1
|
Piotin A, Oulehri W, Charles AL, Tacquard C, Collange O, Mertes PM, Geny B. Oxidative Stress and Mitochondria Are Involved in Anaphylaxis and Mast Cell Degranulation: A Systematic Review. Antioxidants (Basel) 2024; 13:920. [PMID: 39199166 PMCID: PMC11352116 DOI: 10.3390/antiox13080920] [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: 06/14/2024] [Revised: 07/23/2024] [Accepted: 07/24/2024] [Indexed: 09/01/2024] Open
Abstract
Anaphylaxis, an allergic reaction caused by the massive release of active mediators, can lead to anaphylactic shock (AS), the most severe and potentially life-threatening form of anaphylactic reaction. Nevertheless, understanding of its pathophysiology to support new therapies still needs to be improved. We performed a systematic review, assessing the role and the complex cellular interplay of mitochondria and oxidative stress during anaphylaxis, mast cell metabolism and degranulation. After presenting the main characteristics of anaphylaxis, the oxidant/antioxidant balance and mitochondrial functions, we focused this review on the involvement of mitochondria and oxidative stress in anaphylaxis. Then, we discussed the role of oxidative stress and mitochondria following mast cell stimulation by allergens, leading to degranulation, in order to further elucidate mechanistic pathways. Finally, we considered potential therapeutic interventions implementing these findings for the treatment of anaphylaxis. Experimental studies evaluated mainly cardiomyocyte metabolism during AS. Cardiac dysfunction was associated with left ventricle mitochondrial impairment and lipid peroxidation. Studies evaluating in vitro mast cell degranulation, following Immunoglobulin E (IgE) or non-IgE stimulation, revealed that mitochondrial respiratory complex integrity and membrane potential are crucial for mast cell degranulation. Antigen stimulation raises reactive oxygen species (ROS) production from nicotinamide adenine dinucleotide phosphate (NADPH) oxidases and mitochondria, leading to mast cell degranulation. Moreover, mast cell activation involved mitochondrial morphological changes and mitochondrial translocation to the cell surface near exocytosis sites. Interestingly, antioxidant administration reduced degranulation by lowering ROS levels. Altogether, these results highlight the crucial role of oxidative stress and mitochondria during anaphylaxis and mast cell degranulation. New therapeutics against anaphylaxis should probably target oxidative stress and mitochondria, in order to decrease anaphylaxis-induced systemic and major organ deleterious effects.
Collapse
Affiliation(s)
- Anays Piotin
- Physiology and Functional Exploration Service, Strasbourg University Hospital, 67000 Strasbourg, France;
- Division of Asthma and Allergy, Chest Diseases Department, Strasbourg University Hospital, 67000 Strasbourg, France
- Team 3072 “Mitochondria, Oxidative Stress and Muscle Protection”, Translational Medicine Federation of Strasbourg (FMTS), Faculty of Medicine, University of Strasbourg, 67000 Strasbourg, France; (W.O.); (A.-L.C.); (O.C.); (P.-M.M.)
| | - Walid Oulehri
- Team 3072 “Mitochondria, Oxidative Stress and Muscle Protection”, Translational Medicine Federation of Strasbourg (FMTS), Faculty of Medicine, University of Strasbourg, 67000 Strasbourg, France; (W.O.); (A.-L.C.); (O.C.); (P.-M.M.)
- Department of Anesthesia and Intensive Care, Strasbourg University Hospital, 67000 Strasbourg, France;
| | - Anne-Laure Charles
- Team 3072 “Mitochondria, Oxidative Stress and Muscle Protection”, Translational Medicine Federation of Strasbourg (FMTS), Faculty of Medicine, University of Strasbourg, 67000 Strasbourg, France; (W.O.); (A.-L.C.); (O.C.); (P.-M.M.)
| | - Charles Tacquard
- Department of Anesthesia and Intensive Care, Strasbourg University Hospital, 67000 Strasbourg, France;
- Établissement Français du Sang (EFS) Grand Est, French National Institute of Health and Medical Research), (INSERM) BPPS UMR_S1255, Fédération de Médecine Translationnelle de Strasbourg (FMTS), University of Strasbourg, 67000 Strasbourg, France
| | - Olivier Collange
- Team 3072 “Mitochondria, Oxidative Stress and Muscle Protection”, Translational Medicine Federation of Strasbourg (FMTS), Faculty of Medicine, University of Strasbourg, 67000 Strasbourg, France; (W.O.); (A.-L.C.); (O.C.); (P.-M.M.)
- Department of Anesthesia and Intensive Care, Strasbourg University Hospital, 67000 Strasbourg, France;
| | - Paul-Michel Mertes
- Team 3072 “Mitochondria, Oxidative Stress and Muscle Protection”, Translational Medicine Federation of Strasbourg (FMTS), Faculty of Medicine, University of Strasbourg, 67000 Strasbourg, France; (W.O.); (A.-L.C.); (O.C.); (P.-M.M.)
- Department of Anesthesia and Intensive Care, Strasbourg University Hospital, 67000 Strasbourg, France;
| | - Bernard Geny
- Physiology and Functional Exploration Service, Strasbourg University Hospital, 67000 Strasbourg, France;
- Team 3072 “Mitochondria, Oxidative Stress and Muscle Protection”, Translational Medicine Federation of Strasbourg (FMTS), Faculty of Medicine, University of Strasbourg, 67000 Strasbourg, France; (W.O.); (A.-L.C.); (O.C.); (P.-M.M.)
| |
Collapse
|
2
|
Geng Z, Chen W, Lu Q, Fu B, Fu X. UCP2 overexpression activates SIRT3 to regulate oxidative stress and mitochondrial dynamics induced by myocardial injury. Arch Biochem Biophys 2024; 753:109918. [PMID: 38301949 DOI: 10.1016/j.abb.2024.109918] [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: 08/09/2023] [Revised: 01/21/2024] [Accepted: 01/29/2024] [Indexed: 02/03/2024]
Abstract
OBJECTIVE Our previous study found that overexpression of uncoupling protein-2 (UCP2) had a protective effect on lipopolysaccharide (LPS)-induced sepsis cardiomyocytes. The aim of this study was to explore the effect and mechanism of uncoupling protein-2 (UCP2) on myocardial ischemia-reperfusion injury. METHODS In this study, we established hypoxia-reoxygenation (HR) injury model in rats and isolated cardiomyocytes of newborn rats. We also carried out following methods which include virus transfection technology, cell counting Kit-8 (CCK8), flow cytometry, enzyme linked immunosorbent assay (ELISA), Western blot (WB), quantitative reverse transcription PCR (RT qPCR), transmission electron microscopy, fluorescence colocalization and immunoprecipitation. MAIN RESULTS The results of this study showed that hypoxia-reoxygenation treatment in cardiomyocytes increased UCP2, myocardial enzyme and myocardial apoptosis and weakened cardiomyocyte viability. We observed increased cardiomyocyte viability and mitochondrial membrane potential, decreased myocardial enzyme and myocardial apoptosis, Inhibition of oxidative stress when UCP2 was overexpressed in cardiomyocytes. It also can Increase ATP and stabilize mitochondrial dynamics. Further studies founded that Sirtuin-3(SIRT3) changed with the expression of UCP2, which was confirmed by fluorescence co-localization and immunoprecipitation. CONCLUSIONS Our findings revealed that UCP2 and SIRT3 were important targets of anti-myocardial injury by inhibiting cellular oxidative stress and stabilizing mitochondrial dynamics.
Collapse
Affiliation(s)
- Zhengguang Geng
- Department of Critical Care Medicine, Affiliated hospital of Zunyi Medical University, Zunyi, Guizhou, 563000, China; Guizhou Key Laboratory of Anesthesia and Organ Protection& Zunyi Medical University, Zunyi, Guizhou, 563000, China.
| | - Wenbo Chen
- Department of Critical Care Medicine, Affiliated hospital of Zunyi Medical University, Zunyi, Guizhou, 563000, China.
| | - Qinju Lu
- Department of Critical Care Medicine, Affiliated hospital of Zunyi Medical University, Zunyi, Guizhou, 563000, China.
| | - Bao Fu
- Department of Critical Care Medicine, Affiliated hospital of Zunyi Medical University, Zunyi, Guizhou, 563000, China.
| | - Xiaoyun Fu
- Department of Critical Care Medicine, Affiliated hospital of Zunyi Medical University, Zunyi, Guizhou, 563000, China.
| |
Collapse
|
3
|
Tang X, Liu H, Rao R, Huang Y, Dong M, Xu M, Feng S, Shi X, Wang L, Wang Z, Zhou B. Modeling drug-induced mitochondrial toxicity with human primary cardiomyocytes. SCIENCE CHINA. LIFE SCIENCES 2024; 67:301-319. [PMID: 37864082 DOI: 10.1007/s11427-023-2369-3] [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: 01/12/2023] [Accepted: 05/16/2023] [Indexed: 10/22/2023]
Abstract
Mitochondrial toxicity induced by therapeutic drugs is a major contributor for cardiotoxicity, posing a serious threat to pharmaceutical industries and patients' lives. However, mitochondrial toxicity testing is not incorporated into routine cardiac safety screening procedures. To accurately model native human cardiomyocytes, we comprehensively evaluated mitochondrial responses of adult human primary cardiomyocytes (hPCMs) to a nucleoside analog, remdesivir (RDV). Comparison of their response to human pluripotent stem cell-derived cardiomyocytes revealed that the latter utilized a mitophagy-based mitochondrial recovery response that was absent in hPCMs. Accordingly, action potential duration was elongated in hPCMs, reflecting clinical incidences of RDV-induced QT prolongation. In a screen for mitochondrial protectants, we identified mitochondrial ROS as a primary mediator of RDV-induced cardiotoxicity. Our study demonstrates the utility of hPCMs in the detection of clinically relevant cardiac toxicities, and offers a framework for hPCM-based high-throughput screening of cardioprotective agents.
Collapse
Affiliation(s)
- Xiaoli Tang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100037, China
| | - Hong Liu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100037, China
| | - Rongjia Rao
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100037, China
| | - Yafei Huang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100037, China
| | - Mengqi Dong
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100037, China
| | - Miaomiao Xu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100037, China
| | - Shanshan Feng
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100037, China
| | - Xun Shi
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100037, China
| | - Li Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100037, China
- Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen, Shenzhen, 518020, China
| | - Zengwu Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100037, China
- Department of Epidemiology, Cardiovascular Institute and Fuwai Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, 100037, China
| | - Bingying Zhou
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100037, China.
- Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen, Shenzhen, 518020, China.
| |
Collapse
|
4
|
Piamsiri C, Maneechote C, Jinawong K, Arunsak B, Chunchai T, Nawara W, Chattipakorn SC, Chattipakorn N. GSDMD-mediated pyroptosis dominantly promotes left ventricular remodeling and dysfunction in post-myocardial infarction: a comparison across modes of programmed cell death and mitochondrial involvement. J Transl Med 2023; 21:16. [PMID: 36627703 PMCID: PMC9830763 DOI: 10.1186/s12967-023-03873-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 01/02/2023] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Myocardial infarction (MI) has recently accounted for more than one-third of global mortality. Multiple molecular pathological pathways, such as oxidative stress, inflammation, and mitochondrial dysfunction, have been recognized as possible mechanisms in the development of MI. Furthermore, different phases of ischemic injury following the progression of MI were also associated with multiple types of programmed cell death (PCDs), including apoptosis, necroptosis, ferroptosis, and pyroptosis. However, it remains unknown whether which types of PCDs play the most dominant role in post-myocardial infarction (post-MI). METHOD In this study, we used a preclinical rat model of MI induced by permanent left anterior descending coronary artery (LAD) ligation (n = 6) or a sham operated rat model (n = 6). After a 5-week experiment, cardiac function and morphology, mitochondrial studies, and molecular signaling analysis of PCDs were determined. RESULTS Herein, we demonstrated that post-MI rats had considerably impaired cardiac geometry, increased oxidative stress, myocardial injuries, and subsequently contractile dysfunction. They also exhibited worsened cardiac mitochondrial function and dynamic imbalance. More importantly, we found that post-MI mediated abundant myocardial cell death through multiple PCDs, including apoptosis, necroptosis, and pyroptosis, but not ferroptosis. CONCLUSION In this study, we provide the first insights into the mechanism of PCDs by pyroptosis, which is leveraged as the most dominant mode of cell death after MI.
Collapse
Affiliation(s)
- Chanon Piamsiri
- grid.7132.70000 0000 9039 7662Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200 Thailand ,grid.7132.70000 0000 9039 7662Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200 Thailand ,grid.7132.70000 0000 9039 7662Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200 Thailand
| | - Chayodom Maneechote
- grid.7132.70000 0000 9039 7662Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200 Thailand ,grid.7132.70000 0000 9039 7662Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200 Thailand
| | - Kewarin Jinawong
- grid.7132.70000 0000 9039 7662Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200 Thailand ,grid.7132.70000 0000 9039 7662Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200 Thailand ,grid.7132.70000 0000 9039 7662Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200 Thailand
| | - Busarin Arunsak
- grid.7132.70000 0000 9039 7662Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200 Thailand ,grid.7132.70000 0000 9039 7662Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200 Thailand
| | - Titikorn Chunchai
- grid.7132.70000 0000 9039 7662Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200 Thailand ,grid.7132.70000 0000 9039 7662Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200 Thailand
| | - Wichwara Nawara
- grid.7132.70000 0000 9039 7662Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200 Thailand ,grid.7132.70000 0000 9039 7662Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200 Thailand
| | - Siriporn C Chattipakorn
- grid.7132.70000 0000 9039 7662Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200 Thailand ,grid.7132.70000 0000 9039 7662Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200 Thailand ,grid.7132.70000 0000 9039 7662Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai, 50200 Thailand
| | - Nipon Chattipakorn
- grid.7132.70000 0000 9039 7662Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200 Thailand ,grid.7132.70000 0000 9039 7662Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200 Thailand ,grid.7132.70000 0000 9039 7662Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200 Thailand
| |
Collapse
|
5
|
Ischemic Preconditioning and Postconditioning Protect the Heart by Preserving the Mitochondrial Network. BIOMED RESEARCH INTERNATIONAL 2022; 2022:6889278. [PMID: 36203484 PMCID: PMC9532115 DOI: 10.1155/2022/6889278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 09/06/2022] [Indexed: 12/02/2022]
Abstract
Background Mitochondria fuse to form elongated networks which are more tolerable to stress and injury. Ischemic pre- and postconditioning (IPC and IPost, respectively) are established cardioprotective strategies in the preclinical setting. Whether IPC and IPost modulates mitochondrial morphology is unknown. We hypothesize that the protective effects of IPC and IPost may be conferred via preservation of mitochondrial network. Methods IPC and IPost were applied to the H9c2 rat myoblast cells, isolated adult primary murine cardiomyocytes, and the Langendorff-isolated perfused rat hearts. The effects of IPC and IPost on cardiac cell death following ischemia-reperfusion injury (IRI), mitochondrial morphology, and gene expression of mitochondrial-shaping proteins were investigated. Results IPC and IPost successfully reduced cardiac cell death and myocardial infarct size. IPC and IPost maintained the mitochondrial network in both H9c2 and isolated adult primary murine cardiomyocytes. 2D-length measurement of the 3 mitochondrial subpopulations showed that IPC and IPost significantly increased the length of interfibrillar mitochondria (IFM). Gene expression of the pro-fusion protein, Mfn1, was significantly increased by IPC, while the pro-fission protein, Drp1, was significantly reduced by IPost in the H9c2 cells. In the primary cardiomyocytes, gene expression of both Mfn1 and Mfn2 were significantly upregulated by IPC and IPost, while Drp1 was significantly downregulated by IPost. In the Langendorff-isolated perfused heart, gene expression of Drp1 was significantly downregulated by both IPC and IPost. Conclusion IPC and IPost-mediated upregulation of pro-fusion proteins (Mfn1 and Mfn2) and downregulation of pro-fission (Drp1) promote maintenance of the interconnected mitochondrial network, ultimately conferring cardioprotection against IRI.
Collapse
|
6
|
Resveratrol Reestablishes Mitochondrial Quality Control in Myocardial Ischemia/Reperfusion Injury through Sirt1/Sirt3-Mfn2-Parkin-PGC-1α Pathway. Molecules 2022; 27:molecules27175545. [PMID: 36080311 PMCID: PMC9457908 DOI: 10.3390/molecules27175545] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Revised: 08/14/2022] [Accepted: 08/17/2022] [Indexed: 01/18/2023] Open
Abstract
Resveratrol is a natural polyphenol found in various plants. It has been widely studied on cardiovascular disorders. It is known that resveratrol can activate Sirtuin proteins and participate in cellular energy metabolism through a Sirtuin-dependent pathway. Here, we hypothesized that resveratrol may protect against myocardial ischemia/reperfusion injury (MIRI) through the target of Sirt1/Sirt3 on mitochondrial dynamics, cardiac autophagy, bioenergetics and oxidative damage in hypoxia/reoxygenation (H/R)-induced neonatal rat cardiomyocytes. We observed that resveratrol could activate the Sirt1/Sirt3-FoxO pathway on myocardial mitochondria in H/R cardiomyocytes. Subsequently, we found that resveratrol repaired the fission–fusion balance, autophagic flux and mitochondrial biosynthesis compared by H/R group. These changes were followed by increased functional mitochondrial number, mitochondrial bioenergetics and a better mitochondrial antioxidant enzyme system. Meanwhile, these effects were antagonized by co-treatment with Selisistat (Ex527), a Sirtuin inhibitor. Together, our findings uncover the potential contribution of resveratrol in reestablishing a mitochondrial quality control network with Parkin, Mfn2 and PGC-1α as the key nodes.
Collapse
|
7
|
SIRT3 protects bovine mammary epithelial cells from heat stress damage by activating the AMPK signaling pathway. Cell Death Discov 2021; 7:304. [PMID: 34675216 PMCID: PMC8531291 DOI: 10.1038/s41420-021-00695-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 08/26/2021] [Accepted: 09/23/2021] [Indexed: 12/11/2022] Open
Abstract
With global warming, heat stress has become an important challenge for the global dairy industry. Sirtuin 3 (SIRT3), an important mitochondrial NAD+dependent decarboxylase and a major regulator of cellular energy metabolism and antioxidant defense, is integral to maintaining normal mitochondrial function. The aim of this study was to assess the protective effect of SIRT3 on damage to bovine mammary epithelial cells (BMECs) induced by heat stress and to explore its potential mechanism. Our results indicate that SIRT3 is significantly downregulated in heat-stressed mammary tissue and high-temperature-treated BMECs. SIRT3 knockdown significantly increased the expression of HSP70, Bax, and cleaved-caspase 3 and inhibited the production of antioxidases, thus promoting ROS production and cell apoptosis in BMECs. In addition, SIRT3 knockdown can aggravate mitochondrial damage by mediating the expression of genes related to mitochondrial fission and fusion, including dynamin-related protein 1, mitochondrial fission 1 protein, and mitochondrial fusion proteins 1and 2. In addition, SIRT3 knockdown substantially decreased AMPK phosphorylation in BMECs. In contrast, SIRT3 overexpression in high-temperature treatment had the opposite effect to SIRT3 knockdown in BMECs. SIRT3 overexpression reduced mitochondrial damage and weakened the oxidative stress response of BMECs induced by heat stress and promoted the phosphorylation of AMPK. Taken together, our results indicate that SIRT3 can protect BMECs from heat stress damage through the AMPK signaling pathway. Therefore, the reduction of oxidative stress by SIRT3 may be the primary molecular mechanism underlying resistance to heat stress in summer cows.
Collapse
|
8
|
Boulghobra D, Coste F, Geny B, Reboul C. Exercise training protects the heart against ischemia-reperfusion injury: A central role for mitochondria? Free Radic Biol Med 2020; 152:395-410. [PMID: 32294509 DOI: 10.1016/j.freeradbiomed.2020.04.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 04/01/2020] [Accepted: 04/07/2020] [Indexed: 12/11/2022]
Abstract
Ischemic heart disease is one of the main causes of morbidity and mortality worldwide. Physical exercise is an effective lifestyle intervention to reduce the risk factors for cardiovascular disease and also to improve cardiac function and survival in patients with ischemic heart disease. Among the strategies that contribute to reduce heart damages during ischemia and reperfusion, regular physical exercise is efficient both in rodent experimental models and in humans. However, the cellular and molecular mechanisms of the cardioprotective effects of exercise remain unclear. During ischemia and reperfusion, mitochondria are crucial players in cell death, but also in cell survival. Although exercise training can influence mitochondrial function, the consequences on heart sensitivity to ischemic insults remain elusive. In this review, we describe the effects of physical activity on cardiac mitochondria and their potential key role in exercise-induced cardioprotection against ischemia-reperfusion damage. Based on recent scientific data, we discuss the role of different pathways that might help to explain why mitochondria are a key target of exercise-induced cardioprotection.
Collapse
Affiliation(s)
| | - Florence Coste
- LAPEC EA4278, Avignon Université, F-84000, Avignon, France
| | - Bernard Geny
- EA3072, «Mitochondrie, Stress Oxydant, et Protection Musculaire», Université de Strasbourg, 67000, Strasbourg, France
| | - Cyril Reboul
- LAPEC EA4278, Avignon Université, F-84000, Avignon, France.
| |
Collapse
|
9
|
Mitochondrial MiRNA in Cardiovascular Function and Disease. Cells 2019; 8:cells8121475. [PMID: 31766319 PMCID: PMC6952824 DOI: 10.3390/cells8121475] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 11/13/2019] [Accepted: 11/18/2019] [Indexed: 12/19/2022] Open
Abstract
MicroRNAs (miRNAs) are small noncoding RNAs functioning as crucial post-transcriptional regulators of gene expression involved in cardiovascular development and health. Recently, mitochondrial miRNAs (mitomiRs) have been shown to modulate the translational activity of the mitochondrial genome and regulating mitochondrial protein expression and function. Although mitochondria have been verified to be essential for the development and as a therapeutic target for cardiovascular diseases, we are just beginning to understand the roles of mitomiRs in the regulation of crucial biological processes, including energy metabolism, oxidative stress, inflammation, and apoptosis. In this review, we summarize recent findings regarding how mitomiRs impact on mitochondrial gene expression and mitochondrial function, which may help us better understand the contribution of mitomiRs to both the regulation of cardiovascular function under physiological conditions and the pathogenesis of cardiovascular diseases.
Collapse
|
10
|
Oleaga C, Jalilvand G, Legters G, Martin C, Ekman G, McAleer CW, Long CJ, Hickman JJ. A human in vitro platform for the evaluation of pharmacology strategies in cardiac ischemia. APL Bioeng 2019; 3:036103. [PMID: 31431939 PMCID: PMC6692160 DOI: 10.1063/1.5089237] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 07/22/2019] [Indexed: 12/21/2022] Open
Abstract
Cardiac ischemic events increase the risk for arrhythmia, heart attack, heart failure, and death and are the leading mortality condition globally. Reperfusion therapy is the first line of treatment for this condition, and although it significantly reduces mortality, cardiac ischemia remains a significant threat. New therapeutic strategies are under investigation to improve the ischemia survival rate; however, the current preclinical models to validate these fail to predict the human outcome. We report the development of a functional human cardiac in vitro system for the study of conduction velocity under ischemic conditions. The system is a bioMEMs platform formed by human iPSC derived cardiomyocytes patterned on microelectrode arrays and maintained in serum-free conditions. Electrical activity changes of conduction velocity, beat frequency, and QT interval (the QT-interval measures the period from onset of depolarization to the completion of repolarization) or action potential length can be evaluated over time and under the stress of ischemia. The optimized protocol induces >80% reduction in conduction velocity, after a 4 h depletion period, and a partial recovery after 72 h of oxygen and nutrient reintroduction. The sensitivity of the platform for pharmacological interventions was challenged with a gap junction modulator (ZP1609), known to prevent or delay the depression of conduction velocity induced by ischemic metabolic stress. ZP1609 significantly improved the drastic drop in conduction velocity and enabled a greater recovery. This model represents a new preclinical platform for studying cardiac ischemia with human cells, which does not rely on biomarker analysis and has the potential for screening novel cardioprotective drugs with readouts that are closer to the measured clinical parameters.
Collapse
Affiliation(s)
- Carlota Oleaga
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, Florida 32826, USA
| | - Golareh Jalilvand
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, Florida 32826, USA
| | - Gregg Legters
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, Florida 32826, USA
| | - Candace Martin
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, Florida 32826, USA
| | - Gail Ekman
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, Florida 32826, USA
| | | | | | - James J. Hickman
- Author to whom correspondence should be addressed:. Tel.: +1 407-823-1925
| |
Collapse
|
11
|
Hu M, Li T, Bo Z, Xiang F. The protective role of carnosic acid in ischemic/reperfusion injury through regulation of autophagy under T2DM. Exp Biol Med (Maywood) 2019; 244:602-611. [PMID: 30947537 DOI: 10.1177/1535370219840987] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
IMPACT STATEMENT We have provided, for the first time, evidence that carnosic acid (CA) attenuates ischemia-reperfusion injury of diabetic myocardium, i.e. diabetic myocardial ischemia/reperfusion (DMI/R) injury, via enhancement of autophagy. A greater understanding of the target molecule in CA-enhanced autophagy is necessary for the development of potential chemotherapy for DMI/R injury.
Collapse
Affiliation(s)
- Min Hu
- 1 Department of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Tianyu Li
- 1 Department of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zixiang Bo
- 1 Department of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Feixiang Xiang
- 2 Department of Ultrasound, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.,3 Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| |
Collapse
|
12
|
De la Fuente S, Sheu SS. SR-mitochondria communication in adult cardiomyocytes: A close relationship where the Ca 2+ has a lot to say. Arch Biochem Biophys 2019; 663:259-268. [PMID: 30685253 PMCID: PMC6377816 DOI: 10.1016/j.abb.2019.01.026] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Revised: 01/14/2019] [Accepted: 01/22/2019] [Indexed: 02/07/2023]
Abstract
In adult cardiomyocytes, T-tubules, junctional sarcoplasmic reticulum (jSR), and mitochondria juxtapose each other and form a unique and highly repetitive functional structure along the cell. The close apposition between jSR and mitochondria creates high Ca2+ microdomains at the contact sites, increasing the efficiency of the excitation-contraction-bioenergetics coupling, where the Ca2+ transfer from SR to mitochondria plays a critical role. The SR-mitochondria contacts are established through protein tethers, with mitofusin 2 the most studied SR-mitochondrial "bridge", albeit controversial. Mitochondrial Ca2+ uptake is further optimized with the mitochondrial Ca2+ uniporter preferentially localized in the jSR-mitochondria contact sites and the mitochondrial Na+/Ca2+ exchanger localized away from these sites. Despite all these unique features facilitating the privileged transport of Ca2+ from SR to mitochondria in adult cardiomyocytes, the question remains whether mitochondrial Ca2+ concentrations oscillate in synchronicity with cytosolic Ca2+ transients during heartbeats. Proper Ca2+ transfer controls not only the process of mitochondrial bioenergetics, but also of mitochondria-mediated cell death, autophagy/mitophagy, mitochondrial fusion/fission dynamics, reactive oxygen species generation, and redox signaling, among others. Our review focuses specifically on Ca2+ signaling between SR and mitochondria in adult cardiomyocytes. We discuss the physiological and pathological implications of this SR-mitochondrial Ca2+ signaling, research gaps, and future trends.
Collapse
Affiliation(s)
- Sergio De la Fuente
- Center for Translational Medicine, Department of Medicine, Thomas Jefferson University, Philadelphia, PA, 19107, USA.
| | - Shey-Shing Sheu
- Center for Translational Medicine, Department of Medicine, Thomas Jefferson University, Philadelphia, PA, 19107, USA.
| |
Collapse
|
13
|
Li Y, Liu X. Novel insights into the role of mitochondrial fusion and fission in cardiomyocyte apoptosis induced by ischemia/reperfusion. J Cell Physiol 2018. [PMID: 29528108 DOI: 10.1002/jcp.26522] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
As the main source of energy in the body, mitochondria are highly dynamic organelles, which are constantly going through fusion and fission. The fine balance of mitochondrial fusion and fission plays an important role in maintaining the stability of cardiomyocyte homeostasis. The processes of mitochondrial fusion and fission are very complex, which is mediated by fusion and fission proteins. Disruptions in these processes through controlling fusion and fission proteins affect mitochondrial functions and cardiomyocyte survival. Ischemia/reperfusion (I/R) can regulate the expression and post-translational modifications of fusion and fission proteins thereby inducing the abnormality of mitochondrial fusion and fission and cardiomyocyte apoptosis. Furthermore, intervention with the expression and function of fusion and fission proteins influences on cardiomyocyte apoptosis under I/R conditions. In this review, we focus on the current developments in the effects of mitochondrial fusion and fission on cardiomyocyte functions, the implications for cardiomyocyte apoptosis in response to I/R, and possible mechanisms. And we review their roles as a potential therapeutic target for treating I/R-induced cardiomyocyte injury.
Collapse
Affiliation(s)
- YuZhen Li
- Department of Pathophysiology, Institute of Basic Medical Science, PLA General Hospital, Beijing, China
| | - XiuHua Liu
- Department of Pathophysiology, Institute of Basic Medical Science, PLA General Hospital, Beijing, China
| |
Collapse
|
14
|
Zhao Y, Ponnusamy M, Liu C, Tian J, Dong Y, Gao J, Wang C, Zhang Y, Zhang L, Wang K, Li P. MiR-485-5p modulates mitochondrial fission through targeting mitochondrial anchored protein ligase in cardiac hypertrophy. Biochim Biophys Acta Mol Basis Dis 2017; 1863:2871-2881. [DOI: 10.1016/j.bbadis.2017.07.034] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 07/11/2017] [Accepted: 07/31/2017] [Indexed: 12/15/2022]
|
15
|
Liu JD, Chen HJ, Wang DL, Wang H, Deng Q. Pim-1 Kinase Regulating Dynamics Related Protein 1 Mediates Sevoflurane Postconditioning-induced Cardioprotection. Chin Med J (Engl) 2017; 130:309-317. [PMID: 28139514 PMCID: PMC5308013 DOI: 10.4103/0366-6999.198922] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
BACKGROUND It is well documented that sevoflurane postconditioning (SP) has a significant myocardial protection effect. However, the mechanisms underlying SP are still unclear. In the present study, we investigated the hypothesis that the Pim-1 kinase played a key role in SP-induced cardioprotection by regulating dynamics-related protein 1 (Drp1). METHODS A Langendorff model was used in this study. Seventy-two rats were randomly assigned into six groups as follows: CON group, ischemia reperfusion (I/R) group, SP group , SP+proto-oncogene serine/threonine-protein kinase 1 (Pim-1) inhibitor II group, SP+dimethylsufoxide group, and Pim-1 inhibitor II group (n = 12, each). Hemodynamic parameters and infarct size were measured to reflect the extent of myocardial I/R injury. The expressions of Pim-1, B-cell leukemia/lymphoma 2 (Bcl-2) and cytochrome C (Cyt C) in cytoplasm and mitochondria, the Drp1 in mitochondria, and the total Drp1 and p-Drp1ser637 were measured by Western blotting. In addition, transmission electron microscope was used to observe mitochondrial morphology. The experiment began in October 2014 and continued until July 2016. RESULTS SP improved myocardial I/R injury-induced hemodynamic parametric changes, cardiac function, and preserved mitochondrial phenotype and decreased myocardial infarct size (24.49 ± 1.72% in Sev group compared with 41.98 ± 4.37% in I/R group; P< 0.05). However, Pim-1 inhibitor II significantly (P < 0.05) abolished the protective effect of SP. Western blotting analysis demonstrated that, compared with I/R group, the expression of Pim-1 and Bcl-2 in cytoplasm and mitochondria as well as the total p-Drp1ser637 in Sev group (P < 0.05) were upregulated. Meanwhile, SP inhibited Drp1 compartmentalization to the mitochondria followed by a reduction in the release of Cyt C. Pretreatment with Pim-1 inhibitor II significantly (P < 0.05) abolished SP-induced Pim-1/p-Drp1ser637 signaling activation. CONCLUSIONS These findings suggested that SP could attenuate myocardial ischemia-reperfusion injury by increasing the expression of the Pim-1 kinase. Upregulation of Pim-1 might phosphorylate Drp1 and prevent extensive mitochondrial fission through Drp1 cytosolic sequestration.
Collapse
Affiliation(s)
- Jin-Dong Liu
- Department of Anesthesiology, The Affiliated Hospital of Xuzhou Medical University, Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University; Department of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Hui-Juan Chen
- Department of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Da-Liang Wang
- Department of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Hui Wang
- Department of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Qian Deng
- Department of Anesthesiology, The Affiliated Hospital of Xuzhou Medical University, Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University; Department of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| |
Collapse
|
16
|
Lesnefsky EJ, Chen Q, Tandler B, Hoppel CL. Mitochondrial Dysfunction and Myocardial Ischemia-Reperfusion: Implications for Novel Therapies. Annu Rev Pharmacol Toxicol 2017; 57:535-565. [PMID: 27860548 PMCID: PMC11060135 DOI: 10.1146/annurev-pharmtox-010715-103335] [Citation(s) in RCA: 275] [Impact Index Per Article: 34.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Mitochondria have emerged as key participants in and regulators of myocardial injury during ischemia and reperfusion. This review examines the sites of damage to cardiac mitochondria during ischemia and focuses on the impact of these defects. The concept that mitochondrial damage during ischemia leads to cardiac injury during reperfusion is addressed. The mechanisms that translate ischemic mitochondrial injury into cellular damage, during both ischemia and early reperfusion, are examined. Next, we discuss strategies that modulate and counteract these mechanisms of mitochondrial-driven injury. The new concept that mitochondria are not merely stochastic sites of oxidative and calcium-mediated injury but that they activate cellular responses of mitochondrial remodeling and cellular reactions that modulate the balance between cell death and recovery is reviewed, and the therapeutic implications of this concept are discussed.
Collapse
Affiliation(s)
- Edward J Lesnefsky
- Department of Medicine, Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia 23298; ,
- Medical Service, McGuire Veterans Affairs Medical Center, Richmond, Virginia 23249;
| | - Qun Chen
- Department of Medicine, Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia 23298; ,
| | - Bernard Tandler
- Department of Biological Sciences, Case Western Reserve University School of Dental Medicine, Cleveland, Ohio 44106;
| | - Charles L Hoppel
- Department of Pharmacology, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106;
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106
- Center for Mitochondrial Disease, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106
| |
Collapse
|
17
|
Abstract
Mitochondrial dynamics, fission and fusion, were first identified in yeast with investigation in heart cells beginning only in the last 5 to 7 years. In the ensuing time, it has become evident that these processes are not only required for healthy mitochondria, but also, that derangement of these processes contributes to disease. The fission and fusion proteins have a number of functions beyond the mitochondrial dynamics. Many of these functions are related to their membrane activities, such as apoptosis. However, other functions involve other areas of the mitochondria, such as OPA1's role in maintaining cristae structure and preventing cytochrome c leak, and its essential (at least a 10 kDa fragment of OPA1) role in mtDNA replication. In heart disease, changes in expression of these important proteins can have detrimental effects on mitochondrial and cellular function.
Collapse
Affiliation(s)
- A A Knowlton
- Molecular & Cellular Cardiology, Division of Cardiovascular Medicine and Pharmacology Department, University of California, Davis, and The Department of Veteran's Affairs, Northern California VA, Sacramento, California, USA
| | - T T Liu
- Molecular & Cellular Cardiology, Division of Cardiovascular Medicine and Pharmacology Department, University of California, Davis, and The Department of Veteran's Affairs, Northern California VA, Sacramento, California, USA
| |
Collapse
|
18
|
Xiao S, Shaw RM. Cardiomyocyte protein trafficking: Relevance to heart disease and opportunities for therapeutic intervention. Trends Cardiovasc Med 2014; 25:379-89. [PMID: 25649302 DOI: 10.1016/j.tcm.2014.12.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Revised: 12/18/2014] [Accepted: 12/20/2014] [Indexed: 11/30/2022]
Abstract
Cardiomyocytes, the individual contractile units of heart muscle, are long-lived and robust. Given the longevity of these cells, it can be easy to overlook their dynamic intracellular environment that contain rapid protein movements and frequent protein turnover. Critical gene transcription and protein translation occur continuously, as well as trafficking and localization of proteins to specific functional zones of cell membrane. As heart failure becomes an increasingly important clinical entity, growing numbers of investigative teams are examining the cell biology of healthy and diseased cardiomyocytes. In this review, we introduce the major architectural structures and types of protein movements within cardiac cells, and then review recent studies that explore the regulation of such movements. We conclude by introducing current translational directions of the basic studies with a focus on novel areas of therapeutic development.
Collapse
Affiliation(s)
- Shaohua Xiao
- Heart Institute and Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Robin M Shaw
- Heart Institute and Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA; Department of Medicine, University of California Los Angeles, Los Angeles, CA.
| |
Collapse
|
19
|
Ma S, Wang Y, Chen Y, Cao F. The role of the autophagy in myocardial ischemia/reperfusion injury. Biochim Biophys Acta Mol Basis Dis 2014; 1852:271-6. [PMID: 24859226 DOI: 10.1016/j.bbadis.2014.05.010] [Citation(s) in RCA: 232] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Revised: 04/29/2014] [Accepted: 05/12/2014] [Indexed: 02/08/2023]
Abstract
Autophagy is an intracellular process responsible for damaged or unnecessary protein and organelle degradation. In the heart, autophagy occurs at basal level and dysregulated autophagy is associated with a variety of cardiovascular diseases. Autophagy is enhanced in ischemia as well as in the reperfusion phase during cardiac ischemia reperfusion (I/R) injury. More importantly, recent studies revealed that autophagy exerted both beneficial and detrimental effects in pathology of cardiac ischemia reperfusion. This paper is to review the functional significance of autophagy in cardiac ischemia reperfusion injury and discuss underlying signaling pathways. This article is part of a Special Issue entitled: Autophagy and protein quality control in cardiometabolic diseases.
Collapse
Affiliation(s)
- Sai Ma
- Department of Cardiology, Chinese PLA General Hospital, 28# Fuxing Street, Beijing 100852, China; Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127# Changle West Road, Xi'an, Shaanxi 710032, China
| | - Yabin Wang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127# Changle West Road, Xi'an, Shaanxi 710032, China
| | - Yundai Chen
- Department of Cardiology, Chinese PLA General Hospital, 28# Fuxing Street, Beijing 100852, China
| | - Feng Cao
- Department of Cardiology, Chinese PLA General Hospital, 28# Fuxing Street, Beijing 100852, China; Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127# Changle West Road, Xi'an, Shaanxi 710032, China.
| |
Collapse
|