1
|
Xiang KF, Wan JJ, Wang PY, Liu X. Role of glycogen in cardiac metabolic stress. Metabolism 2024; 162:156059. [PMID: 39500406 DOI: 10.1016/j.metabol.2024.156059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 10/22/2024] [Accepted: 10/30/2024] [Indexed: 11/11/2024]
Abstract
Metabolic stress in the myocardium arises from a diverse array of acute and chronic pathophysiological contexts. Glycogen mishandling is a key feature of metabolic stress, while maladaptation in energy-stress situations confers functional deficits. Cardiac glycogen serves as a pivotal reserve for myocardial energy, which is classically described as an energy source and contributes to glucose homeostasis during hypoxia or ischemia. Despite extensive research activity, how glycogen metabolism affects cardiovascular disease remains unclear. In this review, we focus on its regulation across myocardial energy metabolism in response to stress, and its role in metabolism, immunity, and autophagy. We further summarize the cardiovascular-related drugs regulating glycogen metabolism. In this way, we provide current knowledge for the understanding of glycogen metabolism in the myocardium.
Collapse
Affiliation(s)
- Ke-Fa Xiang
- Department of Clinical Pharmacy, School of Pharmacy, Second Military Medical University, Shanghai, China; Department of Cardiology, The 72nd Group Army Hospital, Huzhou University, Huzhou, Zhejiang 313000, China
| | - Jing-Jing Wan
- Department of Clinical Pharmacy, School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Peng-Yuan Wang
- Department of Clinical Pharmacy, School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Xia Liu
- Department of Clinical Pharmacy, School of Pharmacy, Second Military Medical University, Shanghai, China.
| |
Collapse
|
2
|
Zhang J, Li X, Cui W, Lu D, Zhang Y, Liao J, Guo L, Jiao C, Tao L, Xu Y, Shen X. 1,8-cineole ameliorates experimental diabetic angiopathy by inhibiting NLRP3 inflammasome-mediated pyroptosis in HUVECs via SIRT2. Biomed Pharmacother 2024; 177:117085. [PMID: 38972150 DOI: 10.1016/j.biopha.2024.117085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 06/29/2024] [Accepted: 07/01/2024] [Indexed: 07/09/2024] Open
Abstract
Accumulating evidence strongly support the key role of NLRP3-mediated pyroptosis in the pathogenesis and progression of vascular endothelial dysfunction associated with diabetes mellitus. Various studies have demonstrated that the activation or upregulation of Silent Information Regulation 2 homolog 2 (SIRT2) exerts inhibitory effect on the expression of NLRP3. Although 1,8-cineole has been found to protect against endothelial dysfunction and cardiovascular diseases, its role and mechanism in diabetic angiopathy remain unknown. Therefore, the aim of this study was to investigate the ameliorative effect of 1,8-cineole through SIRT2 on pyroptosis associated with diabetic angiopathy in human umbilical vein endothelial cells (HUVECs) and to elucidate the underlying mechanism. The findings revealed that 1,8-cineole exhibited a protective effect against vascular injury and ameliorated pathological alterations in the thoracic aorta of diabetic mice. Moreover, it effectively mitigated pyroptosis induced by palmitic acid-high glucose (PA-HG) in HUVECs. Treatment with 1,8-cineole effectively restored the reduced levels of SIRT2 and suppressed the elevated expression of pyroptosis-associated proteins. Additionally, our findings demonstrated the occurrence of NLRP3 deacetylation and the physical interaction between NLRP3 and SIRT2. The SIRT2 inhibitor AGK2 and siRNA-SIRT2 effectively attenuated the effect of 1,8-cineole on NLRP3 deacetylation in HUVECs and compromised its inhibitory effect against pyroptosis in HUVECs. However, overexpression of SIRT2 inhibited PA-HG-induced pyroptosis in HUVECs. 1,8-Cineole inhibited the deacetylation of NLRP3 by regulating SIRT2, thereby reducing pyroptosis in HUVECs. In conclusion, our findings suggest that PA-HG-induced pyroptosis in HUVECs plays a crucial role in the development of diabetic angiopathy, which can be mitigated by 1,8-cineole.
Collapse
Affiliation(s)
- Jian Zhang
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, No.6 Ankang Avenue,Guiyang city and Guian New District, Guizhou 561113, China; The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province (The high educational key laboratory of Guizhou province for natural medicianl Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guiyang city and Guian New District, Guizhou 561113, China; The Key Laboratory of Optimal Utilization of Natural Medicine Resources,The Department of Pharmacology of Materia Medica, School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guiyang city and Guian New District, Guizhou 561113, China
| | - Xinlin Li
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, No.6 Ankang Avenue,Guiyang city and Guian New District, Guizhou 561113, China; The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province (The high educational key laboratory of Guizhou province for natural medicianl Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guiyang city and Guian New District, Guizhou 561113, China; The Key Laboratory of Optimal Utilization of Natural Medicine Resources,The Department of Pharmacology of Materia Medica, School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guiyang city and Guian New District, Guizhou 561113, China
| | - Wenqing Cui
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, No.6 Ankang Avenue,Guiyang city and Guian New District, Guizhou 561113, China; The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province (The high educational key laboratory of Guizhou province for natural medicianl Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guiyang city and Guian New District, Guizhou 561113, China; The Key Laboratory of Optimal Utilization of Natural Medicine Resources,The Department of Pharmacology of Materia Medica, School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guiyang city and Guian New District, Guizhou 561113, China
| | - Dingchun Lu
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, No.6 Ankang Avenue,Guiyang city and Guian New District, Guizhou 561113, China; The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province (The high educational key laboratory of Guizhou province for natural medicianl Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guiyang city and Guian New District, Guizhou 561113, China; The Key Laboratory of Optimal Utilization of Natural Medicine Resources,The Department of Pharmacology of Materia Medica, School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guiyang city and Guian New District, Guizhou 561113, China
| | - Yanyan Zhang
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, No.6 Ankang Avenue,Guiyang city and Guian New District, Guizhou 561113, China; The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province (The high educational key laboratory of Guizhou province for natural medicianl Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guiyang city and Guian New District, Guizhou 561113, China; The Key Laboratory of Optimal Utilization of Natural Medicine Resources,The Department of Pharmacology of Materia Medica, School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guiyang city and Guian New District, Guizhou 561113, China
| | - Jiajia Liao
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, No.6 Ankang Avenue,Guiyang city and Guian New District, Guizhou 561113, China; The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province (The high educational key laboratory of Guizhou province for natural medicianl Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guiyang city and Guian New District, Guizhou 561113, China; The Key Laboratory of Optimal Utilization of Natural Medicine Resources,The Department of Pharmacology of Materia Medica, School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guiyang city and Guian New District, Guizhou 561113, China
| | - Linlin Guo
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, No.6 Ankang Avenue,Guiyang city and Guian New District, Guizhou 561113, China; The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province (The high educational key laboratory of Guizhou province for natural medicianl Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guiyang city and Guian New District, Guizhou 561113, China; The Key Laboratory of Optimal Utilization of Natural Medicine Resources,The Department of Pharmacology of Materia Medica, School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guiyang city and Guian New District, Guizhou 561113, China
| | - Chunen Jiao
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, No.6 Ankang Avenue,Guiyang city and Guian New District, Guizhou 561113, China; The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province (The high educational key laboratory of Guizhou province for natural medicianl Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guiyang city and Guian New District, Guizhou 561113, China; The Key Laboratory of Optimal Utilization of Natural Medicine Resources,The Department of Pharmacology of Materia Medica, School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guiyang city and Guian New District, Guizhou 561113, China
| | - Ling Tao
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, No.6 Ankang Avenue,Guiyang city and Guian New District, Guizhou 561113, China; The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province (The high educational key laboratory of Guizhou province for natural medicianl Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guiyang city and Guian New District, Guizhou 561113, China
| | - Yini Xu
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, No.6 Ankang Avenue,Guiyang city and Guian New District, Guizhou 561113, China; The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province (The high educational key laboratory of Guizhou province for natural medicianl Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guiyang city and Guian New District, Guizhou 561113, China; The Key Laboratory of Optimal Utilization of Natural Medicine Resources,The Department of Pharmacology of Materia Medica, School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guiyang city and Guian New District, Guizhou 561113, China.
| | - Xiangchun Shen
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, No.6 Ankang Avenue,Guiyang city and Guian New District, Guizhou 561113, China; The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province (The high educational key laboratory of Guizhou province for natural medicianl Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guiyang city and Guian New District, Guizhou 561113, China; The Key Laboratory of Optimal Utilization of Natural Medicine Resources,The Department of Pharmacology of Materia Medica, School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guiyang city and Guian New District, Guizhou 561113, China.
| |
Collapse
|
3
|
Weng H, Zou W, Tian F, Xie H, Liu A, Liu W, Liu Y, Zhou N, Cai X, Wu J, Zheng Y, Shu X. Inhalable cardiac targeting peptide modified nanomedicine prevents pressure overload heart failure in male mice. Nat Commun 2024; 15:6058. [PMID: 39025877 PMCID: PMC11258261 DOI: 10.1038/s41467-024-50312-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 07/08/2024] [Indexed: 07/20/2024] Open
Abstract
Heart failure causes considerable morbidity and mortality worldwide. Clinically applied drugs for the treatment of heart failure are still severely limited by poor delivery efficiency to the heart and off-target consumption. Inspired by the high heart delivery efficiency of inhaled drugs, we present an inhalable cardiac-targeting peptide (CTP)-modified calcium phosphate (CaP) nanoparticle for the delivery of TP-10, a selective inhibitor of PDE10A. The CTP modification significantly promotes cardiomyocyte and fibroblast targeting during the pathological state of heart failure in male mice. TP-10 is subsequently released from TP-10@CaP-CTP and effectively attenuates cardiac remodelling and improved cardiac function. In view of these results, a low dosage (2.5 mg/kg/2 days) of inhaled medication exerted good therapeutic effects without causing severe lung injury after long-term treatment. In addition, the mechanism underlying the amelioration of heart failure is investigated, and the results reveal that the therapeutic effects of this system on cardiomyocytes and cardiac fibroblasts are mainly mediated through the cAMP/AMPK and cGMP/PKG signalling pathways. By demonstrating the targeting capacity of CTP and verifying the biosafety of inhalable CaP nanoparticles in the lung, this work provides a perspective for exploring myocardium-targeted therapy and presents a promising clinical strategy for the long-term management of heart failure.
Collapse
Affiliation(s)
- Haobo Weng
- Department of Echocardiography, Shanghai Institute of Medical Imaging, Zhongshan Hospital, Fudan University, Shanghai, PR China
- Shanghai Key Laboratory of Neuro-Ultrasound for Diagnosis and Treatment, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
- Department of Cardiology, Shanghai Institute of Cardiovascular Disease, Zhongshan Hospital, Fudan University, Shanghai, PR China
| | - Weijuan Zou
- Shanghai Key Laboratory of Neuro-Ultrasound for Diagnosis and Treatment, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
- Department of Ultrasound in Medicine, Shanghai Institute of Ultrasound in Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Fangyan Tian
- Department of Echocardiography, Shanghai Institute of Medical Imaging, Zhongshan Hospital, Fudan University, Shanghai, PR China
- Department of Cardiology, Shanghai Institute of Cardiovascular Disease, Zhongshan Hospital, Fudan University, Shanghai, PR China
- Department of Ultrasound Medicine, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Huilin Xie
- Department of Echocardiography, Shanghai Institute of Medical Imaging, Zhongshan Hospital, Fudan University, Shanghai, PR China
- Department of Cardiology, Shanghai Institute of Cardiovascular Disease, Zhongshan Hospital, Fudan University, Shanghai, PR China
| | - Ao Liu
- Department of Echocardiography, Shanghai Institute of Medical Imaging, Zhongshan Hospital, Fudan University, Shanghai, PR China
- Department of Cardiology, Shanghai Institute of Cardiovascular Disease, Zhongshan Hospital, Fudan University, Shanghai, PR China
| | - Wen Liu
- Department of Echocardiography, Shanghai Institute of Medical Imaging, Zhongshan Hospital, Fudan University, Shanghai, PR China
| | - Yu Liu
- Department of Echocardiography, Shanghai Institute of Medical Imaging, Zhongshan Hospital, Fudan University, Shanghai, PR China
| | - Nianwei Zhou
- Department of Echocardiography, Shanghai Institute of Medical Imaging, Zhongshan Hospital, Fudan University, Shanghai, PR China
| | - Xiaojun Cai
- Shanghai Key Laboratory of Neuro-Ultrasound for Diagnosis and Treatment, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
- Department of Ultrasound in Medicine, Shanghai Institute of Ultrasound in Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Jianrong Wu
- Shanghai Key Laboratory of Neuro-Ultrasound for Diagnosis and Treatment, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, PR China.
- Department of Ultrasound in Medicine, Shanghai Institute of Ultrasound in Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, PR China.
| | - Yuanyi Zheng
- Shanghai Key Laboratory of Neuro-Ultrasound for Diagnosis and Treatment, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, PR China.
- Department of Ultrasound in Medicine, Shanghai Institute of Ultrasound in Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, PR China.
| | - Xianhong Shu
- Department of Echocardiography, Shanghai Institute of Medical Imaging, Zhongshan Hospital, Fudan University, Shanghai, PR China.
- Department of Cardiology, Shanghai Institute of Cardiovascular Disease, Zhongshan Hospital, Fudan University, Shanghai, PR China.
- Department of Ultrasound in Medicine, Shanghai Xuhui District Central Hospital, Shanghai, PR China.
| |
Collapse
|
4
|
Yu Y, Su FF, Xu C. Maximakinin reversed H 2O 2 induced oxidative damage in rat cardiac H9c2 cells through AMPK/Akt and AMPK/ERK1/2 signaling pathways. Biomed Pharmacother 2024; 174:116489. [PMID: 38513595 DOI: 10.1016/j.biopha.2024.116489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 03/16/2024] [Accepted: 03/19/2024] [Indexed: 03/23/2024] Open
Abstract
Maximakinin (MK), a homolog of bradykinin (BK), is extracted from skin venom of the Chinese toad Bombina maxima. Although MK has a good antihypertensive effect, its effect on myocardial cells is unclear. This study investigates the protective effect of MK on hydrogen peroxide (H2O2)-induced oxidative damage in rat cardiac H9c2 cells and explores its mechanism of action. A 3-(4,5-Dimethyl-2-Thiazolyl)-2,5-Diphenyl Tetrazolium Bromide (MTT) assay was selected to detect the effect of MK on H9c2 cell viability, while flow cytometry was used to investigate the influence of MK and H2O2 on intracellular reactive oxygen species (ROS) levels. Protein expression changes were detected by western blot. In addition, specific protein inhibitors were applied to confirm the induction of ROS-related signaling pathways by MK. MTT assay results show that MK significantly reversed H2O2-induced cell growth inhibition. Flow cytometry Dichlorodihydrofluorescein diacetate (DCFH-DA) staining shows that MK significantly reversed H2O2-induced increases in intracellular ROS production in H9c2 cells. Moreover, the addition of specific protein inhibitors suggests that MK reverses H2O2-induced oxidative damage by activating AMP-activated protein kinase (AMPK)/protein kinase B (Akt) and AMPK/extracellular-regulated kinase 1/2 (ERK1/2) pathways. Finally, an inhibitor of bradykinin B2 receptors (B2Rs), HOE-140, was applied to investigate potential targets of MK in H9c2 cells. HOE-140 significantly blocked induction of AMPK/Akt and AMPK/ERK1/2 pathways by MK, suggesting a potentially important role for B2Rs in MK reversing H2O2-induced oxidative damage. Above all, MK protects against oxidative damage by inhibiting H2O2-induced ROS production in H9c2 cells. The protective mechanism of MK may be achieved by activation of B2Rs to activate downstream AMPK/Akt and AMPK/ERK1/2 pathways.
Collapse
Affiliation(s)
- Yang Yu
- Life Science and Biology Pharmacy College, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe, Liaoning, Shenyang 110016, China
| | - Fan-Fan Su
- Life Science and Biology Pharmacy College, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe, Liaoning, Shenyang 110016, China
| | - Cheng Xu
- Life Science and Biology Pharmacy College, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe, Liaoning, Shenyang 110016, China.
| |
Collapse
|
5
|
Zhang Z, Sun M, Jiang W, Yu L, Zhang C, Ma H. Myocardial Metabolic Reprogramming in HFpEF. J Cardiovasc Transl Res 2024; 17:121-132. [PMID: 37650988 DOI: 10.1007/s12265-023-10433-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Accepted: 08/22/2023] [Indexed: 09/01/2023]
Abstract
Heart failure (HF) caused by structural or functional cardiac abnormalities is a significant cause of morbidity and mortality worldwide. While HF with reduced ejection fraction (HErEF) is well understood, more than half of patients have HF with preserved ejection fraction (HFpEF). Currently, the treatment for HFpEF primarily focuses on symptom alleviation, lacking specific drugs. The stressed heart undergoes metabolic switches in substrate preference, which is a compensatory process involved in cardiac pathological remodeling. Although metabolic reprogramming in HF has gained attention in recent years, its role in HFpEF still requires further elucidation. In this review, we present a summary of cardiac mitochondrial dysfunction and cardiac metabolic reprogramming in HFpEF. Additionally, we emphasize potential therapeutic approaches that target metabolic reprogramming for the treatment of HFpEF.
Collapse
Affiliation(s)
- Zihui Zhang
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, People's Republic of China
| | - Mingchu Sun
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, People's Republic of China
| | - Wenhua Jiang
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, People's Republic of China
| | - Lu Yu
- Department of Pathology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Chan Zhang
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, People's Republic of China.
| | - Heng Ma
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, People's Republic of China.
- Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, 710032, People's Republic of China.
| |
Collapse
|
6
|
Yang G, Zhang Q, Dong C, Hou G, Li J, Jiang X, Xin Y. Nrf2 prevents diabetic cardiomyopathy via antioxidant effect and normalization of glucose and lipid metabolism in the heart. J Cell Physiol 2024; 239:e31149. [PMID: 38308838 DOI: 10.1002/jcp.31149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 10/11/2023] [Accepted: 10/25/2023] [Indexed: 02/05/2024]
Abstract
Metabolic disorders and oxidative stress are the main causes of diabetic cardiomyopathy. Activation of nuclear factor erythroid 2-related factor 2 (Nrf2) exerts a powerful antioxidant effect and prevents the progression of diabetic cardiomyopathy. However, the mechanism of its cardiac protection and direct action on cardiomyocytes are not well understood. Here, we investigated in a cardiomyocyte-restricted Nrf2 transgenic mice (Nrf2-TG) the direct effect of Nrf2 on cardiomyocytes in DCM and its mechanism. In this study, cardiomyocyte-restricted Nrf2 transgenic mice (Nrf2-TG) were used to directly observe whether cardiomyocyte-specific overexpression of Nrf2 can prevent diabetic cardiomyopathy and correct glucose and lipid metabolism disorders in the heart. Compared to wild-type mice, Nrf2-TG mice showed resistance to diabetic cardiomyopathy in a streptozotocin-induced type 1 diabetes mouse model. This was primarily manifested as improved echocardiography results as well as reduced myocardial fibrosis, cardiac inflammation, and oxidative stress. These results showed that Nrf2 can directly act on cardiomyocytes to exert a cardioprotective role. Mechanistically, the cardioprotective effects of Nrf2 depend on its antioxidation activity, partially through improving glucose and lipid metabolism by directly targeting lipid metabolic pathway of AMPK/Sirt1/PGC-1α activation via upstream genes of sestrin2 and LKB1, and indirectly enabling AKT/GSK-3β/HK-Ⅱ activity via AMPK mediated p70S6K inhibition.
Collapse
Affiliation(s)
- Ge Yang
- Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| | - Qihe Zhang
- Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| | - Chao Dong
- Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| | - Guowen Hou
- Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| | - Jinjie Li
- Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| | - Xin Jiang
- Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, and Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, Jilin, China
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun, Jilin, China
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, Jilin, China
| | - Ying Xin
- Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| |
Collapse
|
7
|
Hao Y, Li B, Huber SA, Liu W. Bibliometric analysis of trends in cardiac aging research over the past 20 years. Medicine (Baltimore) 2023; 102:e34870. [PMID: 37653740 PMCID: PMC10470686 DOI: 10.1097/md.0000000000034870] [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: 07/04/2023] [Revised: 07/31/2023] [Accepted: 08/01/2023] [Indexed: 09/02/2023] Open
Abstract
BACKGROUND In recent years, many studies have addressed cardiac aging and related diseases. This study aims to understand the research trend of cardiac aging and find new hot issues. METHODS We searched the web of science core collection database for articles published between 2003 and 2022 on the topic of "cardiac aging." Complete information including keywords, publication year, journal title, country, organization, and author were extracted for analysis. The VOS viewer software was used to generate network maps of keywords, countries, institutions, and author relationships for visual network analysis. RESULTS A total of 1002 papers were analyzed in the study. Overall, the number of annual publications on cardiac aging has increased since 2009, and new hot topics are emerging. The top 3 countries with the most publications were the United States (471 articles), China (209 articles) and Italy (101 articles). The University of Washington published the most papers (35 articles). The cluster analysis with author as the keyword found that the connections among different scholars are scattered and clustered in a small range. Network analysis based on keyword co-occurrence and year of publication identified relevant features and trends in cardiac aging research. According to the results of cluster analysis, all the articles are divided into 4 topics: "mechanisms of cardiac aging", "prevention and treatment of cardiac aging", "characteristics of cardiac aging", and "others." In recent years, the mechanism and treatment of cardiac aging have attracted the most attention. In both studies, animal models are used more often than in human populations. Mitochondrial dysfunction, autophagy and mitochondrial autophagy are hotspots in current research. CONCLUSION In this study, bibliometric analysis was used to analyze the research trend of cardiac aging in the past 20 years. The mechanism and treatment of cardiac aging are the most concerned contents. Mitochondrial dysfunction, autophagy and mitophagy are the focus of future research on cardiac aging.
Collapse
Affiliation(s)
- Yan Hao
- Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- Harbin Medical University, Harbin, Heilongjiang, China
| | - Bohan Li
- Harbin Medical University, Harbin, Heilongjiang, China
| | - Sally A. Huber
- Department of Pathology and Laboratory Medicine, University of Vermont, Colchester, VT
| | - Wei Liu
- Harbin Medical University, Harbin, Heilongjiang, China
- Department of Geriatric Cardiovascular Division, Guangdong Provincial Geriatrics Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| |
Collapse
|
8
|
Bruns DR, McNair BD, Peelor FF, Borowik AK, Pranay A, Yusifov A, Miller BF. Skeletal and cardiac muscle have different protein turnover responses in a model of right heart failure. GeroScience 2023; 45:2545-2557. [PMID: 37118350 PMCID: PMC10651599 DOI: 10.1007/s11357-023-00777-7] [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: 02/08/2023] [Accepted: 03/20/2023] [Indexed: 04/30/2023] Open
Abstract
Right heart failure (RHF) is a common and deadly disease in aged populations. Extra-cardiac outcomes of RHF such as skeletal muscle atrophy contribute to morbidity and mortality. Despite the significance of maintaining right ventricular (RV) and muscle function, the mechanisms of RHF and muscle atrophy are unclear. Metformin (MET) improves cardiac and muscle function through the regulation of metabolism and the cellular stress response. However, whether MET is a viable therapeutic for RHF and muscle atrophy is not yet known. We used deuterium oxide labeling to measure individual protein turnover in the RV as well as subcellular skeletal muscle proteostasis in aged male mice subjected to 4 weeks of hypobaric hypoxia (HH)-induced RHF. Mice exposed to HH had elevated RV mass and impaired RV systolic function, neither of which was prevented by MET. HH resulted in a higher content of glycolytic, cardiac, and antioxidant proteins in the RV, most of which were inhibited by MET. The synthesis of these key RV proteins was generally unchanged by MET, suggesting MET accelerated protein breakdown. HH resulted in a loss of skeletal muscle mass due to inhibited protein synthesis alongside myofibrillar protein breakdown. MET did not impact HH-induced muscle protein turnover and did not prevent muscle wasting. Together, we show tissue-dependent responses to HH-induced RHF where the RV undergoes hypertrophic remodeling with higher expression of metabolic and stress response proteins. Skeletal muscle undergoes loss of protein mass and atrophy, primarily due to myofibrillar protein breakdown. MET did not prevent HH-induced RV dysfunction or muscle wasting, suggesting that the identification of other therapies to attenuate RHF and concomitant muscle atrophy is warranted.
Collapse
Affiliation(s)
- Danielle R Bruns
- Division of Kinesiology & Health, University of Wyoming, 1000 E. University Ave, Dept. 3196, Laramie, WY, 82071, USA.
- Wyoming WWAMI Medical Education, Laramie, WY, USA.
| | - Benjamin D McNair
- Division of Kinesiology & Health, University of Wyoming, 1000 E. University Ave, Dept. 3196, Laramie, WY, 82071, USA
| | - Frederick F Peelor
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Agnieszka K Borowik
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Atul Pranay
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Aykhan Yusifov
- Division of Kinesiology & Health, University of Wyoming, 1000 E. University Ave, Dept. 3196, Laramie, WY, 82071, USA
| | - Benjamin F Miller
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
- Oklahoma City VA Medical Center, Oklahoma City, OK, USA
| |
Collapse
|
9
|
Mamic P, Lanfear DE. Gut Microbiome in Cardiovascular Disease and Heart Failure: Seeing the Iceberg Below Its Surface. JACC. HEART FAILURE 2023; 11:822-824. [PMID: 37407156 DOI: 10.1016/j.jchf.2023.05.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 05/04/2023] [Indexed: 07/07/2023]
Affiliation(s)
- Petra Mamic
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford University, Stanford, California, USA; Department of Genetics, Stanford University School of Medicine, Stanford University, Stanford, California, USA
| | - David E Lanfear
- Department of Medicine, Cardiovascular Division, and the Center for Individualized and Genomic Medicine Research, Henry Ford Hospital, Detroit, Michigan, USA.
| |
Collapse
|
10
|
Zhao Z, Cui X, Liao Z. Mechanism of fibroblast growth factor 21 in cardiac remodeling. Front Cardiovasc Med 2023; 10:1202730. [PMID: 37416922 PMCID: PMC10322220 DOI: 10.3389/fcvm.2023.1202730] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Accepted: 06/07/2023] [Indexed: 07/08/2023] Open
Abstract
Cardiac remodeling is a basic pathological process that enables the progression of multiple cardiac diseases to heart failure. Fibroblast growth factor 21 is considered a regulator in maintaining energy homeostasis and shows a positive role in preventing damage caused by cardiac diseases. This review mainly summarizes the effects and related mechanisms of fibroblast growth factor 21 on pathological processes associated with cardiac remodeling, based on a variety of cells of myocardial tissue. The possibility of Fibroblast growth factor 21 as a promising treatment for the cardiac remodeling process will also be discussed.
Collapse
Affiliation(s)
- Zeyu Zhao
- Queen Mary College, Nanchang University, Nanchang, China
| | - Xuemei Cui
- Fourth Clinical Medical College, Nanchang University, Nanchang, China
| | - Zhangping Liao
- Jiangxi Provincial Key Laboratory of Basic Pharmacology School of Pharmaceutical Science, Nanchang University, Nanchang, China
| |
Collapse
|
11
|
Hoehlschen J, Hofreither D, Tomin T, Birner-Gruenberger R. Redox-driven cardioprotective effects of sodium-glucose co-transporter-2 inhibitors: comparative review. Cardiovasc Diabetol 2023; 22:101. [PMID: 37120524 PMCID: PMC10148992 DOI: 10.1186/s12933-023-01822-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 04/03/2023] [Indexed: 05/01/2023] Open
Abstract
Sodium-glucose co-transporter-2 inhibitors are used in the treatment of diabetes but are also emerging as cardioprotective agents in heart diseases even in the absence of type 2 diabetes. In this paper, upon providing a short overview of common pathophysiological features of diabetes, we review the clinically reported cardio- and nephroprotective potential of sodium-glucose co-transporter-2 inhibitors currently available on the market, including Dapagliflozin, Canagliflozin, and Empagliflozin. To that end, we summarize findings of clinical trials that have initially drawn attention to the drugs' organ-protective potential, before providing an overview of their proposed mechanism of action. Since we particularly expect that their antioxidative properties will broaden the application of gliflozins from therapeutic to preventive care, special emphasis was put on this aspect.
Collapse
Affiliation(s)
- Julia Hoehlschen
- Institute of Chemical Technologies and Analytics, TU Wien, Wien, Austria
| | - Dominik Hofreither
- Institute of Chemical Technologies and Analytics, TU Wien, Wien, Austria
| | - Tamara Tomin
- Institute of Chemical Technologies and Analytics, TU Wien, Wien, Austria.
| | - Ruth Birner-Gruenberger
- Institute of Chemical Technologies and Analytics, TU Wien, Wien, Austria.
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria.
| |
Collapse
|
12
|
Zabielska-Kaczorowska MA, Braczko A, Pelikant-Malecka I, Slominska EM, Smolenski RT. Hidden Pool of Cardiac Adenine Nucleotides That Controls Adenosine Production. Pharmaceuticals (Basel) 2023; 16:ph16040599. [PMID: 37111356 PMCID: PMC10142527 DOI: 10.3390/ph16040599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 04/11/2023] [Accepted: 04/14/2023] [Indexed: 04/29/2023] Open
Abstract
Myocardial ischemic adenosine production decreases in subsequent events that may blunt its protective functions. To test the relation between total or mitochondrial cardiac adenine nucleotide pool (TAN) on the energy status with adenosine production, Langendorff perfused rat hearts were subjected to three protocols: 1 min ischemia at 40 min, 10 min ischemia at 50 min, and 1 min ischemia at 85 min in Group I; additional infusion of adenosine (30 µM) for 15 min after 10 min ischemia in Group I-Ado, and 1 min ischemia at 40 and 85 min in the controls (Group No I). A 31P NMR and an HPLC were used for the analysis of nucleotide and catabolite concentrations in the heart and coronary effluent. Cardiac adenosine production in Group I measured after 1 min ischemia at 85 min decreased to less than 15% of that at 40 min in Group I, accompanied by a decrease in cardiac ATP and TAN to 65% of the initial results. Adenosine production at 85 min was restored to 45% of that at 40 min in Group I-Ado, accompanied by a rebound of ATP and TAN by 10% vs. Group I. Mitochondrial TAN and free AMP concentrations paralleled that of total cardiac TAN. Changes in energy equilibrium or mitochondrial function were minor. This study highlights that only a fraction of the cardiac adenine nucleotide pool is available for adenosine production, but further studies are necessary to clarify its nature.
Collapse
Affiliation(s)
- Magdalena A Zabielska-Kaczorowska
- Department of Physiology, Medical University of Gdansk, 80-210 Gdansk, Poland
- Department of Biochemistry, Medical University of Gdansk, 80-210 Gdansk, Poland
| | - Alicja Braczko
- Department of Biochemistry, Medical University of Gdansk, 80-210 Gdansk, Poland
| | - Iwona Pelikant-Malecka
- Division of Medical Laboratory Diagnostics, Medical University of Gdansk, 80-210 Gdansk, Poland
| | - Ewa M Slominska
- Department of Biochemistry, Medical University of Gdansk, 80-210 Gdansk, Poland
| | - Ryszard T Smolenski
- Department of Biochemistry, Medical University of Gdansk, 80-210 Gdansk, Poland
- Heart Science Centre, Imperial College at Harefield Hospital, Harefield UB9 6JH, UK
| |
Collapse
|
13
|
Wang A, Li Z, Sun Z, Liu Y, Zhang D, Ma X. Potential Mechanisms Between HF and COPD: New Insights From Bioinformatics. Curr Probl Cardiol 2023; 48:101539. [PMID: 36528207 DOI: 10.1016/j.cpcardiol.2022.101539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022]
Abstract
Heart failure (HF) and chronic obstructive pulmonary disease (COPD) are closely related in clinical practice. This study aimed to investigate the co-genetic characteristics and potential molecular mechanisms of HF and COPD. HF and COPD datasets were downloaded from gene expression omnibus database. After identifying common differentially expressed genes (DEGs), the functional analysis highlighted the critical role of extracellular matrix and ribosomal signaling pathways in both diseases. In addition, GeneMANIA's results suggested that the 2 diseases were related to immune infiltration, and CIBERSORT suggested the role of macrophages. We also discovered 4 TFs and 1408 miRNAs linked to both diseases, and salbutamol may positively affect them.
Collapse
Affiliation(s)
- Anzhu Wang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China; Graduate School, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhendong Li
- Qingdao West Coast New Area People's Hospital, Qingdao, China
| | - Zhuo Sun
- Qingdao West Coast New Area People's Hospital, Qingdao, China
| | - Yicheng Liu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China; Graduate School, China Academy of Chinese Medical Sciences, Beijing, China
| | - Dawu Zhang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China; National Clinical Research Center for Chinese Medicine Cardiology, Beijing, China
| | - Xiaochang Ma
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China; National Clinical Research Center for Chinese Medicine Cardiology, Beijing, China.
| |
Collapse
|
14
|
Wang T, Yuan C, Liu J, Deng L, Li W, He J, Liu H, Qu L, Wu J, Zou W. Targeting Energy Protection as a Novel Strategy to Disclose Di'ao Xinxuekang against the Cardiotoxicity Caused by Doxorubicin. Int J Mol Sci 2023; 24:ijms24020897. [PMID: 36674413 PMCID: PMC9867483 DOI: 10.3390/ijms24020897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 12/25/2022] [Accepted: 12/28/2022] [Indexed: 01/06/2023] Open
Abstract
Doxorubicin (DOX) can induce myocardial energy metabolism disorder and further worsen heart failure. "Energy protection" is proposed as a new cardiac protection strategy. Previous studies have found that Di'ao Xinxuekang (DXXK) can improve doxorubicin-induced cardiotoxicity in mice by inhibiting ferroptosis. However, there are very few studies associating DXXK and energy protection. This study aims to explore the "energy protection" effect of DXXK on cardiotoxicity induced by DOX. A DOX-induced cardiotoxicity model established in rats and H9c2 cells are used to analyze the therapeutic effects of DXXK on serum indexes, cardiac function indexes and cardiac histopathology. The metabonomic methods were used to explore the potential mechanism of DXXK in treating DOX-induced cardiotoxicity. In addition, we also observed the mitochondrial- and autophagy-related indicators of myocardial cells and the mRNA expression level of the core target regulating energy-metabolism-related pathways. Our results indicated that DXXK can improve cardiac function, reduce myocardial enzymes and alleviate the histological damage of heart tissue caused by DOX. In addition, DXXK can improve mitochondrial damage induced by DOX and inhibit excessive autophagy. Metabonomics analysis showed that DOX can significantly affects the pathways related to energy metabolism of myocardial cells, which are involved in the therapeutic mechanism of DXXK. In conclusion, DXXK can treat DOX-induced cardiotoxicity through the AMPK-mediated energy protection pathway.
Collapse
Affiliation(s)
- Tao Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Chuqiao Yuan
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Jia Liu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Liangyan Deng
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Wei Li
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Junling He
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Honglin Liu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Liping Qu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Jianming Wu
- School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China
- Correspondence: (J.W.); (W.Z.)
| | - Wenjun Zou
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
- Correspondence: (J.W.); (W.Z.)
| |
Collapse
|
15
|
Salidroside attenuates myocardial ischemia/reperfusion injury via AMPK-induced suppression of endoplasmic reticulum stress and mitochondrial fission. Toxicol Appl Pharmacol 2022; 448:116093. [PMID: 35659894 DOI: 10.1016/j.taap.2022.116093] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 05/22/2022] [Accepted: 05/26/2022] [Indexed: 12/19/2022]
Abstract
Ischemic heart disease (IHD) is the primary cause of death worldwide. Salidroside (Sal), the major active compound derived from Rhodiola rosea, is believed to have cardioprotective effects. AMP-activated protein kinase (AMPK), is a pivotal AMP-activated protein kinase in energy metabolism. Whether Sal plays an anti-endoplasmic reticulum stress/mitochondrial fission role through AMPK remains elusive. In this study, we established a myocardial ischemia/reperfusion (I/R) rat model. Rat hearts exposed to Sal with or without compound C were then subjected to I/R. Further, H9c2 cardiomyocytes were subjected to simulated ischemia/reperfusion (SIR) by hypoxia-reoxygenation. The rats and cardiomyocytes were pretreated with Sal, followed by Compound C and AMPK-siRNA to block AMPK activity. We found that Sal significantly ameliorated cardiac function, mitigated infarct size and serum content of lactate dehydrogenase and creatine kinase, improved mitochondrial function, and reduced mitochondrial fission and apoptosis. Furthermore, in cultured H9c2 cardiomyocytes, Sal increased the cell viability and inhibited SIR-induced myocardial apoptosis and mitochondrial fission. Furthermore, the translocation of Drp1 from the cytoplasm to mitochondria induced by salidroside was confirmed both in vivo and in vitro. However, the use of Compound C or AMPK siRNA to block AMPK activity leads to blockade of the protective effects of Sal. In summary, protects against myocardial I/R by activating the AMPK signaling pathway, inhibiting ER stress, and reducing mitochondrial fission and apoptosis.
Collapse
|
16
|
The Beneficial Effects of Chinese Herbal Monomers on Ameliorating Diabetic Cardiomyopathy via Nrf2 Signaling. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:3959390. [PMID: 35656019 PMCID: PMC9155920 DOI: 10.1155/2022/3959390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 04/15/2022] [Accepted: 04/23/2022] [Indexed: 12/05/2022]
Abstract
Diabetic cardiomyopathy (DCM) is the main factor responsible for poor prognosis and survival in patients with diabetes. The highly complex pathogenesis of DCM involves multiple signaling pathways, including nuclear factor-κB (NF-κB) signaling pathway, adenosine monophosphate-activated protein kinase (AMPK) signaling pathway, phosphatidylinositol 3-kinase-protein kinase B (Akt) signaling pathway, mitogen-activated protein kinase (MAPK) signaling pathway, and transforming growth factor-β (TGF-β) signaling pathway. Nuclear factor erythroid-2-related factor 2 (Nrf2) seems essential to the amelioration of the progression of DCM, not only through counterbalancing oxidative stress, but also through interacting with other signaling pathways to combat inflammation, the disorder in energy homeostasis and insulin signaling, and fibrosis. It has been evidenced that Chinese herbal monomers could attenuate DCM through the crosstalk of Nrf2 with other signaling pathways. This article has summarized the pathogenesis of DCM (especially in oxidative stress), the beneficial effects of ameliorating DCM via the Nrf2 signaling pathway and its crosstalk, and examples of Chinese herbal monomers. It will facilitate pharmacological research and development to promote the utilization of traditional Chinese medicine in DCM.
Collapse
|
17
|
García-Díez E, López-Oliva ME, Pérez-Jiménez J, Martín MA, Ramos S. Metabolic regulation of (-)-epicatechin and the colonic metabolite 2,3-dihydroxybenzoic acid on the glucose uptake, lipid accumulation and insulin signalling in cardiac H9c2 cells. Food Funct 2022; 13:5602-5615. [PMID: 35502961 DOI: 10.1039/d2fo00182a] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Epicatechin (EC) and main colonic phenolic acids derived from flavonoid intake have been suggested to exert healthful effects, although their mechanism of action remains unknown. Heart damage is highly prevalent in metabolic diseases, and the failure of this organ is a major cause of death worldwide. In this study, the modulation of the energy metabolism and insulin signalling by the mentioned compounds in cardiac H9c2 cells was evaluated. Incubation of cells with EC (1-20 μM) and 2,3-dihydroxybenzoic acid (DHBA, 10 μM) reduced glucose uptake, and both compounds decreased lipid accumulation at concentrations higher than 0.5 μM. EC and DHBA also increased the tyrosine phosphorylated and total insulin receptor (IR) levels, and activated the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT) pathway in cardiac H9c2 cells. Interestingly, EC and DHBA did not modify glucose transporters (SGLT-1 and GLUT-1) levels, and increased GLUT-4 values. In addition, EC and DHBA decreased cluster of differentiation 36 (CD36) and fatty acid synthase (FAS) values, and enhanced carnitine palmitoyl transferase 1 (CPT1) and proliferator activated receptor α (PPARα) levels. By using specific inhibitors of AKT and 5'-AMP-activated protein kinase (AMPK), the participation of both proteins in EC- and DHBA-mediated regulation on glucose uptake and lipid accumulation was shown. Taken together, EC and DHBA modulate glucose uptake and lipid accumulation via AKT and AMPK, and reinforce the insulin signalling by activating key proteins of this pathway in H9c2 cells.
Collapse
Affiliation(s)
- Esther García-Díez
- Department of Metabolism and Nutrition, Institute of Food Science and Technology and Nutrition (ICTAN), Consejo Superior de Investigaciones Científicas (CSIC), José Antonio Novais 10, Ciudad Universitaria, 28040 Madrid, Spain.
| | - María Elvira López-Oliva
- Sección Departamental de Fisiología. Facultad de Farmacia, Universidad Complutense de Madrid (UCM), Spain
| | - Jara Pérez-Jiménez
- Department of Metabolism and Nutrition, Institute of Food Science and Technology and Nutrition (ICTAN), Consejo Superior de Investigaciones Científicas (CSIC), José Antonio Novais 10, Ciudad Universitaria, 28040 Madrid, Spain.
| | - María Angeles Martín
- Department of Metabolism and Nutrition, Institute of Food Science and Technology and Nutrition (ICTAN), Consejo Superior de Investigaciones Científicas (CSIC), José Antonio Novais 10, Ciudad Universitaria, 28040 Madrid, Spain. .,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), ISCIII, Spain
| | - Sonia Ramos
- Department of Metabolism and Nutrition, Institute of Food Science and Technology and Nutrition (ICTAN), Consejo Superior de Investigaciones Científicas (CSIC), José Antonio Novais 10, Ciudad Universitaria, 28040 Madrid, Spain.
| |
Collapse
|
18
|
Zhu M, Zhang C, Zhang Z, Liao X, Ren D, Li R, Liu S, He X, Dong N. Changes in transcriptomic landscape in human end-stage heart failure with distinct etiology. iScience 2022; 25:103935. [PMID: 35252820 PMCID: PMC8894266 DOI: 10.1016/j.isci.2022.103935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 01/21/2022] [Accepted: 02/14/2022] [Indexed: 11/15/2022] Open
Affiliation(s)
- Miaomiao Zhu
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
- Center for Genomics and Proteomics Research, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
- Hubei Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Chao Zhang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, China
| | - Zhe Zhang
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Xudong Liao
- College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Dongfeng Ren
- College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Rui Li
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, China
| | - Shiliang Liu
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Ximiao He
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
- Center for Genomics and Proteomics Research, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
- Hubei Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
- Corresponding author
| | - Nianguo Dong
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, China
- Corresponding author
| |
Collapse
|
19
|
Lin M, Xiong W, Wang S, Li Y, Hou C, Li C, Li G. The Research Progress of Trastuzumab-Induced Cardiotoxicity in HER-2-Positive Breast Cancer Treatment. Front Cardiovasc Med 2022; 8:821663. [PMID: 35097033 PMCID: PMC8789882 DOI: 10.3389/fcvm.2021.821663] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 12/14/2021] [Indexed: 12/14/2022] Open
Abstract
In recent years, the incidence of breast cancer has been increasing on an annual basis. Human epidermal growth factor receptor-2 (HER-2) is overexpressed in 15-20% human breast cancers, which is associated with poor prognosis and a high recurrence rate. Trastuzumab is the first humanized monoclonal antibody against HER-2. The most significant adverse effect of trastuzumab is cardiotoxicity, which has become an important factor in limiting the safe use of the drug. Unfortunately, the mechanism causing this cardiotoxicity is still not completely understood, and the use of preventive interventions remains controversial. This article focuses on trastuzumab-induced cardiotoxicity, reviewing the clinical application, potential cardiotoxicity, mechanism and discussing the potential interventions through summarizing related researches over the past tens of years.
Collapse
Affiliation(s)
- Mengmeng Lin
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Weiping Xiong
- Department of Cardiology, Shanghai Putuo District Liqun Hospital, Shanghai, China
| | - Shiyuan Wang
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yingying Li
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chunying Hou
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chunyu Li
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Guohui Li
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
20
|
Meng Y, Ding P, Wang H, Yang X, Wang Z, Nie D, Liu J, Huang Y, Su G, Hu J, Su Y, Du X, Dong N, Jia H, Zhang H, Zhang J, Li J. Ca2+/calmodulin-dependent protein kinase II inhibition reduces myocardial fatty acid uptake and oxidation after myocardial infarction. Biochim Biophys Acta Mol Cell Biol Lipids 2022; 1867:159120. [DOI: 10.1016/j.bbalip.2022.159120] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 01/24/2022] [Accepted: 01/26/2022] [Indexed: 12/28/2022]
|
21
|
Effects of Metformin in Heart Failure: From Pathophysiological Rationale to Clinical Evidence. Biomolecules 2021; 11:biom11121834. [PMID: 34944478 PMCID: PMC8698925 DOI: 10.3390/biom11121834] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 11/26/2021] [Accepted: 12/01/2021] [Indexed: 12/20/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a worldwide major health burden and heart failure (HF) is the most common cardiovascular (CV) complication in affected patients. Therefore, identifying the best pharmacological approach for glycemic control, which is also useful to prevent and ameliorate the prognosis of HF, represents a crucial issue. Currently, the choice is between the new drugs sodium/glucose co-transporter 2 inhibitors that have consistently shown in large CV outcome trials (CVOTs) to reduce the risk of HF-related outcomes in T2DM, and metformin, an old medicament that might end up relegated to the background while exerting interesting protective effects on multiple organs among which include heart failure. When compared with other antihyperglycemic medications, metformin has been demonstrated to be safe and to lower morbidity and mortality for HF, even if these results are difficult to interpret as they emerged mainly from observational studies. Meta-analyses of randomized controlled clinical trials have not produced positive results on the risk or clinical course of HF and sadly, large CV outcome trials are lacking. The point of force of metformin with respect to new diabetic drugs is the amount of data from experimental investigations that, for more than twenty years, still continues to provide mechanistic explanations of the several favorable actions in heart failure such as, the improvement of the myocardial energy metabolic status by modulation of glucose and lipid metabolism, the attenuation of oxidative stress and inflammation, and the inhibition of myocardial cell apoptosis, leading to reduced cardiac remodeling and preserved left ventricular function. In the hope that specific large-scale trials will be carried out to definitively establish the metformin benefit in terms of HF failure outcomes, we reviewed the literature in this field, summarizing the available evidence from experimental and clinical studies reporting on effects in heart metabolism, function, and structure, and the prominent pathophysiological mechanisms involved.
Collapse
|
22
|
Chan YT, Wang N, Feng Y. The toxicology and detoxification of Aconitum: traditional and modern views. Chin Med 2021; 16:61. [PMID: 34315520 PMCID: PMC8314510 DOI: 10.1186/s13020-021-00472-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 07/16/2021] [Indexed: 01/23/2023] Open
Abstract
Aconitum carmichaeli Debx.-derived herbal medicine has been used for anti-inflammation and anti-arrhythmia purpose for more than two thousand years. It is processed into Chuanwu (Radix Aconiti praeparata) and Fuzi (Radix Aconiti lateralis praeparata) in Traditional Chinese Medicine, which are two useful drugs but with toxic properties. There have been patients poisoned by accidental ingestion of Aconitum plants or misuse of the herbal drug, and this is of great concern to study in-depth. In this review, we provided the traditional and contemporary practice of using Aconitum herbs as medicine, from functions, processing methods to toxicity in ethnomedicine aspects to discuss the underlying connections of traditional and modern understanding on the toxicity of Aconitum plants. We summarized the functions and toxicology of the herbal drugs are analyzed from chemical and clinical aspects, with the help of traditional and modern knowledge of medicine. The medicinal doses and lethal doses determined by researches are summarized, and the usage and processing methods are updated and reviewed in the modern view. In addition, clinical management of poisoned cases using western medicine is discussed. This review provides insights and awareness of safety when using Aconitum-derived herbal medicine, and the application of modern scientific knowledge to optimize the detoxification processes. We suggest the possibility to renew the current standard processing method from the official Pharmacopoeia all over the world.
Collapse
Affiliation(s)
- Yau-Tuen Chan
- School of Chinese Medicine, The University of Hong Kong, Hong Kong, China
| | - Ning Wang
- School of Chinese Medicine, The University of Hong Kong, Hong Kong, China.
| | - Yibin Feng
- School of Chinese Medicine, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
23
|
Sayour AA, Ruppert M, Oláh A, Benke K, Barta BA, Zsáry E, Ke H, Horváth EM, Merkely B, Radovits T. Left Ventricular SGLT1 Protein Expression Correlates with the Extent of Myocardial Nitro-Oxidative Stress in Rats with Pressure and Volume Overload-Induced Heart Failure. Antioxidants (Basel) 2021; 10:antiox10081190. [PMID: 34439438 PMCID: PMC8388925 DOI: 10.3390/antiox10081190] [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: 06/17/2021] [Revised: 07/22/2021] [Accepted: 07/23/2021] [Indexed: 12/30/2022] Open
Abstract
Myocardial sodium-glucose cotransporter 1 (SGLT1) has been shown to be upregulated in humans with heart failure (HF) with or without diabetes. In vitro studies have linked SGLT1 to increased nitro-oxidative stress in cardiomyocytes. We aimed to assess the relation between left ventricular (LV) SGLT1 expression and the extent of nitro-oxidative stress in two non-diabetic rat models of chronic heart failure (HF) evoked by either pressure (TAC, n = 12) or volume overload (ACF, n = 12). Sham-operated animals (Sham-T and Sham-A, both n = 12) served as controls. Both TAC and ACF induced characteristic LV structural and functional remodeling. Western blotting revealed that LV SGLT1 protein expression was significantly upregulated in both HF models (both p < 0.01), whereas the phosphorylation of ERK1/2 was decreased only in ACF; AMPKα activity was significantly reduced in both models. The protein expression of the Nox4 NADPH oxidase isoform was increased in both TAC and ACF compared with respective controls (both p < 0.01), showing a strong positive correlation with SGLT1 expression (r = 0.855, p < 0.001; and r = 0.798, p = 0.001, respectively). Furthermore, SGLT1 protein expression positively correlated with the extent of myocardial nitro-oxidative stress in failing hearts assessed by 3-nitrotyrosin (r = 0.818, p = 0.006) and 4-hydroxy-2-nonenal (r = 0.733, p = 0.020) immunostaining. Therefore, LV SGLT1 protein expression was upregulated irrespective of the nature of chronic hemodynamic overload, and correlated significantly with the expression of Nox4 and with the level of myocardial nitro-oxidative stress, suggesting a pathophysiological role of SGLT1 in HF.
Collapse
Affiliation(s)
- Alex Ali Sayour
- Heart and Vascular Center, Department of Cardiology, Semmelweis University, Városmajor Str. 68, H-1122 Budapest, Hungary; (M.R.); (A.O.); (K.B.); (B.A.B.); (E.Z.); (B.M.); (T.R.)
- Correspondence:
| | - Mihály Ruppert
- Heart and Vascular Center, Department of Cardiology, Semmelweis University, Városmajor Str. 68, H-1122 Budapest, Hungary; (M.R.); (A.O.); (K.B.); (B.A.B.); (E.Z.); (B.M.); (T.R.)
| | - Attila Oláh
- Heart and Vascular Center, Department of Cardiology, Semmelweis University, Városmajor Str. 68, H-1122 Budapest, Hungary; (M.R.); (A.O.); (K.B.); (B.A.B.); (E.Z.); (B.M.); (T.R.)
| | - Kálmán Benke
- Heart and Vascular Center, Department of Cardiology, Semmelweis University, Városmajor Str. 68, H-1122 Budapest, Hungary; (M.R.); (A.O.); (K.B.); (B.A.B.); (E.Z.); (B.M.); (T.R.)
| | - Bálint András Barta
- Heart and Vascular Center, Department of Cardiology, Semmelweis University, Városmajor Str. 68, H-1122 Budapest, Hungary; (M.R.); (A.O.); (K.B.); (B.A.B.); (E.Z.); (B.M.); (T.R.)
| | - Eszter Zsáry
- Heart and Vascular Center, Department of Cardiology, Semmelweis University, Városmajor Str. 68, H-1122 Budapest, Hungary; (M.R.); (A.O.); (K.B.); (B.A.B.); (E.Z.); (B.M.); (T.R.)
| | - Haoran Ke
- Department of Physiology, Semmelweis University, Tűzoltó Str. 37-47, H-1094 Budapest, Hungary; (H.K.); (E.M.H.)
| | - Eszter Mária Horváth
- Department of Physiology, Semmelweis University, Tűzoltó Str. 37-47, H-1094 Budapest, Hungary; (H.K.); (E.M.H.)
| | - Béla Merkely
- Heart and Vascular Center, Department of Cardiology, Semmelweis University, Városmajor Str. 68, H-1122 Budapest, Hungary; (M.R.); (A.O.); (K.B.); (B.A.B.); (E.Z.); (B.M.); (T.R.)
| | - Tamás Radovits
- Heart and Vascular Center, Department of Cardiology, Semmelweis University, Városmajor Str. 68, H-1122 Budapest, Hungary; (M.R.); (A.O.); (K.B.); (B.A.B.); (E.Z.); (B.M.); (T.R.)
| |
Collapse
|
24
|
Wu B, You S, Qian H, Wu S, Lu S, Zhang Y, Sun Y, Zhang N. The role of SIRT2 in vascular-related and heart-related diseases: A review. J Cell Mol Med 2021; 25:6470-6478. [PMID: 34028177 PMCID: PMC8278089 DOI: 10.1111/jcmm.16618] [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: 02/23/2021] [Revised: 04/01/2021] [Accepted: 04/13/2021] [Indexed: 12/15/2022] Open
Abstract
At present, cardiovascular disease is one of the important factors of human death, and there are many kinds of proteins involved. Sirtuins family proteins are involved in various physiological and pathological activities of the human body. Among them, there are more and more studies on the relationship between sirtuin2 (SIRT2) protein and cardiovascular diseases. SIRT2 can effectively inhibit pathological cardiac hypertrophy. The effect of SIRT2 on ischaemia‐reperfusion injury has different effects under different conditions. SIRT2 can reduce the level of reactive oxygen species (ROS), which may help to reduce the severity of diabetic cardiomyopathy. SIRT2 can affect a variety of cardiovascular diseases, energy metabolism and the ageing of cardiomyocytes, thereby affecting heart failure. SIRT2 also plays an important role in vascular disease. For endothelial cell damage used by oxidative stress, the role of SIRT2 is bidirectional, which is related to the degree of oxidative stress stimulation. When the degree of stimulation is small, SIRT2 plays a protective role, and when the degree of stimulation increases to a certain level, SIRT2 plays a negative role. In addition, SIRT2 is also involved in the remodelling of blood vessels and the repair of skin damage.
Collapse
Affiliation(s)
- Boquan Wu
- Department of Cardiology, The First Hospital of China Medical University, Shenyang, China
| | - Shilong You
- Department of Cardiology, The First Hospital of China Medical University, Shenyang, China
| | - Hao Qian
- Department of Cardiology, The First Hospital of China Medical University, Shenyang, China
| | - Shaojun Wu
- Department of Cardiology, The First Hospital of China Medical University, Shenyang, China
| | - Saien Lu
- Department of Cardiology, The First Hospital of China Medical University, Shenyang, China
| | - Ying Zhang
- Department of Cardiology, The First Hospital of China Medical University, Shenyang, China
| | - Yingxian Sun
- Department of Cardiology, The First Hospital of China Medical University, Shenyang, China
| | - Naijin Zhang
- Department of Cardiology, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
25
|
Li Z, Zhao H, Wang J. Metabolism and Chronic Inflammation: The Links Between Chronic Heart Failure and Comorbidities. Front Cardiovasc Med 2021; 8:650278. [PMID: 34026868 PMCID: PMC8131678 DOI: 10.3389/fcvm.2021.650278] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 03/31/2021] [Indexed: 12/12/2022] Open
Abstract
Heart failure (HF) patients often suffer from multiple comorbidities, such as diabetes, atrial fibrillation, depression, chronic obstructive pulmonary disease, and chronic kidney disease. The coexistance of comorbidities usually leads to multi morbidity and poor prognosis. Treatments for HF patients with multi morbidity are still an unmet clinical need, and finding an effective therapy strategy is of great value. HF can lead to comorbidity, and in return, comorbidity may promote the progression of HF, creating a vicious cycle. This reciprocal correlation indicates there may be some common causes and biological mechanisms. Metabolism remodeling and chronic inflammation play a vital role in the pathophysiological processes of HF and comorbidities, indicating metabolism and inflammation may be the links between HF and comorbidities. In this review, we comprehensively discuss the major underlying mechanisms and therapeutic implications for comorbidities of HF. We first summarize the potential role of metabolism and inflammation in HF. Then, we give an overview of the linkage between common comorbidities and HF, from the perspective of epidemiological evidence to the underlying metabolism and inflammation mechanisms. Moreover, with the help of bioinformatics, we summarize the shared risk factors, signal pathways, and therapeutic targets between HF and comorbidities. Metabolic syndrome, aging, deleterious lifestyles (sedentary behavior, poor dietary patterns, smoking, etc.), and other risk factors common to HF and comorbidities are all associated with common mechanisms. Impaired mitochondrial biogenesis, autophagy, insulin resistance, and oxidative stress, are among the major mechanisms of both HF and comorbidities. Gene enrichment analysis showed the PI3K/AKT pathway may probably play a central role in multi morbidity. Additionally, drug targets common to HF and several common comorbidities were found by network analysis. Such analysis has already been instrumental in drug repurposing to treat HF and comorbidity. And the result suggests sodium-glucose transporter-2 (SGLT-2) inhibitors, IL-1β inhibitors, and metformin may be promising drugs for repurposing to treat multi morbidity. We propose that targeting the metabolic and inflammatory pathways that are common to HF and comorbidities may provide a promising therapeutic strategy.
Collapse
Affiliation(s)
- Zhiwei Li
- Department of Pathophysiology, State Key Laboratory of Medical Molecular Biology Institute of Basic Medicine, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Hongmei Zhao
- Department of Pathophysiology, State Key Laboratory of Medical Molecular Biology Institute of Basic Medicine, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Jing Wang
- Department of Pathophysiology, State Key Laboratory of Medical Molecular Biology Institute of Basic Medicine, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| |
Collapse
|
26
|
Zhang Z, Tian S, Wu C, Yan L, Wan J, Zhang J, Liu X, Zhang W. Comprehensive bioinformatics analysis reveals kinase activity profiling associated with heart failure. J Cell Biochem 2021; 122:1126-1140. [PMID: 33899242 DOI: 10.1002/jcb.29935] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 03/22/2021] [Indexed: 01/27/2023]
Abstract
Heart failure is a complex clinical syndrome originating from cardiac injury, which leads to considerable morbidity and mortality. Among the dynamic molecular adaptations occurring in heart failure development, aggravation of the disease is often attributed to global or local abnormality of the kinase. Therefore, the overall monitoring of kinase activity is indispensable. In this study, a bioinformatics analysis method was developed to conduct deep mining of transcriptome and phosphoproteome in failing heart tissue. A total of 982 differentially expressed genes and 9781 phosphorylation sites on 3252 proteins were identified. Via upstream regulator relations and kinase-substrate relations, a dendrogram of kinases can be constructed to monitor its abnormality. The results show that, on the dendrogram, the distribution of kinases demonstrated complex kinase activity changes and certain rules that occur during heart failure. Finally, we also identified the hub kinases in heart failure and verified the expression of these kinases by reverse-transcription polymerase chain reaction and Western blot analysis. In conclusion, for the first time, we have systematically analyzed the differences in kinases during heart failure and provided an unprecedented breadth of multi-omics data. These results can bring about a sufficient data foundation and novel research perspectives.
Collapse
Affiliation(s)
- Zhen Zhang
- School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Saisai Tian
- School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Chennan Wu
- School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Li Yan
- School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Jingjing Wan
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Jinbo Zhang
- School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Xia Liu
- School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Weidong Zhang
- School of Pharmacy, Second Military Medical University, Shanghai, China
| |
Collapse
|
27
|
Hardy E, Fernandez-Patron C. Targeting MMP-Regulation of Inflammation to Increase Metabolic Tolerance to COVID-19 Pathologies: A Hypothesis. Biomolecules 2021; 11:biom11030390. [PMID: 33800947 PMCID: PMC7998259 DOI: 10.3390/biom11030390] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 02/28/2021] [Accepted: 03/02/2021] [Indexed: 02/06/2023] Open
Abstract
Many individuals infected with the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) develop no or only mild symptoms, but some can go on onto develop a spectrum of pathologies including pneumonia, acute respiratory distress syndrome, respiratory failure, systemic inflammation, and multiorgan failure. Many pathogens, viral and non-viral, can elicit these pathologies, which justifies reconsidering whether the target of therapeutic approaches to fight pathogen infections should be (a) the pathogen itself, (b) the pathologies elicited by the pathogen interaction with the human host, or (c) a combination of both. While little is known about the immunopathology of SARS-CoV-2, it is well-established that the above-mentioned pathologies are associated with hyper-inflammation, tissue damage, and the perturbation of target organ metabolism. Mounting evidence has shown that these processes are regulated by endoproteinases (particularly, matrix metalloproteinases (MMPs)). Here, we review what is known about the roles played by MMPs in the development of COVID-19 and postulate a mechanism by which MMPs could influence energy metabolism in target organs, such as the lung. Finally, we discuss the suitability of MMPs as therapeutic targets to increase the metabolic tolerance of the host to damage inflicted by the pathogen infection, with a focus on SARS-CoV-2.
Collapse
Affiliation(s)
- Eugenio Hardy
- Center for Molecular Immunology, 16040 Havana, Cuba
- Correspondence: (E.H.); (C.F.-P.)
| | - Carlos Fernandez-Patron
- Department of Biochemistry, Mazankowski Alberta Heart Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada
- Correspondence: (E.H.); (C.F.-P.)
| |
Collapse
|
28
|
Moheimani HR, Amiriani T, Alizadeh AM, Jand Y, Shakiba D, Ensan PS, Jafarzadeh F, Rajaei M, Enayati A, Pourabouk M, Aliazadeh S, Pourkhani AH, Mazaheri Z, Zeyghami MA, Dehpour A, Khori V. Preconditioning and anti-apoptotic effects of Metformin and Cyclosporine-A in an isolated bile duct-ligated rat heart. Eur J Pharmacol 2021; 893:173807. [PMID: 33359222 DOI: 10.1016/j.ejphar.2020.173807] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 11/18/2020] [Accepted: 12/08/2020] [Indexed: 12/18/2022]
Abstract
Despite all previous studies relating to the mechanism of cirrhotic cardiomyopathy (CCM), the role of cirrhosis on Ischemic Preconditioning (IPC) has not yet been explored. The present study strives to assess the cardioprotective role of IPC in bile duct ligated (BDL) rats as well as the cardioprotective role of Cyclosporin-A (CsA) and Metformin (Met) in CCM. Cirrhosis was induced by bile duct ligation (BDL). Rats' hearts were isolated and attached to a Langendorff Apparatus. The pharmacological preconditioning with Met and CsA was done before the main ischemia. Myocardial infarct size, hemodynamic and electrophysiological parameters, biochemical markers, and apoptotic indices were determined at the end of the experiment. Infarct size, apoptotic indices, arrhythmia score, and incidence of VF decreased significantly in the IPC group in comparison with the I/R group. These significant decreases were abolished in the IPC (BDL) group. Met significantly decreased the infarct size and apoptotic indices compared with I/R (BDL) and normal groups, while CsA led to similar decreases except in the level of caspase-3 and -8. Met and CsA decreased and increased the arrhythmia score and incidence of VF in the BDL groups, respectively. Functional recovery indices decreased in the I/R (BDL) and IPC (BDL) groups. Met improved these parameters. Therefore, the current study depicted that the cardioprotective effect of Met and CsA on BDL rats is mediated through the balance between pAMPK and apoptosis in the mitochondria.
Collapse
Key Words
- Bile duct ligation
- Caspase
- Cyclosporin-A
- Cyclosporin-A (3S,6S,9S,12R,15S,18S,21S,24S,30S,33S)-30-Ethyl-33-[(E,1R,2R)-1-Hydroxy-2-methylhex-4-enyl]-1,4,7,10,12,15,19,25,28-nonamethyl-6,9,18,24-tetrakis(2-methylpropyl)-3,21-di(propan-2-yl)-1,4,7,10,13,16,19,22,25,28,31undecazacyclotritriacontane-2,5,8,11,14,17,20,23,26,29,32-undecone, PubChemCID: 5284373
- Ischemic preconditioning
- Metformin
- Metformin 3-(diaminomethylidene)-1,1-dimethylguanidine, PubChem CID:4091
Collapse
Affiliation(s)
- Hamid Reza Moheimani
- Ischemic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Taghi Amiriani
- Golestan Research Center of Gastroenterology and Hepatology, Gorgan, Iran
| | - Ali Mohammad Alizadeh
- Cancer Research Center of Institute Cancer, Tehran University of Medical Science, Tehran, Iran
| | - Yahya Jand
- Ischemic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran; Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Delaram Shakiba
- Department of Mechanical Engineering and Material Science, Washington University in St. Louis, St. Louis, MO, USA
| | - Parham Sayyah Ensan
- Ischemic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Fatemeh Jafarzadeh
- Ischemic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Maryam Rajaei
- Ischemic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Ayesheh Enayati
- Ischemic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Mona Pourabouk
- Ischemic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Shahriar Aliazadeh
- Ischemic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Amir Hoshang Pourkhani
- Ischemic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Zohreh Mazaheri
- Basic Medical Science Research Center, Histogenotech Company, Tehran, Iran
| | - Mohammad Ali Zeyghami
- Ischemic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Ahmadreza Dehpour
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Vahid Khori
- Ischemic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran.
| |
Collapse
|
29
|
Mendieta G, Ben-Aicha S, Gutiérrez M, Casani L, Aržanauskaitė M, Carreras F, Sabate M, Badimon L, Vilahur G. Intravenous Statin Administration During Myocardial Infarction Compared With Oral Post-Infarct Administration. J Am Coll Cardiol 2020; 75:1386-1402. [PMID: 32216907 DOI: 10.1016/j.jacc.2020.01.042] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Revised: 01/03/2020] [Accepted: 01/23/2020] [Indexed: 12/20/2022]
Abstract
BACKGROUND Beyond lipid-lowering, statins exert cardioprotective effects. High-dose statin treatment seems to reduce cardiovascular complications in high-risk patients. The ideal timing and administration regime remain unknown. OBJECTIVES This study compared the cardioprotective effects of intravenous statin administration during myocardial infarction (MI) with oral administration immediately post-MI. METHODS Hypercholesterolemic pigs underwent MI induction (90 min of ischemia) and were kept for 42 days. Animals were distributed in 3 arms (A): A1 received an intravenous bolus of atorvastatin during MI; A2 received an intravenous bolus of vehicle during MI; and A3 received oral atorvastatin within 2 h post-MI. A1 and A3 remained on daily oral atorvastatin for the following 42 days. Cardiac magnetic resonance analysis (days 3 and 42 post-MI) and molecular/histological studies were performed. RESULTS At day 3, A1 showed a 10% reduction in infarct size compared with A3 and A2 and a 50% increase in myocardial salvage. At day 42, both A1 and A3 showed a significant decrease in scar size versus A2; however, A1 showed a further 24% reduction versus A3. Functional analyses revealed improved systolic performance in A1 compared with A2 and less wall motion abnormalities in the jeopardized myocardium versus both groups at day 42. A1 showed enhanced collagen content and AMP-activated protein kinase activation in the scar, increased vessel density in the penumbra, higher tumor necrosis factor α plasma levels and lower peripheral blood mononuclear cell activation versus both groups. CONCLUSIONS Intravenous administration of atorvastatin during MI limits cardiac damage, improves cardiac function, and mitigates remodeling to a larger extent than when administered orally shortly after reperfusion. This therapeutic approach deserves to be investigated in ST-segment elevation MI patients.
Collapse
Affiliation(s)
- Guiomar Mendieta
- Cardiovascular Research Center-ICCC, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain; Department of Cardiology, Clinic Hospital, Barcelona, Spain
| | - Soumaya Ben-Aicha
- Cardiovascular Research Center-ICCC, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain
| | - Manuel Gutiérrez
- Radiology, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain
| | - Laura Casani
- Cardiovascular Research Center-ICCC, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain; Radiology, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain
| | - Monika Aržanauskaitė
- Cardiovascular Research Center-ICCC, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain
| | - Francesc Carreras
- Cardiology, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain
| | - Manel Sabate
- Department of Cardiology, Clinic Hospital, Barcelona, Spain
| | - Lina Badimon
- Cardiovascular Research Center-ICCC, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain; CiberCV, Institute Carlos III, Barcelona, Spain
| | - Gemma Vilahur
- Cardiovascular Research Center-ICCC, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain; CiberCV, Institute Carlos III, Barcelona, Spain.
| |
Collapse
|
30
|
Huang Y, Zhang K, Jiang M, Ni J, Chen J, Li L, Deng J, Zhu Y, Mao J, Gao X, Fan G. Regulation of energy metabolism by combination therapy attenuates cardiac metabolic remodeling in heart failure. Int J Biol Sci 2020; 16:3133-3148. [PMID: 33162820 PMCID: PMC7645995 DOI: 10.7150/ijbs.49520] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 09/26/2020] [Indexed: 12/12/2022] Open
Abstract
Cardiac metabolic remodeling is recognized as an important hallmark of heart failure (HF), while strategies that target energy metabolism have therapeutic potential in treating HF. Shen-Fu formula (S-F) is a standardized herbal preparation frequently used in clinical practice and is a promising combinatorial therapy for HF-related metabolic remodeling. Herein, we performed an untargeted multi-omics analysis using transcriptomics, proteomics, and metabolomics on HF mice induced by transverse aortic constriction (TAC). Integrated and pathway-driven analyses were used to reveal the therapeutic targets associated with S-F treatment. The cardioprotective effect and potential mechanism of S-F were verified by the results from echocardiography, hemodynamics, histopathology, and biochemical assays. As a result, S-F significantly alleviated myocardial fibrosis and hypertrophy, thus reducing the loss of heart function during adverse cardiac remodeling in TAC mice. Integrated omics analysis showed that S-F synergistically mediated the metabolic flexibility of fatty acids and glucose in cardiac energy metabolism. These effects of S-F were confirmed by the activation of AMP-activated protein kinase (AMPK) and its downstream targets in the failing heart. Collectively, our results demonstrated that S-F suppressed cardiac metabolic remodeling through activating AMPK-related pathways via energy-dependent mechanisms.
Collapse
Affiliation(s)
- Yuting Huang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China.,State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Kai Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China.,Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, People's Republic of China
| | - Miaomiao Jiang
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Jingyu Ni
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China.,State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Jingrui Chen
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China.,State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Lan Li
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China.,State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Jie Deng
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China.,State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Yan Zhu
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Jingyuan Mao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China.,Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, People's Republic of China
| | - Xiumei Gao
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Guanwei Fan
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China.,State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China.,Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, People's Republic of China
| |
Collapse
|
31
|
Hasan KM, Friedman TC, Parveen M, Espinoza-Derout J, Bautista F, Razipour MM, Shao XM, Jordan MC, Roos KP, Mahata SK, Sinha-Hikim AP. Electronic cigarettes cause alteration in cardiac structure and function in diet-induced obese mice. PLoS One 2020; 15:e0239671. [PMID: 33002059 PMCID: PMC7529198 DOI: 10.1371/journal.pone.0239671] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 08/30/2020] [Indexed: 11/19/2022] Open
Abstract
In spite of the widespread use of electronic cigarettes, also known as e-cigarettes, and the proposed adverse cardiac effects of nicotine, the detrimental effects of e-cigarettes on the heart are not well known. This study examines the detrimental effects of e-cigarettes with nicotine at doses that yield circulating nicotine and cotinine in the ranges similar to the levels found in habitual smokers, and a high fat diet (HFD) on cardiac structure and function in a commonly used model of diet-induced obesity (DIO). C57BL/6J mice on an HFD were exposed to e-cigarette in the presence (2.4% nicotine) or absence (0% nicotine) of nicotine and saline aerosol for 12 weeks. Echocardiographic data demonstrated a decrease in left ventricular (LV) fractional shortening, LV ejection fraction, and velocity of circumferential fiber shortening (VCF) in mice treated with e-cigarette (2.4% nicotine) compared to e-cigarette (0% nicotine) or saline exposed mice. Cardiomyocytes (CMs) of mice treated with e-cigarette (2.4% nicotine) exhibited LV abnormalities, including lipid accumulation (ventricular steatosis), myofibrillar derangement and destruction, and mitochondrial hypertrophy, as revealed by transmission electron microscopy. The detrimental effects of e-cigarettes (2.4% nicotine) on cardiac structure and function was accompanied by increased oxidative stress, plasma free fatty acid levels, CM apoptosis, and inactivation of AMP-activated protein kinase and activation of its downstream target, acetyl-CoA-carboxylase. Our results indicate profound adverse effects of e-cigarettes (2.4% nicotine) on the heart in obese mice and raise questions about the safety of the nicotine e-cigarettes use.
Collapse
Affiliation(s)
- Kamrul M. Hasan
- Division of Endocrinology, Metabolism and Molecular Medicine, Department of Internal Medicine, Charles R. Drew University, Los Angeles, CA, United States of America
- * E-mail: (KMH); (APSH)
| | - Theodore C. Friedman
- Division of Endocrinology, Metabolism and Molecular Medicine, Department of Internal Medicine, Charles R. Drew University, Los Angeles, CA, United States of America
- David Geffen School of Medicine at University of California, Los Angeles, CA, United States of America
| | - Meher Parveen
- Division of Endocrinology, Metabolism and Molecular Medicine, Department of Internal Medicine, Charles R. Drew University, Los Angeles, CA, United States of America
| | - Jorge Espinoza-Derout
- Division of Endocrinology, Metabolism and Molecular Medicine, Department of Internal Medicine, Charles R. Drew University, Los Angeles, CA, United States of America
| | - Francisco Bautista
- Division of Endocrinology, Metabolism and Molecular Medicine, Department of Internal Medicine, Charles R. Drew University, Los Angeles, CA, United States of America
| | - Mohammad M. Razipour
- Division of Endocrinology, Metabolism and Molecular Medicine, Department of Internal Medicine, Charles R. Drew University, Los Angeles, CA, United States of America
| | - Xuesi M. Shao
- Division of Endocrinology, Metabolism and Molecular Medicine, Department of Internal Medicine, Charles R. Drew University, Los Angeles, CA, United States of America
- David Geffen School of Medicine at University of California, Los Angeles, CA, United States of America
| | - Maria C. Jordan
- David Geffen School of Medicine at University of California, Los Angeles, CA, United States of America
| | - Kenneth P. Roos
- David Geffen School of Medicine at University of California, Los Angeles, CA, United States of America
| | - Sushil K. Mahata
- VA San Diego Health Care System and University of California, San Diego, CA, United States of America
| | - Amiya P. Sinha-Hikim
- Division of Endocrinology, Metabolism and Molecular Medicine, Department of Internal Medicine, Charles R. Drew University, Los Angeles, CA, United States of America
- David Geffen School of Medicine at University of California, Los Angeles, CA, United States of America
- * E-mail: (KMH); (APSH)
| |
Collapse
|
32
|
Zhang L, Tian J, Diao S, Zhang G, Xiao M, Chang D. GLP-1 receptor agonist liraglutide protects cardiomyocytes from IL-1β-induced metabolic disturbance and mitochondrial dysfunction. Chem Biol Interact 2020; 332:109252. [PMID: 32898504 DOI: 10.1016/j.cbi.2020.109252] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 08/11/2020] [Accepted: 09/04/2020] [Indexed: 12/27/2022]
Abstract
Cardiac inflammation plays a critical role in the development of heart failure. Inflammation-induced oxidative stress contributes to aberrant cardiac metabolism and mitochondrial function. GLP-1 receptor agonists (GLP-1 RAs) are a type of blood glucose-lowering agent typically used in the treatment of type 2 diabetes. Recent studies have convincingly shown that GLP-1 RAs possess beneficial effects in diabetes-related cardiovascular complications. Liraglutide is a commonly used long-acting agonist that shows promising cardioprotective benefits. In this study, we investigated the protective role of Liraglutide in cultured cardiomyocytes. We found that HL-1 cardiomyocytes moderately expressed the GLP-1 receptor, and co-treatment with Liraglutide ameliorated IL-1β-induced cellular ROS production and NADPH oxidase (NOX)-4 expression. Furthermore, we found that Liraglutide protected cardiomyocytes from IL-1β-induced decreased mitochondrial membrane potential and reduced ATP production. Seahorse analysis revealed that Liraglutide mitigated IL-1β-induced reduced basal and maximum respiration rates as well as spare respiration capacity. Additionally, we found that Liraglutide alleviated IL-1β-induced aberrant triglyceride accumulation and adiponectin secretion. Mechanistically, we showed that Liraglutide ameliorated IL-1β-induced phosphorylation of AMPK and ACC as well as the reduction in PGC-1α, CPT-1, and DGAT1. Finally, through the study we demonstrated that the blockage of AMPK activity by Compound C abolished the ameliorative effect of Liraglutide on IL-1β-induced repressed ATP production and triglyceride accumulation, indicating that the action of Liraglutide was dependent on AMPK activation. In conclusion, this study revealed the molecular mechanism of Liraglutide protection in cultured cardiomyocytes. The GLP-1 RA Liraglutide could have therapeutic implications by modulating cardiac inflammation.
Collapse
Affiliation(s)
- Lili Zhang
- Department of Cardiology, The People's Hospital of Longhua, The Affiliated Hospital of Southern Medical University, Shenzhen City, Guangdong Province, 518109, China; Department of Cardiology, Heilongjiang Academy of Medical Science, Harbin City, Heilongjiang Province, 150086, China
| | - Jiali Tian
- Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin City, Heilongjiang Province, 150001, China
| | - Sujuan Diao
- The Outpatient Department, The People's Hospital of Longhua, The Affiliated Hospital of Southern Medical University, Shenzhen City, Guangdong Province, 518109, China
| | - Guowei Zhang
- Department of Cardiac Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin City, Heilongjiang Province, 150001, China
| | - Mochao Xiao
- Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin City, Heilongjiang Province, 150001, China.
| | - Dong Chang
- Department of Cardiology, Xiamen Cardiovascular Hospital of Xiamen University, Xiamen City, Fujian Province, 361006, China.
| |
Collapse
|
33
|
Abstract
Doxorubicin is a commonly used chemotherapeutic agent for the treatment of a range of cancers, but despite its success in improving cancer survival rates, doxorubicin is cardiotoxic and can lead to congestive heart failure. Therapeutic options for this patient group are limited to standard heart failure medications with the only drug specific for doxorubicin cardiotoxicity to reach FDA approval being dexrazoxane, an iron-chelating agent targeting oxidative stress. However, dexrazoxane has failed to live up to its expectations from preclinical studies while also bringing up concerns about its safety. Despite decades of research, the molecular mechanisms of doxorubicin cardiotoxicity are still poorly understood and oxidative stress is no longer considered to be the sole evil. Mitochondrial impairment, increased apoptosis, dysregulated autophagy and increased fibrosis have also been shown to be crucial players in doxorubicin cardiotoxicity. These cellular processes are all linked by one highly conserved intracellular kinase: adenosine monophosphate-activated protein kinase (AMPK). AMPK regulates mitochondrial biogenesis via PGC1α signalling, increases oxidative mitochondrial metabolism, decreases apoptosis through inhibition of mTOR signalling, increases autophagy through ULK1 and decreases fibrosis through inhibition of TGFβ signalling. AMPK therefore sits at the control point of many mechanisms shown to be involved in doxorubicin cardiotoxicity and cardiac AMPK signalling itself has been shown to be impaired by doxorubicin. In this review, we introduce different agents known to activate AMPK (metformin, statins, resveratrol, thiazolidinediones, AICAR, specific AMPK activators) as well as exercise and dietary restriction, and we discuss the existing evidence for their potential role in cardioprotection from doxorubicin cardiotoxicity.
Collapse
Affiliation(s)
- Kerstin N Timm
- Department of Physiology Anatomy and Genetics, University of Oxford, Oxford, UK.
| | - Damian J Tyler
- Department of Physiology Anatomy and Genetics, University of Oxford, Oxford, UK
- Oxford Centre for Clinical Magnetic Resonance Research, University of Oxford, Oxford, UK
| |
Collapse
|
34
|
Steinberg GR, Carling D. AMP-activated protein kinase: the current landscape for drug development. Nat Rev Drug Discov 2020; 18:527-551. [PMID: 30867601 DOI: 10.1038/s41573-019-0019-2] [Citation(s) in RCA: 399] [Impact Index Per Article: 99.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Since the discovery of AMP-activated protein kinase (AMPK) as a central regulator of energy homeostasis, many exciting insights into its structure, regulation and physiological roles have been revealed. While exercise, caloric restriction, metformin and many natural products increase AMPK activity and exert a multitude of health benefits, developing direct activators of AMPK to elicit beneficial effects has been challenging. However, in recent years, direct AMPK activators have been identified and tested in preclinical models, and a small number have entered clinical trials. Despite these advances, which disease(s) represent the best indications for therapeutic AMPK activation and the long-term safety of such approaches remain to be established.
Collapse
Affiliation(s)
- Gregory R Steinberg
- Centre for Metabolism, Obesity and Diabetes Research, Department of Medicine and Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada.
| | - David Carling
- Cellular Stress Group, Medical Research Council London Institute of Medical Sciences, Hammersmith Hospital, Imperial College, London, UK
| |
Collapse
|
35
|
Urata Y, Salehi R, Lima PDA, Osuga Y, Tsang BK. Neuropeptide Y regulates proliferation and apoptosis in granulosa cells in a follicular stage-dependent manner. J Ovarian Res 2020; 13:5. [PMID: 31915051 PMCID: PMC6950994 DOI: 10.1186/s13048-019-0608-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 12/29/2019] [Indexed: 12/17/2022] Open
Abstract
Background The complex regulatory mechanism involved in ovarian follicular development is not completely understood. Neuronal neuropeptide Y (NPY) is involved in the regulation of feeding behavior, energy homeostasis, and reproduction behavior, while its function in ovarian follicular development is not clear. The objective of this study was to investigate if and how NPY regulates follicle development in the ovary. Methods All experiments were performed using Sprague Dawley rats. To understand NPY expression pattern at different stages of follicular development, NPY content was assessed using immunohistochemistry in individual follicles. NPY and its receptors expression pattern were evaluated in granulosa cells isolated from preantral (PA), early antral (EA) and late antral follicles (LAF). The influence of NPY on granulosa cell proliferation and apoptosis were further assessed in vitro, using Ki67- and TUNEL-positivity assays. To investigate whether NPY induced-proliferation in EA granulosa cells is mediated through the activation of NPY receptor Y5 (NPY5R) and Mitogen-activated protein kinase (MEK) signal pathway, EA granulosa cells were treated with NPY5R antagonist (CGP71683) and MEK inhibitors (PD98059 and U0126), and Ki67-positive cells were assessed. Results NPY protein expression was follicular stage-dependent and cell type-specific. NPY signal intensity in EA was higher than those in PA and LAF. Antral granulosa cells showed the highest signal intensity compared to mural granulosa cells, cumulus cells and theca cells. Granulosa cells NPY protein content and mRNA abundance were higher in EA than in LAF. NPY receptor contents in granulosa cells were follicular stage-dependent. While NPY reduced apoptosis of EA granulosa cells, it increased the proliferation through NPY5R and MEK pathway. In contrast, in LAF granulosa cells, NPY reduced proliferation and increased the number of apoptotic cells, with no significant effects on PA granulosa cells. Conclusion This study is the first to evaluate the intraovarian role of NPY in granulosa cells at various stage of follicular development. These results indicate that NPY regulates granulosa cells proliferation and apoptosis in a follicular stage-dependent and autocrine manner. NPY may play a role in pathogenesis of ovarian follicular disorders.
Collapse
Affiliation(s)
- Yoko Urata
- Departments of Obstetrics & Gynecology and Cellular & Molecular Medicine, Interdisciplinary School of Health Sciences, University of Ottawa; Chronic Disease Program, Ottawa Hospital Research Institute, Critical Care Wing, 3rd floor, Room W3107, 501 Smyth Road, Ottawa, ON, K1H 8L6, Canada. .,Department of Obstetrics and Gynecology, the University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan.
| | - Reza Salehi
- Departments of Obstetrics & Gynecology and Cellular & Molecular Medicine, Interdisciplinary School of Health Sciences, University of Ottawa; Chronic Disease Program, Ottawa Hospital Research Institute, Critical Care Wing, 3rd floor, Room W3107, 501 Smyth Road, Ottawa, ON, K1H 8L6, Canada
| | - Patricia D A Lima
- Departments of Obstetrics & Gynecology and Cellular & Molecular Medicine, Interdisciplinary School of Health Sciences, University of Ottawa; Chronic Disease Program, Ottawa Hospital Research Institute, Critical Care Wing, 3rd floor, Room W3107, 501 Smyth Road, Ottawa, ON, K1H 8L6, Canada.,Queen's Cardiopulmonary Unit, Queen's University, BioSciences Complex, Room 1605, 116 Barrie Street, Kingston, ON, K7L 3N6, Canada
| | - Yutaka Osuga
- Department of Obstetrics and Gynecology, the University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Benjamin K Tsang
- Departments of Obstetrics & Gynecology and Cellular & Molecular Medicine, Interdisciplinary School of Health Sciences, University of Ottawa; Chronic Disease Program, Ottawa Hospital Research Institute, Critical Care Wing, 3rd floor, Room W3107, 501 Smyth Road, Ottawa, ON, K1H 8L6, Canada
| |
Collapse
|
36
|
Wang Z, Wang SP, Shao Q, Li PF, Sun Y, Luo LZ, Yan XQ, Fan ZY, Hu J, Zhao J, Hang PZ, Du ZM. Brain-derived neurotrophic factor mimetic, 7,8-dihydroxyflavone, protects against myocardial ischemia by rebalancing optic atrophy 1 processing. Free Radic Biol Med 2019; 145:187-197. [PMID: 31574344 DOI: 10.1016/j.freeradbiomed.2019.09.033] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 09/26/2019] [Accepted: 09/27/2019] [Indexed: 01/09/2023]
Abstract
Brain-derived neurotrophic factor (BDNF)/tropomyosin-related kinase B (TrkB) pathway is associated with ischemic heart diseases (IHD). 7,8-dihydroxyflavone (7,8-DHF), BDNF mimetic, is a potent agonist of TrkB. We aimed to investigate the effects and the underlying mechanisms of 7,8-DHF on cardiac ischemia. Myocardial ischemic mouse model was induced by ligation of left anterior descending coronary artery. 7,8-DHF (5 mg/kg) was administered intraperitoneally two days after ischemia for four weeks. Echocardiography, HE staining and transmission electron microscope were used to examine the function, histology and ultrastructure of the heart. H9c2 cells were treated with hydrogen peroxide (H2O2), 7,8-DHF or TrkB inhibitor ANA-12. The effects of 7,8-DHF on cell viability, mitochondrial membrane potential (MMP) and mitochondrial superoxide generation were examined. Furthermore, mitochondrial fission and protein expression of mitochondrial dynamics (Mfn2 [mitofusin 2], OPA1 [optic atrophy 1], Drp1 [dynamin-related protein 1] and Fis-1 [fission 1]) was detected by mitotracker green staining and western blot, respectively. 7,8-DHF attenuated cardiac dysfunction and cardiomyocyte abnormality of myocardial ischemic mice. Moreover, 7,8-DHF increased cell viability and reduced cell death accompanied by improving MMP, inhibiting mitochondrial superoxide and preventing excessive mitochondrial fission of H2O2-treated H9c2 cells. The cytoprotective effects of 7,8-DHF were antagonized by ANA-12. Mechanistically, 7,8-DHF repressed OMA1-dependent conversion of L-OPA1 into S-OPA1, which was abolished by Akt inhibitor. In conclusion, 7,8-DHF protects against cardiac ischemic injury by inhibiting the proteolytic cleavage of OPA1. These findings provide a novel pharmacological effect of 7,8-DHF on mitochondrial dynamics and a new potential target for IHD.
Collapse
Affiliation(s)
- Zhen Wang
- Institute of Clinical Pharmacology, The Second Affiliated Hospital (The University Key Laboratory of Drug Research, Heilongjiang Province), Department of Clinical Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, 150086, China
| | - Shi-Peng Wang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Qun Shao
- Department of Cardiology, The Third Affiliated Hospital of Harbin Medical University, Harbin, 150081, China
| | - Pei-Feng Li
- Institute of Clinical Pharmacology, The Second Affiliated Hospital (The University Key Laboratory of Drug Research, Heilongjiang Province), Department of Clinical Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, 150086, China
| | - Yue Sun
- Institute of Clinical Pharmacology, The Second Affiliated Hospital (The University Key Laboratory of Drug Research, Heilongjiang Province), Department of Clinical Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, 150086, China
| | - Lan-Zi Luo
- Institute of Clinical Pharmacology, The Second Affiliated Hospital (The University Key Laboratory of Drug Research, Heilongjiang Province), Department of Clinical Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, 150086, China
| | - Xiu-Qing Yan
- Institute of Clinical Pharmacology, The Second Affiliated Hospital (The University Key Laboratory of Drug Research, Heilongjiang Province), Department of Clinical Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, 150086, China
| | - Zi-Yi Fan
- Institute of Clinical Pharmacology, The Second Affiliated Hospital (The University Key Laboratory of Drug Research, Heilongjiang Province), Department of Clinical Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, 150086, China
| | - Juan Hu
- Institute of Clinical Pharmacology, The Second Affiliated Hospital (The University Key Laboratory of Drug Research, Heilongjiang Province), Department of Clinical Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, 150086, China
| | - Jing Zhao
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University (Key Laboratory of Cardiac Diseases and Heart Failure, Harbin Medical University), Harbin, 150001, China.
| | - Peng-Zhou Hang
- Institute of Clinical Pharmacology, The Second Affiliated Hospital (The University Key Laboratory of Drug Research, Heilongjiang Province), Department of Clinical Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, 150086, China.
| | - Zhi-Min Du
- Institute of Clinical Pharmacology, The Second Affiliated Hospital (The University Key Laboratory of Drug Research, Heilongjiang Province), Department of Clinical Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, 150086, China; State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, China.
| |
Collapse
|
37
|
Vilahur G, Gutiérrez M, Casani L, Lambert C, Mendieta G, Ben-Aicha S, Capdevila A, Pons-Lladó G, Carreras F, Carlsson L, Hidalgo A, Badimon L. P2Y12 antagonists and cardiac repair post-myocardial infarction: global and regional heart function analysis and molecular assessments in pigs. Cardiovasc Res 2019; 114:1860-1870. [PMID: 30124783 DOI: 10.1093/cvr/cvy201] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 08/13/2018] [Indexed: 01/08/2023] Open
Abstract
Aims P2Y12 antagonists are the standard in antiplatelet therapy but their potential effects on functional myocardial recovery and cardioprotection post-myocardial infarction (MI) are unknown. We investigated in a preclinical model of MI whether ticagrelor and clopidogrel differently affect cardiac repair post-MI. Methods and results Pigs either received: (i) clopidogrel (600 mg; 75 mg/qd); (ii) ticagrelor (180 mg; 90 mg/bid); and (iii) placebo control. MI was induced by mid-left anterior descending coronary artery balloon occlusion (60 min) and animals received the maintenance doses for the following 42 days. Serial cardiac magnetic resonance was performed at Day 3 and Day 42 for the assessment of global and regional cardiac parameters. We determined cardiac AMP-activated protein kinase (AMPK), Akt/PKB, aquaporin-4, vascular density, and fibrosis. In comparison to controls, both P2Y12 antagonists limited infarct expansion at Day 3, although ticagrelor induced a further 5% reduction (P < 0.05 vs. clopidogrel) whereas oedema was only reduced by ticagrelor (≈23% P < 0.05). Scar size decreased at Day 42 in ticagrelor-treated pigs vs. controls but not in clopidogrel-treated pigs. Left ventricular ejection fraction was higher 3 days post-MI in ticagrelor-treated pigs and persisted up to Day 42 (P < 0.05 vs. post-MI). Regional analysis revealed that control and clopidogrel-treated pigs had severe and extensive wall motion abnormalities in the jeopardized myocardium and a reduced myocardial viability that was not as evident in ticagrelor-treated pigs (χ2P < 0.05 vs. ticagrelor). Only ticagrelor enhanced myocardial AMPK and Akt/PKB activation and reduced aquaporin-4 levels (P < 0.05 vs. control and clopidogrel). No differences were observed in vessel density and fibrosis markers among groups. Conclusions Ticagrelor is more efficient than clopidogrel in attenuating myocardial structural and functional alterations post-MI and in improving cardiac healing. These benefits are associated with persistent AMPK and Akt/PKB activation.
Collapse
Affiliation(s)
- Gemma Vilahur
- Program ICCC, IR-Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, c/Sant Antoni M a Claret 167, Barcelona, Spain.,CIBERCV, Instituto Salud Carlos III, Madrid, Spain
| | - Manuel Gutiérrez
- Program ICCC, IR-Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, c/Sant Antoni M a Claret 167, Barcelona, Spain.,Radiology Unit, Hospital de la Santa Creu i Sant Pau (HSCSP), Barcelona, Spain
| | - Laura Casani
- Program ICCC, IR-Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, c/Sant Antoni M a Claret 167, Barcelona, Spain.,CIBERCV, Instituto Salud Carlos III, Madrid, Spain
| | - Carmen Lambert
- Program ICCC, IR-Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, c/Sant Antoni Ma Claret 167, Barcelona, Spain
| | - Guiomar Mendieta
- Program ICCC, IR-Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, c/Sant Antoni M a Claret 167, Barcelona, Spain.,Cardiology Department, Hospital Clinico, Barcelona, Spain
| | - Soumaya Ben-Aicha
- Program ICCC, IR-Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, c/Sant Antoni Ma Claret 167, Barcelona, Spain
| | - Antoni Capdevila
- Radiology Unit, Hospital de la Santa Creu i Sant Pau (HSCSP), Barcelona, Spain
| | - Guillem Pons-Lladó
- Cardiology Unit, Hospital de la Santa Creu i Sant Pau (HSCSP), Barcelona, Spain
| | - Francesc Carreras
- Cardiology Unit, Hospital de la Santa Creu i Sant Pau (HSCSP), Barcelona, Spain
| | - Leif Carlsson
- Cardiovascular and Metabolic Diseases, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Mölndal, Sweden
| | - Alberto Hidalgo
- Radiology Unit, Hospital de la Santa Creu i Sant Pau (HSCSP), Barcelona, Spain
| | - Lina Badimon
- Program ICCC, IR-Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, c/Sant Antoni M a Claret 167, Barcelona, Spain.,CIBERCV, Instituto Salud Carlos III, Madrid, Spain.,Cardiovascular Research Chair UAB (Autonomous University of Barcelona), Barcelona, Spain
| |
Collapse
|
38
|
Deidda M, Mercurio V, Cuomo A, Noto A, Mercuro G, Cadeddu Dessalvi C. Metabolomic Perspectives in Antiblastic Cardiotoxicity and Cardioprotection. Int J Mol Sci 2019; 20:ijms20194928. [PMID: 31590338 PMCID: PMC6801977 DOI: 10.3390/ijms20194928] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 09/24/2019] [Accepted: 09/26/2019] [Indexed: 12/23/2022] Open
Abstract
Despite advances in supportive and protective therapy for myocardial function, cardiovascular diseases due to antineoplastic therapy-primarily cardiomyopathy associated with contractile dysfunction-remain a major cause of morbidity and mortality. Because of the limitations associated with current therapies, investigators are searching for alternative strategies that can timely recognise cardiovascular damage-thus permitting a quick therapeutic approach-or prevent the development of the disease. Damage to the heart can result from both traditional chemotherapeutic agents, such as anthracyclines, and new targeted therapies, such as tyrosine kinase inhibitors. In recent years, metabolomics has proved to be a practical tool to highlight fundamental changes in the metabolic state in several pathological conditions. In this article, we present the state-of-the-art technology with regard to the metabolic mechanisms underlying cardiotoxicity and cardioprotection.
Collapse
Affiliation(s)
- Martino Deidda
- Department of Medical Sciences and Public Health, University of Cagliari, 09042 Monserrato-Cagliari, Italy.
| | - Valentina Mercurio
- Department of Translational Medical Sciences, Federico II University, 80131 Naples, Italy.
| | - Alessandra Cuomo
- Department of Translational Medical Sciences, Federico II University, 80131 Naples, Italy.
| | - Antonio Noto
- Department of Medical Sciences and Public Health, University of Cagliari, 09042 Monserrato-Cagliari, Italy.
| | - Giuseppe Mercuro
- Department of Medical Sciences and Public Health, University of Cagliari, 09042 Monserrato-Cagliari, Italy.
| | - Christian Cadeddu Dessalvi
- Department of Medical Sciences and Public Health, University of Cagliari, 09042 Monserrato-Cagliari, Italy.
| |
Collapse
|
39
|
Lysine acetyltransferases and lysine deacetylases as targets for cardiovascular disease. Nat Rev Cardiol 2019; 17:96-115. [DOI: 10.1038/s41569-019-0235-9] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/26/2019] [Indexed: 12/28/2022]
|
40
|
Bailcalin Protects against Diabetic Cardiomyopathy through Keap1/Nrf2/AMPK-Mediated Antioxidative and Lipid-Lowering Effects. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:3206542. [PMID: 31354905 PMCID: PMC6636513 DOI: 10.1155/2019/3206542] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Revised: 01/15/2019] [Accepted: 03/04/2019] [Indexed: 12/16/2022]
Abstract
Previous studies demonstrated that Bailcalin (BAI) prevented cardiac injuries under different disease models. Whether BAI protected against type 2 diabetes mellitus- (T2DM-) associated cardiomyopathy was investigated in this study. T2DM was established by the combination of streptozotocin injection and high-fat diet in mice. BAI was administered daily for 6 months. After evaluating cardiac functions, mice hearts were removed and processed for morphological, biochemical, and molecular mechanism analyses. Neonatal rat cardiomyocytes (NRCM) were isolated and treated with high glucose and palmitate (HG/Pal) for in vitro investigation. BAI significantly ameliorated T2DM-induced cardiomyocyte hypertrophy, interstitial fibrosis, and lipid accumulation accompanied by markedly improved cardiac functions in diabetic mice. Mechanically, BAI restored decreased phosphorylation of AMPK and enhanced expression and nuclei translocation of Nrf2. In in vitro experiments, BAI also prevented NRCM from HG/Pal-induced apoptosis and oxidative stress injuries by increasing p-AMPK and Nrf2 accumulation. The means by which BAI restored p-AMPK seemed to be related to the antioxidative effects of Nrf2 after silencing AMPK or Nrf2 in NRCM. Furthermore, BAI regulated Nrf2 by inhibiting Nrf2 ubiquitination and consequent degradation mediated by Keap1. This study showed that BAI alleviated diabetes-associated cardiac dysfunction and cardiomyocyte injuries in vivo and in vitro via Keap1/Nrf2/AMPK-mediated antioxidation and lipid-lowering effects. BAI might be a potential adjuvant drug for diabetes cardiomyopathy treatment.
Collapse
|
41
|
Sharma AX, Quittner-Strom EB, Lee Y, Johnson JA, Martin SA, Yu X, Li J, Lu J, Cai Z, Chen S, Wang MY, Zhang Y, Pearson MJ, Dorn AC, McDonald JG, Gordillo R, Yan H, Thai D, Wang ZV, Unger RH, Holland WL. Glucagon Receptor Antagonism Improves Glucose Metabolism and Cardiac Function by Promoting AMP-Mediated Protein Kinase in Diabetic Mice. Cell Rep 2019; 22:1760-1773. [PMID: 29444429 PMCID: PMC5978750 DOI: 10.1016/j.celrep.2018.01.065] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 11/30/2017] [Accepted: 01/22/2018] [Indexed: 12/13/2022] Open
Abstract
The antidiabetic potential of glucagon receptor antagonism presents an opportunity for use in an insulin-centric clinical environment. To investigate the metabolic effects of glucagon receptor antagonism in type 2 diabetes, we treated Leprdb/db and Lepob/ob mice with REMD 2.59, a human monoclonal antibody and competitive antagonist of the glucagon receptor. As expected, REMD 2.59 suppresses hepatic glucose production and improves glycemia. Surprisingly, it also enhances insulin action in both liver and skeletal muscle, coinciding with an increase in AMP-activated protein kinase (AMPK)-mediated lipid oxidation. Furthermore, weekly REMD 2.59 treatment over a period of months protects against diabetic cardiomyopathy. These functional improvements are not derived simply from correcting the systemic milieu; nondiabetic mice with cardiac-specific overexpression of lipoprotein lipase also show improvements in contractile function after REMD 2.59 treatment. These observations suggest that hyperglucagonemia enables lipotoxic conditions, allowing the development of insulin resistance and cardiac dysfunction during disease progression.
Collapse
Affiliation(s)
- Ankit X Sharma
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390-8549, USA
| | - Ezekiel B Quittner-Strom
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390-8549, USA
| | - Young Lee
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390-8549, USA; Medical Service, Veteran's Administration North Texas Health Care System, Dallas, TX 75216, USA
| | - Joshua A Johnson
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390-8549, USA
| | - Sarah A Martin
- Department of Molecular Genetics, The University of Texas Southwestern Medical Center, Dallas, TX 75390-8549, USA
| | - Xinxin Yu
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390-8549, USA; Medical Service, Veteran's Administration North Texas Health Care System, Dallas, TX 75216, USA
| | - Jianping Li
- Division of Cardiology, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390-8549, USA
| | - John Lu
- REMD Biotherapeutics Inc., Camarillo, CA 93012, USA
| | | | - Shiuhwei Chen
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390-8549, USA
| | - May-Yun Wang
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390-8549, USA; Medical Service, Veteran's Administration North Texas Health Care System, Dallas, TX 75216, USA
| | - Yiyi Zhang
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390-8549, USA
| | - Mackenzie J Pearson
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390-8549, USA
| | - Andie C Dorn
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390-8549, USA
| | - Jeffrey G McDonald
- Department of Molecular Genetics, The University of Texas Southwestern Medical Center, Dallas, TX 75390-8549, USA; Center for Human Nutrition, The University of Texas Southwestern Medical Center, Dallas, TX 75390-8549, USA
| | - Ruth Gordillo
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390-8549, USA
| | - Hai Yan
- REMD Biotherapeutics Inc., Camarillo, CA 93012, USA
| | - Dung Thai
- REMD Biotherapeutics Inc., Camarillo, CA 93012, USA
| | - Zhao V Wang
- Division of Cardiology, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390-8549, USA
| | - Roger H Unger
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390-8549, USA; Medical Service, Veteran's Administration North Texas Health Care System, Dallas, TX 75216, USA
| | - William L Holland
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390-8549, USA.
| |
Collapse
|
42
|
Antoniou CK, Manolakou P, Magkas N, Konstantinou K, Chrysohoou C, Dilaveris P, Gatzoulis KA, Tousoulis D. Cardiac Resynchronisation Therapy and Cellular Bioenergetics: Effects Beyond Chamber Mechanics. Eur Cardiol 2019; 14:33-44. [PMID: 31131035 PMCID: PMC6523053 DOI: 10.15420/ecr.2019.2.2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Cardiac resynchronisation therapy is a cornerstone in the treatment of advanced dyssynchronous heart failure. However, despite its widespread clinical application, precise mechanisms through which it exerts its beneficial effects remain elusive. Several studies have pointed to a metabolic component suggesting that, both in concert with alterations in chamber mechanics and independently of them, resynchronisation reverses detrimental changes to cellular metabolism, increasing energy efficiency and metabolic reserve. These actions could partially account for the existence of responders that improve functionally but not echocardiographically. This article will attempt to summarise key components of cardiomyocyte metabolism in health and heart failure, with a focus on the dyssynchronous variant. Both chamber mechanics-related and -unrelated pathways of resynchronisation effects on bioenergetics – stemming from the ultramicroscopic level – and a possible common underlying mechanism relating mechanosensing to metabolism through the cytoskeleton will be presented. Improved insights regarding the cellular and molecular effects of resynchronisation on bioenergetics will promote our understanding of non-response, optimal device programming and lead to better patient care.
Collapse
Affiliation(s)
| | - Panagiota Manolakou
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens Athens, Greece
| | - Nikolaos Magkas
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens Athens, Greece
| | - Konstantinos Konstantinou
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens Athens, Greece
| | - Christina Chrysohoou
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens Athens, Greece
| | - Polychronis Dilaveris
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens Athens, Greece
| | - Konstantinos A Gatzoulis
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens Athens, Greece
| | - Dimitrios Tousoulis
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens Athens, Greece
| |
Collapse
|
43
|
Cardiac adaptation to exercise training in health and disease. Pflugers Arch 2019; 472:155-168. [PMID: 31016384 DOI: 10.1007/s00424-019-02266-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2019] [Revised: 02/13/2019] [Accepted: 02/15/2019] [Indexed: 02/08/2023]
Abstract
The heart is the primary pump that circulates blood through the entire cardiovascular system, serving many important functions in the body. Exercise training provides favorable anatomical and physiological changes that reduce the risk of heart disease and failure. Compared with pathological cardiac hypertrophy, exercise-induced physiological cardiac hypertrophy leads to an improvement in heart function. Exercise-induced cardiac remodeling is associated with gene regulatory mechanisms and cellular signaling pathways underlying cellular, molecular, and metabolic adaptations. Exercise training also promotes mitochondrial biogenesis and oxidative capacity leading to a decrease in cardiovascular disease. In this review, we summarized the exercise-induced adaptation in cardiac structure and function to understand cellular and molecular signaling pathways and mechanisms in preclinical and clinical trials.
Collapse
|
44
|
Wu J, Guo S, Zhang Y, Hu J, Lin W, Zhang B, Chen X, Wang H, Mo H, Zhan T. Yiqi Huoxue Decoction modifies the expression of myocardial cytoskeleton-associated proteins by regulating the AMPK signaling pathway in H9c2 cells exposed to hypoxic conditions. JOURNAL OF TRADITIONAL CHINESE MEDICAL SCIENCES 2019. [DOI: 10.1016/j.jtcms.2019.04.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
|
45
|
Lu Q, Li X, Liu J, Sun X, Rousselle T, Ren D, Tong N, Li J. AMPK is associated with the beneficial effects of antidiabetic agents on cardiovascular diseases. Biosci Rep 2019; 39:BSR20181995. [PMID: 30710062 PMCID: PMC6379227 DOI: 10.1042/bsr20181995] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Revised: 01/21/2019] [Accepted: 01/31/2019] [Indexed: 02/06/2023] Open
Abstract
Diabetics have higher morbidity and mortality in cardiovascular disease (CVD). A variety of antidiabetic agents are available for clinical choice. Cardiovascular (CV) safety assessment of these agents is crucial in addition to hypoglycemic effect before clinical prescription. Adenosine 5'-monophosphate-activated protein kinase (AMPK) is an important cell energy sensor, which plays an important role in regulating myocardial energy metabolism, reducing ischemia and ischemia/reperfusion (I/R) injury, improving heart failure (HF) and ventricular remodeling, ameliorating vascular endothelial dysfunction, antichronic inflammation, anti-apoptosis, and regulating autophagy. In this review, we summarized the effects of antidiabetic agents to CVD according to basic and clinical research evidence and put emphasis on whether these agents can play roles in CV system through AMPK-dependent signaling pathways. Metformin has displayed definite CV benefits related to AMPK. Sodium-glucose cotransporter 2 inhibitors also demonstrate sufficient clinical evidence for CV protection, but the mechanisms need further exploration. Glucagon-likepeptide1 analogs, dipeptidyl peptidase-4 inhibitors, α-glucosidase inhibitors and thiazolidinediones also show some AMPK-dependent CV benefits. Sulfonylureas and meglitinides may be unfavorable to CV system. AMPK is becoming a promising target for the treatment of diabetes, metabolic syndrome and CVD. But there are still some questions to be answered.
Collapse
Affiliation(s)
- Qingguo Lu
- Department of Endocrinology and Metabolism, West China Hospital of Sichuan University, 610041 Chengdu, China
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, 39216 Jackson, MS, U.S.A
| | - Xuan Li
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, 39216 Jackson, MS, U.S.A
| | - Jia Liu
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, 39216 Jackson, MS, U.S.A
- Department of Geriatrics, The First Hospital of Jilin University, 130021 Changchun, China
| | - Xiaodong Sun
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, 39216 Jackson, MS, U.S.A
- Department of Endocrinology, Affiliated Hospital of Weifang Medical University, 261000 Weifang, China
| | - Thomas Rousselle
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, 39216 Jackson, MS, U.S.A
| | - Di Ren
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, 39216 Jackson, MS, U.S.A
| | - Nanwei Tong
- Department of Endocrinology and Metabolism, West China Hospital of Sichuan University, 610041 Chengdu, China
| | - Ji Li
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, 39216 Jackson, MS, U.S.A.
| |
Collapse
|
46
|
Zhao J, Du J, Pan Y, Chen T, Zhao L, Zhu Y, Chen Y, Zheng Y, Liu Y, Sun L, Hang P, Du Z. Activation of cardiac TrkB receptor by its small molecular agonist 7,8-dihydroxyflavone inhibits doxorubicin-induced cardiotoxicity via enhancing mitochondrial oxidative phosphorylation. Free Radic Biol Med 2019; 130:557-567. [PMID: 30472367 DOI: 10.1016/j.freeradbiomed.2018.11.024] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 11/09/2018] [Accepted: 11/20/2018] [Indexed: 02/08/2023]
Abstract
Brain-derived neurotrophic factor (BDNF)/tropomyosin-related receptor kinase B (TrkB) pathway has been revealed as a novel therapeutic target for several neurological diseases. Recently, small-molecule TrkB agonist 7,8-dihydroxyflavone (7,8-DHF) has received considerable attention as a novel potential candidate for the treatment of various BDNF-implicated human disorders. However, its roles in cardiac diseases are not fully understood. Here, the present study aimed to clarify the effects and mechanisms of 7,8-DHF on doxorubicin (Dox)-induced cardiotoxicity. Kunming mice and H9c2 cells were employed to investigate the functional role of 7,8-DHF both in vivo and in vitro. 7,8-DHF markedly increased cell viability and reduced cell death of Dox-treated cells. Meanwhile, 7,8-DHF significantly increased mitochondrial respiration, membrane potential, and optic atrophy 1 (OPA1) protein expression. 7,8-DHF improved cardiac function and attenuated cardiac injury in Dox mice model. Expression of AMP-activated protein kinase (AMPK) and signal transducers and activators of transcription 3 (STAT3) was restored by 7,8-DHF. Furthermore, the protective role of 7,8-DHF was abolished by ANA-12 (a specific antagonist of TrkB). In elucidating the molecular mechanism, the phosphorylation of Akt was significantly increased while extracellular regulated protein kinase (ERK) was decreased after 7,8-DHF treatment. The regulatory effects of 7,8-DHF on STAT3 and AMPK was reversed by Akt inhibitor. In summary, 7,8-DHF attenuated Dox-induced cardiotoxicity by activating Akt and increasing mitochondrial oxidative phosphorylation and thereby regulating STAT3, AMPK, and ERK signals. The present study enhanced current understanding of TrkB receptor in the cardiovascular system and provided a novel target for prevention and treatment of heart diseases.
Collapse
Affiliation(s)
- Jing Zhao
- Department of Cardiology, the First Affiliated Hospital of Harbin Medical University (Key Laboratory of Cardiac Diseases and Heart Failure, Harbin Medical University), Harbin 150001, PR China
| | - Jingjing Du
- Institute of Clinical Pharmacology, the Second Affiliated Hospital of Harbin Medical University (The University Key Laboratory of Drug Research, Heilongjiang Province), Harbin 150086, PR China
| | - Yang Pan
- Institute of Clinical Pharmacology, the Second Affiliated Hospital of Harbin Medical University (The University Key Laboratory of Drug Research, Heilongjiang Province), Harbin 150086, PR China
| | - Tingting Chen
- Institute of Clinical Pharmacology, the Second Affiliated Hospital of Harbin Medical University (The University Key Laboratory of Drug Research, Heilongjiang Province), Harbin 150086, PR China
| | - Lihui Zhao
- Institute of Clinical Pharmacology, the Second Affiliated Hospital of Harbin Medical University (The University Key Laboratory of Drug Research, Heilongjiang Province), Harbin 150086, PR China
| | - Yanmeng Zhu
- Department of Pharmacology, Harbin Medical University, Harbin 150081, PR China
| | - Yingfu Chen
- Institute of Clinical Pharmacology, the Second Affiliated Hospital of Harbin Medical University (The University Key Laboratory of Drug Research, Heilongjiang Province), Harbin 150086, PR China
| | - Yuyang Zheng
- Institute of Clinical Pharmacology, the Second Affiliated Hospital of Harbin Medical University (The University Key Laboratory of Drug Research, Heilongjiang Province), Harbin 150086, PR China
| | - Yu Liu
- Institute of Clinical Pharmacology, the Second Affiliated Hospital of Harbin Medical University (The University Key Laboratory of Drug Research, Heilongjiang Province), Harbin 150086, PR China
| | - Lihua Sun
- Department of Pharmacology, Harbin Medical University, Harbin 150081, PR China
| | - Pengzhou Hang
- Institute of Clinical Pharmacology, the Second Affiliated Hospital of Harbin Medical University (The University Key Laboratory of Drug Research, Heilongjiang Province), Harbin 150086, PR China; Department of Clinical Pharmacology, College of Pharmacy, Harbin Medical University, Harbin 150081, PR China.
| | - Zhimin Du
- Institute of Clinical Pharmacology, the Second Affiliated Hospital of Harbin Medical University (The University Key Laboratory of Drug Research, Heilongjiang Province), Harbin 150086, PR China; Department of Clinical Pharmacology, College of Pharmacy, Harbin Medical University, Harbin 150081, PR China.
| |
Collapse
|
47
|
Lee JA, Kim HR, Kim J, Park KD, Kim DJ, Hwang O. The Novel Neuroprotective Compound KMS99220 Has an Early Anti-neuroinflammatory Effect via AMPK and HO-1, Independent of Nrf2. Exp Neurobiol 2018; 27:408-418. [PMID: 30429650 PMCID: PMC6221835 DOI: 10.5607/en.2018.27.5.408] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 10/10/2018] [Accepted: 10/11/2018] [Indexed: 12/17/2022] Open
Abstract
We have previously reported a novel synthetic compound KMS99220 that prevented degeneration of the nigral dopaminergic neurons and the associated motor deficits, suggesting a neuroprotective therapeutic utility for Parkinson's disease. Microglia are closely associated with neuroinflammation, which plays a key role in the pathogenesis of neurodegenerative diseases. In this study, we investigated the effects of KMS99220 on the signaling involving AMP-activated protein kinase (AMPK) and heme oxygenase-1 (HO-1), the enzymes thought to regulate inflammation. KMS99220 was shown to elevate the enzyme activity of purified AMPK, and phosphorylation of cellular AMPK in BV2 microglia. It increased the level of HO-1, and this was attenuated by AMPK inhibitors. KMS99220 lowered phosphorylation of IκB, nuclear translocation of NFκB, induction of inducible nitric oxide synthase, and generation of nitric oxide in BV2 cells that had been challenged with lipopolysaccharide. This anti-inflammatory response involved HO-1, because both its pharmacological inhibition and knockdown of its expression abolished the response. The AMPK inhibitors also reversed the anti-inflammatory effects of KMS99220. The induction of HO-1 by KMS99220 occurred within 1 h, and this appeared not to involve the transcription factor Nrf2, because Nrf2 knockdown did not affect the compound's HO-1 inducing- and anti-inflammatory effects in this time window. These findings indicated that KMS99220 leads to AMPK-induced HO-1 expression in microglia, which in turn plays an important role in early anti-inflammatory signaling. Together with its neuroprotective property, KMS99220 may serve as a feasible therapeutic agent against neuroinflammation and neurodegeneration.
Collapse
Affiliation(s)
- Ji Ae Lee
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Hye Ri Kim
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Jiyoung Kim
- Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea
| | - Ki Duk Park
- Convergence Research Center for Diagnosis, Treatment and Care System of Dementia, Korea Institute of Science and Technology, Seoul 02792, Korea
| | - Dong Jin Kim
- Center for Neuro-Medicine, Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, Korea
| | - Onyou Hwang
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul 05505, Korea
| |
Collapse
|
48
|
Abstract
The nuclear receptor peroxisome proliferator-activated receptor δ (PPARδ) can transcriptionally regulate target genes. PPARδ exerts essential regulatory functions in the heart, which requires constant energy supply. PPARδ plays a key role in energy metabolism, controlling not only fatty acid (FA) and glucose oxidation, but also redox homeostasis, mitochondrial biogenesis, inflammation, and cardiomyocyte proliferation. PPARδ signaling is impaired in the heart under various pathological conditions, such as pathological cardiac hypertrophy, myocardial ischemia/reperfusion, doxorubicin cardiotoxicity and diabetic cardiomyopathy. PPARδ deficiency in the heart leads to cardiac dysfunction, myocardial lipid accumulation, cardiac hypertrophy/remodeling and heart failure. This article provides an up-today overview of this research area and discusses the role of PPARδ in the heart in light of the complex mechanisms of its transcriptional regulation and its potential as a translatable therapeutic target for the treatment of cardiac disorders.
Collapse
Affiliation(s)
- Qinglin Yang
- Cardiovascular Center of Excellence, LSU Healther Science Center, 533 Bolivar St, New Orleans, LA 70112, USA
| | - Qinqiang Long
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, China
| |
Collapse
|
49
|
Lee HW, Ahmad M, Weldrick JJ, Wang HW, Burgon PG, Leenen FHH. Effects of exercise training and TrkB blockade on cardiac function and BDNF-TrkB signaling postmyocardial infarction in rats. Am J Physiol Heart Circ Physiol 2018; 315:H1821-H1834. [PMID: 30311496 DOI: 10.1152/ajpheart.00245.2018] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Exercise training is beneficial for preserving cardiac function postmyocardial infarction (post-MI), but the underlying mechanisms are not well understood. We investigated one possible mechanism, brain-derived neurotrophic factor (BDNF)-tropomyosin-related kinase B (TrkB) signaling, with the TrkB blocker ANA-12 (0.5 mg·kg-1·day-1). Male Wistar rats underwent sham surgery or ligation of the left descending coronary artery. The surviving MI rats were allocated as follows: sedentary MI rats treated with vehicle, exercise-trained MI rats treated with vehicle, and exercise-trained MI rats treated with ANA-12. Exercise training was done 5 days/wk for 4 wk on a motor-driven treadmill. At the end, left ventricular (LV) function was evaluated by echocardiography and a Millar catheter. Mature BDNF and downstream effectors of BDNF-TrkB signaling, Ca2+/calmodulin-dependent protein kinase II (CaMKII), Akt, and AMP-activated protein kinase (AMPK), were assessed in the noninfarct area of the LV by Western blot analysis. Exercise training increased stroke volume and cardiac index and attenuated the decrease in ejection fraction (EF) and increase in LV end-diastolic pressure post-MI. ANA-12 blocked the improvement of EF and attenuated the increases in stroke volume and cardiac index but did not affect LV end-diastolic pressure. Exercise training post-MI prevented decreases in mature BDNF, phosphorylated (p-)CaMKII, p-Akt, and p-AMPKα expression. These effects were all blocked by ANA-12 except for p-AMPKα. In conclusion, the exercise-induced improvement of EF is mediated by the BDNF-TrkB axis and the downstream effectors CaMKII and Akt. BDNF-TrkB signaling appears to contribute to the improvement in systolic function by exercise training. NEW & NOTEWORTHY Exercise training improves ejection fraction and left ventricular end-diastolic pressure (LVEDP) and increases stroke volume and cardiac index in rats postmyocardial infarction (post-MI). The improvement of EF but not LVEDP is mediated by activation of the brain-derived neurotrophic factor (BDNF)-tropomyosin-related kinase B (TrkB) axis and downstream effectors Ca2+/calmodulin-dependent protein kinase II (CaMKII) and Akt. This suggests that activation of BDNF-TrkB signaling and CaMKII and Akt is a promising target to attenuate progressive cardiac dysfunction post-MI.
Collapse
Affiliation(s)
- Heow Won Lee
- Brain and Heart Research Group, University of Ottawa Heart Institute , Ottawa, ON , Canada
| | - Monir Ahmad
- Brain and Heart Research Group, University of Ottawa Heart Institute , Ottawa, ON , Canada
| | - Jonathan J Weldrick
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa , Ottawa, ON , Canada
| | - Hong-Wei Wang
- Brain and Heart Research Group, University of Ottawa Heart Institute , Ottawa, ON , Canada
| | - Patrick G Burgon
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa , Ottawa, ON , Canada
| | - Frans H H Leenen
- Brain and Heart Research Group, University of Ottawa Heart Institute , Ottawa, ON , Canada
| |
Collapse
|
50
|
Dziubak A, Wójcicka G, Wojtak A, Bełtowski J. Metabolic Effects of Metformin in the Failing Heart. Int J Mol Sci 2018; 19:ijms19102869. [PMID: 30248910 PMCID: PMC6213955 DOI: 10.3390/ijms19102869] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 09/11/2018] [Accepted: 09/17/2018] [Indexed: 01/03/2023] Open
Abstract
Accumulating evidence shows that metformin is an insulin-sensitizing antidiabetic drug widely used in the treatment of type 2 diabetes mellitus (T2DM), which can exert favorable effects on cardiovascular risk and may be safely used in patients with heart failure (HF), and even able to reduce the incidence of HF and to reduce HF mortality. In failing hearts, metformin improves myocardial energy metabolic status through the activation of AMP (adenosine monophosphate)-activated protein kinase (AMPK) and the regulation of lipid and glucose metabolism. By increasing nitric oxide (NO) bioavailability, limiting interstitial fibrosis, reducing the deposition of advanced glycation end-products (AGEs), and inhibiting myocardial cell apoptosis metformin reduces cardiac remodeling and hypertrophy, and thereby preserves left ventricular systolic and diastolic functions. While a lot of preclinical and clinical studies showed the cardiovascular safety of metformin therapy in diabetic patients and HF, to confirm observed benefits, the specific large-scale trials configured for HF development in diabetic patients as a primary endpoints are necessary.
Collapse
Affiliation(s)
- Aleksandra Dziubak
- Department of Pathophysiology, Medical University of Lublin, ul. Jaczewskiego 8b, 20-090 Lublin, Poland.
| | - Grażyna Wójcicka
- Department of Pathophysiology, Medical University of Lublin, ul. Jaczewskiego 8b, 20-090 Lublin, Poland.
| | - Andrzej Wojtak
- Department of Vascular Surgery, Medical University of Lubin, 20-090 Lublin, Poland.
| | - Jerzy Bełtowski
- Department of Pathophysiology, Medical University of Lublin, ul. Jaczewskiego 8b, 20-090 Lublin, Poland.
| |
Collapse
|