1
|
Liu HH, Yang F, Zhang L, Zhang XL, Zhao N, Zhang ZY, Zhou JB, Wei TP, Qian LL, Ding LG, Wang RX. Decreased PLK2 promotes atrial fibrillation in diabetic mice through Nrf2/HO-1 pathway. Acta Diabetol 2025:10.1007/s00592-025-02480-9. [PMID: 40080197 DOI: 10.1007/s00592-025-02480-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 02/23/2025] [Indexed: 03/15/2025]
Abstract
BACKGROUND Type 2 diabetes mellitus (T2DM) is associated with an increased incidence of atrial fibrillation (AF). However, the exact mechanisms involved have not yet been fully elucidated. Dysregulation of cardiac potassium channels can trigger AF. This study aimed to investigate the mechanisms of abnormal expression of atrial potassium channel proteins Kv1.5, Kv4.2, and Kv4.3 in type 2 diabetic mice. METHODS The db/db mice and their control littermates were set as the T2DM group and the control (Con) group. Acetylcholine-calcium chloride was injected via the tail veins to induce AF. HL-1 cells were cultured with normal or high-glucose medium and treated with or without Dimethyl Fumarate (DMF) or hemin in vitro. The expression and cellular localization of proteins were evaluated by western blotting and immunofluorescence. RESULTS The results showed that high glucose impaired the expression of Kv1.5, Kv4.2 and Kv4.3 proteins both in vivo and in vitro, in parallel with a significant down-regulation of polo-like kinase 2 (PLK2), nuclear factor erythroid 2-related factor 2 (Nrf2), p-Nrf2 and heme oxygenase-1 (HO-1) proteins. Moreover, immunofluorescence revealed that both high glucose and PLK2 knockdown could result in reduced Nrf2 and p-Nrf2 expression and subsequent nuclear translocation. While overexpression of PLK2, treatment with DMF, an agonist of Nrf2, or hemin, an inducer of HO-1, could restore the reduction of Kv1.5, Kv4.2 and Kv4.3 proteins caused by high glucose. CONCLUSION Diabetes reduces the expression of Kv1.5, Kv4.2 and Kv4.3 proteins in atrial cells through inhibition of PLK2/Nrf2/HO-1 pathway, thereby leading to the increased susceptibility to AF in T2DM.
Collapse
Affiliation(s)
- Huan-Huan Liu
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China
- Department of Cardiology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, 214023, China
| | - Fan Yang
- Department of Cardiology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, 214023, China
| | - Lei Zhang
- Department of Cardiology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, 214023, China
| | - Xiao-Lu Zhang
- Department of Cardiology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, 214023, China
| | - Ning Zhao
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China
| | - Zhen-Ye Zhang
- Department of Cardiology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, 214023, China
| | - Jia-Bin Zhou
- Department of Cardiology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, 214023, China
| | - Tian-Peng Wei
- Department of Cardiology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, 214023, China
| | - Ling-Ling Qian
- Department of Cardiology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, 214023, China.
| | - Li-Gang Ding
- Cardiac Arrhythmia Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China.
| | - Ru-Xing Wang
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China.
- Department of Cardiology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, 214023, China.
| |
Collapse
|
2
|
Ma Y, Guo C, Wang Y, Liu X. Effects of Dl-3-n-butylphthalide on neurological function, hemodynamics and Hcy concentration in cerebral hemorrhage: a systematic review and meta-analysis. Front Pharmacol 2024; 15:1360932. [PMID: 38881880 PMCID: PMC11177091 DOI: 10.3389/fphar.2024.1360932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 05/06/2024] [Indexed: 06/18/2024] Open
Abstract
Background Dl-3-n-Butylphthalide (NBP) has emerged as a potential therapeutic agent for cerebral hemorrhage, despite not being included in current guideline recommendations. Investigating the underlying physiological and pathological mechanisms of Dl-3-n-Butylphthalide in cerebral hemorrhage treatment remains a critical area of research. Objective This review aims to evaluate the efficacy of Dl-3-n-Butylphthalide in cerebral hemorrhage treatment and elucidate its potential biological mechanisms, thereby providing evidence to support treatment optimization. Methods A comprehensive search of seven electronic databases (PubMed, Web of Science, Embase, Cochrane Library, China National Knowledge Infrastructure, VIP, and Wanfang Database) was conducted for studies published up to September 2023. Screening and data extraction were performed by a team of researchers. The Cochrane collaboration tool was utilized for risk bias assessment, and Revman 5.3 along with Stata 17.0 were employed for statistical analysis. Outcomes We searched 254 literature, and 19 were included in this meta-analysis. The results showed that Dl-3-n-Butylphthalide improved the clinical efficacy rate (RR = 1.25, 95% CI 1.19-1.31; p = 0.00), quality of life (MD = 13.93, 95% CI: 11.88-15.98; p = 0.000), increased cerebral blood flow and velocity, reduced cerebral edema volume, Hcy concentration, and did not have obvious adverse reactions (RR = 0.68, 95% CI: 0.39-1.18; p = 0.10). Conclusion This meta-analysis is the first to demonstrate the potential of Dl-3-n-Butylphthalide in treating cerebral hemorrhage. It suggests that Dl-3-n-Butylphthalide may alleviate clinical symptoms by modulating neurological function and improving hemodynamics. Our findings provide robust evidence for incorporating Dl-3-n-Butylphthalide into cerebral hemorrhage treatment strategies, potentially guiding future clinical practice and research. Systematic Review Registration: https://www.crd.york.ac.uk/PROSPERO/ display_record.php?RecordID=355114, Identifier CRD42022355114.
Collapse
Affiliation(s)
- Yingqi Ma
- First School of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Chenchen Guo
- Neck-Shoulder and Lumbocrural Pain Hospital of Shandong First Medical University, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Yiguo Wang
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xinxin Liu
- Centre for Evidence-Based Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
3
|
Yin P, Wu Y, Long X, Zhu S, Chen S, Lu F, Lin K, Xu J. HACE1 expression in heart failure patients might promote mitochondrial oxidative stress and ferroptosis by targeting NRF2. Aging (Albany NY) 2023; 15:13888-13900. [PMID: 38070140 PMCID: PMC10756096 DOI: 10.18632/aging.205272] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 10/17/2023] [Indexed: 12/21/2023]
Abstract
BACKGROUND Heart failure is a prevalent and life-threatening medical condition characterized by abnormal atrial electrical activity, contributing to a higher risk of ischemic stroke. Atrial remodelling, driven by oxidative stress and structural changes, plays a central role in heart failure progression. Recent studies suggest that HACE1, a regulatory gene, may be involved in cardiac protection against heart failure. METHODS Clinical data analysis involved heart failure patients, while an animal model utilized C57BL/6J mice. RT-PCR, microarray analysis, histological examination, ELISA, and Western blot assays were employed to assess gene and protein expression, oxidative stress, and cardiac function. Cell transfection and culture of mouse atrial fibroblasts were performed for in-vitro experiments. RESULTS HACE1 expression was reduced in heart failure patients and correlated negatively with collagen levels. In mouse models, HACE1 up-regulation reduced oxidative stress, mitigated fibrosis, and improved cardiac function. Conversely, HACE1 knockdown exacerbated oxidative stress, fibrosis, and cardiac dysfunction. HACE1 also protected against ferroptosis and mitochondrial damage. NRF2, a transcription factor implicated in oxidative stress, was identified as a target of HACE1, with HACE1 promoting NRF2 activity through ubiquitination. CONCLUSIONS HACE1 emerges as a potential therapeutic target and diagnostic marker for heart failure. It regulates oxidative stress, mitigates cardiac fibrosis, and protects against ferroptosis and mitochondrial damage. The study reveals that HACE1 achieves these effects, at least in part, through NRF2 activation via ubiquitination, offering insights into novel mechanisms for heart failure pathogenesis and potential interventions.
Collapse
Affiliation(s)
- Peiyi Yin
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Yongbin Wu
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Xiang Long
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Shuqiang Zhu
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Shiwei Chen
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Feng Lu
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Kun Lin
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Jianjun Xu
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| |
Collapse
|
4
|
Zhao X, Liu Y, Han X, Wang X, Qu C, Liu X, Yang B. Dapagliflozin attenuates the vulnerability to atrial fibrillation in rats with lipopolysaccharide-induced myocardial injury. Int Immunopharmacol 2023; 125:111038. [PMID: 38149574 DOI: 10.1016/j.intimp.2023.111038] [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: 03/20/2023] [Revised: 09/24/2023] [Accepted: 10/07/2023] [Indexed: 12/28/2023]
Abstract
BACKGROUND Oxidative stress is an essential component participating in the development and maintenance of atrial fibrillation (AF). Dapagliflozin, a SGLT2 inhibitor, has been shown to exert cardioprotective effects by ameliorating oxidative stress in multiple heart disease models. However, its potential to attenuate lipopolysaccharide (LPS)-induced myocardial injury in rats remains unknown. AIM This study aims to investigate the role of dapagliflozin in LPS-induced myocardial injury and the potential mechanisms involved. METHODS Rats were intraperitoneally administered LPS to induce sepsis-like condition. The intervention was conducted with intraperitoneal injection of dapagliflozin or saline 1 h in advance. The effects of dapagliflozin were detected by electrophysiological recordings, western blot, qPCR, ELISA, HE staining, immunohistochemistry and fluorescence. We further validated the mechanism in vitro using HL-1 cells. RESULTS Dapagliflozin significantly improved LPS-induced myocardial injury, reduced susceptibility to AF, and mitigated atrial tissue inflammatory cell infiltration and atrial myocyte apoptosis. These were correlated with the Nrf2/HO-1 signaling pathway, which subsequently reduced oxidative stress. Subsequently, we used a specific inhibitor of the Nrf2/HO-1 pathway in vitro, reversed the anti-oxidative stress effects of dapagliflozin on HL-1 cells, further confirming the Nrf2/HO-1 pathway's pivotal role in dapagliflozin-mediated cardioprotection. CONCLUSION Dapagliflozin ameliorated myocardial injury and susceptibility to AF induced by LPS through anti-oxidative stress, which relied on upregulation of the Nrf2/HO-1 pathway.
Collapse
Affiliation(s)
- Xin Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China
| | - Yating Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China
| | - Xueyu Han
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China
| | - Xiukun Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China
| | - Chuan Qu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China
| | - Xin Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China.
| | - Bo Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China.
| |
Collapse
|
5
|
Yan M, Liu T, Zhong P, Xiong F, Cui B, Wu J, Wu G. Chronic catestatin treatment reduces atrial fibrillation susceptibility via improving calcium handling in post-infarction heart failure rats. Peptides 2023; 159:170904. [PMID: 36375660 DOI: 10.1016/j.peptides.2022.170904] [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: 08/06/2022] [Revised: 11/09/2022] [Accepted: 11/10/2022] [Indexed: 11/13/2022]
Abstract
BACKGROUND Abnormal Ca2+ handling is a pivotal element of atrial fibrillation (AF) substrates. Catestatin (CST) modulates intracellular Ca2+ handling in cardiomyocytes (CMs). We investigated the effects of CST administration on atrial Ca2+ handling and AF susceptibility in rats with post-infarction heart failure (HF). METHODS Myocardial infarction (MI) was established by ligation of the left anterior descending coronary artery in rats. Two-week later, rats with post-infarction HF were randomly treated with saline (MI group) or CST (MI + CST group) for 4-week. Cellular Ca2+ imaging was performed by incubating atrial CMs with Fura-2 AM. An in vitro electrophysiological study was performed to assess the vulnerability to action potential duration (APD) alternans and AF. Ca2+ handling proteins expression was determined using western blotting. RESULTS In atrial CMs, compared with the sham group, the sarcoplasmic reticulum (SR) Ca2+ load, Ca2+ transient (CaT) amplitude, and threshold for Ca2+ alternans were significantly decreased, but the diastolic intracellular Ca2+ level, SR Ca2+ leakage, and spontaneous Ca2+ events were markedly increased in the MI group. However, CST attenuated these Ca2+-handling abnormalities induced by post-infarction HF. Moreover, vulnerability to atrial APD alternans and AF was significantly increased in isolated hearts from the MI group compared to the sham group, whereas all effects were prevented by CST. CST treatment also preserved SR Ca2+-ATPase protein expression but decreased the protein levels of phosphorylated-ryanodine receptor 2 and phosphorylated-Ca2+/calmodulin-dependent protein kinase II in atria from post-infarction HF rats. CONCLUSION Chronic CST treatment reduces AF vulnerability in rats with MI-induced HF by improving Ca2+ handling.
Collapse
Affiliation(s)
- Min Yan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Tao Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan 430060, China.
| | - Peng Zhong
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan 430060, China.
| | - Feng Xiong
- Montreal Heart Institute, Department of Medicine, University of Montreal, Montreal H1T 1C8, Quebec, Canada; Department of Pharmacology and Therapeutics, McGill University, Montreal H3G 1Y6, Quebec, Canada
| | - Bo Cui
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Jinchun Wu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Gang Wu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China; Hubei Key Laboratory of Cardiology, Wuhan 430060, China.
| |
Collapse
|
6
|
Role of Butylphthalide in Immunity and Inflammation: Butylphthalide May Be a Potential Therapy for Anti-Inflammation and Immunoregulation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:7232457. [PMID: 35422893 PMCID: PMC9005281 DOI: 10.1155/2022/7232457] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Accepted: 03/14/2022] [Indexed: 12/14/2022]
Abstract
Inflammation and immunity play an essential role in disease pathogenesis. 3-N-Butylphthalide (NBP), a group of compounds extracted from seeds of Apium graveolens (Chinese celery), has been demonstrated as an efficient and effective therapy for ischemic stroke. The amount of research on NBP protective effect is increasing at pace, such as microcircular reconstruction, alleviating inflammation, ameliorating brain edema and blood-brain barrier (BBB) damage, mitochondrial function protection, antiplatelet aggregation, antithrombosis, decreasing oxidative damage, and reducing neural cell apoptosis. There has been increasing research emphasizing the association between NBP and immunity and inflammation in the past few years. Hence, it is aimed at reviewing the related literature and summarizing the underlying anti-inflammatory and immunoregulatory function of NBP in various disorders.
Collapse
|
7
|
Exosomes from Bone Marrow Mesenchymal Stem Cells with Overexpressed Nrf2 Inhibit Cardiac Fibrosis in Rats with Atrial Fibrillation. Cardiovasc Ther 2022; 2022:2687807. [PMID: 35360547 PMCID: PMC8941574 DOI: 10.1155/2022/2687807] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 02/26/2022] [Accepted: 03/03/2022] [Indexed: 12/18/2022] Open
Abstract
Background Even though nuclear factor-erythroid 2-related factor 2 (Nrf2) signaling has been associated with the pathogenesis of multiple heart conditions, data on roles of Nrf2 within atrial fibrillation (AF) still remain scant. The present investigation had the aim of analyzing Nrf2-overexpressing role/s upon bone mesenchymal stem cell- (BMSC-) derived exosomes in rats with AF. Methods Exosomes were collected from control or Nrf2 lentivirus-transduced BMSCs and then injected into rats with AF through the tail vein. AF duration was observed using electrocardiography. Immunohistochemical staining was then employed for assessing Nrf2, HO-1, α-SMA, collagen I, or TGF-β1 expression profiles within atrial myocardium tissues. Conversely, Masson staining was utilized to evaluate atrial fibrosis whereas apoptosis within myocardia was evaluated through TUNEL assays. In addition, TNF-α, IL-1β, IL-4, or IL-10 serum expression was assessed through ELISA. Results Results of the current study showed significant downregulation of Nrf2/HO-1 within AF rat myocardia. It was found that injection of the control or Lv-Nrf2 exosomes significantly alleviated and lowered AF timespans together with reducing cardiomyocyte apoptosis. Moreover, injection of Lv-Nrf2 exosomes essentially lowered AF-driven atrial fibrosis and also inhibited inflammatory responses in the rats with AF. Conclusion Delivery of BMSC-derived exosomes using overexpressed Nrf2 inhibited AF-induced arrhythmias, myocardial fibrosis, apoptosis, and inflammation via Nrf2/HO-1 pathway triggering.
Collapse
|
8
|
Qiu H, Huang ZY, Cao H, Zhang Z, Ma J, Li XQ, Huang S, Li X, Qiu W, Zhao Z, Ji C, Huang L, Jiang W, Yang ZQ, Xian SX, Wu H, Lu W, Ding C. Deciphering mechanism of the herbal formula WuShen in the treatment of postinfarction heart failure. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 95:153878. [PMID: 34929563 DOI: 10.1016/j.phymed.2021.153878] [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: 09/24/2021] [Revised: 11/15/2021] [Accepted: 12/02/2021] [Indexed: 06/14/2023]
Abstract
BACKGROUND Numerous clinical studies reported the effectiveness of herbal formula WuShen (WS) in treating cardiovascular diseases, yet relevant basic research was rarely conducted. METHODS AND RESULTS Twelve main bioactive compounds of WS decoction were identified using the ultra-performance liquid chromatography-LTQ-Orbitrap mass spectrometer. A total of 137 active compounds with 613 targets were predicted by network pharmacology; their bioinformatic annotation and human microarray data suggested that wounding healing, inflammatory response, and gap junction were potentially the major therapeutic modules. A rat model of post-myocardial infarction (MI) heart failure (HF) was used to study the effects of WS on cardiac function, adverse cardiac remodeling, and experimental arrhythmias. Rats treated with WS led to a significantly improved pump function and reduced susceptibility to both ventricular tachycardia and atrial fibrillation, and restricted adverse cardiac remodeling partly via inhibiting TGFβ1/SMADs mediated extracellular matrix deposition and Rac1/NOX2/CTGF/Connexin43 -involved gap junction remodeling. CONCLUSIONS The present study highlights that WS can be applied to the treatment of heart failure and the upstream therapy for atrial fibrillation and ventricular tachycardia through its preventive effect on adverse cardiac remodeling.
Collapse
Affiliation(s)
- Huiliang Qiu
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510006, P.R. China; Department of Cardiology, Guangdong Provincial Hospital of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, P.R. China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510020, P.R. China; Division of Cardiology, University of California, San Francisco, San Francisco CA 94143, United States of America
| | - Zeng-Yan Huang
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510405, P.R. China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, P.R. China
| | - Haiming Cao
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510006, P.R. China; Department of Cardiology, Guangdong Provincial Hospital of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, P.R. China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510020, P.R. China
| | - Zezhao Zhang
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510006, P.R. China; Department of Cardiology, Guangdong Provincial Hospital of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, P.R. China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510020, P.R. China
| | - Jin Ma
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510006, P.R. China; Department of Cardiology, Guangdong Provincial Hospital of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, P.R. China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510020, P.R. China
| | - Xiao-Qing Li
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510006, P.R. China; Department of Cardiology, Guangdong Provincial Hospital of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, P.R. China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510020, P.R. China
| | - Shen Huang
- Department of Cardiology, Affiliated Hospital of Chengdu University, Chengdu 610081, P.R. China
| | - Xiong Li
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510006, P.R. China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510020, P.R. China
| | - Wencong Qiu
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510006, P.R. China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510020, P.R. China
| | - Zicong Zhao
- Foshan Hospital of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Foshan 528000, P.R. China
| | - Chunlan Ji
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510006, P.R. China; Department of Cardiology, Guangdong Provincial Hospital of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, P.R. China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510020, P.R. China
| | - Lihua Huang
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510006, P.R. China; Department of Cardiology, Guangdong Provincial Hospital of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, P.R. China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510020, P.R. China
| | - Wei Jiang
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510006, P.R. China; Department of Cardiology, Guangdong Provincial Hospital of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, P.R. China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510020, P.R. China; Guangdong Provincial Key Laboratory of Chinese Medicine for Prevention and Treatment of Refractory Chronic Diseases, Guangzhou 510020, P.R. China
| | - Zhong-Qi Yang
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510405, P.R. China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, P.R. China
| | - Shao-Xiang Xian
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510405, P.R. China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, P.R. China
| | - Huanlin Wu
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510006, P.R. China; Department of Cardiology, Guangdong Provincial Hospital of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, P.R. China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510020, P.R. China; Department of internal medicine, Beijing University of Chinese Medicine, Beijing 100000, P.R. China
| | - Weihui Lu
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510006, P.R. China; Department of Cardiology, Guangdong Provincial Hospital of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, P.R. China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510020, P.R. China.
| | - Chunhua Ding
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510006, P.R. China; Department of Cardiology, Guangdong Provincial Hospital of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, P.R. China; Cardiac Department, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing 100049, P.R. China.
| |
Collapse
|
9
|
Yu Y, Zhu Y, Sun X, Li Y, Wang M, Dong B, Sun X, Hou W. DL-3-n-butylphthalide protects H9c2 cardiomyoblasts from ischemia/reperfusion injury by regulating HSP70 expression via PI3K/AKT pathway activation. Exp Ther Med 2021; 22:1008. [PMID: 34345290 PMCID: PMC8311253 DOI: 10.3892/etm.2021.10441] [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: 05/20/2021] [Accepted: 06/21/2021] [Indexed: 02/07/2023] Open
Abstract
DL-3-n-butylphthalide (NBP) is commonly used to treat ischemic strokes due to its antioxidative and anti-inflammatory effects. The present study aimed to examine the protective effects of NBP on myocardial ischemia-reperfusion injury (MIRI) by establishing a MIRI model in H9c2 cells. Cell viability assay using Cell Counting Kit-8, lactate dehydrogenase (LDH) cytotoxicity and lipid peroxidation malondialdehyde (MDA) content were assessed to detect cell activity, degree of cell injury and oxidative stress reaction. Reverse transcription-quantitative PCR was used to quantify the expression of inflammatory factors in H9c2 cells. Western blotting and immunofluorescence staining were used to detect the protein expression of PI3K/AKT and heat shock protein 70 (HSP70). The present results indicated that NBP significantly increased cell viability during ischemia-reperfusion. Moreover, NBP inhibited the release of LDH and the production of MDA. NBP treatment also significantly decreased the expression of inflammatory factors at the mRNA level. Additionally, NBP activated the PI3K/AKT pathway and upregulated the expression of HSP70 compared with cells in the MIRI model. LY294002, a PI3K inhibitor, reversed the protective effects of NBP and suppressed the expression of HSP70. The present study demonstrated that NBP protected H9c2 cells from MIRI by regulating HSP70 expression via PI3K/AKT pathway activation.
Collapse
Affiliation(s)
- Yunchen Yu
- Department of Cardiovascular Surgery, Affiliated Hospital of Weifang Medical University, Weifang, Shandong 261000, P.R. China
- Department of Anesthesiology, Qingdao Fuwai Cardiovascular Hospital, Qingdao, Shandong 266000, P.R. China
- School of Anesthesiology, Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, Weifang Medical University, Weifang, Shandong 261000, P.R. China
| | - Yuying Zhu
- School of Anesthesiology, Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, Weifang Medical University, Weifang, Shandong 261000, P.R. China
| | - Xiaotong Sun
- School of Anesthesiology, Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, Weifang Medical University, Weifang, Shandong 261000, P.R. China
| | - Yongxing Li
- Department of Cardiovascular Surgery, Affiliated Hospital of Weifang Medical University, Weifang, Shandong 261000, P.R. China
| | - Mingling Wang
- School of Anesthesiology, Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, Weifang Medical University, Weifang, Shandong 261000, P.R. China
| | - Bin Dong
- School of Anesthesiology, Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, Weifang Medical University, Weifang, Shandong 261000, P.R. China
| | - Xiaodong Sun
- Department of Endocrinology, Affiliated Hospital of Weifang Medical University, Weifang, Shandong 261000, P.R. China
| | - Wenming Hou
- Department of Cardiovascular Surgery, Affiliated Hospital of Weifang Medical University, Weifang, Shandong 261000, P.R. China
- Correspondence to: Professor Wenming Hou, Department of Cardiovascular Surgery, Affiliated Hospital of Weifang Medical University, 2428 Yuhe Road, Weifang, Shandong 261000, P.R. China
| |
Collapse
|
10
|
Han B, Xu J, Shi X, Zheng Z, Shi F, Jiang F, Han J. DL-3-n-Butylphthalide Attenuates Myocardial Hypertrophy by Targeting Gasdermin D and Inhibiting Gasdermin D Mediated Inflammation. Front Pharmacol 2021; 12:688140. [PMID: 34168567 PMCID: PMC8217660 DOI: 10.3389/fphar.2021.688140] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 05/26/2021] [Indexed: 11/18/2022] Open
Abstract
Pressure overload leads to a hypertrophic milieu that produces deleterious cardiac dysfunction. Inflammation is a key pathophysiological mechanism underpinning myocardial hypertrophy. DL-3-n-butylphthalide (NBP), a neuroprotective agent, also has potent cardioprotective effects. In this study, the potential of NBP to antagonize myocardial hypertrophy was evaluated in C57BL/6 mice in vivo and in rat primary cardiomyocytes in vitro. In mice, NBP treatment reduced cardiac hypertrophy and dysfunction in a transverse aortic constriction (TAC)-induced pressure overload model. In angiotensin (Ang) II-challenged cardiomyocytes, NBP prevents cell size increases and inhibits gasdermin D (GSDMD)-mediated inflammation. Furthermore, overexpression of GSDMD-N reduced the protective effects of NBP against Ang II-induced changes. Using molecular docking and MD simulation, we found that the GSDMD-N protein may be a target of NBP. Our study shows that NBP attenuates myocardial hypertrophy by targeting GSDMD and inhibiting GSDMD-mediated inflammation.
Collapse
Affiliation(s)
- Bingjiang Han
- Department of Cardiology, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Jiajun Xu
- Department of Cardiology, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Xiaowen Shi
- Department of Cardiology, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Zhanxiong Zheng
- Department of Cardiology, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Fengjie Shi
- Department of Cardiology, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Fenfen Jiang
- Department of Cardiology, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Jibo Han
- Department of Cardiology, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| |
Collapse
|
11
|
Jiang J, Gu X, Wang H, Ding S. Resveratrol improves cardiac function and left ventricular fibrosis after myocardial infarction in rats by inhibiting NLRP3 inflammasome activity and the TGF- β1/SMAD2 signaling pathway. PeerJ 2021; 9:e11501. [PMID: 34123595 PMCID: PMC8166236 DOI: 10.7717/peerj.11501] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 05/02/2021] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Several studies have shown that resveratrol (RES), a naturally occurring polyphenol found in many plants, is beneficial for preventing cardiovascular diseases. However, the mechanism underlying the RES-mediated protection against myocardial infarction has not yet been revealed entirely. In this study, we investigated the protective effects of RES on cardiac function in a rat model of acute myocardial infarction (AMI) and the related underlying mechanisms. METHODS Male Sprague-Dawley rats were randomly divided into four groups: Sham (sham operation), Sham-RES, AMI (AMI induction), and AMI-RES. The rat AMI model was established by the permanent ligation of left anterior descending coronary artery method. The rats in the RES-treated groups were gavaged with RES (50 mg/kg/day) daily for 45 days after the Sham operation or AMI induction; rats in the Sham and AMI groups were gavaged with deionized water. Cardiac function was evaluated by echocardiography. Atrial interstitial fibrosis was assessed by hematoxylin-eosin or Masson's trichrome staining. Real-time PCR and western blotting analyses were performed to examine the levels of signaling pathway components. RESULTS RES supplementation decreased the inflammatory cytokine levels, improved the cardiac function, and ameliorated atrial interstitial fibrosis in the rats with AMI. Furthermore, RES supplementation inhibited NLRP3 inflammasome activity, decreased the TGF-β1 production, and downregulated the p-SMAD2/SMAD2 expression in the heart. CONCLUSION RES shows notable cardioprotective effects in a rat model of AMI; the possible mechanisms underlying these effects may involve the improvement of cardiac function and atrial interstitial fibrosis via the RES-mediated suppression of NLRP3 inflammasome activity and inhibition of the TGF-β1/SMAD2 signaling pathway in the heart.
Collapse
Affiliation(s)
- Jinjin Jiang
- Jiangsu Vocational College of Medicine, Yancheng, Jiangsu, China
| | - Xiuping Gu
- Department of Cardiology, General Hospital of TISCO, Taiyuan, Shanxi, China
| | - Huifeng Wang
- Department of Cardiology, General Hospital of TISCO, Taiyuan, Shanxi, China
| | - Shibin Ding
- Jiangsu Vocational College of Medicine, Yancheng, Jiangsu, China
| |
Collapse
|
12
|
Liu Y, Yin Z, Xu X, Liu C, Duan X, Song Q, Tuo Y, Wang C, Yang J, Yin S. Crosstalk between the activated Slit2-Robo1 pathway and TGF-β1 signalling promotes cardiac fibrosis. ESC Heart Fail 2021; 8:447-460. [PMID: 33236535 PMCID: PMC7835586 DOI: 10.1002/ehf2.13095] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 09/27/2020] [Accepted: 10/22/2020] [Indexed: 12/20/2022] Open
Abstract
AIMS Previous reports indicated that the Slit2-Robo signalling pathway is involved in embryonic heart development and fibrosis in other solid organs, but its function in adult cardiac fibrosis has not been investigated. Here, we investigate the role of the Slit2-Robo1 signalling pathway in cardiac fibrosis. METHODS AND RESULTS The right atrial tissue samples were obtained from patients with valvular heart disease complicated by atrial fibrillation during heart valve surgery and from healthy heart donors. The fibrotic animal model is created by performing transverse aortic constriction (TAC) surgery. The Robo1, Slit2, TGF-β1, and collagen I expression levels in human and animal samples were evaluated by immunohistochemistry and western blot analysis. Echocardiography measured the changes in heart size and cardiac functions of animals. Angiotensin II (Ang II), Slit2-siRNA, TGF-β1-siRNA, recombinant Slit2, and recombinant TGF-β1 were transfected to cardiac fibroblasts (CFs) respectively to observe their effects on collagen I expression level. The right atrial appendage of patients with valvular heart disease complicated by atrial fibrillation found significantly up-regulated Slit2, Robo1, TGF-β1, and collagen I expression levels. TAC surgery leads to heart enlargement, cardiac fibrosis, and up-regulation of Slit2, Robo1, TGF-β1, and collagen I expression levels in animal model. Robo1 antagonist R5 and TGF-β1 antagonist SB431542 suppressed cardiac fibrosis in TAC mice. Treatment with 100 nM Ang II in CFs caused significantly increased Slit2, Robo1, Smad2/3, TGF-β1, collagen I, PI3K, and Akt expression levels. Transfecting Slit2-siRNA and TGF-β1-siRNA, respectively, into rat CFs significantly down-regulated Smad2/3 and collagen I expression, inhibiting the effects of Ang II. Recombinant Slit2 activated the TGF-β1/Smad signalling pathway in CFs and up-regulated Periostin, Robo1, and collagen I expression. CONCLUSIONS The Slit2-Robo1 signalling pathway interfered with the TGF-β1/Smad pathway and promoted cardiac fibrosis. Blockade of Slit2-Robo1 might be a new treatment for cardiac fibrosis.
Collapse
Affiliation(s)
- Yunqi Liu
- Department of Cardiac Surgery, The First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
- NCH Key Laboratory of Assisted CirculationSun Yat‐sen UniversityGuangzhouChina
| | - Ziwei Yin
- Division of BiosciencesUniversity College LondonLondonUK
| | - Xueqin Xu
- Department of Cardiac Surgery, The First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
- NCH Key Laboratory of Assisted CirculationSun Yat‐sen UniversityGuangzhouChina
| | - Chen Liu
- NCH Key Laboratory of Assisted CirculationSun Yat‐sen UniversityGuangzhouChina
- Department of Cardiology, The First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Xiaoying Duan
- Department of Cardiac Surgery, The First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
- NCH Key Laboratory of Assisted CirculationSun Yat‐sen UniversityGuangzhouChina
| | - Qinlan Song
- Department of Cardiac Surgery, The First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
- NCH Key Laboratory of Assisted CirculationSun Yat‐sen UniversityGuangzhouChina
| | - Ying Tuo
- Department of Pathology, The First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Cuiping Wang
- Department of Cardiothoracic Surgery ICU, The First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Jing Yang
- Department of Pediatric Dentistry, Stomatological HospitalSouthern Medical UniversityGuangzhouChina
| | - Shengli Yin
- Department of Cardiac Surgery, The First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
- NCH Key Laboratory of Assisted CirculationSun Yat‐sen UniversityGuangzhouChina
| |
Collapse
|
13
|
Marco-Contelles J, Zhang Y. From Seeds of Apium graveolens Linn. to a Cerebral Ischemia Medicine: The Long Journey of 3- n-Butylphthalide. J Med Chem 2020; 63:12485-12510. [PMID: 32672958 DOI: 10.1021/acs.jmedchem.0c00887] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
3-n-Butylphthalide (NBP) as well as its derivatives and analogues (NBPs), in racemic or enantiomerically pure forms, possess potent and diverse pharmacological properties and have shown a great potential therapeutic interest for many human conditions, especially for cerebral ischemia. This Perspective outlines the synthesis and therapeutic applications of NBPs.
Collapse
Affiliation(s)
- José Marco-Contelles
- Laboratory of Medicinal Chemistry, Institute of Organic Chemistry, CSIC, Juan de la Cierva, 3, 28006-Madrid, Spain
| | - Yihua Zhang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, PR China.,Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing 210009, PR China.,Center of Drug Discovery, China Pharmaceutical University, Nanjing 210009, PR China
| |
Collapse
|
14
|
Adaptive Autophagy Offers Cardiorenal Protection in Rats with Acute Myocardial Infarction. Cardiol Res Pract 2020; 2020:7158975. [PMID: 32655948 PMCID: PMC7322605 DOI: 10.1155/2020/7158975] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 04/14/2020] [Accepted: 05/15/2020] [Indexed: 01/19/2023] Open
Abstract
Objective Understanding the multifactorial changes involved in the kidney and heart after acute myocardial infarction (AMI) is prerequisite for further mechanisms and early intervention, especially autophagy changes. Here, we discussed the role of adaptive autophagy in the heart and kidney of rats with AMI. Methods A rat model of AMI was established by ligating the left anterior descending branch of the coronary artery. Animals were sacrificed at 2 and 4 weeks after the operation to assess the morphological and functional changes of the heart and kidney, as well as the autophagy pathway. In vitro, HK-2 and AC16 cell injuries and the autophagy pathway were assayed after autophagy was inhibited by 3-methyladenine (3-MA) in a hypoxia incubator. Results We found that the left ventricular systolic pressure (LVSP) significantly decreased in the model group at weeks 2 and 4. At weeks 2 and 4, the level of urinary kidney injury molecule 1 (uKIM1) of the model group was significantly higher than the sham group. At week 4, urinary neutrophil gelatinase-associated lipocalcin (uNGAL) and urinary albumin also significantly increased. At week 2, microtubule-associated protein 1 light chain 3-II (LC3-II), ATG5, and Beclin1 were significantly elevated in the heart and kidney compared with the sham-operated rats, but there was no change in p62 levels. At week 4, LC3-II did not significantly increase and p62 levels significantly increased. In addition, 3-MA markedly increased KIM1, NGAL, and the activity of caspase-3 in the hypoxic HK-2 and AC16 cell. Conclusion Autophagy will undergo adaptive changes and play a protective role in the heart and kidney of rats after AMI.
Collapse
|
15
|
Deng B, Zhang X, Liang Y, Jiang H, Huang W, Wu Y, Deng W. Nonadherent culture method promotes MSC-mediated vascularization in myocardial infarction via miR-519d/VEGFA pathway. Stem Cell Res Ther 2020; 11:266. [PMID: 32616068 PMCID: PMC7330937 DOI: 10.1186/s13287-020-01780-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 06/12/2020] [Accepted: 06/18/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) can provide therapeutic benefits for myocardial infarction (MI) recovery; however, the molecular mechanism by which MSCs improve the heart function is unclear. METHODS Microarray analysis was performed to examine the expression profiling of human MSCs (hMSCs) grown as adherent cultures (AC-hMSCs) or nonadherent cultures on ultra-low-adherent plates (nonAC-hMSCs). Real-time quantitative polymerase chain reaction (RT-qPCR), western blotting, and enzyme-linked immunosorbent assays (ELISA) were used to assess VEGFA expression and secretion in the AC-hMSCs and nonAC-hMSCs. The paracrine effect of VEGFA-overexpressing AC-MSCs (AC-VEGFA-hMSCs) or VEGFA-knockdown nonAC-hMSCs (nonAC-shVEGFA-hMSCs) on the angiogenic ability of human umbilical vein endothelial cells (HUVECs) was evaluated using tube formation assay. AC-VEGFA-hMSCs or nonAC-shVEGFA-hMSCs were transplanted into myocardial infarction rats to investigate the therapeutic effect of AC-VEGFA-hMSCs or nonAC-shVEGFA-hMSCs. Luciferase reporter assay was used to confirm the association of VEGFA with miR-519d. RESULTS Microarray analysis revealed that VEGFA is downregulated in AC-hMSCs compared to nonAC-hMSCs. Functional assays revealed that high levels of VEGFA produced from AC-VEGFA-hMSCs increased the tube formation capacity of HUVECs in vitro, improved angiogenesis and cardiac performance, and reduced infarct size in a rat MI model. Low levels of VEGFA secretion from nonAC-shVEGFA-hMSCs had the opposite effects. Mechanistically, we found that miR-519d directly targets VEGFA. High levels of VEGFA secreted from VEGFA-overexpressing nonAC-hMSCs abolished the repressive effect of miR-519d on HUVEC angiogenesis. CONCLUSION Our findings indicate that nonadherent culture-induced secretion of VEGFA plays an important role in MSCs via the miR-519d/VEGFA pathway and may provide a novel therapeutic strategy for MI treatment.
Collapse
Affiliation(s)
- Baoping Deng
- Department of Cardiothoracic Surgery, Zhongshan People's Hospital, 2 Sunwen East Road, Zhongshan, Guangdong, 528403, People's Republic of China
- Department of Vascular Surgery, Affiliated Hospital of Guilin Medical University, Guilin, 541001, People's Republic of China
| | - Xianlan Zhang
- Department of Vascular Surgery, Affiliated Hospital of Guilin Medical University, Guilin, 541001, People's Republic of China
| | - Yi Liang
- Department of Cardiothoracic Surgery, Zhongshan People's Hospital, 2 Sunwen East Road, Zhongshan, Guangdong, 528403, People's Republic of China
| | - Haiming Jiang
- Department of Cardiothoracic Surgery, Zhongshan People's Hospital, 2 Sunwen East Road, Zhongshan, Guangdong, 528403, People's Republic of China
| | - Weizhao Huang
- Department of Cardiothoracic Surgery, Zhongshan People's Hospital, 2 Sunwen East Road, Zhongshan, Guangdong, 528403, People's Republic of China
| | - Yinmeng Wu
- Department of Cardiothoracic Surgery, Zhongshan People's Hospital, 2 Sunwen East Road, Zhongshan, Guangdong, 528403, People's Republic of China
| | - Weiping Deng
- Department of Gastroenterology, Taihe Hospital, Hubei University of Medicine, 32 Ren Min South Road, Shiyan, 442000, Hubei, People's Republic of China.
| |
Collapse
|
16
|
Li X, Xue YM, Guo HM, Deng CY, Peng DW, Yang H, Wei W, Liu Y, Liu FZ, Wang ZY, Zhang MZ, Rao F, Wu SL. High hydrostatic pressure induces atrial electrical remodeling through upregulation of inflammatory cytokines. Life Sci 2019; 242:117209. [PMID: 31870776 DOI: 10.1016/j.lfs.2019.117209] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 12/10/2019] [Accepted: 12/18/2019] [Indexed: 01/01/2023]
Abstract
AIMS Hypertension is an independent risk factor for atrial fibrillation (AF). However, the direct effect of hydrostatic pressure on atrial electrical remodeling is unclear. The present study investigated whether hydrostatic pressure is responsible for atrial electrical remodeling and addressed a potential role of inflammation in this pathology. MAIN METHODS Whole-cell patch-clamp recordings and biochemical assays were used to study the regulation and expression of ion channels in left atrial appendages in patients with AF, spontaneously hypertensive rats (SHRs), and atrium-derived cells (HL-1 cells) exposed to standard (0 mmHg) and elevated (20, 40 mmHg) hydrostatic pressure. KEY FINDINGS Both TNF-α and MIF were highly expressed in patients with AF and SHRs. AF inducibility in SHRs was higher after atrial burst pacing, accompanied by a decrease in the L-type calcium current (ICa,L), an increase in the transient outward K+ current (Ito) and ultra-rapid delayed rectifier K+ current (IKur), and a shortened action potential duration (APD), which could be inhibited by atorvastatin. Furthermore, exposure to elevated pressure was associated with electrical remodeling of the HL-1 cells. The peak current density of ICa,L was reduced, while Ito and IKur were increased. Moreover, the expression levels of Kv4.3, Kv1.5, TNF-α, and MIF were upregulated, while the expression of Cav1.2 was downregulated in HL-1 cells after treatment with high hydrostatic pressure (40 mmHg). Atorvastatin alleviated the electrical remodeling and increased inflammatory markers in HL-1 cells induced by high hydrostatic pressure. SIGNIFICANCE Elevated hydrostatic pressure led to atrial electrical remodeling and increased AF susceptibility by upregulating inflammation.
Collapse
Affiliation(s)
- Xin Li
- Guangdong Provincial Key Laboratory of Clinical Pharmacology, Department of Cardiology, Guangdong Cardiovascular Institute, Guangzhou 510080, China; Research Center of Medical Sciences, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Yu-Mei Xue
- Guangdong Provincial Key Laboratory of Clinical Pharmacology, Department of Cardiology, Guangdong Cardiovascular Institute, Guangzhou 510080, China; Research Center of Medical Sciences, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Hui-Ming Guo
- Department of Cardiac Surgery, Guangdong Provincial People's Hospital, Guangzhou 510080, China
| | - Chun-Yu Deng
- Guangdong Provincial Key Laboratory of Clinical Pharmacology, Department of Cardiology, Guangdong Cardiovascular Institute, Guangzhou 510080, China; Research Center of Medical Sciences, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - De-Wei Peng
- Guangdong Provincial Key Laboratory of Clinical Pharmacology, Department of Cardiology, Guangdong Cardiovascular Institute, Guangzhou 510080, China; Research Center of Medical Sciences, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Hui Yang
- Guangdong Provincial Key Laboratory of Clinical Pharmacology, Department of Cardiology, Guangdong Cardiovascular Institute, Guangzhou 510080, China; Research Center of Medical Sciences, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Wei Wei
- Guangdong Provincial Key Laboratory of Clinical Pharmacology, Department of Cardiology, Guangdong Cardiovascular Institute, Guangzhou 510080, China; Research Center of Medical Sciences, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Yang Liu
- Guangdong Provincial Key Laboratory of Clinical Pharmacology, Department of Cardiology, Guangdong Cardiovascular Institute, Guangzhou 510080, China; Research Center of Medical Sciences, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Fang-Zhou Liu
- Guangdong Provincial Key Laboratory of Clinical Pharmacology, Department of Cardiology, Guangdong Cardiovascular Institute, Guangzhou 510080, China; Research Center of Medical Sciences, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Zhao-Yu Wang
- Guangdong Provincial Key Laboratory of Clinical Pharmacology, Department of Cardiology, Guangdong Cardiovascular Institute, Guangzhou 510080, China; Research Center of Medical Sciences, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Meng-Zhen Zhang
- Research Center of Medical Sciences, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Fang Rao
- Guangdong Provincial Key Laboratory of Clinical Pharmacology, Department of Cardiology, Guangdong Cardiovascular Institute, Guangzhou 510080, China; Research Center of Medical Sciences, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China.
| | - Shu-Lin Wu
- Guangdong Provincial Key Laboratory of Clinical Pharmacology, Department of Cardiology, Guangdong Cardiovascular Institute, Guangzhou 510080, China; Research Center of Medical Sciences, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China.
| |
Collapse
|
17
|
Sun W, Wu Y, Gao M, Tian Y, Qi P, Shen Y, Huang L, Shi L, Wang Y, Liu X. C-reactive protein promotes inflammation through TLR4/NF-κB/TGF-β pathway in HL-1 cells. Biosci Rep 2019; 39:BSR20190888. [PMID: 31391207 PMCID: PMC6712437 DOI: 10.1042/bsr20190888] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 07/25/2019] [Accepted: 08/02/2019] [Indexed: 01/08/2023] Open
Abstract
Atrial fibrillation (AF) is the most common type of heart arrhythmia. Currently, the pathogenesis of AF is not fully understood yet. A growing body of evidence highlighted the strong association between inflammation and the pathogenesis of AF. C-reactive protein (CRP) is an inflammation marker with increased expression in AF. Therefore, the aim of this study was to determine if CRP promotes inflammation, which may sequentially mediate the onset of AF and the concurrent atrial fibrosis, through TLR4/NF-κB/TGF-β pathway. HL-1 cells were treated with either 25 or 50 μg/ml recombinant human CRP. TGF-β1 and NF-κB inhibitors were given either solely or together to the 50 μg/ml CRP-treated cells. Cell proliferation, apoptosis, the expression of apoptotic factors and TLR4, IL-6, TGF-β1, Smad2, and the phosphorylation of Smad2 were determined. Data showed that CRP induced dose-dependent inhibition on cell proliferation and promoted cell apoptosis, which was induced through both intrinsic and extrinsic pathways. Such effects were reversed by inhibiting TGF-β1 and/or NF-κB. Inhibition of TGF-β1 and/or NF-κB also reduced the expression of TLR4 and IL-6. Inhibition of NF-κB alone weakened the expression of TGF-β1 and phosphorylation of Smad2. Our study demonstrated that CRP is not only a marker, but also an important mediator in the induction of inflammation and likely the pathogenesis of AF. We for the first time reported CRP-induced activation and cross-talk between TLR4 and NF-κB/TGF-β1 signaling pathway in a cardiomyocyte model. Reducing CRP and targeting TLR4/NF-κB/TGF-β1 pathway may provide new insights in the therapeutic interventions to inflammation-induced AF.
Collapse
Affiliation(s)
- Weiping Sun
- Department of Cardiology, Beijing Chao-yang Hospital, Capital Medical University, Beijing 100037, China
- Department of Cardiology, Beijing Luhe Hospital, Capital Medical University, Beijing 100037, China
| | - Yongquan Wu
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Mingyang Gao
- Department of Cardiology, Beijing Chao-yang Hospital, Capital Medical University, Beijing 100037, China
| | - Ying Tian
- Department of Cardiology, Beijing Chao-yang Hospital, Capital Medical University, Beijing 100037, China
| | - Peng Qi
- Department of Cardiology, Beijing Chao-yang Hospital, Capital Medical University, Beijing 100037, China
| | - Yujing Shen
- Department of Cardiology, Fuwai Hospital, Chinese academy of Medicine sciences, Beijing 100037, China
| | - Lihong Huang
- Department of Cardiology, Beijing Chao-yang Hospital, Capital Medical University, Beijing 100037, China
| | - Liang Shi
- Department of Cardiology, Beijing Chao-yang Hospital, Capital Medical University, Beijing 100037, China
| | - Yanjiang Wang
- Department of Cardiology, Beijing Chao-yang Hospital, Capital Medical University, Beijing 100037, China
| | - Xingpeng Liu
- Department of Cardiology, Beijing Chao-yang Hospital, Capital Medical University, Beijing 100037, China
| |
Collapse
|
18
|
Chen XQ, Qiu K, Liu H, He Q, Bai JH, Lu W. Application and prospects of butylphthalide for the treatment of neurologic diseases. Chin Med J (Engl) 2019; 132:1467-1477. [PMID: 31205106 PMCID: PMC6629339 DOI: 10.1097/cm9.0000000000000289] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Indexed: 01/30/2023] Open
Abstract
OBJECTIVE The 3-N-butylphthalide (NBP) comprises one of the chemical constituents of celery oil. It has a series of pharmacologic mechanisms including reconstructing microcirculation, protecting mitochondrial function, inhibiting oxidative stress, inhibiting neuronal apoptosis, etc. Based on the complex multi-targets of pharmacologic mechanisms of NBP, the clinical application of NBP is increasing and more clinical researches and animal experiments are also focused on NBP. The aim of this review was to comprehensively and systematically summarize the application of NBP on neurologic diseases and briefly summarize its application to non-neurologic diseases. Moreover, recent progress in experimental models of NBP on animals was summarized. DATA SOURCES Literature was collected from PubMed and Wangfang database until November 2018, using the search terms including "3-N-butylphthalide," "microcirculation," "mitochondria," "ischemic stroke," "Alzheimer disease," "vascular dementia," "Parkinson disease," "brain edema," "CO poisoning," "traumatic central nervous system injury," "autoimmune disease," "amyotrophic lateral sclerosis," "seizures," "diabetes," "diabetic cataract," and "atherosclerosis." STUDY SELECTION Literature was mainly derived from English articles or articles that could be obtained with English abstracts and partly derived from Chinese articles. Article type was not limited. References were also identified from the bibliographies of identified articles and the authors' files. RESULTS NBP has become an important adjunct for ischemic stroke. In vascular dementia, the clinical application of NBP to treat severe cognitive dysfunction syndrome caused by the hypoperfusion of brain tissue during cerebrovascular disease is also increasing. Evidence also suggests that NBP has a therapeutic effect for neurodegenerative diseases. Many animal experiments have found that it can also improve symptoms in other neurologic diseases such as epilepsy, cerebral edema, and decreased cognitive function caused by severe acute carbon monoxide poisoning. Moreover, NBP has therapeutic effects for diabetes, diabetes-induced cataracts, and non-neurologic diseases such as atherosclerosis. Mechanistically, NBP mainly improves microcirculation and protects mitochondria. Its broad pharmacologic effects also include inhibiting oxidative stress, nerve cell apoptosis, inflammatory responses, and anti-platelet and anti-thrombotic effects. CONCLUSIONS The varied pharmacologic mechanisms of NBP involve many complex molecular mechanisms; however, there many unknown pharmacologic effects await further study.
Collapse
Affiliation(s)
- Xi-Qian Chen
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | | | | | | | | | | |
Collapse
|
19
|
Qiu H, Ji C, Liu W, Wu Y, Lu Z, Lin Q, Xue Z, Liu X, Wu H, Jiang W, Zou C. Chronic Kidney Disease Increases Atrial Fibrillation Inducibility: Involvement of Inflammation, Atrial Fibrosis, and Connexins. Front Physiol 2018; 9:1726. [PMID: 30564139 PMCID: PMC6288485 DOI: 10.3389/fphys.2018.01726] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 11/16/2018] [Indexed: 12/13/2022] Open
Abstract
Chronic kidney disease (CKD) causes atrial structural remodeling and subsequently increases the incidence of atrial fibrillation (AF). Atrial connexins and inflammatory responses may be involved in this remodeling process. In this study, nephrectomy was used to produce the CKD rat model. Three months post-nephrectomy, cardiac structure, function and AF vulnerability were quantified using echocardiography and electrophysiology methods. The left atrial tissue was tested for quantification of fibrosis and inflammation, and for the distribution and expression of connexin (Cx) 40 and Cx43. An echocardiography showed that CKD resulted in the left atrial enlargement and left ventricular hypertrophy, but had no functional changes. CKD caused a significant increase in the AF inducible rate (91.11% in CKD group vs. 6.67% in sham group, P < 0.001) and the AF duration [107 (0–770) s in CKD vs. 0 (0–70) s in sham, P < 0.001] with prolonged P-wave duration. CKD induced severe interstitial fibrosis, activated the transforming growth factor-β1/Smad2/3 pathway with a massive extracellular matrix deposition of collagen type I and α-smooth muscle actin, and matured the NLR (nucleotide-binding domain leucine-rich repeat-containing receptor) pyrin domain-containing protein 3 (NLRP3) inflammasome with an inflammatory cascade response. CKD resulted in an increase in non-phosphorylated-Cx43, a decrease in Cx40 and phosphorylated-Cx43, and lateralized the distribution of Cx40 and Cx43 proteins with upregulations of Rac-1, connective tissue growth factor and N-cadherin. These findings implicate the transforming growth factor-β1/Smad2/3, the NLRP3 inflammasome and the connexins as potential mediators of increased AF vulnerability in CKD.
Collapse
Affiliation(s)
- Huiliang Qiu
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Cardiology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Chunlan Ji
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Nephrology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Wei Liu
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Cardiology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Yuchi Wu
- Department of Nephrology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Zhaoyu Lu
- Department of Nephrology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Qizhan Lin
- Department of Nephrology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Zheng Xue
- Department of Cardiology, Guangzhou Hospital of Traditional Chinese Medicine, Guangzhou, China
| | - Xusheng Liu
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Nephrology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Huanlin Wu
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Cardiology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China.,Department of internal medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Wei Jiang
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Cardiology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Chuan Zou
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Nephrology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| |
Collapse
|
20
|
Qiu H, Liu W, Lan T, Pan W, Chen X, Wu H, Xu D. Salvianolate reduces atrial fibrillation through suppressing atrial interstitial fibrosis by inhibiting TGF-β1/Smad2/3 and TXNIP/NLRP3 inflammasome signaling pathways in post-MI rats. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2018; 51:255-265. [PMID: 30466624 DOI: 10.1016/j.phymed.2018.09.238] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 09/25/2018] [Accepted: 09/30/2018] [Indexed: 06/09/2023]
Abstract
BACKGROUND Salvianolate is the main water-soluble bioactive compound of Salvia Miltiorrhiza Bunge and is now clinically used in the treatment of cardiovascular diseases in China. However, its applications in the prevention of atrial interstitial fibrosis (AIF) and atrial fibrillation (AF) are not fully revealed. PURPOSES To investigate the preventive effect of salvianolate on the pathogenesis of AF in post-myocardial infarction (MI) rats and to elucidate the potential mechanisms. MATERIALS AND METHODS Rats underwent left anterior descending coronary artery ligation were randomized into four groups and administered intraperitoneally with vehicle (MI group, n = 13), or 10, 20 and 40 mg/kg salvianolate (Sal-L, Sal-M and Sal-H group, n = 13, 14 and 13 respectively) for totally five weeks. Rats underwent sham operation was used as control group (Sham, n = 10). Then, echocardiography and AF inducibility test were detected. Tissues and serum were collected for Sirius red and fast green counter stain or hematoxylin-eosin to assess atrial interstitial fibrosis and hypertrophy, or for western blot and ELISA analysis. RESULTS Salvianolate injection significantly improved cardiac function, reduced left atrial enlargement and P-wave duration, and decreased not only the vulnerability to AF but also AF duration. Histologic analysis showed that salvianolate mitigated AIF and atrial hypertrophy. Western blot analysis found that salvianolate inhibited the TGFβ1/Smad2/3 mediated-collagen deposition and inhibited the TXNIP/NLRP3 inflammasome /IL-1β and IL-18 signal pathway. ELISA analysis showed that salvianolate significantly reduced the serum concentrations of BNP, IL-6, CRP and TGFβ1. CONCLUSIONS Salvianolate may constitute a novel upstream therapy for AF by suppressing AIF. The underlying mechanism may be attributable to its inhibitory effects on TGF-β1/Smad2/3 and TXNIP/NLRP3 inflammasome signaling pathway.
Collapse
Affiliation(s)
- Huiliang Qiu
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510006, China.
| | - Wei Liu
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Taohua Lan
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; Cardiovascular Department, Guangdong Provincial Hospital of Chinese Medicine, 2nd Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, China; Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou 510006, China
| | - Wenjun Pan
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Xiaoling Chen
- Cardiovascular Department, Guangdong Provincial Hospital of Chinese Medicine, 2nd Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, China; Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou 510006, China
| | - Huanlin Wu
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; Cardiovascular Department, Guangdong Provincial Hospital of Chinese Medicine, 2nd Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, China; Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou 510006, China; Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China.
| | - Danping Xu
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; Cardiovascular Department, Guangdong Provincial Hospital of Chinese Medicine, 2nd Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, China; Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou 510006, China.
| |
Collapse
|
21
|
DL-3-n-butylphthalide improves ventricular function, and prevents ventricular remodeling and arrhythmias in post-MI rats. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2018; 391:627-637. [DOI: 10.1007/s00210-018-1490-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Accepted: 03/23/2018] [Indexed: 11/27/2022]
|
22
|
Li N, Dobrev D. Targeting atrial fibrillation promoting atrial structural remodeling: is this a viable strategy in patients with heart failure? Naunyn Schmiedebergs Arch Pharmacol 2018; 391:231-233. [PMID: 29327256 DOI: 10.1007/s00210-018-1463-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 01/05/2018] [Indexed: 12/15/2022]
Affiliation(s)
- Na Li
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA.
- Department of Medicine (Section of Cardiovascular Research), Baylor College of Medicine, Houston, TX, USA.
- Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX, USA.
- Baylor College of Medicine, One Baylor Plaza BCM285, Houston, TX, 77030, USA.
| | - Dobromir Dobrev
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany.
- Faculty of Medicine, University Duisburg-Essen, Hufelandstrasse 55, 45122, Essen, Germany.
| |
Collapse
|