1
|
Yanık Çolak S, Andaç B, Özgün E, Yılmaz Bülbül B, Okur M, Yekdeş AC, Yıldız Ç, Çelik M. The Relationship Between Serum MG53 Levels and the Presence of Metabolic Syndrome and Its Components. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:582. [PMID: 40282872 PMCID: PMC12028786 DOI: 10.3390/medicina61040582] [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: 02/07/2025] [Revised: 02/26/2025] [Accepted: 03/19/2025] [Indexed: 04/29/2025]
Abstract
Background and Objectives: MG53 is a myokine/cardiokine involved in membrane repair. Some preclinical studies suggest that it is associated with insulin resistance. Metabolic syndrome (MS) is manifested by dyslipidemia, hypertension (HT), visceral obesity, hyperinsulinism, and glucose intolerance. We aimed to evaluate the relationship between the MG53 protein and MS and its components. Materials and Methods: This study was conducted among 64 patients with MS and 64 age- and sex-matched healthy participants. MG53 levels were measured using Human-MG53, a commercially available enzyme-linked immunosorbent assay (ELISA) kit (Cat# CSB-EL024511HU, Alfagen laboratory supplies, Cusabio, Bornova, İzmir.). Results: There was no significant connection between serum MG53 levels and the presence of MS (p = 0.969). We found no correlation between serum MG53 levels and the presence of HT, weight, waist circumference, body mass index, HDL-C, fasting blood glucose, and HbA1c levels. Conclusions: This study's results suggest no association between serum MG53 levels and MS parameters in the studied ethnic population. Due to the limited number and controversy of available studies on this subject, our findings may provide perspective for conducting studies with more diverse populations to obtain more comprehensive results.
Collapse
Affiliation(s)
- Serpil Yanık Çolak
- Department of Endocrinology and Metabolism, Trakya University Medical Faculty, Edirne 22000, Turkey; (B.A.)
| | - Burak Andaç
- Department of Endocrinology and Metabolism, Trakya University Medical Faculty, Edirne 22000, Turkey; (B.A.)
| | - Eray Özgün
- Department of Biochemistry, Trakya University Medical Faculty, Edirne 22000, Turkey
| | - Buket Yılmaz Bülbül
- Department of Endocrinology and Metabolism, Trakya University Medical Faculty, Edirne 22000, Turkey; (B.A.)
| | - Mine Okur
- Department of Endocrinology and Metabolism, Trakya University Medical Faculty, Edirne 22000, Turkey; (B.A.)
| | - Ali Cem Yekdeş
- Department of Public Health, Trakya University Medical Faculty, Edirne 22000, Turkey
| | - Çağla Yıldız
- Department of Internal Medicine, Trakya University Medical Faculty, Edirne 22000, Turkey
| | - Mehmet Çelik
- Department of Endocrinology and Metabolism, Trakya University Medical Faculty, Edirne 22000, Turkey; (B.A.)
| |
Collapse
|
2
|
Yuan S, Yu Q, Luo M, Wu J, Wang L. Friend or Foe: The Paradoxical Roles of MG53 in Diabetes. Diabetes 2025; 74:145-152. [PMID: 39535840 DOI: 10.2337/db24-0556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024]
Abstract
MG53 is predominantly expressed in striated muscles. The role of MG53 in diabetes has gradually been elucidated but is still full of controversy. Some reports have indicated that MG53 is upregulated in animal models with metabolic disorders and that muscle-specific MG53 upregulation is sufficient to induce whole-body insulin resistance and metabolic syndrome through targeting both the insulin receptor (IR) and insulin receptor substrate 1 (IRS-1) for ubiquitin-dependent degradation. Additionally, MG53 has been identified as a myokine/cardiokine that is secreted from striated muscles into the bloodstream, and circulating MG53 has further been shown to trigger insulin resistance by binding to the extracellular domain of the IR, thereby allosterically inhibiting insulin signaling. Conversely, findings have been reported from other studies that contradict these results. Specifically, no significant change in MG53 expression in striated muscles or serum has been observed in diabetic models, and the MG53-mediated degradation of IRS-1 may be insufficient to induce insulin resistance due to the compensatory roles of other IRS subtypes. Furthermore, sustained elevation of MG53 levels in serum or systemic administration of recombinant human MG53 (rhMG53) has shown no impact on metabolic function. In this article, we will fully characterize these two disparate views, strive to provide critical insights into their contrasts, and propose several specific experimental approaches that may yield additional evidence. Our goal is to encourage the scientific community to elucidate the effects of MG53 on metabolic diseases and the molecular mechanisms involved, thereby providing the theoretical basis for the treatment of metabolic diseases and the applications of rhMG53.
Collapse
Affiliation(s)
- Shuangshuang Yuan
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Luzhou Municipal Key Laboratory of Thrombosis and Vascular Biology, and Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Qin Yu
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Luzhou Municipal Key Laboratory of Thrombosis and Vascular Biology, and Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Mao Luo
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Luzhou Municipal Key Laboratory of Thrombosis and Vascular Biology, and Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Jianbo Wu
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Luzhou Municipal Key Laboratory of Thrombosis and Vascular Biology, and Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Liqun Wang
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Luzhou Municipal Key Laboratory of Thrombosis and Vascular Biology, and Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
3
|
Nasiri-Valikboni A, Rashid M, Azimi A, Zarei H, Yousefifard M. Protective effect of sevoflurane on myocardial ischemia-reperfusion injury: a systematic review and meta-analysis. Int J Surg 2024; 110:7311-7330. [PMID: 39093878 PMCID: PMC11573079 DOI: 10.1097/js9.0000000000001975] [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: 05/30/2024] [Accepted: 07/14/2024] [Indexed: 08/04/2024]
Abstract
BACKGROUND Myocardial ischemia-reperfusion (I/R) injury significantly impacts recovery in both cardiac and noncardiac surgeries, potentially leading to severe cardiac dysfunction. Sevoflurane, a volatile anesthetic, is reputed for its protective effects against myocardial I/R injury, although evidence remains inconclusive. This systematic review and meta-analysis aim to clarify the cardioprotective efficacy of sevoflurane. METHODS The systematic search of databases including Medline, Embase, Scopus, and Web of Science, was supplemented with a manual search to retrieve studies using rat or mouse models of myocardial I/R injury, comparing sevoflurane pretreatment (≥24 h before I/R), preconditioning (within 24 h before I/R), or postconditioning (after I/R) against nontreated controls. The outcomes were cardiac function, myocardial infarct size, apoptosis, inflammation, oxidative stress, and cardiac biomarkers. Using the random effects model, standardized mean differences (SMD) were pooled to perform meta-analyses. RESULTS Fifty-one studies, encompassing 8189 subjects, were included in the meta-analysis. Pretreatment with Sevoflurane significantly reduced infarct size. Sevoflurane preconditioning exhibited positive effects on left ventricular parameters and ejection fraction, and reduced infarct size, apoptosis, and oxidative stress. Postconditioning with Sevoflurane demonstrated improvements in cardiac function, including enhanced left ventricular parameters and reduced infarct size, apoptosis, inflammation, oxidative stress, and cardiac biomarkers. CONCLUSION Sevoflurane demonstrates a significant protective effect against myocardial I/R injury in animal models. These findings support the potential clinical utility of sevoflurane as an anesthetic choice in preventing and managing myocardial I/R injury during surgeries.
Collapse
Affiliation(s)
| | - Mohamad Rashid
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | - Amir Azimi
- Physiology Research Center, Iran University of Medical Sciences, Tehran
| | - Hamed Zarei
- Physiology Research Center, Iran University of Medical Sciences, Tehran
| | | |
Collapse
|
4
|
Lee KE, Nishi M, Kim J, Murayama T, Dawson Z, Wang X, Zhou X, Tan T, Cai C, Takeshima H, Park KH. MG53's non-physiologic interaction with insulin receptor: lack of effect on insulin-stimulated Akt phosphorylation in muscle, heart and liver tissues. Front Endocrinol (Lausanne) 2024; 15:1425426. [PMID: 39355613 PMCID: PMC11442255 DOI: 10.3389/fendo.2024.1425426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 08/15/2024] [Indexed: 10/03/2024] Open
Abstract
Rationale MG53's known function in facilitating tissue repair and anti-inflammation has broad applications to regenerative medicine. There is controversy regarding MG53's role in the development of type 2 diabetes mellitus. Objective This study aims to address this controversy - whether MG53's myokine function contributes to inhibition of insulin signaling in muscle, heart, and liver tissues. Study design We determined the binding affinity of the recombinant human MG53 (rhMG53) to the insulin receptor extracellular domain (IR-ECD) and found low affinity of interaction with Kd (>480 nM). Using cultured C2C12 myotubes and HepG2 cells, we found no effect of rhMG53 on insulin-stimulated Akt phosphorylation (p-Akt). We performed in vivo assay with C57BL/6J mice subjected to insulin stimulation (1 U/kg, intraperitoneal injection) and observed no effect of rhMG53 on insulin-stimulated p-Akt in muscle, heart and liver tissues. Conclusion Overall, our data suggest that rhMG53 can bind to the IR-ECD, however has a low likelihood of a physiologic role, as the Kd for binding is ~10,000 higher than the physiologic level of MG53 present in the serum of rodents and humans (~10 pM). Our findings question the notion proposed by Xiao and colleagues - whether targeting circulating MG53 opens a new therapeutic avenue for type 2 diabetes mellitus and its complications.
Collapse
Affiliation(s)
- Kyung Eun Lee
- Division of Surgical Sciences, Department of Surgery, University of Virginia, Charlottesville, VA, United States
| | - Miyuki Nishi
- Department of Biological Chemistry, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Jongsoo Kim
- Division of Surgical Sciences, Department of Surgery, University of Virginia, Charlottesville, VA, United States
| | - Takashi Murayama
- Department of Cellular and Molecular Pharmacology, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Zachary Dawson
- Division of Surgical Sciences, Department of Surgery, University of Virginia, Charlottesville, VA, United States
| | - Xiaoliang Wang
- Division of Surgical Sciences, Department of Surgery, University of Virginia, Charlottesville, VA, United States
| | - Xinyu Zhou
- Division of Surgical Sciences, Department of Surgery, University of Virginia, Charlottesville, VA, United States
| | - Tao Tan
- Division of Surgical Sciences, Department of Surgery, University of Virginia, Charlottesville, VA, United States
| | - Chuanxi Cai
- Division of Surgical Sciences, Department of Surgery, University of Virginia, Charlottesville, VA, United States
| | - Hiroshi Takeshima
- Department of Biological Chemistry, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Ki Ho Park
- Division of Surgical Sciences, Department of Surgery, University of Virginia, Charlottesville, VA, United States
| |
Collapse
|
5
|
Boengler K, Eickelmann C, Kleinbongard P. Mitochondrial Kinase Signaling for Cardioprotection. Int J Mol Sci 2024; 25:4491. [PMID: 38674076 PMCID: PMC11049936 DOI: 10.3390/ijms25084491] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 04/15/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
Myocardial ischemia/reperfusion injury is reduced by cardioprotective adaptations such as local or remote ischemic conditioning. The cardioprotective stimuli activate signaling cascades, which converge on mitochondria and maintain the function of the organelles, which is critical for cell survival. The signaling cascades include not only extracellular molecules that activate sarcolemmal receptor-dependent or -independent protein kinases that signal at the plasma membrane or in the cytosol, but also involve kinases, which are located to or within mitochondria, phosphorylate mitochondrial target proteins, and thereby modify, e.g., respiration, the generation of reactive oxygen species, calcium handling, mitochondrial dynamics, mitophagy, or apoptosis. In the present review, we give a personal and opinionated overview of selected protein kinases, localized to/within myocardial mitochondria, and summarize the available data on their role in myocardial ischemia/reperfusion injury and protection from it. We highlight the regulation of mitochondrial function by these mitochondrial protein kinases.
Collapse
Affiliation(s)
- Kerstin Boengler
- Institute of Physiology, Justus-Liebig University, 35392 Giessen, Germany
| | - Chantal Eickelmann
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, 45147 Essen, Germany; (C.E.); (P.K.)
| | - Petra Kleinbongard
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, 45147 Essen, Germany; (C.E.); (P.K.)
| |
Collapse
|
6
|
Farkašová F, Kindernay L, Ferko M, Rajtík T, Szobi A, Ravingerová T. Age-Dependent Effects of Remote Preconditioning in Hypertensive Rat Hearts are Associated With Activation of RISK Signaling. Physiol Res 2023; 72:S11-S22. [PMID: 37294114 DOI: 10.33549/physiolres.935019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/23/2023] Open
Abstract
Remote ischemic preconditioning (RIPC) represents one of the forms of innate cardioprotection. While being effective in animal models, its application in humans has not been always beneficial, which might be attributed to the presence of various comorbidities, such as hypertension, or being related to the confounding factors, such as patients' sex and age. RIPC has been shown to mediate its cardioprotective effects through the activation of Reperfusion Injury Salvage Kinase (RISK) pathway in healthy animals, however, scarce evidence supports this effect of RIPC in the hearts of spontaneously hypertensive (SHR) rats, in particular, in relationship with aging. The study aimed to investigate the effectiveness of RIPC in male SHR rats of different age and to evaluate the role of RISK pathway in the effect of RIPC on cardiac ischemic tolerance. RIPC was performed using three cycles of inflation/deflation of the pressure cuff placed on the hind limb of anesthetized rats aged three, five and eight months. Subsequently, hearts were excised, Langendorff-perfused and exposed to 30-min global ischemia and 2-h reperfusion. Infarct-sparing and antiarrhythmic effects of RIPC were observed only in three and five months-old animals but not in eight months-old rats. Beneficial effects of RIPC were associated with increased activity of RISK and decreased apoptotic signaling only in three and five months-old animals. In conclusion, RIPC showed cardioprotective effects in SHR rats that were partially age-dependent and might be attributed to the differences in the activation of RISK pathway and various aspects of ischemia/reperfusion injury in aging animals.
Collapse
Affiliation(s)
- Farkašová Farkašová
- Institute for Heart Research, Centre of Experimental Medicine Slovak Academy of Sciences, Bratislava, Slovak Republic.
| | | | | | | | | | | |
Collapse
|
7
|
Wang Q, Ju F, Li J, Liu T, Zuo Y, Abbott GW, Hu Z. Empagliflozin protects against renal ischemia/reperfusion injury in mice. Sci Rep 2022; 12:19323. [PMID: 36369319 PMCID: PMC9652474 DOI: 10.1038/s41598-022-24103-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 11/10/2022] [Indexed: 11/13/2022] Open
Abstract
Renal ischemia/reperfusion (I/R) can induce acute kidney injury. Empagliflozin is a newly developed inhibitor of sodium-glucose cotransporter-2 (SGLT2) approved as an antidiabetic medication for patients with type 2 diabetes mellitus. Despite the established cardioprotective functions of empagliflozin, its protective role in renal I/R is unclear. Here, the present study evaluated the renoprotective effects of empagliflozin in a mouse model of renal I/R injury. Male C57/BL6 mice were allocated to sham-operated, I/R, and empagliflozin groups. Kidney pedicles on both sides were clamped for 45 min and were reperfused for 24 h. Empagliflozin (1 mg/kg) was administered to the mice for 2 days preischemia. The GSK-3β inhibitor SB216763 was administered intravenously at the beginning of reperfusion (0.1 mg/kg). Renal function and histological scores were evaluated. The kidneys were taken for immunohistochemical analysis, western blotting and apoptosis measurements. We found that empagliflozin decreased serum levels of creatinine and urea, reduced the average kidney weight-to-tibia length ratio, attenuated tubular damage, reduced renal proinflammatory cytokine expression and inhibited apoptosis in injured kidneys. Furthermore, empagliflozin increased renal glycogen synthase kinase 3β (GSK-3β) phosphorylation post I/R. Pharmacological inhibition of GSK-3β activity mimicked the renal protective effects offered by empagliflozin. In summary, these results support a protective role of empagliflozin against renal I/R injury.
Collapse
Affiliation(s)
- Qifeng Wang
- grid.13291.380000 0001 0807 1581Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan China
| | - Feng Ju
- grid.13291.380000 0001 0807 1581Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan China
| | - Jiaxue Li
- grid.13291.380000 0001 0807 1581Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan China
| | - Ting Liu
- grid.13291.380000 0001 0807 1581Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan China
| | - Yunxia Zuo
- grid.13291.380000 0001 0807 1581Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan China
| | - Geoffrey W. Abbott
- grid.266093.80000 0001 0668 7243Bioelectricity Laboratory, Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA USA
| | - Zhaoyang Hu
- grid.13291.380000 0001 0807 1581Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan China
| |
Collapse
|
8
|
Hu B, Tian T, Hao PP, Liu WC, Chen YG, Jiang TY, Xue FS. The Protective Effect of Sevoflurane Conditionings Against Myocardial Ischemia/Reperfusion Injury: A Systematic Review and Meta-Analysis of Preclinical Trials in in-vivo Models. Front Cardiovasc Med 2022; 9:841654. [PMID: 35571167 PMCID: PMC9095933 DOI: 10.3389/fcvm.2022.841654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 03/16/2022] [Indexed: 11/24/2022] Open
Abstract
Objective Myocardial ischemia/reperfusion injury (IRI) is a common and serious complication in clinical practice. Sevoflurane conditionings have been identified to provide a protection against myocardial IRI in animal experiments, but their true clinical benefits remain controversial. Here, we aimed to analyze the preclinical evidences obtained in animal models of myocardial IRI and explore the possible reasons for controversial clinical benefits. Methods Our primary outcome was the difference in mean infarct size between the sevoflurane and control groups in animal models of myocardial IRI. After searching the databases of PubMed, Embase, Web of Science, and the Cochrane Library, a systematic review retrieved 37 eligible studies, from which 28 studies controlled comparisons of sevoflurane preconditioning (SPreC) and 40 studies controlled comparisons of sevoflurane postconditioning (SPostC) that were made in a pooled random-effects meta-analysis. In total, this analysis included data from 313 control animals and 536 animals subject to sevoflurane conditionings. Results Pooled estimates for primary outcome demonstrated that sevoflurane could significantly reduce the infarct size after myocardial IRI whether preconditioning [weighted mean difference (WMD): −18.56, 95% CI: −23.27 to −13.85, P < 0.01; I2 = 94.1%, P < 0.01] or postconditioning (WMD: −18.35, 95% CI: −20.88 to −15.83, P < 0.01; I2 = 90.5%, P < 0.01) was performed. Interestingly, there was significant heterogeneity in effect size that could not be explained by any of the prespecified variables by meta-regression and stratified analysis. However, sensitivity analysis still identified the cardioprotective benefits of sevoflurane conditionings with robust results. Conclusion Sevoflurane conditionings can significantly reduce infarct size in in-vivo models of myocardial IRI. Given the fact that there is a lack of consistency in the quality and design of included studies, more well-performed in-vivo studies with the detailed characterization of sevoflurane protocols, especially studies in larger animals regarding cardioprotection effects of sevoflurane, are still required.
Collapse
|
9
|
Zhong W, Benissan-Messan DZ, Ma J, Cai C, Lee PHU. Cardiac effects and clinical applications of MG53. Cell Biosci 2021; 11:115. [PMID: 34183055 PMCID: PMC8240287 DOI: 10.1186/s13578-021-00629-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 06/11/2021] [Indexed: 12/18/2022] Open
Abstract
Heart disease remains the leading cause of mortality globally, so further investigation is required to identify its underlying mechanisms and potential targets for treatment and prevention. Mitsugumin 53 (MG53), also known as TRIM72, is a TRIM family protein that was found to be involved in cell membrane repair and primarily found in striated muscle. Its role in skeletal muscle regeneration and myogenesis has been well documented. However, accumulating evidence suggests that MG53 has a potentially protective role in heart tissue, including in ischemia/reperfusion injury of the heart, cardiomyocyte membrane injury repair, and atrial fibrosis. This review summarizes the regulatory role of MG53 in cardiac tissues, current debates regarding MG53 in diabetes and diabetic cardiomyopathy, as well as highlights potential clinical applications of MG53 in treating cardiac pathologies.
Collapse
Affiliation(s)
- Weina Zhong
- Department of Surgery, The Ohio State University, Columbus, OH, USA
| | | | - Jianjie Ma
- Department of Surgery, The Ohio State University, Columbus, OH, USA
| | - Chuanxi Cai
- Department of Surgery, The Ohio State University, Columbus, OH, USA.
| | - Peter H U Lee
- Department of Surgery, The Ohio State University, Columbus, OH, USA.
- Department of Pathology and Laboratory Medicine, Brown University, Campus Box G-E5, 70 Ship Street, Providence, RI, 02912, USA.
- Department of Cardiothoracic Surgery, Southcoast Health, Fall River, MA, USA.
| |
Collapse
|
10
|
Benissan-Messan DZ, Zhu H, Zhong W, Tan T, Ma J, Lee PHU. Multi-Cellular Functions of MG53 in Muscle Calcium Signaling and Regeneration. Front Physiol 2020; 11:583393. [PMID: 33240103 PMCID: PMC7677405 DOI: 10.3389/fphys.2020.583393] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 10/09/2020] [Indexed: 12/28/2022] Open
Abstract
Since its identification in 2009, multiple studies have indicated the importance of MG53 in muscle physiology. The protein is produced in striated muscles but has physiologic implications reaching beyond the confines of striated muscles. Roles in muscle regeneration, calcium homeostasis, excitation-contraction coupling, myogenesis, and the mitochondria highlight the protein's wide-reaching impact. Numerous therapeutic applications could potentially emerge from these physiologic roles. This review summarizes the current literature regarding the role of MG53 in the skeletal muscle. Therapeutic applications are discussed.
Collapse
Affiliation(s)
| | - Hua Zhu
- Department of Surgery, The Ohio State University, Columbus, OH, United States
| | - Weina Zhong
- Department of Surgery, The Ohio State University, Columbus, OH, United States
| | - Tao Tan
- Department of Surgery, The Ohio State University, Columbus, OH, United States
| | - Jianjie Ma
- Department of Surgery, The Ohio State University, Columbus, OH, United States
| | - Peter H. U. Lee
- Department of Surgery, The Ohio State University, Columbus, OH, United States
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, United States
- Department of Cardiothoracic Surgery, Southcoast Health, Fall River, MA, United States
| |
Collapse
|
11
|
Bertocchi I, Foglietta F, Collotta D, Eva C, Brancaleone V, Thiemermann C, Collino M. The hidden role of NLRP3 inflammasome in obesity-related COVID-19 exacerbations: Lessons for drug repurposing. Br J Pharmacol 2020; 177:4921-4930. [PMID: 32776354 PMCID: PMC7436458 DOI: 10.1111/bph.15229] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 07/17/2020] [Accepted: 08/02/2020] [Indexed: 02/07/2023] Open
Abstract
COVID-19, the illness caused by SARS-CoV-2, has a wide-ranging clinical spectrum that, in the worst-case scenario, involves a rapid progression to severe acute respiratory syndrome and death. Epidemiological data show that obesity and diabetes are among the main risk factors associated with high morbidity and mortality. The increased susceptibility to SARS-CoV-2 infection documented in obesity-related metabolic derangements argues for initial defects in defence mechanisms, most likely due to an elevated systemic metabolic inflammation ("metaflammation"). The NLRP3 inflammasome is a master regulator of metaflammation and has a pivotal role in the pathophysiology of either obesity or diabetes. Here, we discuss the most recent findings suggesting contribution of NLRP3 inflammasome to the increase in complications in COVID-19 patients with diabesity. We also review current pharmacological strategies for COVID-19, focusing on treatments whose efficacy could be due, at least in part, to interference with the activation of the NLRP3 inflammasome. LINKED ARTICLES: This article is part of a themed issue on The Pharmacology of COVID-19. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v177.21/issuetoc.
Collapse
Affiliation(s)
- Ilaria Bertocchi
- Department of Neuroscience Rita Levi Montalcini, University of Turin, Turin, Italy.,University of Turin, Neuroscience Institute of the Cavalieri-Ottolenghi Foundation, Orbassano (TORINO), Italy
| | - Federica Foglietta
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Debora Collotta
- Department of Drug Science and Technology, University of Turin, Turin, Italy
| | - Carola Eva
- Department of Neuroscience Rita Levi Montalcini, University of Turin, Turin, Italy.,University of Turin, Neuroscience Institute of the Cavalieri-Ottolenghi Foundation, Orbassano (TORINO), Italy
| | | | - Christoph Thiemermann
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Massimo Collino
- Department of Drug Science and Technology, University of Turin, Turin, Italy
| |
Collapse
|
12
|
Wang Q, Bian Z, Jiang Q, Wang X, Zhou X, Park KH, Hsueh W, Whitson BA, Haggard E, Li H, Chen K, Cai C, Tan T, Zhu H, Ma J. MG53 Does Not Manifest the Development of Diabetes in db/db Mice. Diabetes 2020; 69:1052-1064. [PMID: 32139593 PMCID: PMC7171965 DOI: 10.2337/db19-0807] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 02/25/2020] [Indexed: 12/18/2022]
Abstract
MG53 is a member of the TRIM protein family that is predominantly expressed in striated muscles and participates in cell membrane repair. Controversy exists regarding MG53's role in insulin signaling and manifestation of diabetes. We generated db/db mice with either whole-body ablation or sustained elevation of MG53 in the bloodstream in order to evaluate the physiological function of MG53 in diabetes. To quantify the amount of MG53 protein in circulation, we developed a monoclonal antibody against MG53 with high specificity. Western blot using this antibody revealed lower or no change of serum MG53 levels in db/db mice or patients with diabetes compared with control subjects. Neither whole-body ablation of MG53 nor sustained elevation of MG53 in circulation altered insulin signaling and glucose handling in db/db mice. Instead, mice with ablation of MG53 were more susceptible to streptozotocin-induced dysfunctional handling of glucose compared with the wild-type littermates. Alkaline-induced corneal injury demonstrated delayed healing in db/db mice, which was restored by topical administration of recombinant human (rh)MG53. Daily intravenous administration of rhMG53 in rats at concentrations up to 10 mg/kg did not produce adverse effects on glucose handling. These findings challenge the hypothetical function of MG53 as a causative factor for the development of diabetes. Our data suggest that rhMG53 is a potentially safe and effective biologic to treat diabetic oculopathy in rodents.
Collapse
Affiliation(s)
- Qiang Wang
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH
| | - Zehua Bian
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH
| | - Qiwei Jiang
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH
| | - Xiaoliang Wang
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH
| | - Xinyu Zhou
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH
| | - Ki Ho Park
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH
| | - Willa Hsueh
- Diabetes and Metabolism Research Center, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH
| | - Bryan A Whitson
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH
| | - Erin Haggard
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH
| | - Haichang Li
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH
| | - Ken Chen
- Department of Cardiology, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, China
| | - Chuanxi Cai
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH
| | - Tao Tan
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH
- TRIM-edicine, Inc., Columbus, OH
| | - Hua Zhu
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH
| | - Jianjie Ma
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH
| |
Collapse
|
13
|
Adesanya TMA, Russell M, Park KH, Zhou X, Sermersheim MA, Gumpper K, Koenig SN, Tan T, Whitson BA, Janssen PML, Lincoln J, Zhu H, Ma J. MG 53 Protein Protects Aortic Valve Interstitial Cells From Membrane Injury and Fibrocalcific Remodeling. J Am Heart Assoc 2020; 8:e009960. [PMID: 30741589 PMCID: PMC6405656 DOI: 10.1161/jaha.118.009960] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Background The aortic valve of the heart experiences constant mechanical stress under physiological conditions. Maladaptive valve injury responses contribute to the development of valvular heart disease. Here, we test the hypothesis that MG 53 (mitsugumin 53), an essential cell membrane repair protein, can protect valvular cells from injury and fibrocalcific remodeling processes associated with valvular heart disease. Methods and Results We found that MG 53 is expressed in pig and human patient aortic valves and observed aortic valve disease in aged Mg53-/- mice. Aortic valves of Mg53-/- mice showed compromised cell membrane integrity. In vitro studies demonstrated that recombinant human MG 53 protein protects primary valve interstitial cells from mechanical injury and that, in addition to mediating membrane repair, recombinant human MG 53 can enter valve interstitial cells and suppress transforming growth factor-β-dependent activation of fibrocalcific signaling. Conclusions Together, our data characterize valve interstitial cell membrane repair as a novel mechanism of protection against valvular remodeling and assess potential in vivo roles of MG 53 in preventing valvular heart disease.
Collapse
Affiliation(s)
- T M Ayodele Adesanya
- 1 Department of Surgery The Ohio State University Wexner Medical Center Columbus OH
| | - Melanie Russell
- 1 Department of Surgery The Ohio State University Wexner Medical Center Columbus OH
| | - Ki Ho Park
- 1 Department of Surgery The Ohio State University Wexner Medical Center Columbus OH
| | - Xinyu Zhou
- 1 Department of Surgery The Ohio State University Wexner Medical Center Columbus OH
| | | | - Kristyn Gumpper
- 1 Department of Surgery The Ohio State University Wexner Medical Center Columbus OH
| | - Sara N Koenig
- 2 Department of Physiology and Cell Biology The Ohio State University Wexner Medical Center Columbus OH
| | - Tao Tan
- 1 Department of Surgery The Ohio State University Wexner Medical Center Columbus OH
| | - Bryan A Whitson
- 1 Department of Surgery The Ohio State University Wexner Medical Center Columbus OH
| | - Paul M L Janssen
- 2 Department of Physiology and Cell Biology The Ohio State University Wexner Medical Center Columbus OH
| | - Joy Lincoln
- 3 Center for Cardiovascular Research The Research Institute at Nationwide Children's Hospital Columbus OH
| | - Hua Zhu
- 1 Department of Surgery The Ohio State University Wexner Medical Center Columbus OH
| | - Jianjie Ma
- 1 Department of Surgery The Ohio State University Wexner Medical Center Columbus OH
| |
Collapse
|
14
|
|
15
|
Le Corvoisier P, Gallet R, Lesault PF, Audureau E, Paul M, Ternacle J, Ghostine S, Champagne S, Arrouasse R, Bitari D, Mouillet G, Dubois-Randé JL, Berdeaux A, Ghaleh B, Deux JF, Teiger E. Intra-coronary morphine versus placebo in the treatment of acute ST-segment elevation myocardial infarction: the MIAMI randomized controlled trial. BMC Cardiovasc Disord 2018; 18:193. [PMID: 30340532 PMCID: PMC6194573 DOI: 10.1186/s12872-018-0936-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 10/09/2018] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Experimental studies suggest that morphine may protect the myocardium against ischemia-reperfusion injury by activating salvage kinase pathways. The objective of this two-center, randomized, double-blind, controlled trial was to assess potential cardioprotective effects of intra-coronary morphine in patients with ST-segment elevation myocardial infarction (STEMI) referred for primary percutaneous intervention. METHODS Ninety-one patients with STEMI were randomly assigned to intracoronary morphine (1 mg) or placebo at reperfusion of the culprit coronary artery. The primary endpoint was infarct size/left ventricular mass ratio assessed by magnetic resonance imaging on day 3-5. Secondary endpoints included the areas under the curve (AUC) for troponin T and creatine kinase over three days, left ventricular ejection fraction assessed by echocardiography on days 1 and 6, and clinical outcomes. RESULTS Infarct size/left ventricular mass ratio was not significantly reduced by intracoronary morphine compared to placebo (27.2% ± 15.0% vs. 30.5% ± 10.6%, respectively, p = 0.28). Troponin T and creatine kinase AUCs were similar in the two groups. Morphine did not improve left ventricular ejection fraction on day 1 (49.7 ± 10.3% vs. 49.3 ± 9.3% with placebo, p = 0.84) or day 6 (48.5 ± 10.2% vs. 49.0 ± 8.5% with placebo, p = 0.86). The number of major adverse cardiac events, including stent thrombosis, during the one-year follow-up was similar in the two groups. CONCLUSIONS Intracoronary morphine at reperfusion did not significantly reduce infarct size or improve left ventricular systolic function in patients with STEMI. Presence of comorbidities in some patients may contribute to explain these results. TRIAL REGISTRATION ClinicalTrials.gov, NCT01186445 (date of registration: August 23, 2010).
Collapse
Affiliation(s)
- Philippe Le Corvoisier
- Department VERDI, Inserm, CIC1430, AP-HP, Henri Mondor Hospital, 51 Avenue du Maréchal de Lattre de Tassigny, F-94010, Creteil, France. .,Inserm, U955 team 3, F-94010, Creteil, France.
| | - Romain Gallet
- Inserm, U955 team 3, F-94010, Creteil, France.,Interventional Cardiology Unit, AP-HP, Henri Mondor Hospital, F-94010, Creteil, France
| | | | - Etienne Audureau
- Department of Public Health and CEPIA EA7376, AP-HP, Henri Mondor Hospital, F-94010, Creteil, France
| | - Muriel Paul
- Department of Pharmacy, AP-HP, Henri Mondor Hospital, F-94010, Creteil, France
| | - Julien Ternacle
- Interventional Cardiology Unit, AP-HP, Henri Mondor Hospital, F-94010, Creteil, France
| | - Saïd Ghostine
- Department of Cardiology, Marie-Lannelongue Hospital, F-92350, Le Plessis-Robinson, France
| | - Stéphane Champagne
- Interventional Cardiology Unit, AP-HP, Henri Mondor Hospital, F-94010, Creteil, France
| | - Raphaele Arrouasse
- Department VERDI, Inserm, CIC1430, AP-HP, Henri Mondor Hospital, 51 Avenue du Maréchal de Lattre de Tassigny, F-94010, Creteil, France
| | - Dalila Bitari
- Department VERDI, Inserm, CIC1430, AP-HP, Henri Mondor Hospital, 51 Avenue du Maréchal de Lattre de Tassigny, F-94010, Creteil, France
| | - Gauthier Mouillet
- Interventional Cardiology Unit, AP-HP, Henri Mondor Hospital, F-94010, Creteil, France
| | - Jean-Luc Dubois-Randé
- Inserm, U955 team 3, F-94010, Creteil, France.,Department of Cardiology, AP-HP, Henri Mondor Hospital, F-94010, Creteil, France
| | | | | | - Jean-François Deux
- Department of Radiology, AP-HP, Henri Mondor Hospital, F-94010, Creteil, France
| | - Emmanuel Teiger
- Inserm, U955 team 3, F-94010, Creteil, France.,Interventional Cardiology Unit, AP-HP, Henri Mondor Hospital, F-94010, Creteil, France
| |
Collapse
|
16
|
Yang JT, Wang J, Zhou XR, Xiao C, Lou YY, Tang LH, Zhang FJ, Qian LB. Luteolin alleviates cardiac ischemia/reperfusion injury in the hypercholesterolemic rat via activating Akt/Nrf2 signaling. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2018; 391:719-728. [PMID: 29671020 DOI: 10.1007/s00210-018-1496-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Accepted: 04/05/2018] [Indexed: 12/11/2022]
Abstract
Myocardial ischemia/reperfusion (I/R) injury in hypercholesterolemia is associated with oxidative stress, while luteolin is known to reduce oxidative stress by activating Akt/nuclear factor erythroid-2-related factor 2 (Nrf2) signaling and alleviate cardiac I/R injury. Here, we investigated whether luteolin pretreatment diminishes myocardial I/R injury in hypercholesterolemic rats by activating Akt/Nrf2 signaling. Hypercholesterolemic rats were produced by 2% cholesterol diet for 8 weeks. Luteolin (100 mg/kg/day, i.g.) or LY294002 was administered for the last 2 weeks. The hearts were then isolated and subjected to 30 min of global ischemia followed by 120 min of reperfusion. Pretreatment with luteolin significantly improved left ventricular function throughout reperfusion, increased cardiac tissue viability, reduced coronary lactate dehydrogenase release and the myocardial malondialdehyde level, upregulated p-Akt and p-GSK3β expressions, inhibited nuclear translocation of Fyn, and activated Nrf2 function in hypercholesterolemic I/R rat hearts. All these improving effects of luteolin were significantly attenuated by LY294002. Ca2+-induced mitochondrial permeability transition pore (mPTP) opening and mitochondrial inner membrane potential reduction were significantly inhibited in ventricular myocytes isolated from luteolin-treated hypercholesterolemic rats, which were attenuated by LY294002. These results indicate that luteolin protects the hypercholesterolemic heart against I/R injury due to upregulation of Akt-mediated Nrf2 antioxidative function and inhibition of mPTP.
Collapse
Affiliation(s)
- Jin-Ting Yang
- Department of Anesthesiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Jue Wang
- Department of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, 481 Binwen Road, Hangzhou, 310053, China
| | - Xin-Ru Zhou
- Department of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, 481 Binwen Road, Hangzhou, 310053, China
| | - Chi Xiao
- Department of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, 481 Binwen Road, Hangzhou, 310053, China
| | - Yang-Yun Lou
- Department of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, 481 Binwen Road, Hangzhou, 310053, China
| | - Li-Hui Tang
- Department of Anesthesiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Feng-Jiang Zhang
- Department of Anesthesiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Ling-Bo Qian
- Department of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, 481 Binwen Road, Hangzhou, 310053, China.
| |
Collapse
|
17
|
Sufentanil protects the rat myocardium against ischemia-reperfusion injury via activation of the ERK1/2 pathway. Cytotechnology 2017; 70:169-176. [PMID: 28856530 DOI: 10.1007/s10616-017-0127-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 07/21/2017] [Indexed: 01/29/2023] Open
Abstract
Sufentanil, a lipophilic opioid, is the most frequently used clinical drug for ischemic heart disease. The effects of sufentanil on MAPK signaling in ischemic heart disease were explored. The effects of sufentanil on ischemia-reperfusion (IR)-induced myocardial injury in a rat model were examined. The serum levels of CK, LDH, MDA and SOD, and the activities of Na+-K+-ATPase and Ca2+-Mg2+-ATPase were measured. The levels of total and phosphorylated ERK1/2, JNK, and p38 were measured by western blotting in the heart, and the myocardial H9C2 cell line was studied. Using the Cell Counting Kit-8, the growth rate of H9C2 cells affected by sufentanil was studied. The serum levels of CK, LDH and MDA were higher in the IR group than in the SO and SUF groups. The SOD level, as well as the activities of Na+-K+-ATPase and Ca2+-Mg2+-ATPase, were lower in the SO and SUF groups than in the IR group. The phosphorylated ERK1/2 level was lower in the IR group than in the SO and SUF groups. The growth rate of H9C2 cells increased with the concentration of sufentanil and the exposure time. The phosphorylated ERK level was upregulated by 4-12 h of sufentanil exposure, indicating that the effects were time-dependent. Furthermore, an inhibition of ERK signaling by chemical inhibition suppressed the sufentanil-mediated increase in the growth rate of H9C2 cells. Sufentanil appears to be beneficial for cases of worsening ischemic heart disease. Further studies are necessary before a clinical application is considered.
Collapse
|
18
|
Randomized comparison of sevoflurane versus propofol-remifentanil on the cardioprotective effects in elderly patients with coronary heart disease. BMC Anesthesiol 2017; 17:104. [PMID: 28800722 PMCID: PMC5553920 DOI: 10.1186/s12871-017-0397-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 08/06/2017] [Indexed: 01/22/2023] Open
Abstract
Background It is skeptical about cardioprotective property of sevoflurane in patients undergoing noncardiac surgery, especially in the elderly patients with coronary heart disease. We hypothesized that long duration of sevoflurane inhalation in noncardiac surgery could ameliorate myocardial damage in such patients. Methods This was a randomized, prospective study. One hundred twenty-one elderly patients with coronary heart disease were randomly allocated into two groups. Maintenance of anesthesia was achieved by sevoflurane inhalation (Group S) or propofol-remifentanil respectively (Group PR). Serum cardiac troponin I (cTnI) and brain natriuretic peptide (BNP) were measured before anesthesia induction (T0), 8 h (T1) and 24 h (T2) after anesthesia respectively. The perioperative cardiac output, complications and postoperative 3-month follow-up from end of surgery were recorded. Results Between the two groups, there were no statistical differences in the values of cTnI and BNP during the study. However, The area under the curve of cTnI values over 24 h after operation was less in Group S. Group PR had lower cardiac output and consumed more amount of phenylephrine during the study (P < 0.05). Conclusions Compared with the group PR, sevoflurane had no benefit in the myocardial protection for the elderly patients with CHD. However, Sevoflurane showed advantage in maintaining hemodynamic stability during the operative period. Trial registration Chinese Clinical Trial Registry, ChiCTR-IPR-16008871, 21 July 2016.
Collapse
|
19
|
Wang R, Xi L, Kukreja RC. PDE5 Inhibitor Tadalafil and Hydroxychloroquine Cotreatment Provides Synergistic Protection against Type 2 Diabetes and Myocardial Infarction in Mice. J Pharmacol Exp Ther 2017; 361:29-38. [PMID: 28123046 PMCID: PMC5363764 DOI: 10.1124/jpet.116.239087] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 01/23/2017] [Indexed: 12/15/2022] Open
Abstract
Diabetes is associated with a high risk for ischemic heart disease. We have previously shown that phosphodiesterase 5 inhibitor tadalafil (TAD) induces cardioprotection against ischemia/ reperfusion (I/R) injury in diabetic mice. Hydroxychloroquine (HCQ) is a widely used antimalarial and anti-inflammatory drug that has been reported to reduce hyperglycemia in diabetic patients. Therefore, we hypothesized that a combination of TAD and HCQ may induce synergistic cardioprotection in diabetes. We also investigated the role of insulin-Akt-mammalian target of rapamycin (mTOR) signaling, which regulates protein synthesis and cell survival. Adult male db/db mice were randomized to receive vehicle, TAD (6 mg/kg), HCQ (50 mg/kg), or TAD + HCQ daily by gastric gavage for 7 days. Hearts were isolated and subjected to 30-minute global ischemia, followed by 1-hour reperfusion in Langendorff mode. Cardiac function and myocardial infarct size were determined. Plasma glucose, insulin and lipid levels, and relevant pancreatic and cardiac protein markers were measured. Treatment with TAD + HCQ reduced myocardial infarct size (17.4% ± 4.3% vs. 37.8% ± 4.9% in control group, P < 0.05) and enhanced the production of ATP. The TAD + HCQ combination treatment also reduced fasting blood glucose, plasma free fatty acids, and triglyceride levels. Furthermore, TAD + HCQ increased plasma insulin levels (513 ± 73 vs. 232 ± 30 mU/liter, P < 0.05) with improved insulin sensitivity, larger pancreatic β-cell area, and pancreas mass. Insulin-like growth factor-1 (IGF-1) levels were also elevated by TAD + HCQ (343 ± 14 vs. 262 ± 22 ng/ml, P < 0.05). The increased insulin/IGF-1 resulted in activation of downstream Akt/mTOR cellular survival pathway. These results suggest that combination treatment with TAD and HCQ could be a novel and readily translational pharmacotherapy for reducing cardiovascular risk factors and protecting against myocardial I/R injury in type 2 diabetes.
Collapse
Affiliation(s)
- Rui Wang
- Pauley Heart Center, Division of Cardiology, Virginia Commonwealth University. Richmond, Virginia
| | - Lei Xi
- Pauley Heart Center, Division of Cardiology, Virginia Commonwealth University. Richmond, Virginia
| | - Rakesh C Kukreja
- Pauley Heart Center, Division of Cardiology, Virginia Commonwealth University. Richmond, Virginia
| |
Collapse
|
20
|
Andreadou I, Iliodromitis EK, Lazou A, Görbe A, Giricz Z, Schulz R, Ferdinandy P. Effect of hypercholesterolaemia on myocardial function, ischaemia-reperfusion injury and cardioprotection by preconditioning, postconditioning and remote conditioning. Br J Pharmacol 2017; 174:1555-1569. [PMID: 28060997 DOI: 10.1111/bph.13704] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Revised: 12/16/2016] [Accepted: 12/20/2016] [Indexed: 01/15/2023] Open
Abstract
Hypercholesterolaemia is considered to be a principle risk factor for cardiovascular disease, having direct negative effects on the myocardium itself, in addition to the development of atherosclerosis. Since hypercholesterolaemia affects the global cardiac gene expression profile, among many other factors, it results in increased myocardial oxidative stress, mitochondrial dysfunction and inflammation triggered apoptosis, all of which may account for myocardial dysfunction and increased susceptibility of the myocardium to infarction. In addition, numerous experimental and clinical studies have revealed that hyperlcholesterolaemia may interfere with the cardioprotective potential of conditioning mechanisms. Although not fully elucidated, the underlying mechanisms for the lost cardioprotection in hypercholesterolaemic animals have been reported to involve dysregulation of the endothelial NOS-cGMP, reperfusion injury salvage kinase, peroxynitrite-MMP2 signalling pathways, modulation of ATP-sensitive potassium channels and apoptotic pathways. In this review article, we summarize the current knowledge on the effect of hypercholesterolaemia on the non-ischaemic and ischaemic heart as well as on the cardioprotection induced by drugs or ischaemic preconditioning, postconditioning and remote conditioning. Future perspectives concerning the mechanisms and the design of preclinical and clinical trials are highlighted. LINKED ARTICLES This article is part of a themed section on Redox Biology and Oxidative Stress in Health and Disease. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.12/issuetoc.
Collapse
Affiliation(s)
- Ioanna Andreadou
- Laboratory of Pharmacology, Faculty of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Athens, Greece
| | - Efstathios K Iliodromitis
- Second Department of Cardiology, Medical School, National and Kapodistrian University of Athens, Attikon University Hospital, Athens, Greece
| | - Antigone Lazou
- School of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Anikó Görbe
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary.,Pharmahungary Group, Szeged, Hungary.,Cardiovascular Research Group, Department of Biochemistry, University of Szeged, Szeged, Hungary
| | - Zoltán Giricz
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary.,Pharmahungary Group, Szeged, Hungary
| | - Rainer Schulz
- Cardiovascular Research Group, Department of Biochemistry, University of Szeged, Szeged, Hungary.,Department of Physiology, Justus-Liebig-University, Giessen, Germany
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary.,Pharmahungary Group, Szeged, Hungary.,Cardiovascular Research Group, Department of Biochemistry, University of Szeged, Szeged, Hungary
| |
Collapse
|
21
|
Tan T, Ko YG, Ma J. Dual function of MG53 in membrane repair and insulin signaling. BMB Rep 2017; 49:414-23. [PMID: 27174502 PMCID: PMC5070728 DOI: 10.5483/bmbrep.2016.49.8.079] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Indexed: 12/20/2022] Open
Abstract
MG53 is a member of the TRIM-family protein that acts as a key component of the cell membrane repair machinery. MG53 is also an E3-ligase that ubiquinates insulin receptor substrate-1 and controls insulin signaling in skeletal muscle cells. Since its discovery in 2009, research efforts have been devoted to translate this basic discovery into clinical applications in human degenerative and metabolic diseases. This review article highlights the dual function of MG53 in cell membrane repair and insulin signaling, the mechanism that underlies the control of MG53 function, and the therapeutic value of targeting MG53 function in regenerative medicine. [BMB Reports 2016; 49(8): 414-423]
Collapse
Affiliation(s)
- Tao Tan
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Young-Gyu Ko
- Division of Life Sciences, Korea University, Seoul 02841, Korea
| | - Jianjie Ma
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| |
Collapse
|
22
|
See Hoe LE, May LT, Headrick JP, Peart JN. Sarcolemmal dependence of cardiac protection and stress-resistance: roles in aged or diseased hearts. Br J Pharmacol 2016; 173:2966-91. [PMID: 27439627 DOI: 10.1111/bph.13552] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Revised: 06/27/2016] [Accepted: 06/28/2016] [Indexed: 12/25/2022] Open
Abstract
Disruption of the sarcolemmal membrane is a defining feature of oncotic death in cardiac ischaemia-reperfusion (I-R), and its molecular makeup not only fundamentally governs this process but also affects multiple determinants of both myocardial I-R injury and responsiveness to cardioprotective stimuli. Beyond the influences of membrane lipids on the cytoprotective (and death) receptors intimately embedded within this bilayer, myocardial ionic homeostasis, substrate metabolism, intercellular communication and electrical conduction are all sensitive to sarcolemmal makeup, and critical to outcomes from I-R. As will be outlined in this review, these crucial sarcolemmal dependencies may underlie not only the negative effects of age and common co-morbidities on myocardial ischaemic tolerance but also the on-going challenge of implementing efficacious cardioprotection in patients suffering accidental or surgically induced I-R. We review evidence for the involvement of sarcolemmal makeup changes in the impairment of stress-resistance and cardioprotection observed with ageing and highly prevalent co-morbid conditions including diabetes and hypercholesterolaemia. A greater understanding of membrane changes with age/disease, and the inter-dependences of ischaemic tolerance and cardioprotection on sarcolemmal makeup, can facilitate the development of strategies to preserve membrane integrity and cell viability, and advance the challenging goal of implementing efficacious 'cardioprotection' in clinically relevant patient cohorts. Linked Articles This article is part of a themed section on Molecular Pharmacology of G Protein-Coupled Receptors. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v173.20/issuetoc.
Collapse
Affiliation(s)
- Louise E See Hoe
- Menzies Health Institute Queensland, Griffith University, Gold Coast, Australia.,Critical Care Research Group, The Prince Charles Hospital and The University of Queensland, Chermside, Queensland, Australia
| | - Lauren T May
- Monash Institute of Pharmaceutical Sciences, Monash University, Clayton, VIC, Australia
| | - John P Headrick
- Menzies Health Institute Queensland, Griffith University, Gold Coast, Australia
| | - Jason N Peart
- Menzies Health Institute Queensland, Griffith University, Gold Coast, Australia.
| |
Collapse
|
23
|
Avlas O, Srara S, Shainberg A, Aravot D, Hochhauser E. Silencing cardiomyocyte TLR4 reduces injury following hypoxia. Exp Cell Res 2016; 348:115-122. [PMID: 27448767 DOI: 10.1016/j.yexcr.2016.07.020] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2016] [Revised: 07/17/2016] [Accepted: 07/18/2016] [Indexed: 01/04/2023]
Abstract
Toll-like receptor 4 (TLR4), the receptor for lipopolysaccharide (LPS) of gram-negative pathogens expressed in the heart, is activated by several endogenous ligands associated with tissue injury in response to myocardial infarction (MI). The aim of this study was to investigate the involvement of TLR4 signaling in cardiomyocytes dysfunction following hypoxia (90min) using multiple methodologies such as knocking down TLR4 and small interfering RNA (siTLR4). Cardiomyocytes of C57Bl/6 mice (WT) subjected to hypoxic stress showed increased cardiac release of LDH, HMGB1, IκB, TNF-α and myocardial apoptotic and necrotic markers (BAX, PI) compared to TLR4 knock out mice (TLR4KO). Treating these cardiomyocytes with siRNA against TLR4 decreased the damage markers (LDH, IκB, TNF-α). TLR4 silencing during hypoxic stress resulted in the activation of the p-AKT and p-GSK3β (by ∼25%). The latter is an indicator that there is a reduction of mitochondrial permeability transition pore (mPTP) opening following hypoxic myocardial induced injury leading to preserved mitochondrial membrane potential. Silencing TLR4 in cardiomyocytes improved cell survival following hypoxic injury through activation of the AKT/GSK3β pathway, reduced inflammatory and apoptotic signals. These findings suggest that TLR4 may serve as a potential target in the treatment of ischemic myocardial injury. Moreover, RNA interfering targeting TLR4 expression represents a therapeutic strategy.
Collapse
Affiliation(s)
- Orna Avlas
- The Mina & Everard Goodman Faculty of Life Sciences Bar-Ilan University, Ramat Gan, Israel; The Cardiac Research Laboratory of the Department of Cardiothoracic Surgery, Felsenstein Medical Research Center, Rabin Medical Center, Tel Aviv University, Petah Tikva, Israel
| | - Smadar Srara
- The Mina & Everard Goodman Faculty of Life Sciences Bar-Ilan University, Ramat Gan, Israel
| | - Asher Shainberg
- The Mina & Everard Goodman Faculty of Life Sciences Bar-Ilan University, Ramat Gan, Israel
| | - Dan Aravot
- The Cardiac Research Laboratory of the Department of Cardiothoracic Surgery, Felsenstein Medical Research Center, Rabin Medical Center, Tel Aviv University, Petah Tikva, Israel
| | - Edith Hochhauser
- The Cardiac Research Laboratory of the Department of Cardiothoracic Surgery, Felsenstein Medical Research Center, Rabin Medical Center, Tel Aviv University, Petah Tikva, Israel.
| |
Collapse
|
24
|
Lemoine S, Tritapepe L, Hanouz JL, Puddu PE. The mechanisms of cardio-protective effects of desflurane and sevoflurane at the time of reperfusion: anaesthetic post-conditioning potentially translatable to humans? Br J Anaesth 2016; 116:456-75. [PMID: 26794826 DOI: 10.1093/bja/aev451] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Myocardial conditioning is actually an essential strategy in the management of ischaemia-reperfusion injury. The concept of anaesthetic post-conditioning is intriguing, its action occurring at a pivotal moment (that of reperfusion when ischaemia reperfusion lesions are initiated) where the activation of these cardio-protective mechanisms could overpower the mechanisms leading to ischaemia reperfusion injuries. Desflurane and sevoflurane are volatile anaesthetics frequently used during cardiac surgery. This review focuses on the efficacy of desflurane and sevoflurane administered during early reperfusion as a potential cardio-protective strategy. In the context of experimental studies in animal models and in human atrial tissues in vitro, the mechanisms underlying the cardio-protective effect of these agents and their capacity to induce post-conditioning have been reviewed in detail, underlining the role of reactive oxygen species generation, the activation of the cellular signalling pathways, and the actions on mitochondria along with the translatable actions in humans; this might well be sufficient to set the basis for launching randomized clinical studies, actually needed to confirm this strategy as one of real impact.
Collapse
Affiliation(s)
- S Lemoine
- Department of Anaesthesiology and Intensive Care, France and Faculty of Medicine, Centre Hospitalier Universitaire de Caen, Normandie Université, Pôle d'Anesthésie-Réanimation Chirurgicale - Niveau 6, CHU de Caen, Avenue Cote de Nacre, Caen Cedex 14033, France
| | - L Tritapepe
- Department of Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - J L Hanouz
- Department of Anaesthesiology and Intensive Care, France and Faculty of Medicine, Centre Hospitalier Universitaire de Caen, Normandie Université, Pôle d'Anesthésie-Réanimation Chirurgicale - Niveau 6, CHU de Caen, Avenue Cote de Nacre, Caen Cedex 14033, France
| | - P E Puddu
- Department of Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
25
|
Protective effect of hyperoside on cardiac ischemia reperfusion injury through inhibition of ER stress and activation of Nrf2 signaling. ASIAN PAC J TROP MED 2016; 9:76-80. [DOI: 10.1016/j.apjtm.2015.12.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Revised: 11/20/2015] [Accepted: 12/03/2015] [Indexed: 11/20/2022] Open
|
26
|
Duann P, Li H, Lin P, Tan T, Wang Z, Chen K, Zhou X, Gumpper K, Zhu H, Ludwig T, Mohler PJ, Rovin B, Abraham WT, Zeng C, Ma J. MG53-mediated cell membrane repair protects against acute kidney injury. Sci Transl Med 2015; 7:279ra36. [PMID: 25787762 DOI: 10.1126/scitranslmed.3010755] [Citation(s) in RCA: 108] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Injury to the renal proximal tubular epithelium (PTE) represents the underlying consequence of acute kidney injury (AKI) after exposure to various stressors, including nephrotoxins and ischemia/reperfusion (I/R). Although the kidney has the ability to repair itself after mild injury, insufficient repair of PTE cells may trigger inflammatory and fibrotic responses, leading to chronic renal failure. We report that MG53, a member of the TRIM family of proteins, participates in repair of injured PTE cells and protects against the development of AKI. We show that MG53 translocates to acute injury sites on PTE cells and forms a repair patch. Ablation of MG53 leads to defective membrane repair. MG53-deficient mice develop pronounced tubulointerstitial injury and increased susceptibility to I/R-induced AKI compared to wild-type mice. Recombinant human MG53 (rhMG53) protein can target injury sites on PTE cells to facilitate repair after I/R injury or nephrotoxin exposure. Moreover, in animal studies, intravenous delivery of rhMG53 ameliorates cisplatin-induced AKI without affecting the tumor suppressor efficacy of cisplatin. These findings identify MG53 as a vital component of reno-protection, and targeting MG53-mediated repair of PTE cells represents a potential approach to prevention and treatment of AKI.
Collapse
Affiliation(s)
- Pu Duann
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Haichang Li
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Peihui Lin
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Tao Tan
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Zhen Wang
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing Institute of Cardiology, Chongqing 400042, China
| | - Ken Chen
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing Institute of Cardiology, Chongqing 400042, China
| | - Xinyu Zhou
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Kristyn Gumpper
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Hua Zhu
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Thomas Ludwig
- Department of Molecular and Cellular Biochemistry, The Ohio State University, Columbus, OH 43210, USA
| | - Peter J Mohler
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA.,Department of Medicine, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Brad Rovin
- Department of Medicine, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - William T Abraham
- Department of Medicine, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Chunyu Zeng
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing Institute of Cardiology, Chongqing 400042, China
| | - Jianjie Ma
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA.,Department of Molecular and Cellular Biochemistry, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
27
|
Maladaptive Modulations of NLRP3 Inflammasome and Cardioprotective Pathways Are Involved in Diet-Induced Exacerbation of Myocardial Ischemia/Reperfusion Injury in Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2016:3480637. [PMID: 26788246 PMCID: PMC4691622 DOI: 10.1155/2016/3480637] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 09/09/2015] [Indexed: 12/22/2022]
Abstract
Excessive fatty acids and sugars intake is known to affect the development of cardiovascular diseases, including myocardial infarction. However, the underlying mechanisms are ill defined. Here we investigated the balance between prosurvival and detrimental pathways within the heart of C57Bl/6 male mice fed a standard diet (SD) or a high-fat high-fructose diet (HFHF) for 12 weeks and exposed to cardiac ex vivo ischemia/reperfusion (IR) injury. Dietary manipulation evokes a maladaptive response in heart mice, as demonstrated by the shift of myosin heavy chain isoform content from α to β, the increased expression of the Nlrp3 inflammasome and markers of oxidative metabolism, and the downregulation of the hypoxia inducible factor- (HIF-)2α and members of the Reperfusion Injury Salvage Kinases (RISK) pathway. When exposed to IR, HFHF mice hearts showed greater infarct size and lactic dehydrogenase release in comparison with SD mice. These effects were associated with an exacerbated overexpression of Nlrp3 inflammasome, resulting in marked caspase-1 activation and a compromised activation of the cardioprotective RISK/HIF-2α pathways. The common mechanisms of damage here reported lead to a better understanding of the cross-talk among prosurvival and detrimental pathways leading to the development of cardiovascular disorders associated with metabolic diseases.
Collapse
|
28
|
Ravingerová T, Ledvényiová-Farkašová V, Ferko M, Barteková M, Bernátová I, Pecháňová O, Adameová A, Kolář F, Lazou A. Pleiotropic preconditioning-like cardioprotective effects of hypolipidemic drugs in acute ischemia–reperfusion in normal and hypertensive rats. Can J Physiol Pharmacol 2015; 93:495-503. [DOI: 10.1139/cjpp-2014-0502] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Although pleiotropy, which is defined as multiple effects derived from a single gene, was recognized many years ago, and considerable progress has since been achieved in this field, it is not very clear how much this feature of a drug is clinically relevant. During the last decade, beneficial pleiotropic effects from hypolipidemic drugs (as in, effects that are different from the primary ones) have been associated with reduction of cardiovascular risk. As with statins, the agonists of peroxisome proliferator-activated receptors (PPARs), niacin and fibrates, have been suggested to exhibit pleiotropic activity that could significantly modify the outcome of a cardiovascular ailment. This review examines findings demonstrating the impacts of treatment with hypolipidemic drugs on cardiac response to ischemia in a setting of acute ischemia–reperfusion, in relation to PPAR activation. Specifically, it addresses the issue of susceptibility to ischemia, with particular regard to the preconditioning-like cardioprotection conferred by hypolipidemic drugs, as well as the potential molecular mechanisms behind this cardioprotection. Finally, the involvement of PPAR activation in the mechanisms of non-metabolic cardioprotective effects from hypolipidemic drugs, and their effects on normal and pathologically altered myocardium (in the hearts of hypertensive rats) is also discussed.
Collapse
Affiliation(s)
- Táňa Ravingerová
- Institute for Heart Research, Slovak Academy of Sciences and Centre of Excellence of SAS NOREG, POB 104, Dúbravská cesta 9, 840 05 Bratislava, Slovak Republic
| | - Veronika Ledvényiová-Farkašová
- Institute for Heart Research, Slovak Academy of Sciences and Centre of Excellence of SAS NOREG, POB 104, Dúbravská cesta 9, 840 05 Bratislava, Slovak Republic
| | - Miroslav Ferko
- Institute for Heart Research, Slovak Academy of Sciences and Centre of Excellence of SAS NOREG, POB 104, Dúbravská cesta 9, 840 05 Bratislava, Slovak Republic
| | - Monika Barteková
- Institute for Heart Research, Slovak Academy of Sciences and Centre of Excellence of SAS NOREG, POB 104, Dúbravská cesta 9, 840 05 Bratislava, Slovak Republic
| | - Iveta Bernátová
- Institute of Normal and Pathological Physiology, Slovak Academy of Sciences and Centre of Excellence of SAS NOREG, Bratislava, Slovak Republic
| | - Ol’ga Pecháňová
- Institute of Normal and Pathological Physiology, Slovak Academy of Sciences and Centre of Excellence of SAS NOREG, Bratislava, Slovak Republic
| | - Adriana Adameová
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University, Bratislava, Slovak Republic
| | - František Kolář
- Institute of Physiology, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Antigone Lazou
- School of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
29
|
Effect of metabolic syndrome on mitsugumin 53 expression and function. PLoS One 2015; 10:e0124128. [PMID: 25950605 PMCID: PMC4423930 DOI: 10.1371/journal.pone.0124128] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2014] [Accepted: 02/26/2015] [Indexed: 12/25/2022] Open
Abstract
Metabolic syndrome is a cluster of risk factors, such as obesity, insulin resistance, and hyperlipidemia that increases the individual’s likelihood of developing cardiovascular diseases. Patients inflicted with metabolic disorders also suffer from tissue repair defect. Mitsugumin 53 (MG53) is a protein essential to cellular membrane repair. It facilitates the nucleation of intracellular vesicles to sites of membrane disruption to create repair patches, contributing to the regenerative capacity of skeletal and cardiac muscle tissues upon injury. Since individuals suffering from metabolic syndrome possess tissue regeneration deficiency and MG53 plays a crucial role in restoring membrane integrity, we studied MG53 activity in mice models exhibiting metabolic disorders induced by a 6 month high-fat diet (HFD) feeding. Western blotting showed that MG53 expression is not altered within the skeletal and cardiac muscles of mice with metabolic syndrome. Rather, we found that MG53 levels in blood circulation were actually reduced. This data directly contradicts findings presented by Song et. al that indict MG53 as a causative factor for metabolic syndrome (Nature 494, 375-379). The diminished MG53 serum level observed may contribute to the inadequate tissue repair aptitude exhibited by diabetic patients. Furthermore, immunohistochemical analyses reveal that skeletal muscle fibers of mice with metabolic disorders experience localization of subcellular MG53 around mitochondria. This clustering may represent an adaptive response to oxidative stress resulting from HFD feeding and may implicate MG53 as a guardian to protect damaged mitochondria. Therapeutic approaches that elevate MG53 expression in serum circulation may be a novel method to treat the degenerative tissue repair function of diabetic patients.
Collapse
|
30
|
Ferdinandy P, Hausenloy DJ, Heusch G, Baxter GF, Schulz R. Interaction of risk factors, comorbidities, and comedications with ischemia/reperfusion injury and cardioprotection by preconditioning, postconditioning, and remote conditioning. Pharmacol Rev 2014; 66:1142-74. [PMID: 25261534 DOI: 10.1124/pr.113.008300] [Citation(s) in RCA: 473] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Pre-, post-, and remote conditioning of the myocardium are well described adaptive responses that markedly enhance the ability of the heart to withstand a prolonged ischemia/reperfusion insult and provide therapeutic paradigms for cardioprotection. Nevertheless, more than 25 years after the discovery of ischemic preconditioning, we still do not have established cardioprotective drugs on the market. Most experimental studies on cardioprotection are still undertaken in animal models, in which ischemia/reperfusion is imposed in the absence of cardiovascular risk factors. However, ischemic heart disease in humans is a complex disorder caused by, or associated with, cardiovascular risk factors and comorbidities, including hypertension, hyperlipidemia, diabetes, insulin resistance, heart failure, altered coronary circulation, and aging. These risk factors induce fundamental alterations in cellular signaling cascades that affect the development of ischemia/reperfusion injury per se and responses to cardioprotective interventions. Moreover, some of the medications used to treat these risk factors, including statins, nitrates, and antidiabetic drugs, may impact cardioprotection by modifying cellular signaling. The aim of this article is to review the recent evidence that cardiovascular risk factors and their medication may modify the response to cardioprotective interventions. We emphasize the critical need to take into account the presence of cardiovascular risk factors and concomitant medications when designing preclinical studies for the identification and validation of cardioprotective drug targets and clinical studies. This will hopefully maximize the success rate of developing rational approaches to effective cardioprotective therapies for the majority of patients with multiple risk factors.
Collapse
Affiliation(s)
- Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Cardiovascular Research Group, Department of Biochemistry, University of Szeged, Szeged and Pharmahungary Group, Szeged, Hungary (P.F.); The Hatter Cardiovascular Institute, University College London, London, United Kingdom (D.J.H.); Institute for Pathophysiology, University of Essen Medical School, Essen, Germany (G.H.); Division of Pharmacology, Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, United Kingdom (G.F.B.); and Institute of Physiology, Justus-Liebig University, Giessen, Germany (R.S.)
| | - Derek J Hausenloy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Cardiovascular Research Group, Department of Biochemistry, University of Szeged, Szeged and Pharmahungary Group, Szeged, Hungary (P.F.); The Hatter Cardiovascular Institute, University College London, London, United Kingdom (D.J.H.); Institute for Pathophysiology, University of Essen Medical School, Essen, Germany (G.H.); Division of Pharmacology, Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, United Kingdom (G.F.B.); and Institute of Physiology, Justus-Liebig University, Giessen, Germany (R.S.)
| | - Gerd Heusch
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Cardiovascular Research Group, Department of Biochemistry, University of Szeged, Szeged and Pharmahungary Group, Szeged, Hungary (P.F.); The Hatter Cardiovascular Institute, University College London, London, United Kingdom (D.J.H.); Institute for Pathophysiology, University of Essen Medical School, Essen, Germany (G.H.); Division of Pharmacology, Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, United Kingdom (G.F.B.); and Institute of Physiology, Justus-Liebig University, Giessen, Germany (R.S.)
| | - Gary F Baxter
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Cardiovascular Research Group, Department of Biochemistry, University of Szeged, Szeged and Pharmahungary Group, Szeged, Hungary (P.F.); The Hatter Cardiovascular Institute, University College London, London, United Kingdom (D.J.H.); Institute for Pathophysiology, University of Essen Medical School, Essen, Germany (G.H.); Division of Pharmacology, Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, United Kingdom (G.F.B.); and Institute of Physiology, Justus-Liebig University, Giessen, Germany (R.S.)
| | - Rainer Schulz
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Cardiovascular Research Group, Department of Biochemistry, University of Szeged, Szeged and Pharmahungary Group, Szeged, Hungary (P.F.); The Hatter Cardiovascular Institute, University College London, London, United Kingdom (D.J.H.); Institute for Pathophysiology, University of Essen Medical School, Essen, Germany (G.H.); Division of Pharmacology, Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, United Kingdom (G.F.B.); and Institute of Physiology, Justus-Liebig University, Giessen, Germany (R.S.)
| |
Collapse
|