1
|
Yanjuan L, Shuangyou D, Ying W, Xing C, Yue C, Zixuan Y, Shumeng Z, Lingli C, Jie L. The Research Progress: Cuproptosis and Copper Metabolism in Regulating Cardiovascular Diseases. J Cardiovasc Pharmacol 2025; 85:89-96. [PMID: 39591592 DOI: 10.1097/fjc.0000000000001653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 11/09/2024] [Indexed: 11/28/2024]
Abstract
ABSTRACT Studies have shown an association between cardiovascular disease and abnormal copper metabolism. Cuproptosis is caused by the accumulation of copper in vivo, and is a newly identified form of cell death. It regulates cardiovascular diseases by affecting vascular endothelial function and myocardial energy metabolism through pathways such as oxidative stress, mitochondrial function, and gene expression. The treatment of copper accumulation in Traditional Chinese Medicine primarily involves heat-clearing and detoxification therapy, supplemented with diuretic therapy. In contrast, Western medicine mainly uses copper chelators. Flavonoids are common active ingredients used in the treatment of copper metabolism-related and cardiovascular diseases. In this article, we reviewed the relationship between copper metabolism, cuproptosis, and cardiovascular disease, providing novel strategies for preventing and treating cardiovascular disease; our ultimate aim is to encourage inspiration and contemplation among readers.
Collapse
Affiliation(s)
- Liu Yanjuan
- Hunan University of Chinese Medicine, Changsha, China
- Hunan Provincial Key Laboratory of TCM Diagnosis, Changsha, China
| | - Deng Shuangyou
- Hunan University of Chinese Medicine, Changsha, China
- Hunan Provincial Key Laboratory of TCM Diagnosis, Changsha, China
| | - Wang Ying
- Hunan University of Chinese Medicine, Changsha, China
- Hunan Provincial Key Laboratory of TCM Diagnosis, Changsha, China
| | - Chen Xing
- Hunan University of Chinese Medicine, Changsha, China
- Hunan Provincial Key Laboratory of TCM Diagnosis, Changsha, China
| | - Chen Yue
- Hunan University of Chinese Medicine, Changsha, China
- Hunan Provincial Key Laboratory of TCM Diagnosis, Changsha, China
| | - Yu Zixuan
- Hunan University of Chinese Medicine, Changsha, China
- Hunan Provincial Key Laboratory of TCM Diagnosis, Changsha, China
| | - Zhang Shumeng
- Hunan University of Chinese Medicine, Changsha, China
- Hunan Provincial Key Laboratory of TCM Diagnosis, Changsha, China
| | - Chen Lingli
- Hunan University of Chinese Medicine, Changsha, China
| | - Li Jie
- Hunan University of Chinese Medicine, Changsha, China
- Hunan Provincial Key Laboratory of TCM Diagnosis, Changsha, China
| |
Collapse
|
2
|
Silva CNDF, Bessa ADSMD, Costa JMD, Lopes PR, Neves ÂR, Teles Bombardelli MML, Colugnati DB, Pedrino GR, Mendes EP, Santos RASD, Biancardi MF, Santos FCAD, Castro CH. Mas receptor blockade impairs exercise-induced cardiac hypertrophy. Peptides 2024; 181:171296. [PMID: 39265810 DOI: 10.1016/j.peptides.2024.171296] [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: 07/25/2024] [Revised: 09/06/2024] [Accepted: 09/09/2024] [Indexed: 09/14/2024]
Abstract
Exercise training leads to physiological cardiac hypertrophy and the protective axis of the renin-angiotensin system composed of angiotensin-converting enzyme 2, angiotensin-(1-7), and Mas receptor seems involved in this process. However, the role of the basal activity of the Mas receptor in exercise-induced physiological cardiac hypertrophy is still unclear. We evaluated the effects of the Mas receptor blockade on the left ventricular structure and function of rats submitted to running training. Rats were assigned to 4 groups: sedentary (S), sedentary + A-779 (Mas receptor antagonist, 120 µg/kg/day, i.p.; SA), trained (60-minute treadmill running sessions, five days a week, 8 weeks; T), and trained + A-779 (TA). Systolic blood pressure was higher in sedentary and trained rats treated with A-779 at the end of the experimental period. The A-779 treatment prevented the left ventricular hypertrophy evoked by physical exercise and increased collagen deposition in sedentary and trained rats. Cardiomyocytes from the SA group presented increased length and thickness of the sarcomeres, elongated mitochondria, glycogen deposits, and enlarged cisterns of the sarcoplasmic reticulum. TA group presented a reduced sarcomere thickness and cytoplasm with a degenerative aspect. These findings show that the basal activity of the Mas receptor is essential for the proper turnover of the extracellular matrix in the myocardium and the maintenance of the sarcomeric structure of cardiomyocytes.
Collapse
Affiliation(s)
| | | | | | - Paulo Ricardo Lopes
- Department of Physiological Sciences, Federal University of Goiás, Goiânia, GO, Brazil
| | - Ângela Ribeiro Neves
- Department of Physiological Sciences, Federal University of Goiás, Goiânia, GO, Brazil
| | | | | | | | | | | | | | | | | |
Collapse
|
3
|
Gong S, Sui Y, Xiao M, Fu D, Xiong Z, Zhang L, Tian Q, Fu Y, Xiong W. Canagliflozin Mediates Mitophagy Through the AMPK/PINK1/Parkin Pathway to Alleviate ISO-induced Cardiac Remodeling. J Cardiovasc Pharmacol 2024; 84:496-505. [PMID: 39150485 DOI: 10.1097/fjc.0000000000001625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 07/26/2024] [Indexed: 08/17/2024]
Abstract
ABSTRACT Heart failure has always been a prevalent, disabling, and potentially life-threatening disease. For the treatment of heart failure, controlling cardiac remodeling is very important. In recent years, clinical trials have shown that sodium-glucose cotransporter-2 (SGLT-2) inhibitors not only excel in lowering glucose levels but also demonstrate favorable cardiovascular protective effects. However, the precise mechanisms behind the cardiovascular benefits of SGLT-2 inhibitors remain elusive. In this research, we assessed the impact of canagliflozin (CANA, an SGLT-2 inhibitor) on cardiac remodeling progression in mice and preliminarily elucidated the possible mechanism of action of the SGLT-2 inhibitor. Our results indicate that the administration of canagliflozin significantly attenuates myocardial hypertrophy and fibrosis and enhances cardiac ejection function in mice with isoprenaline (ISO)-induced cardiac remodeling. Notably, excessive mitophagy, along with mitochondrial structural abnormalities observed in ISO-induced cardiac remodeling, was mitigated by canagliflozin treatment, thereby attenuating cardiac remodeling progression. Furthermore, the differential expression of AMPK/PINK1/Parkin pathway-related proteins in ISO-induced cardiac remodeling was effectively reversed by canagliflozin, suggesting the therapeutic potential of targeting this pathway with the drug. Thus, our study indicates that canagliflozin holds promise in mitigating cardiac injury, enhancing cardiac function, and potentially exerting cardioprotective effects by modulating mitochondrial function and mitophagy through the AMPK/PINK1/Parkin pathway.
Collapse
MESH Headings
- Animals
- Male
- Mice
- AMP-Activated Protein Kinases/metabolism
- Canagliflozin/pharmacology
- Disease Models, Animal
- Fibrosis
- Isoproterenol
- Mice, Inbred C57BL
- Mitochondria, Heart/drug effects
- Mitochondria, Heart/enzymology
- Mitochondria, Heart/pathology
- Mitochondria, Heart/metabolism
- Mitophagy/drug effects
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/pathology
- Myocytes, Cardiac/enzymology
- Myocytes, Cardiac/metabolism
- Protein Kinases/metabolism
- Signal Transduction/drug effects
- Sodium-Glucose Transporter 2 Inhibitors/pharmacology
- Stroke Volume/drug effects
- Ubiquitin-Protein Ligases/metabolism
- Ventricular Function, Left/drug effects
- Ventricular Remodeling/drug effects
Collapse
Affiliation(s)
- Shaolin Gong
- Department of Cardiology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China ; and
| | - Yuan Sui
- Department of Cardiology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China ; and
| | - Mengxuan Xiao
- Department of Cardiology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China ; and
| | - Daoyao Fu
- Department of Cardiology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China ; and
| | - Zhiping Xiong
- Department of Cardiology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China ; and
| | - Liuping Zhang
- Department of Cardiology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China ; and
| | - Qingshan Tian
- Department of Cardiology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China ; and
| | - Yongnan Fu
- Department of Cardiology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China ; and
| | - Wenjun Xiong
- Department of Cardiology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China ; and
- Center of Reproduction, Development and Aging and Institute of Translation Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| |
Collapse
|
4
|
Lei XT, Pu DL, Shan G, Wu QN. Atorvastatin ameliorated myocardial fibrosis by inhibiting oxidative stress and modulating macrophage polarization in diabetic cardiomyopathy. World J Diabetes 2024; 15:1070-1073. [PMID: 38983803 PMCID: PMC11229961 DOI: 10.4239/wjd.v15.i6.1070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 03/07/2024] [Accepted: 04/07/2024] [Indexed: 06/11/2024] Open
Abstract
In this editorial, we commented on the article published in the recent issue of the World Journal of Diabetes. Diabetic cardiomyopathy (DCM) is characterized by myocardial fibrosis, ventricular hypertrophy and diastolic dysfunction in diabetic patients, which can cause heart failure and threaten the life of patients. The pathogenesis of DCM has not been fully clarified, and it may involve oxidative stress, inflammatory stimulation, apoptosis, and autophagy. There is lack of effective therapies for DCM in the clinical practice. Statins have been widely used in the clinical practice for years mainly to reduce cholesterol and stabilize arterial plaques, and exhibit definite cardiovascular protective effects. Studies have shown that statins also have anti-inflammatory and antioxidant effects. We were particularly concerned about the recent findings that atorvastatin alleviated myocardial fibrosis in db/db mice by regulating the antioxidant stress and anti-inflammatory effects of macrophage polarization on diabetic myocardium, and thereby improving DCM.
Collapse
Affiliation(s)
- Xiao-Tian Lei
- Department of Endocrinology, The First Affiliated Hospital of the Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Dan-Lan Pu
- Department of Endocrinology, Chongqing Yubei District People's Hospital, Chongqing 400030, China
| | - Geng Shan
- Department of Endocrinology, Dazu Hospital of Chongqing Medical University, The People's Hospital of Dazu, Chongqing 402360, China
| | - Qi-Nan Wu
- Department of Endocrinology, Dazu Hospital of Chongqing Medical University, The People's Hospital of Dazu, Chongqing 402360, China
| |
Collapse
|
5
|
Wang J, Zou J, Shi Y, Zeng N, Guo D, Wang H, Zhao C, Luan F, Zhang X, Sun J. Traditional Chinese medicine and mitophagy: A novel approach for cardiovascular disease management. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 128:155472. [PMID: 38461630 DOI: 10.1016/j.phymed.2024.155472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 02/06/2024] [Accepted: 02/20/2024] [Indexed: 03/12/2024]
Abstract
BACKGROUND Cardiovascular disease (CVD) remains the leading cause of morbidity and mortality worldwide, imposing an enormous economic burden on individuals and human society. Laboratory studies have identified several drugs that target mitophagy for the prevention and treatment of CVD. Only a few of these drugs have been successful in clinical trials, and most studies have been limited to animal and cellular models. Furthermore, conventional drugs used to treat CVD, such as antiplatelet agents, statins, and diuretics, often result in adverse effects on patients' cardiovascular, metabolic, and respiratory systems. In contrast, traditional Chinese medicine (TCM) has gained significant attention for its unique theoretical basis and clinical efficacy in treating CVD. PURPOSE This paper systematically summarizes all the herbal compounds, extracts, and active monomers used to target mitophagy for the treatment of CVD in the last five years. It provides valuable information for researchers in the field of basic cardiovascular research, pharmacologists, and clinicians developing herbal medicines with fewer side effects, as well as a useful reference for future mitophagy research. METHODS The search terms "cardiovascular disease," "mitophagy," "herbal preparations," "active monomers," and "cardiac disease pathogenesis" in combination with "natural products" and "diseases" were used to search for studies published in the past five years until January 2024. RESULTS Studies have shown that mitophagy plays a significant role in the progression and development of CVD, such as atherosclerosis (AS), heart failure (HF), myocardial infarction (MI), myocardial ischemia/reperfusion injury (MI/RI), cardiac hypertrophy, cardiomyopathy, and arrhythmia. Herbal compound preparations, crude extracts, and active monomers have shown potential as effective treatments for these conditions. These substances protect cardiomyocytes by inducing mitophagy, scavenging damaged mitochondria, and maintaining mitochondrial homeostasis. They display notable efficacy in combating CVD. CONCLUSION TCM (including herbal compound preparations, extracts, and active monomers) can treat CVD through various pharmacological mechanisms and signaling pathways by inducing mitophagy. They represent a hotspot for future cardiovascular basic research and a promising candidate for the development of future cardiovascular drugs with fewer side effects and better therapeutic efficacy.
Collapse
Affiliation(s)
- Jinhui Wang
- Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an 712046, Shaanxi, PR China
| | - Junbo Zou
- Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an 712046, Shaanxi, PR China
| | - Yajun Shi
- Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an 712046, Shaanxi, PR China
| | - Nan Zeng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, PR China
| | - Dongyan Guo
- Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an 712046, Shaanxi, PR China
| | - He Wang
- Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an 712046, Shaanxi, PR China
| | - Chongbo Zhao
- Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an 712046, Shaanxi, PR China
| | - Fei Luan
- Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an 712046, Shaanxi, PR China.
| | - Xiaofei Zhang
- Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an 712046, Shaanxi, PR China.
| | - Jing Sun
- Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an 712046, Shaanxi, PR China.
| |
Collapse
|
6
|
Xu G, Xu Y, Zhang Y, Kao G, Li J. miR-1268a Regulates Fatty Acid Metabolism by Targeting CD36 in Angiotensin II-induced Heart Failure. Cell Biochem Biophys 2024; 82:1193-1201. [PMID: 38619643 DOI: 10.1007/s12013-024-01268-y] [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] [Accepted: 04/03/2024] [Indexed: 04/16/2024]
Abstract
Multiple RNAs have been involved in the progress of heart failure. However, the role of miR-1268a in heart failure is still unclear. The differentially expressed miRNAs in heart failure was analyzed based on GEO dataset GSE104150. AC16 cells were treated with Angiotensin II (Ang II) to explore the role of miR-1268a in heart failure. The web tool miRWalk was used to analyze the targets of miR-1268a. miR-1268a was up-regulated in Ang II-treated AC16 cells. Ang II treatment markedly inhibited cell proliferation, ATP production, fatty acid (FA) uptake and enhanced levels of HF markers BNP and ST2, and oxidative stress of AC16 cells. Notably, inhibition of miR-1268a eliminated the inhibiting effect of Ang II on cell proliferation, ATP production, FA uptake and decreased levels of BNP an ST2, and oxidative stress on AC16 cells. Furthermore, CD36 was a target of miR-1268a and the CD36 level was decreased by miR-1268a mimics but increased by miR-1268a inhibitor in AC16 cells. miR-1268a regulates FA metabolism and oxidative stress in myocardial cells by targeting CD36 in heart failure.
Collapse
Affiliation(s)
- Gang Xu
- Department of Cardiovascular Medicine, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing, 400010, China
| | - Yi Xu
- Department of Cardiovascular Medicine, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing, 400010, China
| | - Ying Zhang
- Department of Cardiovascular Medicine, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing, 400010, China
| | - Guoying Kao
- Department of Cardiovascular Medicine, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing, 400010, China.
| | - Jun Li
- Department of Cardiovascular Medicine, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing, 400010, China.
| |
Collapse
|
7
|
Dhakal S, Macreadie IG. Simvastatin, Its Antimicrobial Activity and Its Prevention of Alzheimer's Disease. Microorganisms 2024; 12:1133. [PMID: 38930515 PMCID: PMC11205914 DOI: 10.3390/microorganisms12061133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 05/22/2024] [Accepted: 05/29/2024] [Indexed: 06/28/2024] Open
Abstract
Simvastatin, a blockbuster drug for treating hypercholesterolemia, has multifactorial benefits as an antimicrobial agent and plays a preventative role in reducing the incidence of Alzheimer's Disease (AD). Although most of the beneficial effects of simvastatin have been attributed to its ability to reduce cholesterol levels, recent scientific studies have suggested that its benefits are largely due to its pleiotropic effects in targeting other pathways, e.g., by inhibiting protein lipidation. There are certain pleiotropic effects that can be predicted from the inhibition of the mevalonate pathway; however, some of the effects of simvastatin in proteostasis lead to reduced levels of amyloid beta, the key contributor to AD. This review discusses the use of simvastatin as an antimicrobial agent and anti-AD drug.
Collapse
Affiliation(s)
- Sudip Dhakal
- Health and Biosecurity, Commonwealth Scientific and Industrial Research Organization (CSIRO), Geelong, VIC 3220, Australia;
| | - Ian G. Macreadie
- School of Science, RMIT University, Bundoora, VIC 3063, Australia
| |
Collapse
|
8
|
Peng F, Liao M, Jin W, Liu W, Li Z, Fan Z, Zou L, Chen S, Zhu L, Zhao Q, Zhan G, Ouyang L, Peng C, Han B, Zhang J, Fu L. 2-APQC, a small-molecule activator of Sirtuin-3 (SIRT3), alleviates myocardial hypertrophy and fibrosis by regulating mitochondrial homeostasis. Signal Transduct Target Ther 2024; 9:133. [PMID: 38744811 PMCID: PMC11094072 DOI: 10.1038/s41392-024-01816-1] [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: 08/15/2023] [Revised: 02/20/2024] [Accepted: 03/25/2024] [Indexed: 05/16/2024] Open
Abstract
Sirtuin 3 (SIRT3) is well known as a conserved nicotinamide adenine dinucleotide+ (NAD+)-dependent deacetylase located in the mitochondria that may regulate oxidative stress, catabolism and ATP production. Accumulating evidence has recently revealed that SIRT3 plays its critical roles in cardiac fibrosis, myocardial fibrosis and even heart failure (HF), through its deacetylation modifications. Accordingly, discovery of SIRT3 activators and elucidating their underlying mechanisms of HF should be urgently needed. Herein, we identified a new small-molecule activator of SIRT3 (named 2-APQC) by the structure-based drug designing strategy. 2-APQC was shown to alleviate isoproterenol (ISO)-induced cardiac hypertrophy and myocardial fibrosis in vitro and in vivo rat models. Importantly, in SIRT3 knockout mice, 2-APQC could not relieve HF, suggesting that 2-APQC is dependent on SIRT3 for its protective role. Mechanically, 2-APQC was found to inhibit the mammalian target of rapamycin (mTOR)-p70 ribosomal protein S6 kinase (p70S6K), c-jun N-terminal kinase (JNK) and transforming growth factor-β (TGF-β)/ small mother against decapentaplegic 3 (Smad3) pathways to improve ISO-induced cardiac hypertrophy and myocardial fibrosis. Based upon RNA-seq analyses, we demonstrated that SIRT3-pyrroline-5-carboxylate reductase 1 (PYCR1) axis was closely assoiated with HF. By activating PYCR1, 2-APQC was shown to enhance mitochondrial proline metabolism, inhibited reactive oxygen species (ROS)-p38 mitogen activated protein kinase (p38MAPK) pathway and thereby protecting against ISO-induced mitochondrialoxidative damage. Moreover, activation of SIRT3 by 2-APQC could facilitate AMP-activated protein kinase (AMPK)-Parkin axis to inhibit ISO-induced necrosis. Together, our results demonstrate that 2-APQC is a targeted SIRT3 activator that alleviates myocardial hypertrophy and fibrosis by regulating mitochondrial homeostasis, which may provide a new clue on exploiting a promising drug candidate for the future HF therapeutics.
Collapse
Affiliation(s)
- Fu Peng
- West China School of Pharmacy and Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Minru Liao
- West China School of Pharmacy and Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Wenke Jin
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Wei Liu
- West China School of Pharmacy and Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zixiang Li
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Zhichao Fan
- West China School of Pharmacy and Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Ling Zou
- West China School of Pharmacy and Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen, 518060, China
| | - Siwei Chen
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen, 518060, China
| | - Lingjuan Zhu
- School of Traditional Chinese Materia Medica, Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Qian Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Gu Zhan
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Liang Ouyang
- West China School of Pharmacy and Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Bo Han
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Jin Zhang
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen, 518060, China.
| | - Leilei Fu
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China.
| |
Collapse
|
9
|
Peng Y, Tao Y, Liu L, Zhang J, Wei B. Crosstalk among Reactive Oxygen Species, Autophagy and Metabolism in Myocardial Ischemia and Reperfusion Stages. Aging Dis 2024; 15:1075-1107. [PMID: 37728583 PMCID: PMC11081167 DOI: 10.14336/ad.2023.0823-4] [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: 07/03/2023] [Accepted: 08/23/2023] [Indexed: 09/21/2023] Open
Abstract
Myocardial ischemia is the most common cardiovascular disease. Reperfusion, an important myocardial ischemia tool, causes unexpected and irreversible damage to cardiomyocytes, resulting in myocardial ischemia/reperfusion (MI/R) injury. Upon stress, especially oxidative stress induced by reactive oxygen species (ROS), autophagy, which degrades the intracellular energy storage to produce metabolites that are recycled into metabolic pathways to buffer metabolic stress, is initiated during myocardial ischemia and MI/R injury. Excellent cardioprotective effects of autophagy regulators against MI and MI/R have been reported. Reversing disordered cardiac metabolism induced by ROS also exhibits cardioprotective action in patients with myocardial ischemia. Herein, we review current knowledge on the crosstalk between ROS, cardiac autophagy, and metabolism in myocardial ischemia and MI/R. Finally, we discuss the possible regulators of autophagy and metabolism that can be exploited to harness the therapeutic potential of cardiac metabolism and autophagy in the diagnosis and treatment of myocardial ischemia and MI/R.
Collapse
Affiliation(s)
- Yajie Peng
- Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China; School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China.
| | - Yachuan Tao
- Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China; School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China.
- Department of Pharmacology, School of Pharmaceutical Sciences, Fudan University, Shanghai, China
| | - Lingxu Liu
- Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China; School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China.
| | - Ji Zhang
- The First Affiliated Hospital of Zhengzhou University, Department of Pharmacy, Zhengzhou, Henan, China.
| | - Bo Wei
- Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China; School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
10
|
Chen Y, Li S, Yang Z, Wang T, Yin F, Zhao X, Zhang Y. Value of Bax and Bcl2 expression in peripheral blood mononuclear cells for clinical prognosis of patients with chronic heart failure. Medicine (Baltimore) 2024; 103:e36943. [PMID: 38241555 PMCID: PMC10798729 DOI: 10.1097/md.0000000000036943] [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: 03/20/2023] [Accepted: 07/28/2023] [Indexed: 01/21/2024] Open
Abstract
To investigate the expression of Bax and Bcl2 protein in peripheral blood mononuclear cells (PBMC) of patients with chronic heart failure (CHF), and to analyze their value for predicting major adverse cardiovascular event (MACE) in CHF patients. A total of 154 fasting venous blood samples from CHF patients were collected in our hospital from January 2017 to June 2019, and they were divided into 2 group according to whether MACE occurred during 3 years follow-up, MACE group and No-MACE group. Levels of Bax and Bcl2 protein expression in PBMC of CHF patients using enzyme-linked immunosorbent assay (ELISA), and then evaluated the predictive power of Bax and Bcl2 expression for MACE using logistic regression analysis and ROC curve. 62 (40.26%) of 154 CHF patients occurred MACE during follow-up, and there were significant differences in age, diabetes, LVEF, LDL-C and NYHA grade between MACE group and No-MACE group. Levels of Bax protein expression in PBMC of CHF patients in MACE group were significantly higher than those in No-MACE group, while levels of Bcl2 protein expression were significantly lower than those in No-MACE group, and Bax and Bcl2 protein levels increased and decreased with NYHA grades in MACE group and No-MACE group, respectively. Results of univariate and multivariate logistic regression analysis showed that Bax (OR, 1.026; 95% CI, 1.003-1.049; P = .027) and Bcl2 levels (OR, 0.952; 95% CI, 0.908-0.998; P = .041) were independent predictive factors for MACE in CHF patients. In addition, Bax and Bcl2 levels could be used to differentiate CHF patients at risk for MACE with an AUC of 0.744 (95% CI: 0.660-0.827) and an AUC of 0.743 (95% CI: 0.667-0.819), respectively. Levels of Bax and Bcl2 protein in PBMC could be used as independent predictive factors for MACE in CHF patients.
Collapse
Affiliation(s)
- Yangang Chen
- Department of Internal Medicine-Cardiovascular, Liangzhou Hospital of Wuwei City, Wuwei City, Gansu Province, China
| | - Shuiquan Li
- Department of Internal Medicine-Cardiovascular, Liangzhou Hospital of Wuwei City, Wuwei City, Gansu Province, China
| | - Zhenwen Yang
- Department of Internal Medicine-Cardiovascular, Liangzhou Hospital of Wuwei City, Wuwei City, Gansu Province, China
| | - Tianlu Wang
- Department of Internal Medicine-Cardiovascular, Liangzhou Hospital of Wuwei City, Wuwei City, Gansu Province, China
| | - Fahui Yin
- Department of Internal Medicine-Cardiovascular, Liangzhou Hospital of Wuwei City, Wuwei City, Gansu Province, China
| | - Xiangyu Zhao
- Department of Internal Medicine-Cardiovascular, Liangzhou Hospital of Wuwei City, Wuwei City, Gansu Province, China
| | - Yong Zhang
- Department of Internal Medicine-Cardiovascular, Liangzhou Hospital of Wuwei City, Wuwei City, Gansu Province, China
| |
Collapse
|
11
|
Gao S, Liu XP, Li TT, Chen L, Feng YP, Wang YK, Yin YJ, Little PJ, Wu XQ, Xu SW, Jiang XD. Animal models of heart failure with preserved ejection fraction (HFpEF): from metabolic pathobiology to drug discovery. Acta Pharmacol Sin 2024; 45:23-35. [PMID: 37644131 PMCID: PMC10770177 DOI: 10.1038/s41401-023-01152-0] [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: 04/19/2023] [Accepted: 08/08/2023] [Indexed: 08/31/2023] Open
Abstract
Heart failure (HF) with preserved ejection fraction (HFpEF) is currently a preeminent challenge for cardiovascular medicine. It has a poor prognosis, increasing mortality, and is escalating in prevalence worldwide. Despite accounting for over 50% of all HF patients, the mechanistic underpinnings driving HFpEF are poorly understood, thus impeding the discovery and development of mechanism-based therapies. HFpEF is a disease syndrome driven by diverse comorbidities, including hypertension, diabetes and obesity, pulmonary hypertension, aging, and atrial fibrillation. There is a lack of high-fidelity animal models that faithfully recapitulate the HFpEF phenotype, owing primarily to the disease heterogeneity, which has hampered our understanding of the complex pathophysiology of HFpEF. This review provides an updated overview of the currently available animal models of HFpEF and discusses their characteristics from the perspective of energy metabolism. Interventional strategies for efficiently utilizing energy substrates in preclinical HFpEF models are also discussed.
Collapse
Affiliation(s)
- Si Gao
- Department of Pharmacy, School of Medicine, Guangxi University of Science and Technology, Liuzhou, 545005, China
| | - Xue-Ping Liu
- Department of Pharmacy, School of Medicine, Guangxi University of Science and Technology, Liuzhou, 545005, China
| | - Ting-Ting Li
- Department of Pharmacy, School of Medicine, Guangxi University of Science and Technology, Liuzhou, 545005, China
| | - Li Chen
- Department of Pharmacy, School of Medicine, Guangxi University of Science and Technology, Liuzhou, 545005, China
| | - Yi-Ping Feng
- Department of Pharmacy, School of Medicine, Guangxi University of Science and Technology, Liuzhou, 545005, China
| | - Yu-Kun Wang
- Department of Pharmacy, School of Medicine, Guangxi University of Science and Technology, Liuzhou, 545005, China
| | - Yan-Jun Yin
- School of Pharmacy, Bengbu Medical College, Bengbu, 233000, China
| | - Peter J Little
- School of Pharmacy, University of Queensland, Pharmacy Australia Centre of Excellence, Woolloongabba, QLD, 4102, Australia
| | - Xiao-Qian Wu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China.
| | - Suo-Wen Xu
- Department of Endocrinology, First Affiliated Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China.
| | - Xu-Dong Jiang
- Department of Pharmacy, School of Medicine, Guangxi University of Science and Technology, Liuzhou, 545005, China.
| |
Collapse
|
12
|
Qian K, Tang J, Ling YJ, Zhou M, Yan XX, Xie Y, Zhu LJ, Nirmala K, Sun KY, Qin ZH, Sheng R. Exogenous NADPH exerts a positive inotropic effect and enhances energy metabolism via SIRT3 in pathological cardiac hypertrophy and heart failure. EBioMedicine 2023; 98:104863. [PMID: 37950995 PMCID: PMC10663691 DOI: 10.1016/j.ebiom.2023.104863] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/18/2023] [Accepted: 10/18/2023] [Indexed: 11/13/2023] Open
Abstract
BACKGROUND Therapies are urgently required to ameliorate pathological cardiac hypertrophy and enhance cardiac function in heart failure. Our preliminary experiments have demonstrated that exogenous NADPH exhibits a positive inotropic effect on isolated heart. This study aims to investigate the positive inotropic effects of NADPH in pathological cardiac hypertrophy and heart failure, as well as the underlying mechanisms involved. METHODS Endogenous plasma NADPH contents were determined in patients with chronic heart failure and control adults. The positive inotropic effects of NADPH were investigated in isolated toad heart or rat heart. The effects of NADPH were investigated in isoproterenol (ISO)-induced cardiac hypertrophy or transverse aortic constriction (TAC)-induced heart failure. The underlying mechanisms of NADPH were studied using SIRT3 knockout mice, echocardiography, Western blotting, transmission electron microscopy, and immunoprecipitation. FINDINGS The endogenous NADPH content in the blood of patients and animals with pathological cardiac hypertrophy or heart failure was significantly reduced compared with age-sex matched control subjects. Exogenous NADPH showed positive inotropic effects on the isolated normal and failing hearts, while antagonism of ATP receptor partially abolished the positive inotropic effect of NADPH. Exogenous NADPH administration significantly reduced heart weight indices, and improved cardiac function in the mice with pathological cardiac hypertrophy or heart failure. NADPH increased SIRT3 expression and activity, deacetylated target proteins, improved mitochondrial function and facilitated ATP production in the hypertrophic myocardium. Importantly, inhibition of SIRT3 abolished the positive inotropic effect of NADPH, and the anti-heart failure effect of NADPH was significantly reduced in the SIRT3 Knockout mice. INTERPRETATION Exogenous NADPH shows positive inotropic effect and improves energy metabolism via SIRT3 in pathological cardiac hypertrophy and heart failure. NADPH thus may be one of the potential candidates for the treatment of pathological cardiac hypertrophy or heart failure. FUNDING This work was supported by grants from the National Natural Science Foundation of China (No. 81973315, 82173811, 81730092), Natural Science Foundation of Jiangsu Higher Education (20KJA310008), Jiangsu Key Laboratory of Neuropsychiatric Diseases (BM2013003) and the Priority Academic Program Development of the Jiangsu Higher Education Institutes (PAPD).
Collapse
Affiliation(s)
- Ke Qian
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, Suzhou, 215123, China
| | - Jie Tang
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, Suzhou, 215123, China
| | - Yue-Juan Ling
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, Suzhou, 215123, China
| | - Ming Zhou
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, Suzhou, 215123, China
| | - Xin-Xin Yan
- Department of Cardiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, 215008, China
| | - Yu Xie
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, Suzhou, 215123, China
| | - Lu-Jia Zhu
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, Suzhou, 215123, China
| | - Koju Nirmala
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, Suzhou, 215123, China
| | - Kang-Yun Sun
- Department of Cardiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, 215008, China
| | - Zheng-Hong Qin
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, Suzhou, 215123, China
| | - Rui Sheng
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, Suzhou, 215123, China.
| |
Collapse
|
13
|
Wang R, Luo X, Li S, Wen X, Zhang X, Zhou Y, Xie W. A bibliometric analysis of cardiomyocyte apoptosis from 2014 to 2023: A review. Medicine (Baltimore) 2023; 102:e35958. [PMID: 38013295 PMCID: PMC10681623 DOI: 10.1097/md.0000000000035958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 10/13/2023] [Indexed: 11/29/2023] Open
Abstract
Cardiomyocyte apoptosis is an important factor in cardiac function decline observed in various cardiovascular diseases. To understand the progress in the field of cardiomyocyte apoptosis research, this paper uses bibliometrics to statistically analyze publications in this field. A total of 5939 articles were retrieved from the core Web of Science database, and then VOSviewer and Citespace were used to conduct a scientometric analysis of the authors, countries, institutions, references and keywords included in the articles to determine the cooperative relationships between researchers that study cardiomyocyte apoptosis. At present, the research hotspots in this field mainly include experimental research, molecular mechanisms, pathophysiology and cardiac regeneration of cardiomyocyte apoptosis-related diseases. NOD-like receptor thermal protein domain associated protein 3 inflammasome, circular RNA, and sepsis are the research frontiers in this field and are emerging as new areas of research focus. This work provides insight into research directions and the clinical application value for the continued advancement of cardiomyocyte apoptosis research.
Collapse
Affiliation(s)
- Rui Wang
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xu Luo
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Songyun Li
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xin Wen
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xin Zhang
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yunxiang Zhou
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wen Xie
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
14
|
Li S, Li L, Zhang C, Fu H, Yu S, Zhou M, Guo J, Fang Z, Li A, Zhao M, Zhang M, Wang X. PM2.5 leads to adverse pregnancy outcomes by inducing trophoblast oxidative stress and mitochondrial apoptosis via KLF9/CYP1A1 transcriptional axis. eLife 2023; 12:e85944. [PMID: 37737576 PMCID: PMC10584374 DOI: 10.7554/elife.85944] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 09/21/2023] [Indexed: 09/23/2023] Open
Abstract
Epidemiological studies have demonstrated that fine particulate matter (PM2.5) is associated with adverse obstetric and postnatal metabolic health outcomes, but the mechanism remains unclear. This study aimed to investigate the toxicological pathways by which PM2.5 damaged placental trophoblasts in vivo and in vitro. We confirmed that PM2.5 induced adverse gestational outcomes such as increased fetal mortality rates, decreased fetal numbers and weight, damaged placental structure, and increased apoptosis of trophoblasts. Additionally, PM2.5 induced dysfunction of the trophoblast cell line HTR8/SVneo, including in its proliferation, apoptosis, invasion, migration and angiogenesis. Moreover, we comprehensively analyzed the transcriptional landscape of HTR8/SVneo cells exposed to PM2.5 through RNA-Seq and observed that PM2.5 triggered overexpression of pathways involved in oxidative stress and mitochondrial apoptosis to damage HTR8/SVneo cell biological functions through CYP1A1. Mechanistically, PM2.5 stimulated KLF9, a transcription factor identified as binding to CYP1A1 promoter region, which further modulated the CYP1A1-driven downstream phenotypes. Together, this study demonstrated that the KLF9/CYP1A1 axis played a crucial role in the toxic progression of PM2.5 induced adverse pregnancy outcomes, suggesting adverse effects of environmental pollution on pregnant females and putative targeted therapeutic strategies.
Collapse
Affiliation(s)
- Shuxian Li
- Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Maternal and Child Health Care Hospital of Shandong Province Affiliated to Qingdao UniversityJinanChina
| | - Lingbing Li
- The Second Hospital, Cheeloo College of Medicine, Shandong UniversityJinanChina
| | - Changqing Zhang
- Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Maternal and Child Health Care Hospital of Shandong Province Affiliated to Qingdao UniversityJinanChina
| | - Huaxuan Fu
- Jinan Environmental Monitoring Center of Shandong ProvinceJinanChina
| | - Shuping Yu
- School of Public Health, Weifang Medical UniversityWeifangChina
| | - Meijuan Zhou
- Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Maternal and Child Health Care Hospital of Shandong Province Affiliated to Qingdao UniversityJinanChina
| | - Junjun Guo
- Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Maternal and Child Health Care Hospital of Shandong Province Affiliated to Qingdao UniversityJinanChina
| | - Zhenya Fang
- Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Maternal and Child Health Care Hospital of Shandong Province Affiliated to Qingdao UniversityJinanChina
| | - Anna Li
- Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Maternal and Child Health Care Hospital of Shandong Province Affiliated to Qingdao UniversityJinanChina
| | - Man Zhao
- Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Maternal and Child Health Care Hospital of Shandong Province Affiliated to Qingdao UniversityJinanChina
| | - Meihua Zhang
- Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Maternal and Child Health Care Hospital of Shandong Province Affiliated to Qingdao UniversityJinanChina
| | - Xietong Wang
- Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Maternal and Child Health Care Hospital of Shandong Province Affiliated to Qingdao UniversityJinanChina
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
| |
Collapse
|
15
|
Li Q, Zhang S, Yang G, Wang X, Liu F, Li Y, Chen Y, Zhou T, Xie D, Liu Y, Zhang L. Energy metabolism: A critical target of cardiovascular injury. Biomed Pharmacother 2023; 165:115271. [PMID: 37544284 DOI: 10.1016/j.biopha.2023.115271] [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: 06/06/2023] [Revised: 07/31/2023] [Accepted: 07/31/2023] [Indexed: 08/08/2023] Open
Abstract
Cardiovascular diseases are the main killers threatening human health. Many studies have shown that abnormal energy metabolism plays a key role in the occurrence and development of acute and chronic cardiovascular diseases. Regulating cardiac energy metabolism is a frontier topic in the treatment of cardiovascular diseases. However, we are not very clear about the choice of different substrates, the specific mechanism of energy metabolism participating in the course of cardiovascular disease, and how to develop appropriate drugs to regulate energy metabolism to treat cardiovascular disease. Therefore, this paper reviews how energy metabolism participates in cardiovascular pathophysiological processes and potential drugs aimed at interfering energy metabolism.It is expected to provide good suggestions for promoting the clinical prevention and treatment of cardiovascular diseases from the perspective of energy metabolism.
Collapse
Affiliation(s)
- Qiyang Li
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities, Gansu University of Chinese Medicine, Lanzhou, China
| | - Shangzu Zhang
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities, Gansu University of Chinese Medicine, Lanzhou, China
| | - Gengqiang Yang
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities, Gansu University of Chinese Medicine, Lanzhou, China
| | - Xin Wang
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities, Gansu University of Chinese Medicine, Lanzhou, China
| | - Fuxian Liu
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities, Gansu University of Chinese Medicine, Lanzhou, China
| | - Yangyang Li
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities, Gansu University of Chinese Medicine, Lanzhou, China
| | - Yan Chen
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities, Gansu University of Chinese Medicine, Lanzhou, China
| | - Ting Zhou
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities, Gansu University of Chinese Medicine, Lanzhou, China
| | - Dingxiong Xie
- Gansu Institute of Cardiovascular Diseases, LanZhou, China.
| | - Yongqi Liu
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities, Gansu University of Chinese Medicine, Lanzhou, China; Key Laboratory of Dunhuang Medicine and Transformation Ministry of Education, China.
| | - Liying Zhang
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities, Gansu University of Chinese Medicine, Lanzhou, China; Gansu Institute of Cardiovascular Diseases, LanZhou, China.
| |
Collapse
|
16
|
Ibrahim NA, Buabeid MA, Shaimaa Arafa E, Elmorshedy KE. Zinc's protective role against hydroxychloroquine-induced cardiac effects in adult male albino rats. Saudi J Biol Sci 2023; 30:103733. [PMID: 37521750 PMCID: PMC10374629 DOI: 10.1016/j.sjbs.2023.103733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 06/22/2023] [Accepted: 06/30/2023] [Indexed: 08/01/2023] Open
Abstract
Background Long exposure to Hydroxychloroquine (HCQ) has been complicated by some dangerous though infrequent cardiotoxicity. Methods A total of 40 normal adult male albino rats dispersed into 4 groups were used. Group 1 (Control group), Group II (HCQ treated group), Group III (zinc [Zn]-treated group), and Group IV (HCQ and Zn treated group). Once the experimentation ended, rats were sacrificed and cardiac soft tissue sections were processed twenty-four hours at the end of the experiment for histological study. Results Cardiac-stained sections revealed that HCQ induced widespread necrosis, dilatation, and vacuolar degeneration. However, the combination of HCQ with Zn ameliorated these damaging effects. Cardiac enzyme parameters were also studied in the 4 groups and revealed CK-MB and troponin were considerably elevated in groups II associated to the control group. Conclusion It was concluded that Zn revealed a protective role against HCQ cardiomyopathy in adult male albino rats. This might signify an appreciated means for Zn-based treatment in the upcoming subsequent clinical records to adjust doses and guarantee patient safeguard.
Collapse
Affiliation(s)
- Nihal A. Ibrahim
- Department of Clinical Sciences, College of Pharmacy and Health Sciences, Ajman University, UAE
- Centre of Medical and Bio-allied Health Sciences Research (CMBAHSR), Ajman University, Ajman, UAE
| | | | - El Shaimaa Arafa
- Department of Clinical Sciences, College of Pharmacy and Health Sciences, Ajman University, UAE
- Centre of Medical and Bio-allied Health Sciences Research (CMBAHSR), Ajman University, Ajman, UAE
| | | |
Collapse
|
17
|
Qi Y, Chen Z, Guo B, Liu Z, Wang L, Liu S, Xue L, Ma M, Yin Y, Li Y, Liu G. Speckle-tracking echocardiography provides sensitive measurements of subtle early alterations associated with cardiac dysfunction in T2DM rats. BMC Cardiovasc Disord 2023; 23:266. [PMID: 37217862 DOI: 10.1186/s12872-023-03239-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 04/12/2023] [Indexed: 05/24/2023] Open
Abstract
BACKGROUND Diabetic cardiomyopathy results in cardiac structural and functional abnormalities. Previous studies have demonstrated that inhibiting the RhoA/ROCK signalling pathway increases the injury resistance of cardiomyocytes. The early detection of cardiac structural and functional alterations may facilitate an improved understanding of the pathophysiologic progress and guide therapy. This study aimed to identify the optimal diagnostic measures for the subtle early alterations of cardiac dysfunction in type 2 diabetes mellitus (T2DM) rats. METHODS Twenty-four rat models were divided into four groups and received treatments for 4 weeks: the CON group (control rats), the DM group (T2DM rats), the DMF group (T2DM rats receiving fasudil) and the CONF group (control rats receiving fasudil) group. Left ventricular (LV) structure was quantified by histological staining and transmission electron microscopy. LV function and myocardial deformation were assessed by high-frequency echocardiography. RESULTS Treatment with fasudil, a ROCK inhibitor, significantly protected against diabetes-induced myocardial hypertrophy, fibrosis and mitochondrial dysfunction. Impaired LV performance was found in T2DM rats, as evidenced by significant reductions in the ejection fraction (EF), fractional shortening (FS) and the mitral valve (MV) E/A ratio (which decreased 26%, 34% and 20%, respectively). Fasudil failed to improve the conventional ultrasonic parameters in T2DM rats, but the myocardial deformation measured by speckle-tracking echocardiography (STE) were significantly improved (global circumferential strain, GCS: P = 0.003; GCS rate, GCSR: P = 0.021). When receiver operating characteristic (ROC) curves were used in combination with linear regression analysis, STE parameters were found to be characterized by both optimal prediction of cardiac damage [AUC (95% CI): fractional area change, FAC: 0.927 (0.744, 0.993); GCS: 0.819 (0.610, 0.945); GCSR: 0.899 (0.707, 0.984)] and stronger correlations with cardiac fibrosis (FAC: r = -0.825; GCS: r = 0.772; GCSR: r = 0.829) than conventional parameters. CONCLUSION The results suggest that STE parameters are more sensitive and specific than conventional parameters in predicting the subtle cardiac functional changes that occur in the early stage, providing new insight into the management of diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Yanchao Qi
- Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, 050031, Hebei, People's Republic of China
- Heart Center, The First Hospital of Hebei Medical University, Shijiazhuang, 050031, Hebei, People's Republic of China
| | - Zhiyan Chen
- Heart Center, The First Hospital of Hebei Medical University, Shijiazhuang, 050031, Hebei, People's Republic of China
| | - Bingyan Guo
- Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, People's Republic of China
| | - Zhe Liu
- Heart Center, The First Hospital of Hebei Medical University, Shijiazhuang, 050031, Hebei, People's Republic of China
| | - Lijie Wang
- Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, People's Republic of China
| | - Suyun Liu
- Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, People's Republic of China
| | - Lixiang Xue
- Center of Basic Medical Research, Peking University Third Hospital, Beijing, 100191, People's Republic of China
| | - Meifang Ma
- Department of Cardiology, Handan Central Hospital, Handan, 056008, Hebei, People's Republic of China
| | - Yajuan Yin
- Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, 050031, Hebei, People's Republic of China
- Hebei International Joint Research Center for Structural Heart Disease, Shijiazhuang, 050031, Hebei, People's Republic of China
- Hebei Key Laboratory of Cardiac Injury Repair Mechanism Study, Shijiazhuang, 050031, Hebei, People's Republic of China
- Hebei Key Laboratory of Heart and Metabolism, Shijiazhuang, 050031, People's Republic of China
| | - Yongjun Li
- Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, People's Republic of China.
| | - Gang Liu
- Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, 050031, Hebei, People's Republic of China.
- Heart Center, The First Hospital of Hebei Medical University, Shijiazhuang, 050031, Hebei, People's Republic of China.
- Hebei International Joint Research Center for Structural Heart Disease, Shijiazhuang, 050031, Hebei, People's Republic of China.
- Hebei Key Laboratory of Cardiac Injury Repair Mechanism Study, Shijiazhuang, 050031, Hebei, People's Republic of China.
- Hebei Key Laboratory of Heart and Metabolism, Shijiazhuang, 050031, People's Republic of China.
| |
Collapse
|
18
|
Ma Z, Liu Z, Li X, Zhang H, Han D, Xiong W, Zhou H, Yang X, Zeng Q, Ren H, Xu D. Metformin Collaborates with PINK1/Mfn2 Overexpression to Prevent Cardiac Injury by Improving Mitochondrial Function. BIOLOGY 2023; 12:biology12040582. [PMID: 37106782 PMCID: PMC10135998 DOI: 10.3390/biology12040582] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 04/05/2023] [Accepted: 04/07/2023] [Indexed: 04/29/2023]
Abstract
Both mitochondrial quality control and energy metabolism are critical in maintaining the physiological function of cardiomyocytes. When damaged mitochondria fail to be repaired, cardiomyocytes initiate a process referred to as mitophagy to clear defective mitochondria, and studies have shown that PTEN-induced putative kinase 1 (PINK1) plays an important role in this process. In addition, previous studies indicated that peroxisome proliferator-activated receptor gamma coactivator-1α (PGC-1α) is a transcriptional coactivator that promotes mitochondrial energy metabolism, and mitofusin 2 (Mfn2) promotes mitochondrial fusion, which is beneficial for cardiomyocytes. Thus, an integration strategy involving mitochondrial biogenesis and mitophagy might contribute to improved cardiomyocyte function. We studied the function of PINK1 in mitophagy in isoproterenol (Iso)-induced cardiomyocyte injury and transverse aortic constriction (TAC)-induced myocardial hypertrophy. Adenovirus vectors were used to induce PINK1/Mfn2 protein overexpression. Cardiomyocytes treated with isoproterenol (Iso) expressed high levels of PINK1 and low levels of Mfn2, and the changes were time dependent. PINK1 overexpression promoted mitophagy, attenuated the Iso-induced reduction in MMP, and reduced ROS production and the apoptotic rate. Cardiac-specific overexpression of PINK1 improved cardiac function, attenuated pressure overload-induced cardiac hypertrophy and fibrosis, and facilitated myocardial mitophagy in TAC mice. Moreover, metformin treatment and PINK1/Mfn2 overexpression reduced mitochondrial dysfunction by inhibiting ROS generation leading to an increase in both ATP production and mitochondrial membrane potential in Iso-induced cardiomyocyte injury. Our findings indicate that a combination strategy may help ameliorate myocardial injury by improving mitochondrial quality.
Collapse
Affiliation(s)
- Zhuang Ma
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou 510080, China
- Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou 510515, China
| | - Zuheng Liu
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou 510080, China
- Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou 510515, China
- Xiamen Key Laboratory of Cardiac Electrophysiology, Department of Cardiology, Xiamen Institute of Cardiovascular Diseases, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361013, China
| | - Xudong Li
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou 510080, China
- Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou 510515, China
| | - Hao Zhang
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou 510080, China
- Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou 510515, China
| | - Dunzheng Han
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou 510080, China
- Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou 510515, China
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Wenjun Xiong
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou 510080, China
- Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou 510515, China
| | - Haobin Zhou
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou 510080, China
- Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou 510515, China
| | - Xi Yang
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou 510080, China
- Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou 510515, China
| | - Qingchun Zeng
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou 510080, China
- Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou 510515, China
| | - Hao Ren
- Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou 510515, China
- Department of Rheumatology, Nanfang Hospital, Southern Medical University, Guangzhou 516006, China
| | - Dingli Xu
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou 510080, China
- Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou 510515, China
| |
Collapse
|
19
|
Liu IF, Lin TC, Wang SC, Yen CH, Li CY, Kuo HF, Hsieh CC, Chang CY, Chang CR, Chen YH, Liu YR, Lee TY, Huang CY, Hsu CH, Lin SJ, Liu PL. Long-term administration of Western diet induced metabolic syndrome in mice and causes cardiac microvascular dysfunction, cardiomyocyte mitochondrial damage, and cardiac remodeling involving caveolae and caveolin-1 expression. Biol Direct 2023; 18:9. [PMID: 36879344 PMCID: PMC9987103 DOI: 10.1186/s13062-023-00363-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 02/21/2023] [Indexed: 03/08/2023] Open
Abstract
BACKGROUND Long-term consumption of an excessive fat and sucrose diet (Western diet, WD) has been considered a risk factor for metabolic syndrome (MS) and cardiovascular disease. Caveolae and caveolin-1 (CAV-1) proteins are involved in lipid transport and metabolism. However, studies investigating CAV-1 expression, cardiac remodeling, and dysfunction caused by MS, are limited. This study aimed to investigate the correlation between the expression of CAV-1 and abnormal lipid accumulation in the endothelium and myocardium in WD-induced MS, and the occurrence of myocardial microvascular endothelial cell dysfunction, myocardial mitochondrial remodeling, and damage effects on cardiac remodeling and cardiac function. METHODS We employed a long-term (7 months) WD feeding mouse model to measure the effect of MS on caveolae/vesiculo-vacuolar organelle (VVO) formation, lipid deposition, and endothelial cell dysfunction in cardiac microvascular using a transmission electron microscopy (TEM) assay. CAV-1 and endothelial nitric oxide synthase (eNOS) expression and interaction were evaluated using real-time polymerase chain reaction, Western blot, and immunostaining. Cardiac mitochondrial shape transition and damage, mitochondria-associated endoplasmic reticulum membrane (MAM) disruption, cardiac function change, caspase-mediated apoptosis pathway activation, and cardiac remodeling were examined using TEM, echocardiography, immunohistochemistry, and Western blot assay. RESULTS Our study demonstrated that long-term WD feeding caused obesity and MS in mice. In mice, MS increased caveolae and VVO formation in the microvascular system and enhanced CAV-1 and lipid droplet binding affinity. In addition, MS caused a significant decrease in eNOS expression, vascular endothelial cadherin, and β-catenin interactions in cardiac microvascular endothelial cells, accompanied by impaired vascular integrity. MS-induced endothelial dysfunction caused massive lipid accumulation in the cardiomyocytes, leading to MAM disruption, mitochondrial shape transition, and damage. MS promoted brain natriuretic peptide expression and activated the caspase-dependent apoptosis pathway, leading to cardiac dysfunction in mice. CONCLUSION MS resulted in cardiac dysfunction, remodeling by regulating caveolae and CAV-1 expression, and endothelial dysfunction. Lipid accumulation and lipotoxicity caused MAM disruption and mitochondrial remodeling in cardiomyocytes, leading to cardiomyocyte apoptosis and cardiac dysfunction and remodeling.
Collapse
Affiliation(s)
- I-Fan Liu
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, 112304, Taiwan.,Heart Center, Cheng Hsin General Hospital, Taipei, 112401, Taiwan
| | - Tzu-Chieh Lin
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 807378, Taiwan.,Division of Cardiology, Department of Internal Medicine, Kaohsiung Medical University, Kaohsiung, 807378, Taiwan
| | - Shu-Chi Wang
- Department of Medical Laboratory Science and Biotechnology, Kaohsiung Medical University, Kaohsiung, 807378, Taiwan
| | - Chia-Hung Yen
- Graduate Institute of Natural Products, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, 807378, Taiwan
| | - Chia-Yang Li
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 807378, Taiwan
| | - Hsuan-Fu Kuo
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Medical University, Kaohsiung, 807378, Taiwan.,Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 807378, Taiwan.,Department of Internal Medicine, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, 807378, Taiwan.,Department of Internal Medicine, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 807378, Taiwan
| | - Chong-Chao Hsieh
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 807378, Taiwan.,Division of Cardiovascular Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, 807378, Taiwan.,Department of Surgery, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 807378, Taiwan
| | - Chia-Yuan Chang
- Department of Mechanical Engineering, National Cheng Kung University, Tainan, 701401, Taiwan
| | - Chuang-Rung Chang
- Department of Medical Science, National Tsing Hua University, Hsinchu, 300044, Taiwan.,Institute of Biotechnology, National Tsing Hua University, Hsinchu, 300044, Taiwan
| | - Yung-Hsiang Chen
- Graduate Institute of Integrated Medicine, College of Chinese Medicine, China Medical University, Taichung, 404333, Taiwan.,Department of Psychology, College of Medical and Health Science, Asia University, Taichung, 413305, Taiwan
| | - Yu-Ru Liu
- Department of Respiratory Therapy, College of Medicine, Kaohsiung Medical University, Kaohsiung, 807378, Taiwan
| | - Tsung-Ying Lee
- Department of Respiratory Therapy, College of Medicine, Kaohsiung Medical University, Kaohsiung, 807378, Taiwan
| | - Chi-Yuan Huang
- Department of Respiratory Therapy, College of Medicine, Kaohsiung Medical University, Kaohsiung, 807378, Taiwan
| | - Chih-Hsin Hsu
- Department of Internal Medicine, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan, 701401, Taiwan.
| | - Shing-Jong Lin
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, 112304, Taiwan. .,Cardiovascular Research Center, National Yang Ming Chiao Tung University, Taipei, 112304, Taiwan. .,Department of Medical Research, Taipei Veterans General Hospital, Taipei, 112201, Taiwan. .,Taipei Heart Institute, Taipei Medical University, Taipei, 110301, Taiwan. .,Heart Center, Cheng-Hsin General Hospital, Taipei, 112401, Taiwan.
| | - Po-Len Liu
- Department of Respiratory Therapy, College of Medicine, Kaohsiung Medical University, Kaohsiung, 807378, Taiwan. .,Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, 807378, Taiwan. .,Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung, 807378, Taiwan.
| |
Collapse
|
20
|
Ding X, Hou Y, Liu X, Li X, Liu X, Deng Y, Cao N, Yu W. The role of Sirt3-induced autophagy in renal structural damage caused by periodontitis in rats. J Periodontal Res 2023; 58:97-108. [PMID: 36380567 DOI: 10.1111/jre.13071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 10/14/2022] [Accepted: 10/26/2022] [Indexed: 11/18/2022]
Abstract
OBJECTIVE This research aimed to explore the effect of periodontitis on renal tissues injury in rats and the role of Sirtuin3 (Sirt3) and its regulation of autophagy in this progression. MATERIAL AND METHODS Thirty Wistar rats were assigned into three groups: control, periodontitis (P), and periodontitis with gavage administration of Sirt3 activator resveratrol (P + RSV). To induce periodontitis, the wire ligature was placed around the cervical region of the rat maxillary first molar. After 8 weeks, micro-computed tomography (Micro-CT) and hematoxylin and eosin (HE) were used to evaluate the alveolar bone resorption and periodontal inflammation. Serum and urine biochemical indicators were measured to assess renal function. The pathological changes of the kidney were observed via HE and periodic acid Schiff (PAS) staining. Autophagosome was viewed by transmission electron microscopy (TEM). Real-time PCR and western blot were used to test expressions of Sirt3 and autophagy indicators in renal and periodontal tissues, including mammalian target of rapamycin (mTOR), phosphor-mTOR (p-mTOR), BECN1 (Beclin-1), and microtubule-associated protein 1 light chain 3 (LC3). RESULTS Alveolar bone destruction, resorption, and periodontal inflammation were observed in the P group (compared with the control group), and the above indexes were significantly improved after RSV intervention; the obvious changes in renal tissue structure in the P group were partially recovered after RSV intervention, while renal functional status was not affected (among the three groups); in addition, the levels of Sirt3 and autophagy in kidney and periodontal tissues of P group were inhibited, manifested as a decrease in the number of autophagosomes (renal tissue) and expressions of autophagy marker Beclin-1 and LC3 conversion rate and an increase in the expression of p-mTOR. After Sirt3 activation (RSV), the above indicators were significantly improved. CONCLUSION Periodontitis causes renal structural damage in rats, which may be connected to the effect of Sirt3-induced autophagy.
Collapse
Affiliation(s)
- Xu Ding
- Department of Periodontology, Hospital of Stomatology, Jilin University, Changchun, China
| | - Yubo Hou
- Department of Periodontology, Hospital of Stomatology, Jilin University, Changchun, China
| | - Xinchan Liu
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, China
| | - Xin Li
- Department of Periodontology, Hospital of Stomatology, Jilin University, Changchun, China
| | - Xiaomeng Liu
- Department of Periodontology, Hospital of Stomatology, Jilin University, Changchun, China
| | - Yu Deng
- Department of Periodontology, Hospital of Stomatology, Jilin University, Changchun, China
| | - Niuben Cao
- Department of Periodontology, Hospital of Stomatology, Jilin University, Changchun, China
| | - Weixian Yu
- Department of Periodontology, Hospital of Stomatology, Jilin University, Changchun, China.,Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, China
| |
Collapse
|
21
|
Effect of Thyroxine on the Structural and Dynamic Features of Cardiac Mitochondria and Mitophagy in Rats. Cells 2023; 12:cells12030396. [PMID: 36766738 PMCID: PMC9913912 DOI: 10.3390/cells12030396] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 01/19/2023] [Accepted: 01/19/2023] [Indexed: 01/24/2023] Open
Abstract
This work investigated the effect of thyroxine on the biogenesis and quality control system in rat heart mitochondria. In hyperthyroid rats, the concentrations of free triiodothyronine and thyroxine increased severalfold, indicating the development of hyperthyroidism in these animals. The electron microscopy showed 58% of cardiac mitochondria to be in a swollen state. Some organelles were damaged and had a reduced number of cristae. Multilamellar bodies formed from cristae/membranes were found in the vacuolated part of the mitochondria. The hyperthyroidism caused no changes to mitochondrial biogenesis in the investigated animals. At the same time, the levels of mitochondrial dynamics proteins OPA1 and Drp1 increased in the hyperthyroid rats. The administration of thyroxine to the animals led to a decrease in the amount of PINK1 and Parkin in heart tissue. The data suggest that excess thyroid hormones lead to changes in mitochondrial dynamics and impair Parkin-dependent mitophagy in hyperthyroid rat heart.
Collapse
|
22
|
Guan Z, Chen J, Wang L, Hao M, Dong X, Luo T, Jiang J, Lin Z, Li X, Chen P, Yang Z, Ye X, Wang L, Xian S, Chen Z. Nuanxinkang prevents the development of myocardial infarction-induced chronic heart failure by promoting PINK1/Parkin-mediated mitophagy. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 108:154494. [PMID: 36279758 DOI: 10.1016/j.phymed.2022.154494] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 09/27/2022] [Accepted: 10/05/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND Mitochondrial dysfunction is an important pathological feature of chronic heart failure (CHF). Regulation of mitophagy can effectively maintain mitochondrial homeostasis and energy metabolism, thereby inhibiting the development of CHF. Nuanxinkang (NXK), a Chinese herbal compound preparation, has significant cardioprotective effects on CHF; however, its underlying mechanism on mitophagy has not been completely clarified. This research intended to investigate the mechanism of NXK in treating myocardial infarction (MI)-induced CHF. METHODS The left anterior descending coronary artery (LAD) ligation surgery was performed to establish an MI-induced CHF model in male C57BL/6 mice. From 1 day after surgery, mice were given NXK (0.41, 0.82 or 1.65 g/kg/d), Perindopril (PDPL, 0.607 mg/kg/d), or an equivalent amount of sterile water by gavage for 28 continuous days. Then, mice were examined for cardiac function, myocardial fibrosis, cardiomyocyte apoptosis, mitochondrial structure and mitophagy levels of cardiomyocytes, etc. In addition, a hypoxic injury model was created using HL-1 cardiomyocytes from wild-type (WT) mice. HL-1 cells were pretreated with or without NXK-containing serum. Mitochondrial function and mitophagy levels were examined in HL-1 cells. RESULTS In MI-induced CHF mice, cardiac dysfunction, severe cardiac remodeling, elevated levels of oxidative stress, reduced ATP levels, and inhibition of PINK1/Parkin-mediated mitophagy were observed. High-dose NXK treatment (1.65 g/kg/d) significantly improved myocardial energy metabolism, inhibited cardiac remodeling, improved cardiac function, and restored cardiac PINK1/Parkin-mediated mitophagy levels to some extent in MI mice. In vitro, elevated levels of mitochondrial reactive oxygen species (ROS) with impaired mitochondrial membrane potential (ΔΨm) were observed in hypoxic HL-1 cells. While NXK treatment significantly protected cardiomyocytes from hypoxia-induced mitochondrial dysfunction, which is consistent with the in vivo results. Further studies showed that NXK could increase PINK1/Parkin-mediated mitophagy levels in cardiomyocytes, which could be blocked by the mitophagy inhibitor Mdivi-1. CONCLUSION In conclusion, NXK could prevent cardiac mitochondrial dysfunction and improve cardiac function against MI-induced CHF by promoting Pink1/Parkin-mediated mitophagy, which represents a very prospective strategy for the treatment of CHF.
Collapse
Affiliation(s)
- Zhuoji Guan
- Dongguan Hospital, Guangzhou University of Chinese Medicine, Dongguan 523005, China; Guangzhou University of Chinese Medicine, Guangzhou 510405, China; The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Guangzhou Key Laboratory of Chinese Medicine for Prevention and Treatment of Chronic Heart Failure, Guangzhou 510405, China
| | - Jie Chen
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China; The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Guangzhou Key Laboratory of Chinese Medicine for Prevention and Treatment of Chronic Heart Failure, Guangzhou 510405, China
| | - Linhai Wang
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China; The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Guangzhou Key Laboratory of Chinese Medicine for Prevention and Treatment of Chronic Heart Failure, Guangzhou 510405, China
| | - Mengjiao Hao
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Sun Yat-sen University, Guangzhou 510006, China
| | - Xin Dong
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China; The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Guangzhou Key Laboratory of Chinese Medicine for Prevention and Treatment of Chronic Heart Failure, Guangzhou 510405, China
| | - Tong Luo
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China; The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Guangzhou Key Laboratory of Chinese Medicine for Prevention and Treatment of Chronic Heart Failure, Guangzhou 510405, China
| | - Jialin Jiang
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China; The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Guangzhou Key Laboratory of Chinese Medicine for Prevention and Treatment of Chronic Heart Failure, Guangzhou 510405, China
| | - Zhijun Lin
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China; The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Guangzhou Key Laboratory of Chinese Medicine for Prevention and Treatment of Chronic Heart Failure, Guangzhou 510405, China
| | - Xuan Li
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China; The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Guangzhou Key Laboratory of Chinese Medicine for Prevention and Treatment of Chronic Heart Failure, Guangzhou 510405, China
| | - Pinliang Chen
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China; The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Guangzhou Key Laboratory of Chinese Medicine for Prevention and Treatment of Chronic Heart Failure, Guangzhou 510405, China
| | - Zhongqi Yang
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China; The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Guangzhou Key Laboratory of Chinese Medicine for Prevention and Treatment of Chronic Heart Failure, Guangzhou 510405, China
| | - Xiaohan Ye
- Dongguan Hospital, Guangzhou University of Chinese Medicine, Dongguan 523005, China; Guangzhou University of Chinese Medicine, Guangzhou 510405, China; The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Guangzhou Key Laboratory of Chinese Medicine for Prevention and Treatment of Chronic Heart Failure, Guangzhou 510405, China.
| | - Lingjun Wang
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China; The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Guangzhou Key Laboratory of Chinese Medicine for Prevention and Treatment of Chronic Heart Failure, Guangzhou 510405, China.
| | - Shaoxiang Xian
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China; The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Guangzhou Key Laboratory of Chinese Medicine for Prevention and Treatment of Chronic Heart Failure, Guangzhou 510405, China.
| | - Zixin Chen
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China; The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Guangzhou Key Laboratory of Chinese Medicine for Prevention and Treatment of Chronic Heart Failure, Guangzhou 510405, China.
| |
Collapse
|
23
|
Facts and ideas on statins with respect to their lipophilicity: a focus on skeletal muscle cells and bone besides known cardioprotection. Mol Cell Biochem 2022:10.1007/s11010-022-04621-y. [PMID: 36471123 PMCID: PMC9734727 DOI: 10.1007/s11010-022-04621-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 11/28/2022] [Indexed: 12/12/2022]
Abstract
Statins are known to block cholesterol synthesis in the liver. They also exhibit non-lipid pleiotropic effects due to the inhibition of protein prenylation, thereby modulating various signaling pathways of cellular homeostasis and integrity. Both lipid control and pleiotropic action of statins are clinically used, mainly for treatment of hypercholesterolemia and primary and secondary prevention of cardiovascular diseases. Because the prescription of statins is increasing and statin therapy is often lifelong, in particular in patients with other risk factors, safety issues being associated with polymorbidity and polypragmasia as well as the persistence with and adherence to statins are specific points of attention of clinicians and clinical pharmacologists. Furthermore, because skeletal myocytes have a cholesterol inhibitory sensitivity greater than hepatocytes, a choice of an appropriate statin based on its lipophilicity and the associated likelihood of its side effects on skeletal muscle cells and bone is warranted in such polymorbid patients. These approaches can effectively modulate the risk: benefit ratio and highlight a need for personalized therapy as much as possible, thereby minimizing risk of discontinuation of therapy and poor compliance.
Collapse
|
24
|
Kobara M, Toba H, Nakata T. Roles of autophagy in angiotensin II-induced cardiomyocyte apoptosis. Clin Exp Pharmacol Physiol 2022; 49:1342-1351. [PMID: 36059129 DOI: 10.1111/1440-1681.13719] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 08/25/2022] [Accepted: 09/01/2022] [Indexed: 01/31/2023]
Abstract
Autophagy is a self-degradation process of cytoplasmic components and occurs in the failing heart. Angiotensin II plays a critical role in the progression of heart failure and induces autophagy. We investigated the mechanism underlying angiotensin II-enhanced autophagy and examined the role of autophagy in angiotensin II-induced cardiomyocyte injury. Neonatal rat cardiomyocytes were treated with angiotensin II (1-100 nmol/L). Angiotensin II dose-dependently increased autophagy indicators of microtubule-associated protein 1 light chain (LC) 3-II and monodansylcadaverine-labelled vesicles. It also enhanced the intracellular production of reactive oxygen species (ROS), assessed by H2DCFDA, an intracellular ROS indicator. NADPH oxidase- and mitochondria-derived ROS production was increased by angiotensin II, while angiotensin II-induced LC3-II expression was suppressed by inhibitors of these sources of ROS. Confocal microscopy revealed that superoxide-producing mitochondria colocalized with lysosomes after the angiotensin II stimulation. Myocyte apoptosis was assessed by nuclear staining with DAPI and caspase-3 activity. A 6-h stimulation with angiotensin II did not affect myocyte apoptosis, while a co-treatment with 3-methyl-adenine (3MA), an autophagy inhibitor, augmented apoptosis. These results indicate that autophagy suppressed apoptosis because it removed damaged mitochondria in the early stages of the angiotensin II stimulation. A longer angiotensin II stimulation for 24 h induced apoptosis and propidium iodide-positive lethal myocytes, while the co-treatment with 3MA did not lead to further increases. In conclusion, angiotensin II-induced autophagy removes ROS-producing mitochondria. Autophagy is a beneficial phenomenon against myocyte apoptosis in the early phase, but its benefit was limited in the late phase of angiotensin II stimulation.
Collapse
Affiliation(s)
- Miyuki Kobara
- Department of Clinical Pharmacology, Division of Pathological Science, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Hiroe Toba
- Department of Clinical Pharmacology, Division of Pathological Science, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Tetsuo Nakata
- Department of Clinical Pharmacology, Division of Pathological Science, Kyoto Pharmaceutical University, Kyoto, Japan
| |
Collapse
|
25
|
Wang Y, Xu Y, Guo W, Fang Y, Hu L, Wang R, Zhao R, Guo D, Qi B, Ren G, Ren J, Li Y, Zhang M. Ablation of Shank3 alleviates cardiac dysfunction in aging mice by promoting CaMKII activation and Parkin-mediated mitophagy. Redox Biol 2022; 58:102537. [PMID: 36436456 PMCID: PMC9709154 DOI: 10.1016/j.redox.2022.102537] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 11/08/2022] [Indexed: 11/19/2022] Open
Abstract
Compromised mitophagy and mitochondrial homeostasis are major contributors for the etiology of cardiac aging, although the precise underlying mechanisms remains elusive. Shank3, a heart-enriched protein, has recently been reported to regulate aging-related neurodegenerative diseases. This study aimed to examine the role of Shank3 in the pathogenesis of cardiac senescence and the possible mechanisms involved. Cardiac-specific conditional Shank3 knockout (Shank3CKO) mice were subjected to natural aging. Mitochondrial function and mitophagy activity were determined in vivo, in mouse hearts and in vitro, in cardiomyocytes. Here, we showed that cardiac Shank3 expression exhibited a gradual increase during the natural progression of the aging, accompanied by overtly decreased mitophagy activity and a decline in cardiac function. Ablation of Shank3 promoted mitophagy, reduced mitochondria-derived superoxide (H2O2 and O2•-) production and apoptosis, and protected against cardiac dysfunction in the aged heart. In an in vitro study, senescent cardiomyocytes treated with D-gal exhibited reduced mitophagy and significantly elevated Shank3 expression. Shank3 knock-down restored mitophagy, leading to increased mitochondrial membrane potential, decreased mitochondrial oxidative stress, and reduced apoptosis in senescent cardiomyocytes, whereas Shank3 overexpression mimicked D-gal-induced mitophagy inhibition and mitochondrial dysfunction in normally cultured cardiomyocytes. Mechanistically, the IP assay revealed that Shank3 directly binds to CaMKII, and this interaction was further increased in the aged heart. Enhanced Shank3/CaMKII binding impedes mitochondrial translocation of CaMKII, resulting in the inhibition of parkin-mediated mitophagy, which ultimately leads to mitochondrial dysfunction and cardiac damage in the aged heart. Our study identified Shank3 as a novel contributor to aging-related cardiac damage. Manipulating Shank3/CaMKII-induced mitophagy inhibition could thus be an optional strategy for therapeutic intervention in clinical aging-related cardiac dysfunctions.
Collapse
Affiliation(s)
- Ying Wang
- Department of Cardiology, Tangdu Hospital, Air Force Medical University, Xi’an, 710038, China
| | - Yuerong Xu
- Department of Orthodontics, School of Stomatology, Air Force Medical University, Xi’an, 710032, China
| | - Wangang Guo
- Department of Cardiology, Tangdu Hospital, Air Force Medical University, Xi’an, 710038, China
| | - Yexian Fang
- Department of Cardiology, Tangdu Hospital, Air Force Medical University, Xi’an, 710038, China
| | - Lang Hu
- Department of Cardiology, Tangdu Hospital, Air Force Medical University, Xi’an, 710038, China
| | - Runze Wang
- Department of Cardiology, Tangdu Hospital, Air Force Medical University, Xi’an, 710038, China
| | - Ran Zhao
- Department of Cardiology, Tangdu Hospital, Air Force Medical University, Xi’an, 710038, China
| | - Dong Guo
- Department of Cardiology, Tangdu Hospital, Air Force Medical University, Xi’an, 710038, China
| | - Bingchao Qi
- Department of Cardiology, Tangdu Hospital, Air Force Medical University, Xi’an, 710038, China
| | - Gaotong Ren
- Department of Cardiology, Tangdu Hospital, Air Force Medical University, Xi’an, 710038, China
| | - Jun Ren
- Department of Cardiology and Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China,Corresponding author. Department of Cardiology and Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Yan Li
- Department of Cardiology, Tangdu Hospital, Air Force Medical University, Xi’an, 710038, China,Corresponding author.
| | - Mingming Zhang
- Department of Cardiology, Tangdu Hospital, Air Force Medical University, Xi’an, 710038, China,Corresponding author.
| |
Collapse
|
26
|
Jin L, Geng L, Ying L, Shu L, Ye K, Yang R, Liu Y, Wang Y, Cai Y, Jiang X, Wang Q, Yan X, Liao B, Liu J, Duan F, Sweeney G, Woo CWH, Wang Y, Xia Z, Lian Q, Xu A. FGF21-Sirtuin 3 Axis Confers the Protective Effects of Exercise Against Diabetic Cardiomyopathy by Governing Mitochondrial Integrity. Circulation 2022; 146:1537-1557. [PMID: 36134579 DOI: 10.1161/circulationaha.122.059631] [Citation(s) in RCA: 94] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
BACKGROUND Exercise is an effective nonpharmacological strategy to alleviate diabetic cardiomyopathy (DCM) through poorly defined mechanisms. FGF21 (fibroblast growth factor 21), a peptide hormone with pleiotropic benefits on cardiometabolic homeostasis, has been identified as an exercise responsive factor. This study aims to investigate whether FGF21 signaling mediates the benefits of exercise on DCM, and if so, to elucidate the underlying mechanisms. METHODS The global or hepatocyte-specific FGF21 knockout mice, cardiomyocyte-selective β-klotho (the obligatory co-receptor for FGF21) knockout mice, and their wild-type littermates were subjected to high-fat diet feeding and injection of streptozotocin to induce DCM, followed by a 6-week exercise intervention and assessment of cardiac functions. Cardiac mitochondrial structure and function were assessed by electron microscopy, enzymatic assays, and measurements of fatty acid oxidation and ATP production. Human induced pluripotent stem cell-derived cardiomyocytes were used to investigate the receptor and postreceptor signaling pathways conferring the protective effects of FGF21 against toxic lipids-induced mitochondrial dysfunction. RESULTS Treadmill exercise markedly induced cardiac expression of β-klotho and significantly attenuated diabetes-induced cardiac dysfunction in wild-type mice, accompanied by reduced mitochondrial damage and increased activities of mitochondrial enzymes in hearts. However, such cardioprotective benefits of exercise were largely abrogated in mice with global or hepatocyte-selective ablation of FGF21, or cardiomyocyte-specific deletion of β-klotho. Mechanistically, exercise enhanced the cardiac actions of FGF21 to induce the expression of the mitochondrial deacetylase SIRT3 by AMPK-evoked phosphorylation of FOXO3, thereby reversing diabetes-induced hyperacetylation and functional impairments of a cluster of mitochondrial enzymes. FGF21 prevented toxic lipids-induced mitochondrial dysfunction and oxidative stress by induction of the AMPK/FOXO3/SIRT3 signaling axis in human induced pluripotent stem cell-derived cardiomyocytes. Adeno-associated virus-mediated restoration of cardiac SIRT3 expression was sufficient to restore the responsiveness of diabetic FGF21 knockout mice to exercise in amelioration of mitochondrial dysfunction and DCM. CONCLUSIONS The FGF21-SIRT3 axis mediates the protective effects of exercise against DCM by preserving mitochondrial integrity and represents a potential therapeutic target for DCM. REGISTRATION URL: https://www. CLINICALTRIALS gov; Unique identifier: NCT03240978.
Collapse
Affiliation(s)
- Leigang Jin
- State Key Laboratory of Pharmaceutical Biotechnology (L.J., L.G., L.Y., L.S., R.Y., Y.L., Yao Wang, Y.C., X.J., Q.W., X.Y., B.L., C.W.H.W., Yu Wang, Z.X., Q.L., A.X.), University of Hong Kong, China.,Department of Medicine (L.J., L.G., L.S., R.Y., Y.L., Yao Wang, X.J., Q.W., X.Y., J.L., Z.X., Q.L., A.X.), University of Hong Kong, China.,Department of Pharmacology and Pharmacy (L.J., L.Y., B.L., C.W.H.W., Yu Wang, A.X.), University of Hong Kong, China
| | - Leiluo Geng
- State Key Laboratory of Pharmaceutical Biotechnology (L.J., L.G., L.Y., L.S., R.Y., Y.L., Yao Wang, Y.C., X.J., Q.W., X.Y., B.L., C.W.H.W., Yu Wang, Z.X., Q.L., A.X.), University of Hong Kong, China.,Department of Medicine (L.J., L.G., L.S., R.Y., Y.L., Yao Wang, X.J., Q.W., X.Y., J.L., Z.X., Q.L., A.X.), University of Hong Kong, China
| | - Lei Ying
- State Key Laboratory of Pharmaceutical Biotechnology (L.J., L.G., L.Y., L.S., R.Y., Y.L., Yao Wang, Y.C., X.J., Q.W., X.Y., B.L., C.W.H.W., Yu Wang, Z.X., Q.L., A.X.), University of Hong Kong, China.,Department of Pharmacology and Pharmacy (L.J., L.Y., B.L., C.W.H.W., Yu Wang, A.X.), University of Hong Kong, China
| | - Lingling Shu
- State Key Laboratory of Pharmaceutical Biotechnology (L.J., L.G., L.Y., L.S., R.Y., Y.L., Yao Wang, Y.C., X.J., Q.W., X.Y., B.L., C.W.H.W., Yu Wang, Z.X., Q.L., A.X.), University of Hong Kong, China.,Department of Medicine (L.J., L.G., L.S., R.Y., Y.L., Yao Wang, X.J., Q.W., X.Y., J.L., Z.X., Q.L., A.X.), University of Hong Kong, China
| | - Kevin Ye
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, Canada (K.Y.)
| | - Ranyao Yang
- State Key Laboratory of Pharmaceutical Biotechnology (L.J., L.G., L.Y., L.S., R.Y., Y.L., Yao Wang, Y.C., X.J., Q.W., X.Y., B.L., C.W.H.W., Yu Wang, Z.X., Q.L., A.X.), University of Hong Kong, China.,Department of Medicine (L.J., L.G., L.S., R.Y., Y.L., Yao Wang, X.J., Q.W., X.Y., J.L., Z.X., Q.L., A.X.), University of Hong Kong, China
| | - Yan Liu
- State Key Laboratory of Pharmaceutical Biotechnology (L.J., L.G., L.Y., L.S., R.Y., Y.L., Yao Wang, Y.C., X.J., Q.W., X.Y., B.L., C.W.H.W., Yu Wang, Z.X., Q.L., A.X.), University of Hong Kong, China.,Department of Medicine (L.J., L.G., L.S., R.Y., Y.L., Yao Wang, X.J., Q.W., X.Y., J.L., Z.X., Q.L., A.X.), University of Hong Kong, China
| | - Yao Wang
- State Key Laboratory of Pharmaceutical Biotechnology (L.J., L.G., L.Y., L.S., R.Y., Y.L., Yao Wang, Y.C., X.J., Q.W., X.Y., B.L., C.W.H.W., Yu Wang, Z.X., Q.L., A.X.), University of Hong Kong, China.,Department of Medicine (L.J., L.G., L.S., R.Y., Y.L., Yao Wang, X.J., Q.W., X.Y., J.L., Z.X., Q.L., A.X.), University of Hong Kong, China
| | - Yin Cai
- State Key Laboratory of Pharmaceutical Biotechnology (L.J., L.G., L.Y., L.S., R.Y., Y.L., Yao Wang, Y.C., X.J., Q.W., X.Y., B.L., C.W.H.W., Yu Wang, Z.X., Q.L., A.X.), University of Hong Kong, China.,Department of Health Technology and Informatics, Hong Kong Polytechnic University, China (Y.C.)
| | - Xue Jiang
- State Key Laboratory of Pharmaceutical Biotechnology (L.J., L.G., L.Y., L.S., R.Y., Y.L., Yao Wang, Y.C., X.J., Q.W., X.Y., B.L., C.W.H.W., Yu Wang, Z.X., Q.L., A.X.), University of Hong Kong, China.,Department of Medicine (L.J., L.G., L.S., R.Y., Y.L., Yao Wang, X.J., Q.W., X.Y., J.L., Z.X., Q.L., A.X.), University of Hong Kong, China
| | - Qin Wang
- State Key Laboratory of Pharmaceutical Biotechnology (L.J., L.G., L.Y., L.S., R.Y., Y.L., Yao Wang, Y.C., X.J., Q.W., X.Y., B.L., C.W.H.W., Yu Wang, Z.X., Q.L., A.X.), University of Hong Kong, China.,Department of Medicine (L.J., L.G., L.S., R.Y., Y.L., Yao Wang, X.J., Q.W., X.Y., J.L., Z.X., Q.L., A.X.), University of Hong Kong, China
| | - Xingqun Yan
- State Key Laboratory of Pharmaceutical Biotechnology (L.J., L.G., L.Y., L.S., R.Y., Y.L., Yao Wang, Y.C., X.J., Q.W., X.Y., B.L., C.W.H.W., Yu Wang, Z.X., Q.L., A.X.), University of Hong Kong, China.,Department of Medicine (L.J., L.G., L.S., R.Y., Y.L., Yao Wang, X.J., Q.W., X.Y., J.L., Z.X., Q.L., A.X.), University of Hong Kong, China
| | - Boya Liao
- State Key Laboratory of Pharmaceutical Biotechnology (L.J., L.G., L.Y., L.S., R.Y., Y.L., Yao Wang, Y.C., X.J., Q.W., X.Y., B.L., C.W.H.W., Yu Wang, Z.X., Q.L., A.X.), University of Hong Kong, China.,Department of Pharmacology and Pharmacy (L.J., L.Y., B.L., C.W.H.W., Yu Wang, A.X.), University of Hong Kong, China
| | - Jie Liu
- Department of Medicine (L.J., L.G., L.S., R.Y., Y.L., Yao Wang, X.J., Q.W., X.Y., J.L., Z.X., Q.L., A.X.), University of Hong Kong, China.,Cord Blood Bank, Guangzhou Institute of Eugenics and Perinatology, Women and Children's Medical Center, Guangzhou Medical University, China (J.L., F.D., Q.L.)
| | - Fuyu Duan
- Cord Blood Bank, Guangzhou Institute of Eugenics and Perinatology, Women and Children's Medical Center, Guangzhou Medical University, China (J.L., F.D., Q.L.)
| | - Gary Sweeney
- Department of Biology, York University, Toronto, Canada (G.S.)
| | - Connie Wai Hong Woo
- State Key Laboratory of Pharmaceutical Biotechnology (L.J., L.G., L.Y., L.S., R.Y., Y.L., Yao Wang, Y.C., X.J., Q.W., X.Y., B.L., C.W.H.W., Yu Wang, Z.X., Q.L., A.X.), University of Hong Kong, China.,Department of Pharmacology and Pharmacy (L.J., L.Y., B.L., C.W.H.W., Yu Wang, A.X.), University of Hong Kong, China
| | - Yu Wang
- State Key Laboratory of Pharmaceutical Biotechnology (L.J., L.G., L.Y., L.S., R.Y., Y.L., Yao Wang, Y.C., X.J., Q.W., X.Y., B.L., C.W.H.W., Yu Wang, Z.X., Q.L., A.X.), University of Hong Kong, China.,Department of Pharmacology and Pharmacy (L.J., L.Y., B.L., C.W.H.W., Yu Wang, A.X.), University of Hong Kong, China
| | - Zhengyuan Xia
- State Key Laboratory of Pharmaceutical Biotechnology (L.J., L.G., L.Y., L.S., R.Y., Y.L., Yao Wang, Y.C., X.J., Q.W., X.Y., B.L., C.W.H.W., Yu Wang, Z.X., Q.L., A.X.), University of Hong Kong, China.,Department of Medicine (L.J., L.G., L.S., R.Y., Y.L., Yao Wang, X.J., Q.W., X.Y., J.L., Z.X., Q.L., A.X.), University of Hong Kong, China.,Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China (Z.X.)
| | - Qizhou Lian
- State Key Laboratory of Pharmaceutical Biotechnology (L.J., L.G., L.Y., L.S., R.Y., Y.L., Yao Wang, Y.C., X.J., Q.W., X.Y., B.L., C.W.H.W., Yu Wang, Z.X., Q.L., A.X.), University of Hong Kong, China.,Department of Medicine (L.J., L.G., L.S., R.Y., Y.L., Yao Wang, X.J., Q.W., X.Y., J.L., Z.X., Q.L., A.X.), University of Hong Kong, China.,Cord Blood Bank, Guangzhou Institute of Eugenics and Perinatology, Women and Children's Medical Center, Guangzhou Medical University, China (J.L., F.D., Q.L.)
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical Biotechnology (L.J., L.G., L.Y., L.S., R.Y., Y.L., Yao Wang, Y.C., X.J., Q.W., X.Y., B.L., C.W.H.W., Yu Wang, Z.X., Q.L., A.X.), University of Hong Kong, China.,Department of Medicine (L.J., L.G., L.S., R.Y., Y.L., Yao Wang, X.J., Q.W., X.Y., J.L., Z.X., Q.L., A.X.), University of Hong Kong, China.,Department of Pharmacology and Pharmacy (L.J., L.Y., B.L., C.W.H.W., Yu Wang, A.X.), University of Hong Kong, China
| |
Collapse
|
27
|
Yao M, Liu Y, Sun M, Qin S, Xin W, Guan X, Zhang B, He T, Huang Y. The molecular mechanisms and intervention strategies of mitophagy in cardiorenal syndrome. Front Physiol 2022; 13:1008517. [PMID: 36353377 PMCID: PMC9638141 DOI: 10.3389/fphys.2022.1008517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 10/13/2022] [Indexed: 11/15/2022] Open
Abstract
Cardiorenal syndrome (CRS) is defined as a disorder of the heart and kidney, in which acute or chronic injury of one organ may lead to acute or chronic dysfunction of the other. It is characterized by high morbidity and mortality, resulting in high economic costs and social burdens. However, there is currently no effective drug-based treatment. Emerging evidence implicates the involvement of mitophagy in the progression of CRS, including cardiovascular disease (CVD) and chronic kidney disease (CKD). In this review, we summarized the crucial roles and molecular mechanisms of mitophagy in the pathophysiology of CRS. It has been reported that mitophagy impairment contributes to a vicious loop between CKD and CVD, which ultimately accelerates the progression of CRS. Further, recent studies revealed that targeting mitophagy may serve as a promising therapeutic approach for CRS, including clinical drugs, stem cells and small molecule agents. Therefore, studies focusing on mitophagy may benefit for expanding innovative basic research, clinical trials, and therapeutic strategies for CRS.
Collapse
Affiliation(s)
- Mengying Yao
- Department of Nephrology, The key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yong Liu
- Department of Nephrology, The key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Mengjia Sun
- Department of Cardiology, Institute of Cardiovascular Diseases of PLA, The Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Shaozong Qin
- Department of Nephrology, The key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Wang Xin
- Department of Nephrology, The key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Xu Guan
- Department of Nephrology, The key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Bo Zhang
- Department of Nephrology, The key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Ting He
- Department of Nephrology, The key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
- *Correspondence: Yinghui Huang, ; Ting He,
| | - Yinghui Huang
- Department of Nephrology, The key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
- *Correspondence: Yinghui Huang, ; Ting He,
| |
Collapse
|
28
|
Hu J, Liu T, Fu F, Cui Z, Lai Q, Zhang Y, Yu B, Liu F, Kou J, Li F. Omentin1 ameliorates myocardial ischemia-induced heart failure via SIRT3/FOXO3a-dependent mitochondrial dynamical homeostasis and mitophagy. Lab Invest 2022; 20:447. [PMID: 36192726 PMCID: PMC9531426 DOI: 10.1186/s12967-022-03642-x] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 09/16/2022] [Indexed: 12/03/2022]
Abstract
Background Adipose tissue-derived adipokines are involved in various crosstalk between adipose tissue and other organs. Omentin1, a novel adipokine, exerts vital roles in the maintenance of body metabolism, insulin resistance and the like. However, the protective effect of omentin1 in myocardial ischemia (MI)-induced heart failure (HF) and its specific mechanism remains unclear and to be elucidated. Methods The model of MI-induced HF mice and oxygen glucose deprivation (OGD)-injured cardiomyocytes were performed. Mice with overexpression of omentin1 were constructed by a fat-specific adeno-associated virus (AAV) vector system. Results We demonstrated that circulating omentin1 level diminished in HF patients compared with healthy subjects. Furthermore, the fat-specific overexpression of omentin1 ameliorated cardiac function, cardiac hypertrophy, infarct size and cardiac pathological features, and also enhanced SIRT3/FOXO3a signaling in HF mice. Additionally, administration with AAV-omentin1 increased mitochondrial fusion and decreased mitochondrial fission in HF mice, as evidenced by up-regulated expression of Mfn2 and OPA1, and downregulation of p-Drp1(Ser616). Then, it also promoted PINK1/Parkin-dependent mitophagy. Simultaneously, treatment with recombinant omentin1 strengthened OGD-injured cardiomyocyte viability, restrained LDH release, and enhanced the mitochondrial accumulation of SIRT3 and nucleus transduction of FOXO3a. Besides, omentin1 also ameliorated unbalanced mitochondrial fusion-fission dynamics and activated mitophagy, thereby, improving the damaged mitochondria morphology and controlling mitochondrial quality in OGD-injured cardiomyocytes. Interestingly, SIRT3 played an important role in the improvement effects of omentin1 on mitochondrial function, unbalanced mitochondrial fusion-fission dynamics and mitophagy. Conclusion Omentin1 improves MI-induced HF and myocardial injury by maintaining mitochondrial dynamical homeostasis and activating mitophagy via upregulation of SIRT3/FOXO3a signaling. This study provides evidence for further application of omentin1 in cardiovascular diseases from the perspective of crosstalk between heart and adipose tissue. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-022-03642-x.
Collapse
Affiliation(s)
- Jingui Hu
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Research Center for Traceability and Standardization of TCMs, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Longmian Road, Nanjing, 211198, People's Republic of China
| | - Tao Liu
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Research Center for Traceability and Standardization of TCMs, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Longmian Road, Nanjing, 211198, People's Republic of China
| | - Fei Fu
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Research Center for Traceability and Standardization of TCMs, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Longmian Road, Nanjing, 211198, People's Republic of China
| | - Zekun Cui
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Research Center for Traceability and Standardization of TCMs, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Longmian Road, Nanjing, 211198, People's Republic of China
| | - Qiong Lai
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Research Center for Traceability and Standardization of TCMs, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Longmian Road, Nanjing, 211198, People's Republic of China
| | - Yuanyuan Zhang
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Research Center for Traceability and Standardization of TCMs, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Longmian Road, Nanjing, 211198, People's Republic of China
| | - Boyang Yu
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Research Center for Traceability and Standardization of TCMs, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Longmian Road, Nanjing, 211198, People's Republic of China
| | - Fuming Liu
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Junping Kou
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Research Center for Traceability and Standardization of TCMs, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Longmian Road, Nanjing, 211198, People's Republic of China.
| | - Fang Li
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Research Center for Traceability and Standardization of TCMs, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Longmian Road, Nanjing, 211198, People's Republic of China.
| |
Collapse
|
29
|
Riegger J, Maurer S, Pulasani S, Brenner RE. Simvastatin and fluvastatin attenuate trauma-induced cell death and catabolism in human cartilage. Front Bioeng Biotechnol 2022; 10:965302. [PMID: 36159664 PMCID: PMC9500391 DOI: 10.3389/fbioe.2022.965302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 08/19/2022] [Indexed: 11/15/2022] Open
Abstract
Joint injuries are known to induce pathomechanisms that might lead to posttraumatic osteoarthritis (PTOA). In this regard, statins with their pleiotropic effects could represent potential therapeutic agents in preventing the development of PTOA. Therefore, we investigated the effects of simvastatin and fluvastatin in a drop-tower-based human ex vivo cartilage trauma model. After 7 days, a mechanical impact (0.59 J) resulted in a decrease of the cell viability and increased expression of catabolic enzymes in cartilage explants. Simvastatin and fluvastatin treatment of impacted cartilage demonstrated cell protective effects in a concentration dependent manner. Moreover, statin therapy exhibited chondroprotective effects as demonstrated by attenuated expression of MMP-2 and MMP-13 as well as subsequent breakdown of collagen type II (after impact). Further analysis indicated antioxidative properties of the statins by upregulating the gene expression of SOD2 and suppression that of NOX2 and NOX4. Despite its protective effects, simvastatin impaired the biosynthesis of collagen type II, which was confirmed during chondrogenic redifferentiation of high passage chondrocytes. However, while long-term administration of statins for 4 weeks impaired chondrogenic redifferentiation, addition of simvastatin at low concentrations for 1 week exhibited a slightly promoting effect. In conclusion, our data imply that simvastatin and fluvastatin are suitable in terms of initial harm reduction after cartilage trauma.
Collapse
|
30
|
Lu G, Wang Y, Shi Y, Zhang Z, Huang C, He W, Wang C, Shen H. Autophagy in health and disease: From molecular mechanisms to therapeutic target. MedComm (Beijing) 2022; 3:e150. [PMID: 35845350 PMCID: PMC9271889 DOI: 10.1002/mco2.150] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 06/01/2022] [Accepted: 06/02/2022] [Indexed: 02/05/2023] Open
Abstract
Macroautophagy/autophagy is an evolutionally conserved catabolic process in which cytosolic contents, such as aggregated proteins, dysfunctional organelle, or invading pathogens, are sequestered by the double-membrane structure termed autophagosome and delivered to lysosome for degradation. Over the past two decades, autophagy has been extensively studied, from the molecular mechanisms, biological functions, implications in various human diseases, to development of autophagy-related therapeutics. This review will focus on the latest development of autophagy research, covering molecular mechanisms in control of autophagosome biogenesis and autophagosome-lysosome fusion, and the upstream regulatory pathways including the AMPK and MTORC1 pathways. We will also provide a systematic discussion on the implication of autophagy in various human diseases, including cancer, neurodegenerative disorders (Alzheimer disease, Parkinson disease, Huntington's disease, and Amyotrophic lateral sclerosis), metabolic diseases (obesity and diabetes), viral infection especially SARS-Cov-2 and COVID-19, cardiovascular diseases (cardiac ischemia/reperfusion and cardiomyopathy), and aging. Finally, we will also summarize the development of pharmacological agents that have therapeutic potential for clinical applications via targeting the autophagy pathway. It is believed that decades of hard work on autophagy research is eventually to bring real and tangible benefits for improvement of human health and control of human diseases.
Collapse
Affiliation(s)
- Guang Lu
- Department of Physiology, Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhouChina
| | - Yu Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic MedicineSichuan University and Collaborative Innovation Center for BiotherapyChengduChina
| | - Yin Shi
- Department of BiochemistryZhejiang University School of MedicineHangzhouChina
| | - Zhe Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic MedicineSichuan University and Collaborative Innovation Center for BiotherapyChengduChina
| | - Canhua Huang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic MedicineSichuan University and Collaborative Innovation Center for BiotherapyChengduChina
| | - Weifeng He
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn ResearchSouthwest HospitalArmy Medical UniversityChongqingChina
| | - Chuang Wang
- Department of Pharmacology, Provincial Key Laboratory of PathophysiologyNingbo University School of MedicineNingboZhejiangChina
| | - Han‐Ming Shen
- Department of Biomedical Sciences, Faculty of Health Sciences, Ministry of Education Frontiers Science Center for Precision OncologyUniversity of MacauMacauChina
| |
Collapse
|
31
|
Cherukuri SP, Chikatimalla R, Dasaradhan T, Koneti J, Gadde S, Kalluru R. Breast Cancer and the Cardiovascular Disease: A Narrative Review. Cureus 2022; 14:e27917. [PMID: 36110451 PMCID: PMC9464354 DOI: 10.7759/cureus.27917] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/12/2022] [Indexed: 11/05/2022] Open
Abstract
Breast cancer is the most common malignancy affecting females worldwide and is also among the top causes of all cancer-related deaths. Cardiovascular disease (CVD) is known to have the highest rate of mortality in women. There are several risk factors for both CVD and breast cancer that overlap, such as diet, smoking, and obesity, and also the current breast cancer treatment has a significant detrimental effect on cardiovascular health in general. Patients with exposure to potentially cardiotoxic treatments, including anthracyclines, trastuzumab, and radiation therapy, are more likely to develop CVD than non-cancer controls. Early detection and treatment may reduce the risk of the development of cardiac morbidity and mortality and would increase the number of breast cancer survivors. This article provides a comprehensive overview of breast cancer, identifies shared risk factors among breast cancer and CVD, and the cardiotoxic effects of therapy. It also reviews possible prevention and treatment of CVD in breast cancer patients and reviews literature about chemoprevention of cardiac disease in the setting of breast cancer treatment.
Collapse
|
32
|
Wang Q, Sun Z, Cao S, Lin X, Wu M, Li Y, Yin J, Zhou W, Huang S, Zhang A, Zhang Y, Xia W, Jia Z. Reduced Immunity Regulator MAVS Contributes to Non-Hypertrophic Cardiac Dysfunction by Disturbing Energy Metabolism and Mitochondrial Homeostasis. Front Immunol 2022; 13:919038. [PMID: 35844503 PMCID: PMC9283757 DOI: 10.3389/fimmu.2022.919038] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 06/06/2022] [Indexed: 11/20/2022] Open
Abstract
Cardiac dysfunction is manifested as decline of cardiac systolic function, and multiple cardiovascular diseases (CVDs) can develop cardiac insufficiency. Mitochondrial antiviral signaling (MAVS) is known as an innate immune regulator involved in viral infectious diseases and autoimmune diseases, whereas its role in the heart remains obscure. The alteration of MAVS was analyzed in animal models with non-hypertrophic and hypertrophic cardiac dysfunction. Then, MAVS-deficient mice were generated to examine the heart function, mitochondrial status and energy metabolism. In vitro, CRISPR/Cas9-based gene editing was used to delete MAVS in H9C2 cell lines and the phenotypes of mitochondria and energy metabolism were evaluated. Here we observed reduced MAVS expression in cardiac tissue from several non-hypertrophic cardiac dysfunction models, contrasting to the enhanced MAVS in hypertrophic heart. Furthermore, we examined the heart function in mice with partial or total MAVS deficiency and found spontaneously developed cardiac pump dysfunction and cardiac dilation as assessed by echocardiography parameters. Metabonomic results suggested MAVS deletion probably promoted cardiac dysfunction by disturbing energy metabolism, especially lipid metabolism. Disordered and mitochondrial homeostasis induced by mitochondrial oxidative stress and mitophagy impairment also advanced the progression of cardiac dysfunction of mice without MAVS. Knockout of MAVS using CRISPR/Cas9 in cardiomyocytes damaged mitochondrial structure and function, as well as increased mitochondrial ROS production. Therefore, reduced MAVS contributed to the pathogenesis of non-hypertrophic cardiac dysfunction, which reveals a link between a key regulator of immunity (MAVS) and heart function.
Collapse
Affiliation(s)
- Qian Wang
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
- Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Zhenzhen Sun
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
- Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Shihan Cao
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
- Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Xiuli Lin
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
- Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Mengying Wu
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
- Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Yuanyuan Li
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
- Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Jie Yin
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
- Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Wei Zhou
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
- Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Songming Huang
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
- Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Aihua Zhang
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
- Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Yue Zhang
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
- Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing, China
- *Correspondence: Zhanjun Jia, ; Weiwei Xia, ; Yue Zhang,
| | - Weiwei Xia
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
- Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing, China
- *Correspondence: Zhanjun Jia, ; Weiwei Xia, ; Yue Zhang,
| | - Zhanjun Jia
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
- Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing, China
- *Correspondence: Zhanjun Jia, ; Weiwei Xia, ; Yue Zhang,
| |
Collapse
|
33
|
Wu Y, Jiang T, Hua J, Xiong Z, Dai K, Chen H, Li L, Peng J, Peng X, Zheng Z, Xiong W. PINK1/Parkin-mediated mitophagy in cardiovascular disease: From pathogenesis to novel therapy. Int J Cardiol 2022; 361:61-69. [PMID: 35594994 DOI: 10.1016/j.ijcard.2022.05.025] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 04/29/2022] [Accepted: 05/11/2022] [Indexed: 12/22/2022]
Abstract
Cardiovascular disease(CVD)is one of the predominant causes of death and morbidity. Mitochondria play a key role in maintaining cardiac energy metabolism. However, mitochondrial dysfunction leads to excessive production of ROS, resulting in oxidative damage to cardiomyocytes and contributing to a variety of cardiovascular diseases. In such a case, the clearance of impaired mitochondria is necessary. Currently, most studies have indicated an essential role for mitophagy in maintaining cardiac homeostasis and regulating CVD-related metabolic transition. Recent studies have implicated that PTEN-induced putative kinase 1 (PINK1)/Parkin-mediated mitophagy has been implicated in maintaining cardiomyocyte homeostasis. Here, we discuss the physiological and pathological roles of PINK1/Parkin-mediated mitophagy in the cardiovascular system, as well as potential therapeutic strategies based on PINK1/Parkin-mediated mitophagy modulation, which are of great significance for the prevention and treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Yanze Wu
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China; Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Ting Jiang
- Department of Hospital Infection Control, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Jinghai Hua
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Zhiping Xiong
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Kai Dai
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Hui Chen
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Lei Li
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Jingtian Peng
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Xiaoping Peng
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Zeqi Zheng
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Wenjun Xiong
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China.
| |
Collapse
|
34
|
The Regulatory Mechanism and Effect of RIPK3 on PE-induced Cardiomyocyte Hypertrophy. J Cardiovasc Pharmacol 2022; 80:236-250. [PMID: 35561290 DOI: 10.1097/fjc.0000000000001293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 04/26/2022] [Indexed: 12/07/2022]
Abstract
ABSTRACT As a critical regulatory molecule, receptor-interacting protein kinase 3 (RIPK3) can mediate the signaling pathway of programmed necrosis. Calcium/calmodulin-dependent protein kinase II (CaMKII) has been proved as a new substrate for RIPK3-induced necroptosis. In the present study, we aimed to investigate the regulatory mechanism of RIPK3 on phenylephrine (PE)-induced cardiomyocyte hypertrophy. Cardiomyocyte hypertrophy was induced by exposure to PE (100 μM) for 48 h. Primary cardiomyocytes were pretreated with RIPK3 inhibitor GSK'872 (10 μM), and RIPK3 siRNA was used to deplete the intracellular expression of RIPK3. The indexes related to myocardial hypertrophy, cell injury, necroptosis, CaMKII activation, gene expression, oxidative stress, and mitochondrial membrane potential were measured. We found that after cardiomyocytes were stimulated by PE, the expressions of hypertrophy markers, atrial and brain natriuretic peptides (ANP and BNP), were increased, the release of lactate dehydrogenase (LDH) was increased, the level of adenosine triphosphate (ATP)was decreased, the oxidation and phosphorylation levels of CaMKII were increased, and CaMKIIδ alternative splicing was disturbed. However, both GSK'872 and depletion of RIPK3 could reduce myocardial dysfunction, inhibit CaMKII activation and necroptosis, and finally alleviate myocardial hypertrophy. In addition, the pretreatment of RIPK3 could also lessen the accumulation of reactive oxygen species (ROS) induced by PE and stabilize the membrane potential of mitochondria. These results indicated that targeted inhibition of RIPK3 could suppress the activation of CaMKII and reduce necroptosis and oxidative stress, leading to alleviated myocardial hypertrophy. Collectively, our findings provided valuable insights into the clinical treatment of hypertrophic cardiomyopathy.
Collapse
|
35
|
Zhang C, Zhang B, Zhang X, Wang M, Sun X, Sun G. Panax notoginseng Saponin Protects Against Diabetic Cardiomyopathy Through Lipid Metabolism Modulation. J Am Heart Assoc 2022; 11:e023540. [PMID: 35112884 PMCID: PMC9245810 DOI: 10.1161/jaha.121.023540] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Background People with diabetes are more likely to develop cardiovascular diseases. Lipotoxicity plays a key role in the development of diabetic cardiomyopathy. Panax notoginseng saponin (PNS) has been used to treat diabetes and obesity. However, the role of PNS in diabetic cardiomyopathy remains unclear. Methods and Results Diabetic db/db mice received high‐dose (200 mg/kg per day) or medium‐dose (100 mg/kg per day) PNS by gavage for 12 weeks until week 36. Lipid accumulation and cardiac function in diabetic mice were detected and possible mechanisms involved were explored. PNS significantly improved body weight, body fat content, serum lipids, adipocytokines, and antioxidative function in db/db mice. Lipid accumulation in adipose tissue, liver, and heart were also alleviated by PNS treatment. Cardiac function and mitochondrial structure were also improved by PNS. H9c2 cells were treated with palmitate acid, and PNS pretreatment reduced lipid accumulation, mitochondrial reactive oxygen species, as well as improved mitochondrial membrane potential and mitochondrial oxygen consumption rate. Levels of proteins and expression of genes related to glucose and lipid metabolism, antioxidative function, and mitochondrial dynamics were also improved by PNS administration. Conclusions PNS attenuated heart dysfunction in diabetic mice by reducing lipotoxicity as well as modulating oxidative stress and improving mitochondrial function.
Collapse
Affiliation(s)
- Chenyang Zhang
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational MedicineInstitute of Medicinal Plant DevelopmentPeking Union Medical College and Chinese Academy of Medical SciencesBeijingChina
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal MedicineMinistry of EducationBeijingChina
- Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolic DisordersState Administration of Traditional Chinese MedicineBeijingChina
- NMPA Key Laboratory for Research and Evaluation of PharmacovigilanceBeijingChina
- Zhongguancun Open Laboratory of the Research and Development of Natural Medicine and Health ProductsBeijingChina
- Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine PrescriptionChinese Academy of Medical SciencesBeijingChina
| | - Bin Zhang
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational MedicineInstitute of Medicinal Plant DevelopmentPeking Union Medical College and Chinese Academy of Medical SciencesBeijingChina
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal MedicineMinistry of EducationBeijingChina
- Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolic DisordersState Administration of Traditional Chinese MedicineBeijingChina
- NMPA Key Laboratory for Research and Evaluation of PharmacovigilanceBeijingChina
- Zhongguancun Open Laboratory of the Research and Development of Natural Medicine and Health ProductsBeijingChina
- Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine PrescriptionChinese Academy of Medical SciencesBeijingChina
| | - Xuelian Zhang
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational MedicineInstitute of Medicinal Plant DevelopmentPeking Union Medical College and Chinese Academy of Medical SciencesBeijingChina
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal MedicineMinistry of EducationBeijingChina
- Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolic DisordersState Administration of Traditional Chinese MedicineBeijingChina
- NMPA Key Laboratory for Research and Evaluation of PharmacovigilanceBeijingChina
- Zhongguancun Open Laboratory of the Research and Development of Natural Medicine and Health ProductsBeijingChina
- Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine PrescriptionChinese Academy of Medical SciencesBeijingChina
| | - Min Wang
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational MedicineInstitute of Medicinal Plant DevelopmentPeking Union Medical College and Chinese Academy of Medical SciencesBeijingChina
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal MedicineMinistry of EducationBeijingChina
- Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolic DisordersState Administration of Traditional Chinese MedicineBeijingChina
- NMPA Key Laboratory for Research and Evaluation of PharmacovigilanceBeijingChina
- Zhongguancun Open Laboratory of the Research and Development of Natural Medicine and Health ProductsBeijingChina
- Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine PrescriptionChinese Academy of Medical SciencesBeijingChina
| | - Xiaobo Sun
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational MedicineInstitute of Medicinal Plant DevelopmentPeking Union Medical College and Chinese Academy of Medical SciencesBeijingChina
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal MedicineMinistry of EducationBeijingChina
- Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolic DisordersState Administration of Traditional Chinese MedicineBeijingChina
- NMPA Key Laboratory for Research and Evaluation of PharmacovigilanceBeijingChina
- Zhongguancun Open Laboratory of the Research and Development of Natural Medicine and Health ProductsBeijingChina
- Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine PrescriptionChinese Academy of Medical SciencesBeijingChina
| | - Guibo Sun
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational MedicineInstitute of Medicinal Plant DevelopmentPeking Union Medical College and Chinese Academy of Medical SciencesBeijingChina
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal MedicineMinistry of EducationBeijingChina
- Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolic DisordersState Administration of Traditional Chinese MedicineBeijingChina
- NMPA Key Laboratory for Research and Evaluation of PharmacovigilanceBeijingChina
- Zhongguancun Open Laboratory of the Research and Development of Natural Medicine and Health ProductsBeijingChina
- Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine PrescriptionChinese Academy of Medical SciencesBeijingChina
| |
Collapse
|
36
|
Lipid Droplet-a New Target in Ischemic Heart Disease. J Cardiovasc Transl Res 2022; 15:730-739. [PMID: 34984637 DOI: 10.1007/s12265-021-10204-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 12/22/2021] [Indexed: 10/19/2022]
Abstract
Lipid droplet (LD) is a kind of subcellular organelle, which originates from the endoplasmic reticulum (ER). LDs can move flexibly between other organelles and store energy in the cells. In recent years, LDs and lipid droplet-associated proteins have attracted added attention at home and abroad, especially in cardiovascular diseases. Cardiovascular diseases, especially ischemic heart disease (IHD), have always been the focus of attention because of their high morbidity and mortality. Atherosclerosis and myocardial remodeling are two important pathologic processes of IHD, and LDs and other organelles are involved in the development of the disease. The interaction between LDs and ER is involved in the formation of foam cells in atherosclerosis. And LDs, mitochondria, and lysosomes also affect the remodeling of cardiomyocytes by affecting ROS production and regulating PI3K/AKT pathways. In this article, we will review the role of LDs in IHD.
Collapse
|
37
|
He X, Liu J, Zang WJ. Mitochondrial homeostasis and redox status in cardiovascular diseases: Protective role of the vagal system. Free Radic Biol Med 2022; 178:369-379. [PMID: 34906725 DOI: 10.1016/j.freeradbiomed.2021.12.255] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/21/2021] [Accepted: 12/09/2021] [Indexed: 01/01/2023]
Abstract
Mitochondria participate in essential cellular functions, including energy production, metabolism, redox homeostasis regulation, intracellular Ca2+ handling, apoptosis, and cell fate determination. Disruption of mitochondrial homeostasis under pathological conditions results in mitochondrial reactive oxygen species (ROS) generation and energy insufficiency, which further disturb mitochondrial and cellular homeostasis in a deleterious loop. Mitochondrial redox status has therefore become a potential target for therapy against cardiovascular diseases. In this review, we highlight recent progress in determining the roles of mitochondrial processes in regulating mitochondrial redox status, including mitochondrial dynamics (fusion-fission pathways), mitochondrial cristae remodeling, mitophagy, biogenesis, and mitochondrion-organelle interactions (endoplasmic reticulum-mitochondrion interactions, nucleus-mitochondrion communication, and lipid droplet-mitochondrion interactions). The strategies that activate vagal system include direct vagal activation (electrical vagal stimulation and administration of vagal neurotransmitter acetylcholine) and pharmacological modulation (choline and cholinesterase inhibitors). The vagal system plays an important role in maintaining mitochondrial homeostasis and suppressing mitochondrial oxidative stress by promoting mitochondrial biogenesis and mitophagy, moderating mitochondrial fusion and fission, strengthening mitochondrial cristae stabilization, regulating mitochondrion-organelle interactions, and inhibiting mitochondrial Ca2+ overload. Therefore, enhancement of vagal activity can maintain mitochondrial homeostasis and represents a promising therapeutic strategy for cardiovascular diseases.
Collapse
Affiliation(s)
- Xi He
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, PR China
| | - Jiankang Liu
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology and Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, PR China; University of Health and Rehabilitation Sciences, Qingdao, PR China
| | - Wei-Jin Zang
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, PR China.
| |
Collapse
|
38
|
Bhullar S, Shah A, Dhalla N. Mechanisms for the development of heart failure and improvement of cardiac function by angiotensin-converting enzyme inhibitors. SCRIPTA MEDICA 2022. [DOI: 10.5937/scriptamed53-36256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
Angiotensin-converting enzyme (ACE) inhibitors, which prevent the conversion of angiotensin I to angiotensin II, are well-known for the treatments of cardiovascular diseases, such as heart failure, hypertension and acute coronary syndrome. Several of these inhibitors including captopril, enalapril, ramipril, zofenopril and imidapril attenuate vasoconstriction, cardiac hypertrophy and adverse cardiac remodeling, improve clinical outcomes in patients with cardiac dysfunction and decrease mortality. Extensive experimental and clinical research over the past 35 years has revealed that the beneficial effects of ACE inhibitors in heart failure are associated with full or partial prevention of adverse cardiac remodeling. Since cardiac function is mainly determined by coordinated activities of different subcellular organelles, including sarcolemma, sarcoplasmic reticulum, mitochondria and myofibrils, for regulating the intracellular concentration of Ca2+ and myocardial metabolism, there is ample evidence to suggest that adverse cardiac remodelling and cardiac dysfunction in the failing heart are the consequence of subcellular defects. In fact, the improvement of cardiac function by different ACE inhibitors has been demonstrated to be related to the attenuation of abnormalities in subcellular organelles for Ca2+-handling, metabolic alterations, signal transduction defects and gene expression changes in failing cardiomyocytes. Various ACE inhibitors have also been shown to delay the progression of heart failure by reducing the formation of angiotensin II, the development of oxidative stress, the level of inflammatory cytokines and the occurrence of subcellular defects. These observations support the view that ACE inhibitors improve cardiac function in the failing heart by multiple mechanisms including the reduction of oxidative stress, myocardial inflammation and Ca2+-handling abnormalities in cardiomyocytes.
Collapse
|
39
|
Simvastatin potentiates the cell-killing activity of imatinib in imatinib-resistant chronic myeloid leukemia cells mainly through PI3K/AKT pathway attenuation and Myc downregulation. Eur J Pharmacol 2021; 913:174633. [PMID: 34843676 DOI: 10.1016/j.ejphar.2021.174633] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 11/11/2021] [Accepted: 11/11/2021] [Indexed: 12/28/2022]
Abstract
Constitutively activated BCR-ABL kinase is considered the driver event responsible in the initiation and development of chronic myeloid leukemia (CML). The advent of the first BCR-ABL inhibitor imatinib has significantly improved the clinical outcome of CML cases. However, resistance to imatinib occurs in 25-30% of CML patients. Due to the lack of effective therapeutic strategies, novel treatment approaches are urgently required for imatinib-resistant CML. Simvastatin, a well-known HMG-CoA reductase inhibitor that confers tremendous clinical benefits in cardiovascular diseases, has attracted mounting attentions for its potent antitumor effects on multiple tumor types. In this study, we demonstrated that simvastatin monotherapy was effective in diminishing cell viability in both imatinib-sensitive and imatinib-resistant CML cells, including T351I mutated cells, with the latter being less vulnerable to the simvastatin than the former. Notably, we found that simvastatin acted as a robust cytotoxic sensitizer of imatinib to kill imatinib-resistant and T315I mutated CML cells in vitro and in vivo. Mechanistically, the cooperative interaction of simvastatin and imatinib was associated with the inactivation of the PI3K/Akt signaling pathway, which was a classical downstream pro-survival cascade of the BCR-ABL kinase. In addition, this drug combination obviously decreased Myc expression through attenuation of canonical Wnt/β-catenin signaling and increased H3K27 trimethylation. Taken together, we provide attractive preclinical results for the combinatorial regimen of simvastatin and imatinib against imatinib-resistant and T315I mutated CML cells. This combined regimens warrants further clinical investigations in patients with imatinib-resistant CML.
Collapse
|
40
|
Wang C, Cui R, Niu C, Zhong X, Zhu Q, Ji D, Li X, Zhang H, Liu C, Zhou L, Li Y, Xu G, Wei Y. Low-dose PCB126 exposure disrupts cardiac metabolism and causes hypertrophy and fibrosis in mice. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 290:118079. [PMID: 34488161 DOI: 10.1016/j.envpol.2021.118079] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 08/29/2021] [Accepted: 08/30/2021] [Indexed: 06/13/2023]
Abstract
The residue of polychlorinated biphenyls (PCBs) exists throughout the environment and humans are subject to long-term exposure. As such, the potential environmental and health risk caused by low-dose exposure to PCBs has attracted much attention. 3, 3', 4, 4', 5-pentachlorobiphenyl (PCB126), the highest toxicity compound among dioxin-like-PCBs, has been widely used and mass-produced. Cardiotoxicity is PCB126's crucial adverse effect. Maintaining proper metabolism underlies heart health, whereas the impact of PCB126 exposure on cardiac metabolic patterns has yet to be elucidated. In this study, we administered 0.5 and 50 μg/kg bw of PCB126 to adult male mice weekly by gavage for eight weeks. Pathological results showed that low-dose PCB126 exposure induced heart injury. Metabolomic analysis of the heart tissue exposed to low-dose PCB126 identified 59 differential metabolites that were involved in lipid metabolism, amino acid metabolism, and the tricarboxylic acid (TCA) cycle. Typical metabolomic characteristic of cardiac hypertrophy was reflected by accumulation of fatty acids (e.g. palmitic, palmitoleic, and linoleic acid), and disturbance of carbohydrates including D-glucose and intermediates in TCA cycle (fumaric, succinic, and citric acid). Low-dose PCB126 exposure increased glycine and threonine, the amino acids necessary for the productions of collagen and elastin. Besides, PCB126-exposed mice exhibited upregulation of collagen synthesis enzymes and extracellular matrix proteins, indicative of cardiac fibrosis. Moreover, the expression of genes related to TGFβ/PPARγ/MMP-2 signaling pathway was perturbed in the PCB126-treated hearts. Together, our results reveal that low-dose PCB126 exposure disrupts cardiac metabolism correlated with hypertrophy and fibrosis. This study sheds light on the underlying mechanism of PCBs' cardiotoxicity and identifies potential sensitive biomarkers for environmental monitoring.
Collapse
Affiliation(s)
- Can Wang
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Ruina Cui
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Congying Niu
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Xiali Zhong
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Qicheng Zhu
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Di Ji
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Xianjie Li
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Hongxia Zhang
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Chunqiao Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China
| | - Lina Zhou
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Yanli Li
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Guowang Xu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Yanhong Wei
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China.
| |
Collapse
|
41
|
Kuo HF, Liu IF, Li CY, Tsai CS, Chen YH, Lian WS, Lin TC, Liu YR, Lee TY, Huang CY, Hsieh CC, Hsu CH, Lin FY, Liu PL. Endocardial Endothelial Dysfunction and Unknown Polymorphic Composite Accumulation in Heart Failure. Biomedicines 2021; 9:biomedicines9101465. [PMID: 34680582 PMCID: PMC8533412 DOI: 10.3390/biomedicines9101465] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 10/04/2021] [Accepted: 10/11/2021] [Indexed: 01/03/2023] Open
Abstract
The accumulation of unknown polymorphic composites in the endocardium damages the endocardial endothelium (EE). However, the composition and role of unknown polymorphic composites in heart failure (HF) progression remain unclear. Here, we aimed to explore composite deposition during endocardium damage and HF progression. Adult male Sprague–Dawley rats were divided into two HF groups—angiotensin II-induced HF and left anterior descending artery ligation-induced HF. Heart tissues from patients who had undergone coronary artery bypass graft surgery (non-HF) and those with dilated cardiomyopathy (DCM) and ischemic cardiomyopathy (ICM) were collected. EE damage, polymorphic unknown composite accumulation, and elements in deposits were examined. HF progression reduced the expression of CD31 in the endocardium, impaired endocardial integrity, and exposed the myofibrils and mitochondria. The damaged endocardial surface showed the accumulation of unknown polymorphic composites. In the animal HF model, especially HF caused by myocardial infarction, the weight and atomic percentages of O, Na, and N in the deposited composites were significantly higher than those of the other groups. The deposited composites in the human HF heart section (DCM) had a significantly higher percentage of Na and S than the other groups, whereas the percentage of C and Na in the DCM and ICM groups was significantly higher than those of the control group. HF causes widespread EE dysfunction, and EndMT was accompanied by polymorphic composites of different shapes and elemental compositions, which further damage and deteriorate heart function.
Collapse
Affiliation(s)
- Hsuan-Fu Kuo
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (H.-F.K.); (C.-Y.L.); (T.-C.L.)
- Department of Internal Medicine, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Internal Medicine, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - I-Fan Liu
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan;
- Heart Center, Cheng Hsin General Hospital, Taipei 112, Taiwan
| | - Chia-Yang Li
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (H.-F.K.); (C.-Y.L.); (T.-C.L.)
| | - Chien-Sung Tsai
- Division of Cardiovascular Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan;
- Department and Graduate Institute of Pharmacology, National Defense Medical Center, Taipei 114, Taiwan
| | - Yung-Hsiang Chen
- Graduate Institute of Integrated Medicine, College of Chinese Medicine, China Medical University, Taichung 404, Taiwan;
- Department of Psychology, College of Medical and Health Science, Asia University, Taichung 413, Taiwan
| | - Wei-Shiung Lian
- Core Laboratory for Phenomics and Diagnostic, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan;
- Department of Medical Research, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
| | - Tzu-Chieh Lin
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (H.-F.K.); (C.-Y.L.); (T.-C.L.)
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Yu-Ru Liu
- Department of Respiratory Therapy, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (Y.-R.L.); (T.-Y.L.); (C.-Y.H.)
| | - Tsung-Ying Lee
- Department of Respiratory Therapy, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (Y.-R.L.); (T.-Y.L.); (C.-Y.H.)
| | - Chi-Yuan Huang
- Department of Respiratory Therapy, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (Y.-R.L.); (T.-Y.L.); (C.-Y.H.)
| | - Chong-Chao Hsieh
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Division of Cardiovascular Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Surgery, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Correspondence: (C.-C.H.); (C.-H.H.); (F.-Y.L.); (P.-L.L.)
| | - Chih-Hsin Hsu
- Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 740, Taiwan
- Correspondence: (C.-C.H.); (C.-H.H.); (F.-Y.L.); (P.-L.L.)
| | - Feng-Yen Lin
- Department of Internal Medicine and Taipei Heart Institute, Taipei Medical University, Taipei 106, Taiwan
- Division of Cardiology and Cardiovascular Research Center, Taipei Medical University Hospital, Taipei 106, Taiwan
- Correspondence: (C.-C.H.); (C.-H.H.); (F.-Y.L.); (P.-L.L.)
| | - Po-Len Liu
- Department of Respiratory Therapy, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (Y.-R.L.); (T.-Y.L.); (C.-Y.H.)
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
- Correspondence: (C.-C.H.); (C.-H.H.); (F.-Y.L.); (P.-L.L.)
| |
Collapse
|
42
|
Zhan H, Huang F, Niu Q, Jiao M, Han X, Zhang K, Ma W, Mi S, Guo S, Zhao Z. Downregulation of miR-128 Ameliorates Ang II-Induced Cardiac Remodeling via SIRT1/PIK3R1 Multiple Targets. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:8889195. [PMID: 34646427 PMCID: PMC8505057 DOI: 10.1155/2021/8889195] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 08/22/2021] [Accepted: 09/01/2021] [Indexed: 12/14/2022]
Abstract
Recent studies reported that miR-128 was differentially expressed in cardiomyocytes in response to pathologic stress. However, its function and mechanism remain to be fully elucidated. The aim of the present study was to investigate the role of miR-128 in chronic angiotensin II (Ang II) infusion-induced cardiac remodeling and its underlying mechanism. The cardiac remodeling and heart failure in vivo were established in C57BL/6 mice by chronic subcutaneous Ang II delivery. Knocking down miR-128 was conducted in the hearts of the mice by intravenous injection of HBAAV2/9-miR-128-GFP sponge (miR-128 inhibitor). In vitro experiments of cardiac hypertrophy, apoptosis, and aberrant autophagy were performed in cultured cells after Ang II treatment or transfection of miR-128 antagomir. Our results showed that chronic Ang II delivery for 28 days induced cardiac dysfunction, hypertrophy, fibrosis, apoptosis, and oxidative stress in the mice, while the miR-128 expression was notably enhanced in the left ventricle. Silencing miR-128 in the hearts of mice ameliorated Ang II-induced cardiac dysfunction, hypertrophy, fibrosis apoptosis, and oxidative stress injury. Moreover, Ang II induced excessive autophagy in the mouse hearts, which was suppressed by miR-128 knockdown. In cultured cells, Ang II treatment induced a marked elevation in the miR-128 expression. Downregulation of miR-128 in the cells by transfection with miR-128 antagomir attenuated Ang II-induced apoptosis and oxidative injury probably via directly targeting on the SIRT1/p53 pathway. Intriguingly, we found that miR-128 inhibition activated PIK3R1/Akt/mTOR pathway and thereby significantly damped Ang II-stimulated pathological autophagy in cardiomyocytes, which consequently mitigated cell oxidative stress and apoptosis. In conclusion, downregulation of miR-128 ameliorates Ang II-provoked cardiac oxidative stress, hypertrophy, fibrosis, apoptosis, and dysfunction in mice, likely through targeting on PIK3R1/Akt/mTORC1 and/or SIRT1/p53 pathways. These results indicate that miR-128 inhibition might be a potent therapeutic strategy for maladaptive cardiac remodeling and heart failure.
Collapse
Affiliation(s)
- Heqin Zhan
- Department of Pharmacology, School of Basic Medicine Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China
- Department of Pharmacology, College of Pharmacy, Xinxiang Medical University, Xinxiang, Henan 453003, China
| | - Feng Huang
- Department of Pharmacology, College of Pharmacy, Xinxiang Medical University, Xinxiang, Henan 453003, China
| | - Qian Niu
- Department of Pharmacology, College of Pharmacy, Xinxiang Medical University, Xinxiang, Henan 453003, China
- Department of Pharmacy, Sanmenxia Central Hospital, Sanmenxia, Henan 472000, China
| | - Mingli Jiao
- Department of Pharmacology, College of Pharmacy, Xinxiang Medical University, Xinxiang, Henan 453003, China
| | - Xumeng Han
- Department of Pharmacology, College of Pharmacy, Xinxiang Medical University, Xinxiang, Henan 453003, China
| | - Kaina Zhang
- Department of Pharmacology, School of Basic Medicine Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China
| | - WenZhuo Ma
- Department of Pharmacology, School of Basic Medicine Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China
| | - Shan Mi
- Department of Pharmacology, College of Pharmacy, Xinxiang Medical University, Xinxiang, Henan 453003, China
| | - Shiyu Guo
- Department of Pharmacology, College of Pharmacy, Xinxiang Medical University, Xinxiang, Henan 453003, China
| | - Zhenghang Zhao
- Department of Pharmacology, School of Basic Medicine Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China
| |
Collapse
|
43
|
miR-29a-3p/THBS2 Axis Regulates PAH-Induced Cardiac Fibrosis. Int J Mol Sci 2021; 22:ijms221910574. [PMID: 34638915 PMCID: PMC8509017 DOI: 10.3390/ijms221910574] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 09/23/2021] [Accepted: 09/26/2021] [Indexed: 12/21/2022] Open
Abstract
Pulmonary artery hypertension (PAH) pathology involves extracellular matrix (ECM) remodeling in cardiac tissues, thus promoting cardiac fibrosis progression. miR-29a-3p reportedly inhibits lung progression and liver fibrosis by regulating ECM protein expression; however, its role in PAH-induced fibrosis remains unclear. In this study, we aimed to investigate the role of miR-29a-3p in cardiac fibrosis progression in PAH and its influence on ECM protein thrombospondin-2 (THBS2) expression. The diagnostic and prognostic values of miR-29a-3p and THBS2 in PAH were evaluated. The expressions and effects of miR-29a-3p and THBS2 were assessed in cell culture, monocrotaline-induced PAH mouse model, and patients with PAH. The levels of circulating miR-29a-3p and THBS2 in patients and mice with PAH decreased and increased, respectively. miR-29a-3p directly targets THBS2 and regulates THBS2 expression via a direct anti-fibrotic effect on PAH-induced cardiac fibrosis. The circulating levels of miR-29a-3p and THBS2 were correlated with PAH diagnostic parameters, suggesting their independent prognostic value. miR-29a-3p targeted THBS2 expression via a direct anti-fibrotic effect on PAH-induced cardiac fibrosis, indicating miR-29a-3p acts as a messenger with promising therapeutic effects.
Collapse
|
44
|
Bhullar SK, Shah AK, Dhalla NS. Role of angiotensin II in the development of subcellular remodeling
in heart failure. EXPLORATION OF MEDICINE 2021. [DOI: 10.37349/emed.2021.00054] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The development of heart failure under various pathological conditions such as myocardial infarction (MI), hypertension and diabetes are accompanied by adverse cardiac remodeling and cardiac dysfunction. Since heart function is mainly determined by coordinated activities of different subcellular organelles including sarcolemma, sarcoplasmic reticulum, mitochondria and myofibrils for regulating the intracellular concentration of Ca2+, it has been suggested that the occurrence of heart failure is a consequence of subcellular remodeling, metabolic alterations and Ca2+-handling abnormalities in cardiomyocytes. Because of the elevated plasma levels of angiotensin II (ANG II) due to activation of the renin-angiotensin system (RAS) in heart failure, we have evaluated the effectiveness of treatments with angiotensin converting enzyme (ACE) inhibitors and ANG II type 1 receptor (AT1R) antagonists in different experimental models of heart failure. Attenuation of marked alterations in subcellular activities, protein content and gene expression were associated with improvement in cardiac function in MI-induced heart failure by treatment with enalapril (an ACE inhibitor) or losartan (an AT1R antagonist). Similar beneficial effects of ANG II blockade on subcellular remodeling and cardiac performance were also observed in failing hearts due to pressure overload, volume overload or chronic diabetes. Treatments with enalapril and losartan were seen to reduce the degree of RAS activation as well as the level of oxidative stress in failing hearts. These observations provide evidence which further substantiate to support the view that activation of RAS and high level of plasma ANG II play a critical role in inducing subcellular defects and cardiac dys-function during the progression of heart failure.
Collapse
Affiliation(s)
- Sukhwinder K. Bhullar
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, University of Manitoba, Winnipeg, Manitoba R2H 2A6, Canada
| | - Anureet K. Shah
- School of Kinesiology, Nutrition and Food Science, California State University, Los Angeles, CA 90032, USA
| | - Naranjan S. Dhalla
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, University of Manitoba, Winnipeg, Manitoba R2H 2A6, Canada; Department of Physiology and Pathophysiology, Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba R3E 3P5, Canada
| |
Collapse
|
45
|
Correction. Br J Pharmacol 2021; 178:3845-3846. [PMID: 34431101 DOI: 10.1111/bph.15624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
|
46
|
An D, Zeng Q, Zhang P, Ma Z, Zhang H, Liu Z, Li J, Ren H, Xu D. Alpha-ketoglutarate ameliorates pressure overload-induced chronic cardiac dysfunction in mice. Redox Biol 2021; 46:102088. [PMID: 34364218 PMCID: PMC8353361 DOI: 10.1016/j.redox.2021.102088] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 07/27/2021] [Accepted: 07/29/2021] [Indexed: 12/11/2022] Open
Abstract
Increasing evidence indicates the involvement of myocardial oxidative injury and mitochondrial dysfunction in the pathophysiology of heart failure (HF). Alpha-ketoglutarate (AKG) is an intermediate metabolite of the tricarboxylic acid (TCA) cycle that participates in different cellular metabolic and regulatory pathways. The circulating concentration of AKG was found to decrease with ageing and is elevated after acute exercise and resistance exercise and in HF. Recent studies in experimental models have shown that dietary AKG reduces reactive oxygen species (ROS) production and systemic inflammatory cytokine levels, regulates metabolism, extends lifespan and delays the occurrence of age-related decline. However, the effects of AKG on HF remain unclear. In the present study, we explored the effects of AKG on left ventricular (LV) systolic function, the myocardial ROS content and mitophagy in mice with transverse aortic constriction (TAC). AKG supplementation inhibited pressure overload-induced myocardial hypertrophy and fibrosis and improved cardiac systolic dysfunction; in vitro, AKG decreased the Ang II-induced upregulation of β-MHC and ANP, reduced ROS production and cardiomyocyte apoptosis, and repaired Ang II-mediated injury to the mitochondrial membrane potential (MMP). These benefits of AKG in the TAC mice may have been obtained by enhanced mitophagy, which cleared damaged mitochondria. In summary, our study suggests that AKG improves myocardial hypertrophy remodelling, fibrosis and LV systolic dysfunction in the pressure-overloaded heart by promoting mitophagy to clear damaged mitochondria and reduce ROS production; thus, AKG may have therapeutic potential for HF.
Collapse
Affiliation(s)
- Dongqi An
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou, China
| | - Qingchun Zeng
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou, China
| | - Peijian Zhang
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou, China
| | - Zhuang Ma
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou, China
| | - Hao Zhang
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou, China
| | - Zuheng Liu
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou, China; Department of Cardiology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Jiaying Li
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou, China
| | - Hao Ren
- Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou, China; Department of Rheumatology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Dingli Xu
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou, China.
| |
Collapse
|
47
|
Dual Role of Mitophagy in Cardiovascular Diseases. J Cardiovasc Pharmacol 2021; 78:e30-e39. [PMID: 34232224 DOI: 10.1097/fjc.0000000000001046] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 04/08/2021] [Indexed: 01/13/2023]
Abstract
ABSTRACT Mitophagy is involved in the development of various cardiovascular diseases, such as atherosclerosis, heart failure, myocardial ischemia/reperfusion injury, and hypertension. Mitophagy is essential for maintaining intracellular homeostasis and physiological function in most cardiovascular origin cells, such as cardiomyocytes, endothelial cells, and vascular smooth muscle cells. Mitophagy is crucial to ensuring energy supply by selectively removing dysfunctional mitochondria, maintaining a balance in the number of mitochondria in cells, ensuring the integrity of mitochondrial structure and function, maintaining homeostasis, and promoting cell survival. Substantial research has indicated a "dual" effect of mitophagy on cardiac function, with inadequate and increased mitochondrial degradation both likely to influence the progression of cardiovascular disease. This review summarizes the main regulatory pathways of mitophagy and emphasizes that an appropriate amount of mitophagy can prevent endothelial cell injury, vascular smooth muscle cell proliferation, macrophage polarization, and cardiomyocyte apoptosis, avoiding further progression of cardiovascular diseases.
Collapse
|
48
|
Collins HE, Kane MS, Litovsky SH, Darley-Usmar VM, Young ME, Chatham JC, Zhang J. Mitochondrial Morphology and Mitophagy in Heart Diseases: Qualitative and Quantitative Analyses Using Transmission Electron Microscopy. FRONTIERS IN AGING 2021; 2:670267. [PMID: 35822027 PMCID: PMC9261312 DOI: 10.3389/fragi.2021.670267] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Accepted: 03/26/2021] [Indexed: 01/21/2023]
Abstract
Transmission electron microscopy (TEM) has long been an important technique, capable of high degree resolution and visualization of subcellular structures and organization. Over the last 20 years, TEM has gained popularity in the cardiovascular field to visualize changes at the nanometer scale in cardiac ultrastructure during cardiovascular development, aging, and a broad range of pathologies. Recently, the cardiovascular TEM enabled the studying of several signaling processes impacting mitochondrial function, such as mitochondrial fission/fusion, autophagy, mitophagy, lysosomal degradation, and lipophagy. The goals of this review are to provide an overview of the current usage of TEM to study cardiac ultrastructural changes; to understand how TEM aided the visualization of mitochondria, autophagy, and mitophagy under normal and cardiovascular disease conditions; and to discuss the overall advantages and disadvantages of TEM and potential future capabilities and advancements in the field.
Collapse
Affiliation(s)
- Helen E. Collins
- Division of Environmental Medicine, Department of Medicine, University of Louisville, KY, United States
| | - Mariame Selma Kane
- Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Silvio H. Litovsky
- Division of Anatomic Pathology, Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Victor M. Darley-Usmar
- Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Martin E. Young
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - John C. Chatham
- Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Jianhua Zhang
- Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
49
|
He Y, Huang W, Zhang C, Chen L, Xu R, Li N, Wang F, Han L, Yang M, Zhang D. Energy metabolism disorders and potential therapeutic drugs in heart failure. Acta Pharm Sin B 2021; 11:1098-1116. [PMID: 34094822 PMCID: PMC8144890 DOI: 10.1016/j.apsb.2020.10.007] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 08/27/2020] [Accepted: 09/07/2020] [Indexed: 02/07/2023] Open
Abstract
Heart failure (HF) is a global public health problem with high morbidity and mortality. A large number of studies have shown that HF is caused by severe energy metabolism disorders, which result in an insufficient heart energy supply. This deficiency causes cardiac pump dysfunction and systemic energy metabolism failure, which determine the development of HF and recovery of heart. Current HF therapy acts by reducing heart rate and cardiac preload and afterload, treating the HF symptomatically or delaying development of the disease. Drugs aimed at cardiac energy metabolism have not yet been developed. In this review, we outline the main characteristics of cardiac energy metabolism in healthy hearts, changes in metabolism during HF, and related pathways and targets of energy metabolism. Finally, we discuss drugs that improve cardiac function via energy metabolism to provide new research ideas for the development and application of drugs for treating HF.
Collapse
|
50
|
Liu WL, Li CY, Cheng WC, Chang CY, Chen YH, Lu CY, Wang SC, Liu YR, Cheng MH, Chong IW, Liu PL. High Mobility Group Box 1 Promotes Lung Cancer Cell Migration and Motility via Regulation of Dynamin-Related Protein 1. Int J Mol Sci 2021; 22:ijms22073628. [PMID: 33807275 PMCID: PMC8036886 DOI: 10.3390/ijms22073628] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 03/25/2021] [Accepted: 03/26/2021] [Indexed: 01/08/2023] Open
Abstract
High mobility group box 1 (HMGB1) has been demonstrated to promote the migration and invasion of non-small cell lung cancer (NSCLC). However, the mechanism of action of HMGB1 in regulating tumor mobility remains unclear. Therefore, we aimed to investigate whether HMGB1 affects mitochondria distribution and regulates dynamin-related protein 1 (DRP1)-mediated lamellipodia/filopodia formation to promote NSCLC migration. The regulation of mitochondrial membrane tension, dynamics, polarization, fission process, and cytoskeletal rearrangements in lung cancer cells by HMGB1 was analyzed using confocal microscopy. The HMGB1-mediated regulation of DRP1 phosphorylation and colocalization was determined using immunostaining and co-immunoprecipitation assays. The tumorigenic potential of HMGB1 was assessed in vivo and further confirmed using NSCLC patient samples. Our results showed that HMGB1 increased the polarity and mobility of cells (mainly by regulating the cytoskeletal system actin and microtubule dynamics and distribution), promoted the formation of lamellipodia/filopodia, and enhanced the expression and phosphorylation of DRP1 in both the nucleus and cytoplasm. In addition, HMGB1 and DRP1 expressions were positively correlated and exhibited poor prognosis and survival in patients with lung cancer. Collectively, HMGB1 plays a key role in the formation of lamellipodia and filopodia by regulating cytoskeleton dynamics and DRP1 expression to promote lung cancer migration.
Collapse
Affiliation(s)
- Wei-Lun Liu
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei City 242, Taiwan;
- Division of Critical Care Medicine, Department of Emergency and Critical Care Medicine, Fu Jen Catholic University Hospital, Fu Jen Catholic University, New Taipei City 243, Taiwan
| | - Chia-Yang Li
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Wei-Chung Cheng
- Graduate Institute of Biomedical Science, Research Center for Cancer Biology, China Medical University, Taichung 404, Taiwan;
| | - Chia-Yuan Chang
- Department of Mechanical Engineering, National Cheng Kung University, Tainan 701, Taiwan;
| | - Yung-Hsiang Chen
- Graduate Institute of Integrated Medicine, College of Chinese Medicine, China Medical University, Taichung 404, Taiwan;
- Department of Psychology, College of Medical and Health Science, Asia University, Taichung 413, Taiwan
| | - Chi-Yu Lu
- Department of Biochemistry, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Shu-Chi Wang
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Department of Medical Laboratory Science and Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Center for Liquid Biopsy, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Yu-Ru Liu
- Department of Respiratory Therapy, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Meng-Hsuan Cheng
- Department of Respiratory Therapy, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
- School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Correspondence: (M.-H.C.); (I.-W.C.); (P.-L.L.); Tel.: +886-7-312-1101 (ext. 5651) (M.-H.C. & I.-W.C.); +886-7-312-1101 (ext. 2801) (P.-L.L.)
| | - Inn-Wen Chong
- Department of Respiratory Therapy, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Correspondence: (M.-H.C.); (I.-W.C.); (P.-L.L.); Tel.: +886-7-312-1101 (ext. 5651) (M.-H.C. & I.-W.C.); +886-7-312-1101 (ext. 2801) (P.-L.L.)
| | - Po-Len Liu
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Department of Respiratory Therapy, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Correspondence: (M.-H.C.); (I.-W.C.); (P.-L.L.); Tel.: +886-7-312-1101 (ext. 5651) (M.-H.C. & I.-W.C.); +886-7-312-1101 (ext. 2801) (P.-L.L.)
| |
Collapse
|