1
|
Bagheri SM, Allahtavakoli M, Hakimizadeh E. Neuroprotective effect of ischemic postconditioning against hyperperfusion and its mechanisms of neuroprotection. JOURNAL OF RESEARCH IN MEDICAL SCIENCES : THE OFFICIAL JOURNAL OF ISFAHAN UNIVERSITY OF MEDICAL SCIENCES 2024; 29:31. [PMID: 39239075 PMCID: PMC11376715 DOI: 10.4103/jrms.jrms_341_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 05/13/2023] [Accepted: 06/27/2023] [Indexed: 09/07/2024]
Abstract
Background In recent years, stroke and ischemia-reperfusion injury has motivated researchers to find new ways to reduce the complications. Although reperfusion is essential for brain survival, it is like a double-edged sword that may cause further damage to the brain. Ischemic postconditioning (IPostC) refers to the control of blood flow in postischemia-reperfusion that can reduce ischemia-reperfusion injuries. Materials and Methods Articles were collected by searching for the terms: Ischemic postconditioning and neuroprotective and ischemic postconditioning and hyperperfusion. Suitable articles were collected from electronic databases, including ISI Web of Knowledge, Medline/PubMed, ScienceDirect, Embase, Scopus, Biological Abstract, Chemical Abstract, and Google Scholar. Results New investigations show that IPostC has protection against hyperperfusion by reducing the amount of blood flow during reperfusion and thus reducing infarction volume, preventing the blood-brain barrier damage, and reducing the rate of apoptosis through the activation of innate protective systems. Numerous mechanisms have been suggested for IPostC, which include reduction of free radical production, apoptosis, inflammatory factors, and activation of endogenous protective pathways. Conclusion It seems that postconditioning can prevent damage to the brain by reducing the flow and blood pressure caused by hyperperfusion. It can protect the brain against damages such as stroke and hyperperfusion by activating various endogenous protection systems. In the present review article, we tried to evaluate both useful aspects of IPostC, neuroprotective effects, and fight against hyperperfusion.
Collapse
Affiliation(s)
- Seyyed Majid Bagheri
- Physiology-Pharmacology Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Mohammad Allahtavakoli
- Physiology-Pharmacology Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Department of Physiology and Pharmacology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Elham Hakimizadeh
- Physiology-Pharmacology Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| |
Collapse
|
2
|
Neuroprotective Effect of Piclamilast-Induced Post-Ischemia Pharmacological Treatment in Mice. Neurochem Res 2022; 47:2230-2243. [PMID: 35482135 DOI: 10.1007/s11064-022-03609-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 04/13/2022] [Accepted: 04/13/2022] [Indexed: 01/10/2023]
Abstract
Various studies have evidenced the neuroprotective role of PDE4 inhibitors. However, whether PDE4 inhibitor, Piclamilast pharmacological post-treatment is protective during cerebral ischemia reperfusion-induced injury remains unknown. Therefore, this study design included testing the hypothesis that Piclamilast administered at the beginning of a reperfusion phase (Piclamilast pPost-trt) shows protective effects and explores & probes underlying downstream mechanisms. Swiss albino male mice were subjected to global ischemic and reperfusion injury for 17 min. The animals examined cerebral infarct size, biochemical parameters, inflammatory mediators, and motor coordination. For memory, assessment mice were subjected to morris water maze (MWM) and elevated plus maze (EPM) test. Histological changes were assessed using HE staining. Piclamilast pPost-trt significantly reduced I/R injury-induced deleterious effects on biochemical parameters of oxidative stress, inflammatory parameters, infarct size, and histopathological changes, according to the findings. These neuroprotective effects of pPost-trt are significantly abolished by pre-treatment with selective CREB inhibitor, 666-15. Current study concluded that induced neuroprotective benefits of Piclamilast Post-trt, in all probability, maybe mediated through CREB activation. Hence, its neuroprotective effects can be further explored in clinical settings.
Collapse
|
3
|
Ye R, Jneid H, Alam M, Uretsky BF, Atar D, Kitakaze M, Davidson SM, Yellon DM, Birnbaum Y. Do We Really Need Aspirin Loading for STEMI? Cardiovasc Drugs Ther 2022; 36:1221-1238. [PMID: 35171384 DOI: 10.1007/s10557-022-07327-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/11/2022] [Indexed: 12/12/2022]
Abstract
Aspirin loading (chewable or intravenous) as soon as possible after presentation is a class I recommendation by current ST elevation myocardial infarction (STEMI) guidelines. Earlier achievement of therapeutic antiplatelet effects by aspirin loading has long been considered the standard of care. However, the effects of the loading dose of aspirin (alone or in addition to a chronic maintenance oral dose) have not been studied. A large proportion of myocardial cell death occurs upon and after reperfusion (reperfusion injury). Numerous agents and interventions have been shown to limit infarct size in animal models when administered before or immediately after reperfusion. However, these interventions have predominantly failed to show significant protection in clinical studies. In the current review, we raise the hypothesis that aspirin loading may be the culprit. Data obtained from animal models consistently show that statins, ticagrelor, opiates, and ischemic postconditioning limit myocardial infarct size. In most of these studies, aspirin was not administered. However, when aspirin was administered before reperfusion (as is the case in the majority of studies enrolling STEMI patients), the protective effects of statin, ticagrelor, morphine, and ischemic postconditioning were attenuated, which can be plausibly attributable to aspirin loading. We therefore suggest studying the effects of aspirin loading before reperfusion on the infarct size limiting effects of statins, ticagrelor, morphine, and/ or postconditioning in large animal models using long reperfusion periods (at least 24 h). If indeed aspirin attenuates the protective effects, clinical trials should be conducted comparing aspirin loading to alternative antiplatelet regimens without aspirin loading in patients with STEMI undergoing primary percutaneous coronary intervention.
Collapse
Affiliation(s)
- Regina Ye
- University of Texas at Austin, Austin, TX, USA
| | - Hani Jneid
- Department of Medicine Baylor College of Medicine, 7200 Cambridge Street Houston, Texas, 77030, USA
| | - Mahboob Alam
- Department of Medicine Baylor College of Medicine, 7200 Cambridge Street Houston, Texas, 77030, USA
| | - Barry F Uretsky
- University of Arkansas for Medical Sciences, Central Arkansas Veterans Health System, Little Rock, AR, USA
| | - Dan Atar
- Department of Cardiology, Oslo University Hospital Ulleval, Oslo, Norway, and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Masafumi Kitakaze
- Center of Medical Innovation and Translational Research, Department of Medical Data Science, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Sean M Davidson
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London, WC1E 6HX, UK
| | - Derek M Yellon
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London, WC1E 6HX, UK
| | - Yochai Birnbaum
- Department of Medicine Baylor College of Medicine, 7200 Cambridge Street Houston, Texas, 77030, USA.
| |
Collapse
|
4
|
Verhaar N, de Buhr N, von Köckritz-Blickwede M, Hewicker-Trautwein M, Pfarrer C, Mazzuoli-Weber G, Schulte H, Kästner S. Ischaemic postconditioning reduces apoptosis in experimental jejunal ischaemia in horses. BMC Vet Res 2021; 17:175. [PMID: 33902575 PMCID: PMC8077964 DOI: 10.1186/s12917-021-02877-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 04/12/2021] [Indexed: 03/06/2023] Open
Abstract
BACKGROUND Ischaemic postconditioning (IPoC) refers to brief periods of reocclusion of blood supply following an ischaemic event. This has been shown to ameliorate ischaemia reperfusion injury in different tissues, and it may represent a feasible therapeutic strategy for ischaemia reperfusion injury following strangulating small intestinal lesions in horses. The objective of this study was to assess the degree cell death, inflammation, oxidative stress, and heat shock response in an equine experimental jejunal ischaemia model with and without IPoC. METHODS In this randomized, controlled, experimental in vivo study, 14 horses were evenly assigned to a control group and a group subjected to IPoC. Under general anaesthesia, segmental ischaemia with arterial and venous occlusion was induced in 1.5 m jejunum. Following ischaemia, the mesenteric vessels were repeatedly re-occluded in group IPoC only. Full thickness intestinal samples and blood samples were taken at the end of the pre-ischaemia period, after ischaemia, and after 120 min of reperfusion. Immunohistochemical staining or enzymatic assays were performed to determine the selected variables. RESULTS The mucosal cleaved-caspase-3 and TUNEL cell counts were significantly increased after reperfusion in the control group only. The cleaved-caspase-3 cell count was significantly lower in group IPoC after reperfusion compared to the control group. After reperfusion, the tissue myeloperoxidase activity and the calprotectin positive cell counts in the mucosa were increased in both groups, and only group IPoC showed a significant increase in the serosa. Tissue malondialdehyde and superoxide dismutase as well as blood lactate levels showed significant progression during ischaemia or reperfusion. The nuclear immunoreactivity of Heat shock protein-70 increased significantly during reperfusion. None of these variables differed between the groups. The neuronal cell counts in the myenteric plexus ganglia were not affected by the ischaemia model. CONCLUSIONS A reduced apoptotic cell count was found in the group subjected to IPoC. None of the other tested variables were significantly affected by IPoC. Therefore, the clinical relevance and possible protective mechanism of IPoC in equine intestinal ischaemia remains unclear. Further research on the mechanism of action and its effect in clinical cases of strangulating colic is needed.
Collapse
Affiliation(s)
- Nicole Verhaar
- Clinic for Horses, University of Veterinary Medicine Hannover, Hannover, Germany.
| | - Nicole de Buhr
- Department of Biochemistry, University of Veterinary Medicine Hannover, Hannover, Germany
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Maren von Köckritz-Blickwede
- Department of Biochemistry, University of Veterinary Medicine Hannover, Hannover, Germany
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, Hannover, Germany
| | | | - Christiane Pfarrer
- Institute for Anatomy, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Gemma Mazzuoli-Weber
- Institute for Physiology and Cell Biology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Henri Schulte
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hannover, Germany
| | - Sabine Kästner
- Clinic for Horses, University of Veterinary Medicine Hannover, Hannover, Germany
- Small Animal Clinic, University of Veterinary Medicine Hannover, Hannover, Germany
| |
Collapse
|
5
|
Taha HSED, Shaker MM. Percutaneous management of reperfusion arrhythmias during primary percutaneous coronary intervention: a case report. Egypt Heart J 2021; 73:30. [PMID: 33765245 PMCID: PMC7994471 DOI: 10.1186/s43044-021-00158-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 03/16/2021] [Indexed: 12/02/2022] Open
Abstract
Background Myocardial reperfusion may cause profound electrophysiological alterations and can lead to serious reperfusion arrhythmias (RA). Management of RA and the accompanying electrical storm that may occur remains a problem. To our knowledge, the role of balloon re-inflation of the infarct-related artery (IRA) has never been addressed as a treatment modality for RA presenting as ventricular tachycardia (VT) with pulse or supraventricular tachycardia (SVT). Case presentation Six patients presenting with ST elevation myocardial infarction (STEMI) in the first 12 h, who underwent successful primary percutaneous coronary intervention (PCI), developed RA in the cathlab after restoration of flow in the IRA. The RA was in the form of VT with pulse, except in one patient who had SVT. In four patients, the RA was associated with hemodynamic instability. The mean age of the studied patients was 59.16 ± 7.94 years, and four were males. Coronary artery disease risk factors were prevalent, with four patients being hypertensive, two dyslipidemic, one diabetic, and 2 current smokers. One patient had a history of prior myocardial infarction (MI), and none had a history of congestive heart failure. The coronary angiography showed 100% occlusion of IRA in all patients and 2–3-vessel disease was present in 50%. PCI was successful with restoration of thrombolysis in myocardial infarction (TIMI) 2–3 flow in IRA in all cases. The mean time to revascularization from the onset of chest pain was 4.88 ± 2.68 h. In all cases, balloon re-inflation was successful in terminating the arrhythmias. None of the patients needed direct current cardioversion or anti-arrhythmic drugs for management of the acute arrhythmia. Conclusion Balloon re-inflation of IRA was successful in terminating RA that develop in the form of VT with pulse or SVT.
Collapse
Affiliation(s)
- Hesham Salah El Din Taha
- Department of Cardiology, Faculty of Medicine, Cairo University, 27 Nafezet Sheem El Shafae St Kasr Al Ainy, Cairo, 11562, Egypt.
| | - Mirna Mamdouh Shaker
- Department of Cardiology, Faculty of Medicine, Cairo University, 27 Nafezet Sheem El Shafae St Kasr Al Ainy, Cairo, 11562, Egypt
| |
Collapse
|
6
|
Guerrero-Orriach JL, Malo-Manso A, Ramirez-Aliaga M, Florez Vela AI, Galán-Ortega M, Moreno-Cortes I, Gonzalez-Almendros I, Ramirez-Fernandez A, Ariza-Villanueva D, Escalona-Belmonte JJ, Quesada-Muñoz G, Sepúlveda-Haro E, Romero-Molina S, Bellido-Estevez I, Gomez-Luque A, Rubio-Navarro M, Alcaide-Torres J, Santiago-Fernandez C, Garrido-Sanchez L, Cruz-Mañas J. Renal and Neurologic Benefit of Levosimendan vs Dobutamine in Patients With Low Cardiac Output Syndrome After Cardiac Surgery: Clinical Trial FIM-BGC-2014-01. Front Pharmacol 2020; 11:1331. [PMID: 32982742 PMCID: PMC7479222 DOI: 10.3389/fphar.2020.01331] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 08/10/2020] [Indexed: 12/19/2022] Open
Abstract
Background Low-cardiac output syndrome (LCOS) after cardiac surgery secondary to systemic hypoperfusion is associated with a higher incidence of renal and neurological damage. A range of effective therapies are available for LCOS. The beneficial systemic effects of levosimendan persist even after cardiac output is restored, which suggests an independent cardioprotective effect. Methods A double-blind clinical trial was conducted in patients with a confirmed diagnosis of LCOS randomized into two treatment groups (levosimendan vs. dobutamine). Monitoring of hemodynamic (cardiac index, systolic volume index, heart rate, mean arterial pressure, central venous pressure, central venous saturation); biochemical (e.g. creatinine, S100B protein, NT-proBNP, troponin I); and renal parameters was performed using acute kidney injury scale (AKI scale) and renal and brain ultrasound measurements [vascular resistance index (VRI)] at diagnosis and during the first 48 h. Results Significant differences were observed between groups in terms of cardiac index, systolic volume index, NT-proBNP, and kidney injury stage at diagnosis. In the levosimendan group, there were significant variations in AKI stage after 24 and 48 h. No significant differences were observed in the other parameters studied. Conclusion Levosimendan showed a beneficial effect on renal function in LCOS patients after cardiac surgery that was independent from cardiac output and vascular tone. This effect is probably achieved by pharmacological postconditioning. Clinical Trial Registration EUDRA CT, identifier 2014-001461-27. https://www.clinicaltrialsregister.eu/ctr-search/search?query=2014-001461-27.
Collapse
Affiliation(s)
- Jose L Guerrero-Orriach
- Institute of Biomedical Research in Malaga [IBIMA], Malaga, Spain.,Department of Anaesthesiology, Virgen de la Victoria University Hospital, Malaga, Spain.,Department of Pharmacology and Pediatrics, School of Medicine, University of Malaga, Malaga, Spain
| | - Alfredo Malo-Manso
- Institute of Biomedical Research in Malaga [IBIMA], Malaga, Spain.,Department of Anaesthesiology, Virgen de la Victoria University Hospital, Malaga, Spain
| | - Marta Ramirez-Aliaga
- Institute of Biomedical Research in Malaga [IBIMA], Malaga, Spain.,Department of Anaesthesiology, Virgen de la Victoria University Hospital, Malaga, Spain
| | | | - Manuel Galán-Ortega
- Institute of Biomedical Research in Malaga [IBIMA], Malaga, Spain.,Department of Anaesthesiology, Virgen de la Victoria University Hospital, Malaga, Spain
| | - Isabel Moreno-Cortes
- Department of Anaesthesiology, Virgen de la Victoria University Hospital, Malaga, Spain
| | | | - Alicia Ramirez-Fernandez
- Institute of Biomedical Research in Malaga [IBIMA], Malaga, Spain.,Department of Anaesthesiology, Virgen de la Victoria University Hospital, Malaga, Spain
| | | | - Juan Jose Escalona-Belmonte
- Institute of Biomedical Research in Malaga [IBIMA], Malaga, Spain.,Department of Anaesthesiology, Virgen de la Victoria University Hospital, Malaga, Spain
| | - Guillermo Quesada-Muñoz
- Institute of Biomedical Research in Malaga [IBIMA], Malaga, Spain.,Department of Anaesthesiology, Virgen de la Victoria University Hospital, Malaga, Spain
| | | | | | - Inmaculada Bellido-Estevez
- Institute of Biomedical Research in Malaga [IBIMA], Malaga, Spain.,Department of Pharmacology and Pediatrics, School of Medicine, University of Malaga, Malaga, Spain
| | - Aurelio Gomez-Luque
- Institute of Biomedical Research in Malaga [IBIMA], Malaga, Spain.,Department of Anaesthesiology, Virgen de la Victoria University Hospital, Malaga, Spain.,Department of Pharmacology and Pediatrics, School of Medicine, University of Malaga, Malaga, Spain
| | - Manuel Rubio-Navarro
- Department of Anaesthesiology, Virgen de la Victoria University Hospital, Malaga, Spain
| | - Juan Alcaide-Torres
- Institute of Biomedical Research in Malaga [IBIMA], Malaga, Spain.,Unidad de Gestión Clínica de Endocrinología y Nutrición, Virgen de la Victoria University Hospital, Málaga, Spain
| | - Concepcion Santiago-Fernandez
- Institute of Biomedical Research in Malaga [IBIMA], Malaga, Spain.,Unidad de Gestión Clínica de Endocrinología y Nutrición, Virgen de la Victoria University Hospital, Málaga, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición-CIBEROBN, Instituto de Salud Carlos III, Málaga, Spain
| | - Lourdes Garrido-Sanchez
- Institute of Biomedical Research in Malaga [IBIMA], Malaga, Spain.,Unidad de Gestión Clínica de Endocrinología y Nutrición, Virgen de la Victoria University Hospital, Málaga, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición-CIBEROBN, Instituto de Salud Carlos III, Málaga, Spain
| | - Jose Cruz-Mañas
- Department of Anaesthesiology, Virgen de la Victoria University Hospital, Malaga, Spain
| |
Collapse
|
7
|
Remote Ischemic Postconditioning Inhibits Hippocampal Neuronal Apoptosis and Mitophagy After Cardiopulmonary Resuscitation in Rats. Shock 2020; 55:74-82. [PMID: 32590695 DOI: 10.1097/shk.0000000000001596] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
BACKGROUND Studies have shown that remote ischemic post-conditioning can improve brain damage caused by ischemia and hypoxia. However, the specific mechanism underlying this phenomenon is still unclear. The purpose of this study was to investigate the effects of remote ischemic post-conditioning on neuronal apoptosis and mitophagy after cardiopulmonary resuscitation (CPR) in rats. METHODS Male Sprague-Dawley rats were used to establish an asphyxia cardiac arrest model by clamping the tracheal duct. First, the expression levels of P53, Cytochrome c (Cytc), and Parkin in the cytoplasm and mitochondria were observed at 3, 6, 24, and 72 h after the restoration of spontaneous circulation (ROSC). Then neurological deficit scores, hippocampal neuron apoptosis, mitochondrial P53 and Parkin, cytoplasmic Cytc, and neuron ultrastructure were evaluated 24 h after ROSC. RESULTS P53 and Parkin can translocate from the cytoplasm to the mitochondria, promoting the translocation of cytoplasmic Cytc to mitochondria after CPR, reaching a peak at 24 h after the ROSC. The P53 inhibitor Pifithrin-μ reduced apoptosis induced by P53 mitochondrial translocation. Apoptosis was induced after cardiac arrest and attenuated by remote ischemic postconditioning via inhibiting P53 mitochondrial translocation and the release of Cytc to the cytoplasm. In addition, remote ischemic postconditioning could inhibit Parkin-mediated mitophagy. CONCLUSION Taken together, our results show that remote ischemic post-conditioning improves neural function after CPR by inhibiting P53 mitochondrial translocation-induced apoptosis and Parkin-mediated mitophagy.
Collapse
|
8
|
Wang Z, Wen J, Zhou C, Wang Z, Wei M. Gene expression profiling analysis to investigate the role of remote ischemic postconditioning in ischemia-reperfusion injury in rats. BMC Genomics 2019; 20:361. [PMID: 31072368 PMCID: PMC6509872 DOI: 10.1186/s12864-019-5743-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 04/29/2019] [Indexed: 12/23/2022] Open
Abstract
Background Blood flow restoration is a definitive therapy for salvaging the myocardium following ischemic injury. Nevertheless, the sudden restoration of blood flow to the ischemic myocardium can induce ischemia-reperfusion injury (IRI). Results Herein, we investigated the cardioprotective effect of remote ischemic postconditioning (RPostC) through our in vivo rat model of myocardial IRI. The study included three groups: the control group, the IRI group, and the IRI + RPostC group. Ischemia-reperfusion treatment led to an increase in the myocardial infarction area, which was inhibited by RPostC. In contrast to that in the control group, the myocardial apoptosis level was enhanced in the IRI group, whereas RPostC treatment decreased IRI-induced cellular apoptosis. Affymetrix Rat Gene 2.0 ST chip data identified a total of 265 upregulated genes and 267 downregulated genes between the IRI and IRI + RPostC groups. A group of differentially expressed noncoding RNAs (ncRNAs), such as MTA_TC0600002772.mm, MTA_TC1300002394.mm, U7 small nuclear RNA (Rnu7) and RGD7543256_1, were identified. Gene Ontology (GO) enrichment analysis indicated that the positive regulation of some molecular functions, such as GTPase activity, GTP binding, cyclic-nucleotide phosphodiesterase activity and cytokine activity, may contribute to the cardioprotective role of RPostC. Moreover, pathway enrichment analysis using the Kyoto Encyclopedia of Genes and Genomes (KEGG) suggested the potential implication of the TNF signaling pathway and Toll-like receptor signaling pathway. Global signal transduction network analysis, co-expression network analysis and quantitative real-time polymerase chain reaction analysis further identified several core genes, including Pdgfra, Stat1, Lifr and Stfa3. Conclusion Remote ischemic postconditioning treatment can decrease IRI-mediated myocardial apoptosis by regulating multiple processes and pathways, such as GTPase activity, cytokine activity, and the TNF and Toll-like receptor signaling pathways. The potential role of the above ncRNAs and core genes in IRI-induced cardiac damage merits further study as well. Electronic supplementary material The online version of this article (10.1186/s12864-019-5743-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Zanxin Wang
- Department of Cardiac Surgery, Fuwai Hospital Chinese Academy of Medical Sciences Shenzhen, 12 Langshan Road, Nanshan District, Shenzhen, 518057, Guangdong Province, People's Republic of China. .,Department of Cardiac Surgery, Shenzhen Sun Yat-sen Cardiovascular Hospital, Shenzhen, People's Republic of China. .,Department of Cardiovascular Surgery, Tianjin Medical University General Hospital, Tianjin, People's Republic of China.
| | - Junmin Wen
- Department of Intensive Care, Fuwai Hospital Chinese Academy of Medical Sciences Shenzhen, Shenzhen, Guangdong, People's Republic of China.,Department of Intensive Care, Shenzhen Sun Yat-sen Cardiovascular Hospital, Shenzhen, People's Republic of China
| | - Chuzhi Zhou
- Department of Intensive Care, Fuwai Hospital Chinese Academy of Medical Sciences Shenzhen, Shenzhen, Guangdong, People's Republic of China.,Department of Intensive Care, Shenzhen Sun Yat-sen Cardiovascular Hospital, Shenzhen, People's Republic of China
| | - Zhiwei Wang
- Department of Cardiac Surgery, Fuwai Hospital Chinese Academy of Medical Sciences Shenzhen, 12 Langshan Road, Nanshan District, Shenzhen, 518057, Guangdong Province, People's Republic of China.,Department of Cardiac Surgery, Shenzhen Sun Yat-sen Cardiovascular Hospital, Shenzhen, People's Republic of China
| | - Minxin Wei
- Department of Cardiac Surgery, Fuwai Hospital Chinese Academy of Medical Sciences Shenzhen, 12 Langshan Road, Nanshan District, Shenzhen, 518057, Guangdong Province, People's Republic of China. .,Department of Cardiac Surgery, Shenzhen Sun Yat-sen Cardiovascular Hospital, Shenzhen, People's Republic of China. .,Department of Cardiovascular Surgery, Tianjin Medical University General Hospital, Tianjin, People's Republic of China.
| |
Collapse
|
9
|
Ding M, Hu L, Yang H, Gao C, Zeng K, Yu M, Feng J, Qiu J, Liu C, Fu F, Li Y. Reduction of SIRT1 blunts the protective effects of ischemic post-conditioning in diabetic mice by impairing the Akt signaling pathway. Biochim Biophys Acta Mol Basis Dis 2019; 1865:1677-1689. [PMID: 30954556 DOI: 10.1016/j.bbadis.2019.04.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 12/20/2018] [Accepted: 01/06/2019] [Indexed: 01/01/2023]
Abstract
Ischemic post-conditioning (IPO) activates Akt signaling to confer cardioprotection. The responsiveness of diabetic hearts to IPO is impaired. We hypothesized that decreased cardiac SIRT1, a positive regulator of Akt, may be responsible for the impaired responsiveness of diabetic hearts to IPO-mediated cardioprotection. High-fat diet and streptozotocin-induced diabetic mice were subjected to myocardial ischemia/reperfusion (MI/R, 30 min ischemia and 180 min reperfusion) or IPO (three cycles of 10 s of reperfusion and ischemia at the onset of reperfusion). Adenoviral vectors encoding GFP or SIRT1 (Ad-SIRT1) were administered by direct injection into the left ventricular. Our results showed that IPO activated the Akt signaling pathway and reduced MI/R injury in non-diabetic hearts but not in diabetic hearts, in which reduced expression of SIRT1 and increased Akt acetylation were observed. Delivery of Ad-SIRT1 into the diabetic hearts reduced Akt acetylation and restored the cardioprotective effects of IPO by modulating Akt signaling pathway. In contrast, cardiac-specific SIRT1 knockout increased Akt acetylation and blunted the cardioprotective effects of IPO. In in vitro study, transfection with wild-type SIRT1 but not inactive mutant SIRT1 reduced the expression of Akt acetylation and restored the protective effects of hypoxic post-conditioning in high glucose-incubated cardiomyocytes. Moreover, the cardiomyocytes transfected with constitutive Akt acetylation showed repressed Akt phosphorylation and blunted protective effects against hypoxia/reoxygenation injury. These findings demonstrate that the reduction of SIRT1 blunts the protective effects of IPO by impairing Akt signaling pathway and that SIRT1 up-regulation restores IPO-mediated cardioprotection in diabetic mice via deacetylation-dependent activation of Akt signaling pathway.
Collapse
Affiliation(s)
- Mingge Ding
- Department of Cardiology, Xi'an Central Hospital, Xi'an Jiaotong University, China; Department of Physiology and Pathophysiology, Fourth Military Medical University, China
| | - Lang Hu
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, China
| | - Hongyan Yang
- School of Aerospace Medicine, Fourth Military Medical University, China
| | - Chao Gao
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, China
| | - Ke Zeng
- Department of Physiology and Pathophysiology, Fourth Military Medical University, China
| | - Mingzhe Yu
- Department of Physiology and Pathophysiology, Fourth Military Medical University, China
| | - Jiahao Feng
- Department of Physiology and Pathophysiology, Fourth Military Medical University, China
| | - Jihuan Qiu
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, China
| | - Chaoyang Liu
- School of Life Sciences, Northwest University, Xi'an 710000, China
| | - Feng Fu
- Department of Physiology and Pathophysiology, Fourth Military Medical University, China.
| | - Yan Li
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, China.
| |
Collapse
|
10
|
Buchholz B, Kelly J, Muñoz M, Bernatené EA, Méndez Diodati N, González Maglio DH, Dominici FP, Gelpi RJ. Vagal stimulation mimics preconditioning and postconditioning of ischemic myocardium in mice by activating different protection mechanisms. Am J Physiol Heart Circ Physiol 2018; 314:H1289-H1297. [PMID: 29631370 DOI: 10.1152/ajpheart.00286.2017] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Vagal stimulation (VS) during myocardial ischemia and reperfusion has beneficial effects. However, it is not known whether short-term VS applied before ischemia or at the onset of reperfusion protects the ischemic myocardium. This study was designed to determine whether short-term VS applied before ischemia or at the onset of reperfusion reduces myocardial infarct size (IS), mimicking classic preconditioning and postconditioning. A second objective was to study the participation of muscarinic and nicotinic receptors in the protection of both preischemic and reperfusion stimulation. FVB mice were subjected to 30 min of regional myocardial ischemia followed by 2 h of reperfusion without VS, with 10-min preischemic VS (pVS), or with VS during the first 10 min of reperfusion (rVS). pVS reduced IS, and this effect was abolished by atropine and wortmannin. rVS also reduced IS in a similar manner, and this effect was abolished by the α7-nicotinic acetylcholine receptor blocker methyllycaconitine. pVS increased Akt and glycogen synthase kinase (GSK)-3β phosphorylation. No changes in Akt and GSK-3β phosphorylation were observed in rVS. Stimulation-mediated IS protection was abolished with the JAK2 blocker AG490. rVS did not modify IL-6 and IL-10 levels in the plasma or myocardium. Splenic denervation and splenectomy did not abolish the protective effect of rVS. In conclusion, pVS and rVS reduced IS by different mechanisms: pVS activated the Akt/GSK-3β muscarinic pathway, whereas rVS activated α7-nicotinic acetylcholine receptors and JAK2, independently of the cholinergic anti-inflammatory pathway. NEW & NOTEWORTHY Our data suggest, for the first time, that vagal stimulation applied briefly either before ischemia or at the beginning of reperfusion mimics classic preconditioning and postconditioning and reduces myocardial infarction, activating different mechanisms. We also infer an important role of α7-nicotinic receptors for myocardial protection independent of the cholinergic anti-inflammatory pathway.
Collapse
Affiliation(s)
- Bruno Buchholz
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Patología, Instituto de Fisiopatología Cardiovascular , Buenos Aires , Argentina.,Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Instituto de Bioquímica y Medicina Molecular, Facultad de Medicina , Buenos Aires , Argentina
| | - Jazmín Kelly
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Patología, Instituto de Fisiopatología Cardiovascular , Buenos Aires , Argentina.,Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Instituto de Bioquímica y Medicina Molecular, Facultad de Medicina , Buenos Aires , Argentina
| | - Marina Muñoz
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Química Biológica, Instituto de Química y Fisicoquímica Biológicas , Buenos Aires , Argentina
| | - Eduardo A Bernatené
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Patología, Instituto de Fisiopatología Cardiovascular , Buenos Aires , Argentina.,Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Instituto de Bioquímica y Medicina Molecular, Facultad de Medicina , Buenos Aires , Argentina
| | - Nahuel Méndez Diodati
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Patología, Instituto de Fisiopatología Cardiovascular , Buenos Aires , Argentina.,Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Instituto de Bioquímica y Medicina Molecular, Facultad de Medicina , Buenos Aires , Argentina
| | - Daniel H González Maglio
- Cátedra de Inmunología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires , Buenos Aires , Argentina.,Instituto de Estudios de la Inmunidad Humoral, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires , Argentina
| | - Fernando P Dominici
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Química Biológica, Instituto de Química y Fisicoquímica Biológicas , Buenos Aires , Argentina
| | - Ricardo J Gelpi
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Patología, Instituto de Fisiopatología Cardiovascular , Buenos Aires , Argentina.,Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Instituto de Bioquímica y Medicina Molecular, Facultad de Medicina , Buenos Aires , Argentina
| |
Collapse
|
11
|
Hu X, Lv T, Yang SF, Zhang XH, Miao YF. Limb remote ischemic post‑conditioning reduces injury and improves long‑term behavioral recovery in rats following subarachnoid hemorrhage: Possible involvement of the autophagic process. Mol Med Rep 2018; 17:21-30. [PMID: 29115588 PMCID: PMC5780133 DOI: 10.3892/mmr.2017.7858] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 03/16/2017] [Indexed: 12/14/2022] Open
Abstract
Hemorrhage‑related neurologic injury is a primary cause of disability and mortality following subarachnoid hemorrhage (SAH). The aim of the present study was to investigate the potential neuroprotective effect and the possible role of autophagy in limb remote ischemic post‑conditioning (RIPostC) using an endovascular puncture rat model of SAH. RIPostC was induced by three cycles of occlusion (10 min) and release (10 min) in the bilateral femoral artery using an aneurysm clip. Early RIPostC began immediately following SAH, delayed RIPostC began following a 30 min delay and the repeated RIPostC group underwent the protocol every day for 3 days. Brain water content, SAH grading, terminal deoxynucleotidyl transferase dUTP nick end labeling‑DAPI staining, transmission electron microscopy, and neurological and behavioral tests were conducted three days following surgery. Long term outcomes of behavior and memory were assessed using a rotarod test and Morris water maze test 1 month subsequently. Biomarkers of autophagy, including Beclin‑1 and light chain 3 (LC3), were assessed using western blotting. The results of the present study demonstrated that, compared with other groups, repeated RIPostC was able to alleviate brain edema, prevent neuronal apoptosis, and improve short term and long term neurological function and memory. Beclin‑1 and LC3 in the cortex were upregulated following treatment with repeated RIPostC. Autolysosomes increased 3 days following SAH and were maintained for 1 month in the repeated RIPostC group. Therefore, the present study indicated that the optimized repeated RIPostC may provide a noninvasive strategy to induce neuroprotection, and improve the short and long term outcomes of SAH‑related cerebral injury, possibly involving the autophagy pathway.
Collapse
Affiliation(s)
- Xiang Hu
- Department of Neurosurgery, AoYoung Hospital, Zhangjiagang, Jiangsu 215617, P.R. China
| | - Tao Lv
- Department of Neurosurgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Shao-Feng Yang
- Department of Neurosurgery, Ren Ji Hospital South Campus, School of Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 201112, P.R. China
| | - Xiao-Hua Zhang
- Department of Neurosurgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Yi-Feng Miao
- Department of Neurosurgery, Ren Ji Hospital South Campus, School of Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 201112, P.R. China
| |
Collapse
|
12
|
Matsuura TR, Bartos JA, Tsangaris A, Shekar KC, Olson MD, Riess ML, Bienengraeber M, Aufderheide TP, Neumar RW, Rees JN, McKnite SH, Dikalova AE, Dikalov SI, Douglas HF, Yannopoulos D. Early Effects of Prolonged Cardiac Arrest and Ischemic Postconditioning during Cardiopulmonary Resuscitation on Cardiac and Brain Mitochondrial Function in Pigs. Resuscitation 2017; 116:8-15. [PMID: 28408349 PMCID: PMC5552370 DOI: 10.1016/j.resuscitation.2017.03.033] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Revised: 03/20/2017] [Accepted: 03/27/2017] [Indexed: 11/21/2022]
Abstract
BACKGROUND Out-of-hospital cardiac arrest (CA) is a prevalent medical crisis resulting in severe injury to the heart and brain and an overall survival of less than 10%. Mitochondrial dysfunction is predicted to be a key determinant of poor outcomes following prolonged CA. However, the onset and severity of mitochondrial dysfunction during CA and cardiopulmonary resuscitation (CPR) is not fully understood. Ischemic postconditioning (IPC), controlled pauses during the initiation of CPR, has been shown to improve cardiac function and neurologically favorable outcomes after 15min of CA. We tested the hypothesis that mitochondrial dysfunction develops during prolonged CA and can be rescued with IPC during CPR (IPC-CPR). METHODS A total of 63 swine were randomized to no ischemia (Naïve), 19min of ventricular fibrillation (VF) CA without CPR (Untreated VF), or 15min of CA with 4min of reperfusion with either standard CPR (S-CPR) or IPC-CPR. Mitochondria were isolated from the heart and brain to quantify respiration, rate of ATP synthesis, and calcium retention capacity (CRC). Reactive oxygen species (ROS) production was quantified from fresh frozen heart and brain tissue. RESULTS Compared to Naïve, Untreated VF induced cardiac and brain ROS overproduction concurrent with decreased mitochondrial respiratory coupling and CRC, as well as decreased cardiac ATP synthesis. Compared to Untreated VF, S-CPR attenuated brain ROS overproduction but had no other effect on mitochondrial function in the heart or brain. Compared to Untreated VF, IPC-CPR improved cardiac mitochondrial respiratory coupling and rate of ATP synthesis, and decreased ROS overproduction in the heart and brain. CONCLUSIONS Fifteen minutes of VF CA results in diminished mitochondrial respiration, ATP synthesis, CRC, and increased ROS production in the heart and brain. IPC-CPR attenuates cardiac mitochondrial dysfunction caused by prolonged VF CA after only 4min of reperfusion, suggesting that IPC-CPR is an effective intervention to reduce cardiac injury. However, reperfusion with both CPR methods had limited effect on mitochondrial function in the brain, emphasizing an important physiological divergence in post-arrest recovery between those two vital organs.
Collapse
Affiliation(s)
- Timothy R Matsuura
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA
| | - Jason A Bartos
- Department of Medicine-Cardiovascular Division, University of Minnesota, Minneapolis, MN, USA
| | - Adamantios Tsangaris
- Department of Medicine-Cardiovascular Division, University of Minnesota, Minneapolis, MN, USA
| | | | - Matthew D Olson
- Department of Medicine-Cardiovascular Division, University of Minnesota, Minneapolis, MN, USA
| | - Matthias L Riess
- Department of Anesthesiology, TVHS VA Medical Center, Nashville, TN, USA; Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Martin Bienengraeber
- Departments of Anesthesiology and Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Tom P Aufderheide
- Department of Emergency Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Robert W Neumar
- Department of Emergency Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Jennifer N Rees
- Department of Medicine-Cardiovascular Division, University of Minnesota, Minneapolis, MN, USA
| | - Scott H McKnite
- Department of Medicine-Cardiovascular Division, University of Minnesota, Minneapolis, MN, USA
| | - Anna E Dikalova
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Sergey I Dikalov
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Hunter F Douglas
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Demetris Yannopoulos
- Department of Medicine-Cardiovascular Division, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
13
|
Feyzizadeh S, Badalzadeh R. Application of ischemic postconditioning's algorithms in tissues protection: response to methodological gaps in preclinical and clinical studies. J Cell Mol Med 2017; 21:2257-2267. [PMID: 28402080 PMCID: PMC5618671 DOI: 10.1111/jcmm.13159] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 02/13/2017] [Indexed: 12/11/2022] Open
Abstract
Ischaemic postconditioning (IPostC) was introduced for the first time by Zhao et al. as a feasible method for reduction of myocardial ischaemia–reperfusion (IR) injury. The cardioprotection by this protocol has been extensively evaluated in various species. Then, further research revealed that IPostC is a safe and convenient approach in limiting IR injury of non‐myocardial tissues such as lung, liver, kidney, intestine, skeletal muscle, brain and spinal cord. IPostC has been conducted with different algorithms, resulting in diverse effects. The possible important factors leading to these differences are the difference in activation levels of signalling pathways and protective mediators by any algorithm, presence or absence of IPostC effectors in each tissue, or intrinsic characteristics of the tissues as well as the methodological biases. Also, the conflicting results have been shown with the application of the same algorithm of IPostC in certain tissues or animal species. The effectiveness of IPostC may depend upon various parameters including the species and the tissues characteristics. For example, different heart rates and metabolic rates of the species and unequal amounts of perfusion and blood flow of the tissues should be considered as the important determinants of IPostC effectiveness and should be thought about in designing IPostC algorithms for future studies. Due to these discrepancies, there is still no optimal single IPostC algorithm applicable to any tissue or any species. This issue is the main topic of the present article.
Collapse
Affiliation(s)
- Saeid Feyzizadeh
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Badalzadeh
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
14
|
Inhibition of endogenous thioredoxin-1 in the heart of transgenic mice does not confer cardioprotection in ischemic postconditioning. Int J Biochem Cell Biol 2016; 81:315-322. [PMID: 27682518 DOI: 10.1016/j.biocel.2016.09.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 09/21/2016] [Accepted: 09/24/2016] [Indexed: 12/29/2022]
Abstract
Thioredoxin-1 maintains the cellular redox status and decreases the infarct size in ischemia/reperfusion injury. However, whether the increase of thioredoxin-1 expression or its lack of activity modifies the protection conferred by ischemic postconditioning has not been yet elucidated. The aim was to evaluate if the thioredoxin-1 overexpression enhances the posctconditioning protective effect, and whether the lack of the activity abolishes the reduction of the infarct size. Wild type mice hearts, transgenic mice hearts overexpressing thioredoxin-1, and a dominant negative mutant (C32S/C35S) of thioredoxin-1 were used. The hearts were subjected to 30min of ischemia and 120min of reperfusion (Langendorff) (I/R group) or to postconditioning protocol (PostC group). The infarct size in the Wt-PostC group decreased in comparison to the Wt-I/R group (54.6±2.4 vs. 39.2±2.1%, p<0.05), but this protection was abolished in DN-Trx1-PostC group (49.7±1.1%). The ischemia/reperfusion and postconditioning in mice overexpressing thioredoxin-1 reduced infarct size at the same magnitude (35.9±2.1 and 38.4±1.3%, p<0.05 vs. Wt-I/R). In Wt-PostC, Trx1-I/R and Trx1- PostC, Akt and GSK3β phosphorylation increased compared to Wt-I/R, without changes in DN-Trx1 groups. In conclusion, given that the cardioprotection conferred by thioredoxin-1 overexpression and postconditioning, is accomplished through the activation of the Akt/GSK3β survival pathway, no synergic effect was evidenced. Thioredoxin-1 plays a key role in the postconditioning, given that when this protein is inactive the cardioprotective mechanism was abolished. Thus, diverse comorbidities or situations modifying the thioredoxin activity, could explain the absence of this strong mechanism of protection in different clinical situations.
Collapse
|
15
|
Epps JA, Smart NA. Remote ischaemic conditioning in the context of type 2 diabetes and neuropathy: the case for repeat application as a novel therapy for lower extremity ulceration. Cardiovasc Diabetol 2016; 15:130. [PMID: 27613524 PMCID: PMC5018170 DOI: 10.1186/s12933-016-0444-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 08/19/2016] [Indexed: 02/07/2023] Open
Abstract
An emerging treatment modality for reducing damage caused by ischaemia–reperfusion injury is ischaemic conditioning. This technique induces short periods of ischaemia that have been found to protect against a more significant ischaemic insult. Remote ischaemic conditioning (RIC) can be administered more conveniently and safely, by inflation of a pneumatic blood pressure cuff to a suprasystolic pressure on a limb. Protection is then transferred to a remote organ via humoral and neural pathways. The diabetic state is particularly vulnerable to ischaemia–reperfusion injury, and ischaemia is a significant cause of many diabetic complications, including the diabetic foot. Despite this, studies utilising ischaemic conditioning and RIC in type 2 diabetes have often been disappointing. A newer strategy, repeat RIC, involves the repeated application of short periods of limb ischaemia over days or weeks. It has been demonstrated that this improves endothelial function, skin microcirculation, and modulates the systemic inflammatory response. Repeat RIC was recently shown to be beneficial for healing in lower extremity diabetic ulcers. This article summarises the mechanisms of RIC, and the impact that type 2 diabetes may have upon these, with the role of neural mechanisms in the context of diabetic neuropathy a focus. Repeat RIC may show more promise than RIC in type 2 diabetes, and its potential mechanisms and applications will also be explored. Considering the high costs, rates of chronicity and serious complications resulting from diabetic lower extremity ulceration, repeat RIC has the potential to be an effective novel advanced therapy for this condition.
Collapse
Affiliation(s)
- J A Epps
- School of Science and Technology, The University of New England, Armidale, NSW, 2351, Australia
| | - N A Smart
- School of Science and Technology, The University of New England, Armidale, NSW, 2351, Australia.
| |
Collapse
|
16
|
Ischemic postconditioning confers cardioprotection and prevents reduction of Trx-1 in young mice, but not in middle-aged and old mice. Mol Cell Biochem 2016; 415:67-76. [PMID: 26932791 DOI: 10.1007/s11010-016-2677-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2015] [Accepted: 02/18/2016] [Indexed: 01/04/2023]
Abstract
Thioredoxin-1 (Trx-1) is part of an antioxidant system that maintains the cell redox homeostasis but their role on ischemic postconditioning (PostC) is unknown. The aim of this work was to determine whether Trx-1 participates in the cardioprotective mechanism of PostC in young, middle-aged, and old mice. Male FVB young (Y: 3 month-old), middle-aged (MA: 12 month-old), and old (O: 20 month-old) mice were used. Langendorff-perfused hearts were subjected to 30 min of ischemia and 120 min of reperfusion (I/R group). After ischemia, we performed 6 cycles of R/I (10 s each) followed by 120 min of reperfusion (PostC group). We measured the infarct size (triphenyltetrazolium); Trx-1, total and phosphorylated Akt, and GSK3β expression (Western blot); and the GSH/GSSG ratio (HPLC). PostC reduced the infarct size in young mice (I/R-Y: 52.3 ± 2.4 vs. PostC-Y: 40.0 ± 1.9, p < 0.05), but this protection was abolished in the middle-aged and old mice groups. Trx-1 expression decreased after I/R, and the PostC prevented the protein degradation in young animals (I/R-Y: 1.05 ± 0.1 vs. PostC-Y: 0.52 ± .0.07, p < 0.05). These changes were accompanied by an improvement in the GSH/GSSG ratio (I/R-Y: 1.25 ± 0.30 vs. PostC-Y: 7.10 ± 2.10, p < 0.05). However, no changes were observed in the middle-aged and old groups. Cytosolic Akt and GSK3β phosphorylation increased in the PostC compared with the I/R group only in young animals. Our results suggest that PostC prevents Trx-1 degradation, decreasing oxidative stress and allowing the activation of Akt and GSK3β to exert its cardioprotective effect. This protection mechanism is not activated in middle-aged and old animals.
Collapse
|
17
|
Jiang X, Guo CX, Zeng XJ, Li HH, Chen BX, Du FH. A soluble receptor for advanced glycation end-products inhibits myocardial apoptosis induced by ischemia/reperfusion via the JAK2/STAT3 pathway. Apoptosis 2016; 20:1033-47. [PMID: 25894538 DOI: 10.1007/s10495-015-1130-4] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
sRAGE can protect cardiomyocytes from apoptosis induced by ischemia/reperfusion (I/R). However, the signaling mechanisms in cardioprotection by sRAGE are currently unknown. We investigated the cardioprotective effect and potential molecular mechanisms of sRAGE inhibition on apoptosis in the mouse myocardial I/R as an in vivo model and neonatal rat cardiomyocyte subjected to ischemic buffer as an in vitro model. Cardiac function and myocardial infarct size following by I/R were evaluated with echocardiography and Evans blue/2,3,5-triphenyltetrazolium chloride. Apoptosis was detected by TUNEL staining and caspase-3 activity. Expression of the apoptosis-related proteins p53, Bax, Bcl-2, JAK2/p-JAK2, STAT3/p-STAT3, AKT/p-AKT, ERK/p-ERK, STAT5A/p-STAT5A and STAT6/p-STAT6 were detected by western blot analysis in the presence and absence of the JAK2 inhibitor AG 490. sRAGE (100 µg/day) improved the heart function in mice with I/R: the left ventricular ejection fraction and fractional shortening were increased by 42 and 57%, respectively; the infarct size was decreased by 52%, the TUNEL-positive myocytes by 66%, and activity of caspase-3 by 24%, the protein expression of p53 and ratio of Bax to Bcl-2 by 29 and 88%, respectively; protein expression of the p-JAK2, p-STAT3 and p-AKT were increased by 92, 280 and 31%, respectively. sRAGE have no effect on protein expression of p-ERK1/2, p-STAT5A and p-STAT6 following by I/R. sRAGE (900 nmol/L) exhibited anti-apoptotic effects in cardiomyocytes by decreasing TUNEL-positive myocytes by 67% and caspase-3 activity by 20%, p53 protein level and the Bax/Bcl-2 ratio by 58 and 86%, respectively; increasing protein expression of the p-JAK2 and p-STAT3 by 26 and 156%, respectively, p-AKT protein level by 33%. The anti-apoptotic effects of sRAGE following I/R were blocked by JAK2 inhibitor AG 490. The effect of sRAGE reduction on TUNEL-positive myocytes and caspase-3 activity were abolished by PI3K inhibitor LY294002, but not ERK 1/2 inhibitor PD98059. These results suggest that sRAGE protects cardiomyocytes from apoptosis induced by I/R in vitro and in vivo by activating the JAK2/STAT3 signaling pathway.
Collapse
Affiliation(s)
- Xue Jiang
- Department of Cardiology, Beijing Tian Tan Hospital, Capital Medical University, 6 Tiantan Xili, Dongcheng District, Beijing, 100050, China
| | | | | | | | | | | |
Collapse
|
18
|
Gabrielová E, Křen V, Jabůrek M, Modrianský M. Silymarin component 2,3-dehydrosilybin attenuates cardiomyocyte damage following hypoxia/reoxygenation by limiting oxidative stress. Physiol Res 2014; 64:79-91. [PMID: 25194130 DOI: 10.33549/physiolres.932703] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Ischemic postconditioning and remote conditioning are potentially useful tools for protecting ischemic myocardium. This study tested the hypothesis that 2,3-dehydrosilybin (DHS), a flavonolignan component of Silybum marianum, could attenuate cardiomyocyte damage following hypoxia/reoxygenation by decreasing the generation of reactive oxygen species (ROS). After 5-6 days of cell culture in normoxic conditions the rat neonatal cardiomyocytes were divided into four groups. Control group (9 h at normoxic conditions), hypoxia/reoxygenation group (3 h at 1 % O₂, 94 % N₂and 5 % CO₂followed by 10 min of 10 micromol·l⁻¹DHS and 6 h of reoxygenation in normoxia) and postconditioning group (3 h of hypoxia, three cycles of 5 min reoxygenation and 5 min hypoxia followed by 6 h of normoxia). Cell viability assessed by propidium iodide staining was decreased after DHS treatment consistent with increased levels of lactatedehydrogenase (LDH) after reoxygenation. LDH leakage was significantly reduced when cardiomyocytes in the H/Re group were exposed to DHS. DHS treatment reduced H₂O₂production and also decreased the generation of ROS in the H/Re group as evidenced by a fluorescence indicator. DHS treatment reduces reoxygenation-induced injury in cardiomyocytes by attenuation of ROS generation, H₂O₂and protein carbonyls levels. In addition, we found that both the postconditioning protocol and the DHS treatment are associated with restored ratio of phosphorylated/total protein kinase C epsilon, relative to the H/Re group. In conclusion, our data support the protective role of DHS in hypoxia/reperfusion injury and indicate that DHS may act as a postconditioning mimic.
Collapse
Affiliation(s)
- E Gabrielová
- Department of Medical Chemistry and Biochemistry, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic.
| | | | | | | |
Collapse
|
19
|
Theisen MM, Schlottmann S, August C, Herzog C, Theilmeier G, Maas M, Blumenstiel JM, Weber TP, Van Aken HK, Kaerlein KT. Detection and distribution of opioid peptide receptors in porcine myocardial tissue. Pharmacol Res 2014; 84:45-9. [PMID: 24788078 DOI: 10.1016/j.phrs.2014.04.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Revised: 04/12/2014] [Accepted: 04/20/2014] [Indexed: 12/16/2022]
Abstract
There is growing evidence that opioid peptide receptors (OPRs) play an important role in cardiovascular function. Many studies have been conducted in swine, in view of their anatomic and physiologic similarities to humans. Until now, the presence and particularly distribution of OPRs has been unclear. Porcine myocardial tissue was obtained from both the left and right atria and ventricles. Expression of mRNA for μ-, δ- and κ-OPR was determined by reverse transcription PCR. OPR proteins were detected by Western blot, distribution and cellular location were identified using immunohistochemistry. Homogenous expression of mRNA and protein for δ- and κ-OPRs were demonstrated in all porcine myocardial tissue tested, whereas expression of μ-OPR mRNA was not demonstrated in any of the tissues tested. This study demonstrates the expression of δ- and κ-OPRs in porcine myocardial tissue. No differences in distribution of δ- and κ-OPRs were found between the four heart cavities. Modulation of cardiac function by δ- and κ-OPR agonists or antagonists is therefore possible, while μ-OPR-mediated direct cardiac effects appear unlikely, due to nonexpression of the receptor. This study demonstrates that porcine studies can further elucidate the role of OPRs in cardiac (patho-)physiology.
Collapse
Affiliation(s)
- Marc Michael Theisen
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, Albert-Schweitzer-Campus 1, D-48149 Muenster, Germany; Department of Anesthesiology, Intensive Care and Pain Medicine, Raphaelsklinik Muenster, Loerstraße 23, D-48143 Muenster, Germany.
| | - Silke Schlottmann
- Department of Anesthesiology and Intensive Care, University of Ulm, Albert-Einstein-Allee 23, D-89081 Ulm, Germany
| | - Christian August
- Gerhard-Domagk-Institute of Pathology, University Hospital Muenster, Albert-Schweitzer-Campus 1, D-48149 Muenster, Germany
| | - Christine Herzog
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, Albert-Schweitzer-Campus 1, D-48149 Muenster, Germany
| | - Gregor Theilmeier
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, Albert-Schweitzer-Campus 1, D-48149 Muenster, Germany
| | - Matthias Maas
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, Albert-Schweitzer-Campus 1, D-48149 Muenster, Germany
| | - Jonas Martin Blumenstiel
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, Albert-Schweitzer-Campus 1, D-48149 Muenster, Germany
| | - Thomas Peter Weber
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, Albert-Schweitzer-Campus 1, D-48149 Muenster, Germany
| | - Hugo Karel Van Aken
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, Albert-Schweitzer-Campus 1, D-48149 Muenster, Germany
| | - Kristoffer Tim Kaerlein
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, Albert-Schweitzer-Campus 1, D-48149 Muenster, Germany
| |
Collapse
|