1
|
Burda R, Burda J, Morochovič R. Ischemic Tolerance—A Way to Reduce the Extent of Ischemia–Reperfusion Damage. Cells 2023; 12:cells12060884. [PMID: 36980225 PMCID: PMC10047660 DOI: 10.3390/cells12060884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 03/08/2023] [Accepted: 03/11/2023] [Indexed: 03/14/2023] Open
Abstract
Individual tissues have significantly different resistance to ischemia–reperfusion damage. There is still no adequate treatment for the consequences of ischemia–reperfusion damage. By utilizing ischemic tolerance, it is possible to achieve a significant reduction in the extent of the cell damage due to ischemia–reperfusion injury. Since ischemia–reperfusion damage usually occurs unexpectedly, the use of preconditioning is extremely limited. In contrast, postconditioning has wider possibilities for use in practice. In both cases, the activation of ischemic tolerance can also be achieved by the application of sublethal stress on a remote organ. Despite very encouraging and successful results in animal experiments, the clinical results have been disappointing so far. To avoid the factors that prevent the activation of ischemic tolerance, the solution has been to use blood plasma containing tolerance effectors. This plasma is taken from healthy donors in which, after exposure to two sublethal stresses within 48 h, effectors of ischemic tolerance occur in the plasma. Application of this activated plasma to recipient animals after the end of lethal ischemia prevents cell death and significantly reduces the consequences of ischemia–reperfusion damage. Until there is a clear chemical identification of the end products of ischemic tolerance, the simplest way of enhancing ischemic tolerance will be the preparation of activated plasma from young healthy donors with the possibility of its immediate use in recipients during the initial treatment.
Collapse
Affiliation(s)
- Rastislav Burda
- Department of Trauma Surgery, Faculty of Medicine, Pavol Jozef Šafárik University in Košice, Rastislavova 43, 040 01 Košice, Slovakia
- Department of Trauma Surgery, Louis Pasteur University Hospital, Rastislavova 43, 040 01 Košice, Slovakia
- Correspondence:
| | - Jozef Burda
- Institute of Neurobiology, Slovak Academy of Sciences, 040 01 Košice, Slovakia
| | - Radoslav Morochovič
- Department of Trauma Surgery, Faculty of Medicine, Pavol Jozef Šafárik University in Košice, Rastislavova 43, 040 01 Košice, Slovakia
- Department of Trauma Surgery, Louis Pasteur University Hospital, Rastislavova 43, 040 01 Košice, Slovakia
| |
Collapse
|
2
|
Aslan G, Atessahin A, Sahna E. The inhibition of apoptosis through myocardial postconditioning by affecting Fas/FasIg signaling through miR139-3p and miR181a-1. J Card Surg 2020; 35:564-570. [PMID: 31945231 DOI: 10.1111/jocs.14426] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
BACKGROUND AND AIM OF THE STUDY Ischemic postconditioning (PostC) is considered to be one of the strongest mechanisms limiting the extent of myocardial infarction, and reducing ischemia-reperfusion (I/R) injury. I/R-induced myocardial injury results in apoptosis, autophagy, and necrosis. The aim of the present study was to investigate the roles of the necrotic gene cytochrome b-245 beta chain (Cybb); Cybb-related microRNA miR139-3p; the autophagy gene Beclin-1 (Becn1); proapoptotic genes Fas, Faslg and growth arrest and DNA-damage-inducible 45 alpha (Gadd45a); and apoptosis-related microRNA miR181a-1 levels on I/R injury, as well as, the potential protective effects of PostC through this gene and microRNAs. METHODS The left main coronary artery was subjected to ischemia for 30 minutes, followed by reperfusion for 120 minutes. PostC involved three cycles of I/R, each lasting 10 seconds. Gene and microRNA levels were analyzed using a quantitative reverse transcription-polymerase chain reaction. RESULTS Although an increase was observed in the expression levels of the Cybb, Fas, Faslg and Gadd45a genes, the miR139-3p, miR181a-1, and Becn1 expression levels were found to decrease with I/R injury. PostC was determined to restore the expression of all the genes to the normal levels. CONCLUSIONS The abovementioned genes can be used as important prognostic markers in the diagnosis of reperfusion injury and in the evaluation of treatment efficacy. It was further noted that increased expression of CYBB, which is one of the target genes for miR139-3p, and a decreased expression of miR181a-1 may cause apoptosis by affecting Fas and Faslg signaling. PostC can inhibit apoptosis by increasing miR139-3p and miR181a-1 levels.
Collapse
Affiliation(s)
- Gulnur Aslan
- Department of Medical Pharmacology, Faculty of Medicine, Firat University, Elazig, Turkey
| | - Ahmet Atessahin
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Firat University, Elazig, Turkey
| | - Engin Sahna
- Department of Medical Pharmacology, Faculty of Medicine, Firat University, Elazig, Turkey
| |
Collapse
|
3
|
Sanz MN, Farine E, Niederberger P, Méndez-Carmona N, Wyss RK, Arnold M, Gulac P, Fiedler GM, Gressette M, Garnier A, Carrel TP, Tevaearai Stahel HT, Longnus SL. Cardioprotective reperfusion strategies differentially affect mitochondria: Studies in an isolated rat heart model of donation after circulatory death (DCD). Am J Transplant 2019; 19:331-344. [PMID: 30019521 DOI: 10.1111/ajt.15024] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2017] [Revised: 06/22/2018] [Accepted: 07/05/2018] [Indexed: 01/25/2023]
Abstract
Donation after circulatory death (DCD) holds great promise for improving cardiac graft availability; however, concerns persist regarding injury following warm ischemia, after donor circulatory arrest, and subsequent reperfusion. Application of preischemic treatments is limited for ethical reasons; thus, cardioprotective strategies applied at graft procurement (reperfusion) are of particular importance in optimizing graft quality. Given the key role of mitochondria in cardiac ischemia-reperfusion injury, we hypothesize that 3 reperfusion strategies-mild hypothermia, mechanical postconditioning, and hypoxia, when briefly applied at reperfusion onset-provoke mitochondrial changes that may underlie their cardioprotective effects. Using an isolated, working rat heart model of DCD, we demonstrate that all 3 strategies improve oxygen-consumption-cardiac-work coupling and increase tissue adenosine triphosphate content, in parallel with increased functional recovery. These reperfusion strategies, however, differentially affect mitochondria; mild hypothermia also increases phosphocreatine content, while mechanical postconditioning stimulates mitochondrial complex I activity and reduces cytochrome c release (marker of mitochondrial damage), whereas hypoxia upregulates the expression of peroxisome proliferator-activated receptor-gamma coactivator (regulator of mitochondrial biogenesis). Characterization of the role of mitochondria in cardioprotective reperfusion strategies should aid in the identification of new, mitochondrial-based therapeutic targets and the development of effective reperfusion strategies that could ultimately facilitate DCD heart transplantation.
Collapse
Affiliation(s)
- Maria N Sanz
- Department of Cardiovascular Surgery, Inselspital, Bern University Hospital, Bern, Switzerland.,Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Emilie Farine
- Department of Cardiovascular Surgery, Inselspital, Bern University Hospital, Bern, Switzerland.,Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Petra Niederberger
- Department of Cardiovascular Surgery, Inselspital, Bern University Hospital, Bern, Switzerland.,Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Natalia Méndez-Carmona
- Department of Cardiovascular Surgery, Inselspital, Bern University Hospital, Bern, Switzerland.,Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Rahel K Wyss
- Department of Cardiovascular Surgery, Inselspital, Bern University Hospital, Bern, Switzerland.,Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Maria Arnold
- Department of Cardiovascular Surgery, Inselspital, Bern University Hospital, Bern, Switzerland.,Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Patrik Gulac
- Department of Cardiovascular Surgery, Inselspital, Bern University Hospital, Bern, Switzerland.,Department for BioMedical Research, University of Bern, Bern, Switzerland.,Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University, Bratislava, Slovak Republic
| | - Georg M Fiedler
- Center of Laboratory Medicine, University Institute of Clinical Chemistry, Inselspital, Bern University Hospital, Bern, Switzerland
| | - Mélanie Gressette
- UMR-S 1180, INSERM, Université Paris-Sud, Université Paris-Saclay, Châtenay-Malabry, France
| | - Anne Garnier
- UMR-S 1180, INSERM, Université Paris-Sud, Université Paris-Saclay, Châtenay-Malabry, France
| | - Thierry P Carrel
- Department of Cardiovascular Surgery, Inselspital, Bern University Hospital, Bern, Switzerland.,Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Hendrik T Tevaearai Stahel
- Department of Cardiovascular Surgery, Inselspital, Bern University Hospital, Bern, Switzerland.,Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Sarah L Longnus
- Department of Cardiovascular Surgery, Inselspital, Bern University Hospital, Bern, Switzerland.,Department for BioMedical Research, University of Bern, Bern, Switzerland
| |
Collapse
|
4
|
Pagliaro P, Femminò S, Popara J, Penna C. Mitochondria in Cardiac Postconditioning. Front Physiol 2018; 9:287. [PMID: 29632499 PMCID: PMC5879113 DOI: 10.3389/fphys.2018.00287] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 03/12/2018] [Indexed: 12/11/2022] Open
Abstract
Mitochondria play a pivotal role in cardioprotection. Here we report some fundamental studies which considered the role of mitochondrial components (connexin 43, mitochondrial KATP channels and mitochondrial permeability transition pore) in postconditioning cardioprotection. We briefly discuss the role of mitochondria, reactive oxygen species and gaseous molecules in postconditioning. Also the effects of anesthetics-used as cardioprotective substances-is briefly considered in the context of postconditioning. The role of mitochondrial postconditioning signaling in determining the limitation of cell death is underpinned. Issues in clinical translation are briefly considered. The aim of the present mini-review is to discuss in a historical perspective the role of main mitochondria mechanisms in cardiac postconditioning.
Collapse
Affiliation(s)
- Pasquale Pagliaro
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Saveria Femminò
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Jasmin Popara
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Claudia Penna
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| |
Collapse
|
5
|
Jahandiez V, Cour M, Bochaton T, Abrial M, Loufouat J, Gharib A, Varennes A, Ovize M, Argaud L. Fast therapeutic hypothermia prevents post-cardiac arrest syndrome through cyclophilin D-mediated mitochondrial permeability transition inhibition. Basic Res Cardiol 2017; 112:35. [PMID: 28492973 DOI: 10.1007/s00395-017-0624-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 05/02/2017] [Indexed: 03/14/2023]
Abstract
The opening of the mitochondrial permeability transition pore (PTP), which is regulated by the matrix protein cyclophilin D (CypD), plays a key role in the pathophysiology of post-cardiac arrest (CA) syndrome. We hypothesized that therapeutic hypothermia could prevent post-CA syndrome through a CypD-mediated PTP inhibition in both heart and brain. In addition, we investigated whether specific pharmacological PTP inhibition would confer additive protection to cooling. Adult male New Zealand White rabbits underwent 15 min of CA followed by 120 min of reperfusion. Five groups (n = 10-15/group) were studied: control group (CA only), hypothermia group (HT, hypothermia at 32-34 °C induced by external cooling at reperfusion), NIM group (injection at reperfusion of 2.5 mg/kg NIM811, a specific CypD inhibitor), HT + NIM, and sham group. The following measurements were taken: hemodynamics, echocardiography, and cellular damage markers (including S100β protein and troponin Ic). Oxidative phosphorylation and PTP opening were assessed on mitochondria isolated from both brain and heart. Acetylation of CypD was measured by immunoprecipitation in both the cerebral cortex and myocardium. Hypothermia and NIM811 significantly prevented cardiovascular dysfunction, pupillary areflexia, and early tissue damage. Hypothermia and NIM811 preserved oxidative phosphorylation, limited PTP opening in both brain and heart mitochondria and prevented increase in CypD acetylation in brain. There were no additive beneficial effects in the combination of NIM811 and therapeutic hypothermia. In conclusion, therapeutic hypothermia limited post-CA syndrome by preventing mitochondrial permeability transition mainly through a CypD-dependent mechanism.
Collapse
Affiliation(s)
- Vincent Jahandiez
- Hospices Civils de Lyon, Hôpital Edouard Herriot, Service de Réanimation Médicale, 5 Place d'Arsonval, 69437, Lyon Cedex 03, France
- Université de Lyon, Université Claude Bernard Lyon 1, Faculté de médecine Lyon-Est, 69373, Lyon, France
- INSERM UMR 1060, CarMeN, Equipe 5 "Cardioprotection", 69373, Lyon, France
| | - Martin Cour
- Hospices Civils de Lyon, Hôpital Edouard Herriot, Service de Réanimation Médicale, 5 Place d'Arsonval, 69437, Lyon Cedex 03, France
- Université de Lyon, Université Claude Bernard Lyon 1, Faculté de médecine Lyon-Est, 69373, Lyon, France
- INSERM UMR 1060, CarMeN, Equipe 5 "Cardioprotection", 69373, Lyon, France
| | - Thomas Bochaton
- INSERM UMR 1060, CarMeN, Equipe 5 "Cardioprotection", 69373, Lyon, France
| | - Maryline Abrial
- INSERM UMR 1060, CarMeN, Equipe 5 "Cardioprotection", 69373, Lyon, France
| | - Joseph Loufouat
- INSERM UMR 1060, CarMeN, Equipe 5 "Cardioprotection", 69373, Lyon, France
| | - Abdallah Gharib
- INSERM UMR 1060, CarMeN, Equipe 5 "Cardioprotection", 69373, Lyon, France
| | - Annie Varennes
- Hospices Civils de Lyon, Groupement Hospitalier Edouard Herriot, Laboratoire de Biochimie, 69437, Lyon, France
| | - Michel Ovize
- INSERM UMR 1060, CarMeN, Equipe 5 "Cardioprotection", 69373, Lyon, France
| | - Laurent Argaud
- Hospices Civils de Lyon, Hôpital Edouard Herriot, Service de Réanimation Médicale, 5 Place d'Arsonval, 69437, Lyon Cedex 03, France.
- Université de Lyon, Université Claude Bernard Lyon 1, Faculté de médecine Lyon-Est, 69373, Lyon, France.
- INSERM UMR 1060, CarMeN, Equipe 5 "Cardioprotection", 69373, Lyon, France.
| |
Collapse
|
6
|
The Citrus Flavanone Naringenin Protects Myocardial Cells against Age-Associated Damage. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:9536148. [PMID: 28386313 PMCID: PMC5366223 DOI: 10.1155/2017/9536148] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Revised: 02/06/2017] [Accepted: 02/28/2017] [Indexed: 12/21/2022]
Abstract
In recent years, the health-promoting effects of the citrus flavanone naringenin have been examined. The results have provided evidence for the modulation of some key mechanisms involved in cellular damage by this compound. In particular, naringenin has been revealed to have protective properties such as an antioxidant effect in cardiometabolic disorders. Very recently, beneficial effects of naringenin have been demonstrated in old rats. Because aging has been demonstrated to be directly related to the occurrence of cardiac disorders, in the present study, the ability of naringenin to prevent cardiac cell senescence was investigated. For this purpose, a cellular model of senescent myocardial cells was set up and evaluated using colorimetric, fluorimetric, and immunometric techniques. Relevant cellular senescence markers, such as X-gal staining, cell cycle regulator levels, and the percentage of cell cycle-arrested cells, were found to be reduced in the presence of naringenin. In addition, cardiac markers of aging-induced damage, including radical oxidative species levels, mitochondrial metabolic activity, mitochondrial calcium buffer capacity, and estrogenic signaling functions, were also modulated by the compound. These results suggested that naringenin has antiaging effects on myocardial cells.
Collapse
|
7
|
Cour M, Jahandiez V, Loufouat J, Ovize M, Argaud L. Minor Changes in Core Temperature Prior to Cardiac Arrest Influence Outcomes: An Experimental Study. J Cardiovasc Pharmacol Ther 2014; 20:407-13. [PMID: 25540058 DOI: 10.1177/1074248414562911] [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: 09/24/2014] [Accepted: 10/15/2014] [Indexed: 02/05/2023]
Abstract
AIM To investigate whether slight variations in core temperature prior to cardiac arrest (CA) influence short-term outcomes and mitochondrial functions. METHODS AND MATERIALS Three groups of New Zealand White rabbits (n = 12/group) were submitted to 15 minutes of CA at 38°C (T-38 group), 39°C (T-39), or 40°C (T 40) and 120 minutes of reperfusion. A Sham-operated group (n = 6) underwent only surgery. Restoration of spontaneous circulation (ROSC), survival, hemodynamics, and pupillary reactivity were recorded. Animals surviving to the end of the observation period were euthanized to assess fresh brain and heart mitochondrial functions (permeability transition and oxidative phosphorylation). Markers of brain and heart damages were also measured. RESULTS The duration of asphyxia required to induce CA was significantly lower in the T-40 group when compared to the T-38 group (P < .05). The rate of ROSC was >80% in all groups (P = nonsignificant [ns]). Survival significantly differed among the T-38, T-39, and T-40 groups: 10 (83%) of 12, 7 (58%) of 12, and 4 (33%) of 12, respectively (log-rank test, P = .027). At the end of the protocol, none of the animals in the T-40 group had pupillary reflexes compared to 8 (67%) of 12 in the T-38 group (P < .05). Troponin and protein S100B were significantly higher in the T-40 versus T-38 group (P < .05). Cardiac arrest significantly impaired both inner mitochondrial membrane integrity and oxidative phosphorylation in all groups. Brain mitochondria disorders were significantly more severe in the T-40 group compared to the T-38 group (P < .05). CONCLUSION Small changes in body temperature prior to asphyxial CA significantly influence brain mitochondrial functions and short-term outcomes in rabbits.
Collapse
Affiliation(s)
- Martin Cour
- Hospices Civils de Lyon, Groupement Hospitalier Edouard Herriot, Service de Réanimation Médicale, Lyon, France Faculté de médecine Lyon-Est, Université de Lyon, Université Claude Bernard Lyon 1, Lyon, France INSERM UMR 1060, CarMeN, Lyon, France
| | - Vincent Jahandiez
- Hospices Civils de Lyon, Groupement Hospitalier Edouard Herriot, Service de Réanimation Médicale, Lyon, France Faculté de médecine Lyon-Est, Université de Lyon, Université Claude Bernard Lyon 1, Lyon, France INSERM UMR 1060, CarMeN, Lyon, France
| | | | - Michel Ovize
- INSERM UMR 1060, CarMeN, Lyon, France Hospices Civils de Lyon, Groupement Hospitalier Est, Explorations Fonctionnelles Cardiovasculaires & Centre d'Investigations Cliniques de Lyon, Lyon, France
| | - Laurent Argaud
- Hospices Civils de Lyon, Groupement Hospitalier Edouard Herriot, Service de Réanimation Médicale, Lyon, France Faculté de médecine Lyon-Est, Université de Lyon, Université Claude Bernard Lyon 1, Lyon, France INSERM UMR 1060, CarMeN, Lyon, France
| |
Collapse
|
8
|
Abstract
The reduction or cessation of the blood supply to an organ results in tissue ischemia. Ischemia can cause significant tissue damage, and is observed as a result of a thrombosis, as part of a disease process, and during surgery. However, the restoration of the blood supply often causes more damage to the tissue than the ischemic episode itself. Research is therefore focused on identifying the cellular pathways involved in the protection of organs from the damage incurred by this process of ischemia reperfusion (I/R). The hypoxia-inducible factors (HIFs) are a family of heterodimeric transcription factors that are stabilized during ischemia. The genes that are expressed downstream of HIF activity enhance oxygen-independent ATP generation, cell survival, and angiogenesis, amongst other phenotypes. They are, therefore, important factors in the protection of tissues from I/R injury. Interestingly, a number of the mechanisms already known to induce organ protection against I/R injury, including preconditioning, postconditioning, and activation of signaling pathways such as adenosine receptor signaling, converge on the HIF system. This review describes the evidence for HIFs playing a role in I/R protection mediated by these factors, highlights areas that require further study, and discuss whether HIFs themselves are good therapeutic targets for protecting tissues from I/R injury.
Collapse
Affiliation(s)
- Neil J Howell
- Department of Cardiothoracic Surgery, University Hospital Birmingham, Edgbaston, Birmingham, UK
| | - Daniel A Tennant
- College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| |
Collapse
|
9
|
Oosterlinck W, Herijgers P. Cardiomyocyte changes in the metabolic syndrome and implications for endogeneous protective strategies. Expert Rev Cardiovasc Ther 2014; 12:331-43. [DOI: 10.1586/14779072.2014.893825] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
10
|
Zhong GQ, Tu RH, Zeng ZY, Li QJ, He Y, Li S, He Y, Xiao F. Novel functional role of heat shock protein 90 in protein kinase C-mediated ischemic postconditioning. J Surg Res 2014; 189:198-206. [PMID: 24742623 DOI: 10.1016/j.jss.2014.01.038] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Revised: 01/19/2014] [Accepted: 01/24/2014] [Indexed: 10/25/2022]
Abstract
BACKGROUND Previous studies have shown that heat shock protein 90 (HSP90) plays a vital role in ischemic preconditioning. The present study was designed to explore whether HSP90 might be responsible for cardioprotection in ischemic postconditioning (PostC). MATERIALS AND METHODS Rat hearts underwent 30 min of regional ischemia and 2 h of reperfusion in situ, and PostC was effected with three cycles of 30-s reperfusion and 30-s coronary artery occlusion at the end of ischemia. Ninety rats were randomized into five groups: sham; ischemia-reperfusion (I/R); PostC; 1 mg/kg HSP90 inhibitor geldanamycin (GA) plus PostC (PostC + GA1); and 5 mg/kg GA plus PostC (PostC + GA5). The GA was administered 10 min before reperfusion. RESULTS Compared with the I/R group, the PostC group exhibited lower infarct size (46.7 ± 3.0% versus 27.4 ± 4.0%, respectively), release of lactate dehydrogenase and creatine kinase-MB (2252.6 ± 350.8 versus 1713.7 ± 202.4 IU/L, 2804.3 ± 315.7 versus 1846.2 ± 238.0 IU/L, respectively), cardiomyocyte apoptosis (48.4 ± 5.6% versus 27.6 ± 3.8%, respectively), and mitochondrial damage. These beneficial effects were accompanied by an increase in mitochondrial Bcl-2 levels and a decrease in Bax levels. In addition, mitochondrial protein kinase Cepsilon (PKCepsilon) was relatively low in the I/R group but significantly higher in the PostC group, whereas cytosolic PKCepsilon was relatively high in the I/R group but significantly lower in the PostC group, suggesting the translocation of PKCepsilon from cytosol to mitochondria during PostC. However, blocking HSP90 function with GA inhibited the protection of PostC and PKCepsilon mitochondrial translocation. CONCLUSIONS HSP90 is critical in PostC-induced cardioprotection, and its activity might be linked to mitochondrial targeting of PKCepsilon, the activation of which results in upregulation of its target gene, Bcl-2, and the inhibition of proapoptotic Bax in mitochondria.
Collapse
Affiliation(s)
- Guo-Qiang Zhong
- Department of Geriatric Cardiology, First Affiliated Hospital, Guang Xi Medical University, Nanning, China; Department of Cardiology, First Affiliated Hospital, Guang Xi Medical University, Nanning, China.
| | - Rong-Hui Tu
- Department of Geriatric Cardiology, First Affiliated Hospital, Guang Xi Medical University, Nanning, China
| | - Zhi-Yu Zeng
- Department of Geriatric Cardiology, First Affiliated Hospital, Guang Xi Medical University, Nanning, China
| | - Qing-Jie Li
- Department of Cardiology, First Affiliated Hospital, Guang Xi Medical University, Nanning, China
| | - Yan He
- Department of Geriatric Cardiology, First Affiliated Hospital, Guang Xi Medical University, Nanning, China
| | - Shuo Li
- Department of Cardiology, First Affiliated Hospital, Guang Xi Medical University, Nanning, China
| | - Yan He
- Department of Geriatric Cardiology, First Affiliated Hospital, Guang Xi Medical University, Nanning, China
| | - Fei Xiao
- Department of Cardiology, First Affiliated Hospital, Guang Xi Medical University, Nanning, China
| |
Collapse
|
11
|
Protection tissulaire: une nouvelle piste. MEDECINE INTENSIVE REANIMATION 2014. [DOI: 10.1007/s13546-013-0817-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
12
|
Redox balance and cardioprotection. Basic Res Cardiol 2013; 108:392. [DOI: 10.1007/s00395-013-0392-7] [Citation(s) in RCA: 101] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2013] [Revised: 09/24/2013] [Accepted: 10/14/2013] [Indexed: 12/11/2022]
|
13
|
Abstract
Aquaporins are a group of proteins with high-selective permeability for water. A subgroup called aquaglyceroporins is also permeable to glycerol, urea and a few other solutes. Aquaporin function has mainly been studied in the brain, kidney, glands and skeletal muscle, while the information about aquaporins in the heart is still scarce. The current review explores the recent advances in this field, bringing aquaporins into focus in the context of myocardial ischemia, reperfusion, and blood osmolarity disturbances. Since the amount of data on aquaporins in the heart is still limited, examples and comparisons from better-studied areas of aquaporin biology have been used. The human heart expresses aquaporin-1, -3, -4 and -7 at the protein level. The potential roles of aquaporins in the heart are discussed, and some general phenomena that the myocardial aquaporins share with aquaporins in other organs are elaborated. Cardiac aquaporin-1 is mostly distributed in the microvasculature. Its main role is transcellular water flux across the endothelial membranes. Aquaporin-4 is expressed in myocytes, both in cardiac and in skeletal muscle. In addition to water flux, its function is connected to the calcium signaling machinery. It may play a role in ischemia-reperfusion injury. Aquaglyceroporins, especially aquaporin-7, may serve as a novel pathway for nutrient delivery into the heart. They also mediate toxicity of various poisons. Aquaporins cannot influence permeability by gating, therefore, their function is regulated by changes of expression-on the levels of transcription, translation (by microRNAs), post-translational modification, membrane trafficking, ubiquitination and subsequent degradation. Studies using mice genetically deficient for aquaporins have shown rather modest changes in the heart. However, they might still prove to be attractive targets for therapy directed to reduce myocardial edema and injury caused by ischemia and reperfusion.
Collapse
|
14
|
Diazoxide postconditioning induces mitochondrial protein S-Nitrosylation and a redox-sensitive mitochondrial phosphorylation/translocation of RISK elements: no role for SAFE. Basic Res Cardiol 2013; 108:371. [DOI: 10.1007/s00395-013-0371-z] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2012] [Revised: 06/28/2013] [Accepted: 07/05/2013] [Indexed: 02/07/2023]
|
15
|
Fauconnier J, Roberge S, Saint N, Lacampagne A. Type 2 ryanodine receptor: A novel therapeutic target in myocardial ischemia/reperfusion. Pharmacol Ther 2013; 138:323-32. [DOI: 10.1016/j.pharmthera.2013.01.015] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Accepted: 01/22/2013] [Indexed: 10/27/2022]
|
16
|
Penna C, Perrelli MG, Pagliaro P. Mitochondrial pathways, permeability transition pore, and redox signaling in cardioprotection: therapeutic implications. Antioxid Redox Signal 2013; 18:556-99. [PMID: 22668069 DOI: 10.1089/ars.2011.4459] [Citation(s) in RCA: 125] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Reperfusion therapy is the indispensable treatment of acute myocardial infarction (AMI) and must be applied as soon as possible to attenuate the ischemic insult. However, reperfusion is responsible for additional myocardial damage likely involving opening of the mitochondrial permeability transition pore (mPTP). A great part of reperfusion injury occurs during the first minute of reperfusion. The prolonged opening of mPTP is considered one of the endpoints of the cascade to myocardial damage, causing loss of cardiomyocyte function and viability. Opening of mPTP and the consequent oxidative stress due to reactive oxygen and nitrogen species (ROS/RNS) are considered among the major mechanisms of mitochondrial and myocardial dysfunction. Kinases and mitochondrial components constitute an intricate network of signaling molecules and mitochondrial proteins, which interact in response to stressors. Cardioprotective pathways are activated by stimuli such as preconditioning and postconditioning (PostC), obtained with brief intermittent ischemia or with pharmacological agents, which drastically reduce the lethal ischemia/reperfusion injury. The protective pathways converging on mitochondria may preserve their function. Protection involves kinases, adenosine triphosphate-dependent potassium channels, ROS signaling, and the mPTP modulation. Some clinical studies using ischemic PostC during angioplasty support its protective effects, and an interesting alternative is pharmacological PostC. In fact, the mPTP desensitizer, cyclosporine A, has been shown to induce appreciable protections in AMI patients. Several factors and comorbidities that might interfere with cardioprotective signaling are considered. Hence, treatments adapted to the characteristics of the patient (i.e., phenotype oriented) might be feasible in the future.
Collapse
Affiliation(s)
- Claudia Penna
- Department of Clinical and Biological Sciences, University of Turin, Orbassano, Italy
| | | | | |
Collapse
|
17
|
Guo C, Zeng X, Song J, Zhang M, Wang H, Xu X, Du F, Chen B. A soluble receptor for advanced glycation end-products inhibits hypoxia/reoxygenation-induced apoptosis in rat cardiomyocytes via the mitochondrial pathway. Int J Mol Sci 2012; 13:11923-11940. [PMID: 23109892 PMCID: PMC3472784 DOI: 10.3390/ijms130911923] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2012] [Revised: 09/05/2012] [Accepted: 09/11/2012] [Indexed: 12/14/2022] Open
Abstract
Severe myocardial dysfunction and tissue damage resulting from ischemia/reperfusion (I/R) is a common clinical scenario in patients with certain types of heart diseases and therapies such as thrombolysis, percutaneous coronary intervention, coronary artery bypass grafting, and cardiac transplantation. The underlining mechanism of endogenous cardiac protection after I/R injury has been a focus of current research. Growing evidences suggests that soluble receptor for advanced glycation end-products (sRAGE) has a cardioprotective effect; however, its role in I/R injury remains unclear. We hypothesized that exogenous administration of sRAGE during hypoxia/reoxygenation (H/R) induces cardioprotection by inhibiting cardiomyocyte apoptosis via multiple signals, involving mitochondrial membrane potential (MMP), the mitochondrial permeability transition pore (mPTP), mitochondrial cytochrome c, caspase-3, Bcl-2 and Bax. Neonatal rat cardiomyocytes underwent hypoxia for 3-h followed by 2-h reoxygenation or were treated with sRAGE for 10 min before H/R. Compared with H/R alone, sRAGE pretreatment reduced H/R-induced cardiomyocyte apoptosis from 27.9% ± 5.9% to 9.4% ± 0.7% (p < 0.05). In addition, sRAGE treatment significantly inhibited H/R-induced mitochondrial depolarization and mPTP opening, reduced mitochondrial cytochrome c leakage, caspase-3 and caspase-9 activity, and decreased the ratio of Bax to Bcl-2. Therefore, we conclude that the exogenous administration of sRAGE during H/R is involved in cardioprotection by inhibiting apoptosis via the mitochondrial pathway, which, if further confirmed in vivo, may have important clinical implications during H/R.
Collapse
Affiliation(s)
- Caixia Guo
- Department of Cardiology, Beijing Tian Tan Hospital, Capital Medical University, Beijing 100050, China; E-Mails: (J.S.); (M.Z.); (X.X.); (F.D.); (B.C.)
- Author to whom correspondence should be addressed. E-Mail: ; Tel.: +86-10-6709-6562; Fax: +86-10-6709-6567
| | - Xiangjun Zeng
- Department of Pathophysiology, Capital Medical University, Beijing 100069, China; E-Mails: (X.Z.); (H.W.)
| | - Juanjuan Song
- Department of Cardiology, Beijing Tian Tan Hospital, Capital Medical University, Beijing 100050, China; E-Mails: (J.S.); (M.Z.); (X.X.); (F.D.); (B.C.)
| | - Min Zhang
- Department of Cardiology, Beijing Tian Tan Hospital, Capital Medical University, Beijing 100050, China; E-Mails: (J.S.); (M.Z.); (X.X.); (F.D.); (B.C.)
| | - Hongxia Wang
- Department of Pathophysiology, Capital Medical University, Beijing 100069, China; E-Mails: (X.Z.); (H.W.)
| | - Xiaowei Xu
- Department of Cardiology, Beijing Tian Tan Hospital, Capital Medical University, Beijing 100050, China; E-Mails: (J.S.); (M.Z.); (X.X.); (F.D.); (B.C.)
| | - Fenghe Du
- Department of Cardiology, Beijing Tian Tan Hospital, Capital Medical University, Beijing 100050, China; E-Mails: (J.S.); (M.Z.); (X.X.); (F.D.); (B.C.)
| | - Buxing Chen
- Department of Cardiology, Beijing Tian Tan Hospital, Capital Medical University, Beijing 100050, China; E-Mails: (J.S.); (M.Z.); (X.X.); (F.D.); (B.C.)
| |
Collapse
|
18
|
Blockade of electron transport before ischemia protects mitochondria and decreases myocardial injury during reperfusion in aged rat hearts. Transl Res 2012; 160:207-16. [PMID: 22698829 PMCID: PMC3423471 DOI: 10.1016/j.trsl.2012.01.024] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2011] [Revised: 01/25/2012] [Accepted: 01/26/2012] [Indexed: 12/29/2022]
Abstract
Myocardial injury is increased in the aged heart following ischemia and reperfusion (I-R) in both humans and experimental models. Hearts from aged 24-month-old Fischer 344 rats sustain greater cell death and decreased contractile recovery after I-R compared with 6-month-old adult controls. Cardiac mitochondria incur damage during I-R contributing to cell death. Aged rats have a defect in complex III of the mitochondrial electron transport chain (ETC) localized to the interfibrillar population of cardiac mitochondria (IFM), situated in the interior of the cardiomyocyte among the myofibrils. The defect involves the quinol oxidation site (Qo) and increases the production of reactive oxygen species (ROS) in the baseline state. Ischemia further decreases complex III activity via functional inactivation of the iron-sulfur subunit. We studied the contribution of ischemia-induced defects in complex III with the increased cardiac injury in the aged heart. The reversible blockade of the ETC proximal to complex III during ischemia using amobarbital protects mitochondria against ischemic damage, removing the ischemia component of mitochondrial dysfunction. Reperfusion of the aged heart in the absence of ischemic mitochondrial damage decreases net ROS production from mitochondria and reduces cell death. Thus, even despite the persistence of the age-related defects in electron transport, protection against ischemic damage to mitochondria can reduce injury in the aged heart. The direct therapeutic targeting of mitochondria protects against ischemic damage and decreases cardiac injury during reperfusion in the high risk elderly heart.
Collapse
|
19
|
STUMPNER J, SMUL TM, REDEL A, HILZ T, TISCHER-ZEITZ T, EISENBARTH H, SCHICK MA, KEHL F, ROEWER N, LANGE M. Desflurane-induced and ischaemic postconditioning against myocardial infarction are mediated by Pim-1 kinase. Acta Anaesthesiol Scand 2012; 56:904-13. [PMID: 22385356 DOI: 10.1111/j.1399-6576.2012.02657.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/11/2012] [Indexed: 11/30/2022]
Abstract
BACKGROUND Anaesthetic-induced (APOST) and ischaemic postconditioning (IPOST) against myocardial infarction are mediated via phosphatidylinositol-3-kinase/Akt. Pim-1 kinase is acting downstream of Akt and has recently been demonstrated to enhance cardiomyocyte survival. We tested the hypothesis that both APOST and IPOST are mediated by Pim-1 kinase. METHODS Pentobarbital-anaesthetized male C57BL/6 mice were subjected to 45-min coronary artery occlusion (CAO) and 3-h reperfusion. Animals received either no intervention, the Pim-1 kinase inhibitor II (10 μg/g intraperitoneally) or its vehicle dimethy sulfoxide (10 μl/g intraperitoneally). Three minutes prior to the end of CAO, 1.0 minimum alveolar concentration desflurane was administered for 18 min alone or in combination with Pim-1 kinase inhibitor II. IPOST was induced by three cycles of each 10-s ischaemia/reperfusion, and animals received either IPOST alone or in combination with Pim-1 kinase inhibitor II. Infarct size was determined with triphenyltetrazolium chloride and area at risk with Evans blue. Protein expression of Pim-1 kinase, Bad, phospho-Bad(Ser112) and B-cell lymphoma 2 was determined using Western immunoblotting analysis. RESULTS Infarct size in control animals (CON) was 46 ± 3%. Dimethylsulfoxide (47 ± 3%) and Pim-1 kinase inhibitor II (44 ± 5%) did not significantly reduce infarct size. Desflurane (16 ± 2%*; *P < 0.05 vs. CON) and IPOST (21 ± 2%*) significantly reduced infarct size compared with CON. Inhibition of Pim-1 kinase abolished desflurane-induced postconditioning (46 ± 4%) and IPOST (44 ± 5%). Western blot analysis revealed that only desflurane enhances phosphorylation of Bad at serine 112 that was abrogated by Pim-1 kinase inhibitor II. CONCLUSION These data suggest that Pim-1 kinase mediates both desflurane-induced postconditioning and IPOST in mice.
Collapse
Affiliation(s)
- J. STUMPNER
- Department of Anaesthesia and Critical Care; University of Würzburg; Würzburg; Germany
| | - T. M. SMUL
- Department of Anaesthesia and Critical Care; University of Würzburg; Würzburg; Germany
| | | | - T. HILZ
- Department of Anaesthesia and Critical Care; University of Würzburg; Würzburg; Germany
| | - T. TISCHER-ZEITZ
- Department of Anaesthesia and Critical Care; University of Würzburg; Würzburg; Germany
| | - H. EISENBARTH
- Department of Forensic Psychiatry and Psychotherapy; University of Regensburg; Regensburg; Germany
| | - M. A. SCHICK
- Department of Anaesthesia and Critical Care; University of Würzburg; Würzburg; Germany
| | - F. KEHL
- Department of Anesthesiology and Critical Care; Hospital of Karlsruhe; Karlsruhe; Germany
| | - N. ROEWER
- Department of Anaesthesia and Critical Care; University of Würzburg; Würzburg; Germany
| | | |
Collapse
|
20
|
Wang HX, Zhang DM, Zeng XJ, Mu J, Yang H, Lu LQ, Zhang LK. Upregulation of cytochrome P450 2J3/11,12-epoxyeicosatrienoic acid inhibits apoptosis in neonatal rat cardiomyocytes by a caspase-dependent pathway. Cytokine 2012; 60:360-8. [PMID: 22717287 DOI: 10.1016/j.cyto.2012.04.029] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2011] [Revised: 02/10/2012] [Accepted: 04/18/2012] [Indexed: 11/19/2022]
Abstract
Short, nonlethal ischemic episodes administered to hearts directly after ischemic events (ischemic postconditioning, IPost) have an advantage over ischemic preconditioning (IPC). The endogenous cytochrome P450 2J3/11,12-epoxyeicosatrienoic acid (CYP2J3/11,12-EET) is upregulated by IPost, but not IPC, in the rat heart. The CYP epoxygenase inhibitor N-methylsulphonyl-6-(2-propargyloxyphenyl) hexanamide (MS-PPOH) reduces the cardioprotective effects of IPost, but not IPC. We proposed that upregulation of CYP2J3/11,12-EET during IPost induces cardioprotection by inhibiting cardiomyocyte apoptosis and that multiple apoptotic signals, including changes in mitochondrial membrane potential (MMP) and mitochondrial permeability transition pore (mPTP) opening, mitochondrial cytochrome c leakage, caspase-3 levels, and levels of protective kinases such as Bcl-2 and Bax, are involved in the process. Neonatal rat cardiomyocytes underwent 3-h hypoxia followed by 2-, 5-, or 6-h reoxygenation (H/R) or three cycles of 5-min reoxygenation followed by 5-min hypoxia before 90-min reoxygenation (HPost); or were transfected with pcDNA3.1-CYP2J3 for 48 h before H/R; or were treated with MS-PPOH for 10 min before HPost. For HPost alone, pcDNA3.1-CYP2J3 transfection attenuated cardiomyocyte apoptosis to 68.4% (p<0.05) of that with H/R. pcDNA3.1-CYP2J3 transfection significantly decreased MMP and inhibited mPTP opening induced by H/R, reduced mitochondrial cytochrome c leakage, cleaved caspase-3 protein expression, and increased the ratio of Bcl-2 to Bax expression. MS-PPOH abolished this effect. Therefore, upregulation of CYP2J3/11,12-EET during HPost is involved in cardioprotection by inhibiting apoptosis via a caspase-dependent pathway, and the apoptosis-suppressive effect may have important clinical implications during HPost.
Collapse
MESH Headings
- 8,11,14-Eicosatrienoic Acid/analogs & derivatives
- 8,11,14-Eicosatrienoic Acid/metabolism
- Amides/pharmacology
- Animals
- Animals, Newborn
- Apoptosis/drug effects
- Caspase 3/metabolism
- Cell Survival/drug effects
- Cytochrome P-450 Enzyme Inhibitors
- Cytochrome P-450 Enzyme System/metabolism
- Cytochromes c/metabolism
- Hypoxia/enzymology
- Hypoxia/pathology
- Membrane Potential, Mitochondrial/drug effects
- Mitochondrial Membrane Transport Proteins/metabolism
- Mitochondrial Permeability Transition Pore
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/enzymology
- Myocytes, Cardiac/pathology
- Oxygen/metabolism
- Rats
- Rats, Wistar
- Up-Regulation/drug effects
- bcl-2-Associated X Protein/metabolism
Collapse
Affiliation(s)
- Hong-Xia Wang
- Department of Pathophysiology, Capital Medical University, Beijing 100069, China
| | | | | | | | | | | | | |
Collapse
|
21
|
Abstract
Cardiac ischemia damages the mitochondrial electron transport chain and the damage persists during reperfusion. Ischemic postconditioning (PC), applied during early reperfusion, decreases cardiac injury. This finding suggests that the ischemia-damaged mitochondria can be regulated to decrease cardiac injury. The reversible blockade of electron transport during ischemia prevents damage to mitochondria. We propose that the targets of PC cytoprotective signaling are mitochondria damaged by ischemia. Thus, if ischemia-mediated mitochondrial damage is prevented, PC at the onset of reperfusion will not result in additional protection. Isolated, Langendorff-perfused adult rat hearts underwent 25-minute global ischemia and 30-minute reperfusion. Amobarbital (2.5 mM) was used to reversibly inhibit electron transport during ischemia. PC (6 cycles of 10-second ischemia-reperfusion) was applied at the onset of reperfusion. Subsarcolemmal and interfibrillar mitochondria were isolated after reperfusion. Blockade of electron transport with amobarbital only during ischemia preserved oxidative phosphorylation and decreased myocardial injury. PC, after untreated ischemia, decreased cardiac injury without improvement of oxidative phosphorylation. Blockade of electron transport during ischemia or PC improved calcium tolerance and inner membrane potential in subsarcolemmal mitochondria after reperfusion. In hearts treated with amobarbital before ischemia, PC did not provide further protection. Thus, PC protects myocardium via the regulation of ischemia-damaged mitochondria during early reperfusion.
Collapse
|
22
|
Post-cardiac arrest myocardial dysfunction is improved with cyclosporine treatment at onset of resuscitation but not in the reperfusion phase. Resuscitation 2012; 82 Suppl 2:S41-7. [PMID: 22208177 DOI: 10.1016/s0300-9572(11)70150-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
AIM OF STUDY Significant myocardial dysfunction and high mortality occur after whole-body ischaemia-eperfusion injuries in the post-cardiac arrest status. The inhibition of mitochondrial permeability transition pore (mPTP) opening during ischaemia-reperfusion can ameliorate injuries in the specific organs. We investigated the effect and therapeutic window of pharmacological inhibition of mPTP opening in cardiac arrest. METHODS Forty male Wistar rats were resuscitated after cardiac arrest induced by 8.5 min of asphyxia. Cyclosporine (10 mg/kg) was administered intravenously at onset of resuscitation in protocol 1 study and administered 3 min after ROSC in protocol 2 with placebo control in both. RESULTS Left ventricular systolic (dP/dt 40), diastolic (maximal negative dP/dt) functions and cardiac output were improved in the group with cyclosporine treatment at onset of resuscitation compared to control group (p < 0.01, respectively). Seventy-two hour survival was better in the group with cyclosporine treatment at onset of resuscitation compared to control (p = 0.046). Left ventricular systolic and diastolic function, cardiac output and 72 h survival were not improved in the group with cyclosporine treatment 3 min after ROSC. The severity of mitochondrial damage under electronic microscopy, mPTP opening, mitochondrial respiratory control ratio and ADP:O ratio were ameliorated in the group with cyclosporine treatment at onset of resuscitation (p< 0.05, respectively) but not in the group with cyclosporine treatment at 3 min after ROSC. CONCLUSIONS Post-cardiac arrest myocardial dysfunction and survival can be improved by cyclosporine treatment at onset of resuscitation, but not by the cyclosporine treatment at 3 min after ROSC.
Collapse
|
23
|
Sloan RC, Moukdar F, Frasier CR, Patel HD, Bostian PA, Lust RM, Brown DA. Mitochondrial permeability transition in the diabetic heart: Contributions of thiol redox state and mitochondrial calcium to augmented reperfusion injury. J Mol Cell Cardiol 2012; 52:1009-18. [DOI: 10.1016/j.yjmcc.2012.02.009] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2011] [Revised: 02/13/2012] [Accepted: 02/23/2012] [Indexed: 12/23/2022]
|
24
|
Excitation-contraction coupling in ventricular myocytes is enhanced by paracrine signaling from mesenchymal stem cells. J Mol Cell Cardiol 2012; 52:1249-56. [PMID: 22465692 DOI: 10.1016/j.yjmcc.2012.03.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2011] [Revised: 02/14/2012] [Accepted: 03/12/2012] [Indexed: 12/17/2022]
Abstract
In clinical trials mesenchymal stem cells (MSCs) are transplanted into cardiac ischemic regions to decrease infarct size and improve contractility. However, the mechanism and time course of MSC-mediated cardioprotection are incompletely understood. We tested the hypothesis that paracrine signaling by MSCs promotes changes in cardiac excitation-contraction (EC) coupling that protects myocytes from cell death and enhances contractility. Isolated mouse ventricular myocytes (VMs) were treated with control tyrode, MSC conditioned-tyrode (ConT) or co-cultured with MSCs. The Ca handling properties of VMs were monitored by laser scanning confocal microscopy and whole cell voltage clamp. ConT superfusion of VMs resulted in a time dependent increase of the Ca transient amplitude (ConT(15min): ΔF/F(0)=3.52±0.38, n=14; Ctrl(15min): ΔF/F(0)=2.41±0.35, n=14) and acceleration of the Ca transient decay (τ: ConT: 269±18ms n=14; vs. Ctrl: 315±57ms, n=14). Voltage clamp recordings confirmed a ConT induced increase in I(Ca,L) (ConT: -5.9±0.5 pA/pF n=11; vs. Ctrl: -4.04±0.3 pA/pF, n=12). The change of τ resulted from increased SERCA activity. Changes in the Ca transient amplitude and τ were prevented by the PI3K inhibitors Wortmannin (100nmol/L) and LY294002 (10μmol/L) and the Akt inhibitor V (20μmol/L) indicating regulation through PI3K signal transduction and Akt activation which was confirmed by western blotting. A change in τ was also prevented in eNOS(-/-) myocytes or by inhibition of eNOS suggesting an NO mediated regulation of SERCA activity. Since paracrine signaling further resulted in increased survival of VMs we propose that the Akt induced change in Ca signaling is also a mechanism by which MSCs mediate an anti-apoptotic effect.
Collapse
|
25
|
Zeng Z, Huang HF, Chen MQ, Song F, Zhang YJ. Contributions of heme oxygenase-1 in postconditioning-protected ischemia-reperfusion injury in rat liver transplantation. Transplant Proc 2012; 43:2517-23. [PMID: 21911116 DOI: 10.1016/j.transproceed.2011.04.021] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2010] [Revised: 03/18/2011] [Accepted: 04/21/2011] [Indexed: 12/12/2022]
Abstract
BACKGROUND Heme oxygenase-1 (HO-1), an oxidative stress-response gene up-regulated by various physiological and exogenous stimuli, has cytoprotective activities. Ischemic postconditioning (Postcon) can protect an organ from ischemia-reperfusion (I/R) injury. In the present study, we investigated the potential contributions of HO-1 to Postcon-dependent protection against I/R injury in rat liver transplantation models. MATERIALS AND METHODS Adult male Sprague-Dawley rats were randomly divided into four groups: sham group with laparotomy for liver exposure; I/R group with 24-hour cold ischemia of the donor liver; Postcon group with the same treatment as the I/R group plus ischemic Postcon; and zinc protoporphyrin (ZnPP HO-1 inhibitor) + Postcon group treated the same as the Postcon cohort with donors pretreated using ZnPP 24 hours before the I/R injury. We measured liver tissue and peripheral blood samples collected at 6 hours after reperfusion and serum transaminase levels, histopathology, liver tissue malondialdehyde (MDA) content, superoxide dismutase (SOD) activity and HO-1 expression in the liver. RESULTS Postcon significantly diminished the elevation of serum transaminases levels after I/R injury when compared with I/R and ZnPP+Postcon groups. Postcon treated rats showed significantly lower MDA production and higher SOD activity. HO-1 was induced in rat livers exposed to Postcon; its levels were obviously overexpressed after 6 hours in Postcon rats. Inhibiting the expression of HO-1, negated the protective effects of Postcon. CONCLUSIONS Induction of HO-1 in the Postcon condition played a protective role against hepatic I/R injury and enhanced the early antioxidative activity. The protective effects of Postcon were significantly associated with greater intrahepatic HO-1 expression.
Collapse
Affiliation(s)
- Z Zeng
- Organ Transplant Center, The First Affiliated Hospital of Kunming Medical College, Kunming, China.
| | | | | | | | | |
Collapse
|
26
|
Gomez L, Paillard M, Price M, Chen Q, Teixeira G, Spiegel S, Lesnefsky EJ. A novel role for mitochondrial sphingosine-1-phosphate produced by sphingosine kinase-2 in PTP-mediated cell survival during cardioprotection. Basic Res Cardiol 2011; 106:1341-53. [PMID: 22002221 DOI: 10.1007/s00395-011-0223-7] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2011] [Revised: 08/15/2011] [Accepted: 09/09/2011] [Indexed: 01/15/2023]
Abstract
Although mitochondria are key determinants of myocardial injury during ischemia-reperfusion (I/R), their interaction with critical cytoprotective signaling systems is not fully understood. Sphingosine-1-phosphate (S1P) produced by sphingosine kinase-1 protects the heart from I/R damage. Recently a new role for mitochondrial S1P produced by a second isoform of sphingosine kinase, SphK2, was described to regulate complex IV assembly and respiration via interaction with mitochondrial prohibitin-2. Here we investigated the role of SphK2 in cardioprotection by preconditioning. Littermate (WT) and sphk2 (-/-) mice underwent 45 min of in vivo ischemia and 24 h reperfusion. Mice received no intervention (I/R) or preconditioning (PC) via 5 min I/R before the index ischemia. Despite the activation of PC-cytoprotective signaling pathways in both groups, infarct size in sphk2 (-/-) mice was not reduced by PC (42 ± 3% PC vs. 43 ± 4% I/R, p = ns) versus WT (24 ± 3% PC vs. 43 ± 3% I/R, p < 0.05). sphk2 (-/-) mitochondria exhibited decreased oxidative phosphorylation and increased susceptibility to permeability transition (PTP). Unlike WT, PC did not prevent ischemic damage to electron transport or the increased susceptibility to PTP. To evaluate the direct contribution to the resistance of mitochondria to cytoprotection, SphK2, PHB2 or cytochrome oxidase subunit IV was depleted in cardiomyoblasts. PC protection was abolished by each knockdown concomitant with decreased PTP resistance. These results point to a new action of S1P in cardioprotection and suggest that the mitochondrial S1P produced by SphK2 is required for the downstream protective modulation of PTP as an effector of preconditioning protection.
Collapse
Affiliation(s)
- Ludovic Gomez
- Department of Medicine, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, USA
| | | | | | | | | | | | | |
Collapse
|
27
|
Yin X, Zheng Y, Zhai X, Zhao X, Cai L. Diabetic inhibition of preconditioning- and postconditioning-mediated myocardial protection against ischemia/reperfusion injury. EXPERIMENTAL DIABETES RESEARCH 2011; 2012:198048. [PMID: 21822424 PMCID: PMC3148591 DOI: 10.1155/2012/198048] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2011] [Accepted: 05/31/2011] [Indexed: 01/04/2023]
Abstract
Ischemic preconditioning (IPC) or postconditioning (Ipost) is proved to efficiently prevent ischemia/reperfusion injuries. Mortality of diabetic patients with acute myocardial infarction was found to be 2-6 folds higher than that of non-diabetic patients with same myocardial infarction, which may be in part due to diabetic inhibition of IPC- and Ipost-mediated protective mechanisms. Both IPC- and Ipost-mediated myocardial protection is predominantly mediated by stimulating PI3K/Akt and associated GSK-3β pathway while diabetes-mediated pathogenic effects are found to be mediated by inhibiting PI3K/Akt and associated GSK-3β pathway. Therefore, this review briefly introduced the general features of IPC- and Ipost-mediated myocardial protection and the general pathogenic effects of diabetes on the myocardium. We have collected experimental evidence that indicates the diabetic inhibition of IPC- and Ipost-mediated myocardial protection. Increasing evidence implies that diabetic inhibition of IPC- and Ipost-mediated myocardial protection may be mediated by inhibiting PI3K/Akt and associated GSK-3β pathway. Therefore any strategy to activate PI3K/Akt and associated GSK-3β pathway to release the diabetic inhibition of both IPC and Ipost-mediated myocardial protection may provide the protective effect against ischemia/reperfusion injuries.
Collapse
Affiliation(s)
- Xia Yin
- The Cardiovascular Center, The First Hospital of Jilin University, 71 Xinmin Street, Changchun 130021, China
- KCHRI, The Department of Pediatrics, University of Louisville, Louisville, KY 40202, USA
| | - Yang Zheng
- The Cardiovascular Center, The First Hospital of Jilin University, 71 Xinmin Street, Changchun 130021, China
| | - Xujie Zhai
- Breast Surgery, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Xin Zhao
- The Cardiovascular Center, The First Hospital of Jilin University, 71 Xinmin Street, Changchun 130021, China
| | - Lu Cai
- The Cardiovascular Center, The First Hospital of Jilin University, 71 Xinmin Street, Changchun 130021, China
- KCHRI, The Department of Pediatrics, University of Louisville, Louisville, KY 40202, USA
| |
Collapse
|
28
|
Perrelli MG, Pagliaro P, Penna C. Ischemia/reperfusion injury and cardioprotective mechanisms: Role of mitochondria and reactive oxygen species. World J Cardiol 2011; 3:186-200. [PMID: 21772945 PMCID: PMC3139040 DOI: 10.4330/wjc.v3.i6.186] [Citation(s) in RCA: 236] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2011] [Revised: 05/11/2011] [Accepted: 05/18/2011] [Indexed: 02/06/2023] Open
Abstract
Reperfusion therapy must be applied as soon as possible to attenuate the ischemic insult of acute myocardial infarction (AMI). However reperfusion is responsible for additional myocardial damage, which likely involves opening of the mitochondrial permeability transition pore (mPTP). In reperfusion injury, mitochondrial damage is a determining factor in causing loss of cardiomyocyte function and viability. Major mechanisms of mitochondrial dysfunction include the long lasting opening of mPTPs and the oxidative stress resulting from formation of reactive oxygen species (ROS). Several signaling cardioprotective pathways are activated by stimuli such as preconditioning and postconditioning, obtained with brief intermittent ischemia or with pharmacological agents. These pathways converge on a common target, the mitochondria, to preserve their function after ischemia/reperfusion. The present review discusses the role of mitochondria in cardioprotection, especially the involvement of adenosine triphosphate-dependent potassium channels, ROS signaling, and the mPTP. Ischemic postconditioning has emerged as a new way to target the mitochondria, and to drastically reduce lethal reperfusion injury. Several clinical studies using ischemic postconditioning during angioplasty now support its protective effects, and an interesting alternative is pharmacological postconditioning. In fact ischemic postconditioning and the mPTP desensitizer, cyclosporine A, have been shown to induce comparable protection in AMI patients.
Collapse
Affiliation(s)
- Maria-Giulia Perrelli
- Maria-Giulia Perrelli, Pasquale Pagliaro, Claudia Penna, Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy
| | | | | |
Collapse
|
29
|
Vinten-Johansen J, Granfeldt A, Mykytenko J, Undyala VV, Dong Y, Przyklenk K. The multidimensional physiological responses to postconditioning. Antioxid Redox Signal 2011; 14:791-810. [PMID: 20618066 DOI: 10.1089/ars.2010.3396] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Reperfusion is the definitive treatment to reduce infarct size and other manifestations of postischemic injury. However, reperfusion contributes to postischemic injury, and, therefore, reperfusion therapies do not achieve the optimal salvage of myocardium. Other tissues as well undergo injury after reperfusion, notably, the coronary vascular endothelium. Postconditioning has been shown to have salubrious effects on different tissue types within the heart (cardiomyocytes, endothelium) and to protect against various pathologic processes, including necrosis, apoptosis, contractile dysfunction, arrhythmias, and microvascular injury or "no-reflow." The mechanisms by which postconditioning alters the pathophysiology of reperfusion injury is exceedingly complex and involves physiological mechanisms (e.g., delaying re-alkalinization of tissue pH, triggering release of autacoids, and opening and closing of various channels) and molecular mechanisms (activation of kinases) that affect cellular and subcellular targets or effectors. The physiologic responses to postconditioning are not isolated or mutually exclusive, but are interactive, with one response affecting another in an integrated manner. This integrated response on multiple targets differs from the monotherapy approach by drugs that have failed to reduce reperfusion injury on a consistent basis and may underlie the efficacy of this therapeutic approach across species and in human trials.
Collapse
Affiliation(s)
- Jakob Vinten-Johansen
- Department of Surgery (Cardiothoracic), Carlyle Fraser Heart Center, Emory University, 550 Peachtree Street NE, Atlanta, GA 30308-2225, USA.
| | | | | | | | | | | |
Collapse
|
30
|
Abstract
Several signal transduction pathways are activated by cardioprotective stimuli, including ischemic or pharmacological postconditioning. These pathways converge on a common target, the mitochondria, and cardioprotection by postconditioning is associated with preserved mitochondrial function after ischemia/reperfusion. The present review discusses the role of mitochondria in cardioprotection, especially the involvement of ATP-dependent potassium channels, reactive oxygen species, and the mitochondrial permeability transition pore, and focuses on the effects of postconditioning on mitochondrial function (i.e., their oxygen consumption and calcium retention capacity). The contribution of mitochondria to loss of protection by postconditioning in diseased or aged myocardium is also addressed.
Collapse
Affiliation(s)
- Kerstin Boengler
- Institut für Pathophysiologie, Universitätsklinikum Essen, Hufelandstrasse 55, Essen, Germany.
| | | | | |
Collapse
|
31
|
Giulivi C, Ross-Inta C, Omanska-Klusek A, Napoli E, Sakaguchi D, Barrientos G, Allen PD, Pessah IN. Basal bioenergetic abnormalities in skeletal muscle from ryanodine receptor malignant hyperthermia-susceptible R163C knock-in mice. J Biol Chem 2011; 286:99-113. [PMID: 20978128 PMCID: PMC3013050 DOI: 10.1074/jbc.m110.153247] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2010] [Revised: 10/13/2010] [Indexed: 12/25/2022] Open
Abstract
Malignant hyperthermia (MH) and central core disease in humans have been associated with mutations in the skeletal ryanodine receptor (RyR1). Heterozygous mice expressing the human MH/central core disease RyR1 R163C mutation exhibit MH when exposed to halothane or heat stress. Considering that many MH symptoms resemble those that could ensue from a mitochondrial dysfunction (e.g. metabolic acidosis and hyperthermia) and that MH-susceptible mice or humans have a higher than normal cytoplasmic Ca(2+) concentration at rest, we evaluated the role of mitochondria in skeletal muscle from R163C compared with wild type mice under basal (untriggered) conditions. R163C skeletal muscle exhibited a significant increase in matrix Ca(2+), increased reactive oxygen species production, lower expression of mitochondrial proteins, and higher mtDNA copy number. These changes, in conjunction with lower myoglobin and glycogen contents, Myh4 and GAPDH transcript levels, GAPDH activity, and lower glucose utilization suggested a switch to a compromised bioenergetic state characterized by both low oxidative phosphorylation and glycolysis. The shift in bioenergetic state was accompanied by a dysregulation of Ca(2+)-responsive signaling pathways regulated by calcineurin and ERK1/2. Chronically elevated resting Ca(2+) in R163C skeletal muscle elicited the maintenance of a fast-twitch fiber program and the development of insulin resistance-like phenotype as part of a metabolic adaptation to the R163C RyR1 mutation.
Collapse
Affiliation(s)
- Cecilia Giulivi
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, California 95616, USA.
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Argaud L, Cour M. Ischémie-reperfusion myocardique — Postconditionnement: protéger de l’ischémie à la reperfusion. MEDECINE INTENSIVE REANIMATION 2011. [DOI: 10.1007/s13546-010-0104-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
33
|
Yu GG, Zeng XJ, Wang HX, Lu LQ, Zheng SP, Ma LQ, Chang J, Wang J, Zhang DM, Du FH, Zhang LK. Cytochrome P450 2J3/epoxyeicosatrienoic acids mediate the cardioprotection induced by ischaemic post-conditioning, but not preconditioning, in the rat. Clin Exp Pharmacol Physiol 2010; 38:63-70. [DOI: 10.1111/j.1440-1681.2010.05464.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
34
|
Abstract
Infarct size is determined not only by the severity of ischemia but also by pathological processes initiated at reperfusion. Accumulating experimental evidence indicates that lethal reperfusion injury might account for up to half of the final size of the myocardial infarct. Ischemic postconditioning (brief repeated periods of ischemia-reperfusion applied at the onset of coronary reflow) has been recently described as a powerful cardioprotection mechanism that prevents lethal reperfusion injury. This is the first method proven to reduce the final infarct size by about 50% in several in vivo models and to be confirmed in recent preliminary human studies. The molecular pathways are incompletely mapped but they probably converge to a mitochondrial key target: the mitochondrial permeability transition pore (PTP) which opening during early reperfusion is an event that promotes myocardial cell death. In different animal models and experimental settings, pharmacological PTP inhibition at the onset of reperfusion reproduces all the cardioprotective effects of ischemic postconditioning. In a recent proof-of-concept trial, the administration (just before percutaneous coronary intervention) of cyclosporine A, a potent PTP inhibitor, was associated with smaller infarct size. This review will focus on the physiological preclinical data on both ischemic and pharmacological postconditioning that are relevant to their translation to clinical therapeutics.
Collapse
|
35
|
Bell RM, Yellon DM. There is More to Life than Revascularization: Therapeutic Targeting of Myocardial Ischemia/Reperfusion Injury. Cardiovasc Ther 2010; 29:e67-79. [DOI: 10.1111/j.1755-5922.2010.00190.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
|
36
|
Morkuniene R, Arandarcikaite O, Ivanoviene L, Borutaite V. Estradiol-induced protection against ischemia-induced heart mitochondrial damage and caspase activation is mediated by protein kinase G. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2010; 1797:1012-7. [DOI: 10.1016/j.bbabio.2010.03.027] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2009] [Revised: 03/28/2010] [Accepted: 03/29/2010] [Indexed: 10/19/2022]
|
37
|
Ovize M, Baxter GF, Di Lisa F, Ferdinandy P, Garcia-Dorado D, Hausenloy DJ, Heusch G, Vinten-Johansen J, Yellon DM, Schulz R. Postconditioning and protection from reperfusion injury: where do we stand? Position paper from the Working Group of Cellular Biology of the Heart of the European Society of Cardiology. Cardiovasc Res 2010; 87:406-23. [PMID: 20448097 DOI: 10.1093/cvr/cvq129] [Citation(s) in RCA: 430] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Ischaemic postconditioning (brief periods of ischaemia alternating with brief periods of reflow applied at the onset of reperfusion following sustained ischaemia) effectively reduces myocardial infarct size in all species tested so far, including humans. Ischaemic postconditioning is a simple and safe manoeuvre, but because reperfusion injury is initiated within minutes of reflow, postconditioning must be applied at the onset of reperfusion. The mechanisms of protection by postconditioning include: formation and release of several autacoids and cytokines; maintained acidosis during early reperfusion; activation of protein kinases; preservation of mitochondrial function, most strikingly the attenuation of opening of the mitochondrial permeability transition pore (MPTP). Exogenous recruitment of some of the identified signalling steps can induce cardioprotection when applied at the time of reperfusion in animal experiments, but more recently cardioprotection was also observed in a proof-of-concept clinical trial. Indeed, studies in patients with an acute myocardial infarction showed a reduction of infarct size and improved left ventricular function when they underwent ischaemic postconditioning or pharmacological inhibition of MPTP opening during interventional reperfusion. Further animal studies and large-scale human studies are needed to determine whether patients with different co-morbidities and co-medications respond equally to protection by postconditioning. Also, our understanding of the underlying mechanisms must be improved to develop new therapeutic strategies to be applied at reperfusion with the ultimate aim of limiting the burden of ischaemic heart disease and potentially providing protection for other organs at risk of reperfusion injury, such as brain and kidney.
Collapse
Affiliation(s)
- Michel Ovize
- Service d'Explorations Fonctionnelles Cardiovasculaires and Inserm U886, Hospices Civils de Lyon, University of Lyon, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Inhibition of mitochondrial permeability transition pore opening: the Holy Grail of cardioprotection. Basic Res Cardiol 2010; 105:151-4. [PMID: 20066536 DOI: 10.1007/s00395-009-0080-9] [Citation(s) in RCA: 237] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
39
|
Attenuation of myocardial injury by postconditioning: role of hypoxia inducible factor-1alpha. Basic Res Cardiol 2009; 105:109-18. [PMID: 19597757 DOI: 10.1007/s00395-009-0044-0] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2009] [Revised: 06/19/2009] [Accepted: 06/26/2009] [Indexed: 01/12/2023]
Abstract
Postconditioning (PostC) has regenerated interest as a mechanical intervention against myocardial ischemia/reperfusion injury, but its molecular mechanisms remain elusive. This study tested the hypothesis that hypoxia inducible factor-1alpha (HIF-1alpha) plays a role in PostC-induced cardioprotection. Male Wistar rats were subjected to 30 min ischemia followed by 3 h of reperfusion (Control). PostC with 3 cycles of 10 s reperfusion and 10 s re-occlusion was applied at the onset of reperfusion. Relative to the Sham group, HIF-1alpha protein level was increased by 2.9-fold in the Control group, but its level was enhanced by 5.8-fold with PostC (P < 0.01 vs. Control). However, HIF-1alpha protein level was further augmented by 2.0-fold and 3.3-fold, respectively, when the prolyl hydroxylase inhibitor, dimethyloxalylglycine (DMOG, 40 mg/kg, i.p.) was given at 24 h before ischemia in both Control and PostC groups. PostC reduced infarct size by 24% compared with the Control (27 +/- 4.2% vs. 36 +/- 5.2%, P < 0.01), consistent with significant lower levels of plasma creatine kinase activity, index of cardiomyocyte apoptosis and caspase-3 activity. Although pretreatment with DMOG significantly reduced infarct size relative to the Control, the infarct-sparing effect of PostC was remarkably enhanced when DMOG was given before PostC (18 +/- 2.0% vs. 27 +/- 4.2% in PostC alone, P < 0.05). There was a significant linear inverse relationship between HIF-1alpha protein level and infarct size (r = -0.799, P < 0.01) among all groups. Furthermore, along with up-regulated HIF-1alpha expression, the levels of iNOS mRNA and protein were significantly increased in the PostC alone and DMOG plus PostC groups. In conclusion, these data suggest that HIF-1alpha is involved in cardioprotection by PostC and pharmacological augmentation of HIF-1alpha expression that enhances the infarct-sparing effect of PostC; iNOS, the downstream gene of HIF-1alpha, may participate in signaling pathways in mediating PostC's protection.
Collapse
|
40
|
Skyschally A, van Caster P, Iliodromitis EK, Schulz R, Kremastinos DT, Heusch G. Ischemic postconditioning: experimental models and protocol algorithms. Basic Res Cardiol 2009; 104:469-83. [PMID: 19543787 DOI: 10.1007/s00395-009-0040-4] [Citation(s) in RCA: 157] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2009] [Revised: 04/30/2009] [Accepted: 05/27/2009] [Indexed: 02/07/2023]
Abstract
Ischemic postconditioning, a simple mechanical maneuver at the onset of reperfusion, reduces infarct size after ischemia/reperfusion. After its first description in 2003 by Zhao et al. numerous experimental studies have investigated this protective phenomenon. Whereas the underlying mechanisms and signal transduction are not yet understood in detail, infarct size reduction by ischemic postconditioning was confirmed in all species tested so far, including man. We have now reviewed the literature with focus on experimental models and protocols to better understand the determinants of protection by ischemic postconditioning or lack of it. Only studies with infarct size as unequivocal endpoint were considered. In all species and models, the duration of index ischemia and the protective protocol algorithm impact on the outcome of ischemic postconditioning, and gender, age, and myocardial temperature contribute.
Collapse
Affiliation(s)
- Andreas Skyschally
- Institut für Pathophysiologie, Universitätsklinikum Essen, Essen, Germany.
| | | | | | | | | | | |
Collapse
|
41
|
Granfeldt A, Lefer DJ, Vinten-Johansen J. Protective ischaemia in patients: preconditioning and postconditioning. Cardiovasc Res 2009; 83:234-46. [PMID: 19398470 DOI: 10.1093/cvr/cvp129] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Infarct size can be limited by reducing the determinants of infarct size or increasing collateral blood flow by treatment initiated before the ischaemic event. Reperfusion is the definitive treatment for permanently reducing infarct size and restoring some degree of contractile function to the affected myocardium. Innate survival mechanisms in the heart can be stimulated by short, non-lethal periods of ischaemia and reperfusion, applied either before or after the ischaemic event. Preconditioning, a series of transient intervals of ischaemia and reperfusion applied before the lethal 'index' ischaemic event, sets in motion molecular and cellular mechanisms that increase cardiomyocyte survival to a degree that had not hitherto been seen before. The cardioprotective ischaemic-reperfusion protocol applied at onset of reperfusion, termed 'postconditioning' (Postcon), is also associated with significant cardioprotection that can be applied at the point of reperfusion treatment in the catheterization laboratory or operating room. Both preconditioning and Postcon have been successfully applied to the clinical setting and have been found to reduce infarct size and other attributes of post-ischaemic injury. This review will summarize the physiological preclinical data on preconditioning and Postcon that are relevant to their translation to clinical therapeutics and treatment.
Collapse
Affiliation(s)
- Asger Granfeldt
- Department of Anesthesiology and Critical Care, Aarhus University, Aarhus, Denmark
| | | | | |
Collapse
|
42
|
Penna C, Perrelli MG, Raimondo S, Tullio F, Merlino A, Moro F, Geuna S, Mancardi D, Pagliaro P. Postconditioning induces an anti-apoptotic effect and preserves mitochondrial integrity in isolated rat hearts. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2009; 1787:794-801. [PMID: 19328770 DOI: 10.1016/j.bbabio.2009.03.013] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2008] [Revised: 03/16/2009] [Accepted: 03/18/2009] [Indexed: 01/01/2023]
Abstract
Postconditioning (PostC) may limit mitochondrial damage and apoptotic signaling. We studied markers of apoptosis and mitochondrial protection in isolated rat hearts, which underwent a) perfusion without ischemia (Sham), b) 30-min ischemia (I) plus 2-hour reperfusion (R), or c) PostC protocol (5 intermittent cycles of 10-s reperfusion and 10-s ischemia immediately after the 30-min ischemia). Markers were studied in cytosolic (CF) and/or mitochondrial (MF) fractions. In CF, while pro-apoptotic factors (cytochrome c and caspase-3) were reduced, the anti-apoptotic markers (Bcl-2 and Pim-1) were increased by PostC, compared to the I/R group. Accordingly, phospho-GSK-3beta and Bcl-2 levels increased in mitochondria of PostC group. Moreover, I/R reduced the level of mitochondrial structural protein (HSP-60) in MF and increased in CF, thus suggesting mitochondrial damage and HSP-60 release in cytosol, which were prevented by PostC. Electron microscopy confirmed that I/R markedly damaged cristae and mitochondrial membranes; damage was markedly reduced by PostC. Finally, total connexin-43 (Cx43) levels were reduced in the CF of the I/R group, whereas phospho-Cx43 level resulted in higher levels in the MF of the I/R group than the Sham group. PostC limited the I/R-induced increase of mitochondrial phospho-Cx43. Data suggest that PostC i) increases the levels of anti-apoptotic markers, including the cardioprotective kinase Pim-1, ii) decreases the pro-apoptotic markers, e.g. cytochrome c, iii) preserves the mitochondrial structure, and iv) limits the migration of phospho-Cx43 to mitochondria.
Collapse
Affiliation(s)
- Claudia Penna
- Department of Clinical and Biological Sciences, University of Turin, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Hausenloy DJ, Ong SB, Yellon DM. The mitochondrial permeability transition pore as a target for preconditioning and postconditioning. Basic Res Cardiol 2009; 104:189-202. [PMID: 19242644 DOI: 10.1007/s00395-009-0010-x] [Citation(s) in RCA: 209] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2009] [Revised: 01/25/2009] [Accepted: 01/27/2009] [Indexed: 12/19/2022]
Abstract
The experimental evidence supporting the mitochondrial permeability transition pore (mPTP) as a major mediator of lethal myocardial reperfusion injury and therefore a critical target for cardioprotection is persuasive. Although, its molecular identity eludes investigators, it is generally accepted that mitochondrial cyclophilin-D, the target for the inhibitory effects of cyclosporine-A on the mPTP, is a regulatory component of the mPTP. Animal myocardial infarction studies and a recent clinical proof-of-concept study have demonstrated that pharmacologically inhibiting its opening at the onset of myocardial reperfusion reduces myocardial infarct size in the region of 30-50%. Interestingly, the inhibition of mPTP opening at this time appears to underpin the infarct-limiting effects of the endogenous cardioprotective strategies of ischemic preconditioning (IPC) and postconditioning (IPost). However, the mechanism underlying this inhibitory action of IPC and IPost on mPTP opening is unclear. The objectve of this review article will be to explore the potential mechanisms which link IPC and IPost to mPTP inhibition in the reperfused heart.
Collapse
Affiliation(s)
- Derek J Hausenloy
- The Hatter Institute and Center for Cardiology, University College London Hospitals and Medical School, Grafton Way, London, UK.
| | | | | |
Collapse
|