1
|
Bertero E, Popoiu TA, Maack C. Mitochondrial calcium in cardiac ischemia/reperfusion injury and cardioprotection. Basic Res Cardiol 2024; 119:569-585. [PMID: 38890208 PMCID: PMC11319510 DOI: 10.1007/s00395-024-01060-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 05/31/2024] [Accepted: 06/01/2024] [Indexed: 06/20/2024]
Abstract
Mitochondrial calcium (Ca2+) signals play a central role in cardiac homeostasis and disease. In the healthy heart, mitochondrial Ca2+ levels modulate the rate of oxidative metabolism to match the rate of adenosine triphosphate consumption in the cytosol. During ischemia/reperfusion (I/R) injury, pathologically high levels of Ca2+ in the mitochondrial matrix trigger the opening of the mitochondrial permeability transition pore, which releases solutes and small proteins from the matrix, causing mitochondrial swelling and ultimately leading to cell death. Pharmacological and genetic approaches to tune mitochondrial Ca2+ handling by regulating the activity of the main Ca2+ influx and efflux pathways, i.e., the mitochondrial Ca2+ uniporter and sodium/Ca2+ exchanger, represent promising therapeutic strategies to protect the heart from I/R injury.
Collapse
Affiliation(s)
- Edoardo Bertero
- Department of Translational Research, Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, Am Schwarzenberg 15, Haus A15, 97078, Würzburg, Germany
- Chair of Cardiovascular Disease, Department of Internal Medicine and Specialties (Di.M.I.), University of Genoa, Genoa, Italy
| | - Tudor-Alexandru Popoiu
- Department of Translational Research, Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, Am Schwarzenberg 15, Haus A15, 97078, Würzburg, Germany
- "Victor Babes" University of Medicine and Pharmacy, Timisoara, Romania
| | - Christoph Maack
- Department of Translational Research, Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, Am Schwarzenberg 15, Haus A15, 97078, Würzburg, Germany.
| |
Collapse
|
2
|
Shi RR, He TQ, Lin MS, Xu J, Gu JH, Xu H. O-GlcNAcylation in ischemic diseases. Front Pharmacol 2024; 15:1377235. [PMID: 38783961 PMCID: PMC11113977 DOI: 10.3389/fphar.2024.1377235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Accepted: 04/15/2024] [Indexed: 05/25/2024] Open
Abstract
Protein glycosylation is an extensively studied field, with the most studied forms being oxygen or nitrogen-linked N-acetylglucosamine (O-GlcNAc or N-GlcNAc) glycosylation. Particular residues on proteins are targeted by O-GlcNAcylation, which is among the most intricate post-translational modifications. Significantly contributing to an organism's proteome, it influences numerous factors affecting protein stability, function, and subcellular localization. It also modifies the cellular function of target proteins that have crucial responsibilities in controlling pathways related to the central nervous system, cardiovascular homeostasis, and other organ functions. Under conditions of acute stress, changes in the levels of O-GlcNAcylation of these proteins may have a defensive function. Nevertheless, deviant O-GlcNAcylation nullifies this safeguard and stimulates the advancement of several ailments, the prognosis of which relies on the cellular milieu. Hence, this review provides a concise overview of the function and comprehension of O-GlcNAcylation in ischemia diseases, aiming to facilitate the discovery of new therapeutic targets for efficient treatment, particularly in patients with diabetes.
Collapse
Affiliation(s)
- Rui-Rui Shi
- Nantong Institute of Genetics and Reproductive Medicine, Affiliated Maternity and Child Healthcare Hospital of Nantong University, Nantong, China
| | - Tian-Qi He
- Nantong Institute of Genetics and Reproductive Medicine, Affiliated Maternity and Child Healthcare Hospital of Nantong University, Nantong, China
- Department of Pharmacy, Affiliated Maternity and Child Healthcare Hospital of Nantong University, Nantong, China
| | - Meng-Si Lin
- Prenatal Screening and Diagnosis Center, Affiliated Maternity and Child Healthcare Hospital of Nantong University, Nantong, China
| | - Jian Xu
- Nantong Institute of Genetics and Reproductive Medicine, Affiliated Maternity and Child Healthcare Hospital of Nantong University, Nantong, China
- Department of Pharmacy, Affiliated Maternity and Child Healthcare Hospital of Nantong University, Nantong, China
| | - Jin-Hua Gu
- Nantong Institute of Genetics and Reproductive Medicine, Affiliated Maternity and Child Healthcare Hospital of Nantong University, Nantong, China
- Department of Pharmacy, Affiliated Maternity and Child Healthcare Hospital of Nantong University, Nantong, China
| | - Hui Xu
- Nantong Institute of Genetics and Reproductive Medicine, Affiliated Maternity and Child Healthcare Hospital of Nantong University, Nantong, China
| |
Collapse
|
3
|
Appleby S, Aitken-Buck HM, Holdaway MS, Byers MS, Frampton CM, Paton LN, Richards AM, Lamberts RR, Pemberton CJ. Cardiac effects of myoregulin in ischemia-reperfusion. Peptides 2024; 174:171156. [PMID: 38246425 DOI: 10.1016/j.peptides.2024.171156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 01/15/2024] [Accepted: 01/17/2024] [Indexed: 01/23/2024]
Abstract
Myoregulin is a recently discovered micropeptide that controls calcium levels by inhibiting the intracellular calcium pump sarco-endoplasmic reticulum Ca2+-ATPase (SERCA). Keeping calcium levels balanced in the heart is essential for normal heart functioning, thus myoregulin has the potential to be a crucial regulator of cardiac muscle performance by reducing the rate of intracellular Ca2+ uptake. We provide the first report of myoregulin mRNA expression in human heart tissue, absence of expression in human plasma, and the effects of myoregulin on cardiac hemodynamics in an ex vivo Langendorff isolated rat heart model of ischemia/reperfusion. In this preliminary study, myoregulin provided a cardio-protective effect, as assessed by preservation of left ventricular contractility and relaxation, during ischemia/reperfusion. This study provides the foundation for future research in this area.
Collapse
Affiliation(s)
- Sarah Appleby
- Christchurch Heart Institute, University of Otago, Christchurch, 2 Riccarton Avenue, Christchurch 8011, New Zealand.
| | - Hamish M Aitken-Buck
- Department of Physiology, HeartOtago, University of Otago, 270 Great King St, Dunedin 9016, New Zealand.
| | - Mark S Holdaway
- Christchurch Heart Institute, University of Otago, Christchurch, 2 Riccarton Avenue, Christchurch 8011, New Zealand.
| | - Mathew S Byers
- Christchurch Heart Institute, University of Otago, Christchurch, 2 Riccarton Avenue, Christchurch 8011, New Zealand.
| | - Chris M Frampton
- Christchurch Heart Institute, University of Otago, Christchurch, 2 Riccarton Avenue, Christchurch 8011, New Zealand.
| | - Louise N Paton
- Christchurch Heart Institute, University of Otago, Christchurch, 2 Riccarton Avenue, Christchurch 8011, New Zealand.
| | - A Mark Richards
- Christchurch Heart Institute, University of Otago, Christchurch, 2 Riccarton Avenue, Christchurch 8011, New Zealand; Department of Cardiology, Te Whatu Ora Waitaha, 2 Riccarton Avenue, Christchurch 8011, New Zealand; Cardiovascular Research Institute, National University of Singapore, 1E Kent Ridge Road, Singapore.
| | - Regis R Lamberts
- Department of Physiology, HeartOtago, University of Otago, 270 Great King St, Dunedin 9016, New Zealand.
| | - Christopher J Pemberton
- Christchurch Heart Institute, University of Otago, Christchurch, 2 Riccarton Avenue, Christchurch 8011, New Zealand.
| |
Collapse
|
4
|
Dimitrov AG. Resting membrane state as an interplay of electrogenic transporters with various pumps. Pflugers Arch 2023; 475:1113-1128. [PMID: 37468808 DOI: 10.1007/s00424-023-02838-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 06/26/2023] [Accepted: 07/06/2023] [Indexed: 07/21/2023]
Abstract
In this study, a new idea that electrogenic transporters determine cell resting state is presented. The previous assumption was that pumps, especially the sodium one, determine it. The latter meets difficulties, because it violates the law of conservation of energy; also a significant deficit of pump activity is reported. The amount of energy carried by a single ATP molecule reflects the potential of the inner mitochondrial membrane, which is about -200 mV. If pumps enforce a resting membrane potential that is more than twice smaller, then the majority of energy stored in ATP would be dissipated by each pump turning. However, this problem could be solved if control is transferred from pumps to something else, e.g., electrogenic transporters. Then pumps would transfer the energy to the ionic gradient without losses, while the cell surface membrane potential would be associated with the reversal potential of some electrogenic transporters. A minimal scheme of this type would include a sodium-calcium exchanger as well as sodium and calcium pumps. However, note that calcium channels and pumps are positioned along both intracellular organelles and the surface membrane. Therefore, the above-mentioned scheme would involve them as well as possible intercellular communications. Such schemes where various kinds of pumps are assumed to work in parallel may explain, to a great extent, the slow turning rate of the individual members. Interaction of pumps and transporters positioned at distant biological membranes with various forms of energy transfer between them may thus result in hypoxic/reperfusion injury, different kinds of muscle fatigue, and nerve-glia interactions.
Collapse
Affiliation(s)
- A G Dimitrov
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., Bl. 105, 1113, Sofia, Bulgaria.
| |
Collapse
|
5
|
Lozano O, Marcos P, Salazar-Ramirez FDJ, Lázaro-Alfaro AF, Sobrevia L, García-Rivas G. Targeting the mitochondrial Ca 2+ uniporter complex in cardiovascular disease. Acta Physiol (Oxf) 2023; 237:e13946. [PMID: 36751976 DOI: 10.1111/apha.13946] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 02/02/2023] [Accepted: 02/03/2023] [Indexed: 02/09/2023]
Abstract
Cardiovascular diseases (CVDs), the leading cause of death worldwide, share in common mitochondrial dysfunction, in specific a dysregulation of Ca2+ uptake dynamics through the mitochondrial Ca2+ uniporter (MCU) complex. In particular, Ca2+ uptake regulates the mitochondrial ATP production, mitochondrial dynamics, oxidative stress, and cell death. Therefore, modulating the activity of the MCU complex to regulate Ca2+ uptake, has been suggested as a potential therapeutic approach for the treatment of CVDs. Here, the role and implications of the MCU complex in CVDs are presented, followed by a review of the evidence for MCU complex modulation, genetically and pharmacologically. While most approaches have aimed within the MCU complex for the modulation of the Ca2+ pore channel, the MCU subunit, its intra- and extra- mitochondrial implications, including Ca2+ dynamics, oxidative stress, post-translational modifications, and its repercussions in the cardiac function, highlight that targeting the MCU complex has the translational potential for novel CVDs therapeutics.
Collapse
Affiliation(s)
- Omar Lozano
- Cátedra de Cardiología y Medicina Vascular, School of Medicine and Health Sciences, Tecnologico de Monterrey, Monterrey, Mexico
- Biomedical Research Center, Hospital Zambrano-Hellion, TecSalud, Tecnologico de Monterrey, San Pedro Garza García, Mexico
- The Institute for Obesity Research, Tecnologico de Monterrey, Monterrey, Mexico
| | - Patricio Marcos
- Cátedra de Cardiología y Medicina Vascular, School of Medicine and Health Sciences, Tecnologico de Monterrey, Monterrey, Mexico
| | - Felipe de Jesús Salazar-Ramirez
- Cátedra de Cardiología y Medicina Vascular, School of Medicine and Health Sciences, Tecnologico de Monterrey, Monterrey, Mexico
| | - Anay F Lázaro-Alfaro
- Cátedra de Cardiología y Medicina Vascular, School of Medicine and Health Sciences, Tecnologico de Monterrey, Monterrey, Mexico
| | - Luis Sobrevia
- The Institute for Obesity Research, Tecnologico de Monterrey, Monterrey, Mexico
- Cellular and Molecular Physiology Laboratory, Department of Obstetrics, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
- Department of Physiology, Faculty of Pharmacy, Universidad de Sevilla, Seville, Spain
- University of Queensland Centre for Clinical Research (UQCCR), Faculty of Medicine and Biomedical Sciences, University of Queensland, Herston, Queensland, Australia
| | - Gerardo García-Rivas
- Cátedra de Cardiología y Medicina Vascular, School of Medicine and Health Sciences, Tecnologico de Monterrey, Monterrey, Mexico
- Biomedical Research Center, Hospital Zambrano-Hellion, TecSalud, Tecnologico de Monterrey, San Pedro Garza García, Mexico
- The Institute for Obesity Research, Tecnologico de Monterrey, Monterrey, Mexico
- Center of Functional Medicine, Hospital Zambrano-Hellion, TecSalud, Tecnologico de Monterrey, San Pedro Garza García, Mexico
| |
Collapse
|
6
|
Alsahly MB, Zakari MO, Koch LG, Britton S, Katwa LC, Fisher-Wellman K, Lust RM. Augmented Cardiac Mitochondrial Capacity in High Capacity Aerobic Running "Disease-Resistant" Phenotype at Rest Is Lost Following Ischemia Reperfusion. Front Cardiovasc Med 2021; 8:752640. [PMID: 34805308 PMCID: PMC8595288 DOI: 10.3389/fcvm.2021.752640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 09/30/2021] [Indexed: 11/13/2022] Open
Abstract
Rationale: Regular active exercise is considered therapeutic for cardiovascular disease, in part by increasing mitochondrial respiratory capacity, but a significant amount of exercise capacity is determined genetically. Animal models, demonstrating either high capacity aerobic running (HCR) or low capacity aerobic running (LCR) phenotypes, have been developed to study the intrinsic contribution, with HCR rats subsequently characterized as "disease resistant" and the LCRs as "disease prone." Enhanced cardioprotection in HCRs has been variable and mutifactoral, but likely includes a metabolic component. These studies were conducted to determine the influence of intrinsic aerobic phenotype on cardiac mitochondrial function before and after ischemia and reperfusion. Methods: A total of 34 HCR and LCR rats were obtained from the parent colony at the University of Toledo, housed under sedentary conditions, and fed normal chow. LCR and HCR animals were randomly assigned to either control or ischemia-reperfusion (IR). On each study day, one HCR/LCR pair was anesthetized, and hearts were rapidly excised. In IR animals, the hearts were immediately flushed with iced hyperkalemic, hyperosmotic, cardioplegia solution, and subjected to global hypothermic ischemic arrest (80 min). Following the arrest, the hearts underwent warm reperfusion (120 min) using a Langendorff perfusion system. Following reperfusion, the heart was weighed and the left ventricle (LV) was isolated. A midventricular ring was obtained to estimate infarction size [triphenyltetrazolium chloride (TTC)] and part of the remaining tissue (~150 mg) was transferred to a homogenation buffer on ice. Isolated mitochondria (MITO) samples were prepared and used to determine respiratory capacity under different metabolic conditions. In control animals, MITO were obtained and prepared similarly immediately following anesthesia and heart removal, but without IR. Results: In the control rats, both resting and maximally stimulated respiratory rates were higher (32 and 40%, respectively; p < 0.05) in HCR mitochondria compared to LCR. After IR, resting MITO respiratory rates were decreased to about 10% of control in both strains, and the augmented capacity in HCRs was absent. Maximally stimulated rates also were decreased more than 50% from control and were no longer different between phenotypes. Ca++ retention capacity and infarct size were not significantly different between HCR and LCR (49.2 ± 5.6 vs. 53.7 ± 4.9%), nor was average coronary flow during reperfusion or arrhythmogenesis. There was a significant loss of mitochondria following IR, which was coupled with decreased function in the remaining mitochondria in both strains. Conclusion: Cardiac mitochondrial capacity from HCR was significantly higher than LCR in the controls under each condition. After IR insult, the cardiac mitochondrial respiratory rates were similar between phenotypes, as was Ca++ retention capacity, infarct size, and arrhythmogenicity, despite the increased mitochondrial capacity in the HCRs before ischemia. Relatively, the loss of respiratory capacity was actually greater in HCR than LCR. These data could suggest limits in the extent to which the HCR phenotype might be "protective" against acute tissue stressors. The extent to which any of these deficits could be "rescued" by adding an active exercise component to the intrinsic phenotype is unknown.
Collapse
Affiliation(s)
- Musaad B. Alsahly
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, United States
- East Carolina Diabetes and Obesity Center, East Carolina University, Greenville, NC, United States
| | - Madaniah O. Zakari
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, United States
- Department of Physiology, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Lauren G. Koch
- Department of Physiology, College of Medicine, Taibah University, Medina, Saudi Arabia
| | - Steven Britton
- Department of Physiology and Pharmacology, University of Toledo, Toledo, OH, United States
| | - Laxmansa C. Katwa
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, United States
| | - Kelsey Fisher-Wellman
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, United States
- Departments of Anesthesiology and Molecular and Integrative Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Robert M. Lust
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, United States
- Departments of Anesthesiology and Molecular and Integrative Medicine, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
7
|
Dynamic Regulation of Cysteine Oxidation and Phosphorylation in Myocardial Ischemia-Reperfusion Injury. Cells 2021; 10:cells10092388. [PMID: 34572037 PMCID: PMC8469016 DOI: 10.3390/cells10092388] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 09/06/2021] [Accepted: 09/07/2021] [Indexed: 02/02/2023] Open
Abstract
Myocardial ischemia-reperfusion (I/R) injury significantly alters heart function following infarct and increases the risk of heart failure. Many studies have sought to preserve irreplaceable myocardium, termed cardioprotection, but few, if any, treatments have yielded a substantial reduction in clinical I/R injury. More research is needed to fully understand the molecular pathways that govern cardioprotection. Redox mechanisms, specifically cysteine oxidations, are acute and key regulators of molecular signaling cascades mediated by kinases. Here, we review the role of reactive oxygen species in modifying cysteine residues and how these modifications affect kinase function to impact cardioprotection. This exciting area of research may provide novel insight into mechanisms and likely lead to new treatments for I/R injury.
Collapse
|
8
|
Zhao T, Wu W, Sui L, Huang Q, Nan Y, Liu J, Ai K. Reactive oxygen species-based nanomaterials for the treatment of myocardial ischemia reperfusion injuries. Bioact Mater 2021; 7:47-72. [PMID: 34466716 PMCID: PMC8377441 DOI: 10.1016/j.bioactmat.2021.06.006] [Citation(s) in RCA: 134] [Impact Index Per Article: 44.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 05/09/2021] [Accepted: 06/02/2021] [Indexed: 02/06/2023] Open
Abstract
Interventional coronary reperfusion strategies are widely adopted to treat acute myocardial infarction, but morbidity and mortality of acute myocardial infarction are still high. Reperfusion injuries are inevitable due to the generation of reactive oxygen species (ROS) and apoptosis of cardiac muscle cells. However, many antioxidant and anti-inflammatory drugs are largely limited by pharmacokinetics and route of administration, such as short half-life, low stability, low bioavailability, and side effects for treatment myocardial ischemia reperfusion injury. Therefore, it is necessary to develop effective drugs and technologies to address this issue. Fortunately, nanotherapies have demonstrated great opportunities for treating myocardial ischemia reperfusion injury. Compared with traditional drugs, nanodrugs can effectively increase the therapeutic effect and reduces side effects by improving pharmacokinetic and pharmacodynamic properties due to nanodrugs’ size, shape, and material characteristics. In this review, the biology of ROS and molecular mechanisms of myocardial ischemia reperfusion injury are discussed. Furthermore, we summarized the applications of ROS-based nanoparticles, highlighting the latest achievements of nanotechnology researches for the treatment of myocardial ischemia reperfusion injury. Cardiovascular diseases are the leading cause of death worldwide. Researches of the myocardial infarction pathology and development of new treatments have very important scientific significance in the biomedical field. Many nanomaterials have shown amazing therapeutic effects to reduce myocardial damage by eliminating ROS. Nanomaterials effectively reduced myocardial damage through eliminating ROS from NOXs, M-ETC, M-Ca2+, M-mPTP, and RIRR.
Collapse
Affiliation(s)
- Tianjiao Zhao
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410087, China.,Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410008, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410087, China
| | - Wei Wu
- Department of Geriatric Surgery, Xiangya Hospital, Central South University, Changsha, 410087, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410087, China
| | - Lihua Sui
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410008, China.,Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410008, China
| | - Qiong Huang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410087, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410087, China
| | - Yayun Nan
- Geriatric Medical Center, Ningxia People's Hospital, Yinchuan, 750003, China
| | - Jianhua Liu
- Department of Radiology, The Second Hospital of Jilin University, Changchun, 130041, China
| | - Kelong Ai
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410008, China.,Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410008, China
| |
Collapse
|
9
|
Daiber A, Steven S, Euler G, Schulz R. Vascular and Cardiac Oxidative Stress and Inflammation as Targets for Cardioprotection. Curr Pharm Des 2021; 27:2112-2130. [PMID: 33550963 DOI: 10.2174/1381612827666210125155821] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 11/11/2020] [Indexed: 11/22/2022]
Abstract
Cardiac and vascular diseases are often associated with increased oxidative stress and inflammation, and both may contribute to the disease progression. However, successful applications of antioxidants in the clinical setting are very rare and specific anti-inflammatory therapeutics only emerged recently. Reasons for this rely on the great diversity of oxidative stress and inflammatory cells that can either act as cardioprotective or cause tissue damage in the heart. Recent large-scale clinical trials found that highly specific anti-inflammatory therapies using monoclonal antibodies against cytokines resulted in lower cardiovascular mortality in patients with pre-existing atherosclerotic disease. In addition, unspecific antiinflammatory medication and established cardiovascular drugs with pleiotropic immunomodulatory properties such as angiotensin converting enzyme (ACE) inhibitors or statins have proven beneficial cardiovascular effects. Normalization of oxidative stress seems to be a common feature of these therapies, which can be explained by a close interaction/crosstalk of the cellular redox state and inflammatory processes. In this review, we give an overview of cardiac reactive oxygen species (ROS) sources and processes of cardiac inflammation as well as the connection of ROS and inflammation in ischemic cardiomyopathy in order to shed light on possible cardioprotective interventions.
Collapse
Affiliation(s)
- Andreas Daiber
- Department of Cardiology, Molecular Cardiology, University Medical Center Mainz, Mainz, Germany
| | - Sebastian Steven
- Department of Cardiology, Molecular Cardiology, University Medical Center Mainz, Mainz, Germany
| | - Gerhild Euler
- Institute of Physiology, Justus-Liebig University, Giessen, Germany
| | - Rainer Schulz
- Institute of Physiology, Justus-Liebig University, Giessen, Germany
| |
Collapse
|
10
|
Luo Y, Li Z, Ge P, Guo H, Li L, Zhang G, Xu C, Chen H. Comprehensive Mechanism, Novel Markers and Multidisciplinary Treatment of Severe Acute Pancreatitis-Associated Cardiac Injury - A Narrative Review. J Inflamm Res 2021; 14:3145-3169. [PMID: 34285540 PMCID: PMC8286248 DOI: 10.2147/jir.s310990] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 06/15/2021] [Indexed: 12/12/2022] Open
Abstract
Acute pancreatitis (AP) is one of the common acute abdominal inflammatory diseases in clinic with acute onset and rapid progress. About 20% of the patients will eventually develop into severe acute pancreatitis (SAP) characterized by a large number of inflammatory cells infiltration, gland flocculus flaky necrosis and hemorrhage, finally inducing systemic inflammatory response syndrome (SIRS) and multiple organ dysfunction syndrome (MODS). Pancreatic enzyme activation, intestinal endotoxemia (IETM), cytokine activation, microcirculation disturbance, autonomic nerve dysfunction and autophagy dysregulation all play an essential role in the occurrence and progression of SAP. Organ dysfunction is the main cause of early death in SAP. Acute kidney injury (AKI) and acute lung injury (ALI) are common, while cardiac injury (CI) is not, but the case fatality risk is high. Many basic studies have observed obvious ultrastructure change of heart in SAP, including myocardial edema, cardiac hypertrophy, myocardial interstitial collagen deposition. Moreover, in clinical practice, patients with SAP often presented various abnormal electrocardiogram (ECG) and cardiac function. Cases complicated with acute myocardial infarction and pericardial tamponade have also been reported and even result in stress cardiomyopathy. Due to the molecular mechanisms underlying SAP-associated cardiac injury (SACI) remain poorly understood, and there is no complete, unified treatment and sovereign remedy at present, this article reviews reports referring to the pathogenesis, potential markers and treatment methods of SACI in recent years, in order to improve the understanding of cardiac injury in severe pancreatitis.
Collapse
Affiliation(s)
- YaLan Luo
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, Liaoning, People's Republic of China.,Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, People's Republic of China.,Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, People's Republic of China
| | - ZhaoXia Li
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, People's Republic of China
| | - Peng Ge
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, Liaoning, People's Republic of China.,Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, People's Republic of China.,Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, People's Republic of China
| | - HaoYa Guo
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, Liaoning, People's Republic of China.,Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, People's Republic of China.,Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, People's Republic of China
| | - Lei Li
- Department of Vascular Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, People's Republic of China
| | - GuiXin Zhang
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, People's Republic of China
| | - CaiMing Xu
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, People's Republic of China
| | - HaiLong Chen
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, People's Republic of China
| |
Collapse
|
11
|
Seidlmayer LK, Hanson BJ, Thai PN, Schaefer S, Bers DM, Dedkova EN. PK11195 Protects From Cell Death Only When Applied During Reperfusion: Succinate-Mediated Mechanism of Action. Front Physiol 2021; 12:628508. [PMID: 34149440 PMCID: PMC8212865 DOI: 10.3389/fphys.2021.628508] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 04/28/2021] [Indexed: 11/13/2022] Open
Abstract
Aim: Reperfusion after myocardial ischemia causes cellular injury, in part due to changes in mitochondrial Ca2+ handling, oxidative stress, and myocyte energetics. We have previously shown that the 18-kDa translocator protein of the outer mitochondrial membrane (TSPO) can modulate Ca2+ handling. Here, we aim to evaluate the role of the TSPO in ischemia/reperfusion (I/R) injury. Methods: Rabbit ventricular myocytes underwent simulated acute ischemia (20 min) and reperfusion (at 15 min, 1 h, and 3 h) in the absence and presence of 50 μM PK11195, a TSPO inhibitor. Cell death was measured by lactate dehydrogenase (LDH) assay, while changes in mitochondrial Ca2+, membrane potential (ΔΨm), and reactive oxygen species (ROS) generation were monitored using confocal microscopy in combination with fluorescent indicators. Substrate utilization was measured with Biolog mitochondrial plates. Results: Cell death was increased by ~200% following I/R compared to control untreated ventricular myocytes. Incubation with 50 μM PK11195 during both ischemia and reperfusion did not reduce cell death but increased mitochondrial Ca2+ uptake and ROS generation. However, application of 50 μM PK11195 only at the onset and during reperfusion effectively protected against cell death. The large-scale oscillations in ΔΨm observed after ~1 h of reperfusion were significantly delayed by 1 μM cyclosporin A and almost completely prevented by 50 μM PK11195 applied during 3 h of reperfusion. After an initial increase, mitochondrial Ca2+, measured with Myticam, rapidly declined during 3 h of reperfusion after the initial transient increase. This decline was prevented by application of PK11195 at the onset and during reperfusion. PK11195 prevented a significant increase in succinate utilization following I/R and succinate-induced forward-mode ROS generation. Treatment with PK11195 was also associated with a significant increase in glutamate and a decrease in leucine utilization. Conclusion: PK11195 administered specifically at the moment of reperfusion limited ROS-induced ROS release and cell death, likely in part, by a shift from succinate to glutamate utilization. These data demonstrate a unique mechanism to limit cardiac injury after I/R.
Collapse
Affiliation(s)
- Lea K Seidlmayer
- Department of Cardiology, University Hospital Olbenburg, Olbenburg, Germany
| | - Benjamin J Hanson
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| | - Phung N Thai
- Department of Internal Medicine, Division of Cardiovascular Medicine, School of Medicine, University of California, Davis, Davis, CA, United States
| | - Saul Schaefer
- Department of Internal Medicine, Division of Cardiovascular Medicine, School of Medicine, University of California, Davis, Davis, CA, United States
| | - Donald M Bers
- Department of Pharmacology, School of Medicine, University of California, Davis, Davis, CA, United States
| | - Elena N Dedkova
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| |
Collapse
|
12
|
Regulation of Mitochondrial Homeostasis by sAC-Derived cAMP Pool: Basic and Translational Aspects. Cells 2021; 10:cells10020473. [PMID: 33671810 PMCID: PMC7926680 DOI: 10.3390/cells10020473] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 02/17/2021] [Accepted: 02/19/2021] [Indexed: 01/21/2023] Open
Abstract
In contrast to the traditional view of mitochondria being solely a source of cellular energy, e.g., the "powerhouse" of the cell, mitochondria are now known to be key regulators of numerous cellular processes. Accordingly, disturbance of mitochondrial homeostasis is a basic mechanism in several pathologies. Emerging data demonstrate that 3'-5'-cyclic adenosine monophosphate (cAMP) signalling plays a key role in mitochondrial biology and homeostasis. Mitochondria are equipped with an endogenous cAMP synthesis system involving soluble adenylyl cyclase (sAC), which localizes in the mitochondrial matrix and regulates mitochondrial function. Furthermore, sAC localized at the outer mitochondrial membrane contributes significantly to mitochondrial biology. Disturbance of the sAC-dependent cAMP pools within mitochondria leads to mitochondrial dysfunction and pathology. In this review, we discuss the available data concerning the role of sAC in regulating mitochondrial biology in relation to diseases.
Collapse
|
13
|
Abstract
Unlike acute myocardial infarction with reperfusion, in which infarct size is the end point reflecting irreversible injury, myocardial stunning and hibernation result from reversible myocardial ischaemia-reperfusion injury, and contractile dysfunction is the obvious end point. Stunned myocardium is characterized by a disproportionately long-lasting, yet fully reversible, contractile dysfunction that follows brief bouts of myocardial ischaemia. Reperfusion precipitates a burst of reactive oxygen species formation and alterations in excitation-contraction coupling, which interact and cause the contractile dysfunction. Hibernating myocardium is characterized by reduced regional contractile function and blood flow, which both recover after reperfusion or revascularization. Short-term myocardial hibernation is an adaptation of contractile function to the reduced blood flow such that energy and substrate metabolism recover during the ongoing ischaemia. Chronic myocardial hibernation is characterized by severe morphological alterations and altered expression of metabolic and pro-survival proteins. Myocardial stunning is observed clinically and must be recognized but is rarely haemodynamically compromising and does not require treatment. Myocardial hibernation is clinically identified with the use of imaging techniques, and the myocardium recovers after revascularization. Several trials in the past two decades have challenged the superiority of revascularization over medical therapy for symptomatic relief and prognosis in patients with chronic coronary syndromes. A better understanding of the pathophysiology of myocardial stunning and hibernation is important for a more precise indication of revascularization and its consequences. Therefore, this Review summarizes the current knowledge of the pathophysiology of these characteristic reperfusion phenomena and highlights their clinical implications.
Collapse
|
14
|
Dissecting Cellular Mechanisms of Long-Chain Acylcarnitines-Driven Cardiotoxicity: Disturbance of Calcium Homeostasis, Activation of Ca 2+-Dependent Phospholipases, and Mitochondrial Energetics Collapse. Int J Mol Sci 2020; 21:ijms21207461. [PMID: 33050414 PMCID: PMC7589681 DOI: 10.3390/ijms21207461] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 10/05/2020] [Accepted: 10/07/2020] [Indexed: 01/16/2023] Open
Abstract
Long-chain acylcarnitines (LCAC) are implicated in ischemia-reperfusion (I/R)-induced myocardial injury and mitochondrial dysfunction. Yet, molecular mechanisms underlying involvement of LCAC in cardiac injury are not sufficiently studied. It is known that in cardiomyocytes, palmitoylcarnitine (PC) can induce cytosolic Ca2+ accumulation, implicating L-type calcium channels, Na+/Ca2+ exchanger, and Ca2+-release from sarcoplasmic reticulum (SR). Alternatively, PC can evoke dissipation of mitochondrial potential (ΔΨm) and mitochondrial permeability transition pore (mPTP). Here, to dissect the complex nature of PC action on Ca2+ homeostasis and oxidative phosphorylation (OXPHOS) in cardiomyocytes and mitochondria, the methods of fluorescent microscopy, perforated path-clamp, and mitochondrial assays were used. We found that LCAC in dose-dependent manner can evoke Ca2+-sparks and oscillations, long-living Ca2+ enriched microdomains, and, finally, Ca2+ overload leading to hypercontracture and cardiomyocyte death. Collectively, PC-driven cardiotoxicity involves: (I) redistribution of Ca2+ from SR to mitochondria with minimal contribution of external calcium influx; (II) irreversible inhibition of Krebs cycle and OXPHOS underlying limited mitochondrial Ca2+ buffering; (III) induction of mPTP reinforced by PC-calcium interplay; (IV) activation of Ca2+-dependent phospholipases cPLA2 and PLC. Based on the inhibitory analysis we may suggest that simultaneous inhibition of both phospholipases could be an effective strategy for protection against PC-mediated toxicity in cardiomyocytes.
Collapse
|
15
|
Decreased Mitochondrial Function, Biogenesis, and Degradation in Peripheral Blood Mononuclear Cells from Amyotrophic Lateral Sclerosis Patients as a Potential Tool for Biomarker Research. Mol Neurobiol 2020; 57:5084-5102. [PMID: 32840822 PMCID: PMC7541388 DOI: 10.1007/s12035-020-02059-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 08/07/2020] [Indexed: 12/11/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a multifactorial and progressive neurodegenerative disease of unknown etiology. Due to ALS’s unpredictable onset and progression rate, the search for biomarkers that allow the detection and tracking of its development and therapeutic efficacy would be of significant medical value. Considering that alterations of energy supply are one of ALS’s main hallmarks and that a correlation has been established between gene expression in human brain tissue and peripheral blood mononuclear cells (PBMCs), the present work investigates whether changes in mitochondrial function could be used to monitor ALS. To achieve this goal, PBMCs from ALS patients and control subjects were used; blood sampling is a quite non-invasive method and is cost-effective. Different parameters were evaluated, namely cytosolic calcium levels, mitochondrial membrane potential, oxidative stress, and metabolic compounds levels, as well as mitochondrial dynamics and degradation. Altogether, we observed lower mitochondrial calcium uptake/retention, mitochondria depolarization, and redox homeostasis deregulation, in addition to a decrease in critical metabolic genes, a diminishment in mitochondrial biogenesis, and an augmentation in mitochondrial fission and autophagy-related gene expression. All of these changes can contribute to the decreased ATP and pyruvate levels observed in ALS PBMCs. Our data indicate that PBMCs from ALS patients show a significant mitochondrial dysfunction, resembling several findings from ALS’ neural cells/models, which could be exploited as a powerful tool in ALS research. Our findings can also guide future studies on new pharmacological interventions for ALS since assessments of brain samples are challenging and represent a relevant limited strategy. Graphical abstract ![]()
Collapse
|
16
|
Tibenska V, Benesova A, Vebr P, Liptakova A, Hejnová L, Elsnicová B, Drahota Z, Hornikova D, Galatík F, Kolar D, Vybiral S, Alánová P, Novotný J, Kolar F, Novakova O, Zurmanova JM. Gradual cold acclimation induces cardioprotection without affecting β-adrenergic receptor-mediated adenylyl cyclase signaling. J Appl Physiol (1985) 2020; 128:1023-1032. [DOI: 10.1152/japplphysiol.00511.2019] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Novel strategies are needed that can stimulate endogenous signaling pathways to protect the heart from myocardial infarction. The present study tested the hypothesis that appropriate regimen of cold acclimation (CA) may provide a promising approach for improving myocardial resistance to ischemia/reperfusion (I/R) injury without negative side effects. We evaluated myocardial I/R injury, mitochondrial swelling, and β-adrenergic receptor (β-AR)-adenylyl cyclase-mediated signaling. Male Wistar rats were exposed to CA (8°C, 8 h/day for a week, followed by 4 wk at 8°C for 24 h/day), while the recovery group (CAR) was kept at 24°C for an additional 2 wk. The myocardial infarction induced by coronary occlusion for 20 min followed by 3-h reperfusion was reduced from 56% in controls to 30% and 23% after CA and CAR, respectively. In line, the rate of mitochondrial swelling at 200 μM Ca2+ was decreased in both groups. Acute administration of metoprolol decreased infarction in control group and did not affect the CA-elicited cardiprotection. Accordingly, neither β1-AR-Gsα-adenylyl cyclase signaling, stimulated with specific ligands, nor p-PKA/PKA ratios were affected after CA or CAR. Importantly, Western blot and immunofluorescence analyses revealed β2- and β3-AR protein enrichment in membranes in both experimental groups. We conclude that gradual cold acclimation results in a persisting increase of myocardial resistance to I/R injury without hypertension and hypertrophy. The cardioprotective phenotype is associated with unaltered adenylyl cyclase signaling and increased mitochondrial resistance to Ca2+-overload. The potential role of upregulated β2/β3-AR pathways remains to be elucidated. NEW & NOTEWORTHY We present a new model of mild gradual cold acclimation increasing tolerance to myocardial ischemia/reperfusion injury without hypertension and hypertrophy. Cardioprotective phenotype is accompanied by unaltered adenylyl cyclase signaling and increased mitochondrial resistance to Ca2+-overload. The potential role of upregulated β2/β3-adrenoreceptor activation is considered. These findings may stimulate the development of novel preventive and therapeutic strategies against myocardial ischemia/reperfusion injury.
Collapse
Affiliation(s)
- V. Tibenska
- Department of Physiology, Faculty of Science, Charles University, Prague, Czech Republic
| | - A. Benesova
- Department of Physiology, Faculty of Science, Charles University, Prague, Czech Republic
| | - P. Vebr
- Department of Physiology, Faculty of Science, Charles University, Prague, Czech Republic
| | - A. Liptakova
- Department of Physiology, Faculty of Science, Charles University, Prague, Czech Republic
| | - L. Hejnová
- Department of Physiology, Faculty of Science, Charles University, Prague, Czech Republic
| | - B. Elsnicová
- Department of Physiology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Z. Drahota
- Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - D. Hornikova
- Department of Physiology, Faculty of Science, Charles University, Prague, Czech Republic
| | - F. Galatík
- Department of Physiology, Faculty of Science, Charles University, Prague, Czech Republic
| | - D. Kolar
- Department of Physiology, Faculty of Science, Charles University, Prague, Czech Republic
| | - S. Vybiral
- Department of Physiology, Faculty of Science, Charles University, Prague, Czech Republic
| | - P. Alánová
- Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - J. Novotný
- Department of Physiology, Faculty of Science, Charles University, Prague, Czech Republic
| | - F. Kolar
- Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - O. Novakova
- Department of Physiology, Faculty of Science, Charles University, Prague, Czech Republic
- Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - J. M. Zurmanova
- Department of Physiology, Faculty of Science, Charles University, Prague, Czech Republic
| |
Collapse
|
17
|
Wu L, Tan JL, Chen ZY, Huang G. Cardioprotection of post-ischemic moderate ROS against ischemia/reperfusion via STAT3-induced the inhibition of MCU opening. Basic Res Cardiol 2019; 114:39. [DOI: 10.1007/s00395-019-0747-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Accepted: 08/19/2019] [Indexed: 12/20/2022]
|
18
|
Wang P, Wang SC, Yang H, Lv C, Jia S, Liu X, Wang X, Meng D, Qin D, Zhu H, Wang YF. Therapeutic Potential of Oxytocin in Atherosclerotic Cardiovascular Disease: Mechanisms and Signaling Pathways. Front Neurosci 2019; 13:454. [PMID: 31178679 PMCID: PMC6537480 DOI: 10.3389/fnins.2019.00454] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 04/23/2019] [Indexed: 12/12/2022] Open
Abstract
Coronary artery disease (CAD) is a major cardiovascular disease responsible for high morbidity and mortality worldwide. The major pathophysiological basis of CAD is atherosclerosis in association with varieties of immunometabolic disorders that can suppress oxytocin (OT) receptor (OTR) signaling in the cardiovascular system (CVS). By contrast, OT not only maintains cardiovascular integrity but also has the potential to suppress and even reverse atherosclerotic alterations and CAD. These protective effects of OT are associated with its protection of the heart and blood vessels from immunometabolic injuries and the resultant inflammation and apoptosis through both peripheral and central approaches. As a result, OT can decelerate the progression of atherosclerosis and facilitate the recovery of CVS from these injuries. At the cellular level, the protective effect of OT on CVS involves a broad array of OTR signaling events. These signals mainly belong to the reperfusion injury salvage kinase pathway that is composed of phosphatidylinositol 3-kinase-Akt-endothelial nitric oxide synthase cascades and extracellular signal-regulated protein kinase 1/2. Additionally, AMP-activated protein kinase, Ca2+/calmodulin-dependent protein kinase signaling and many others are also implicated in OTR signaling in the CVS protection. These signaling events interact coordinately at many levels to suppress the production of inflammatory cytokines and the activation of apoptotic pathways. A particular target of these signaling events is endoplasmic reticulum (ER) stress and mitochondrial oxidative stress that interact through mitochondria-associated ER membrane. In contrast to these protective effects and machineries, rare but serious cardiovascular disturbances were also reported in labor induction and animal studies including hypotension, reflexive tachycardia, coronary spasm or thrombosis and allergy. Here, we review our current understanding of the protective effect of OT against varieties of atherosclerotic etiologies as well as the approaches and underlying mechanisms of these effects. Moreover, potential cardiovascular disturbances following OT application are also discussed to avoid unwanted effects in clinical trials of OT usages.
Collapse
Affiliation(s)
- Ping Wang
- Department of Genetics, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Stephani C Wang
- Department of Medicine, Albany Medical Center, Albany, NY, United States
| | - Haipeng Yang
- Department of Pediatrics, The Forth Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Chunmei Lv
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Shuwei Jia
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Xiaoyu Liu
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Xiaoran Wang
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Dexin Meng
- Department of Physiology, Jiamusi University, Jiamusi, China
| | - Danian Qin
- Department of Physiology, Shantou University of Medical College, Shantou, China
| | - Hui Zhu
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Yu-Feng Wang
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| |
Collapse
|
19
|
Is there an effect of ischemic conditioning on myocardial contractile function following acute myocardial ischemia/reperfusion injury? Biochim Biophys Acta Mol Basis Dis 2019; 1865:822-830. [PMID: 30660684 DOI: 10.1016/j.bbadis.2018.12.020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 12/18/2018] [Accepted: 12/19/2018] [Indexed: 10/27/2022]
Abstract
Ischemic conditioning induces cardioprotection; the final infarct size following a myocardial ischemic event is reduced. However, whether ischemic conditioning has long-term beneficial effects on myocardial contractile function following such an ischemic event needs further elucidation. To date, ex vivo studies have shown that ischemic conditioning improves the contractile recovery of isolated ventricular papillary muscle or atrial trabeculae following simulated ischemia. However, in vivo animal studies and studies in patients undergoing elective cardiac surgery show conflicting results. At the subcellular level, it is known that ischemic conditioning improved energy metabolism, preserved mitochondrial respiration, ATP production, and Ca2+ homeostasis in isolated mitochondria from the myocardium. Ischemic conditioning also presents with post-translational modifications of proteins in the contractile machinery of the myocardium. The beneficial effects on myocardial contractile function need further elucidation. This article is part of a Special Issue entitled: The power of metabolism: Linking energy supply and demand to contractile function edited by Torsten Doenst, Michael Schwarzer and Christine Des Rosiers.
Collapse
|
20
|
Rinaldi L, Pozdniakova S, Jayarajan V, Troidl C, Abdallah Y, Aslam M, Ladilov Y. Protective role of soluble adenylyl cyclase against reperfusion-induced injury of cardiac cells. Biochim Biophys Acta Mol Basis Dis 2018; 1865:252-260. [PMID: 30044950 DOI: 10.1016/j.bbadis.2018.07.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Revised: 06/15/2018] [Accepted: 07/17/2018] [Indexed: 12/16/2022]
Abstract
AIMS Disturbance of mitochondrial function significantly contributes to the myocardial injury that occurs during reperfusion. Increasing evidence suggests a role of intra-mitochondrial cyclic AMP (cAMP) signaling in promoting respiration and ATP synthesis. Mitochondrial levels of cAMP are controlled by type 10 soluble adenylyl cyclase (sAC) and phosphodiesterase 2 (PDE2), however their role in the reperfusion-induced injury remains unknown. Here we aimed to examine whether sAC may support cardiomyocyte survival during reperfusion. METHODS AND RESULTS Adult rat cardiomyocytes or rat cardiac H9C2 cells were subjected to metabolic inhibition and recovery as a model of simulated ischemia and reperfusion. Cytosolic Ca2+, pH, mitochondrial cAMP (live-cell imaging), and cell viability were analyzed during a 15-min period of reperfusion. Suppression of sAC activity in cardiomyocytes and H9C2 cells, either by sAC knockdown, by pharmacological inhibition or by withdrawal of bicarbonate, a natural sAC activator, compromised cell viability and recovery of cytosolic Ca2+ homeostasis during reperfusion. Contrariwise, overexpression of mitochondria-targeted sAC in H9C2 cells suppressed reperfusion-induced cell death. Analyzing cAMP concentration in mitochondrial matrix we found that inhibition of PDE2, a predominant mitochondria-localized PDE isoform in mammals, during reperfusion significantly increased cAMP level in mitochondrial matrix, but not in cytosol. Accordingly, PDE2 inhibition attenuated reperfusion-induced cardiomyocyte death and improved recovery of the cytosolic Ca2+ homeostasis. CONCLUSION sAC plays an essential role in supporting cardiomyocytes viability during reperfusion. Elevation of mitochondrial cAMP pool either by sAC overexpression or by PDE2 inhibition beneficially affects cardiomyocyte survival during reperfusion.
Collapse
Affiliation(s)
- Laura Rinaldi
- Department of Cardiology and Angiology, Justus Liebig University, Giessen, Germany
| | | | | | - Christian Troidl
- Department of Cardiology and Angiology, Justus Liebig University, Giessen, Germany
| | - Yaser Abdallah
- Department of Cardiology and Angiology, Justus Liebig University, Giessen, Germany
| | - Muhammad Aslam
- Department of Cardiology and Angiology, Justus Liebig University, Giessen, Germany; DZHK (German Centre for Cardiovascular Research), partner site Rhein-Main, Bad Nauheim, Germany
| | - Yury Ladilov
- Center for Cardiovascular Research, Charité, Berlin, Germany; DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany.
| |
Collapse
|
21
|
Holland NA, Fraiser CR, Sloan RC, Devlin RB, Brown DA, Wingard CJ. Ultrafine Particulate Matter Increases Cardiac Ischemia/Reperfusion Injury via Mitochondrial Permeability Transition Pore. Cardiovasc Toxicol 2018; 17:441-450. [PMID: 28194639 DOI: 10.1007/s12012-017-9402-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Ultrafine particulate matter (UFP) has been associated with increased cardiovascular morbidity and mortality. However, the mechanisms that drive PM-associated cardiovascular disease and dysfunction remain unclear. We examined the impact of oropharyngeal aspiration of 100 μg UFP from the Chapel Hill, NC, air shed in Sprague-Dawley rats on cardiac function, arrhythmogenesis, and cardiac ischemia/reperfusion (I/R) injury using a Langendorff working heart model. We found that exposure to UFP was capable of significantly exacerbating cardiac I/R injury without changing overall cardiac function or major changes in arrhythmogenesis. Cardiac I/R injury was attenuable with administration of cyclosporin A (CsA), suggesting a role for the mitochondrial permeability transition pore (mPTP) in UFP-associated cardiovascular toxicity. Isolated cardiac mitochondria displayed decreased Ca2+ buffering before opening of the mPTP. These findings suggest that UFP-induced expansion of cardiac I/R injury may be a result of mPTP Ca2+ sensitization resulting in increased mitochondrial permeability transition and potential initiation of mPTP-associated cell death pathways.
Collapse
Affiliation(s)
- Nathan A Holland
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, USA
| | - Chad R Fraiser
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, USA
| | - Ruben C Sloan
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, USA
| | - Robert B Devlin
- National Health and Environmental Effects Research Laboratory, Environmental Public Health Division, US Environmental Protection Agency, Chapel Hill, NC, USA
| | - David A Brown
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, USA
| | - Christopher J Wingard
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, USA. .,Lansing School of Nursing and Health Sciences, Physical Therapy Department, Bellarmine University, 2001 Newburg Rd, Louisville, KY, 40205, USA.
| |
Collapse
|
22
|
Holland NA, Francisco JT, Johnson SC, Morgan JS, Dennis TJ, Gadireddy NR, Tulis DA. Cyclic Nucleotide-Directed Protein Kinases in Cardiovascular Inflammation and Growth. J Cardiovasc Dev Dis 2018; 5:E6. [PMID: 29367584 PMCID: PMC5872354 DOI: 10.3390/jcdd5010006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 01/17/2018] [Accepted: 01/19/2018] [Indexed: 02/08/2023] Open
Abstract
Cardiovascular disease (CVD), including myocardial infarction (MI) and peripheral or coronary artery disease (PAD, CAD), remains the number one killer of individuals in the United States and worldwide, accounting for nearly 18 million (>30%) global deaths annually. Despite considerable basic science and clinical investigation aimed at identifying key etiologic components of and potential therapeutic targets for CVD, the number of individuals afflicted with these dreaded diseases continues to rise. Of the many biochemical, molecular, and cellular elements and processes characterized to date that have potential to control foundational facets of CVD, the multifaceted cyclic nucleotide pathways continue to be of primary basic science and clinical interest. Cyclic adenosine monophosphate (cyclic AMP) and cyclic guanosine monophosphate (cyclic GMP) and their plethora of downstream protein kinase effectors serve ubiquitous roles not only in cardiovascular homeostasis but also in the pathogenesis of CVD. Already a major target for clinical pharmacotherapy for CVD as well as other pathologies, novel and potentially clinically appealing actions of cyclic nucleotides and their downstream targets are still being discovered. With this in mind, this review article focuses on our current state of knowledge of the cyclic nucleotide-driven serine (Ser)/threonine (Thr) protein kinases in CVD with particular emphasis on cyclic AMP-dependent protein kinase (PKA) and cyclic GMP-dependent protein kinase (PKG). Attention is given to the regulatory interactions of these kinases with inflammatory components including interleukin 6 signals, with G protein-coupled receptor and growth factor signals, and with growth and synthetic transcriptional platforms underlying CVD pathogenesis. This article concludes with a brief discussion of potential future directions and highlights the importance for continued basic science and clinical study of cyclic nucleotide-directed protein kinases as emerging and crucial controllers of cardiac and vascular disease pathologies.
Collapse
Affiliation(s)
- Nathan A Holland
- Department of Physiology, Brody School of Medicine, East Carolina University, 600 Moye Boulevard, Greenville, NC 27834, USA.
| | - Jake T Francisco
- Department of Physiology, Brody School of Medicine, East Carolina University, 600 Moye Boulevard, Greenville, NC 27834, USA.
| | - Sean C Johnson
- Department of Physiology, Brody School of Medicine, East Carolina University, 600 Moye Boulevard, Greenville, NC 27834, USA.
| | - Joshua S Morgan
- Department of Physiology, Brody School of Medicine, East Carolina University, 600 Moye Boulevard, Greenville, NC 27834, USA.
| | - Troy J Dennis
- Department of Physiology, Brody School of Medicine, East Carolina University, 600 Moye Boulevard, Greenville, NC 27834, USA.
| | - Nishitha R Gadireddy
- Department of Physiology, Brody School of Medicine, East Carolina University, 600 Moye Boulevard, Greenville, NC 27834, USA.
| | - David A Tulis
- Department of Physiology, Brody School of Medicine, East Carolina University, 600 Moye Boulevard, Greenville, NC 27834, USA.
| |
Collapse
|
23
|
Francois AA, Obasanjo-Blackshire K, Clark JE, Boguslavskyi A, Holt MR, Parker PJ, Marber MS, Heads RJ. Loss of Protein Kinase Novel 1 (PKN1) is associated with mild systolic and diastolic contractile dysfunction, increased phospholamban Thr17 phosphorylation, and exacerbated ischaemia-reperfusion injury. Cardiovasc Res 2018; 114:138-157. [PMID: 29045568 PMCID: PMC5815577 DOI: 10.1093/cvr/cvx206] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 03/17/2017] [Accepted: 10/13/2017] [Indexed: 01/20/2023] Open
Abstract
Aims PKN1 is a stress-responsive protein kinase acting downstream of small GTP-binding proteins of the Rho/Rac family. The aim was to determine its role in endogenous cardioprotection. Methods and results Hearts from PKN1 knockout (KO) or wild type (WT) littermate control mice were perfused in Langendorff mode and subjected to global ischaemia and reperfusion (I/R). Myocardial infarct size was doubled in PKN1 KO hearts compared to WT hearts. PKN1 was basally phosphorylated on the activation loop Thr778 PDK1 target site which was unchanged during I/R. However, phosphorylation of p42/p44-MAPK was decreased in KO hearts at baseline and during I/R. In cultured neonatal rat ventricular cardiomyocytes (NRVM) and NRVM transduced with kinase dead (KD) PKN1 K644R mutant subjected to simulated ischaemia/reperfusion (sI/R), PhosTag® gel analysis showed net dephosphorylation of PKN1 during sI and early R despite Thr778 phosphorylation. siRNA knockdown of PKN1 in NRVM significantly decreased cell survival and increased cell injury by sI/R which was reversed by WT- or KD-PKN1 expression. Confocal immunofluorescence analysis of PKN1 in NRVM showed increased localization to the sarcoplasmic reticulum (SR) during sI. GC-MS/MS and immunoblot analysis of PKN1 immunoprecipitates following sI/R confirmed interaction with CamKIIδ. Co-translocation of PKN1 and CamKIIδ to the SR/membrane fraction during sI correlated with phospholamban (PLB) Thr17 phosphorylation. siRNA knockdown of PKN1 in NRVM resulted in increased basal CamKIIδ activation and increased PLB Thr17 phosphorylation only during sI. In vivo PLB Thr17 phosphorylation, Sarco-Endoplasmic Reticulum Ca2+ ATPase (SERCA2) expression and Junctophilin-2 (Jph2) expression were also basally increased in PKN1 KO hearts. Furthermore, in vivo P-V loop analysis of the beat-to-beat relationship between rate of LV pressure development or relaxation and end diastolic P (EDP) showed mild but significant systolic and diastolic dysfunction with preserved ejection fraction in PKN1 KO hearts. Conclusion Loss of PKN1 in vivo significantly reduces endogenous cardioprotection and increases myocardial infarct size following I/R injury. Cardioprotection by PKN1 is associated with reduced CamKIIδ-dependent PLB Thr17 phosphorylation at the SR and therefore may stabilize the coupling of SR Ca2+ handling and contractile function, independent of its kinase activity.
Collapse
Affiliation(s)
- Asvi A Francois
- Department of Cardiology, School of Cardiovascular Medicine and Sciences, British Heart Foundation Centre for Research Excellence, Faculty of Life Sciences and Medicine, The Rayne Institute, King’s College London, St Thomas’s Hospital, Lambeth Palace Road, London, UK
| | - Kofo Obasanjo-Blackshire
- Department of Cardiology, School of Cardiovascular Medicine and Sciences, British Heart Foundation Centre for Research Excellence, Faculty of Life Sciences and Medicine, The Rayne Institute, King’s College London, St Thomas’s Hospital, Lambeth Palace Road, London, UK
| | - James E Clark
- Department of Cardiology, School of Cardiovascular Medicine and Sciences, British Heart Foundation Centre for Research Excellence, Faculty of Life Sciences and Medicine, The Rayne Institute, King’s College London, St Thomas’s Hospital, Lambeth Palace Road, London, UK
| | - Andrii Boguslavskyi
- Department of Cardiology, School of Cardiovascular Medicine and Sciences, British Heart Foundation Centre for Research Excellence, Faculty of Life Sciences and Medicine, The Rayne Institute, King’s College London, St Thomas’s Hospital, Lambeth Palace Road, London, UK
| | - Mark R Holt
- Department of Cardiology, School of Cardiovascular Medicine and Sciences, British Heart Foundation Centre for Research Excellence, Faculty of Life Sciences and Medicine, The Rayne Institute, King’s College London, St Thomas’s Hospital, Lambeth Palace Road, London, UK
- Randall Division of Cell and Molecular Biophysics, King’s College London, New Hunt’s House, Guy’s Hospital Campus, London, UK
| | - Peter J Parker
- Division of Cancer Studies, School of Cancer and Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King’s College London, New Hunt’s House, Guy’s Hospital Campus, London, UK
- Protein Phosphorylation Laboratory, Francis Crick Institute, Lincoln’s Inn Fields, London, UK
| | - Michael S Marber
- Department of Cardiology, School of Cardiovascular Medicine and Sciences, British Heart Foundation Centre for Research Excellence, Faculty of Life Sciences and Medicine, The Rayne Institute, King’s College London, St Thomas’s Hospital, Lambeth Palace Road, London, UK
| | - Richard J Heads
- Department of Cardiology, School of Cardiovascular Medicine and Sciences, British Heart Foundation Centre for Research Excellence, Faculty of Life Sciences and Medicine, The Rayne Institute, King’s College London, St Thomas’s Hospital, Lambeth Palace Road, London, UK
| |
Collapse
|
24
|
Lumkwana D, Botha A, Samodien E, Hanser S, Lopes J. Laminin, laminin-entactin and extracellular matrix are equally appropriate adhesive substrates for isolated adult rat cardiomyocyte culture and experimentation. Cell Adh Migr 2017; 12:503-511. [PMID: 29091577 DOI: 10.1080/19336918.2017.1387693] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
Although techniques for isolating and culturing adult cardiomyocytes were developed four decades ago, it still remains a challenge to isolate high yields of healthy viable cardiomyocytes, to maintain them in culture, and to use them successfully in experiments. This is due to the difficulty in deciding which adhesive substrate and buffer composition to use. Therefore this study aimed to (1) identify a robust experimental buffer to sustain survival of cultured adult rat cardiomyocytes (ARCMs) during control normoxic conditions, and (2) to identify an adhesive substrate that provides optimal cell adherence, not only during normoxia, but especially during simulated ischemia-reperfusion (SIR) experiments. Adhesion and viability of ARCMs were evaluated on laminin, laminin-entactin (LE), and extracellular matrix (ECM) at concentrations between 25-200 ug/ml, in three different normoxic experimental buffers and under SIR conditions. Differences in normoxic buffer composition had no effect on the adherence of ARCMs, but had a significant effect on mitochondrial function and thus cell viability. HEPES buffered PBS supplemented with 10 mM glucose was not sufficient to sustain cell viability unless 2 mM pyruvate was added, yet a cocktail of PBS and M199 provided an even greater viability. Finally, laminin, LE, and ECM retained similar numbers of ARCMs per concentration, but only provided efficient adhesion at concentrations ≥ 100 ug/ml.
Collapse
Affiliation(s)
- D Lumkwana
- a Department of Biomedical Sciences , Division of Medical Physiology, Faculty of Health Sciences and Medicine, Stellenbosch University , Tygerberg , Cape Town , South Africa
| | - A Botha
- a Department of Biomedical Sciences , Division of Medical Physiology, Faculty of Health Sciences and Medicine, Stellenbosch University , Tygerberg , Cape Town , South Africa
| | - E Samodien
- a Department of Biomedical Sciences , Division of Medical Physiology, Faculty of Health Sciences and Medicine, Stellenbosch University , Tygerberg , Cape Town , South Africa
| | - S Hanser
- a Department of Biomedical Sciences , Division of Medical Physiology, Faculty of Health Sciences and Medicine, Stellenbosch University , Tygerberg , Cape Town , South Africa
| | - J Lopes
- a Department of Biomedical Sciences , Division of Medical Physiology, Faculty of Health Sciences and Medicine, Stellenbosch University , Tygerberg , Cape Town , South Africa
| |
Collapse
|
25
|
Mallet RT, Olivencia-Yurvati AH, Bünger R. Pyruvate enhancement of cardiac performance: Cellular mechanisms and clinical application. Exp Biol Med (Maywood) 2017; 243:198-210. [PMID: 29154687 DOI: 10.1177/1535370217743919] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Cardiac contractile function is adenosine-5'-triphosphate (ATP)-intensive, and the myocardium's high demand for oxygen and energy substrates leaves it acutely vulnerable to interruptions in its blood supply. The myriad cardioprotective properties of the natural intermediary metabolite pyruvate make it a potentially powerful intervention against the complex injury cascade ignited by myocardial ischemia-reperfusion. A readily oxidized metabolic substrate, pyruvate augments myocardial free energy of ATP hydrolysis to a greater extent than the physiological fuels glucose, lactate and fatty acids, particularly when it is provided at supra-physiological plasma concentrations. Pyruvate also exerts antioxidant effects by detoxifying reactive oxygen and nitrogen intermediates, and by increasing nicotinamide adenine dinucleotide phosphate reduced form (NADPH) production to maintain glutathione redox state. These enhancements of free energy and antioxidant defenses combine to augment sarcoplasmic reticular Ca2+ release and re-uptake central to cardiac mechanical performance and to restore β-adrenergic signaling of ischemically stunned myocardium. By minimizing Ca2+ mismanagement and oxidative stress, pyruvate suppresses inflammation in post-ischemic myocardium. Thus, pyruvate administration stabilized cardiac performance, augmented free energy of ATP hydrolysis and glutathione redox systems, and/or quelled inflammation in a porcine model of cardiopulmonary bypass, a canine model of cardiac arrest-resuscitation, and a caprine model of hypovolemia and hindlimb ischemia-reperfusion. Pyruvate's myriad benefits in preclinical models provide the mechanistic framework for its clinical application as metabolic support for myocardium at risk. Phase one trials have demonstrated pyruvate's safety and efficacy for intravenous resuscitation for septic shock, intracoronary infusion for heart failure and as a component of cardioplegia for cardiopulmonary bypass. The favorable outcomes of these trials, which argue for expanded, phase three investigations of pyruvate therapy, mirror findings in isolated, perfused hearts, underscoring the pivotal role of preclinical research in identifying clinical interventions for cardiovascular diseases. Impact statement This article reviews pyruvate's cardioprotective properties as an energy-yielding metabolic fuel, antioxidant and anti-inflammatory agent in mammalian myocardium. Preclinical research has shown these properties make pyruvate a powerful intervention to curb the complex injury cascade ignited by ischemia and reperfusion. In ischemically stunned isolated hearts and in large mammal models of cardiopulmonary bypass, cardiac arrest-resuscitation and hypovolemia, intracoronary pyruvate supports recovery of myocardial contractile function, intracellular Ca2+ homeostasis and free energy of ATP hydrolysis, and its antioxidant actions restore β-adrenergic signaling and suppress inflammation. The first clinical trials of pyruvate for cardiopulmonary bypass, fluid resuscitation and intracoronary intervention for congestive heart failure have been reported. Receiver operating characteristic analyses show remarkable concordance between pyruvate's beneficial functional and metabolic effects in isolated, perfused hearts and in patients recovering from cardiopulmonary bypass in which they received pyruvate- vs. L-lactate-fortified cardioplegia. This research exemplifies the translation of mechanism-oriented preclinical studies to clinical application and outcomes.
Collapse
Affiliation(s)
- Robert T Mallet
- 1 Department of Integrative Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX 76107-2699, USA
| | - Albert H Olivencia-Yurvati
- 1 Department of Integrative Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX 76107-2699, USA.,2 Department of Medical Education, University of North Texas Health Science Center, Fort Worth, TX 76107-2699, USA
| | - Rolf Bünger
- 3 Emeritus Member of the American Physiological Society, McLean, VA 22101, USA
| |
Collapse
|
26
|
Gündüz D, Hamm CW, Aslam M. Simultaneous Isolation of High Quality Cardiomyocytes, Endothelial Cells, and Fibroblasts from an Adult Rat Heart. J Vis Exp 2017. [PMID: 28570510 DOI: 10.3791/55601] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
The rat is an important animal model used in cardiovascular research, and rat cardiac cells are used routinely for in vitro analysis of the molecular mechanisms of cardiovascular disease progression such as cardiac hypertrophy, fibrosis, and atherosclerosis. Although several attempts with variable success have been made to develop immortalized cell lines from the cardiovascular system to understand these cellular mechanisms, primary cells offer a more natural and close to in vivo environment for such studies. Therefore, different laboratories working on a particular cell type have developed protocols to isolate individual types of rat cardiac cells of interest. A protocol that allows the isolation of more than one cell type, however, is missing. Here an optimized protocol is described that allows the isolation of high-quality major cardiac cell types (cardiomyocytes, endothelial cells, and fibroblasts) from a single preparation and enables their use for cellular analyses. This permits the most efficient use of available resources, which may save time and reduce research costs.
Collapse
Affiliation(s)
- Dursun Gündüz
- Internal Medicine, Cardiology and Angiology, University Hospital Giessen
| | - Christian W Hamm
- Internal Medicine, Cardiology and Angiology, University Hospital Giessen
| | - Muhammad Aslam
- Internal Medicine, Cardiology and Angiology, University Hospital Giessen;
| |
Collapse
|
27
|
Mitochondrial Cx43 hemichannels contribute to mitochondrial calcium entry and cell death in the heart. Basic Res Cardiol 2017; 112:27. [PMID: 28364353 DOI: 10.1007/s00395-017-0618-1] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 03/30/2017] [Indexed: 10/19/2022]
Abstract
Mitochondrial connexin 43 (Cx43) plays a key role in cardiac cytoprotection caused by repeated exposure to short periods of non-lethal ischemia/reperfusion, a condition known as ischemic preconditioning. Cx43 also forms calcium (Ca2+)-permeable hemichannels that may potentially lead to mitochondrial Ca2+ overload and cell death. Here, we studied the role of Cx43 in facilitating mitochondrial Ca2+ entry and investigated its downstream consequences. To that purpose, we used various connexin-targeting peptides interacting with extracellular (Gap26) and intracellular (Gap19, RRNYRRNY) Cx43 domains, and tested their effect on mitochondrial dye- and Ca2+-uptake, electrophysiological properties of plasmalemmal and mitochondrial Cx43 channels, and cell injury/cell death. Our results in isolated mice cardiac subsarcolemmal mitochondria indicate that Cx43 forms hemichannels that contribute to Ca2+ entry and may trigger permeability transition and cell injury/death. RRNYRRNY displayed the strongest effects in all assays and inhibited plasma membrane as well as mitochondrial Cx43 hemichannels. RRNYRRNY also strongly reduced the infarct size in ex vivo cardiac ischemia-reperfusion studies. These results indicate that Cx43 contributes to mitochondrial Ca2+ homeostasis and is involved in triggering cell injury/death pathways that can be inhibited by RRNYRRNY peptide.
Collapse
|
28
|
Zhou JQ, Qiu T, Zhang L, Chen ZB, Wang ZS, Ma XX, Li D. Allopurinol preconditioning attenuates renal ischemia/reperfusion injury by inhibiting HMGB1 expression in a rat model. Acta Cir Bras 2016; 31:176-82. [PMID: 27050788 DOI: 10.1590/s0102-865020160030000005] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 02/15/2016] [Indexed: 01/15/2023] Open
Abstract
PURPOSE To investigate the potential effects of pretreatment with allopurinol on renal ischemia/reperfusion injury (IRI) in a rat model. METHODS Twenty four rats were subjected to right kidney uninephrectomy were randomly distributed into the following three groups (n=8): Group A (sham-operated group); Group B (ischemic group) with 30 min of renal ischemia after surgery; and Group C (allopurinol + ischemia group) pretreated with allopurinol at 50 mg/kg for 14 days. At 72 h after renal reperfusion, the kidney was harvested to assess inflammation and apoptosis. RESULTS Pretreatment with allopurinol significantly improved renal functional and histological grade scores following I/R injury (p<0.05). Compared with Group B, the expression levels of caspase-3 and Bax were markedly reduced in Group C, meanwhile, whereas expression of bcl-2 was clearly increased (p<0.05). A newly described marker of inflammation, High Mobility Group Box 1(HMGB1), showed reduced expression in Group C (p<0.05). CONCLUSION Pretreatment with allopurinol had a protective effect on kidney ischemia/reperfusion injury, which might be related to the inhibition of HMGB1 expression.
Collapse
Affiliation(s)
- Jiang-qiao Zhou
- Department of Organ Transplantation, Renmin Hospital, Wuhan University, Wuhan, Hubei, China
| | - Tao Qiu
- Department of Organ Transplantation, Renmin Hospital, Wuhan University, Wuhan, Hubei, China
| | - Lu Zhang
- Department of Urology, Renmin Hospital, Wuhan University, Wuhan, Hubei, China
| | - Zhong-bao Chen
- Department of Organ Transplantation, Renmin Hospital, Wuhan University, Wuhan, Hubei, China
| | - Zhi-shun Wang
- Department of Urology, Renmin Hospital, Wuhan University, Wuhan, Hubei, China
| | - Xiao-xiong Ma
- Department of Organ Transplantation, Renmin Hospital, Wuhan University, Wuhan, Hubei, China
| | - Dongyu Li
- Intensive Care Unit, PuAi Hospital, Anlu, Hubei, China
| |
Collapse
|
29
|
[Cardioprotection via the arm? : How a blood pressure cuff decreases infarct sizes]. Herz 2016; 42:565-572. [PMID: 27785525 DOI: 10.1007/s00059-016-4490-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 09/10/2016] [Accepted: 09/30/2016] [Indexed: 01/05/2023]
Abstract
Cardiovascular diseases and especially myocardial infarctions are responsible for a high morbidity and mortality throughout Europe. An essential aspect of myocardial infarction is ischemia/reperfusion injury which represents the necrosis of myocytes following reperfusion. One possible option to counteract ischemia/reperfusion injury is the much researched process of remote ischemic conditioning (RIC), whereby a certain tissue (e.g. skeletal muscle) is subjected to several cycles of short periods (e.g. 5 min) of ischemia and reperfusion and leads to the protection of another organ (e.g. the heart). Despite substantial efforts to elucidate the underlying mechanisms during the last decades, this phenomenon is not yet completely understood. Clinical studies mainly concentrated on laboratory and radiological parameters, which led to better understanding of RIC; however, large clinical studies evaluating the possible influence on mortality are still lacking. This review article provides an introduction to RIC and summarizes the current understanding of known pathomechanisms and the results of important clinical studies.
Collapse
|
30
|
Shaikh S, Troncoso R, Criollo A, Bravo-Sagua R, García L, Morselli E, Cifuentes M, Quest AFG, Hill JA, Lavandero S. Regulation of cardiomyocyte autophagy by calcium. Am J Physiol Endocrinol Metab 2016; 310:E587-E596. [PMID: 26884385 PMCID: PMC4835942 DOI: 10.1152/ajpendo.00374.2015] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 02/10/2016] [Indexed: 11/22/2022]
Abstract
Calcium signaling plays a crucial role in a multitude of events within the cardiomyocyte, including cell cycle control, growth, apoptosis, and autophagy. With respect to calcium-dependent regulation of autophagy, ion channels and exchangers, receptors, and intracellular mediators play fundamental roles. In this review, we discuss calcium-dependent regulation of cardiomyocyte autophagy, a lysosomal mechanism that is often cytoprotective, serving to defend against disease-related stress and nutrient insufficiency. We also highlight the importance of the subcellular distribution of calcium and related proteins, interorganelle communication, and other key signaling events that govern cardiomyocyte autophagy.
Collapse
Affiliation(s)
- Soni Shaikh
- Advanced Center for Chronic Disease and Center for Molecular Studies of the Cell, Facultad de Ciencias Quimicas y Farmaceuticas and Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Rodrigo Troncoso
- Advanced Center for Chronic Disease and Center for Molecular Studies of the Cell, Facultad de Ciencias Quimicas y Farmaceuticas and Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Instituto de Nutrición y Tecnología de los Alimentos, Universidad de Chile, Santiago, Chile
| | - Alfredo Criollo
- Advanced Center for Chronic Disease and Center for Molecular Studies of the Cell, Facultad de Ciencias Quimicas y Farmaceuticas and Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Instituto de Investigación en Ciencias Odontológicas, Facultad de Odontología, Universidad de Chile, Santiago, Chile
| | - Roberto Bravo-Sagua
- Advanced Center for Chronic Disease and Center for Molecular Studies of the Cell, Facultad de Ciencias Quimicas y Farmaceuticas and Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Lorena García
- Advanced Center for Chronic Disease and Center for Molecular Studies of the Cell, Facultad de Ciencias Quimicas y Farmaceuticas and Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Departamento de Bioquimica y Biologia Molecular, Facultad de Ciencias Quimicas y Farmaceuticas, Universidad de Chile, Santiago, Chile
| | - Eugenia Morselli
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Mariana Cifuentes
- Instituto de Nutrición y Tecnología de los Alimentos, Universidad de Chile, Santiago, Chile
| | - Andrew F G Quest
- Advanced Center for Chronic Disease and Center for Molecular Studies of the Cell, Facultad de Ciencias Quimicas y Farmaceuticas and Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Programa de Biología Celular y Molecular, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile; and
| | - Joseph A Hill
- Departments of Internal Medicine (Cardiology Division) and
- Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Sergio Lavandero
- Advanced Center for Chronic Disease and Center for Molecular Studies of the Cell, Facultad de Ciencias Quimicas y Farmaceuticas and Facultad de Medicina, Universidad de Chile, Santiago, Chile;
- Departamento de Bioquimica y Biologia Molecular, Facultad de Ciencias Quimicas y Farmaceuticas, Universidad de Chile, Santiago, Chile
- Programa de Biología Celular y Molecular, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile; and
- Departments of Internal Medicine (Cardiology Division) and
| |
Collapse
|
31
|
Lejay A, Fang F, John R, Van JA, Barr M, Thaveau F, Chakfe N, Geny B, Scholey JW. Ischemia reperfusion injury, ischemic conditioning and diabetes mellitus. J Mol Cell Cardiol 2016; 91:11-22. [DOI: 10.1016/j.yjmcc.2015.12.020] [Citation(s) in RCA: 125] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Revised: 11/15/2015] [Accepted: 12/20/2015] [Indexed: 01/08/2023]
|
32
|
Teixeira G, Chiari P, Fauconnier J, Abrial M, Couture-Lepetit E, Harisseh R, Pillot B, Lacampagne A, Tourneur Y, Gharib A, Ovize M. Involvement of Cyclophilin D and Calcium in Isoflurane-induced Preconditioning. Anesthesiology 2015; 123:1374-84. [PMID: 26460965 DOI: 10.1097/aln.0000000000000876] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
BACKGROUND The mitochondrial permeability transition pore (PTP) has been established as an important mediator of ischemia-reperfusion-induced cell death. The matrix protein cyclophilin D (CypD) is the best known regulator of PTP opening. Therefore, the authors hypothesized that isoflurane, by inhibiting the respiratory chain complex I, another regulator of PTP, might reinforce the myocardial protection afforded by CypD inhibition. METHODS Adult mouse or isolated cardiomyocytes from wild-type or CypD knockout (CypD-KO) mice were subjected to ischemia or hypoxia followed by reperfusion or reoxygenation. Infarct size was assessed in vivo. Mitochondrial membrane potential and PTP opening were assessed using tetramethylrhodamine methyl ester perchlorate and calcein-cobalt fluorescence, respectively. Fluo-4 AM and rhod-2 AM staining allowed the measurement, by confocal microscopy, of Ca transient and Ca transfer from sarcoplasmic reticulum (SR) to mitochondria after caffeine stimulation. RESULTS Both inhibition of CypD and isoflurane significantly reduced infarct size (-50 and -37%, respectively) and delayed PTP opening (+63% each). Their combination had no additive effect (n = 6/group). CypD-KO mice displayed endogenous protection against ischemia-reperfusion. Isoflurane depolarized the mitochondrial membrane (-28%, n = 5), decreased oxidative phosphorylation (-59%, n = 5), and blunted the caffeine-induced Ca transfer from SR to mitochondria (-22%, n = 7) in the cardiomyocytes of wild-type mice. Importantly, this transfer was spontaneously decreased in the cardiomyocytes of CypD-KO mice (-25%, n = 4 to 5). CONCLUSIONS The results suggest that the partial inhibitory effect of isoflurane on respiratory complex I is insufficient to afford a synergy to CypD-induced protection. Isoflurane attenuates the Ca transfer from SR to mitochondria, which is also the prominent role of CypD, and finally prevents PTP opening.
Collapse
Affiliation(s)
- Geoffrey Teixeira
- From INSERM UMR-1060, CarMeN Laboratory, Université Lyon-1, Faculté de Médecine Rockefeller, Lyon, France (G.T., P.C., M.A., E.C.-L., R.H., B.P., Y.T., A.G., M.O.); Service d'Anesthésie Réanimation, Hôpital Louis Pradel, Hospices Civils de Lyon, Lyon, France (P.C.); INSERM UMR-1046, Université Montpellier 1, Université Montpellier 2, Centre Hospitalier Universitaire de Montpellier, Montpellier, France (J.F., A.L.); and Service d'Explorations Fonctionnelles Cardiovasculaires and CIC de Lyon, Hôpital Louis Pradel, Hospices Civils de Lyon, Lyon, France (M.O.)
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Schreckenberg R, Rebelo M, Deten A, Weber M, Rohrbach S, Pipicz M, Csonka C, Ferdinandy P, Schulz R, Schlüter KD. Specific Mechanisms Underlying Right Heart Failure: The Missing Upregulation of Superoxide Dismutase-2 and Its Decisive Role in Antioxidative Defense. Antioxid Redox Signal 2015; 23:1220-32. [PMID: 25978844 PMCID: PMC4657518 DOI: 10.1089/ars.2014.6139] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
AIMS Research into right ventricular (RV) physiology and identification of pathomechanisms underlying RV failure have been neglected for many years, because function of the RV is often considered less important for overall hemodynamics and maintenance of blood circulation. In view of this, this study focuses on identifying specific adaptive mechanisms of the RV and left ventricle (LV) during a state of chronic nitric oxide (NO) deficiency, one of the main causes of cardiac failure. NO deficiency was induced in rats by L-NAME feeding over a 4 week period. The cardiac remodeling was then characterized separately for the RV/LV using quantitative real-time polymerase chain reaction, histology, and functional measurements. RESULTS Only the RV underwent remodeling that corresponded morphologically and functionally with the pattern of dilated cardiomyopathy. Symptoms in the LV were subtle and consisted primarily of moderate hypertrophy. A massive increase in reactive oxygen species (ROS) (+4.5±0.8-fold, vs. control) and a higher degree of oxidized tropomyosin (+46%±4% vs. control) and peroxynitrite (+32%±2% vs. control) could be identified as the cause of both RV fibrosis and contractile dysfunction. The expression of superoxide dismutase-2 was specifically increased in the LV by 51%±3% and prevented the ROS increase and the corresponding structural and functional remodeling. INNOVATION This study identified the inability of the RV to increase its antioxidant capacity as an important risk factor for developing RV failure. CONCLUSION Unlike the LV, the RV did not display the necessary adaptive mechanisms to cope with increased oxidative stress during a state of chronic NO deficiency.
Collapse
Affiliation(s)
- Rolf Schreckenberg
- 1 Physiologisches Institut , Justus-Liebig-Universität Gießen, Giessen, Germany
| | - Manuel Rebelo
- 1 Physiologisches Institut , Justus-Liebig-Universität Gießen, Giessen, Germany
| | - Alexander Deten
- 2 Fraunhofer-Institut für Zelltherapie und Immunologie , Leipzig, Germany
| | - Martin Weber
- 1 Physiologisches Institut , Justus-Liebig-Universität Gießen, Giessen, Germany
| | - Susanne Rohrbach
- 1 Physiologisches Institut , Justus-Liebig-Universität Gießen, Giessen, Germany
| | - Márton Pipicz
- 3 Pharmahungary Group , Szeged, Hungary .,4 Cardiovascular Research Group, Department of Biochemistry, University of Szeged , Szeged, Hungary
| | - Csaba Csonka
- 3 Pharmahungary Group , Szeged, Hungary .,4 Cardiovascular Research Group, Department of Biochemistry, University of Szeged , Szeged, Hungary
| | - Péter Ferdinandy
- 3 Pharmahungary Group , Szeged, Hungary .,5 Department of Pharmacology and Pharmacotherapy, Semmelweis University , Budapest, Hungary
| | - Rainer Schulz
- 1 Physiologisches Institut , Justus-Liebig-Universität Gießen, Giessen, Germany
| | | |
Collapse
|
34
|
Altamirano F, Wang ZV, Hill JA. Cardioprotection in ischaemia-reperfusion injury: novel mechanisms and clinical translation. J Physiol 2015; 593:3773-88. [PMID: 26173176 PMCID: PMC4575567 DOI: 10.1113/jp270953] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2015] [Accepted: 06/23/2015] [Indexed: 12/29/2022] Open
Abstract
In recent decades, robust successes have been achieved in conquering the acutely lethal manifestations of heart disease. Nevertheless, the prevalence of heart disease, especially heart failure, continues to rise. Among the precipitating aetiologies, ischaemic disease is a leading cause of heart failure. In the context of ischaemia, the myocardium is deprived of oxygen and nutrients, which elicits a cascade of events that provokes cell death. This ischaemic insult is typically coupled with reperfusion, either spontaneous or therapeutically imposed, wherein blood supply is restored to the previously ischaemic tissue. While this intervention limits ischaemic injury, it triggers a new cascade of events that is also harmful, viz. reperfusion injury. In recent years, novel insights have emerged regarding mechanisms of ischaemia-reperfusion injury, and some hold promise as targets of therapeutic relevance. Here, we review a select number of these pathways, focusing on recent discoveries and highlighting prospects for therapeutic manipulation for clinical benefit.
Collapse
Affiliation(s)
- Francisco Altamirano
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical CenterDallas, TX, 75390, USA
| | - Zhao V Wang
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical CenterDallas, TX, 75390, USA
| | - Joseph A Hill
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical CenterDallas, TX, 75390, USA
- Department of Molecular Biology, University of Texas Southwestern Medical CenterDallas, TX, 75390, USA
| |
Collapse
|
35
|
Inhibition of PKCβ2 overexpression ameliorates myocardial ischaemia/reperfusion injury in diabetic rats via restoring caveolin-3/Akt signaling. Clin Sci (Lond) 2015; 129:331-44. [PMID: 25849791 DOI: 10.1042/cs20140789] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Activation of PKCβ (protein kinase Cβ) plays a critical role in myocardial I/R (ischaemia/reperfusion) injury in non-diabetic rodents. In the myocardium of diabetes, PKCβ2 overexpression is associated with increased vulnerability to post-ischaemic I/R injury with concomitantly impaired cardiomyocyte Cav (caveolin)-3 and Akt signalling compared with non-diabetic rats. We hypothesized that myocardial PKCβ overexpression in diabetes exacerbates myocardial I/R injury through impairing Cav-3/Akt signalling. Streptozotocin-induced diabetic rats were treated with the selective PKCβ inhibitor ruboxistaurin (RBX, 1 mg/kg per day) for 4 weeks, starting from 1 week after diabetes induction, before inducing myocardial I/R achieved by occluding the left descending coronary artery followed by reperfusion. Cardiac function was measured using a pressure-volume conductance system. In an in vitro study, cardiac H9C2 cells were exposed to high glucose (30 mmol/l) and subjected to hypoxia followed by reoxygenation (H/R) in the presence or absence of the selective PKCβ2 inhibitor CGP53353 (1 μmol/l), siRNAs of PKCβ2 or Cav-3 or Akt. Cell apoptosis and mitochondrial membrane potential were assessed by TUNEL (terminal deoxynucleotidyltransferase-mediated dUTP nick-end labelling) and JC-1 staining respectively. RBX significantly decreased post-ischaemic myocardial infarct size (35±5% compared with 49±3% in control, P<0.05) and attenuated cardiac dysfunction, and prevented the reduction in cardiac Cav-3 and enhanced phosphorylated/activated Akt (p-Akt) in diabetic rats (P<0.05). H/R increased cardiomyocyte injury under high glucose conditions as was evident by increased TUNEL-positive and increased JC-1 monomeric cells (P<0.05 compared with control), accompanied with increased PKCβ2 phosphorylation/activation and decreased Cav-3 expression. Either CGP53353 or PKCβ2 siRNA significantly attenuated all of these changes and enhanced p-Akt. Cav-3 gene knockdown significantly reduced p-Akt and increased post-hypoxic cellular and mitochondrial injury despite a concomitant reduction in PKCβ2 phosphorylation. PKCβ2 inhibition with RBX protects diabetic hearts from myocardial I/R injury through Cav-3-dependent activation of Akt.
Collapse
|
36
|
Abstract
Reperfusion is mandatory to salvage ischemic myocardium from infarction, but reperfusion per se contributes to injury and ultimate infarct size. Therefore, cardioprotection beyond that by timely reperfusion is needed to reduce infarct size and improve the prognosis of patients with acute myocardial infarction. The conditioning phenomena provide such cardioprotection, insofar as brief episodes of coronary occlusion/reperfusion preceding (ischemic preconditioning) or following (ischemic postconditioning) sustained myocardial ischemia with reperfusion reduce infarct size. Even ischemia/reperfusion in organs remote from the heart provides cardioprotection (remote ischemic conditioning). The present review characterizes the signal transduction underlying the conditioning phenomena, including their physical and chemical triggers, intracellular signal transduction, and effector mechanisms, notably in the mitochondria. Cardioprotective signal transduction appears as a highly concerted spatiotemporal program. Although the translation of ischemic postconditioning and remote ischemic conditioning protocols to patients with acute myocardial infarction has been fairly successful, the pharmacological recruitment of cardioprotective signaling has been largely disappointing to date.
Collapse
Affiliation(s)
- Gerd Heusch
- From the Institute for Pathophysiology, West German Heart and Vascular Centre, University of Essen Medical School, Essen, Germany.
| |
Collapse
|
37
|
Inserte J, Garcia-Dorado D. The cGMP/PKG pathway as a common mediator of cardioprotection: translatability and mechanism. Br J Pharmacol 2015; 172:1996-2009. [PMID: 25297462 DOI: 10.1111/bph.12959] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Revised: 09/16/2014] [Accepted: 09/26/2014] [Indexed: 12/24/2022] Open
Abstract
Cardiomyocyte cell death occurring during myocardial reperfusion (reperfusion injury) contributes to final infarct size after transient coronary occlusion. Different interrelated mechanisms of reperfusion injury have been identified, including alterations in cytosolic Ca(2+) handling, sarcoplasmic reticulum-mediated Ca(2+) oscillations and hypercontracture, proteolysis secondary to calpain activation and mitochondrial permeability transition. All these mechanisms occur during the initial minutes of reperfusion and are inhibited by intracellular acidosis. The cGMP/PKG pathway modulates the rate of recovery of intracellular pH, but has also direct effect on Ca(2+) oscillations and mitochondrial permeability transition. The cGMP/PKG pathway is depressed in cardiomyocytes by ischaemia/reperfusion and preserved by ischaemic postconditioning, which importantly contributes to postconditioning protection. The present article reviews the mechanisms and consequences of the effect of ischaemic postconditioning on the cGMP/PKG pathway, the different pharmacological strategies aimed to stimulate it during myocardial reperfusion and the evidence, limitations and promise of translation of these strategies to the clinical practice. Overall, the preclinical and clinical evidence suggests that modulation of the cGMP/PKG pathway may be a therapeutic target in the context of myocardial infarction.
Collapse
Affiliation(s)
- Javier Inserte
- Cardiology Department, Vall d'Hebron University Hospital and Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | | |
Collapse
|
38
|
Defective sarcoplasmic reticulum-mitochondria calcium exchange in aged mouse myocardium. Cell Death Dis 2014; 5:e1573. [PMID: 25522267 PMCID: PMC4454162 DOI: 10.1038/cddis.2014.526] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Revised: 10/21/2014] [Accepted: 11/04/2014] [Indexed: 02/06/2023]
Abstract
Mitochondrial alterations are critically involved in increased vulnerability to disease during aging. We investigated the contribution of mitochondria–sarcoplasmic reticulum (SR) communication in cardiomyocyte functional alterations during aging. Heart function (echocardiography) and ATP/phosphocreatine (NMR spectroscopy) were preserved in hearts from old mice (>20 months) with respect to young mice (5–6 months). Mitochondrial membrane potential and resting O2 consumption were similar in mitochondria from young and old hearts. However, maximal ADP-stimulated O2 consumption was specifically reduced in interfibrillar mitochondria from aged hearts. Second generation proteomics disclosed an increased mitochondrial protein oxidation in advanced age. Because energy production and oxidative status are regulated by mitochondrial Ca2+, we investigated the effect of age on mitochondrial Ca2+ uptake. Although no age-dependent differences were found in Ca2+ uptake kinetics in isolated mitochondria, mitochondrial Ca2+ uptake secondary to SR Ca2+ release was significantly reduced in cardiomyocytes from old hearts, and this effect was associated with decreased NAD(P)H regeneration and increased mitochondrial ROS upon increased contractile activity. Immunofluorescence and proximity ligation assay identified the defective communication between mitochondrial voltage-dependent anion channel and SR ryanodine receptor (RyR) in cardiomyocytes from aged hearts associated with altered Ca2+ handling. Age-dependent alterations in SR Ca2+ transfer to mitochondria and in Ca2+ handling could be reproduced in cardiomyoctes from young hearts after interorganelle disruption with colchicine, at concentrations that had no effect in aged cardiomyocytes or isolated mitochondria. Thus, defective SR–mitochondria communication underlies inefficient interorganelle Ca2+ exchange that contributes to energy demand/supply mistmach and oxidative stress in the aged heart.
Collapse
|
39
|
Role of soluble adenylyl cyclase in cell death and growth. Biochim Biophys Acta Mol Basis Dis 2014; 1842:2646-55. [PMID: 25010002 DOI: 10.1016/j.bbadis.2014.06.034] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Revised: 06/26/2014] [Accepted: 06/27/2014] [Indexed: 12/13/2022]
Abstract
cAMP signaling is an evolutionarily conserved intracellular communication system controlling numerous cellular functions. Until recently, transmembrane adenylyl cyclase (tmAC) was considered the major source for cAMP in the cell, and the role of cAMP signaling was therefore attributed exclusively to the activity of this family of enzymes. However, increasing evidence demonstrates the role of an alternative, intracellular source of cAMP produced by type 10 soluble adenylyl cyclase (sAC). In contrast to tmAC, sAC produces cAMP in various intracellular microdomains close to specific cAMP targets, e.g., in nucleus and mitochondria. Ongoing research demonstrates involvement of sAC in diverse physiological and pathological processes. The present review is focused on the role of cAMP signaling, particularly that of sAC, in cell death and growth. Although the contributions of sAC to the regulation of these cellular functions have only recently been discovered, current data suggest that sAC plays key roles in mitochondrial bioenergetics and the mitochondrial apoptosis pathway, as well as cell proliferation and development. Furthermore, recent reports suggest the importance of sAC in several pathologies associated with apoptosis as well as in oncogenesis. This article is part of a Special Issue entitled: The role of soluble adenylyl cyclase in health and disease.
Collapse
|
40
|
Inserte J, Hernando V, Ruiz-Meana M, Poncelas-Nozal M, Fernández C, Agulló L, Sartorio C, Vilardosa Ú, Garcia-Dorado D. Delayed phospholamban phosphorylation in post-conditioned heart favours Ca2+ normalization and contributes to protection. Cardiovasc Res 2014; 103:542-53. [DOI: 10.1093/cvr/cvu163] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
41
|
Garcia-Dorado D, Rodríguez-Sinovas A, Ruiz-Meana M, Inserte J. Protección contra el daño miocárdico por isquemia-reperfusión en la práctica clínica. Rev Esp Cardiol 2014. [DOI: 10.1016/j.recesp.2014.01.011] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
42
|
Garcia-Dorado D, Rodríguez-Sinovas A, Ruiz-Meana M, Inserte J. Protection against myocardial ischemia-reperfusion injury in clinical practice. ACTA ACUST UNITED AC 2014; 67:394-404. [PMID: 24774733 DOI: 10.1016/j.rec.2014.01.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Accepted: 01/22/2014] [Indexed: 12/28/2022]
Abstract
Even when reperfusion therapy is applied as early as possible, survival and quality of life are compromised in a considerable number of patients with ST-segment elevation acute myocardial infarction. Some cell death following transient coronary occlusion occurs during reperfusion, due to poor handling of calcium in the sarcoplasmic reticulum-mitochondria system, calpain activation, oxidative stress, and mitochondrial failure, all promoted by rapid normalization of intracellular pH. Various clinical trials have shown that infarct size can be limited by nonpharmacological strategies--such as ischemic postconditioning and remote ischemic conditioning--or by drugs--such as cyclosporine, insulin, glucagon-like peptide-1 agonists, beta-blockers, or stimulation of cyclic guanosine monophosphate synthesis. However, some clinical studies have yielded negative results, largely due to a lack of consistent preclinical data or a poor design, especially delayed administration. Large-scale clinical trials are therefore necessary, particularly those with primary clinical variables and combined therapies that consider age, sex, and comorbidities, to convert protection against reperfusion injury into a standard treatment for patients with ST-segment elevation acute myocardial infarction.
Collapse
Affiliation(s)
- David Garcia-Dorado
- Hospital Universitario e Institut de Recerca Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain.
| | - Antonio Rodríguez-Sinovas
- Hospital Universitario e Institut de Recerca Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Marisol Ruiz-Meana
- Hospital Universitario e Institut de Recerca Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Javier Inserte
- Hospital Universitario e Institut de Recerca Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
43
|
Martindale JJ, Metzger JM. Uncoupling of increased cellular oxidative stress and myocardial ischemia reperfusion injury by directed sarcolemma stabilization. J Mol Cell Cardiol 2014; 67:26-37. [PMID: 24362314 PMCID: PMC3920738 DOI: 10.1016/j.yjmcc.2013.12.008] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Revised: 12/09/2013] [Accepted: 12/11/2013] [Indexed: 12/31/2022]
Abstract
Myocardial ischemia/reperfusion (I/R) injury is a major clinical problem leading to cardiac dysfunction and myocyte death. It is widely held that I/R causes damage to membrane phospholipids, and is a significant mechanism of cardiac I/R injury. Molecular dissection of sarcolemmal damage in I/R, however, has been difficult to address experimentally. We studied here cardiac I/R injury under conditions targeting gain- or loss-of sarcolemma integrity. To implement gain-in-sarcolemma integrity during I/R, synthetic copolymer-based sarcolemmal stabilizers (CSS), including Poloxamer 188 (P188), were used as a tool to directly stabilize the sarcolemma. Consistent with the hypothesis of sarcolemmal stabilization, cellular markers of necrosis and apoptosis evident in untreated myocytes were fully blocked in sarcolemma stabilized myocytes. Unexpectedly, sarcolemmal stabilization of adult cardiac myocytes did not affect the status of myocyte-generated oxidants or lipid peroxidation in two independent assays. We also investigated the loss of sarcolemmal integrity using two independent genetic mouse models, dystrophin-deficient mdx or dysferlin knockout (Dysf KO) mice. Both models of sarcolemmal loss-of-function were severely affected by I/R injury ex vivo, and this was lessened by CSS. In vivo studies also showed that infarct size was significantly reduced in CSS-treated hearts. Mechanistically, these findings support a model whereby I/R-mediated increased myocyte oxidative stress is uncoupled from myocyte injury. Because the sarcolemma stabilizers used here do not transit across the myocyte membrane this is evidence that intracellular targets of oxidants are not sufficiently altered to affect cell death when sarcolemma integrity is preserved by synthetic stabilizers. These findings, in turn, suggest that sarcolemma destabilization, and consequent Ca(2+) mishandling, as a focal initiating mechanism underlying myocardial I/R injury.
Collapse
Affiliation(s)
- Joshua J Martindale
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Joseph M Metzger
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, USA.
| |
Collapse
|
44
|
Wang Z, Cai F, Hu L, Lu Y. The role of mitochondrial permeability transition pore in regulating the shedding of the platelet GPIbα ectodomain. Platelets 2013; 25:373-81. [DOI: 10.3109/09537104.2013.821604] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
45
|
DeSantiago J, Bare DJ, Banach K. Ischemia/Reperfusion injury protection by mesenchymal stem cell derived antioxidant capacity. Stem Cells Dev 2013; 22:2497-507. [PMID: 23614555 DOI: 10.1089/scd.2013.0136] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Mesenchymal stem cell (MSC) transplantation after ischemia/reperfusion (I/R) injury reduces infarct size and improves cardiac function. We used mouse ventricular myocytes (VMs) in an in vitro model of I/R to determine the mechanism by which MSCs prevent reperfusion injury by paracrine signaling. Exposure of mouse VMs to an ischemic challenge depolarized their mitochondrial membrane potential (Ψmito), increased their diastolic Ca(2+), and significantly attenuated cell shortening. Reperfusion of VMs with Ctrl tyrode or MSC-conditioned tyrode (ConT) resulted in a transient increase of the Ca(2+) transient amplitudes in all cells. ConT-reperfused cells exhibited a decreased number early after depolarization (EADs) (ConT: 6.3% vs. Ctrl: 28.4%) and prolonged survival (ConT: 58% vs. Ctrl: 33%). Ψmito rapidly recovered in Ctrl as well as ConT-treated VMs on reperfusion; however, in Ctrl solution, an exaggerated hyperpolarization of Ψmito was determined that preceded the collapse of Ψmito. The ability of ConT to attenuate the hyperpolarization of Ψmito was suppressed on inhibition of the PI3K/Akt signaling pathway or IK,ATP. However, protection of Ψmito was best mimicked by the reactive oxygen species (ROS) scavenger mitoTEMPO. Analysis of ConT revealed a significant antioxidant capacity that was linked to the presence of extracellular superoxide dismutase (SOD3) in ConT. In conclusion, MSC ConT protects VMs from simulated I/R injury by its SOD3-mediated antioxidant capacity and by delaying the recovery of Ψmito through Akt-mediated opening of IK,ATP. These changes attenuate reperfusion-induced ROS production and prevent the opening of the permeability transition pore and arrhythmic Ca(2+) release.
Collapse
Affiliation(s)
- Jaime DeSantiago
- Section of Cardiology, Department of Medicine, Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, Illinois 60612-7323, USA
| | | | | |
Collapse
|
46
|
Fauconnier J, Roberge S, Saint N, Lacampagne A. Type 2 ryanodine receptor: A novel therapeutic target in myocardial ischemia/reperfusion. Pharmacol Ther 2013; 138:323-32. [DOI: 10.1016/j.pharmthera.2013.01.015] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Accepted: 01/22/2013] [Indexed: 10/27/2022]
|
47
|
Schriewer JM, Peek CB, Bass J, Schumacker PT. ROS-mediated PARP activity undermines mitochondrial function after permeability transition pore opening during myocardial ischemia-reperfusion. J Am Heart Assoc 2013; 2:e000159. [PMID: 23598272 PMCID: PMC3647275 DOI: 10.1161/jaha.113.000159] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Background Ischemia–reperfusion (I/R) studies have implicated oxidant stress, the mitochondrial permeability transition pore (mPTP), and poly(ADP‐ribose) polymerase (PARP) as contributing factors in myocardial cell death. However, the interdependence of these factors in the intact, blood‐perfused heart is not known. We therefore wanted to determine whether oxidant stress, mPTP opening, and PARP activity contribute to the same death pathway after myocardial I/R. Methods and Results A murine left anterior descending coronary artery (LAD) occlusion (30 minutes) and release (1 to 4 hours) model was employed. Experimental groups included controls and antioxidant‐treated, mPTP‐inhibited, or PARP‐inhibited hearts. Antioxidant treatment prevented oxidative damage, mPTP opening, ATP depletion, and PARP activity, placing oxidant stress as the proximal death trigger. Genetic deletion of cyclophilin D (CypD−/−) prevented loss of total NAD+ and PARP activity, and mPTP‐mediated loss of mitochondrial function. Control hearts showed progressive mitochondrial depolarization and loss of ATP from 1.5 to 4 hours of reperfusion, but not outer mitochondrial membrane rupture. Neither genetic deletion of PARP‐1 nor its pharmacological inhibition prevented the initial mPTP‐mediated depolarization or loss of ATP, but PARP ablation did allow mitochondrial recovery by 4 hours of reperfusion. Conclusions These results indicate that oxidant stress, the mPTP, and PARP activity contribute to a single death pathway after I/R in the heart. PARP activation undermines cell survival by preventing mitochondrial recovery after mPTP opening early in reperfusion. This suggests that PARP‐mediated prolongation of mitochondrial depolarization contributes significantly to cell death via an energetic crisis rather than by mitochondrial outer membrane rupture.
Collapse
Affiliation(s)
- Jacqueline M Schriewer
- Division of Neonatology, Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | | | | | | |
Collapse
|
48
|
Shahzad T, Kasseckert SA, Iraqi W, Johnson V, Schulz R, Schlüter KD, Dörr O, Parahuleva M, Hamm C, Ladilov Y, Abdallah Y. Mechanisms involved in postconditioning protection of cardiomyocytes against acute reperfusion injury. J Mol Cell Cardiol 2013; 58:209-16. [PMID: 23328483 DOI: 10.1016/j.yjmcc.2013.01.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2012] [Revised: 12/26/2012] [Accepted: 01/07/2013] [Indexed: 12/12/2022]
Abstract
Experimental and clinical studies demonstrated that postconditioning confers protection against myocardial ischemia/reperfusion injury. However the underlying cellular mechanisms responsible for the beneficial effect of postconditioning are still poorly understood. The aim of the present study was to examine the role of cytosolic and mitochondrial Ca(2+)-handling. For this purpose adult rat cardiomyocytes were subjected to simulated in vitro ischemia (glucose-free hypoxia at pH6.4) followed by simulated reperfusion with a normoxic buffer (pH7.4; 2.5 mmol/L glucose). Postconditioning, i.e., 2 repetitive cycles of normoxic (5s) and hypoxic (2.5 min) superfusion, was applied during the first 5 min of reoxygenation. Mitochondrial membrane potential (ΔΨm), cytosolic and mitochondrial Ca(2+) concentrations, cytosolic pH and necrosis were analysed applying JC-1, fura-2, fura-2/manganese, BCECF and propidium iodide, respectively. Mitochondrial permeability transition pore (MPTP) opening was detected by calcein release. Hypoxic treatment led to a reduction of ΔΨm, an increase in cytosolic and mitochondrial Ca(2+) concentration, and acidification of cardiomyocytes. During the first minutes of reoxygenation, ΔΨm transiently recovered, but irreversibly collapsed after 7 min of reoxygenation, which was accompanied by MPTP opening. Simultaneously, mitochondrial Ca(2+) increased during reperfusion and cardiomyocytes developed spontaneous cytosolic Ca(2+) oscillations and severe contracture followed by necrosis after 25 min of reoxygenation. In postconditioned cells, the collapse in ΔΨm as well as the leak of calcein, the increase in mitochondrial Ca(2+), cytosolic Ca(2+) oscillations, contracture and necrosis were significantly reduced. Furthermore postconditioning delayed cardiomyocyte pH recovery. Postconditioning by hypoxia/reoxygenation was as protective as treatment with cyclosporine A. Combining cyclosporine A and postconditioning had no additive effect. The data of the present study demonstrate that postconditioning by hypoxia/reoxygenation prevents reperfusion injury by limiting mitochondrial Ca(2+) load and thus opening of the MPTP in isolated cardiomyocytes. These effects seem to be supported by postconditioning-induced delay in pH recovery and suppression of Ca(2+) oscillations.
Collapse
Affiliation(s)
- Tayyab Shahzad
- Department of Cardiology and Angiology, University Hospital Giessen and Marburg Klinikstr. 33, 35392 Giessen, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Kleinbongard P, Baars T, Heusch G. Calcium antagonists in myocardial ischemia/reperfusion—update 2012. Wien Med Wochenschr 2012; 162:302-10. [DOI: 10.1007/s10354-012-0113-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2012] [Accepted: 04/03/2012] [Indexed: 01/10/2023]
|
50
|
Yaniv Y, Spurgeon HA, Lyashkov AE, Yang D, Ziman BD, Maltsev VA, Lakatta EG. Crosstalk between mitochondrial and sarcoplasmic reticulum Ca2+ cycling modulates cardiac pacemaker cell automaticity. PLoS One 2012; 7:e37582. [PMID: 22666369 PMCID: PMC3362629 DOI: 10.1371/journal.pone.0037582] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2012] [Accepted: 04/22/2012] [Indexed: 01/02/2023] Open
Abstract
Background Mitochondria dynamically buffer cytosolic Ca2+ in cardiac ventricular cells and this affects the Ca2+ load of the sarcoplasmic reticulum (SR). In sinoatrial-node cells (SANC) the SR generates periodic local, subsarcolemmal Ca2+ releases (LCRs) that depend upon the SR load and are involved in SANC automaticity: LCRs activate an inward Na+-Ca2+ exchange current to accelerate the diastolic depolarization, prompting the ensemble of surface membrane ion channels to generate the next action potential (AP). Objective To determine if mitochondrial Ca2+ (Ca2+m), cytosolic Ca2+ (Ca2+c)-SR-Ca2+ crosstalk occurs in single rabbit SANC, and how this may relate to SANC normal automaticity. Results Inhibition of mitochondrial Ca2+ influx into (Ru360) or Ca2+ efflux from (CGP-37157) decreased [Ca2+]m to 80±8% control or increased [Ca2+]m to 119±7% control, respectively. Concurrent with inhibition of mitochondrial Ca2+ influx or efflux, the SR Ca2+ load, and LCR size, duration, amplitude and period (imaged via confocal linescan) significantly increased or decreased, respectively. Changes in total ensemble LCR Ca2+ signal were highly correlated with the change in the SR Ca2+ load (r2 = 0.97). Changes in the spontaneous AP cycle length (Ru360, 111±1% control; CGP-37157, 89±2% control) in response to changes in [Ca2+]m were predicted by concurrent changes in LCR period (r2 = 0.84). Conclusion A change in SANC Ca2+m flux translates into a change in the AP firing rate by effecting changes in Ca2+c and SR Ca2+ loading, which affects the characteristics of spontaneous SR Ca2+ release.
Collapse
Affiliation(s)
- Yael Yaniv
- Laboratory of Cardiovascular Science, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, United States of America
| | - Harold A. Spurgeon
- Laboratory of Cardiovascular Science, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, United States of America
| | - Alexey E. Lyashkov
- Laboratory of Cardiovascular Science, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, United States of America
| | - Dongmei Yang
- Laboratory of Cardiovascular Science, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, United States of America
| | - Bruce D. Ziman
- Laboratory of Cardiovascular Science, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, United States of America
| | - Victor A. Maltsev
- Laboratory of Cardiovascular Science, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, United States of America
| | - Edward G. Lakatta
- Laboratory of Cardiovascular Science, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|