1
|
Qian Y, Qi Y, Lin J, Zhang T, Mo L, Xue Q, Zheng N, Niu Y, Dong X, Shi Y, Jiang Y. AdipoRon ameliorates chronic ethanol induced cardiac necroptosis by reducing ceramide mediated mtROS. Free Radic Biol Med 2025; 229:237-250. [PMID: 39805512 DOI: 10.1016/j.freeradbiomed.2025.01.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 12/30/2024] [Accepted: 01/11/2025] [Indexed: 01/16/2025]
Abstract
Chronic ethanol (EtOH) consumption has been widely recognized as a significant contributor to cardiotoxicity. However, no specific treatment is currently available to ameliorate chronic ethanol induced cardiotoxicity. Adiponectin receptor agonist AdipoRon exerts protective effects in multiple organs through alleviating lipotoxicity. Our previous study showed that chronic ethanol consumption increased de novo ceramide synthesis and necroptosis in myocardium. In this study, we investigated the role of AdipoRon on ceramide metabolism and necroptosis in chronic ethanol-treated myocardium. Eight-week-old C57/BL6J mice were fed with a Lieber-Decarli diet containing vehicle or AdipoRon for 12 weeks. Cardiac function, histology and oxidative stress were assessed. We found that chronic ethanol treatment decreased expression of AdipoR2 in myocardium and H9c2 cells, whereas AdipoRon improved cardiac function, reduced myocardium ceramide levels and suppressed necroptosis. By pharmacological interventions, RNA interference and point mutations in AdipoR2, we demonstrated that AdipoRon reduced ceramide levels through PPARα mediated lipid metabolism rather than AdipoR2's ceramidase activity. Using transmission electron microscope and reactive oxygen species (ROS) staining, we showed that chronic ethanol induced myocardium mitochondria damage and mitochondrial reactive oxygen species (mtROS) accumulation. Meanwhile, we found that AdipoRon ameliorated chronic ethanol induced cardiac necroptosis via the SIRT3-SOD2-mtROS pathway. Moreover, C6 ceramide treatment recapitulated chronic ethanol in inducing mtROS and necroptosis, whereas the ceramide synthesis inhibitors myriocin (MYR) and fumonisin B1 (FB1) attenuated chronic ethanol induced mtROS and necroptosis. Collectively, AdipoRon ameliorates chronic ethanol induced cardiac necroptosis by reducing ceramide de novo synthesis and mtROS, which highlights the therapeutic potential of targeting ceramide metabolism and oxidative stress pathways in treating ethanol induced cardiotoxicity.
Collapse
Affiliation(s)
- Yile Qian
- Department of Forensic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Yanyu Qi
- School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Junyi Lin
- Department of Forensic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Tianyi Zhang
- Department of Forensic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Lingjie Mo
- Department of Forensic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Qiupeng Xue
- Forensic Science and Information Technology Research Centre of Supreme People's Procuratorate, Beijing, 100726, China
| | - Nianchang Zheng
- Department of Forensic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Yaqin Niu
- Department of Forensic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Xiaoru Dong
- Department of Forensic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Yan Shi
- Academy of Forensic Science Shanghai Key Laboratory of Forensic Medicine, Shanghai, 200063, China.
| | - Yan Jiang
- Department of Forensic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
2
|
Gianopoulos I, Mantzoros CS, Daskalopoulou SS. Adiponectin and Adiponectin Receptors in Atherosclerosis. Endocr Rev 2025; 46:1-25. [PMID: 39106421 PMCID: PMC11720176 DOI: 10.1210/endrev/bnae021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 05/14/2024] [Accepted: 08/02/2024] [Indexed: 08/09/2024]
Abstract
Adiponectin is an abundantly secreted hormone that communicates information between the adipose tissue, and the immune and cardiovascular systems. In metabolically healthy individuals, adiponectin is usually found at high levels and helps improve insulin responsiveness of peripheral tissues, glucose tolerance, and fatty acid oxidation. Beyond its metabolic functions in insulin-sensitive tissues, adiponectin plays a prominent role in attenuating the development of atherosclerotic plaques, partially through regulating macrophage-mediated responses. In this context, adiponectin binds to its receptors, adiponectin receptor 1 (AdipoR1) and AdipoR2 on the cell surface of macrophages to activate a downstream signaling cascade and induce specific atheroprotective functions. Notably, macrophages modulate the stability of the plaque through their ability to switch between proinflammatory responders, and anti-inflammatory proresolving mediators. Traditionally, the extremes of the macrophage polarization spectrum span from M1 proinflammatory and M2 anti-inflammatory phenotypes. Previous evidence has demonstrated that the adiponectin-AdipoR pathway influences M1-M2 macrophage polarization; adiponectin promotes a shift toward an M2-like state, whereas AdipoR1- and AdipoR2-specific contributions are more nuanced. To explore these concepts in depth, we discuss in this review the effect of adiponectin and AdipoR1/R2 on 1) metabolic and immune responses, and 2) M1-M2 macrophage polarization, including their ability to attenuate atherosclerotic plaque inflammation, and their potential as therapeutic targets for clinical applications.
Collapse
Affiliation(s)
- Ioanna Gianopoulos
- Division of Experimental Medicine, Department of Medicine, Faculty of Medicine, Research Institute of the McGill University Health Centre, McGill University, Montreal, Quebec H4A 3J1, Canada
| | - Christos S Mantzoros
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
- Section of Endocrinology, Diabetes and Metabolism, Boston VA Healthcare System, Boston, MA 02130, USA
| | - Stella S Daskalopoulou
- Division of Experimental Medicine, Department of Medicine, Faculty of Medicine, Research Institute of the McGill University Health Centre, McGill University, Montreal, Quebec H4A 3J1, Canada
- Division of Internal Medicine, Department of Medicine, Faculty of Medicine, McGill University Health Centre, McGill University, Montreal, Quebec H4A 3J1, Canada
| |
Collapse
|
3
|
Han R, Huang H, Zhu J, Jin X, Wang Y, Xu Y, Xia Z. Adipokines and their potential impacts on susceptibility to myocardial ischemia/reperfusion injury in diabetes. Lipids Health Dis 2024; 23:372. [PMID: 39538244 PMCID: PMC11558907 DOI: 10.1186/s12944-024-02357-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024] Open
Abstract
Coronary artery disease has a high mortality rate and is a striking public health concern, affecting a substantial portion of the global population. On the early onset of myocardial ischemia, thrombolytic therapy and coronary revascularization could promptly restore the bloodstream and nutrient supply to the ischemic tissue, efficiently preserving less severely injured myocardium. However, the abrupt re-establishment of blood flow triggers the significant discharge of previously accumulated oxidative substances and inflammatory cytokines, leading to further harm referred to as ischemia/reperfusion (I/R) injury. Diabetes significantly raises the vulnerability of the heart to I/R injury due to disrupted glucose and lipid processing, impaired insulin sensitivity and metabolic signaling, and increased inflammatory responses. Numerous studies have indicated that adipokines are crucial in the etiology and pathogenesis of obesity, diabetes, hyperlipidemia, hypertension, and coronary artery disease. Adipokines such as adiponectin, adipsin, visfatin, chemerin, omentin, and apelin, which possess protective properties against inflammatory activity and insulin resistance, have been shown to confer myocardial protection in conditions such as atherosclerosis, myocardial hypertrophy, myocardial I/R injury, and diabetic complications. On the other hand, adipokines such as leptin and resistin, known for their pro-inflammatory characteristics, have been linked to elevated cardiac lipid deposition, insulin resistance, and fibrosis. Meteorin-like (metrnl) exhibits opposite effects in various pathological conditions. However, the data on adipokines in myocardial I/R, especially in diabetes, is still incomplete and controversial. This review focuses on recent research regarding the categorization and function of adipokines in the heart muscle, and the identification of different signaling pathways involved in myocardial I/R injury under diabetic conditions, aiming to facilitate the exploration of therapeutic strategies against myocardial I/R injury in diabetes.
Collapse
Affiliation(s)
- Ronghui Han
- Faculty of Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macao, PR, China
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Guangdong, China
| | - Hemeng Huang
- Department of Emergency, Affiliated Hospital of Guangdong Medical University, Guangdong, China
| | - Jianyu Zhu
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Guangdong, China
| | - Xiaogao Jin
- Department of Anesthesiology, The Second Affiliated Hospital of Guangdong Medical University, Guangdong, China
| | - Yongyan Wang
- Faculty of Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macao, PR, China
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Guangdong, China
| | - Youhua Xu
- Faculty of Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macao, PR, China.
- Macau University of Science and Technology Zhuhai MUST Science and Technology Research Institute, Hengqin, Zhuhai, People's Republic of China.
- Faculty of Pharmacy, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macao, People's Republic of China.
| | - Zhengyuan Xia
- Faculty of Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macao, PR, China.
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Guangdong, China.
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Medicine, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
4
|
Fu L, Du J, Furkert D, Shipton ML, Liu X, Aguirre T, Chin AC, Riley AM, Potter BVL, Fiedler D, Zhang X, Zhu Y, Fu C. Depleting inositol pyrophosphate 5-InsP7 protected the heart against ischaemia-reperfusion injury by elevating plasma adiponectin. Cardiovasc Res 2024; 120:954-970. [PMID: 38252884 PMCID: PMC11218692 DOI: 10.1093/cvr/cvae017] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 11/20/2023] [Accepted: 11/30/2023] [Indexed: 01/24/2024] Open
Abstract
AIMS Adiponectin is an adipocyte-derived circulating protein that exerts cardiovascular and metabolic protection. Due to the futile degradation of endogenous adiponectin and the challenges of exogenous administration, regulatory mechanisms of adiponectin biosynthesis are of significant pharmacological interest. METHODS AND RESULTS Here, we report that 5-diphosphoinositol 1,2,3,4,6-pentakisphosphate (5-InsP7) generated by inositol hexakisphosphate kinase 1 (IP6K1) governed circulating adiponectin levels via thiol-mediated protein quality control in the secretory pathway. IP6K1 bound to adiponectin and DsbA-L and generated 5-InsP7 to stabilize adiponectin/ERp44 and DsbA-L/Ero1-Lα interactions, driving adiponectin intracellular degradation. Depleting 5-InsP7 by either IP6K1 deletion or pharmacological inhibition blocked intracellular adiponectin degradation. Whole-body and adipocyte-specific deletion of IP6K1 boosted plasma adiponectin levels, especially its high molecular weight forms, and activated AMPK-mediated protection against myocardial ischaemia-reperfusion injury. Pharmacological inhibition of 5-InsP7 biosynthesis in wild-type but not adiponectin knockout mice attenuated myocardial ischaemia-reperfusion injury. CONCLUSION Our findings revealed that 5-InsP7 is a physiological regulator of adiponectin biosynthesis that is amenable to pharmacological intervention for cardioprotection.
Collapse
Affiliation(s)
- Lin Fu
- Tianjin Key Laboratory of Metabolic Diseases, Department of Physiology and Pathophysiology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, 22 Qixiangtai Road, Heping District, Tianjin 300070, China
| | - Jimin Du
- Tianjin Key Laboratory of Metabolic Diseases, Department of Physiology and Pathophysiology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, 22 Qixiangtai Road, Heping District, Tianjin 300070, China
| | - David Furkert
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany
| | - Megan L Shipton
- Medicinal Chemistry and Drug Discovery, Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
| | - Xiaoqi Liu
- Tianjin Key Laboratory of Metabolic Diseases, Department of Physiology and Pathophysiology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, 22 Qixiangtai Road, Heping District, Tianjin 300070, China
| | - Tim Aguirre
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany
| | - Alfred C Chin
- Weill Cornell/Rockefeller/Sloan Kettering Tri-Institutional MD-PhD Program, New York, NY, USA
| | - Andrew M Riley
- Medicinal Chemistry and Drug Discovery, Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
| | - Barry V L Potter
- Medicinal Chemistry and Drug Discovery, Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
| | - Dorothea Fiedler
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany
| | - Xu Zhang
- Tianjin Key Laboratory of Metabolic Diseases, Department of Physiology and Pathophysiology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, 22 Qixiangtai Road, Heping District, Tianjin 300070, China
| | - Yi Zhu
- Tianjin Key Laboratory of Metabolic Diseases, Department of Physiology and Pathophysiology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, 22 Qixiangtai Road, Heping District, Tianjin 300070, China
| | - Chenglai Fu
- Tianjin Key Laboratory of Metabolic Diseases, Department of Physiology and Pathophysiology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, 22 Qixiangtai Road, Heping District, Tianjin 300070, China
- Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Yangpu District, Shanghai 200092, China
| |
Collapse
|
5
|
Yue H, Zhang Q, Chang S, Zhao X, Wang M, Li W. Adiponectin protects against myocardial ischemia-reperfusion injury: a systematic review and meta-analysis of preclinical animal studies. Lipids Health Dis 2024; 23:51. [PMID: 38368320 PMCID: PMC10874037 DOI: 10.1186/s12944-024-02028-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Accepted: 01/22/2024] [Indexed: 02/19/2024] Open
Abstract
BACKGROUND Myocardial ischemia-reperfusion injury (MIRI) is widespread in the treatment of ischemic heart disease, and its treatment options are currently limited. Adiponectin (APN) is an adipocytokine with cardioprotective properties; however, the mechanisms of APN in MIRI are unclear. Therefore, based on preclinical (animal model) evidence, the cardioprotective effects of APN and the underlying mechanisms were explored. METHODS The literature was searched for the protective effect of APN on MIRI in six databases until 16 November 2023, and data were extracted according to selection criteria. The outcomes were the size of the myocardial necrosis area and hemodynamics. Markers of oxidation, apoptosis, and inflammation were secondary outcome indicators. The quality evaluation was performed using the animal study evaluation scale recommended by the Systematic Review Center for Laboratory animal Experimentation statement. Stata/MP 14.0 software was used for the summary analysis. RESULTS In total, 20 papers with 426 animals were included in this study. The pooled analysis revealed that APN significantly reduced myocardial infarct size [weighted mean difference (WMD) = 16.67 (95% confidence interval (CI) = 13.18 to 20.16, P < 0.001)] and improved hemodynamics compared to the MIRI group [Left ventricular end-diastolic pressure: WMD = 5.96 (95% CI = 4.23 to 7.70, P < 0.001); + dP/dtmax: WMD = 1393.59 (95% CI = 972.57 to 1814.60, P < 0.001); -dP/dtmax: WMD = 850.06 (95% CI = 541.22 to 1158.90, P < 0.001); Left ventricular ejection fraction: WMD = 9.96 (95% CI = 7.29 to 12.63, P < 0.001)]. Apoptosis indicators [caspase-3: standardized mean difference (SMD) = 3.86 (95% CI = 2.97 to 4.76, P < 0.001); TUNEL-positive cells: WMD = 13.10 (95% CI = 8.15 to 18.05, P < 0.001)], inflammatory factor levels [TNF-α: SMD = 4.23 (95% CI = 2.48 to 5.98, P < 0.001)], oxidative stress indicators [Superoxide production: SMD = 4.53 (95% CI = 2.39 to 6.67, P < 0.001)], and lactate dehydrogenase levels [SMD = 2.82 (95% CI = 1.60 to 4.04, P < 0.001)] were significantly reduced. However, the superoxide dismutase content was significantly increased [SMD = 1.91 (95% CI = 1.17 to 2.65, P < 0.001)]. CONCLUSION APN protects against MIRI via anti-inflammatory, antiapoptotic, and antioxidant effects, and this effect is achieved by activating different signaling pathways.
Collapse
Affiliation(s)
- Hongyi Yue
- Engineering Research Center of Tibetan Medicine Detection Technology, Ministry of Education, School of Medicine, Xizang Minzu University, Xianyang, 712082, Shaanxi, China
| | - Qunhui Zhang
- The First Affiliated Hospital, Department of Cardiology, Hengyang Medical School, University of South China, Hunan, 421001, China
- Hunan Provincial Key Laboratory of Multi-omics And Artificial Intelligence of Cardiovascular Diseases, University of South China, Hunan, 421001, China
| | - Senhao Chang
- Engineering Research Center of Tibetan Medicine Detection Technology, Ministry of Education, School of Medicine, Xizang Minzu University, Xianyang, 712082, Shaanxi, China
| | - Xinjie Zhao
- Engineering Research Center of Tibetan Medicine Detection Technology, Ministry of Education, School of Medicine, Xizang Minzu University, Xianyang, 712082, Shaanxi, China
| | - Mengjie Wang
- Engineering Research Center of Tibetan Medicine Detection Technology, Ministry of Education, School of Medicine, Xizang Minzu University, Xianyang, 712082, Shaanxi, China
| | - Wenhua Li
- Engineering Research Center of Tibetan Medicine Detection Technology, Ministry of Education, School of Medicine, Xizang Minzu University, Xianyang, 712082, Shaanxi, China.
| |
Collapse
|
6
|
Meng Z, Liang B, Wu Y, Liu C, Wang H, Du Y, Gan L, Gao E, Lau WB, Christopher TA, Lopez BL, Koch WJ, Ma X, Zhao F, Wang Y, Zhao J. Hypoadiponectinemia-induced upregulation of microRNA449b downregulating Nrf-1 aggravates cardiac ischemia-reperfusion injury in diabetic mice. J Mol Cell Cardiol 2023; 182:1-14. [PMID: 37437402 PMCID: PMC10566306 DOI: 10.1016/j.yjmcc.2023.06.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 06/23/2023] [Accepted: 06/25/2023] [Indexed: 07/14/2023]
Abstract
Diabetes enhances myocardial ischemic/reperfusion (MI/R) injury via an incompletely understood mechanism. Adiponectin (APN) is a cardioprotective adipokine suppressed by diabetes. However, how hypoadiponectinemia exacerbates cardiac injury remains incompletely understood. Dysregulation of miRNAs plays a significant role in disease development. However, whether hypoadiponectinemia alters cardiac miRNA profile, contributing to diabetic heart injury, remains unclear. Methods and Results: Wild-type (WT) and APN knockout (APN-KO) mice were subjected to MI/R. A cardiac microRNA profile was determined. Among 23 miRNAs increased in APN-KO mice following MI/R, miR-449b was most significantly upregulated (3.98-fold over WT mice). Administrating miR-449b mimic increased apoptosis, enlarged infarct size, and impaired cardiac function in WT mice. In contrast, anti-miR-449b decreased apoptosis, reduced infarct size, and improved cardiac function in APN-KO mice. Bioinformatic analysis predicted 73 miR-449b targeting genes, and GO analysis revealed oxidative stress as the top pathway regulated by these genes. Venn analysis followed by luciferase assay identified Nrf-1 and Ucp3 as the two most important miR-449b targets. In vivo administration of anti-miR-449b in APN-KO mice attenuated MI/R-stimulated superoxide overproduction. In vitro experiments demonstrated that high glucose/high lipid and simulated ischemia/reperfusion upregulated miR-449b and inhibited Nrf-1 and Ucp3 expression. These pathological effects were attenuated by anti-miR-449b or Nrf-1 overexpression. In a final attempt to validate our finding in a clinically relevant model, high-fat diet (HFD)-induced diabetic mice were subjected to MI/R and treated with anti-miR-449b or APN. Diabetes significantly increased miR-449b expression and downregulated Nrf-1 and Ucp3 expression. Administration of anti-miR-449b or APN preserved cardiac Nrf-1 expression, reduced cardiac oxidative stress, decreased apoptosis and infarct size, and improved cardiac function. Conclusion: We demonstrated for the first time that hypoadiponectinemia upregulates miR-449b and suppresses Nrf-1/Ucp3 expression, promoting oxidative stress and exacerbating MI/R injury in this population. Dysregulated APN/miR-449b/oxidative stress pathway is a potential therapeutic target against diabetic MI/R injury.
Collapse
Affiliation(s)
- Zhijun Meng
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107, United States of America
| | - Bin Liang
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107, United States of America
| | - Yalin Wu
- Department of Biomedical Engineering, University of Alabama at Birmingham, AL 35294, United States of America
| | - Caihong Liu
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107, United States of America
| | - Han Wang
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107, United States of America
| | - Yunhui Du
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107, United States of America
| | - Lu Gan
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107, United States of America
| | - Erhe Gao
- Center of Translational Medicine, Temple University School of Medicine, Philadelphia, PA 19140, United States of America
| | - Wayne B Lau
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107, United States of America
| | - Theodore A Christopher
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107, United States of America
| | - Bernard L Lopez
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107, United States of America
| | - Walter J Koch
- Center of Translational Medicine, Temple University School of Medicine, Philadelphia, PA 19140, United States of America
| | - Xinliang Ma
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107, United States of America
| | - Fujie Zhao
- Department of Biomedical Engineering, University of Alabama at Birmingham, AL 35294, United States of America
| | - Yajing Wang
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107, United States of America; Department of Biomedical Engineering, University of Alabama at Birmingham, AL 35294, United States of America.
| | - Jianli Zhao
- Department of Biomedical Engineering, University of Alabama at Birmingham, AL 35294, United States of America.
| |
Collapse
|
7
|
Shawky NM, Soliman E, Abdel-Rahman AA, Rezq S. Editorial: Cardiorenal dysregulation in endocrine disorders: innovative mechanisms and therapeutic interventions. Front Pharmacol 2023; 14:1275919. [PMID: 37693912 PMCID: PMC10486096 DOI: 10.3389/fphar.2023.1275919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 08/21/2023] [Indexed: 09/12/2023] Open
Affiliation(s)
- Noha M. Shawky
- Department of Pharmacology and Toxicology, Mississippi Center of Excellence in Perinatal Research, Women’s Health Research Center, Cardiovascular Renal Research Center, University of Mississippi Medical Center, Jackson, MS, United States
| | - Eman Soliman
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, Blacksburg, VA, United States
| | - Abdel A. Abdel-Rahman
- Department of Pharmacology and Toxicology, Brody School of Medicine, East Carolina University, Greenville, NC, United States
| | - Samar Rezq
- Department of Pharmacology and Toxicology, Mississippi Center of Excellence in Perinatal Research, Women’s Health Research Center, Cardiovascular Renal Research Center, University of Mississippi Medical Center, Jackson, MS, United States
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| |
Collapse
|
8
|
Ren C, Yi W, Jiang B, Gao E, Liang J, Zhang B, Yang Z, Zheng D, Zhang Y. Diminished AdipoR1/APPL1 Interaction Mediates Reduced Cardioprotective Actions of Adiponectin against Myocardial Ischemia/Reperfusion Injury in Type-2 Diabetic Mice. Stem Cells Int 2023; 2023:7441367. [PMID: 36860545 PMCID: PMC9970717 DOI: 10.1155/2023/7441367] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 09/19/2022] [Accepted: 09/26/2022] [Indexed: 02/22/2023] Open
Abstract
Background Obesity-related diseases have important implications for the occurrence, severity, and outcome of ischemic heart disease. Patients with obesity, hyperlipidemia, and diabetes mellitus (metabolic syndrome) are at increased risk of heart attack with decreased plasma lipocalin levels, and lipocalin is negatively correlated with heart attack incidence. APPL1 is a signaling protein with multiple functional structural domains and plays an important role in the APN signaling pathway. There are two known subtypes of lipocalin membrane receptors, AdipoR1 and AdipoR2. AdioR1 is mainly distributed in skeletal muscle while AdipoR2 is mainly distributed in the liver. Objective To clarify whether the AdipoR1-APPL1 signaling pathway mediates the effect of lipocalin in reducing myocardial ischemia/reperfusion injury and its mechanism will provide us with a new approach to treat myocardial ischemia/reperfusion injury using lipocalin as an intervention and therapeutic target. Methods (1) Induction of hypoxia/reoxygenation in SD mammary rat cardiomyocytes to simulate myocardial ischemia/reperfusion; (2) downregulation of APPL1 expression in cardiomyocytes to observe the effect of lipocalin on myocardial ischemia/reperfusion and its mechanism of action. Results (1) Primary mammary rat cardiomyocytes were isolated and cultured and induced to simulate MI/R by hypoxia/reoxygenation; (2) lipocalin inhibited H/R-induced apoptosis in cardiomyocytes; and (3) APN attenuated MI/R injury through AdipoR1-APPL1 and the possible mechanism. Conclusion This study demonstrates for the first time that lipocalin can attenuate myocardial ischemia/reperfusion injury through the AdipoR1-APPL1 signaling pathway and that the reduction of AdipoR1/APPL1 interaction plays an important role in cardiac APN resistance to MI/R injury in diabetic mice.
Collapse
Affiliation(s)
- Chao Ren
- Department of Cardiac Surgery, No. 960 Hospital of PLA, Jinan City, Shandong Province, China
| | - Wei Yi
- Department of Cardiovascular surgery, Xijing Hospital, Air Force Military Medical University, Shaanxi, China
| | - Bo Jiang
- Department of Cardiac surgery, General Hospital of Ningxia Medical University, Ningxia Medical University, Ningxia, China
| | - Erhe Gao
- Center for Translational Medicine, Temple University, Pennsylvania, USA
| | - Jiali Liang
- Department of Cardiac Surgery, No. 960 Hospital of PLA, Jinan City, Shandong Province, China
| | - Bo Zhang
- Department of Cardiac Surgery, No. 960 Hospital of PLA, Jinan City, Shandong Province, China
| | - Zhe Yang
- Department of Cardiac Surgery, No. 960 Hospital of PLA, Jinan City, Shandong Province, China
| | - Dezhi Zheng
- Department of Cardiac Surgery, No. 960 Hospital of PLA, Jinan City, Shandong Province, China
| | - Yong Zhang
- Department of Cardiac Surgery, No. 960 Hospital of PLA, Jinan City, Shandong Province, China
| |
Collapse
|
9
|
Martínez-Huenchullán SF, Fox SL, Tam CS, Maharjan BR, Olaya-Agudo LF, Ehrenfeld P, Williams PF, Mclennan SV, Twigg SM. Constant-moderate versus high-intensity interval training on heart adiponectin levels in high-fat fed mice: a preventive and treatment approach. Arch Physiol Biochem 2023; 129:41-45. [PMID: 32715774 DOI: 10.1080/13813455.2020.1797098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
CONTEXT Research has described that adiponectin plays a key role in cardiomyocytes metabolism, however, the effects of exercise during obesity on cardiac adiponectin levels is unclear. OBJECTIVE To investigate the effects of constant-moderate endurance (END) and high-intensity interval training (HIIT), on heart adiponectin levels in mice. MATERIAL AND METHODS Two experiments were conducted: (1) preventive (EX1): 10 week-old male mice were fed standard (CHOW) or high-fat diet (HFD;45% fat) and simultaneously trained with END and HIIT for 10 weeks; (2) Treatment (EX2): after 10 weeks of dietary intervention, another cohort of 10 week-old mice were trained by both programmes for 10 weeks. RESULTS In EX1, END and HIIT decreased low-molecular weight adiponectin (∼0.5-fold; p < 0.05) and increased GLUT4 levels (∼2-fold; p < .05). In EX2, HFD significantly decreased high-molecular weight adiponectin (∼0.7-fold; p < .05), and END reversed this change.Discussion and conclusion: HFD and exercise influence heart adiponectin isoforms and therefore might impact cardiomyocyte metabolism.
Collapse
Affiliation(s)
- Sergio F Martínez-Huenchullán
- Greg Brown Diabetes & Endocrinology Laboratory, Charles Perkins Centre, Faculty of Medicine and Health, University of Sydney, Sydney, Australia
- School of Physical Therapy, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile
| | - Sarah L Fox
- Greg Brown Diabetes & Endocrinology Laboratory, Charles Perkins Centre, Faculty of Medicine and Health, University of Sydney, Sydney, Australia
| | - Charmaine S Tam
- Northern Clinical School and Centre for Translational Data Science, University of Sydney, Sydney, Australia
| | - Babu Raja Maharjan
- Department of Biochemistry, School of Medicine, Patan Academy of Health Sciences, Lalitpur, Nepal
| | - Luisa F Olaya-Agudo
- Greg Brown Diabetes & Endocrinology Laboratory, Charles Perkins Centre, Faculty of Medicine and Health, University of Sydney, Sydney, Australia
| | - Pamela Ehrenfeld
- Laboratory of Cellular Pathology. Institute of Anatomy, Histology & Pathology, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile
- Centro Interdisciplinario de Estudios del Sistema Nervioso (CISNe), Valdivia, Chile
| | - Paul F Williams
- Greg Brown Diabetes & Endocrinology Laboratory, Charles Perkins Centre, Faculty of Medicine and Health, University of Sydney, Sydney, Australia
| | - Susan V Mclennan
- Greg Brown Diabetes & Endocrinology Laboratory, Charles Perkins Centre, Faculty of Medicine and Health, University of Sydney, Sydney, Australia
- New South Wales Health Pathology, New South Wales, Australia
- Department of Endocrinology, Royal Prince Alfred Hospital, Sydney, Australia
| | - Stephen M Twigg
- Greg Brown Diabetes & Endocrinology Laboratory, Charles Perkins Centre, Faculty of Medicine and Health, University of Sydney, Sydney, Australia
- Department of Endocrinology, Royal Prince Alfred Hospital, Sydney, Australia
| |
Collapse
|
10
|
Xia W, Li X, Wu Q, Xu A, Zhang L, Xia Z. The importance of caveolin as a target in the prevention and treatment of diabetic cardiomyopathy. Front Immunol 2022; 13:951381. [PMID: 36405687 PMCID: PMC9666770 DOI: 10.3389/fimmu.2022.951381] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 10/21/2022] [Indexed: 08/30/2023] Open
Abstract
The diabetic population has been increasing in the past decades and diabetic cardiomyopathy (DCM), a pathology that is defined by the presence of cardiac remodeling and dysfunction without conventional cardiac risk factors such as hypertension and coronary heart diseases, would eventually lead to fatal heart failure in the absence of effective treatment. Impaired insulin signaling, commonly known as insulin resistance, plays an important role in the development of DCM. A family of integral membrane proteins named caveolins (mainly caveolin-1 and caveolin-3 in the myocardium) and a protein hormone adiponectin (APN) have all been shown to be important for maintaining normal insulin signaling. Abnormalities in caveolins and APN have respectively been demonstrated to cause DCM. This review aims to summarize recent research findings of the roles and mechanisms of caveolins and APN in the development of DCM, and also explore the possible interplay between caveolins and APN.
Collapse
Affiliation(s)
- Weiyi Xia
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Guangdong, China
- Department of Orthopaedics and Traumatology, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Xia Li
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qingping Wu
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Liangqing Zhang
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Guangdong, China
| | - Zhengyuan Xia
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Guangdong, China
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| |
Collapse
|
11
|
Weber BY, Brenner GB, Kiss B, Gergely TG, Sayour NV, Tian H, Makkos A, Görbe A, Ferdinandy P, Giricz Z. Rosiglitazone Does Not Show Major Hidden Cardiotoxicity in Models of Ischemia/Reperfusion but Abolishes Ischemic Preconditioning-Induced Antiarrhythmic Effects in Rats In Vivo. Pharmaceuticals (Basel) 2022; 15:ph15091055. [PMID: 36145276 PMCID: PMC9503202 DOI: 10.3390/ph15091055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 08/19/2022] [Accepted: 08/22/2022] [Indexed: 11/16/2022] Open
Abstract
Clinical observations are highly inconsistent with the use of the antidiabetic rosiglitazone regarding its associated increased risk of myocardial infarction. This may be due to its hidden cardiotoxic properties that have only become evident during post-marketing studies. Therefore, we aimed to investigate the hidden cardiotoxicity of rosiglitazone in ischemia/reperfusion (I/R) injury models. Rats were treated orally with either 0.8 mg/kg/day rosiglitazone or vehicle for 28 days and subjected to I/R with or without cardioprotective ischemic preconditioning (IPC). Rosiglitazone did not affect mortality, arrhythmia score, or infarct size during I/R. However, rosiglitazone abolished the antiarrhythmic effects of IPC. To investigate the direct effect of rosiglitazone on cardiomyocytes, we utilized adult rat cardiomyocytes (ARCMs), AC16, and differentiated AC16 (diffAC16) human cardiac cell lines. These were subjected to simulated I/R in the presence of rosiglitazone. Rosiglitazone improved cell survival of ARCMs at 0.3 μM. At 0.1 and 0.3 μM, rosiglitazone improved cell survival of AC16s but not that of diffAC16s. This is the first demonstration that chronic administration of rosiglitazone does not result in major hidden cardiotoxic effects in myocardial I/R injury models. However, the inhibition of the antiarrhythmic effects of IPC may have some clinical relevance that needs to be further explored.
Collapse
Affiliation(s)
- Bennet Y. Weber
- MTA-SE System Pharmacology Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1089 Budapest, Hungary
| | - Gábor B. Brenner
- MTA-SE System Pharmacology Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1089 Budapest, Hungary
| | - Bernadett Kiss
- MTA-SE System Pharmacology Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1089 Budapest, Hungary
| | - Tamás G. Gergely
- MTA-SE System Pharmacology Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1089 Budapest, Hungary
| | - Nabil V. Sayour
- MTA-SE System Pharmacology Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1089 Budapest, Hungary
| | - Huimin Tian
- MTA-SE System Pharmacology Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1089 Budapest, Hungary
| | - András Makkos
- MTA-SE System Pharmacology Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1089 Budapest, Hungary
| | - Anikó Görbe
- MTA-SE System Pharmacology Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1089 Budapest, Hungary
- Pharmahungary Group, H-6722 Szeged, Hungary
| | - Péter Ferdinandy
- MTA-SE System Pharmacology Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1089 Budapest, Hungary
- Pharmahungary Group, H-6722 Szeged, Hungary
| | - Zoltán Giricz
- MTA-SE System Pharmacology Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1089 Budapest, Hungary
- Pharmahungary Group, H-6722 Szeged, Hungary
- Correspondence:
| |
Collapse
|
12
|
Zhu D, Zhang Z, Zhao J, Liu D, Gan L, Lau WB, Xie D, Meng Z, Yao P, Tsukuda J, Christopher TA, Lopez BL, Gao E, Koch WJ, Wang Y, Ma XL. Targeting Adiponectin Receptor 1 Phosphorylation Against Ischemic Heart Failure. Circ Res 2022; 131:e34-e50. [PMID: 35611695 PMCID: PMC9308652 DOI: 10.1161/circresaha.121.319976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
BACKGROUND Despite significantly reduced acute myocardial infarction (MI) mortality in recent years, ischemic heart failure continues to escalate. Therapeutic interventions effectively reversing pathological remodeling are an urgent unmet medical need. We recently demonstrated that AdipoR1 (APN [adiponectin] receptor 1) phosphorylation by GRK2 (G-protein-coupled receptor kinase 2) contributes to maladaptive remodeling in the ischemic heart. The current study clarified the underlying mechanisms leading to AdipoR1 phosphorylative desensitization and investigated whether blocking AdipoR1 phosphorylation may restore its protective signaling, reversing post-MI remodeling. METHODS Specific sites and underlying molecular mechanisms responsible for AdipoR1 phosphorylative desensitization were investigated in vitro (neonatal and adult cardiomyocytes). The effects of AdipoR1 phosphorylation inhibition upon APN post-MI remodeling and heart failure progression were investigated in vivo. RESULTS Among 4 previously identified sites sensitive to GRK2 phosphorylation, alanine substitution of Ser205 (AdipoR1S205A), but not other 3 sites, rescued GRK2-suppressed AdipoR1 functions, restoring APN-induced cell salvage kinase activation and reducing oxidative cell death. The molecular investigation followed by functional determination demonstrated that AdipoR1 phosphorylation promoted clathrin-dependent (not caveolae) endocytosis and lysosomal-mediated (not proteasome) degradation, reducing AdipoR1 protein level and suppressing AdipoR1-mediated cytoprotective action. GRK2-induced AdipoR1 endocytosis and degradation were blocked by AdipoR1S205A overexpression. Moreover, AdipoR1S205E (pseudophosphorylation) phenocopied GRK2 effects, promoted AdipoR1 endocytosis and degradation, and inhibited AdipoR1 biological function. Most importantly, AdipoR1 function was preserved during heart failure development in AdipoR1-KO (AdipoR1 knockout) mice reexpressing hAdipoR1S205A. APN administration in the failing heart reversed post-MI remodeling and improved cardiac function. However, reexpressing hAdipoR1WT in AdipoR1-KO mice failed to restore APN cardioprotection. CONCLUSIONS Ser205 is responsible for AdipoR1 phosphorylative desensitization in the failing heart. Blockade of AdipoR1 phosphorylation followed by pharmacological APN administration is a novel therapy effective in reversing post-MI remodeling and mitigating heart failure progression.
Collapse
Affiliation(s)
- Di Zhu
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107
| | - Zhen Zhang
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107
| | - Jianli Zhao
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107
| | - Demin Liu
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107
| | - Lu Gan
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107
| | - Wayne Bond Lau
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107
| | - Dina Xie
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107
| | - Zhijun Meng
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107
| | - Peng Yao
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107
| | - Jumpei Tsukuda
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107
| | | | - Bernard L. Lopez
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107
| | - Erhe Gao
- Department of Cardiovascular Sciences, Center for Translational Medicine, Temple University, Philadelphia, PA 19104
| | - Walter J. Koch
- Department of Cardiovascular Sciences, Center for Translational Medicine, Temple University, Philadelphia, PA 19104
| | - Yajing Wang
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107
- Corresponding Authors: Xinliang (Xin) Ma, M.D., Ph.D, Department of Medicine and, Department of Emergency Medicine, 1025 Walnut Street, College Building 300, Thomas Jefferson University, Philadelphia, PA 19107, Tel: 215-955-4994, Or Yajing Wang, MD,PhD, Department of Emergency Medicine, 1025 Walnut Street, College Building 325, Thomas Jefferson University, Philadelphia, PA 19107, Tel: 215-955-8895,
| | - Xin-Liang Ma
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19107
- Corresponding Authors: Xinliang (Xin) Ma, M.D., Ph.D, Department of Medicine and, Department of Emergency Medicine, 1025 Walnut Street, College Building 300, Thomas Jefferson University, Philadelphia, PA 19107, Tel: 215-955-4994, Or Yajing Wang, MD,PhD, Department of Emergency Medicine, 1025 Walnut Street, College Building 325, Thomas Jefferson University, Philadelphia, PA 19107, Tel: 215-955-8895,
| |
Collapse
|
13
|
Sevoflurane preconditioning protects against acute MI/R injury via enhancing AdipoR1-Cav3 interaction and alleviating endoplasmic reticulum stress. Exp Cell Res 2022; 417:113217. [DOI: 10.1016/j.yexcr.2022.113217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/17/2022] [Accepted: 05/17/2022] [Indexed: 11/23/2022]
|
14
|
Liu J, Li X, Wang X, Peng L, Song G, He J. Angiotensin(1-7) Improves Islet Function in Diabetes Through Reducing JNK/Caspase-3 Signaling. Horm Metab Res 2022; 54:250-258. [PMID: 35413746 DOI: 10.1055/a-1796-9286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
The aim of this study is to investigate whether Angiotensin (1-7), the physiological antagonist of Angiotensin II (AngII), has antidiabetic activity and the possible mechanism. Male Wistar rats were randomly divided into 3 groups: control group fed the normal diet, DM group fed high-fat diet and injected with STZ, and Angiotensin (1-7) group receiving injection of STZ followed by Angiotensin (1-7) treatment. Serum Ang II, fasting blood glucose, insulin, HOMA-IR, and HOMA-beta were determined in control, diabetes and Angiotensin (1-7) groups. The increased AngII and insulin resistance in diabetes group were accompanied by changes in islet histopathology. However, Angiotensin (1-7) improved the islet function and histopathology in diabetes without affecting the level of AngII. Western blot confirmed that Angiotensin (1-7) decreased the cleaved caspase 3 levels in pancreas of DM. The increased expression of JNK, Bax, and Bcl2 genes under diabetic conditions were partially reversed after Angiotensin (1-7) administration in pancreas. Immunofluorescence analysis showed that p-JNK was markedly increased in islet of DM rats, which was markedly alleviated after Angiotensin (1-7) treatment. Furthermore, Angiotensin (1-7) reversed high glucose(HG) induced mitochondrial apoptosis augments. Finally, Angiotensin (1-7) attenuated the apoptosis of INS-1 cells through reducing JNK activation in diabetes, which was blocked by anisomycin (a potent agonist of JNK). Our findings provide supporting evidence that Angiotensin (1-7) improved the islet beta-cells apoptosis by JNK-mediated mitochondrial dysfunction, which might be a novel target for the treatment and prevention of beta-cells dysfunction in DM.
Collapse
Affiliation(s)
- Jing Liu
- Department of Endocrinology, Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Xing Li
- Department of Endocrinology, Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Xiaoyan Wang
- Department of Endocrinology, Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Lina Peng
- Department of Endocrinology, Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Guoning Song
- Department of Endocrinology, Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Junhua He
- Department of Endocrinology, Second Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
15
|
Zarkasi KA, Abdul Murad NA, Ahmad N, Jamal R, Abdullah N. Coronary Heart Disease in Type 2 Diabetes Mellitus: Genetic Factors and Their Mechanisms, Gene-Gene, and Gene-Environment Interactions in the Asian Populations. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:647. [PMID: 35055468 PMCID: PMC8775550 DOI: 10.3390/ijerph19020647] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 01/03/2022] [Accepted: 01/04/2022] [Indexed: 02/04/2023]
Abstract
Asians are more susceptible to type 2 diabetes mellitus (T2D) and its coronary heart disease (CHD) complications than the Western populations, possibly due to genetic factors, higher degrees of obesity, insulin resistance, and endothelial dysfunction that could occur even in healthy individuals. The genetic factors and their mechanisms, along with gene-gene and gene-environment interactions associated with CHD in T2D Asians, are yet to be explored. Therefore, the objectives of this paper were to review the current evidence of genetic factors for CHD, summarize the proposed mechanisms of these genes and how they may associate with CHD risk, and review the gene-gene and gene-environment interactions in T2D Asians with CHD. The genetic factors can be grouped according to their involvement in the energy and lipoprotein metabolism, vascular and endothelial pathology, antioxidation, cell cycle regulation, DNA damage repair, hormonal regulation of glucose metabolism, as well as cytoskeletal function and intracellular transport. Meanwhile, interactions between single nucleotide polymorphisms (SNPs) from different genes, SNPs within a single gene, and genetic interaction with environmental factors including obesity, smoking habit, and hyperlipidemia could modify the gene's effect on the disease risk. Collectively, these factors illustrate the complexities of CHD in T2D, specifically among Asians.
Collapse
Affiliation(s)
- Khairul Anwar Zarkasi
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia (UKM), Kuala Lumpur 56000, Malaysia; (K.A.Z.); (N.A.A.M.); (R.J.)
- Biochemistry Unit, Preclinical Department, Faculty of Medicine and Defence Health, Universiti Pertahanan Nasional Malaysia, Kuala Lumpur 57000, Malaysia
| | - Nor Azian Abdul Murad
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia (UKM), Kuala Lumpur 56000, Malaysia; (K.A.Z.); (N.A.A.M.); (R.J.)
| | - Norfazilah Ahmad
- Epidemiology and Statistics Unit, Department of Community Health, Faculty of Medicine, Universiti Kebangsaan Malaysia (UKM), Kuala Lumpur 56000, Malaysia;
| | - Rahman Jamal
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia (UKM), Kuala Lumpur 56000, Malaysia; (K.A.Z.); (N.A.A.M.); (R.J.)
| | - Noraidatulakma Abdullah
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia (UKM), Kuala Lumpur 56000, Malaysia; (K.A.Z.); (N.A.A.M.); (R.J.)
- Faculty of Health Sciences, Universiti Kebangsaan Malaysia (UKM), Kuala Lumpur 50300, Malaysia
| |
Collapse
|
16
|
Taha HSED, Kandil H, Farag N, Oraby A, Sharkawy ME, Fawzy F, Mahrous H, Bahgat J, Samy M, Aboul M, Abdrabou M, Shaker MM. Egyptian practical guidance in hypertriglyceridemia management 2021. Egypt Heart J 2021; 73:107. [PMID: 34928475 PMCID: PMC8688602 DOI: 10.1186/s43044-021-00235-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 12/14/2021] [Indexed: 11/10/2022] Open
Abstract
Hypertriglyceridemia (HTG) is a very common, yet underappreciated problem in clinical practice. Elevated triglyceride (TG) levels are independently associated with atherosclerotic cardiovascular disease (ASCVD) risk. Furthermore, severe HTG may lead to acute pancreatitis. Although LDL-guided statin therapy has improved ASCVD outcomes, residual risk remains. Recent trials have demonstrated that management of high TG levels, in patients already on statin therapy, reduces the rate of major vascular events. Few guidelines were issued, providing important recommendations for HTG management strategies. The goal of treatment is to reduce the risk of ASCVD and acute pancreatitis. The management stands on lifestyle modification, detection of secondary causes of HTG and pharmacological therapy, when indicated. In this guidance we review the causes and classification of HTG and summarize the current methods for risk estimation, diagnosis and treatment. The present guidance provides a focused update on the management of HTG, outlined in a simple user-friendly format, with an emphasis on the latest available data.
Collapse
Affiliation(s)
- Hesham Salah El Din Taha
- Department of Cardiology, Faculty of Medicine, Cairo University, 27 Nafezet Sheem El Shafae St Kasr Al Ainy, Cairo, 11562 Egypt
| | - Hossam Kandil
- Department of Cardiology, Faculty of Medicine, Cairo University, 27 Nafezet Sheem El Shafae St Kasr Al Ainy, Cairo, 11562 Egypt
| | | | | | | | - Fouad Fawzy
- Department of Cardiology, Faculty of Medicine, Cairo University, 27 Nafezet Sheem El Shafae St Kasr Al Ainy, Cairo, 11562 Egypt
| | - Hossam Mahrous
- Department of Cardiology, Faculty of Medicine, Cairo University, 27 Nafezet Sheem El Shafae St Kasr Al Ainy, Cairo, 11562 Egypt
| | - Juliette Bahgat
- Department of Cardiology, Faculty of Medicine, Cairo University, 27 Nafezet Sheem El Shafae St Kasr Al Ainy, Cairo, 11562 Egypt
| | - Mina Samy
- Department of Cardiology, Faculty of Medicine, Cairo University, 27 Nafezet Sheem El Shafae St Kasr Al Ainy, Cairo, 11562 Egypt
| | - Mohamed Aboul
- Department of Cardiology, Faculty of Medicine, Cairo University, 27 Nafezet Sheem El Shafae St Kasr Al Ainy, Cairo, 11562 Egypt
| | - Mostafa Abdrabou
- Department of Cardiology, Faculty of Medicine, Cairo University, 27 Nafezet Sheem El Shafae St Kasr Al Ainy, Cairo, 11562 Egypt
| | - Mirna Mamdouh Shaker
- Department of Cardiology, Faculty of Medicine, Cairo University, 27 Nafezet Sheem El Shafae St Kasr Al Ainy, Cairo, 11562 Egypt
| |
Collapse
|
17
|
de Alencar AKN, Wang H, de Oliveira GMM, Sun X, Zapata-Sudo G, Groban L. Crossroads between Estrogen Loss, Obesity, and Heart Failure with Preserved Ejection Fraction. Arq Bras Cardiol 2021; 117:1191-1201. [PMID: 34644788 PMCID: PMC8757160 DOI: 10.36660/abc.20200855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 12/16/2020] [Accepted: 01/27/2021] [Indexed: 11/24/2022] Open
Abstract
The prevalence of obesity and heart failure with preserved ejection fraction (HFpEF) increases significantly in postmenopausal women. Although obesity is a risk factor for left ventricular diastolic dysfunction (LVDD), the mechanisms that link the cessation of ovarian hormone production, and particularly estrogens, to the development of obesity, LVDD, and HFpEF in aging females are unclear. Clinical, and epidemiologic studies show that postmenopausal women with abdominal obesity (defined by waist circumference) are at greater risk for developing HFpEF than men or women without abdominal obesity. The study presents a review of clinical data that support a mechanistic link between estrogen loss plus obesity and left ventricular remodeling with LVDD. It also seeks to discuss potential cell and molecular mechanisms for estrogen-mediated protection against adverse adipocyte cell types, tissue depots, function, and metabolism that may contribute to LVDD and HFpEF.
Collapse
Affiliation(s)
| | - Hao Wang
- Wake Forest School of MedicineDepartments of AnesthesiologyWinston-SalemNorth CarolinaEstados Unidos da AméricaWake Forest School of Medicine - Departments of Anesthesiology, Winston-Salem, North Carolina - Estados Unidos da América
- Wake Forest School of MedicineWinston-SalemNorth CarolinaEstados Unidos da AméricaWake Forest School of Medicine - Internal Medicine-Section of Molecular Medicine, Winston-Salem, North Carolina - Estados Unidos da América
| | - Gláucia Maria Moraes de Oliveira
- Universidade Federal do Rio de JaneiroDepartamento de Clínica MédicaFaculdade de MedicinaRio de JaneiroRJBrasilUniversidade Federal do Rio de Janeiro - Departamento de Clínica Médica, Faculdade de Medicina, Rio de Janeiro, RJ - Brasil
| | - Xuming Sun
- Wake Forest School of MedicineDepartments of AnesthesiologyWinston-SalemNorth CarolinaEstados Unidos da AméricaWake Forest School of Medicine - Departments of Anesthesiology, Winston-Salem, North Carolina - Estados Unidos da América
| | - Gisele Zapata-Sudo
- Universidade Federal do Rio de JaneiroInstituto de Ciências BiomédicasRio de JaneiroRJBrasilUniversidade Federal do Rio de Janeiro - Instituto de Ciências Biomédicas, Rio de Janeiro, RJ - Brasil
- Universidade Federal do Rio de JaneiroInstituto de Cardiologia Edson SaadFaculdade de MedicinaRio de JaneiroRJBrasilUniversidade Federal do Rio de Janeiro - Instituto de Cardiologia Edson Saad, Faculdade de Medicina, Rio de Janeiro, RJ - Brasil
| | - Leanne Groban
- Wake Forest School of MedicineDepartments of AnesthesiologyWinston-SalemNorth CarolinaEstados Unidos da AméricaWake Forest School of Medicine - Departments of Anesthesiology, Winston-Salem, North Carolina - Estados Unidos da América
- Wake Forest School of MedicineWinston-SalemNorth CarolinaEstados Unidos da AméricaWake Forest School of Medicine - Internal Medicine-Section of Molecular Medicine, Winston-Salem, North Carolina - Estados Unidos da América
| |
Collapse
|
18
|
Meng Z, Liang H, Zhao J, Gao J, Liu C, Ma X, Liu J, Liang B, Jiao X, Cao J, Wang Y. HMOX1 upregulation promotes ferroptosis in diabetic atherosclerosis. Life Sci 2021; 284:119935. [PMID: 34508760 DOI: 10.1016/j.lfs.2021.119935] [Citation(s) in RCA: 110] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 08/27/2021] [Accepted: 08/30/2021] [Indexed: 12/26/2022]
Abstract
OBJECTIVE Atherosclerotic vascular disease remains the principal cause of death and disability among patients with type 2 diabetes. Unfortunately, the problem is not adequately resolved by therapeutic strategies with currently available drugs or approaches that solely focus on optimal glycemic control. To identify the key contributors and better understand the mechanism of diabetic atherosclerotic vascular disease, we aimed to elucidate the key genetic characteristics and pathological pathways in atherosclerotic vascular disease through nonbiased bioinformatics analysis and subsequent experimental demonstration and exploration in diabetic atherosclerotic vascular disease. METHODS AND RESULTS Sixty-eight upregulated and 23 downregulated genes were identified from the analysis of gene expression profiles (GSE30169 and GSE6584). A comprehensive bioinformatic assay further identified that ferroptosis, a new type of programmed cell death and HMOX1 (a gene that encodes heme oxygenase), were vital factors in atherosclerotic vascular disease. We further demonstrated that diabetes significantly increased ferroptosis and HMOX1 levels compared to normal controls. Importantly, the ferroptosis inhibitor ferrostatin-1 (Fer-1) effectively attenuated diabetic atherosclerosis, suggesting the causative role of ferroptosis in diabetic atherosclerosis development. At the cellular level, Fer-1 ameliorated high glucose high lipid-induced lipid peroxidation and downregulated ROS production. More importantly, HMOX1 knockdown attenuated Fe2+ overload, reduced iron content and ROS, and alleviated lipid peroxidation, which led to a reduction in ferroptosis in diabetic human endothelial cells. CONCLUSIONS We demonstrated that HMOX1 upregulation is responsible for the increased ferroptosis in diabetic atherosclerosis development, suggesting that HMOX1 may serve as a potential therapeutic or drug development target for diabetic atherosclerosis.
Collapse
Affiliation(s)
- Zhijun Meng
- Department of Physiology, Key Laboratory of Cellular Physiology, Shanxi Medical University, Taiyuan, Shanxi, China; Clinical Laboratory, Shanxi Provincial People's Hospital of Shanxi Medical University, Taiyuan, Shanxi, China; Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA, United States of America
| | - Hongping Liang
- Clinical Laboratory, Shanxi Provincial People's Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Jianli Zhao
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA, United States of America
| | - Jia Gao
- Department of Physiology, Key Laboratory of Cellular Physiology, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Caihong Liu
- Department of Physiology, Key Laboratory of Cellular Physiology, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xinliang Ma
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA, United States of America
| | - Jing Liu
- Department of Physiology, Key Laboratory of Cellular Physiology, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Bin Liang
- Department of Cardiology, The Second Affiliated Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xiangying Jiao
- Department of Physiology, Key Laboratory of Cellular Physiology, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Jimin Cao
- Department of Physiology, Key Laboratory of Cellular Physiology, Shanxi Medical University, Taiyuan, Shanxi, China.
| | - Yajing Wang
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA, United States of America.
| |
Collapse
|
19
|
Sharma A, Mah M, Ritchie RH, De Blasio MJ. The adiponectin signalling pathway - A therapeutic target for the cardiac complications of type 2 diabetes? Pharmacol Ther 2021; 232:108008. [PMID: 34610378 DOI: 10.1016/j.pharmthera.2021.108008] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 08/17/2021] [Accepted: 09/23/2021] [Indexed: 12/11/2022]
Abstract
Diabetes is associated with an increased risk of heart failure (HF). This is commonly termed diabetic cardiomyopathy and is often characterised by increased cardiac fibrosis, pathological hypertrophy, increased oxidative and endoplasmic reticulum stress as well as diastolic dysfunction. Adiponectin is a cardioprotective adipokine that is downregulated in settings of type 2 diabetes (T2D) and obesity. Furthermore, both adiponectin receptors (AdipoR1 and R2) are also downregulated in these settings which further results in impaired cardiac adiponectin signalling and reduced cardioprotection. In many cardiac pathologies, adiponectin signalling has been shown to protect against cardiac remodelling and lipotoxicity, however its cardioprotective actions in T2D-induced cardiomyopathy remain unresolved. Diabetic cardiomyopathy has historically lacked effective treatment options. In this review, we summarise the current evidence for links between the suppressed adiponectin signalling pathway and cardiac dysfunction, in diabetes. We describe adiponectin receptor-mediated signalling pathways that are normally associated with cardioprotection, as well as current and potential future therapeutic approaches that could target this pathway as possible interventions for diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Abhipree Sharma
- Heart Failure Pharmacology, Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Michael Mah
- Heart Failure Pharmacology, Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Rebecca H Ritchie
- Heart Failure Pharmacology, Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia; Department of Pharmacology, Monash University, Clayton, VIC 3800, Australia; Department of Medicine, Monash University, Clayton, VIC 3800, Australia
| | - Miles J De Blasio
- Heart Failure Pharmacology, Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia; Department of Pharmacology, Monash University, Clayton, VIC 3800, Australia.
| |
Collapse
|
20
|
McClung JA, Levy L, Garcia V, Stec DE, Peterson SJ, Abraham NG. Heme-oxygenase and lipid mediators in obesity and associated cardiometabolic diseases: Therapeutic implications. Pharmacol Ther 2021; 231:107975. [PMID: 34499923 DOI: 10.1016/j.pharmthera.2021.107975] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 07/08/2021] [Accepted: 07/27/2021] [Indexed: 02/08/2023]
Abstract
Obesity-mediated metabolic syndrome remains the leading cause of death worldwide. Among many potential targets for pharmacological intervention, a promising strategy involves the heme oxygenase (HO) system, specifically its inducible form, HO-1. This review collects and updates much of the current knowledge relevant to pharmacology and clinical medicine concerning HO-1 in metabolic diseases and its effect on lipid metabolism. HO-1 has pleotropic effects that collectively reduce inflammation, while increasing vasodilation and insulin and leptin sensitivity. Recent reports indicate that HO-1 with its antioxidants via the effect of bilirubin increases formation of biologically active lipid metabolites such as epoxyeicosatrienoic acid (EET), omega-3 and other polyunsaturated fatty acids (PUFAs). Similarly, HO-1and bilirubin are potential therapeutic targets in the treatment of fat-induced liver diseases. HO-1-mediated upregulation of EET is capable not only of reversing endothelial dysfunction and hypertension, but also of reversing cardiac remodeling, a hallmark of the metabolic syndrome. This process involves browning of white fat tissue (i.e. formation of healthy adipocytes) and reduced lipotoxicity, which otherwise will be toxic to the heart. More importantly, this review examines the activity of EET in biological systems and a series of pathways that explain its mechanism of action and discusses how these might be exploited for potential therapeutic use. We also discuss the link between cardiac ectopic fat deposition and cardiac function in humans, which is similar to that described in obese mice and is regulated by HO-1-EET-PGC1α signaling, a potent negative regulator of the inflammatory adipokine NOV.
Collapse
Affiliation(s)
- John A McClung
- Department of Medicine, New York Medical College, Valhalla, NY 10595, United States of America
| | - Lior Levy
- Department of Medicine, New York Medical College, Valhalla, NY 10595, United States of America
| | - Victor Garcia
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595, United States of America
| | - David E Stec
- Department of Physiology and Biophysics, Cardiorenal and Metabolic Diseases Research Center, University of Mississippi Medical Center, Jackson, MS 39216, United States of America.
| | - Stephen J Peterson
- Department of Medicine, Weill Cornell Medicine, New York, NY 10065, United States of America; New York Presbyterian Brooklyn Methodist Hospital, Brooklyn, NY 11215, United States of America
| | - Nader G Abraham
- Department of Medicine, New York Medical College, Valhalla, NY 10595, United States of America; Department of Pharmacology, New York Medical College, Valhalla, NY 10595, United States of America.
| |
Collapse
|
21
|
Nesti L, Tricò D, Mengozzi A, Natali A. Rethinking pioglitazone as a cardioprotective agent: a new perspective on an overlooked drug. Cardiovasc Diabetol 2021; 20:109. [PMID: 34006325 PMCID: PMC8130304 DOI: 10.1186/s12933-021-01294-7] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 04/29/2021] [Indexed: 12/13/2022] Open
Abstract
Since 1985, the thiazolidinedione pioglitazone has been widely used as an insulin sensitizer drug for type 2 diabetes mellitus (T2DM). Although fluid retention was early recognized as a safety concern, data from clinical trials have not provided conclusive evidence for a benefit or a harm on cardiac function, leaving the question unanswered. We reviewed the available evidence encompassing both in vitro and in vivo studies in tissues, isolated organs, animals and humans, including the evidence generated by major clinical trials. Despite the increased risk of hospitalization for heart failure due to fluid retention, pioglitazone is consistently associated with reduced risk of myocardial infarction and ischemic stroke both in primary and secondary prevention, without any proven direct harm on the myocardium. Moreover, it reduces atherosclerosis progression, in-stent restenosis after coronary stent implantation, progression rate from persistent to permanent atrial fibrillation, and reablation rate in diabetic patients with paroxysmal atrial fibrillation after catheter ablation. In fact, human and animal studies consistently report direct beneficial effects on cardiomyocytes electrophysiology, energetic metabolism, ischemia–reperfusion injury, cardiac remodeling, neurohormonal activation, pulmonary circulation and biventricular systo-diastolic functions. The mechanisms involved may rely either on anti-remodeling properties (endothelium protective, inflammation-modulating, anti-proliferative and anti-fibrotic properties) and/or on metabolic (adipose tissue metabolism, increased HDL cholesterol) and neurohormonal (renin–angiotensin–aldosterone system, sympathetic nervous system, and adiponectin) modulation of the cardiovascular system. With appropriate prescription and titration, pioglitazone remains a useful tool in the arsenal of the clinical diabetologist.
Collapse
Affiliation(s)
- Lorenzo Nesti
- Metabolism, Nutrition, and Atherosclerosis Laboratory, Department of Clinical and Experimental Medicine, University of Pisa, Via Savi 10, 56126, Pisa, Italy. .,Cardiopulmonary Laboratory, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy.
| | - Domenico Tricò
- Metabolism, Nutrition, and Atherosclerosis Laboratory, Department of Clinical and Experimental Medicine, University of Pisa, Via Savi 10, 56126, Pisa, Italy.,Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, Pisa, Italy
| | - Alessandro Mengozzi
- Institute of Life Sciences, Sant'Anna School of Advanced Studies, Pisa, Italy
| | - Andrea Natali
- Metabolism, Nutrition, and Atherosclerosis Laboratory, Department of Clinical and Experimental Medicine, University of Pisa, Via Savi 10, 56126, Pisa, Italy.,Cardiopulmonary Laboratory, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| |
Collapse
|
22
|
Kim YS, Cho HH, Cho DI, Jeong HY, Lim SY, Jun JH, Kim MR, Kang BG, Cho M, Kang HJ, Kang WS, Oh GT, Ahn Y. The adipokine Retnla deficiency increases responsiveness to cardiac repair through adiponectin-rich bone marrow cells. Cell Death Dis 2021; 12:307. [PMID: 33753732 PMCID: PMC7985519 DOI: 10.1038/s41419-021-03593-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 01/19/2021] [Indexed: 02/07/2023]
Abstract
Resistin-like alpha (Retnla) is a member of the resistin family and known to modulate fibrosis and inflammation. Here, we investigated the role of Retnla in the cardiac injury model. Myocardial infarction (MI) was induced in wild type (WT), Retnla knockout (KO), and Retnla transgenic (TG) mice. Cardiac function was assessed by echocardiography and was significantly preserved in the KO mice, while worsened in the TG group. Angiogenesis was substantially increased in the KO mice, and cardiomyocyte apoptosis was markedly suppressed in the KO mice. By Retnla treatment, the expression of p21 and the ratio of Bax to Bcl2 were increased in cardiomyocytes, while decreased in cardiac fibroblasts. Interestingly, the numbers of cardiac macrophages and unsorted bone marrow cells (UBCs) were higher in the KO mice than in the WT mice. Besides, phosphorylated histone H3(+) cells were more frequent in bone marrow of KO mice. Moreover, adiponectin in UBCs was notably higher in the KO mice compared with WT mice. In an adoptive transfer study, UBCs were isolated from KO mice to transplant to the WT infarcted heart. Cardiac function was better in the KO-UBCs transplanted group in the WT-UBCs transplanted group. Taken together, proliferative and adiponectin-rich bone marrow niche was associated with substantial cardiac recovery by suppression of cardiac apoptosis and proliferation of cardiac fibroblast.
Collapse
Affiliation(s)
- Yong Sook Kim
- Cell Regeneration Research Center, Chonnam National University Hospital, Gwangju, Republic of Korea.,Biomedical Research Center, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Hyang Hee Cho
- Cell Regeneration Research Center, Chonnam National University Hospital, Gwangju, Republic of Korea.,Department of Molecular Medicine, Graduate School, Chonnam National University, Gwangju, Republic of Korea
| | - Dong Im Cho
- Cell Regeneration Research Center, Chonnam National University Hospital, Gwangju, Republic of Korea.,Biomedical Research Center, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Hye-Yun Jeong
- Cell Regeneration Research Center, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Soo Yeon Lim
- Cell Regeneration Research Center, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Ju Hee Jun
- Cell Regeneration Research Center, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Mi Ra Kim
- Cell Regeneration Research Center, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Bo Gyeong Kang
- Cell Regeneration Research Center, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Meeyoung Cho
- Cell Regeneration Research Center, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Hye-Jin Kang
- Cell Regeneration Research Center, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Wan Seok Kang
- Cell Regeneration Research Center, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Goo Taeg Oh
- Department of Life Sciences, Ewha Womans University, Seoul, Republic of Korea
| | - Youngkeun Ahn
- Cell Regeneration Research Center, Chonnam National University Hospital, Gwangju, Republic of Korea. .,Department of Cardiovascular Medicine, Chonnam National University Medical School, Gwangju, Republic of Korea. .,Department of Cardiology, Chonnam National University Hospital, Gwangju, Republic of Korea.
| |
Collapse
|
23
|
Penna C, Alloatti G, Crisafulli A. Mechanisms Involved in Cardioprotection Induced by Physical Exercise. Antioxid Redox Signal 2020; 32:1115-1134. [PMID: 31892282 DOI: 10.1089/ars.2019.8009] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Significance: Regular exercise training can reduce myocardial damage caused by acute ischemia/reperfusion (I/R). Exercise can reproduce the phenomenon of ischemic preconditioning, due to the capacity of brief periods of ischemia to reduce myocardial damage caused by acute I/R. In addition, exercise may also activate the multiple kinase cascade responsible for cardioprotection even in the absence of ischemia. Recent Advances: Animal and human studies highlighted the fact that, besides to reduce risk factors related to cardiovascular disease, the beneficial effects of exercise are also due to its ability to induce conditioning of the heart. Exercise behaves as a physiological stress that triggers beneficial adaptive cellular responses, inducing a protective phenotype in the heart. The factors contributing to the exercise-induced heart preconditioning include stimulation of the anti-radical defense system and nitric oxide production, opioids, myokines, and adenosine-5'-triphosphate (ATP) dependent potassium channels. They appear to be also involved in the protective effect exerted by exercise against cardiotoxicity related to chemotherapy. Critical Issues and Future Directions: Although several experimental evidences on the protective effect of exercise have been obtained, the mechanisms underlying this phenomenon have not yet been fully clarified. Further studies are warranted to define precise exercise prescriptions in patients at risk of myocardial infarction or undergoing chemotherapy.
Collapse
Affiliation(s)
- Claudia Penna
- National Institute for Cardiovascular Research (INRC), Bologna, Italy.,Department of Clinical and Biological Sciences, University of Turin, Torino, Italy
| | | | - Antonio Crisafulli
- Department of Medical Sciences and Public Health, Sports Physiology Lab., University of Cagliari, Cagliari, Italy
| |
Collapse
|
24
|
Chen X, Wang C, Yang P, Shi L, Wang H. Ube2s-stabilized β-catenin protects against myocardial ischemia/reperfusion injury by activating HIF-1α signaling. Aging (Albany NY) 2020; 12:5716-5732. [PMID: 32250966 PMCID: PMC7185123 DOI: 10.18632/aging.102960] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 02/22/2020] [Indexed: 04/12/2023]
Abstract
The activation of hypoxia-inducible factor (HIF) is an important event for mediating the adaptive response to myocardial ischemia/reperfusion (MI/R) injury. The ubiquitin-conjugating enzyme E2S (Ube2s) catalyzes ubiquitin conjugation to target proteins. Here, we report the positive regulation of HIF-1α signaling by Ube2s via stabilizing β-catenin, by which Ube2s acts to protect against MI/R injury. We show that Ube2s expression is upregulated in the hearts of mice subjected to MI/R injury. Functionally, Ube2s depletion exacerbates and its overexpression ameliorates MI/R injury. In addition, Ube2s augments the activation of HIF-1α and reduces myocardial apoptosis. Moreover, Ube2s induces the accumulation of β-Catenin through increasing its stabilization. Importantly, β-Catenin knockdown abrogates Ube2s-augmented HIF-1α activation, and meanwhile, diminishes the protective effect of Ube2s on MI/R injury, thus establishing a causal link between Ube2s-stabilized β-catenin and HIF-1α-mediated myocardial protection. Altogether, this study identifies the Ube2s/β-catenin/HIF-1α axis as a novel protective regulator involved in MI/R injury, and also implies that it might represent a potential therapeutic target for ameliorating MI/R injury.
Collapse
Affiliation(s)
- Xi Chen
- Department of Pharmacy, The Second Affiliated Hospital of Air Force Medical University, Xi’an, China
| | - Chiyao Wang
- Department of Cardiology, The Second Affiliated Hospital of Air Force Medical University, Xi’an, China
| | - Pei Yang
- Department of Cardiology, The Second Affiliated Hospital of Air Force Medical University, Xi’an, China
| | - Lei Shi
- Department of Pharmacy, The Second Affiliated Hospital of Air Force Medical University, Xi’an, China
| | - Haiyan Wang
- Department of Cardiology, The Second Affiliated Hospital of Air Force Medical University, Xi’an, China
| |
Collapse
|
25
|
Liu J, Meng Z, Gan L, Guo R, Gao J, Liu C, Zhu D, Liu D, Zhang L, Zhang Z, Xie D, Jiao X, Lau WB, Lopez BL, Christopher TA, Ma X, Cao J, Wang Y. C1q/TNF-related protein 5 contributes to diabetic vascular endothelium dysfunction through promoting Nox-1 signaling. Redox Biol 2020; 34:101476. [PMID: 32122792 PMCID: PMC7327962 DOI: 10.1016/j.redox.2020.101476] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 02/19/2020] [Accepted: 02/21/2020] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVE Dysregulated adipokine profiles contribute to the pathogenesis of diabetic cardiovascular complications. Endothelial cell (EC) dysfunction, a common pathological alteration in cardiovascular disorders, is exaggerated in diabetes. However, it is unclear whether and how dysregulated adipokines may contribute to diabetic EC dysfunction. METHODS AND RESULTS Serum C1q/TNF-Related Protein 5 (CTRP5) were determined in control/diabetes patients, and control/diabetic mice (high-fat diet, HFD). We observed for the first time that serum total CTRP5 was increased, high molecular weight (HMW) form was decreased, but the globular form (gCTRP5) was significantly increased in diabetic patients. These pathological alterations were reproduced in diabetic mice. To determine the pathological significance of increased gCTRP5 in diabetes, in vivo, ex vivo and in vitro experiments were performed. Diabetic atherosclerosis and EC dysfunction were significantly attenuated by the in vivo administration of CTRP5 neutralization antibody (CTRP5Ab). EC apoptosis was significantly increased in diabetic EC (isolated from HFD animal aorta) or high glucose high lipid (HGHL) cultured HUVECs. These pathological alterations were further potentiated by gCTRP5 and attenuated by CTRP5Ab. Pathway specific discovery-driven approach revealed that Nox1 expression was one of the signaling molecules commonly activated by HFD, HGHL, and gCTRP5. Treatment with CTRP5Ab reversed HFD-induced Nox1 upregulation. Finally, Nox1siRNA was used to determine the causative role of Nox1 in gCTRP5 induced EC apoptosis in diabetes. Results showed that gCTRP5 activated the mitochondrial apoptotic signal of EC in diabetes, which was blocked by the silencing Nox1 gene. CONCLUSION We demonstrated for the first time that gCTRP5 is a novel molecule contributing to diabetic vascular EC dysfunction through Nox1-mediated mitochondrial apoptosis, suggesting that interventions blocking gCTRP5 may protect diabetic EC function, ultimately attenuate diabetic cardiovascular complications.
Collapse
Affiliation(s)
- Jing Liu
- Department of Physiology, Shanxi Medical University, Shanxi, China; Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| | - Zhijun Meng
- Department of Physiology, Shanxi Medical University, Shanxi, China; Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| | - Lu Gan
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| | - Rui Guo
- Department of Physiology, Shanxi Medical University, Shanxi, China
| | - Jia Gao
- Department of Physiology, Shanxi Medical University, Shanxi, China
| | - Caihong Liu
- Department of Physiology, Shanxi Medical University, Shanxi, China
| | - Di Zhu
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| | - Demin Liu
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| | - Ling Zhang
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| | - Zhen Zhang
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| | - Dina Xie
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| | - Xiangying Jiao
- Department of Physiology, Shanxi Medical University, Shanxi, China
| | - Wayne Bond Lau
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| | - Bernard L Lopez
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| | | | - Xinliang Ma
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| | - Jimin Cao
- Department of Physiology, Shanxi Medical University, Shanxi, China.
| | - Yajing Wang
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
26
|
Vuong E, Nothling J, Lombard C, Jewkes R, Peer N, Abrahams N, Seedat S. Peripheral adiponectin levels in anxiety, mood, trauma- and stressor-related disorders: A systematic review and meta-analysis. J Affect Disord 2020; 260:372-409. [PMID: 31539673 DOI: 10.1016/j.jad.2019.09.050] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Revised: 07/01/2019] [Accepted: 09/08/2019] [Indexed: 02/08/2023]
Abstract
BACKGROUND Anxiety, mood, trauma- and stressor-related disorders confer increased risk for metabolic disease. Adiponectin, a cytokine released by adipose tissue is associated with these disorders and obesity via inflammatory processes. Available data describing associations with mental disorders remain limited and conflicted. METHODS A systematic search was conducted for English, peer-reviewed articles from inception until February 2019 that assessed for serum or plasma adiponectin levels in adults with an anxiety, mood or trauma-related disorder. Diagnoses were determined by psychiatric interview, based on DSM-IV, DSM-5 or ICD-10 criteria. Analyses were performed using STATA 15 and Standardized mean difference (SMD) with 95% confidence interval was applied to pool the effect size of meta-analysis studies. RESULTS In total 65 eligible studies were included in the systematic review and 30 studies in this meta-analysis. 19,178 participants (11,262 females and 7916 males), comprising healthy adults and adults with anxiety, mood and trauma-related disorders, were included. Overall results indicated an inverse association between adiponectin levels and examined mental disorders. Specifically, patients with an anxiety disorder (SMD = -1.18 µg/mL, 95% CI, -2.34; -0.01, p = 0.047); trauma or stressor-related disorder (SMD = -0.34 µg/mL, 95% CI, -0.52; -0.17, p = 0.0000) or bipolar disorder (SMD = -0.638 µg/mL, 95% CI, -1.16, -0.12, p = 0.017) had significant lower adiponectin levels compared to healthy adults. LIMITATIONS Heterogeneity, potential publication bias, and lack of control for important potential confounders were significant limitations. CONCLUSION Peripheral adiponectin levels appear to be inversely associated with anxiety, mood, trauma- and stressor related disorders and may be a promising biomarker for diagnosis and disease monitoring.
Collapse
Affiliation(s)
- E Vuong
- South African Research Chairs Initiative (SARChI), PTSD Program, Department of Psychiatry, Stellenbosch University, South Africa; Department of Psychiatry, Stellenbosch University, Stellenbosch, South Africa.
| | - J Nothling
- South African Research Chairs Initiative (SARChI), PTSD Program, Department of Psychiatry, Stellenbosch University, South Africa; Department of Psychiatry, Stellenbosch University, Stellenbosch, South Africa
| | - C Lombard
- Biostatistics Unit, South Africa Medical Research Council, Cape Town, South Africa
| | - R Jewkes
- Gender and Health Research Unit, South African Medical Research Council, Tygerberg, South Africa
| | - N Peer
- Non-Communicable Diseases Research Unit, South African Medical Research Council, Durban, South Africa
| | - N Abrahams
- Gender and Health Research Unit, South African Medical Research Council, Tygerberg, South Africa
| | - S Seedat
- South African Research Chairs Initiative (SARChI), PTSD Program, Department of Psychiatry, Stellenbosch University, South Africa; Department of Psychiatry, Stellenbosch University, Stellenbosch, South Africa
| |
Collapse
|
27
|
Xia Y, Zhang F, Zhao S, Li Y, Chen X, Gao E, Xu X, Xiong Z, Zhang X, Zhang J, Zhao H, Wang W, Wang H, Guo Y, Liu Y, Li C, Wang S, Zhang L, Yan W, Tao L. Adiponectin determines farnesoid X receptor agonism-mediated cardioprotection against post-infarction remodelling and dysfunction. Cardiovasc Res 2019; 114:1335-1349. [PMID: 29668847 DOI: 10.1093/cvr/cvy093] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 04/12/2018] [Indexed: 12/16/2022] Open
Abstract
Aims The farnesoid X receptor (FXR) is a member of the metabolic nuclear receptor superfamily that plays a critical regulatory role in cardiovascular physiology/pathology. However, the role of systemic FXR activation in the chronic phase in myocardial infarction (MI)-induced cardiac remodelling and dysfunction remains unclear. In this study, we aimed to elucidate the role of long-term FXR activation on post-MI cardiac remodelling and dysfunction. Methods and results Mice underwent either MI surgery or sham operation. At 1 week after MI, both sham and MI mice were gavaged with 25 mg/kg/d of a synthetic FXR agonist (GW4064) or a vehicle control for 7 weeks, and cardiac performance was assessed by consecutive echocardiography studies. Administration of GW4064 significantly increased left ventricular ejection fraction at 4 weeks and 8 weeks after MI (both P < 0.01). Moreover, GW4064 treatment increased angiogenesis and mitochondrial biogenesis, reduced cardiomyocyte loss and inflammation, and ameliorated cardiac remodelling as evidenced by heart weight, lung weight, atrial natriuretic peptide/brain natriuretic peptide levels, and myocardial fibrosis at 8 weeks post-MI. At the molecular level, GW4064 significantly increased FXR mRNA expression and transcriptional activity in heart tissue. Moreover, over-expression of myocardial FXR failed to exert significant cardioprotection in vivo, indicating that GW4064 improved post-MI heart remodelling and function independent of myocardial FXR expression/activity. Among the four down-stream soluble molecules of FXR, plasma adiponectin was most significantly increased by GW4064. In cultured adipocytes, GW4064 increased mRNA levels and protein expression of adiponectin. Conditioned medium of GW4064-treated adipocytes activated AMPK-PGC-1α signalling and reduced hypoxia-induced cardiomyocyte apoptosis, all of which were attenuated by an adiponectin neutralizing anti-body. More importantly, when knocking-out adiponectin in mice, the cardioprotective effects of GW4064 were attenuated. Conclusions We are the first to show that FXR agonism ameliorated post-MI cardiac dysfunction and remodelling by stimulating adiponectin secretion. Thus, we demonstrated that FXR agonism is a potential therapeutic strategy in post-MI heart failure.
Collapse
Affiliation(s)
- Yunlong Xia
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Road, Xi'an 710032, China
| | - Fuyang Zhang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Road, Xi'an 710032, China
| | - Shihao Zhao
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Road, Xi'an 710032, China.,Department of Cardiology, Hainan Branch of PLA General Hospital, Sanya 572013, China
| | - Yueyang Li
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Road, Xi'an 710032, China
| | - Xiyao Chen
- Department of Geriatrics, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Erhe Gao
- Center for Translational Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Xinyue Xu
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases, Department of Periodontology, School of Stomatology, Fourth Military Medical University, Xi'an 710032, China
| | - Zhenyu Xiong
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Road, Xi'an 710032, China
| | - Xiaomeng Zhang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Road, Xi'an 710032, China
| | - Jinglong Zhang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Road, Xi'an 710032, China
| | - Huishou Zhao
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Road, Xi'an 710032, China
| | - Wei Wang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Road, Xi'an 710032, China
| | - Helin Wang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Road, Xi'an 710032, China
| | - Yanjie Guo
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Road, Xi'an 710032, China
| | - Yi Liu
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Road, Xi'an 710032, China
| | - Congye Li
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Road, Xi'an 710032, China
| | - Shan Wang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Road, Xi'an 710032, China
| | - Ling Zhang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Road, Xi'an 710032, China
| | - Wenjun Yan
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Road, Xi'an 710032, China
| | - Ling Tao
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Road, Xi'an 710032, China
| |
Collapse
|
28
|
Guo R, Gan L, Lau WB, Yan Z, Xie D, Gao E, Christopher TA, Lopez BL, Ma X, Wang Y. Withaferin A Prevents Myocardial Ischemia/Reperfusion Injury by Upregulating AMP-Activated Protein Kinase-Dependent B-Cell Lymphoma2 Signaling. Circ J 2019; 83:1726-1736. [PMID: 31217391 DOI: 10.1253/circj.cj-18-1391] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND Withaferin A (WFA), an anticancer constituent of the plant Withania somnifera, inhibits tumor growth in association with apoptosis induction. However, the potential role of WFA in the cardiovascular system is little-studied and controversial. METHODS AND RESULTS Two different doses of WFA were tested to determine their cardioprotective effects in myocardial ischemia/reperfusion (MI/R) injury through evaluation of cardiofunction in wild-type and AMP-activated protein kinase domain negative (AMPK-DN) gentransgenic mice. Surprisingly, cardioprotective effects (improved cardiac function and reduced infarct size) were observed with low-dose WFA (1 mg/kg) delivery but not high-dose (5 mg/kg). Mechanistically, low-dose WFA attenuated myocardial apoptosis. It decreased MI/R-induced activation of caspase 9, the indicator of the intrinsic mitochondrial pathway, but not caspase 8. It also upregulated the level of AMP-activated protein kinase (AMPK) phosphorylation and increased the MI/R inhibited ratio of Bcl2/Bax. In AMPK-deficient mice, WFA did not ameliorate MI/R-induced cardiac dysfunction, attenuate infarct size, or restore the Bcl2/Bax (B-cell lymphoma2/Mcl-2-like protein 4) ratio. CONCLUSIONS These results demonstrated for the first time that low-dose WFA is cardioprotective via upregulation of the anti-apoptotic mitochondrial pathway in an AMPK-dependent manner.
Collapse
Affiliation(s)
- Rui Guo
- Department of Physiology, Shanxi Medical University
- Department of Emergency Medicine, Thomas Jefferson University
| | - Lu Gan
- Department of Emergency Medicine, Thomas Jefferson University
| | - Wayne Bond Lau
- Department of Emergency Medicine, Thomas Jefferson University
| | - Zheyi Yan
- Department of Physiology, Shanxi Medical University
- Department of Emergency Medicine, Thomas Jefferson University
| | - Dina Xie
- Department of Emergency Medicine, Thomas Jefferson University
| | - Erhe Gao
- Center for Translational Medicine, Temple University
| | | | - Bernard L Lopez
- Department of Emergency Medicine, Thomas Jefferson University
| | - Xinliang Ma
- Department of Emergency Medicine, Thomas Jefferson University
| | - Yajing Wang
- Department of Physiology, Shanxi Medical University
- Department of Emergency Medicine, Thomas Jefferson University
| |
Collapse
|
29
|
Lei X, Wu Q, Leng W, Wu M, Chen L, Liang Z. Exenatide reduces cardiomyocyte apoptosis by stimulating adiponectin secretion and activating APPL1-AMPK-PPARα axis. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:326. [PMID: 31475196 DOI: 10.21037/atm.2019.06.17] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Background To explore the mechanism that exenatide reduces cardiomyocyte apoptosis via the adiponectin pathway in vitro. Methods Cardiomyocytes were randomly divided into the control group (group C), diabetic group (group D), diabetic + exenatide treatment group (group DE), diabetic + exenatide treatment + APPL1 overexpression group (group OE), and diabetic + exenatide treatment + APPL1 knock-down group (group KD). After 48 h culture, the apoptosis rate, the adiponectin level in the cell culture fluid, and the expression levels of APPL1, p-AMPK, PPARα and NF-κB were detected by TUNEL, ELISA, and Western blotting, respectively. Results Compared to group C, the apoptosis rate was markedly increased, the adiponectin level was decreased, the expression of APPL1, p-AMPK and PPARα was down-regulated and that of NF-κB was up-regulated in group D (P<0.05); in group DE, the apoptosis rate was significantly decreased, the expression of APPL1, p-AMPK and PPARα was up-regulated and that of NF-κB was down-regulated, as compared with group D (P<0.05). The apoptosis rate in group OE was lower than that in group DE, the expression of APPL1, p-AMPK and PPARα was up-regulated and that of NF-κB was down-regulated (P<0.05). In group KD, the adiponectin level was elevated and the cardiomyocyte apoptosis rate was increased, as compared to group D (P<0.05). Furthermore, the expression of APPL1, p-AMPK and PPARα was down-regulated and that of NF-κB was up-regulated compared with group DE (P<0.05). Conclusions Exenatide can activate the "APPL1-AMPK-PPARα" anti-apoptosis signaling axis by promoting adiponectin expression in cardiomyocytes and reducing the apoptosis of diabetic cardiomyocytes, thus protecting cardiomyocytes.
Collapse
Affiliation(s)
- Xiaotian Lei
- Department of Endocrinology, Southwest Hospital, the Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Qinan Wu
- Department of Endocrine Nephropathy, Chongqing University Cancer Hospital and Chongqing Cancer Institute and Chongqing Cancer Hospital, Chongqing 400038, China
| | - Weiling Leng
- Department of Endocrinology, Southwest Hospital, the Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Minxia Wu
- Health Management Center, Southwest Hospital, the Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Liu Chen
- Department of Endocrinology, Southwest Hospital, the Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Ziwen Liang
- Department of Endocrinology, Southwest Hospital, the Third Military Medical University (Army Medical University), Chongqing 400038, China
| |
Collapse
|
30
|
Abstract
Adiponectin is one of the most widely studied adipokines to date. First described in the mid-1990's, studying its regulation, biogenesis and physiological effects has proven to be extremely insightful and improved our understanding of the mechanisms that ensure systemic metabolic homeostasis. Here, we provide a brief overview of the current state of the field with respect to adiponectin, its history, sites and mechanisms of action, and the critical questions that will need to be addressed in the future.
Collapse
Affiliation(s)
- Leon G Straub
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Philipp E Scherer
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
31
|
Yan Z, Guo R, Gan L, Lau WB, Cao X, Zhao J, Ma X, Christopher TA, Lopez BL, Wang Y. Withaferin A inhibits apoptosis via activated Akt-mediated inhibition of oxidative stress. Life Sci 2018; 211:91-101. [PMID: 30213729 DOI: 10.1016/j.lfs.2018.09.020] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 09/07/2018] [Accepted: 09/09/2018] [Indexed: 12/16/2022]
Abstract
Withaferin A (WFA), a withanolide derived from medicinal plant Withania somnifera, possesses anti-tumorigenic and immunomodulatory activities against various cancer cells. However, the role of WFA in myocardial ischemia reperfusion (MI/R) injury remains unclear. In the present study, we determined whether WFA may regulate cardiac ischemia reperfusion injury and elucidate the underlying mechanisms. We demonstrated that WFA enhanced H9c2 cells survival ability against simulated ischemia/reperfusion (SI/R) or hydrogen peroxide (H2O2)-induced cell apoptosis. In addition, the enhanced oxidative stress induced by SI/R was inhibited by WFA. Among the multiple antioxidant molecules determined, antioxidants SOD2, SOD3, Prdx-1 was obviously upregulated by WFA. When Akt inhibitor IV was administrated, WFA's suppression effect on oxidative stress was obviously abolished. Additional experiments demonstrated that WFA successfully inhibited H2O2 induced upregulation of SOD2, SOD3, and Prdx-1, ameliorated cardiomyocyte caspase-3 activity via an Akt dependent manner. Collectively, these results support the therapeutic potential of WFA against cardiac ischemia reperfusion injury and highlight the application of WFA in cardiovascular diseases holding great promise for the future.
Collapse
Affiliation(s)
- Zheyi Yan
- Department of Physiology, Shanxi Medical University, Shanxi, China; Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA, United States of America; Department of Ophthalmology, The First Affiliated Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Rui Guo
- Department of Physiology, Shanxi Medical University, Shanxi, China; Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA, United States of America
| | - Lu Gan
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA, United States of America
| | - Wayne Bond Lau
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA, United States of America
| | - Xiaoming Cao
- Department of Physiology, Shanxi Medical University, Shanxi, China
| | - Jianli Zhao
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA, United States of America
| | - Xinliang Ma
- Department of Physiology, Shanxi Medical University, Shanxi, China; Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA, United States of America
| | - Theodore A Christopher
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA, United States of America
| | - Bernard L Lopez
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA, United States of America
| | - Yajing Wang
- Department of Physiology, Shanxi Medical University, Shanxi, China; Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA, United States of America.
| |
Collapse
|
32
|
Guo R, Li G. Tanshinone modulates the expression of Bcl-2 and Bax in cardiomyocytes and has a protective effect in a rat model of myocardial ischemia-reperfusion. Hellenic J Cardiol 2018; 59:323-328. [PMID: 29412156 DOI: 10.1016/j.hjc.2017.11.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2017] [Revised: 11/13/2017] [Accepted: 11/21/2017] [Indexed: 11/24/2022] Open
Abstract
OBJECTIVE To investigate the protective activity of tanshinone in a rat model of myocardial ischemia-reperfusion and determine its effect on the expression of Bcl-2 and Bax in cardiomyocytes. METHODS We established a rat model of myocardial ischemia-reperfusion. Rats were randomly divided into blank (no surgery); saline; and low-dose (2 mg/ml), medium-dose (15 mg/ml), and high-dose (50 mg/ml) tanshinone groups. We measured heart rate and troponin (cTnI) levels, performed TUNEL to detect cardiomyocyte apoptosis, and detected LDH and CK-MB activities in serum by ELISA. We performed RT-qPCR and western blot to detect the expression of Bcl-2 and Bax mRNA and protein in cardiomyocytes. RESULTS Rats treated with tanshinone experienced more stable heart rate after ischemia-reperfusion compared with those in the saline control group. cTnI decreased after ischemia-reperfusion in mice injected with tanshinone, while cTnI in saline-treated mice increased significantly compared with that in the blank control group. TUNEL staining showed that there were greater apoptotic cardiomyocytes in the saline group, but the tanshinone groups showed fewer apoptotic cardiomyocytes. LDH and CK-MB activities were significantly increased after reperfusion in the saline group (p<0.01) and also in the low- and medium-dose tanshinone groups (p<0.05). However, no significant differences were found in the high-dose tanshinone group (p>0.05). The expression levels of Bcl-2 mRNA and protein in cardiomyocytes of rats were higher in the three tanshinone groups in a dose-sensitive manner than those in the blank and saline groups (p<0.05). By contrast, the expression levels of Bax mRNA and protein were reduced in the three tanshinone groups in a dose-sensitive manner compared to those in the blank and saline groups (p<0.05). CONCLUSION Tanshinone shows a protective effect in a dose-dependent manner in a rat model of myocardial ischemia-reperfusion, suggesting its potential therapeutic use.
Collapse
Affiliation(s)
- Rui Guo
- Department of Pharmacy, Hospital of Chengdu Office of People's Government of Tibetan Autonomous Region, Chengdu, 610041, China.
| | - Gen Li
- Department of Pharmacy, Chengdu Women & Children Central Hospital, Chengdu, 610091, China
| |
Collapse
|
33
|
Cardiac-derived CTRP9 protects against myocardial ischemia/reperfusion injury via calreticulin-dependent inhibition of apoptosis. Cell Death Dis 2018; 9:723. [PMID: 29925877 PMCID: PMC6010444 DOI: 10.1038/s41419-018-0726-3] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2018] [Revised: 05/10/2018] [Accepted: 05/17/2018] [Indexed: 12/17/2022]
Abstract
Cardiokines play an essential role in maintaining normal cardiac functions and responding to acute myocardial injury. Studies have demonstrated the heart itself is a significant source of C1q/TNF-related protein 9 (CTRP9). However, the biological role of cardiac-derived CTRP9 remains unclear. We hypothesize cardiac-derived CTRP9 responds to acute myocardial ischemia/reperfusion (MI/R) injury as a cardiokine. We explored the role of cardiac-derived CTRP9 in MI/R injury via genetic manipulation and a CTRP9-knockout (CTRP9-KO) animal model. Inhibition of cardiac CTRP9 exacerbated, whereas its overexpression ameliorated, left ventricular dysfunction and myocardial apoptosis. Endothelial CTRP9 expression was unchanged while cardiomyocyte CTRP9 levels decreased after simulated ischemia/`reperfusion (SI/R) in vitro. Cardiomyocyte CTRP9 overexpression inhibited SI/R-induced apoptosis, an effect abrogated by CTRP9 antibody. Mechanistically, cardiac-derived CTRP9 activated anti-apoptotic signaling pathways and inhibited endoplasmic reticulum (ER) stress-related apoptosis in MI/R injury. Notably, CTRP9 interacted with the ER molecular chaperone calreticulin (CRT) located on the cell surface and in the cytoplasm of cardiomyocytes. The CTRP9-CRT interaction activated the protein kinase A-cAMP response element binding protein (PKA-CREB) signaling pathway, blocked by functional neutralization of the autocrine CTRP9. Inhibition of either CRT or PKA blunted cardiac-derived CTRP9's anti-apoptotic actions against MI/R injury. We further confirmed these findings in CTRP9-KO rats. Together, these results demonstrate that autocrine CTRP9 of cardiomyocyte origin protects against MI/R injury via CRT association, activation of the PKA-CREB pathway, ultimately inhibiting cardiomyocyte apoptosis.
Collapse
|
34
|
|
35
|
Liu J, Sui H, Zhao J, Wang Y. Osmotin Protects H9c2 Cells from Simulated Ischemia-Reperfusion Injury through AdipoR1/PI3K/AKT Signaling Pathway. Front Physiol 2017; 8:611. [PMID: 28993734 PMCID: PMC5622187 DOI: 10.3389/fphys.2017.00611] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 08/09/2017] [Indexed: 01/04/2023] Open
Abstract
Objective: This study aimed to investigate the effect of osmotin on myocardial ischemia/reperfusion (I/R), as well as the underlying mechanisms. Methods:In vitro I/R injury model was established on rat cardiac myoblast H9c2 cells by oxygen and glucose deprivation followed by reperfusion (OGD/R). Cells were administrated with osmotin, and transfected with small interfering RNAs (siRNAs) which specifically target adiponectin receptor 1 or 2 (AdipoR1/2). Besides, the cells were incubated with or without LY294002 as inhibitor of phosphatidylinositol 3-kinase (PI3K) under OGD/R condition. Cell viability, apoptosis, expressions of apoptosis-related proteins and inflammatory factors were analyzed. Results: The results showed that osmotin significantly increased H9c2 cells viability compared with the cells treated with vehicle (P < 0.05), and decreased H9c2 cells apoptosis by regulating expressions of apoptosis-related proteins. Moreover, we observed that osmotin statistically reduced the release of proinflammatory factors and increased the release of anti-inflammatory factors in H9c2 cells (P < 0.05). However, these effects were markedly reversed by AdipoR1 silence but not AdipoR2. Furthermore, osmotin dramatically upregulated the phosphorylation levels of PI3K, AKT, ERK, and downregulated the phosphorylation level of NF-κB (P < 0.05). While administration of LY294002 reduced cell viability, increased cell apoptosis, and aggravated inflammatory response (P < 0.05). Conclusion: Our results suggested that the protective effect of osmotin on the simulated OGD/R injured H9c2 cells might be associated with AdipoR1/PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Jianhua Liu
- Department of Cardiology, Xinxiang Central HospitalXinxiang, Henan, China
| | - Hua Sui
- Department of Endocrinology, Xinxiang Central HospitalXinxiang, Henan, China
| | - Jianlin Zhao
- Department of Endocrinology, Xinxiang Central HospitalXinxiang, Henan, China
| | - Yan Wang
- Department of Cardiology, Xinxiang Central HospitalXinxiang, Henan, China
| |
Collapse
|
36
|
Guo B, Li Y, Jin X, Liu S, Miao C. Nitric oxide/cyclic GMP pathway mediates the endothelin-1-upregulation of adiponectin expression in rat cardiomyocytes. Biomed Rep 2017; 7:267-271. [PMID: 28808570 DOI: 10.3892/br.2017.953] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 09/21/2016] [Indexed: 11/06/2022] Open
Abstract
Endothelin-1 (ET-1) serves an important role in the development of cardiac dysfunction and heart failure. ET-1 and angiotensin II (AngII) comprise a mutually reciprocal signalling network in the myocardium and serve similar or additive roles in the development of heart failure. Our previous study previously demonstrated that AngII upregulated the expression of APN in cardiomyocytes through an AngII receptor type 2/nitric oxide (NO)/cyclic GMP(cGMP)-dependent pathway. The purpose of the present study was to determine the effects of ET-1 and the additive effects of ET-1 and AngII on the gene expression and secretion of APN, and the underlying mechanisms involved. ELISA was used to determine the secretion of adiponectin (APN) and reverse transcription-quantitative polymerase chain reaction was used to evaluate the gene expression of APN. ET-1 induced APN secretion in a time- and dose-dependent manner, and induced APN secretion with AngII simultaneously, as determined via APN mRNA analyses. ETA and ETB receptors were also involved. The use of a NO synthase inhibitor and an analogue of cGMP antagonist resulted in a diminished ET-1- and/or AngII-mediated APN induction in cardiomyocytes. These results suggested that ET-1, as well as AngII, upregulated the gene expression and secretion of APN via the common NO/cGMP-mediated mechanism.
Collapse
Affiliation(s)
- Bingyan Guo
- Department of Cardiovascular Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| | - Yongjun Li
- Department of Cardiovascular Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| | - Xin Jin
- Department of Cardiovascular Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| | - Suyun Liu
- Department of Cardiovascular Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| | - Chenglong Miao
- Department of Cardiovascular Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| |
Collapse
|
37
|
Li X, Dai Y, Yan S, Shi Y, Han B, Li J, Cha L, Mu J. Down-regulation of lncRNA KCNQ1OT1 protects against myocardial ischemia/reperfusion injury following acute myocardial infarction. Biochem Biophys Res Commun 2017; 491:1026-1033. [PMID: 28780351 DOI: 10.1016/j.bbrc.2017.08.005] [Citation(s) in RCA: 123] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 08/01/2017] [Indexed: 12/31/2022]
Abstract
This study aimed to investigate the protective effects of long non-coding RNA KCNQ1OT1 against myocardial ischemia/reperfusion (I/R) injury following acute myocardial infarction, as well as its regulatory mechanism. We used the cardiac muscle H9c2 cells under condition of oxygen glucose deprivation followed by reperfusion (OGD/R) to induce myocardial I/R injury. Then H9C2 cells were transfected with si-NC, si-KCNQ1OT1, pc-NC, pc-KCNQ1OT1, si-AdipoR1 and si-AdipoR2, respectively. The myocardial cell viability and apoptosis were respectively detected. In addition, the expression levels of inflammatory factors, apoptosis-related proteins and p38 MAPK/NF-κB pathway-related proteins were detected. Besides, an inhibitor of p38 MAPK/NF-κB pathway SB203580 was used to treat cells to verify the relationship between KCNQ1OT1 and p38 MAPK/NF-κB pathway. The expression of KCNQ1OT1 was significantly up-regulated in OGD/R-induced myocardial H9C2 cells. The OGD/R-induced decreased cell viability and AdipoR1 expression could be reversed after suppression of KCNQ1OT1. In addition, suppression of KCNQ1OT1 reduced OGD/R-induced increased expressions of TNF-α, IL-6 and IL-1β and OGD/R-induced increased cell apoptosis, which were reversed after knockdown of AdipoR1. Besides, suppression of KCNQ1OT1 significantly down-regulated the OGD/R-induced increased expression of p-p38 and p-NF-κB, which were also reversed after knockdown of AdipoR1. Moreover, SB203580, an inhibitor of p38 MAPK/NF-κB signal pathway, could further enhance the inhibitory effects of KCNQ1OT1 suppression on the expression of p-p38, TNF-α, IL-6, IL-1β and p-NF-κB in OGD/R-induced myocardial H9C2 cells. Suppression of KCNQ1OT1 may prevent myocardial I/R injury following acute myocardial infarction via regulating AdipoR1 and involving in p38 MAPK/NF-κB signal pathway.
Collapse
Affiliation(s)
- Xin Li
- Department of Cardiology, The First Affiliated Hospital of Medical College of Xi'AN JIAOTONG University, Xian, Shanxi 710061, China; Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, China
| | - Yingnan Dai
- Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, China
| | - Shujun Yan
- Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, China
| | - Yanli Shi
- Department of Record Room, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, China
| | - Baihe Han
- Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, China
| | - Jingxiu Li
- Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, China
| | - Li Cha
- Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, China
| | - Jianjun Mu
- Department of Cardiology, The First Affiliated Hospital of Medical College of Xi'AN JIAOTONG University, Xian, Shanxi 710061, China.
| |
Collapse
|
38
|
Liang Y, Li X, Zhang Y, Yeung SC, Zhen Z, Ip MSM, Tse HF, Lian Q, Mak JCW. Induced Pluripotent Stem Cells-Derived Mesenchymal Stem Cells Attenuate Cigarette Smoke-Induced Cardiac Remodeling and Dysfunction. Front Pharmacol 2017; 8:501. [PMID: 28804458 PMCID: PMC5532447 DOI: 10.3389/fphar.2017.00501] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 07/14/2017] [Indexed: 12/11/2022] Open
Abstract
The strong relationship between cigarette smoking and cardiovascular disease (CVD) has been well-documented, but the mechanisms by which smoking increases CVD risk appear to be multifactorial and incompletely understood. Mesenchymal stem cells (MSCs) are regarded as an important candidate for cell-based therapy in CVD. We hypothesized that MSCs derived from induced pluripotent stem cell (iPSC-MSCs) or bone marrow (BM-MSCs) might alleviate cigarette smoke (CS)-induced cardiac injury. This study aimed to investigate the effects of BM-MSCs or iPSC-MSCs on CS-induced changes in serum and cardiac lipid profiles, oxidative stress and inflammation as well as cardiac function in a rat model of passive smoking. Male Sprague-Dawley rats were randomly selected for exposure to either sham air (SA) as control or 4% CS for 1 h per day for 56 days. On day 29 and 43, human adult BM-MSCs, iPSC-MSCs or PBS were administered intravenously to CS-exposed rats. Results from echocardiography, serum and cardiac lipid profiles, cardiac antioxidant capacity, cardiac pro- and anti-inflammatory cytokines and cardiac morphological changes were evaluated at the end of treatment. iPSC-MSC-treated group showed a greater effect in the improvement of CS-induced cardiac dysfunction over BM-MSCs-treated group as shown by increased percentage left ventricular ejection fraction and percentage fractional shortening, in line with the greater reversal of cardiac lipid abnormality. In addition, iPSC-MSCs administration attenuated CS-induced elevation of cardiac pro-inflammatory cytokines as well as restoration of anti-inflammatory cytokines and anti-oxidative markers, leading to ameliorate cardiac morphological abnormalities. These data suggest that iPSC-MSCs on one hand may restore CS-induced cardiac lipid abnormality and on the other hand may attenuate cardiac oxidative stress and inflammation via inhibition of CS-induced NF-κB activation, leading to improvement of cardiac remodeling and dysfunction. Thus, iPSC-MSCs may be a promising candidate in cell-based therapy to prevent cardiac complications in smokers.
Collapse
Affiliation(s)
- Yingmin Liang
- Department of Medicine, The University of Hong KongPok Fu Lam, Hong Kong.,Shenzhen Institute of Research and Innovation, The University of Hong KongPok Fu Lam, Hong Kong
| | - Xiang Li
- Department of Medicine, The University of Hong KongPok Fu Lam, Hong Kong.,Shenzhen Institute of Research and Innovation, The University of Hong KongPok Fu Lam, Hong Kong
| | - Yuelin Zhang
- Department of Medicine, The University of Hong KongPok Fu Lam, Hong Kong
| | - Sze Chun Yeung
- Department of Medicine, The University of Hong KongPok Fu Lam, Hong Kong.,Shenzhen Institute of Research and Innovation, The University of Hong KongPok Fu Lam, Hong Kong
| | - Zhe Zhen
- Department of Medicine, The University of Hong KongPok Fu Lam, Hong Kong
| | - Mary S M Ip
- Department of Medicine, The University of Hong KongPok Fu Lam, Hong Kong.,Shenzhen Institute of Research and Innovation, The University of Hong KongPok Fu Lam, Hong Kong.,Research Centre of Heart, Brain, Hormone and Healthy Aging, The University of Hong KongPok Fu Lam, Hong Kong
| | - Hung Fat Tse
- Department of Medicine, The University of Hong KongPok Fu Lam, Hong Kong
| | - Qizhou Lian
- Department of Medicine, The University of Hong KongPok Fu Lam, Hong Kong.,Shenzhen Institute of Research and Innovation, The University of Hong KongPok Fu Lam, Hong Kong.,Research Centre of Heart, Brain, Hormone and Healthy Aging, The University of Hong KongPok Fu Lam, Hong Kong.,Department of Ophthalmology, The University of Hong KongPok Fu Lam, Hong Kong
| | - Judith C W Mak
- Department of Medicine, The University of Hong KongPok Fu Lam, Hong Kong.,Shenzhen Institute of Research and Innovation, The University of Hong KongPok Fu Lam, Hong Kong.,Research Centre of Heart, Brain, Hormone and Healthy Aging, The University of Hong KongPok Fu Lam, Hong Kong.,Department of Pharmacology and Pharmacy, The University of Hong KongPok Fu Lam, Hong Kong
| |
Collapse
|
39
|
Wang Y, Ma XL, Lau WB. Cardiovascular Adiponectin Resistance: The Critical Role of Adiponectin Receptor Modification. Trends Endocrinol Metab 2017; 28:519-530. [PMID: 28473178 PMCID: PMC6391995 DOI: 10.1016/j.tem.2017.03.004] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2016] [Revised: 03/01/2017] [Accepted: 03/28/2017] [Indexed: 12/23/2022]
Abstract
For the past two decades, a great deal of research has been published concerning adiponectin (APN), an abundant protein responsible for regulating numerous biologic functions including antioxidative, antinitrative, anti-inflammatory, and cardioprotective effects. A review of APN and its two major receptors is timely because of new findings concerning the mechanisms by which APN signaling may be altered in pathologic processes such as diabetes and heart failure. In this review we elaborate on currently known information regarding the physiologic role of APN and the known mechanisms underlying pathologic APN resistance - namely, APN receptor downregulation and phosphorylation - and provide insight regarding the future directions of APN research including an assessment of the clinical applicability of preventing pathologic post-translational modification of the APN receptor.
Collapse
Affiliation(s)
- Yajing Wang
- Department of Emergency Medicine, Thomas Jefferson University, 1025 Walnut Street, Philadelphia, PA 19107, USA
| | - Xin L Ma
- Department of Emergency Medicine, Thomas Jefferson University, 1025 Walnut Street, Philadelphia, PA 19107, USA
| | - Wayne Bond Lau
- Department of Emergency Medicine, Thomas Jefferson University, 1025 Walnut Street, Philadelphia, PA 19107, USA.
| |
Collapse
|
40
|
Abstract
Cardiovascular disease (CVD) is the greatest cause of death, accounting for nearly one-third of all deaths worldwide. The increase in obesity rates over 3 decades is widespread and threatens the public health in both developed and developing countries. Obesity, the excessive accumulation of visceral fat, causes the clustering of metabolic disorders, such as type 2 diabetes, dyslipidemia, and hypertension, culminating in the development of CVD. Adipose tissue is not only an energy storage organ, but an active endocrine tissue producing various biologically active proteins known as adipokines. Since leptin, a central regulator of food intake and energy expenditure, was demonstrated to be an adipose-specific adipokine, attention has focused on the identification and characterization of unknown adipokines to clarify the mechanisms underlying obesity-related disorders. Numerous adipokines have been identified in the past 2 decades; most adipokines are upregulated in the obese state. Adipokines such as tumor necrosis factor (TNF)-α, interleukin (IL)-6, IL-1β, and resistin are pro-inflammatory, and exacerbate various metabolic and cardiovascular diseases. However, a small number of adipokines, including adiponectin, are decreased by obesity, and generally exhibit antiinflammatory properties and protective functions against obesity-related diseases. Collectively, an imbalance in the production of pro- and antiinflammatory adipokines in the obese condition results in multiple complications. In this review, we focus on the pathophysiologic roles of adipokines with cardiovascular protective properties.
Collapse
Affiliation(s)
- Wayne Bond Lau
- Department of Emergency Medicine, Thomas Jefferson University
| | - Koji Ohashi
- Molecular Cardiovascular Medicine, Nagoya University Graduate School of Medicine
| | - Yajing Wang
- Department of Emergency Medicine, Thomas Jefferson University
| | - Hayato Ogawa
- Department of Cardiology, Nagoya University Graduate School of Medicine
| | - Toyoaki Murohara
- Department of Cardiology, Nagoya University Graduate School of Medicine
| | - Xin-Liang Ma
- Department of Emergency Medicine, Thomas Jefferson University
| | - Noriyuki Ouchi
- Molecular Cardiovascular Medicine, Nagoya University Graduate School of Medicine
| |
Collapse
|
41
|
Zhang J, Xia L, Zhang F, Zhu D, Xin C, Wang H, Zhang F, Guo X, Lee Y, Zhang L, Wang S, Guo X, Huang C, Gao F, Liu Y, Tao L. A novel mechanism of diabetic vascular endothelial dysfunction: Hypoadiponectinemia-induced NLRP3 inflammasome activation. Biochim Biophys Acta Mol Basis Dis 2017; 1863:1556-1567. [DOI: 10.1016/j.bbadis.2017.02.012] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 02/05/2017] [Accepted: 02/09/2017] [Indexed: 12/15/2022]
|
42
|
Wang H, Wu W, Duan J, Ma M, Kong W, Ke Y, Li G, Zheng J. Cardioprotection of ischemic preconditioning in rats involves upregulating adiponectin. J Mol Endocrinol 2017; 58:155-165. [PMID: 28219936 DOI: 10.1530/jme-16-0163] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 01/27/2017] [Indexed: 11/08/2022]
Abstract
It has been reported that ischemic preconditioning (IPC) and adiponectin (APN) are cardioprotective in many cardiovascular disorders. However, whether APN mediates the effect of IPC on myocardial injury has not been elucidated. This study was conducted to investigate whether IPC affects myocardial ischemic injury by increasing APN expression. Male adult rats with cardiac knockdowns of APN and its receptors via intramyocardial small-interfering RNA injection were subjected to IPC and then myocardial infarction (MI) at 24 h after IPC. Globular APN (gAd) was injected at 10 min before MI. APN mRNA and protein levels in myocardium as well as the plasma APN concentration were markedly high at 6 and 12 h after IPC. IPC ameliorated myocardial injury as evidenced by improved cardiac functions and a reduced infarct size. Compared with the control MI group, rats in the IPC + MI group had elevated levels of left ventricular ejection fraction and fractional shortening and a smaller MI size (P < 0.05). However, the aforementioned protective effects were ameliorated in the absence of APN and APN receptors, followed by the inhibition of AMP-activated protein kinase (AMPK) phosphorylation, but reversed by gAd treatment in wild-type rats, and AMPK phosphorylation increased (P < 0.05). Overall, our results suggest that the cardioprotective effects of IPC are partially due to upregulation of APN and provide a further insight into IPC-mediated signaling effects.
Collapse
Affiliation(s)
- Hui Wang
- Department of Intensive Care UnitChina-Japan Friendship Hospital, Beijing, People's Republic of China
| | - Wenjing Wu
- Department of CardiologyChina-Japan Friendship Hospital, Beijing, People's Republic of China
| | - Jun Duan
- Department of Intensive Care UnitChina-Japan Friendship Hospital, Beijing, People's Republic of China
| | - Ming Ma
- Department of Plastic and CosmetologyBeijing Haidian Hospital, Beijing, People's Republic of China
| | - Wei Kong
- Department of Physiology and PathophysiologySchool of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, People's Republic of China
| | - Yuannan Ke
- Department of CardiologyChina-Japan Friendship Hospital, Beijing, People's Republic of China
| | - Gang Li
- Department of Intensive Care UnitChina-Japan Friendship Hospital, Beijing, People's Republic of China
| | - Jingang Zheng
- Department of CardiologyChina-Japan Friendship Hospital, Beijing, People's Republic of China
| |
Collapse
|
43
|
Sun Y, Zhao D, Yang Y, Gao C, Zhang X, Ma Z, Jiang S, Zhao L, Chen W, Ren K, Yi W, Gao F. Adiponectin exerts cardioprotection against ischemia/reperfusion injury partially via calreticulin mediated anti-apoptotic and anti-oxidative actions. Apoptosis 2016; 22:108-117. [DOI: 10.1007/s10495-016-1304-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
44
|
Adiponectin protects the rats liver against chronic intermittent hypoxia induced injury through AMP-activated protein kinase pathway. Sci Rep 2016; 6:34151. [PMID: 27678302 PMCID: PMC5039704 DOI: 10.1038/srep34151] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 09/05/2016] [Indexed: 12/25/2022] Open
Abstract
This study was performed to assess the effect of chronic intermittent hypoxia (CIH) on the liver, the associated mechanisms and the potential therapeutic roles of adiponectin (Ad). Sixty rats were randomly assigned to four groups: the normal control (NC), NC and Ad supplement (NC + Ad), CIH, and CIH and Ad supplement (CIH + Ad) groups. The rats in the CIH and CIH + Ad groups were exposed to a hypoxic environment for 4 months. Rats in the NC + Ad and CIH + Ad groups were also treated with an intravenous injection of Ad (10 ug), twice a week. The plasma levels of hepatic enzymes, serum triglyceride, liver triglyceride, fasting blood glucose and hepatic cell apoptosis in hepatic tissue, were higher in the CIH group than in the NC and NC + Ad groups. However, the Ad supplementation in the CIH + Ad group rescued the hepatic tissue insult by activating the AMP-activated protein kinase (AMPK) pathway. In conclusion, Ad could protect against CIH-induced hepatic injury partly through the AMPK pathway.
Collapse
|
45
|
Li H, Yao W, Liu Z, Xu A, Huang Y, Ma XL, Irwin MG, Xia Z. Hyperglycemia Abrogates Ischemic Postconditioning Cardioprotection by Impairing AdipoR1/Caveolin-3/STAT3 Signaling in Diabetic Rats. Diabetes 2016; 65:942-55. [PMID: 26718505 DOI: 10.2337/db15-0782] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Accepted: 12/12/2015] [Indexed: 11/13/2022]
Abstract
Signal transducer and activator of transcription 3 (STAT3) activation is key for ischemic postconditioning (IPo) to attenuate myocardial ischemia-reperfusion injury (MIRI), but IPo loses cardioprotection in diabetes in which cardiac STAT3 activation is impaired and adiponectin (APN) reduced. We found that IPo increased postischemic cardiomyocyte-derived APN, activated mitochondrial STAT3 (mitoSTAT3), improved mitochondrial function, and attenuated MIRI in wild-type but not in APN knockout (Adipo(-/-)) mice subjected to 30 min coronary occlusion, followed by 2 or 24 h of reperfusion. Hypoxic postconditioning-induced protection against hypoxia/reoxygenation injury was lost in Adipo(-/-) cardiomyocytes but restored by recombinant APN, but this APN beneficial effect was abolished by specific STAT3 or APN receptor 1 (AdipoR1) gene knockdown, or caveolin-3 (Cav3) disruption. APN activated cardiac STAT3 and restored IPo cardioprotection in 4-week diabetic rats where AdipoR1 and Cav3 were functionally interactive but not in 8-week diabetic rats whose cardiac Cav3 was severely reduced and AdipoR1/Cav3 signaling impaired. We concluded that IPo activates mitoSTAT3 through APN/AdipoR1/Cav3 pathway to confer cardioprotection, whereas in diabetes, IPo loses cardioprotection due to impaired APN/AdipoR1/Cav3 signaling. Therefore, effective means that may concomitantly activate APN and repair APN signaling (i.e., AdipoR1/Cav3) in diabetes may represent promising avenues in the treatment of MIRI in diabetes.
Collapse
Affiliation(s)
- Haobo Li
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China Department of Anesthesiology, The University of Hong Kong, Hong Kong, China
| | - Weifeng Yao
- Department of Anesthesiology, The University of Hong Kong, Hong Kong, China
| | - Zipeng Liu
- Department of Anesthesiology, The University of Hong Kong, Hong Kong, China Centre for Genomic Sciences, The University of Hong Kong, Hong Kong, China
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Yu Huang
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China Institute of Vascular Medicine and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Xin-Liang Ma
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA
| | - Michael G Irwin
- Department of Anesthesiology, The University of Hong Kong, Hong Kong, China
| | - Zhengyuan Xia
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China Department of Anesthesiology, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
46
|
Jung CC, Su HJ. Long-term allergen exposure induces adipose tissue inflammation and circulatory system injury. Cell Immunol 2016; 303:34-42. [PMID: 27004794 DOI: 10.1016/j.cellimm.2016.03.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 03/05/2016] [Accepted: 03/07/2016] [Indexed: 11/30/2022]
Abstract
The purpose of this study was to study whether allergen exposure can induce inflammation and lower the anti-inflammation levels in serum and in adipose tissues, and further develop cardiovascular injury. Our data showed that heart rate was significantly higher in the OVA-challenged mice compared to control mice. Moreover, there were higher expressions of pro-inflammation genes in the OVA-challenged mice in adipose tissues, and the expressions of anti-inflammation genes were lower. The levels of inflammation mediators were associated in serum and adipose tissues. The level of circulatory injury lactate dehydrogenase was significantly associated with the levels of E-selectin, resistin and adiponectin in the serum. The hematoxylin and eosin and immunohistochemistry stains indicated the OVA-challenged mice had higher levels of inflammation. In summary, the current study demonstrated allergen exposure can cause cardiovascular injury, and inflammatory mediators in adipose tissues play an important role in the pathogenesis of cardiovascular injury.
Collapse
Affiliation(s)
- Chien-Cheng Jung
- Department of Environmental and Occupational Health, Out-Patient Building, College of Medicine, National Cheng Kung University, 138 Sheng-Li Road, Tainan 70403, Taiwan
| | - Huey-Jen Su
- Department of Environmental and Occupational Health, Out-Patient Building, College of Medicine, National Cheng Kung University, 138 Sheng-Li Road, Tainan 70403, Taiwan.
| |
Collapse
|
47
|
Silva-Palacios A, Königsberg M, Zazueta C. Nrf2 signaling and redox homeostasis in the aging heart: A potential target to prevent cardiovascular diseases? Ageing Res Rev 2016; 26:81-95. [PMID: 26732035 DOI: 10.1016/j.arr.2015.12.005] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Revised: 12/09/2015] [Accepted: 12/21/2015] [Indexed: 10/22/2022]
Abstract
Aging process is often accompanied with a high incidence of cardiovascular diseases (CVD) due to the synergistic effects of age-related changes in heart morphology/function and prolonged exposure to injurious effects of CVD risk factors. Oxidative stress, considered a hallmark of aging, is also an important feature in pathologies that predispose to CVD development, like hypertension, diabetes and obesity. Approaches directed to prevent the occurrence of CVD during aging have been explored both in experimental models and in controlled clinical trials, in order to improve health span, reduce hospitalizations and increase life quality during elderly. In this review we discuss oxidative stress role as a main risk factor that relates CVD with aging. As well as interventions that aim to reduce oxidative stress by supplementing with exogenous antioxidants. In particular, strategies of improving the endogenous antioxidant defenses through activating the nuclear factor related-2 factor (Nrf2) pathway; one of the best studied molecules in cellular redox homeostasis and a master regulator of the antioxidant and phase II detoxification response.
Collapse
|
48
|
Wang LL, Miller D, Wanders D, Nanayakkara G, Amin R, Judd R, Morrison EE, Zhong JM. Adiponectin downregulation is associated with volume overload-induced myocyte dysfunction in rats. Acta Pharmacol Sin 2016; 37:187-95. [PMID: 26616727 DOI: 10.1038/aps.2015.84] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 08/26/2015] [Indexed: 12/13/2022]
Abstract
AIM Adiponectin has been reported to exert protective effects during pathological ventricular remodeling, but the role of adiponectin in volume overload-induced heart failure remains unclear. In this study we investigated the effect of adiponectin on cardiac myocyte contractile dysfunction following volume overload in rats. METHODS Volume overload was surgically induced in rats by infrarenal aorta-vena cava fistula. The rats were intravenously administered adenoviral adiponectin at 2-, 6- and 9-weeks following fistula. The protein expression of adiponectin, adiponectin receptors (AdipoR1/R2 and T-cadherin) and AMPK activity were measured using Western blot analyses. Isolated ventricular myocytes were prepared at 12 weeks post-fistula to examine the contractile performance of myocytes and intracellular Ca(2+) transient. RESULTS A-V fistula resulted in significant reductions in serum and myocardial adiponectin levels, myocardial adiponectin receptor (AdipoR1/R2 and T-cadherin) levels, as well as myocardial AMPK activity. Consistent with these changes, the isolated myocytes exhibited significant depression in cell shortening and intracellular Ca(2+) transient. Administration of adenoviral adiponectin significantly increased serum adiponectin levels and prevented myocyte contractile dysfunction in fistula rats. Furthermore, pretreatment of isolated myocytes with recombinant adiponectin (2.5 μg/mL) significantly improved their contractile performance in fistula rats, but had no effects in control or adenoviral adiponectin-administered rats. CONCLUSION These results demonstrate a positive correlation between adiponectin downregulation and volume overload-induced ventricular remodeling. Adiponectin plays a protective role in volume overload-induced heart failure.
Collapse
|
49
|
Gao C, Liu Y, Yu Q, Yang Q, Li B, Sun L, Yan W, Cai X, Gao E, Xiong L, Wang H, Tao L. TNF-α antagonism ameliorates myocardial ischemia-reperfusion injury in mice by upregulating adiponectin. Am J Physiol Heart Circ Physiol 2015; 308:H1583-91. [PMID: 25888509 DOI: 10.1152/ajpheart.00346.2014] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Accepted: 03/23/2015] [Indexed: 11/22/2022]
Abstract
Tumor necrosis factor-α (TNF-α) antagonism alleviates myocardial ischemia-reperfusion (MI/R) injury. However, the mechanisms by which the downstream mediators of TNF-α change after acute antagonism during MI/R remain unclear. Adiponectin (APN) exerts anti-ischemic effects, but it is downregulated during MI/R. This study was conducted to investigate whether TNF-α is responsible for the decrease of APN, and whether antagonizing TNF-α affects MI/R injury by increasing APN. Male adult wild-type (WT), APN knockout (APN KO) mice, and those with cardiac knockdowns of APN receptors via siRNA injection were subjected to 30 min of MI followed by reperfusion. The TNF-α antagonist etanercept or globular domain of APN (gAD) was injected 10 min before reperfusion. Etanercept ameliorated MI/R injury in WT mice as evidenced by improved cardiac function, and reduced infarct size and cardiomyocyte apoptosis. APN concentrations were augmented in response to etanercept, followed by an increase in AMP-activated protein kinase phosphorylation. Etanercept still increased cardiac function and reduced infarct size and apoptosis in both APN KO and APN receptors knockdown mice. However, its potential was significantly weakened in these mice compared with the WT mice. TNF-α is responsible for the decrease in APN during MI/R. The cardioprotective effects of TNF-α neutralization are partially due to the upregulation of APN. The results provide more insight into the TNF-α-mediated signaling effects during MI/R and support the need for clinical trials to validate the efficacy of acute TNF-α antagonism in the treatment of MI/R injury.
Collapse
Affiliation(s)
- Chao Gao
- Department of Cardiology, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Yi Liu
- Department of Cardiology, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Qiujun Yu
- Department of Cardiology, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Qiang Yang
- Department of Cardiology, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Bing Li
- Department of Dermatology, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Lu Sun
- Department of Cardiology, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Wenjun Yan
- Department of Cardiology, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Xiaoqing Cai
- Department of Physiology, The Fourth Military Medical University, Xi'an, China; and
| | - Erhe Gao
- Department of Physiology, The Fourth Military Medical University, Xi'an, China; and
| | - Lize Xiong
- Department of Anesthesiology, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Haichang Wang
- Department of Cardiology, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Ling Tao
- Department of Cardiology, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| |
Collapse
|
50
|
Yuan Y, Lau WB, Su H, Sun Y, Yi W, Du Y, Christopher T, Lopez B, Wang Y, Ma XL. C1q-TNF-related protein-9, a novel cardioprotetcive cardiokine, requires proteolytic cleavage to generate a biologically active globular domain isoform. Am J Physiol Endocrinol Metab 2015; 308:E891-8. [PMID: 25783894 PMCID: PMC4436995 DOI: 10.1152/ajpendo.00450.2014] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Accepted: 03/10/2015] [Indexed: 12/23/2022]
Abstract
Prevalence and severity of postmyocardial infarction heart failure continually escalate in type 2 diabetes via incompletely understood mechanisms. The discovery of the cardiac secretomes, collectively known as "cardiokines", has significantly enhanced appreciation of the local microenvironment's influence on disease development. Recent studies demonstrated that C1q-TNF-related protein-9 (CTRP9), a newly discovered adiponectin (APN) paralog, is highly expressed in the heart. However, its relationship with APN (concerning diabetic cardiovascular injury in particular) remains unknown. Plasma CTRP9 levels are elevated in APN knockout and reduced in diabetic mice. In contrast to APN, which circulates as full-length multimers, CTRP9 circulates in the plasma primarily in the globular domain isoform (gCTRP9). Recombinant full-length CTRP9 (fCTRP9) was cleaved when incubated with cardiac tissue extracts, generating gCTRP9, a process inhibited by protease inhibitor cocktail. gCTRP9 rapidly activates cardiac survival kinases, including AMPK, Akt, and endothelial NOS. However, fCTRP9-mediated kinase activation is much less potent and significantly delayed. Kinase activation by fCTRP9, but not gCTRP9, is inhibited by protease inhibitor cocktail. These results demonstrate for the first time that the novel cardiokine CTRP9 undergoes proteolytic cleavage to generate gCTRP9, the dominant circulatory and actively cardioprotective isoform. Enhancing cardiac CTRP9 production and/or its proteolytic posttranslational modification are of therapeutic potential, attenuating diabetic cardiac injury.
Collapse
Affiliation(s)
- Yuexing Yuan
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania; Zhejiang Provincial Hospital of Chinese Traditional Medicine, Hangzhou, Zhejiang Province, China
| | - Wayne Bond Lau
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Hui Su
- Department of Geriatrics, Xijing Hospital, the Fourth Military Medical University, Xi'an, China
| | - Yang Sun
- Department of Geriatrics, Xijing Hospital, the Fourth Military Medical University, Xi'an, China
| | - Wei Yi
- Department of Cardiovascular Surgery, Xijing Hospital, the Fourth Military Medical University, Xi'an, China
| | - Yunhui Du
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Theodore Christopher
- Department of Geriatrics, Xijing Hospital, the Fourth Military Medical University, Xi'an, China
| | - Bernard Lopez
- Department of Geriatrics, Xijing Hospital, the Fourth Military Medical University, Xi'an, China
| | - Yajing Wang
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Xin-Liang Ma
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania; Center for Translational Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania; and
| |
Collapse
|