: 10.14336/ad.2019.0522] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 05/22/2019] [Indexed: 12/21/2022] Open
Abstract
Cardiac function of the human heart changes with age. The age-related change of systolic function is subtle under normal conditions, but abrupt under stress or in a pathogenesis state. Aging decreases the cardiac tolerance to stress and increases susceptibility to ischemia, which caused by aging-induced Ca2+ transient impairment and metabolic dysfunction. The changes of contractility proteins and the relative molecules are in a non-linear fashion. Specifically, the expression and activation of cMLCK increase first then fall during ischemia and reperfusion (I/R). This change is responsible for the nonmonotonic contractility alteration in I/R which the underlying mechanism is still unclear. Contractility recovery in I/R is also attenuated by age. The age-related change in cardiac contractility influences the therapeutic effect and intervention timepoint. For most cardiac ischemia therapies, the therapeutic result in the elderly is not identical to the young. Anti-aging treatment has the potential to prevent the development of ischemic injury and improves cardiac function. In this review we discuss the mechanism underlying the contractility changes in the aged heart and age-induced ischemic injury. The potential mechanism underlying the increased susceptibility to ischemic injury in advanced age is highlighted. Furthermore, we discuss the effect of age and the administration time for intervention in cardiac ischemia therapies.
Collapse
Affiliation(s)
- Ming Dong
- Department of Pathophysiology, Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Shenzhen University Health Science Center, Guangdong, China
| | - Ziyi Yang
- Department of Pathophysiology, Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Shenzhen University Health Science Center, Guangdong, China
| | - Hongcheng Fang
- Shenzhen Shajing Hospital, Affiliated of Guangzhou Medical University, Shenzhen, Guangdong, China
| | - Jiaqing Xiang
- Department of Pathophysiology, Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Shenzhen University Health Science Center, Guangdong, China
| | - Cong Xu
- Department of Pathophysiology, Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Shenzhen University Health Science Center, Guangdong, China
| | - Yanqing Zhou
- Department of Pathophysiology, Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Shenzhen University Health Science Center, Guangdong, China
| | - Qianying Wu
- Department of Pathophysiology, Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Shenzhen University Health Science Center, Guangdong, China
| | - Jie Liu
- Department of Pathophysiology, Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Shenzhen University Health Science Center, Guangdong, China
| |
Collapse
|
18
|
Stroik DR, Ceholski DK, Bidwell PA, Mleczko J, Thanel PF, Kamdar F, Autry JM, Cornea RL, Thomas DD. Viral expression of a SERCA2a-activating PLB mutant improves calcium cycling and synchronicity in dilated cardiomyopathic hiPSC-CMs. J Mol Cell Cardiol 2019; 138:59-65. [PMID: 31751570 DOI: 10.1016/j.yjmcc.2019.11.147] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 11/04/2019] [Accepted: 11/10/2019] [Indexed: 12/19/2022]
Abstract
There is increasing momentum toward the development of gene therapy for heart failure (HF) that is defined by impaired calcium (Ca2+) transport and reduced contractility. We have used FRET (fluorescence resonance energy transfer) between fluorescently-tagged SERCA2a (the cardiac Ca2+ pump) and PLB (phospholamban, ventricular peptide inhibitor of SERCA) to test directly the effectiveness of loss-of-inhibition/gain-of-binding (LOI/GOB) PLB mutants (PLBM) that were engineered to compete with the binding of inhibitory wild-type PLB (PLBWT). Our therapeutic strategy is to relieve PLBWT inhibition of SERCA2a by using the reserve adrenergic capacity mediated by PLB to enhance cardiac contractility. Using a FRET assay, we determined that the combination of a LOI PLB mutation (L31A) and a GOB PLB mutation (I40A) results in a novel engineered LOI/GOB PLBM (L31A/I40A) that effectively competes with PLBWT binding to cardiac SERCA2a in HEK293-6E cells. We demonstrated that co-expression of PLBM enhances SERCA Ca-ATPase activity by increasing enzyme Ca2+ affinity (1/KCa) in PLBWT-inhibited HEK293 cell homogenates. For an initial assessment of PLBM physiological effectiveness, we used human induced pluripotent stem cell derived cardiomyocytes (hiPSC-CMs) from a healthy individual. In this system, we observed that adeno-associated virus 2 (rAAV2)-driven expression of PLBM enhances the amplitude of SR Ca2+ release and the rate of SR Ca2+ re-uptake. To assess therapeutic potential, we used a hiPSC-CM model of dilated cardiomyopathy (DCM) containing PLB mutation R14del, where we observed that rAAV2-driven expression of PLBM rescues arrhythmic Ca2+ transients and alleviates decreased Ca2+ transport. Thus, we propose that PLBM transgene expression is a promising gene therapy strategy that directly targets the underlying pathophysiology of abnormal Ca2+ transport and thus contractility in underlying systolic heart failure.
Collapse
Affiliation(s)
- Daniel R Stroik
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN 55455, United States of America
| | - Delaine K Ceholski
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York City, New York 10029, United States of America
| | - Philip A Bidwell
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN 55455, United States of America; Division of Cardiology, Department of Medicine, University of Minnesota, Minneapolis, MN 55455, United States of America
| | - Justyna Mleczko
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York City, New York 10029, United States of America
| | - Paul F Thanel
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN 55455, United States of America
| | - Forum Kamdar
- Division of Cardiology, Department of Medicine, University of Minnesota, Minneapolis, MN 55455, United States of America
| | - Joseph M Autry
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN 55455, United States of America
| | - Razvan L Cornea
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN 55455, United States of America
| | - David D Thomas
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN 55455, United States of America.
| |
Collapse
|
19
|
Hamilton S, Terentyev D. Altered Intracellular Calcium Homeostasis and Arrhythmogenesis in the Aged Heart. Int J Mol Sci 2019; 20:ijms20102386. [PMID: 31091723 PMCID: PMC6566636 DOI: 10.3390/ijms20102386] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 04/29/2019] [Accepted: 05/08/2019] [Indexed: 12/17/2022] Open
Abstract
Aging of the heart is associated with a blunted response to sympathetic stimulation, reduced contractility, and increased propensity for arrhythmias, with the risk of sudden cardiac death significantly increased in the elderly population. The altered cardiac structural and functional phenotype, as well as age-associated prevalent comorbidities including hypertension and atherosclerosis, predispose the heart to atrial fibrillation, heart failure, and ventricular tachyarrhythmias. At the cellular level, perturbations in mitochondrial function, excitation-contraction coupling, and calcium homeostasis contribute to this electrical and contractile dysfunction. Major determinants of cardiac contractility are the intracellular release of Ca2+ from the sarcoplasmic reticulum by the ryanodine receptors (RyR2), and the following sequestration of Ca2+ by the sarco/endoplasmic Ca2+-ATPase (SERCa2a). Activity of RyR2 and SERCa2a in myocytes is not only dependent on expression levels and interacting accessory proteins, but on fine-tuned regulation via post-translational modifications. In this paper, we review how aberrant changes in intracellular Ca2+ cycling via these proteins contributes to arrhythmogenesis in the aged heart.
Collapse
Affiliation(s)
- Shanna Hamilton
- Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH 43210, USA.
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH 43210, USA.
| | - Dmitry Terentyev
- Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH 43210, USA.
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
20
|
Targeting protein-protein interactions for therapeutic discovery via FRET-based high-throughput screening in living cells. Sci Rep 2018; 8:12560. [PMID: 30135432 PMCID: PMC6105598 DOI: 10.1038/s41598-018-29685-z] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 07/16/2018] [Indexed: 01/16/2023] Open
Abstract
We have developed a structure-based high-throughput screening (HTS) method, using time-resolved fluorescence resonance energy transfer (TR-FRET) that is sensitive to protein-protein interactions in living cells. The membrane protein complex between the cardiac sarcoplasmic reticulum Ca-ATPase (SERCA2a) and phospholamban (PLB), its Ca-dependent regulator, is a validated therapeutic target for reversing cardiac contractile dysfunction caused by aberrant calcium handling. However, efforts to develop compounds with SERCA2a-PLB specificity have yet to yield an effective drug. We co-expressed GFP-SERCA2a (donor) in the endoplasmic reticulum membrane of HEK293 cells with RFP-PLB (acceptor), and measured FRET using a fluorescence lifetime microplate reader. We screened a small-molecule library and identified 21 compounds (Hits) that changed FRET by >3SD. 10 of these Hits reproducibly alter SERCA2a-PLB structure and function. One compound increases SERCA2a calcium affinity in cardiac membranes but not in skeletal, suggesting that the compound is acting specifically on the SERCA2a-PLB complex, as needed for a drug to mitigate deficient calcium transport in heart failure. The excellent assay quality and correlation between structural and functional assays validate this method for large-scale HTS campaigns. This approach offers a powerful pathway to drug discovery for a wide range of protein-protein interaction targets that were previously considered “undruggable”.
Collapse
|
21
|
Cannata' A, Merlo M, Artico J, Gentile P, Camparini L, Cristallini J, Porcari A, Loffredo F, Sinagra G. Cardiovascular aging: the unveiled enigma from bench to bedside. J Cardiovasc Med (Hagerstown) 2018; 19:517-526. [PMID: 30024423 DOI: 10.2459/jcm.0000000000000694] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
: The rapid increase in the median age of the world's population requires particular attention towards older and more fragile people. Cardiovascular risk factors, time and comorbidities play a vicious role in the development of heart failure, both with reduced and preserved ejection fraction, in the elderly. Understanding the mechanisms underlying the pathophysiological processes observed with aging is pivotal to target those patients and their therapeutic needs properly. This review aims to investigate and to dissect the main pathways leading to the aging cardiomyopathy, helping to understand the relationship from bench to bedside of the clinical phenotype.
Collapse
Affiliation(s)
- Antonio Cannata'
- Cardiovascular and Thoracic Department, Azienda Sanitaria Universitaria Integrata di Trieste and University of Trieste.,International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| | - Marco Merlo
- Cardiovascular and Thoracic Department, Azienda Sanitaria Universitaria Integrata di Trieste and University of Trieste
| | - Jessica Artico
- Cardiovascular and Thoracic Department, Azienda Sanitaria Universitaria Integrata di Trieste and University of Trieste
| | - Piero Gentile
- Cardiovascular and Thoracic Department, Azienda Sanitaria Universitaria Integrata di Trieste and University of Trieste
| | - Luca Camparini
- International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| | - Jacopo Cristallini
- Cardiovascular and Thoracic Department, Azienda Sanitaria Universitaria Integrata di Trieste and University of Trieste
| | - Aldostefano Porcari
- Cardiovascular and Thoracic Department, Azienda Sanitaria Universitaria Integrata di Trieste and University of Trieste
| | - Francesco Loffredo
- Cardiovascular and Thoracic Department, Azienda Sanitaria Universitaria Integrata di Trieste and University of Trieste.,International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| | - Gianfranco Sinagra
- Cardiovascular and Thoracic Department, Azienda Sanitaria Universitaria Integrata di Trieste and University of Trieste
| |
Collapse
|
22
|
Guo W, Pencina KM, Gagliano-Jucá T, Jasuja R, Morris N, O'Connell KE, Westmoreland S, Bhasin S. Effects of an ActRIIB.Fc Ligand Trap on Cardiac Function in Simian Immunodeficiency Virus-Infected Male Rhesus Macaques. J Endocr Soc 2018; 2:817-831. [PMID: 30019021 PMCID: PMC6041778 DOI: 10.1210/js.2018-00099] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 05/22/2018] [Indexed: 12/13/2022] Open
Abstract
An important safety consideration in the use of antagonists of myostatin and activins is whether these drugs induce myocardial hypertrophy and impair cardiac function. The current study evaluated the effects of a soluble ActRIIB receptor Fc fusion protein (ActRIIB.Fc), a ligand trap for TGF-β/activin family members including myostatin, on myocardial mass and function in simian immunodeficiency virus (SIV)-infected juvenile rhesus macaques (Macaca mulatta). Fourteen pair-housed, juvenile male rhesus macaques were inoculated with SIVmac239; 4 weeks postinoculation, they were treated with weekly injections of 10 mg/kg ActRIIB.Fc or saline for 12 weeks. Myocardial mass and function were evaluated using two-dimensional echocardiography at baseline and after 12 weeks. The administration of ActRIIB.Fc was associated with a significantly greater increase in thickness of left ventricular posterior wall and interventricular septum both in diastole and systole. Cardiac output and cardiac index increased with time, more in animals treated with ActRIIB.Fc than in those treated with saline, but the difference was not statistically significant. The changes in ejection fraction, fractional shortening, and stroke volume did not differ significantly between groups. The changes in end-diastolic and end-systolic volumes did not differ between groups. In addition to a large reduction in IGF1 mRNA expression in the ActRIIB.Fc-treated animals, complex changes were detected in the myocardial expression of proteins related to calcium transport and storage. In conclusion, ActRIIB.Fc administration for 12 weeks was associated with increased myocardial mass but did not adversely affect myocardial function in juvenile SIV-infected rhesus macaques. Further studies are necessary to establish long-term cardiac safety.
Collapse
Affiliation(s)
- Wen Guo
- Research Program in Men's Health: Aging and Metabolism, Boston Claude D. Pepper Older Americans Independence Center, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Karol M Pencina
- Research Program in Men's Health: Aging and Metabolism, Boston Claude D. Pepper Older Americans Independence Center, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Thiago Gagliano-Jucá
- Research Program in Men's Health: Aging and Metabolism, Boston Claude D. Pepper Older Americans Independence Center, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Ravi Jasuja
- Research Program in Men's Health: Aging and Metabolism, Boston Claude D. Pepper Older Americans Independence Center, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Nancy Morris
- Division of Comparative Pathology, New England Primate Research Center, Southborough, Massachusetts
| | - Karyn E O'Connell
- Division of Comparative Pathology, New England Primate Research Center, Southborough, Massachusetts
| | - Susan Westmoreland
- Division of Comparative Pathology, New England Primate Research Center, Southborough, Massachusetts
| | - Shalender Bhasin
- Research Program in Men's Health: Aging and Metabolism, Boston Claude D. Pepper Older Americans Independence Center, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
23
|
Zhai Y, Luo Y, Wu P, Li D. New insights into SERCA2a gene therapy in heart failure: pay attention to the negative effects of B-type natriuretic peptides. J Med Genet 2018; 55:287-296. [PMID: 29478009 DOI: 10.1136/jmedgenet-2017-105120] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 01/30/2018] [Accepted: 02/05/2018] [Indexed: 12/28/2022]
Abstract
Sarcoplasmic/endoplasmic reticulum calcium ATPase 2a (SERCA2a) is a target of interest in gene therapy for heart failure with reduced ejection fraction (HFrEF). However, the results of an important clinical study, the Calcium Upregulation by Percutaneous Administration of Gene Therapy in Cardiac Disease (CUPID) trial, were controversial. Promising results were observed in the CUPID 1 trial, but the results of the CUPID 2 trial were negative. The factors that caused the controversial results remain unclear. Importantly, enrolled patients were required to have a higher plasma level of B-type natriuretic peptide (BNP) in the CUPID 2 trial. Moreover, BNP was shown to inhibit SERCA2a expression. Therefore, it is possible that high BNP levels interact with treatment effects of SERCA2a gene transfer and accordingly lead to negative results of CUPID 2 trial. From this point of view, effects of SERCA2a gene therapy should be explored in heart failure with preserved ejection fraction, which is characterised by lower BNP levels compared with HFrEF. In this review, we summarise the current knowledge of SERCA2a gene therapy for heart failure, analyse potential interaction between BNP levels and therapeutic effects of SERCA2a gene transfer and provide directions for future research to solve the identified problems.
Collapse
Affiliation(s)
- Yuting Zhai
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yuanyuan Luo
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Pei Wu
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Dongye Li
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, Jiangsu, China
| |
Collapse
|
24
|
Mattila M, Koskenvuo J, Söderström M, Eerola K, Savontaus M. Intramyocardial injection of SERCA2a-expressing lentivirus improves myocardial function in doxorubicin-induced heart failure. J Gene Med 2018; 18:124-33. [PMID: 27203155 DOI: 10.1002/jgm.2885] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Revised: 04/19/2016] [Accepted: 05/17/2016] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Doxorubicin is an effective anticancer drug. The major limitation to its use is the induction of dose-dependent cardiomyopathy. No specific treatment exists for doxorubicin-induced cardiomyopathy and treatments used for other forms of heart failure have only limited beneficial effects. The contraction-relaxation cycle of the heart is controlled by cytosolic calcium concentrations, which, in turn, are critically regulated by the activity of the sarcoplasmic reticulum Ca(2) (+) ATPase (SERCA2a) pump. We hypothesized that SERCA2a gene transfer would ameliorate doxorubicin-induced cardiomyopathy. METHODS Lentiviral vectors LV-SERCA2a-GFP and LV-GFP were constructed and in vitro gene transfer of LV-SERCA2a-GFP confirmed SERCA2a expression by western blot analysis. Heart failure was induced by giving a single intraperitoneal injection of doxorubicin. LV-SERCA2a-GFP, LV-GFP vectors and phosphate-buffered saline (PBS) were injected under echocardiographic control to the anterior wall of the left ventricle. RESULTS Echocardiography analyses were performed on the injection day and 28 days postinjection. On the injection day, there were no significant differences in the average ejection fractions (EFs) among SERCA2a (40.0%), GFP (41.1%) and PBS (39.4%) injected animals. On day 28, EF in the SERCA2a group had increased by 16.6 ± 6.7% to 46.4 ± 2.1%. By contrast, EFs in the GFP (40.2 ± 1.3%) and PBS (40.6 ± 1.4%) groups remained at pre-injection levels. In addition, end systolic and end diastolic left ventricle volumes were significantly smaller in the SERCA2a group compared to controls. CONCLUSIONS SERCA2a gene transfer significantly improves left ventricle function and dimensions in doxorubicin-induced cardiomyopathy, thus making LV-SERCA2a gene transfer an attractive treatment modality for doxorubicin-induced heart failure. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Minttu Mattila
- Turku Centre for Biotechnology, University of Turku, Turku, Finland.,Department of Medical Biochemistry and Genetics, University of Turku, Turku, Finland.,Department of Pharmacology, Drug Development and Therapeutics, University of Turku, Turku, Finland.,Drug Research Doctoral Program, University of Turku, Turku, Finland
| | - Juha Koskenvuo
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland.,Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, University of Turku, Turku, Finland
| | - Mirva Söderström
- Department of Pathology, Turku University Hospital, University of Turku, Turku, Finland
| | - Kim Eerola
- Turku Centre for Biotechnology, University of Turku, Turku, Finland.,Department of Pharmacology, Drug Development and Therapeutics, University of Turku, Turku, Finland.,Drug Research Doctoral Program, University of Turku, Turku, Finland
| | - Mikko Savontaus
- Turku Centre for Biotechnology, University of Turku, Turku, Finland.,Department of Medical Biochemistry and Genetics, University of Turku, Turku, Finland.,Heart Centre, Turku University Hospital, University of Turku, Turku, Finland
| |
Collapse
|
25
|
Clarke JD, Caldwell JL, Pearman CM, Eisner DA, Trafford AW, Dibb KM. Increased Ca buffering underpins remodelling of Ca 2+ handling in old sheep atrial myocytes. J Physiol 2017; 595:6263-6279. [PMID: 28752958 PMCID: PMC5621500 DOI: 10.1113/jp274053] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2017] [Accepted: 07/26/2017] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS Ageing is associated with an increased risk of cardiovascular disease and arrhythmias, with the most common arrhythmia being found in the atria of the heart. Little is known about how the normal atria of the heart remodel with age and thus why dysfunction might occur. We report alterations to the atrial systolic Ca2+ transient that have implications for the function of the atrial in the elderly. We describe a novel mechanism by which increased Ca buffering can account for changes to systolic Ca2+ in the old atria. The present study helps us to understand how the processes regulating atrial contraction are remodelled during ageing and provides a basis for future work aiming to understand why dysfunction develops. ABSTRACT Many cardiovascular diseases, including those affecting the atria, are associated with advancing age. Arrhythmias, including those in the atria, can arise as a result of electrical remodelling or alterations in Ca2+ homeostasis. In the atria, age-associated changes in the action potential have been documented. However, little is known about remodelling of intracellular Ca2+ homeostasis in the healthy aged atria. Using single atrial myocytes from young and old Welsh Mountain sheep, we show the free Ca2+ transient amplitude and rate of decay of systolic Ca2+ decrease with age, whereas sarcoplasmic reticulum (SR) Ca content increases. An increase in intracellular Ca buffering explains both the decrease in Ca2+ transient amplitude and decay kinetics in the absence of any change in sarcoendoplasmic reticulum calcium transport ATPase function. Ageing maintained the integrated Ca2+ influx via ICa-L but decreased peak ICa-L . Decreased peak ICa-L was found to be responsible for the age-associated increase in SR Ca content but not the decrease in Ca2+ transient amplitude. Instead, decreased peak ICa-L offsets increased SR load such that Ca2+ release from the SR was maintained during ageing. The results of the present study highlight a novel mechanism by which increased Ca buffering decreases systolic Ca2+ in old atria. Furthermore, for the first time, we have shown that SR Ca content is increased in old atrial myocytes.
Collapse
Affiliation(s)
- Jessica D. Clarke
- Unit of Cardiac Physiology, Manchester Academic Health Sciences Centre, Central Manchester Foundation Trust, 3.14 Core Technology FacilityUniversity of ManchesterManchesterUK
| | - Jessica L. Caldwell
- Unit of Cardiac Physiology, Manchester Academic Health Sciences Centre, Central Manchester Foundation Trust, 3.14 Core Technology FacilityUniversity of ManchesterManchesterUK
| | - Charles M. Pearman
- Unit of Cardiac Physiology, Manchester Academic Health Sciences Centre, Central Manchester Foundation Trust, 3.14 Core Technology FacilityUniversity of ManchesterManchesterUK
| | - David A. Eisner
- Unit of Cardiac Physiology, Manchester Academic Health Sciences Centre, Central Manchester Foundation Trust, 3.14 Core Technology FacilityUniversity of ManchesterManchesterUK
| | - Andrew W. Trafford
- Unit of Cardiac Physiology, Manchester Academic Health Sciences Centre, Central Manchester Foundation Trust, 3.14 Core Technology FacilityUniversity of ManchesterManchesterUK
| | - Katharine M. Dibb
- Unit of Cardiac Physiology, Manchester Academic Health Sciences Centre, Central Manchester Foundation Trust, 3.14 Core Technology FacilityUniversity of ManchesterManchesterUK
| |
Collapse
|
26
|
Verjans R, van Bilsen M, Schroen B. MiRNA Deregulation in Cardiac Aging and Associated Disorders. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2017; 334:207-263. [PMID: 28838539 DOI: 10.1016/bs.ircmb.2017.03.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The prevalence of age-related diseases is increasing dramatically, among which cardiac disease represents the leading cause of death. Aging of the heart is characterized by various molecular and cellular hallmarks impairing both cardiomyocytes and noncardiomyocytes, and resulting in functional deteriorations of the cardiac system. The aging process includes desensitization of β-adrenergic receptor (βAR)-signaling and decreased calcium handling, altered growth signaling and cardiac hypertrophy, mitochondrial dysfunction and impaired autophagy, increased programmed cell death, low-grade inflammation of noncanonical inflammatory cells, and increased ECM deposition. MiRNAs play a fundamental role in regulating the processes underlying these detrimental changes in the cardiac system, indicating that MiRNAs are crucially involved in aging. Among others, MiR-34, MiR-146a, and members of the MiR-17-92 cluster, are deregulated during senescence and drive cardiac aging processes. It is therefore suggested that MiRNAs form possible therapeutic targets to stabilize the aged failing myocardium.
Collapse
Affiliation(s)
- Robin Verjans
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Marc van Bilsen
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Blanche Schroen
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands.
| |
Collapse
|
27
|
Ryu T, Song SY. Perioperative management of left ventricular diastolic dysfunction and heart failure: an anesthesiologist's perspective. Korean J Anesthesiol 2017; 70:3-12. [PMID: 28184260 PMCID: PMC5296384 DOI: 10.4097/kjae.2017.70.1.3] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 12/19/2016] [Accepted: 12/19/2016] [Indexed: 12/12/2022] Open
Abstract
Anesthesiologists frequently see asymptomatic patients with diastolic dysfunction or heart failure for various surgeries. These patients typically show normal systolic function but abnormal diastolic parameters in their preoperative echocardiographic evaluations. The symptoms that are sometimes seen are similar to those of chronic obstructive pulmonary disease. Patients with diastolic dysfunction, and even with diastolic heart failure, have the potential to develop a hypertensive crisis or pulmonary congestion. Thus, in addition to conventional perioperative risk quantification, it may be important to consider the results of diastolic assessment for predicting the postoperative outcome and making better decisions. If anesthesiologists see female patients older than 70 years of age who have hypertension, diabetes, chronic renal disease, recent weight gain, or exercise intolerance, they should focus on the patient's diastologic echocardiography indicators such as left atrial enlargement or left ventricular hypertrophy. In addition, there is a need for perioperative strategies to mitigate diastolic dysfunction-related morbidity. Specifically, hypertension should be controlled, keeping pulse pressure below diastolic blood pressure, maintaining a sinus rhythm and normovolemia, and avoiding tachycardia and myocardial ischemia. There is no need to classify these diastolic dysfunction, but it is important to manage this condition to avoid worsening outcomes.
Collapse
Affiliation(s)
- Taeha Ryu
- Department of Anesthesiology and Pain Medicine, School of Medicine, Catholic University of Daegu, Daegu, Korea
| | - Seok-Young Song
- Department of Anesthesiology and Pain Medicine, School of Medicine, Catholic University of Daegu, Daegu, Korea
| |
Collapse
|
28
|
Cannatà A, Camparini L, Sinagra G, Giacca M, Loffredo FS. Pathways for salvage and protection of the heart under stress: novel routes for cardiac rejuvenation. Cardiovasc Res 2016; 111:142-53. [DOI: 10.1093/cvr/cvw106] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 05/10/2016] [Indexed: 01/07/2023] Open
|
29
|
Roh J, Rhee J, Chaudhari V, Rosenzweig A. The Role of Exercise in Cardiac Aging: From Physiology to Molecular Mechanisms. Circ Res 2016; 118:279-95. [PMID: 26838314 DOI: 10.1161/circresaha.115.305250] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Aging induces structural and functional changes in the heart that are associated with increased risk of cardiovascular disease and impaired functional capacity in the elderly. Exercise is a diagnostic and therapeutic tool, with the potential to provide insights into clinical diagnosis and prognosis, as well as the molecular mechanisms by which aging influences cardiac physiology and function. In this review, we first provide an overview of how aging impacts the cardiac response to exercise, and the implications this has for functional capacity in older adults. We then review the underlying molecular mechanisms by which cardiac aging contributes to exercise intolerance, and conversely how exercise training can potentially modulate aging phenotypes in the heart. Finally, we highlight the potential use of these exercise models to complement models of disease in efforts to uncover new therapeutic targets to prevent or treat heart disease in the aging population.
Collapse
Affiliation(s)
- Jason Roh
- From the Cardiovascular Division (J. Roh, J. Rhee, V.C., A.R.) and Department of Anesthesiology, Critical Care, and Pain Medicine (J. Rhee), Massachusetts General Hospital and Harvard Medical School, Boston
| | - James Rhee
- From the Cardiovascular Division (J. Roh, J. Rhee, V.C., A.R.) and Department of Anesthesiology, Critical Care, and Pain Medicine (J. Rhee), Massachusetts General Hospital and Harvard Medical School, Boston
| | - Vinita Chaudhari
- From the Cardiovascular Division (J. Roh, J. Rhee, V.C., A.R.) and Department of Anesthesiology, Critical Care, and Pain Medicine (J. Rhee), Massachusetts General Hospital and Harvard Medical School, Boston
| | - Anthony Rosenzweig
- From the Cardiovascular Division (J. Roh, J. Rhee, V.C., A.R.) and Department of Anesthesiology, Critical Care, and Pain Medicine (J. Rhee), Massachusetts General Hospital and Harvard Medical School, Boston.
| |
Collapse
|
30
|
Pan B, Xu ZW, Xu Y, Liu LJ, Zhu J, Wang X, Nan C, Zhang Z, Shen W, Huang XP, Tian J. Diastolic dysfunction and cardiac troponin I decrease in aging hearts. Arch Biochem Biophys 2016; 603:20-8. [PMID: 27184165 DOI: 10.1016/j.abb.2016.05.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2016] [Revised: 05/07/2016] [Accepted: 05/11/2016] [Indexed: 11/28/2022]
Abstract
Cardiac tropnoin I (cTnI) plays a critical role in the regulation of diastolic function, and its low expression may result in cardiac diastolic dysfunction, which is the most common form of cardiovascular disorders in older adults. In this study, cTnI expression levels were determined in mice at various ages and cardiac function was measured and compared between young adult mice (3 and 10 months) and older mice (18 months). The data indicated that the cTnI levels reached a peak high in young adult hearts (3 months), but decreased in older hearts (18 months). Furthermore, the older hearts showed a significant diastolic dysfunction observed by P-V loop and echocardiography measurements. To further define the mechanism underlying the cTnI decrease in aging hearts, we tested DNA methylation and histone acetylation modifications of cTnI gene. We found that acetylation of histone near the promoter region of cTnI gene played an important role in regulation of cTnI expression in the heart at different ages. Our study indicates that epigenetic modification caused cTnI expression decrease is one of the possible causes that result in a reduced cTnI level and diastolic dysfunction in the older hearts.
Collapse
Affiliation(s)
- B Pan
- Heart Centre, Children's Hospital of Chongqing Medical University, Chongqing, PR China; Key Laboratory of Developmental Disease in Childhood (Chongqing Medical University), Ministry of Education, Chongqing, PR China; Key Laboratory of Pediatrics in Chongqing, PR China; Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, PR China
| | - Z W Xu
- Heart Centre, Children's Hospital of Chongqing Medical University, Chongqing, PR China; Key Laboratory of Developmental Disease in Childhood (Chongqing Medical University), Ministry of Education, Chongqing, PR China; Key Laboratory of Pediatrics in Chongqing, PR China; Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, PR China
| | - Y Xu
- Heart Centre, Children's Hospital of Chongqing Medical University, Chongqing, PR China; Key Laboratory of Developmental Disease in Childhood (Chongqing Medical University), Ministry of Education, Chongqing, PR China; Key Laboratory of Pediatrics in Chongqing, PR China; Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, PR China
| | - L J Liu
- Key Laboratory of Developmental Disease in Childhood (Chongqing Medical University), Ministry of Education, Chongqing, PR China; Key Laboratory of Pediatrics in Chongqing, PR China; Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, PR China
| | - J Zhu
- Key Laboratory of Developmental Disease in Childhood (Chongqing Medical University), Ministry of Education, Chongqing, PR China; Key Laboratory of Pediatrics in Chongqing, PR China; Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, PR China
| | - X Wang
- Department of Biomedical Science, Charlie E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, USA
| | - C Nan
- Department of Biomedical Science, Charlie E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, USA
| | - Z Zhang
- Department of Biomedical Science, Charlie E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, USA
| | - W Shen
- Department of Biomedical Science, Charlie E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, USA
| | - X P Huang
- Department of Biomedical Science, Charlie E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, USA.
| | - J Tian
- Heart Centre, Children's Hospital of Chongqing Medical University, Chongqing, PR China.
| |
Collapse
|
31
|
Abstract
Cardiovascular disease (CVD) is a common problem in the elderly. In particular, the morbidity and mortality of patients with heart failure (HF) increase with age. The poor outcomes of elderly patients with HF can be explained partly by cardiac aging at the cellular and organ levels. Moreover, recent evidence has demonstrated that functional evaluation, which may reflect the status of individual aging, predicts mortality in patients with HF. Age-related changes occur throughout the body and in virtually all organ systems. Thus, we should pay more attention to geriatric conditions when treating patients with HF. Frailty represents a complex clinical syndrome that results from multiple impairments across different organs and is characterized by decreased physiological reserves and increased vulnerability to stressors. Frail patients with CVD have a worse prognosis than non-frail patients. Evidence demonstrates that frailty is an independent risk factor for incident HF among older people. The ways in which cellular senescence promotes age-related CVD and frailty remain an important issue in the biology of aging and clinical geriatrics. Senescent cells that have acquired a senescence-associated secretory phenotype (SASP) can cause local and potentially systemic inflammation. SASP might be a key phenomenon in the association between cellular senescence and the development of age-related CVD and frailty. Frailty is a dynamic and potentially reversible state; therefore, translational research efforts are focused on obtaining mechanistic insights into the pathobiology of frailty, the development of novel therapeutics, and the identification of biomarkers for frailty. This is particularly important in developed countries that are confronted with an aging society.
Collapse
|
32
|
Rammos C, Hendgen-Cotta UB, Totzeck M, Pohl J, Lüdike P, Flögel U, Deenen R, Köhrer K, French BA, Gödecke A, Kelm M, Rassaf T. Impact of dietary nitrate on age-related diastolic dysfunction. Eur J Heart Fail 2016; 18:599-610. [PMID: 27118445 DOI: 10.1002/ejhf.535] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 11/30/2015] [Accepted: 12/30/2015] [Indexed: 12/28/2022] Open
Abstract
AIMS Diastolic dysfunction is highly prevalent, and ageing is the main contributor due to impairments in active cardiac relaxation, ventriculo-vascular stiffening, and endothelial dysfunction. Nitric oxide (NO) affects cardiovascular functions, and NO bioavailability is critically reduced with ageing. Whether replenishment of NO deficiency with dietary inorganic nitrate would offer a novel approach to reverse age-related cardiovascular alterations was not known. METHODS AND RESULTS A dietary nitrate supplementation was applied to young (6 month) and old (20 month) wild-type mice for 8 weeks and compared with controls. High-resolution ultrasound, pressure-volume catheter techniques, and isolated heart measurements were applied to assess cardiac diastolic and vascular functions. Cardiac manganese-enhanced magnetic resonance imaging was performed to study the effects of dietary nitrate on myocyte calcium handling. In aged mice with preserved systolic function, dietary nitrate supplementation improved LV diastolic function, arterial compliance, and coronary flow reserve. Mechanistically, improved cardiovascular functions were associated with an accelerated cardiomyocyte calcium handling and augmented NO/cyclic guanosine monophosphate/protein kinase G signalling, while enhanced nitrate reduction was related to age-related differences in the oral microbiome. CONCLUSION Dietary inorganic nitrate reverses age-related LV diastolic dysfunction and improves vascular functions. Our results highlight the potential of a dietary approach in the therapy of age-related cardiovascular alterations.
Collapse
Affiliation(s)
- Christos Rammos
- West-German Heart and Vascular Center Essen, Department of Medicine, Division of Cardiology, Medical Faculty, University Hospital Essen, Essen, Germany
| | - Ulrike B Hendgen-Cotta
- West-German Heart and Vascular Center Essen, Department of Medicine, Division of Cardiology, Medical Faculty, University Hospital Essen, Essen, Germany
| | - Matthias Totzeck
- West-German Heart and Vascular Center Essen, Department of Medicine, Division of Cardiology, Medical Faculty, University Hospital Essen, Essen, Germany
| | - Julia Pohl
- West-German Heart and Vascular Center Essen, Department of Medicine, Division of Cardiology, Medical Faculty, University Hospital Essen, Essen, Germany
| | - Peter Lüdike
- West-German Heart and Vascular Center Essen, Department of Medicine, Division of Cardiology, Medical Faculty, University Hospital Essen, Essen, Germany
| | - Ulrich Flögel
- Department of Molecular Cardiology, Heinrich-Heine-University, Düsseldorf, Germany
| | - René Deenen
- Biological and Medical Research Center (BMFZ), Genomics and Transcriptomics Laboratory, Heinrich-Heine-University, Düsseldorf, Germany
| | - Karl Köhrer
- Biological and Medical Research Center (BMFZ), Genomics and Transcriptomics Laboratory, Heinrich-Heine-University, Düsseldorf, Germany
| | - Brent A French
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Axel Gödecke
- Department of Cardiovascular Physiology, Heinrich-Heine-University, Düsseldorf, Germany
| | - Malte Kelm
- Department of Medicine, Division of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Tienush Rassaf
- West-German Heart and Vascular Center Essen, Department of Medicine, Division of Cardiology, Medical Faculty, University Hospital Essen, Essen, Germany
| |
Collapse
|
33
|
Alencar AK, da Silva JS, Lin M, Silva AM, Sun X, Ferrario CM, Cheng C, Sudo RT, Zapata-Sudo G, Wang H, Groban L. Effect of Age, Estrogen Status, and Late-Life GPER Activation on Cardiac Structure and Function in the Fischer344×Brown Norway Female Rat. J Gerontol A Biol Sci Med Sci 2016; 72:152-162. [PMID: 27006078 DOI: 10.1093/gerona/glw045] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 02/25/2016] [Indexed: 01/08/2023] Open
Abstract
Age-associated changes in cardiac structure and function, together with estrogen loss, contribute to the progression of heart failure with preserved ejection fraction in older women. To investigate the effects of aging and estrogen loss on the development of its precursor, asymptomatic left ventricular diastolic dysfunction, echocardiograms were performed in 10 middle-aged (20 months) and 30 old-aged (30 months) female Fischer344×Brown-Norway rats, 4 and 8 weeks after ovariectomy (OVX) and sham procedures (gonads left intact). The cardioprotective potential of administering chronic G1, the selective agonist to the new G-protein-coupled estrogen receptor (GPER), was further evaluated in old rats (Old-OVX+G1) versus age-matched, vehicle-treated OVX and gonadal intact rats. Advanced age and estrogen loss led to decreases in myocardial relaxation and elevations in filling pressure, in part, due to reductions in phosphorylated phospholamban and increases in cardiac collagen deposition. Eight weeks of G-protein-coupled estrogen receptor activation in Old-OVX+G1 rats reversed the adverse effects of age and estrogen loss on myocardial relaxation through increases in sarcoplasmic reticulum Ca2+ ATPase expression and reductions in interstitial fibrosis. These findings may explain the preponderance of heart failure with preserved ejection fraction in older postmenopausal women and provide a promising, late-life therapeutic target to reverse or halt the progression of left ventricular diastolic dysfunction.
Collapse
Affiliation(s)
- Allan K Alencar
- Department of Anesthesiology, Wake Forest School of Medicine, Winston-Salem, North Carolina.,Programa de Pesquisa em Desenvolvimento de Fármacos, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Jaqueline S da Silva
- Programa de Pesquisa em Desenvolvimento de Fármacos, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marina Lin
- Department of Anesthesiology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Ananssa M Silva
- Programa de Pesquisa em Desenvolvimento de Fármacos, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Xuming Sun
- Department of Anesthesiology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Carlos M Ferrario
- Department of Internal Medicine, Section on Nephrology, Wake Forest School of Medicine, Winston-Salem, North Carolina.,Department of Surgery, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Cheping Cheng
- Department of Internal Medicine, Section on Cardiovascular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Roberto T Sudo
- Programa de Pesquisa em Desenvolvimento de Fármacos, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Gisele Zapata-Sudo
- Programa de Pesquisa em Desenvolvimento de Fármacos, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Hao Wang
- Department of Anesthesiology, Wake Forest School of Medicine, Winston-Salem, North Carolina.,Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Leanne Groban
- Department of Anesthesiology, Wake Forest School of Medicine, Winston-Salem, North Carolina. .,Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina.,The Hypertension and Vascular Research Center, Wake Forest School of Medicine, Winston-Salem, North Carolina.,The Sticht Center on Aging, Wake Forest School of Medicine, Winston-Salem, North Carolina
| |
Collapse
|
34
|
Ye LF, Zheng YR, Wang LH. Effects of Shenmai injection and its bioactive components following ischemia/reperfusion in cardiomyocytes. Exp Ther Med 2015; 10:1348-1354. [PMID: 26622490 PMCID: PMC4578100 DOI: 10.3892/etm.2015.2662] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 06/24/2015] [Indexed: 12/20/2022] Open
Abstract
The aim of the present study was to determine whether the myocardial protective function of Shenmai injection (SM) during ischemia/reperfusion (I/R) is attributable to its regulation of intracellular calcium (Ca2+) and phospholamban (PLB) levels. Cultured neonatal Sprague Dawley rat cardiomyocytes were used to compare the effects of normoxia, total saponins of Panax ginseng (TSPG), ginsenoside Rg1 (Rg1) and SM treatments in rat myocardial cells following I/R. For each of these treatment groups, the mRNA and protein levels of PLB and the sarco/endoplasmic reticulum Ca2+ ATPase (SERCA) were evaluated, in addition to the cytoplasmic Ca2+ concentration [Ca2+]i and the rate of apoptosis. The results indicated that I/R markedly decreased phosphorylated PLB and SERCA expression and that SM was able to mitigate this effect, while TPSG and Rg1 were not. Furthermore, SM appeared to prevent aberrant apoptosis and restore the depleted [Ca2+]i resulting from I/R. The protective efficacy of SM against heart disease following I/R may, therefore, be due in part to its effect on intracellular Ca2+ homeostasis. SM may exert its protective effects by relieving PLB inhibition, and the pharmacodynamic actions of SM appear to be significantly more effective than those of its bioreactive components, TPSG and Rgl.
Collapse
Affiliation(s)
- Li-Fang Ye
- Department of Cardiology, First Affiliated Hospital, Medical College of Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| | - Ya-Ru Zheng
- Department of Cardiology, First Affiliated Hospital, Medical College of Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| | - Li-Hong Wang
- Department of Cardiology, People's Hospital of Zhejiang Province, Hangzhou, Zhejiang 310014, P.R. China
| |
Collapse
|
35
|
Feridooni HA, Dibb KM, Howlett SE. How cardiomyocyte excitation, calcium release and contraction become altered with age. J Mol Cell Cardiol 2015; 83:62-72. [PMID: 25498213 DOI: 10.1016/j.yjmcc.2014.12.004] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Revised: 12/02/2014] [Accepted: 12/04/2014] [Indexed: 11/29/2022]
Abstract
Cardiovascular disease is the main cause of death globally, accounting for over 17 million deaths each year. As the incidence of cardiovascular disease rises markedly with age, the overall risk of cardiovascular disease is expected to increase dramatically with the aging of the population such that by 2030 it could account for over 23 million deaths per year. It is therefore vitally important to understand how the heart remodels in response to normal aging for at least two reasons: i) to understand why the aged heart is increasingly susceptible to disease; and ii) since it may be possible to modify treatment of disease in older adults if the underlying substrate upon which the disease first develops is fully understood. It is well known that age modulates cardiac function at the level of the individual cardiomyocyte. Generally, in males, aging reduces cell shortening, which is associated with a decrease in the amplitude of the systolic Ca(2+) transient. This may arise due to a decrease in peak L-type Ca(2+) current. Sarcoplasmic reticulum (SR) Ca(2+) load appears to be maintained during normal aging but evidence suggests that SR function is disrupted, such that the rate of sarco/endoplasmic reticulum Ca(2+)-ATPase (SERCA)-mediated Ca(2+) removal is reduced and the properties of SR Ca(2+) release in terms of Ca(2+) sparks are altered. Interestingly, Ca(2+) handling is modulated by age to a lesser degree in females. Here we review how cellular contraction is altered as a result of the aging process by considering expression levels and functional properties of key proteins involved in controlling intracellular Ca(2+). We consider how changes in both electrical properties and intracellular Ca(2+) handling may interact to modulate cardiomyocyte contraction. We also reflect on why cardiovascular risk may differ between the sexes by highlighting sex-specific variation in the age-associated remodeling process. This article is part of a Special Issue entitled CV Aging.
Collapse
Affiliation(s)
- Hirad A Feridooni
- Department of Pharmacology, Dalhousie University, PO Box 15000, 5850 College St, Halifax, NS B3H 4R2, Canada.
| | - Katharine M Dibb
- Institute of Cardiovascular Sciences, University of Manchester, Manchester, UK.
| | - Susan E Howlett
- Department of Pharmacology, Dalhousie University, PO Box 15000, 5850 College St, Halifax, NS B3H 4R2, Canada; Department of Medicine (Geriatric Medicine), Dalhousie University, PO Box 15000, 5850 College St, Halifax, NS B3H 4R2, Canada; Institute of Cardiovascular Sciences, University of Manchester, Manchester, UK.
| |
Collapse
|
36
|
Revuelta-López E, Cal R, Herraiz-Martínez A, de Gonzalo-Calvo D, Nasarre L, Roura S, Gálvez-Montón C, Bayes-Genis A, Badimon L, Hove-Madsen L, Llorente-Cortés V. Hypoxia-driven sarcoplasmic/endoplasmic reticulum calcium ATPase 2 (SERCA2) downregulation depends on low-density lipoprotein receptor-related protein 1 (LRP1)-signalling in cardiomyocytes. J Mol Cell Cardiol 2015; 85:25-36. [PMID: 25968337 DOI: 10.1016/j.yjmcc.2015.04.028] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 04/29/2015] [Accepted: 04/30/2015] [Indexed: 01/30/2023]
Abstract
The maintenance of sarcoplasmic reticulum Ca(2+) ATPase (SERCA2) activity is crucial for cardiac function and SERCA2 is dramatically reduced in the heart exposed to hypoxic/ischemic conditions. Previous work from our group showed that hypoxia upregulates the phosphorylated form of the Ca(2+)-dependent nonreceptor protein tyrosine kinase (PTK) proline-rich tyrosine kinase 2 (pPyk2) protein levels in a low-density lipoprotein receptor-related protein (LRP1)-dependent manner. Pyk2 in turn may modulate SERCA2 in cardiomyocytes although this remains controversial. We therefore aimed to investigate the role of LRP1 on hypoxia-induced SERCA2 depletion in cardiomyocytes and to establish LRP1 signalling mechanisms involved. Western blot analysis showed that hypoxia reduced SERCA2 concomitantly with a sustained increase in LRP1 and pPyk2 protein levels in HL-1 cardiomyocytes. By impairing hypoxia-induced Pyk2 phosphorylation and HIF-1α accumulation, LRP1 deficiency prevented SERCA2 depletion and reduction of the sarcoplasmic reticulum calcium content in cardiomyocytes. Moreover, the inhibition of Pyk2 phosphorylation (with the Src-family inhibitor PP2) or the specific silencing of Pyk2 (with siRNA-anti Pyk2) preserved low HIF-1α and high SERCA2 levels in HL-1 cardiomyocytes exposed to hypoxia. We determined that the LRP1/Pyk2 axis represses SERCA2 mRNA expression via HIF-1α since HIF-1α overexpression abolished the protective effect of LRP1 deficiency on SERCA2 depletion. Our findings show a crucial role of LRP1/Pyk2/HIF-1α in hypoxia-induced cardiomyocyte SERCA2 downregulation, a pathophysiological process closely associated with heart failure.
Collapse
Affiliation(s)
| | - Roi Cal
- Cardiovascular Research Center, CSIC-ICCC, IIB-Sant Pau, Barcelona, Spain
| | | | | | - Laura Nasarre
- Cardiovascular Research Center, CSIC-ICCC, IIB-Sant Pau, Barcelona, Spain
| | - Santiago Roura
- ICREC Research Program, Health Science Research Institute Germans Trias i Pujol, Can Ruti Campus, Badalona, Spain
| | - Carolina Gálvez-Montón
- ICREC Research Program, Health Science Research Institute Germans Trias i Pujol, Can Ruti Campus, Badalona, Spain
| | - Antoni Bayes-Genis
- ICREC Research Program, Health Science Research Institute Germans Trias i Pujol, Can Ruti Campus, Badalona, Spain
| | - Lina Badimon
- Cardiovascular Research Center, CSIC-ICCC, IIB-Sant Pau, Barcelona, Spain
| | - Leif Hove-Madsen
- Cardiovascular Research Center, CSIC-ICCC, IIB-Sant Pau, Barcelona, Spain
| | | |
Collapse
|
37
|
Butler L, Cros C, Oldman KL, Harmer AR, Pointon A, Pollard CE, Abi-Gerges N. Enhanced Characterization of Contractility in Cardiomyocytes During Early Drug Safety Assessment. Toxicol Sci 2015; 145:396-406. [DOI: 10.1093/toxsci/kfv062] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
|
38
|
Overexpression of cyclic adenosine monophosphate effluent protein MRP4 induces an altered response to β-adrenergic stimulation in the senescent rat heart. Anesthesiology 2015; 122:334-42. [PMID: 25383567 DOI: 10.1097/aln.0000000000000526] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND In the senescent heart, the positive inotropic response to β-adrenoceptor stimulation is reduced, partly by dysregulation of β1- and β3-adrenoceptors. The multidrug resistance protein 4 (MRP4) takes part in the control of intracellular cyclic adenosine monophosphate concentration by controlling its efflux but the role of MRP4 in the β-adrenergic dysfunction of the senescent heart remains unknown. METHODS The β-adrenergic responses to isoproterenol were investigated in vivo (stress echocardiography) and in vitro (isolated cardiomyocyte by Ionoptix with sarcomere shortening and calcium transient) in young (3 months old) and senescent (24 months old) rats pretreated or not with MK571, a specific MRP4 inhibitor. MRP4 was quantified in left ventricular homogenates by Western blotting. Data are mean ± SD expressed as percent of baseline value. RESULTS The positive inotropic effect of isoproterenol was reduced in senescent rats in vivo (left ventricular shortening fraction 120 ± 16% vs. 158 ± 20%, P < 0.001, n = 16 rats) and in vitro (sarcomere shortening 129 ± 37% vs. 148 ± 35%, P = 0.004, n = 41 or 43 cells) as compared to young rats. MRP4 expression increased 3.6-fold in senescent compared to young rat myocardium (P = 0.012, n = 8 rats per group). In senescent rats, inhibition of MRP4 by MK571 restored the positive inotropic effect of isoproterenol in vivo (143 ± 11%, n = 8 rats). In vitro in senescent cardiomyocytes pretreated with MK571, both sarcomere shortening (161 ± 45% vs. 129 ± 37%, P = 0.007, n = 41 cells per group) and calcium transient amplitude (132 ± 25% vs. 113 ± 27%, P = 0.007) increased significantly. CONCLUSION MRP4 overexpression contributes to the reduction of the positive inotropic response to β-adrenoceptor stimulation in the senescent heart.
Collapse
|
39
|
Loffredo FS, Nikolova AP, Pancoast JR, Lee RT. Heart failure with preserved ejection fraction: molecular pathways of the aging myocardium. Circ Res 2014; 115:97-107. [PMID: 24951760 DOI: 10.1161/circresaha.115.302929] [Citation(s) in RCA: 134] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Age-related diastolic dysfunction is a major factor in the epidemic of heart failure. In patients hospitalized with heart failure, HFpEF is now as common as heart failure with reduced ejection fraction. We now have many successful treatments for heart failure with reduced ejection fraction, while specific treatment options for HFpEF patients remain elusive. The lack of treatments for HFpEF reflects our very incomplete understanding of this constellation of diseases. There are many pathophysiological factors in HFpEF, but aging appears to play an important role. Here, we propose that aging of the myocardium is itself a specific pathophysiological process. New insights into the aging heart, including hormonal controls and specific molecular pathways, such as microRNAs, are pointing to myocardial aging as a potentially reversible process. While the overall process of aging remains mysterious, understanding the molecular pathways of myocardial aging has never been more important. Unraveling these pathways could lead to new therapies for the enormous and growing problem of HFpEF.
Collapse
Affiliation(s)
- Francesco S Loffredo
- From the Department of Stem Cell and Regenerative Biology, Harvard University, Brigham Regenerative Medicine Center, Cambridge, MA; Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA; and Harvard Stem Cell Institute, Cambridge, MA
| | - Andriana P Nikolova
- From the Department of Stem Cell and Regenerative Biology, Harvard University, Brigham Regenerative Medicine Center, Cambridge, MA; Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA; and Harvard Stem Cell Institute, Cambridge, MA
| | - James R Pancoast
- From the Department of Stem Cell and Regenerative Biology, Harvard University, Brigham Regenerative Medicine Center, Cambridge, MA; Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA; and Harvard Stem Cell Institute, Cambridge, MA
| | - Richard T Lee
- From the Department of Stem Cell and Regenerative Biology, Harvard University, Brigham Regenerative Medicine Center, Cambridge, MA; Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA; and Harvard Stem Cell Institute, Cambridge, MA.
| |
Collapse
|
40
|
Gladden JD, Linke WA, Redfield MM. Heart failure with preserved ejection fraction. Pflugers Arch 2014; 466:1037-53. [PMID: 24663384 PMCID: PMC4075067 DOI: 10.1007/s00424-014-1480-8] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Revised: 02/07/2014] [Accepted: 02/08/2014] [Indexed: 01/06/2023]
Abstract
As part of this series devoted to heart failure (HF), we review the epidemiology, diagnosis, pathophysiology, and treatment of HF with preserved ejection fraction (HFpEF). Gaps in knowledge and needed future research are discussed.
Collapse
Affiliation(s)
- James D. Gladden
- Division of Cardiovascular Diseases, Department of Medicine, Mayo Clinic, Guggenheim 9, 200 First Street, Southwest Rochester, MN 55905, USA
| | - Wolfgang A. Linke
- Department of Cardiovascular Physiology, Ruhr University Bochum, Bochum, Germany
| | - Margaret M. Redfield
- Division of Cardiovascular Diseases, Department of Medicine, Mayo Clinic, Guggenheim 9, 200 First Street, Southwest Rochester, MN 55905, USA,
| |
Collapse
|
41
|
Qin F, Siwik DA, Pimentel DR, Morgan RJ, Biolo A, Tu VH, Kang YJ, Cohen RA, Colucci WS. Cytosolic H2O2 mediates hypertrophy, apoptosis, and decreased SERCA activity in mice with chronic hemodynamic overload. Am J Physiol Heart Circ Physiol 2014; 306:H1453-63. [PMID: 24633550 DOI: 10.1152/ajpheart.00084.2014] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Oxidative stress in the myocardium plays an important role in the pathophysiology of hemodynamic overload. The mechanism by which reactive oxygen species (ROS) in the cardiac myocyte mediate myocardial failure in hemodynamic overload is not known. Accordingly, our goals were to test whether myocyte-specific overexpression of peroxisomal catalase (pCAT) that localizes in the sarcoplasm protects mice from hemodynamic overload-induced failure and prevents oxidation and inhibition of sarco(endo)plasmic reticulum Ca(2+)-ATPase (SERCA), an important sarcoplasmic protein. Chronic hemodynamic overload was caused by ascending aortic constriction (AAC) for 12 wk in mice with myocyte-specific transgenic expression of pCAT. AAC caused left ventricular hypertrophy and failure associated with a generalized increase in myocardial oxidative stress and specific oxidative modifications of SERCA at cysteine 674 and tyrosine 294/5. pCAT overexpression ameliorated myocardial hypertrophy and apoptosis, decreased pathological remodeling, and prevented the progression to heart failure. Likewise, pCAT prevented oxidative modifications of SERCA and increased SERCA activity without changing SERCA expression. Thus cardiac myocyte-restricted expression of pCAT effectively ameliorated the structural and functional consequences of chronic hemodynamic overload and increased SERCA activity via a post-translational mechanism, most likely by decreasing inhibitory oxidative modifications. In pressure overload-induced heart failure cardiac myocyte cytosolic ROS play a pivotal role in mediating key pathophysiologic events including hypertrophy, apoptosis, and decreased SERCA activity.
Collapse
Affiliation(s)
- Fuzhong Qin
- From the Cardiovascular Medicine Section, Department of Medicine, and the Myocardial and Vascular Biology Units, Boston University Medical Center, Boston, Massachusetts
| | - Deborah A Siwik
- From the Cardiovascular Medicine Section, Department of Medicine, and the Myocardial and Vascular Biology Units, Boston University Medical Center, Boston, Massachusetts
| | - David R Pimentel
- From the Cardiovascular Medicine Section, Department of Medicine, and the Myocardial and Vascular Biology Units, Boston University Medical Center, Boston, Massachusetts
| | - Robert J Morgan
- From the Cardiovascular Medicine Section, Department of Medicine, and the Myocardial and Vascular Biology Units, Boston University Medical Center, Boston, Massachusetts
| | - Andreia Biolo
- From the Cardiovascular Medicine Section, Department of Medicine, and the Myocardial and Vascular Biology Units, Boston University Medical Center, Boston, Massachusetts
| | - Vivian H Tu
- From the Cardiovascular Medicine Section, Department of Medicine, and the Myocardial and Vascular Biology Units, Boston University Medical Center, Boston, Massachusetts
| | - Y James Kang
- From the Cardiovascular Medicine Section, Department of Medicine, and the Myocardial and Vascular Biology Units, Boston University Medical Center, Boston, Massachusetts
| | - Richard A Cohen
- From the Cardiovascular Medicine Section, Department of Medicine, and the Myocardial and Vascular Biology Units, Boston University Medical Center, Boston, Massachusetts
| | - Wilson S Colucci
- From the Cardiovascular Medicine Section, Department of Medicine, and the Myocardial and Vascular Biology Units, Boston University Medical Center, Boston, Massachusetts
| |
Collapse
|
42
|
Winter J, Brack KE, Ng GA. Cardiac contractility modulation in the treatment of heart failure: initial results and unanswered questions. Eur J Heart Fail 2014; 13:700-10. [DOI: 10.1093/eurjhf/hfr042] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- James Winter
- Department of Cardiovascular Sciences; University of Leicester; Clinical Sciences Wing, Glenfield Hospital Leicester LE3 9QP UK
| | - Kieran E. Brack
- Department of Cardiovascular Sciences; University of Leicester; Clinical Sciences Wing, Glenfield Hospital Leicester LE3 9QP UK
| | - G. André Ng
- Department of Cardiovascular Sciences; University of Leicester; Clinical Sciences Wing, Glenfield Hospital Leicester LE3 9QP UK
- Leicester NIHR Biomedical Research Unit in Cardiovascular Disease; Glenfield Hospital; Leicester LE3 9QP UK
| |
Collapse
|
43
|
Akasheva DU, Plokhova EV, Strazhesko ID, Dudinskaya EN, Tkacheva ON. HEART AND AGE (PART III): MODIFYING AGEING PROCESSES. КАРДИОВАСКУЛЯРНАЯ ТЕРАПИЯ И ПРОФИЛАКТИКА 2013. [DOI: 10.15829/1728-8800-2013-5-91-96] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
|
44
|
Qin F, Siwik DA, Lancel S, Zhang J, Kuster GM, Luptak I, Wang L, Tong X, Kang YJ, Cohen RA, Colucci WS. Hydrogen peroxide-mediated SERCA cysteine 674 oxidation contributes to impaired cardiac myocyte relaxation in senescent mouse heart. J Am Heart Assoc 2013; 2:e000184. [PMID: 23963753 PMCID: PMC3828801 DOI: 10.1161/jaha.113.000184] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Background A hallmark of aging of the cardiac myocyte is impaired sarcoplasmic reticulum (SR) calcium uptake and relaxation due to decreased SR calcium ATPase (SERCA) activity. We tested the hypothesis that H2O2‐mediated oxidation of SERCA contributes to impaired myocyte relaxation in aging. Methods and Results Young (5‐month‐old) and senescent (21‐month‐old) FVB wild‐type (WT) or transgenic mice with myocyte‐specific overexpression of catalase were studied. In senescent mice, myocyte‐specific overexpression of catalase (1) prevented oxidative modification of SERCA as evidenced by sulfonation at Cys674, (2) preserved SERCA activity, (3) corrected impaired calcium handling and relaxation in isolated cardiac myocytes, and (4) prevented impaired left ventricular relaxation and diastolic dysfunction. Nitroxyl, which activates SERCA via S‐glutathiolation at Cys674, failed to activate SERCA in freshly isolated ventricular myocytes from senescent mice. Finally, in adult rat ventricular myocytes in primary culture, adenoviral overexpression of SERCA in which Cys674 is mutated to serine partially preserved SERCA activity during exposure to H2O2. Conclusion Oxidative modification of SERCA at Cys674 contributes to decreased SERCA activity and impaired myocyte relaxation in the senescent heart. Strategies to decrease oxidant levels and/or protect target proteins such as SERCA may be of value to preserve diastolic function in the aging heart.
Collapse
Affiliation(s)
- Fuzhong Qin
- Cardiovascular Medicine Section, Department of Medicine, The Myocardial Biology Unit and Vascular Biology Section, Boston University Medical Center, Boston, MA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Dhalla NS, Rangi S, Babick AP, Zieroth S, Elimban V. Cardiac remodeling and subcellular defects in heart failure due to myocardial infarction and aging. Heart Fail Rev 2013; 17:671-81. [PMID: 21850540 DOI: 10.1007/s10741-011-9278-7] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Although several risk factors including hypertension, cardiac hypertrophy, coronary artery disease, and diabetes are known to result in heart failure, elderly subjects are more susceptible to myocardial infarction and more likely to develop heart failure. This article is intended to discuss that cardiac dysfunction in hearts failing due to myocardial infarction and aging is associated with cardiac remodeling and defects in the subcellular organelles such as sarcolemma (SL), sarcoplasmic reticulum (SR), and myofibrils. Despite some differences in the pattern of heart failure due to myocardial infarction and aging with respect to their etiology and sequence of events, evidence has been presented to show that subcellular remodeling plays a critical role in the occurrence of intracellular Ca(2+)-overload and development of cardiac dysfunction in both types of failing heart. In particular, alterations in gene expression for SL and SR proteins induce Ca(2+)-handling abnormalities in cardiomyocytes, whereas those for myofibrillar proteins impair the interaction of Ca(2+) with myofibrils in hearts failing due to myocardial infarction and aging. In addition, different phosphorylation mechanisms, which regulate the activities of Ca(2+)-cycling proteins in SL and SR membranes as well as Ca(2+)-binding proteins in myofibrils, become defective in the failing heart. Accordingly, it is suggested that subcellular remodeling involving defects in Ca(2+)-handling and Ca(2+)-binding proteins as well as their regulatory mechanisms is intimately associated with cardiac remodeling and heart failure due to myocardial infarction and aging.
Collapse
Affiliation(s)
- Naranjan S Dhalla
- Institute of Cardiovascular Sciences, St. Boniface General Hospital Research Centre, 351 Tache Avenue, Winnipeg, MB, Canada.
| | | | | | | | | |
Collapse
|
46
|
Tang T, Hammond HK. Gene transfer for congestive heart failure: update 2013. Transl Res 2013; 161:313-20. [PMID: 23261978 PMCID: PMC3602385 DOI: 10.1016/j.trsl.2012.11.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2012] [Revised: 11/19/2012] [Accepted: 11/27/2012] [Indexed: 01/08/2023]
Abstract
Congestive heart failure is a major cause of morbidity and mortality with increasing social and economic costs. There have been no new high impact therapeutic agents for this devastating disease for more than a decade. However, many pivotal regulators of cardiac function have been identified using cardiac-directed transgene expression and gene deletion in preclinical studies. Some of these increase function of the failing heart. Altering the expression of these pivotal regulators using gene transfer is now either being tested in clinical gene transfer trials, or soon will be. In this review, we summarize recent progress in cardiac gene transfer for clinical congestive heart failure.
Collapse
Affiliation(s)
- Tong Tang
- Department of Medicine, University of California San Diego, and VA San Diego Healthcare System, San Diego, Calif., USA
| | | |
Collapse
|
47
|
Wright DB, Trian T, Siddiqui S, Pascoe CD, Johnson JR, Dekkers BG, Dakshinamurti S, Bagchi R, Burgess JK, Kanabar V, Ojo OO. Phenotype modulation of airway smooth muscle in asthma. Pulm Pharmacol Ther 2013; 26:42-9. [DOI: 10.1016/j.pupt.2012.08.005] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2012] [Revised: 08/11/2012] [Accepted: 08/13/2012] [Indexed: 01/26/2023]
|
48
|
Siddiqui S, Redhu NS, Ojo OO, Liu B, Irechukwu N, Billington C, Janssen L, Moir LM. Emerging airway smooth muscle targets to treat asthma. Pulm Pharmacol Ther 2012; 26:132-44. [PMID: 22981423 DOI: 10.1016/j.pupt.2012.08.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2012] [Revised: 07/28/2012] [Accepted: 08/27/2012] [Indexed: 11/26/2022]
Abstract
Asthma is characterized in part by variable airflow obstruction and non-specific hyperresponsiveness to a variety of bronchoconstrictors, both of which are mediated by the airway smooth muscle (ASM). The ASM is also involved in the airway inflammation and airway wall remodeling observed in asthma. For all these reasons, the ASM provides an important target for the treatment of asthma. Several classes of drugs were developed decades ago which targeted the ASM - including β-agonists, anti-cholinergics, anti-histamines and anti-leukotrienes - but no substantially new class of drug has appeared recently. In this review, we summarize the on-going work of several laboratories aimed at producing novel targets and/or tools for the treatment of asthma. These range from receptors and ion channels on the ASM plasmalemma, to intracellular effectors (particularly those related to cyclic nucleotide signaling, calcium-homeostasis and phosphorylation cascades), to anti-IgE therapy and outright destruction of the ASM itself.
Collapse
Affiliation(s)
- Sana Siddiqui
- Meakins-Christie Laboratories, Department of Medicine, McGill University, 3626 St Urbain, Montréal, Québec H2X 2P2, Canada
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Babušíková E, Lehotský J, Dobrota D, Račay P, Kaplán P. Age-associated changes in Ca(2+)-ATPase and oxidative damage in sarcoplasmic reticulum of rat heart. Physiol Res 2012; 61:453-60. [PMID: 22881224 DOI: 10.33549/physiolres.932320] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Altered Ca(2+) handling may be responsible for the development of cardiac contractile dysfunctions with advanced age. In the present study, we investigated the roles of oxidative damage to sarcoplasmic reticulum (SR) and expression of Ca(2+)-ATPase (SERCA 2a) and phospholamban in age-associated dysfunction of cardiac SR. SR vesicles were prepared from hearts of 2-, 6-, 15-, and 26-month-old Wistar rats. Although activity of Ca(2+)-ATPase decreased with advancing age, no differences in relative amounts of SERCA 2a and phospholamban protein were observed. On the other hand, significant accumulation of protein oxidative damage occurred with aging. The results of this study suggest that age-related alteration in Ca(2+)-ATPase activity in the rat heart is not a consequence of decreased protein levels of SERCA 2a and phospholamban, but could arise from oxidative modifications of SR proteins. Cellular oxidative damage caused by reactive oxygen species could contribute to age-related alternations in myocardial relaxation.
Collapse
Affiliation(s)
- E Babušíková
- Department of Medical Biochemistry, Jessenius Faculty of Medicine, Comenius University, Martin, Slovak Republic
| | | | | | | | | |
Collapse
|
50
|
ElGuindy A, Yacoub MH. Heart failure with preserved ejection fraction. Glob Cardiol Sci Pract 2012; 2012:10. [PMID: 25610841 PMCID: PMC4239812 DOI: 10.5339/gcsp.2012.10] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2012] [Accepted: 05/23/2012] [Indexed: 12/13/2022] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) has recently emerged as a major cause of cardiovascular morbidity and mortality. Contrary to initial beliefs, HFpEF is now known to be as common as heart failure with reduced ejection fraction (HFrEF) and carries an unacceptably high mortality rate. With a prevalence that has been steadily rising over the past two decades, it is very likely that HFpEF will represent the dominant heart failure phenotype over the coming few years. The scarcity of trials in this semi-discrete form of heart failure and lack of unified enrolment criteria in the studies conducted to date might have contributed to the current absence of specific therapies. Understanding the epidemiological, pathophysiological and molecular differences (and similarities) between these two forms of heart failure is cornerstone to the development of targeted therapies. Carefully designed studies that adhere to unified diagnostic criteria with the recruitment of appropriate controls and adoption of practical end-points are urgently needed to help identify effective treatment strategies.
Collapse
Affiliation(s)
- Ahmed ElGuindy
- Department of Cardiology, Aswan Heart Centre, Aswan, Egypt
| | - Magdi H Yacoub
- Harefield Heart Science Centre, National Heart and Lung Institute, Imperial College London, UK
| |
Collapse
|