1
|
Deng RM, Zhou J. The role of PI3K/AKT signaling pathway in myocardial ischemia-reperfusion injury. Int Immunopharmacol 2023; 123:110714. [PMID: 37523969 DOI: 10.1016/j.intimp.2023.110714] [Citation(s) in RCA: 55] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 07/09/2023] [Accepted: 07/24/2023] [Indexed: 08/02/2023]
Abstract
Myocardial ischemia has a high incidence and mortality rate, and reperfusion is currently the standard intervention. However, reperfusion may lead to further myocardial damage, known as myocardial ischemia/reperfusion injury (MIRI). There are currently no effective clinical treatments for MIRI. The PI3K/Akt signaling pathway is involved in cardiovascular health and disease and plays an important role in reducing myocardial infarct size and restoring cardiac function after MIRI. Activation of the PI3K/Akt pathway provides myocardial protection through synergistic upregulation of antioxidant, anti-inflammatory, and autophagy activities and inhibition of mitochondrial dysfunction and cardiomyocyte apoptosis. Many studies have shown that PI3K/Akt has a significant protective effect against MIRI. Here, we reviewed the molecular regulation of PI3K/Akt in MIRI and summarized the molecular mechanism by which PI3K/Akt affects MIRI, the effects of ischemic preconditioning and ischemic postconditioning, and the role of related drugs or activators targeting PI3K/Akt in MIRI, providing novel insights for the formulation of myocardial protection strategies. This review provides evidence of the role of PI3K/Akt activation in MIRI and supports its use as a therapeutic target.
Collapse
Affiliation(s)
- Rui-Ming Deng
- Department of Anesthesiology, Ganzhou People's Hospital, 16 Meiguan Avenue, Ganzhou, Jiangxi Province 341000, PR China; The Affiliated Ganzhou Hospital of Nanchang University, 16 Meiguan Avenue, Ganzhou, Jiangxi Province 341000, PR China
| | - Juan Zhou
- Department of thyroid and Breast Surgery, Ganzhou People's Hospital, 16 Meiguan Avenue, Ganzhou, Jiangxi Province 341000, PR China; The Affiliated Ganzhou Hospital of Nanchang University, 16 Meiguan Avenue, Ganzhou, Jiangxi Province 341000, PR China.
| |
Collapse
|
2
|
Wang X, Ji X. Interactions between remote ischemic conditioning and post-stroke sleep regulation. Front Med 2021; 15:867-876. [PMID: 34811643 DOI: 10.1007/s11684-021-0887-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 07/31/2021] [Indexed: 12/31/2022]
Abstract
Sleep disturbances are common in patients with stroke, and sleep quality has a critical role in the onset and outcome of stroke. Poor sleep exacerbates neurological injury, impedes nerve regeneration, and elicits serious complications. Thus, exploring a therapy suitable for patients with stroke and sleep disturbances is imperative. As a multi-targeted nonpharmacological intervention, remote ischemic conditioning can reduce the ischemic size of the brain, improve the functional outcome of stroke, and increase sleep duration. Preclinical/clinical evidence showed that this method can inhibit the inflammatory response, mediate the signal transductions of adenosine, activate the efferents of the vagal nerve, and reset the circadian clocks, all of which are involved in sleep regulation. In particular, cytokines tumor necrosis factor α (TNFα) and adenosine are sleep factors, and electrical vagal nerve stimulation can improve insomnia. On the basis of the common mechanisms of remote ischemic conditioning and sleep regulation, a causal relationship was proposed between remote ischemic conditioning and post-stroke sleep quality.
Collapse
Affiliation(s)
- Xian Wang
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, 100069, China
| | - Xunming Ji
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, 100069, China. .,Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine, Beijing, 100069, China.
| |
Collapse
|
3
|
Ikeda M, Ide T, Tadokoro T, Miyamoto HD, Ikeda S, Okabe K, Ishikita A, Sato M, Abe K, Furusawa S, Ishimaru K, Matsushima S, Tsutsui H. Excessive Hypoxia-Inducible Factor-1α Expression Induces Cardiac Rupture via p53-Dependent Apoptosis After Myocardial Infarction. J Am Heart Assoc 2021; 10:e020895. [PMID: 34472375 PMCID: PMC8649270 DOI: 10.1161/jaha.121.020895] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Background Apoptosis plays a pivotal role in cardiac rupture after myocardial infarction (MI), and p53 is a key molecule in apoptosis during cardiac rupture. Hif‐1α (hypoxia‐inducible factor‐1α), upregulated under hypoxia, is a known p53 inducer. However, the role of Hif‐1α in the regulatory mechanisms underlying p53 upregulation, apoptosis, and cardiac rupture after MI is unclear. Methods and Results We induced MI in mice by ligating the left anterior descending artery. Hif‐1α and p53 expressions were upregulated in the border zone at day 5 after MI, accompanied by apoptosis. In rat neonatal cardiomyocytes, treatment with cobalt chloride (500 μmol/L), which mimics severe hypoxia by inhibiting PHD (prolyl hydroxylase domain‐containing protein), increased Hif‐1α and p53, accompanied by myocyte death with caspase‐3 cleavage. Silencing Hif‐1α or p53 inhibited caspase‐3 cleavage, and completely prevented myocyte death under PHD inhibition. In cardiac‐specific Hif‐1α hetero‐knockout mice, expression of p53 and cleavage of caspase‐3 and poly (ADP‐ribose) polymerase were reduced, and apoptosis was suppressed on day 5. Furthermore, the cleavage of caspase‐8 and IL‐1β (interleukin‐1β) was also suppressed in hetero knockout mice, accompanied by reduced macrophage infiltration and matrix metalloproteinase/tissue inhibitor of metalloproteinase activation. Although there was no intergroup difference in infarct size, the cardiac rupture and survival rates were significantly improved in the hetero knockout mice until day 10 after MI. Conclusions Hif‐1α plays a pivotal role in apoptosis, inflammation, and cardiac rupture after MI, in which p53 is a critical mediator, and may be a prospective therapeutic target for preventing cardiac rupture.
Collapse
Affiliation(s)
- Masataka Ikeda
- Department of Cardiovascular Medicine Faculty of Medical Sciences Kyushu University Fukuoka Japan.,Division of Cardiovascular Medicine Research Institute of Angiocardiology Faculty of Medical Sciences Kyushu University Fukuoka Japan
| | - Tomomi Ide
- Department of Cardiovascular Medicine Faculty of Medical Sciences Kyushu University Fukuoka Japan.,Division of Cardiovascular Medicine Research Institute of Angiocardiology Faculty of Medical Sciences Kyushu University Fukuoka Japan
| | - Tomonori Tadokoro
- Department of Cardiovascular Medicine Faculty of Medical Sciences Kyushu University Fukuoka Japan.,Division of Cardiovascular Medicine Research Institute of Angiocardiology Faculty of Medical Sciences Kyushu University Fukuoka Japan
| | - Hiroko Deguchi Miyamoto
- Department of Cardiovascular Medicine Faculty of Medical Sciences Kyushu University Fukuoka Japan.,Division of Cardiovascular Medicine Research Institute of Angiocardiology Faculty of Medical Sciences Kyushu University Fukuoka Japan
| | - Soichiro Ikeda
- Department of Cardiovascular Medicine Faculty of Medical Sciences Kyushu University Fukuoka Japan.,Division of Cardiovascular Medicine Research Institute of Angiocardiology Faculty of Medical Sciences Kyushu University Fukuoka Japan
| | - Kosuke Okabe
- Department of Cardiovascular Medicine Faculty of Medical Sciences Kyushu University Fukuoka Japan.,Division of Cardiovascular Medicine Research Institute of Angiocardiology Faculty of Medical Sciences Kyushu University Fukuoka Japan
| | - Akihito Ishikita
- Department of Cardiovascular Medicine Faculty of Medical Sciences Kyushu University Fukuoka Japan.,Division of Cardiovascular Medicine Research Institute of Angiocardiology Faculty of Medical Sciences Kyushu University Fukuoka Japan
| | - Midori Sato
- Department of Cardiovascular Medicine Faculty of Medical Sciences Kyushu University Fukuoka Japan.,Division of Cardiovascular Medicine Research Institute of Angiocardiology Faculty of Medical Sciences Kyushu University Fukuoka Japan
| | - Ko Abe
- Department of Cardiovascular Medicine Faculty of Medical Sciences Kyushu University Fukuoka Japan.,Division of Cardiovascular Medicine Research Institute of Angiocardiology Faculty of Medical Sciences Kyushu University Fukuoka Japan
| | - Shun Furusawa
- Department of Cardiovascular Medicine Faculty of Medical Sciences Kyushu University Fukuoka Japan.,Division of Cardiovascular Medicine Research Institute of Angiocardiology Faculty of Medical Sciences Kyushu University Fukuoka Japan
| | - Kosei Ishimaru
- Department of Cardiovascular Medicine Faculty of Medical Sciences Kyushu University Fukuoka Japan.,Division of Cardiovascular Medicine Research Institute of Angiocardiology Faculty of Medical Sciences Kyushu University Fukuoka Japan
| | - Shouji Matsushima
- Department of Cardiovascular Medicine Faculty of Medical Sciences Kyushu University Fukuoka Japan.,Division of Cardiovascular Medicine Research Institute of Angiocardiology Faculty of Medical Sciences Kyushu University Fukuoka Japan
| | - Hiroyuki Tsutsui
- Department of Cardiovascular Medicine Faculty of Medical Sciences Kyushu University Fukuoka Japan.,Division of Cardiovascular Medicine Research Institute of Angiocardiology Faculty of Medical Sciences Kyushu University Fukuoka Japan
| |
Collapse
|
4
|
Effects of remote ischemic postconditioning on HIF-1α and other markers in on-pump cardiac surgery. Gen Thorac Cardiovasc Surg 2021; 70:239-247. [PMID: 34378159 DOI: 10.1007/s11748-021-01690-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 08/04/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND There is a lack of data about the effects of remote ischemic postconditioning (RIPostC) on hypoxia-inducible factor-1α (HIF-1α) plasma levels after on-pump cardiac surgery (OPCS). This study aimed to measure the effects of RIPostC on postoperative HIF-1α plasma levels, cardiac markers and arterial oxygenation in patients undergoing OPCS. METHODS This single-centre randomized, double blind, controlled trial, enrolled 70 patients (35 control and 35 RIPostC). RIPostC was performed by 3 cycles (5 min of ischemia followed by 5 min of reperfusion) administered in upper arm immediately after the pump period. The primary outcome was to measure HIF-1α plasma levels: before surgery (T0), and 2 h (T1), 8 h (T2), 24 h (T3), 36 h (T4) and 48 h (T5) after RIPostC. As secondary endpoint, Troponin T, CK-MB, CPK plasma levels and PaO2/FiO2 ratio were measured. RESULTS HIF-1α plasma levels were increased at T1-T3 compared to T0 in both groups (P < 0.001). In the RIPostC group HIF-1α increased compared to the control group: differences between means (95% CI) were 0.034 (0.006-0.06) P = 0.019 at T1; 0.041 (0.013-0.069) P = 0.005 at T2; and 0.021 (0.001-0.042) P = 0.045 at T3. PaO2/FiO2 was higher in the RIPostC group than in the control group: at T3, T4 and T5. Moreover, Troponin T, CK-MB and CPK values decreased in the RIPostC group compared to the control group. CONCLUSIONS HIF-1α plasma levels increased in control patients during for at least 36 h after OPCS. RIPostC resulted in even higher HIF-1α levels during at least the first 24 h and improved arterial oxygenation and cardiac markers.
Collapse
|
5
|
Cullen PP, Tsui SS, Caplice NM, Hinchion JA. A state-of-the-art review of the current role of cardioprotective techniques in cardiac transplantation. Interact Cardiovasc Thorac Surg 2021; 32:683-694. [PMID: 33971665 DOI: 10.1093/icvts/ivaa333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 11/24/2020] [Accepted: 12/06/2020] [Indexed: 11/14/2022] Open
Abstract
OBJECTIVES The use of 'extended criteria' donor hearts and reconditioned hearts from donation after circulatory death has corresponded with an increase in primary graft dysfunction, with ischaemia-reperfusion injury being a major contributing factor in its pathogenesis. Limiting ischaemia-reperfusion injury through optimising donor heart preservation may significantly improve outcomes. We sought to review the literature to evaluate the evidence for this. METHODS A review of the published literature was performed to assess the potential impact of organ preservation optimisation on cardiac transplantation outcomes. RESULTS Ischaemia-reperfusion injury is a major factor in myocardial injury during transplantation with multiple potential therapeutic targets. Innate survival pathways have been identified, which can be mimicked with pharmacological conditioning. Although incompletely understood, discoveries in this domain have yielded extremely encouraging results with one of the most exciting prospects being the synergistic effect of selected agents. Ex situ heart perfusion is an additional promising adjunct. CONCLUSIONS Cardiac transplantation presents a unique opportunity to perfuse the whole heart before, or immediately after, the onset of ischaemia, thus maximising the potential for global cardioprotection while limiting possible systemic side effects. While clinical translation in the setting of myocardial infarction has often been disappointing, cardiac transplantation may afford the opportunity for cardioprotection to finally deliver on its preclinical promise.
Collapse
Affiliation(s)
- Paul P Cullen
- Department of Cardiothoracic Surgery, Cork University Hospital, Cork, Ireland
| | - Steven S Tsui
- Department of Transplantation, Royal Papworth Hospital NHS Foundation Trust, Cambridge, UK
| | - Noel M Caplice
- Centre for Research in Vascular Biology, Biosciences Institute, University College Cork, Cork, Ireland
| | - John A Hinchion
- Department of Cardiothoracic Surgery, Cork University Hospital, Cork, Ireland
| |
Collapse
|
6
|
Pearce L, Davidson SM, Yellon DM. Does remote ischaemic conditioning reduce inflammation? A focus on innate immunity and cytokine response. Basic Res Cardiol 2021; 116:12. [PMID: 33629195 PMCID: PMC7904035 DOI: 10.1007/s00395-021-00852-0] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 02/04/2021] [Indexed: 02/07/2023]
Abstract
The benefits of remote ischaemic conditioning (RIC) have been difficult to translate to humans, when considering traditional outcome measures, such as mortality and heart failure. This paper reviews the recent literature of the anti-inflammatory effects of RIC, with a particular focus on the innate immune response and cytokine inhibition. Given the current COVID-19 pandemic, the inflammatory hypothesis of cardiac protection is an attractive target on which to re-purpose such novel therapies. A PubMed/MEDLINE™ search was performed on July 13th 2020, for the key terms RIC, cytokines, the innate immune system and inflammation. Data suggest that RIC attenuates inflammation in animals by immune conditioning, cytokine inhibition, cell survival and the release of anti-inflammatory exosomes. It is proposed that RIC inhibits cytokine release via a reduction in nuclear factor kappa beta (NF-κB)-mediated NLRP3 inflammasome production. In vivo, RIC attenuates pro-inflammatory cytokine release in myocardial/cerebral infarction and LPS models of endotoxaemia. In the latter group, cytokine inhibition is associated with a profound survival benefit. Further clinical trials should establish whether the benefits of RIC in inflammation can be observed in humans. Moreover, we must consider whether uncomplicated MI and elective surgery are the most suitable clinical conditions in which to test this hypothesis.
Collapse
Affiliation(s)
- Lucie Pearce
- The Hatter Cardiovascular Institute, 67 Chenies Mews, London, WC1E 6HX, UK
| | - Sean M Davidson
- The Hatter Cardiovascular Institute, 67 Chenies Mews, London, WC1E 6HX, UK
| | - Derek M Yellon
- The Hatter Cardiovascular Institute, 67 Chenies Mews, London, WC1E 6HX, UK.
| |
Collapse
|
7
|
Wu JW, Hu H, Li D, Ma LK. Hypoxia-inducible factor 2-alpha-dependent induction of IL-6 protects the heart from ischemia/reperfusion injury. Aging (Albany NY) 2021; 13:3443-3458. [PMID: 33428604 PMCID: PMC7906200 DOI: 10.18632/aging.202276] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 09/05/2020] [Indexed: 12/21/2022]
Abstract
Myocardial ischemia-reperfusion injury (MIRI) results in increased myocardial infarct size and leads to poor clinical outcomes. Hypoxia-inducible factor 2-alpha (HIF2α) exerts myocardial protective effects during MIRI through as yet unclear mechanisms. Here, we show that knockdown of HIF2α with cardiotropic recombinant adeno-associated virus serotype 9 (rAAV9) in mouse hearts significantly increased the infarct sizes during myocardial ischemia/reperfusion (MI/R). In addition, HIF2α transcriptionally regulated the expression of interleukin 6 (IL-6) in cardiomyocytes to elicit cardioprotection. Likewise, IL-6 deficiency aggravated MIRI, while treatment with recombinant IL-6 had cardioprotective effects and rescued the mice with HIF2α knockdown. Furthermore, IL-6 treatment significantly activated the PI3K/Akt and STAT3 signaling pathways in the myocardium during MI/R, and the specific inhibitors wortmannin (specific phosphoinositide 3-kinase inhibitor) and Stattic (specific STAT3 inhibitor) substantially abolished HIF2α/IL-6-induced cardioprotection. These studies suggest that HIF2α transcription regulates the expression of IL-6 in cardiomyocytes and plays a protective role during MI/R.
Collapse
Affiliation(s)
- Jia-Wei Wu
- Department of Cardiology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Hao Hu
- Department of Cardiology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Dan Li
- Department of Cardiology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Li-Kun Ma
- Department of Cardiology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei 230001, China
| |
Collapse
|
8
|
Zhang Y, Liu J, Li Y, Tan N, Du K, Zhao H, Wang J, Zhang J, Wang W, Wang Y. Protective Role of Enalapril in Anthracycline-Induced Cardiotoxicity: A Systematic Review. Front Pharmacol 2020; 11:788. [PMID: 32536868 PMCID: PMC7266978 DOI: 10.3389/fphar.2020.00788] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Accepted: 05/12/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Evidence of the preventive and therapeutic effects of enalapril on cardiotoxicity caused by chemotherapy needs to be further confirmed and updated. METHODS We performed a systematic review of studies from electronic databases that were searched from inception to January 29, 2019, and included relevant studies analyzing enalapril as a cardioprotective agent before or during the use of anthracyclines by oncology patients. Homogeneous results from different studies were pooled using RevMan 5.3 software. The Cochrane risk-of-bias tool was used to determine the quality of the studies. RESULTS We examined and screened 626 studies according to specific criteria and ultimately included seven studies that were relevant to the indicated topic. Among them, three studies reported the incidence of death during 6- and 12-month follow-up periods. Six of the seven included studies showed possible positive results, suggesting that enalapril plays a cardioprotective role, while five of these studies showed that there was a significant difference in the left ventricular ejection fraction (LVEF) between an enalapril group and a control group (weighted mean difference (WMD) = 7.18, 95% CI: 2.49-11.87, I2 = 96%, P < .001). Moreover, enalapril was beneficial in reducing troponin I (TnI), creatine kinase myocardial band (CK-MB) and N-terminal pro-b-type natriuretic peptide (NT-proBNP) levels in cancer patients treated with anthracycline. CONCLUSIONS Although a protective effect of enalapril on myocardial toxicity was observed in terms of the LVEF values and TnI, CK-MB and NT-proBNP levels, its use in the prevention and treatment of cardiotoxicity caused by anthracycline needs to be investigated by more scientific research.
Collapse
Affiliation(s)
- Yili Zhang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Junjie Liu
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yuan Li
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Nannan Tan
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Kangjia Du
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Huihui Zhao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
- Ministry of Education Key Laboratory of TCM Syndrome and Formula & Beijing Key Laboratory of TCM Syndrome and Formula, Beijing, China
| | - Juan Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
- Ministry of Education Key Laboratory of TCM Syndrome and Formula & Beijing Key Laboratory of TCM Syndrome and Formula, Beijing, China
| | - Jian Zhang
- School of Life Science, Beijing University of Chinese Medicine, Beijing, China
| | - Wei Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
- Ministry of Education Key Laboratory of TCM Syndrome and Formula & Beijing Key Laboratory of TCM Syndrome and Formula, Beijing, China
| | - Yong Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
9
|
Moulin S, Thomas A, Arnaud C, Arzt M, Wagner S, Maier LS, Pépin JL, Godin-Ribuot D, Gaucher J, Belaidi E. Cooperation Between Hypoxia-Inducible Factor 1α and Activating Transcription Factor 4 in Sleep Apnea-Mediated Myocardial Injury. Can J Cardiol 2020; 36:936-940. [PMID: 32387037 DOI: 10.1016/j.cjca.2020.04.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 04/01/2020] [Accepted: 04/01/2020] [Indexed: 01/21/2023] Open
Abstract
Chronic intermittent hypoxia (CIH) occurring during sleep apnea amplifies infarct size owing to ischemia-reperfusion. CIH activates hypoxia-inducible factor 1 (HIF-1) and activating transcription factor 4 (ATF4). However, whether HIF-1 and ATF4 interact to promote cardiomyocyte death remains unexplored. For the first time, we observed that in myocardium from apneic patients, CCAAT enhancer-binding protein homologous protein (CHOP) expression is increased and HIF-1α expression is correlated with sleep apnea severity. In mice, single-allele deletion of HIF-1α prevents CIH increase in CHOP expression and infarct size. We uncovered a physical interaction between HIF-1α and ATF4 in CIH that may represent a novel cardiomyocyte death complex.
Collapse
Affiliation(s)
- Sophie Moulin
- Laboratoire HP2, Institut National de la Santé et de la Recherche Médicale, Université Grenoble Alpes, Grenoble, France; Institut National de la Santé et de la Recherche Médicale U1042, Grenoble, France
| | - Amandine Thomas
- Laboratoire HP2, Institut National de la Santé et de la Recherche Médicale, Université Grenoble Alpes, Grenoble, France; Institut National de la Santé et de la Recherche Médicale U1042, Grenoble, France
| | - Claire Arnaud
- Laboratoire HP2, Institut National de la Santé et de la Recherche Médicale, Université Grenoble Alpes, Grenoble, France; Institut National de la Santé et de la Recherche Médicale U1042, Grenoble, France
| | - Michael Arzt
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Stefan Wagner
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Lars S Maier
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Jean-Louis Pépin
- Laboratoire HP2, Institut National de la Santé et de la Recherche Médicale, Université Grenoble Alpes, Grenoble, France; Institut National de la Santé et de la Recherche Médicale U1042, Grenoble, France; Centre Hospitalier Universitaire des Alpes, Grenoble, France
| | - Diane Godin-Ribuot
- Laboratoire HP2, Institut National de la Santé et de la Recherche Médicale, Université Grenoble Alpes, Grenoble, France; Institut National de la Santé et de la Recherche Médicale U1042, Grenoble, France
| | - Jonathan Gaucher
- Laboratoire HP2, Institut National de la Santé et de la Recherche Médicale, Université Grenoble Alpes, Grenoble, France; Institut National de la Santé et de la Recherche Médicale U1042, Grenoble, France
| | - Elise Belaidi
- Laboratoire HP2, Institut National de la Santé et de la Recherche Médicale, Université Grenoble Alpes, Grenoble, France; Institut National de la Santé et de la Recherche Médicale U1042, Grenoble, France.
| |
Collapse
|
10
|
Naryzhnaya NV, Maslov LN, Oeltgen PR. Pharmacology of mitochondrial permeability transition pore inhibitors. Drug Dev Res 2019; 80:1013-1030. [PMID: 31823411 DOI: 10.1002/ddr.21593] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 08/08/2019] [Accepted: 08/12/2019] [Indexed: 12/13/2022]
Affiliation(s)
- Natalia V. Naryzhnaya
- Laboratory of Experimental CardiologyCardiology Research Institute, Tomsk National Research Medical Center of the Russian Academy of Science Tomsk Russia
| | - Leonid N. Maslov
- Laboratory of Experimental CardiologyCardiology Research Institute, Tomsk National Research Medical Center of the Russian Academy of Science Tomsk Russia
| | - Peter R. Oeltgen
- Department of PathologyUniversity of Kentucky College of Medicine Lexington Kentucky
| |
Collapse
|
11
|
Wang H, Lyu Y, Liao Q, Jin L, Xu L, Hu Y, Yu Y, Guo K. Effects of Remote Ischemic Preconditioning in Patients Undergoing Off-Pump Coronary Artery Bypass Graft Surgery. Front Physiol 2019; 10:495. [PMID: 31110480 PMCID: PMC6501551 DOI: 10.3389/fphys.2019.00495] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 04/08/2019] [Indexed: 01/15/2023] Open
Abstract
Purpose This study aimed to evaluate effects of remote ischemic preconditioning (RIPC) on myocardial injury in patients undergoing off-pump coronary artery bypass graft surgery (OPCABG). Methods Sixty-five patients scheduled for the OPCABG were randomly assigned to control (n = 32) or RIPC group (n = 33). All patients received general anesthesia. Before the surgical incision, RIPC was induced on an upper limb with repeated 5-min ischemia and 5-min reperfusion for four times. Blood samples were collected from right internal jugular vein. Plasma levels of IL-6, IL-8, IL-10, TNF-α, cTnT, HFABP, IMA, and MDA were detected at pre-operatively and 0, 6, 18, 24, 48, 72, and 120 h after the surgery. Left internal mammary artery (LIMA) and great saphenous vein (GSV) was cut into 2–3 mm for Western blot analysis of Hif-1α. Results In the present study, RIPC treatment significantly reduced plasma levels of cardiac troponin T (p < 0.05), heart-type fatty acid binding protein (p < 0.05), ischemia modified albumin (p < 0.05), malondialdehyde (p < 0.05), as well as plasma levels of pro-inflammatory cytokines including IL-6, IL-8, and TNF-α (P < 0.05, respectively). RIPC treatment significantly increased hypoxia-inducible factor-1α (p < 0.05) expression as well. Mechanical ventilation time for postoperative patients was shortened in RIPC group than those in control group (17.4 ± 3.8 h vs. 19.7 ± 2.9 h, respectively, p < 0.05). Conclusion RIPC by upper limb ischemia shortens mechanical ventilation time in patients undergoing OPCABG. RIPC treatment reduces postoperative myocardial enzyme expression and pro-inflammatory cytokine production. RIPC is a protective therapeutic approach in the coronary artery bypass graft surgery.
Collapse
Affiliation(s)
- Huilin Wang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yi Lyu
- Department of Anesthesiology, Yunnan Baoshan Anli Hospital, Baoshan, China
| | - Qingwu Liao
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Lin Jin
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Liying Xu
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yan Hu
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ying Yu
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Kefang Guo
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
12
|
Ndongson-Dongmo B, Lang GP, Mece O, Hechaichi N, Lajqi T, Hoyer D, Brodhun M, Heller R, Wetzker R, Franz M, Levy FO, Bauer R. Reduced ambient temperature exacerbates SIRS-induced cardiac autonomic dysregulation and myocardial dysfunction in mice. Basic Res Cardiol 2019; 114:26. [DOI: 10.1007/s00395-019-0734-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 04/12/2019] [Indexed: 12/13/2022]
|
13
|
Things get broken: the hypoxia-inducible factor prolyl hydroxylases in ischemic heart disease. Basic Res Cardiol 2019; 114:16. [PMID: 30859331 DOI: 10.1007/s00395-019-0725-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 03/01/2019] [Indexed: 12/19/2022]
Abstract
A major challenge in developing new treatments for myocardial infarction (MI) is an improved understanding of the pathophysiology of hypoxic tissue damage and the activation of endogenous adaptive programs to hypoxia. Due to the relevance of oxygen in metabolism, molecular adaptation to hypoxia driven by the hypoxia-inducible factors (HIFs) and the HIF-regulating prolyl hydroxylase domain enzymes (PHDs) is pivotal for the survival of cells and tissue under hypoxia. The heart under ischemic stress will extensively rely on these mechanisms of endogenous cardiac protection until hypoxia becomes too severe. In the past, work from several laboratories has provided evidence that inhibition of HIF-regulating PHDs might improve the outcome in ischemic heart disease (IHD) potentially because the adaptive mechanisms are boosted early and vigorously. Here, we review the role of the HIF hydroxylase pathway in IHD and highlight the potential of PHD inhibitors as a new treatment for MI with special regard to acute ischemia, reperfusion, and regeneration of the heart.
Collapse
|
14
|
Krylatov AV, Maslov LN, Voronkov NS, Boshchenko AA, Popov SV, Gomez L, Wang H, Jaggi AS, Downey JM. Reactive Oxygen Species as Intracellular Signaling Molecules in the Cardiovascular System. Curr Cardiol Rev 2018; 14:290-300. [PMID: 29962348 PMCID: PMC6300799 DOI: 10.2174/1573403x14666180702152436] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 06/13/2018] [Accepted: 06/20/2018] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Redox signaling plays an important role in the lives of cells. This signaling not only becomes apparent in pathologies but is also thought to be involved in maintaining physiological homeostasis. Reactive Oxygen Species (ROS) can activate protein kinases: CaMKII, PKG, PKA, ERK, PI3K, Akt, PKC, PDK, JNK, p38. It is unclear whether it is a direct interaction of ROS with these kinases or whether their activation is a consequence of inhibition of phosphatases. ROS have a biphasic effect on the transport of Ca2+ in the cell: on one hand, they activate the sarcoplasmic reticulum Ca2+-ATPase, which can reduce the level of Ca2+ in the cell, and on the other hand, they can inactivate Ca2+-ATPase of the plasma membrane and open the cation channels TRPM2, which promote Ca2+-loading and subsequent apoptosis. ROS inhibit the enzyme PHD2, which leads to the stabilization of HIF-α and the formation of the active transcription factor HIF. CONCLUSION Activation of STAT3 and STAT5, induced by cytokines or growth factors, may include activation of NADPH oxidase and enhancement of ROS production. Normal physiological production of ROS under the action of cytokines activates the JAK/STAT while excessive ROS production leads to their inhibition. ROS cause the activation of the transcription factor NF-κB. Physiological levels of ROS control cell proliferation and angiogenesis. ROS signaling is also involved in beneficial adaptations to survive ischemia and hypoxia, while further increases in ROS can trigger programmed cell death by the mechanism of apoptosis or autophagy. ROS formation in the myocardium can be reduced by moderate exercise.
Collapse
Affiliation(s)
| | - Leonid N. Maslov
- Address correspondence to this author at the Cardiology Research Institute, Tomsk National Research Medical Center of the Russian Academy of
Science, Tomsk, Russia; Tel: 3822 262174; Fax: 3822 555057;
E-mail:
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Gedik N, Kottenberg E, Thielmann M, Frey UH, Jakob H, Peters J, Heusch G, Kleinbongard P. Potential humoral mediators of remote ischemic preconditioning in patients undergoing surgical coronary revascularization. Sci Rep 2017; 7:12660. [PMID: 28978919 PMCID: PMC5627278 DOI: 10.1038/s41598-017-12833-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 09/15/2017] [Indexed: 01/03/2023] Open
Abstract
Remote ischemic preconditioning (RIPC) by repeated brief cycles of limb ischemia/reperfusion may reduce myocardial ischemia/reperfusion injury and improve patients‘ prognosis after elective coronary artery bypass graft (CABG) surgery. The signal transducer and activator of transcription (STAT)5 activation in left ventricular myocardium is associated with RIPC´s cardioprotection. Cytokines and growth hormones typically activate STATs and could therefore act as humoral transfer factors of RIPC´s cardioprotection. We here determined arterial plasma concentrations of 25 different cytokines, growth hormones, and other factors which have previously been associated with cardioprotection, before (baseline)/after RIPC or placebo (n = 23/23), respectively, and before/after ischemic cardioplegic arrest in CABG patients. RIPC-induced protection was reflected by a 35% reduction of serum troponin I release. With the exception of interleukin-1α, none of the humoral factors changed in their concentrations after RIPC or placebo, respectively. Interleukin-1α, when normalized to baseline, increased after RIPC (280 ± 56%) but not with placebo (97 ± 15%). The interleukin-1α concentration remained increased until after ischemic cardioplegic arrest and was also higher than with placebo in absolute concentrations (25 ± 6 versus 16 ± 3 pg/mL). Only interleukin-1α possibly fulfills the criteria which would be expected from a substance to be released in response to RIPC and to protect the myocardium during ischemic cardioplegic arrest.
Collapse
Affiliation(s)
- Nilgün Gedik
- Institute for Pathophysiology, West German Heart and Vascular Center Essen, Universitätsklinikum Essen, Universität Duisburg- Essen, Essen, Germany
| | - Eva Kottenberg
- Klinik für Anästhesiologie und Intensivmedizin, Universitätsklinikum Essen, Universität Duisburg-Essen, Essen, Germany
| | - Matthias Thielmann
- Department of Thoracic and Cardiovascular Surgery, West German Heart and Vascular Center Essen, Universitätsklinikum Essen, Universität Duisburg- Essen, Essen, Germany
| | - Ulrich H Frey
- Klinik für Anästhesiologie und Intensivmedizin, Universitätsklinikum Essen, Universität Duisburg-Essen, Essen, Germany
| | - Heinz Jakob
- Department of Thoracic and Cardiovascular Surgery, West German Heart and Vascular Center Essen, Universitätsklinikum Essen, Universität Duisburg- Essen, Essen, Germany
| | - Jürgen Peters
- Klinik für Anästhesiologie und Intensivmedizin, Universitätsklinikum Essen, Universität Duisburg-Essen, Essen, Germany
| | - Gerd Heusch
- Institute for Pathophysiology, West German Heart and Vascular Center Essen, Universitätsklinikum Essen, Universität Duisburg- Essen, Essen, Germany
| | - Petra Kleinbongard
- Institute for Pathophysiology, West German Heart and Vascular Center Essen, Universitätsklinikum Essen, Universität Duisburg- Essen, Essen, Germany.
| |
Collapse
|
16
|
Belaidi E, Thomas A, Bourdier G, Moulin S, Lemarié E, Levy P, Pépin JL, Korichneva I, Godin-Ribuot D, Arnaud C. Endoplasmic reticulum stress as a novel inducer of hypoxia inducible factor-1 activity: its role in the susceptibility to myocardial ischemia-reperfusion induced by chronic intermittent hypoxia. Int J Cardiol 2016; 210:45-53. [PMID: 26922713 DOI: 10.1016/j.ijcard.2016.02.096] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Revised: 02/09/2016] [Accepted: 02/14/2016] [Indexed: 12/30/2022]
Abstract
BACKGROUND Obstructive sleep apnea (OSA) is a highly prevalent disease and a risk factor for myocardial infarction expansion in humans. Intermittent hypoxia (IH) is known to be the most important OSA feature in terms of cardiovascular morbi-mortality. Since ER stress and HIF-1 are known to be involved in cardiomyocyte life or death, this study investigates the role of ER stress on HIF-1 activation in myocardial susceptibility to ischemia-reperfusion (I/R) induced by IH. METHODS C57Bl6J, HIF-1α(+/-) and their respective control mice were exposed to 14 days of IH (21-5% FiO2, 60 scycle, 8h/day). Myocardial inter-organelle calcium exchanges, ER stress and HIF-1 activity were investigated and in vivo I/R was performed to measure infarct size. In additional groups, tauroursodeoxycholic acid (TUDCA, 75 mg·kg(-1)), an ER stress inhibitor, was administered daily during exposure. RESULTS In C57Bl6J mice, chronic IH induced an increase in ER-Ca(2+) content, ER stress markers and HIF-1 activity, associated with an enhanced infarct size (33.7 ± 9.4 vs. 61.0 ± 5.6% in N and IH, respectively, p<0.05). IH failed to increase infarct size in HIF-1α deficient mice (42.4 ± 2.7 and 24.7 ± 3.4% N and IH, respectively). Finally, TUDCA totally abolished the IH-induced increase in HIF-1 activity (1.3 ± 0.04 vs. 0.14 ± 0.02 fold increase in IH vs. IH-TUDCA respectively, p<0.0001) and in infarct size (55.5 ± 7.6 vs. 49.9 ± 3.0 in N-TUDCA and IH-TUDCA, respectively). CONCLUSION This novel regulatory mechanism of HIF-1 activity by ER stress should be considered as a potential diagnostic tool for cardiovascular complications in OSA patients as well as a therapeutic target to limit myocardial ischemic damage.
Collapse
Affiliation(s)
- Elise Belaidi
- Université Grenoble Alpes, Laboratoire HP2, Grenoble F-38042, France; INSERM, U1042, Grenoble F-38042, France.
| | - Amandine Thomas
- Université Grenoble Alpes, Laboratoire HP2, Grenoble F-38042, France; INSERM, U1042, Grenoble F-38042, France
| | - Guillaume Bourdier
- Université Grenoble Alpes, Laboratoire HP2, Grenoble F-38042, France; INSERM, U1042, Grenoble F-38042, France
| | - Sophie Moulin
- Université Grenoble Alpes, Laboratoire HP2, Grenoble F-38042, France; INSERM, U1042, Grenoble F-38042, France
| | - Emeline Lemarié
- Université Grenoble Alpes, Laboratoire HP2, Grenoble F-38042, France; INSERM, U1042, Grenoble F-38042, France
| | - Patrick Levy
- Université Grenoble Alpes, Laboratoire HP2, Grenoble F-38042, France; INSERM, U1042, Grenoble F-38042, France
| | - Jean-Louis Pépin
- Université Grenoble Alpes, Laboratoire HP2, Grenoble F-38042, France; INSERM, U1042, Grenoble F-38042, France
| | - Irina Korichneva
- Université Picardie, Laboratoire de biologie cellulaire moléculaire, Amiens 80000, France
| | - Diane Godin-Ribuot
- Université Grenoble Alpes, Laboratoire HP2, Grenoble F-38042, France; INSERM, U1042, Grenoble F-38042, France
| | - Claire Arnaud
- Université Grenoble Alpes, Laboratoire HP2, Grenoble F-38042, France; INSERM, U1042, Grenoble F-38042, France
| |
Collapse
|
17
|
Cabrera-Fuentes HA, Alba-Alba C, Aragones J, Bernhagen J, Boisvert WA, Bøtker HE, Cesarman-Maus G, Fleming I, Garcia-Dorado D, Lecour S, Liehn E, Marber MS, Marina N, Mayr M, Perez-Mendez O, Miura T, Ruiz-Meana M, Salinas-Estefanon EM, Ong SB, Schnittler HJ, Sanchez-Vega JT, Sumoza-Toledo A, Vogel CW, Yarullina D, Yellon DM, Preissner KT, Hausenloy DJ. Meeting report from the 2nd International Symposium on New Frontiers in Cardiovascular Research. Protecting the cardiovascular system from ischemia: between bench and bedside. Basic Res Cardiol 2016; 111:7. [PMID: 26667317 PMCID: PMC4679108 DOI: 10.1007/s00395-015-0527-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Accepted: 11/26/2015] [Indexed: 12/17/2022]
Abstract
Recent advances in basic cardiovascular research as well as their translation into the clinical situation were the focus at the last "New Frontiers in Cardiovascular Research meeting". Major topics included the characterization of new targets and procedures in cardioprotection, deciphering new players and inflammatory mechanisms in ischemic heart disease as well as uncovering microRNAs and other biomarkers as versatile and possibly causal factors in cardiovascular pathogenesis. Although a number of pathological situations such as ischemia-reperfusion injury or atherosclerosis can be simulated and manipulated in diverse animal models, also to challenge new drugs for intervention, patient studies are the ultimate litmus test to obtain unequivocal information about the validity of biomedical concepts and their application in the clinics. Thus, the open and bidirectional exchange between bench and bedside is crucial to advance the field of ischemic heart disease with a particular emphasis of understanding long-lasting approaches in cardioprotection.
Collapse
Affiliation(s)
- Hector A Cabrera-Fuentes
- Institute of Biochemistry, Medical School, Justus-Liebig University, Giessen, Germany
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore, Singapore, Singapore
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, Singapore
- Department of Microbiology, Kazan Federal University, Kazan, Russian Federation
- Escuela de Ingeniería y Ciencias, Centro de Biotecnología-FEMSA, Tecnológico de Monterrey, Monterrey, NL, México
| | - Corina Alba-Alba
- Institute of Genetics, Univeristy of the Sea. Puerto Escondido Campus, Oaxaca Oaxacan System of State Universities (SUNEO), Oaxaca, México
| | - Julian Aragones
- Research Unit, Hospital of Santa Cristina, Research Institute Princesa (IP), Autonomous University of Madrid, Madrid, Spain
| | - Jürgen Bernhagen
- Institute of Biochemistry and Molecular Cell Biology, RWTH Aachen University, Aachen, Germany
| | - William A Boisvert
- Department of Microbiology, Kazan Federal University, Kazan, Russian Federation
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, USA
| | - Hans E Bøtker
- Department of Cardiology, Aarhus University Hospital, Skejby, Aarhus N, Denmark
| | | | - Ingrid Fleming
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe-University, Frankfurt, Germany
| | | | - Sandrine Lecour
- Hatter Institute and MRC Inter-University Cape Heart Unit, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Elisa Liehn
- Institute for Molecular Cardiovascular Research, RWTH University Hospital Aachen, Aachen, Germany
| | - Michael S Marber
- Department of Cardiology, The Rayne Institute, St Thomas' Campus, King's College London, London, UK
| | - Nephtali Marina
- Department of Clinical Pharmacology, University College London, London, UK
| | - Manuel Mayr
- The James Black Centre, King's College, University of London, London, UK
| | - Oscar Perez-Mendez
- Department of Molecular Biology, National Institute of Cardiology, Mexico City, Mexico
| | - Tetsuji Miura
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Marisol Ruiz-Meana
- Valld'Hebron University Hospital and Research Institute, Barcelona, Spain
| | | | - Sang-Bing Ong
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore, Singapore, Singapore
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, Singapore
| | - Hans J Schnittler
- Institute of Anatomy and Vascular Biology, Westfalian-Wilhelms-University, Münster, Germany
| | - Jose T Sanchez-Vega
- Laboratory of Parasitology, Department of Microbiology and Parasitology, Faculty of Medicine, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Adriana Sumoza-Toledo
- Laboratorio Multidisciplinario de Ciencias Biomédicas, Instituto de Investigaciones Medico-Biológicas, Universidad Veracruzana campus Veracruz, Veracruz, Mexico
| | - Carl-Wilhelm Vogel
- Department of Pathology, John A. Burns School of Medicine, University of Hawaii, Honolulu, USA
| | - Dina Yarullina
- Department of Microbiology, Kazan Federal University, Kazan, Russian Federation
| | - Derek M Yellon
- The Hatter Cardiovascular Institute, University College London, London, UK
- The National Institute of Health Research University College London Hospitals Biomedical Research Centre, London, UK
| | - Klaus T Preissner
- Institute of Biochemistry, Medical School, Justus-Liebig University, Giessen, Germany
| | - Derek J Hausenloy
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore, Singapore, Singapore.
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, Singapore.
- The Hatter Cardiovascular Institute, University College London, London, UK.
- The National Institute of Health Research University College London Hospitals Biomedical Research Centre, London, UK.
| |
Collapse
|
18
|
Abstract
Reperfusion is mandatory to salvage ischemic myocardium from infarction, but reperfusion per se contributes to injury and ultimate infarct size. Therefore, cardioprotection beyond that by timely reperfusion is needed to reduce infarct size and improve the prognosis of patients with acute myocardial infarction. The conditioning phenomena provide such cardioprotection, insofar as brief episodes of coronary occlusion/reperfusion preceding (ischemic preconditioning) or following (ischemic postconditioning) sustained myocardial ischemia with reperfusion reduce infarct size. Even ischemia/reperfusion in organs remote from the heart provides cardioprotection (remote ischemic conditioning). The present review characterizes the signal transduction underlying the conditioning phenomena, including their physical and chemical triggers, intracellular signal transduction, and effector mechanisms, notably in the mitochondria. Cardioprotective signal transduction appears as a highly concerted spatiotemporal program. Although the translation of ischemic postconditioning and remote ischemic conditioning protocols to patients with acute myocardial infarction has been fairly successful, the pharmacological recruitment of cardioprotective signaling has been largely disappointing to date.
Collapse
Affiliation(s)
- Gerd Heusch
- From the Institute for Pathophysiology, West German Heart and Vascular Centre, University of Essen Medical School, Essen, Germany.
| |
Collapse
|
19
|
Peng C, Pei H, Wei F, Tian X, Deng J, Yan C, Li Y, Sun M, Zhang J, Liu D, Rong J, Wang J, Gao E, Li S, Han Y. Cellular repressor of E1A-stimulated gene overexpression in bone mesenchymal stem cells protects against rat myocardial infarction. Int J Cardiol 2015; 183:232-41. [DOI: 10.1016/j.ijcard.2015.01.059] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Revised: 12/14/2014] [Accepted: 01/25/2015] [Indexed: 12/13/2022]
|
20
|
Plasma from human volunteers subjected to remote ischemic preconditioning protects human endothelial cells from hypoxia-induced cell damage. Basic Res Cardiol 2015; 110:17. [PMID: 25716080 PMCID: PMC4341024 DOI: 10.1007/s00395-015-0474-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Revised: 02/17/2015] [Accepted: 02/18/2015] [Indexed: 02/06/2023]
Abstract
Short repeated cycles of peripheral ischemia/reperfusion (I/R) can protect distant organs from subsequent prolonged I/R injury; a phenomenon known as remote ischemic preconditioning (RIPC). A RIPC-mediated release of humoral factors might play a key role in this protection and vascular endothelial cells are potential targets for these secreted factors. In the present study, RIPC-plasma obtained from healthy male volunteers was tested for its ability to protect human umbilical endothelial cells (HUVEC) from hypoxia–induced cell damage. 10 healthy male volunteers were subjected to a RIPC-protocol consisting of 4 × 5 min inflation/deflation of a blood pressure cuff located at the upper arm. Plasma was collected before (T0; control), directly after (T1) and 1 h after (T2) the RIPC procedure. HUVEC were subjected to 24 h hypoxia damage and simultaneously incubated with 5 % of the respective RIPC-plasma. Cell damage was evaluated by lactate dehydrogenase (LDH)-measurements. Western blot experiments of hypoxia inducible factor 1 alpha (HIF1alpha), phosphorylated signal transducer and activator of transcription 5 (STAT5), protein kinase B (AKT) and extracellular signal-related kinase 1/2 (ERK-1/2) were performed. Furthermore, the concentrations of hVEGF were evaluated in the RIPC-plasma by sandwich ELISA. Hypoxia–induced cell damage was significantly reduced by plasma T1 (p = 0.02 vs T0). The protective effect of plasma T1 was accompanied by an augmentation of the intracellular HIF1alpha (p = 0.01 vs T0) and increased phosphorylation of ERK-1/2 (p = 0.03 vs T0). Phosphorylation of AKT and STAT5 remained unchanged. Analysis of the protective RIPC-plasma T1 showed significantly reduced levels of hVEGF (p = 0.01 vs T0). RIPC plasma protects endothelial cells from hypoxia–induced cell damage and humoral mediators as well as intracellular HIF1alpha may be involved.
Collapse
|
21
|
Cuadrado-Berrocal I, Gómez-Gaviro MV, Benito Y, Barrio A, Bermejo J, Fernández-Santos ME, Sánchez PL, Desco M, Fernández-Avilés F, Fernández-Velasco M, Boscá L, de Las Heras B. A labdane diterpene exerts ex vivo and in vivo cardioprotection against post-ischemic injury: involvement of AKT-dependent mechanisms. Biochem Pharmacol 2015; 93:428-439. [PMID: 25557296 DOI: 10.1016/j.bcp.2014.12.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Revised: 12/17/2014] [Accepted: 12/18/2014] [Indexed: 02/07/2023]
Abstract
Therapeutic approaches to protect the heart from ischemia/reperfusion (I/R) injury are an area of intense research, as myocardial infarction is a major cause of mortality and morbidity. Diterpenes are bioactive natural products with great therapeutic potential. In the present study, we have investigated the in vivo cardioprotective effects of a labdane diterpene (DT1) against cardiac I/R injury and the molecular mechanisms involved. DT1 attenuates post-ischemic injury via an AKT-dependent activation of HIF-1α, survival pathways and inhibition of NF-κB signaling. Myocardial infarction (MI) was induced in Wistar rats occluding the left coronary artery (LCA) for 30min followed by 72h reperfusion. DT1 (5mg/kg) was intravenously administered at reperfusion. In addition, we investigated the mechanisms of cardioprotection in the Langendorff-perfused model. Cardioprotection was observed when DT1 was administered after myocardial injury. The molecular mechanisms involved the activation of the survival pathway PDK-1, AKT and AMPK, a reduced phosphorylation of PKD1/2 and sustained HIF-1α activity, leading to increased expression of anti-apoptotic proteins and decreased caspase-3 activation. Pharmacological inhibition of AKT following MI and prior to DT1 challenge significantly decreased the cardioprotection afforded by DT1 therapy at reperfusion. Cardiac function after MI was significantly improved after DT1-treatment, as evidenced by hemodynamic recovery and decreased myocardial infarct size. These findings demonstrate an efficient in vivo cardioprotection by diterpene DT1 against I/R when administered at reperfusion, opening new therapeutic strategies as adjunctive therapy for the pharmacological management of I/R injury.
Collapse
Affiliation(s)
- Irene Cuadrado-Berrocal
- Departamento de Farmacología, Facultad de Farmacia, Universidad Complutense de Madrid, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain
| | - María V Gómez-Gaviro
- Servicio de Cardiología, Instituto de Investigación Sanitaria Gregorio Marañón, C/ Doctor Esquerdo 46, 28007 Madrid, Spain; Unidad de Medicina y Cirugía Experimental, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain; Departamento de Bioingeniería e Ingeniería Aerospacial, Universidad Carlos III, Madrid, Spain
| | - Yolanda Benito
- Servicio de Cardiología, Instituto de Investigación Sanitaria Gregorio Marañón, C/ Doctor Esquerdo 46, 28007 Madrid, Spain
| | - Alicia Barrio
- Servicio de Cardiología, Instituto de Investigación Sanitaria Gregorio Marañón, C/ Doctor Esquerdo 46, 28007 Madrid, Spain
| | - Javier Bermejo
- Servicio de Cardiología, Instituto de Investigación Sanitaria Gregorio Marañón, C/ Doctor Esquerdo 46, 28007 Madrid, Spain
| | - María Eugenia Fernández-Santos
- Servicio de Cardiología, Instituto de Investigación Sanitaria Gregorio Marañón, C/ Doctor Esquerdo 46, 28007 Madrid, Spain
| | - Pedro L Sánchez
- Servicio de Cardiología, Instituto de Investigación Sanitaria Gregorio Marañón, C/ Doctor Esquerdo 46, 28007 Madrid, Spain
| | - Manuel Desco
- Unidad de Medicina y Cirugía Experimental, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain; Departamento de Bioingeniería e Ingeniería Aerospacial, Universidad Carlos III, Madrid, Spain
| | - Francisco Fernández-Avilés
- Servicio de Cardiología, Instituto de Investigación Sanitaria Gregorio Marañón, C/ Doctor Esquerdo 46, 28007 Madrid, Spain
| | - María Fernández-Velasco
- Instituto de Investigaciones Biomédicas 'Alberto Sols' (CSIC-UAM), Arturo Duperier 4, 28029 Madrid, Spain; Instituto de Investigación Hospital Universitario La PAZ, IDIPAZ, Paseo de la Castellana, 28029 Madrid, Spain
| | - Lisardo Boscá
- Instituto de Investigaciones Biomédicas 'Alberto Sols' (CSIC-UAM), Arturo Duperier 4, 28029 Madrid, Spain; Instituto de Investigación Hospital Universitario La PAZ, IDIPAZ, Paseo de la Castellana, 28029 Madrid, Spain.
| | - Beatriz de Las Heras
- Departamento de Farmacología, Facultad de Farmacia, Universidad Complutense de Madrid, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain.
| |
Collapse
|
22
|
Penna C, Pasqua T, Amelio D, Perrelli MG, Angotti C, Tullio F, Mahata SK, Tota B, Pagliaro P, Cerra MC, Angelone T. Catestatin increases the expression of anti-apoptotic and pro-angiogenetic factors in the post-ischemic hypertrophied heart of SHR. PLoS One 2014; 9:e102536. [PMID: 25099124 PMCID: PMC4123866 DOI: 10.1371/journal.pone.0102536] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Accepted: 06/19/2014] [Indexed: 12/15/2022] Open
Abstract
Background In the presence of comorbidities the effectiveness of many cardioprotective strategies is blunted. The goal of this study was to assess in a hypertensive rat model if the early reperfusion with anti-hypertensive and pro-angiogenic Chromogranin A-derived peptide, Catestatin (CST:hCgA352–372; CST-Post), protects the heart via Reperfusion-Injury-Salvage-Kinases (RISK)-pathway activation, limiting infarct-size and apoptosis, and promoting angiogenetic factors (e.g., hypoxia inducible factor, HIF-1α, and endothelial nitric oxide synthase, eNOS, expression). Methods and Results The effects of CST-Post on infarct-size, apoptosis and pro-angiogenetic factors were studied in isolated hearts of spontaneously hypertensive rats (SHR), which underwent the following protocols: (a) 30-min ischemia and 120-min reperfusion (I/R); (b) 30-min ischemia and 20-min reperfusion (I/R-short), both with and without CST-Post (75 nM for 20-min at the beginning of reperfusion). In unprotected Wistar-Kyoto hearts, used as normal counterpart, infarct-size resulted smaller than in SHR. CST-Post reduced significantly infarct-size and improved post-ischemic cardiac function in both strains. After 20-min reperfusion, CST-Post induced S-nitrosylation of calcium channels and phosphorylation of RISK-pathway in WKY and SHR hearts. Yet specific inhibitors of the RISK pathway blocked the CST-Post protective effects against infarct in the 120-min reperfusion groups. Moreover, apoptosis (evaluated by TUNEL, ARC and cleaved caspase) was reduced by CST-Post. Importantly, CST-Post increased expression of pro-angiogenetic factors (i.e., HIF-1α and eNOS expression) after two-hour reperfusion. Conclusions CST-Post limits reperfusion damages and reverses the hypertension-induced increase of I/R susceptibility. Moreover, CST-Post triggers antiapoptotic and pro-angiogenetic factors suggesting that CST-Post can be used as an anti-maladaptive remodeling treatment.
Collapse
Affiliation(s)
- Claudia Penna
- Department of Clinical and Biological Sciences, University of Turin, Torino, Italy
| | - Teresa Pasqua
- Department of Biology, Ecology and Earth Sciences, University of Calabria, Arcavacata di Rende, Italy
| | - Daniela Amelio
- Department of Biology, Ecology and Earth Sciences, University of Calabria, Arcavacata di Rende, Italy
| | | | - Carmelina Angotti
- Department of Clinical and Biological Sciences, University of Turin, Torino, Italy
| | - Francesca Tullio
- Department of Clinical and Biological Sciences, University of Turin, Torino, Italy
| | - Sushil K. Mahata
- VA San Diego Healthcare System, University of California, San Diego, California, United States of America
| | - Bruno Tota
- Department of Biology, Ecology and Earth Sciences, University of Calabria, Arcavacata di Rende, Italy
- National Institute of Cardiovascular Research, Bologna, Italy
| | - Pasquale Pagliaro
- Department of Clinical and Biological Sciences, University of Turin, Torino, Italy
- National Institute of Cardiovascular Research, Bologna, Italy
- * E-mail: (PP); (MCC)
| | - Maria C. Cerra
- Department of Biology, Ecology and Earth Sciences, University of Calabria, Arcavacata di Rende, Italy
- National Institute of Cardiovascular Research, Bologna, Italy
- * E-mail: (PP); (MCC)
| | - Tommaso Angelone
- Department of Biology, Ecology and Earth Sciences, University of Calabria, Arcavacata di Rende, Italy
- National Institute of Cardiovascular Research, Bologna, Italy
| |
Collapse
|
23
|
Role of hypoxia inducible factor-1α in remote limb ischemic preconditioning. J Mol Cell Cardiol 2013; 65:98-104. [PMID: 24140799 DOI: 10.1016/j.yjmcc.2013.10.001] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Revised: 10/04/2013] [Accepted: 10/04/2013] [Indexed: 11/23/2022]
Abstract
Remote ischemic preconditioning (RIPC) has emerged as a feasible and attractive therapeutic procedure for heart protection against ischemia/reperfusion (I/R) injury. However, its molecular mechanisms remain poorly understood. Hypoxia inducible factor-1α (HIF-1α) is a transcription factor that plays a key role in the cellular adaptation to hypoxia and ischemia. This study's aim was to test whether RIPC-induced cardioprotection requires HIF-1α upregulation to be effective. In the first study, wild-type mice and mice heterozygous for HIF1a (gene encoding the HIF-1α protein) were subjected to RIPC immediately before myocardial infarction (MI). RIPC resulted in a robust HIF-1α activation in the limb and acute cardioprotection in wild-type mice. RIPC-induced cardioprotection was preserved in heterozygous mice, despite the low HIF-1α expression in their limbs. In the second study, the role of HIF-1α in RIPC was evaluated using cadmium (Cd), a pharmacological HIF-1α inhibitor. Rats were subjected to MI (MI group) or to RIPC immediately prior to MI (R-MI group). Cd was injected 18 0min before RIPC (Cd-R-MI group). RIPC induced robust HIF-1α activation in rat limbs and significantly reduced infarct size (IS). Despite Cd's inhibition of HIF-1α activation, RIPC-induced cardioprotection was preserved in the Cd-R-MI group. RIPC applied immediately prior to MI increased HIF-1α expression and attenuated IS in rats and wild-type mice. However, RIPC-induced cardioprotection was preserved in partially HIF1a-deficient mice and in rats pretreated with Cd. When considered together, these results suggest that HIF-1α upregulation is unnecessary in acute RIPC.
Collapse
|
24
|
Remote ischemic preconditioning regulates HIF-1α levels, apoptosis and inflammation in heart tissue of cardiosurgical patients: a pilot experimental study. Basic Res Cardiol 2012. [PMID: 23203207 DOI: 10.1007/s00395-012-0314-0] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Transient episodes of ischemia in a remote organ (remote ischemic preconditioning, RIPC) bears the potential to attenuate myocardial injury, but the underlying mechanisms are only poorly understood. In the pilot experimental study presented we investigated cellular and molecular effects of RIPC in heart tissue of cardiosurgical patients with cardiopulmonary bypass (CPB) and focussed on apoptotic events, local and systemic inflammation as well as the regulation of the hypoxia induced factor-1α (HIF-1α). RIPC was induced by four 5-min cycles of transient upper limb ischemia/reperfusion using a blood-pressure cuff. Right atrial tissue and serum were obtained from patients receiving RIPC (N = 32) and control patients (N = 29) before and after CPB. RIPC patients showed reduced troponin T serum concentrations in the first 48 h after surgery (P < 0.05 vs. control) indicating cardioprotective effects of RIPC. Samples from RIPC patients that were collected before CPB contained significantly increased amounts of HIF-1α and procaspase-3 (HIF-1α: P < 0.05 vs. control, procaspase-3: P < 0.05 vs. control), whereas activities of caspases 3 and 7 were by trend reduced. Samples from RIPC patients that were taken after CPB showed an increased activity of myeloperoxidase (P < 0.05 vs. control; P < 0.05 vs. RIPC before CPB) as well as elevated tissue concentrations of the interleukin (IL)-1β (P < 0.05 vs. RIPC before CPB). Serum levels of IL-8, IL-1β and TNFα were significantly increased in RIPC patients before CPB (P < 0.05 vs. control before CPB). In summary, RIPC regulates HIF-1α levels, apoptosis and inflammation in the myocardium of cardiosurgical patients and leads to increased concentrations of circulating cytokines.
Collapse
|