1
|
Zhou R, Barnes K, Gibson S, Fillmore N. Dual-edged role of SIRT1 in energy metabolism and cardiovascular disease. Am J Physiol Heart Circ Physiol 2024; 327:H1162-H1173. [PMID: 39269450 DOI: 10.1152/ajpheart.00001.2024] [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: 01/01/2024] [Revised: 08/13/2024] [Accepted: 09/06/2024] [Indexed: 09/15/2024]
Abstract
Regulation of energy metabolism is pivotal in the development of cardiovascular diseases. Dysregulation in mitochondrial fatty acid oxidation has been linked to cardiac lipid accumulation and diabetic cardiomyopathy. Sirtuin 1 (SIRT1) is a deacetylase that regulates the acetylation of various proteins involved in mitochondrial energy metabolism. SIRT1 mediates energy metabolism by directly and indirectly affecting multiple aspects of mitochondrial processes, such as mitochondrial biogenesis. SIRT1 interacts with essential mitochondrial energy regulators such as peroxisome proliferator-activated receptor-α (PPARα), PPARγ coactivator-1α, estrogen-related receptor-α, and their downstream targets. Apart from that, SIRT1 regulates additional proteins, including forkhead box protein O1 and AMP-activated protein kinase in cardiac disease. Interestingly, studies have also shown that the expression of SIRT1 plays a dual-edged role in energy metabolism. Depending on the physiological state, SIRT1 expression can be detrimental or protective. This review focuses on the molecular pathways through which SIRT1 regulates energy metabolism in cardiovascular diseases. We will review SIRT1 and discuss its role in cardiac energy metabolism and its benefits and detrimental effects in heart disease.
Collapse
Affiliation(s)
- Redemptor Zhou
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health and Human Sciences, North Dakota State University, Fargo, North Dakota, United States
| | - Kaleb Barnes
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health and Human Sciences, North Dakota State University, Fargo, North Dakota, United States
| | - Savannah Gibson
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health and Human Sciences, North Dakota State University, Fargo, North Dakota, United States
| | - Natasha Fillmore
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health and Human Sciences, North Dakota State University, Fargo, North Dakota, United States
| |
Collapse
|
2
|
Zhang J, Wang G, Shi Y, Liu X, Liu S, Chen W, Ning Y, Cao Y, Zhao Y, Li M. Growth differentiation factor 11 regulates high glucose-induced cardiomyocyte pyroptosis and diabetic cardiomyopathy by inhibiting inflammasome activation. Cardiovasc Diabetol 2024; 23:160. [PMID: 38715043 PMCID: PMC11077721 DOI: 10.1186/s12933-024-02258-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 05/01/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND Diabetic cardiomyopathy (DCM) is a crucial complication of long-term chronic diabetes that can lead to myocardial hypertrophy, myocardial fibrosis, and heart failure. There is increasing evidence that DCM is associated with pyroptosis, a form of inflammation-related programmed cell death. Growth differentiation factor 11 (GDF11) is a member of the transforming growth factor β superfamily, which regulates oxidative stress, inflammation, and cell survival to mitigate myocardial hypertrophy, myocardial infarction, and vascular injury. However, the role of GDF11 in regulating pyroptosis in DCM remains to be elucidated. This research aims to investigate the role of GDF11 in regulating pyroptosis in DCM and the related mechanism. METHODS AND RESULTS Mice were injected with streptozotocin (STZ) to induce a diabetes model. H9c2 cardiomyocytes were cultured in high glucose (50 mM) to establish an in vitro model of diabetes. C57BL/6J mice were preinjected with adeno-associated virus 9 (AAV9) intravenously via the tail vein to specifically overexpress myocardial GDF11. GDF11 attenuated pyroptosis in H9c2 cardiomyocytes after high-glucose treatment. In diabetic mice, GDF11 alleviated cardiomyocyte pyroptosis, reduced myocardial fibrosis, and improved cardiac function. Mechanistically, GDF11 inhibited pyroptosis by preventing inflammasome activation. GDF11 achieved this by specifically binding to apoptosis-associated speck-like protein containing a CARD (ASC) and preventing the assembly and activation of the inflammasome. Additionally, the expression of GDF11 during pyroptosis was regulated by peroxisome proliferator-activated receptor α (PPARα). CONCLUSION These findings demonstrate that GDF11 can treat diabetic cardiomyopathy by alleviating pyroptosis and reveal the role of the PPARα-GDF11-ASC pathway in DCM, providing ideas for new strategies for cardioprotection.
Collapse
Affiliation(s)
- Jing Zhang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Shandong University, 250012, Jinan, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, 250012, Jinan, Shandong, China
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, 250012, Jinan, Shandong, China
- Shandong Technology Innovation Center for Reproductive Health, 250012, Jinan, Shandong, China
- Shandong Provincial Clinical Research Center for Reproductive Health, 250012, Jinan, Shandong, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 250012, Jinan, Shandong, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No. 2021RU001), 250012, Jinan, Shandong, China
| | - Guolong Wang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Shandong University, 250012, Jinan, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, 250012, Jinan, Shandong, China
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, 250012, Jinan, Shandong, China
- Shandong Technology Innovation Center for Reproductive Health, 250012, Jinan, Shandong, China
- Shandong Provincial Clinical Research Center for Reproductive Health, 250012, Jinan, Shandong, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 250012, Jinan, Shandong, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No. 2021RU001), 250012, Jinan, Shandong, China
| | - Yuxuan Shi
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Shandong University, 250012, Jinan, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, 250012, Jinan, Shandong, China
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, 250012, Jinan, Shandong, China
- Shandong Technology Innovation Center for Reproductive Health, 250012, Jinan, Shandong, China
- Shandong Provincial Clinical Research Center for Reproductive Health, 250012, Jinan, Shandong, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 250012, Jinan, Shandong, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No. 2021RU001), 250012, Jinan, Shandong, China
| | - Xin Liu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Shandong University, 250012, Jinan, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, 250012, Jinan, Shandong, China
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, 250012, Jinan, Shandong, China
- Shandong Technology Innovation Center for Reproductive Health, 250012, Jinan, Shandong, China
- Shandong Provincial Clinical Research Center for Reproductive Health, 250012, Jinan, Shandong, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 250012, Jinan, Shandong, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No. 2021RU001), 250012, Jinan, Shandong, China
| | - Shuang Liu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Shandong University, 250012, Jinan, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, 250012, Jinan, Shandong, China
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, 250012, Jinan, Shandong, China
- Shandong Technology Innovation Center for Reproductive Health, 250012, Jinan, Shandong, China
- Shandong Provincial Clinical Research Center for Reproductive Health, 250012, Jinan, Shandong, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 250012, Jinan, Shandong, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No. 2021RU001), 250012, Jinan, Shandong, China
| | - Wendi Chen
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Shandong University, 250012, Jinan, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, 250012, Jinan, Shandong, China
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, 250012, Jinan, Shandong, China
- Shandong Technology Innovation Center for Reproductive Health, 250012, Jinan, Shandong, China
- Shandong Provincial Clinical Research Center for Reproductive Health, 250012, Jinan, Shandong, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 250012, Jinan, Shandong, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No. 2021RU001), 250012, Jinan, Shandong, China
| | - Yunna Ning
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Shandong University, 250012, Jinan, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, 250012, Jinan, Shandong, China
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, 250012, Jinan, Shandong, China
- Shandong Technology Innovation Center for Reproductive Health, 250012, Jinan, Shandong, China
- Shandong Provincial Clinical Research Center for Reproductive Health, 250012, Jinan, Shandong, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 250012, Jinan, Shandong, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No. 2021RU001), 250012, Jinan, Shandong, China
| | - Yongzhi Cao
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Shandong University, 250012, Jinan, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, 250012, Jinan, Shandong, China
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, 250012, Jinan, Shandong, China
- Shandong Technology Innovation Center for Reproductive Health, 250012, Jinan, Shandong, China
- Shandong Provincial Clinical Research Center for Reproductive Health, 250012, Jinan, Shandong, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 250012, Jinan, Shandong, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No. 2021RU001), 250012, Jinan, Shandong, China
| | - Yueran Zhao
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Shandong University, 250012, Jinan, Shandong, China.
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, 250012, Jinan, Shandong, China.
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, 250012, Jinan, Shandong, China.
- Shandong Technology Innovation Center for Reproductive Health, 250012, Jinan, Shandong, China.
- Shandong Provincial Clinical Research Center for Reproductive Health, 250012, Jinan, Shandong, China.
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 250012, Jinan, Shandong, China.
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No. 2021RU001), 250012, Jinan, Shandong, China.
| | - Ming Li
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Shandong University, 250012, Jinan, Shandong, China.
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, 250012, Jinan, Shandong, China.
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, 250012, Jinan, Shandong, China.
- Shandong Technology Innovation Center for Reproductive Health, 250012, Jinan, Shandong, China.
- Shandong Provincial Clinical Research Center for Reproductive Health, 250012, Jinan, Shandong, China.
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 250012, Jinan, Shandong, China.
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No. 2021RU001), 250012, Jinan, Shandong, China.
| |
Collapse
|
3
|
Zeng Y, Li Y, Jiang W, Hou N. Molecular mechanisms of metabolic dysregulation in diabetic cardiomyopathy. Front Cardiovasc Med 2024; 11:1375400. [PMID: 38596692 PMCID: PMC11003275 DOI: 10.3389/fcvm.2024.1375400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 03/08/2024] [Indexed: 04/11/2024] Open
Abstract
Diabetic cardiomyopathy (DCM), one of the most serious complications of diabetes mellitus, has become recognized as a cardiometabolic disease. In normoxic conditions, the majority of the ATP production (>95%) required for heart beating comes from mitochondrial oxidative phosphorylation of fatty acids (FAs) and glucose, with the remaining portion coming from a variety of sources, including fructose, lactate, ketone bodies (KB) and branched chain amino acids (BCAA). Increased FA intake and decreased utilization of glucose and lactic acid were observed in the diabetic hearts of animal models and diabetic patients. Moreover, the polyol pathway is activated, and fructose metabolism is enhanced. The use of ketones as energy sources in human diabetic hearts also increases significantly. Furthermore, elevated BCAA levels and impaired BCAA metabolism were observed in the hearts of diabetic mice and patients. The shift in energy substrate preference in diabetic hearts results in increased oxygen consumption and impaired oxidative phosphorylation, leading to diabetic cardiomyopathy. However, the precise mechanisms by which impaired myocardial metabolic alterations result in diabetes mellitus cardiac disease are not fully understood. Therefore, this review focuses on the molecular mechanisms involved in alterations of myocardial energy metabolism. It not only adds more molecular targets for the diagnosis and treatment, but also provides an experimental foundation for screening novel therapeutic agents for diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Yue Zeng
- Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- Department of Pharmacy, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, China
| | - Yilang Li
- Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- Department of Pharmacy, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, China
| | - Wenyue Jiang
- Department of Pharmacy, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, China
| | - Ning Hou
- Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- Department of Pharmacy, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, China
| |
Collapse
|
4
|
Guo D, Zhang M, Qi B, Peng T, Liu M, Li Z, Fu F, Guo Y, Li C, Wang Y, Hu L, Li Y. Lipid overload-induced RTN3 activation leads to cardiac dysfunction by promoting lipid droplet biogenesis. Cell Death Differ 2024; 31:292-308. [PMID: 38017147 PMCID: PMC10923887 DOI: 10.1038/s41418-023-01241-x] [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: 03/07/2023] [Revised: 11/07/2023] [Accepted: 11/14/2023] [Indexed: 11/30/2023] Open
Abstract
Lipid droplet (LD) accumulation is a notable feature of obesity-induced cardiomyopathy, while underlying mechanism remains poorly understood. Here we show that mice fed with high-fat diet (HFD) exhibited significantly increase in cardiac LD and RTN3 expression, accompanied by cardiac function impairment. Multiple loss- and gain-of function experiments indicate that RTN3 is critical to HFD-induced cardiac LD accumulation. Mechanistically, RTN3 directly bonds with fatty acid binding protein 5 (FABP5) to facilitate the directed transport of fatty acids to endoplasmic reticulum, thereby promoting LD biogenesis in a diacylglycerol acyltransferase 2 dependent way. Moreover, lipid overload-induced RTN3 upregulation is due to increased expression of CCAAT/enhancer binding protein α (C/EBPα), which positively regulates RTN3 transcription by binding to its promoter region. Notably, above findings were verified in the myocardium of obese patients. Our findings suggest that manipulating LD biogenesis by modulating RTN3 may be a potential strategy for treating cardiac dysfunction in obese patients.
Collapse
Affiliation(s)
- Dong Guo
- Department of Cardiology, Tangdu Hospital, Airforce Medical University, Xi'an, 710032, China
| | - Mingming Zhang
- Department of Cardiology, Tangdu Hospital, Airforce Medical University, Xi'an, 710032, China
| | - Bingchao Qi
- Department of Cardiology, Tangdu Hospital, Airforce Medical University, Xi'an, 710032, China
| | - Tingwei Peng
- Department of Cardiology, Tangdu Hospital, Airforce Medical University, Xi'an, 710032, China
| | - Mingchuan Liu
- Department of Cardiology, Tangdu Hospital, Airforce Medical University, Xi'an, 710032, China
| | - Zhelong Li
- Department of Ultrasound Diagnostics, Tangdu Hospital, Airforce Medical University, Xi'an, 710032, China
| | - Feng Fu
- Department of Physiology and Pathophysiology, Airforce Medical University, Xi'an, 710032, China
| | - Yanjie Guo
- Department of Cardiology, Xi'an International Medical Center Hospital, Xi'an, 710100, China
| | - Congye Li
- Department of Cardiology, Xijing Hospital, Airforce Medical University, 710032, Xi'an, China
| | - Ying Wang
- Department of Cardiology, Tangdu Hospital, Airforce Medical University, Xi'an, 710032, China
| | - Lang Hu
- Department of Cardiology, Tangdu Hospital, Airforce Medical University, Xi'an, 710032, China.
| | - Yan Li
- Department of Cardiology, Tangdu Hospital, Airforce Medical University, Xi'an, 710032, China.
| |
Collapse
|
5
|
Xie SY, Liu SQ, Zhang T, Shi WK, Xing Y, Fang WX, Zhang M, Chen MY, Xu SC, Fan MQ, Li LL, Zhang H, Zhao N, Zeng ZX, Chen S, Zeng XF, Deng W, Tang QZ. USP28 Serves as a Key Suppressor of Mitochondrial Morphofunctional Defects and Cardiac Dysfunction in the Diabetic Heart. Circulation 2024; 149:684-706. [PMID: 37994595 DOI: 10.1161/circulationaha.123.065603] [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: 05/17/2023] [Accepted: 10/30/2023] [Indexed: 11/24/2023]
Abstract
BACKGROUND The majority of people with diabetes are susceptible to cardiac dysfunction and heart failure, and conventional drug therapy cannot correct diabetic cardiomyopathy progression. Herein, we assessed the potential role and therapeutic value of USP28 (ubiquitin-specific protease 28) on the metabolic vulnerability of diabetic cardiomyopathy. METHODS The type 2 diabetes mouse model was established using db/db leptin receptor-deficient mice and high-fat diet/streptozotocin-induced mice. Cardiac-specific knockout of USP28 in the db/db background mice was generated by crossbreeding db/m and Myh6-Cre+/USP28fl/fl mice. Recombinant adeno-associated virus serotype 9 carrying USP28 under cardiac troponin T promoter was injected into db/db mice. High glucose plus palmitic acid-incubated neonatal rat ventricular myocytes and human induced pluripotent stem cell-derived cardiomyocytes were used to imitate diabetic cardiomyopathy in vitro. The molecular mechanism was explored through RNA sequencing, immunoprecipitation and mass spectrometry analysis, protein pull-down, chromatin immunoprecipitation sequencing, and chromatin immunoprecipitation assay. RESULTS Microarray profiling of the UPS (ubiquitin-proteasome system) on the basis of db/db mouse hearts and diabetic patients' hearts demonstrated that the diabetic ventricle presented a significant reduction in USP28 expression. Diabetic Myh6-Cre+/USP28fl/fl mice exhibited more severe progressive cardiac dysfunction, lipid accumulation, and mitochondrial disarrangement, compared with their controls. On the other hand, USP28 overexpression improved systolic and diastolic dysfunction and ameliorated cardiac hypertrophy and fibrosis in the diabetic heart. Adeno-associated virus serotype 9-USP28 diabetic mice also exhibited less lipid storage, reduced reactive oxygen species formation, and mitochondrial impairment in heart tissues than adeno-associated virus serotype 9-null diabetic mice. As a result, USP28 overexpression attenuated cardiac remodeling and dysfunction, lipid accumulation, and mitochondrial impairment in high-fat diet/streptozotocin-induced type 2 diabetes mice. These results were also confirmed in neonatal rat ventricular myocytes and human induced pluripotent stem cell-derived cardiomyocytes. RNA sequencing, immunoprecipitation and mass spectrometry analysis, chromatin immunoprecipitation assays, chromatin immunoprecipitation sequencing, and protein pull-down assay mechanistically revealed that USP28 directly interacted with PPARα (peroxisome proliferator-activated receptor α), deubiquitinating and stabilizing PPARα (Lys152) to promote Mfn2 (mitofusin 2) transcription, thereby impeding mitochondrial morphofunctional defects. However, such cardioprotective benefits of USP28 were largely abrogated in db/db mice with PPARα deletion and conditional loss-of-function of Mfn2. CONCLUSIONS Our findings provide a USP28-modulated mitochondria homeostasis mechanism that involves the PPARα-Mfn2 axis in diabetic hearts, suggesting that USP28 activation or adeno-associated virus therapy targeting USP28 represents a potential therapeutic strategy for diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Sai-Yang Xie
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China (S.-y.X, S.-q.L., T.Z., W.-k.S., Y.X., W.-x.F., M.Z., M.-Y.C., L.-l.L., H.Z., N.Z., W.D., Q.z.T.)
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, P.R. China (S.-y.X, S.-q.L., T.Z., W.-k.S., Y.X., W.-x.F., M.Z., M.-Y.C., L.-l.L., H.Z., N.Z., W.D., Q.z.T.)
| | - Shi-Qiang Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China (S.-y.X, S.-q.L., T.Z., W.-k.S., Y.X., W.-x.F., M.Z., M.-Y.C., L.-l.L., H.Z., N.Z., W.D., Q.z.T.)
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, P.R. China (S.-y.X, S.-q.L., T.Z., W.-k.S., Y.X., W.-x.F., M.Z., M.-Y.C., L.-l.L., H.Z., N.Z., W.D., Q.z.T.)
| | - Tong Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China (S.-y.X, S.-q.L., T.Z., W.-k.S., Y.X., W.-x.F., M.Z., M.-Y.C., L.-l.L., H.Z., N.Z., W.D., Q.z.T.)
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, P.R. China (S.-y.X, S.-q.L., T.Z., W.-k.S., Y.X., W.-x.F., M.Z., M.-Y.C., L.-l.L., H.Z., N.Z., W.D., Q.z.T.)
| | - Wen-Ke Shi
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China (S.-y.X, S.-q.L., T.Z., W.-k.S., Y.X., W.-x.F., M.Z., M.-Y.C., L.-l.L., H.Z., N.Z., W.D., Q.z.T.)
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, P.R. China (S.-y.X, S.-q.L., T.Z., W.-k.S., Y.X., W.-x.F., M.Z., M.-Y.C., L.-l.L., H.Z., N.Z., W.D., Q.z.T.)
| | - Yun Xing
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China (S.-y.X, S.-q.L., T.Z., W.-k.S., Y.X., W.-x.F., M.Z., M.-Y.C., L.-l.L., H.Z., N.Z., W.D., Q.z.T.)
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, P.R. China (S.-y.X, S.-q.L., T.Z., W.-k.S., Y.X., W.-x.F., M.Z., M.-Y.C., L.-l.L., H.Z., N.Z., W.D., Q.z.T.)
| | - Wen-Xi Fang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China (S.-y.X, S.-q.L., T.Z., W.-k.S., Y.X., W.-x.F., M.Z., M.-Y.C., L.-l.L., H.Z., N.Z., W.D., Q.z.T.)
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, P.R. China (S.-y.X, S.-q.L., T.Z., W.-k.S., Y.X., W.-x.F., M.Z., M.-Y.C., L.-l.L., H.Z., N.Z., W.D., Q.z.T.)
| | - Min Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China (S.-y.X, S.-q.L., T.Z., W.-k.S., Y.X., W.-x.F., M.Z., M.-Y.C., L.-l.L., H.Z., N.Z., W.D., Q.z.T.)
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, P.R. China (S.-y.X, S.-q.L., T.Z., W.-k.S., Y.X., W.-x.F., M.Z., M.-Y.C., L.-l.L., H.Z., N.Z., W.D., Q.z.T.)
| | - Meng-Ya Chen
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China (S.-y.X, S.-q.L., T.Z., W.-k.S., Y.X., W.-x.F., M.Z., M.-Y.C., L.-l.L., H.Z., N.Z., W.D., Q.z.T.)
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, P.R. China (S.-y.X, S.-q.L., T.Z., W.-k.S., Y.X., W.-x.F., M.Z., M.-Y.C., L.-l.L., H.Z., N.Z., W.D., Q.z.T.)
| | - Si-Chi Xu
- Department of Cardiology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, P.R. China (S.-c.X.)
| | - Meng-Qi Fan
- College of Life Sciences, Wuhan University, P.R. China (M.-q.F.)
| | - Lan-Lan Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China (S.-y.X, S.-q.L., T.Z., W.-k.S., Y.X., W.-x.F., M.Z., M.-Y.C., L.-l.L., H.Z., N.Z., W.D., Q.z.T.)
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, P.R. China (S.-y.X, S.-q.L., T.Z., W.-k.S., Y.X., W.-x.F., M.Z., M.-Y.C., L.-l.L., H.Z., N.Z., W.D., Q.z.T.)
| | - Heng Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China (S.-y.X, S.-q.L., T.Z., W.-k.S., Y.X., W.-x.F., M.Z., M.-Y.C., L.-l.L., H.Z., N.Z., W.D., Q.z.T.)
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, P.R. China (S.-y.X, S.-q.L., T.Z., W.-k.S., Y.X., W.-x.F., M.Z., M.-Y.C., L.-l.L., H.Z., N.Z., W.D., Q.z.T.)
| | - Nan Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China (S.-y.X, S.-q.L., T.Z., W.-k.S., Y.X., W.-x.F., M.Z., M.-Y.C., L.-l.L., H.Z., N.Z., W.D., Q.z.T.)
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, P.R. China (S.-y.X, S.-q.L., T.Z., W.-k.S., Y.X., W.-x.F., M.Z., M.-Y.C., L.-l.L., H.Z., N.Z., W.D., Q.z.T.)
| | - Zhao-Xiang Zeng
- Department of Vascular Surgery, Shanghai General Hospital, Shanghai Jiaotong University, P.R. China (Z.-x.Z)
- Department of Cardiac Surgery, Changhai Hospital, Navy Medical University, Shanghai, P.R. China (Z.-x.Z)
| | - Si Chen
- Cardiovascular Research Institute of Wuhan University, P.R. China (S.C., X.-f.Z.)
| | - Xiao-Feng Zeng
- Cardiovascular Research Institute of Wuhan University, P.R. China (S.C., X.-f.Z.)
| | - Wei Deng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China (S.-y.X, S.-q.L., T.Z., W.-k.S., Y.X., W.-x.F., M.Z., M.-Y.C., L.-l.L., H.Z., N.Z., W.D., Q.z.T.)
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, P.R. China (S.-y.X, S.-q.L., T.Z., W.-k.S., Y.X., W.-x.F., M.Z., M.-Y.C., L.-l.L., H.Z., N.Z., W.D., Q.z.T.)
| | - Qi-Zhu Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China (S.-y.X, S.-q.L., T.Z., W.-k.S., Y.X., W.-x.F., M.Z., M.-Y.C., L.-l.L., H.Z., N.Z., W.D., Q.z.T.)
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, P.R. China (S.-y.X, S.-q.L., T.Z., W.-k.S., Y.X., W.-x.F., M.Z., M.-Y.C., L.-l.L., H.Z., N.Z., W.D., Q.z.T.)
| |
Collapse
|
6
|
Xu W, Billon C, Li H, Wilderman A, Qi L, Graves A, Rideb JRDC, Zhao Y, Hayes M, Yu K, Losby M, Hampton CS, Adeyemi CM, Hong SJ, Nasiotis E, Fu C, Oh TG, Fan W, Downes M, Welch RD, Evans RM, Milosavljevic A, Walker JK, Jensen BC, Pei L, Burris T, Zhang L. Novel Pan-ERR Agonists Ameliorate Heart Failure Through Enhancing Cardiac Fatty Acid Metabolism and Mitochondrial Function. Circulation 2024; 149:227-250. [PMID: 37961903 PMCID: PMC10842599 DOI: 10.1161/circulationaha.123.066542] [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: 08/09/2023] [Accepted: 10/20/2023] [Indexed: 11/15/2023]
Abstract
BACKGROUND Cardiac metabolic dysfunction is a hallmark of heart failure (HF). Estrogen-related receptors ERRα and ERRγ are essential regulators of cardiac metabolism. Therefore, activation of ERR could be a potential therapeutic intervention for HF. However, in vivo studies demonstrating the potential usefulness of ERR agonist for HF treatment are lacking, because compounds with pharmacokinetics appropriate for in vivo use have not been available. METHODS Using a structure-based design approach, we designed and synthesized 2 structurally distinct pan-ERR agonists, SLU-PP-332 and SLU-PP-915. We investigated the effect of ERR agonist on cardiac function in a pressure overload-induced HF model in vivo. We conducted comprehensive functional, multi-omics (RNA sequencing and metabolomics studies), and genetic dependency studies both in vivo and in vitro to dissect the molecular mechanism, ERR isoform dependency, and target specificity. RESULTS Both SLU-PP-332 and SLU-PP-915 significantly improved ejection fraction, ameliorated fibrosis, and increased survival associated with pressure overload-induced HF without affecting cardiac hypertrophy. A broad spectrum of metabolic genes was transcriptionally activated by ERR agonists, particularly genes involved in fatty acid metabolism and mitochondrial function. Metabolomics analysis showed substantial normalization of metabolic profiles in fatty acid/lipid and tricarboxylic acid/oxidative phosphorylation metabolites in the mouse heart with 6-week pressure overload. ERR agonists increase mitochondria oxidative capacity and fatty acid use in vitro and in vivo. Using both in vitro and in vivo genetic dependency experiments, we show that ERRγ is the main mediator of ERR agonism-induced transcriptional regulation and cardioprotection and definitively demonstrated target specificity. ERR agonism also led to downregulation of cell cycle and development pathways, which was partially mediated by E2F1 in cardiomyocytes. CONCLUSIONS ERR agonists maintain oxidative metabolism, which confers cardiac protection against pressure overload-induced HF in vivo. Our results provide direct pharmacologic evidence supporting the further development of ERR agonists as novel HF therapeutics.
Collapse
Affiliation(s)
- Weiyi Xu
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, TX (W.X., H.L., A.W., L.Q., A.G., J.R.D.C.R., Y.Z., K.Y., M.L., E.N., A.M., L.Z.)
| | - Cyrielle Billon
- Department of Pharmaceutical and Administrative Sciences, University of Health Sciences and Pharmacy, St Louis, MO (C.B., M.H., T.B.)
- Center for Clinical Pharmacology, St Louis College of Pharmacy, Washington University School of Medicine, St Louis, MO (C.B., M.H., T.B.)
| | - Hui Li
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, TX (W.X., H.L., A.W., L.Q., A.G., J.R.D.C.R., Y.Z., K.Y., M.L., E.N., A.M., L.Z.)
| | - Andrea Wilderman
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, TX (W.X., H.L., A.W., L.Q., A.G., J.R.D.C.R., Y.Z., K.Y., M.L., E.N., A.M., L.Z.)
| | - Lei Qi
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, TX (W.X., H.L., A.W., L.Q., A.G., J.R.D.C.R., Y.Z., K.Y., M.L., E.N., A.M., L.Z.)
| | - Andrea Graves
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, TX (W.X., H.L., A.W., L.Q., A.G., J.R.D.C.R., Y.Z., K.Y., M.L., E.N., A.M., L.Z.)
| | - Jernie Rae Dela Cruz Rideb
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, TX (W.X., H.L., A.W., L.Q., A.G., J.R.D.C.R., Y.Z., K.Y., M.L., E.N., A.M., L.Z.)
| | - Yuanbiao Zhao
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, TX (W.X., H.L., A.W., L.Q., A.G., J.R.D.C.R., Y.Z., K.Y., M.L., E.N., A.M., L.Z.)
| | - Matthew Hayes
- Department of Pharmaceutical and Administrative Sciences, University of Health Sciences and Pharmacy, St Louis, MO (C.B., M.H., T.B.)
- Center for Clinical Pharmacology, St Louis College of Pharmacy, Washington University School of Medicine, St Louis, MO (C.B., M.H., T.B.)
| | - Keyang Yu
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, TX (W.X., H.L., A.W., L.Q., A.G., J.R.D.C.R., Y.Z., K.Y., M.L., E.N., A.M., L.Z.)
| | - McKenna Losby
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, TX (W.X., H.L., A.W., L.Q., A.G., J.R.D.C.R., Y.Z., K.Y., M.L., E.N., A.M., L.Z.)
| | - Carissa S Hampton
- Department of Pharmacology and Physiology, St Louis University School of Medicine, MO (C.S.H., C.M.A., J.K.W.)
| | - Christiana M Adeyemi
- Department of Pharmacology and Physiology, St Louis University School of Medicine, MO (C.S.H., C.M.A., J.K.W.)
| | - Seok Jae Hong
- McAllister Heart Institute (S.J.H., B.C.J.), University of North Carolina, Chapel Hill
| | - Eleni Nasiotis
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, TX (W.X., H.L., A.W., L.Q., A.G., J.R.D.C.R., Y.Z., K.Y., M.L., E.N., A.M., L.Z.)
| | - Chen Fu
- Department of Psychiatry, University of Massachusetts Chan Medical School, Worcester, MA (C.F.)
- University Hospitals Cleveland Medical Center, OH (C.F.)
| | - Tae Gyu Oh
- Howard Hughes Medical Institute, Salk Institute for Biological Studies, La Jolla, CA (T.G.O., W.F., M.D., R.M.E.)
| | - Weiwei Fan
- Howard Hughes Medical Institute, Salk Institute for Biological Studies, La Jolla, CA (T.G.O., W.F., M.D., R.M.E.)
| | - Michael Downes
- Howard Hughes Medical Institute, Salk Institute for Biological Studies, La Jolla, CA (T.G.O., W.F., M.D., R.M.E.)
| | - Ryan D Welch
- Biology and Chemistry Department, Blackburn College, Carlinville, IL (R.D.W.)
| | - Ronald M Evans
- Howard Hughes Medical Institute, Salk Institute for Biological Studies, La Jolla, CA (T.G.O., W.F., M.D., R.M.E.)
| | - Aleksandar Milosavljevic
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, TX (W.X., H.L., A.W., L.Q., A.G., J.R.D.C.R., Y.Z., K.Y., M.L., E.N., A.M., L.Z.)
| | - John K Walker
- Department of Pharmacology and Physiology, St Louis University School of Medicine, MO (C.S.H., C.M.A., J.K.W.)
| | - Brian C Jensen
- McAllister Heart Institute (S.J.H., B.C.J.), University of North Carolina, Chapel Hill
- Department of Medicine, Division of Cardiology (B.C.J.), University of North Carolina, Chapel Hill
| | - Liming Pei
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, and University of Pennsylvania, Philadelphia (L.P.)
| | - Thomas Burris
- Department of Pharmaceutical and Administrative Sciences, University of Health Sciences and Pharmacy, St Louis, MO (C.B., M.H., T.B.)
- Center for Clinical Pharmacology, St Louis College of Pharmacy, Washington University School of Medicine, St Louis, MO (C.B., M.H., T.B.)
| | - Lilei Zhang
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, TX (W.X., H.L., A.W., L.Q., A.G., J.R.D.C.R., Y.Z., K.Y., M.L., E.N., A.M., L.Z.)
| |
Collapse
|
7
|
Walker MA, Chen H, Yadav A, Ritterhoff J, Villet O, McMillen T, Wang Y, Purcell H, Djukovic D, Raftery D, Isoherranen N, Tian R. Raising NAD + Level Stimulates Short-Chain Dehydrogenase/Reductase Proteins to Alleviate Heart Failure Independent of Mitochondrial Protein Deacetylation. Circulation 2023; 148:2038-2057. [PMID: 37965787 PMCID: PMC10842390 DOI: 10.1161/circulationaha.123.066039] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 10/04/2023] [Indexed: 11/16/2023]
Abstract
BACKGROUND Strategies to increase cellular NAD+ (oxidized nicotinamide adenine dinucleotide) level have prevented cardiac dysfunction in multiple models of heart failure, but molecular mechanisms remain unclear. Little is known about the benefits of NAD+-based therapies in failing hearts after the symptoms of heart failure have appeared. Most pretreatment regimens suggested mechanisms involving activation of sirtuin, especially Sirt3 (sirtuin 3), and mitochondrial protein acetylation. METHODS We induced cardiac dysfunction by pressure overload in SIRT3-deficient (knockout) mice and compared their response with nicotinamide riboside chloride treatment with wild-type mice. To model a therapeutic approach, we initiated the treatment in mice with established cardiac dysfunction. RESULTS We found nicotinamide riboside chloride improved mitochondrial function and blunted heart failure progression. Similar benefits were observed in wild-type and knockout mice. Boosting NAD+ level improved the function of NAD(H) redox-sensitive SDR (short-chain dehydrogenase/reductase) family proteins. Upregulation of Mrpp2 (mitochondrial ribonuclease P protein 2), a multifunctional SDR protein and a subunit of mitochondrial ribonuclease P, improves mitochondrial DNA transcripts processing and electron transport chain function. Activation of SDRs in the retinol metabolism pathway stimulates RXRα (retinoid X receptor α)/PPARα (proliferator-activated receptor α) signaling and restores mitochondrial oxidative metabolism. Downregulation of Mrpp2 and impaired mitochondrial ribonuclease P were found in human failing hearts, suggesting a shared mechanism of defective mitochondrial biogenesis in mouse and human heart failure. CONCLUSIONS These findings identify SDR proteins as important regulators of mitochondrial function and molecular targets of NAD+-based therapy. Furthermore, the benefit is observed regardless of Sirt3-mediated mitochondrial protein deacetylation, a widely held mechanism for NAD+-based therapy for heart failure. The data also show that NAD+-based therapy can be useful in pre-existing heart failure.
Collapse
Affiliation(s)
- Matthew A. Walker
- Mitochondria and Metabolism Center, Department of
Anesthesiology & Pain Medicine, University of Washington, Seattle, WA
98109
| | - Hongye Chen
- Mitochondria and Metabolism Center, Department of
Anesthesiology & Pain Medicine, University of Washington, Seattle, WA
98109
| | - Aprajita Yadav
- Department of Pharmaceutics, School of Pharmacy, University
of Washington, Seattle, WA 98195
| | - Julia Ritterhoff
- Mitochondria and Metabolism Center, Department of
Anesthesiology & Pain Medicine, University of Washington, Seattle, WA
98109
| | - Outi Villet
- Mitochondria and Metabolism Center, Department of
Anesthesiology & Pain Medicine, University of Washington, Seattle, WA
98109
| | - Tim McMillen
- Mitochondria and Metabolism Center, Department of
Anesthesiology & Pain Medicine, University of Washington, Seattle, WA
98109
| | - Yuliang Wang
- Department of Computer Science & Engineering,
University of Washington, Seattle, WA 98195
| | - Hayley Purcell
- Mitochondria and Metabolism Center, Department of
Anesthesiology & Pain Medicine, University of Washington, Seattle, WA
98109
| | - Danijel Djukovic
- Mitochondria and Metabolism Center, Department of
Anesthesiology & Pain Medicine, University of Washington, Seattle, WA
98109
| | - Daniel Raftery
- Mitochondria and Metabolism Center, Department of
Anesthesiology & Pain Medicine, University of Washington, Seattle, WA
98109
| | - Nina Isoherranen
- Department of Pharmaceutics, School of Pharmacy, University
of Washington, Seattle, WA 98195
| | - Rong Tian
- Mitochondria and Metabolism Center, Department of
Anesthesiology & Pain Medicine, University of Washington, Seattle, WA
98109
| |
Collapse
|
8
|
Su S, Ji X, Li T, Teng Y, Wang B, Han X, Zhao M. The changes of cardiac energy metabolism with sodium-glucose transporter 2 inhibitor therapy. Front Cardiovasc Med 2023; 10:1291450. [PMID: 38124893 PMCID: PMC10731052 DOI: 10.3389/fcvm.2023.1291450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Accepted: 11/13/2023] [Indexed: 12/23/2023] Open
Abstract
Background/aims To investigate the specific effects of s odium-glucose transporter 2 inhibitor (SGLT2i) on cardiac energy metabolism. Methods A systematic literature search was conducted in eight databases. The retrieved studies were screened according to the inclusion and exclusion criteria, and relevant information was extracted according to the purpose of the study. Two researchers independently screened the studies, extracted information, and assessed article quality. Results The results of the 34 included studies (including 10 clinical and 24 animal studies) showed that SGLT2i inhibited cardiac glucose uptake and glycolysis, but promoted fatty acid (FA) metabolism in most disease states. SGLT2i upregulated ketone metabolism, improved the structure and functions of myocardial mitochondria, alleviated oxidative stress of cardiomyocytes in all literatures. SGLT2i increased cardiac glucose oxidation in diabetes mellitus (DM) and cardiac FA metabolism in heart failure (HF). However, the regulatory effects of SGLT2i on cardiac FA metabolism in DM and cardiac glucose oxidation in HF varied with disease types, stages, and intervention duration of SGLT2i. Conclusion SGLT2i improved the efficiency of cardiac energy production by regulating FA, glucose and ketone metabolism, improving mitochondria structure and functions, and decreasing oxidative stress of cardiomyocytes under pathological conditions. Thus, SGLT2i is deemed to exert a benign regulatory effect on cardiac metabolic disorders in various diseases. Systematic review registration https://www.crd.york.ac.uk/, PROSPERO (CRD42023484295).
Collapse
Affiliation(s)
- Sha Su
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Xiang Ji
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Tong Li
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Yu Teng
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Baofu Wang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Xiaowan Han
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
- Department of Cardiology, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Mingjing Zhao
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
9
|
Wang H, Liu X, Zhou Q, Liu L, Jia Z, Qi Y, Xu F, Zhang Y. Current status and emerging trends of cardiac metabolism from the past 20 years: A bibliometric study. Heliyon 2023; 9:e21952. [PMID: 38045208 PMCID: PMC10692779 DOI: 10.1016/j.heliyon.2023.e21952] [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: 04/21/2023] [Revised: 10/30/2023] [Accepted: 11/01/2023] [Indexed: 12/05/2023] Open
Abstract
Background Abnormal cardiac metabolism is a key factor in the development of cardiovascular diseases. Consequently, there has been considerable emphasis on researching and developing drugs that regulate metabolism. This study employed bibliometric methods to comprehensively and objectively analyze the relevant literature, offering insights into the knowledge dynamics in this field. Methods The data source for this study was the Web of Science Core Collection (WoSCC), from which the collected data were imported into bibliometric software for analysis. Results The United States was the leading contributor, accounting for 38.33 % of publications. The University of Washington and Damian J. Tyler were the most active institution and author, respectively. The American Journal of Physiology-Heart and Circulatory Physiology, Journal of Molecular and Cellular Cardiology, Cardiovascular Research, Circulation Research, and American Journal of Physiology-Endocrinology and Metabolism were highly influential journals that published numerous high-quality articles on cardiac metabolism. Common keywords in this research area included heart failure, insulin resistance, skeletal muscle, mitochondria, as well as topic words such as cardiac metabolism, fatty acid oxidation, glucose metabolism, and myocardial metabolism. Co-citation analysis has shown that research on heart failure and in vitro modeling of cardiovascular disease has gained prominence in recent years and making it a research hotspot. Conclusion Research on cardiac metabolism is steadily growing, with a specific focus on heart failure and the interplay between mitochondrial dysfunction, insulin resistance, and cardiac metabolism. An emerging trend in this field involves the enhancement of maturation in human induced pluripotent stem cell-derived cardiomyocyte (hiPSC-CM) through the manipulation of cardiac metabolism.
Collapse
Affiliation(s)
- Hongqin Wang
- Institute of Geriatric, Xiyuan Hospital, Beijing, China
- China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaolin Liu
- Institute of Geriatric, Xiyuan Hospital, Beijing, China
- China Academy of Chinese Medical Sciences, Beijing, China
| | - Qingbing Zhou
- Institute of Geriatric, Xiyuan Hospital, Beijing, China
- China Academy of Chinese Medical Sciences, Beijing, China
| | - Li Liu
- Institute of Geriatric, Xiyuan Hospital, Beijing, China
- China Academy of Chinese Medical Sciences, Beijing, China
| | - Zijun Jia
- Institute of Geriatric, Xiyuan Hospital, Beijing, China
- Beijing University of Chinese Medicine, Beijing, China
| | - Yifei Qi
- Institute of Geriatric, Xiyuan Hospital, Beijing, China
- China Academy of Chinese Medical Sciences, Beijing, China
| | - Fengqin Xu
- Institute of Geriatric, Xiyuan Hospital, Beijing, China
- China Academy of Chinese Medical Sciences, Beijing, China
| | - Ying Zhang
- Institute of Geriatric, Xiyuan Hospital, Beijing, China
- China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
10
|
Higuchi S, Wood C, Nasiri RH, Giddla LJ, Molina V, Diarra R, DiPatrizio NV, Kawamura A, Haeusler RA. The 16α-hydroxylated Bile Acid, Pythocholic Acid Decreases Food Intake and Increases Oleoylethanolamide in Male Mice. Endocrinology 2023; 164:bqad116. [PMID: 37490843 PMCID: PMC10407715 DOI: 10.1210/endocr/bqad116] [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: 01/17/2023] [Revised: 07/20/2023] [Accepted: 07/21/2023] [Indexed: 07/27/2023]
Abstract
Modulation of bile acid (BA) structure is a potential strategy for obesity and metabolic disease treatment. BAs act not only as signaling molecules involved in energy expenditure and glucose homeostasis, but also as regulators of food intake. The structure of BAs, particularly the position of the hydroxyl groups of BAs, impacts food intake partly by intestinal effects: (1) modulating the activity of N-acyl phosphatidylethanolamine phospholipase D, which produces the anorexigenic bioactive lipid oleoylethanolamide (OEA) or (2) regulating lipid absorption and the gastric emptying-satiation pathway. We hypothesized that 16α-hydroxylated BAs uniquely regulate food intake because of the long intermeal intervals in snake species in which these BAs are abundant. However, the effects of 16α-hydroxylated BAs in mammals are completely unknown because they are not naturally found in mammals. To test the effect of 16α-hydroxylated BAs on food intake, we isolated the 16α-hydroxylated BA pythocholic acid from ball pythons (Python regius). Pythocholic acid or deoxycholic acid (DCA) was given by oral gavage in mice. DCA is known to increase N-acyl phosphatidylethanolamine phospholipase D activity better than other mammalian BAs. We evaluated food intake, OEA levels, and gastric emptying in mice. We successfully isolated pythocholic acid from ball pythons for experimental use. Pythocholic acid treatment significantly decreased food intake in comparison to DCA treatment, and this was associated with increased jejunal OEA, but resulted in no change in gastric emptying or lipid absorption. The exogenous BA pythocholic acid is a novel regulator of food intake and the satiety signal for OEA in the mouse intestine.
Collapse
Affiliation(s)
- Sei Higuchi
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
- Naomi Berrie Diabetes Center and Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
| | - Courtney Wood
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA 92521, USA
| | - Raidah H Nasiri
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| | - Leela J Giddla
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| | - Valentina Molina
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| | - Rokia Diarra
- Department of Biological Sciences, St. John's University, Queens, NY 11439, USA
| | - Nicholas V DiPatrizio
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA 92521, USA
| | - Akira Kawamura
- Department of Chemistry, Hunter College of CUNY, New York, NY 10065, USA
| | - Rebecca A Haeusler
- Naomi Berrie Diabetes Center and Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
| |
Collapse
|
11
|
Darby JR, Zhang S, Holman SL, Muhlhausler BS, McMillen IC, Morrison JL. Cardiac growth and metabolism of the fetal sheep are not vulnerable to a 10 day increase in fetal glucose and insulin concentrations during late gestation. Heliyon 2023; 9:e18292. [PMID: 37519661 PMCID: PMC10372399 DOI: 10.1016/j.heliyon.2023.e18292] [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: 10/09/2022] [Revised: 07/06/2023] [Accepted: 07/13/2023] [Indexed: 08/01/2023] Open
Abstract
Aims To evaluate the effects of fetal glucose infusion in late gestation on the mRNA expression and protein abundance of molecules involved in the regulation of cardiac growth and metabolism. Main methods Either saline or glucose was infused into fetal sheep from 130 to 140 days (d) gestation (term, 150 d). At 140 d gestation, left ventricle tissue samples were collected. Quantitative real-time RT-PCR and Western blot were used to determine the mRNA expression and protein abundance of key signalling molecules within the left ventricle of the fetal heart. Key findings Although intra-fetal glucose infusion increased fetal plasma glucose and insulin concentrations, there was no change in the expression of molecules within the signalling pathways that regulate proliferation, hypertrophy, apoptosis or fibrosis in the fetal heart. Cardiac Solute carrier family 2 member 1 (SLC2A1) mRNA expression was decreased by glucose infusion. Glucose infusion increased cardiac mRNA expression of both Peroxisome proliferator activated receptor alpha (PPARA) and peroxisome proliferator activated receptor gamma (PPARG). However, there was no change in the mRNA expression of PPAR cofactors or molecules with PPAR response elements. Furthermore, glucose infusion did not impact the protein abundance of the 5 oxidative phosphorylation complexes of the electron transport chain. Significance Despite a 10-day doubling of fetal plasma glucose and insulin concentrations, the present study suggests that within the fetal left ventricle, the mRNA and protein expression of the signalling molecules involved in cardiac growth, development and metabolism are relatively unaffected.
Collapse
Affiliation(s)
| | | | | | | | | | - Janna L. Morrison
- Corresponding author. Australian Research Council Future Fellow, Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, GPO Box 2471, Adelaide, SA, 5001, Australia,
| |
Collapse
|
12
|
Werbner B, Tavakoli-Rouzbehani OM, Fatahian AN, Boudina S. The dynamic interplay between cardiac mitochondrial health and myocardial structural remodeling in metabolic heart disease, aging, and heart failure. THE JOURNAL OF CARDIOVASCULAR AGING 2023; 3:9. [PMID: 36742465 PMCID: PMC9894375 DOI: 10.20517/jca.2022.42] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
This review provides a holistic perspective on the bi-directional relationship between cardiac mitochondrial dysfunction and myocardial structural remodeling in the context of metabolic heart disease, natural cardiac aging, and heart failure. First, a review of the physiologic and molecular drivers of cardiac mitochondrial dysfunction across a range of increasingly prevalent conditions such as metabolic syndrome and cardiac aging is presented, followed by a general review of the mechanisms of mitochondrial quality control (QC) in the heart. Several important mechanisms by which cardiac mitochondrial dysfunction triggers or contributes to structural remodeling of the heart are discussed: accumulated metabolic byproducts, oxidative damage, impaired mitochondrial QC, and mitochondrial-mediated cell death identified as substantial mechanistic contributors to cardiac structural remodeling such as hypertrophy and myocardial fibrosis. Subsequently, the less studied but nevertheless important reverse relationship is explored: the mechanisms by which cardiac structural remodeling feeds back to further alter mitochondrial bioenergetic function. We then provide a condensed pathogenesis of several increasingly important clinical conditions in which these relationships are central: diabetic cardiomyopathy, age-associated declines in cardiac function, and the progression to heart failure, with or without preserved ejection fraction. Finally, we identify promising therapeutic opportunities targeting mitochondrial function in these conditions.
Collapse
Affiliation(s)
- Benjamin Werbner
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT 84112, USA
| | | | - Amir Nima Fatahian
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT 84112, USA
| | - Sihem Boudina
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
13
|
Naasner L, Froese N, Hofmann W, Galuppo P, Werlein C, Shymotiuk I, Szaroszyk M, Erschow S, Amanakis G, Bähre H, Kühnel MP, Jonigk DD, Geffers R, Seifert R, Ricke-Hoch M, Wende AR, Blaner WS, Abel ED, Bauersachs J, Riehle C. Vitamin A preserves cardiac energetic gene expression in a murine model of diet-induced obesity. Am J Physiol Heart Circ Physiol 2022; 323:H1352-H1364. [PMID: 36399384 DOI: 10.1152/ajpheart.00514.2022] [Citation(s) in RCA: 2] [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: 09/07/2022] [Revised: 11/07/2022] [Accepted: 11/08/2022] [Indexed: 11/19/2022]
Abstract
Perturbed vitamin-A metabolism is associated with type 2 diabetes and mitochondrial dysfunction that are pathophysiologically linked to the development of diabetic cardiomyopathy (DCM). However, the mechanism, by which vitamin A might regulate mitochondrial energetics in DCM has previously not been explored. To test the hypothesis that vitamin-A deficiency accelerates the onset of cardiomyopathy in diet-induced obesity (DIO), we subjected mice with lecithin retinol acyltransferase (Lrat) germline deletion, which exhibit impaired vitamin-A stores, to vitamin A-deficient high-fat diet (HFD) feeding. Wild-type mice fed with a vitamin A-sufficient HFD served as controls. Cardiac structure, contractile function, and mitochondrial respiratory capacity were preserved despite vitamin-A deficiency following 20 wk of HFD feeding. Gene profiling by RNA sequencing revealed that vitamin A is required for the expression of genes involved in cardiac fatty acid oxidation, glycolysis, tricarboxylic acid cycle, and mitochondrial oxidative phosphorylation in DIO as expression of these genes was relatively preserved under vitamin A-sufficient HFD conditions. Together, these data identify a transcriptional program, by which vitamin A preserves cardiac energetic gene expression in DIO that might attenuate subsequent onset of mitochondrial and contractile dysfunction.NEW & NOTEWORTHY The relationship between vitamin-A status and the pathogenesis of diabetic cardiomyopathy has not been studied in detail. We assessed cardiac mitochondrial respiratory capacity, contractile function, and gene expression by RNA sequencing in a murine model of combined vitamin-A deficiency and diet-induced obesity. Our study identifies a role for vitamin A in preserving cardiac energetic gene expression that might attenuate subsequent development of mitochondrial and contractile dysfunction in diet-induced obesity.
Collapse
Affiliation(s)
- Lea Naasner
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Natali Froese
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Winfried Hofmann
- Department of Human Genetics, Hannover Medical School, Hannover, Germany
| | - Paolo Galuppo
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | | | - Ivanna Shymotiuk
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Malgorzata Szaroszyk
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Sergej Erschow
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Georgios Amanakis
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Heike Bähre
- Research Core Unit Metabolomics, Institute of Pharmacology, Hannover Medical School, Hannover, Germany
| | - Mark P Kühnel
- Institute of Pathology, Hannover Medical School, Hannover, Germany
- Biomedical Research in End-stage and Obstructive Lung Disease Hannover (BREATH), German Lung Research Centre (DZL), Hannover, Germany
| | - Danny D Jonigk
- Institute of Pathology, Hannover Medical School, Hannover, Germany
- Biomedical Research in End-stage and Obstructive Lung Disease Hannover (BREATH), German Lung Research Centre (DZL), Hannover, Germany
| | - Robert Geffers
- Research Group Genome Analytics, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Roland Seifert
- Research Core Unit Metabolomics, Institute of Pharmacology, Hannover Medical School, Hannover, Germany
| | - Melanie Ricke-Hoch
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Adam R Wende
- Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama
| | - William S Blaner
- Department of Medicine, Institute of Human Nutrition, College of Physicians and Surgeons, Columbia University, New York, New York
| | - E Dale Abel
- Department of Medicine, David Geffen School of Medicine and UCLA Health, Los Angeles, California
| | - Johann Bauersachs
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Christian Riehle
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
14
|
Chen J, You R, Lv Y, Liu H, Yang G. Conjugated linoleic acid regulates adipocyte fatty acid binding protein expression via peroxisome proliferator-activated receptor α signaling pathway and increases intramuscular fat content. Front Nutr 2022; 9:1029864. [PMID: 36523338 PMCID: PMC9745092 DOI: 10.3389/fnut.2022.1029864] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Accepted: 10/19/2022] [Indexed: 06/22/2024] Open
Abstract
Intramuscular fat (IMF) is correlated positively with meat tenderness, juiciness and taste that affected sensory meat quality. Conjugated linoleic acid (CLA) has been extensively researched to increase IMF content in animals, however, the regulatory mechanism remains unclear. Adipocyte fatty acid binding protein (A-FABP) gene has been proposed as candidates for IMF accretion. The purpose of this study is to explore the molecular regulatory pathways of CLA on intramuscular fat deposition. Here, our results by cell lines indicated that CLA treatment promoted the expression of A-FABP through activated the transcription factor of peroxisome proliferator-activated receptor α (PPARα). Moreover, in an animal model, we discovered that dietary supplemental with CLA significantly enhanced IMF deposition by up-regulating the mRNA and protein expression of PPARα and A-FABP in the muscle tissues of mice. In addition, our current study also demonstrated that dietary CLA increased mRNA expression of genes and enzymes involved in fatty acid synthesis and lipid metabolism the muscle tissues of mice. These findings suggest that CLA mainly increases the expression of A-FABP through PPARα signaling pathway and regulates the expression of genes and enzymes related to IMF deposition, thus increasing IMF content. These results contribute to better understanding the molecular mechanism of IMF accretion in animals for the improvement of meat quality.
Collapse
Affiliation(s)
| | | | | | | | - Guoqing Yang
- Laboratory of Animal Gene Engineering, College of Life Sciences, Henan Agricultural University, Zhengzhou, China
| |
Collapse
|
15
|
Fatty Acid Amide Hydrolase Deficiency Is Associated with Deleterious Cardiac Effects after Myocardial Ischemia and Reperfusion in Mice. Int J Mol Sci 2022; 23:ijms232012690. [PMID: 36293543 PMCID: PMC9604059 DOI: 10.3390/ijms232012690] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/11/2022] [Accepted: 10/19/2022] [Indexed: 11/06/2022] Open
Abstract
Ischemic cardiomyopathy leads to inflammation and left ventricular (LV) dysfunction. Animal studies provided evidence for cardioprotective effects of the endocannabinoid system, including cardiomyocyte adaptation, inflammation, and remodeling. Cannabinoid type-2 receptor (CB2) deficiency led to increased apoptosis and infarctions with worsened LV function in ischemic cardiomyopathy. The aim of our study was to investigate a possible cardioprotective effect of endocannabinoid anandamide (AEA) after ischemia and reperfusion (I/R). Therefore, fatty acid amide hydrolase deficient (FAAH)−/− mice were subjected to repetitive, daily, 15 min, left anterior descending artery (LAD) occlusion over 3 and 7 consecutive days. Interestingly, FAAH−/− mice showed stigmata such as enhanced inflammation, cardiomyocyte loss, stronger remodeling, and persistent scar with deteriorated LV function compared to wild-type (WT) littermates. As endocannabinoids also activate PPAR-α (peroxisome proliferator-activated receptor), PPAR-α mediated effects of AEA were eliminated with PPAR-α antagonist GW6471 i.v. in FAAH−/− mice. LV function was assessed using M-mode echocardiography. Immunohistochemical analysis revealed apoptosis, macrophage accumulation, collagen deposition, and remodeling. Hypertrophy was determined by cardiomyocyte area and heart weight/tibia length. Molecular analyses involved Taqman® RT-qPCR and immune cells were analyzed with fluorescence-activated cell sorting (FACS). Most importantly, collagen deposition was reduced to WT levels when FAAH−/− mice were treated with GW6471. Chemokine ligand-2 (CCL2) expression was significantly higher in FAAH−/− mice compared to WT, followed by higher macrophage infiltration in infarcted areas, both being reversed by GW6471 treatment. Besides restoring antioxidative properties and contractile elements, PPAR-α antagonism also reversed hypertrophy and remodeling in FAAH−/− mice. Finally, FAAH−/−-mice showed more substantial downregulation of PPAR-α compared to WT, suggesting a compensatory mechanism as endocannabinoids are also ligands for PPAR-α, and its activation causes lipotoxicity leading to cardiomyocyte apoptosis. Our study gives novel insights into the role of endocannabinoids acting via PPAR-α. We hypothesize that the increase in endocannabinoids may have partially detrimental effects on cardiomyocyte survival due to PPAR-α activation.
Collapse
|
16
|
Cardiomyocyte peroxisome proliferator-activated receptor α is essential for energy metabolism and extracellular matrix homeostasis during pressure overload-induced cardiac remodeling. Acta Pharmacol Sin 2022; 43:1231-1242. [PMID: 34376812 PMCID: PMC9061810 DOI: 10.1038/s41401-021-00743-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 07/11/2021] [Indexed: 01/03/2023] Open
Abstract
Peroxisome proliferator-activated receptor α (PPARα), a ligand-activated nuclear receptor critical for systemic lipid homeostasis, has been shown closely related to cardiac remodeling. However, the roles of cardiomyocyte PPARα in pressure overload-induced cardiac remodeling remains unclear because of lacking a cardiomyocyte-specific Ppara-deficient (PparaΔCM) mouse model. This study aimed to determine the specific role of cardiomyocyte PPARα in transverse aortic constriction (TAC)-induced cardiac remodeling using an inducible PparaΔCM mouse model. PparaΔCM and Pparafl/fl mice were randomly subjected to sham or TAC for 2 weeks. Cardiomyocyte PPARα deficiency accelerated TAC-induced cardiac hypertrophy and fibrosis. Transcriptome analysis showed that genes related to fatty acid metabolism were dramatically downregulated, but genes critical for glycolysis were markedly upregulated in PparaΔCM hearts. Moreover, the hypertrophy-related genes, including genes involved in extracellular matrix (ECM) remodeling, cell adhesion, and cell migration, were upregulated in hypertrophic PparaΔCM hearts. Western blot analyses demonstrated an increased HIF1α protein level in hypertrophic PparaΔCM hearts. PET/CT analyses showed an enhanced glucose uptake in hypertrophic PparaΔCM hearts. Bioenergetic analyses further revealed that both basal and maximal oxygen consumption rates and ATP production were significantly increased in hypertrophic Pparafl/fl hearts; however, these increases were markedly blunted in PparaΔCM hearts. In contrast, hypertrophic PparaΔCM hearts exhibited enhanced extracellular acidification rate (ECAR) capacity, as reflected by increased basal ECAR and glycolysis but decreased glycolytic reserve. These results suggest that cardiomyocyte PPARα is crucial for the homeostasis of both energy metabolism and ECM during TAC-induced cardiac remodeling, thus providing new insights into potential therapeutics of cardiac remodeling-related diseases.
Collapse
|
17
|
Molecular mechanisms of coronary microvascular endothelial dysfunction in diabetes mellitus: focus on mitochondrial quality surveillance. Angiogenesis 2022; 25:307-329. [PMID: 35303170 DOI: 10.1007/s10456-022-09835-8] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 03/07/2022] [Indexed: 12/12/2022]
Abstract
Coronary microvascular endothelial dysfunction is both a culprit and a victim of diabetes, and can accelerate diabetes-related microvascular and macrovascular complications by promoting vasoconstrictive, pro-inflammatory and pro-thrombotic responses. Perturbed mitochondrial function induces oxidative stress, disrupts metabolism and activates apoptosis in endothelial cells, thus exacerbating the progression of coronary microvascular complications in diabetes. The mitochondrial quality surveillance (MQS) system responds to stress by altering mitochondrial metabolism, dynamics (fission and fusion), mitophagy and biogenesis. Dysfunctional mitochondria are prone to fission, which generates two distinct types of mitochondria: one with a normal and the other with a depolarized mitochondrial membrane potential. Mitochondrial fusion and mitophagy can restore the membrane potential and homeostasis of defective mitochondrial fragments. Mitophagy-induced decreases in the mitochondrial population can be reversed by mitochondrial biogenesis. MQS abnormalities induce pathological mitochondrial fission, delayed mitophagy, impaired metabolism and defective biogenesis, thus promoting the accumulation of unhealthy mitochondria and the activation of mitochondria-dependent apoptosis. In this review, we examine the effects of MQS on mitochondrial fitness and explore the association of MQS disorders with coronary microvascular endothelial dysfunction in diabetes. We also discuss the potential to treat diabetes-related coronary microvascular endothelial dysfunction using novel MQS-altering drugs.
Collapse
|
18
|
Heart Failure and Drug Therapies: A Metabolic Review. Int J Mol Sci 2022; 23:ijms23062960. [PMID: 35328390 PMCID: PMC8950643 DOI: 10.3390/ijms23062960] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 03/01/2022] [Accepted: 03/01/2022] [Indexed: 02/07/2023] Open
Abstract
Cardiovascular disease is the leading cause of mortality globally with at least 26 million people worldwide living with heart failure (HF). Metabolism has been an active area of investigation in the setting of HF since the heart demands a high rate of ATP turnover to maintain homeostasis. With the advent of -omic technologies, specifically metabolomics and lipidomics, HF pathologies have been better characterized with unbiased and holistic approaches. These techniques have identified novel pathways in our understanding of progression of HF and potential points of intervention. Furthermore, sodium-glucose transport protein 2 inhibitors, a drug that has changed the dogma of HF treatment, has one of the strongest types of evidence for a potential metabolic mechanism of action. This review will highlight cardiac metabolism in both the healthy and failing heart and then discuss the metabolic effects of heart failure drugs.
Collapse
|
19
|
Exploring the Pattern of Metabolic Alterations Causing Energy Imbalance via PPARα Dysregulation in Cardiac Muscle During Doxorubicin Treatment. Cardiovasc Toxicol 2022; 22:436-461. [PMID: 35157213 DOI: 10.1007/s12012-022-09725-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 02/02/2022] [Indexed: 12/14/2022]
Abstract
Cardiotoxicity by anthracycline antineoplastic drug doxorubicin is one of the systemic toxicity of the cardiovascular system. The mechanism responsible for doxorubicin cardiotoxicity and lipid metabolism remains elusive. The current study tested the hypotheses that the role of peroxisome proliferator-activated receptor α (PPARα) in the progress of doxorubicin-induced cardiomyopathy and its mechanism behind lipid metabolism. In the present study, male rats were subjected to intraperitoneal injection (5-week period) of doxorubicin with different dosages such as low dosage (1.5 mg/kg body weight) and high dosage (15 mg/kg body weight) to induce doxorubicin cardiomyopathy. Myocardial PPARα was impaired in both low dosage and high dosage of doxorubicin-treated rats in a dose-dependent manner. The attenuated level of PPARα impairs the expression of the genes involved in mitochondrial transporter, fatty acid transportation, lipolysis, lipid metabolism, and fatty acid oxidation. Moreover, it disturbs the reverse triacylglycerol transporter apolipoprotein B-100 (APOB) in the myocardium. Doxorubicin elevates the circulatory lipid profile and glucose. Further aggravated lipid profile in circulation impedes the metabolism of lipid in cardiac tissue, which causes a lipotoxic condition in the heart and subsequently associated disease for the period of doxorubicin treatment. Elevated lipids in the circulation translocate into the heart dysregulates lipid metabolism in the heart, which causes augmented oxidative stress and necro-apoptosis and mediates lipotoxic conditions. This finding determines the mechanistic role of doxorubicin-disturbed lipid metabolism via PPARα, which leads to cardiac dysfunction.
Collapse
|
20
|
Hong JH, Zhang HG. Transcription Factors Involved in the Development and Prognosis of Cardiac Remodeling. Front Pharmacol 2022; 13:828549. [PMID: 35185581 PMCID: PMC8849252 DOI: 10.3389/fphar.2022.828549] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 01/14/2022] [Indexed: 01/09/2023] Open
Abstract
To compensate increasing workload, heart must work harder with structural changes, indicated by increasing size and changing shape, causing cardiac remodeling. However, pathological and unlimited compensated cardiac remodeling will ultimately lead to decompensation and heart failure. In the past decade, numerous studies have explored many signaling pathways involved in cardiac remodeling, but the complete mechanism of cardiac remodeling is still unrecognized, which hinders effective treatment and drug development. As gene transcriptional regulators, transcription factors control multiple cellular activities and play a critical role in cardiac remodeling. This review summarizes the regulation of fetal gene reprogramming, energy metabolism, apoptosis, autophagy in cardiomyocytes and myofibroblast activation of cardiac fibroblasts by transcription factors, with an emphasis on their potential roles in the development and prognosis of cardiac remodeling.
Collapse
|
21
|
Yamamoto T, Sano M. Deranged Myocardial Fatty Acid Metabolism in Heart Failure. Int J Mol Sci 2022; 23:996. [PMID: 35055179 PMCID: PMC8779056 DOI: 10.3390/ijms23020996] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 01/12/2022] [Accepted: 01/14/2022] [Indexed: 01/27/2023] Open
Abstract
The heart requires fatty acids to maintain its activity. Various mechanisms regulate myocardial fatty acid metabolism, such as energy production using fatty acids as fuel, for which it is known that coordinated control of fatty acid uptake, β-oxidation, and mitochondrial oxidative phosphorylation steps are important for efficient adenosine triphosphate (ATP) production without unwanted side effects. The fatty acids taken up by cardiomyocytes are not only used as substrates for energy production but also for the synthesis of triglycerides and the replacement reaction of fatty acid chains in cell membrane phospholipids. Alterations in fatty acid metabolism affect the structure and function of the heart. Recently, breakthrough studies have focused on the key transcription factors that regulate fatty acid metabolism in cardiomyocytes and the signaling systems that modify their functions. In this article, we reviewed the latest research on the role of fatty acid metabolism in the pathogenesis of heart failure and provide an outlook on future challenges.
Collapse
Affiliation(s)
| | - Motoaki Sano
- Department of Cardiology, Keio University School of Medicine, Tokyo 160-8582, Japan;
| |
Collapse
|
22
|
Umbarawan Y, Kawakami R, Syamsunarno MRAA, Obinata H, Yamaguchi A, Hanaoka H, Hishiki T, Hayakawa N, Koitabashi N, Sunaga H, Matsui H, Kurabayashi M, Iso T. Reduced Fatty Acid Use from CD36 Deficiency Deteriorates Streptozotocin-Induced Diabetic Cardiomyopathy in Mice. Metabolites 2021; 11:881. [PMID: 34940639 PMCID: PMC8707002 DOI: 10.3390/metabo11120881] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 12/10/2021] [Indexed: 01/17/2023] Open
Abstract
Cardiac dysfunction is induced by multifactorial mechanisms in diabetes. Deranged fatty acid (FA) utilization, known as lipotoxicity, has long been postulated as one of the upstream events in the development of diabetic cardiomyopathy. CD36, a transmembrane glycoprotein, plays a major role in FA uptake in the heart. CD36 knockout (CD36KO) hearts exhibit reduced rates of FA transport with marked enhancement of glucose use. In this study, we explore whether reduced FA use by CD36 ablation suppresses the development of streptozotocin (STZ)-induced diabetic cardiomyopathy. We found that cardiac contractile dysfunction had deteriorated 16 weeks after STZ treatment in CD36KO mice. Although accelerated glucose uptake was not reduced in CD36KO-STZ hearts, the total energy supply, estimated by the pool size in the TCA cycle, was significantly reduced. The isotopomer analysis with 13C6-glucose revealed that accelerated glycolysis, estimated by enrichment of 13C2-citrate and 13C2-malate, was markedly suppressed in CD36KO-STZ hearts. Levels of ceramides, which are cardiotoxic lipids, were not elevated in CD36KO-STZ hearts compared to wild-type-STZ ones. Furthermore, increased energy demand by transverse aortic constriction resulted in synergistic exacerbation of contractile dysfunction in CD36KO-STZ mice. These findings suggest that CD36KO-STZ hearts are energetically compromised by reduced FA use and suppressed glycolysis; therefore, the limitation of FA utilization is detrimental to cardiac energetics in this model of diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Yogi Umbarawan
- Department of Cardiovascular Medicine, Gunma University Graduate School of Medicine, 3-39-22 Showa-Machi, Maebashi 371-8511, Japan; (Y.U.); (R.K.); (M.R.A.A.S.); (N.K.); (H.S.); (M.K.)
- Department of Internal Medicine, Faculty of Medicine Universitas Indonesia, Jl. Salemba Raya no. 6, Jakarta 10430, Indonesia
| | - Ryo Kawakami
- Department of Cardiovascular Medicine, Gunma University Graduate School of Medicine, 3-39-22 Showa-Machi, Maebashi 371-8511, Japan; (Y.U.); (R.K.); (M.R.A.A.S.); (N.K.); (H.S.); (M.K.)
| | - Mas Rizky A. A. Syamsunarno
- Department of Cardiovascular Medicine, Gunma University Graduate School of Medicine, 3-39-22 Showa-Machi, Maebashi 371-8511, Japan; (Y.U.); (R.K.); (M.R.A.A.S.); (N.K.); (H.S.); (M.K.)
- Department of Biomedical Sciences, Universitas Padjadjaran, Jl. Raya Bandung Sumedang KM 21, Jatinangor 45363, Indonesia
| | - Hideru Obinata
- Education and Research Support Center, Gunma University Graduate School of Medicine, 3-39-22 Showa-Machi, Maebashi 371-8511, Japan;
| | - Aiko Yamaguchi
- Department of Bioimaging Information Analysis, Gunma University Graduate School of Medicine, 3-39-22 Showa-Machi, Maebashi 371-8511, Japan; (A.Y.); (H.H.)
| | - Hirofumi Hanaoka
- Department of Bioimaging Information Analysis, Gunma University Graduate School of Medicine, 3-39-22 Showa-Machi, Maebashi 371-8511, Japan; (A.Y.); (H.H.)
| | - Takako Hishiki
- Department of Biochemistry, Keio University School of Medicine, 35 Shinano-Machi, Shinjuku-Ku, Tokyo 160-8582, Japan; (T.H.); (N.H.)
- Clinical and Translational Research Center, Keio University School of Medicine, 35 Shinano-Machi, Shinjuku-Ku, Tokyo 160-8582, Japan
| | - Noriyo Hayakawa
- Department of Biochemistry, Keio University School of Medicine, 35 Shinano-Machi, Shinjuku-Ku, Tokyo 160-8582, Japan; (T.H.); (N.H.)
- Clinical and Translational Research Center, Keio University School of Medicine, 35 Shinano-Machi, Shinjuku-Ku, Tokyo 160-8582, Japan
| | - Norimichi Koitabashi
- Department of Cardiovascular Medicine, Gunma University Graduate School of Medicine, 3-39-22 Showa-Machi, Maebashi 371-8511, Japan; (Y.U.); (R.K.); (M.R.A.A.S.); (N.K.); (H.S.); (M.K.)
| | - Hiroaki Sunaga
- Department of Cardiovascular Medicine, Gunma University Graduate School of Medicine, 3-39-22 Showa-Machi, Maebashi 371-8511, Japan; (Y.U.); (R.K.); (M.R.A.A.S.); (N.K.); (H.S.); (M.K.)
- Center for Liberal Arts and Sciences, Ashikaga University, 268-1 Omae-Machi, Ashikaga 326-8558, Japan
| | - Hiroki Matsui
- Department of Laboratory Sciences, Gunma University Graduate School of Health Sciences, 3-39-22 Showa-Machi, Maebashi 371-8511, Japan;
| | - Masahiko Kurabayashi
- Department of Cardiovascular Medicine, Gunma University Graduate School of Medicine, 3-39-22 Showa-Machi, Maebashi 371-8511, Japan; (Y.U.); (R.K.); (M.R.A.A.S.); (N.K.); (H.S.); (M.K.)
| | - Tatsuya Iso
- Department of Cardiovascular Medicine, Gunma University Graduate School of Medicine, 3-39-22 Showa-Machi, Maebashi 371-8511, Japan; (Y.U.); (R.K.); (M.R.A.A.S.); (N.K.); (H.S.); (M.K.)
- Department of Medical Technology and Clinical Engineering, Gunma University of Health and Welfare, 191-1 Kawamagari-Machi, Maebashi 371-0823, Japan
| |
Collapse
|
23
|
Kawaguchi S, Okada M. Cardiac Metabolism in Sepsis. Metabolites 2021; 11:metabo11120846. [PMID: 34940604 PMCID: PMC8707959 DOI: 10.3390/metabo11120846] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 12/02/2021] [Accepted: 12/02/2021] [Indexed: 12/11/2022] Open
Abstract
The mechanism of sepsis-induced cardiac dysfunction is believed to be different from that of myocardial ischemia. In sepsis, chemical mediators, such as endotoxins, cytokines, and nitric oxide, cause metabolic abnormalities, mitochondrial dysfunction, and downregulation of β-adrenergic receptors. These factors inhibit the production of ATP, essential for myocardial energy metabolism, resulting in cardiac dysfunction. This review focuses on the metabolic changes in sepsis, particularly in the heart. In addition to managing inflammation, interventions focusing on metabolism may be a new therapeutic strategy for cardiac dysfunction due to sepsis.
Collapse
Affiliation(s)
- Satoshi Kawaguchi
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Bloomington, IN 46202, USA;
| | - Motoi Okada
- Department of Emergency Medicine, Asahikawa Medical University, Asahikawa 078-8510, Japan
- Correspondence: ; Tel.: +81-166-68-2852
| |
Collapse
|
24
|
Montaigne D, Butruille L, Staels B. PPAR control of metabolism and cardiovascular functions. Nat Rev Cardiol 2021; 18:809-823. [PMID: 34127848 DOI: 10.1038/s41569-021-00569-6] [Citation(s) in RCA: 359] [Impact Index Per Article: 119.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/10/2021] [Indexed: 12/22/2022]
Abstract
Peroxisome proliferator-activated receptor-α (PPARα), PPARδ and PPARγ are transcription factors that regulate gene expression following ligand activation. PPARα increases cellular fatty acid uptake, esterification and trafficking, and regulates lipoprotein metabolism genes. PPARδ stimulates lipid and glucose utilization by increasing mitochondrial function and fatty acid desaturation pathways. By contrast, PPARγ promotes fatty acid uptake, triglyceride formation and storage in lipid droplets, thereby increasing insulin sensitivity and glucose metabolism. PPARs also exert antiatherogenic and anti-inflammatory effects on the vascular wall and immune cells. Clinically, PPARγ activation by glitazones and PPARα activation by fibrates reduce insulin resistance and dyslipidaemia, respectively. PPARs are also physiological master switches in the heart, steering cardiac energy metabolism in cardiomyocytes, thereby affecting pathological heart failure and diabetic cardiomyopathy. Novel PPAR agonists in clinical development are providing new opportunities in the management of metabolic and cardiovascular diseases.
Collapse
Affiliation(s)
- David Montaigne
- University of Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Laura Butruille
- University of Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Bart Staels
- University of Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France.
| |
Collapse
|
25
|
Yan A, Xie G, Ding X, Wang Y, Guo L. Effects of Lipid Overload on Heart in Metabolic Diseases. Horm Metab Res 2021; 53:771-778. [PMID: 34891207 PMCID: PMC8664556 DOI: 10.1055/a-1693-8356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Metabolic diseases are often associated with lipid and glucose metabolism abnormalities, which increase the risk of cardiovascular disease. Diabetic cardiomyopathy (DCM) is an important development of metabolic diseases and a major cause of death. Lipids are the main fuel for energy metabolism in the heart. The increase of circulating lipids affects the uptake and utilization of fatty acids and glucose in the heart, and also affects mitochondrial function. In this paper, the mechanism of lipid overload in metabolic diseases leading to cardiac energy metabolism disorder is discussed.
Collapse
Affiliation(s)
- An Yan
- Tianjin University of Traditional Chinese Medicine, Tianjin,
China
| | - Guinan Xie
- Tianjin University of Traditional Chinese Medicine, Tianjin,
China
| | - Xinya Ding
- Tianjin University of Traditional Chinese Medicine, Tianjin,
China
| | - Yi Wang
- Tianjin University of Traditional Chinese Medicine, Tianjin,
China
- Correspondence Yi Wang Institute of Traditional Chinese MedicineTianjin University of Traditional Chinese Medicine300193 TianjinChina+86-22-59596555
| | - Liping Guo
- Tianjin Academy of Traditional Chinese Medicine, Tianjin,
China
- Liping Guo Tianjin Academy of Traditional Chinese Medicine300120 TianjinChina
| |
Collapse
|
26
|
Karwi QG, Sun Q, Lopaschuk GD. The Contribution of Cardiac Fatty Acid Oxidation to Diabetic Cardiomyopathy Severity. Cells 2021; 10:cells10113259. [PMID: 34831481 PMCID: PMC8621814 DOI: 10.3390/cells10113259] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 11/15/2021] [Accepted: 11/17/2021] [Indexed: 12/17/2022] Open
Abstract
Diabetes is a major risk factor for the development of cardiovascular disease via contributing and/or triggering significant cellular signaling and metabolic and structural alterations at the level of the heart and the whole body. The main cause of mortality and morbidity in diabetic patients is cardiovascular disease including diabetic cardiomyopathy. Therefore, understanding how diabetes increases the incidence of diabetic cardiomyopathy and how it mediates the major perturbations in cell signaling and energy metabolism should help in the development of therapeutics to prevent these perturbations. One of the significant metabolic alterations in diabetes is a marked increase in cardiac fatty acid oxidation rates and the domination of fatty acids as the major energy source in the heart. This increased reliance of the heart on fatty acids in the diabetic has a negative impact on cardiac function and structure through a number of mechanisms. It also has a detrimental effect on cardiac efficiency and worsens the energy status in diabetes, mainly through inhibiting cardiac glucose oxidation. Furthermore, accelerated cardiac fatty acid oxidation rates in diabetes also make the heart more vulnerable to ischemic injury. In this review, we discuss how cardiac energy metabolism is altered in diabetic cardiomyopathy and the impact of cardiac insulin resistance on the contribution of glucose and fatty acid to overall cardiac ATP production and cardiac efficiency. Furthermore, how diabetes influences the susceptibility of the myocardium to ischemia/reperfusion injury and the role of the changes in glucose and fatty acid oxidation in mediating these effects are also discussed.
Collapse
Affiliation(s)
- Qutuba G. Karwi
- Cardiovascular Research Centre, Department of Pediatrics, University of Alberta, Edmonton, AB T6G 2S2, Canada; (Q.G.K.); (Q.S.)
| | - Qiuyu Sun
- Cardiovascular Research Centre, Department of Pediatrics, University of Alberta, Edmonton, AB T6G 2S2, Canada; (Q.G.K.); (Q.S.)
| | - Gary D. Lopaschuk
- 423 Heritage Medical Research Centre, University of Alberta, Edmonton, AB T6G 2S2, Canada
- Correspondence: ; Tel.: +1-780-492-2170; Fax: +1-780-492-9753
| |
Collapse
|
27
|
Nuclear Receptors in Myocardial and Cerebral Ischemia-Mechanisms of Action and Therapeutic Strategies. Int J Mol Sci 2021; 22:ijms222212326. [PMID: 34830207 PMCID: PMC8617737 DOI: 10.3390/ijms222212326] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 11/08/2021] [Accepted: 11/10/2021] [Indexed: 12/12/2022] Open
Abstract
Nearly 18 million people died from cardiovascular diseases in 2019, of these 85% were due to heart attack and stroke. The available therapies although efficacious, have narrow therapeutic window and long list of contraindications. Therefore, there is still an urgent need to find novel molecular targets that could protect the brain and heart against ischemia without evoking major side effects. Nuclear receptors are one of the promising targets for anti-ischemic drugs. Modulation of estrogen receptors (ERs) and peroxisome proliferator-activated receptors (PPARs) by their ligands is known to exert neuro-, and cardioprotective effects through anti-apoptotic, anti-inflammatory or anti-oxidant action. Recently, it has been shown that the expression of aryl hydrocarbon receptor (AhR) is strongly increased after brain or heart ischemia and evokes an activation of apoptosis or inflammation in injury site. We hypothesize that activation of ERs and PPARs and inhibition of AhR signaling pathways could be a promising strategy to protect the heart and the brain against ischemia. In this Review, we will discuss currently available knowledge on the mechanisms of action of ERs, PPARs and AhR in experimental models of stroke and myocardial infarction and future perspectives to use them as novel targets in cardiovascular diseases.
Collapse
|
28
|
Wei DZ, Li D, Zheng DM, An ZN, Xing XJ, Jiang DW, Mei XF, Liu C. Curcumin Conjugated Gold Nanoclusters as Perspective Therapeutics for Diabetic Cardiomyopathy. Front Chem 2021; 9:763892. [PMID: 34765588 PMCID: PMC8576376 DOI: 10.3389/fchem.2021.763892] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 10/12/2021] [Indexed: 12/23/2022] Open
Abstract
Accumulation of lipids in the myocardium contributes to the development of cardiac dysfunctions and various chronic diseases, such as diabetic cardiomyopathy (DCM). Curcumin (Cur) can relieve lipid accumulation problems, but its efficiency is limited by poor water solubility and biocompatibility. Herein, gold nanoclusters (AuNCs) were used to improve the efficiency of Cur, and the conjugates Curcumin-AuNCs (AuCur) were developed. In the treatment of high-fat-induced myocardial cell damage, we found that AuCur could effectively reduce intracellular lipid accumulation, the increase of reactive oxygen species (ROS), the increase of mitochondrial division, and the increase of apoptosis compared with Cur. AuCur decreased the expression of the peroxisome proliferator-activated receptors-α subtype (PPARα), and the therapeutic effect of AuCur was canceled when the expression of PPARα was enhanced. For the above reasons, AuCur treated the toxic effect of high lipid on cardiomyocytes by regulating PPARα, providing a new idea and method for the treatment of DCM.
Collapse
Affiliation(s)
- Dong-Zhuo Wei
- Clinical Discipline of Chinese and Western Integrative Medicine, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Dan Li
- Public Basic Academy, Jinzhou Medical University, Jinzhou, China
| | - Dan-Meng Zheng
- Department of Endocrinology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Zhen-Ni An
- Department of Endocrinology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Xue-Jiao Xing
- Department of Endocrinology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Ding-Wen Jiang
- Department of Endocrinology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Xi-Fan Mei
- Department of Orthopedics, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Chang Liu
- Department of Endocrinology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| |
Collapse
|
29
|
de Wit-Verheggen VHW, van de Weijer T. Changes in Cardiac Metabolism in Prediabetes. Biomolecules 2021; 11:1680. [PMID: 34827678 PMCID: PMC8615987 DOI: 10.3390/biom11111680] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 10/28/2021] [Accepted: 11/08/2021] [Indexed: 01/05/2023] Open
Abstract
In type 2 diabetes mellitus (T2DM), there is an increased prevalence of cardiovascular disease (CVD), even when corrected for atherosclerosis and other CVD risk factors. Diastolic dysfunction is one of the early changes in cardiac function that precedes the onset of cardiac failure, and it occurs already in the prediabetic state. It is clear that these changes are closely linked to alterations in cardiac metabolism; however, the exact etiology is unknown. In this narrative review, we provide an overview of the early cardiac changes in fatty acid and glucose metabolism in prediabetes and its consequences on cardiac function. A better understanding of the relationship between metabolism, mitochondrial function, and cardiac function will lead to insights into the etiology of the declined cardiac function in prediabetes.
Collapse
Affiliation(s)
- Vera H. W. de Wit-Verheggen
- Department of Nutrition and Movement Sciences, School for Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, 6200 MD Maastricht, The Netherlands;
| | - Tineke van de Weijer
- Department of Nutrition and Movement Sciences, School for Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, 6200 MD Maastricht, The Netherlands;
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, 6200 MD Maastricht, The Netherlands
| |
Collapse
|
30
|
Hwang S, Chung KW. Targeting fatty acid metabolism for fibrotic disorders. Arch Pharm Res 2021; 44:839-856. [PMID: 34664210 DOI: 10.1007/s12272-021-01352-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 10/11/2021] [Indexed: 02/06/2023]
Abstract
Fibrosis is defined by abnormal accumulation of extracellular matrix, which can affect virtually every organ system under diseased conditions. Fibrotic tissue remodeling often leads to organ dysfunction and is highly associated with increased morbidity and mortality. The disease burden caused by fibrosis is substantial, and the medical need for effective antifibrotic therapies is essential. Significant progress has been made in understanding the molecular mechanism and pathobiology of fibrosis, such as transforming growth factor-β (TGF-β)-mediated signaling pathways. However, owing to the complex and dynamic properties of fibrotic disorders, there are currently no therapeutic options that can prevent or reverse fibrosis. Recent studies have revealed that alterations in fatty acid metabolic processes are common mechanisms and core pathways that play a central role in different fibrotic disorders. Excessive lipid accumulation or defective fatty acid oxidation is associated with increased lipotoxicity, which directly contributes to the development of fibrosis. Genetic alterations or pharmacologic targeting of fatty acid metabolic processes have great potential for the inhibition of fibrosis development. Furthermore, mechanistic studies have revealed active interactions between altered metabolic processes and fibrosis development. Several well-known fibrotic factors change the lipid metabolic processes, while altered metabolic processes actively participate in fibrosis development. This review summarizes the recent evidence linking fatty acid metabolism and fibrosis, and provides new insights into the pathogenesis of fibrotic diseases for the development of drugs for fibrosis prevention and treatment.
Collapse
Affiliation(s)
- Seonghwan Hwang
- College of Pharmacy, Pusan National University, Busan, 46214, Republic of Korea
| | - Ki Wung Chung
- College of Pharmacy, Pusan National University, Busan, 46214, Republic of Korea.
| |
Collapse
|
31
|
Ritterhoff J, McMillen TS, Villet O, Young S, Kolwicz SC, Senn T, Caudal A, Tian R. Increasing fatty acid oxidation elicits a sex-dependent response in failing mouse hearts. J Mol Cell Cardiol 2021; 158:1-10. [PMID: 33989657 PMCID: PMC8405556 DOI: 10.1016/j.yjmcc.2021.05.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 05/06/2021] [Accepted: 05/07/2021] [Indexed: 01/12/2023]
Abstract
BACKGROUND Reduced fatty acid oxidation (FAO) is a hallmark of metabolic remodeling in heart failure. Enhancing mitochondrial long-chain fatty acid uptake by Acetyl-CoA carboxylase 2 (ACC2) deletion increases FAO and prevents cardiac dysfunction during chronic stresses, but therapeutic efficacy of this approach has not been determined. METHODS Male and female ACC2 f/f-MCM (ACC2KO) and their respective littermate controls were subjected to chronic pressure overload by TAC surgery. Tamoxifen injection 3 weeks after TAC induced ACC2 deletion and increased FAO in ACC2KO mice with pathological hypertrophy. RESULTS ACC2 deletion in mice with pre-existing cardiac pathology promoted FAO in female and male hearts, but improved cardiac function only in female mice. In males, pressure overload caused a downregulation in the mitochondrial oxidative function. Stimulating FAO by ACC2 deletion caused unproductive acyl-carnitine accumulation, which failed to improve cardiac energetics. In contrast, mitochondrial oxidative capacity was sustained in female pressure overloaded hearts and ACC2 deletion improved myocardial energetics. Mechanistically, we revealed a sex-dependent regulation of PPARα signaling pathway in heart failure, which accounted for the differential response to ACC2 deletion. CONCLUSION Metabolic remodeling in the failing heart is sex-dependent which could determine the response to metabolic intervention. The findings suggest that both mitochondrial oxidative capacity and substrate preference should be considered for metabolic therapy of heart failure.
Collapse
Affiliation(s)
- Julia Ritterhoff
- Mitochondria and Metabolism Center, Department of Anesthesiology and Pain Medicine, University of Washington, Republican Street 850, 98109 Seattle, WA, USA
| | - Timothy S. McMillen
- Mitochondria and Metabolism Center, Department of Anesthesiology and Pain Medicine, University of Washington, Republican Street 850, 98109 Seattle, WA, USA
| | - Outi Villet
- Mitochondria and Metabolism Center, Department of Anesthesiology and Pain Medicine, University of Washington, Republican Street 850, 98109 Seattle, WA, USA
| | - Sara Young
- Mitochondria and Metabolism Center, Department of Anesthesiology and Pain Medicine, University of Washington, Republican Street 850, 98109 Seattle, WA, USA
| | - Stephen C. Kolwicz
- Mitochondria and Metabolism Center, Department of Anesthesiology and Pain Medicine, University of Washington, Republican Street 850, 98109 Seattle, WA, USA.,Heart and Muscle Metabolism Laboratory, Health and Exercise Physiology, Ursinus College, Collegeville, PA 19426, USA
| | - Taurence Senn
- Department of Medicinal Chemistry, School of Pharmacy, University of Washington, H172 Health Science Building, 98195 Seattle, WA, USA
| | - Arianne Caudal
- Mitochondria and Metabolism Center, Department of Anesthesiology and Pain Medicine, University of Washington, Republican Street 850, 98109 Seattle, WA, USA
| | - Rong Tian
- Mitochondria and Metabolism Center, Department of Anesthesiology and Pain Medicine, University of Washington, Republican Street 850, 98109 Seattle, WA, USA.,Corresponding author at: Mitochondria and Metabolism Center, University of Washington School of Medicine, 850 Republican Street, Seattle, WA 98109
| |
Collapse
|
32
|
Zhuang L, Mao Y, Liu Z, Li C, Jin Q, Lu L, Tao R, Yan X, Chen K. FABP3 Deficiency Exacerbates Metabolic Derangement in Cardiac Hypertrophy and Heart Failure via PPARα Pathway. Front Cardiovasc Med 2021; 8:722908. [PMID: 34458345 PMCID: PMC8387950 DOI: 10.3389/fcvm.2021.722908] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 07/12/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Cardiac hypertrophy was accompanied by various cardiovascular diseases (CVDs), and due to the high global incidence and mortality of CVDs, it has become increasingly critical to characterize the pathogenesis of cardiac hypertrophy. We aimed to determine the metabolic roles of fatty acid binding protein 3 (FABP3) on transverse aortic constriction (TAC)-induced cardiac hypertrophy. Methods and Results: Transverse aortic constriction or Ang II treatment markedly upregulated Fabp3 expression. Notably, Fabp3 ablation aggravated TAC-induced cardiac hypertrophy and cardiac dysfunction. Multi-omics analysis revealed that Fabp3-deficient hearts exhibited disrupted metabolic signatures characterized by increased glycolysis, toxic lipid accumulation, and compromised fatty acid oxidation and ATP production under hypertrophic stimuli. Mechanistically, FABP3 mediated metabolic reprogramming by directly interacting with PPARα, which prevented its degradation and synergistically modulated its transcriptional activity on Mlycd and Gck. Finally, treatment with the PPARα agonist, fenofibrate, rescued the pro-hypertrophic effects of Fabp3 deficiency. Conclusions: Collectively, these findings reveal the indispensable roles of the FABP3-PPARα axis on metabolic homeostasis and the development of hypertrophy, which sheds new light on the treatment of hypertrophy.
Collapse
Affiliation(s)
- Lingfang Zhuang
- Department of Vascular and Cardiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ye Mao
- Department of Health Management Center, Ruijin Hospital Lu Wan Branch, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zizhu Liu
- Department of Vascular and Cardiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chenni Li
- Department of Vascular and Cardiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qi Jin
- Department of Vascular and Cardiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lin Lu
- Department of Vascular and Cardiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rong Tao
- Department of Vascular and Cardiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoxiang Yan
- Department of Vascular and Cardiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kang Chen
- Department of Vascular and Cardiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
33
|
Cardiac-Specific Overexpression of ERRγ in Mice Induces Severe Heart Dysfunction and Early Lethality. Int J Mol Sci 2021; 22:ijms22158047. [PMID: 34360813 PMCID: PMC8348522 DOI: 10.3390/ijms22158047] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 07/23/2021] [Accepted: 07/24/2021] [Indexed: 11/17/2022] Open
Abstract
Proper cardiac function depends on the coordinated expression of multiple gene networks related to fuel utilization and mitochondrial ATP production, heart contraction, and ion transport. Key transcriptional regulators that regulate these gene networks have been identified. Among them, estrogen-related receptors (ERRs) have emerged as crucial modulators of cardiac function by regulating cellular metabolism and contraction machinery. Consistent with this role, lack of ERRα or ERRγ results in cardiac derangements that lead to functional maladaptation in response to increased workload. Interestingly, metabolic inflexibility associated with diabetic cardiomyopathy has been recently associated with increased mitochondrial fatty acid oxidation and expression of ERRγ, suggesting that sustained expression of this nuclear receptor could result in a cardiac pathogenic outcome. Here, we describe the generation of mice with cardiac-specific overexpression of ERRγ, which die at young ages due to heart failure. ERRγ transgenic mice show signs of dilated cardiomyopathy associated with cardiomyocyte hypertrophy, increased cell death, and fibrosis. Our results suggest that ERRγ could play a role in mediating cardiac pathogenic responses.
Collapse
|
34
|
Paredes A, Santos-Clemente R, Ricote M. Untangling the Cooperative Role of Nuclear Receptors in Cardiovascular Physiology and Disease. Int J Mol Sci 2021; 22:ijms22157775. [PMID: 34360540 PMCID: PMC8346021 DOI: 10.3390/ijms22157775] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 07/13/2021] [Accepted: 07/16/2021] [Indexed: 12/12/2022] Open
Abstract
The heart is the first organ to acquire its physiological function during development, enabling it to supply the organism with oxygen and nutrients. Given this early commitment, cardiomyocytes were traditionally considered transcriptionally stable cells fully committed to contractile function. However, growing evidence suggests that the maintenance of cardiac function in health and disease depends on transcriptional and epigenetic regulation. Several studies have revealed that the complex transcriptional alterations underlying cardiovascular disease (CVD) manifestations such as myocardial infarction and hypertrophy is mediated by cardiac retinoid X receptors (RXR) and their partners. RXRs are members of the nuclear receptor (NR) superfamily of ligand-activated transcription factors and drive essential biological processes such as ion handling, mitochondrial biogenesis, and glucose and lipid metabolism. RXRs are thus attractive molecular targets for the development of effective pharmacological strategies for CVD treatment and prevention. In this review, we summarize current knowledge of RXR partnership biology in cardiac homeostasis and disease, providing an up-to-date view of the molecular mechanisms and cellular pathways that sustain cardiomyocyte physiology.
Collapse
|
35
|
Shirif AZ, Kovačević S, Brkljačić J, Teofilović A, Elaković I, Djordjevic A, Matić G. Decreased Glucocorticoid Signaling Potentiates Lipid-Induced Inflammation and Contributes to Insulin Resistance in the Skeletal Muscle of Fructose-Fed Male Rats Exposed to Stress. Int J Mol Sci 2021; 22:ijms22137206. [PMID: 34281257 PMCID: PMC8269441 DOI: 10.3390/ijms22137206] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 06/25/2021] [Accepted: 06/28/2021] [Indexed: 01/03/2023] Open
Abstract
The modern lifestyle brings both excessive fructose consumption and daily exposure to stress which could lead to metabolic disturbances and type 2 diabetes. Muscles are important points of glucose and lipid metabolism, with a crucial role in the maintenance of systemic energy homeostasis. We investigated whether 9-week fructose-enriched diet, with and without exposure to 4-week unpredictable stress, disturbs insulin signaling in the skeletal muscle of male rats and evaluated potential contributory roles of muscle lipid metabolism, glucocorticoid signaling and inflammation. The combination of fructose-enriched diet and stress increased peroxisome proliferator-activated receptors-α and -δ and stimulated lipid uptake, lipolysis and β-oxidation in the muscle of fructose-fed stressed rats. Combination of treatment also decreased systemic insulin sensitivity judged by lower R-QUICKI, and lowered muscle protein content and stimulatory phosphorylations of insulin receptor supstrate-1 and Akt, as well as the level of 11β-hydroxysteroid dehydrogenase type 1 and glucocorticoid receptor. At the same time, increased levels of protein tyrosine phosphatase-1B, nuclear factor-κB, tumor necrosis factor-α, were observed in the muscle of fructose-fed stressed rats. Based on these results, we propose that decreased glucocorticoid signaling in the skeletal muscle can make a setting for lipid-induced inflammation and the development of insulin resistance in fructose-fed stressed rats.
Collapse
|
36
|
Salvatore T, Pafundi PC, Galiero R, Albanese G, Di Martino A, Caturano A, Vetrano E, Rinaldi L, Sasso FC. The Diabetic Cardiomyopathy: The Contributing Pathophysiological Mechanisms. Front Med (Lausanne) 2021; 8:695792. [PMID: 34277669 PMCID: PMC8279779 DOI: 10.3389/fmed.2021.695792] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 06/07/2021] [Indexed: 12/12/2022] Open
Abstract
Individuals with diabetes mellitus (DM) disclose a higher incidence and a poorer prognosis of heart failure (HF) than non-diabetic people, even in the absence of other HF risk factors. The adverse impact of diabetes on HF likely reflects an underlying “diabetic cardiomyopathy” (DM–CMP), which may by exacerbated by left ventricular hypertrophy and coronary artery disease (CAD). The pathogenesis of DM-CMP has been a hot topic of research since its first description and is still under active investigation, as a complex interplay among multiple mechanisms may play a role at systemic, myocardial, and cellular/molecular levels. Among these, metabolic abnormalities such as lipotoxicity and glucotoxicity, mitochondrial damage and dysfunction, oxidative stress, abnormal calcium signaling, inflammation, epigenetic factors, and others. These disturbances predispose the diabetic heart to extracellular remodeling and hypertrophy, thus leading to left ventricular diastolic and systolic dysfunction. This Review aims to outline the major pathophysiological changes and the underlying mechanisms leading to myocardial remodeling and cardiac functional derangement in DM-CMP.
Collapse
Affiliation(s)
- Teresa Salvatore
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Pia Clara Pafundi
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Raffaele Galiero
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Gaetana Albanese
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Anna Di Martino
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Alfredo Caturano
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Erica Vetrano
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Luca Rinaldi
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Ferdinando Carlo Sasso
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Naples, Italy
| |
Collapse
|
37
|
Mana MD, Hussey AM, Tzouanas CN, Imada S, Barrera Millan Y, Bahceci D, Saiz DR, Webb AT, Lewis CA, Carmeliet P, Mihaylova MM, Shalek AK, Yilmaz ÖH. High-fat diet-activated fatty acid oxidation mediates intestinal stemness and tumorigenicity. Cell Rep 2021; 35:109212. [PMID: 34107251 PMCID: PMC8258630 DOI: 10.1016/j.celrep.2021.109212] [Citation(s) in RCA: 84] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 03/01/2021] [Accepted: 05/12/2021] [Indexed: 12/19/2022] Open
Abstract
Obesity is an established risk factor for cancer in many tissues. In the mammalian intestine, a pro-obesity high-fat diet (HFD) promotes regeneration and tumorigenesis by enhancing intestinal stem cell (ISC) numbers, proliferation, and function. Although PPAR (peroxisome proliferator-activated receptor) nuclear receptor activity has been proposed to facilitate these effects, their exact role is unclear. Here we find that, in loss-of-function in vivo models, PPARα and PPARδ contribute to the HFD response in ISCs. Mechanistically, both PPARs do so by robustly inducing a downstream fatty acid oxidation (FAO) metabolic program. Pharmacologic and genetic disruption of CPT1A (the rate-controlling enzyme of mitochondrial FAO) blunts the HFD phenotype in ISCs. Furthermore, inhibition of CPT1A dampens the pro-tumorigenic consequences of a HFD on early tumor incidence and progression. These findings demonstrate that inhibition of a HFD-activated FAO program creates a therapeutic opportunity to counter the effects of a HFD on ISCs and intestinal tumorigenesis.
Collapse
Affiliation(s)
- Miyeko D Mana
- Department of Biology, The David H. Koch Institute for Integrative Cancer Research at MIT, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA.
| | - Amanda M Hussey
- Department of Biology, The David H. Koch Institute for Integrative Cancer Research at MIT, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Constantine N Tzouanas
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Institute for Medical Engineering & Science, Department of Chemistry, and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge MA 02139, USA; Program in Health Sciences & Technology, Harvard Medical School, Boston, MA 02115, USA
| | - Shinya Imada
- Department of Biology, The David H. Koch Institute for Integrative Cancer Research at MIT, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | | - Dorukhan Bahceci
- Department of Biology, The David H. Koch Institute for Integrative Cancer Research at MIT, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Dominic R Saiz
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
| | - Anna T Webb
- Department of Biology, The David H. Koch Institute for Integrative Cancer Research at MIT, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Caroline A Lewis
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, and Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven 3000, Belgium; State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou 510060, Guangdong, P.R. China; Department of Biomedicine, Aarhus University, Aarhus 8000, Denmark
| | - Maria M Mihaylova
- Department of Biological Chemistry and Pharmacology, Ohio State University, Columbus, OH 43210, USA; The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Alex K Shalek
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Institute for Medical Engineering & Science, Department of Chemistry, and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge MA 02139, USA; Program in Health Sciences & Technology, Harvard Medical School, Boston, MA 02115, USA
| | - Ömer H Yilmaz
- Department of Biology, The David H. Koch Institute for Integrative Cancer Research at MIT, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
38
|
The Mystery of Diabetic Cardiomyopathy: From Early Concepts and Underlying Mechanisms to Novel Therapeutic Possibilities. Int J Mol Sci 2021; 22:ijms22115973. [PMID: 34205870 PMCID: PMC8198766 DOI: 10.3390/ijms22115973] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 05/26/2021] [Accepted: 05/30/2021] [Indexed: 02/07/2023] Open
Abstract
Diabetic patients are predisposed to diabetic cardiomyopathy, a specific form of cardiomyopathy which is characterized by the development of myocardial fibrosis, cardiomyocyte hypertrophy, and apoptosis that develops independently of concomitant macrovascular and microvascular diabetic complications. Its pathophysiology is multifactorial and poorly understood and no specific therapeutic guideline has yet been established. Diabetic cardiomyopathy is a challenging diagnosis, made after excluding other potential entities, treated with different pharmacotherapeutic agents targeting various pathophysiological pathways that need yet to be unraveled. It has great clinical importance as diabetes is a disease with pandemic proportions. This review focuses on the potential mechanisms contributing to this entity, diagnostic options, as well as on potential therapeutic interventions taking in consideration their clinical feasibility and limitations in everyday practice. Besides conventional therapies, we discuss novel therapeutic possibilities that have not yet been translated into clinical practice.
Collapse
|
39
|
Zhang X, Fan J, Li H, Chen C, Wang Y. CD36 Signaling in Diabetic Cardiomyopathy. Aging Dis 2021; 12:826-840. [PMID: 34094645 PMCID: PMC8139204 DOI: 10.14336/ad.2020.1217] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Accepted: 12/17/2020] [Indexed: 12/14/2022] Open
Abstract
Cluster of differentiation 36 (CD36), also referred to as scavenger receptor B2, has been shown to serve multiple functions in lipid metabolism, inflammatory signaling, oxidative stress, and energy reprogramming. As a scavenger receptor, CD36 interacts with various ligands, such as oxidized low-density lipoprotein (oxLDL), thrombospondin 1 (TSP-1), and fatty acid (FA), thereby activating specific downstream signaling pathways. Cardiac CD36 is mostly expressed on the surface of cardiomyocytes and endothelial cells. The pathophysiological process of diabetic cardiomyopathy (DCM) encompasses diverse metabolic abnormalities, such as enhanced transfer of cardiac myocyte sarcolemmal FA, increased levels of advanced glycation end-products, elevation in oxidative stress, impaired insulin signaling cascade, disturbance in calcium handling, and microvascular rarefaction which are closely related to CD36 signaling. This review presents a summary of the CD36 signaling pathway that acts mainly as a long-chain FA transporter in cardiac myocytes and functions as a receptor to bind to numerous ligands in endothelial cells. Finally, we summarize the recent basic research and clinical findings regarding CD36 signaling in DCM, suggesting a promising strategy to treat this condition.
Collapse
Affiliation(s)
- Xudong Zhang
- Division of Cardiology, Tongji Hospital, Tongji Medical College and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, China
| | - Jiahui Fan
- Division of Cardiology, Tongji Hospital, Tongji Medical College and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, China
| | - Huaping Li
- Division of Cardiology, Tongji Hospital, Tongji Medical College and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, China
| | - Chen Chen
- Division of Cardiology, Tongji Hospital, Tongji Medical College and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Wang
- Division of Cardiology, Tongji Hospital, Tongji Medical College and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
40
|
Sithara T, Drosatos K. Metabolic Complications in Cardiac Aging. Front Physiol 2021; 12:669497. [PMID: 33995129 PMCID: PMC8116539 DOI: 10.3389/fphys.2021.669497] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 04/07/2021] [Indexed: 11/13/2022] Open
Abstract
Aging is a process that can be accompanied by molecular and cellular alterations that compromise cardiac function. Although other metabolic disorders with increased prevalence in aged populations, such as diabetes mellitus, dyslipidemia, and hypertension, are associated with cardiovascular complications; aging-related cardiomyopathy has some unique features. Healthy hearts oxidize fatty acids, glucose, lactate, ketone bodies, and amino acids for producing energy. Under physiological conditions, cardiac mitochondria use fatty acids and carbohydrate mainly to generate ATP, 70% of which is derived from fatty acid oxidation (FAO). However, relative contribution of nutrients in ATP synthesis is altered in the aging heart with glucose oxidation increasing at the expense of FAO. Cardiac aging is also associated with impairment of mitochondrial abundance and function, resulting in accumulation of reactive oxygen species (ROS) and activation of oxidant signaling that eventually leads to further mitochondrial damage and aggravation of cardiac function. This review summarizes the main components of pathophysiology of cardiac aging, which pertain to cardiac metabolism, mitochondrial function, and systemic metabolic changes that affect cardiac function.
Collapse
Affiliation(s)
- Thomas Sithara
- Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Konstantinos Drosatos
- Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| |
Collapse
|
41
|
Lin KL, Chen SD, Lin KJ, Liou CW, Chuang YC, Wang PW, Chuang JH, Lin TK. Quality Matters? The Involvement of Mitochondrial Quality Control in Cardiovascular Disease. Front Cell Dev Biol 2021; 9:636295. [PMID: 33829016 PMCID: PMC8019794 DOI: 10.3389/fcell.2021.636295] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 03/02/2021] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular diseases are one of the leading causes of death and global health problems worldwide. Multiple factors are known to affect the cardiovascular system from lifestyles, genes, underlying comorbidities, and age. Requiring high workload, metabolism of the heart is largely dependent on continuous power supply via mitochondria through effective oxidative respiration. Mitochondria not only serve as cellular power plants, but are also involved in many critical cellular processes, including the generation of intracellular reactive oxygen species (ROS) and regulating cellular survival. To cope with environmental stress, mitochondrial function has been suggested to be essential during bioenergetics adaptation resulting in cardiac pathological remodeling. Thus, mitochondrial dysfunction has been advocated in various aspects of cardiovascular pathology including the response to ischemia/reperfusion (I/R) injury, hypertension (HTN), and cardiovascular complications related to type 2 diabetes mellitus (DM). Therefore, mitochondrial homeostasis through mitochondrial dynamics and quality control is pivotal in the maintenance of cardiac health. Impairment of the segregation of damaged components and degradation of unhealthy mitochondria through autophagic mechanisms may play a crucial role in the pathogenesis of various cardiac disorders. This article provides in-depth understanding of the current literature regarding mitochondrial remodeling and dynamics in cardiovascular diseases.
Collapse
Affiliation(s)
- Kai-Lieh Lin
- Center for Mitochondrial Research and Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan.,Department of Anesthesiology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Shang-Der Chen
- Center for Mitochondrial Research and Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan.,Department of Neurology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan.,Center of Parkinson's Disease, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Kai-Jung Lin
- Center for Mitochondrial Research and Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Chia-Wei Liou
- Center for Mitochondrial Research and Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan.,Department of Neurology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan.,Center of Parkinson's Disease, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Yao-Chung Chuang
- Center for Mitochondrial Research and Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan.,Department of Neurology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan.,Center of Parkinson's Disease, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Pei-Wen Wang
- Center for Mitochondrial Research and Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan.,Department of Metabolism, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Jiin-Haur Chuang
- Center for Mitochondrial Research and Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan.,Department of Pediatric Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Tsu-Kung Lin
- Center for Mitochondrial Research and Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan.,Department of Neurology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan.,Center of Parkinson's Disease, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| |
Collapse
|
42
|
Saw EL, Pearson JT, Schwenke DO, Munasinghe PE, Tsuchimochi H, Rawal S, Coffey S, Davis P, Bunton R, Van Hout I, Kai Y, Williams MJA, Kakinuma Y, Fronius M, Katare R. Activation of the cardiac non-neuronal cholinergic system prevents the development of diabetes-associated cardiovascular complications. Cardiovasc Diabetol 2021; 20:50. [PMID: 33618724 PMCID: PMC7898760 DOI: 10.1186/s12933-021-01231-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 01/29/2021] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Acetylcholine (ACh) plays a crucial role in the function of the heart. Recent evidence suggests that cardiomyocytes possess a non-neuronal cholinergic system (NNCS) that comprises of choline acetyltransferase (ChAT), choline transporter 1 (CHT1), vesicular acetylcholine transporter (VAChT), acetylcholinesterase (AChE) and type-2 muscarinic ACh receptors (M2AChR) to synthesize, release, degrade ACh as well as for ACh to transduce a signal. NNCS is linked to cardiac cell survival, angiogenesis and glucose metabolism. Impairment of these functions are hallmarks of diabetic heart disease (DHD). The role of the NNCS in DHD is unknown. The aim of this study was to examine the effect of diabetes on cardiac NNCS and determine if activation of cardiac NNCS is beneficial to the diabetic heart. METHODS Ventricular samples from type-2 diabetic humans and db/db mice were used to measure the expression pattern of NNCS components (ChAT, CHT1, VAChT, AChE and M2AChR) and glucose transporter-4 (GLUT-4) by western blot analysis. To determine the function of the cardiac NNCS in the diabetic heart, a db/db mouse model with cardiac-specific overexpression of ChAT gene was generated (db/db-ChAT-tg). Animals were followed up serially and samples collected at different time points for molecular and histological analysis of cardiac NNCS components and prosurvival and proangiogenic signaling pathways. RESULTS Immunoblot analysis revealed alterations in the components of cardiac NNCS and GLUT-4 in the type-2 diabetic human and db/db mouse hearts. Interestingly, the dysregulation of cardiac NNCS was followed by the downregulation of GLUT-4 in the db/db mouse heart. Db/db-ChAT-tg mice exhibited preserved cardiac and vascular function in comparison to db/db mice. The improved function was associated with increased cardiac ACh and glucose content, sustained angiogenesis and reduced fibrosis. These beneficial effects were associated with upregulation of the PI3K/Akt/HIF1α signaling pathway, and increased expression of its downstream targets-GLUT-4 and VEGF-A. CONCLUSION We provide the first evidence for dysregulation of the cardiac NNCS in DHD. Increased cardiac ACh is beneficial and a potential new therapeutic strategy to prevent or delay the development of DHD.
Collapse
Affiliation(s)
- Eng Leng Saw
- Department of Physiology, HeartOtago, School of Biomedical Sciences, University of Otago, 270, Great King Street, Dunedin, 9016, New Zealand
| | - James T Pearson
- Department of Cardiac Physiology, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan
- Biomedicine Discovery Institute and Department of Physiology, Monash University, Melbourne, VIC, Australia
| | - Daryl O Schwenke
- Department of Physiology, HeartOtago, School of Biomedical Sciences, University of Otago, 270, Great King Street, Dunedin, 9016, New Zealand
| | - Pujika Emani Munasinghe
- Department of Physiology, HeartOtago, School of Biomedical Sciences, University of Otago, 270, Great King Street, Dunedin, 9016, New Zealand
| | - Hirotsugu Tsuchimochi
- Department of Cardiac Physiology, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan
| | - Shruti Rawal
- Department of Physiology, HeartOtago, School of Biomedical Sciences, University of Otago, 270, Great King Street, Dunedin, 9016, New Zealand
| | - Sean Coffey
- Department of Medicine, School of Medicine, University of Otago, Dunedin, New Zealand
| | - Philip Davis
- Department of Cardiothoracic Surgery, School of Medicine, University of Otago, Dunedin, New Zealand
| | - Richard Bunton
- Department of Cardiothoracic Surgery, School of Medicine, University of Otago, Dunedin, New Zealand
| | - Isabelle Van Hout
- Department of Physiology, HeartOtago, School of Biomedical Sciences, University of Otago, 270, Great King Street, Dunedin, 9016, New Zealand
| | - Yuko Kai
- Department of Bioregulatory Science, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Michael J A Williams
- Department of Medicine, School of Medicine, University of Otago, Dunedin, New Zealand
| | - Yoshihiko Kakinuma
- Department of Bioregulatory Science, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan.
| | - Martin Fronius
- Department of Physiology, HeartOtago, School of Biomedical Sciences, University of Otago, 270, Great King Street, Dunedin, 9016, New Zealand.
| | - Rajesh Katare
- Department of Physiology, HeartOtago, School of Biomedical Sciences, University of Otago, 270, Great King Street, Dunedin, 9016, New Zealand.
| |
Collapse
|
43
|
Pasqua T, Rocca C, Giglio A, Angelone T. Cardiometabolism as an Interlocking Puzzle between the Healthy and Diseased Heart: New Frontiers in Therapeutic Applications. J Clin Med 2021; 10:721. [PMID: 33673114 PMCID: PMC7918460 DOI: 10.3390/jcm10040721] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/05/2021] [Accepted: 02/07/2021] [Indexed: 12/14/2022] Open
Abstract
Cardiac metabolism represents a crucial and essential connecting bridge between the healthy and diseased heart. The cardiac muscle, which may be considered an omnivore organ with regard to the energy substrate utilization, under physiological conditions mainly draws energy by fatty acids oxidation. Within cardiomyocytes and their mitochondria, through well-concerted enzymatic reactions, substrates converge on the production of ATP, the basic chemical energy that cardiac muscle converts into mechanical energy, i.e., contraction. When a perturbation of homeostasis occurs, such as an ischemic event, the heart is forced to switch its fatty acid-based metabolism to the carbohydrate utilization as a protective mechanism that allows the maintenance of its key role within the whole organism. Consequently, the flexibility of the cardiac metabolic networks deeply influences the ability of the heart to respond, by adapting to pathophysiological changes. The aim of the present review is to summarize the main metabolic changes detectable in the heart under acute and chronic cardiac pathologies, analyzing possible therapeutic targets to be used. On this basis, cardiometabolism can be described as a crucial mechanism in keeping the physiological structure and function of the heart; furthermore, it can be considered a promising goal for future pharmacological agents able to appropriately modulate the rate-limiting steps of heart metabolic pathways.
Collapse
Affiliation(s)
- Teresa Pasqua
- Department of Health Science, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy;
| | - Carmine Rocca
- Laboratory of Cellular and Molecular Cardiovascular Pathophysiology, Department of Biology, E. and E.S. (Di.B.E.S.T.), University of Calabria, 87036 Rende (CS), Italy
| | - Anita Giglio
- Department of Biology, E. and E.S. (Di.B.E.S.T.), University of Calabria, 87036 Rende (CS), Italy;
| | - Tommaso Angelone
- Laboratory of Cellular and Molecular Cardiovascular Pathophysiology, Department of Biology, E. and E.S. (Di.B.E.S.T.), University of Calabria, 87036 Rende (CS), Italy
- National Institute of Cardiovascular Research (I.N.R.C.), 40126 Bologna, Italy
| |
Collapse
|
44
|
Kyriazis ID, Hoffman M, Gaignebet L, Lucchese AM, Markopoulou E, Palioura D, Wang C, Bannister TD, Christofidou-Solomidou M, Oka SI, Sadoshima J, Koch WJ, Goldberg IJ, Yang VW, Bialkowska AB, Kararigas G, Drosatos K. KLF5 Is Induced by FOXO1 and Causes Oxidative Stress and Diabetic Cardiomyopathy. Circ Res 2021; 128:335-357. [PMID: 33539225 PMCID: PMC7870005 DOI: 10.1161/circresaha.120.316738] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 11/30/2020] [Indexed: 12/12/2022]
Abstract
RATIONALE Diabetic cardiomyopathy (DbCM) is a major complication in type-1 diabetes, accompanied by altered cardiac energetics, impaired mitochondrial function, and oxidative stress. Previous studies indicate that type-1 diabetes is associated with increased cardiac expression of KLF5 (Krüppel-like factor-5) and PPARα (peroxisome proliferator-activated receptor) that regulate cardiac lipid metabolism. OBJECTIVE In this study, we investigated the involvement of KLF5 in DbCM and its transcriptional regulation. METHODS AND RESULTS KLF5 mRNA levels were assessed in isolated cardiomyocytes from cardiovascular patients with diabetes and were higher compared with nondiabetic individuals. Analyses in human cells and diabetic mice with cardiomyocyte-specific FOXO1 (Forkhead box protein O1) deletion showed that FOXO1 bound directly on the KLF5 promoter and increased KLF5 expression. Diabetic mice with cardiomyocyte-specific FOXO1 deletion had lower cardiac KLF5 expression and were protected from DbCM. Genetic, pharmacological gain and loss of KLF5 function approaches and AAV (adeno-associated virus)-mediated Klf5 delivery in mice showed that KLF5 induces DbCM. Accordingly, the protective effect of cardiomyocyte FOXO1 ablation in DbCM was abolished when KLF5 expression was rescued. Similarly, constitutive cardiomyocyte-specific KLF5 overexpression caused cardiac dysfunction. KLF5 caused oxidative stress via direct binding on NADPH oxidase (NOX)4 promoter and induction of NOX4 (NADPH oxidase 4) expression. This was accompanied by accumulation of cardiac ceramides. Pharmacological or genetic KLF5 inhibition alleviated superoxide formation, prevented ceramide accumulation, and improved cardiac function in diabetic mice. CONCLUSIONS Diabetes-mediated activation of cardiomyocyte FOXO1 increases KLF5 expression, which stimulates NOX4 expression, ceramide accumulation, and causes DbCM.
Collapse
Affiliation(s)
- Ioannis D. Kyriazis
- Lewis Katz School of Medicine at Temple University, Center for Translational Medicine, Philadelphia, PA, 19131, USA
| | - Matthew Hoffman
- Lewis Katz School of Medicine at Temple University, Center for Translational Medicine, Philadelphia, PA, 19131, USA
| | - Lea Gaignebet
- Charité – Universitätsmedizin Berlin, Berlin 10115, Germany
| | - Anna Maria Lucchese
- Lewis Katz School of Medicine at Temple University, Center for Translational Medicine, Philadelphia, PA, 19131, USA
| | - Eftychia Markopoulou
- Lewis Katz School of Medicine at Temple University, Center for Translational Medicine, Philadelphia, PA, 19131, USA
| | - Dimitra Palioura
- Lewis Katz School of Medicine at Temple University, Center for Translational Medicine, Philadelphia, PA, 19131, USA
| | - Chao Wang
- The Scripps Research Institute, Jupiter, FL, 33458m USA
| | | | - Melpo Christofidou-Solomidou
- Pulmonary, Allergy, and Critical Care Division, University of Pennsylvania School of Medicine, Philadelphia, PA, 19104, USA
| | - Shin-ichi Oka
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ, 07101, USA
| | - Junichi Sadoshima
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ, 07101, USA
| | - Walter J. Koch
- Lewis Katz School of Medicine at Temple University, Center for Translational Medicine, Philadelphia, PA, 19131, USA
| | - Ira J. Goldberg
- Division of Endocrinology, Diabetes and Metabolism, New York University School of Medicine, New York, NY, 10016, USA
| | - Vincent W. Yang
- School of Medicine, Stony Brook University, Stony Brook, NY, 11794, USA
| | | | - Georgios Kararigas
- Charité – Universitätsmedizin Berlin, Berlin 10115, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin 10785, Germany
- Department of Physiology, Faculty of Medicine, University of Iceland, 101 Reykjavík, Iceland
| | - Konstantinos Drosatos
- Lewis Katz School of Medicine at Temple University, Center for Translational Medicine, Philadelphia, PA, 19131, USA
| |
Collapse
|
45
|
Effects of Three-Month Feeding High Fat Diets with Different Fatty Acid Composition on Myocardial Proteome in Mice. Nutrients 2021; 13:nu13020330. [PMID: 33498641 PMCID: PMC7911225 DOI: 10.3390/nu13020330] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/17/2021] [Accepted: 01/20/2021] [Indexed: 01/04/2023] Open
Abstract
Westernized diet is characterized by a high content of saturated fatty acids (SFA) and a low level of omega-3 polyunsaturated fatty acids (PUFA), often accompanied by an imbalance in the omega-6/omega-3 PUFA ratio. Since increased intake of SFA and n-6 PUFA is considered as a cardiovascular disease risk factor, this study was conducted to determine whether a three-month dietary supplementation of high-fat diets (HFDs) with saturated fatty acids and a significant proportion of various n-6 and n-3 PUFA ratios would affect the architecture and protein expression patterns of the murine heart. Therefore, three HFD (n = 6) feeding groups: rich in SFA, dominated by PUFA with the n-6/n-3–14:1, and n-6/n-3–5:1, ratios were compared to animals fed standard mouse chow. For this purpose, we performed two-dimensional electrophoresis with MALDI-ToF mass spectrometry-based identification of differentially expressed cardiac proteins, and a histological examination of cardiac morphology. The results indicated that mice fed with all HFDs developed signs of hypertrophy and cardiac fibrosis. Animals fed SFA-rich HFD manifested the most severe cardiac hypertrophy and fibrosis lesions, whereas less pronounced changes were observed in the group of animals that ingested the highest amount of omega-3 FA. In general, all HFDs, regardless of FA composition, evoked a comparable pattern of cardiac protein changes and affected the following biological processes: lipid metabolism and FA β-oxidation, glycolysis, TCA cycle, respiratory chain, myocardium contractility, oxidative stress and PUFA eicosanoid metabolism. However, it should be noted that three proteins, namely IDH3A, LDHB, and AK1, were affected differently by various FA contents. High expression of these myocardial proteins found in the group of animals fed a HFD with the highest n-3 PUFA content could be closely related to the observed development of hypertrophy.
Collapse
|
46
|
Kuo MW, Tsai HH, Wang SH, Chen YY, Yu AL, Yu J. Yulink, predicted from evolutionary analysis, is involved in cardiac function. J Biomed Sci 2021; 28:7. [PMID: 33423678 PMCID: PMC7798328 DOI: 10.1186/s12929-020-00701-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 12/21/2020] [Indexed: 11/11/2022] Open
Abstract
Background The comparative evolutionary genomics analysis was used to study the functions of novel Ka/Ks-predicted human exons in a zebrafish model. The Yulink (MIOS, Entrez Gene: 54,468), a conserved gene from zebrafish to human with WD40 repeats at N-terminus, was identified and found to encode an 875 amino acid in human. The biological function of this Yulink gene in cardiomyocytes remains unexplored. The purpose of this study is to determine the involvement of Yulink in the functions of cardiomyocytes and to investigate its molecular regulatory mechanism. Methods Knockdown of Yulink was performed using morpholino or shRNA in zebrafish, mouse HL-1 cardiomyocytes, and human iPSC-derived cardiomyocytes. The expression levels of mRNA and protein were quantified by qPCR and western blots. Other methods including DNA binding, ligand uptake, agonists treatment and Ca2+ imaging assays were used to study the molecular regulatory mechanism by Yulink. Statistical data were shown as mean ± SD or mean ± standard error. Results The knockdown of yulink with three specific morpholinos in zebrafish resulted in cardiac dysfunctions with pericardial edema, decreased heart beats and cardiac output. The Yulink knockdown in mouse HL-1 cardiomyocytes disrupted Ca2+ cycling, reduced DNA binding activity of PPARγ (peroxisome proliferator-activated receptor gamma) and resulted in a reduction of Serca2 (sarcoplasmic reticulum Ca2+ ATPase 2) expression. Expression of Serca2 was up-regulated by PPARγ agonists and down-regulated by PPARγ-shRNA knockdown, suggesting that Yulink regulates SERCA2 expression through PPARγ in mouse HL-1 cardiomyocytes. On the other hand, YULINK, PPARγ or SERCA2 over-expression rescued the phenotypes of Yulink KD cells. In addition, knockdown of YULINK in human iPSC-derived cardiomyocytes also disrupted Ca2+ cycling via decreased SERCA2 expression. Conclusions Overall, our data showed that Yulink is an evolutionarily conserved gene from zebrafish to human. Mechanistically Yulink regulated Serca2 expression in cardiomyocytes, presumably mediated through PPARγ nuclear entry. Deficiency of Yulink in mouse and human cardiomyocytes resulted in irregular Ca2+ cycling, which may contribute to arrhythmogenesis.
Collapse
Affiliation(s)
- Ming-Wei Kuo
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Taoyuan, 333, Taiwan
| | - Hsiu-Hui Tsai
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Taoyuan, 333, Taiwan
| | - Sheng-Hung Wang
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Taoyuan, 333, Taiwan
| | - Yi-Yin Chen
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Taoyuan, 333, Taiwan
| | - Alice L Yu
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Taoyuan, 333, Taiwan.,Department of Pediatrics, University of California, San Diego, CA, USA
| | - John Yu
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Taoyuan, 333, Taiwan. .,Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan.
| |
Collapse
|
47
|
Wang L, Cai Y, Jian L, Cheung CW, Zhang L, Xia Z. Impact of peroxisome proliferator-activated receptor-α on diabetic cardiomyopathy. Cardiovasc Diabetol 2021; 20:2. [PMID: 33397369 PMCID: PMC7783984 DOI: 10.1186/s12933-020-01188-0] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 12/02/2020] [Indexed: 12/21/2022] Open
Abstract
The prevalence of cardiomyopathy is higher in diabetic patients than those without diabetes. Diabetic cardiomyopathy (DCM) is defined as a clinical condition of abnormal myocardial structure and performance in diabetic patients without other cardiac risk factors, such as coronary artery disease, hypertension, and significant valvular disease. Multiple molecular events contribute to the development of DCM, which include the alterations in energy metabolism (fatty acid, glucose, ketone and branched chain amino acids) and the abnormalities of subcellular components in the heart, such as impaired insulin signaling, increased oxidative stress, calcium mishandling and inflammation. There are no specific drugs in treating DCM despite of decades of basic and clinical investigations. This is, in part, due to the lack of our understanding as to how heart failure initiates and develops, especially in diabetic patients without an underlying ischemic cause. Some of the traditional anti-diabetic or lipid-lowering agents aimed at shifting the balance of cardiac metabolism from utilizing fat to glucose have been shown inadequately targeting multiple aspects of the conditions. Peroxisome proliferator-activated receptor α (PPARα), a transcription factor, plays an important role in mediating DCM-related molecular events. Pharmacological targeting of PPARα activation has been demonstrated to be one of the important strategies for patients with diabetes, metabolic syndrome, and atherosclerotic cardiovascular diseases. The aim of this review is to provide a contemporary view of PPARα in association with the underlying pathophysiological changes in DCM. We discuss the PPARα-related drugs in clinical applications and facts related to the drugs that may be considered as risky (such as fenofibrate, bezafibrate, clofibrate) or safe (pemafibrate, metformin and glucagon-like peptide 1-receptor agonists) or having the potential (sodium-glucose co-transporter 2 inhibitor) in treating DCM.
Collapse
Affiliation(s)
- Lin Wang
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
- Department of Anaesthesiology, The University of Hong Kong, Hong Kong, SAR, China
| | - Yin Cai
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
- Department of Anaesthesiology, The University of Hong Kong, Hong Kong, SAR, China
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, SAR, China
| | - Liguo Jian
- Department of Cardiology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chi Wai Cheung
- Department of Anaesthesiology, The University of Hong Kong, Hong Kong, SAR, China
| | - Liangqing Zhang
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China.
| | - Zhengyuan Xia
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China.
- Department of Anaesthesiology, The University of Hong Kong, Hong Kong, SAR, China.
| |
Collapse
|
48
|
Du H, Zhao Y, Yin Z, Wang DW, Chen C. The role of miR-320 in glucose and lipid metabolism disorder-associated diseases. Int J Biol Sci 2021; 17:402-416. [PMID: 33613101 PMCID: PMC7893589 DOI: 10.7150/ijbs.53419] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 11/21/2020] [Indexed: 02/06/2023] Open
Abstract
Glucose and lipids are important nutrients that provide the majority of energy for each organ to maintain homeostasis of the body. With the continuous improvement in living standards, the incidence of metabolic disorder-associated diseases, such as diabetes, hyperlipidemia, and atherosclerosis, is increasing worldwide. Among them, diabetes, which could be induced by both glucose and lipid metabolic disorders, is one of the five diseases with the highest incidence and mortality worldwide. However, the detailed molecular mechanisms underlying glucose and lipid metabolism disorders and target-organ damage are still not fully defined. MicroRNAs (miRNAs) are small, non-coding, single-stranded RNAs, which usually affect their target mRNAs in the cytoplasm by post-transcriptional regulation. Previously, we have found that miR-320 contributed to glucose and lipid metabolism via different signaling pathways. Most importantly, we identified that nuclear miR-320 mediated diabetes-induced cardiac dysfunction by activating the transcription of fatty acid metabolic genes to cause lipotoxicity in the heart. Here, we reviewed the roles of miR-320 in glucose and lipid metabolism and target-organ damage.
Collapse
Affiliation(s)
| | | | | | | | - Chen Chen
- Division of Cardiology, Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
49
|
ALTamimi JZ, BinMowyna MN, AlFaris NA, Alagal RI, El-kott AF, AL-Farga AM. Fisetin protects against streptozotocin-induced diabetic cardiomyopathy in rats by suppressing fatty acid oxidation and inhibiting protein kinase R. Saudi Pharm J 2021; 29:27-42. [PMID: 33603537 PMCID: PMC7873759 DOI: 10.1016/j.jsps.2020.12.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Accepted: 12/03/2020] [Indexed: 01/04/2023] Open
Abstract
This study examined if the Fisetin against streptozotocin-induced diabetic cardiomyopathy (DC) in rats involves regulating cardiac metabolism and suppressing protein kinase R (PKR). Male rats were divided (12/groups) as control (non-diabetic), control + Fisetin, T1DM, and T1DM + Fisetin. Fisetin was administered orally at a final dose of 2.5 mg/kg for 12 weeks. In T1DM1-induced rats, Fisetin prevented heart and final body weights loss, lowered circulatory levels troponin I and creatinine kinase-MB (CK-MB), increased fasting insulin levels, and improved ventricular systolic and diastolic functions. It also preserved the structure of the cardiomyocytes and reduced oxidative stress, fibrosis, protein levels of transforming growth factor-β1 (TGF-β1), collagenase 1A, caspase-3, and the activation of JNK, p53, and p38 MAPK. In the control and diabetic rats, Fisetin attenuated fasting hyperglycaemia, the increases in glucose levels after the oral and insulin tolerance tests, and HOMA-IR. It also increased cardiac glucose oxidation by increasing the activity of private dehydrogenase (PDH), phosphofructokinase (PFK), protein levels of PPAR-α and suppressed cardiac inflammation by inhibiting NF-κB. These effects were associated with a reduction in the activity of PKR and subsequent increase in the activity of eeukaryotic initiation factor 2 (eIF2) with a parallel increase in protein levels of p67, a cellular inhibitor of PKR. In cultured cardiomyocytes, Fisetin, prevented high glucose (HG)-induced activation of PKR and reduction in p67, in a dose-dependent manner. However, the effect of Fisetin on PKR was diminished in LG and HG-treated cardiomyocytes with p67-siRNA. In conclusion, Fisetin protects against DC in rats by improving cardiac glucose metabolism and suppressing PKR.
Collapse
Affiliation(s)
- Jozaa Z. ALTamimi
- Nutrition and Food Science (PHD), Department of Physical Sport Science, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Mona N. BinMowyna
- College of Applied Medical Sciences, Shaqra University, Shaqra, Saudi Arabia
| | - Nora A. AlFaris
- Nutrition and Food Science (PHD), Department of Physical Sport Science, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Reham I. Alagal
- Nutrition and Food Science (PHD), Department of Physical Sport Science, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Attalla F. El-kott
- Department of Biology, College of Science, King Khalid University, Abha, Saudi Arabia
| | - Ammar M. AL-Farga
- Department of Biochemistry, College of Sciences, University of Jeddah, Jeddah, Saudi Arabia
| |
Collapse
|
50
|
Bo B, Zhou Y, Zheng Q, Wang G, Zhou K, Wei J. The Molecular Mechanisms Associated with Aerobic Exercise-Induced Cardiac Regeneration. Biomolecules 2020; 11:biom11010019. [PMID: 33375497 PMCID: PMC7823705 DOI: 10.3390/biom11010019] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/20/2020] [Accepted: 12/24/2020] [Indexed: 12/13/2022] Open
Abstract
The leading cause of heart failure is cardiomyopathy and damage to the cardiomyocytes. Adult mammalian cardiomyocytes have the ability to regenerate, but this cannot wholly compensate for myocardial cell loss after myocardial injury. Studies have shown that exercise has a regulatory role in the activation and promotion of regeneration of healthy and injured adult cardiomyocytes. However, current research on the effects of aerobic exercise in myocardial regeneration is not comprehensive. This review discusses the relationships between aerobic exercise and the regeneration of cardiomyocytes with respect to complex molecular and cellular mechanisms, paracrine factors, transcriptional factors, signaling pathways, and microRNAs that induce cardiac regeneration. The topics discussed herein provide a knowledge base for physical activity-induced cardiomyocyte regeneration, in which exercise enhances overall heart function and improves the efficacy of cardiac rehabilitation.
Collapse
Affiliation(s)
- Bing Bo
- School of Physical Education, Henan University, Kaifeng 475001, Henan, China; (B.B.); (Y.Z.); (Q.Z.); (G.W.); (K.Z.)
- Sports Reform and Development Research Center, Henan University, Kaifeng 475001, Henan, China
- School of Life Sciences, Henan University, Kaifeng 475001, Henan, China
| | - Yang Zhou
- School of Physical Education, Henan University, Kaifeng 475001, Henan, China; (B.B.); (Y.Z.); (Q.Z.); (G.W.); (K.Z.)
| | - Qingyun Zheng
- School of Physical Education, Henan University, Kaifeng 475001, Henan, China; (B.B.); (Y.Z.); (Q.Z.); (G.W.); (K.Z.)
- Sports Reform and Development Research Center, Henan University, Kaifeng 475001, Henan, China
| | - Guandong Wang
- School of Physical Education, Henan University, Kaifeng 475001, Henan, China; (B.B.); (Y.Z.); (Q.Z.); (G.W.); (K.Z.)
| | - Ke Zhou
- School of Physical Education, Henan University, Kaifeng 475001, Henan, China; (B.B.); (Y.Z.); (Q.Z.); (G.W.); (K.Z.)
- Sports Reform and Development Research Center, Henan University, Kaifeng 475001, Henan, China
| | - Jianshe Wei
- School of Life Sciences, Henan University, Kaifeng 475001, Henan, China
- Correspondence: ; Tel.: +86-13938625812
| |
Collapse
|