1
|
Cai K, Jiang H, Zou Y, Song C, Cao K, Chen S, Wu Y, Zhang Z, Geng D, Zhang N, Liu B, Sun G, Tang M, Li Z, Zhang Y, Sun Y, Zhang Y. Programmed death of cardiomyocytes in cardiovascular disease and new therapeutic approaches. Pharmacol Res 2024; 206:107281. [PMID: 38942341 DOI: 10.1016/j.phrs.2024.107281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 06/21/2024] [Accepted: 06/21/2024] [Indexed: 06/30/2024]
Abstract
Cardiovascular diseases (CVDs) have a complex pathogenesis and pose a major threat to human health. Cardiomyocytes have a low regenerative capacity, and their death is a key factor in the morbidity and mortality of many CVDs. Cardiomyocyte death can be regulated by specific signaling pathways known as programmed cell death (PCD), including apoptosis, necroptosis, autophagy, pyroptosis, and ferroptosis, etc. Abnormalities in PCD can lead to the development of a variety of cardiovascular diseases, and there are also molecular-level interconnections between different PCD pathways under the same cardiovascular disease model. Currently, the link between programmed cell death in cardiomyocytes and cardiovascular disease is not fully understood. This review describes the molecular mechanisms of programmed death and the impact of cardiomyocyte death on cardiovascular disease development. Emphasis is placed on a summary of drugs and potential therapeutic approaches that can be used to treat cardiovascular disease by targeting and blocking programmed cell death in cardiomyocytes.
Collapse
Affiliation(s)
- Kexin Cai
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China
| | - Haoyue Jiang
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China
| | - Yuanming Zou
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China
| | - Chunyu Song
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China
| | - Kexin Cao
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China
| | - Shuxian Chen
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China
| | - Yanjiao Wu
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China
| | - Zhaobo Zhang
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China
| | - Danxi Geng
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China
| | - Naijin Zhang
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China; Institute of health sciences, China medical university, 77 Puhe Road, Shenbei New District, Shenyang, Liaoning 110001, People's Republic of China; Key Laboratory of Reproductive and Genetic Medicine (China Medical University), National Health Commission, 77 Puhe Road, Shenbei New District, Shenyang, Liaoning 110001, People's Republic of China
| | - Bo Liu
- The first hospital of China Medical University, Department of cardiac surgery, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China.
| | - Guozhe Sun
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China.
| | - Man Tang
- Department of clinical pharmacology, College of Pharmacy, China medical university, 77 Puhe Road, Shenbei New District, Shenyang, Liaoning 110001, People's Republic of China.
| | - Zhao Li
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China.
| | - Yixiao Zhang
- Department of Urology Surgery, Shengjing Hospital of China Medical University, No.36 Sanhao Street, Heping District, Shenyang, Liaoning 110004, People's Republic of China.
| | - Yingxian Sun
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China; Institute of health sciences, China medical university, 77 Puhe Road, Shenbei New District, Shenyang, Liaoning 110001, People's Republic of China; Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention, Ministry of Education, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang, Liaoning 110001, People's Republic of China.
| | - Ying Zhang
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China; Institute of health sciences, China medical university, 77 Puhe Road, Shenbei New District, Shenyang, Liaoning 110001, People's Republic of China.
| |
Collapse
|
2
|
Chen Z, Liu Y, Ma R, Zhang M, Wu X, Pen H, Gui F, Liu Y, Xia H, Hu N, Ai B, Xiong J, Xia H, Li W, Ai F. Protective Effect of Long Noncoding RNA OXCT1-AS1 on Doxorubicin-Induced Apoptosis of Human Myocardial Cells by the Competitive Endogenous RNA Pattern. Arq Bras Cardiol 2024; 121:e20230675. [PMID: 38958296 PMCID: PMC11216341 DOI: 10.36660/abc.20230675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 01/26/2024] [Accepted: 03/11/2024] [Indexed: 07/04/2024] Open
Abstract
BACKGROUND The anthracycline chemotherapeutic antibiotic doxorubicin (DOX) can induce cumulative cardiotoxicity and lead to cardiac dysfunction. Long non-coding RNAs (lncRNAs) can function as important regulators in DOX-induced myocardial injury. OBJECTIVE This study aims to investigate the functional role and molecular mechanism of lncRNA OXCT1 antisense RNA 1 (OXCT1-AS1) in DOX-induced myocardial cell injury in vitro. METHODS Human cardiomyocytes (AC16) were stimulated with DOX to induce a myocardial cell injury model. OXCT1-AS1, miR-874-3p, and BDH1 expression in AC16 cells were determined by RT-qPCR. AC16 cell viability was measured by XTT assay. Flow cytometry was employed to assess the apoptosis of AC16 cells. Western blotting was used to evaluate protein levels of apoptosis-related markers. Dual-luciferase reporter assay was conducted to verify the binding ability between miR-874-3p and OXCT1-AS1 and between miR-874-3p and BDH1. The value of p<0.05 indicated statistical significance. RESULTS OXCT1-AS1 expression was decreased in DOX-treated AC16 cells. Overexpression of OXCT1-AS1 reversed the reduction of cell viability and promotion of cell apoptosis caused by DOX. OXCT1-AS1 is competitively bound to miR-874-3p to upregulate BDH1. BDH1 overexpression restored AC16 cell viability and suppressed cell apoptosis under DOX stimulation. Knocking down BDH1 reversed OXCT1-AS1-mediated attenuation of AC16 cell apoptosis under DOX treatment. CONCLUSION LncRNA OXCT1-AS1 protects human myocardial cells AC16 from DOX-induced apoptosis via the miR-874-3p/BDH1 axis.
Collapse
Affiliation(s)
- Zhen Chen
- Huazhong University of Science and TechnologyTongji Medical CollegeThe Central Hospital of WuhanWuhanChinaDepartment of Emergency – The Central Hospital of Wuhan – Tongji Medical College – Huazhong University of Science and Technology, Wuhan – China
| | - Yijue Liu
- Huazhong University of Science and TechnologyTongji Medical CollegeThe Central Hospital of WuhanWuhanChinaDepartment of Emergency – The Central Hospital of Wuhan – Tongji Medical College – Huazhong University of Science and Technology, Wuhan – China
| | - Rui Ma
- Hubei University of MedicineSinopharm Dongfeng General HospitalDepartment of Geriatric MedicineShiyanChinaDepartment of Geriatric Medicine – Sinopharm Dongfeng General Hospital – Hubei University of Medicine, Shiyan – China
| | - Mengli Zhang
- Huazhong University of Science and TechnologyTongji Medical CollegeThe Central Hospital of WuhanWuhanChinaDepartment of Emergency – The Central Hospital of Wuhan – Tongji Medical College – Huazhong University of Science and Technology, Wuhan – China
| | - Xian Wu
- Huazhong University of Science and TechnologyTongji Medical CollegeThe Central Hospital of WuhanWuhanChinaDepartment of Emergency – The Central Hospital of Wuhan – Tongji Medical College – Huazhong University of Science and Technology, Wuhan – China
| | - Huan Pen
- Huazhong University of Science and TechnologyTongji Medical CollegeThe Central Hospital of WuhanWuhanChinaDepartment of Emergency – The Central Hospital of Wuhan – Tongji Medical College – Huazhong University of Science and Technology, Wuhan – China
| | - Feng Gui
- Huazhong University of Science and TechnologyTongji Medical CollegeThe Central Hospital of WuhanWuhanChinaDepartment of Emergency – The Central Hospital of Wuhan – Tongji Medical College – Huazhong University of Science and Technology, Wuhan – China
| | - Yafeng Liu
- Renmin Hospital of Wuhan UniversityDepartment of EmergencyWuhanChinaDepartment of Emergency – Renmin Hospital of Wuhan University, Wuhan – China
| | - Hao Xia
- Renmin Hospital of Wuhan UniversityDepartment of CardiologyWuhanChinaDepartment of Cardiology – Renmin Hospital of Wuhan University, Wuhan – China
| | - Niandan Hu
- Renmin Hospital of Wuhan UniversityDepartment of EmergencyWuhanChinaDepartment of Emergency – Renmin Hospital of Wuhan University, Wuhan – China
| | - Bo Ai
- Renmin Hospital of Wuhan UniversityDepartment of EmergencyWuhanChinaDepartment of Emergency – Renmin Hospital of Wuhan University, Wuhan – China
| | - Jun Xiong
- Renmin Hospital of Wuhan UniversityDepartment of EmergencyWuhanChinaDepartment of Emergency – Renmin Hospital of Wuhan University, Wuhan – China
| | - Hongxia Xia
- Renmin Hospital of Wuhan UniversityDepartment of EmergencyWuhanChinaDepartment of Emergency – Renmin Hospital of Wuhan University, Wuhan – China
| | - Wenqiang Li
- Renmin Hospital of Wuhan UniversityDepartment of EmergencyWuhanChinaDepartment of Emergency – Renmin Hospital of Wuhan University, Wuhan – China
| | - Fen Ai
- Huazhong University of Science and TechnologyTongji Medical CollegeThe Central Hospital of WuhanWuhanChinaDepartment of Emergency – The Central Hospital of Wuhan – Tongji Medical College – Huazhong University of Science and Technology, Wuhan – China
| |
Collapse
|
3
|
Zhao L, Qin Y, Liu Y, An L, Liu W, Zhang C, Song Q, Dai C, Zhang J, Li A. The total xanthones extracted from Gentianella acuta alleviates HFpEF by activating the IRE1α/Xbp1s pathway. J Cell Mol Med 2024; 28:e18466. [PMID: 38847482 PMCID: PMC11157675 DOI: 10.1111/jcmm.18466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 05/11/2024] [Accepted: 05/17/2024] [Indexed: 06/10/2024] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a clinical syndrome characterized by pulmonary and systemic congestion resulting from left ventricular diastolic dysfunction and increased filling pressure. Currently, however, there is no evidence on effective pharmacotherapy for HFpEF. In this study, we aimed to investigate the therapeutic effect of total xanthones extracted from Gentianella acuta (TXG) on HFpEF by establishing an high-fat diet (HFD) + L-NAME-induced mouse model. Echocardiography was employed to assess the impact of TXG on the cardiac function in HFpEF mice. Haematoxylin and eosin staining, wheat germ agglutinin staining, and Masson's trichrome staining were utilized to observe the histopathological changes following TXG treatment. The results demonstrated that TXG alleviated HFpEF by reducing the expressions of genes associated with myocardial hypertrophy, fibrosis and apoptosis. Furthermore, TXG improved cardiomyocyte apoptosis by inhibiting the expression of apoptosis-related proteins. Mechanistic investigations revealed that TXG could activate the inositol-requiring enzyme 1α (IRE1α)/X-box-binding protein 1 (Xbp1s) signalling pathway, but the knockdown of IRE1α using the IRE1α inhibitor STF083010 or siRNA-IRE1α impaired the ability of TXG to ameliorate cardiac remodelling in HFpEF models. In conclusion, TXG alleviates myocardial hypertrophy, fibrosis and apoptosis through the activation of the IRE1α/Xbp1s signalling pathway, suggesting its potential beneficial effects on HFpEF patients.
Collapse
Affiliation(s)
- Linna Zhao
- Hebei Key Laboratory of Chinese Medicine Research on Cardio‐Cerebrovascular DiseaseShijiazhuangHebeiChina
| | - Yiping Qin
- Hebei Key Laboratory of Chinese Medicine Research on Cardio‐Cerebrovascular DiseaseShijiazhuangHebeiChina
| | - Yangong Liu
- The First Hospital of Hebei Medical UniversityShijiazhuangHebeiChina
| | - Liping An
- College of Basic MedicineHebei University of Chinese MedicineShijiazhuangHebeiChina
| | - Weizhe Liu
- Hebei Key Laboratory of Chinese Medicine Research on Cardio‐Cerebrovascular DiseaseShijiazhuangHebeiChina
- College of Basic MedicineHebei University of Chinese MedicineShijiazhuangHebeiChina
| | - Chuang Zhang
- Department of TechnologyHebei University of Chinese MedicineShijiazhuangHebeiChina
| | - Qiuhang Song
- Hebei Key Laboratory of Chinese Medicine Research on Cardio‐Cerebrovascular DiseaseShijiazhuangHebeiChina
| | - Cheng Dai
- Hebei Key Laboratory of Chinese Medicine Research on Cardio‐Cerebrovascular DiseaseShijiazhuangHebeiChina
| | - Juanjuan Zhang
- Hebei Key Laboratory of Chinese Medicine Research on Cardio‐Cerebrovascular DiseaseShijiazhuangHebeiChina
- Faculty of NursingHebei University of Chinese MedicineShijiazhuangHebeiChina
| | - Aiying Li
- Hebei Key Laboratory of Chinese Medicine Research on Cardio‐Cerebrovascular DiseaseShijiazhuangHebeiChina
- College of Basic MedicineHebei University of Chinese MedicineShijiazhuangHebeiChina
| |
Collapse
|
4
|
Hou Z, Yang F, Chen K, Wang Y, Qin J, Liang F. hUC-MSC-EV-miR-24 enhances the protective effect of dexmedetomidine preconditioning against myocardial ischemia-reperfusion injury through the KEAP1/Nrf2/HO-1 signaling. Drug Deliv Transl Res 2024; 14:143-157. [PMID: 37540334 DOI: 10.1007/s13346-023-01388-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/07/2023] [Indexed: 08/05/2023]
Abstract
The cardioprotective effect of microRNAs (miRNAs) on myocardial ischemic-reperfusion (I/R) injury has been documented. Here, we aim to decipher the mechanism of miR-24 delivered by human umbilical cord mesenchymal stem cell-derived extracellular vesicles (hUC-MSC-EVs) in myocardial I/R injury after dexmedetomidine (DEX) preconditioning. We collected and identified hUC-MSCs and extracted EVs, which were co-cultured with DEX-preconditioned hypoxia/reoxygenation (H/R) cardiomyocyte models or injected into I/R mouse models. The cardiomyocytes and myocardial injury were evaluated by molecular biology experiments. miR-24 was highly expressed in hUC-MSC-EVs. hUC-MSC-EVs could transfer miR-24 into cardiomyocytes where miR-24 augmented cell viability and inhibited cell apoptosis after DEX preconditioning. In the co-culture system of RAW264.7 macrophages with hUC-MSC-EVs, miR-24 promoted M2-type polarization of macrophages and reduced M1-type macrophage polarization. Mechanistically, miR-24 targeted KEAP1 and inhibited its expression, resulting in disruption of the Nrf2/HO-1 signaling. In vivo data confirmed that miR-24 delivered by hUC-MSC-EVs enhanced the suppressing effect of DEX preconditioning on inflammation and apoptosis in rats following myocardial I/R injury. Overall, miR-24 delivered by hUC-MSC-EVs can promote M2 polarization of macrophages and enhance the protective effect of DEX preconditioning on myocardial I/R injury by down-regulating the KEAP1/Nrf2/HO-1 signaling axis.
Collapse
Affiliation(s)
- Zixin Hou
- Department of Anesthesiology, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, 421001, People's Republic of China
| | - Fengrui Yang
- Department of Anesthesiology, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, 421001, People's Republic of China
- Department of Anesthesiology, Hengyang Medical School, Affiliated Huaihua Hospital, University of South China, Huaihua, 418000, People's Republic of China
| | - Kemin Chen
- Department of Anesthesiology, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, 421001, People's Republic of China
| | - Yuxia Wang
- Department of Anesthesiology, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, 421001, People's Republic of China
| | - Jie Qin
- Department of Anesthesiology, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, 421001, People's Republic of China
| | - Feng Liang
- Department of Anesthesiology, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, 421001, People's Republic of China.
| |
Collapse
|
5
|
Xu Q, Cao Y, Zhong X, Qin X, Feng J, Peng H, Su Y, Ma Z, Zhou S. Riboflavin protects against heart failure via SCAD-dependent DJ-1-Keap1-Nrf2 signalling pathway. Br J Pharmacol 2023; 180:3024-3044. [PMID: 37377111 DOI: 10.1111/bph.16184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 06/14/2023] [Accepted: 06/15/2023] [Indexed: 06/29/2023] Open
Abstract
BACKGROUND AND PURPOSE Our recent studies have shown that flavin adenine dinucleotide (FAD) exerts cardiovascular protective effects by supplementing short-chain acyl-CoA dehydrogenase (SCAD). The current study aimed to elucidate whether riboflavin (the precursor of FAD) could improve heart failure via activating SCAD and the DJ-1-Keap1-Nrf2 signalling pathway. EXPERIMENTAL APPROACH Riboflavin treatment was given to the mouse transverse aortic constriction (TAC)-induced heart failure model. Cardiac structure and function, energy metabolism and apoptosis index were assessed, and relevant signalling proteins were analysed. The mechanisms underlying the cardioprotection by riboflavin were analysed in the cell apoptosis model induced by tert-butyl hydroperoxide (tBHP). KEY RESULTS In vivo, riboflavin ameliorated myocardial fibrosis and energy metabolism, improved cardiac dysfunction and inhibited oxidative stress and cardiomyocyte apoptosis in TAC-induced heart failure. In vitro, riboflavin ameliorated cell apoptosis in H9C2 cardiomyocytes by decreasing reactive oxygen species (ROS). At the molecular level, riboflavin significantly restored FAD content, SCAD expression and enzymatic activity, activated DJ-1 and inhibited the Keap1-Nrf2/HO1 signalling pathway in vivo and in vitro. SCAD knockdown exaggerated the tBHP-induced DJ-1 decrease and Keap1-Nrf2/HO1 signalling pathway activation in H9C2 cardiomyocytes. The knockdown of SCAD abolished the anti-apoptotic effects of riboflavin on H9C2 cardiomyocytes. DJ-1 knockdown hindered SCAD overexpression anti-apoptotic effects and regulation on Keap1-Nrf2/HO1 signalling pathway in H9C2 cardiomyocytes. CONCLUSIONS AND IMPLICATIONS Riboflavin exerts cardioprotective effects on heart failure by improving oxidative stress and cardiomyocyte apoptosis via FAD to stimulate SCAD and then activates the DJ-1-Keap1-Nrf2 signalling pathway.
Collapse
Affiliation(s)
- Qingping Xu
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, China
- Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Yuhong Cao
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, China
- Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Xiaoyi Zhong
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, China
- Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Xue Qin
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, China
- Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Jingyun Feng
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, China
- Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Huan Peng
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, China
- Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Yongshao Su
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, China
- Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Zhichao Ma
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, China
- Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Sigui Zhou
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, China
- Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
6
|
Meng T, Zhang D, Zhang Y, Tian P, Chen J, Liu A, Li Y, Song C, Zheng Y, Su G. Tamoxifen induced cardiac damage via the IL-6/p-STAT3/PGC-1α pathway. Int Immunopharmacol 2023; 125:110978. [PMID: 37925944 DOI: 10.1016/j.intimp.2023.110978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 09/12/2023] [Accepted: 09/20/2023] [Indexed: 11/07/2023]
Abstract
Tamoxifen (TAM) is an effective anticancer drug for breast and ovarian cancer. However, increased risk of cardiotoxicity is a long-term clinical problem associated with TAM, while the underlying mechanisms remain unclear. Here, we performed experiments in cardiomyocytes and tumor-bearing or nontumor-bearing mice, and demonstrated that TAM induced cardiac injury via the IL-6/p-STAT3/PGC-1α/IL-6 feedback loop, which is responsible for reactive oxygen species (ROS) accumulation. Compared with non-tumor bearing mice, tumor-bearing mice showed stronger cardiac toxicity after TAM injection, although there was no significant difference. In vitro experiments demonstrated STAT3 phosphorylation inhibitor can increase PGC-1α expression and protect cardiomyocyte via decreasing ROS. Since tumor has higher STAT3 phosphorylation and IL-6 expression level, our research results indicated combining TAM and STAT3 inhibitor might be an effective treatment strategy which can provide both tumor killing and cardioprotective function. Further in vivo research is needed to fully elucidate the effect and mechanisms of the combination therapy of TAM and STAT3 inhibitor.
Collapse
Affiliation(s)
- Tingting Meng
- Research Center of Translational Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Dan Zhang
- Jinan Central Hospital, Jinan, Shandong, China
| | - Yu Zhang
- Research Center of Translational Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China; Research Center of Translational Medicine, Jinan Central Hospital, Shandong University, Jinan, Shandong, China
| | - Peng Tian
- Research Center of Translational Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China; Research Center of Translational Medicine, Jinan Central Hospital, Shandong University, Jinan, Shandong, China
| | - Jianlin Chen
- Research Center of Translational Medicine, Jinan Central Hospital, Weifang Medical University, Weifang, China
| | - Anbang Liu
- Research Center of Translational Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Ying Li
- Research Center of Translational Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Chunhong Song
- Laboratory Animal Center, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Yan Zheng
- Research Center of Translational Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China; Research Center of Translational Medicine, Jinan Central Hospital, Shandong University, Jinan, Shandong, China.
| | - Guohai Su
- Research Center of Translational Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
| |
Collapse
|
7
|
Wei XH, Liu WJ, Jiang W, Lan TH, Pan H, Ma MY, You LZ, Shang HC. XinLi formula, a traditional Chinese decoction, alleviates chronic heart failure via regulating the interaction of AGTR1 and AQP1. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 113:154722. [PMID: 36867964 DOI: 10.1016/j.phymed.2023.154722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 02/10/2023] [Accepted: 02/18/2023] [Indexed: 06/18/2023]
Abstract
BACKGROUND XinLi formula (XLF) is a traditional Chinese medicine used in clinical practice to treat chronic heart failure (CHF) in humans, with remarkable curative effect. However, the mechanism remains unknown. PURPOSE The goal of the current investigation was to determine how XLF affected CHF in a rat model of the condition brought on by ligation of the left anterior descending coronary artery, and to investigate the underlying mechanism. STUDY DESIGN AND METHODS Cardiac function was detected by echocardiography. The contents of myocardial enzymes, Ang II, ALD, TGF-β1, and inflammatory factors were measured by ELISA. Myocardial injury and myocardial fibrosis were evaluated by HE and Masson staining. Myocardial edema was assessed by cardiac mass index and transmission electron microscopy. Using Western blot and immunohistochemistry to examining the protein expression of inflammasome, TGF-β1, AGTR1, and AQP1 in the left ventricle. Furthermore, the interaction of AGTR1 and AQP1 was evaluated by co-immunoprecipitation. RESULTS XLF attenuated myocardial enzymes and myocardial injury, and improved cardiac function in rats with CHF after myocardial infarction. It also reduced Ang II and ALD levels in CHF rats, and suppressed the expression of AGTR1 and TGF-β1, finally alleviated myocardial fibrosis. By mechanism, XLF inhibited the expression of NLRP3 inflammasome proteins, reduced the plasma contents of IL-1β, IL-18, IL-6 and TNF-α. Additionally, XLF inhibited the expression of AQP1 and the interaction of AGTR1 and AQP1, alleviating myocardial edema. The common structure of the main chemical constituents of XLF were glycoside compounds with glycosyl. CONCLUSION XLF ameliorated CHF, which was evidenced by the alleviation of myocardial fibrosis by inhibiting AGTR1/NLRP3 signal, as well as the attenuation of myocardial edema by suppressing the interaction of AGTR1 and AQP1.
Collapse
Affiliation(s)
- Xiao-Hong Wei
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China; Guangdong Provincial Key Laboratory of Chinese Medicine for Prevention and Treatment of Refractory Chronic Diseases, Guangzhou 510020, China
| | - Wen-Jing Liu
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Wei Jiang
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510020, China; Guangdong Provincial Key Laboratory of Chinese Medicine for Prevention and Treatment of Refractory Chronic Diseases, Guangzhou 510020, China; Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510020, China
| | - Tao-Hua Lan
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510020, China; Guangdong Provincial Key Laboratory of Chinese Medicine for Prevention and Treatment of Refractory Chronic Diseases, Guangzhou 510020, China; Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510020, China
| | - Hai'e Pan
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Ming-Yue Ma
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Liang-Zhen You
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Hong-Cai Shang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China.
| |
Collapse
|
8
|
Yu H, Peng Y, Dong W, Shen B, Yang G, Nie Q, Tian Y, Qin L, Song C, Chen B, Zhao Y, Li L, Hong S. Nrf2 attenuates methamphetamine-induced myocardial injury by regulating oxidative stress and apoptosis in mice. Hum Exp Toxicol 2023; 42:9603271231219488. [PMID: 38031934 DOI: 10.1177/09603271231219488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2023]
Abstract
OBJECTIVES Methamphetamine (MA) abuse is a serious social problem worldwide. Cardiovascular complications were the second leading cause of death among MA abusers. We aimed to clarify the effects of MA on myocardial injury, oxidative stress, and apoptosis in myocardial cells and to explore the potential mechanism of nuclear factor-erythroid factor 2-related factor 2 (Nrf2) in MA-induced oxidative stress and apoptosis. METHODS An acute cardiac toxicity model of MA was established by intraperitoneal injection of MA (2 mg/kg) for 5 days. Nrf2 activation (by sulforaphane (SFN) 1 h before MA injection) and Nrf2 gene knockout were performed to explore the regulatory effects of Nrf2 on cardiac toxicity. RESULTS The protein expressions of Nrf2 (p < .001) and heme oxygenase-1 (HO-1) were increased (p < .01), suggesting that MA activated the Nrf2/HO-1 pathway. In the MA group, cardiac injury score (p < .001) and cardiac troponin I (cTnI) protein expression increased (p < .01). Malondialdehyde (MDA) content increased (p < .001), superoxide dismutase (SOD) activity decreased (p < .05). Protein expressions of Caspase-3 (p < .001) and Bax (p < .001) increased, and Bcl-2 decreased (p < .001) as well. These changes were reversed by activation of Nrf2 but became more pronounced after Nrf2 knockout, suggested that the activation and knockout of Nrf2 attenuated and aggravated MA-induced myocardial injury, oxidative stress and apoptosis in myocardial cells, respectively. CONCLUSIONS MA administration induced myocardial injury, oxidative stress, and apoptosis in mice. Nrf2 attenuated MA-induced myocardial injury by regulating oxidative stress and apoptosis, thus playing a protective role.
Collapse
Affiliation(s)
- Hao Yu
- NHC Key Laboratory of Drug Addiction Medicine, School of Forensic Medicine, Kunming Medical University, Kunming, China
- West China Hospital, Sichuan University, Chengdu, China
| | - Yanxia Peng
- NHC Key Laboratory of Drug Addiction Medicine, School of Forensic Medicine, Kunming Medical University, Kunming, China
| | - Wenjuan Dong
- NHC Key Laboratory of Drug Addiction Medicine, School of Forensic Medicine, Kunming Medical University, Kunming, China
| | - Baoyu Shen
- NHC Key Laboratory of Drug Addiction Medicine, School of Forensic Medicine, Kunming Medical University, Kunming, China
| | - Genmeng Yang
- NHC Key Laboratory of Drug Addiction Medicine, School of Forensic Medicine, Kunming Medical University, Kunming, China
| | - Qianyun Nie
- NHC Key Laboratory of Drug Addiction Medicine, School of Forensic Medicine, Kunming Medical University, Kunming, China
| | - Yan Tian
- NHC Key Laboratory of Drug Addiction Medicine, School of Forensic Medicine, Kunming Medical University, Kunming, China
| | - Lixiang Qin
- NHC Key Laboratory of Drug Addiction Medicine, School of Forensic Medicine, Kunming Medical University, Kunming, China
| | - Chunhui Song
- NHC Key Laboratory of Drug Addiction Medicine, School of Forensic Medicine, Kunming Medical University, Kunming, China
| | - Bingzheng Chen
- NHC Key Laboratory of Drug Addiction Medicine, School of Forensic Medicine, Kunming Medical University, Kunming, China
| | - Yongna Zhao
- Key Laboratory of Natural Medicine Pharmacology of Yunnan Province, Kunming Medical University, Kunming, China
| | - Lihua Li
- NHC Key Laboratory of Drug Addiction Medicine, School of Forensic Medicine, Kunming Medical University, Kunming, China
| | - Shijun Hong
- NHC Key Laboratory of Drug Addiction Medicine, School of Forensic Medicine, Kunming Medical University, Kunming, China
| |
Collapse
|
9
|
Xie B, Wu B, Zhao M, Liu Y, Xu G, Li Q. Efficacy and safety of Yiqi Huoxue Yangxin decoction combined with western medicine in patients with chronic coronary heart disease: A protocol for systematic review and meta-analysis. Medicine (Baltimore) 2022; 101:e32283. [PMID: 36595820 PMCID: PMC9794337 DOI: 10.1097/md.0000000000032283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND There is no evidence-based data to confirm the efficacy of Yiqi Huoxue Yangxin Decoction combined with Western medicine in patients with coronary heart disease (CHD). Therefore, in order to provide new medical evidence for clinical treatment, we used this protocol to conduct a systematic review and meta-analysis on the effectiveness and safety of Yiqi Huoxue Yangxin Decoction combined with Western medicine in patients with CHD. METHODS The systematic review protocol is registered in PROSPERO under registration number CRD42022372800. The systematic literature review will follow PRISMA guidelines (Preferred Reporting Items for Systematic Reviews and Meta-Analyses). The following search terms will be used in PUBMED, Scopus, EMBASE, Cochrane Library, CNKI, Wan Fang, and VIP on December 1, 2022. Two independent authors extract the following descriptive raw information from selected studies: study characteristics such as authors, year of publication, study design; patient demographic information such as number of patients, mean age, body mass index, and sex ratio. The primary outcome of interest is symptom scores. Secondary outcomes include ECG effective rate, improvement of blood lipid index, and adverse events. The Cochrane Bias Risk Tool is used independently by 2 reviewers to assess the risk of bias in included randomized controlled trials. The quality of retrospective studies will be assessed using the Newcastle-Ottawa scale. CONCLUSIONS The results of our review will be reported strictly following the PRISMA criteria and the review will add to the existing literature by showing compelling evidence and improved guidance in clinic settings.
Collapse
Affiliation(s)
- Bingxin Xie
- Department of Cardiovascular Medicine, Southern Branch of Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Bao Wu
- Department of Cardiovascular Medicine, Southern Branch of Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Mingzhang Zhao
- Department of emergency, Southern Branch of Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yili Liu
- Department of Ultrasonography, Southern Branch of Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Guolei Xu
- Department of Cardiovascular Medicine, Southern Branch of Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qiao Li
- Department of Endocrinology, Southern Branch of Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- * Correspondence: Qiao Li, MD, Department of Endocrinology, Southern Branch of Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China (e-mail: )
| |
Collapse
|
10
|
Ye Z, Wang Q, Dai S, Ji X, Cao P, Xu C, Bao G. The Berberis vulgaris L. extract berberine exerts its anti-oxidant effects to ameliorate cholesterol overloading-induced cell apoptosis in the primary mice hepatocytes: an in vitro study. In Vitro Cell Dev Biol Anim 2022; 58:855-866. [PMID: 36481977 DOI: 10.1007/s11626-022-00737-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 11/03/2022] [Indexed: 12/14/2022]
Abstract
Cholesterol overloading stress damages normal cellular functions in hepatocytes and induces metabolic disorders to facilitate the development of multiple diseases, including cardiovascular diseases, which seriously degrades the life quality of human beings. Recent data suggest that the Berberis vulgaris L. extract berberine is capable of regulating cholesterol homeostasis, which is deemed as potential therapeutic drug for the treatment of cholesterol overloading-associated diseases, but its detailed functions and molecular mechanisms are still largely unknown. In the present study, we evidenced that berberine suppressed cell apoptosis in high-cholesterol-diet mice liver and cholesterol-overloaded mice hepatocytes. Also, cholesterol overloading promoted reactive oxygen species (ROS) generation to trigger oxidative damages in hepatocytes, which were reversed by co-treating cells with both berberine and the ROS scavenger N-acetylcysteine (NAC). Moreover, the underlying mechanisms were uncovered, and we validated that berberine downregulated Keap1, and upregulated Nrf2 to activate the anti-oxidant Nrf2/HO-1 signaling pathway in cholesterol overloading-treated hepatocytes, and both Keap1 upregulation and Nrf2 downregulation abrogated the suppressing effects of berberine on cell apoptosis in the hepatocytes with cholesterol exposure. Taken together, we concluded that berberine activated the anti-oxidant Keap1/Nrf2/HO-1 pathway to eliminate cholesterol overloading-induced oxidative stress and apoptotic cell death in mice hepatocytes, and those evidences hinted that berberine might be used as putative therapeutic drug for the treatment of cholesterol overloading-associated cardiovascular diseases.
Collapse
Affiliation(s)
- Zhengchen Ye
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Kunming Medical University, Xichang Road No. 295, Kunming, 650032, Yunnan Province, China
| | - Qiang Wang
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Kunming Medical University, Xichang Road No. 295, Kunming, 650032, Yunnan Province, China
| | - Shupeng Dai
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Kunming Medical University, Xichang Road No. 295, Kunming, 650032, Yunnan Province, China
| | - Xiang Ji
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Kunming Medical University, Xichang Road No. 295, Kunming, 650032, Yunnan Province, China
| | - Pingli Cao
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Kunming Medical University, Xichang Road No. 295, Kunming, 650032, Yunnan Province, China
| | - Chenglei Xu
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Kunming Medical University, Xichang Road No. 295, Kunming, 650032, Yunnan Province, China
| | - Guoqing Bao
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Kunming Medical University, Xichang Road No. 295, Kunming, 650032, Yunnan Province, China.
| |
Collapse
|
11
|
The Effect of Yiqi Huoxue Tongluo Decoction on Spinal Cord Microglia Activation and ASK1-MKK3-p38 Signal Pathway in Rats with Diabetic Neuropathic Pain. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:2408265. [PMID: 35646150 PMCID: PMC9135525 DOI: 10.1155/2022/2408265] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 03/23/2022] [Accepted: 04/13/2022] [Indexed: 02/06/2023]
Abstract
Diabetic neuropathic pain (DNP) is one of the most common chronic peripheral neuropathies in diabetes mellitus (DM). Objective. To observe the underlying mechanism of the effects of Yiqi Huoxue Tongluo Decoction (YQHX) on DNP rats. Methods. SD rats were intraperitoneally injected with 35 mg/kg streptozotocin (STZ) to prepare DNP models and were treated with YQHX for 8 weeks. Results. Studies have shown that the drug restores some levels of MWT, TWL, and MNCV, downregulates the levels of inflammatory factors IL-6, IL-1β, and TNF-α, downregulates the levels of ASK1-MKK3-p38, and weakens the level of OX42 activation. Conclusion. Yiqi Huoxue Tongluo Decoction can relieve DNP by affecting the activity of spinal cord microglia and the ASK1-MKK3-p38 signaling pathway, thereby reducing the central sensitization caused by the inflammatory response of DNP rats.
Collapse
|
12
|
Wang L, Wang S, Jia T, Sun X, Xing Z, Liu H, Yao J, Chen Y. Dexmedetomidine prevents cardiomyocytes from hypoxia/reoxygenation injury via modulating tetmethylcytosine dioxygenase 1-mediated DNA demethylation of Sirtuin1. Bioengineered 2022; 13:9369-9386. [PMID: 35387565 PMCID: PMC9161963 DOI: 10.1080/21655979.2022.2054762] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
Myocardial hypoxia/reoxygenation (H/R) injury is a common pathological change in patients with acute myocardial infarction undergoing reperfusion therapy. Dexmedetomidine (DEX) has been found to substantially improve ischemia-mediated cell damage. Here, we focus on probing the role and mechanism of DEX in ameliorating myocardial H/R injury. Oxygen–glucose deprivation and reoxygenation (OGD/R) were applied to construct the H/R injury model in human myocardial cell lines. After different concentrations of DEX’s treatment, cell counting kit-8 (CCK-8) assay and BrdU assay were employed to test cell viability. The profiles of apoptosis-related proteins Bcl2, Bax, Bad and Caspase3, 8, 9 were determined by Western blot (WB). The expression of inflammatory factors interleukin 1β (IL-1β) and tumor necrosis factor-α (TNF-α) was checked by reverse transcription-polymerase chain reaction (RT-PCR). By conducting WB, we examined the expression of NF-κB, Sirt1, Tet methylcytosine dioxygenase 1 (TET1) and DNA methylation-related proteins (DNA methyltransferase 1, DNMT1; DNA methyltransferase 3 alpha, DNMT3A; and DNA methyltransferase 3 beta, DNMT3B). Our data showed that OGD/R stimulation distinctly hampered the viability and elevated apoptosis and inflammatory factor expression in cardiomyocytes. DEX treatment notably impeded myocardial apoptosis and inflammation and enhanced cardiomyocyte viability. OGD/R enhanced total DNA methylation levels in cardiomyocytes, while DEX curbed DNA methylation. In terms of mechanism, inhibiting TET1 or Sirtuin1 (Sirt1) curbed the DEX-mediated myocardial protection. TET1 strengthened demethylation of the Sirt1 promoter and up-regulated Sirt1. DEX up-regulates Sirt1 by accelerating TET1 and mediating demethylation of the Sirt1 promoter and improves H/R-mediated myocardial injury.
Collapse
Affiliation(s)
- Li Wang
- Department of Anesthesiology. First Affiliated Hospital of Hebei North College, Zhangjiakou, China
| | - Shaowei Wang
- Department of Anesthesiology. First Affiliated Hospital of Hebei North College, Zhangjiakou, China
| | - Tong Jia
- Department of Anesthesiology. First Affiliated Hospital of Hebei North College, Zhangjiakou, China
| | - Xiaojia Sun
- Department of Anesthesiology. First Affiliated Hospital of Hebei North College, Zhangjiakou, China
| | - Zhen Xing
- Department of Anesthesiology. First Affiliated Hospital of Hebei North College, Zhangjiakou, China
| | - Hui Liu
- Department of Anesthesiology. First Affiliated Hospital of Hebei North College, Zhangjiakou, China
| | - Jie Yao
- Department of Anesthesiology. First Affiliated Hospital of Hebei North College, Zhangjiakou, China
| | - Yanlin Chen
- Department of Anesthesiology. First Affiliated Hospital of Hebei North College, Zhangjiakou, China
| |
Collapse
|
13
|
Wang S, Lv T, Chen Q, Yang Y, Xu L, Zhang X, Wang E, Hu X, Liu Y. Transcriptome sequencing and lncRNA-miRNA-mRNA network construction in cardiac fibrosis and heart failure. Bioengineered 2022; 13:7118-7133. [PMID: 35235759 PMCID: PMC8974171 DOI: 10.1080/21655979.2022.2045839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Cardiac fibrosis (CF) and heart failure (HF) are common heart diseases, and severe CF can lead to HF. In this study, we tried to find their common potential molecular markers, which may help the diagnosis and treatment of CF and HF. RNA library construction and high-throughput sequencing were performed. The DESeq2 package in R was used to screen differentially expressed mRNAs (DEmRNAs), differentially expressed lncRNA (DElncRNAs) and differentially expressed miRNA (DEmiRNAs) between different samples. The common DEmRNAs, DElncRNAs and DEmiRNAs for the two diseases were obtained. The ConsensusPathDB (CPDB) was used to perform biological function enrichment for common DEmRNAs. Gene interaction network was constructed to screen out key genes. Subsequently, real-time polymerase chain reaction (RT-PCR) verification was performed. Lastly, GSE104150 and GSE21125 data sets were utilized for expression validation and diagnostic analysis. There were 1477 DEmRNAs, 502 DElncRNAs and 36 DEmiRNAs between CF and healthy control group. There were 607 DEmRNAs, 379DElncRNAs,s and 42 DEmiRNAs between HF and healthy control group. CH and FH shared 146 DEmRNAs, 80 DElncRNAs, and 6 DEmiRNAs. Hsa-miR-144-3p, CCNE2, C9orf72, MAP3K20-AS1, LEF1-AS1, AC243772.2, FLJ46284, and AC239798.2 were key molecules in lncRNA-miRNA-mRNA network. In addition, hsa-miR-144-3p and CCNE2 may be considered as potential diagnostic gene biomarkers in HF. In this study, the identification of common biomarkers of CF and HF may help prevent CF to HF transition as early as possible.
Collapse
Affiliation(s)
- Shuo Wang
- Department of Cardiovasology, Shijiazhuang People's Hospital, Shijiazhaung, HB, China
| | - Tianjie Lv
- Department of Cardiovasology, Shijiazhuang People's Hospital, Shijiazhaung, HB, China
| | - Qincong Chen
- Department of Cardiovasology, Shijiazhuang People's Hospital, Shijiazhaung, HB, China
| | - Yan Yang
- Department of Cardiovasology, Shijiazhuang People's Hospital, Shijiazhaung, HB, China
| | - Lei Xu
- Department of Cardiovasology, Shijiazhuang People's Hospital, Shijiazhaung, HB, China
| | - Xiaolei Zhang
- Department of Cardiovasology, Shijiazhuang People's Hospital, Shijiazhaung, HB, China
| | - Enmao Wang
- Department of Cardiovasology, Shijiazhuang People's Hospital, Shijiazhaung, HB, China
| | - Xitian Hu
- Department of Cardiovasology, Shijiazhuang People's Hospital, Shijiazhaung, HB, China
| | - Yuying Liu
- Department of Cardiovasology, Shijiazhuang People's Hospital, Shijiazhaung, HB, China
| |
Collapse
|
14
|
Qi Y, Tang Y, Yin L, Ding K, Zhao C, Yan W, Yao Y. miR-129-5p restores cardiac function in rats with chronic heart failure by targeting the E3 ubiquitin ligase Smurf1 and promoting PTEN expression. Bioengineered 2022; 13:2371-2386. [PMID: 35034538 PMCID: PMC8974089 DOI: 10.1080/21655979.2021.2024335] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Chronic heart failure (CHF) is a prevalent health concern with complex pathogenesis. This current study set out to estimate the function of the miR-129-5p/Smurf1/PTEN axis on cardiac function injury in CHF. The model of CHF in rats was established. The cardiac function indexes, myocardial tissue damage, and oxidative stress-related factors in CHF rats were evaluated after the interference of Smurf1/miR-129-5p/PTEN. The targeting relationships between miR-129-5p and Smurf1 and between PTEN and Smurf1 were verified. It was found that that after modeling, cardiac functions were impaired, heart/left ventricular/lung weight and the myocardial structure was destroyed, and the degree of fibrosis of myocardial tissue was increased. After Smurf1 knockdown, the cardiac function, myocardial structure, and oxidative stress were improved, and the fibrosis in myocardial tissue was decreased. Smurf1 was a target of miR-129-5p. miR-129-5p could annul the protective effect of Smurf1 silencing on CHF rats. Smurf1 inhibited PTEN expression by promoting PTEN ubiquitination, while miR-129-5p enhanced PTEN expression by inhibiting Smurf1. Meanwhile, overexpression of PTEN annulled the cardiac dysfunction in CHF rats induced by Smurf1. In conclusion, miR-129-5p targeted Smurf1 and repressed the ubiquitination of PTEN, and promoted PTEN expression, thus improving the cardiac function of CHF rats.
Collapse
Affiliation(s)
- Yuan Qi
- Department of Cardiology, Gongli Hospital of Shanghai Pudong New Area, Shanghai, China
| | - Yan Tang
- Department of Cardiology, Tongji Hospital Affiliated to Tongji University, Shanghai, China
| | - Lu Yin
- Department of Cardiology, Gongli Hospital of Shanghai Pudong New Area, Shanghai, China
| | - Keke Ding
- Department of Cardiology, Tongji Hospital Affiliated to Tongji University, Shanghai, China
| | - Cuimei Zhao
- Department of Cardiology, Tongji Hospital Affiliated to Tongji University, Shanghai, China
| | - Wenwen Yan
- Department of Cardiology, Tongji Hospital Affiliated to Tongji University, Shanghai, China
| | - Yi'an Yao
- Department of Cardiology, Tongji Hospital Affiliated to Tongji University, Shanghai, China
| |
Collapse
|
15
|
Wang Q, Ma M, Yu H, Yu H, Zhang S, Li R. Mirtazapine prevents cell activation, inflammation, and oxidative stress against isoflurane exposure in microglia. Bioengineered 2022; 13:521-530. [PMID: 34964706 PMCID: PMC8805817 DOI: 10.1080/21655979.2021.2009971] [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: 10/15/2021] [Accepted: 11/18/2021] [Indexed: 11/23/2022] Open
Abstract
Mirtazapine is an antidepressant drug that has been proven to possess a cognitive enhancer efficiency. In this study, we evaluated the potential protective effects of mirtazapine on BV2 microglia in response to isoflurane exposure. Our results show that mirtazapine attenuated isoflurane-induced expression of microglia-specific protein Iba1 in BV2 microglia. Mirtazapine prevented isoflurane-induced production of the pro-inflammatory factors interleukin (IL)-1β and IL-18 by inhibiting the activation of the nod-like receptor family protein 3 (NLRP3) inflammasome in BV2 microglia. The increased reactive oxygen species (ROS) production and elevated expression level of NADPH oxidase 4 (NOX4) in isoflurane-induced BV2 microglia were mitigated by mirtazapine. Isoflurane exposure reduced triggering receptor expressed on myeloid cells 2 (TREM2) expression in BV2 microglia, which was restored by mirtazapine. Moreover, silencing of TREM2 abolished the inhibitory effects of mirtazapine on ionized calcium-binding adapter molecule 1 (Iba1) expression and inflammation in BV2 microglia. From these results, we could infer that mirtazapine exerted a protective effect on BV2 microglia against isoflurane exposure-caused microglia activation, neuroinflammation, and oxidative stress via inducing TREM2 activation. Hence, mirtazapine might be a potential intervention strategy to prevent isoflurane exposure-caused cognitive dysfunction in clinical practice.
Collapse
Affiliation(s)
- Qi Wang
- Department of Anesthesiology, Cangzhou Central Hospital, Cangzhou, Hebei, China
| | - Meina Ma
- Department of Anesthesiology, Cangzhou Central Hospital, Cangzhou, Hebei, China
| | - Hong Yu
- Department of Anesthesiology, Cangzhou Central Hospital, Cangzhou, Hebei, China
| | - Hongmei Yu
- Department of Anesthesiology, Cangzhou Central Hospital, Cangzhou, Hebei, China
| | - Shuai Zhang
- Department of Anesthesiology, Cangzhou Central Hospital, Cangzhou, Hebei, China
| | - Rui Li
- Department of Anesthesiology, Cangzhou Central Hospital, Cangzhou, Hebei, China
| |
Collapse
|
16
|
Nie S, Cui X, Guo J, Ma X, Zhi H, Li S, Li Y. Long non-coding RNA AK006774 inhibits cardiac ischemia-reperfusion injury via sponging miR-448. Bioengineered 2021; 12:4972-4982. [PMID: 34369259 PMCID: PMC8806428 DOI: 10.1080/21655979.2021.1954135] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
In recent years, the incidence and mortality of myocardial infarction (MI) have been increasing throughout the world, threatening public health. Non-coding RNAs (ncRNAs), including microRNAs (miRNAs) and long non-coding RNAs (lncRNAs), play critical roles in the progression of MI. The present study aimed to investigate the role of lncRNA AK006774 in the progression of myocardial infarction and find out novel therapeutic or diagnostic target of myocardial infarction. A mouse ischemia/reperfusion (I/R) model and 2,3,5-Triphenyte-trazoliumchloride (TTC) staining were performed to evaluate the effects of AK006774 on I/R injury in vivo. Hypoxia/reoxygenation (H/R) models using primary cardiomyocytes have been established. Flow cytometry and Terminal Deoxynucleotide Transferase dUTP Nick End Labeling (TUNEL) assays were performed to evaluate the effects of AK006774 on cardiomyocyte apoptosis. Luciferase and RNA pull-down assays were performed to verify the interaction between miR-448 and its targets. Western blotting and quantitative PCR were performed to determine protein and gene expression, respectively. We first found that AK006774 overexpression reduced I/R-induced infarct area and cardiomyocyte apoptosis in vivo. Accordingly, AK006774 inhibited apoptosis and oxidative stress in cardiomyocytes subjected to H/R treatment in vitro. Mechanistically, AK006774 modulated the expression of bcl-2 by sponging miR-448. Overexpression of miR-448 antagonized the effects of AK006774 on cardiomyocyte apoptosis. The AK006774/miR-448/bcl-2 signaling axis acts as a key regulator of I/R injury and may be a potential therapeutic or diagnostic target for the treatment of MI.
Collapse
Affiliation(s)
- Shen Nie
- Department of Emergency, Cangzhou Central Hospital, Cangzhou, China
| | - Xiaoya Cui
- Department of Emergency, Cangzhou Central Hospital, Cangzhou, China
| | - Jinping Guo
- Department of Emergency, Cangzhou Central Hospital, Cangzhou, China
| | - Xiaohua Ma
- Department of Emergency, Cangzhou Central Hospital, Cangzhou, China
| | - Haijun Zhi
- Department of Emergency, Cangzhou Central Hospital, Cangzhou, China
| | - Shilei Li
- Department of Emergency, Cangzhou Central Hospital, Cangzhou, China
| | - Yong Li
- Department of Emergency, Cangzhou Central Hospital, Cangzhou, China
| |
Collapse
|