1
|
Wu N, Chi J, Cai H, Hu J, Lai Y, Lin C, Kang L, Sun J, Huang J, Li M, Xu L. Traditional Chinese medication qili qiangxin capsule protects against myocardial ischemia-reperfusion injury through suppressing autophagy via the phosphoinositide 3-kinase/protein kinase B/forkhead box O3 axis. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118821. [PMID: 39265794 DOI: 10.1016/j.jep.2024.118821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 08/30/2024] [Accepted: 09/09/2024] [Indexed: 09/14/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Positive evidence from clinical trials highlights the promising potential of traditional Chinese medication, Qili qiangxin capsule (QLQX), on chronic heart failure; however, limited data are available regarding its effects and mechanism in myocardial ischemia-reperfusion injury (MIRI). Herein, we aimed to explore cardioprotective effects and the underlying mechanism of QLQX in MIRI in vivo and in vitro. MATERIALS AND METHODS Mice were subjected to left anterior descending coronary artery ligation for 30 min followed by 24 h of reperfusion with or without 7-day pretreatment with QLQX (0.234, 0.468, or 0.936 g/kg). Cardiac function, myocardial infarction, and morphological changes were evaluated. The mechanism underlying the cardio-protection of QLQX on MIRI was determined by network pharmacology based on the common genes of potential targets of QLQX and MIRI-related genes, further validated by H9c2 cardiomyocytes exposing hypoxia/reoxygenation (H/R). The viability, apoptosis, as well as autophagy and relevant signaling proteins in H9c2 were analyzed. RESULTS QLQX pretreatment markedly improved cardiac function and decreased myocardium infarct size, apoptotic cardiomyocyte number, and LHD, CK-MB, and TnT levels in MIRI mice. QLQX could mitigate H/R-induced H9c2 cardiomyocyte injury, as evidenced by decreased cell apoptosis and LDH release and increased ATP production. QLQX effectively attenuates excessive autophagy in cardiomyocytes both in vivo and in vitro. Mechanically, network pharmacology analysis demonstrated the cardio-protection of QLQX on MIRI involving in PI3K/Akt signaling; the effects of QLQX on H/R-induced H9c2 cardiomyocytes were abolished by a specific PI3K inhibitor. CONCLUSION QLQX protects against cardiomyocyte apoptosis and excessive autophagy via PI3K/Akt signaling during MIRI.
Collapse
Affiliation(s)
- Ningxia Wu
- Cardiovascular Department, The Affiliated Traditional Chinese Medicine Hospital, Guangzhou Medical University, Guangzhou, 510130, China; Department of Geriatric Cardiology, General Hospital of Southern Theatre Command, Guangzhou, 510040, China; Graduate School, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| | - Jianing Chi
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China.
| | - Hua Cai
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China.
| | - Jiaman Hu
- School of Public Health, Guangdong Pharmaceutical University, Guangzhou, 510315, China.
| | - Yingying Lai
- School of Public Health, Guangdong Pharmaceutical University, Guangzhou, 510315, China.
| | - Cailong Lin
- School of Public Health, Guangdong Pharmaceutical University, Guangzhou, 510315, China.
| | - Liang Kang
- Graduate School, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| | - Jingping Sun
- Graduate School, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| | - Jianyu Huang
- Department of Geriatric Cardiology, General Hospital of Southern Theatre Command, Guangzhou, 510040, China.
| | - Min Li
- Department of Geriatric Cardiology, General Hospital of Southern Theatre Command, Guangzhou, 510040, China.
| | - Lin Xu
- Department of Geriatric Cardiology, General Hospital of Southern Theatre Command, Guangzhou, 510040, China.
| |
Collapse
|
2
|
He F, Wang F, Xiang H, Ma Y, Lu Q, Xia Y, Zhou H, Wang Y, Ke J. Activation of adenosine A2B receptor alleviates myocardial ischemia-reperfusion injury by inhibiting endoplasmic reticulum stress and restoring autophagy flux. Arch Biochem Biophys 2024; 754:109945. [PMID: 38395121 DOI: 10.1016/j.abb.2024.109945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 02/10/2024] [Accepted: 02/20/2024] [Indexed: 02/25/2024]
Abstract
Myocardial ischemia-reperfusion injury (MIRI) poses a significant threat to patients with coronary heart disease. Adenosine A2A receptors have been known as a protective role in MIRI by regulating autophagy, so we assumed that activation of adenosine A2B receptor (A2BAR) might exert a similar effect during MIRI and underlying mechanism be related to proteostasis maintenance as well. In situ hearts were subjected to 30 min of ischemia and 120 min of reperfusion (IR), while invitro cardiomyocytes from neonatal rats experienced 6 h of oxygen-glucose deprivation followed by 12 h of reoxygenation (OGDR). Initially, we observed that post-ischemia-reperfusion induced autophagy flux blockade and ERS both in vivo and in vitro, evident through the increased expression of p62, LC3II, and BIP, which indicated the deteriorated proteostasis. We used a selective A2BAR agonist, Bay 60-6583, to explore the positive effects of A2BAR on cardiomyocytes and found that A2BAR activation rescued damaged cardiac function and morphological changes in the IR group and improved frail cell viability in the OGDR group. The A2BAR agonist also alleviated the blockage of autophagic flux, coupled with augmented ERS in the IR/OGDR group, which was reassured by using an autophagy inhibitor chloroquine (CQ) and ERS inhibitor (4-PBA) in vitro. Additionally, considering cAMP/PKA as a well-known downstream effector of A2BAR, we utilized H89, a selective PKA inhibitor. We observed that the positive efficacy of Bay 60-6583 was inhibited by H89. Collectively, our findings demonstrate that the A2BAR/cAMP/PKA signaling pathway exerts a protective role in MIRI by mitigating impaired autophagic flux and excessive ERS.
Collapse
Affiliation(s)
- Feng He
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Fuyu Wang
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Hanmin Xiang
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yunna Ma
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Qing Lu
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yun Xia
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Huimin Zhou
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yanlin Wang
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, China.
| | - Jianjuan Ke
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
3
|
Öz-Arslan D, Durer ZA, Kan B. G protein-coupled receptor-mediated autophagy in health and disease. Br J Pharmacol 2024. [PMID: 38501194 DOI: 10.1111/bph.16345] [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: 09/01/2023] [Revised: 01/05/2024] [Accepted: 01/27/2024] [Indexed: 03/20/2024] Open
Abstract
G protein-coupled receptors (GPCRs) constitute the largest and most diverse superfamily of mammalian transmembrane proteins. These receptors are involved in a wide range of physiological functions and are targets for more than a third of available drugs in the market. Autophagy is a cellular process involved in degrading damaged proteins and organelles and in recycling cellular components. Deficiencies in autophagy are involved in a variety of pathological conditions. Both GPCRs and autophagy are essential in preserving homeostasis and cell survival. There is emerging evidence suggesting that GPCRs are direct regulators of autophagy. Additionally, autophagic machinery is involved in the regulation of GPCR signalling. The interplay between GPCR and autophagic signalling mechanisms significantly impacts on health and disease; however, there is still an incomplete understanding of the underlying mechanisms and therapeutic implications in different tissues and disease contexts. This review aims to discuss the interactions between GPCR and autophagy signalling. Studies on muscarinic receptors, beta-adrenoceptors, taste receptors, purinergic receptors and adhesion GPCRs are summarized, in relation to autophagy.
Collapse
Affiliation(s)
- Devrim Öz-Arslan
- Department of Biophysics, Acibadem MAA University, School of Medicine, Istanbul, Turkey
| | - Zeynep Aslıhan Durer
- Department of Biophysics, Acibadem MAA University, School of Medicine, Istanbul, Turkey
- Department of Biochemistry, Acibadem MAA University, School of Pharmacy, Istanbul, Turkey
| | - Beki Kan
- Department of Biophysics, Acibadem MAA University, School of Medicine, Istanbul, Turkey
| |
Collapse
|
4
|
Yacouba Moukeila MB, Thokerunga E, He F, Bongolo CC, Xia Y, Wang F, Gado AF, Mamoudou H, Khan S, Ousseina B, Ousmane HA, Diarra D, Ke J, Zhang Z, Wang Y. Adenosine 2 receptor regulates autophagy and apoptosis to alleviate ischemia reperfusion injury in type 2 diabetes via IRE-1 signaling. BMC Cardiovasc Disord 2023; 23:154. [PMID: 36964482 PMCID: PMC10039586 DOI: 10.1186/s12872-023-03116-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 02/08/2023] [Indexed: 03/26/2023] Open
Abstract
PURPOSE This study aimed to determine the effect and mechanism of action of adenosine 2 receptor (A2R) activation on myocardial ischemia reperfusion injury (MIRI) under diabetic conditions. METHODS MIRI type 2 diabetic rats and H9C2 cardiomyocytes were treated with A2R agonist and then subjected to hypoxia for 6 h and reoxygenation for 18 h. Myocardial damage, and infarct size were determined by cardiac ultrasound. Indicators of cardiomyocyte injury, creatine kinase-MB and cardiac troponin I were detected by Enzyme Linked Immunosorbent Assay. Endoplasmic reticulum stress (ERS) was determined through measuring the expression levels of ERS related genes GRP78, p-IRE1/IRE1, and p-JNKJNK. The mechanism of A2R cardio protection in MIRI through regulating ERS induced autophagy was determined by investigating the ER resident protein IRE-1. The ER-stress inducer Tunicamycin, and the IRE-1 inhibitor STF in combination with the A2R agonist NECA were used, and the cellular responses were assessed through autophagy proteins expression Beclin-1, p62, LC3 and apoptosis. RESULTS NECA improved left ventricular function post MIRI, limited myocardial infarct size, reduced myocardial damage, decreased cardiomyocytes apoptosis, and attenuated ERS induced autophagy through regulating the IRE-XBP1s-CHOP pathway. These actions resulted into overall protection of the myocardium against MIRI. CONCLUSION In summary, A2R activation by NECA prior to ischemia attenuates apoptosis, reduces ERS induced autophagy and restores left ventricular function. This protective effect occurs through regulating the IRE1-XBPs-CHOP related mechanisms. NECA is thus a potential target for the treatment of MIRI in patient with type 2 diabetes.
Collapse
Affiliation(s)
| | - Erick Thokerunga
- Program and Department of Clinical Laboratory Medicine, Center for Gene Diagnosis, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China
| | - Feng He
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China
| | - Christian Cedric Bongolo
- Program and Department of Clinical Laboratory Medicine, Center for Gene Diagnosis, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China
| | - Yun Xia
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China
| | - Fuyu Wang
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China
| | - Adamou Foumakoye Gado
- Department of Anesthesia and Intensive Care, Hôpital Général de Référence Niamey, Niamey, Niger
| | - Hama Mamoudou
- Department of Anesthesia and Intensive Care, National Hospital of Niamey, Niamey, Niger
| | - Shahzad Khan
- Department of Pathology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China
| | - Bonkano Ousseina
- Department of Cardiovascular and Internal Medicine, Niamey Amirou Boubacar Diallo National Hospital, Abdou Moumouni University, Niamey, Niger
| | | | - Drissa Diarra
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Jianjuan Ke
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China.
| | - Zongze Zhang
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China.
| | - Yanlin Wang
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China.
| |
Collapse
|
5
|
Zhang W, Qiao W, Zuo L. A1 and A2b adenosine receptors regulate GPX4 against ferroptosis of cardiomyocytes in myocardial infarction rat model and in vitro. Tissue Cell 2022; 77:101828. [DOI: 10.1016/j.tice.2022.101828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 05/16/2022] [Accepted: 05/17/2022] [Indexed: 10/18/2022]
|
6
|
Xia Y, He F, Moukeila Yacouba MB, Zhou H, Li J, Xiong Y, Zhang J, Li H, Wang Y, Ke J. Adenosine A2a Receptor Regulates Autophagy Flux and Apoptosis to Alleviate Ischemia-Reperfusion Injury via the cAMP/PKA Signaling Pathway. Front Cardiovasc Med 2022; 9:755619. [PMID: 35571159 PMCID: PMC9099415 DOI: 10.3389/fcvm.2022.755619] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 03/23/2022] [Indexed: 12/12/2022] Open
Abstract
Exploring effective methods to lessen myocardial ischemia-reperfusion injury still has positive significance. The adenosine A2a receptor (A2aR) has played a crucial part in cardiac ischemia-reperfusion injury. Previous studies revealed that the adenosine A2a receptor regulated autophagy, but the specific mechanism in myocardial ischemia-reperfusion injury was still unclear. We established an ischemia-reperfusion model (30 min of ischemia and 2 h of reperfusion) in vivo and a model with oxygen-glucose deprivation for 6 h and reoxygenation for 18 h (OGDR) in vitro. The ischemia-reperfusion injury resulted in prolonged QTc interval, left ventricular systolic dysfunction, and myocardial infarction. In vitro model, we found that the OGDR-induced autophagosomes and apoptosis caused myocardial cell death, as evidenced by a significant increase in the generation of lactate dehydrogenase and creatine kinase-MB. Furthermore, overactivated autophagy with rapamycin showed an anti-apoptotic effect. The interaction between autophagy and apoptosis in myocardial ischemia-reperfusion injury was complex and variable. We discovered that the activation of adenosine A2a receptor could promote the expression of Bcl-2 to inhibit the levels of Beclin-1 and LC3II. The number of autophagosomes exceeded that of autolysosomes under OGDR, but the result reversed after A2aR activation. Activated A2aR with its agonist CGS21680 before reperfusion saved cellular survival through anti-apoptosis and anti-autophagy effect, thus improving ventricular contraction disorders, and visibly reducing myocardial infarction size. The myocardial protection of adenosine A2a receptor after ischemia may involve the cAMP-PKA signaling pathway and the interaction of Bcl-2-Beclin-1.
Collapse
|
7
|
Qin GW, Lu P, Peng L, Jiang W. Ginsenoside Rb1 Inhibits Cardiomyocyte Autophagy via PI3K/Akt/mTOR Signaling Pathway and Reduces Myocardial Ischemia/Reperfusion Injury. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2021; 49:1913-1927. [PMID: 34775933 DOI: 10.1142/s0192415x21500907] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Myocardial ischemia/reperfusion injury (MIRI) is the major cause of myocardial cell damage in acute myocardial infarction, and its treatment remains a clinical challenge. Ginsenoside Rb1 showed protective effects on the cardiovascular system; however, the underlying mechanism remains largely unclear. Effects of Ginsenoside Rb1 on rat MIRI-induced myocardial infarct size were evaluated through TTC staining. TUNEL assay and flow cytometry analysis were employed to estimate cell apoptosis. Apoptosis, autophagy and PI3K/Akt/mTOR pathway-related proteins were estimated via western blot. Expression of Beclin1 in myocardial tissues were examined by immunohistochemical analysis. Expression levels of IL-1[Formula: see text], TNF-[Formula: see text] and IL-6 were tested by enzyme-linked immunosorbent assay (ELISA). Here, we found that Ginsenoside Rb1 treatment not only alleviated MIRI in rats but also protected H9C2 cells against hypoxia/reoxygenation induced damage. Ginsenoside Rb1 abolished the MIRI-induced activation of autophagy. Meanwhile, we found that treatment of 3-MA (autophagy inhibitor) could enhance the protective effects of Ginsenoside Rb1 on H9C2 cells during H/R. Moreover, Ginsenoside Rb1 treatment resulted in the activation of the PI3K/Akt/mTOR pathway, and treatment of LY294002 (PI3K/Akt pathway repressor) abolished the protective effects of Ginsenoside Rb1 on myocardial in vitro and in vivo. Our results suggest that Ginsenoside Rb1 functions as a protector against MIRI by repressing cardiomyocyte autophagy through the PI3K/Akt/mTOR signaling pathway.
Collapse
Affiliation(s)
- Guo-Wei Qin
- Department of Science and Technology, Guilin Medical University, Guilin 541001, Guangxi Zhuang Autonomous Region, P. R. China
| | - Pan Lu
- Department of Cardiology, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi Zhuang Autonomous Region, P. R. China
| | - Li Peng
- Department of Cardiology, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi Zhuang Autonomous Region, P. R. China
| | - Wei Jiang
- Department of Traditional Chinese Medicine, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi Zhuang Autonomous Region, P. R. China
| |
Collapse
|
8
|
Ding HS, Yang J, Yang J, Guo X, Tang YH, Huang Y, Chen Z, Fan ZX, Huang CX. Fluvastatin attenuated ischemia/reperfusion-induced autophagy and apoptosis in cardiomyocytes through down-regulation HMGB1/TLR4 signaling pathway. Mol Biol Rep 2021; 48:3893-3901. [PMID: 34032975 DOI: 10.1007/s11033-021-06326-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 03/31/2021] [Indexed: 11/10/2022]
Abstract
Fluvastatin, a traditional fat-decreasing drug, is widely used for curing cardiovascular disease. Previous reports demonstrated that fluvastatin pretreatment protected against myocardial ischemia/reperfusion (I/R) by inhibiting TLR4 signaling pathway and/or reducing proinflammatory cytokines. However, whether fluvastatin has a cardioprotective effect against apoptosis and autophagy remains unknown. This study aims to evaluate whether the cardioprotective role of fluvastatin in I/R is mediated by high-mobility group box 1 (HMGB1)/toll-like receptor 4 (TLR4) pathway via anti-apoptotic and anti-autophagic functions. Sprague-Dawley rats were anesthetized, artificially ventilated and subjected to 30 min of coronary occlusion, followed by 4 h of reperfusion. The animals were randomized into four groups: (i) Sham operation; (ii) I/R; (iii) I/R + low-dosage fluvastatin (10 mg/kg); and (iv) I/R + high-dosage fluvastatin (20 mg/kg). After reperfusion, the hemodynamic parameters, myocardial infarct size, structural alteration of myocardium, apoptosis index, pro-inflammatory cytokine production, Beclin-1, Light chain 3 (LC3), HMGB1, TLR4 and Nuclear factor kappa B (NF-κB) protein levels were measured and recorded. It was found that fluvastatin preconditioning improved left ventricular dysfunction, reduced HMGB1/TLR4/NF-κB expressions, and inhibited cardiomyocyte apoptosis, autophagy, and inflammation reaction. Moreover, treatment with fluvastatin ameliorated myocardial injury by reducing infarct size, causing less damage to cardiac structure, downregulating autophagy-related protein expression and releasing pro-inflammation mediators. Our findings indicate that fluvastatin exerts beneficial effects on cardiac ischemic damage, which may be associated with its anti-autophagic and anti-apoptotic functions via inhibition of HMGB1/TLR4-related pathway during I/R injury.
Collapse
Affiliation(s)
- Hua-Sheng Ding
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, 430060, People's Republic of China.,Institute of Cardiovascular Diseases, Wuhan University, Wuhan, 430060, People's Republic of China.,Hubei Key Laboratory of Cardiology, Wuhan, 430060, People's Republic of China
| | - Jun Yang
- Institute of Cardiovascular Diseases, China Three Gorges University, Yichang, 443000, People's Republic of China
| | - Jian Yang
- Institute of Cardiovascular Diseases, China Three Gorges University, Yichang, 443000, People's Republic of China
| | - Xin Guo
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, 430060, People's Republic of China.,Institute of Cardiovascular Diseases, Wuhan University, Wuhan, 430060, People's Republic of China.,Hubei Key Laboratory of Cardiology, Wuhan, 430060, People's Republic of China
| | - Yan-Hong Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, 430060, People's Republic of China.,Institute of Cardiovascular Diseases, Wuhan University, Wuhan, 430060, People's Republic of China.,Hubei Key Laboratory of Cardiology, Wuhan, 430060, People's Republic of China
| | - Yan Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, 430060, People's Republic of China.,Institute of Cardiovascular Diseases, Wuhan University, Wuhan, 430060, People's Republic of China.,Hubei Key Laboratory of Cardiology, Wuhan, 430060, People's Republic of China
| | - Zhen Chen
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, 430060, People's Republic of China.,Institute of Cardiovascular Diseases, Wuhan University, Wuhan, 430060, People's Republic of China.,Hubei Key Laboratory of Cardiology, Wuhan, 430060, People's Republic of China
| | - Zhi-Xing Fan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, 430060, People's Republic of China.,Institute of Cardiovascular Diseases, Wuhan University, Wuhan, 430060, People's Republic of China.,Hubei Key Laboratory of Cardiology, Wuhan, 430060, People's Republic of China
| | - Cong-Xin Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, 430060, People's Republic of China. .,Institute of Cardiovascular Diseases, Wuhan University, Wuhan, 430060, People's Republic of China. .,Hubei Key Laboratory of Cardiology, Wuhan, 430060, People's Republic of China.
| |
Collapse
|
9
|
Fu M, Xie D, Sun Y, Pan Y, Zhang Y, Chen X, Shi Y, Deng S, Cheng B. Exosomes derived from MSC pre-treated with oridonin alleviates myocardial IR injury by suppressing apoptosis via regulating autophagy activation. J Cell Mol Med 2021; 25:5486-5496. [PMID: 33955654 PMCID: PMC8184716 DOI: 10.1111/jcmm.16558] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 03/26/2021] [Accepted: 04/01/2021] [Indexed: 12/16/2022] Open
Abstract
This study aimed to investigate the molecular mechanisms underlying the role of bone marrow mesenchymal stem cells (BMMSCs)‐derived exosomes in ischaemia/reperfusion (IR)‐induced damage, and the role of oridonin in the treatment of IR. Exosomes were isolated from BMMSCs. Western blot analysis was done to examine the expression of proteins including CD63, CD8, apoptotic‐linked gene product 2 interacting protein X (AliX), Beclin‐1, ATG13, B‐cell lymphoma‐2 (Bcl‐2), apoptotic peptidase activating factor 1 (Apaf1) and Bcl2‐associated X (Bax) in different treatment groups. Accordingly, the expression of CD63, CD81 and AliX was higher in BMMSCs‐EXOs and IR + BMMSCs‐EXOs + ORI groups compared with that in the BMMSCs group. And BMMSCs‐derived exosomes inhibited the progression of IR‐induced myocardial damage, while this protective effect was boosted by the pre‐treatment with oridonin. Moreover, Beclin‐1, ATG13 and Bcl‐2 were significantly down‐regulated while Apaf1 and Bax were significantly up‐regulated in IR rats. And the presence of BMMSCs‐derived exosomes partly alleviated IR‐induced dysregulation of these proteins, while the oridonin pre‐treatment boosted the effect of these BMMSCs‐derived exosomes. The inhibited proliferation and promoted apoptosis of H9c2 cells induced by hypoxia/reperfusion (HR) were mitigated by the administration of BMMSCs‐derived exosomes. Meanwhile, HR also induced down‐regulation of Beclin‐1, ATG13 and Bcl‐2 expression and up‐regulation of Apaf1 and Bax, which were mitigated by the administration of BMMSCs‐derived exosomes. And oridonin pre‐treatment boosted the effect of BMMSCs‐derived exosomes. In conclusion, our results validated that BMMSCs‐derived exosomes suppressed the IR‐induced damages by participating in the autophagy process, while the pre‐treatment with oridonin could boost the protective effect of BMMSCs‐derived exosomes.
Collapse
Affiliation(s)
- Minghuan Fu
- Department of Geriatric Cardiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Dili Xie
- Department of Geriatric Cardiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Ying Sun
- Department of Geriatric Cardiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Yuanyuan Pan
- Department of Geriatric Cardiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Yunhe Zhang
- Department of Geriatric Cardiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Xiaohan Chen
- Department of Geriatric Cardiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Yong Shi
- Department of Geriatric Cardiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Shengnan Deng
- Department of Geriatric Cardiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Biao Cheng
- Department of Geriatric Cardiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
10
|
Boknik P, Eskandar J, Hofmann B, Zimmermann N, Neumann J, Gergs U. Role of Cardiac A 2A Receptors Under Normal and Pathophysiological Conditions. Front Pharmacol 2021; 11:627838. [PMID: 33574762 PMCID: PMC7871008 DOI: 10.3389/fphar.2020.627838] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 12/15/2020] [Indexed: 12/12/2022] Open
Abstract
This review presents an overview of cardiac A2A-adenosine receptors The localization of A2A-AR in the various cell types that encompass the heart and the role they play in force regulation in various mammalian species are depicted. The putative signal transduction systems of A2A-AR in cells in the living heart, as well as the known interactions of A2A-AR with membrane-bound receptors, will be addressed. The possible role that the receptors play in some relevant cardiac pathologies, such as persistent or transient ischemia, hypoxia, sepsis, hypertension, cardiac hypertrophy, and arrhythmias, will be reviewed. Moreover, the cardiac utility of A2A-AR as therapeutic targets for agonistic and antagonistic drugs will be discussed. Gaps in our knowledge about the cardiac function of A2A-AR and future research needs will be identified and formulated.
Collapse
Affiliation(s)
- P. Boknik
- Institut für Pharmakologie und Toxikologie, Medizinische Fakultät, Westfälische Wilhelms-Universität, Münster, Germany
| | - J. Eskandar
- Institut für Pharmakologie und Toxikologie, Medizinische Fakultät, Westfälische Wilhelms-Universität, Münster, Germany
| | - B. Hofmann
- Cardiac Surgery, Medizinische Fakultät, Martin-Luther-Universität Halle-Wittenberg, Halle, Germany
| | - N. Zimmermann
- Bundesinstitut für Arzneimittel und Medizinprodukte, Bonn, Germany
| | - J. Neumann
- Institut für Pharmakologie und Toxikologie, Medizinische Fakultät, Martin-Luther-Universität Halle-Wittenberg, Halle, Germany
| | - U. Gergs
- Institut für Pharmakologie und Toxikologie, Medizinische Fakultät, Martin-Luther-Universität Halle-Wittenberg, Halle, Germany
| |
Collapse
|
11
|
Bellis A, Mauro C, Barbato E, Di Gioia G, Sorriento D, Trimarco B, Morisco C. The Rationale of Neprilysin Inhibition in Prevention of Myocardial Ischemia-Reperfusion Injury during ST-Elevation Myocardial Infarction. Cells 2020; 9:cells9092134. [PMID: 32967374 PMCID: PMC7565478 DOI: 10.3390/cells9092134] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 09/17/2020] [Accepted: 09/18/2020] [Indexed: 12/11/2022] Open
Abstract
During the last three decades, timely myocardial reperfusion using either thrombolytic therapy or primary percutaneous intervention (pPCI) has allowed amazing improvements in outcomes with a more than halving in 1-year ST-elevation myocardial infarction (STEMI) mortality. However, mortality and left ventricle (LV) remodeling remain substantial in these patients. As such, novel therapeutic interventions are required to reduce myocardial infarction size, preserve LV systolic function, and improve survival in reperfused-STEMI patients. Myocardial ischemia-reperfusion injury (MIRI) prevention represents the main goal to reach in order to reduce STEMI mortality. There is currently no effective therapy for MIRI prevention in STEMI patients. A significant reason for the weak and inconsistent results obtained in this field may be the presence of multiple, partially redundant, mechanisms of cell death during ischemia-reperfusion, whose relative importance may depend on the conditions. Therefore, it is always more recognized that it is important to consider a "multi-targeted cardioprotective therapy", defined as an additive or synergistic cardioprotective agents or interventions directed to distinct targets with different timing of application (before, during, or after pPCI). Given that some neprilysin (NEP) substrates (natriuretic peptides, angiotensin II, bradykinin, apelins, substance P, and adrenomedullin) exert a cardioprotective effect against ischemia-reperfusion injury, it is conceivable that antagonism of proteolytic activity by this enzyme may be considered in a multi-targeted strategy for MIRI prevention. In this review, by starting from main pathophysiological mechanisms promoting MIRI, we discuss cardioprotective effects of NEP substrates and the potential benefit of NEP pharmacological inhibition in MIRI prevention.
Collapse
Affiliation(s)
- Alessandro Bellis
- Dipartimento di Scienze Biomediche Avanzate, Università FEDERICO II, 80131 Napoli, Italy; (A.B.); (E.B.); (G.D.G.); (D.S.); (B.T.)
- Unità Operativa Complessa Cardiologia con UTIC ed Emodinamica—Dipartimento Emergenza Accettazione, Azienda Ospedaliera “Antonio Cardarelli”, 80131 Napoli, Italy;
| | - Ciro Mauro
- Unità Operativa Complessa Cardiologia con UTIC ed Emodinamica—Dipartimento Emergenza Accettazione, Azienda Ospedaliera “Antonio Cardarelli”, 80131 Napoli, Italy;
| | - Emanuele Barbato
- Dipartimento di Scienze Biomediche Avanzate, Università FEDERICO II, 80131 Napoli, Italy; (A.B.); (E.B.); (G.D.G.); (D.S.); (B.T.)
| | - Giuseppe Di Gioia
- Dipartimento di Scienze Biomediche Avanzate, Università FEDERICO II, 80131 Napoli, Italy; (A.B.); (E.B.); (G.D.G.); (D.S.); (B.T.)
- Cardiac Catheterization Laboratory, Montevergine Clinic, 83013 Mercogliano (AV), Italy
| | - Daniela Sorriento
- Dipartimento di Scienze Biomediche Avanzate, Università FEDERICO II, 80131 Napoli, Italy; (A.B.); (E.B.); (G.D.G.); (D.S.); (B.T.)
| | - Bruno Trimarco
- Dipartimento di Scienze Biomediche Avanzate, Università FEDERICO II, 80131 Napoli, Italy; (A.B.); (E.B.); (G.D.G.); (D.S.); (B.T.)
| | - Carmine Morisco
- Dipartimento di Scienze Biomediche Avanzate, Università FEDERICO II, 80131 Napoli, Italy; (A.B.); (E.B.); (G.D.G.); (D.S.); (B.T.)
- Correspondence: ; Tel.: +39-081-746-2253; Fax: +39-081-746-2256
| |
Collapse
|
12
|
Inhibition of A 2B Adenosine Receptor Attenuates Intestinal Injury in a Rat Model of Necrotizing Enterocolitis. Mediators Inflamm 2020; 2020:1562973. [PMID: 32714089 PMCID: PMC7354672 DOI: 10.1155/2020/1562973] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 06/06/2020] [Accepted: 06/17/2020] [Indexed: 12/27/2022] Open
Abstract
Necrotizing enterocolitis (NEC) is a lethal gastrointestinal tract disease that occurs in premature infants. Adenosine receptor A2B (A2BR) regulates the inflammation cytokine secretion and immune cell infiltration in the colonic pathophysiology conditions. In the present study, we aim to determine the roles of A2BR in the development of NEC. A NEC rat model was established and treated with A2BR agonist-BAY60-6583 or A2BR antagonist-PSB1115. Animals in the control group were free from any interventions. Our results showed that the inhibition of A2BR PSB1115 improved intestinal injury and inflammation in newborn NEC rats. The expression levels of caspase-3 and the ratio of apoptotic cells were upregulated in NEC rats, and these indices were downregulated after treating with PSB1115 but further upregulated by BAY60-6583. Meanwhile, a similar trend was also witnessed in the changes of MPO activities and proinflammatory cytokines including IL-6, IFN-γ, and TNF-α. However, the anti-inflammatory cytokine IL-10 in the NECP group was significantly higher than that in the NEC and NECB groups (p < 0.05, respectively). Moreover, the expression of Ki67 was significantly increased in the NECP group as compared with those of the NEC and the NECB groups (p < 0.05, respectively). Collectively, our study suggested that the inhibition of A2BR attenuates NEC in the neonatal rat, at least partially through the modulation of inflammation and the induction of epithelial cell proliferation.
Collapse
|
13
|
Li X, Hu X, Wang J, Xu W, Yi C, Ma R, Jiang H. Inhibition of autophagy via activation of PI3K/Akt/mTOR pathway contributes to the protection of hesperidin against myocardial ischemia/reperfusion injury. Int J Mol Med 2018; 42:1917-1924. [PMID: 30066841 PMCID: PMC6108872 DOI: 10.3892/ijmm.2018.3794] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Accepted: 07/24/2018] [Indexed: 01/21/2023] Open
Abstract
Hesperidin has been reported to attenuate myocardial ischemia/reperfusion (I/R) injury; however, its effect on autophagy during myocardial I/R and the underlying mechanism remains unknown. The present study aimed to investigate whether hesperidin inhibited I/R-induced excessive myocardial autophagy through activating the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt)/mammalian target of rapamycin (mTOR) pathway. Male adult rats were pretreated with hesperidin for a total of 3 days prior to ischemia in the absence or presence of LY294002, a PI3K inhibitor, and then subjected to ischemia for 30 min followed by reperfusion for 4 h. Myocardial infarct size was measured by Evans blue/triphenyltetrazolium chloride staining. Hematoxylin and eosin staining was used for observing the histological changes in the heart, and the serum levels of creatine kinase-MB (CK-MB) and cardiac troponin I (cTnI) were measured by enzyme-linked immunosorbent assay. Additionally, the protein levels of light chain (LC) 3II, Beclin1, phosphorylated (p)-mTOR, p-Akt and p-PI3K were determined by western blot analysis. Hesperidin pretreatment significantly decreased the myocardial infarct size, myocardial damage and serum levels of CK-MB and cTnI. Furthermore, the expression levels of LC3II and Beclin1 were significantly downregulated and the expression levels of p-mTOR, p-Akt and p-PI3K were markedly upregulated by hesperidin. However, the aforementioned effects as a result of hesperidin were significantly reversed by the presence of LY294002. These results demonstrated that hesperidin reduced myocardial I/R injury by suppressing excessive autophagy. Activation of the PI3K/Akt/mTOR pathway contributed to the inhibitory effect of hesperidin on excessive autophagy.
Collapse
Affiliation(s)
- Xuefei Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Xiaorong Hu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Jichun Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Weipan Xu
- Department of Cardiology, Huangshi Center Hospital, Affiliated Hospital of Hubei Polytechnic University, Huangshi, Hubei 435000, P.R. China
| | - Chunfeng Yi
- Department of Cardiology, Wuhan No. 1 Hospital, Wuhan, Hubei 430060, P.R. China
| | - Ruisong Ma
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Hong Jiang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
14
|
Lin XL, Xiao WJ, Xiao LL, Liu MH. Molecular mechanisms of autophagy in cardiac ischemia/reperfusion injury (Review). Mol Med Rep 2018; 18:675-683. [PMID: 29845269 DOI: 10.3892/mmr.2018.9028] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 05/10/2018] [Indexed: 11/05/2022] Open
Abstract
Autophagy is a maintenance process for recycling long-lived proteins and cytoplasmic organelles. The level of this process is enhanced during ischemia/reperfusion (I/R) injury. Autophagy can trigger survival signaling in myocardial ischemia, whereas defective autophagy during reperfusion is detrimental. Autophagy can be regulated through multiple signaling pathways in I/R, including Beclin‑1/class III phosphatidylinositol‑3 kinase (PI‑3K), adenosine monophosphate activated protein kinase/mammalian target of rapamycin (mTOR), and PI‑3K/protein kinase B/mTOR pathways, which consequently lead to different functions. Thus, autophagy has both protective and detrimental functions, which are determined by different signaling pathways and conditions. Targeting the activation of autophagy can be a promising new therapeutic strategy for treating cardiovascular disease.
Collapse
Affiliation(s)
- Xiao-Long Lin
- Department of Pathology, Hui Zhou Third People's Hospital, Guangzhou Medical University, Huizhou, Guangdong 516002, P.R. China
| | - Wei-Jin Xiao
- Department of Pathology, The Central Hospital of Shaoyang, Hunan 422000, P.R. China
| | - Le-Le Xiao
- School of Medicine, Huzhou University, Huzhou, Zhejiang 313000, P.R. China
| | - Mi-Hua Liu
- Department of Infectious Diseases, Centre for Lipid Research and Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400016, P.R. China
| |
Collapse
|
15
|
Impaired autophagic flux is associated with the severity of trauma and the role of A 2AR in brain cells after traumatic brain injury. Cell Death Dis 2018; 9:252. [PMID: 29449536 PMCID: PMC5833790 DOI: 10.1038/s41419-018-0316-4] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 12/10/2017] [Accepted: 01/12/2018] [Indexed: 02/06/2023]
Abstract
Recent studies have shown that after traumatic brain injury (TBI), the number of autophagosomes is markedly increased in brain cells surrounding the wound; however, whether autophagy is enhanced or suppressed by TBI remains controversial. In our study, we used a controlled cortical impact system to establish models of mild, moderate and severe TBI. In the mild TBI model, the levels of autophagy-related protein 6 (Beclin1) and autophagy-related protein 12 (ATG12)-autophagy-related protein 5 (ATG5) conjugates were increased, indicating the enhanced initiation of autophagy. Furthermore, the level of the autophagic substrate sequestosome 1 (SQSTM1) was decreased in the ipsilateral cortex. This result, together with the results observed in tandem mRFP-GFP-LC3 adeno-associated virus (AAV)-infected mice, indicates that autophagosome clearance was also increased after mild TBI. Conversely, following moderate and severe TBI, there was no change in the initiation of autophagy, and autophagosome accumulation was observed. Next, we used chloroquine (CQ) to artificially impair autophagic flux in the injured cortex of the mild TBI model and found that the severity of trauma was obviously exacerbated. In addition, autophagic flux and trauma severity were significantly improved in adenosine A2A receptor (A2AR) knockout (KO) mice subjected to moderate TBI. Thus, A2AR may be involved in regulating the impairment of autophagic flux in response to brain injury. Our findings suggest that whether autophagy is increased after TBI is associated with whether autophagic flux is impaired, and the impairment of autophagic flux exacerbates the severity of trauma. Furthermore, A2AR may be a target for alleviating the impairment in autophagic flux after TBI.
Collapse
|
16
|
Xing F, Han H, He Y, Zhang Y, Jing L, Xu Z, Xi J. Roles of Endoplasmic Reticulum Stress in NECA-Induced Cardioprotection against Ischemia/Reperfusion Injury. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:2490501. [PMID: 29391923 PMCID: PMC5748120 DOI: 10.1155/2017/2490501] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2017] [Revised: 08/26/2017] [Accepted: 09/14/2017] [Indexed: 12/19/2022]
Abstract
OBJECTIVE This study aimed to investigate whether the nonselective A2 adenosine receptor agonist NECA induces cardioprotection against myocardial ischemia/reperfusion (I/R) injury via glycogen synthase kinase 3β (GSK-3β) and the mitochondrial permeability transition pore (mPTP) through inhibition of endoplasmic reticulum stress (ERS). METHODS AND RESULTS H9c2 cells were exposed to H2O2 for 20 minutes. NECA significantly prevented H2O2-induced TMRE fluorescence reduction, indicating that NECA inhibited the mPTP opening. NECA blocked H2O2-induced GSK-3β phosphorylation and GRP94 expression. NECA increased GSK-3β phosphorylation and decreased GRP94 expression, which were prevented by both ERS inductor 2-DG and PKG inhibitor KT5823, suggesting that NECA may induce cardioprotection through GSK-3β and cGMP/PKG via ERS. In isolated rat hearts, both NECA and the ERS inhibitor TUDCA decreased myocardial infarction, increased GSK-3β phosphorylation, and reversed GRP94 expression at reperfusion, suggesting that NECA protected the heart by inhibiting GSK-3β and ERS. Transmission electron microscopy showed that NECA and TUDCA reduced mitochondrial swelling and endoplasmic reticulum expansion, further supporting that NECA protected the heart by preventing the mPTP opening and ERS. CONCLUSION These data suggest that NECA prevents the mPTP opening through inactivation of GSK-3β via ERS inhibition. The cGMP/PKG signaling pathway is responsible for GSK-3β inactivation by NECA.
Collapse
Affiliation(s)
- Fengmei Xing
- College of Nursing and Rehabilitation, North China University of Science and Technology, Tangshan 063000, China
| | - Hui Han
- College of Nursing and Rehabilitation, North China University of Science and Technology, Tangshan 063000, China
| | - Yonggui He
- Heart Institute, North China University of Science and Technology, Tangshan 063000, China
| | - Yidong Zhang
- Heart Institute, North China University of Science and Technology, Tangshan 063000, China
| | - Liwei Jing
- College of Nursing and Rehabilitation, North China University of Science and Technology, Tangshan 063000, China
| | - Zhelong Xu
- Heart Institute, North China University of Science and Technology, Tangshan 063000, China
| | - Jinkun Xi
- Heart Institute, North China University of Science and Technology, Tangshan 063000, China
| |
Collapse
|
17
|
Kloner RA, Brown DA, Csete M, Dai W, Downey JM, Gottlieb RA, Hale SL, Shi J. New and revisited approaches to preserving the reperfused myocardium. Nat Rev Cardiol 2017; 14:679-693. [PMID: 28748958 PMCID: PMC5991096 DOI: 10.1038/nrcardio.2017.102] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Early coronary artery reperfusion improves outcomes for patients with ST-segment elevation myocardial infarction (STEMI), but morbidity and mortality after STEMI remain unacceptably high. The primary deficits seen in these patients include inadequate pump function, owing to rapid infarction of muscle in the first few hours of treatment, and adverse remodelling of the heart in the months that follow. Given that attempts to further reduce myocardial infarct size beyond early reperfusion in clinical trials have so far been disappointing, effective therapies are still needed to protect the reperfused myocardium. In this Review, we discuss several approaches to preserving the reperfused heart, such as therapies that target the mechanisms involved in mitochondrial bioenergetics, pyroptosis, and autophagy, as well as treatments that harness the cardioprotective properties of inhaled anaesthetic agents. We also discuss potential therapies focused on correcting the no-reflow phenomenon and its effect on healing and adverse left ventricular remodelling.
Collapse
Affiliation(s)
- Robert A Kloner
- Cardiovascular Research Institute, Huntington Medical Research Institutes, 99 North El Molino Avenue, Pasadena, California 91101, USA
- Division of Cardiovascular Medicine and Department of Medicine, Keck School of Medicine, University of Southern California, 1975 Zonal Avenue, Los Angeles, California 90033, USA
| | - David A Brown
- Department of Human Nutrition, Foods, and Exercise, 1981 Kraft Drive, Blacksburg, Virginia 24060, USA
- Virginia Tech Center for Drug Discovery, Virginia Tech, 1981 Kraft Drive, Blacksburg, Virginia 24060, USA
- Virginia Tech Metabolic Phenotyping Core, Virginia Tech, 1981 Kraft Drive, Blacksburg, Virginia 24060, USA
| | - Marie Csete
- Cardiovascular Research Institute, Huntington Medical Research Institutes, 99 North El Molino Avenue, Pasadena, California 91101, USA
- Department of Anesthesiology, Keck School of Medicine, University of Southern California, Los Angeles, California 90017, USA
| | - Wangde Dai
- Cardiovascular Research Institute, Huntington Medical Research Institutes, 99 North El Molino Avenue, Pasadena, California 91101, USA
- Division of Cardiovascular Medicine and Department of Medicine, Keck School of Medicine, University of Southern California, 1975 Zonal Avenue, Los Angeles, California 90033, USA
| | - James M Downey
- Department of Physiology and Cell Biology, University of South Alabama, 5851 USA Drive North, Mobile, Alabama 36688, USA
| | - Roberta A Gottlieb
- Department of Medicine, Barbra Streisand Women's Heart Center, Heart Institute of Cedars-Sinai, Cedars-Sinai Medical Center, 127 South San Vicente Boulevard, Los Angeles, California 90048, USA
| | - Sharon L Hale
- Cardiovascular Research Institute, Huntington Medical Research Institutes, 99 North El Molino Avenue, Pasadena, California 91101, USA
| | - Jianru Shi
- Cardiovascular Research Institute, Huntington Medical Research Institutes, 99 North El Molino Avenue, Pasadena, California 91101, USA
- Division of Cardiovascular Medicine and Department of Medicine, Keck School of Medicine, University of Southern California, 1975 Zonal Avenue, Los Angeles, California 90033, USA
| |
Collapse
|
18
|
Burnstock G. Purinergic Signalling: Therapeutic Developments. Front Pharmacol 2017; 8:661. [PMID: 28993732 PMCID: PMC5622197 DOI: 10.3389/fphar.2017.00661] [Citation(s) in RCA: 281] [Impact Index Per Article: 40.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 09/05/2017] [Indexed: 12/15/2022] Open
Abstract
Purinergic signalling, i.e., the role of nucleotides as extracellular signalling molecules, was proposed in 1972. However, this concept was not well accepted until the early 1990's when receptor subtypes for purines and pyrimidines were cloned and characterised, which includes four subtypes of the P1 (adenosine) receptor, seven subtypes of P2X ion channel receptors and 8 subtypes of the P2Y G protein-coupled receptor. Early studies were largely concerned with the physiology, pharmacology and biochemistry of purinergic signalling. More recently, the focus has been on the pathophysiology and therapeutic potential. There was early recognition of the use of P1 receptor agonists for the treatment of supraventricular tachycardia and A2A receptor antagonists are promising for the treatment of Parkinson's disease. Clopidogrel, a P2Y12 antagonist, is widely used for the treatment of thrombosis and stroke, blocking P2Y12 receptor-mediated platelet aggregation. Diquafosol, a long acting P2Y2 receptor agonist, is being used for the treatment of dry eye. P2X3 receptor antagonists have been developed that are orally bioavailable and stable in vivo and are currently in clinical trials for the treatment of chronic cough, bladder incontinence, visceral pain and hypertension. Antagonists to P2X7 receptors are being investigated for the treatment of inflammatory disorders, including neurodegenerative diseases. Other investigations are in progress for the use of purinergic agents for the treatment of osteoporosis, myocardial infarction, irritable bowel syndrome, epilepsy, atherosclerosis, depression, autism, diabetes, and cancer.
Collapse
Affiliation(s)
- Geoffrey Burnstock
- Autonomic Neuroscience Centre, University College Medical SchoolLondon, United Kingdom
- Department of Pharmacology and Therapeutics, The University of Melbourne, MelbourneVIC, Australia
| |
Collapse
|
19
|
Hypoxic postconditioning attenuates apoptosis via activation of adenosine A 2a receptors on dermal microvascular endothelial cells of human flaps. J Surg Res 2017; 217:144-152. [DOI: 10.1016/j.jss.2017.05.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2017] [Revised: 04/08/2017] [Accepted: 05/02/2017] [Indexed: 11/23/2022]
|
20
|
Burnstock G. Purinergic Signaling in the Cardiovascular System. Circ Res 2017; 120:207-228. [PMID: 28057794 DOI: 10.1161/circresaha.116.309726] [Citation(s) in RCA: 281] [Impact Index Per Article: 40.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Revised: 11/21/2016] [Accepted: 11/23/2016] [Indexed: 02/07/2023]
Abstract
There is nervous control of the heart by ATP as a cotransmitter in sympathetic, parasympathetic, and sensory-motor nerves, as well as in intracardiac neurons. Centers in the brain control heart activities and vagal cardiovascular reflexes involve purines. Adenine nucleotides and nucleosides act on purinoceptors on cardiomyocytes, AV and SA nodes, cardiac fibroblasts, and coronary blood vessels. Vascular tone is controlled by a dual mechanism. ATP, released from perivascular sympathetic nerves, causes vasoconstriction largely via P2X1 receptors. Endothelial cells release ATP in response to changes in blood flow (via shear stress) or hypoxia, to act on P2 receptors on endothelial cells to produce nitric oxide, endothelium-derived hyperpolarizing factor, or prostaglandins to cause vasodilation. ATP is also released from sensory-motor nerves during antidromic reflex activity, to produce relaxation of some blood vessels. Purinergic signaling is involved in the physiology of erythrocytes, platelets, and leukocytes. ATP is released from erythrocytes and platelets, and purinoceptors and ectonucleotidases are expressed by these cells. P1, P2Y1, P2Y12, and P2X1 receptors are expressed on platelets, which mediate platelet aggregation and shape change. Long-term (trophic) actions of purine and pyrimidine nucleosides and nucleotides promote migration and proliferation of vascular smooth muscle and endothelial cells via P1 and P2Y receptors during angiogenesis, vessel remodeling during restenosis after angioplasty and atherosclerosis. The involvement of purinergic signaling in cardiovascular pathophysiology and its therapeutic potential are discussed, including heart failure, infarction, arrhythmias, syncope, cardiomyopathy, angina, heart transplantation and coronary bypass grafts, coronary artery disease, diabetic cardiomyopathy, hypertension, ischemia, thrombosis, diabetes mellitus, and migraine.
Collapse
Affiliation(s)
- Geoffrey Burnstock
- From the Autonomic Neuroscience Institute, Royal Free and University College Medical School, London, United Kingdom.
| |
Collapse
|
21
|
Chen Z, Wu D, Li L, Chen L. Apelin/APJ System: A Novel Therapeutic Target for Myocardial Ischemia/Reperfusion Injury. DNA Cell Biol 2016; 35:766-775. [DOI: 10.1089/dna.2016.3391] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Affiliation(s)
- Zhe Chen
- Institute of Pharmacy and Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drugs Study, University of South China, Hengyang, China
| | - Di Wu
- Institute of Pharmacy and Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drugs Study, University of South China, Hengyang, China
| | - Lanfang Li
- Institute of Pharmacy and Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drugs Study, University of South China, Hengyang, China
| | - Linxi Chen
- Institute of Pharmacy and Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drugs Study, University of South China, Hengyang, China
| |
Collapse
|
22
|
Activation of Adenosine 2A receptor inhibits neutrophil apoptosis in an autophagy-dependent manner in mice with systemic inflammatory response syndrome. Sci Rep 2016; 6:33614. [PMID: 27647162 PMCID: PMC5028892 DOI: 10.1038/srep33614] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Accepted: 08/30/2016] [Indexed: 01/13/2023] Open
Abstract
Systemic inflammatory response syndrome (SIRS) is an overwhelming whole body inflammation caused by infectious diseases or sterile insults. Neutrophils are the dominant participants during inflammation, and their survival and death determine the initiation as well as resolution of SIRS. Apoptosis and autophagy are two fundamental cellular processes that modulating cell fate, but their correlation and regulators in neutrophils under SIRS condition have not been elucidated. In this study, we demonstrated that high dose of LPS induced both apoptosis and autophagy of neutrophils in a mouse SIRS model and LPS-stimulated neutrophils in vitro. Moreover, we found that the adenosine 2A receptor (A2AR), a known anti-inflammatory G protein-coupled receptor (GPCR), could inhibit LPS-induced neutrophil apoptosis by suppressing the LPS-induced autophagy. Activation of A2AR suppressed LPS-induced autophagy by inhibiting the ROS-JNK pathway as well as promoting GPCR βϒ subunit–AKT signaling. The A2AR-inhibited autophagy suppressed apoptosis of neutrophils by blocking caspase8, caspase3 and PARP signaling. These findings not only increase our understandings of neutrophils’ fate and function in response to systemic inflammation, but also identify a novel anti-inflammatory role of A2AR in modulating neutrophils’ survival during inflammation.
Collapse
|
23
|
Guo X, Jiang H, Yang J, Chen J, Yang J, Ding JW, Li S, Wu H, Ding HS. Radioprotective 105 kDa protein attenuates ischemia/reperfusion-induced myocardial apoptosis and autophagy by inhibiting the activation of the TLR4/NF-κB signaling pathway in rats. Int J Mol Med 2016; 38:885-93. [PMID: 27431018 DOI: 10.3892/ijmm.2016.2686] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Accepted: 07/06/2016] [Indexed: 11/10/2022] Open
|
24
|
Obata T, Nakashima M. Fluvastatin, an HMG-CoA reductase inhibitor, facilitate adenosine production in the rat hearts via activation of ecto-5'-nucleotidase. Microvasc Res 2016; 107:1-5. [PMID: 27102210 DOI: 10.1016/j.mvr.2016.04.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 04/16/2016] [Accepted: 04/16/2016] [Indexed: 01/02/2023]
Abstract
OBJECTIVE The present study was examined whether fluvastatin, 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitor, can increase the production of interstitial adenosine via activation of ecto-5'-nucleotidase in the ventricular myocardium, with use of microdialysis techniques in in situ rat hearts. METHODS Adenosine in the dialysate collected during perfusion with Tyrode's solution containing 100μM AMP (through the probe) originated from the hydrolysis of AMP catalyzed by endogenous ecto-5'-nucleotidase, so that the level of adenosine reflected the activity of ecto-5'-nucleotidase in this tissue. RESULTS Fluvastatin (100μM), an inhibitor of low-density lipoprotein (LDL) oxidation, significantly increased the concentration of adenosine measured in the presence of 100μM AMP (i.e., the activity of ecto-5'-nucleotidase) by 154.7±16.0% (n=6, P<0.05), an increase which inhibited an antagonist of the α1-adrenoceptor (prazosin, 50μM) or of protein kinase C (PKC; chelerythrine, 10μM). Fluvastatin (10-500μM) increased the level of AMP-primed dialysate adenosine in a concentration-dependent manner. CONCLUSION These results indicate that fluvastatin increases in adenosine concentrations in the dialysate which resulted from activation of PKC, mediated by stimulation of α1-adrenoceptors, through activation of ecto-5'-nucleotidase.
Collapse
Affiliation(s)
- Toshio Obata
- School of Nursing, Faculty of Health Sciences, Osaka Aoyama University, 2-11-1 Niina, Mino, City, Japan.
| | - Michiko Nakashima
- Department of Nursing, School of Health Sciences, Asahi University, 1851 Hozumi Mizuho City Gifu, Japan
| |
Collapse
|
25
|
17-Methoxyl-7-Hydroxy-Benzene-Furanchalcone Ameliorates Myocardial Ischemia/Reperfusion Injury in Rat by Inhibiting Apoptosis and Autophagy Via the PI3K–Akt Signal Pathway. Cardiovasc Toxicol 2016; 17:79-87. [DOI: 10.1007/s12012-016-9358-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|