1
|
Barrère-Lemaire S, Vincent A, Jorgensen C, Piot C, Nargeot J, Djouad F. Mesenchymal stromal cells for improvement of cardiac function following acute myocardial infarction: a matter of timing. Physiol Rev 2024; 104:659-725. [PMID: 37589393 DOI: 10.1152/physrev.00009.2023] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 07/05/2023] [Accepted: 08/16/2023] [Indexed: 08/18/2023] Open
Abstract
Acute myocardial infarction (AMI) is the leading cause of cardiovascular death and remains the most common cause of heart failure. Reopening of the occluded artery, i.e., reperfusion, is the only way to save the myocardium. However, the expected benefits of reducing infarct size are disappointing due to the reperfusion paradox, which also induces specific cell death. These ischemia-reperfusion (I/R) lesions can account for up to 50% of final infarct size, a major determinant for both mortality and the risk of heart failure (morbidity). In this review, we provide a detailed description of the cell death and inflammation mechanisms as features of I/R injury and cardioprotective strategies such as ischemic postconditioning as well as their underlying mechanisms. Due to their biological properties, the use of mesenchymal stromal/stem cells (MSCs) has been considered a potential therapeutic approach in AMI. Despite promising results and evidence of safety in preclinical studies using MSCs, the effects reported in clinical trials are not conclusive and even inconsistent. These discrepancies were attributed to many parameters such as donor age, in vitro culture, and storage time as well as injection time window after AMI, which alter MSC therapeutic properties. In the context of AMI, future directions will be to generate MSCs with enhanced properties to limit cell death in myocardial tissue and thereby reduce infarct size and improve the healing phase to increase postinfarct myocardial performance.
Collapse
Affiliation(s)
- Stéphanie Barrère-Lemaire
- Institut de Génomique Fonctionnelle, Université de Montpellier, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Montpellier, France
- LabEx Ion Channel Science and Therapeutics, Université de Nice, Nice, France
| | - Anne Vincent
- Institut de Génomique Fonctionnelle, Université de Montpellier, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Montpellier, France
- LabEx Ion Channel Science and Therapeutics, Université de Nice, Nice, France
| | - Christian Jorgensen
- Institute of Regenerative Medicine and Biotherapies, Université de Montpellier, Institut National de la Santé et de la Recherche Médicale, Montpellier, France
- Centre Hospitalier Universitaire Montpellier, Montpellier, France
| | - Christophe Piot
- Département de Cardiologie Interventionnelle, Clinique du Millénaire, Montpellier, France
| | - Joël Nargeot
- Institut de Génomique Fonctionnelle, Université de Montpellier, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Montpellier, France
- LabEx Ion Channel Science and Therapeutics, Université de Nice, Nice, France
| | - Farida Djouad
- Institute of Regenerative Medicine and Biotherapies, Université de Montpellier, Institut National de la Santé et de la Recherche Médicale, Montpellier, France
- Centre Hospitalier Universitaire Montpellier, Montpellier, France
| |
Collapse
|
2
|
Citric Acid Cycle Metabolites Predict Infarct Size in Pigs Submitted to Transient Coronary Artery Occlusion and Treated with Succinate Dehydrogenase Inhibitors or Remote Ischemic Perconditioning. Int J Mol Sci 2021; 22:ijms22084151. [PMID: 33923786 PMCID: PMC8072915 DOI: 10.3390/ijms22084151] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 04/13/2021] [Accepted: 04/14/2021] [Indexed: 12/02/2022] Open
Abstract
Succinate dehydrogenase (SDH) inhibition with malonate during reperfusion reduced myocardial infarction in animals, whereas its endogenous substrate, succinate, is detected in plasma from STEMI patients. We investigated whether protection by SDH inhibition is additive to that of remote ischemic perconditioning (RIC) in pigs submitted to transient coronary artery occlusion, and whether protective maneuvers influence plasma levels of citric acid cycle metabolites. Forty pigs were submitted to 40 min coronary occlusion and reperfusion, and allocated to four groups (controls, sodium malonate 10 mmol/L, RIC, and malonate + RIC). Plasma was obtained from femoral and great cardiac veins and analyzed by LC-MS/MS. Malonate, RIC, and malonate + RIC reduced infarct size (24.67 ± 5.98, 25.29 ± 3.92 and 29.83 ± 4.62% vs. 46.47 ± 4.49% in controls, p < 0.05), but no additive effects were detected. Enhanced concentrations of succinate, fumarate, malate and citrate were observed in controls during initial reperfusion in the great cardiac vein, and most were reduced by cardioprotective maneuvers. Concentrations of succinate, fumarate, and malate significantly correlated with infarct size. In conclusion, despite the combination of SDH inhibition during reperfusion and RIC did not result in additive protection, plasma concentrations of selected citric acid cycle metabolites are attenuated by protective maneuvers, correlate with irreversible injury, and might become a prognosis tool in STEMI patients.
Collapse
|
3
|
[Perioperative cardioprotection - From bench to bedside : Current experimental evidence and possible reasons for the limited translation into the clinical setting]. Anaesthesist 2021; 70:401-412. [PMID: 33464375 PMCID: PMC8099823 DOI: 10.1007/s00101-020-00912-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/21/2020] [Indexed: 12/30/2022]
Abstract
Hintergrund Ziel der perioperativen Kardioprotektion ist es, die Auswirkungen eines Ischämie- und Reperfusionsschadens zu minimieren. Aus anästhesiologischer Sicht spielt dieser Aspekt insbesondere in der Herzchirurgie bei Patienten mit Einsatz der Herz-Lungen-Maschine, aber auch allgemein bei längerfristigen hypotensiven Phasen oder perioperativen ischämischen Ereignissen im nichtkardiochirurgischen Setting eine wichtige Rolle. Im Laufe der letzten Jahre konnten diverse pharmakologische sowie nichtpharmakologische Strategien der Kardioprotektion identifiziert werden. Die Ergebnisse von Studien an isoliertem Gewebe sowie von tierexperimentellen In-vivo-Studien sind vielversprechend. Eine Translation dieser kardioprotektiven Strategien in die klinische Praxis ist bislang jedoch nicht gelungen. Große klinische Studien konnten keine signifikante Verbesserung des Outcome der Patienten zeigen. Ziel der Arbeit Dieser Übersichtsartikel gibt einen Überblick über die aktuelle experimentelle Evidenz pharmakologischer und nichtpharmakologischer Kardioprotektion. Außerdem sollen mögliche Gründe für die limitierte Translation diskutiert werden. Schließlich werden Möglichkeiten aufgezeigt, wie der Schritt „from bench to bedside“ in Zukunft doch noch gelingen könnte. Material und Methoden Narrative Übersichtsarbeit. Ergebnisse und Diskussion Trotz der vielversprechenden präklinischen experimentellen Ansätze zum Thema Kardioprotektion besteht nach wie vor eine große Diskrepanz zu den Ergebnissen aus großen klinischen Studien in der perioperativen Phase. Mögliche Gründe für die limitierte Translation könnten insbesondere Komorbiditäten und Komedikationen, die Wahl des Anästhesieverfahrens, aber auch die Wahl des Studiendesigns sein. Eine sorgfältige Studienplanung mit Berücksichtigung der genannten Probleme sowie ein simultaner Einsatz mehrerer kardioprotektiver Strategien mit dem Ziel eines additiven bzw. synergistischen Effekts stellen mögliche Ansätze für die Zukunft dar.
Collapse
|
4
|
Zeng H, Wang L, Zhang J, Pan T, Yu Y, Lu J, Zhou P, Yang H, Li P. Activated PKB/GSK-3 β synergizes with PKC- δ signaling in attenuating myocardial ischemia/reperfusion injury via potentiation of NRF2 activity: Therapeutic efficacy of dihydrotanshinone-I. Acta Pharm Sin B 2021; 11:71-88. [PMID: 33532181 PMCID: PMC7838031 DOI: 10.1016/j.apsb.2020.09.006] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 08/30/2020] [Accepted: 08/31/2020] [Indexed: 12/16/2022] Open
Abstract
Disrupted redox status primarily contributes to myocardial ischemia/reperfusion injury (MIRI). NRF2, the endogenous antioxidant regulator, might provide therapeutic benefits. Dihydrotanshinone-I (DT) is an active component in Salvia miltiorrhiza with NRF2 induction potency. This study seeks to validate functional links between NRF2 and cardioprotection of DT and to investigate the molecular mechanism particularly emphasizing on NRF2 cytoplasmic/nuclear translocation. DT potently induced NRF2 nuclear accumulation, ameliorating post-reperfusion injuries via redox alterations. Abrogated cardioprotection in NRF2-deficient mice and cardiomyocytes strongly supports NRF2-dependent cardioprotection of DT. Mechanistically, DT phosphorylated NRF2 at Ser40, rendering its nuclear-import by dissociating from KEAP1 and inhibiting degradation. Importantly, we identified PKC-δ-(Thr505) phosphorylation as primary upstream event triggering NRF2-(Ser40) phosphorylation. Knockdown of PKC-δ dramatically retained NRF2 in cytoplasm, convincing its pivotal role in mediating NRF2 nuclear-import. NRF2 activity was further enhanced by activated PKB/GSK-3β signaling via nuclear-export signal blockage independent of PKC-δ activation. By demonstrating independent modulation of PKC-δ and PKB/GSK-3β/Fyn signaling, we highlight the ability of DT to exploit both nuclear import and export regulation of NRF2 in treating reperfusion injury harboring redox homeostasis alterations. Coactivation of PKC and PKB phenocopied cardioprotection of DT in vitro and in vivo, further supporting the potential applicability of this rationale.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Hua Yang
- Corresponding authors. Tel./fax: +86 25 83271379.
| | - Ping Li
- Corresponding authors. Tel./fax: +86 25 83271379.
| |
Collapse
|
5
|
Effects of remote ischemic preconditioning on prognosis in patients with lung injury: A meta-analysis. J Clin Anesth 2020; 63:109795. [DOI: 10.1016/j.jclinane.2020.109795] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 02/09/2020] [Accepted: 03/14/2020] [Indexed: 01/29/2023]
|
6
|
Min F, Jia XJ, Gao Q, Niu F, Hu ZY, Han YL, Shi HJ, Yu Y. Remote ischemic post-conditioning protects against myocardial ischemia/reperfusion injury by inhibiting the Rho-kinase signaling pathway. Exp Ther Med 2019; 19:99-106. [PMID: 31853278 PMCID: PMC6909662 DOI: 10.3892/etm.2019.8176] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 10/11/2019] [Indexed: 12/14/2022] Open
Abstract
The aim of the present study was to observe the effect of Rho-kinase on remote ischemic post-conditioning (RIPostC) and explore the underlying mechanisms. Male Sprague Dawley rats (n=32) were randomly distributed into four groups: Sham group, ischemia/reperfusion (I/R) group, RIPostC group and I/R with fasudil group (I/R+Fas). Infarction size was detected by triphenyltetrazolium chloride staining. The levels of creatine kinase (CK), lactate dehydrogenase (LDH), superoxide dismutase (SOD), malondialdehyde (MDA) and cardiac troponin I (cTnI) were measured using an ultraviolet spectrophotometer. The mRNA expression levels of Rho-associated coiled-coil containing protein kinase (ROCK)-1 and ROCK2, B-cell lymphoma 2 (Bcl-2) and Bcl-2-associated X protein (Bax) were detected via reverse transcription-PCR. The protein expression levels of phosphorylated-myosin phosphatase target subunit (p-MYPT1) and phosphorylated-myosin light chain (p-MLC) were assessed by western blotting. The results demonstrated that RIPostC could decrease the infarct size, the levels of CK, LDH, cTnI and MDA and increase the activity of SOD compared with the I/R group. In addition, the mRNA expression of ROCK1 and ROCK2 was downregulated, the protein expression of p-MYPT1 and p-MLC was decreased, and the ratio of Bcl-2/Bax was elevated in the RIPostC groups compared with the I/R group. Notably, the aforementioned index in I/R with Fas group was similar to the RIPostC group and no significant difference was observed between RIPostC and I/R+Fas. These results revealed that RIPostC could attenuate I/R injury and the underlying mechanisms might be associated with a reduction in myocardial apoptosis and the suppression of the Rho-kinase signaling pathway.
Collapse
Affiliation(s)
- Feng Min
- Department of Epidemiology and Statistics, Bengbu Medical College, Bengbu, Anhui 233030, P.R. China.,Research Center of Bengbu Medical College, Bengbu, Anhui 233030, P.R. China
| | - Xian Jie Jia
- Department of Epidemiology and Statistics, Bengbu Medical College, Bengbu, Anhui 233030, P.R. China
| | - Qin Gao
- Research Center of Bengbu Medical College, Bengbu, Anhui 233030, P.R. China.,Department of Physiology, Bengbu Medical College, Bengbu, Anhui 233030, P.R. China
| | - Fang Niu
- Department of Epidemiology and Statistics, Bengbu Medical College, Bengbu, Anhui 233030, P.R. China
| | - Zhi Yuan Hu
- Research Center of Bengbu Medical College, Bengbu, Anhui 233030, P.R. China
| | - Ya Ling Han
- Department of Epidemiology and Statistics, Bengbu Medical College, Bengbu, Anhui 233030, P.R. China
| | - Hong Jie Shi
- Department of Epidemiology and Statistics, Bengbu Medical College, Bengbu, Anhui 233030, P.R. China
| | - Ying Yu
- Research Center of Bengbu Medical College, Bengbu, Anhui 233030, P.R. China.,Department of Physiology, Bengbu Medical College, Bengbu, Anhui 233030, P.R. China
| |
Collapse
|
7
|
Davidson SM, Ferdinandy P, Andreadou I, Bøtker HE, Heusch G, Ibáñez B, Ovize M, Schulz R, Yellon DM, Hausenloy DJ, Garcia-Dorado D. Multitarget Strategies to Reduce Myocardial Ischemia/Reperfusion Injury: JACC Review Topic of the Week. J Am Coll Cardiol 2019; 73:89-99. [PMID: 30621955 DOI: 10.1016/j.jacc.2018.09.086] [Citation(s) in RCA: 477] [Impact Index Per Article: 79.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 09/20/2018] [Indexed: 01/04/2023]
Abstract
Many treatments have been identified that confer robust cardioprotection in experimental animal models of acute ischemia and reperfusion injury. However, translation of these cardioprotective therapies into the clinical setting of acute myocardial infarction (AMI) for patient benefit has been disappointing. One important reason might be that AMI is multifactorial, causing cardiomyocyte death via multiple mechanisms, as well as affecting other cell types, including platelets, fibroblasts, endothelial and smooth muscle cells, and immune cells. Many cardioprotective strategies act through common end-effectors and may be suboptimal in patients with comorbidities. In this regard, emerging data suggest that optimal cardioprotection may require the combination of additive or synergistic multitarget therapies. This review will present an overview of the state of cardioprotection today and provide a roadmap for how we might progress towards successful clinical use of cardioprotective therapies following AMI, focusing on the rational combination of judiciously selected, multitarget therapies. This paper emerged as part of the discussions of the European Union (EU)-CARDIOPROTECTION Cooperation in Science and Technology (COST) Action, CA16225.
Collapse
Affiliation(s)
- Sean M Davidson
- The Hatter Cardiovascular Institute, University College London, London, United Kingdom.
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary; Pharmahungary Group, Szeged, Hungary
| | - Ioanna Andreadou
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Hans Erik Bøtker
- Department of Cardiology, Aarhus University Hospital Skejby, Aarhus N, Denmark
| | - Gerd Heusch
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany
| | - Borja Ibáñez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain; CIBER de Enfermedades CardioVasculares, Madrid, Spain; IIS-Fundación Jiménez Díaz University Hospital, Madrid, Spain
| | - Michel Ovize
- INSERM U1060, CarMeN Laboratory, Université de Lyon and Explorations Fonctionnelles Cardiovasculaires, Hôpital Louis Pradel, Hospices Civils de Lyon, Lyon, France
| | - Rainer Schulz
- Institute of Physiology, Justus-Liebig University Giessen, Giessen, Germany
| | - Derek M Yellon
- The Hatter Cardiovascular Institute, University College London, London, United Kingdom
| | - Derek J Hausenloy
- The Hatter Cardiovascular Institute, University College London, London, United Kingdom; Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore; National Heart Research Institute Singapore, National Heart Centre, Singapore; Yong Loo Lin School of Medicine, National University Singapore, Singapore; Tecnologico de Monterrey, Escuela de Ingenieria y Ciencias, Centro de Biotecnologia-FEMSA, Nuevo Leon, México
| | - David Garcia-Dorado
- IIS-Fundación Jiménez Díaz University Hospital, Madrid, Spain; Department of Cardiology, Vascular Biology and Metabolism Area, Vall d'Hebron University Hospital and Research Institute (VHIR), Barcelona, Spain; Universitat Autónoma de Barcelona, Barcelona, Spain.
| |
Collapse
|
8
|
Zhao JJ, Xiao H, Zhao WB, Zhang XP, Xiang Y, Ye ZJ, Mo MM, Peng XT, Wei L. Remote Ischemic Postconditioning for Ischemic Stroke: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Chin Med J (Engl) 2018; 131:956-965. [PMID: 29664057 PMCID: PMC5912063 DOI: 10.4103/0366-6999.229892] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Background: Remote ischemic postconditioning (RIPostC) appears to protect distant organs from ischemia-reperfusion injury (IRI). However, cerebral protection results have remained inconclusive. In the present study, a meta-analysis was performed to compare stroke patients with and without RIPostC. Methods: CNKI, WanFang, VIP, CBM, PubMed, and Cochrane Library databases were searched up to July 2016. Data were analyzed using both fixed-effects and random-effects models by Review Manager. For each outcome, risk ratio (RR) and mean difference (MD) with 95% confidence interval (CI) were calculated. Results: A total of 13 randomized controlled trials that enrolled a total of 794 study participants who suffered from or are at risk for brain IRI were selected. Compared with controls, RIPostC significantly reduced the recurrence of stroke or transient ischemic attacks (RR = 0.37; 95% CI: 0.26–0.55; P < 0.00001). Moreover, it can reduce the levels of the National Institutes of Health Stroke Scale score (MD: 1.96; 95% CI: 2.18–1.75; P < 0.00001), modified Rankin Scale score (MD: 0.73; 95% CI: 1.20–0.25; P = 0.00300), and high-sensitivity C-reactive protein (MD: 4.17; 95% CI: 4.71–3.62; P < 0.00001) between the two groups. There was no side effect of RIPostC using tourniquet cuff around the limb on ischemic stroke treating based on different intervention duration. Conclusion: The present meta-analysis suggests that RIPostC might offer cerebral protection for stroke patients suffering from or are at risk of brain IRI.
Collapse
Affiliation(s)
- Jing-Jing Zhao
- Department of Nursing, Chongqing Medical and Pharmaceutical College, Chongqing 401331, China
| | - Hui Xiao
- Nursing Department, Guangdong Province Hospital of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510120, China
| | - Wen-Bo Zhao
- Department of Nephrology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510632, China
| | - Xiao-Pei Zhang
- Department of Neurology, Guangdong Province Hospital of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510120, China
| | - Yu Xiang
- Department of Neurology, Huaihua Hospital of Traditional Chinese Medicine, Huaihua, Hunan 418000, China
| | - Zeng-Jie Ye
- Department of Economics and Management, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510120, China
| | - Miao-Miao Mo
- Department of Neurology, Guangdong Province Hospital of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510120, China
| | - Xue-Ting Peng
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510120, China
| | - Lin Wei
- Department of Neurology, Guangdong Province Hospital of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510120, China
| |
Collapse
|
9
|
Hausenloy DJ, Garcia-Dorado D, Bøtker HE, Davidson SM, Downey J, Engel FB, Jennings R, Lecour S, Leor J, Madonna R, Ovize M, Perrino C, Prunier F, Schulz R, Sluijter JPG, Van Laake LW, Vinten-Johansen J, Yellon DM, Ytrehus K, Heusch G, Ferdinandy P. Novel targets and future strategies for acute cardioprotection: Position Paper of the European Society of Cardiology Working Group on Cellular Biology of the Heart. Cardiovasc Res 2018; 113:564-585. [PMID: 28453734 DOI: 10.1093/cvr/cvx049] [Citation(s) in RCA: 254] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2016] [Accepted: 03/15/2017] [Indexed: 02/06/2023] Open
Abstract
Ischaemic heart disease and the heart failure that often results, remain the leading causes of death and disability in Europe and worldwide. As such, in order to prevent heart failure and improve clinical outcomes in patients presenting with an acute ST-segment elevation myocardial infarction and patients undergoing coronary artery bypass graft surgery, novel therapies are required to protect the heart against the detrimental effects of acute ischaemia/reperfusion injury (IRI). During the last three decades, a wide variety of ischaemic conditioning strategies and pharmacological treatments have been tested in the clinic-however, their translation from experimental to clinical studies for improving patient outcomes has been both challenging and disappointing. Therefore, in this Position Paper of the European Society of Cardiology Working Group on Cellular Biology of the Heart, we critically analyse the current state of ischaemic conditioning in both the experimental and clinical settings, provide recommendations for improving its translation into the clinical setting, and highlight novel therapeutic targets and new treatment strategies for reducing acute myocardial IRI.
Collapse
Affiliation(s)
- Derek J Hausenloy
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London WC1E 6HX, UK; The National Institute of Health Research University College London Hospitals Biomedical Research Centre, 149 Tottenham Court Road London, W1T 7DN, UK; Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore, 8 College Road, Singapore 169857; National Heart Research Institute Singapore, National Heart Centre Singapore, 5 Hospital Dr, Singapore 169609, Singapore; Yong Loo Lin School of Medicine, National University Singapore, Singapore; Barts Heart Centre, St Bartholomew's Hospital, London, UK
| | - David Garcia-Dorado
- Department of Cardiology, Vall d Hebron University Hospital and Research Institute. Universitat Autònoma, Passeig de la Vall d'Hebron, 119-129, 08035 Barcelona, Spain
| | - Hans Erik Bøtker
- Department of Cardiology, Aarhus University Hospital Skejby, Palle Juul-Jensens Boulevard 99, 8200 Aarhus N, Denmark
| | - Sean M Davidson
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London WC1E 6HX, UK
| | - James Downey
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, 5851 USA Dr. N., MSB 3074, Mobile, AL 36688, USA
| | - Felix B Engel
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nßrnberg, Schloßplatz 4, 91054 Erlangen, Germany
| | - Robert Jennings
- Department of Cardiology, Duke University, Durham, NC 27708, USA
| | - Sandrine Lecour
- Department of Medicine, Hatter Institute for Cardiovascular Research in Africa and South African Medical Research Council Inter-University Cape Heart Group, Faculty of Health Sciences, University of Cape Town, Chris Barnard Building, Anzio Road, Observatory, 7925, Cape Town, Western Cape, South Africa
| | - Jonathan Leor
- Tamman Cardiovascular Research Institute, Sheba Medical Center, Tel Hashomer, Israel; Neufeld Cardiac Research Institute, Tel-Aviv University, Sheba Medical Center, Tel Hashomer, 5265601, Israel; Sheba Center for Regenerative Medicine, Stem Cell, and Tissue Engineering, Tel Hashomer, 5265601, Israel
| | - Rosalinda Madonna
- Center of Aging Sciences and Translational Medicine - CESI-MeT, "G. d'Annunzio" University, Chieti, Italy; Institute of Cardiology, Department of Neurosciences, Imaging, and Clinical Sciences, "G. d'Annunzio University, Chieti, Italy; Texas Heart Institute and University of Texas Medical School in Houston, Department of Internal Medicine, 6770 Bertner Avenue, Houston, Texas 77030 USA
| | - Michel Ovize
- Explorations Fonctionnelles Cardiovasculaires, Hôpital Louis Pradel, 28 Avenue du Doyen Jean Lépine, 69500 Bron, France; UMR 1060 (CarMeN), Université Claude Bernard Lyon, 43 Boulevard du 11 Novembre 1918, 69100 Villeurbanne, France
| | - Cinzia Perrino
- Department of Advanced Biomedical Sciences, Division of Cardiology, Federico II University Corso Umberto I, 40, 80138 Napoli, Italy
| | - Fabrice Prunier
- Department of Cardiology, University of Angers, University Hospital of Angers, 4 Rue Larrey, 49100 Angers, France
| | - Rainer Schulz
- Institute of Physiology, Justus-Liebig, University of Giessen, Ludwigstraße 23, 35390 Gießen, Germany
| | - Joost P G Sluijter
- Cardiology and UMC Utrecht Regenerative Medicine Center, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, Netherlands
| | - Linda W Van Laake
- Division Heart and Lungs, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, Netherlands
| | - Jakob Vinten-Johansen
- Division of Cardiothoracic Surgery, Department of Surgery, Emory University, 201 Dowman Dr, Atlanta, GA 30322, USA
| | - Derek M Yellon
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London WC1E 6HX, UK; The National Institute of Health Research University College London Hospitals Biomedical Research Centre, 149 Tottenham Court Road London, W1T 7DN, UK
| | - Kirsti Ytrehus
- Cardiovascular Research Group, Department of Medical Biology, UiT The Arctic University of Norway, Hansine Hansens veg 18, 9019 Tromsø, Norway
| | - Gerd Heusch
- Institute for Pathophysiology, West-German Heart and Vascular Center, University Hospital Essen, Hufelandstrasse 55, 45147 Essen, Germany
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Nagyvárad tér 4, 1089 Hungary; Pharmahungary Group, Graphisoft Park, 7 Záhony street, Budapest, H-1031, Hungary
| |
Collapse
|
10
|
Saeid F, Aniseh J, Reza B, Manouchehr VS. Signaling mediators modulated by cardioprotective interventions in healthy and diabetic myocardium with ischaemia-reperfusion injury. Eur J Prev Cardiol 2018; 25:1463-1481. [PMID: 29442529 DOI: 10.1177/2047487318756420] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Ischaemic heart diseases are one of the major causes of death in the world. In most patients, ischaemic heart disease is coincident with other risk factors such as diabetes. Patients with diabetes are more prone to cardiac ischaemic dysfunctions including ischaemia-reperfusion injury. Ischaemic preconditioning, postconditioning and remote conditionings are reliable interventions to protect the myocardium against ischaemia-reperfusion injuries through activating various signaling pathways and intracellular mediators. Diabetes can disrupt the intracellular signaling cascades involved in these myocardial protections, and studies have revealed that cardioprotective effects of the conditioning interventions are diminished in the diabetic condition. The complex pathophysiology and poor prognosis of ischaemic heart disease among people with diabetes necessitate the investigation of the interaction of diabetes with ischaemia-reperfusion injury and cardioprotective mechanisms. Reducing the outcomes of ischaemia-reperfusion injury using targeted strategies would be particularly helpful in this population. In this study, we review the protective interventional signaling pathways and mediators which are activated by ischaemic conditioning strategies in healthy and diabetic myocardium with ischaemia-reperfusion injury.
Collapse
Affiliation(s)
- Feyzizadeh Saeid
- 1 Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,2 Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,3 Department of Biochemistry and Clinical Laboratories, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Javadi Aniseh
- 4 Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Badalzadeh Reza
- 1 Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,5 Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Vafaee S Manouchehr
- 6 Department of Nuclear Medicine, Odense University Hospital, Odense-Denmark.,7 Institute of Clinical Research, Department of Psychiatry, University of Southern Denmark, Odense-Denmark.,8 Neuroscience Research Centre, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
11
|
Remote Ischemic Postconditioning Protects against Myocardial Ischemia-Reperfusion Injury by Inhibition of the RAGE-HMGB1 Pathway. BIOMED RESEARCH INTERNATIONAL 2018; 2018:4565630. [PMID: 29789792 PMCID: PMC5896327 DOI: 10.1155/2018/4565630] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 11/23/2017] [Accepted: 12/24/2017] [Indexed: 01/26/2023]
Abstract
Background The aim of the present study was to observe the effect of RAGE-HMGB1 signal pathway on remote ischemic postconditioning in mice with myocardial ischemia reperfusion injury. Methods Mice model of MIRI was established and randomly divided into three groups: control group, ischemia reperfusion group, and remote ischemic postconditioning group. Infarction size was detected by Evans blue and TTC staining. Cardiac function was detected by echocardiography measurement. The protein levels of RAGE, HMGB1, P-AKT, and ERK1/2 were detected by Western blot 120 min following reperfusion. Results RIPostC could decrease the infarct size and increase LVEF and FS compared with I/R group. Two hours after myocardial ischemia reperfusion, the levels of RAGE and HMGB1 were significantly decreased in RIPostC group compared with those in I/R group. The level of p-AKT was significantly higher in the RIPostC group than in the I/R group. LY294002 significantly attenuated RIPostC-increased levels of Akt phosphorylation. Conclusion RIPostC may inhibit the expression of RAGE and HMGB1 and activate PI3K/Akt signaling pathway to extenuate ischemic reperfusion injury in mice. It could further suppress the oxidative stress, have antiapoptosis effect, and reduce inflammatory reaction, but this effect has certain timeliness.
Collapse
|
12
|
Costa FLDS, Yamaki VN, Teixeira RKC, Feijó DH, Valente AL, Carvalho LTFD, Yasojima EY, Brito MVH. Perconditioning combined with postconditioning on kidney ischemia and reperfusion. Acta Cir Bras 2017; 32:599-606. [PMID: 28902935 DOI: 10.1590/s0102-865020170080000001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 07/17/2017] [Indexed: 11/22/2022] Open
Abstract
Purpose: To evaluate if combination of perconditioning and postconditioning provides improved renal protection compared to perconditioning alone in a model of renal reperfusion injury. Methods: Thirty rats were assigned into 6 groups: normality; sham; ischemia and reperfusion; postconditioning; perconditioning; perconditioning + postconditioning. Animals were subjected to right nephrectomy and left renal ischemia for 30 minutes. Postconditioning consisted of 3 cycles of 5 min renal perfusion followed by 5 min of renal ischemia after major ischemic period. Perconditioning consisted of 3 cycles of 5 min hindlimb ischemia followed by 5 min of hindlimb perfusion contemporaneously to renal major ischemic period. After 24 hours, kidney was harvested and blood collected to measure urea and creatinine. Results: Perconditioning obtained better values for creatinine and urea level than only postconditioning (p<0.01); performing both techniques contemporaneously had no increased results (p>0.05). Regarding tissue structure, perconditioning was the only technique to protect the glomerulus and tubules (p<0.05), while postconditioning protected only the glomerulus (p<0.05). Combination of both techniques shows no effect on glomerulus or tubules (p>0.05). Conclusions: Perconditioning had promising results on ischemia and reperfusion induced kidney injury, enhanced kidney function and protected glomerulus and tubules. There was no additive protection when postconditioning and perconditioning were combined.
Collapse
Affiliation(s)
- Felipe Lobato da Silva Costa
- MD, Department of Experimental Surgery, School of Medicine, Universidade do Estado do Pará (UEPA), Belem-PA, Brazil. Conception, design, intellectual and scientific content of the study; interpretation of data; manuscript writing
| | - Vitor Nagai Yamaki
- MD, School of Medicine, UEPA, Belem-PA, Brazil. Acquisition and interpretation of data, statistical analysis
| | - Renan Kleber Costa Teixeira
- Fellow Master degree, Department of Experimental Surgery, UEPA, Belem-PA, Brazil. Interpretation of data, manuscript writing, critical revision
| | - Daniel Haber Feijó
- Graduate student, School of Medicine, UEPA, Belem-PA, Brazil. Interpretation of data, manuscript preparation
| | - André Lopes Valente
- Graduate student, School of Medicine, UEPA, Belem-PA, Brazil. Acquisition and interpretation of data, manuscript preparation
| | - Luan Teles Ferreira de Carvalho
- Graduate student, School of Medicine, UEPA, Belem-PA, Brazil. Acquisition and interpretation of data, manuscript preparation
| | - Edson Yuzur Yasojima
- PhD, Associate Professor, Department of Experimental Surgery, School of Medicine, UEPA, Belem-PA, Brazil. Conception, design, intellectual and scientific content of the study; critical revision
| | - Marcus Vinicius Henriques Brito
- PhD, Full Professor, Department of Experimental Surgery, School of Medicine, UEPA, Belem-PA, Brazil. Conception, design, intellectual and scientific content of the study; critical revision
| |
Collapse
|
13
|
Yu Y, Zhou CH, Yao YT, Li LH. Downregulation of Na +/Ca 2+ Exchanger Isoform 1 Protects Isolated Hearts by Sevoflurane Postconditioning but Not by Delayed Remote Ischemic Preconditioning in Rats. Chin Med J (Engl) 2017; 130:2226-2233. [PMID: 28875959 PMCID: PMC5598336 DOI: 10.4103/0366-6999.213967] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Background: Calcium regulatory proteins-L-type Ca2+ channels (LTCCs), ryanodine receptor 2 (RyR2), and Na+/Ca2+ exchanger isoform 1 (NCX1) have been recognized as important protective mechanisms during myocardial ischemia-reperfusion injury (I/RI). Both sevoflurane postconditioning (SevoPoC) and delayed remote ischemic preconditioning (DRIPC) have been shown to protect the heart against I/RI. In this study, we aimed to compare the effects of SevoPoC and DRIPC on the expression of the three calcium regulatory proteins in an isolated rat heart model. Methods: After 30-min balanced perfusion, isolated hearts from rats were subjected to 30-min ischemia followed by 60-min reperfusion. Totally 40 isolated hearts were randomly assigned to four groups (n = 10/group): time control group, I/RI group, SevoPoC group, and DRIPC group. The effect of SevoPoC (3% v/v) and DRIPC were observed. Myocardial infarct size (IS), cardiac troponin I level, and heart function were measured. The protein and messenger RNA levels of LTCCs, RyR2, and NCX1 were determined. Results: Both SevoPoC and DRIPC improved the recovery of myocardial function, and reduced cardiac troponin I release after I/RI. The decrease in IS was more significant in the SevoPoC group than that in the DRIPC group (16.50% ± 4.54% in the SevoPoC group [P = 0.0006], and 22.34% ± 4.02% in the DRIPC group [P = 0.0007] vs. 35.00% ± 5.24% in the I/RI group, respectively). SevoPoC, but not DRIPC significantly inhibited the activity of NCX1 (0.59 ± 0.09 in the I/RI group vs. 0.32 ± 0.16 in the SevoPoC group, P = 0.006; vs. 0.57 ± 0.14 in the DRIPC group, P = 0.072). No statistical significant differences were observed in the expression of LTCCs and RyR2 between SevoPoC and DRIPC. In addition, subsequent correlation analysis showed a significantly positive relationship between the cardiac troponin I level and the protein expression of NCX1 (r = 0.505, P = 0.023). Conclusion: SevoPoC may be more effective in the cardioprotection than DRIPC partly due to the deactivation of NCX1.
Collapse
Affiliation(s)
- Yang Yu
- Department of Anesthesiology, Fuwai Cardiovascular Hospital, Beijing 100037, China
| | - Cheng-Hui Zhou
- Department of Anesthesiology, Fuwai Cardiovascular Hospital, Beijing 100037, China
| | - Yun-Tai Yao
- Department of Anesthesiology, Fuwai Cardiovascular Hospital, Beijing 100037, China
| | - Li-Huan Li
- Department of Anesthesiology, Fuwai Cardiovascular Hospital, Beijing 100037, China
| |
Collapse
|
14
|
Youn YJ, Yoo BS, Son JW, Lee JW, Ahn MS, Ahn SG, Kim JY, Lee SH, Yoon J, Eom YW, Oh JE, Choi SK. Remote Ischemic Conditioning by Effluent Collected from a Novel Isolated Hindlimb Model Reduces Infarct Size in an Isolated Heart Model. Korean Circ J 2017; 47:714-726. [PMID: 28955390 PMCID: PMC5614948 DOI: 10.4070/kcj.2017.0092] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 05/16/2017] [Accepted: 05/22/2017] [Indexed: 02/03/2023] Open
Abstract
Background and Objectives Experimental protocols for remote ischemic conditioning (RIC) utilize models in which a tourniquet is placed around the hindlimb or effluent is collected from an isolated heart. In analyzing the humoral factors that act as signal transducers in these models, sampled blood can be influenced by systemic responses, while the effluent from an isolated heart might differ from that of the hindlimb. Thus, we designed a new isolated hindlimb model for RIC and tested whether the effluent from this model could affect ischemia/reperfusion (IR) injury and if the reperfusion injury salvage kinase (RISK) and survivor activating factor enhancement (SAFE) pathways are involved in RIC. Materials and Methods After positioning needles into the right iliac artery and vein of rats, Krebs-Henseleit buffer was perfused using a Langendorff apparatus, and effluent was collected. The RIC protocol consisted of 3 cycles of IR for 5 minutes. In the RIC effluent group, collected effluent was perfused in an isolated heart for 10 minutes before initiating IR injury. Results Compared with the control group, the infarct area in the RIC effluent group was significantly smaller (31.2%±3.8% vs. 20.6%±1.8%, p<0.050), while phosphorylation of signal transducer and activation of transcription-3 (STAT-3) was significantly increased. However, there was a trend of increased phosphorylation of extracellular signal-regulated kinase (ERK) 1/2 in this group. Conclusion This is the first study to investigate the effect of effluent from a new isolated hindlimb model after RIC on IR injury in an isolated heart model. The RIC effluent was effective in reducing the IR injury, and the cardioprotective effect was associated with activation of the SAFE pathway.
Collapse
Affiliation(s)
- Young Jin Youn
- Division of Cardiology, Department of Internal Medicine, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Byung-Su Yoo
- Division of Cardiology, Department of Internal Medicine, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Jung-Woo Son
- Division of Cardiology, Department of Internal Medicine, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Jun-Won Lee
- Division of Cardiology, Department of Internal Medicine, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Min-Soo Ahn
- Division of Cardiology, Department of Internal Medicine, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Sung Gyun Ahn
- Division of Cardiology, Department of Internal Medicine, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Jang-Young Kim
- Division of Cardiology, Department of Internal Medicine, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Seung-Hwan Lee
- Division of Cardiology, Department of Internal Medicine, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Junghan Yoon
- Division of Cardiology, Department of Internal Medicine, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Young Woo Eom
- Cell Therapy and Tissue Engineering Center, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Ji-Eun Oh
- Cell Therapy and Tissue Engineering Center, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Seong-Kyung Choi
- Animal Core, Central Research Laboratory, Yonsei University Wonju College of Medicine, Wonju, Korea
| |
Collapse
|
15
|
Maslov LN, Podoksenov YK, Tsibul’nikov SY, Gorbunov AS, Tsepokina AV, Khutornaya MV, Kutikhin AG, Zhang Y, Pei JM. The Phenomenon of Remote Preconditioning of the Heart and Its Main Manifestations. NEUROSCIENCE AND BEHAVIORAL PHYSIOLOGY 2017; 47:667-674. [DOI: 10.1007/s11055-017-0453-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
16
|
Zhou CC, Yao WT, Ge YZ, Xu LW, Wu R, Gao XF, Song KW, Jiang XM, Wang M, Huang WJ, Zhu YP, Li LP, Zhou LH, Xu ZL, Zhang SL, Zhu JG, Li WC, Jia RP. Remote ischemic conditioning for the prevention of contrast-induced acute kidney injury in patients undergoing intravascular contrast administration: a meta-analysis and trial sequential analysis of 16 randomized controlled trials. Oncotarget 2017; 8:79323-79336. [PMID: 29108311 PMCID: PMC5668044 DOI: 10.18632/oncotarget.18106] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 05/12/2017] [Indexed: 12/15/2022] Open
Abstract
Objective We conducted this meta-analysis to examine the effect of remote ischemic conditioning (RIC) on contrast-induced acute kidney injury (CI-AKI) in patients undergoing intravascular contrast administrationon. Methods Pubmed, Embase, and Cochrane Library were comprehensively searched to identify all eligible studies by 15th March, 2017. Risk ratio (RR) and weighted mean difference with the corresponding 95% confidence intervals (CI) were used to examine the treatment effect. The heterogeneity and statistical significance were assessed with Q-test and Z-test, respectively. Results A total of 16 RCTs including 2175 patients were eventually analyzed. Compared with the control group, RIC could significantly decrease the incidence of CI-AKI (RR=0.58; 95% CI: 0.46, 0.74; P < 0.001), which was further confirmed by the trial sequential analysis. Subgroup analyses showed that remote ischemic preconditioning (RIPrC) and remote ischemic postconditioning (RIPoC) were both obviously effective, and perioperative hydration might enhance the efficiency of RIC. RIC also significantly reduced the major adverse cardiovascular events within six months. Conclusion RIC, whether RIPrC or RIPoC, could effectively exert renoprotective role in intravascular contrast administration and reduce the incidence of relevant adverse events.
Collapse
Affiliation(s)
- Chang-Cheng Zhou
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Wen-Tao Yao
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yu-Zheng Ge
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Lu-Wei Xu
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Ran Wu
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Xiao-Fei Gao
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Kai-Wei Song
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Xiao-Min Jiang
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Min Wang
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Wen-Juan Huang
- Department of Nephrology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yun-Peng Zhu
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Liang-Peng Li
- Department of Cardiothoracic Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Liu-Hua Zhou
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Zhong-Le Xu
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China.,Department of Radiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Sheng-Li Zhang
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Jia-Geng Zhu
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Wen-Cheng Li
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Rui-Peng Jia
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
17
|
Brito MVH, Yasojima EY, Percário S, Ribeiro Júnior RFG, Cavalcante LCDC, Monteiro AM, Couteiro RP, Rodrigues IADS, Santos HAGD. Effects of hypertonic saline solution associated to remote ischemic perconditioning in kidney ischemia/reperfusion injury in rats. Acta Cir Bras 2017; 32:211-218. [DOI: 10.1590/s0102-865020170030000005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 02/22/2017] [Indexed: 11/22/2022] Open
|
18
|
Zheng W, Zhang Z, Liu S, Bi J, Zhang J, Du L, Ding X, Liu C. Remote ischemic conditioning protects against acetaminophen-induced acute liver injury in mice. Hepatol Res 2017; 47:234-245. [PMID: 26990366 DOI: 10.1111/hepr.12702] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2015] [Revised: 02/22/2016] [Accepted: 03/14/2016] [Indexed: 12/20/2022]
Abstract
AIM Acetaminophen (APAP) overdose is a major cause of drug-induced acute liver failure. Studies have shown that remote ischemic pre- and post-conditioning (R-IPC and R-IPOST) can protect the liver against ischemia-reperfusion (I/R) and lipopolysaccharide-induced injuries. The aim of this study was to investigate the effect of R-IPC and R-IPOST on APAP-induced hepatotoxicity in mice. METHODS Mice were randomized (n = 6 per group) to seven major groups: (i) normal control; (ii) sham operated; (iii) APAP; (iv) R-IPC + APAP; (v) R-IPC + APAP + zinc protoporphyrin (ZnPP); (vi) R-IPOST + APAP; and (vii) R-IPOST + APAP + ZnPP. Sixteen hours after APAP treatment, mouse liver and serum were collected to determine the severity of liver injury. RESULTS The results showed that R-IPC and R-IPOST significantly decreased APAP-induced serum levels of alanine aminotransferase, aspartate aminotransferase, tumor necrosis factor-α, interleukin-6, and hepatic malondialdehyde, as well as nitrotyrosine formation. Both R-IPC and R-IPOST could improve the hepatic superoxide dismutase, glutathione, and glutathione peroxidase activities and depress the expressions of pro-inflammatory associated proteins, such as inducible nitric oxide synthetase and nuclear factor-κB. They could also increase heme oxygenase-1 expression; however, ZnPP could counteract this protective effect. CONCLUSION Remote ischemic conditioning has significant therapeutic potential in APAP-induced hepatotoxicity by inhibiting oxidative stress and inflammation and promoting heme oxygenase-1 expression.
Collapse
Affiliation(s)
- Wei Zheng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University.,Department of Hepatobiliary Surgery, Shaanxi Provincial People's Hospital
| | - Zhiyong Zhang
- Department of Hepatobiliary Surgery, Shaanxi Provincial People's Hospital
| | - Sushun Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University
| | - Jianbin Bi
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University
| | - Jingyao Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University
| | - Lixue Du
- Department of Hepatobiliary Surgery, Shaanxi Provincial People's Hospital
| | - Xiaoming Ding
- Department of Renal Transplantation, Hospital of Nephrology, The First Affiliated Hospital of Xi'an Jiaotong University
| | - Chang Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University
| |
Collapse
|
19
|
Lavi S, Abu-Romeh N, Wall S, Alemayehu M, Lavi R. Long-term outcome following remote ischemic postconditioning during percutaneous coronary interventions-the RIP-PCI trial long-term follow-up. Clin Cardiol 2017; 40:268-274. [PMID: 28075499 DOI: 10.1002/clc.22668] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 12/02/2016] [Accepted: 12/03/2016] [Indexed: 12/11/2022] Open
Abstract
The clinical value of ischemic conditioning during percutaneous coronary intervention (PCI) and mode of administration is controversial. Our aim was to assess the long-term effect of remote ischemic postconditioning among patients undergoing PCI. We randomized 360 patients undergoing PCI who presented with a negative troponin T at baseline into 3 groups: 2 groups received remote ischemic postconditioning (with ischemia applied to the arm in 1 group and to the thigh in the other group), and the third group acted as a control group. Remote ischemic postconditioning was applied during PCI immediately following stent deployment, by 3, 5-minute cycles of blood pressure cuff inflation to >200 mm Hg on the arm or thigh (20 mm Hg to the arm in the control), with 5-minute breaks between each cycle. There were no differences in baseline characteristics among the 3 groups. Periprocedural myocardial injury occurred in 33% (P = 0.64). After 1 year, there was no difference between groups in death (P = 0.91), myocardial infarction (P = 0.78), or repeat revascularization (P = 0.86). During 3 years of follow-up, there was no difference in death, myocardial infarction, and revascularization among the groups (P = 0.45). Remote ischemic postconditioning during PCI did not affect long-term cardiovascular outcome. A similar effect was obtained when remote ischemia was induced to the upper or lower limb. ClinicalTrials.gov Identifier: NCT00970827.
Collapse
Affiliation(s)
- Shahar Lavi
- Western University, London, Ontario, Canada
- London Health Sciences Centre, London, Ontario, Canada
| | | | - Sabrina Wall
- London Health Sciences Centre, London, Ontario, Canada
| | | | - Ronit Lavi
- Western University, London, Ontario, Canada
- London Health Sciences Centre, London, Ontario, Canada
| |
Collapse
|
20
|
Cardioprotection by remote ischemic conditioning and its signal transduction. Pflugers Arch 2016; 469:159-181. [DOI: 10.1007/s00424-016-1922-6] [Citation(s) in RCA: 107] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Accepted: 11/28/2016] [Indexed: 12/23/2022]
|
21
|
Basalay MV, Mastitskaya S, Mrochek A, Ackland GL, Del Arroyo AG, Sanchez J, Sjoquist PO, Pernow J, Gourine AV, Gourine A. Glucagon-like peptide-1 (GLP-1) mediates cardioprotection by remote ischaemic conditioning. Cardiovasc Res 2016; 112:669-676. [PMID: 27702763 PMCID: PMC5157137 DOI: 10.1093/cvr/cvw216] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 08/21/2016] [Accepted: 09/23/2016] [Indexed: 01/22/2023] Open
Abstract
Aims Although the nature of the humoral factor which mediates cardioprotection established by remote ischaemic conditioning (RIc) remains unknown, parasympathetic (vagal) mechanisms appear to play a critical role. As the production and release of many gut hormones is modulated by the vagus nerve, here we tested the hypothesis that RIc cardioprotection is mediated by the actions of glucagon-like peptide-1 (GLP-1). Methods and results A rat model of myocardial infarction (coronary artery occlusion followed by reperfusion) was used. Remote ischaemic pre- (RIPre) or perconditioning (RIPer) was induced by 15 min occlusion of femoral arteries applied prior to or during the myocardial ischaemia. The degree of RIPre and RIPer cardioprotection was determined in conditions of cervical or subdiaphragmatic vagotomy, or following blockade of GLP-1 receptors (GLP-1R) using specific antagonist Exendin(9–39). Phosphorylation of PI3K/AKT and STAT3 was assessed. RIPre and RIPer reduced infarct size by ∼50%. In conditions of bilateral cervical or subdiaphragmatic vagotomy RIPer failed to establish cardioprotection. GLP-1R blockade abolished cardioprotection induced by either RIPre or RIPer. Exendin(9–39) also prevented RIPre-induced AKT phosphorylation. Cardioprotection induced by GLP-1R agonist Exendin-4 was preserved following cervical vagotomy, but was abolished in conditions of M3 muscarinic receptor blockade. Conclusions These data strongly suggest that GLP-1 functions as a humoral factor of remote ischaemic conditioning cardioprotection. This phenomenon requires intact vagal innervation of the visceral organs and recruitment of GLP-1R-mediated signalling. Cardioprotection induced by GLP-1R activation is mediated by a mechanism involving M3 muscarinic receptors.
Collapse
Affiliation(s)
- Marina V Basalay
- Centre for Cardiovascular and Metabolic Neuroscience, Neuroscience, Physiology & Pharmacology, University College London, Gower Street, London WC1E 6BT, UK.,Research Centre Cardiology, Luxemburg Street 110, Minsk 220026, Belarus
| | - Svetlana Mastitskaya
- Centre for Cardiovascular and Metabolic Neuroscience, Neuroscience, Physiology & Pharmacology, University College London, Gower Street, London WC1E 6BT, UK
| | | | - Gareth L Ackland
- Centre for Cardiovascular and Metabolic Neuroscience, Neuroscience, Physiology & Pharmacology, University College London, Gower Street, London WC1E 6BT, UK.,William Harvey Research Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK; and
| | - Ana Gutierrez Del Arroyo
- William Harvey Research Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK; and
| | - Jenifer Sanchez
- William Harvey Research Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK; and
| | - Per-Ove Sjoquist
- Karolinska Institute, Division of Cardiology, Karolinska University Hospital, Solna 171 76, Stockholm, Sweden
| | - John Pernow
- Karolinska Institute, Division of Cardiology, Karolinska University Hospital, Solna 171 76, Stockholm, Sweden
| | - Alexander V Gourine
- Centre for Cardiovascular and Metabolic Neuroscience, Neuroscience, Physiology & Pharmacology, University College London, Gower Street, London WC1E 6BT, UK;
| | - Andrey Gourine
- Karolinska Institute, Division of Cardiology, Karolinska University Hospital, Solna 171 76, Stockholm, Sweden
| |
Collapse
|
22
|
Johnsen J, Pryds K, Salman R, Løfgren B, Kristiansen SB, Bøtker HE. The remote ischemic preconditioning algorithm: effect of number of cycles, cycle duration and effector organ mass on efficacy of protection. Basic Res Cardiol 2016; 111:10. [DOI: 10.1007/s00395-016-0529-6] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 01/05/2016] [Indexed: 11/29/2022]
|
23
|
Chen K, Yan M, Wu P, Qing Y, Li S, Li Y, Dong Z, Xia H, Huang D, Xin P, Li J, Wei M. Combination of remote ischemic perconditioning and remote ischemic postconditioning fails to increase protection against myocardial ischemia/reperfusion injury, compared with either alone. Mol Med Rep 2015; 13:197-205. [PMID: 26572069 PMCID: PMC4686025 DOI: 10.3892/mmr.2015.4533] [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: 01/09/2015] [Accepted: 10/28/2015] [Indexed: 01/07/2023] Open
Abstract
Remote ischemic perconditioning (RIPerC) and remote ischemic postconditioning (RIPostC) have been previously demonstrated to protect the myocardium against ischemia/reperfusion (IR) injury. However, their combined effects remain to be fully elucidated. In order to investigate this, the present study used an in vivo rat model to assess whether synergistic effects are produced when RIPerC is combined with RIPostC. The rats were randomly assigned to the following groups: Sham, IR, RIPerC, RIPostC and RIPerC + RIPostC groups. The IR model was established by performing 40 min of left coronary artery occlusion, followed by 2 h of reperfusion. RIPerC and RIPostC were induced via four cycles of 5 min occlusion and 5 min reperfusion of the hindlimbs, either during or subsequent to myocardial ischemia. On measurement of infarct sizes, compared with the IR group (49.45±6.59%), the infarct sizes were significantly reduced in the RIPerC (34.36±5.87%) and RIPostC (36.04±6.16%) groups (P<0.05). However, no further reduction in infarct size was observed in the RIPerC + RIPostC group (31.43±5.43%; P>0.05), compared with the groups treated with either RIPerC or RIPostC alone. Activation of the reperfusion injury salvage kinase (RISK) Akt, extracellular signal-regulated kinase 1/2 and glycogen synthase kinase-3β, and survivor activating factor enhancement (SAFE) signal transducer and activator of transcription-3 pathways were enhanced in the RIPerC, RIPostC and the RIPerC + RIPostC groups, compared with the IR group, with no difference among the three groups. Therefore, whereas RIPerC and RIPostC were equally effective in providing protection against myocardial IR injury, the combination of RIPerC and RIPostC failed to provide further protection than treatment with either alone. The cardioprotective effects were found to be associated with increased activation of the RISK and SAFE pathways.
Collapse
Affiliation(s)
- Kankai Chen
- Department of Cardiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Meiling Yan
- Department of Cardiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Penglong Wu
- Department of Cardiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Yanwei Qing
- Department of Cardiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Shuai Li
- Department of Cardiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Yongguang Li
- Department of Cardiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Zhifeng Dong
- Department of Cardiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Hongjuan Xia
- Department of Cardiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Dong Huang
- Department of Cardiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Ping Xin
- Department of Cardiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Jingbo Li
- Department of Cardiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Meng Wei
- Department of Cardiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| |
Collapse
|
24
|
Eitel I, Stiermaier T, Rommel KP, Fuernau G, Sandri M, Mangner N, Linke A, Erbs S, Lurz P, Boudriot E, Mende M, Desch S, Schuler G, Thiele H. Cardioprotection by combined intrahospital remote ischaemic perconditioning and postconditioning in ST-elevation myocardial infarction: the randomized LIPSIA CONDITIONING trial. Eur Heart J 2015; 36:3049-57. [PMID: 26385956 DOI: 10.1093/eurheartj/ehv463] [Citation(s) in RCA: 181] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Accepted: 08/19/2015] [Indexed: 11/13/2022] Open
Abstract
AIMS Remote ischaemic conditioning (RIC) and postconditioning (PostC) are both potent activators of innate protection against ischaemia-reperfusion injury and have demonstrated cardioprotection in experimental and clinical ST-elevation myocardial infarction (STEMI) trials. However, their combined effects have not been studied in detail. The aim of this study was to evaluate if the co-application of intrahospital RIC and PostC has a more powerful effect on myocardial salvage compared with either PostC alone or control. METHODS AND RESULTS This prospective, controlled, single-centre study randomized 696 STEMI patients to one of the following three groups: (i) combined intrahospital RIC + PostC in addition to primary percutaneous coronary intervention (PCI); (ii) PostC in addition to PCI; and (iii) conventional PCI (control). The primary endpoint myocardial salvage index was assessed by cardiac magnetic resonance (CMR) imaging within 3 days after infarction. Secondary endpoints included infarct size and microvascular obstruction (MVO) assessed by CMR. The combined clinical endpoint consisted of death, reinfarction, and new congestive heart failure within 6 months. The primary endpoint myocardial salvage index was significantly greater in the combined RIC + PostC group when compared with the control group (49 [interquartile range 30-72] vs. 40 [interquartile range 16-68], P = 0.02). Postconditioning alone failed to improve myocardial salvage when compared with conventional PCI (P = 0.39). The secondary endpoints, including infarct size and MVO, showed no significant differences between groups. Clinical follow-up at 6 months revealed no differences in the combined clinical endpoint between groups (P = 0.44). CONCLUSION Combined intrahospital RIC + PostC in conjunction with PCI in STEMI significantly improves myocardial salvage in comparison with control and PostC. CLINICALTRIALSGOV NCT02158468.
Collapse
Affiliation(s)
- Ingo Eitel
- University Heart Center Lübeck, Medical Clinic II (Cardiology/Angiology/Intensive Care Medicine), University Hospital Schleswig-Holstein, Ratzeburger Allee 160, 23538 Lübeck, Germany German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Lübeck, Germany
| | - Thomas Stiermaier
- Department of Internal Medicine/Cardiology, University of Leipzig - Heart Center, Leipzig, Germany
| | - Karl P Rommel
- Department of Internal Medicine/Cardiology, University of Leipzig - Heart Center, Leipzig, Germany
| | - Georg Fuernau
- University Heart Center Lübeck, Medical Clinic II (Cardiology/Angiology/Intensive Care Medicine), University Hospital Schleswig-Holstein, Ratzeburger Allee 160, 23538 Lübeck, Germany German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Lübeck, Germany
| | - Marcus Sandri
- Department of Internal Medicine/Cardiology, University of Leipzig - Heart Center, Leipzig, Germany
| | - Norman Mangner
- Department of Internal Medicine/Cardiology, University of Leipzig - Heart Center, Leipzig, Germany
| | - Axel Linke
- Department of Internal Medicine/Cardiology, University of Leipzig - Heart Center, Leipzig, Germany
| | - Sandra Erbs
- Department of Internal Medicine/Cardiology, University of Leipzig - Heart Center, Leipzig, Germany
| | - Phillip Lurz
- Department of Internal Medicine/Cardiology, University of Leipzig - Heart Center, Leipzig, Germany
| | - Enno Boudriot
- Department of Internal Medicine/Cardiology, University of Leipzig - Heart Center, Leipzig, Germany
| | - Meinhard Mende
- Clinical Trial Center Leipzig, University of Leipzig, Leipzig, Germany
| | - Steffen Desch
- University Heart Center Lübeck, Medical Clinic II (Cardiology/Angiology/Intensive Care Medicine), University Hospital Schleswig-Holstein, Ratzeburger Allee 160, 23538 Lübeck, Germany German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Lübeck, Germany
| | - Gerhard Schuler
- Department of Internal Medicine/Cardiology, University of Leipzig - Heart Center, Leipzig, Germany
| | - Holger Thiele
- University Heart Center Lübeck, Medical Clinic II (Cardiology/Angiology/Intensive Care Medicine), University Hospital Schleswig-Holstein, Ratzeburger Allee 160, 23538 Lübeck, Germany German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Lübeck, Germany
| |
Collapse
|
25
|
McCafferty K, Forbes S, Thiemermann C, Yaqoob MM. The challenge of translating ischemic conditioning from animal models to humans: the role of comorbidities. Dis Model Mech 2015; 7:1321-33. [PMID: 25481012 PMCID: PMC4257001 DOI: 10.1242/dmm.016741] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Following a period of ischemia (local restriction of blood supply to a tissue), the restoration of blood supply to the affected area causes significant tissue damage. This is known as ischemia-reperfusion injury (IRI) and is a central pathological mechanism contributing to many common disease states. The medical complications caused by IRI in individuals with cerebrovascular or heart disease are a leading cause of death in developed countries. IRI is also of crucial importance in fields as diverse as solid organ transplantation, acute kidney injury and following major surgery, where post-operative organ dysfunction is a major cause of morbidity and mortality. Given its clinical impact, novel interventions are urgently needed to minimize the effects of IRI, not least to save lives but also to reduce healthcare costs. In this Review, we examine the experimental technique of ischemic conditioning, which entails exposing organs or tissues to brief sub-lethal episodes of ischemia and reperfusion, before, during or after a lethal ischemic insult. This approach has been found to confer profound tissue protection against IRI. We discuss the translation of ischemic conditioning strategies from bench to bedside, and highlight where transition into human clinical studies has been less successful than in animal models, reviewing potential reasons for this. We explore the challenges that preclude more extensive clinical translation of these strategies and emphasize the role that underlying comorbidities have in altering the efficacy of these strategies in improving patient outcomes.
Collapse
Affiliation(s)
- Kieran McCafferty
- Translational Medicine and Therapeutics, William Harvey Research Institute, Queen Mary University London, London, EC1M 6BQ, UK.
| | - Suzanne Forbes
- Translational Medicine and Therapeutics, William Harvey Research Institute, Queen Mary University London, London, EC1M 6BQ, UK
| | - Christoph Thiemermann
- Translational Medicine and Therapeutics, William Harvey Research Institute, Queen Mary University London, London, EC1M 6BQ, UK
| | - Muhammad M Yaqoob
- Translational Medicine and Therapeutics, William Harvey Research Institute, Queen Mary University London, London, EC1M 6BQ, UK
| |
Collapse
|
26
|
Liu Z, Gong R. Remote ischemic preconditioning for kidney protection: GSK3β-centric insights into the mechanism of action. Am J Kidney Dis 2015; 66:846-56. [PMID: 26271146 DOI: 10.1053/j.ajkd.2015.06.026] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 06/22/2015] [Indexed: 12/13/2022]
Abstract
Preventing acute kidney injury (AKI) in high-risk patients following medical interventions is a paramount challenge for clinical practice. Recent data from animal experiments and clinical trials indicate that remote ischemic preconditioning, represented by limb ischemic preconditioning, confers a protective action on the kidney. Ischemic preconditioning is effective in reducing the risk for AKI following cardiovascular interventions and the use of iodinated radiocontrast media. Nevertheless, the underlying mechanisms for this protective effect are elusive. A protective signal is conveyed from the remote site undergoing ischemic preconditioning, such as the limb, to target organs, such as the kidney, by multiple potential communication pathways, which may involve humoral, neuronal, and systemic mechanisms. Diverse transmitting pathways trigger a variety of signaling cascades, including the reperfusion injury salvage kinase and survivor activating factor enhancement pathways, all of which converge on glycogen synthase kinase 3β (GSK3β). Inhibition of GSK3β subsequent to ischemic preconditioning reinforces the Nrf2-mediated antioxidant defense, diminishes the nuclear factor-κB-dependent proinflammatory response, and exerts prosurvival effects ensuing from the desensitized mitochondria permeability transition. Thus, therapeutic targeting of GSK3β by ischemic preconditioning or by pharmacologic preconditioning with existing US Food and Drug Administration-approved drugs having GSK3β-inhibitory activities might represent a pragmatic and cost-effective adjuvant strategy for kidney protection and prophylaxis against AKI.
Collapse
Affiliation(s)
- Zhangsuo Liu
- Department of Nephrology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Rujun Gong
- Division of Kidney Disease and Hypertension, Department of Medicine, Rhode Island Hospital, Brown University School of Medicine, Providence, RI.
| |
Collapse
|
27
|
Crisafulli A, Mancardi D, Marongiu E, Rastaldo R, Penna C, Pagliaro P. Preconditioning cardioprotection and exercise performance: a radical point of view. SPORT SCIENCES FOR HEALTH 2015. [DOI: 10.1007/s11332-015-0225-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
28
|
Alburquerque-Béjar JJ, Barba I, Inserte J, Miró-Casas E, Ruiz-Meana M, Poncelas M, Vilardosa Ú, Valls-Lacalle L, Rodríguez-Sinovas A, Garcia-Dorado D. Combination therapy with remote ischaemic conditioning and insulin or exenatide enhances infarct size limitation in pigs. Cardiovasc Res 2015; 107:246-54. [PMID: 26045476 DOI: 10.1093/cvr/cvv171] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Accepted: 05/23/2015] [Indexed: 12/18/2022] Open
Abstract
AIMS Remote ischaemic conditioning (RIC) has been shown to reduce myocardial infarct size in patients. Our objective was to investigate whether the combination of RIC with either exenatide or glucose-insulin-potassium (GIK) is more effective than RIC alone. METHODS AND RESULTS Pigs were submitted to 40 min of coronary occlusion followed by reperfusion, and received (i) no treatment, (ii) one of the following treatments: RIC (5 min ischemia/5 min reperfusion × 4), GIK, or exenatide (at doses reducing infarct size in clinical trials), or (iii) a combination of two of these treatments (RIC + GIK or RIC + exenatide). After 5 min of reperfusion (n = 4/group), prominent phosphorylation of Akt and endothelial nitric oxide synthase (eNOS) was observed, both in control and reperfused myocardium, in animals receiving GIK, and mitochondria from these hearts showed reduced ADP-stimulated respiration. (1)H NMR-based metabonomics disclosed a shift towards increased glycolysis in GIK and exenatide groups. In contrast, oxidative stress (myocardial nitrotyrosine levels) and eNOS uncoupling were significantly reduced only by RIC. In additional experiments (n = 7-10/group), ANOVA demonstrated a significant effect of the number of treatments after 2 h of reperfusion on infarct size (triphenyltetrazolium, % of the area at risk; 59.21 ± 3.34, 36.64 ± 3.03, and 21.04 ± 2.38% for none, one, and two treatments, respectively), and significant differences between one and two treatments (P = 0.004) but not among individual treatments or between RIC + GIK and RIC + exenatide. CONCLUSIONS GIK and exenatide activate cardioprotective pathways different from those of RIC, and have additive effects with RIC on infarct size reduction in pigs.
Collapse
Affiliation(s)
- Juan José Alburquerque-Béjar
- Laboratory of Experimental Cardiology, Department of Cardiology, Vall d'Hebron University Hospital and Research Institute, Universitat Autònoma de Barcelona, Pg. Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Ignasi Barba
- Laboratory of Experimental Cardiology, Department of Cardiology, Vall d'Hebron University Hospital and Research Institute, Universitat Autònoma de Barcelona, Pg. Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Javier Inserte
- Laboratory of Experimental Cardiology, Department of Cardiology, Vall d'Hebron University Hospital and Research Institute, Universitat Autònoma de Barcelona, Pg. Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Elisabet Miró-Casas
- Laboratory of Experimental Cardiology, Department of Cardiology, Vall d'Hebron University Hospital and Research Institute, Universitat Autònoma de Barcelona, Pg. Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Marisol Ruiz-Meana
- Laboratory of Experimental Cardiology, Department of Cardiology, Vall d'Hebron University Hospital and Research Institute, Universitat Autònoma de Barcelona, Pg. Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Marcos Poncelas
- Laboratory of Experimental Cardiology, Department of Cardiology, Vall d'Hebron University Hospital and Research Institute, Universitat Autònoma de Barcelona, Pg. Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Úrsula Vilardosa
- Laboratory of Experimental Cardiology, Department of Cardiology, Vall d'Hebron University Hospital and Research Institute, Universitat Autònoma de Barcelona, Pg. Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Laura Valls-Lacalle
- Laboratory of Experimental Cardiology, Department of Cardiology, Vall d'Hebron University Hospital and Research Institute, Universitat Autònoma de Barcelona, Pg. Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Antonio Rodríguez-Sinovas
- Laboratory of Experimental Cardiology, Department of Cardiology, Vall d'Hebron University Hospital and Research Institute, Universitat Autònoma de Barcelona, Pg. Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - David Garcia-Dorado
- Laboratory of Experimental Cardiology, Department of Cardiology, Vall d'Hebron University Hospital and Research Institute, Universitat Autònoma de Barcelona, Pg. Vall d'Hebron 119-129, 08035 Barcelona, Spain
| |
Collapse
|
29
|
Liang Y, Li YP, He F, Liu XQ, Zhang JY. Long-term, regular remote ischemic preconditioning improves endothelial function in patients with coronary heart disease. ACTA ACUST UNITED AC 2015; 48:568-76. [PMID: 25923462 PMCID: PMC4470317 DOI: 10.1590/1414-431x20144452] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Accepted: 12/19/2014] [Indexed: 02/14/2023]
Abstract
Remote ischemic preconditioning (RIPre) can prevent myocardial injury. The purpose of
this study was to assess the beneficial effects of long-term regular RIPre on human
arteries. Forty patients scheduled for coronary artery bypass graft (CABG) surgery
were assigned randomly to a RIPre group (n=20) or coronary heart disease (CHD) group
(n=20). Twenty patients scheduled for mastectomy were enrolled as a control group.
RIPre was achieved by occluding arterial blood flow 5 min with a mercury
sphygmomanometer followed by a 5-min reperfusion period, and this was repeated 4
times. The RIPre procedure was repeated 3 times a day for 20 days. In all patients,
arterial fragments discarded during surgery were collected to evaluate endothelial
function by flow-mediated dilation (FMD), CD34+ monocyte count, and
endothelial nitric oxide synthase (eNOS expression). Phosphorylation levels of STAT-3
and Akt were also assayed to explore the underlying mechanisms. Compared with the CHD
group, long-term regular RIPre significantly improved FMD after 20 days (8.5±2.4
vs 4.9±4.2%, P<0.05) and significantly reduced troponin after
CABG surgery (0.72±0.31 and 1.64±0.19, P<0.05). RIPre activated STAT-3 and
increased CD34+ endothelial progenitor cell counts found in arteries.
Long-term, regular RIPre improved endothelial function in patients with CHD, possibly
due to STAT-3 activation, and this may have led to an increase in endothelial
progenitor cells.
Collapse
Affiliation(s)
- Y Liang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Y P Li
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - F He
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - X Q Liu
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - J Y Zhang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
30
|
Abstract
Reperfusion is mandatory to salvage ischemic myocardium from infarction, but reperfusion per se contributes to injury and ultimate infarct size. Therefore, cardioprotection beyond that by timely reperfusion is needed to reduce infarct size and improve the prognosis of patients with acute myocardial infarction. The conditioning phenomena provide such cardioprotection, insofar as brief episodes of coronary occlusion/reperfusion preceding (ischemic preconditioning) or following (ischemic postconditioning) sustained myocardial ischemia with reperfusion reduce infarct size. Even ischemia/reperfusion in organs remote from the heart provides cardioprotection (remote ischemic conditioning). The present review characterizes the signal transduction underlying the conditioning phenomena, including their physical and chemical triggers, intracellular signal transduction, and effector mechanisms, notably in the mitochondria. Cardioprotective signal transduction appears as a highly concerted spatiotemporal program. Although the translation of ischemic postconditioning and remote ischemic conditioning protocols to patients with acute myocardial infarction has been fairly successful, the pharmacological recruitment of cardioprotective signaling has been largely disappointing to date.
Collapse
Affiliation(s)
- Gerd Heusch
- From the Institute for Pathophysiology, West German Heart and Vascular Centre, University of Essen Medical School, Essen, Germany.
| |
Collapse
|
31
|
Xu YC, Li RP, Xue FS, Cui XL, Wang SY, Liu GP, Yang GZ, Sun C, Liao X. κ-Opioid receptors are involved in enhanced cardioprotection by combined fentanyl and limb remote ischemic postconditioning. J Anesth 2015; 29:535-43. [DOI: 10.1007/s00540-015-1998-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Accepted: 03/04/2015] [Indexed: 11/29/2022]
|
32
|
Inserte J, Garcia-Dorado D. The cGMP/PKG pathway as a common mediator of cardioprotection: translatability and mechanism. Br J Pharmacol 2015; 172:1996-2009. [PMID: 25297462 DOI: 10.1111/bph.12959] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Revised: 09/16/2014] [Accepted: 09/26/2014] [Indexed: 12/24/2022] Open
Abstract
Cardiomyocyte cell death occurring during myocardial reperfusion (reperfusion injury) contributes to final infarct size after transient coronary occlusion. Different interrelated mechanisms of reperfusion injury have been identified, including alterations in cytosolic Ca(2+) handling, sarcoplasmic reticulum-mediated Ca(2+) oscillations and hypercontracture, proteolysis secondary to calpain activation and mitochondrial permeability transition. All these mechanisms occur during the initial minutes of reperfusion and are inhibited by intracellular acidosis. The cGMP/PKG pathway modulates the rate of recovery of intracellular pH, but has also direct effect on Ca(2+) oscillations and mitochondrial permeability transition. The cGMP/PKG pathway is depressed in cardiomyocytes by ischaemia/reperfusion and preserved by ischaemic postconditioning, which importantly contributes to postconditioning protection. The present article reviews the mechanisms and consequences of the effect of ischaemic postconditioning on the cGMP/PKG pathway, the different pharmacological strategies aimed to stimulate it during myocardial reperfusion and the evidence, limitations and promise of translation of these strategies to the clinical practice. Overall, the preclinical and clinical evidence suggests that modulation of the cGMP/PKG pathway may be a therapeutic target in the context of myocardial infarction.
Collapse
Affiliation(s)
- Javier Inserte
- Cardiology Department, Vall d'Hebron University Hospital and Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | | |
Collapse
|
33
|
Transient carotid ischemia as a remote conditioning stimulus for myocardial protection in anesthetized rabbits: Insights into intracellular signaling. Int J Cardiol 2015; 184:140-151. [PMID: 25703422 DOI: 10.1016/j.ijcard.2015.01.079] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 12/09/2014] [Accepted: 01/25/2015] [Indexed: 12/16/2022]
Abstract
BACKGROUND We investigated the effectiveness of perconditioning (Perc) applied at different time points along with the role of RISK, SAFE, STAT5 and eNOS pathways. METHODS AND RESULTS Anesthetized rabbits were subjected to 30-min ischemia/3-hour reperfusion. Perc, consisted of 4 cycles of 1-min ischemia/reperfusion, was applied in the carotid artery at different time points. Perc was started and ended during ischemia, started during ischemia and ended at the beginning of reperfusion, started at the end of ischemia and ended at reperfusion and started and ended during reperfusion. The PI3K inhibitor wortmannin, or the JAK-2 inhibitor AG490, was also applied and the infarct size was assessed. In another series assigned to the previous groups, the phosphorylation of Akt, PI3K, ERKs1/2, GSK3β, STAT3, and STAT5 was evaluated. All Perc groups had smaller infarction compared to those without Perc, independently of PI3K or JAK-2 inhibition. STAT5 was the only molecule that was phosphorylated in parallel with cardioprotection. Since Src and angiotensin II mediate the STAT5 pathway, we administered the Scr inhibitor PP1 and the angiotensin II receptor antagonist valsartan. PP1 and valsartan prevented STAT5 phosphorylation, but did not abrogate the effect of Perc. Furthermore, the NOS inhibitor L-NAME was administered and abrogated the infarct size limiting effect of Perc. In parallel, the expression of cleaved caspase-3 was elevated only in the control and Perc-A-L-NAME groups. CONCLUSION Perc reduces infarction independently of RISK, SAFE and STAT5 pathways. Src kinase and angiotensin II play a predominant role in STAT5 activation. eNOS may protect the myocardium through inhibition of apoptosis.
Collapse
|
34
|
Zhao B, Gao W, Hou J, Wu Y, Xia Z. Ischemic postconditioning enhances glycogen synthase kinase-3β expression and alleviates cerebral ischemia/reperfusion injury. Neural Regen Res 2015; 7:1507-12. [PMID: 25657687 PMCID: PMC4308783 DOI: 10.3969/j.issn.1673-5374.2012.19.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2011] [Accepted: 05/07/2012] [Indexed: 11/18/2022] Open
Abstract
The present study established global brain ischemia using the four-vessel occlusion method. Following three rounds of reperfusion for 30 seconds, and occlusion for 10 seconds, followed by reperfusion for 48 hours, infarct area, the number of TUNEL-positive cells and Bcl-2 expression were significantly reduced. However, glycogen synthase kinase-3β activity, cortical Bax and caspase-3 expression significantly increased, similar to results following ischemic postconditioning. Our results indicated that ischemic postconditioning may enhance glycogen synthase kinase-3β activity, a downstream molecule of the phosphatase and tensin homolog deleted on chromosome 10/phosphatidylinositol 3-kinase/protein kinase B signaling pathway, which reduces caspase-3 expression to protect the brain against ischemic injury.
Collapse
Affiliation(s)
- Bo Zhao
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China
| | - Wenwei Gao
- Department of Intensive Care Unit, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China
| | - Jiabao Hou
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China
| | - Yang Wu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China
| | - Zhongyuan Xia
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China
| |
Collapse
|
35
|
Heusch G, Bøtker HE, Przyklenk K, Redington A, Yellon D. Remote ischemic conditioning. J Am Coll Cardiol 2015; 65:177-95. [PMID: 25593060 PMCID: PMC4297315 DOI: 10.1016/j.jacc.2014.10.031] [Citation(s) in RCA: 497] [Impact Index Per Article: 49.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Revised: 10/16/2014] [Accepted: 10/22/2014] [Indexed: 12/12/2022]
Abstract
In remote ischemic conditioning (RIC), brief, reversible episodes of ischemia with reperfusion in one vascular bed, tissue, or organ confer a global protective phenotype and render remote tissues and organs resistant to ischemia/reperfusion injury. The peripheral stimulus can be chemical, mechanical, or electrical and involves activation of peripheral sensory nerves. The signal transfer to the heart or other organs is through neuronal and humoral communications. Protection can be transferred, even across species, with plasma-derived dialysate and involves nitric oxide, stromal derived factor-1α, microribonucleic acid-144, but also other, not yet identified factors. Intracardiac signal transduction involves: adenosine, bradykinin, cytokines, and chemokines, which activate specific receptors; intracellular kinases; and mitochondrial function. RIC by repeated brief inflation/deflation of a blood pressure cuff protects against endothelial dysfunction and myocardial injury in percutaneous coronary interventions, coronary artery bypass grafting, and reperfused acute myocardial infarction. RIC is safe and effective, noninvasive, easily feasible, and inexpensive.
Collapse
Affiliation(s)
- Gerd Heusch
- Institute for Pathophysiology, West German Heart and Vascular Centre Essen, University of Essen Medical School, Essen, Germany.
| | - Hans Erik Bøtker
- Department of Cardiology, Aarhus University Hospital Skejby, Aarhus, Denmark
| | - Karin Przyklenk
- Cardiovascular Research Institute, Wayne State University School of Medicine, Detroit, Michigan
| | - Andrew Redington
- Hospital for Sick Children and University of Toronto, Toronto, Ontario, Canada
| | - Derek Yellon
- The Hatter Cardiovascular Institute, University College London, London, United Kingdom
| |
Collapse
|
36
|
Remote ischemic preconditioning reduces perioperative cardiac and renal events in patients undergoing elective coronary intervention: a meta-analysis of 11 randomized trials. PLoS One 2014; 9:e115500. [PMID: 25551671 PMCID: PMC4281209 DOI: 10.1371/journal.pone.0115500] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Accepted: 10/27/2014] [Indexed: 12/17/2022] Open
Abstract
Background Results from randomized controlled trials (RCT) concerning cardiac and renal effect of remote ischemic preconditioning(RIPC) in patients with stable coronary artery disease(CAD) are inconsistent. The aim of this study was to explore whether RIPC reduce cardiac and renal events after elective percutaneous coronary intervention (PCI). Methods and Results RCTs with data on cardiac or renal effect of RIPC in PCI were searched from Pubmed, EMBase, and Cochrane library (up to July 2014). Meta-regression and subgroup analysis were performed to identify the potential sources of significant heterogeneity(I2≥40%). Eleven RCTs enrolling a total of 1713 study subjects with stable CAD were selected. Compared with controls, RIPC significantly reduced perioperative incidence of myocardial infarction (MI) [odds ratio(OR) = 0.68; 95% CI, 0.51 to 0.91; P = 0.01; I2 = 41.0%] and contrast-induced acute kidney injury(AKI) (OR = 0.61; 95% CI, 0.38 to 0.98; P = 0.04; I2 = 39.0%). Meta-regression and subgroup analyses confirmed that the major source of heterogeneity for the incidence of MI was male proportion (coefficient = −0.049; P = 0.047; adjusted R2 = 0.988; P = 0.02 for subgroup difference). Conclusions The present meta-analysis of RCTs suggests that RIPC may offer cardiorenal protection by reducing the incidence of MI and AKI in patients undergoing elective PCI. Moreover, this effect on MI is more pronounced in male subjects. Future high-quality, large-scale clinical trials should focus on the long-term clinical effect of RIPC.
Collapse
|
37
|
Huang D, Wang FB, Guo M, Li S, Yan ML, Yu T, Wei M, Li JB. Effect of combined treatment with rosuvastatin and protein kinase Cβ2 inhibitor on angiogenesis following myocardial infarction in diabetic rats. Int J Mol Med 2014; 35:829-38. [PMID: 25524396 DOI: 10.3892/ijmm.2014.2043] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2014] [Accepted: 12/12/2014] [Indexed: 11/06/2022] Open
Abstract
The aim of the present study was to investigate the effects of combined treatment with rosuvastatin and LY333531, a selective protein kinase C (PKC)β2 inhibitor, on angiogenesis under hyperglycemic conditions. Human umbilical vein endothelial cells (HUVECs) cultured in medium containing a normal or high concentration of glucose (33.3 mmol/l) were treated with rosuvastatin (0.1 µmol/l) alone or in combination with LY333531 (10 nmol/l). HUVEC migration and tube formation were assessed. Furthermore, rats with streptozotocin-induced diabetes were randomly divided into groups and treated with either rosuvastatin alone (5 mg/kg/day) or in combination with LY333531 (10 mg/kg/day) for 4 weeks following the induction of myocardial infarction (MI). Echocardiographic patterns, the extent of myocardial fibrosis, capillary density in myocardial tissue, the phosphorylation of Akt and endothelial nitric oxide synthase (eNOS), as well as the expression levels of vascular endothelial growth factor (VEGF) and hypoxia-inducible factor 1-α (HIF‑1α) were assessed. The results from the in vitro experiment revealed that the tube-forming and migration ability of the HUVECs exposed to high-glucose medium was significantly improved in the group treated with the combination of rosuvastatin and LY333531. In vivo, the combination of rosuvastatin and LY333531 significantly improved left ventricular function, reduced the extent of myocardial fibrosis and increased myocardial capillary density compared to treatment with rosuvastatin alone. In addition, the expression levels of VEGF, and Akt and eNOS phosphorylation were significantly higher in the group exposed to the combination treatment than in the group treated with rosuvastatin alone. The results of the present study indicate that, compared to treatment with rosuvastatin alone, combined treatment with rosuvastatin and LY333531 promotes a greater level of angiogenesis in diabetic rats with MI. This effect is likely mediated through the upregulation of the VEGF‑dependent Akt/eNOS signaling pathway.
Collapse
Affiliation(s)
- Dong Huang
- Division of Cardiology, The Sixth People's Hospital Affiliated to Shanghai Jiaotong University, Shanghai Jiaotong University School of Medicine, State Key Discipline Division, Shanghai 200233, P.R. China
| | - Fa-Bin Wang
- Division of Cardiology, Tengzhou Central People's Hospital, Tengzhou, Shandong 277500, P.R. China
| | - Ming Guo
- Division of Cardiology, Tengzhou Central People's Hospital, Tengzhou, Shandong 277500, P.R. China
| | - Shuai Li
- Division of Cardiology, The Sixth People's Hospital Affiliated to Shanghai Jiaotong University, Shanghai Jiaotong University School of Medicine, State Key Discipline Division, Shanghai 200233, P.R. China
| | - Mei-Ling Yan
- Division of Cardiology, The Sixth People's Hospital Affiliated to Shanghai Jiaotong University, Shanghai Jiaotong University School of Medicine, State Key Discipline Division, Shanghai 200233, P.R. China
| | - Tao Yu
- Division of Cardiology, The Sixth People's Hospital Affiliated to Shanghai Jiaotong University, Shanghai Jiaotong University School of Medicine, State Key Discipline Division, Shanghai 200233, P.R. China
| | - Meng Wei
- Division of Cardiology, The Sixth People's Hospital Affiliated to Shanghai Jiaotong University, Shanghai Jiaotong University School of Medicine, State Key Discipline Division, Shanghai 200233, P.R. China
| | - Jing-Bo Li
- Division of Cardiology, The Sixth People's Hospital Affiliated to Shanghai Jiaotong University, Shanghai Jiaotong University School of Medicine, State Key Discipline Division, Shanghai 200233, P.R. China
| |
Collapse
|
38
|
Barsukevich V, Basalay M, Sanchez J, Mrochek A, Whittle J, Ackland GL, Gourine AV, Gourine A. Distinct cardioprotective mechanisms of immediate, early and delayed ischaemic postconditioning. Basic Res Cardiol 2014; 110:452. [PMID: 25449894 PMCID: PMC4250560 DOI: 10.1007/s00395-014-0452-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Revised: 10/10/2014] [Accepted: 10/24/2014] [Indexed: 12/18/2022]
Abstract
Cardioprotection against ischaemia/reperfusion injury in mice can be achieved by delayed ischaemic postconditioning (IPost) applied as late as 30 min after the onset of reperfusion. We determined the efficacy of delayed IPost in a rat model of myocardial infarction (MI) and investigated potential underlying mechanisms of this phenomenon. Rats were subjected to 20, 30 or 45 min of coronary artery occlusion followed by 120 min of reperfusion (I/R). Immediate and early IPost included six cycles of I/R (10/10 s) applied 10 s or 10 min after reperfusion onset. In the second series of experiments, the rats were subjected to 30 min of coronary occlusion followed by IPost applied 10 s, 10, 30, 45 or 60 min after the onset of reperfusion. Immediate and early IPost (applied 10 s or 10 min of reperfusion) established cardioprotection only when applied after a period of myocardial ischaemia lasting 30 min. Delayed IPost applied after 30 or 45 min of reperfusion reduced infarct sizes by 36 and 41 %, respectively (both P < 0.01). IPost applied 60 min after reperfusion onset was ineffective. Inhibition of RISK pathway (administration of ERK1/2 inhibitor PD-98059 or PI3K inhibitor LY-294002) abolished cardioprotection established by immediate IPost but had no effect on cardioprotection conferred by early IPost. Blockade of SAFE pathway using JAK/STAT inhibitor AG490 had no effect on the immediate or early IPost cardioprotection. Blockade of mitochondrial KATP (mitoKATP) channels (with 5-Hydroxydecanoate) abolished cardioprotection achieved by immediate and early IPost, but had no effect on cardioprotection when IPost was applied 30 or 45 min into the reperfusion period. Immediate IPost increased phosphorylation of PI3K-AKT and ERK1/2. Early or delayed IPost had no effect on phosphorylation of PI3K-AKT, ERK1/2 or STAT3. These data show that in the rat model, delayed IPost confers significant cardioprotection even if applied 45 min after onset of reperfusion. Cardioprotection induced by immediate and early postconditioning involves recruitment of RISK pathway and/or mitoKATP channels, while delayed postconditioning appears to rely on a different mechanism.
Collapse
|
39
|
Dong Z, Gong K, Huang D, Zhu W, Sun W, Zhang Y, Xin P, Shen Y, Wu P, Li J, Lu Z, Zhang X, Wei M. Myocardial infarction accelerates glomerular injury and microalbuminuria in diabetic rats via local hemodynamics and immunity. Int J Cardiol 2014; 179:397-408. [PMID: 25464495 DOI: 10.1016/j.ijcard.2014.11.033] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2014] [Revised: 11/02/2014] [Accepted: 11/04/2014] [Indexed: 02/08/2023]
Abstract
BACKGROUND Clinically, approximately one-third of patients with chronic heart failure (CHF) exhibit some degree of renal dysfunction. This renal dysfunction is referred to as cardiorenal syndrome (CRS) and plays an important role in the poor prognosis of CHF. Mounting evidence suggests that diabetes is the most common underlying risk factor for CRS. However, the underlying pathophysiological mechanisms are poorly understood. METHODS We performed the following comparisons in two separate protocols: 1) surgically induced myocardial infarction rats (MI, n=10), sham operation rats (Ctr, n=10) and MI rats treated with Fasudil, a Rho-kinase inhibitor (MI+Fas, n=9); and 2) STZ-induced type 1 diabetic rats (DB, n=10), DB+MI rats (n=10) and DB+MI rats treated with Fasudil (DB+MI+Fas, n=9). Renal hemodynamics and vasoconstrictor reactivity were evaluated using the DMT myograph system. Renal immunity was evaluated by flow cytometry, electron microscopy, immunofluorescence, etc. RESULTS Twelve weeks after the operation, compared with DB or MI rats, DB+MI rats exhibited the following characteristics: 1) significantly increased glomerular enlargement, fibrosis, glomerulosclerosis, podocyte injury and microalbuminuria; 2) significantly increased vasoconstrictor reactivity of the renal interlobular arteries and renal venous pressure; 3) significantly increased infiltration of CD₃+ and CD₄+ T cells and decreased Treg/Th17 ratios; and 4) significantly increased glomerular deposition of IgG and C₄. In contrast, rats with MI only showed mildly accelerated glomerular remodeling and microalbuminuria, with little change in renal hemodynamics and immunity. Fasudil treatment significantly improved the renal lesions in DB+MI rats but not MI rats. CONCLUSIONS Post-MI cardiac dysfunction significantly accelerated glomerular remodeling, podocyte injury and microalbuminuria in STZ-induced diabetic rats. These changes were accompanied by altered local hemodynamics and immunity.
Collapse
Affiliation(s)
- Zhifeng Dong
- Department of Cardiology, Shanghai Sixth Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200233, China
| | - Kaizheng Gong
- Department of Cardiology, The Second Clinical Medical College, Yangzhou University, Yangzhou 225001, China
| | - Dong Huang
- Department of Cardiology, Shanghai Sixth Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200233, China
| | - Wei Zhu
- Department of Cardiology, Shanghai Sixth Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200233, China
| | - Wanfeng Sun
- Affiliated Yancheng Hospital of Medical School, Southeast University, Yancheng 224001, China
| | - Ying Zhang
- Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Ping Xin
- Department of Cardiology, Shanghai Sixth Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200233, China
| | - Yuan Shen
- Affiliated Yancheng Hospital of Medical School, Southeast University, Yancheng 224001, China
| | - Penglong Wu
- Department of Cardiology, Shanghai Sixth Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200233, China
| | - Jingbo Li
- Department of Cardiology, Shanghai Sixth Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200233, China
| | - Zhigang Lu
- Department of Cardiology, Shanghai Sixth Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200233, China
| | - Xiaoming Zhang
- Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China.
| | - Meng Wei
- Department of Cardiology, Shanghai Sixth Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200233, China.
| |
Collapse
|
40
|
YU YING, JIA XIANJIE, ZONG QIAOFENG, ZHANG GUANJUN, YE HONGWEI, HU JIE, GAO QIN, GUAN SUDONG. Remote ischemic postconditioning protects the heart by upregulating ALDH2 expression levels through the PI3K/Akt signaling pathway. Mol Med Rep 2014; 10:536-42. [DOI: 10.3892/mmr.2014.2156] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2013] [Accepted: 03/18/2014] [Indexed: 11/06/2022] Open
|
41
|
Lavi S, D'Alfonso S, Diamantouros P, Camuglia A, Garg P, Teefy P, Jablonsky G, Sridhar K, Lavi R. Remote ischemic postconditioning during percutaneous coronary interventions: remote ischemic postconditioning-percutaneous coronary intervention randomized trial. Circ Cardiovasc Interv 2014; 7:225-32. [PMID: 24692535 DOI: 10.1161/circinterventions.113.000948] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Remote ischemic preconditioning may result in reduction in infarct size during percutaneous coronary intervention (PCI). It is unclear whether remote ischemic postconditioning (RIPost) will reduce the incidence of myocardial injury after PCI, and whether ischemic conditioning of a larger remote organ (thigh versus arm) would provide further myocardial protection. METHODS AND RESULTS We randomized 360 patients presenting with stable or unstable angina (28% of patients) and negative Troponin T at baseline to 3 groups: 2 groups received RIPost (induced by ischemia to upper or lower limb), and a third was the control group. RIPost was applied during PCI immediately after stent deployment, by three 5-minute cycles of blood pressure cuff inflation to >200 mm Hg in the arm or thigh (20 mm Hg in the control) with 5-minute breaks between each cycle. The primary end-point was the proportion of patients with Troponin T levels >3×ULN postprocedure (at 6 or 18-24 hours), where ULN stands for upper limit of normal. A total of 120 patients were randomized to each group. There were no differences in baseline characteristics between the 3 groups. The primary outcome occurred in 30%, 35%, and 35% of the arm, thigh, and control groups, respectively (P=0.64). There were no differences in creatine kinase or high sensitivity C-reactive protein levels after PCI or in the incidence of acute kidney injury between the groups. CONCLUSIONS RIPost during PCI did not reduce the incidence of periprocedural myocardial injury. Similar effect was obtained when remote ischemia was induced to the upper or lower limb. CLINICAL TRIAL REGISTRATION URL http://www.clinicaltrials.gov. Unique identifier: NCT00970827.
Collapse
Affiliation(s)
- Shahar Lavi
- From the Western University (S.L., P.D., A.C., P.G., P.T., G.J., K.S., R.L.), London, Ontario, Canada; and London Health Sciences Centre (S.L., S.D., P.D., A.C., P.G., P.T., G.J., K.S., R.L.), London, Ontario, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Zhou C, Li H, Yao Y, Li L. Delayed remote ischemic preconditioning produces an additive cardioprotection to sevoflurane postconditioning through an enhanced heme oxygenase 1 level partly via nuclear factor erythroid 2-related factor 2 nuclear translocation. J Cardiovasc Pharmacol Ther 2014; 19:558-66. [PMID: 24651515 DOI: 10.1177/1074248414524479] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Although both sevoflurane postconditioning (SPoC) and delayed remote ischemic preconditioning (DRIPC) have been proved effective in various animal and human studies, the combined effect of these 2 strategies remains unclear. Therefore, this study was designed to investigate this effect and elucidate the related signal mechanisms in a Langendorff perfused rat heart model. After 30-minute balanced perfusion, isolated hearts were subjected to 30-minute ischemia followed by 60-minute reperfusion except 90-minute perfusion for control. A synergic cardioprotective effect of SPoC (3% v/v) and DRIPC (4 cycles 5-minute occlusion/5-minute reflow at the unilateral hindlimb once per day for 3 days before heart isolation) was observed with facilitated cardiac functional recovery and decreased cardiac enzyme release. The infarct size-limiting effect was more pronounced in the combined group (6.76% ± 2.18%) than in the SPoC group (16.50% ± 4.55%, P < .001) or in the DRIPC group (10.22% ± 2.57%, P = .047). Subsequent analysis revealed that an enhanced heme oxygenase 1 (HO-1) expression, but not protein kinase B/AKt or extracellular signal-regulated kinase 1 and 2 activation, was involved in the synergic cardioprotective effect, which was further confirmed in the messenger RNA level of HO-1. Such trend was also observed in the nuclear factor erythroid 2-related factor 2 (Nrf2) nuclear translocation, an upstream regulation of HO-1. In addition, correlation analysis showed a significantly positive relationship between HO-1 expression and Nrf2 translocation (r = 0.729, P < .001). Hence, we conclude that DRIPC may produce an additive cardioprotection to SPoC through an enhanced HO-1 expression partly via Nrf2 translocation.
Collapse
Affiliation(s)
- Chenghui Zhou
- Department of Anesthesiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Huatong Li
- Department of Anesthesiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuntai Yao
- Department of Anesthesiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lihuan Li
- Department of Anesthesiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
43
|
Sachdeva J, Dai W, Gerczuk PZ, Kloner RA. Combined remote perconditioning and postconditioning failed to attenuate infarct size and contractile dysfunction in a rat model of coronary artery occlusion. J Cardiovasc Pharmacol Ther 2014; 19:567-73. [PMID: 24607766 DOI: 10.1177/1074248413518967] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Although preconditioning remains one of the most powerful maneuvers to reduce myocardial infarct size, it is not feasible in the clinical setting to pretreat patients prior to acute myocardial infarction (MI). The purpose of this study was to investigate the effect of more clinically relevant therapies of remote perconditioning, postconditioning, and the combined effect of remote perconditioning and postconditioning on myocardial infarct size in an anesthetized rat model. METHODS Anesthetized rats were subjected to 45 minutes of proximal left coronary artery occlusion followed by 2 hours of reperfusion. Remote perconditioning was performed 5 minutes after left coronary occlusion with 4 cycles of 5 minutes of occlusion and reperfusion of both the femoral arteries. Postconditioning was applied immediately prior to 2 hours of full reperfusion with 6 cycles of 10 seconds occlusion-reperfusion of the coronary artery. The combined effect was produced by preceding the postconditioning regimen with remote perconditioning, after 5 minutes of left coronary occlusion. RESULTS Remote perconditioning and postconditioning alone failed to reduce infarct size expressed as percentage of the risk zone (42.2% ± 3.9% and 45.0% ± 4.3%). The combination of remote perconditioning and postconditioning also failed to reduce infarct size (45.3% ± 4.1%) as compared to the untreated ischemia-reperfusion group (48.7% ± 3.4%). Hemodynamics including left ventricular end-systole and end-diastolic pressures, +dP/dt, -dP/dt, and heart rate did not show any improvement in the conditioning groups. CONCLUSION This study shows that remote perconditioning and postconditioning alone or combined neither improve hemodynamics nor reduce infarct size in the rat model of MI.
Collapse
Affiliation(s)
- Jaspreet Sachdeva
- The Heart Institute of Good Samaritan Hospital, and Division of Cardiovascular Medicine of the Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Wangde Dai
- The Heart Institute of Good Samaritan Hospital, and Division of Cardiovascular Medicine of the Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Paul Z Gerczuk
- The Heart Institute of Good Samaritan Hospital, and Division of Cardiovascular Medicine of the Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Robert A Kloner
- The Heart Institute of Good Samaritan Hospital, and Division of Cardiovascular Medicine of the Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
44
|
Prunier F, Angoulvant D, Saint Etienne C, Vermes E, Gilard M, Piot C, Roubille F, Elbaz M, Ovize M, Bière L, Jeanneteau J, Delépine S, Benard T, Abi-Khalil W, Furber A. The RIPOST-MI study, assessing remote ischemic perconditioning alone or in combination with local ischemic postconditioning in ST-segment elevation myocardial infarction. Basic Res Cardiol 2014; 109:400. [PMID: 24407359 DOI: 10.1007/s00395-013-0400-y] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2013] [Revised: 12/09/2013] [Accepted: 12/20/2013] [Indexed: 11/30/2022]
Abstract
Local ischemic postconditioning (IPost) and remote ischemic perconditioning (RIPer) are promising cardioprotective therapies in ST-elevation myocardial infarction (STEMI). We aimed: (1) to investigate whether RIPer initiated at the catheterization laboratory would reduce infarct size, as measured using serum creatine kinase-MB isoenzyme (CK-MB) release as a surrogate marker; (2) to assess if the combination of RIPer and IPost would provide an additional reduction. Patients (n = 151) were randomly allocated to one of the following groups: (1) control group, percutaneous transluminal coronary angioplasty (PTCA) alone; (2) RIPer group, PTCA combined with RIPer, consisting of three cycles of 5-min inflation and 5-min deflation of an upper-arm blood-pressure cuff initiated before reperfusion; (3) RIPer+IPost group, PTCA combined with RIPer and IPost, consisting of four cycles of 1-min inflation and 1-min deflation of the angioplasty balloon. The CK-MB area under the curve (AUC) over 72 h was reduced in RIPer, and RIPer+IPost groups, by 31 and 29 %, respectively, compared to the Control group; however, CK-MB AUC differences between the three groups were not statistically significant (p = 0.06). Peak CK-MB, CK-MB AUC to area at risk (AAR) ratio, and peak CK-MB level to AAR ratio were all significantly reduced in the RIPer and RIPer+IPost groups, compared to the Control group. On the contrary, none of these parameters was significantly different between RIPer+IPost and RIPer groups. To conclude, starting RIPer therapy immediately prior to revascularization was shown to reduce infarct size in STEMI patients, yet combining this therapy with an IPost strategy did not lead to further decrease in infarct size.
Collapse
Affiliation(s)
- Fabrice Prunier
- Service de Cardiologie, EA 3860, Laboratoire Cardioprotection, Remodelage et Thrombose, Université Angers, CHU Angers, rue Haute de Reculée, 49045, Angers, France,
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Czigány Z, Turóczi Z, Ónody P, Harsányi L, Lotz G, Hegedüs V, Szijártó A. Remote ischemic perconditioning protects the liver from ischemia-reperfusion injury. J Surg Res 2013; 185:605-13. [PMID: 23953788 DOI: 10.1016/j.jss.2013.07.018] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Revised: 06/24/2013] [Accepted: 07/08/2013] [Indexed: 12/16/2022]
Abstract
BACKGROUND Ischemia-reperfusion (IR)-induced injury is a frequent sequel of major liver resections. IR injury after prolonged surgical interventions could be the source of increased risk of postoperative morbidity and mortality. Hepatoprotective effects of this new feasible method called remote ischemic perconditioning (RIPER) were investigated in our rat model of IR injury. MATERIALS AND METHODS Male Wistar rats underwent ischemia for 60 min on two-thirds of their livers, followed by 1, 6, and 24 h of reperfusion (n = 72, 8 per group). During liver ischemia, but before reperfusion, rats in the treated groups received four cycles of brief infrarenal aortic clamping as perconditioning. Liver microcirculation was monitored by laser Doppler flowmeter parallel with mean arterial pressure measurements. Liver tissue injury and redox homeostasis were investigated. Furthermore, serum tumor necrosis factor alpha (TNF-α) levels were measured. RESULTS In the RIPER group, compared with the IR group, serum transaminase levels were significantly lower after each reperfusion period (alanine aminotransferase: 1 h, P < 0.001; 6 h, P < 0.05; 24 h, P < 0.01 and aspartate aminotransferase: 1 h, P < 0.001; 6 h, P < 0.05; 24 h, P < 0.05). Reperfusion microcirculatory parameters significantly improved in the perconditioned group compared with those in the IR group (reperfusion area: P = 0.005; maximal plateau: P = 0.0002). Regarding TNF-α levels, significant differences were detected between the two IR injured groups (RIPER versus IR: 1 h, 34.3 ± 12.8 pg/mL versus 205.7 ± 60.9 pg/mL, P < 0.001; 6 h, 60.6 ± 11.7 pg/mL versus 110.4 ± 21.6 pg/mL, P < 0.05). Results of the histologic assessment and redox state measurements also showed favorable changes. CONCLUSIONS Our team firstly reported the protective effects of RIPER on liver morphology, redox homeostasis, and microcirculation and proposed the changes of TNF-α expression.
Collapse
Affiliation(s)
- Zoltán Czigány
- 1st Department of Surgery, Semmelweis University, Budapest, Hungary
| | | | | | | | | | | | | |
Collapse
|
46
|
Endoplasmic Reticulum Stress Pathway Involvement in Local and Remote Myocardial Ischemic Conditioning. Shock 2013; 39:433-9. [DOI: 10.1097/shk.0b013e31828e4f80] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
47
|
Matos RI, Watson RS, Nadkarni VM, Huang HH, Berg RA, Meaney PA, Carroll CL, Berens RJ, Praestgaard A, Weissfeld L, Spinella PC. Duration of Cardiopulmonary Resuscitation and Illness Category Impact Survival and Neurologic Outcomes for In-hospital Pediatric Cardiac Arrests. Circulation 2013; 127:442-51. [DOI: 10.1161/circulationaha.112.125625] [Citation(s) in RCA: 196] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background—
Pediatric cardiopulmonary resuscitation (CPR) for >20 minutes has been considered futile after pediatric in-hospital cardiac arrests. This concept has recently been questioned, although the effect of CPR duration on outcomes has not recently been described. Our objective was to determine the relationship between CPR duration and outcomes after pediatric in-hospital cardiac arrests.
Methods and Results—
We examined the effect of CPR duration for pediatric in-hospital cardiac arrests from the Get With The Guidelines–Resuscitation prospective, multicenter registry of in-hospital cardiac arrests. We included 3419 children from 328 US and Canadian Get With The Guidelines–Resuscitation sites with an in-hospital cardiac arrest between January 2000 and December 2009. Patients were stratified into 5 patient illness categories: surgical cardiac, medical cardiac, general medical, general surgical, and trauma. Survival to discharge was 27.9%, but only 19.0% of all cardiac arrest patients had favorable neurological outcomes. Between 1 and 15 minutes of CPR, survival decreased linearly by 2.1% per minute, and rates of favorable neurological outcome decreased by 1.2% per minute. Adjusted probability of survival was 41% for CPR duration of 1 to 15 minutes and 12% for >35 minutes. Among survivors, favorable neurological outcome occurred in 70% undergoing <15 minutes of CPR and 60% undergoing CPR >35 minutes. Compared with general medical patients, surgical cardiac patients had the highest adjusted odds ratios for survival and favorable neurological outcomes, 2.5 (95% confidence interval, 1.8–3.4) and 2.7 (95% confidence interval, 2.0–3.9), respectively.
Conclusions—
CPR duration was independently associated with survival to hospital discharge and neurological outcome. Among survivors, neurological outcome was favorable for the majority of patients. Performing CPR for >20 minutes is not futile in some patient illness categories.
Collapse
Affiliation(s)
- Renée I. Matos
- From the CRISMA Center, Pittsburgh, PA (R.I.M., R.S.W, L.W.); Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA (R.I.M., R.S.W.); Department of Pediatrics, San Antonio Military Medical Center, San Antonio, TX (R.I.M.); The Children’s Hospital of Philadelphia, Philadelphia, PA (V.M.N., R.A.B., P.A.M.); Department of Biostatistics, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA (H.-H.H., L.W.); Connecticut Children’s Medical
| | - R. Scott Watson
- From the CRISMA Center, Pittsburgh, PA (R.I.M., R.S.W, L.W.); Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA (R.I.M., R.S.W.); Department of Pediatrics, San Antonio Military Medical Center, San Antonio, TX (R.I.M.); The Children’s Hospital of Philadelphia, Philadelphia, PA (V.M.N., R.A.B., P.A.M.); Department of Biostatistics, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA (H.-H.H., L.W.); Connecticut Children’s Medical
| | - Vinay M. Nadkarni
- From the CRISMA Center, Pittsburgh, PA (R.I.M., R.S.W, L.W.); Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA (R.I.M., R.S.W.); Department of Pediatrics, San Antonio Military Medical Center, San Antonio, TX (R.I.M.); The Children’s Hospital of Philadelphia, Philadelphia, PA (V.M.N., R.A.B., P.A.M.); Department of Biostatistics, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA (H.-H.H., L.W.); Connecticut Children’s Medical
| | - Hsin-Hui Huang
- From the CRISMA Center, Pittsburgh, PA (R.I.M., R.S.W, L.W.); Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA (R.I.M., R.S.W.); Department of Pediatrics, San Antonio Military Medical Center, San Antonio, TX (R.I.M.); The Children’s Hospital of Philadelphia, Philadelphia, PA (V.M.N., R.A.B., P.A.M.); Department of Biostatistics, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA (H.-H.H., L.W.); Connecticut Children’s Medical
| | - Robert A. Berg
- From the CRISMA Center, Pittsburgh, PA (R.I.M., R.S.W, L.W.); Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA (R.I.M., R.S.W.); Department of Pediatrics, San Antonio Military Medical Center, San Antonio, TX (R.I.M.); The Children’s Hospital of Philadelphia, Philadelphia, PA (V.M.N., R.A.B., P.A.M.); Department of Biostatistics, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA (H.-H.H., L.W.); Connecticut Children’s Medical
| | - Peter A. Meaney
- From the CRISMA Center, Pittsburgh, PA (R.I.M., R.S.W, L.W.); Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA (R.I.M., R.S.W.); Department of Pediatrics, San Antonio Military Medical Center, San Antonio, TX (R.I.M.); The Children’s Hospital of Philadelphia, Philadelphia, PA (V.M.N., R.A.B., P.A.M.); Department of Biostatistics, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA (H.-H.H., L.W.); Connecticut Children’s Medical
| | - Christopher L. Carroll
- From the CRISMA Center, Pittsburgh, PA (R.I.M., R.S.W, L.W.); Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA (R.I.M., R.S.W.); Department of Pediatrics, San Antonio Military Medical Center, San Antonio, TX (R.I.M.); The Children’s Hospital of Philadelphia, Philadelphia, PA (V.M.N., R.A.B., P.A.M.); Department of Biostatistics, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA (H.-H.H., L.W.); Connecticut Children’s Medical
| | - Richard J. Berens
- From the CRISMA Center, Pittsburgh, PA (R.I.M., R.S.W, L.W.); Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA (R.I.M., R.S.W.); Department of Pediatrics, San Antonio Military Medical Center, San Antonio, TX (R.I.M.); The Children’s Hospital of Philadelphia, Philadelphia, PA (V.M.N., R.A.B., P.A.M.); Department of Biostatistics, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA (H.-H.H., L.W.); Connecticut Children’s Medical
| | - Amy Praestgaard
- From the CRISMA Center, Pittsburgh, PA (R.I.M., R.S.W, L.W.); Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA (R.I.M., R.S.W.); Department of Pediatrics, San Antonio Military Medical Center, San Antonio, TX (R.I.M.); The Children’s Hospital of Philadelphia, Philadelphia, PA (V.M.N., R.A.B., P.A.M.); Department of Biostatistics, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA (H.-H.H., L.W.); Connecticut Children’s Medical
| | - Lisa Weissfeld
- From the CRISMA Center, Pittsburgh, PA (R.I.M., R.S.W, L.W.); Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA (R.I.M., R.S.W.); Department of Pediatrics, San Antonio Military Medical Center, San Antonio, TX (R.I.M.); The Children’s Hospital of Philadelphia, Philadelphia, PA (V.M.N., R.A.B., P.A.M.); Department of Biostatistics, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA (H.-H.H., L.W.); Connecticut Children’s Medical
| | - Philip C. Spinella
- From the CRISMA Center, Pittsburgh, PA (R.I.M., R.S.W, L.W.); Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA (R.I.M., R.S.W.); Department of Pediatrics, San Antonio Military Medical Center, San Antonio, TX (R.I.M.); The Children’s Hospital of Philadelphia, Philadelphia, PA (V.M.N., R.A.B., P.A.M.); Department of Biostatistics, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA (H.-H.H., L.W.); Connecticut Children’s Medical
| |
Collapse
|
48
|
Yu T, Zhu W, Gu B, Li S, Wang F, Liu M, Wei M, Li J. Simvastatin attenuates sympathetic hyperinnervation to prevent atrial fibrillation during the postmyocardial infarction remodeling process. J Appl Physiol (1985) 2012; 113:1937-44. [PMID: 22984252 DOI: 10.1152/japplphysiol.00451.2012] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Statin, as a 3-hydroxy-3-methyl-glutaryl-CoA reductase inhibitor, has been shown to prevent atrial fibrillation (AF) due to its anti-inflammatory and antioxidant effects. However, it is still not known whether statin can improve autonomic remodeling to prevent AF. In the present study, using an in vivo rat myocardial infarction (MI) model, we aimed to test whether simvastatin can attenuate nerve sprouting and sympathetic hyperinnervation to prevent AF during the post-MI remodeling process. Our data demonstrate that simvastatin, delivered 3 days after MI for 4 wk, can result in significant decreases in plasma levels of both TNF-α (239 ± 23 pg/ml) and IL-1β (123 ± 11 pg/ml) compared with MI rats without therapy (TNF-α, 728 ± 57 pg/ml; IL-1β, 213 ± 21 pg/ml; P < 0.05), which, however, were still higher than sham-operated rats (TNF-α, 194 ± 20 pg/ml; IL-1β, 75 ± 8 pg/ml; P < 0.05). The similar pattern of changes in inflammation responses was also observed in TNF-α and IL-1β protein expression in the left atrium free wall. The suppressed inflammation responses were associated with reduced superoxide and malondialdehyde generation in the atrium. These changes account for decreases in neural growth factor expression at levels of both mRNA (1.2 ± 0.09 AU vs. MI group, 1.78 ± 0.16 AU) and protein (1.57 ± 0.17 AU vs. MI group, 2.24 ± 0.19 AU; P < 0.05), thus resulting in reduced nerve sprouting and sympathetic hyperinnervation. Accordingly, the rate adaptation of the atrial effective refractory period also recovered, leading to the decreased inducibility of AF. These data suggest that simvastatin administration after MI can prevent AF through reduced sympathetic hyperinnervation.
Collapse
Affiliation(s)
- Tao Yu
- Division of Cardiology, Shanghai Sixth Hospital, Shanghai Jiaotong University School of Medicine, State Key Discipline Division, Shanghai, China
| | - Wei Zhu
- Division of Cardiology, Shanghai Sixth Hospital, Shanghai Jiaotong University School of Medicine, State Key Discipline Division, Shanghai, China
| | - Beiyin Gu
- Division of Cardiology, Shanghai Sixth Hospital, Shanghai Jiaotong University School of Medicine, State Key Discipline Division, Shanghai, China
| | - Shuai Li
- Division of Cardiology, Shanghai Sixth Hospital, Shanghai Jiaotong University School of Medicine, State Key Discipline Division, Shanghai, China
| | - Fabing Wang
- Division of Cardiology, Shanghai Sixth Hospital, Shanghai Jiaotong University School of Medicine, State Key Discipline Division, Shanghai, China
| | - Mingya Liu
- Division of Cardiology, Shanghai Sixth Hospital, Shanghai Jiaotong University School of Medicine, State Key Discipline Division, Shanghai, China
| | - Meng Wei
- Division of Cardiology, Shanghai Sixth Hospital, Shanghai Jiaotong University School of Medicine, State Key Discipline Division, Shanghai, China
| | - Jingbo Li
- Division of Cardiology, Shanghai Sixth Hospital, Shanghai Jiaotong University School of Medicine, State Key Discipline Division, Shanghai, China
| |
Collapse
|
49
|
Czigány Z, Turóczi Z, Bulhardt O, Hegedüs V, Lotz G, Rakonczay Z, Balla Z, Harsányi L, Szijártó A. [Remote ischemic conditioning: short-term effects on rat liver ischemic-reperfusion injury]. Orv Hetil 2012; 153:1579-87. [PMID: 23022881 DOI: 10.1556/oh.2012.29469] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Several techniques have been developed to reduce ischemic-reperfusion injury. A novel method is the remote ischemic perconditioning, applied parallel with target organ ischemia. AIM The aim of the study was to determine the extent of liver ischemic-reperfusion injury via the application of this novel method. METHODS Male Wistar rats (n = 30, 10/group) were subjected to 60-minute partial liver ischemia and 60-minute reperfusion. Rats in the perconditioned group received conditioning treatment during the last 40 minutes of liver ischemia by infrarenal aortic clamping. Hepatic and lower limb microcirculation was monitored by laser Doppler flowmeter during reperfusion. After reperfusion, liver samples were taken for routine histological examination and redox-state assessment. Serum transaminase activities and liver tissue heat-shock protein-72 expression were measured. RESULTS Parameters of microcirculation showed significant (p<0.05) improvement in the perconditioned group in comparison with the control. Besides the significant improvement observed in the serum alanine amino-transferase activities, significantly milder tissue injury was detected histologically in the liver sections of the perconditioned group. Moreover, significant improvement was found in the redox-state parameters. CONCLUSION Perconditioning may be a reasonable possibility to reduce liver ischemic-reperfusion injury.
Collapse
Affiliation(s)
- Zoltán Czigány
- Semmelweis Egyetem, Általános Orvostudományi Kar I. Sebészeti Klinika Budapest
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Yamaki VN, Gonçalves TB, Coelho JVB, Pontes RVS, Costa FLDS, Brito MVH. Efeito protetor do per-condicionamento isquêmico remoto nas lesões da síndrome de isquemia e reperfusão renal em ratos. Rev Col Bras Cir 2012; 39:529-33. [DOI: 10.1590/s0100-69912012000600014] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2012] [Accepted: 07/13/2012] [Indexed: 11/22/2022] Open
Abstract
OBJETIVO: Avaliar o efeito protetor do per-condicionamento isquêmico remoto nas lesões de isquemia e reperfusão renal induzida. MÉTODOS: Quinze ratos (Rattus Novergicus) foram randomizados em três grupos (n=5): Grupo Normalidade (GN), Grupo Controle - Isquemia e Reperfusão (GIR) e Grupo Per-condicionamento isquêmico remoto (GPER). Com exceção do grupo GN, todos os demais foram submetidos à isquemia renal de 30 minutos. No grupo GPER, foi realizado o per-condicionamento isquêmico remoto, constituído de três ciclos de isquemia e reperfusão de cinco minutos cada aplicado, durante o período de isquemia, no membro posterior esquerdo dos ratos, por meio de torniquete. Para quantificar as lesões, foram dosados os níveis séricos de ureia e creatinina, bem como, analisada a histopatologia renal. RESULTADOS: O grupo GPER apresentou-se com melhores níveis de ureia (83,74 ± 14,58%) e creatinina (0,72 ± 26,14%) quando comparado ao grupo GIR, se aproximando do grupo GN. Na histopatologia, os menores níveis de degeneração hidrópica e congestão medular foram encontrados no grupo GPER. CONCLUSÃO: O per-condicionamento isquêmico remoto apresentou importante efeito protetor na lesão de isquemia e reperfusão renal.
Collapse
|