201
|
Bubnova MG, Aronov DM. Efficacy of trimetazidine - an inhibitor of free fatty acids oxidation in the treatment of patients with stable angina pectoris and heart failure. KARDIOLOGIIA 2021; 61:65-76. [PMID: 34882080 DOI: 10.18087/cardio.2021.11.n1801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 09/09/2021] [Indexed: 06/13/2023]
Abstract
Aim To evaluate efficacy of modified-release trimetazidine (TMZ) included into the standard therapy for patients with stable angina and chronic heart failure (CHF) as a part of a subgroup analysis in the PERSPECTIVE study.Material and methods The study included 806 patients: group 1 (n=691), patients receiving a standard therapy and modified-release TMZ (TMZ group); and group 2 (n=115), patients receiving a standard therapy (control group). Total duration of the study was 12 months.Results In the TMZ group, the weekly number of angina attacks decreased by 41.9% (p<0.0001) in 2 months and by 69.6 % (from baseline, р<0.0001) in 12 months, and the frequency of nitroglycerine dosing decreased by 40.8 % (р<0.0001) and 67.7 % (р<0.0001), respectively. In the control group, the respective values did not change. In the TMZ group compared to the control group, the QT interval was shorter (7.9 %; р<0.05), the left ventricular (LV) end-systolic dimension was reduced (13.4 %; р<0.01), interventricular septal thickness and LV posterior wall thickness were decreased (9.5 %; р<0.01 and 12.2 %; р<0.01, respectively), and the ejection fraction was increased (11.4; р<0.05). Following the TMZ treatment, the leukocyte count in peripheral blood was decreased (5.3 %; р<0.01) and the serum concentration of high-sensitivity C-reactive protein was decreased (30.7 %; р<0.01) vs. increases of these indexes in the control group (17.9 %; р<0.05 and 17.8 %; р<0.05, respectively). The proportion of patients hospitalized for exacerbation of CHF or angina for 12 months was 8.6 % in the TMZ group and 15.7 % in the control group (p=0,001).Conclusion In patients with stable angina and CHF, inclusion of modified-release TMZ into the standard therapy decreases the number of angina attacks, reduces the activity of inflammatory factors, and improves the course of disease.
Collapse
Affiliation(s)
- M G Bubnova
- National Medical Research Center for Therapy and Preventive Medicine, Moscow
| | - D M Aronov
- National Medical Research Center for Therapy and Preventive Medicine, Moscow
| |
Collapse
|
202
|
Zhou Q, Deng J, Yao J, Song J, Meng D, Zhu Y, Xu M, Liang Y, Xu J, Sluijter JP, Xiao J. Exercise downregulates HIPK2 and HIPK2 inhibition protects against myocardial infarction. EBioMedicine 2021; 74:103713. [PMID: 34837851 PMCID: PMC8626841 DOI: 10.1016/j.ebiom.2021.103713] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 10/11/2021] [Accepted: 11/09/2021] [Indexed: 12/13/2022] Open
Abstract
Background Exercise can protect myocardial infarction (MI) and downregulate cardiac Homeodomain-Interacting Protein Kinase 2 (HIPK2). However, the role of HIPK2 in MI is unclear. Methods HIPK2–/– mice and miR-222–/– rats, HIPK2 inhibitor (PKI1H) and adeno-associated virus serotype 9 (AAV9) carrying miR-222 were applied in the study. Animals were subjected to running, swimming, acute MI or post-MI remodeling. HIPK2 inhibition and P53 activator were used in neonatal rat cardiomyocytes (NRCMs) and human embryonic stem cell-derived cardiomyocytes (hESC-CMs) subjected to oxygen glucose deprivation/reperfusion (OGD/R). Serum miR-222 levels were analyzed in healthy people and MI patients that were survival or readmitted to the hospital and/or died. Findings Cardiac HIPK2 protein levels were reduced by exercise while increased in MI. In vitro, HIPK2 suppression by lentiviral vectors or inhibitor prevented apoptosis induced by OGD/R in NRCMs and hESC-CMs. HIPK2 inhibitor-treated mice and HIPK2–/– mice reduced infarct size after acute MI, and preserved cardiac function in MI remodeling. Mechanistically, protective effect against apoptosis by HIPK2 suppression was reversed by P53 activators. Furthermore, increasing levels of miR-222, targeting HIPK2, protected post-MI cardiac dysfunction, whereas cardiac dysfunction post-MI was aggravated in miR-222–/– rats. Moreover, serum miR-222 levels were significantly reduced in MI patients, as well as in MI patients that were readmitted to the hospital and/or died compared to those not. Interpretation Exercise-induced HIPK2 suppression attenuates cardiomyocytes apoptosis and protects MI by decreasing P-P53. Inhibition of HIPK2 represents a potential novel therapeutic intervention for MI. Funding This work was supported by the grants from National Key Research and Development Project (2018YFE0113500 to JJ Xiao), National Natural Science Foundation of China (82020108002, 81722008, and 81911540486 to JJ Xiao, 81400647 to MJ Xu, 81800265 to YJ Liang), Innovation Program of Shanghai Municipal Education Commission (2017-01-07-00-09-E00042 to JJ Xiao), the grant from Science and Technology Commission of Shanghai Municipality (18410722200 and 17010500100 to JJ Xiao), the “Dawn” Program of Shanghai Education Commission (19SG34 to JJ Xiao), Shanghai Sailing Program (21YF1413200 to QL Zhou). JS is supported by Horizon2020 ERC-2016-COG EVICARE (725229).
Collapse
Affiliation(s)
- Qiulian Zhou
- Shanghai Engineering Research Center of Organ Repair, Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China; Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, Shanghai, 200444, China
| | - Jiali Deng
- Shanghai Engineering Research Center of Organ Repair, Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China
| | - Jianhua Yao
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Jiaxin Song
- Shanghai Engineering Research Center of Organ Repair, Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China
| | - Danni Meng
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, Shanghai, 200444, China
| | - Yujiao Zhu
- Shanghai Engineering Research Center of Organ Repair, Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China
| | - Minjun Xu
- Shanghai Engineering Research Center of Organ Repair, Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China
| | - Yajun Liang
- Shanghai Engineering Research Center of Organ Repair, Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China
| | - Jiahong Xu
- Department of Cardiology, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Joost Pg Sluijter
- Department of Cardiology, Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht, 3508GA, the Netherlands; UMC Utrecht Regenerative Medicine Center, University Medical Center, Utrecht University, Utrecht, 3508GA, the Netherlands
| | - Junjie Xiao
- Shanghai Engineering Research Center of Organ Repair, Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China; Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, Shanghai, 200444, China.
| |
Collapse
|
203
|
Network Pharmacology and Molecular Docking-Based Analysis on Bioactive Anticoronary Heart Disease Compounds in Trichosanthes kirilowii Maxim and Bulbus allii Macrostemi. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:6704798. [PMID: 34824593 PMCID: PMC8610662 DOI: 10.1155/2021/6704798] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 11/05/2021] [Indexed: 11/17/2022]
Abstract
Trichosanthes kirilowii Maxim. and Bulbus allii Macrostemi are the components of Gualou Xiebai decoction (GLXB), a commonly used herbal combination for the treatment of coronary heart disease (CHD) in traditional Chinese medicine. Although GLXB is associated with a good clinical effect, its active compounds and mechanism of action remain unclear, which limits its clinical application and the development of novel drugs. In this study, we explored key compounds, targets, and mechanisms of action for GLXB in the treatment of CHD using the network pharmacology approach. We identified 18 compounds and 21 action targets via database screening. Enrichment analysis indicated that the effects of GLXB in patients with CHD are primarily associated with the regulation of signalling pathways for tumour necrosis factor, nuclear factor-kappa B, hypoxia-inducible factor-1, arachidonic acid metabolism, and insulin resistance. GLXB thus exerts anti-inflammatory, antihypoxic, and antiagglutinating effects; regulates lipid metabolism; and combats insulin resistance in CHD via these pathways, respectively. After reverse targeting, we observed that the main active compounds of GLXB in the treatment of CHD were quercetin, naringenin, β-sitosterol, ethyl linolenate, ethyl linoleate, and prostaglandin B1. To explore the potential of these compounds in the treatment of CHD, we verified the affinity of the compounds and targets via molecular docking analysis. Our study provides a bridge for the transformation of natural herbs and molecular compounds into novel drug therapies for CHD.
Collapse
|
204
|
Xu D, Li L, Tang L, Guo M, Yang J. Perfluorooctane sulfonate induces heart toxicity involving cardiac apoptosis and inflammation in rats. Exp Ther Med 2021; 23:14. [PMID: 34815766 PMCID: PMC8593915 DOI: 10.3892/etm.2021.10936] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 06/11/2021] [Indexed: 02/07/2023] Open
Abstract
Perfluorooctane sulfonate (PFOS) is a persistent pollutant that exerts toxicity and induces cardiogenesis in humans and animals. Yet, the effect of PFOS exposure on cardiac toxicity in adult rats has, to our knowledge, not been reported and the mechanism still remains unknown. The present study aimed to investigate the toxicity of PFOS on rat hearts and any associated mechanisms. Rats were exposed to 0 (control), 1 and 10 mg/kg PFOS every other day for 14 days. Body weight and heart weight were recorded. The serum levels of lactic dehydrogenase (LDH), creatine kinase (CK), creatine kinase-isoenzyme-MB (CK-MB) and cardiac troponin-T (cTn-T) in heart tissues were measured using biochemical assays. TUNEL staining and western blotting were applied to analyze levels of apoptosis in rat hearts. Pathological assessment and immunohistochemistry analysis of heart tissues were used to evaluate the levels of PFOS-induced cardiotoxicity and inflammatory infiltration. PFOS exposure at the dosage of 10 mg/kg significantly increased the percentage of heart to body weight; however, it did not alter the body weight. At 10 mg/kg, PFOS significantly increased expression levels of myocardial injury markers, such as cTn-T, LDH, CK and CK-MB, while 1 mg/kg PFOS upregulated the expression level of cTn-T in rats. Notably, cardiac fibrosis and myocardiac hypertrophy appeared in the 10 mg/kg PFOS group. In addition, TUNEL-positive cells were significantly increased by exposure to 10 mg/kg PFOS in rat heart tissues. The protein expressions profiles of p53 and Bax were also significantly upregulated in the 10 mg/kg PFOS group. Inflammatory infiltration, detected by anaylzing expression levels of IL-1β and TNF-α, was significantly raised by 10 mg/kg PFOS exposure. In conclusion, these results demonstrated that 10 mg/kg PFOS-induced cardiac toxicity in rats, which was associated with an increase in apoptosis and the expression of proinflammatory cytokines.
Collapse
Affiliation(s)
- Dongmin Xu
- Department of Pharmacy, Chun'an First People's Hospital, Zhejiang Province People's Hospital Chun'an Branch, Hangzhou, Zhejiang 311700, P.R. China
| | - Li Li
- Department of Pharmacy, Chun'an First People's Hospital, Zhejiang Province People's Hospital Chun'an Branch, Hangzhou, Zhejiang 311700, P.R. China
| | - Leilei Tang
- Department of Pharmacy, Zhejiang Xiaoshan Hospital, Hangzhou, Zhejiang 311200, P.R. China
| | - Ming Guo
- Department of Cardiology, Zhejiang Xiaoshan Hospital, Hangzhou, Zhejiang 311200, P.R. China
| | - Jie Yang
- Department of Cardiology, Zhejiang Xiaoshan Hospital, Hangzhou, Zhejiang 311200, P.R. China
| |
Collapse
|
205
|
Zhao Y, Wang P, Chen Z, Li M, Zhang D, Yang L, Li H. Research Progress of Electrical Stimulation in Ischemic Heart Disease. Front Cardiovasc Med 2021; 8:761877. [PMID: 34805318 PMCID: PMC8595213 DOI: 10.3389/fcvm.2021.761877] [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: 08/20/2021] [Accepted: 10/08/2021] [Indexed: 11/13/2022] Open
Abstract
Ischemic heart disease (IHD) is a considerable health burden worldwide with high mortality and morbidity. Treatments for IHD are mainly focused on decreasing oxygen demand or increasing myocardial oxygen supply, including pharmacological, interventional, and surgical treatment, but there are also some limitations. Therefore, it is important to find a simple, effective, and economical treatment. As non-invasive and safe physiotherapy, electrical stimulation (ES) has a promising application in the treatment of IHD. Current studies suggest that ES can affect the occurrence and development of IHD by promoting angiogenesis, regulating autophagy and apoptosis, inhibiting the inflammatory response and oxidative stress. In this review, we focus predominantly on the mechanism of ES and the current progress of ES therapy in IHD, furthermore, give a brief introduction to the forms of ES in clinical application.
Collapse
Affiliation(s)
- Ying Zhao
- Department of Pathophysiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Pengyu Wang
- Department of Pathophysiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Zhe Chen
- Department of Infectious Diseases, Beidahuang Group General Hospital, Harbin, China
| | - Manman Li
- Department of Pathophysiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Dengfeng Zhang
- Department of Pathophysiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Liming Yang
- Department of Pathophysiology, Harbin Medical University-Daqing, Daqing, China
| | - Hong Li
- Department of Pathophysiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| |
Collapse
|
206
|
Zhou J, Dong Y, Cai X, Yang H, Guo T. Identification and Validation of Autophagy-Related Genes as Potential Biomarkers and Therapeutic Targets in Atrial Fibrillation. Int J Gen Med 2021; 14:7783-7796. [PMID: 34785936 PMCID: PMC8580288 DOI: 10.2147/ijgm.s337855] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 10/27/2021] [Indexed: 12/11/2022] Open
Abstract
Background Autophagy is an evolutionary conserved important process for the turnover of intracellular substances in eukaryotes and is closely related to the development of atrial fibrillation (AF). The aim of this study is to identify and validate potential autophagy-related genes (ARGs) of AF through bioinformatics analysis and experimental validation. Methods We downloaded two data sets from the Gene Expression Omnibus (GEO) database, GSE14975 and GSE31821. After merging the data of the two microarrays, adjusting the batch effect, and integrating the differentially expressed genes (DEGs) with ARGs to obtain differentially expressed autophagy-related genes (DEARGs). Functional and pathway enrichment analyses were carried out based on Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG). Use the STRING database to construct a protein–protein interaction (PPI) network. Finally, mRNA expression levels of DEARGs were validated in right atrial tissue samples from AF patients and non-AF controls by qRT-PCR. Results Through bioinformatics analysis, we finally identified 11 DEARGs (CDKN1A, CXCR4, DIRAS3, HSP90AB1, ITGA3, PRKCD, TP53INP2, DAPK2, IFNG, PTK6, and TNFSF10) in AF using [log2 (fold change)] > 0.5 and P < 0.05. In the pathway enrichment analysis, the most significantly enriched pathway was the autophagy pathway. The results of validation showed that the expression levels of CXCR4, DAPK2, and TNFSF10 corroborating with our computational findings, and the results were statistically significant (P<0.05). Conclusion Our study demonstrates that these 11 potential crucial ARGs, especially CXCR4, DAPK2, and TNFSF10, may be potential biomarkers and therapeutic targets in AF, which will help the personalized treatment of AF patients.
Collapse
Affiliation(s)
- Jiao Zhou
- Department of Cardiology, Affiliated Cardiovascular Hospital of Kunming Medical University, Kunming, Yunnan, People's Republic of China.,Department of Cardiology, Fuwai Yunnan Cardiovascular Hospital, Kunming, Yunnan, People's Republic of China
| | - Yunlong Dong
- Department of Gastroenterology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, People's Republic of China
| | - Xiang Cai
- Department of Cardiology, Affiliated Cardiovascular Hospital of Kunming Medical University, Kunming, Yunnan, People's Republic of China.,Department of Cardiology, Fuwai Yunnan Cardiovascular Hospital, Kunming, Yunnan, People's Republic of China
| | - Hongbo Yang
- Department of Cardiology, Affiliated Cardiovascular Hospital of Kunming Medical University, Kunming, Yunnan, People's Republic of China.,Department of Cardiology, Fuwai Yunnan Cardiovascular Hospital, Kunming, Yunnan, People's Republic of China
| | - Tao Guo
- Department of Cardiology, Affiliated Cardiovascular Hospital of Kunming Medical University, Kunming, Yunnan, People's Republic of China.,Department of Cardiology, Fuwai Yunnan Cardiovascular Hospital, Kunming, Yunnan, People's Republic of China
| |
Collapse
|
207
|
Chen X, Xie Q, Zhu Y, Xu J, Lin G, Liu S, Su Z, Lai X, Li Q, Xie J, Yang X. Cardio-protective effect of tetrahydrocurcumin, the primary hydrogenated metabolite of curcumin in vivo and in vitro: Induction of apoptosis and autophagy via PI3K/AKT/mTOR pathways. Eur J Pharmacol 2021; 911:174495. [PMID: 34555398 DOI: 10.1016/j.ejphar.2021.174495] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 08/14/2021] [Accepted: 09/06/2021] [Indexed: 12/14/2022]
Abstract
Tetrahydrocurcumin (THC) is an essential metabolite of curcumin, a major active component of the Curcuma species, which have been used traditionally for the treatment of cardiovascular diseases. The PI3K/AKT/mTOR signaling pathways serve a vital role during myocardial ischemia-reperfusion (MI/R) injury. The aim of the present study was to investigate the cardioprotective potential and mechanism of THC. In the in vivo study, an animal model of MI/R was induced by coronary occlusion. Results indicated that THC (50 mg/kg/day) protected the rat hearts from MI/R-induced heart failure by increasing ejection fraction (EF) and fractional shortening (FS) and decreasing left ventricular end systolic diameter (LVESD) and left ventricular end systolic volume (LVESV). THC also reduced myocardial infarct size and apoptosis. Furthermore, H9c2 cells were incubated with THC (20 μM) to explore its potential effect following exposure to hypoxia and reoxygenation (H/R). THC post-treatment significantly augmented cell viability and prevented lactate dehydrogenase (LDH) release after H/R exposure. THC effectively improved antioxidant activity by increasing SOD and CAT activities and decreasing MDA level. THC also enhanced mitochondrial membrane potential, inhibited apoptotic cell death, diminished the Bax/Bcl-2 ratio and cleaved caspase-3 level relative to the H/R model. In addition, THC effectively decreased Beclin1 expression and LC3 II/LC3 I ratio, but increased p62 expression, compared with the H/R model group, and decreased the formation of H/R-induced autophagosomes and autolysosomes. Furthermore, THC promoted the phosphorylation of PI3K/AKT/mTOR and induced the expression of hypoxia-inducible factor 1α (HIF-1α) after H/R. However, these effects on H9c2 cells were notably abolished by the PI3K inhibitor LY294002 and mTOR inhibitor rapamycin. In conclusion, THC effectively inhibited H/R-induced autophagy and apoptosis via, at least partially, activating the PI3K/AKT/mTOR pathways. THC might have the potential to be further developed into a potential candidate for the treatment of MI/R injury.
Collapse
Affiliation(s)
- Xiaoying Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, PR China
| | - Qingfeng Xie
- Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, PR China; The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, PR China
| | - Ying Zhu
- The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, PR China
| | - Jiamin Xu
- The Second School of Medicine, Guangzhou University of Chinese Medicine, Guangzhou, PR China
| | - Guoshu Lin
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, PR China
| | - Shujun Liu
- The Second School of Medicine, Guangzhou University of Chinese Medicine, Guangzhou, PR China
| | - Ziren Su
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, PR China; Dongguan Institute of Guangzhou University of Chinese Medicine, Dongguan, PR China
| | - Xiaoping Lai
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, PR China; Dongguan Institute of Guangzhou University of Chinese Medicine, Dongguan, PR China.
| | - Qian Li
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, PR China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, PR China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou, PR China
| | - Jianhui Xie
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, PR China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, PR China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou, PR China
| | - Xiaobo Yang
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, PR China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, PR China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou, PR China.
| |
Collapse
|
208
|
Detection of Herbal Combinations and Pharmacological Mechanisms of Clinical Prescriptions for Coronary Heart Disease Using Data Mining and Network Pharmacology. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:9234984. [PMID: 34725557 PMCID: PMC8557045 DOI: 10.1155/2021/9234984] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 09/24/2021] [Accepted: 09/27/2021] [Indexed: 11/24/2022]
Abstract
Though widely used in the treatment of coronary heart disease (CHD), the mechanism of traditional Chinese medicine (TCM) is still unclear because of its complex prescription rules. This study prospectively collected 715 prescriptions of TCM for the treatment of CHD. The characteristics of TCM in prescriptions were described and analyzed, and the rules of prescriptions were analyzed by using association rules. Frequency statistics showed that the high-frequency herbs with a frequency of more than 60% were Gan-cao, Huang-qi, Dang-gui, Chuan-xiong, Yan-hu-suo, and San-qi. The high-frequency herb combinations were summarized by using association rules. By using the method of the “Top N groups” to excavate the empirical prescriptions, the basic prescriptions for treating CHD were summarized. We named the intersection herbs of the basic prescriptions and the high frequency herbs as the core herbal prescription. To explore the possible mechanisms underlying the anti-CHD effect of the core herbal prescription, the bioactive components of core herbal prescription and their targets were screened out by using network pharmacology. Molecular docking was performed between the bioactive components and core targets. A total of 28 potential active ingredients and 5 core targets were identified for the treatment of CHD with core herbal prescription. The enrichment analysis results indicated that the mechanism of action mainly involved neuroactive ligand-receptor interaction and calcium signaling pathway. The commonly used herbal pairs for CHD with qi deficiency and blood stasis syndrome were Huang-qi and Dang-gui. The mechanism of action of common herbal pairs was also studied by network pharmacology. This study summarized the prescription rule of TCM in the treatment of CHD and may provide a new idea for the treatment of CHD.
Collapse
|
209
|
Sun M, Ye Y, Huang Y, Yin W, Yu Z, Wang S. Salvianolic acid B improves autophagic dysfunction and decreases the apoptosis of cholesterol crystal‑induced macrophages via inhibiting the Akt/mTOR signaling pathway. Mol Med Rep 2021; 24:763. [PMID: 34490483 PMCID: PMC8430306 DOI: 10.3892/mmr.2021.12403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 05/07/2021] [Indexed: 11/26/2022] Open
Abstract
Progressive macrophage dysfunction and apoptosis are some of the major events that occur during atherogenesis. To further investigate the intrinsic association between atherosclerosis (AS) and macrophage apoptosis and autophagy, cholesterol crystals (CHCs) were used to stimulate RAW264.7 macrophages to establish a macrophage model of advanced AS. Cells in the CHC group were treated with salvianolic acid B (Sal B) to evaluate its protective effects and reveal its underlying molecular mechanism. The results demonstrated that treatments with Sal B significantly improved autophagy dysfunction and reduced the apoptotic rate of CHC‑induced macrophages. Furthermore, Sal B significantly attenuated CHC‑induced release of proinflammatory factors (TNF‑α and IL‑6) by macrophages. Treatment of macrophages with a specific inhibitor of autophagy (3‑methyladenine) significantly reversed Sal B‑mediated effects on autophagy, suggesting that Sal B‑induced autophagy may display a protective effect in CHC‑induced macrophages. Furthermore, pretreatment of CHC‑induced macrophages with insulin significantly decreased Sal B‑induced autophagy, indicating that the Akt/mTOR signaling pathway may serve as a critical mediator in regulating Sal B‑mediated cell death. Taken together, the present study demonstrated that Sal B improved autophagic dysfunction and reduced the apoptosis of CHC‑induced macrophages via inhibiting the Akt/mTOR signaling pathway.
Collapse
Affiliation(s)
- Mengqi Sun
- Drug Clinical Trial Institution, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Yun Ye
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Yilan Huang
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Wenxian Yin
- Department of Pharmacy, The Affiliated Hospital of Traditional Chinese Medicine Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Zhaolan Yu
- Department of Nephrology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Shurong Wang
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| |
Collapse
|
210
|
Güler MC, Tanyeli A, Erdoğan DG, Eraslan E, Çomaklı S, Polat E, Doğanay S. Urapidil alleviates ovarian torsion detorsion injury via regulating oxidative stress, apoptosis, autophagia, and inflammation. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2021; 24:935-942. [PMID: 34712424 PMCID: PMC8528257 DOI: 10.22038/ijbms.2021.57196.12736] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 06/22/2021] [Indexed: 01/10/2023]
Abstract
Objective(s): This study aimed to determine anti-inflammatory, antioxidant, and antiapoptotic properties of urapidil (Ura) against ovarian torsion detorsion (T/D) injury in rats. Materials and Methods: 40 female Wistar albino rats were grouped as sham, T/D, T/D+dimethyl sulfoxide (DMSO), T/D+Urapidil (Ura) 0.5 mg/kg (low dose), and T/D+Urapidil (Ura) 5 mg/kg (high dose) groups. In treatment groups, Ura was administered intraperitoneally just before detorsion. Biochemical parameters (TAS, TOS, MDA, MPO, and SOD) and immunohistochemical (IL-1β, TNF-α, NF-κB, LC3B, and Caspase-3) analyzes were performed. Results: In the T/D group, OSI and MPO levels were elevated significantly while TAS values decreased compared with the sham group. A significant difference occurred in the low dose treatment group in TAS and OSI levels compared with the T/D group. In the high dose treatment group, significant elevation in TAS but reduction in OSI and MDA levels were observed compared with the T/D group. Immunohistochemical staining resulted in IL-1β, TNF-α, NF-κB, LC3B, and caspase-3 immunopositivity in the T/D group, while Ura treatment decreased those parameters. Intensive congestion and hemorrhage were observed in the T/D group, but contrary to this, treatment groups had alleviated congestion and hemorrhage. Conclusion: These results suggest that Ura demonstrated protective effects against ovarian T/D injury via anti-oxidative, anti-inflammatory, and anti-apoptotic features.
Collapse
Affiliation(s)
- Mustafa Can Güler
- Department of Physiology, Atatürk University, Faculty of Medicine, Erzurum, Turkey
| | - Ayhan Tanyeli
- Department of Physiology, Atatürk University, Faculty of Medicine, Erzurum, Turkey
| | - Derya Güzel Erdoğan
- Department of Physiology, Sakarya University, Faculty of Medicine, Sakarya, Turkey
| | - Ersen Eraslan
- Department of Physiology, Yozgat Bozok University, Faculty of Medicine, Yozgat, Turkey
| | - Selim Çomaklı
- Department of Pathology, Atatürk University, Veterinary Faculty, Erzurum, Turkey
| | - Elif Polat
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Erzurum Technical University, Erzurum, Turkey
| | - Songül Doğanay
- Department of Physiology, Sakarya University, Faculty of Medicine, Sakarya, Turkey
| |
Collapse
|
211
|
Li FH, Guo SW, Zhan TW, Mo HR, Chen X, Wang H, Wei LL, Feng PF, Wu JG, Li P. Integrating network pharmacology and experimental evidence to decipher the cardioprotective mechanism of Yiqihuoxue decoction in rats after myocardial infarction. JOURNAL OF ETHNOPHARMACOLOGY 2021; 279:114062. [PMID: 33771641 DOI: 10.1016/j.jep.2021.114062] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 03/16/2021] [Accepted: 03/18/2021] [Indexed: 05/15/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE "Qi deficiency and blood stasis" syndrome is one of the most common syndromes treated with Traditional Chinese Medicine among ischemic heart disease (IHD) patients in clinic. As a Chinese herbal formula with the function of tonifying Qi and activating blood, Yiqihuoxue Decoction (YQHX) has been frequently proven to be effective in the clinical treatment of IHD. AIM OF THE STUDY The cardioprotective mechanisms of YQHX in treating ischemic heart disease were investigated, with emphasis on the key targets and pathways. MATERIALS AND METHODS In the present study, the potential targets of compounds identified in YQHX were predicted using PharmMapper, Symmap, and STITCH databases, and a "herb-compound-target" network was constructed using Cytoscape. Subsequently, the GO and KEGG functional enrichment analyses were analyzed using the DAVID database. Furthermore, a protein-protein interaction network was constructed using STRING to obtain the key target information. Besides, we used a myocardial ischemia rat model to investigate the cardioprotective effects of YQHX. Transmission electron microscopy and Western blotting were used to observe apoptotic bodies and confirm protein expressions of key candidate targets, respectively. RESULTS Network pharmacology showed that a total of 141 potential targets were obtained from these databases. The functional analysis results revealed that the targets of YQHX were largely associated with apoptosis, and the PI3K-AKT and MAPK pathways might represent key functional pathways. The hub genes of network include ALB, TP53, AKT1, TNF, VEGFA, EGFR, MAPK1, CASP3, JUN, FN1, MMP9, and MAPK8. In vivo, YQHX significantly improved cardiac function and suppressed apoptosis in ischemic rat myocardium. Furthermore, YQHX could significantly upregulate Nrf2 and HO-1 expression, and inhibit JNK phosphorylation. CONCLUSIONS Based on network pharmacology and experimental evidence, this study proves that the cardioprotective effects and mechanisms of YQHX depend on multi-component, multi-target, and multi-pathway. In particular, YQHX exerts anti-apoptotic effects potentially by regulating the Nrf2/HO-1 and JNK-MAPK pathways.
Collapse
Affiliation(s)
- Fang-He Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, PR China; The 3rd Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Shu-Wen Guo
- Fangshan Hospital, Beijing University of Chinese Medicine, Beijing, 102400, PR China.
| | - Tian-Wei Zhan
- Science and Technology College of Jiangxi University of Traditional Chinese Medicine, Nanchang, 330000, PR China
| | - Han-Rong Mo
- Beijing Friendship Hospital, Capital Medical University, Beijing, 100029, PR China
| | - Xi Chen
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, PR China
| | - Hui Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, PR China
| | - Lu-Lu Wei
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, PR China
| | - Peng-Fei Feng
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, PR China
| | - Jian-Gong Wu
- Department of Traditional Chinese Medicine, Peking University People's Hospital, Beijing, 100029, PR China
| | - Ping Li
- The 3rd Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, 100029, China.
| |
Collapse
|
212
|
Liu P, Huang J, Mei W, Zeng X, Wang C, Wen C, Xu J. Epigallocatechin-3-gallate protects cardiomyocytes from hypoxia-reoxygenation damage via raising autophagy related 4C expression. Bioengineered 2021; 12:9496-9506. [PMID: 34699312 PMCID: PMC8810140 DOI: 10.1080/21655979.2021.1996018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Myocardial ischemia/reperfusion (I/R) injury is a serious issue during the therapy of myocardial infarction. Herein, we explored the beneficial influence of Epigallocatechin-3-gallate (EGCG) on hypoxia/reoxygenation (H/R)-stimulated cardiomyocyte H9c2 cells damage, along with possible internal molecular mechanism related autophagy related 4C (ATG4C). H9c2 cells were subjected to H/R stimulation and/or EGCG treatment. ATG4C mRNA expression was measured via q-PCR assay. ATG4C overexpression plasmid (OE-ATG4C) was transfected to arise ATG4C level. Cell viability, apoptosis, reactive oxygen species (ROS) production, ATP level were tested via CCK-8 assay, Annexin V-FITC/PI staining, DCFH-DA staining and ATP Assay Kit, respectively. Western blotting was performed to test Cleaved-caspase 3, Cleaved-caspase 9, cytochrome C, and LC3B protein levels. H/R stimulation resulted in H9c2 cell viability loss, promoted cell apoptosis, and ROS overproduction, as well as lowered ATP level in cells. EGCG treatment alleviated H/R-resulted H9c2 cell viability loss, cell apoptosis, ROS overproduction, and reduction of ATP level. Moreover, H/R stimulation reduced the ATG4C expression in H9c2 cells, while EGCG raised the ATG4C expression. Overexpression of ATG4C strengthened the beneficial influence of EGCG on H/R-stimulated H9c2 cell viability, apoptosis and ROS production. Besides, ATG4C overexpression weakened the H/R-stimulated H9c2 cell autophagy via reducing LC3B II/I expression. EGCG exerted beneficial influence on H/R-stimulated cardiomyocytes, which protected cardiomyocytes from H/R-stimulated viability loss, apoptosis, and ROS overproduction via enhancing ATG4C expression.
Collapse
Affiliation(s)
- Ping Liu
- Clinical Nursing Teaching and Research Section, The Second Xiangya Hospital, Central South University, Changsha, China.,Department of Pediatric Neurology and Cardiovasology, Children's Medical Center, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jin Huang
- Clinical Nursing Teaching and Research Section, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Wanzhen Mei
- Clinical Nursing Teaching and Research Section, The Second Xiangya Hospital, Central South University, Changsha, China.,Department of Pediatric Neurology and Cardiovasology, Children's Medical Center, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xingfang Zeng
- Clinical Nursing Teaching and Research Section, The Second Xiangya Hospital, Central South University, Changsha, China.,Department of Pediatric Neurology and Cardiovasology, Children's Medical Center, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Cheng Wang
- Department of Pediatric Neurology and Cardiovasology, Children's Medical Center, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Chuan Wen
- Department of Pediatric Hematology and Oncology, Children's Medical Center, the Second Xiangya Hospital, Central South University, Changsha, China
| | - Jing Xu
- Clinical Nursing Teaching and Research Section, The Second Xiangya Hospital, Central South University, Changsha, China.,Department of Pediatric Hematology and Oncology, Children's Medical Center, the Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
213
|
Pai PY, Lin YY, Yu SH, Lin CY, Liou YF, Wu XB, Wong JKS, Huang CY, Lee SD. Angiotensin II receptor blocker irbesartan attenuates sleep apnea-induced cardiac apoptosis and enhances cardiac survival and Sirtuin 1 upregulation. Sleep Breath 2021; 26:1161-1172. [PMID: 34626328 DOI: 10.1007/s11325-021-02499-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 09/17/2021] [Accepted: 09/24/2021] [Indexed: 11/25/2022]
Abstract
BACKGROUND The purpose of this study was to investigate whether or not angiotensin II type 1 receptor blocker irbesartan (ARB) with a partial agonist of PPAR-γ could protect against chronic nocturnal intermittent hypoxia (CIH)-induced cardiac Fas/FasL-mediated to mitochondria-mediated apoptosis. METHODS Sprague-Dawley rats were in a normoxic control group (CON-G), or rats were in a chronic nocturnal intermittent hypoxia group (HP-G, from 3 to 7% oxygen versus 21% oxygen per forty seconds cycle, nocturnally 8 h per day for 1 month), or rats were in a chronic nocturnal intermittent hypoxia group pretreated with ARB (50 mg/kg/day, S.C.) (ARB-HP-G). Echocardiography, H&E staining, TUNEL staining, and Western blotting were measured in the left ventricle. RESULTS Hypoxia-induced SIRT1 degradation, Fas receptors, FADD, active caspase-8 and caspase-3 (Fas/FasL apoptotic pathway) and Bax, tBid, active caspase-9 and -3 (mitochondrial apoptotic pathway) and TUNEL-positive apoptosis were reduced in ARB-HP-G when compared with HP-G. IGF-I, IGF1 receptor, p-PI3k, p-Akt, Bcl2, and Bcl-XL (IGF1/PI3K/AKT pro-survival pathway) were increased in ARB-HP-G compared to HP-G. CONCLUSIONS Our findings suggest that the ARB may prevent cardiac Fas/FasL to mitochondrial apoptotic pathways and enhance cardiac IGF1/PI3K/AKT pro-survival pathway in the sleep apnea model associated with JNK de-activation and SIRT1 upregulation. ARB prevents chronic sleep apnea-enhanced cardiac apoptosis via enhancing survival pathways.
Collapse
Affiliation(s)
- Pei-Ying Pai
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan
- Division of Cardiology, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Yi-Yuan Lin
- Department of Exercise and Health Science, National Taipei University of Nursing and Health Sciences, Taipei, Taiwan
| | - Shao-Hong Yu
- College of Rehabilitation, Shandong University of Traditional Chinese Medicine, Shandong, China
| | - Ching-Yuang Lin
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan
- Clinical Immunology Center, China Medical University Hospital, Taichung, Taiwan
| | - Yi-Fan Liou
- Division of Cardiology, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Xu-Bo Wu
- Department of Rehabilitation, Seventh People's Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - James K S Wong
- Department of Cardiology, Asia University Hospital, Taichung, Taiwan
| | - Chih-Yang Huang
- Graduate Institute of Chinese Medical Science, China Medical University, Taichung, Taiwan
- Department of Biotechnology, Asia University, Taichung, Taiwan
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Shin-Da Lee
- Department of Rehabilitation, Seventh People's Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
- Department of Physical Therapy, Asia University, Taichung, Taiwan.
- Department of Physical Therapy, Graduate Institute of Rehabilitation Science, China Medical University, Taichung, Taichung, 40402, Taiwan.
| |
Collapse
|
214
|
Yang Y, Yeung KF, Liu J. CoMM-S 4: A Collaborative Mixed Model Using Summary-Level eQTL and GWAS Datasets in Transcriptome-Wide Association Studies. Front Genet 2021; 12:704538. [PMID: 34616426 PMCID: PMC8488198 DOI: 10.3389/fgene.2021.704538] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 09/03/2021] [Indexed: 11/13/2022] Open
Abstract
Motivation: Genome-wide association studies (GWAS) have achieved remarkable success in identifying SNP-trait associations in the last decade. However, it is challenging to identify the mechanisms that connect the genetic variants with complex traits as the majority of GWAS associations are in non-coding regions. Methods that integrate genomic and transcriptomic data allow us to investigate how genetic variants may affect a trait through their effect on gene expression. These include CoMM and CoMM-S2, likelihood-ratio-based methods that integrate GWAS and eQTL studies to assess expression-trait association. However, their reliance on individual-level eQTL data render them inapplicable when only summary-level eQTL results, such as those from large-scale eQTL analyses, are available. Result: We develop an efficient probabilistic model, CoMM-S4, to explore the expression-trait association using summary-level eQTL and GWAS datasets. Compared with CoMM-S2, which uses individual-level eQTL data, CoMM-S4 requires only summary-level eQTL data. To test expression-trait association, an efficient variational Bayesian EM algorithm and a likelihood ratio test were constructed. We applied CoMM-S4 to both simulated and real data. The simulation results demonstrate that CoMM-S4 can perform as well as CoMM-S2 and S-PrediXcan, and analyses using GWAS summary statistics from Biobank Japan and eQTL summary statistics from eQTLGen and GTEx suggest novel susceptibility loci for cardiovascular diseases and osteoporosis. Availability and implementation: The developed R package is available at https://github.com/gordonliu810822/CoMM.
Collapse
Affiliation(s)
- Yi Yang
- Centre for Quantitative Medicine, Program in Health Services and Systems Research, Duke-NUS Medical School, Singapore, Singapore
| | - Kar-Fu Yeung
- Centre for Quantitative Medicine, Program in Health Services and Systems Research, Duke-NUS Medical School, Singapore, Singapore
| | - Jin Liu
- Centre for Quantitative Medicine, Program in Health Services and Systems Research, Duke-NUS Medical School, Singapore, Singapore
| |
Collapse
|
215
|
Wang T, Li W, Huang Q, Yuan C, Qu L, Xiao X, Zou W. The Clinical Efficacy and Safety of 11 Commonly Used Treatment Strategies Improving Arrhythmia of CHD in China: A Network Meta-Analysis. Front Pharmacol 2021; 12:741716. [PMID: 34616301 PMCID: PMC8488134 DOI: 10.3389/fphar.2021.741716] [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: 07/21/2021] [Accepted: 09/06/2021] [Indexed: 11/26/2022] Open
Abstract
Purpose: Arrhythmia which as a common complication of CHD, has a high incidence. At present, more and more anti-arrhythmic drugs are used in clinical practice. However, which drug has the best efficacy and high safety is still unknown. Therefore, we decided to use NMA to solve this problem. Method: We searched CNKI, Wanfang database, VIP database, Pubmed, Embase and Cochrane libraries, and collected all RCTs of arrhythmia of CHD, and used RevMan (5.3) and Stata (13.0) to carry out this NMA. The primary outcome indicator of this study is efficiency; the secondary outcome indicator is the incidence of adverse reactions. Result: A total of 134 RCTs, 13,951 patients, and 11 treatment strategies were included in this NMA. The results show that all treatment strategies can effectively improve the arrhythmia of patients. Among them, PMA+AM, AM+AT, AM+WG have higher effective rates, and PMA+AM, WG+ME, SC+ME have better safety. The effectiveness and safety of the treatment strategies which combined TCM and chemical drugs, are significantly better than that of using chemical drugs alone. Conclusion: The treatment strategy of combination of multiple drugs usually has higher efficiency and safety. PMA+AM seems to be the most recommended treatment strategy. In addition, the rational combination of TCM and chemical drugs may provide potential benefit. Systematic Review Registration: https://www.crd.york.ac.uk/PROSPERO/, identifier CRD42021229693.
Collapse
Affiliation(s)
- Tao Wang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wei Li
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qianqian Huang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chuqiao Yuan
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Liping Qu
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaohe Xiao
- The Fifth Medical Center of PLA General Hospital, Beijing, China
| | - Wenjun Zou
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- *Correspondence: Wenjun Zou,
| |
Collapse
|
216
|
Dysregulated Circulating Apoptosis- and Autophagy-Related lncRNAs as Diagnostic Markers in Coronary Artery Disease. BIOMED RESEARCH INTERNATIONAL 2021; 2021:5517786. [PMID: 34513991 PMCID: PMC8426068 DOI: 10.1155/2021/5517786] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 04/03/2021] [Accepted: 08/16/2021] [Indexed: 12/24/2022]
Abstract
Objective Increasing evidence emphasizes the implications of dysregulated apoptosis and autophagy cellular processes in coronary artery disease (CAD). Herein, we aimed to explore apoptosis- and autophagy-related long noncoding RNAs (lncRNAs) in peripheral blood of CAD patients. Methods The mRNA and lncRNA expression profiles were retrieved from the Gene Expression Omnibus (GEO) database. With ∣fold change | >1.5 and adjusted p value < 0.05, differentially expressed apoptosis- and autophagy-related mRNAs were screened between CAD and healthy blood samples. Also, differentially expressed lncRNAs were identified for CAD. Using the psych package, apoptosis- and autophagy-related lncRNAs were defined with Spearson's correlation analysis. Receiver operating characteristic (ROC) curves were conducted for the assessment of the diagnosed efficacy of these apoptosis- and autophagy-related lncRNAs. Results Our results showed that 24 apoptosis- and autophagy-related mRNAs were abnormally expressed in CAD than normal controls. 12 circulating upregulated and 1 downregulated apoptosis- and autophagy-related lncRNAs were identified for CAD. The ROCs confirmed that AC004485.3 (AUC = 0.899), AC004920.3 (AUC = 0.93), AJ006998.2 (AUC = 0.776), H19 (AUC = 0.943), RP5-902P8.10 (AUC = 0.956), RP5-1114G22.2 (AUC = 0.883), RP11-247A12.1 (AUC = 0.885), RP11-288L9.4 (AUC = 0.928), RP11-344B5.2 (AUC = 0.858), RP11-452C8.1 (AUC = 0.929), RP11-565A3.1 (AUC = 0.893), and XXbac-B33L19.4 (AUC = 0.932) exhibited good performance in differentiating CAD from healthy controls. Conclusion Collectively, our findings proposed that circulating apoptosis- and autophagy-related lncRNAs could become underlying diagnostic markers for CAD in clinical practice.
Collapse
|
217
|
Calderon-Dominguez M, Mangas A, Belmonte T, Quezada-Feijoo M, Ramos M, Toro R. Fisiopatología de la miocardiopatía dilatada isquémica a través del microRNA-16-5p. Rev Esp Cardiol 2021. [DOI: 10.1016/j.recesp.2020.08.030] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
218
|
Sun YF, Han CH, Han J, Zhou XY, Hu MH, Shen JL. Two Dy(III) complexes: Crystal structures and treatment activity on cardiovascular disease. J SOLID STATE CHEM 2021. [DOI: 10.1016/j.jssc.2021.122347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
219
|
Chen G, Wang M, Ruan Z, Zhu L, Tang C. Mesenchymal stem cell-derived exosomal miR-143-3p suppresses myocardial ischemia-reperfusion injury by regulating autophagy. Life Sci 2021; 280:119742. [PMID: 34166712 DOI: 10.1016/j.lfs.2021.119742] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 06/08/2021] [Accepted: 06/09/2021] [Indexed: 10/21/2022]
Abstract
AIMS Myocardial ischemia-reperfusion (I/R) injury is considered as a major obstacle of myocardial perfusion to save acute myocardial infarction, and causes a serious threat to human health. An extensive body of evidence has unveiled that mesenchymal stem cells (MSCs) as adult stem cells play a vital role in the field of damaged myocardial regeneration and repair. However, the biological role of MSCs derived-exosomes in the protection of myocardial I/R injury has not been elucidated. MAIN METHODS In this study, we isolated and characterized MSCs from the bone marrow of rats femur and tibia. H9c2 cells were administrated to established the cellular hypoxia-reoxygenation (H/R) model, and co-cultured with MSCs and MSCs-derived exosomes. KEY FINDINGS Functional experiments revealed that MSCs and MSCs-derived exosomes inhibited H/R-induced cell apoptosis and cell autophagy. Interestingly, rapamycin as an activator of autophagy reversed the positive effects of MSCs-derived exosomes, while 3-methyladenine (3-MA) as autophagy inhibitor further promoted the effects of MSCs-derived exosomes, indicating MSCs exerted its function on H/R injury by mediating autophagy. Subsequently, we found that CHK2-Beclin2 pathway participated in H/R-induced autophagy. Mechanistically, miR-143-3p directly targeted CHK2 and negatively regulated CHK2 expression. Moreover, repression of exosomal miR-143-3p promoted H/R-induced autophagy via CHK2-Beclin2 pathway. Consistent with the results of in vitro experiments, in vivo experiments confirmed that exosomal miR-143-3p effectively reduced cell apoptosis by regulating autophagy via CHK2-Beclin2 pathway. SIGNIFICANCE Collectively, our results indicated that MSCs-derived exosomal miR-143-3p might represent a promising option for the treatment of I/R injury.
Collapse
Affiliation(s)
- Gecai Chen
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing Jiangsu Province 210009, China; Department of Cardiology, Taizhou People's Hospital, Taizhou, Jiangsu Province 225300, China.
| | - Meixiang Wang
- Department of Cardiology, Taizhou People's Hospital, Taizhou, Jiangsu Province 225300, China
| | - Zhongbao Ruan
- Department of Cardiology, Taizhou People's Hospital, Taizhou, Jiangsu Province 225300, China
| | - Li Zhu
- Department of Cardiology, Taizhou People's Hospital, Taizhou, Jiangsu Province 225300, China
| | - Chengchun Tang
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing Jiangsu Province 210009, China.
| |
Collapse
|
220
|
Kim Y, Zharkinbekov Z, Sarsenova M, Yeltay G, Saparov A. Recent Advances in Gene Therapy for Cardiac Tissue Regeneration. Int J Mol Sci 2021; 22:9206. [PMID: 34502115 PMCID: PMC8431496 DOI: 10.3390/ijms22179206] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 08/16/2021] [Accepted: 08/16/2021] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular diseases (CVDs) are responsible for enormous socio-economic impact and the highest mortality globally. The standard of care for CVDs, which includes medications and surgical interventions, in most cases, can delay but not prevent the progression of disease. Gene therapy has been considered as a potential therapy to improve the outcomes of CVDs as it targets the molecular mechanisms implicated in heart failure. Cardiac reprogramming, therapeutic angiogenesis using growth factors, antioxidant, and anti-apoptotic therapies are the modalities of cardiac gene therapy that have led to promising results in preclinical studies. Despite the benefits observed in animal studies, the attempts to translate them to humans have been inconsistent so far. Low concentration of the gene product at the target site, incomplete understanding of the molecular pathways of the disease, selected gene delivery method, difference between animal models and humans among others are probable causes of the inconsistent results in clinics. In this review, we discuss the most recent applications of the aforementioned gene therapy strategies to improve cardiac tissue regeneration in preclinical and clinical studies as well as the challenges associated with them. In addition, we consider ongoing gene therapy clinical trials focused on cardiac regeneration in CVDs.
Collapse
Affiliation(s)
| | | | | | | | - Arman Saparov
- Department of Medicine, School of Medicine, Nazarbayev University, Nur-Sultan 010000, Kazakhstan; (Y.K.); (Z.Z.); (M.S.); (G.Y.)
| |
Collapse
|
221
|
Li J, Chen J, Zhang F, Li J, An S, Cheng M, Li J. LncRNA CDKN2B-AS1 hinders the proliferation and facilitates apoptosis of ox-LDL-induced vascular smooth muscle cells via the ceRNA network of CDKN2B-AS1/miR-126-5p/PTPN7. Int J Cardiol 2021; 340:79-87. [PMID: 34384839 DOI: 10.1016/j.ijcard.2021.08.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 07/27/2021] [Accepted: 08/04/2021] [Indexed: 11/19/2022]
Abstract
OBJECTIVE The patterns of lncRNA CDKN2B-AS1 in coronary heart disease (CHD) have been extensively studied. This study investigated the competing endogenous RNA (ceRNA) network of CDKN2B-AS1 in coronary atherosclerosis (CAS). METHODS Microarray analyses were performed to screen out the CHD-related lncRNAs (CDKN2B-AS1) and the downstream microRNAs (miR-126-5p). The expression of CDKN2B-AS1 in serum of patients with CHD and healthy volunteers was detected. Vascular smooth muscle cells (VSMCs) were treated with oxidized low density lipoprotein (ox-LDL) to establish the cell model. Then pcDNA-CDKN2B-AS1 and/or miR-126-5p mimic were transfected into ox-LDL-treated VSMCs to estimate cell proliferation, apoptosis and inflammation. The ceRNA network of CDKN2B-AS1 along with the possible pathway in CHD was testified. RESULTS CDKN2B-AS1 expression was low in patients with CHD and ox-LDL-treated VSMCs. Upon CDKN2B-AS1 overexpression, TNF-α, NF-κB and IL-1β levels in VSMCs were decreased, the proliferation of VSMCs was inhibited and the apoptosis rate was increased. Overexpression of miR-126-5p could reverse these trends. CDKN2B-AS1 as a ceRNA competitively bound to miR-126-5p to upregulate PTPN7. CDKN2B-AS1 inhibited VSMC proliferation and accelerated apoptosis by inhibiting the PI3K-Akt pathway. CONCLUSION LncRNA CDKN2B-AS1 upregulates PTPN7 by absorbing miR-126-5p and inhibits the PI3K-Akt pathway, thus hindering the proliferation and accelerating apoptosis of VSMCs induced by ox-LDL, thus being a therapeutic approach for CAS.
Collapse
Affiliation(s)
- Jie Li
- Department of Cardiac Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang, China
| | - Jia Chen
- Department of Cardiac Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang, China
| | - Fan Zhang
- Department of Cardiac Vascular Surgery, Linfen City Center Hospital, Linfen 041000, Shanxi, China
| | - Jianfeng Li
- Department of Cardiac Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang, China
| | - Shoukuan An
- Department of Cardiac Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang, China
| | - Ming Cheng
- Department of Cardiac Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang, China.
| | - Junquan Li
- Department of Cardiac Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang, China.
| |
Collapse
|
222
|
Liu XW, Lu MK, Zhong HT, Liu JJ, Fu YP. Panax Notoginseng Saponins Protect H9c2 Cells From Hypoxia-reoxygenation Injury Through the Forkhead Box O3a Hypoxia-inducible Factor-1 Alpha Cell Signaling Pathway. J Cardiovasc Pharmacol 2021; 78:e681-e689. [PMID: 34354001 PMCID: PMC8584197 DOI: 10.1097/fjc.0000000000001120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 07/14/2021] [Indexed: 11/26/2022]
Abstract
ABSTRACT Panax notoginseng saponins (PNS) are commonly used in the treatment of cardiovascular diseases. Whether PNS can protect myocardial ischemia-reperfusion injury by regulating the forkhead box O3a hypoxia-inducible factor-1 alpha (FOXO3a/HIF-1α) cell signaling pathway remains unclear. The purpose of this study was to investigate the protective effect of PNS on H9c2 cardiomyocytes through the FOXO3a/HIF-1α cell signaling pathway. Hypoxia and reoxygenation of H9C2 cells were used to mimic MIRI in vitro, and the cells were treated with PNS, 2-methoxyestradiol (2ME2), and LY294002." Cell proliferation, lactate dehydrogenase, and malonaldehyde were used to evaluate the degree of cell injury. The level of reactive oxygen species was detected with a fluorescence microscope. The apoptosis rate was detected by flow cytometry. The expression of autophagy-related proteins and apoptosis-related proteins was detected by western blot assay. PNS could reduce H9c2 hypoxia-reoxygenation injury by promoting autophagy and inhibiting apoptosis through the HIF-1α/FOXO3a cell signaling pathway. Furthermore, the protective effects of PNS were abolished by HIF-1α inhibitor 2ME2 and PI3K/Akt inhibitor LY294002. PNS could reduce H9c2 hypoxia-reoxygenation injury by promoting autophagy and inhibiting apoptosis through the HIF-1α/FOXO3a cell signaling pathway.
Collapse
Affiliation(s)
- Xin-Wen Liu
- Department of Pharmacy, Affiliated Hospital of Shaoxing University, Shaoxing, PR China;
| | - Meng-Kai Lu
- Department of Pharmacy, Affiliated Hospital of Shaoxing University, Shaoxing, PR China;
| | - Hui-Ting Zhong
- Department of Research, Affiliated Hospital of Shaoxing University, Shaoxing, PR China; and
| | - Jing-Jing Liu
- Department of Cardiovascular Medicine, Affiliated Hospital of Shaoxing University, Shaoxing, PR China.
| | - Yong-Ping Fu
- Department of Cardiovascular Medicine, Affiliated Hospital of Shaoxing University, Shaoxing, PR China.
| |
Collapse
|
223
|
Overexpression of miR-1298 attenuates myocardial ischemia-reperfusion injury by targeting PP2A. J Thromb Thrombolysis 2021; 53:136-148. [PMID: 34351558 DOI: 10.1007/s11239-021-02540-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/20/2021] [Indexed: 10/20/2022]
Abstract
Previous studies reported that microRNA-1298 was abnormally expressed in the myocardium of rat hearts after hypoxia/normoxia injury. This study aims to investigate the function and specific mechanism of miR-1298 in myocardial ischemia/reperfusion (IR) injury. Neonatal rat cardiomyocytes (NRCMs) were isolated from neonatal rat hearts and subjected to oxygen/glucose deprivation/reperfusion (OGD/R) to induce I/R injury. The rat model with I/R injury was induced by ligating the proximal left anterior descending artery (LAD). MiR-1298 expression was detected by qRT-PCR. The levels of PP2A, Bcl-2, Bax, and AMPK signaling members (p-AMPK, p-GSK3β) was detected by Western blot. Cell apoptosis was evaluated by TUNEL staining assay and flow cytometry. The infarct size of rat hearts was assessed by TTC staining assay. Premature and mature MiR-1298 were significantly downregulated while PP2A was significantly upregulated during I/R injury both in vitro and in vivo. The prediction of Starbase suggested that PP2A was a potential target of miR-1298. MiR-1298 overexpression significantly reduced cardiomyocyte apoptosis in vitro, and its protective effect was obviously attenuated by PP2A overexpression. Luciferase reporter assay showed that miR-1298 targeted PP2A directly. In addition, miR-1298 overexpression significantly reduced infarct size and cardiomyocyte apoptosis in the hearts of rats received with I/R injury in vivo. Moreover, miR-1298 overexpression significantly elevated the levels of Bcl-2 and AMPK signaling members (p-AMPK, p-GSK3β) while decreased Bax level, and these effects were partially reversed by PP2A overexpression. MiR-1298 participated in myocardial I/R injury by targeting the PP2A/AMPK/GSK3β signaling pathway, suggesting that miR-1298 might be a potential therapeutic target for myocardial I/R injury.
Collapse
|
224
|
Fang Y, Duan C, Chen S, Liu Z, Jiang B, Ai W, Wang L, Xie P, Fang H. Tanshinone‑IIA inhibits myocardial infarct via decreasing of the mitochondrial apoptotic signaling pathway in myocardiocytes. Int J Mol Med 2021; 48:158. [PMID: 34212981 PMCID: PMC8262657 DOI: 10.3892/ijmm.2021.4991] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 05/14/2021] [Indexed: 11/06/2022] Open
Abstract
Myocardial ischemia triggers an inflammatory reaction and oxidative stress that increases apoptosis of myocardiocytes. It has been evidenced that tanshinone‑IIA (Tan‑IIA) protects against heart failure post‑myocardial infarction via inhibition of the apoptotic pathway. The purpose of the present study was to investigate the therapeutic effect of Tan‑IIA in a rat model of myocardial ischemia, and explore the possible mechanism of Tan‑IIA in myocardiocytes. The rat model of myocardial ischemia was established by left anterior descending coronary artery and rats received treatment with either Tan‑IIA (10 mg/kg) or PBS for 20 days continuously. The cardiac function in the experimental rat model was detected using the Sequoia 512 echocardiography system on day 21. The cell viability of myocardiocytes was assessed by CCK‑8 assay. Apoptosis of myocardiocytes and myocardial tissue was evaluated by TUNEL assay. The infarct size of the myocardial ischemia rat was determined through 2,3,5‑triphenyltetrazolium chloride (TTC) and Evan blue double staining assay. The expression levels of apoptotic factors were assessed by immunohistochemistry, western blotting and immunofluorescence. The results demonstrated that Tan‑IIA reduced myocardial infarct size and improved the myocardial function in myocardial ischemia rats. Compared with PBS, Tan‑IIA treatment decreased myocardial tissue apoptosis and the expression levels of caspase‑3, Cyto c and Apaf‑1 in myocardial tissue. Tan‑IIA increased the viability of impaired myocardiocytes, inhibited apoptosis of impaired myocardiocytes and increased Bcl‑2 and Bak expression in myocardiocytes. In addition, Tan‑IIA increased Bim and CHOP, decreased TBARS, ROS and H2O2 production, decreased ATF4 and IRE1α expression, and reduced intracellular calcium and oxidative stress in myocardiocytes. Furthermore, caspase‑3 overexpression blocked Tan‑IIA‑decreased apoptosis of myocardiocytes. In conclusion, the data in the present study indicated that Tan‑IIA improved myocardial infarct and apoptosis via the endoplasmic reticulum stress‑dependent pathway and mitochondrial apoptotic signaling pathway.
Collapse
Affiliation(s)
- Yeqing Fang
- Department of Cardiology, Shenzhen Nanshan People's Hospital, Shenzhen, Guangdong 518000, P.R. China
- Shenzhen Nanshan Medical Group Headquarters, Shenzhen, Guangdong 518052, P.R. China
| | - Chengcheng Duan
- Department of Cardiology, Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Shenzhen, Guangdong 518000, P.R. China
| | - Shaoyuan Chen
- Department of Cardiology, Shenzhen Nanshan People's Hospital, Shenzhen, Guangdong 518000, P.R. China
| | - Zhenguo Liu
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 51027, USA
| | - Bimei Jiang
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 51027, USA
- Department of Burns and Plastic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Wen Ai
- Department of Cardiology, Shenzhen Nanshan People's Hospital, Shenzhen, Guangdong 518000, P.R. China
| | - Lei Wang
- Department of Cardiology, Shenzhen Nanshan People's Hospital, Shenzhen, Guangdong 518000, P.R. China
| | - Peiyi Xie
- Department of Cardiology, Shenzhen Nanshan People's Hospital, Shenzhen, Guangdong 518000, P.R. China
| | - Hongcheng Fang
- Department of Cardiology, Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Shenzhen, Guangdong 518000, P.R. China
| |
Collapse
|
225
|
Xu J, Bian X, Zhao H, Sun Y, Tian Y, Li X, Tian W. Morphine Prevents Ischemia/Reperfusion-Induced Myocardial Mitochondrial Damage by Activating δ-opioid Receptor/EGFR/ROS Pathway. Cardiovasc Drugs Ther 2021; 36:841-857. [PMID: 34279751 DOI: 10.1007/s10557-021-07215-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/07/2021] [Indexed: 10/20/2022]
Abstract
OBJECTIVE The purpose of this study was to determine whether the epidermal growth factor receptor (EGFR), which is a classical receptor tyrosine kinase, is involved in the protective effect of morphine against ischemia/reperfusion (I/R)-induced myocardial mitochondrial damage. METHODS Isolated rats hearts were subjected to global ischemia followed by reperfusion. Cardiac H9c2 cells were exposed to a simulated ischemia solution followed by Tyrode's solution to induce hypoxia/reoxygenation (H/R) injury. Triphenyltetrazolium chloride (TTC) was used to measure infarct size. The mitochondrial morphological and functional changes were determined using transmission election microscopy (TEM), mitochondrial stress assay, and mitochondrial swelling, respectively. Mitochondrial fluorescence indicator JC-1, DCFH-DA, and Mitosox Red were used to determine mitochondrial membrane potential (△Ψm), intracellular reactive oxygen species (ROS) and mitochondrial superoxide. A TUNUL assay kit was used to detect the level of apoptosis. Western blotting analysis was used to measure the expression of proteins. RESULTS Treatment of isolated rat hearts with morphine prevented I/R-induced myocardial mitochondrial injury, which was inhibited by the selective EGFR inhibitor AG1478, suggesting that EGFR is involved in the mitochondrial protective effect of morphine under I/R conditions. In support of this hypothesis, the selective EGFR agonist epidermal growth factor (EGF) reduced mitochondrial morphological and functional damage similarly to morphine. Further study demonstrated that morphine may alleviate I/R-induced cardiac damage by inhibiting autophagy but not apoptosis. Morphine increased protein kinase B (Akt), extracellular regulated protein kinases (ERK) and signal transducer and activator of transcription-3 (STAT-3) phosphorylation, which was inhibited by AG1478, and EGF had similar effects, indicating that morphine may activate Akt, ERK, and STAT-3 via EGFR. Morphine and EGF increased intracellular reactive oxygen species (ROS) generation. This effect of morphine was inhibited by AG1478, indicating that morphine promotes intracellular ROS generation by activating EGFR. However, morphine did not increase ROS generation when cells were transfected with siRNA against EGFR. In addition, EGFR activity was markedly increased by morphine, but the effect of morphine was reversed by naltrindole. These results suggest that morphine may activate EGFR via δ-opioid receptor activation. CONCLUSIONS Morphine may prevent I/R-induced myocardial mitochondrial damage by activating EGFR through δ-opioid receptors, in turn increasing RISK and SAFE pathway activity via intracellular ROS. Moreover, morphine may reduce myocardial injury by regulating autophagy but not apoptosis.
Collapse
Affiliation(s)
- Jingman Xu
- School of Public Health, North China University of Science and Technology, 21 Bohai Avenue, Caofeidian District, Tangshan, 063000, Hebei, China.
| | - Xiyun Bian
- Central Laboratory, The Fifth Central Hospital of Tianjin, 300, Tianjin, ,450, China
| | - Huanhuan Zhao
- Department of Physiology and Pathophysiology, Tianjin Medical University, 300, Tianjin, ,010, China
| | - Yujie Sun
- Department of Neurology, Kailuan Hospital, Tangshan, 063000, Hebei Province, China
| | - Yanyi Tian
- School of Public Health, North China University of Science and Technology, 21 Bohai Avenue, Caofeidian District, Tangshan, 063000, Hebei, China
| | - Xiaodong Li
- School of Public Health, North China University of Science and Technology, 21 Bohai Avenue, Caofeidian District, Tangshan, 063000, Hebei, China
| | - Wei Tian
- School of Public Health, North China University of Science and Technology, 21 Bohai Avenue, Caofeidian District, Tangshan, 063000, Hebei, China.
| |
Collapse
|
226
|
Maiese K. New Challenges and Strategies for Cardiac Disease: Autophagy, mTOR, and AMP-activated Protein Kinase. Curr Neurovasc Res 2021; 17:111-112. [PMID: 32036783 DOI: 10.2174/1567202617999200207153935] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
227
|
He Z, Zeng X, Zhou D, Liu P, Han D, Xu L, Bu T, Wang J, Ke M, Pan X, Du Y, Xue H, Lu D, Luo B. LncRNA Chaer Prevents Cardiomyocyte Apoptosis From Acute Myocardial Infarction Through AMPK Activation. Front Pharmacol 2021; 12:649398. [PMID: 34335241 PMCID: PMC8322763 DOI: 10.3389/fphar.2021.649398] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 03/04/2021] [Indexed: 11/30/2022] Open
Abstract
Long non-coding RNA (lncRNA) is widely reported to be involved in cardiac (patho)physiology. Acute myocardial infarction, in which cardiomyocyte apoptosis plays an important role, is a life-threatening disease. Here, we report the lncRNA Chaer that is anti-apoptotic in cardiomyocytes during Acute myocardial infarction. Importantly, lncRNA Chaer is significantly downregulated in both oxygen-glucose deprivation (oxygen-glucose deprivation)-treated cardiomyocytes in vitro and AMI heart. In vitro, overexpression of lncRNA Chaer with adeno virus reduces cardiomyocyte apoptosis induced by OGD-treated while silencing of lncRNA Chaer increases cardiomyocyte apoptosis instead. In vivo, forced expression of lncRNA Chaer with AAV9 attenuates cardiac apoptosis, reduces infarction area and improves mice heart function in AMI. Interestingly, overexpression of lncRNA Chaer promotes the phosphorylation of AMPK, and AMPK inhibitor Compound C reverses the overexpression of lncRNA Chaer effect of reducing cardiomyocyte apoptosis under OGD-treatment. In summary, we identify the novel ability of lncRNA Chaer in regulating cardiomyocyte apoptosis by promoting phosphorylation of AMPK in AMI.
Collapse
Affiliation(s)
- Zhiyu He
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiaojun Zeng
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Deke Zhou
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Traditional Chinese Medicine Hospital of Gaozhou, Department of Cardiology, Gaozhou, China
| | - Peiying Liu
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Dunzheng Han
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Lingling Xu
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Tong Bu
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jinping Wang
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Mengmeng Ke
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiudi Pan
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yipeng Du
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Hao Xue
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Dongfeng Lu
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Bihui Luo
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
228
|
Jo JH, Park HS, Lee DH, Han JH, Heo KS, Myung CS. Rosuvastatin Inhibits the Apoptosis of Platelet-Derived Growth Factor-Stimulated Vascular Smooth Muscle Cells by Inhibiting p38 via Autophagy. J Pharmacol Exp Ther 2021; 378:10-19. [PMID: 33846234 DOI: 10.1124/jpet.121.000539] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 04/06/2021] [Indexed: 12/13/2022] Open
Abstract
The secretion of platelet-derived growth factors (PDGFs) into vascular smooth muscle cells (VSMCs) induced by specific stimuli, such as oxidized low-density lipoprotein (LDL) cholesterol, initially increases the proliferation and migration of VSMCs, and continuous stimulation leads to VSMC apoptosis, resulting in the formation of atheroma. Autophagy suppresses VSMC apoptosis, and statins can activate autophagy. Thus, this study aimed to investigate the mechanism of the autophagy-mediated vasoprotective activity of rosuvastatin, one of the most potent statins, in VSMCs continuously stimulated with PDGF-BB, a PDGF isoform, at a high concentration (100 ng/ml) to induce phenotypic switching of VSMC. Rosuvastatin inhibited apoptosis in a concentration-dependent manner by reducing cleaved caspase-3 and interleukin-1β (IL-1β) levels and reduced intracellular reactive oxygen species (ROS) levels in PDGF-stimulated VSMCs. It also inhibited PDGF-induced p38 phosphorylation and increased the expression of microtubule-associated protein light chain 3 (LC3) and the conversion of LC3-I to LC3-II in PDGF-stimulated VSMCs. The ability of rosuvastatin to inhibit apoptosis and p38 phosphorylation was suppressed by treatment with 3-methyladenine (an autophagy inhibitor) but promoted by rapamycin (an autophagy activator) treatment. SB203580, a p38 inhibitor, reduced the PDGF-induced increase in intracellular ROS levels and inhibited the formation of cleaved caspase-3, indicating the suppression of apoptosis. In carotid ligation model mice, rosuvastatin decreased the thickness and area of the intima and increased the area of the lumen. In conclusion, our observations suggest that rosuvastatin inhibits p38 phosphorylation through autophagy and subsequently reduces intracellular ROS levels, leading to its vasoprotective activity. SIGNIFICANCE STATEMENT: This study shows the mechanism responsible for the vasoprotective activity of rosuvastatin in vascular smooth muscle cells under prolonged platelet-derived growth factor stimulation. Rosuvastatin inhibits p38 activation through autophagy, thereby suppressing intracellular reactive oxygen species levels, leading to the inhibition of apoptosis and reductions in the intima thickness and area. Overall, these results suggest that rosuvastatin can be used as a novel treatment to manage chronic vascular diseases such as atherosclerosis.
Collapse
Affiliation(s)
- Jun-Hwan Jo
- Department of Pharmacology, Chungnam National University College of Pharmacy, Daejeon, Republic of Korea
| | - Hyun-Soo Park
- Department of Pharmacology, Chungnam National University College of Pharmacy, Daejeon, Republic of Korea
| | - Do-Hyung Lee
- Department of Pharmacology, Chungnam National University College of Pharmacy, Daejeon, Republic of Korea
| | - Joo-Hui Han
- Department of Pharmacology, Chungnam National University College of Pharmacy, Daejeon, Republic of Korea
| | - Kyung-Sun Heo
- Department of Pharmacology, Chungnam National University College of Pharmacy, Daejeon, Republic of Korea
| | - Chang-Seon Myung
- Department of Pharmacology, Chungnam National University College of Pharmacy, Daejeon, Republic of Korea
| |
Collapse
|
229
|
Shi M, Cao X, Zhuang J, Chen X. The cardioprotective effect and mechanism of bioactive glass on myocardial reperfusion injury. Biomed Mater 2021; 16. [PMID: 34049296 DOI: 10.1088/1748-605x/ac067e] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 05/28/2021] [Indexed: 02/06/2023]
Abstract
Myocardial reperfusion treatment for ischemic infarction may cause lethal injury of cardiomyocytes, which is known as ischemia/reperfusion (I/R) injury. As a kind of prospective biomaterial with superior properties, the application of bioactive glasses (BGs) in myocardial tissue engineering have received great interests. In this study, the cardioprotective effect and relevant mechanism of BG on myocardial reperfusion injury were investigatedin vitro. H9c2 cardiomyocytes were pretreated with BG extracts and then cultured in hypoxic environment for 30 min followed by reoxygenation for 1 h. The activity of released lactate dehydrogenase (LDH) and the content of malondialdehyde (MDA) in H9c2 cells were tested by assay kits. Cell viability was analyzed by Live/Dead staining assay and the number of living cells was detected by Cell Counting Kit-8 (CCK-8) assay. The cytoskeletal protein F-actin was stained and observed under inverted fluorescence microscope. Mitochondrial membrane potential (MMP) level, reactive oxygen species (ROS) production and apoptosis ratio were evaluated by fluorescent observation and flow cytometry simultaneously. The gene expressions relevant to apoptosis were detected by quantitative real time polymerase chain reaction (qRT-PCR) analysis. The results showed that BG extracts effectively inhibited hypoxia/reoxygenation (H/R)-induced cell injury by suppressing oxidative stress and mitochondrial permeability transition (MPT) within H9c2 cells. Meanwhile, apoptosis caused by H/R injury was alleviated and three classic apoptotic signaling pathways were proved to be regulated by BG extracts. Further analysis showed that phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) signaling pathway was up-regulated in H/R-induced H9c2 cells by BG extracts, leading to relieved cellular apoptosis. These results indicated that BG might exert cardioprotective effect in reperfusion injury when applied in myocardial tissue regeneration and repair.
Collapse
Affiliation(s)
- Miao Shi
- School of Medicine, South China University of Technology, Guangzhou 510006, People's Republic of China.,National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, People's Republic of China
| | - Xiaodong Cao
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, People's Republic of China
| | - Jian Zhuang
- School of Medicine, South China University of Technology, Guangzhou 510006, People's Republic of China.,Guangdong General Hospital, Guangzhou 510080, People's Republic of China
| | - Xiaofeng Chen
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, People's Republic of China
| |
Collapse
|
230
|
Jin T, Lin J, Gong Y, Bi X, Hu S, Lv Q, Chen J, Li X, Chen J, Zhang W, Wang M, Fu G. iPLA 2β Contributes to ER Stress-Induced Apoptosis during Myocardial Ischemia/Reperfusion Injury. Cells 2021; 10:1446. [PMID: 34207793 PMCID: PMC8227999 DOI: 10.3390/cells10061446] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 06/02/2021] [Accepted: 06/07/2021] [Indexed: 01/09/2023] Open
Abstract
Both calcium-independent phospholipase A2 beta (iPLA2β) and endoplasmic reticulum (ER) stress regulate important pathophysiological processes including inflammation, calcium homeostasis and apoptosis. However, their roles in ischemic heart disease are poorly understood. Here, we show that the expression of iPLA2β is increased during myocardial ischemia/reperfusion (I/R) injury, concomitant with the induction of ER stress and the upregulation of cell death. We further show that the levels of iPLA2β in serum collected from acute myocardial infarction (AMI) patients and in samples collected from both in vivo and in vitro I/R injury models are significantly elevated. Further, iPLA2β knockout mice and siRNA mediated iPLA2β knockdown are employed to evaluate the ER stress and cell apoptosis during I/R injury. Additionally, cell surface protein biotinylation and immunofluorescence assays are used to trace and locate iPLA2β. Our data demonstrate the increase of iPLA2β augments ER stress and enhances cardiomyocyte apoptosis during I/R injury in vitro and in vivo. Inhibition of iPLA2β ameliorates ER stress and decreases cell death. Mechanistically, iPLA2β promotes ER stress and apoptosis by translocating to ER upon myocardial I/R injury. Together, our study suggests iPLA2β contributes to ER stress-induced apoptosis during myocardial I/R injury, which may serve as a potential therapeutic target against ischemic heart disease.
Collapse
Affiliation(s)
- Tingting Jin
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310020, China; (T.J.); (J.L.); (Y.G.); (X.B.); (S.H.); (Q.L.); (X.L.)
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310027, China; (J.C.); (J.C.)
| | - Jun Lin
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310020, China; (T.J.); (J.L.); (Y.G.); (X.B.); (S.H.); (Q.L.); (X.L.)
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310027, China; (J.C.); (J.C.)
| | - Yingchao Gong
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310020, China; (T.J.); (J.L.); (Y.G.); (X.B.); (S.H.); (Q.L.); (X.L.)
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310027, China; (J.C.); (J.C.)
| | - Xukun Bi
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310020, China; (T.J.); (J.L.); (Y.G.); (X.B.); (S.H.); (Q.L.); (X.L.)
| | - Shasha Hu
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310020, China; (T.J.); (J.L.); (Y.G.); (X.B.); (S.H.); (Q.L.); (X.L.)
| | - Qingbo Lv
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310020, China; (T.J.); (J.L.); (Y.G.); (X.B.); (S.H.); (Q.L.); (X.L.)
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310027, China; (J.C.); (J.C.)
| | - Jiaweng Chen
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310027, China; (J.C.); (J.C.)
| | - Xiaoting Li
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310020, China; (T.J.); (J.L.); (Y.G.); (X.B.); (S.H.); (Q.L.); (X.L.)
| | - Jiaqi Chen
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310027, China; (J.C.); (J.C.)
| | - Wenbin Zhang
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310020, China; (T.J.); (J.L.); (Y.G.); (X.B.); (S.H.); (Q.L.); (X.L.)
| | - Meihui Wang
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310027, China; (J.C.); (J.C.)
| | - Guosheng Fu
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310020, China; (T.J.); (J.L.); (Y.G.); (X.B.); (S.H.); (Q.L.); (X.L.)
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310027, China; (J.C.); (J.C.)
| |
Collapse
|
231
|
Curcumin-mediated photodynamic therapy inhibits the phenotypic transformation, migration, and foaming of oxidized low-density lipoprotein-treated vascular smooth muscle cells by promoting autophagy. J Cardiovasc Pharmacol 2021; 78:308-318. [PMID: 34091481 PMCID: PMC8340951 DOI: 10.1097/fjc.0000000000001069] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 05/02/2021] [Indexed: 02/05/2023]
Abstract
Supplemental Digital Content is Available in the Text. Vascular smooth muscle cells (VSMCs) are becoming a hot spot and target of atherosclerosis research. This study aimed to observe the specific effects of curcumin (CUR)-mediated photodynamic therapy (CUR-PDT) on oxidized low-density lipoprotein (ox-LDL)-treated VSMCs and confirm whether these effects are mediated by autophagy. In this study, the mouse aortic smooth muscle cell line and A7r5 cell lines were used for parallel experiments. VSMC viability was evaluated by Cell Counting Kit-8 assay. VSMCs were treated with ox-LDL to establish a model of atherosclerosis in vitro. The autophagy level and the expression of proteins related to phenotypic transformation were detected by western blotting. The migration ability of the cells was detected by using transwell assay. The presence of intracellular lipid droplets was detected by Oil Red O staining. The results showed that VSMCs transformed from the contraction phenotype to the synthetic phenotype when stimulated by ox-LDL, during which autophagy was inhibited. However, CUR-PDT treatment significantly promoted the level of autophagy and inhibited the process of phenotypic transformation induced by ox-LDL. In addition, ox-LDL significantly promoted VSMC migration and increased the number of lipid droplets, whereas CUR-PDT treatment significantly reduced the ox-LDL-induced increase in the migration ability of, and lipid droplet numbers in, VSMCs. When the VSMCs were pretreated with the autophagy inhibitor 3-methyladenine for 24 hours, the effects of CUR-PDT were reversed. Therefore, our study indicated that CUR-PDT can inhibit the phenotypic transformation, migration, and foaming of ox-LDL–treated VSMCs by inducing autophagy.
Collapse
|
232
|
Inoue H, Shiga Y, Norimatsu K, Tashiro K, Futami M, Suematsu Y, Sugihara M, Nishikawa H, Katsuda Y, Miura SI. Associations between High-Density Lipoprotein Functionality and Major Adverse Cardiovascular Events in Patients Who Have Undergone Coronary Computed Tomography Angiography. J Clin Med 2021; 10:jcm10112431. [PMID: 34070835 PMCID: PMC8199292 DOI: 10.3390/jcm10112431] [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: 04/25/2021] [Revised: 05/22/2021] [Accepted: 05/26/2021] [Indexed: 01/02/2023] Open
Abstract
The present study aimed to investigate the associations between high-density lipoprotein (HDL) functionality and major adverse cardiovascular events (MACE) in patients who have undergone coronary computed tomography angiography (CCTA). We performed a prospective cohort study and enrolled 151 patients who underwent CCTA and had a follow-up of up to 5 years. We measured cholesterol efflux capacity (CEC), caspase-3/7 activity and monocyte chemoattractant protein-1 (MCP-1) secretion as bioassays of HDL functionality. The patients were divided into MACE(−) (n = 138) and MACE(+) (n = 13) groups. While there was no significant difference in %CEC, caspase-3/7 activity or MCP-1 secretion between the MACE(−) and MACE(+) groups, total CEC and HDL cholesterol (HDL-C) in the MACE(+) group were significantly lower than those in the MACE(−) group. Total CEC was correlated with HDL-C. A receiver-operating characteristic curve analysis showed that there was no significant difference between the areas under the curves for total CEC and HDL-C. In conclusion, total CEC in addition to HDL-C, but not %CEC, was associated with the presence of MACE. On the other hand, HDL functionality with regard to anti-inflammatory and anti-apoptosis effects was not associated with MACE.
Collapse
Affiliation(s)
- Hiroko Inoue
- Department of Cardiology, Fukuoka University Nishijin Hospital, Fukuoka 814-8522, Japan; (H.I.); (K.N.); (M.F.); (H.N.); (Y.K.)
| | - Yuhei Shiga
- Department of Cardiology, Fukuoka University School of Medicine, Fukuoka 814-0180, Japan; (K.T.); (Y.S.); (M.S.)
- Correspondence: (Y.S.); (S.-i.M.); Tel.: +81-92-801-1011 (Y.S. & S.-i.M.)
| | - Kenji Norimatsu
- Department of Cardiology, Fukuoka University Nishijin Hospital, Fukuoka 814-8522, Japan; (H.I.); (K.N.); (M.F.); (H.N.); (Y.K.)
- Department of Cardiology, Fukuoka University School of Medicine, Fukuoka 814-0180, Japan; (K.T.); (Y.S.); (M.S.)
| | - Kohei Tashiro
- Department of Cardiology, Fukuoka University School of Medicine, Fukuoka 814-0180, Japan; (K.T.); (Y.S.); (M.S.)
| | - Makito Futami
- Department of Cardiology, Fukuoka University Nishijin Hospital, Fukuoka 814-8522, Japan; (H.I.); (K.N.); (M.F.); (H.N.); (Y.K.)
- Department of Cardiology, Fukuoka University School of Medicine, Fukuoka 814-0180, Japan; (K.T.); (Y.S.); (M.S.)
| | - Yasunori Suematsu
- Department of Cardiology, Fukuoka University School of Medicine, Fukuoka 814-0180, Japan; (K.T.); (Y.S.); (M.S.)
| | - Makoto Sugihara
- Department of Cardiology, Fukuoka University School of Medicine, Fukuoka 814-0180, Japan; (K.T.); (Y.S.); (M.S.)
| | - Hiroaki Nishikawa
- Department of Cardiology, Fukuoka University Nishijin Hospital, Fukuoka 814-8522, Japan; (H.I.); (K.N.); (M.F.); (H.N.); (Y.K.)
| | - Yousuke Katsuda
- Department of Cardiology, Fukuoka University Nishijin Hospital, Fukuoka 814-8522, Japan; (H.I.); (K.N.); (M.F.); (H.N.); (Y.K.)
| | - Shin-ichiro Miura
- Department of Cardiology, Fukuoka University Nishijin Hospital, Fukuoka 814-8522, Japan; (H.I.); (K.N.); (M.F.); (H.N.); (Y.K.)
- Department of Cardiology, Fukuoka University School of Medicine, Fukuoka 814-0180, Japan; (K.T.); (Y.S.); (M.S.)
- Correspondence: (Y.S.); (S.-i.M.); Tel.: +81-92-801-1011 (Y.S. & S.-i.M.)
| |
Collapse
|
233
|
Copper Preserves Vasculature Structure and Function by Protecting Endothelial Cells from Apoptosis in Ischemic Myocardium. J Cardiovasc Transl Res 2021; 14:1146-1155. [PMID: 33999373 DOI: 10.1007/s12265-021-10128-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 04/15/2021] [Indexed: 02/08/2023]
Abstract
The present study was undertaken to investigate whether Cu protects vasculatures from ischemic injury in the heart. C57/B6 mice were introduced to myocardial ischemia (MI) by permanent ligation of the left anterior descending (LAD) coronary artery. Two hours post-LAD ligation, mice were intravenously injected with a Cu-albumin (Cu-alb) solution, or saline as control. At 1, 4, or 7 days post-MI, hearts were collected for further analysis. A dramatic decrease in CD31-positive endothelial cells concomitantly with abundant apoptosis, along with obstruction of blood flow, was observed in ischemic myocardium 1 day post-MI. The early Cu-alb treatment protected CD31-positive cells from apoptosis, along with a preservation of micro-vessels and a decrease in infarct size. This early vasculature preservation ensured myocardial blood perfusion and protected cardiac contractile function until 28 days post-MI. This strategy of Cu-alb treatment immediately following MI would help develop a therapeutic approach for acute heart attack patients in a clinical setting.
Collapse
|
234
|
Liu L, Huang S, Xu M, Gong Y, Li D, Wan C, Wu H, Tang Q. Isoquercitrin protects HUVECs against high glucose‑induced apoptosis through regulating p53 proteasomal degradation. Int J Mol Med 2021; 48:122. [PMID: 33982778 PMCID: PMC8121554 DOI: 10.3892/ijmm.2021.4955] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 04/12/2021] [Indexed: 02/06/2023] Open
Abstract
High glucose (HG)-induced endothelial apoptosis serves an important role in the vascular dysfunction associated with diabetes mellitus (DM). It has been reported that isoquercitrin (IQC), a flavonoid glucoside, possesses an anti-DM effect, but the mechanism requires further investigation. The present study investigated the effect of IQC against HG-induced apoptosis in human umbilical vein endothelial cells (HUVECs) and explored its molecular mechanism. HUVECs were treated with 5 or 30 mM glucose for 48 h. Endothelial cell viability was monitored using the Cell Counting Kit-8 assay. Mitochondrial membrane potential was detected by JC-1 staining. Apoptosis was observed by TUNEL staining and flow cytometry. Western blotting was used for the analysis of apoptosis-associated proteins Bax, Bcl-2, cleaved (C)-caspase3, total-caspase3, p53 and phosphorylated p53. Reverse transcription-quantitative PCR was used to analyze the mRNA expression levels of Bax, Bcl-2 and p53. Immunofluorescence staining was utilized to detect the expression levels and distribution of p53 and ubiquitin specific peptidase 10 (USP10) in HUVECs. The results revealed that IQC significantly attenuated HG-induced endothelial apoptosis, as shown by decreased apoptotic cells observed by TUNEL, JC-1 staining and flow cytometry. Moreover, under HG stress, IQC treatment markedly inhibited the increased expression levels of the pro-apoptotic proteins p53, Bax and C-caspase3, and increased the expression levels of the anti-apoptotic protein Bcl-2 in HUVECs. However, the anti-apoptotic effect of IQC against HG was partially blunted by increasing p53 protein levels in vitro. IQC influenced the mRNA expression levels of Bax and Bcl-2 in response to HG, but it did not affect the transcription of p53. Notably, IQC inhibited the HG-induced phosphorylation of p53 at Ser15 and the nuclear transport of USP10, destabilizing p53 and increasing the proteasomal degradation of the p53 protein. The current findings revealed that IQC exerted a protective effect against the HG-induced apoptosis of endothelial cells by regulating the proteasomal degradation of the p53 protein, suggesting that IQC may be used as a novel therapeutic compound to ameliorate DM-induced vascular complications.
Collapse
Affiliation(s)
- Libo Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Sihui Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Man Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Yan Gong
- Pharmacy Department, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Dan Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Chunxia Wan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Haiming Wu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Qizhu Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
235
|
Abu-Halima M, Wagner V, Becker LS, Ayesh BM, Abd El-Rahman M, Fischer U, Meese E, Abdul-Khaliq H. Integrated microRNA and mRNA Expression Profiling Identifies Novel Targets and Networks Associated with Ebstein's Anomaly. Cells 2021; 10:cells10051066. [PMID: 33946378 PMCID: PMC8146150 DOI: 10.3390/cells10051066] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 04/27/2021] [Accepted: 04/28/2021] [Indexed: 02/06/2023] Open
Abstract
Little is known about abundance level changes of circulating microRNAs (miRNAs) and messenger RNAs (mRNA) in patients with Ebstein’s anomaly (EA). Here, we performed an integrated analysis to identify the differentially abundant miRNAs and mRNA targets and to identify the potential therapeutic targets that might be involved in the mechanisms underlying EA. A large panel of human miRNA and mRNA microarrays were conducted to determine the genome-wide expression profiles in the blood of 16 EA patients and 16 age and gender-matched healthy control volunteers (HVs). Differential abundance level of single miRNA and mRNA was validated by Real-Time quantitative PCR (RT-qPCR). Enrichment analyses of altered miRNA and mRNA abundance levels were identified using bioinformatics tools. Altered miRNA and mRNA abundance levels were observed between EA patients and HVs. Among the deregulated miRNAs and mRNAs, 76 miRNAs (49 lower abundance and 27 higher abundance, fold-change of ≥2) and 29 mRNAs (25 higher abundance and 4 lower abundance, fold-change of ≥1.5) were identified in EA patients compared to HVs. Bioinformatics analysis identified 37 pairs of putative miRNA-mRNA interactions. The majority of the correlations were detected between the lower abundance level of miRNA and higher abundance level of mRNA, except for let-7b-5p, which showed a higher abundance level and their target gene, SCRN3, showed a lower abundance level. Pathway enrichment analysis of the deregulated mRNAs identified 35 significant pathways that are mostly involved in signal transduction and cellular interaction pathways. Our findings provide new insights into a potential molecular biomarker(s) for the EA that may guide the development of novel targeting therapies.
Collapse
Affiliation(s)
- Masood Abu-Halima
- Institute of Human Genetics, Saarland University, 66421 Homburg, Germany; (V.W.); (L.S.B.); (U.F.); (E.M.)
- Department of Pediatric Cardiology, Saarland University Medical Center, 66421 Homburg, Germany; (M.A.E.-R.); (H.A.-K.)
- Correspondence:
| | - Viktoria Wagner
- Institute of Human Genetics, Saarland University, 66421 Homburg, Germany; (V.W.); (L.S.B.); (U.F.); (E.M.)
- Center for Clinical Bioinformatics, Saarland University, 66123 Saarbrücken, Germany
| | - Lea Simone Becker
- Institute of Human Genetics, Saarland University, 66421 Homburg, Germany; (V.W.); (L.S.B.); (U.F.); (E.M.)
| | - Basim M. Ayesh
- Department of Laboratory Medical Sciences, Alaqsa University, Gaza 4051, Palestine;
| | - Mohammed Abd El-Rahman
- Department of Pediatric Cardiology, Saarland University Medical Center, 66421 Homburg, Germany; (M.A.E.-R.); (H.A.-K.)
| | - Ulrike Fischer
- Institute of Human Genetics, Saarland University, 66421 Homburg, Germany; (V.W.); (L.S.B.); (U.F.); (E.M.)
| | - Eckart Meese
- Institute of Human Genetics, Saarland University, 66421 Homburg, Germany; (V.W.); (L.S.B.); (U.F.); (E.M.)
| | - Hashim Abdul-Khaliq
- Department of Pediatric Cardiology, Saarland University Medical Center, 66421 Homburg, Germany; (M.A.E.-R.); (H.A.-K.)
| |
Collapse
|
236
|
ZHAI GY, QIE SY, GUO QY, QI Y, ZHOU YJ. sDR5-Fc inhibits macrophage M1 polarization by blocking the glycolysis. J Geriatr Cardiol 2021; 18:271-280. [PMID: 33995506 PMCID: PMC8100429 DOI: 10.11909/j.issn.1671-5411.2021.04.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/01/2023] Open
Abstract
BACKGROUND M1 polarization of macrophages is an important pathological process in myocardial ischemia reperfusion injury, which is the major obstacle for the treatment of acute myocardial infarction. Currently, the strategies and mechanisms of inhibiting M1 polarization are poorly explored. This study aims to investigate the role of soluble death receptor 5-Fc (sDR5-Fc) in regulating M1 polarization of macrophages under extreme conditions and explore the mechanisms from the aspect of glycolysis. METHODS Extreme conditions were induced in RAW264.7 cells. Real-time quantitative polymerase chain reaction and western blot were used to detect the expression of mRNA and proteins, respectively. Cell counting kit-8 was used to investigate the proliferation activity of cells. Expression levels of inflammatory cytokines were determined by enzyme-linked immunosorbent assay. RESULTS We found that sDR5-Fc rescues the proliferation of macrophages under extreme conditions, including nutrition deficiency, excessive peroxide, and ultraviolet irradiation. In addition, administration of sDR5-Fc inhibits the M1 polarization of macrophages induced by lipopolysaccharide (LPS) and interferon-gamma (IFN-γ), as the expression of M1 polarization markers CD86, CXC motif chemokine ligand 10, matrix metalloproteinase 9, and tumor necrosis factor-α, as well as the secretion of inflammatory factors interleukin (IL)-1β and IL-6, were significantly decreased. By further investigation of the mechanisms, the results showed that sDR5-Fc can recover the LPS and IFN-γ induced pH reduction, lactic acid elevation, and increased expression of hexokinase 2 and glucose transporter 1, which were markers of glycolysis in macrophages. CONCLUSIONS sDR5-Fc inhibits the M1 polarization of macrophages by blocking the glycolysis, which provides a new direction for the development of strategies in the treatment of myocardial ischemia reperfusion injury.
Collapse
Affiliation(s)
- Guang-Yao ZHAI
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, Capital Medical University, Beijing, China
| | - Shu-Yan QIE
- Department of Rehabilitation, Beijing Rehabilitation Hospital of Capital Medical University, Beijing, China
| | - Qian-Yun GUO
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, Capital Medical University, Beijing, China
| | - Yue QI
- Department of Epidemiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing, China
| | - Yu-Jie ZHOU
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, Capital Medical University, Beijing, China
| |
Collapse
|
237
|
Wang T, Wang L, Zhang Y, Sun J, Xie Y, Yuan Y, Gu J, Bian J, Liu Z, Zou H. Puerarin Restores Autophagosome-Lysosome Fusion to Alleviate Cadmium-Induced Autophagy Blockade via Restoring the Expression of Rab7 in Hepatocytes. Front Pharmacol 2021; 12:632825. [PMID: 33935722 PMCID: PMC8079953 DOI: 10.3389/fphar.2021.632825] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 03/01/2021] [Indexed: 01/02/2023] Open
Abstract
Autophagic dysfunction is one of the main mechanisms by which the environmental pollutant cadmium (Cd) induces cell injury. Puerarin (Pue, a monomeric Chinese herbal medicine extract) has been reported to alleviate Cd-induced cell injury by regulating autophagy pathways; however, its detailed mechanisms are unclear. In the present study, to investigate the detailed mechanisms by which Pue targets autophagy to alleviate Cd hepatotoxicity, alpha mouse liver 12 (AML12) cells were used to construct a model of Cd-induced hepatocyte injury in vitro. First, the protective effect of Pue on Cd-induced cell injury was confirmed by changes in cell proliferation, cell morphology, and cell ultrastructure. Next, we found that Pue activated autophagy and mitigated Cd-induced autophagy blockade. In this process, the lysosome was further activated and the lysosomal degradation capacity was strengthened. We also found that Pue restored the autophagosome-lysosome fusion and the expression of Rab7 in Cd-exposed hepatocytes. However, the fusion of autophagosomes with lysosomes and autophagic flux were inhibited after knocking down Rab7, and were further inhibited after combined treatment with Cd. In addition, after knocking down Rab7, the protective effects of Pue on restoring autophagosome-lysosome fusion and alleviating autophagy blockade in Cd-exposed cells were inhibited. In conclusion, Pue-mediated alleviation of Cd-induced hepatocyte injury was related to the activation of autophagy and the alleviation of autophagy blockade. Pue also restored the fusion of autophagosomes and lysosomes by restoring the protein expression of Rab7, thereby alleviating Cd-induced autophagy blockade in hepatocytes.
Collapse
Affiliation(s)
- Tao Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, School of Horticulture and Plant Protection, Yangzhou University, Yangzhou, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Li Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, School of Horticulture and Plant Protection, Yangzhou University, Yangzhou, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Yi Zhang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, School of Horticulture and Plant Protection, Yangzhou University, Yangzhou, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Jian Sun
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, School of Horticulture and Plant Protection, Yangzhou University, Yangzhou, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Yilin Xie
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Yan Yuan
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, School of Horticulture and Plant Protection, Yangzhou University, Yangzhou, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Jianhong Gu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, School of Horticulture and Plant Protection, Yangzhou University, Yangzhou, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Jianchun Bian
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, School of Horticulture and Plant Protection, Yangzhou University, Yangzhou, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Zongping Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, School of Horticulture and Plant Protection, Yangzhou University, Yangzhou, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Hui Zou
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, School of Horticulture and Plant Protection, Yangzhou University, Yangzhou, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| |
Collapse
|
238
|
Sun W, Dong S, Lu H, Wang N, Zhao Y, An J, Sun L, Lu D. Beclin-1 overexpression regulates NLRP3 activation by promoting TNFAIP3 in microvascular injury following myocardial reperfusion. Cell Signal 2021; 84:110008. [PMID: 33848581 DOI: 10.1016/j.cellsig.2021.110008] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Revised: 04/04/2021] [Accepted: 04/08/2021] [Indexed: 11/26/2022]
Abstract
Innate immune response contributes significantly to ischemia reperfusion (I/R) injury. Targeting innate immunity seems to be a promising method for protecting the microvascular injury in ST-elevation myocardial infarction (STEMI) patients following myocardial I/R injury (MI/R). NLRP3 inflammasome is a central part of the innate immune system involved in the pathophysiological process of MI/R. However, the mechanisms regulating NLRP3 activation are yet to be clarified. Recently, autophagy has been related to the regulation of NLRP3 activation. Thus, how Beclin-1/Becn1 overexpression influences NLRP3 activation in microvascular endothelial cells (CMECs) after MI/R is yet to be investigated. The present study showed that Becn1 overexpression exhibits a significant increase in NLRP3 and IL-1β in CMEC responses to MI/R. Interestingly, Becn1 overexpression promoted TNFAIP3 expression, which restricted NLRP3 activation in vitro and in vivo. The current study also showed that inflammatory cells (CD68) and B (CDB220) lymphocytes were decreased in transgenic mice with overexpression of Beclin-1 (BECN1-Tg) in the spleen and heart. These findings highlighted Becn1 as a prospective target for treating NLRP3 mediated microvascular injury following MI/R.
Collapse
Affiliation(s)
- Wenjing Sun
- Department of Cardiology, Henan Provincial People's Hospital, Zhengzhou 450000, China
| | - Shujuan Dong
- Department of Cardiology, Henan Provincial People's Hospital, Zhengzhou 450000, China
| | - Hongquan Lu
- Department of Nuclear Medicine, Third People's Hospital of Honghe State, Honghe 661000, China
| | - Nan Wang
- Department of Technology Transfer Center, Kunming Medical University, Kunming 650500, China
| | - Yu Zhao
- Department of Technology Transfer Center, Kunming Medical University, Kunming 650500, China
| | - Jingshuo An
- Department of Technology Transfer Center, Kunming Medical University, Kunming 650500, China
| | - Lin Sun
- Department of Cardiology, the Second Affiliated Hospital of Kunming Medical University, Kunming 650501, China.
| | - Di Lu
- Department of Technology Transfer Center, Kunming Medical University, Kunming 650500, China.
| |
Collapse
|
239
|
Liu Z, Gao Z, Zeng L, Liang Z, Zheng D, Wu X. Nobiletin ameliorates cardiac impairment and alleviates cardiac remodeling after acute myocardial infarction in rats via JNK regulation. Pharmacol Res Perspect 2021; 9:e00728. [PMID: 33660406 PMCID: PMC7931132 DOI: 10.1002/prp2.728] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 01/12/2021] [Indexed: 01/07/2023] Open
Abstract
Nobiletin was found to protect against acute myocardial infarction (AMI)-induced cardiac function decline and myocardial remodeling, although the dose-effect relationship and underlying pathways remained unclear. In the current research, different doses of Nobiletin (7.5, 15 and 30 mg/kg/day) were administered to AMI rat model for 21 days. Survival rate, echocardiography, and histological analysis were assessed in vivo. In addition, MTT assay, flow cytometry, and Western blotting were conducted to explore Nobiletin's cytotoxicity and antiapoptotic effect on H9C2 cells. Mechanistically, the activation of MAPK effectors and p38 in vivo was studied. The results showed medium- and high-dose Nobiletin could significantly improve survival rate and cardiac function and reduce the area of infarction and cardiac fibrosis. Medium dose showed the best protection on cardiac functions, whereas high dose showed the best protective effect on cellular apoptosis and histological changes. JNK activation was significantly inhibited by Nobiletin in vivo, which could help to explain the partial contribution of autophagy to AMI-induced apoptosis and the discrepancy on dose-effect relationships. Together, our study suggested that JNK inhibition plays an important role in Nobiletin-induced antiapoptotic effect in myocardial infarction, and medium-dose Nobiletin demonstrated the strongest effect in vivo.
Collapse
Affiliation(s)
- Zumei Liu
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory DiseaseSchool of Pharmaceutical SciencesThe Fifth Affiliated HospitalGuangzhou Medical UniversityGuangzhouPR China
- Department of Central LaboratoryGuangdong Second Provincial General HospitalGuangzhouGuangdongPR China
| | - Zhimin Gao
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory DiseaseSchool of Pharmaceutical SciencesThe Fifth Affiliated HospitalGuangzhou Medical UniversityGuangzhouPR China
| | - Lihuan Zeng
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory DiseaseSchool of Pharmaceutical SciencesThe Fifth Affiliated HospitalGuangzhou Medical UniversityGuangzhouPR China
| | - Zhenye Liang
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory DiseaseSchool of Pharmaceutical SciencesThe Fifth Affiliated HospitalGuangzhou Medical UniversityGuangzhouPR China
| | - Dechong Zheng
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory DiseaseSchool of Pharmaceutical SciencesThe Fifth Affiliated HospitalGuangzhou Medical UniversityGuangzhouPR China
- State Key Laboratory of Quality Research in Chinese MedicineMacau University of Science and TechnologyMacauPR China
| | - Xiaoqian Wu
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory DiseaseSchool of Pharmaceutical SciencesThe Fifth Affiliated HospitalGuangzhou Medical UniversityGuangzhouPR China
| |
Collapse
|
240
|
Liu A, Jia K, Liang H, Jin Q. Comprehensive analysis of autophagy-related genes and patterns of immune cell infiltration in valvular atrial fibrillation. BMC Cardiovasc Disord 2021; 21:132. [PMID: 33706714 PMCID: PMC7948357 DOI: 10.1186/s12872-021-01939-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 02/28/2021] [Indexed: 12/18/2022] Open
Abstract
Background The development of atrial fibrillation (AF) following valvular heart disease (VHD) remains a common disease and is associated with substantial adverse complications. However, valid molecular diagnostic and therapeutic tools for post-VHD AF have not been fully established. This study was conducted to discover the molecular mechanisms and immune microenvironment underlying AF following VHD. Methods Gene expression profiles of the GSE41177 dataset were assessed to construct a protein–protein interaction network, and then, autophagy-related hub genes were identified. In addition, to determine the functions of immune cell infiltration in valvular AF, we used the CIBERSORT algorithm to estimate the composition of 22 immune cell types in valvular heart disease. Finally, correlation analysis was carried out to identify the relationship between differentially expressed autophagy-related genes (DEARGs) and significant immune cell subpopulations to reveal potential regulatory pathways. Results A total of 153 DEARGs were identified in AF-VHD patients compared with controlled donors. Moreover, we screened the top ten hub nodes with the highest degrees through a network analysis. The ten hub nodes were considered hub genes related to AF genesis and progression. Then, we revealed six significant immune cell subpopulations through the CIBERSORT algorithm. Finally, correlation analysis was performed, and six DEARGs (BECN1, GAPDH, ATG7, MAPK3, BCL2L1, and MYC) and three immune cell subpopulations (T cells CD4 memory resting, T cells follicular helper, and neutrophils) were identified as the most significant potential regulators. Conclusion The DEARGs and immune cells identified in our study may be critical in AF development following VHD and provide potential predictive markers and therapeutic targets for determining a treatment strategy for AF patients. Supplementary Information The online version contains supplementary material available at 10.1186/s12872-021-01939-1.
Collapse
Affiliation(s)
- Ao Liu
- Department of Cardiology, Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197, Shanghai Ruijin Er Road, Shanghai, 200025, China
| | - Kangni Jia
- Department of Cardiology, Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197, Shanghai Ruijin Er Road, Shanghai, 200025, China
| | - Huaibin Liang
- Department of Neurology, Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qi Jin
- Department of Cardiology, Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197, Shanghai Ruijin Er Road, Shanghai, 200025, China.
| |
Collapse
|
241
|
Long X, You G, Wu Q, Zhou Y, Xiao Y, Yu F, Deng S, Mo R, Song F, Huang J, Tian M. HomeoboxC6 affects the apoptosis of human vascular endothelial cells and is involved in atherosclerosis. J Cell Physiol 2021; 236:1913-1925. [PMID: 32740941 DOI: 10.1002/jcp.29974] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 06/23/2020] [Accepted: 07/16/2020] [Indexed: 12/18/2022]
Abstract
Apoptosis of vascular endothelial cells (VECs) is highly important in the occurrence and development of atherosclerosis (AS). HomeboxC6 (HOXC6) is expressed in higher levels in multiple malignant tissues, and it influences the malignant biological behavior of the cancer cells. However, the effects of HOXC6 on AS and the apoptosis of VECs have not been fully elucidated. In this study, we demonstrated that HOXC6 expression was increased in aortic wall of AS rats and peripheral blood monocytes of patients with coronary heart disease. Furthermore, it was uncovered that BAX expression was upregulated, while BCL-2 expression was downregulated in the aortic wall of AS rats. The apoptosis of human VECs (HVECs) cultured normally or treated with oxidized low-density lipoprotein in vitro was decreased after transfection with HOXC6-siRNA. Moreover, the results of Western blot analysis unveiled that the expressions of proapoptotic proteins, such as BAX, caspase-3, cleaved-caspase-3, and caspase-9 were reduced, while the expression of antiapoptotic protein, BCL-2, was elevated. Meanwhile, mRNA and protein expressions of phospholipase C beta (PLCβ) were decreased, the phosphorylation levels of protein kinase C zeta (PKCζ) and nuclear transcription factor-κB-p65 (NF-κBp65) and the membrane translocation of PKCζ were reduced as well. Besides, the expression of interleukin-18 (IL-18) protein was downregulated. However, after overexpression of HOXC6, the opposite trends of the abovementioned indices were observed. Furthermore, the inhibition of apoptosis induced by HOXC6-siRNA was reversed by lysophosphatidylcholine, an activator of PKCζ. Taken together, our results indicated that HOXC6 can promote the apoptosis of HVECs and may be involved in the occurrence and development of AS, which may be partially associated with the activation of PLCβ/PKCζ/NF-κBp65/IL-18 signaling pathway.
Collapse
Affiliation(s)
- Xiangshu Long
- Medical College, Guizhou University, Guiyang, Guizhou, China
- Department of Cardiology, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
- People's Hospital of Guizhou University, Guiyang, Guizhou, China
| | - Ganhua You
- Department of Cardiology, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
- People's Hospital of Guizhou University, Guiyang, Guizhou, China
| | - Qiang Wu
- Department of Cardiology, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
- People's Hospital of Guizhou University, Guiyang, Guizhou, China
| | - Yu Zhou
- Medical College, Guizhou University, Guiyang, Guizhou, China
- Department of Cardiology, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
| | - Yan Xiao
- Department of Cardiology, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
| | - Fuxun Yu
- Department of Research Laboratory Center, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
| | - Shiyan Deng
- Department of Cardiology, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
| | - Rui Mo
- Department of Cardiology, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
| | - Fang Song
- Department of Cardiology, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
| | - Jing Huang
- Department of Cardiology, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
| | - Maobo Tian
- Department of Cardiology, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
| |
Collapse
|
242
|
Men H, Cai H, Cheng Q, Zhou W, Wang X, Huang S, Zheng Y, Cai L. The regulatory roles of p53 in cardiovascular health and disease. Cell Mol Life Sci 2021; 78:2001-2018. [PMID: 33179140 PMCID: PMC11073000 DOI: 10.1007/s00018-020-03694-6] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 10/22/2020] [Accepted: 10/29/2020] [Indexed: 02/06/2023]
Abstract
Cardiovascular disease (CVD) remains the leading cause of mortality globally, so further investigation is required to identify its underlying mechanisms and potential targets for its prevention. The transcription factor p53 functions as a gatekeeper, regulating a myriad of genes to maintain normal cell functions. It has received a great deal of research attention as a tumor suppressor. In the past three decades, evidence has also shown a regulatory role for p53 in the heart. Basal p53 is essential for embryonic cardiac development; it is also necessary to maintain normal heart architecture and physiological function. In pathological cardiovascular circumstances, p53 expression is elevated in both patient samples and animal models. Elevated p53 plays a regulatory role via anti-angiogenesis, pro-programmed cell death, metabolism regulation, and cell cycle arrest regulation. This largely promotes the development of CVDs, particularly cardiac remodeling in the infarcted heart, hypertrophic cardiomyopathy, dilated cardiomyopathy, and diabetic cardiomyopathy. Roles for p53 have also been found in atherosclerosis and chemotherapy-induced cardiotoxicity. However, it has different roles in cardiomyocytes and non-myocytes, even in the same model. In this review, we describe the different effects of p53 in cardiovascular physiological and pathological conditions, in addition to potential CVD therapies targeting p53.
Collapse
Affiliation(s)
- Hongbo Men
- Department of Cardiovascular Disease, First Hospital of Jilin University, Jilin University, Changchun, 130021, China
- Department of Pediatrics, Pediatric Research Institute, University of Louisville, Louisville, KY, 40202, USA
| | - He Cai
- Department of Cardiovascular Disease, First Hospital of Jilin University, Jilin University, Changchun, 130021, China
| | - Quanli Cheng
- Department of Cardiovascular Disease, First Hospital of Jilin University, Jilin University, Changchun, 130021, China
| | - Wenqian Zhou
- Department of Cardiovascular Disease, First Hospital of Jilin University, Jilin University, Changchun, 130021, China
- Department of Pediatrics, Pediatric Research Institute, University of Louisville, Louisville, KY, 40202, USA
| | - Xiang Wang
- Department of Cardiovascular Disease, First Hospital of Jilin University, Jilin University, Changchun, 130021, China
- Department of Pediatrics, Pediatric Research Institute, University of Louisville, Louisville, KY, 40202, USA
| | - Shan Huang
- Department of Cardiovascular Disease, First Hospital of Jilin University, Jilin University, Changchun, 130021, China
- Department of Pediatrics, Pediatric Research Institute, University of Louisville, Louisville, KY, 40202, USA
| | - Yang Zheng
- Department of Cardiovascular Disease, First Hospital of Jilin University, Jilin University, Changchun, 130021, China.
| | - Lu Cai
- Department of Pediatrics, Pediatric Research Institute, University of Louisville, Louisville, KY, 40202, USA.
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, 40202, USA.
| |
Collapse
|
243
|
Li F, Wang J, Song Y, Shen D, Zhao Y, Li C, Fu M, Wang Y, Qi B, Han X, Sun A, Zhou J, Ge J. Qiliqiangxin alleviates Ang II-induced CMECs apoptosis by downregulating autophagy via the ErbB2-AKT-FoxO3a axis. Life Sci 2021; 273:119239. [PMID: 33652033 DOI: 10.1016/j.lfs.2021.119239] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 01/26/2021] [Accepted: 02/10/2021] [Indexed: 12/11/2022]
Abstract
Our previous work revealed the protective effect of Qiliqiangxin (QLQX) on cardiac microvascular endothelial cells (CMECs), but the underlying mechanisms remain unclear. We aimed to investigate whether QLQX exerts its protective effect against high-concentration angiotensin II (Ang II)-induced CMEC apoptosis through the autophagy machinery. CMECs were cultured in high-concentration Ang II (1 μM) medium in the presence or absence of QLQX for 48 h. We found that QLQX obviously inhibited Ang II-triggered autophagosome synthesis and apoptosis in cultured CMECs. QLQX-mediated protection against Ang II-induced CMEC apoptosis was reversed by the autophagy activator rapamycin. Specifically, deletion of ATG7 in cultured CMECs indicated a detrimental role of autophagy in Ang II-induced CMEC apoptosis. QLQX reversed Ang II-mediated ErbB2 phosphorylation impairment. Furthermore, inhibition of ErbB2 phosphorylation with lapatinib in CMECs revealed that QLQX-induced downregulation of Ang II-activated autophagy and apoptosis was ErbB2 phosphorylation-dependent via the AKT-FoxO3a axis. Activation of ErbB2 phosphorylation by Neuregulin-1β achieved a similar CMEC-protective effect as QLQX in high-concentration Ang II medium, and this effect was also abolished by autophagy activation. These results show that the CMEC-protective effect of QLQX under high-concentration Ang II conditions could be partly attributable to QLQX-mediated ErbB2 phosphorylation-dependent downregulation of autophagy via the AKT-FoxO3a axis.
Collapse
Affiliation(s)
- Fuhai Li
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
| | - Jingfeng Wang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
| | - Yu Song
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
| | - Dongli Shen
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
| | - Yongchao Zhao
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
| | - Chaofu Li
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
| | - Mingqiang Fu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
| | - Yanyan Wang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
| | - Baozheng Qi
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
| | - Xueting Han
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
| | - Aijun Sun
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China; Institute of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Jingmin Zhou
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China.
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China; Institute of Biomedical Sciences, Fudan University, Shanghai 200032, China.
| |
Collapse
|
244
|
Xu J, Shi J, Cai X, Huang S, Li G, Xu Y. [ Fuxinfang improves hypoxia-induced injury of human aortic endothelial cells by regulating c-Fos-NR4A1-p38 pathway]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2021; 41:200-209. [PMID: 33624592 DOI: 10.12122/j.issn.1673-4254.2021.02.06] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To explore the molecular mechanism of Fuxinfang for improving injury of human aortic endothelial progenitor cells (HAECs). OBJECTIVE Serum samples were collected from male SD rats treated with Fuxinfang (n=8) or saline (n= 5). HAECs cultured in normoxia or hypoxic condition (2% O2) were treated with serum from normal rats or with diluted serum (1% and 10%) from rats treated with Fuxinfang. The differentially expressed genes (DEGs) between Fuxinfang-treated and control cells were detected using high-throughput sequencing to screen the target DEGs that participated in arterial endothelial cell injury and underwent changes in response to both hypoxia and Fuxinfang treatment. AmiGo and String databases were used to infer the interactions among the target genes, and the expressions of the genes were analyzed in HAECs with different treatments using enzyme-linked immunosorbent assay (ELISA) and Western blotting. OBJECTIVE HAECs cultured in hypoxia did not show obvious changes in cell morphology or expressions of hypoxia-related factors in response to treatment with 1% or 10% serum from Fuxinfang-treated rats. The results of high-throughput sequencing showed a total of 7134 DEGs (4205 up-regulated and 2929 down-regulated genes) in HAECs in hypoxia model group and 762 DEGs (305 upregulated and 457 down-regulated genes) in Fuxinfang-treated HAECs. Analysis of AmiGo and String databases and the constructed protein-protein interaction network identified c-Fos, NR4A1, and p38MAPK as the target genes. The results of ELISA and Western blotting showed that the expressions of c-Fos, NR4A1, p38MAPK and pp38MAPK increased significantly in cells with hypoxic exposure (P < 0.05); treatment with the serum containing Fuxinfang significantly reduced the expression levels of c-Fos, NR4A1 and p-p38MAPK in hypoxic HAECs in a concentration-dependent manner (P < 0.05). OBJECTIVE The serum from Fuxinfang-treated rats can concentration-dependently inhibit the expressions of the DEGs occurring in hypoxia. Fuxinfang improves hypoxic injuries of HAECs possibly by down-regulating the expression of c-Fos to inhibit NR4A1 expression and suppressing hypoxia-induced p38 phosphorylation.
Collapse
Affiliation(s)
- J Xu
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200021, China
| | - J Shi
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200021, China
| | - X Cai
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200021, China
| | - S Huang
- Shanghai Pudong TCM Hospital Luoshan Branch, Shanghai 200136, China
| | - G Li
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200021, China
| | - Y Xu
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200021, China
| |
Collapse
|
245
|
Qiao S, Zhao WJ, Li HQ, Ao GZ, An JZ, Wang C, Zhang HL. Necrostatin-1 Analog DIMO Exerts Cardioprotective Effect against Ischemia Reperfusion Injury by Suppressing Necroptosis via Autophagic Pathway in Rats. Pharmacology 2021; 106:189-201. [PMID: 33621976 DOI: 10.1159/000510864] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 07/25/2020] [Indexed: 11/19/2022]
Abstract
AIM It has been reported that necrostatin-1 (Nec-1) is a specific necroptosis inhibitor that could attenuate programmed cell death induced by myocardial ischemia/reperfusion (I/R) injury. This study aimed to observe the effect and mechanism of novel Nec-1 analog (Z)-5-(3,5-dimethoxybenzyl)-2-imine-1-methylimidazolin-4-1 (DIMO) on myocardial I/R injury. METHODS Male SD rats underwent I/R injury with or without different doses of DIMO (1, 2, or 4 mg/kg) treatment. Isolated neonatal rat cardiomyocytes were subjected to oxygen-glucose deprivation/reoxygenation (OGD/R) treatment with or without DIMO (0.1, 1, 10, or 100 μM). Myocardial infarction was measured by TTC staining. Cardiomyocyte injury was assessed by lactate dehydrogenase assay (LDH) and flow cytometry. Receptor-interacting protein 1 kinase (RIP1K) and autophagic markers were detected by co-immunoprecipitation and Western blotting analysis. Molecular docking of DIMO into the ATP binding site of RIP1K was performed using GLIDE. RESULTS DIMO at doses of 1 or 2 mg/kg improved myocardial infarct size. However, the DIMO 4 mg/kg dose was ineffective. DIMO at the dose of 0.1 μM decreased LDH leakage and the ratio of PI-positive cells followed by OGD/R treatment. I/R or OGD/R increased RIP1K expression and in its interaction with RIP3K, as well as impaired myocardial autophagic flux evidenced by an increase in LC3-II/I ratio, upregulated P62 and Beclin-1, and activated cathepsin B and L. In contrast, DIMO treatment reduced myocardial cell death and reversed the above mentioned changes in RIP1K and autophagic flux caused by I/R and OGD/R. DIMO binds to RIP1K and inhibits RIP1K expression in a homology modeling and ligand docking. CONCLUSION DIMO exerts cardioprotection against I/R- or OGD/R-induced injury, and its mechanisms may be associated with the reduction in RIP1K activation and restoration impaired autophagic flux.
Collapse
Affiliation(s)
- Shigang Qiao
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Department of Pharmacology and Laboratory of Cerebrovascular Pharmacology, College of Pharmaceutical Science, Soochow University, Suzhou, China
- Institute of Clinical Medicine Research, The Affiliated Suzhou Science & Technology Town Hospital of Nanjing Medical University, Suzhou, China
| | - Wen-Jie Zhao
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Department of Pharmacology and Laboratory of Cerebrovascular Pharmacology, College of Pharmaceutical Science, Soochow University, Suzhou, China
| | - Huan-Qiu Li
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Department of Pharmacology and Laboratory of Cerebrovascular Pharmacology, College of Pharmaceutical Science, Soochow University, Suzhou, China
| | - Gui-Zhen Ao
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Department of Pharmacology and Laboratory of Cerebrovascular Pharmacology, College of Pharmaceutical Science, Soochow University, Suzhou, China
| | - Jian-Zhong An
- Institute of Clinical Medicine Research, The Affiliated Suzhou Science & Technology Town Hospital of Nanjing Medical University, Suzhou, China
| | - Chen Wang
- Institute of Clinical Medicine Research, The Affiliated Suzhou Science & Technology Town Hospital of Nanjing Medical University, Suzhou, China,
- Department of Anesthesiology and Perioperative Medicine, The Affiliated Suzhou Science & Technology Town Hospital of Nanjing Medical University, Suzhou, China,
| | - Hui-Ling Zhang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Department of Pharmacology and Laboratory of Cerebrovascular Pharmacology, College of Pharmaceutical Science, Soochow University, Suzhou, China
| |
Collapse
|
246
|
Tsai KL, Chou WC, Cheng HC, Huang YT, Chang MS, Chan SH. Anti-IL-20 Antibody Protects against Ischemia/Reperfusion-Impaired Myocardial Function through Modulation of Oxidative Injuries, Inflammation and Cardiac Remodeling. Antioxidants (Basel) 2021; 10:antiox10020275. [PMID: 33578994 PMCID: PMC7916786 DOI: 10.3390/antiox10020275] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 01/29/2021] [Accepted: 01/29/2021] [Indexed: 12/14/2022] Open
Abstract
Acute myocardial infarction (AMI) is the most critical event in the disease spectrum of coronary artery disease. To rescue cardiomyocytes in AMI, it is important to restore blood supply as soon as possible to reduce ischemia-induced injury. However, worse damage can occur during the reperfusion phase, called the reperfusion injury. Under ischemia/reperfusion (I/R) injury, elevated oxidative stress plays a critical role in regulation of apoptosis, inflammation and remodeling of myocardium. Our previous study has demonstrated that interleukin (IL)-20 is increased during hypoxia/reoxygenation stimulation and promotes apoptosis in cardiomyocytes. This study was, therefore, designed to investigate whether IL-20 antibody could reduce I/R-induced myocardial dysfunction. Results from this study revealed that IL-20 antibody treatment significantly suppressed I/R-induced nicotinamide adenine dinucleotide phosphate oxidase, oxidative stress, apoptosis, proinflammatory responses, cardiac fibrosis, and expression of cardiac remodeling markers in Sprague-Dawley rats. Plasma B-type natriuretic peptide level was also reduced by IL-20 antibody injection. IL-20 antibody treatment appeared to restore cardiac function under the I/R injury in terms of greater values of ejection fraction and fractional shortening compared to the control group. Two commonly used indicators of cardiac injury, lactate dehydrogenase and creatine kinase-MB, were also lower in the IL-20 antibody injection group. Taken together, our results suggested that IL-20 antibody holds the potential to reduce the I/R-elicited cardiac dysfunction by preventing cardiac remodeling.
Collapse
Affiliation(s)
- Kun-Ling Tsai
- Department of Physical Therapy, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan; (K.-L.T.); (W.-C.C.); (H.-C.C.); (Y.-T.H.)
- Institute of Allied Health Sciences, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
| | - Wan-Ching Chou
- Department of Physical Therapy, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan; (K.-L.T.); (W.-C.C.); (H.-C.C.); (Y.-T.H.)
| | - Hui-Ching Cheng
- Department of Physical Therapy, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan; (K.-L.T.); (W.-C.C.); (H.-C.C.); (Y.-T.H.)
| | - Yu-Ting Huang
- Department of Physical Therapy, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan; (K.-L.T.); (W.-C.C.); (H.-C.C.); (Y.-T.H.)
| | - Ming-Shi Chang
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
- Correspondence: (M.-S.C.); (S.-H.C.)
| | - Shih-Hung Chan
- Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
- Correspondence: (M.-S.C.); (S.-H.C.)
| |
Collapse
|
247
|
Hsu CN, Tain YL. Preventing Developmental Origins of Cardiovascular Disease: Hydrogen Sulfide as a Potential Target? Antioxidants (Basel) 2021; 10:antiox10020247. [PMID: 33562763 PMCID: PMC7914659 DOI: 10.3390/antiox10020247] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 01/26/2021] [Accepted: 02/01/2021] [Indexed: 12/15/2022] Open
Abstract
The cardiovascular system can be programmed by a diversity of early-life insults, leading to cardiovascular disease (CVD) in adulthood. This notion is now termed developmental origins of health and disease (DOHaD). Emerging evidence indicates hydrogen sulfide (H2S), a crucial regulator of cardiovascular homeostasis, plays a pathogenetic role in CVD of developmental origins. Conversely, early H2S-based interventions have proved beneficial in preventing adult-onset CVD in animal studies via reversing programming processes by so-called reprogramming. The focus of this review will first summarize the current knowledge on H2S implicated in cardiovascular programming. This will be followed by supporting evidence for the links between H2S signaling and underlying mechanisms of cardiovascular programming, such as oxidative stress, nitric oxide deficiency, dysregulated nutrient-sensing signals, activation of the renin–angiotensin system, and gut microbiota dysbiosis. It will also provide an overview from animal models regarding how H2S-based reprogramming interventions, such as precursors of H2S and H2S donors, may prevent CVD of developmental origins. A better understanding of cardiovascular programming and recent advances in H2S-based interventions might provide the answers to bring down the global burden of CVD.
Collapse
Affiliation(s)
- Chien-Ning Hsu
- Department of Pharmacy, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan;
- School of Pharmacy, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - You-Lin Tain
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan
- Correspondence: ; Tel.: +886-975-056-995; Fax: +886-7733-8009
| |
Collapse
|
248
|
Tsai CF, Su HH, Chen K, Liao JM, Yao YT, Chen YH, Wang M, Chu YC, Wang YH, Huang SS. Paeonol Protects Against Myocardial Ischemia/Reperfusion-Induced Injury by Mediating Apoptosis and Autophagy Crosstalk. Front Pharmacol 2021; 11:586498. [PMID: 33551799 PMCID: PMC7858273 DOI: 10.3389/fphar.2020.586498] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 11/30/2020] [Indexed: 01/07/2023] Open
Abstract
Many studies have shown that crosstalk exists between apoptosis and autophagy, despite differences in mechanisms between these processes. Paeonol, a major phenolic compound isolated from Moutan Cortex Radicis, the root bark of Paeonia × suffruticosa Andrews (Paeoniaceae), is widely used in traditional Chinese medicine as an antipyretic, analgesic and anti-inflammatory agent. In this study, we investigated the detailed molecular mechanisms of the crosstalk between apoptosis and autophagy underlying the cardioprotective effects of paeonol in rats subjected to myocardial ischemia/reperfusion (I/R) injury. Myocardial I/R injury was induced by occlusion of the left anterior descending coronary artery (LAD) for 1 h followed by 3 h of reperfusion. Paeonol was intravenously administered 15 min before LAD ligation. We found that paeonol significantly improved cardiac function after myocardial I/R injury and significantly decreased myocardial I/R-induced arrhythmia and mortality. Paeonol also significantly decreased myocardial infarction and plasma LDH activity and Troponin-I levels in carotid blood after I/R. Compared with vehicle treatment, paeonol significantly upregulated Bcl-2 protein expression and significantly downregulated the cleaved forms of caspase-8, caspase-9, caspase-3 and PARP protein expression in the I/R injured myocardium. Myocardial I/R-induced autophagy, including the increase of Beclin-1, p62, LC3-I, and LC3-II protein expression in the myocardium was significantly reversed by paeonol treatment. Paeonol also significantly increased the Bcl-2/Bax and Bcl-2/Beclin-1 ratios in the myocardium after I/R injury. The cardioprotective role of paeonol during I/R injury may be due to its mediation of crosstalk between apoptotic and autophagic signaling pathways, which inhibits apoptosis and autophagic cell death.
Collapse
Affiliation(s)
- Chin-Feng Tsai
- Division of Cardiology, Department of Internal Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan,School of Medicine, Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Hsing-Hui Su
- Department of Pharmacology, Chung Shan Medical University, Taichung, Taiwan
| | - Ke‐Min Chen
- Department of Parasitology, Chung Shan Medical University, Taichung, Taiwan
| | - Jiuan-Miaw Liao
- Department of Physiology, Chung Shan Medical University and Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Yi-Ting Yao
- School of Medicine, Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Yi-Hung Chen
- Graduate Institute of Acupuncture Science, China Medical University, Taichung, Taiwan,Research Center for Chinese Medicine and Acupuncture, China Medical University, Taichung, Taiwan,Department of Photonics and Communication Engineering, Asia University, Taichung, Taiwan
| | - Meilin Wang
- Department of Microbiology and Immunology, School of Medicine, Chung-Shan Medical University, Taichung, Taiwan
| | - Ya-Chun Chu
- Department of Anesthesiology, Taipei Veterans General Hospital and National Yang-Ming University, Taipei, Taiwan,*Correspondence: Ya-Chun Chu, ; Yi-Hsin Wang, ; Shiang-Suo Huang,
| | - Yi-Hsin Wang
- School of Medicine, Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan,Department of Pharmacology, Chung Shan Medical University, Taichung, Taiwan,*Correspondence: Ya-Chun Chu, ; Yi-Hsin Wang, ; Shiang-Suo Huang,
| | - Shiang-Suo Huang
- School of Medicine, Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan,Department of Pharmacology, Chung Shan Medical University, Taichung, Taiwan,Department of Pharmacy, Chung Shan Medical University Hospital, Taichung, Taiwan,*Correspondence: Ya-Chun Chu, ; Yi-Hsin Wang, ; Shiang-Suo Huang,
| |
Collapse
|
249
|
Liu Y, Song A, Wu H, Sun Y, Dai M. Paeonol inhibits apoptosis of vascular smooth muscle cells via up-regulation of autophagy by activating class III PI3K/Beclin-1 signaling pathway. Life Sci 2021; 264:118714. [PMID: 33157088 DOI: 10.1016/j.lfs.2020.118714] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 10/30/2020] [Accepted: 11/02/2020] [Indexed: 12/23/2022]
Abstract
AIMS The cross talk between autophagy and apoptosis of vascular smooth muscle cells (VSMCs) plays a vital role in the development of atherosclerosis (AS). Paeonol is isolated from the radix of Cortex Moutan with anti-atherosclerotic and anti-apoptosis effects. However, the mechanisms of paeonol on VSMCs apoptosis are still not fully understood. In this study, we aimed to explore whether paeonol could inhibit VSMCs apoptosis though modulating VSMCs autophagy. MATERIALS AND METHODS The proteins expressions were detected by western blotting. Autophagosomes and apoptoticbody formation in VSMCs was observed by transmission electron microscopy (TEM). VSMCs autophagy was detected by monodansylcadaverine (MDC) staining using fluorescence microscopy, while VSMCs apoptosis was determined by 4',6-diamidino-2-phenylindole (DAPI) and flow cytometry. KEY FINDINGS We found that paeonol could significantly increase LC3II protein level, decrease p62 and cleaved caspase-3 proteins levels in aorta of AS mice and ox-LDL-injured VSMCs. Paeonol could augment the number of autophagosomes and reduce the amount of apoptotic bodies in ox-LDL-injured VSMCs. Moreover, paeonol obviously induced VSMCs autophagy compared to ox-LDL group and remarkably suppressed VSMCs apoptosis. However, the effects of paeonol on VSMCs apoptosis could be reversed obviously by 3-MA, the autophagy inhibitor. Furthermore, paeonol could activate class III PI3K-Beclin-1 pathway significantly. Gene silencing of either class III PI3K or Beclin-1 could reverse the effects of paeonol on VSMCs autophagy and apoptosis. SIGNIFICANCE Based on our results, paeonol could induce VSMCs autophagy by activating class III PI3K/Beclin-1 signaling pathway, thus ultimately inhibiting VSMCs apoptosis.
Collapse
Affiliation(s)
- Yarong Liu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui 230012, China; Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei, Anhui 230038, China; Anhui Key Laboratory for Research and Development of Traditional Chinese Medicine, Hefei, Anhui 230012, China
| | - Aiwei Song
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui 230012, China
| | - Hongfei Wu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui 230012, China; Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei, Anhui 230038, China; Anhui Key Laboratory for Research and Development of Traditional Chinese Medicine, Hefei, Anhui 230012, China
| | - Yin Sun
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui 230012, China
| | - Min Dai
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui 230012, China; Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei, Anhui 230038, China; Anhui Key Laboratory for Research and Development of Traditional Chinese Medicine, Hefei, Anhui 230012, China.
| |
Collapse
|
250
|
Li W. Evaluation of left ventricular diastolic function of patients with coronary heart disease by ultrasound images on bilateral filtering image noise reduction algorithm combined with electrocardiogram. Pak J Med Sci 2021; 37:1699-1704. [PMID: 34712309 PMCID: PMC8520376 DOI: 10.12669/pjms.37.6-wit.4886] [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: 04/08/2021] [Revised: 06/14/2021] [Accepted: 07/07/2021] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE To explore the evaluation of left ventricular diastolic function (LVDF) in patients with coronary heart disease (CHD) using ultrasound images (UI) combined with electrocardiogram (ECG) on bilateral filtering image noise reduction algorithm (BFINRA). METHODS A BFINRA was constructed, and 60 subjects who were investigated were divided into a control group (CG) from June 2019 to November 2019 in Taizhou People's Hospital, a myocardial infarction group (MIG), and an angina pectoris group (APG). The patient's LVDF was examined by two-dimensional electrophoresis (2DE) and real-time three-dimensional echocardiography (RT-3DE) combined with ECG. The results showed BFINRA could improve UI quality. RESULTS Clinical data indicated there were no substantial differences in age, gender, and fasting blood glucose of all subjects. 2DE examination results showed the left ventricular end-diastolic volume (LVEDV), left ventricular end-systolic volume (LVESV), and early diastolic mitral blood flow velocity / early diastolic mitral annulus velocity (E/E') of MIG were much higher than CG (P<0.05), while the left ventricular ejection fraction (LVEF), E / late diastolic mitral blood flow velocity (E/A) and E' peak value were sharply decreased (P<0.05);LVESV and E/E' of APG were increased dramatically (P<0.05), while E peak, E/A and E' peak were decreased greatly. RT-3DE examination results indicated LVEDV and LVESV of MIG were considerably higher than CG (P<0.05), while LVEF and macrophage resistance factor (MRF) were enormously decreased (P<0.05);LVEDV and LVESV of APG were greatly increased (P<0.05). However, LVEF and MRF were not changed significantly (P>0.05). LVEDV had a remarkable difference (P<0.05), but LVESV and LVEF had no obvious differences (P>0.05). The electrocardiogram results illustrated the increase in QT dispersion (QTd) of MIG and APG was statistically significant (P<0.05) compared with CG, while the negative increase of P-wave terminal force in lead V1 (PTFV1) also had a statistical significance (P<0.05). Correlation analysis revealed that MRF and PTFV1 had positive correlation, while MRF and QTd showed a negative correlation. CONCLUSION The combination of UI and ECG could better assess LVDF in CHD patients.
Collapse
Affiliation(s)
- Wen Li
- Wen Li, Master of Medicine. Electrocardiogram Room, Taizhou People’s Hospital, Taizhou, 225300, Jiangsu, China
| |
Collapse
|