1
|
Deng M, Chen W, Wang H, Wang Y, Zhou W, Yu T. The disappearance of IPO in myocardium of diabetes mellitus rats is associated with the increase of succinate dehydrogenase-flavin protein. BMC Cardiovasc Disord 2021; 21:142. [PMID: 33731005 PMCID: PMC7968298 DOI: 10.1186/s12872-021-01949-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 03/01/2021] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND The aim of the present study was to investigate whether the disappearance of ischemic post-processing (IPO) in the myocardium of diabetes mellitus (DM) is associated with the increase of succinate dehydrogenase-flavin protein (SDHA). METHODS A total of 50 Sprague Dawley rats, weighing 300-400 g, were divided into 5 groups according to the random number table method, each with 10 rats. After DM rats were fed a high-fat and -sugar diet for 4 weeks, they were injected with Streptozotocin to establish the diabetic rat model. Normal rats were fed the same regular diet for the same number of weeks. Next, the above rats were taken to establish a cardiopulmonary bypass (CPB) model. Intraperitoneal glucose tolerance test (IPGTT) and oral glucose tolerance test (OGTT) were used to detect whether the DM rat model was established successfully. Taking blood from the femoral artery to collect the blood-gas analysis indicators, and judged whether the CPB model is established. After perfusion was performed according to the experimental strategy, the area of myocardial infarction (MI), and serum creatine kinase isoenzyme (CK-MB) and cardiac troponin (CTnI) levels were measured. Finally, the relative mRNA and protein expression of SDHA was detected. RESULTS The OGTT and IPGTT suggested that the DM rat model was successfully established. The arterial blood gas analysis indicated that the CPB model was successfully established. As compared with the N group, the heart function of the IR group was significantly reduced, the levels of myocardial enzyme markers, the area of MI, as well as the relative mRNA and protein expression of SDHA, were all increased. As compared with the IR group, the CK-MB and CTnI levels in the IPO group, the MI area, relative mRNA and protein expression of SDHA decreased. As compared with the IPO group, the myocardial enzyme content in the DM + IPO group, the MI area and the relative mRNA and protein expression of SDHA increased. As compared with the DM + IPO group, in the DM + IPO + dme group, the myocardial enzyme content, area of MI and relative mRNA and protein expression were all decreased. CONCLUSION IPO can inhibit the expression of SDHA, reduce MIRI and exert a cardioprotective effect in the normal rats. However, the protective effect of IPO disappears in the diabetic rats. The inhibitor dme combined with IPO can increase the expression of SDHA and restore the protective effect of IPO in DM myocardia.
Collapse
Affiliation(s)
- Mengyuan Deng
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Huichuan District, Zunyi, 563003, Guizhou, People's Republic of China
| | - Wei Chen
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Huichuan District, Zunyi, 563003, Guizhou, People's Republic of China
| | - Haiying Wang
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Huichuan District, Zunyi, 563003, Guizhou, People's Republic of China.
| | - Yan Wang
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Huichuan District, Zunyi, 563003, Guizhou, People's Republic of China
| | - Wenjing Zhou
- Anesthesia Laboratory, Affiliated Hospital of Zunyi Medical University, Zunyi, People's Republic of China
| | - Tian Yu
- Zunyi Medical University, Zunyi, People's Republic of China
| |
Collapse
|
2
|
Halapas A, Kapelouzou A, Chrissoheris M, Pattakos G, Cokkinos DV, Spargias K. The effect of Remote Ischemic Preconditioning (RIPC) on myocardial injury and inflammation in patients with severe aortic valve stenosis undergoing Transcatheter Aortic Valve Replacement (TAVΙ). Hellenic J Cardiol 2021; 62:423-428. [PMID: 33617961 DOI: 10.1016/j.hjc.2021.02.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 01/01/2021] [Accepted: 02/12/2021] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Remote ischemic preconditioning (RIPC) is being evaluated as a strategy to reduce cardiac injury and inflammation in patients undergoing diverse cardiac invasive and surgical procedures. However, it is unclear whether RIPC has protective effects in patients undergoing the transfemoral- transcatheter aortic valve implantation (TF-TAVΙ) procedure. METHODS Between September 2013 and September 2015, 55 random consecutive patients were prospectively assigned to receive SHAM preconditioning (SHAM, 22 patients) or Remote Ischemic Preconditioning (RIPC) (4 cycles of 5 min intermittent leg ischemia and 5 min reperfusion, 33 patients) prior to TF-TAVI. The primary endpoint was to determine the serum levels of: hs-cTn-I (necrosis), CK-18 (apoptosis), and IL-1b (inflammation). Quantification was performed using commercially available ELISA kits. Patients were sampled 1-day pre TF-TAVΙ and 24-hours post TF-TAVΙ. Secondary endpoints included: total mortality, incidence of periprocedural clinical acute myocardial infarction (AMI), acute kidney injury (AKI), and stroke. RESULTS 22 SHAM patients and 33 RIPC patients were finally analyzed. Our data revealed no significant difference in serum levels of hs-cTn-I and CK-18 among various groups. However, in the RIPC group, the increase in IL1b level was significantly lower for 24-h post TF-TAVΙ, (p < 0.01). There were no significant differences between groups in the secondary endpoints at the follow-up interval of one month. RIPC-related adverse events were not observed. CONCLUSIONS Our data suggest that RIPC did not exhibit significant cardiac or kidney protective effects regarding necrosis and apoptosis in patients undergoing TF-TAVΙ. However, an important anti-inflammatory effect was detected in the RIPC group.
Collapse
Affiliation(s)
- Antonios Halapas
- THV Department, Heart Team Hygeia Hospital Athens Greece, Er. Stavrou 9, Marousi, Athens, Greece.
| | - Alkistis Kapelouzou
- Center of Clinical, Experimental Surgery, & Translation Research, Biomedical Research Foundation Academy of Athens (BRFAA), Soranou Efesiou 4, 11527, Athens, Greece
| | - Michael Chrissoheris
- THV Department, Heart Team Hygeia Hospital Athens Greece, Er. Stavrou 9, Marousi, Athens, Greece
| | - Gregory Pattakos
- THV Department, Heart Team Hygeia Hospital Athens Greece, Er. Stavrou 9, Marousi, Athens, Greece
| | - Dennis V Cokkinos
- Center of Clinical, Experimental Surgery, & Translation Research, Biomedical Research Foundation Academy of Athens (BRFAA), Soranou Efesiou 4, 11527, Athens, Greece
| | - Konstantinos Spargias
- THV Department, Heart Team Hygeia Hospital Athens Greece, Er. Stavrou 9, Marousi, Athens, Greece
| |
Collapse
|
3
|
Sabet Sarvestani F, Azarpira N, Al-Abdullah IH, Tamaddon AM. microRNAs in liver and kidney ischemia reperfusion injury: insight to improve transplantation outcome. Biomed Pharmacother 2020; 133:110944. [PMID: 33227704 DOI: 10.1016/j.biopha.2020.110944] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 10/03/2020] [Accepted: 10/25/2020] [Indexed: 12/26/2022] Open
Abstract
Ischemia reperfusion injury (IRI) is a condition that occurs wherever blood flow and oxygen is reduced or absent, such as trauma, vascular disease, stroke, and solid organ transplantation. This condition can lead to tissue damage, especially during organ transplantation. Under such circumstances, some signaling pathways are activated, leading to up- or down- regulation of several genes such as microRNAs (miRNAs) that might attenuate or ameliorate this status. Therefore, by manipulating miRNAs level, they can be used as a biomarker for early diagnosis of IRI or suggestive to be therapeutic agents in clinical situation in future.
Collapse
Affiliation(s)
| | - Negar Azarpira
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Ismail H Al-Abdullah
- Department of Translational Research and Cellular Therapeutics, Diabetes and Metabolism Research Institute, Beckman Research Institute of City of Hope, Duarte, USA.
| | - Ali-Mohammad Tamaddon
- Department of Pharmaceutics and Center for Nanotechnology in Drug Delivery, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
4
|
Hausenloy DJ, Botker HE, Engstrom T, Erlinge D, Heusch G, Ibanez B, Kloner RA, Ovize M, Yellon DM, Garcia-Dorado D. Targeting reperfusion injury in patients with ST-segment elevation myocardial infarction: trials and tribulations. Eur Heart J 2018; 38:935-941. [PMID: 27118196 PMCID: PMC5381598 DOI: 10.1093/eurheartj/ehw145] [Citation(s) in RCA: 145] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 03/15/2016] [Indexed: 02/07/2023] Open
Affiliation(s)
- Derek J Hausenloy
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Graduate Medical School, 8 College Road, Singapore 169857, Singapore.,National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore 169609, Singapore.,The Hatter Cardiovascular Institute, Institute of Cardiovascular Science, University College London, London, UK.,National Institute of Health Research University College London Hospitals Biomedical Research Centre, London W1T 7DN, UK
| | - Hans Erik Botker
- Department of Cardiology, Aarhus University Hospital Skejby, DK-8200 Aarhus N, Denmark
| | - Thomas Engstrom
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - David Erlinge
- Department of Cardiology, Lund University, Lund, Sweden
| | - Gerd Heusch
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany
| | - Borja Ibanez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain.,IIS-Fundación Jiménez Díaz Hospital, Madrid, Spain
| | - Robert A Kloner
- Huntington Medical Research Institutes, Pasadena, CA, USA.,Division of Cardiovascular Medicine, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Michel Ovize
- Explorations Fonctionnelles Cardiovasculaires, Hôpital Louis Pradel, Lyon, France.,UMR 1060 (CarMeN), Université Claude Bernard, Lyon, France
| | - Derek M Yellon
- The Hatter Cardiovascular Institute, Institute of Cardiovascular Science, University College London, London, UK.,National Institute of Health Research University College London Hospitals Biomedical Research Centre, London W1T 7DN, UK
| | - David Garcia-Dorado
- Department of Cardiology, Vall d'Hebron University Hospital and Research Institute, Universitat Autònoma, Pg Vall d'Hebron 119-129, 08035 Barcelona, Spain
| |
Collapse
|
5
|
Corcoran D, Young R, Cialdella P, McCartney P, Bajrangee A, Hennigan B, Collison D, Carrick D, Shaukat A, Good R, Watkins S, McEntegart M, Watt J, Welsh P, Sattar N, McConnachie A, Oldroyd KG, Berry C. The effects of remote ischaemic preconditioning on coronary artery function in patients with stable coronary artery disease. Int J Cardiol 2018; 252:24-30. [PMID: 29249435 PMCID: PMC5761717 DOI: 10.1016/j.ijcard.2017.10.082] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2017] [Revised: 10/09/2017] [Accepted: 10/19/2017] [Indexed: 01/06/2023]
Abstract
Background Remote ischaemic preconditioning (RIPC) is a cardioprotective intervention invoking intermittent periods of ischaemia in a tissue or organ remote from the heart. The mechanisms of this effect are incompletely understood. We hypothesised that RIPC might enhance coronary vasodilatation by an endothelium-dependent mechanism. Methods We performed a prospective, randomised, sham-controlled, blinded clinical trial. Patients with stable coronary artery disease (CAD) undergoing elective invasive management were prospectively enrolled, and randomised to RIPC or sham (1:1) prior to angiography. Endothelial-dependent vasodilator function was assessed in a non-target coronary artery with intracoronary infusion of incremental acetylcholine doses (10− 6, 10− 5, 10− 4 mol/l). Venous blood was sampled pre- and post-RIPC or sham, and analysed for circulating markers of endothelial function. Coronary luminal diameter was assessed by quantitative coronary angiography. The primary outcome was the between-group difference in the mean percentage change in coronary luminal diameter following the maximal acetylcholine dose (Clinicaltrials.gov identifier: NCT02666235). Results 75 patients were enrolled. Following angiography, 60 patients (mean ± SD age 57.5 ± 8.5 years; 80% male) were eligible and completed the protocol (n = 30 RIPC, n = 30 sham). The mean percentage change in coronary luminal diameter was − 13.3 ± 22.3% and − 2.0 ± 17.2% in the sham and RIPC groups respectively (difference 11.32%, 95%CI: 1.2– 21.4, p = 0.032). This remained significant when age and sex were included as covariates (difference 11.01%, 95%CI: 1.01– 21.0, p = 0.035). There were no between-group differences in endothelial-independent vasodilation, ECG parameters or circulating markers of endothelial function. Conclusions RIPC attenuates the extent of vasoconstriction induced by intracoronary acetylcholine infusion. This endothelium-dependent mechanism may contribute to the cardioprotective effects of RIPC.
Collapse
Affiliation(s)
- D Corcoran
- British Heart Foundation Glasgow Cardiovascular Research Centre, University of Glasgow, Scotland, UK; West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, Glasgow, Scotland, UK
| | - R Young
- Robertson Centre for Biostatistics, University of Glasgow, Scotland, UK
| | - P Cialdella
- West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, Glasgow, Scotland, UK
| | - P McCartney
- British Heart Foundation Glasgow Cardiovascular Research Centre, University of Glasgow, Scotland, UK; West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, Glasgow, Scotland, UK
| | - A Bajrangee
- West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, Glasgow, Scotland, UK
| | - B Hennigan
- British Heart Foundation Glasgow Cardiovascular Research Centre, University of Glasgow, Scotland, UK; West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, Glasgow, Scotland, UK
| | - D Collison
- British Heart Foundation Glasgow Cardiovascular Research Centre, University of Glasgow, Scotland, UK
| | - D Carrick
- West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, Glasgow, Scotland, UK
| | - A Shaukat
- West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, Glasgow, Scotland, UK
| | - R Good
- West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, Glasgow, Scotland, UK
| | - S Watkins
- West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, Glasgow, Scotland, UK
| | - M McEntegart
- West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, Glasgow, Scotland, UK
| | - J Watt
- West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, Glasgow, Scotland, UK
| | - P Welsh
- British Heart Foundation Glasgow Cardiovascular Research Centre, University of Glasgow, Scotland, UK
| | - N Sattar
- British Heart Foundation Glasgow Cardiovascular Research Centre, University of Glasgow, Scotland, UK
| | - A McConnachie
- Robertson Centre for Biostatistics, University of Glasgow, Scotland, UK
| | - K G Oldroyd
- West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, Glasgow, Scotland, UK
| | - C Berry
- British Heart Foundation Glasgow Cardiovascular Research Centre, University of Glasgow, Scotland, UK; West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, Glasgow, Scotland, UK.
| |
Collapse
|
6
|
Truesdell AG, Tehrani B, Singh R, Desai S, Saulino P, Barnett S, Lavanier S, Murphy C. 'Combat' Approach to Cardiogenic Shock. Interv Cardiol 2018; 13:81-86. [PMID: 29928313 DOI: 10.15420/icr.2017:35:3] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
The incidence of cardiogenic shock is rising, patient complexity is increasing and patient survival has plateaued. Mirroring organisational innovations of elite military units, our multidisciplinary medical specialists at the INOVA Heart and Vascular Institute aim to combine the adaptability, agility and cohesion of small teams across our large healthcare system. We advocate for widespread adoption of our 'combat' methodology focused on: increased disease awareness, early multidisciplinary shock team activation, group decision-making, rapid initiation of mechanical circulatory support (as appropriate), haemodynamic-guided management, strict protocol adherence, complete data capture and regular after action reviews, with a goal of ending preventable death from cardiogenic shock.
Collapse
Affiliation(s)
- Alexander G Truesdell
- Virginia Heart, Falls Church VA, USA.,INOVA Heart and Vascular Institute, Falls Church VA, USA
| | - Behnam Tehrani
- INOVA Heart and Vascular Institute, Falls Church VA, USA
| | - Ramesh Singh
- INOVA Heart and Vascular Institute, Falls Church VA, USA
| | - Shashank Desai
- INOVA Heart and Vascular Institute, Falls Church VA, USA
| | | | - Scott Barnett
- INOVA Heart and Vascular Institute, Falls Church VA, USA
| | | | - Charles Murphy
- INOVA Heart and Vascular Institute, Falls Church VA, USA
| |
Collapse
|
7
|
Lescano de Souza Junior A, Mancini Filho J, Pavan Torres R, Irigoyen MC, Curi R. Pretreatment with fish oil attenuates heart ischaemia consequences in rats. Exp Physiol 2017; 102:1459-1473. [PMID: 28879655 DOI: 10.1113/ep086332] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2017] [Accepted: 08/31/2017] [Indexed: 12/15/2022]
Abstract
NEW FINDINGS What is the central question of this study? We investigated whether pretreatment with fish oil could prevent the major consequences of ischaemic injury to the heart. What is the main finding and its importance? Fish oil pretreatment attenuated the consequences of ischaemic injury as indicated by the small infarction area and the preservation of systolic function and coronary blood flow. These findings support the use of fish oil in order to reduce the impact of heart ischaemia. ω-3 Polyunsaturated fatty acid (ω-3 PUFA)-rich fish oil supplementation has protective effects on heart ischaemic injury. Left ventricular (LV) ischaemia was induced in rats by permanent ligation of the left descending coronary artery. Saline, fish oil or soybean oil was administered daily by gavage [3 g (kg body weight)-1 ] for 20 days before inducing ischaemia. Outcomes were assessed 24 h after left descending coronary artery ligation. Pretreatment with fish oil decreased the ω-6/ω-3 fatty acid ratio in the LV. A reduction in infarct size and in the intensity of ventricular systolic dysfunction was found in the fish oil group compared with the saline or soybean oil groups through echocardiographic evaluation. Before infarction, LV glycogen concentrations were decreased in the fish oil group compared with the saline group. Soybean oil pretreatment led to a further increase in the LV levels of CINC-2/αβ, IL-1β and TNF-α induced by the heart infarction. In heart-infarcted rats, fish oil pretreatment decreased creatine kinase and caspase-3 activities; prevented the decrease in the coronary blood flow; increased LV contents of ATP and lactate; increased the mRNA levels of iNOS, eNOS, HIF1α, GLUT1, VEGF-α and p53 in the LV as measured by RT-PCR; and did not change LV pro-inflammatory cytokine concentrations compared with the control group. Fish oil protected the heart from ischaemia, as indicated by the decrease in the heart infarction area and systolic dysfunction associated with increased LV ATP concentrations and maintenance of the coronary blood flow with no change in pro-inflammatory cytokine levels.
Collapse
Affiliation(s)
| | - Jorge Mancini Filho
- Department of Food and Experimental Nutrition, Faculty of Pharmaceutical Sciences, University of Sao Paulo, São Paulo, Brazil
| | - Rosângela Pavan Torres
- Department of Food and Experimental Nutrition, Faculty of Pharmaceutical Sciences, University of Sao Paulo, São Paulo, Brazil
| | | | - Rui Curi
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, São Paulo, Brazil.,Post-Graduate Program in Interdisciplinary Health Sciences, Institute of Physical Activity Sciences and Sports, Cruzeiro do Sul University, Sao Paulo, Brazil
| |
Collapse
|
8
|
Javadov S, Jang S, Parodi-Rullán R, Khuchua Z, Kuznetsov AV. Mitochondrial permeability transition in cardiac ischemia-reperfusion: whether cyclophilin D is a viable target for cardioprotection? Cell Mol Life Sci 2017; 74:2795-2813. [PMID: 28378042 PMCID: PMC5977999 DOI: 10.1007/s00018-017-2502-4] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Revised: 02/28/2017] [Accepted: 03/06/2017] [Indexed: 12/13/2022]
Abstract
Growing number of studies provide strong evidence that the mitochondrial permeability transition pore (PTP), a non-selective channel in the inner mitochondrial membrane, is involved in the pathogenesis of cardiac ischemia-reperfusion and can be targeted to attenuate reperfusion-induced damage to the myocardium. The molecular identity of the PTP remains unknown and cyclophilin D is the only protein commonly accepted as a major regulator of the PTP opening. Therefore, cyclophilin D is an attractive target for pharmacological or genetic therapies to reduce ischemia-reperfusion injury in various animal models and humans. Most animal studies demonstrated cardioprotective effects of PTP inhibition; however, a recent large clinical trial conducted by international groups demonstrated that cyclosporine A, a cyclophilin D inhibitor, failed to protect the heart in patients with myocardial infarction. These studies, among others, raise the question of whether cyclophilin D, which plays an important physiological role in the regulation of cell metabolism and mitochondrial bioenergetics, is a viable target for cardioprotection. This review discusses previous studies to provide comprehensive information on the physiological role of cyclophilin D as well as PTP opening in the cell that can be taken into consideration for the development of new PTP inhibitors.
Collapse
Affiliation(s)
- Sabzali Javadov
- Department of Physiology, School of Medicine, University of Puerto Rico, San Juan, PR 00936-5067, Puerto Rico.
| | - Sehwan Jang
- Department of Physiology, School of Medicine, University of Puerto Rico, San Juan, PR 00936-5067, Puerto Rico
| | - Rebecca Parodi-Rullán
- Department of Physiology, School of Medicine, University of Puerto Rico, San Juan, PR 00936-5067, Puerto Rico
| | - Zaza Khuchua
- Cincinnati Children's Research Foundation, University of Cincinnati, 240 Albert Sabin Way, Cincinnati, OH, 54229, USA
| | - Andrey V Kuznetsov
- Cardiac Surgery Research Laboratory, Department of Cardiac Surgery, Innsbruck Medical University, Innsbruck, Austria
| |
Collapse
|
9
|
Jang S, Lewis TS, Powers C, Khuchua Z, Baines CP, Wipf P, Javadov S. Elucidating Mitochondrial Electron Transport Chain Supercomplexes in the Heart During Ischemia-Reperfusion. Antioxid Redox Signal 2017; 27:57-69. [PMID: 27604998 PMCID: PMC5488255 DOI: 10.1089/ars.2016.6635] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
AIMS Mitochondrial supercomplexes (SCs) are the large supramolecular assembly of individual electron transport chain (ETC) complexes that apparently provide highly efficient ATP synthesis and reduce electron leakage and reactive oxygen species (ROS) production. Oxidative stress during cardiac ischemia-reperfusion (IR) can result in degradation of SCs through oxidation of cardiolipin (CL). Also, IR induces calcium overload and enhances reactive oxygen species (mitROS) in mitochondria that result in the opening of the nonselective permeability transition pores (PTP). The opening of the PTP further compromises cellular energetics and increases mitROS ultimately leading to cell death. Here, we examined the role of PTP-induced mitROS in disintegration of SCs during cardiac IR. The relationship between mitochondrial PTP, ROS, and SCs was investigated using Langendorff-perfused rat hearts subjected to global ischemia (25 min) followed by short-time (5 min) or long-time (60 min) reperfusion in the presence or absence of the PTP inhibitor, sanglifehrin A (SfA), and the mitochondrial targeted ROS and electron scavenger, XJB-5-131. Also, the effects of CL deficiency on SC degradation, PTP, and mitROS were investigated in tafazzin knockdown (TazKD) mice. RESULTS Cardiac IR induced PTP opening and mitROS generation, inhibited by SfA. Percent distributions of SCs were significantly affected by IR, and the effects were dependent on the reperfusion time and reversed by SfA and XJB-5-131. TazKD mice demonstrated a 40% lower SC I + III+IV with reduced basal mitochondrial PTP, ROS, and ETC complex activity. Innovation and Conclusion: Sustained reperfusion after cardiac ischemia induces disintegration of mitochondrial SCs, and PTP-induced ROS presumably play a causal role in SC disassembly. Antioxid. Redox Signal. 27, 57-69.
Collapse
Affiliation(s)
- Sehwan Jang
- Department of Physiology, University of Puerto Rico School of Medicine, San Juan, Puerto Rico
| | - Taber S. Lewis
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Corey Powers
- The Heart Institute, Cincinnati Children's Medical Center and University of Cincinnati, Cincinnati, Ohio
| | - Zaza Khuchua
- The Heart Institute, Cincinnati Children's Medical Center and University of Cincinnati, Cincinnati, Ohio
| | - Christopher P. Baines
- Department of Biomedical Sciences, University of Missouri-Columbia, Columbia, Missouri
| | - Peter Wipf
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Sabzali Javadov
- Department of Physiology, University of Puerto Rico School of Medicine, San Juan, Puerto Rico
| |
Collapse
|
10
|
Abstract
The size of the myocardial infarction remains an important therapeutic target, because heart attack size correlates with mortality and heart failure. In this era, myocardial infarct size is reduced primarily by timely reperfusion of the infarct related coronary artery. Whereas numerous pre-clinical studies have shown that certain pharmacologic agents and therapeutic maneuvers reduce myocardial infarction size greater than reperfusion alone, very few of these therapies have translated to successful clinical trials or standard clinical use. In this review we discuss both the recent successes as well as recent disappointments, and describe some of the newer potential therapies from the preclinical literature that have not yet been tested in clinical trials.
Collapse
|
11
|
Delay C, Paradis S, Charles AL, Thaveau F, Chenesseau B, Zoll J, Chakfe N, Geny B, Lejay A. [Skeletal muscle ischemia-reperfusion and ischemic conditioning pathophysiology-clinical applications for the vascular surgeon]. JOURNAL DE MEDECINE VASCULAIRE 2017; 42:29-38. [PMID: 27989659 DOI: 10.1016/j.jmv.2016.11.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 10/15/2016] [Indexed: 06/06/2023]
Abstract
Ischemia-reperfusion, which is characterized by deficient oxygen supply and subsequent restoration of blood flow, can cause irreversible damage to tissue. The vascular surgeon is daily faced with ischemia-reperfusion situations. Indeed, arterial clamping induces ischemia, followed by reperfusion when declamping. Mechanisms underlying ischemia-reperfusion injury are complex and multifactorial. Increases in cellular calcium and reactive oxygen species, initiated during ischemia and then amplified upon reperfusion are thought to be the main mediators of reperfusion injury. Mitochondrial dysfunction also plays an important role. Extensive research has focused on increasing skeletal muscle tolerance to ischemia-reperfusion injury, especially through the use of ischemic conditioning strategies. The purpose of this review is to focus on the cellular responses associated with ischemia-reperfusion, as well as to discuss the effects of ischemic conditioning strategies. This would help the vascular surgeon in daily practice, in order to try to improve surgical outcome in the setting of ischemia-reperfusion.
Collapse
Affiliation(s)
- C Delay
- Service de chirurgie vasculaire et transplantation rénale, nouvel hôpital civil, 1, place de l'Hôpital, BP 426, 67091 Strasbourg, France
| | - S Paradis
- Unité EA 3072 « Mitochondries, stress oxydant et protection musculaire », université de Strasbourg, 4, rue Kirschleger, 67000 Strasbourg, France
| | - A L Charles
- Unité EA 3072 « Mitochondries, stress oxydant et protection musculaire », université de Strasbourg, 4, rue Kirschleger, 67000 Strasbourg, France
| | - F Thaveau
- Service de chirurgie vasculaire et transplantation rénale, nouvel hôpital civil, 1, place de l'Hôpital, BP 426, 67091 Strasbourg, France
| | - B Chenesseau
- Service de chirurgie vasculaire et transplantation rénale, nouvel hôpital civil, 1, place de l'Hôpital, BP 426, 67091 Strasbourg, France
| | - J Zoll
- Unité EA 3072 « Mitochondries, stress oxydant et protection musculaire », université de Strasbourg, 4, rue Kirschleger, 67000 Strasbourg, France
| | - N Chakfe
- Service de chirurgie vasculaire et transplantation rénale, nouvel hôpital civil, 1, place de l'Hôpital, BP 426, 67091 Strasbourg, France; Unité EA 3072 « Mitochondries, stress oxydant et protection musculaire », université de Strasbourg, 4, rue Kirschleger, 67000 Strasbourg, France
| | - B Geny
- Unité EA 3072 « Mitochondries, stress oxydant et protection musculaire », université de Strasbourg, 4, rue Kirschleger, 67000 Strasbourg, France
| | - A Lejay
- Service de chirurgie vasculaire et transplantation rénale, nouvel hôpital civil, 1, place de l'Hôpital, BP 426, 67091 Strasbourg, France; Unité EA 3072 « Mitochondries, stress oxydant et protection musculaire », université de Strasbourg, 4, rue Kirschleger, 67000 Strasbourg, France.
| |
Collapse
|
12
|
Pan D, Li D. At the crossroads from bench to bedside: luteolin is a promising pharmacological agent against myocardial ischemia reperfusion injury. ANNALS OF TRANSLATIONAL MEDICINE 2017; 4:475. [PMID: 28090531 DOI: 10.21037/atm.2016.11.56] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Affiliation(s)
- Defeng Pan
- The First Clinical College, Nanjing Traditional Chinese Medicine University, Nanjing, China
| | - Dongye Li
- The First Clinical College, Nanjing Traditional Chinese Medicine University, Nanjing, China;; Institute of cardiovascular diseases, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
13
|
Abstract
A core feature of ischemic heart disease is injury to cardiomyocytes (CMC). Ischemic CMC manifest the molecular mechanisms to undergo the major forms of cell injury and death, namely, oncotic necrosis, necroptosis, apoptosis and unregulated autophagy. Important modulators of ischemic injury are reperfusion and conditioning. Mitochondria have a major role in mediating the injury to CMC through membrane protein complexes referred to as death channels. Apoptosis is mediated by activation of a channel regulated by the Bcl-2 protein family leading to mitochondrial outer membrane permeabilization (MOMP). Oncotic type injury is mediated by opening of the mitochondrial permeability transition pore (mPTP). Mitochondria also have a reperfusion salvage kinase pathway (RISK). With cyclosporine A serving as a prototype, ongoing research is aimed at developing pharmacological approaches to condition and preserve mitochondrial integrity in order to promote CMC survival during episodes of myocardial ischemia.
Collapse
|
14
|
See Hoe LE, Schilling JM, Busija AR, Haushalter KJ, Ozberk V, Keshwani MM, Roth DM, Toit ED, Headrick JP, Patel HH, Peart JN. Chronic β1-adrenoceptor blockade impairs ischaemic tolerance and preconditioning in murine myocardium. Eur J Pharmacol 2016; 789:1-7. [PMID: 27373851 DOI: 10.1016/j.ejphar.2016.06.054] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Revised: 06/28/2016] [Accepted: 06/29/2016] [Indexed: 01/20/2023]
Abstract
β-adrenoceptor antagonists are commonly used in ischaemic heart disease (IHD) patients, yet may impair signalling and efficacy of 'cardioprotective' interventions. We assessed effects of chronic β1-adrenoceptor antagonism on myocardial resistance to ischaemia-reperfusion (IR) injury and the ability of cardioprotective interventions [classic ischaemic preconditioning (IPC); novel sustained ligand-activated preconditioning (SLP)] to reduce IR injury in murine hearts. Young male C57Bl/6 mice were untreated or received atenolol (0.5g/l in drinking water) for 4 weeks. Subsequently, two cardioprotective stimuli were evaluated: morphine pellets implanted (to induce SLP, controls received placebo) 5 days prior to Langendorff heart perfusion, and IPC in perfused hearts (3×1.5min ischaemia/2min reperfusion). Atenolol significantly reduced in vivo heart rate. Untreated control hearts exhibited substantial left ventricular dysfunction (~50% pressure development recovery, ~20mmHg diastolic pressure rise) with significant release of lactate dehydrogenase (LDH, tissue injury indicator) after 25min ischaemia/45min reperfusion. Contractile dysfunction and elevated LDH were reduced >50% with IPC and SLP. While atenolol treatment did not modify baseline contractile function, post-ischaemic function was significantly depressed compared to untreated hearts. Atenolol pre-treatment abolished beneficial effects of IPC, whereas SLP protection was preserved. These data indicate that chronic β1-adrenoceptor blockade can exert negative effects on functional IR tolerance and negate conventional IPC (implicating β1-adrenoceptors in IR injury and IPC signalling). However, novel morphine-induced SLP is resistant to inhibition by β1-adrenoceptor antagonism.
Collapse
Affiliation(s)
- Louise E See Hoe
- Menzies Health Institute Queensland, Griffith University, Southport, Australia
| | - Jan M Schilling
- VA San Diego Healthcare System, San Diego, USA; Department of Anesthesiology, University of California San Diego, USA
| | - Anna R Busija
- VA San Diego Healthcare System, San Diego, USA; Department of Anesthesiology, University of California San Diego, USA
| | - Kristofer J Haushalter
- VA San Diego Healthcare System, San Diego, USA; Department of Anesthesiology, University of California San Diego, USA; Department of Chemistry and Biochemistry, University of California San Diego, USA
| | - Victoria Ozberk
- Menzies Health Institute Queensland, Griffith University, Southport, Australia
| | - Malik M Keshwani
- Department of Pharmacology, University of California San Diego, USA
| | - David M Roth
- VA San Diego Healthcare System, San Diego, USA; Department of Anesthesiology, University of California San Diego, USA
| | - Eugene Du Toit
- Menzies Health Institute Queensland, Griffith University, Southport, Australia
| | - John P Headrick
- Menzies Health Institute Queensland, Griffith University, Southport, Australia
| | - Hemal H Patel
- VA San Diego Healthcare System, San Diego, USA; Department of Anesthesiology, University of California San Diego, USA
| | - Jason N Peart
- Menzies Health Institute Queensland, Griffith University, Southport, Australia.
| |
Collapse
|
15
|
Rihal CS, Naidu SS, Givertz MM, Szeto WY, Burke JA, Kapur NK, Kern M, Garratt KN, Goldstein JA, Dimas V, Tu T. 2015 SCAI/ACC/HFSA/STS Clinical Expert Consensus Statement on the Use of Percutaneous Mechanical Circulatory Support Devices in Cardiovascular Care (Endorsed by the American Heart Association, the Cardiological Society of India, and Sociedad Latino Americana de Cardiologia Intervencion; Affirmation of Value by the Canadian Association of Interventional Cardiology-Association Canadienne de Cardiologie d'intervention). J Card Fail 2016; 21:499-518. [PMID: 26036425 DOI: 10.1016/j.cardfail.2015.03.002] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Although historically the intra-aortic balloon pump has been the only mechanical circulatory support device available to clinicians, a number of new devices have become commercially available and have entered clinical practice. These include axial flow pumps, such as Impella(®); left atrial to femoral artery bypass pumps, specifically the TandemHeart; and new devices for institution of extracorporeal membrane oxygenation. These devices differ significantly in their hemodynamic effects, insertion, monitoring, and clinical applicability. This document reviews the physiologic impact on the circulation of these devices and their use in specific clinical situations. These situations include patients undergoing high-risk percutaneous coronary intervention, those presenting with cardiogenic shock, and acute decompensated heart failure. Specialized uses for right-sided support and in pediatric populations are discussed and the clinical utility of mechanical circulatory support devices is reviewed, as are the American College of Cardiology/American Heart Association clinical practice guidelines.
Collapse
Affiliation(s)
- Charanjit S Rihal
- Division of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota.
| | - Srihari S Naidu
- Division of Cardiology, Winthrop University Hospital, Mineola, New York
| | - Michael M Givertz
- Cardiovascular Division, Brigham and Women's Hospital, Boston, Massachusetts
| | - Wilson Y Szeto
- Department of Surgery, University of Pennsylvania, Philadelphia, Pennsylvania
| | - James A Burke
- Division of Cardiology, Lehigh Valley Heart Specialists, Allentown, Pennsylvania
| | - Navin K Kapur
- Cardiology, Tufts Medical Center, Boston, Massachusetts
| | - Morton Kern
- Division of Cardiology, UCI Medical Center, Orange, California
| | - Kirk N Garratt
- Department of Cardiac and Vascular Services, Heart and Vascular Institute of New York, Lenox Hill Hospital, New York, New York
| | - James A Goldstein
- Division of Cardiology, Beaumont Heart Center Clinic, Royal Oak, Michigan
| | - Vivian Dimas
- Pediatric Cardiology, UT Southwestern, Dallas, Texas
| | - Thomas Tu
- Louisville Cardiology Group, Interventional Cardiology, Louisville, Kentucky
| | | | | | | | | | | |
Collapse
|
16
|
|
17
|
Pathobiology of Ischemic Heart Disease: Past, Present and Future. Cardiovasc Pathol 2016; 25:214-220. [PMID: 26897485 DOI: 10.1016/j.carpath.2016.01.007] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2016] [Accepted: 01/28/2016] [Indexed: 12/21/2022] Open
Abstract
This review provides a perspective on knowledge of ischemic heart disease (IHD) obtained from the contemporary era of research which began in the 1960s and has continued to the present day. Important discoveries have been made by basic and translational scientists and clinicians. Pathologists have contributed significantly to insights obtained from experimental studies and clinicopathological studies in humans. The review also provides a perspective for future directions in research in IHD aimed at increasing basic knowledge and developing additional therapeutic options for patients with IHD.
Collapse
|
18
|
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: 13.9] [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]
|
19
|
Sihag S, Haas MS, Kim KM, Guerrero JL, Beaudoin J, Alicot EM, Schuerpf F, Gottschall JD, Puro RJ, Madsen JC, Sachs DH, Newman W, Carroll MC, Allan JS. Natural IgM Blockade Limits Infarct Expansion and Left Ventricular Dysfunction in a Swine Myocardial Infarct Model. Circ Cardiovasc Interv 2016; 9:e002547. [PMID: 26671971 PMCID: PMC4687758 DOI: 10.1161/circinterventions.115.002547] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Accepted: 11/16/2015] [Indexed: 01/25/2023]
Abstract
BACKGROUND Acute coronary syndrome is the leading cause of mortality worldwide. However, treatment of acute coronary occlusion inevitably results in ischemia-reperfusion injury. Circulating natural IgM has been shown to play a significant role in mouse models of ischemia-reperfusion injury. A highly conserved self-antigen, nonmuscle myosin heavy chain II, has been identified as a target of pathogenic IgM. We hypothesized that a monoclonal antibody (m21G6) directed against nonmuscle myosin heavy chain II may inhibit IgM binding and reduce injury in a preclinical model of myocardial infarction. Thus, our objective was to evaluate the efficacy of intravenous m21G6 treatment in limiting infarct expansion, troponin release, and left ventricular dysfunction in a swine myocardial infarction model. METHODS AND RESULTS Massachusetts General Hospital miniature swine underwent occlusion of the midleft anterior descending coronary artery for 60 minutes, followed by 1 hour, 5-day, or 21-day reperfusion. Specificity and localization of m21G6 to injured myocardium were confirmed using fluorescently labeled m21G6. Treatment with m21G6 before reperfusion resulted in a 49% reduction in infarct size (P<0.005) and a 61% reduction in troponin-T levels (P<0.05) in comparison with saline controls at 5-day reperfusion. Furthermore, m21G6-treated animals recovered 85.4% of their baseline left ventricular function as measured by 2-dimensional transthoracic echocardiography in contrast to 67.1% in controls at 21-day reperfusion (P<0.05). CONCLUSIONS Treatment with m21G6 significantly reduced infarct size and troponin-T release, and led to marked preservation of cardiac function in our study. Overall, these findings suggest that pathogenic IgM blockade represents a valid therapeutic strategy in mitigating myocardial ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Smita Sihag
- From the Transplantation Biology Research Center, Massachusetts General Hospital, Charlestown (S.S., J.D.G., J.C.M., D.H.S., J.S.A.); Cardiac Surgery Research Laboratory, Massachusetts General Hospital, Boston, (J.L.G., J.B., J.S.A.); DecImmune Therapeutics, Cambridge, MA (M.S.H., E.M.A., F.S., R.J.P., W.N.); Program in Cellular and Molecular Medicine, Boston Children's Hospital, MA (M.C.C.); Department of Pediatrics, Harvard Medical School, Boston, MA (M.C.C.); and Division of Cardiovascular Surgery, Hospital of the University of Pennsylvania, Philadelphia (K.M.K.)
| | - Michael S Haas
- From the Transplantation Biology Research Center, Massachusetts General Hospital, Charlestown (S.S., J.D.G., J.C.M., D.H.S., J.S.A.); Cardiac Surgery Research Laboratory, Massachusetts General Hospital, Boston, (J.L.G., J.B., J.S.A.); DecImmune Therapeutics, Cambridge, MA (M.S.H., E.M.A., F.S., R.J.P., W.N.); Program in Cellular and Molecular Medicine, Boston Children's Hospital, MA (M.C.C.); Department of Pediatrics, Harvard Medical School, Boston, MA (M.C.C.); and Division of Cardiovascular Surgery, Hospital of the University of Pennsylvania, Philadelphia (K.M.K.)
| | - Karen M Kim
- From the Transplantation Biology Research Center, Massachusetts General Hospital, Charlestown (S.S., J.D.G., J.C.M., D.H.S., J.S.A.); Cardiac Surgery Research Laboratory, Massachusetts General Hospital, Boston, (J.L.G., J.B., J.S.A.); DecImmune Therapeutics, Cambridge, MA (M.S.H., E.M.A., F.S., R.J.P., W.N.); Program in Cellular and Molecular Medicine, Boston Children's Hospital, MA (M.C.C.); Department of Pediatrics, Harvard Medical School, Boston, MA (M.C.C.); and Division of Cardiovascular Surgery, Hospital of the University of Pennsylvania, Philadelphia (K.M.K.)
| | - J Luis Guerrero
- From the Transplantation Biology Research Center, Massachusetts General Hospital, Charlestown (S.S., J.D.G., J.C.M., D.H.S., J.S.A.); Cardiac Surgery Research Laboratory, Massachusetts General Hospital, Boston, (J.L.G., J.B., J.S.A.); DecImmune Therapeutics, Cambridge, MA (M.S.H., E.M.A., F.S., R.J.P., W.N.); Program in Cellular and Molecular Medicine, Boston Children's Hospital, MA (M.C.C.); Department of Pediatrics, Harvard Medical School, Boston, MA (M.C.C.); and Division of Cardiovascular Surgery, Hospital of the University of Pennsylvania, Philadelphia (K.M.K.)
| | - Jonathan Beaudoin
- From the Transplantation Biology Research Center, Massachusetts General Hospital, Charlestown (S.S., J.D.G., J.C.M., D.H.S., J.S.A.); Cardiac Surgery Research Laboratory, Massachusetts General Hospital, Boston, (J.L.G., J.B., J.S.A.); DecImmune Therapeutics, Cambridge, MA (M.S.H., E.M.A., F.S., R.J.P., W.N.); Program in Cellular and Molecular Medicine, Boston Children's Hospital, MA (M.C.C.); Department of Pediatrics, Harvard Medical School, Boston, MA (M.C.C.); and Division of Cardiovascular Surgery, Hospital of the University of Pennsylvania, Philadelphia (K.M.K.)
| | - Elisabeth M Alicot
- From the Transplantation Biology Research Center, Massachusetts General Hospital, Charlestown (S.S., J.D.G., J.C.M., D.H.S., J.S.A.); Cardiac Surgery Research Laboratory, Massachusetts General Hospital, Boston, (J.L.G., J.B., J.S.A.); DecImmune Therapeutics, Cambridge, MA (M.S.H., E.M.A., F.S., R.J.P., W.N.); Program in Cellular and Molecular Medicine, Boston Children's Hospital, MA (M.C.C.); Department of Pediatrics, Harvard Medical School, Boston, MA (M.C.C.); and Division of Cardiovascular Surgery, Hospital of the University of Pennsylvania, Philadelphia (K.M.K.)
| | - Franziska Schuerpf
- From the Transplantation Biology Research Center, Massachusetts General Hospital, Charlestown (S.S., J.D.G., J.C.M., D.H.S., J.S.A.); Cardiac Surgery Research Laboratory, Massachusetts General Hospital, Boston, (J.L.G., J.B., J.S.A.); DecImmune Therapeutics, Cambridge, MA (M.S.H., E.M.A., F.S., R.J.P., W.N.); Program in Cellular and Molecular Medicine, Boston Children's Hospital, MA (M.C.C.); Department of Pediatrics, Harvard Medical School, Boston, MA (M.C.C.); and Division of Cardiovascular Surgery, Hospital of the University of Pennsylvania, Philadelphia (K.M.K.)
| | - James D Gottschall
- From the Transplantation Biology Research Center, Massachusetts General Hospital, Charlestown (S.S., J.D.G., J.C.M., D.H.S., J.S.A.); Cardiac Surgery Research Laboratory, Massachusetts General Hospital, Boston, (J.L.G., J.B., J.S.A.); DecImmune Therapeutics, Cambridge, MA (M.S.H., E.M.A., F.S., R.J.P., W.N.); Program in Cellular and Molecular Medicine, Boston Children's Hospital, MA (M.C.C.); Department of Pediatrics, Harvard Medical School, Boston, MA (M.C.C.); and Division of Cardiovascular Surgery, Hospital of the University of Pennsylvania, Philadelphia (K.M.K.)
| | - Robyn J Puro
- From the Transplantation Biology Research Center, Massachusetts General Hospital, Charlestown (S.S., J.D.G., J.C.M., D.H.S., J.S.A.); Cardiac Surgery Research Laboratory, Massachusetts General Hospital, Boston, (J.L.G., J.B., J.S.A.); DecImmune Therapeutics, Cambridge, MA (M.S.H., E.M.A., F.S., R.J.P., W.N.); Program in Cellular and Molecular Medicine, Boston Children's Hospital, MA (M.C.C.); Department of Pediatrics, Harvard Medical School, Boston, MA (M.C.C.); and Division of Cardiovascular Surgery, Hospital of the University of Pennsylvania, Philadelphia (K.M.K.)
| | - Joren C Madsen
- From the Transplantation Biology Research Center, Massachusetts General Hospital, Charlestown (S.S., J.D.G., J.C.M., D.H.S., J.S.A.); Cardiac Surgery Research Laboratory, Massachusetts General Hospital, Boston, (J.L.G., J.B., J.S.A.); DecImmune Therapeutics, Cambridge, MA (M.S.H., E.M.A., F.S., R.J.P., W.N.); Program in Cellular and Molecular Medicine, Boston Children's Hospital, MA (M.C.C.); Department of Pediatrics, Harvard Medical School, Boston, MA (M.C.C.); and Division of Cardiovascular Surgery, Hospital of the University of Pennsylvania, Philadelphia (K.M.K.)
| | - David H Sachs
- From the Transplantation Biology Research Center, Massachusetts General Hospital, Charlestown (S.S., J.D.G., J.C.M., D.H.S., J.S.A.); Cardiac Surgery Research Laboratory, Massachusetts General Hospital, Boston, (J.L.G., J.B., J.S.A.); DecImmune Therapeutics, Cambridge, MA (M.S.H., E.M.A., F.S., R.J.P., W.N.); Program in Cellular and Molecular Medicine, Boston Children's Hospital, MA (M.C.C.); Department of Pediatrics, Harvard Medical School, Boston, MA (M.C.C.); and Division of Cardiovascular Surgery, Hospital of the University of Pennsylvania, Philadelphia (K.M.K.)
| | - Walter Newman
- From the Transplantation Biology Research Center, Massachusetts General Hospital, Charlestown (S.S., J.D.G., J.C.M., D.H.S., J.S.A.); Cardiac Surgery Research Laboratory, Massachusetts General Hospital, Boston, (J.L.G., J.B., J.S.A.); DecImmune Therapeutics, Cambridge, MA (M.S.H., E.M.A., F.S., R.J.P., W.N.); Program in Cellular and Molecular Medicine, Boston Children's Hospital, MA (M.C.C.); Department of Pediatrics, Harvard Medical School, Boston, MA (M.C.C.); and Division of Cardiovascular Surgery, Hospital of the University of Pennsylvania, Philadelphia (K.M.K.)
| | - Michael C Carroll
- From the Transplantation Biology Research Center, Massachusetts General Hospital, Charlestown (S.S., J.D.G., J.C.M., D.H.S., J.S.A.); Cardiac Surgery Research Laboratory, Massachusetts General Hospital, Boston, (J.L.G., J.B., J.S.A.); DecImmune Therapeutics, Cambridge, MA (M.S.H., E.M.A., F.S., R.J.P., W.N.); Program in Cellular and Molecular Medicine, Boston Children's Hospital, MA (M.C.C.); Department of Pediatrics, Harvard Medical School, Boston, MA (M.C.C.); and Division of Cardiovascular Surgery, Hospital of the University of Pennsylvania, Philadelphia (K.M.K.)
| | - James S Allan
- From the Transplantation Biology Research Center, Massachusetts General Hospital, Charlestown (S.S., J.D.G., J.C.M., D.H.S., J.S.A.); Cardiac Surgery Research Laboratory, Massachusetts General Hospital, Boston, (J.L.G., J.B., J.S.A.); DecImmune Therapeutics, Cambridge, MA (M.S.H., E.M.A., F.S., R.J.P., W.N.); Program in Cellular and Molecular Medicine, Boston Children's Hospital, MA (M.C.C.); Department of Pediatrics, Harvard Medical School, Boston, MA (M.C.C.); and Division of Cardiovascular Surgery, Hospital of the University of Pennsylvania, Philadelphia (K.M.K.)
| |
Collapse
|
20
|
Casós K, Pérez ML, Blasco-Lucas A, Ferrer-Curriu G, Gracia-Baena JM, Sureda C, Permanyer E, Igual A, Galiñanes M. Ischemic postconditioning of the isolated human myocardium: Role of the applied protocol. IJC HEART & VASCULATURE 2015; 8:55-61. [PMID: 28785680 PMCID: PMC5497256 DOI: 10.1016/j.ijcha.2015.05.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 05/03/2015] [Indexed: 01/06/2023]
Abstract
Background Ischemic postconditioning (IPostC), has been proposed as a useful approach to reduce infarct size in all species, but its clinical utility remains unclear. Objective To investigate the role played by the protocol used on the efficacy of IPostC in protecting the diseased human myocardium. Methods Myocardial atrial samples from patients were subjected to a 90 min ischemia/120 min reoxygenation followed by different IPostC protocols to investigate the role of the time of ischemia (30, 60, 90 and 120 s) and the number of cycles (1, 2, 3 and 4) with 60 and 120 s of total ischemic time. Muscles were also subjected to ischemic preconditioning (IPreC). The release of lactate dehydrogenase (LDH) and the measurement of tetrazolium bromide (MTT) were determined. Results IPostC increased the LDH and decreased the MTT values from those of control, independently of the duration of the conditioning ischemia. LDH and MTT values also worsened by augmenting the number of IPostC cycles whereas they were significantly improved by IPreC. However, analysis of individual results indicated that in approximately 1/3 of the cases IPostC exhibited some degree of protection especially in the presence of increased ischemic injury. Conclusions The present findings show that IPostC of the human myocardium may be influenced by the protocol used and also by the degree of the preceding ischemic injury. IPostC was beneficial in approximately 1/3 of the cases; however in the remaining cases it increased ischemic damage and, therefore, these results raise a word of caution on its broad clinical use.
Collapse
Affiliation(s)
- Kelly Casós
- Department of Cardiac Surgery and Reparative Therapy of the Heart, Vall d'Hebron Research Institute, University Hospital Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - María-Llanos Pérez
- Department of Cardiac Surgery and Reparative Therapy of the Heart, Vall d'Hebron Research Institute, University Hospital Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Arnau Blasco-Lucas
- Department of Cardiac Surgery and Reparative Therapy of the Heart, Vall d'Hebron Research Institute, University Hospital Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Gemma Ferrer-Curriu
- Department of Cardiac Surgery and Reparative Therapy of the Heart, Vall d'Hebron Research Institute, University Hospital Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Juan Manuel Gracia-Baena
- Department of Cardiac Surgery and Reparative Therapy of the Heart, Vall d'Hebron Research Institute, University Hospital Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Carlos Sureda
- Department of Cardiac Surgery and Reparative Therapy of the Heart, Vall d'Hebron Research Institute, University Hospital Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Eduard Permanyer
- Department of Cardiac Surgery and Reparative Therapy of the Heart, Vall d'Hebron Research Institute, University Hospital Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Alberto Igual
- Department of Cardiac Surgery and Reparative Therapy of the Heart, Vall d'Hebron Research Institute, University Hospital Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Manuel Galiñanes
- Department of Cardiac Surgery and Reparative Therapy of the Heart, Vall d'Hebron Research Institute, University Hospital Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
21
|
2015 SCAI/ACC/HFSA/STS Clinical Expert Consensus Statement on the Use of Percutaneous Mechanical Circulatory Support Devices in Cardiovascular Care. J Am Coll Cardiol 2015; 65:e7-e26. [DOI: 10.1016/j.jacc.2015.03.036] [Citation(s) in RCA: 354] [Impact Index Per Article: 35.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
22
|
Fiorentino F, Angelini GD, Suleiman MS, Rahman A, Anderson J, Bryan AJ, Culliford LA, Moscarelli M, Punjabi PP, Reeves BC. Investigating the effect of remote ischaemic preconditioning on biomarkers of stress and injury-related signalling in patients having isolated coronary artery bypass grafting or aortic valve replacement using cardiopulmonary bypass: study protocol for a randomized controlled trial. Trials 2015; 16:181. [PMID: 25899533 PMCID: PMC4425928 DOI: 10.1186/s13063-015-0696-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Accepted: 03/30/2015] [Indexed: 01/14/2023] Open
Abstract
Background Ischaemia-reperfusion injury occurs during heart surgery that uses cardiopulmonary bypass (CPB) and cardioplegic arrest. It is hypothesised that remote ischaemic preconditioning (RIPC) protects the heart against such injury. Despite the numerous studies investigating the protective effects of RIPC, there is still uncertainty about the interpretation of the findings as well as conflicting results between studies. The objective of this trial is to investigate the cardioprotective effect of RIPC in patients having coronary artery bypass grafting (CABG) or aortic valve replacement surgery. This will be achieved by estimating the effect of the intervention in the two groups of pathologies and by investigating the signalling mechanisms that may underpin the cardioprotective effect. Methods/Design A two-centre randomised controlled trial will be used to investigate the effects of RIPC in two pathologies: patients having isolated CABG and those having aortic valve replacement surgery (AVR) with CPB. Participants will be randomised to RIPC or control (sham RIPC), stratified by surgical stratum. The intervention will be delivered by a research nurse. Data will be collected by a research nurse blinded to the intervention. The patient and the theatre staff are also blinded to the allocation. Markers of myocardial injury and inflammation will be measured in myocardial biopsies and in blood samples at different times. Discussion This trial is designed to investigate whether RIPC will reduce myocardial injury and inflammation following heart surgery and whether there is a difference in effect between participants having CABG or AVR. This trial is a unique opportunity to study the mechanisms associated with RIPC using human myocardial tissue and blood, and to relate these to the extent of myocardial injury/protection. Trial registration Current Controlled Trials ISRCTN33084113 (25 March 2013). Electronic supplementary material The online version of this article (doi:10.1186/s13063-015-0696-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Francesca Fiorentino
- National Heart and Lung Institute, Cardiothoracic Surgery Department, Imperial College London, Du Cane Road, W12 0NN, London, UK.
| | - Gianni D Angelini
- National Heart and Lung Institute, Cardiothoracic Surgery Department, Imperial College London, Du Cane Road, W12 0NN, London, UK. .,Bristol Heart Institute, University of Bristol, Bristol Royal Infirmary, Marlborough Street, BS2 8HW, Bristol, UK.
| | - M-Saadeh Suleiman
- Bristol Heart Institute, University of Bristol, Bristol Royal Infirmary, Marlborough Street, BS2 8HW, Bristol, UK.
| | - Alima Rahman
- National Heart and Lung Institute, Cardiothoracic Surgery Department, Imperial College London, Du Cane Road, W12 0NN, London, UK.
| | - Jon Anderson
- National Heart and Lung Institute, Cardiothoracic Surgery Department, Imperial College London, Du Cane Road, W12 0NN, London, UK.
| | - Alan J Bryan
- Bristol Heart Institute, University of Bristol, Bristol Royal Infirmary, Marlborough Street, BS2 8HW, Bristol, UK.
| | - Lucy A Culliford
- Bristol Heart Institute, University of Bristol, Bristol Royal Infirmary, Marlborough Street, BS2 8HW, Bristol, UK.
| | - Marco Moscarelli
- National Heart and Lung Institute, Cardiothoracic Surgery Department, Imperial College London, Du Cane Road, W12 0NN, London, UK.
| | - Prakash P Punjabi
- National Heart and Lung Institute, Cardiothoracic Surgery Department, Imperial College London, Du Cane Road, W12 0NN, London, UK.
| | - Barnaby C Reeves
- Bristol Heart Institute, University of Bristol, Bristol Royal Infirmary, Marlborough Street, BS2 8HW, Bristol, UK.
| |
Collapse
|
23
|
Rihal CS, Naidu SS, Givertz MM, Szeto WY, Burke JA, Kapur NK, Kern M, Garratt KN, Goldstein JA, Dimas V, Tu T. 2015 SCAI/ACC/HFSA/STS clinical expert consensus statement on the use of percutaneous mechanical circulatory support devices in cardiovascular care (Endorsed by the American heart assocation, the cardiological society of India, and sociedad latino America. Catheter Cardiovasc Interv 2015; 85:E175-96. [DOI: 10.1002/ccd.25720] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Accepted: 10/25/2014] [Indexed: 12/20/2022]
Affiliation(s)
| | - Srihari S. Naidu
- Division of Cardiology; Winthrop University Hospital; Mineola New York
| | | | - Wilson Y. Szeto
- Department of Surgery; University of Pennsylvania; Philadelphia Pennsylvania
| | - James A. Burke
- Division of Cardiology; Lehigh Valley Heart Specialists; Allentown, PA
| | | | - Morton Kern
- Division of Cardiology; UCI Medical Center; Orange CA
| | - Kirk N. Garratt
- Department of Cardiac and Vascular Services, Heart and Vascular Institute of New York; Lenox Hill Hospital; New York New York
| | - James A. Goldstein
- Division of Cardiology; Beaumont Heart Center Clinic; Royal Oak Michigan
| | - Vivian Dimas
- Pediatric Cardiology; UT Southwestern; Dallas Texas
| | - Thomas Tu
- Louisville Cardiology Group; Interventional Cardiology; Louisville Kentucky
| | | |
Collapse
|
24
|
DeGracia DJ, Tri Anggraini F, Taha DTM, Huang ZF. Inductive and Deductive Approaches to Acute Cell Injury. INTERNATIONAL SCHOLARLY RESEARCH NOTICES 2014; 2014:859341. [PMID: 27437490 PMCID: PMC4897055 DOI: 10.1155/2014/859341] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Accepted: 06/25/2014] [Indexed: 11/28/2022]
Abstract
Many clinically relevant forms of acute injury, such as stroke, traumatic brain injury, and myocardial infarction, have resisted treatments to prevent cell death following injury. The clinical failures can be linked to the currently used inductive models based on biological specifics of the injury system. Here we contrast the application of inductive and deductive models of acute cell injury. Using brain ischemia as a case study, we discuss limitations in inductive inferences, including the inability to unambiguously assign cell death causality and the lack of a systematic quantitative framework. These limitations follow from an overemphasis on qualitative molecular pathways specific to the injured system. Our recently developed nonlinear dynamical theory of cell injury provides a generic, systematic approach to cell injury in which attractor states and system parameters are used to quantitatively characterize acute injury systems. The theoretical, empirical, and therapeutic implications of shifting to a deductive framework are discussed. We illustrate how a deductive mathematical framework offers tangible advantages over qualitative inductive models for the development of therapeutics of acutely injured biological systems.
Collapse
Affiliation(s)
- Donald J. DeGracia
- Department of Physiology, Wayne State University, 4116 Scott Hall, 540 East Canfield Avenue, Detroit, MI 48201, USA
| | - Fika Tri Anggraini
- Department of Physiology, Wayne State University, 4116 Scott Hall, 540 East Canfield Avenue, Detroit, MI 48201, USA
| | | | - Zhi-Feng Huang
- Department of Physics and Astronomy, Wayne State University, Detroit, MI 48201, USA
| |
Collapse
|
25
|
Abstract
With the advent of thrombolytic therapy and angioplasty, it has become possible to reduce myocardial infarct size through early reperfusion. Enormous effort has been expended to find therapies that can further reduce infarct size after early intervention. Animal studies have identified many cardioprotective pathways that have the potential to reduce infarct size if activated before the onset of ischemia. More recently, interventions effective at the onset of reperfusion have been described. Although basic research has identified many targets, most has been conducted in rodent models which may not be directly applicable to human disease and even promising agents have been disappointing in large-scale clinical trials. There are many potential explanations for this failure which is the subject of this review. Potential factors include (1) the variability inherent in the patient population, whereas animal studies usually use single sex homogeneous groups maintained on standard diets in carefully controlled environments; (2) the duration of ischemia is generally shorter in animal studies, resulting in potentially more salvageable myocardium than is often the case in patients; (3) that the animals are usually young without comorbidities, whereas the patient population is generally older and has significant comorbidities; (4) animals are not treated with medications a priori, whereas the patient population is often taking medications that may affect ischemic injury; and (5) animal studies may not involve thorough assessment of effects on organs other than the heart, whereas patients can experience adverse effects of treatment in other organs that can preclude clinical use.
Collapse
Affiliation(s)
- Richard S Vander Heide
- Department of Pathology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA.
| | | |
Collapse
|
26
|
Abstract
There is continued interest in the concept of limiting myocardial infarct size with adjunctive agents administered along with reperfusion injury; however, there remains considerable controversy in the literature. The purpose of this article is to review the medical literature on clinical trials performed during the past 3 years that have attempted to reduce myocardial infarct size by administration of adjunctive therapies along with reperfusion therapy. A PubMed-driven literature search revealed a host of clinical trials focusing on the following prominent types of therapies: endogenous conditioning (postconditioning and remote ischemic conditioning); rapid cooling; pharmacological therapy (cyclosporine, abciximab, clopidogrel, tirofiban, erythropoietin, thrombus aspiration, adenosine, glucose-insulin-potassium, statins, antidiabetic agents, FX06, iron chelation, and ranolazine). Although there remains some controversy, quite a few of these studies showed that adjunctive therapy further reduced myocardial infarct size when coupled with reperfusion. Antiplatelet agents are emerging as some of the newest agents that seem to have cardioprotective capabilities. Postconditioning has become a bit more controversial in the clinical literature; remote conditioning, early and rapid cooling, adenosine, and ranolazine are intriguing therapies deserving of larger studies. Certain agents and maneuvers, such as erythropoietin, protein kinase C δ inhibitors, iron chelation, and intra-aortic balloon counterpulsation, perhaps should be retired. The correct adjunctive therapy administered along with reperfusion has the capability of further reducing myocardial injury during ST-segment-elevation myocardial infarction.
Collapse
Affiliation(s)
- Robert A Kloner
- Division of Cardiovascular Medicine, Department of Medicine, Heart Institute, Good Samaritan Hospital, Los Angeles, CA 90017, USA.
| |
Collapse
|
27
|
Translational Research: An Ongoing Challenge in Cardiac Arrest. Resuscitation 2014. [DOI: 10.1007/978-88-470-5507-0_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
28
|
Brown DA, Sabbah HN, Shaikh SR. Mitochondrial inner membrane lipids and proteins as targets for decreasing cardiac ischemia/reperfusion injury. Pharmacol Ther 2013; 140:258-66. [DOI: 10.1016/j.pharmthera.2013.07.005] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Accepted: 07/09/2013] [Indexed: 01/06/2023]
|
29
|
Brown DA, Hale SL, Baines CP, del Rio CL, Hamlin RL, Yueyama Y, Kijtawornrat A, Yeh ST, Frasier CR, Stewart LM, Moukdar F, Shaikh SR, Fisher-Wellman KH, Neufer PD, Kloner RA. Reduction of early reperfusion injury with the mitochondria-targeting peptide bendavia. J Cardiovasc Pharmacol Ther 2013; 19:121-32. [PMID: 24288396 DOI: 10.1177/1074248413508003] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
We recently showed that Bendavia, a novel mitochondria-targeting peptide, reduced infarction and no-reflow across several experimental models. The purpose of this study was to determine the therapeutic timing and mechanism of action that underlie Bendavia's cytoprotective property. In rabbits exposed to in vivo ischemia/reperfusion (30/180 min), Bendavia administered 20 minutes prior to reperfusion (0.05 mg/kg/h, intravenously) reduced myocardial infarct size by ∼50% when administered for either 1 or 3 hours of reperfusion. However, when Bendavia perfusion began just 10 minutes after the onset of reperfusion, the protection against infarction and no-reflow was completely lost, indicating that the mechanism of protection is occurring early in reperfusion. Experiments in isolated mouse liver mitochondria found no discernible effect of Bendavia on blocking the permeability transition pore, and studies in isolated heart mitochondria showed no effect of Bendavia on respiratory rates. As Bendavia significantly lowered reactive oxygen species (ROS) levels in isolated heart mitochondria, the ROS-scavenging capacity of Bendavia was compared to well-known ROS scavengers using in vitro (cell-free) systems that enzymatically generate ROS. Across doses ranging from 1 nmol/L to 1 mmol/L, Bendavia showed no discernible ROS-scavenging properties, clearly differentiating itself from prototypical scavengers. In conclusion, Bendavia is a promising candidate to reduce cardiac injury when present at the onset of reperfusion but not after reperfusion has already commenced. Given that both infarction and no-reflow are related to increased cellular ROS, Bendavia's protective mechanism of action likely involves reduced ROS generation (as opposed to augmented scavenging) by endothelial and myocyte mitochondria.
Collapse
Affiliation(s)
- David A Brown
- 1Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Brieger DB, Redfern J. Contemporary themes in acute coronary syndrome management: from acute illness to secondary prevention. Med J Aust 2013; 199:174-8. [PMID: 23909538 DOI: 10.5694/mja12.11224] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2012] [Accepted: 06/30/2013] [Indexed: 12/22/2022]
Abstract
Acute coronary syndrome (ACS; myocardial infarction and unstable angina) is the leading cause of mortality in Australia, and those who survive one ACS event are at significant risk of experiencing another. Access to evidence-based and optimal ACS management in both the acute and long-term periods is of great importance. Management of ACS should include appropriate timely revascularisation, medical therapy and ongoing secondary prevention. A key consideration in selecting acute antithrombotic therapies is a careful determination of the risk of bleeding versus risk of recurrent ischaemia. Although there is a strong evidence base for the urgency of delivery and the quality of acute care, knowledge translation is suboptimal. There remains a need for ongoing research and policy development aimed at improving ease and equity of access to evidence-based care. Despite universal guideline recommendations for ongoing secondary prevention strategies, research indicates suboptimal use of evidence-based medications, poor adherence to lifestyle recommendations, and low levels of participation in traditional cardiac rehabilitation. Contemporary secondary prevention programs are evolving into flexible, multifaceted interventions to provide maximal clinical benefits to a majority of patients.
Collapse
|
31
|
Unique transcriptional profile of sustained ligand-activated preconditioning in pre- and post-ischemic myocardium. PLoS One 2013; 8:e72278. [PMID: 23991079 PMCID: PMC3749099 DOI: 10.1371/journal.pone.0072278] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2012] [Accepted: 07/15/2013] [Indexed: 01/07/2023] Open
Abstract
Background Opioidergic SLP (sustained ligand-activated preconditioning) induced by 3–5 days of opioid receptor (OR) agonism induces persistent protection against ischemia-reperfusion (I-R) injury in young and aged hearts, and is mechanistically distinct from conventional preconditioning responses. We thus applied unbiased gene-array interrogation to identify molecular effects of SLP in pre- and post-ischemic myocardium. Methodology/Principal Findings Male C57Bl/6 mice were implanted with 75 mg morphine or placebo pellets for 5 days. Resultant SLP did not modify cardiac function, and markedly reduced dysfunction and injury in perfused hearts subjected to 25 min ischemia/45 min reperfusion. Microarray analysis identified 14 up- and 86 down-regulated genes in normoxic hearts from SLP mice (≥1.3-fold change, FDR≤5%). Induced genes encoded sarcomeric/contractile proteins (Myh7, Mybpc3,Myom2,Des), natriuretic peptides (Nppa,Nppb) and stress-signaling elements (Csda,Ptgds). Highly repressed genes primarily encoded chemokines (Ccl2,Ccl4,Ccl7,Ccl9,Ccl13,Ccl3l3,Cxcl3), cytokines (Il1b,Il6,Tnf) and other proteins involved in inflammation/immunity (C3,Cd74,Cd83, Cd86,Hla-dbq1,Hla-drb1,Saa1,Selp,Serpina3), together with endoplasmic stress proteins (known: Dnajb1,Herpud1,Socs3; putative: Il6, Gadd45g,Rcan1) and transcriptional controllers (Egr2,Egr3, Fos,Hmox1,Nfkbid). Biological themes modified thus related to inflammation/immunity, together with cellular/cardiovascular movement and development. SLP also modified the transcriptional response to I-R (46 genes uniquely altered post-ischemia), which may influence later infarction/remodeling. This included up-regulated determinants of cellular resistance to oxidant (Mgst3,Gstm1,Gstm2) and other forms of stress (Xirp1,Ankrd1,Clu), and repression of stress-response genes (Hspa1a,Hspd1,Hsp90aa,Hsph1,Serpinh1) and Txnip. Conclusions Protection via SLP is associated with transcriptional repression of inflammation/immunity, up-regulation of sarcomeric elements and natriuretic peptides, and modulation of cell stress, growth and development, while conventional protective molecules are unaltered.
Collapse
|
32
|
Reduction of myocardial infarct size with ischemic "conditioning": physiologic and technical considerations. Anesth Analg 2013; 117:891-901. [PMID: 23960036 DOI: 10.1213/ane.0b013e318294fc63] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
A wealth of evidence has revealed that the heart can be "conditioned" and rendered less vulnerable to ischemia-reperfusion injury via the upregulation of endogenous protective signaling pathways. Three distinct conditioning strategies have been identified: (1) preconditioning, the phenomenon where brief episodes of myocardial ischemia (too brief to cause cardiomyocyte death) limit necrosis caused by a subsequent sustained ischemic insult; (2) postconditioning, the concept that relief of myocardial ischemia in a staged or stuttered manner attenuates lethal ischemia-reperfusion injury; and (3) remote conditioning, or upregulation of a cardioprotective phenotype initiated by ischemia in a remote organ or tissue and "transported" to the heart. Progress has been made in defining the technical requirements and limitations of each of the 3 ischemic conditioning models (including the timing and severity of the protective stimulus), as well as elucidating the molecular mechanisms (in particular, the receptor-mediated signaling pathways) responsible for conditioning-induced myocardial protection. Moreover, phase III clinical trials are in progress, seeking to capitalize on the protection that can be achieved by postconditioning and remote conditioning, and applying these strategies in patients undergoing cardiac surgery or angioplasty for the treatment of acute myocardial infarction. There is, however, a potentially important caveat to the clinical translation of myocardial conditioning: emerging data suggest that the efficacy of ischemic conditioning is compromised in aging, diabetic, and hypertensive cohorts, the specific populations in which myocardial protection is most relevant. Successful clinical application of myocardial conditioning will therefore require an understanding of the potential confounding consequences of these comorbidities on the "conditioned" phenotype.
Collapse
|
33
|
Yeh CH, Chen TP, Wang YC, Fang SW, Wun TC. Potent cardioprotection from ischemia-reperfusion injury by a two-domain fusion protein comprising annexin V and Kunitz protease inhibitor. J Thromb Haemost 2013; 11:1454-63. [PMID: 23746209 PMCID: PMC3752160 DOI: 10.1111/jth.12314] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2013] [Accepted: 05/25/2013] [Indexed: 12/16/2022]
Abstract
BACKGROUND Considerable evidence suggests that coagulation proteases (tissue factor [TF]/activated factor VII [FVIIa]/FXa/thrombin) and their target protease activated receptors (PAR-1/PAR-2) play important roles in myocardial ischemia-reperfusion (I-R) injury. We hypothesized that localized inhibition of TF/FVIIa on the membrane surfaces of ischemic cells could effectively block coagulation cascade and subsequent PAR-1/PAR-2 cell signaling, thereby protecting the myocardium from I-R injury. OBJECTIVES We recently developed an annexin V-Kunitz inhibitor fusion protein (ANV-6L15) that could specifically bind to anionic phospholipids on the membrane surfaces of apoptotic cells and efficiently inhibit the membrane-anchored TF/FVIIa. In this study, we investigated the cardioprotective effect of ANV-6L15 in a rat cardiac I-R model in comparison with that of hirudin. METHODS Left coronary artery occlusion was maintained for 45 min followed by 4 h of reperfusion in anesthetized Sprague-Dawley rats. One minute before or 2 min after coronary ligation, rats received an intravenous bolus injection of ANV-6L15 (2.5-250 μg kg(-1) ), vehicle, or hirudin via bolus injection and continuous infusion. RESULTS AND CONCLUSIONS ANV-6L15 dose-dependently reduced infarct size by up to 87% and decreased plasma levels of cardiac troponin I, tumor necrosis factor-α, and soluble intercellular adhesion molecule-1, by up to 97%, 96%, and 66%, respectively, with little impact on the coagulation parameters. ANV-6L15 also ameliorated hemodynamic derangements, attenuated neutrophil infiltration and reduced Terminal deoxynucleotidyl transferase dUTP nick end labeling-positive apoptotic cardiomyocytes. Hirudin was less efficacious even at supraclinical dose. ANV-6L15 confers exceptionally potent cardioprotection and is a promising drug candidate for the prevention of myocardial I-R injury.
Collapse
Affiliation(s)
- Chi-Hsiao Yeh
- Division of Thoracic & Cardiovascular Surgery, Chang Gung Memorial Hospital-Keelung, 222 Mai-Chin Road, Keelung, Taiwan ROC 204
- College of Medicine, Chang Gung University, 259 Wen-Hwa 1st Road, Kwei-Shan Tao-Yuan, Taiwan ROC 333
| | - Tzu-Ping Chen
- Division of Thoracic & Cardiovascular Surgery, Chang Gung Memorial Hospital-Keelung, 222 Mai-Chin Road, Keelung, Taiwan ROC 204
| | - Yao-Chang Wang
- Division of Thoracic & Cardiovascular Surgery, Chang Gung Memorial Hospital-Keelung, 222 Mai-Chin Road, Keelung, Taiwan ROC 204
| | - Shu-Wen Fang
- Division of Thoracic & Cardiovascular Surgery, Chang Gung Memorial Hospital-Keelung, 222 Mai-Chin Road, Keelung, Taiwan ROC 204
| | - Tze-Chein Wun
- EVAS Therapeutics, LLC, 613 Huntley Heights Drive, Ballwin MO 63021, USA
| |
Collapse
|
34
|
Oerlemans MI, Koudstaal S, Chamuleau SA, de Kleijn DP, Doevendans PA, Sluijter JP. Targeting cell death in the reperfused heart: Pharmacological approaches for cardioprotection. Int J Cardiol 2013; 165:410-22. [PMID: 22459400 DOI: 10.1016/j.ijcard.2012.03.055] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2011] [Revised: 01/30/2012] [Accepted: 03/03/2012] [Indexed: 02/08/2023]
|
35
|
Fröhlich GM, Meier P, White SK, Yellon DM, Hausenloy DJ. Myocardial reperfusion injury: looking beyond primary PCI. Eur Heart J 2013; 34:1714-22. [PMID: 23536610 DOI: 10.1093/eurheartj/eht090] [Citation(s) in RCA: 293] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Coronary heart disease (CHD) is the leading cause of death and disability in Europe. For patients presenting with an acute ST-segment elevation myocardial infarction (STEMI), timely myocardial reperfusion using either thrombolytic therapy or primary percutaneous coronary intervention (PPCI) is the most effective therapy for limiting myocardial infarct (MI) size, preserving left-ventricular systolic function and reducing the onset of heart failure. Despite this, the morbidity and mortality of STEMI patients remain significant, and novel therapeutic interventions are required to improve clinical outcomes in this patient group. Paradoxically, the process of myocardial reperfusion can itself induce cardiomyocyte death-a phenomenon which has been termed 'myocardial reperfusion injury' (RI), the irreversible consequences of which include microvascular obstruction and myocardial infarction. Unfortunately, there is currently no effective therapy for preventing myocardial RI in STEMI patients making it an important residual target for cardioprotection. Previous attempts to translate cardioprotective therapies (antioxidants, calcium-channel blockers, and anti-inflammatory agents) for reducing RI into the clinic, have been unsuccessful. An improved understanding of the pathophysiological mechanisms underlying RI has resulted in the identification of several promising mechanical (ischaemic post-conditioning, remote ischaemic pre-conditioning, therapeutic hypothermia, and hyperoxaemia), and pharmacological (atrial natriuretic peptide, cyclosporin-A, and exenatide) therapeutic strategies, for preventing myocardial RI, many of which have shown promise in initial proof-of-principle clinical studies. In this article, we review the pathophysiology underlying myocardial RI, and highlight the potential therapeutic interventions which may be used in the future to prevent RI and improve clinical outcomes in patients with CHD.
Collapse
Affiliation(s)
- Georg M Fröhlich
- The Heart Hospital, University College London Hospitals, 16-18 Westmoreland Street, W1G 8PH, London, UK
| | | | | | | | | |
Collapse
|
36
|
Madonna R, Cevik C, Nasser M. Electrical plasticity and cardioprotection in myocardial ischemia--role of selective sodium channel blockers. Clin Cardiol 2013; 36:255-61. [PMID: 23529949 DOI: 10.1002/clc.22113] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2013] [Accepted: 02/11/2013] [Indexed: 11/08/2022] Open
Abstract
The concept of electrical protection of the ischemic myocardium is in constant evolution and has recently been supported by experimental and clinical studies. Historically, antiplatelet agents, angiotensin-converting enzyme inhibitors, β-blockers, and statins have been all proposed as drugs conferring anti-ischemic cardioprotection. This was supported by the evidence consistently indicating that all these drugs were capable of reducing mortality and the risk of repeat myocardial infarction. The electrical plasticity paradigm is, however, a novel concept that depicts the benefits of improved sodium channel blockade with drugs such as ranolazine and cariporide. Although it has been hypothesized that the protective role of ranolazine depends on decreased fatty acid β-oxidation affecting preconditioning, we speculate against such a hypothesis, because inhibition of β-oxidation requires higher concentrations of the drug, above the therapeutic range. Rather, we discuss the key role of calcium overload reduction through inhibition of the late sodium current (I(Na)). Mechanisms driving cardioprotection involve the block of a cascade of complex ionic exchanges that can result in intracellular acidosis, excess cytosolic calcium, myocardial cellular dysfunction, and eventually cell injury and death. In this review we discuss the studies that demonstrate how electrical plasticity through sodium channel blockers can promote cardioprotection against ischemia in coronary heart disease.
Collapse
Affiliation(s)
- Rosalinda Madonna
- Texas Heart Institute and St. Luke's Episcopal Hospital, Department of adult cardiiology, Houston, Texas, USA.
| | | | | |
Collapse
|
37
|
Hausenloy DJ, Erik Bøtker H, Condorelli G, Ferdinandy P, Garcia-Dorado D, Heusch G, Lecour S, van Laake LW, Madonna R, Ruiz-Meana M, Schulz R, Sluijter JPG, Yellon DM, Ovize M. Translating cardioprotection for patient benefit: position paper from the Working Group of Cellular Biology of the Heart of the European Society of Cardiology. Cardiovasc Res 2013; 98:7-27. [PMID: 23334258 DOI: 10.1093/cvr/cvt004] [Citation(s) in RCA: 180] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Coronary heart disease (CHD) is the leading cause of death and disability worldwide. Despite current therapy, the morbidity and mortality for patients with CHD remains significant. The most important manifestations of CHD arise from acute myocardial ischaemia-reperfusion injury (IRI) in terms of cardiomyocyte death and its long-term consequences. As such, new therapeutic interventions are required to protect the heart against the detrimental effects of acute IRI and improve clinical outcomes. Although a large number of cardioprotective therapies discovered in pre-clinical studies have been investigated in CHD patients, few have been translated into the clinical setting, and a significant number of these have failed to show any benefit in terms of reduced myocardial infarction and improved clinical outcomes. Because of this, there is currently no effective therapy for protecting the heart against the detrimental effects of acute IRI in patients with CHD. One major factor for this lack of success in translating cardioprotective therapies into the clinical setting can be attributed to problems with the clinical study design. Many of these clinical studies have not taken into consideration the important data provided from previously published pre-clinical and clinical studies. The overall aim of this ESC Working Group Cellular Biology of the Heart Position Paper is to provide recommendations for optimizing the design of clinical cardioprotection studies, which should hopefully result in new and effective therapeutic interventions for the future benefit of CHD patients.
Collapse
Affiliation(s)
- Derek J Hausenloy
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London WC1E 6HX, UK.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Koufaki M, Fotopoulou T, Iliodromitis EK, Bibli SI, Zoga A, Kremastinos DT, Andreadou I. Discovery of 6-[4-(6-nitroxyhexanoyl)piperazin-1-yl)]-9H-purine, as pharmacological post-conditioning agent. Bioorg Med Chem 2012; 20:5948-56. [DOI: 10.1016/j.bmc.2012.07.037] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2012] [Revised: 07/05/2012] [Accepted: 07/23/2012] [Indexed: 10/28/2022]
|
39
|
Uryash A, Wu H, Bassuk J, Kurlansky P, Adams JA. Preconditioning with periodic acceleration (pGz) provides second window of cardioprotection. Life Sci 2012; 91:178-85. [DOI: 10.1016/j.lfs.2012.06.031] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2012] [Revised: 05/30/2012] [Accepted: 06/27/2012] [Indexed: 11/27/2022]
|
40
|
Kloner RA, Hale SL, Dai W, Gorman RC, Shuto T, Koomalsingh KJ, Gorman JH, Sloan RC, Frasier CR, Watson CA, Bostian PA, Kypson AP, Brown DA. Reduction of ischemia/reperfusion injury with bendavia, a mitochondria-targeting cytoprotective Peptide. J Am Heart Assoc 2012; 1:e001644. [PMID: 23130143 PMCID: PMC3487333 DOI: 10.1161/jaha.112.001644] [Citation(s) in RCA: 114] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2012] [Accepted: 04/20/2012] [Indexed: 12/26/2022]
Abstract
Background Manifestations of reperfusion injury include myocyte death leading to infarction, contractile dysfunction, and vascular injury characterized by the “no-reflow” phenomenon. Mitochondria-produced reactive oxygen species are believed to be centrally involved in each of these aspects of reperfusion injury, although currently no therapies reduce reperfusion injury by targeting mitochondria specifically. Methods and Results We investigated the cardioprotective effects of a mitochondria-targeted peptide, Bendavia (Stealth Peptides), across a spectrum of experimental cardiac ischemia/reperfusion models. Postischemic administration of Bendavia reduced infarct size in an in vivo sheep model by 15% (P=0.02) and in an ex vivo guinea pig model by 38% to 42% (P<0.05). In an in vivo rabbit model, the extent of coronary no-reflow was assessed with Thioflavin S staining and was significantly smaller in the Bendavia group for any given ischemic risk area than in the control group (P=0.0085). Myocardial uptake of Bendavia was ≈25% per minute, and uptake remained consistent throughout reperfusion. Postischemic recovery of cardiac hemodynamics was not influenced by Bendavia in any of the models studied. Isolated myocytes exposed to hypoxia/reoxygenation showed improved survival when treated with Bendavia. This protection appeared to be mediated by lowered reactive oxygen species–mediated cell death during reoxygenation, associated with sustainment of mitochondrial membrane potential in Bendavia-treated myocytes. Conclusions Postischemic administration of Bendavia protected against reperfusion injury in several distinct models of injury. These data suggest that Bendavia is a mitochondria-targeted therapy that reduces reperfusion injury by maintaining mitochondrial energetics and suppressing cellular reactive oxygen species levels. (J Am Heart Assoc. 2012;1:e001644 doi: 10.1161/JAHA.112.001644.)
Collapse
Affiliation(s)
- Robert A Kloner
- Heart Institute of Good Samaritan Hospital, University of Southern California, Los Angeles (R.A.K., S.H., W.D.) ; Keck School of Medicine, Division of Cardiovascular Medicine, University of Southern California, Los Angeles (R.A.K., W.D.)
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Dow J, Painovich J, Hale SL, Tjen-A-Looi S, Longhurst JC, Kloner RA. Absence of actions of commonly used Chinese herbal medicines and electroacupuncture on myocardial infarct size. J Cardiovasc Pharmacol Ther 2012; 17:403-11. [PMID: 22549008 DOI: 10.1177/1074248412443310] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
BACKGROUND Some studies have suggested that certain Chinese herbal remedies and acupuncture could limit ischemia/reperfusion damage. We sought to determine whether the commonly used single herb Danshen (DS), either alone or in combination with Jiang Xiang (JX), or electroacupuncture (EA) reduces myocardial infarct size. METHODS An anesthetized rat model of proximal left coronary artery occlusion (30 minutes) and reperfusion (180 minutes) was used to measure infarct size (triphenyltetrazolium chloride) and ischemic risk zone (blue dye technique). Rats were either untreated (saline) or received an infusion of DS or DS + JX, starting 30 minutes prior to coronary occlusion. In a separate protocol, rats were untreated, received static needle (ND) placement without stimulation or EA at P5-P6 acupuncture points in the rat forearm starting 5 minutes before occlusion and lasting for 40 minutes, or starting 30 minutes before occlusion and lasting for 90 minutes. RESULTS In the herbal experiments, myocardial infarct size expressed as a fraction of the ischemic risk zone was 0.43 ± 0.06 in controls, 0.39 ± 0.05 in the DS group, and 0.42 ± 0.04 in the Danshen + JX groups (P = not significant [NS]). In the acupuncture study, there was no significant difference in infarct size as a fraction of the risk zone among the control group (0.38 ± 0.04), the ND group (0.47 ± 0.04), or the EA group (0.32 ± 0.05). When EA was started 30 minutes prior to coronary occlusion and continued for 30 minutes into reperfusion, infarct size was 0.41 ± 0.07 in controls and 0.38 ± 0.10 in EA (P = NS). Neither herbs nor EA altered heart rate or blood pressure. In a separate study of 5 minutes of coronary occlusion plus reperfusion, EA failed to reduce ventricular arrhythmias. CONCLUSION Our studies do not suggest a cardioprotective effect of DS or DS + JX or EA in an experimental model of myocardial ischemia/reperfusion.
Collapse
Affiliation(s)
- Joan Dow
- Heart Institute, Good Samaritan Hospital, Los Angeles, CA, USA
| | | | | | | | | | | |
Collapse
|