1
|
Al-Awar A, Hussain S. Interplay of Reactive Oxygen Species (ROS) and Epigenetic Remodelling in Cardiovascular Diseases Pathogenesis: A Contemporary Perspective. FRONT BIOSCI-LANDMRK 2024; 29:398. [PMID: 39614429 DOI: 10.31083/j.fbl2911398] [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: 05/24/2024] [Revised: 07/20/2024] [Accepted: 07/24/2024] [Indexed: 12/01/2024]
Abstract
Cardiovascular diseases (CVDs) continue to be the leading cause of mortality worldwide, necessitating the development of novel therapies. Despite therapeutic advancements, the underlying mechanisms remain elusive. Reactive oxygen species (ROS) show detrimental effects at high concentrations but act as essential signalling molecules at physiological levels, playing a critical role in the pathophysiology of CVD. However, the link between pathologically elevated ROS and CVDs pathogenesis remains poorly understood. Recent research has highlighted the remodelling of the epigenetic landscape as a crucial factor in CVD pathologies. Epigenetic changes encompass alterations in DNA methylation, post-translational histone modifications, adenosine triphosphate (ATP)-dependent chromatin modifications, and noncoding RNA transcripts. Unravelling the intricate link between ROS and epigenetic changes in CVD is challenging due to the complexity of epigenetic signals in gene regulation. This review aims to provide insights into the role of ROS in modulating the epigenetic landscape within the cardiovascular system. Understanding these interactions may offer novel therapeutic strategies for managing CVD by targeting ROS-induced epigenetic changes. It has been widely accepted that epigenetic modifications are established during development and remain fixed once the lineage-specific gene expression pattern is achieved. However, emerging evidence has unveiled its remarkable dynamism. Consequently, it is now increasingly recognized that epigenetic modifications may serve as a crucial link between ROS and the underlying mechanisms implicated in CVD.
Collapse
Affiliation(s)
- Amin Al-Awar
- Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg University, 41345 Gothenburg, Sweden
| | - Shafaat Hussain
- Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg University, 41345 Gothenburg, Sweden
| |
Collapse
|
2
|
Yijian L, Weihan S, Lin Y, Heng Z, Yu W, Lin S, Shuo M, Mengyang L, Jianxun W. CircNCX1 modulates cardiomyocyte proliferation through promoting ubiquitination of BRG1. Cell Signal 2024; 120:111193. [PMID: 38679350 DOI: 10.1016/j.cellsig.2024.111193] [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: 01/23/2024] [Revised: 04/05/2024] [Accepted: 04/25/2024] [Indexed: 05/01/2024]
Abstract
In mammal, the myocardium loss cannot be recovered spontaneously due to the negligible proliferation ability of mature mammalian cardiomyocyte. However, accumulated evidence has shown that terminally differentiated mammalian cardiomyocyte also has proliferation potency, which can be mediated by several mechanisms. Here, we reported that circNCX1, the most abundant circular RNA in mammalian hearts, can affect the proliferation of murine cardiomyocytes. The level of circNCX1 is significantly elevated during heart development. Forced expression of circNCX1 inhibits cardiomyocyte proliferation, while silencing of endogenous circNCX1 in cardiomyocyte shows reversed effect in vitro. Mechanistically, circNCX1 functions via negatively regulating transcription activator BRG1. It bridges BRG1 and FBXW7 to enhance the ubiquitination and degradation of BRG1, decreasing the expression of BMP10 to lead cell cycle arrest. In summary, our study first revealed that circNCX1 is a modulator of cardiomyocyte proliferation.
Collapse
Affiliation(s)
- Lu Yijian
- School of Basic Medicine, Qingdao University, Qingdao 266071, China
| | - Sun Weihan
- School of Basic Medicine, Qingdao University, Qingdao 266071, China
| | - Ye Lin
- School of Basic Medicine, Qingdao University, Qingdao 266071, China
| | - Zhang Heng
- School of Basic Medicine, Qingdao University, Qingdao 266071, China
| | - Wang Yu
- School of Basic Medicine, Qingdao University, Qingdao 266071, China
| | - Song Lin
- School of Basic Medicine, Qingdao University, Qingdao 266071, China
| | - Miao Shuo
- School of Basic Medicine, Qingdao University, Qingdao 266071, China
| | - Li Mengyang
- School of Basic Medicine, Qingdao University, Qingdao 266071, China.
| | - Wang Jianxun
- School of Basic Medicine, Qingdao University, Qingdao 266071, China.
| |
Collapse
|
3
|
Chen Z, Lai X, Li J, Yuan X, Li Y, Zhang X, Kang Z, Ouyang Z, Zeng J, Hou N, Liu X. BRG1 Deficiency Promotes Cardiomyocyte Inflammation and Apoptosis by Activating the cGAS-STING Signaling in Diabetic Cardiomyopathy. Inflammation 2024:10.1007/s10753-024-02058-7. [PMID: 38867118 DOI: 10.1007/s10753-024-02058-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 05/06/2024] [Accepted: 05/19/2024] [Indexed: 06/14/2024]
Abstract
Brahma-related gene 1 (BRG1) has been implicated in the repair of DNA double-strand breaks (DSBs). Downregulation of BRG1 impairs DSBs repair leading to accumulation of double-stranded DNA (dsDNA). Currently, the role of BRG1 in diabetic cardiomyopathy (DCM) has not been clarified. In this study, we aimed to explore the function and molecular by which BRG1 regulates DCM using mice and cell models. We found that BRG1 was downregulated in the cardiac tissues of DCM mice and in cardiomyocytes cultured with high glucose and palmitic acid (HG/PA), which was accompanied by accumulation of dsDNA and activation of the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) signaling pathway. shRNA-mediated Brg1 knockdown aggravated DCM mice cardiac functions, enhanced dsDNA accumulation, cGAS-STING signaling activation, which induced inflammation and apoptosis. In addition, the results were further verified in HG/PA-treated primary neonatal rat cardiomyocytes (NRCMs). Overexpression of BRG1 in NRCMs yielded opposite results. Furthermore, a selective cGAS inhibitor RU.521 or STING inhibitor C-176 partially reversed the BRG1 knockdown-induced inflammation and apoptosis in vitro. In conclusion, our results demonstrate that BRG1 is downregulated during DCM in vivo and in vitro, resulting in cardiomyocyte inflammation and apoptosis due to dsDNA accumulation and cGAS-STING signaling activation. Therefore, targeting the BRG1-cGAS-STING pathway may represent a novel therapeutic strategy for improving cardiac function of patients with DCM.
Collapse
Affiliation(s)
- Ziying Chen
- Department of Pharmacy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan 511518, China
- Guangdong Key Laboratory of Molecular Target & Clinical Pharmacology, the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Xiangmao Lai
- Department of Urology, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China
- Guangdong Engineering Technology Research Center of Urinary Continence and Reproductive Medicine, Guangzhou Medical University, Qingyuan, 511518, China
| | - Jingxuan Li
- Department of Pharmacy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan 511518, China
- Guangdong Key Laboratory of Molecular Target & Clinical Pharmacology, the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Xun Yuan
- Guangdong Key Laboratory of Molecular Target & Clinical Pharmacology, the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Yilang Li
- Guangdong Key Laboratory of Molecular Target & Clinical Pharmacology, the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Xiaojing Zhang
- Department of Pharmacy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan 511518, China
| | - Zhanfang Kang
- Guangdong Engineering Technology Research Center of Urinary Continence and Reproductive Medicine, Guangzhou Medical University, Qingyuan, 511518, China
- Department of Basic Medical Research, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China
| | - Zizhang Ouyang
- Department of Pharmacy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan 511518, China
| | - Jianwen Zeng
- Department of Urology, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China.
- Guangdong Engineering Technology Research Center of Urinary Continence and Reproductive Medicine, Guangzhou Medical University, Qingyuan, 511518, China.
| | - Ning Hou
- Guangdong Key Laboratory of Molecular Target & Clinical Pharmacology, the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China.
| | - Xiaoping Liu
- Department of Pharmacy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan 511518, China.
- Guangdong Key Laboratory of Molecular Target & Clinical Pharmacology, the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China.
| |
Collapse
|
4
|
Ma X, Zhao J, Feng Y. Epicardial SMARCA4 deletion exacerbates cardiac injury in myocardial infarction and is related to the inhibition of epicardial epithelial-mesenchymal transition. J Mol Cell Cardiol 2024; 191:76-87. [PMID: 38718920 DOI: 10.1016/j.yjmcc.2024.05.001] [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: 11/06/2023] [Revised: 05/01/2024] [Accepted: 05/04/2024] [Indexed: 05/12/2024]
Abstract
The reactivated adult epicardium produces epicardium-derived cells (EPDCs) via epithelial-mesenchymal transition (EMT) to benefit the recovery of the heart after myocardial infarction (MI). SMARCA4 is the core catalytic subunit of the chromatin re-modeling complex, which has the potential to target some reactivated epicardial genes in MI. However, the effects of epicardial SMARCA4 on MI remain uncertain. This study found that SMARCA4 was activated over time in epicardial cells following MI, and some of activated cells belonged to downstream differentiation types of EPDCs. This study used tamoxifen to induce lineage tracing and SMARCA4 deletion from epicardial cells in Wt1-CreER;Smarca4fl/fl;Rosa26-RFP adult mice. Epicardial SMARCA4 deletion reduces the number of epicardial cells in adult mice, which was related to changes in the activation, proliferation, and apoptosis of epicardial cells. Epicardial SMARCA4 deletion reduced collagen deposition and angiogenesis in the infarcted area, exacerbated cardiac injury in MI. The exacerbation of cardiac injury was related to the inhibition of generation and differentiation of EPDCs. The alterations in EPDCs were associated with inhibited transition between E-CAD and N-CAD during the epicardial EMT, coupled with the down-regulation of WT1, SNAIL1, and PDGF signaling. In conclusion, this study suggests that Epicardial SMARCA4 plays a critical role in cardiac injury caused by MI, and its regulatory mechanism is related to epicardial EMT. Epicardial SMARCA4 holds potential as a novel molecular target for treating MI.
Collapse
Affiliation(s)
- Xingyu Ma
- College of Life Science and Technology, Jinan University, Guangzhou, China.
| | - Jianjun Zhao
- College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Yi Feng
- College of Life Science and Technology, Jinan University, Guangzhou, China
| |
Collapse
|
5
|
Lee S. Cardiovascular Disease and miRNAs: Possible Oxidative Stress-Regulating Roles of miRNAs. Antioxidants (Basel) 2024; 13:656. [PMID: 38929095 PMCID: PMC11200533 DOI: 10.3390/antiox13060656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 05/23/2024] [Accepted: 05/25/2024] [Indexed: 06/28/2024] Open
Abstract
MicroRNAs (miRNAs) have been highlighted as key players in numerous diseases, and accumulating evidence indicates that pathological expressions of miRNAs contribute to both the development and progression of cardiovascular diseases (CVD), as well. Another important factor affecting the development and progression of CVD is reactive oxygen species (ROS), as well as the oxidative stress they may impose on the cells. Considering miRNAs are involved in virtually every biological process, it is not unreasonable to assume that miRNAs also play critical roles in the regulation of oxidative stress. This narrative review aims to provide mechanistic insights on possible oxidative stress-regulating roles of miRNAs in cardiovascular diseases based on differentially expressed miRNAs reported in various cardiovascular diseases and their empirically validated targets that have been implicated in the regulation of oxidative stress.
Collapse
Affiliation(s)
- Seahyoung Lee
- Department of Convergence Science, College of Medicine, Catholic Kwandong University, Gangneung-si 25601, Republic of Korea
| |
Collapse
|
6
|
Li J, Ma ZY, Cui YF, Cui YT, Dong XH, Wang YZ, Fu YY, Xue YD, Tong TT, Ding YZ, Zhu YM, Huang HJ, Zhao L, Lv HZ, Xiong LZ, Zhang K, Han YX, Ban T, Huo R. Cardiac-specific deletion of BRG1 ameliorates ventricular arrhythmia in mice with myocardial infarction. Acta Pharmacol Sin 2024; 45:517-530. [PMID: 37880339 PMCID: PMC10834533 DOI: 10.1038/s41401-023-01170-y] [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: 05/22/2023] [Accepted: 09/14/2023] [Indexed: 10/27/2023] Open
Abstract
Malignant ventricular arrhythmia (VA) after myocardial infarction (MI) is mainly caused by myocardial electrophysiological remodeling. Brahma-related gene 1 (BRG1) is an ATPase catalytic subunit that belongs to a family of chromatin remodeling complexes called Switch/Sucrose Non-Fermentable Chromatin (SWI/SNF). BRG1 has been reported as a molecular chaperone, interacting with various transcription factors or proteins to regulate transcription in cardiac diseases. In this study, we investigated the potential role of BRG1 in ion channel remodeling and VA after ischemic infarction. Myocardial infarction (MI) mice were established by ligating the left anterior descending (LAD) coronary artery, and electrocardiogram (ECG) was monitored. Epicardial conduction of MI mouse heart was characterized in Langendorff-perfused hearts using epicardial optical voltage mapping. Patch-clamping analysis was conducted in single ventricular cardiomyocytes isolated from the mice. We showed that BRG1 expression in the border zone was progressively increased in the first week following MI. Cardiac-specific deletion of BRG1 by tail vein injection of AAV9-BRG1-shRNA significantly ameliorated susceptibility to electrical-induced VA and shortened QTc intervals in MI mice. BRG1 knockdown significantly enhanced conduction velocity (CV) and reversed the prolonged action potential duration in MI mouse heart. Moreover, BRG1 knockdown improved the decreased densities of Na+ current (INa) and transient outward potassium current (Ito), as well as the expression of Nav1.5 and Kv4.3 in the border zone of MI mouse hearts and in hypoxia-treated neonatal mouse ventricular cardiomyocytes. We revealed that MI increased the binding among BRG1, T-cell factor 4 (TCF4) and β-catenin, forming a transcription complex, which suppressed the transcription activity of SCN5A and KCND3, thereby influencing the incidence of VA post-MI.
Collapse
Affiliation(s)
- Jing Li
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Baojian Road, Nangang District, Harbin, 150081, China
| | - Zi-Yue Ma
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Baojian Road, Nangang District, Harbin, 150081, China
| | - Yun-Feng Cui
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Baojian Road, Nangang District, Harbin, 150081, China
| | - Ying-Tao Cui
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Baojian Road, Nangang District, Harbin, 150081, China
| | - Xian-Hui Dong
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Baojian Road, Nangang District, Harbin, 150081, China
| | - Yong-Zhen Wang
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Baojian Road, Nangang District, Harbin, 150081, China
| | - Yu-Yang Fu
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Baojian Road, Nangang District, Harbin, 150081, China
| | - Ya-Dong Xue
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Baojian Road, Nangang District, Harbin, 150081, China
| | - Ting-Ting Tong
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Baojian Road, Nangang District, Harbin, 150081, China
| | - Ying-Zi Ding
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Baojian Road, Nangang District, Harbin, 150081, China
| | - Ya-Mei Zhu
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Baojian Road, Nangang District, Harbin, 150081, China
| | - Hai-Jun Huang
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Baojian Road, Nangang District, Harbin, 150081, China
| | - Ling Zhao
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Baojian Road, Nangang District, Harbin, 150081, China
| | - Hong-Zhao Lv
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Baojian Road, Nangang District, Harbin, 150081, China
| | - Ling-Zhao Xiong
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Baojian Road, Nangang District, Harbin, 150081, China
| | - Kai Zhang
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Baojian Road, Nangang District, Harbin, 150081, China
| | - Yu-Xuan Han
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Baojian Road, Nangang District, Harbin, 150081, China
| | - Tao Ban
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Baojian Road, Nangang District, Harbin, 150081, China.
- Heilongjiang Academy of Medical Sciences, Baojian Road, Nangang District, Harbin, 150081, China.
| | - Rong Huo
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Baojian Road, Nangang District, Harbin, 150081, China.
| |
Collapse
|
7
|
Liang G, Fang J, Zhang P, Ding S, Zhao Y, Feng Y. Metformin plus L-carnitine enhances brown/beige adipose tissue activity via Nrf2/HO-1 signaling to reduce lipid accumulation and inflammation in murine obesity. Open Med (Wars) 2024; 19:20240900. [PMID: 38463531 PMCID: PMC10921440 DOI: 10.1515/med-2024-0900] [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: 06/27/2023] [Revised: 11/13/2023] [Accepted: 12/18/2023] [Indexed: 03/12/2024] Open
Abstract
This study investigated how Metformin (Met) combined with L-carnitine (L-car) modulates brown adipose tissue (BAT) to affect obesity. High-fat-induced obese rats received daily oral gavage with Met and/or L-car, followed by serum biochemical analysis, histopathological observation on adipose tissues, and immunochemistry test for the abdominal expression of BAT-specific uncoupling protein 1 (UCP1). Mouse-embryonic-fibroblast cells were induced into adipocytes, during which Met plus L-car was added with/without saturated fatty acid (SFA). The role of nuclear factor erythroid 2-related factor 2 (Nrf2) in adipocyte browning was investigated by gene silencing. Mitochondria biogenesis in adipocytes was inspected by Mitotracker staining. Nrf2/heme oxygenase-1 (HO-1)/BAT-related genes/proinflammatory marker expressions in adipose tissues and/or adipocytes were analyzed by Western blot, qRT-PCR, and/or immunofluorescence test. Met or L-car improved metabolic disorders, reduced adipocyte vacuolization and swelling, upregulated levels of BAT-related genes including UCP1 and downregulated proinflammatory marker expressions, and activated the Nrf2/HO-1 pathway in adipose tissues of obese rats. Met and L-car functioned more strongly than alone. In adipocytes, Met plus L-car upregulated BAT-related gene levels and protected against SFA-caused inflammation promotion and mitochondria degeneration, which yet was attenuated by Nrf2 silencing. Met plus L-car enhances BAT activity and white adipose tissue browning via the Nrf2/HO-1 pathway to reduce lipid accumulation and inflammation in obese rats.
Collapse
Affiliation(s)
- Guojin Liang
- Anesthesiology Department, Ningbo First Hospital, Ningbo, China
| | - Jie Fang
- Paediatrics Department, Ningbo Women and Children’s Hospital, Zhejiang, 315000, China
| | - Pingping Zhang
- Paediatrics Department, Ningbo Women and Children’s Hospital, Zhejiang, 315000, China
| | - Shuxia Ding
- Paediatrics Department, Ningbo Women and Children’s Hospital, Zhejiang, 315000, China
| | - Yudan Zhao
- Paediatrics Department, Ningbo Women and Children’s Hospital, Zhejiang, 315000, China
| | - Yueying Feng
- Paediatrics Department, Ningbo Women and Children’s Hospital, No. 339 Liuting Street, Ningbo, Zhejiang, 315000, China
| |
Collapse
|
8
|
Fang T, Wang X, Huangfu N. Superfamily II helicases: the potential therapeutic target for cardiovascular diseases. Front Cardiovasc Med 2023; 10:1309491. [PMID: 38152606 PMCID: PMC10752008 DOI: 10.3389/fcvm.2023.1309491] [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: 10/16/2023] [Accepted: 11/29/2023] [Indexed: 12/29/2023] Open
Abstract
Cardiovascular diseases (CVDs) still maintain high morbidity and mortality globally. Helicases, a unique class of enzymes, are extensively implicated in the processes of nucleic acid (NA) metabolism across various organisms. They play a pivotal role in gene expression, inflammatory response, lipid metabolism, and so forth. However, abnormal helicase expression has been associated with immune response, cancer, and intellectual disability in humans. Superfamily II (SFII) is one of the largest and most diverse of the helicase superfamilies. Increasing evidence has implicated SFⅡ helicases in the pathogenesis of multiple CVDs. In this review, we comprehensively review the regulation mechanism of SFⅡ helicases in CVDs including atherosclerosis, myocardial infarction, cardiomyopathies, and heart failure, which will contribute to the investigation of ideal therapeutic targets for CVDs.
Collapse
Affiliation(s)
- Tianxiang Fang
- Health Science Center, Ningbo University, Ningbo, China
- Department of Cardiology, The First Affiliated Hospital of Ningbo University, Ningbo, China
- Department of Cardiology, Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang Province, Ningbo, China
- Clinical Medicine Research Centre for Cardiovascular Disease of Ningbo, Ningbo, China
| | - Xizhi Wang
- Department of Cardiology, Lihuili Hospital Affiliated to Ningbo University, Ningbo, China
| | - Ning Huangfu
- Department of Cardiology, The First Affiliated Hospital of Ningbo University, Ningbo, China
- Department of Cardiology, Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang Province, Ningbo, China
- Clinical Medicine Research Centre for Cardiovascular Disease of Ningbo, Ningbo, China
| |
Collapse
|
9
|
Fu K, Dai S, Li Y, Ma C, Xue X, Zhang S, Wang C, Zhou H, Zhang Y, Li Y. The protective effect of forsythiaside A on 3,5-diethoxycarbonyl-1,4-dihydrocollidine-induced cholestatic liver injury in mice: Based on targeted metabolomics and molecular biology technology. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166822. [PMID: 37523877 DOI: 10.1016/j.bbadis.2023.166822] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 07/17/2023] [Accepted: 07/24/2023] [Indexed: 08/02/2023]
Abstract
Cholestasis is a disorder of bile secretion and excretion caused by a variety of etiologies. At present, there is a lack of functional foods or drugs that can be used for intervention. Forsythiaside A (FTA) is a natural phytochemical component isolated from the medicinal plant Forsythia suspensa (Thunb.) Vahl, which has a significant hepatoprotective effect. In this study, we investigated whether FTA could alleviate liver injury induced by cholestasis. In vitro, FTA reversed the decrease in viability of human intrahepatic bile duct epithelial cells, the decrease in antioxidant enzymes (SOD1, CAT and GSH-Px), and cell apoptosis induced by lithocholic acid. In vivo, FTA protected mice from 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC)-induced liver injury, abnormal serum biochemical indexes, abnormal bile duct hyperplasia, and inflammatory infiltration. Furthermore, FTA treatment alleviated liver fibrosis by inhibiting collagen deposition and HSC activation. The metabonomic results showed that DDC-induced bile acid disorders in the liver and serum were reversed after FTA treatment, which may benefit from the activation of the FXR/BSEP axis. In addition, FTA treatment increased the levels of antioxidant enzymes in the serum and liver. Meanwhile, FTA treatment inhibited ROS and MDA levels and cleaved caspase 3 protein expression, thereby reducing DDC-induced hepatic oxidative stress and apoptosis. Further studies showed that the antioxidant effects of FTA were dependent on the activation of the BRG1/NRF2/HO-1 axis. In a word, FTA has a significant hepatoprotective effect on cholestatic liver injury, and can be further developed as a functional food or drug to prevent and treat cholestatic liver injury.
Collapse
Affiliation(s)
- Ke Fu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Shu Dai
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yanzhi Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Cheng Ma
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Xinyan Xue
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Shenglin Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Cheng Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Honglin Zhou
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yafang Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yunxia Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| |
Collapse
|
10
|
Ma ZY, Li J, Dong XH, Cui YT, Cui YF, Ban T, Huo R. The role of BRG1 in epigenetic regulation of cardiovascular diseases. Eur J Pharmacol 2023; 957:176039. [PMID: 37678658 DOI: 10.1016/j.ejphar.2023.176039] [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: 05/25/2023] [Revised: 08/28/2023] [Accepted: 08/30/2023] [Indexed: 09/09/2023]
Abstract
Cardiovascular diseases have been closely linked to abnormal epigenetic regulation. In the context of epigenetic regulation, BRG1, a pivotal SWI/SNF chromatin remodeling enzyme, emerges as a key epigenetic regulator with significant impact on the development and progression of cardiovascular disorders. From the perspective of epigenetic regulation of cardiovascular diseases, BRG1 emerges as a pivotal SWI/SNF chromatin remodeling enzyme, functioning as a key epigenetic regulator. It exerts substantial influence on the development and progression of cardiovascular disorders by exerting precise control over gene expression and protein levels. Therefore, a comprehensive understanding of BRG1's epigenetic regulatory role in cardiovascular disease is essential for unraveling its underlying pathophysiological mechanisms. This paper summarizes and discusses the function of BRG1 in the epigenetic regulation of cardiovascular diseases.
Collapse
Affiliation(s)
- Zi-Yue Ma
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Baojian Road, Nangang District, Harbin, 150081, PR China
| | - Jing Li
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Baojian Road, Nangang District, Harbin, 150081, PR China
| | - Xian-Hui Dong
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Baojian Road, Nangang District, Harbin, 150081, PR China
| | - Ying-Tao Cui
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Baojian Road, Nangang District, Harbin, 150081, PR China
| | - Yun-Feng Cui
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Baojian Road, Nangang District, Harbin, 150081, PR China
| | - Tao Ban
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Baojian Road, Nangang District, Harbin, 150081, PR China; National-Local Joint Engineering Laboratory of Drug Research and Development of Cardiovascular and Cerebrovascular Diseases in Frigid Zone, The National Development and Reform Commission, Baojian Road, Nangang District, Harbin, 150081, PR China; Heilongjiang Academy of Medical Sciences, Baojian Road, Nangang District, Harbin, 150081, PR China
| | - Rong Huo
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Baojian Road, Nangang District, Harbin, 150081, PR China.
| |
Collapse
|
11
|
Mao X, Yan B, Chen H, Lai P, Ma J. BRG1 mediates protective ability of spermidine to ameliorate osteoarthritic cartilage by Nrf2/KEAP1 and STAT3 signaling pathway. Int Immunopharmacol 2023; 122:110593. [PMID: 37423156 DOI: 10.1016/j.intimp.2023.110593] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 06/20/2023] [Accepted: 06/30/2023] [Indexed: 07/11/2023]
Abstract
BACKGROUND Spermidine (SPD) is a natural polyamine that shows beneficial effects on osteoarthritis (OA). However, the effect of SPD on cartilage inflammation remains unknown. This study aimed to investigate the potential mechanisms underlying the protective effect of SPD against OA-induced articular cartilage degradation. METHOD SW1353 human chondrocytes were treated with hydrogen peroxide and lipopolysaccharide to induce models of inflammation and oxidative stress, followed by different dose of SPD intervention. Moreover, mice that underwent anterior cruciate ligament transection were bred and treated with SPD. The effects of SPD were observed using a CCK-8 kit, real-time polymerase chain reaction, immunoblotting, and immunofluorescent assays. RESULT SPD significantly increased the expression of antioxidant proteins, chondrogenic genes, and inflammatory factors both in vivo and in vitro. And injury of the mouse cartilage was also reduced by SPD. Moreover, SPD activated the Nrf2/KEAP1 pathway and inhibited STAT3 phosphorylation. BRG1 expression was decreased in osteoarthritic mouse cartilage, whereas SPD treatment caused an upregulation. However, when BRG1 was specifically inhibited by an adeno-associated virus and small interfering RNA, the antioxidant and anti-inflammatory effects of SPD were significantly diminished both in vitro and in vivo. CONCLUSION We found that SPD ameliorated cartilage damage in OA by activating the BRG1-mediated Nrf2/KEAP1 pathway. SPD and BRG1 may provide new therapeutic options or targets for the treatment of OA.
Collapse
Affiliation(s)
- Xinjie Mao
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bing Yan
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hongjie Chen
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Peng Lai
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jinzhong Ma
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
12
|
Wang L, Chen P, Pan Y, Wang Z, Xu J, Wu X, Yang Q, Long M, Liu S, Huang W, Ou C, Wu Y. Injectable photocurable Janus hydrogel delivering hiPSC cardiomyocyte-derived exosome for post-heart surgery adhesion reduction. SCIENCE ADVANCES 2023; 9:eadh1753. [PMID: 37540739 PMCID: PMC10403204 DOI: 10.1126/sciadv.adh1753] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 07/06/2023] [Indexed: 08/06/2023]
Abstract
Postsurgical pericardial adhesions pose increased risks of sequelae, prolonged reoperation time, and reduced visibility in the surgical field. Here, we introduce an injectable Janus hydrogel, which exhibits asymmetric adhesiveness properties after photocrosslinking, sustained delivering induced pluripotent stem cell-derived cardiomyocyte exosomes (iCM-EXOs) for post-heart surgery adhesion reduction. Our findings reveal that iCM-EXOs effectively attenuate oxidative stress in hydrogen peroxide-treated primary cardiomyocytes by inhibiting the activation of the transcription factor nuclear factor erythroid 2-related factor 2. Notably, in rat cardiac postsurgery models, the Janus hydrogels loaded with iCM-EXOs demonstrate dual functionality, acting as antioxidants and antipericardial adhesion agents. These hydrogels effectively protect iCM-EXOs from GATA6+ cavity macrophage clearance by inhibiting the recruitment of macrophages from the thoracic cavity. These results highlight the promising potential of iCM-EXO-laden Janus hydrogels for clinical safety and efficacy validation in trials involving heart surgery patients, with the ultimate goal of routine administration during open-heart surgeries.
Collapse
Affiliation(s)
- Ling Wang
- Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou 510515, China
- Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics, Department of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Peier Chen
- Affiliated Dongguan Hospital, Southern Medical University (Dongguan People’s Hospital), Dongguan 523058, China
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Yuxuan Pan
- Affiliated Dongguan Hospital, Southern Medical University (Dongguan People’s Hospital), Dongguan 523058, China
| | - Zihan Wang
- Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics, Department of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Jie Xu
- Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou 510515, China
| | - Xiaoqi Wu
- Department of Urology and Andrology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200001, China
| | - Qiao Yang
- Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou 510515, China
| | - Meng Long
- Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics, Department of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Sitian Liu
- Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics, Department of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Wenhua Huang
- Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics, Department of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Caiwen Ou
- Affiliated Dongguan Hospital, Southern Medical University (Dongguan People’s Hospital), Dongguan 523058, China
| | - Yaobin Wu
- Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics, Department of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
13
|
Guo K, Shang Y, Wang Z, Li Y, Chen J, Zhu B, Zhang D, Chen J. BRG1 alleviates microglial activation by promoting the KEAP1-NRF2/HO-1 signaling pathway and minimizing oxidative damage in cerebral ischemia-reperfusion. Int Immunopharmacol 2023; 119:110201. [PMID: 37172425 DOI: 10.1016/j.intimp.2023.110201] [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: 01/10/2023] [Revised: 03/28/2023] [Accepted: 04/12/2023] [Indexed: 05/15/2023]
Abstract
BRG1 is a key factor in the process of apoptosis and oxidative damage; however, its role in the pathophysiology of ischemic stroke is unclear. Here, we discovered that during middle cerebral artery occlusion (MCAO) reperfusion in mice, microglia were significantly activated in the cerebral cortex of the infarct area, and BRG1 expression was increased in the mouse MCAO/R model, peaking at 4 days. In microglia subjected to OGD/R, BRG1 expression increased and peaked at 12 h after reoxygenation. After ischemic stroke, in vitro changing the expression of BRG1 expression levels greatly altered the activation of microglia and the production of antioxidant and pro-oxidant proteins. Knocking down BRG1 expression levels in vitro increased the inflammatory response, promoted microglial activation, and decreased the expression of the NRF2/HO-1 signaling pathway after ischemic stroke. In contrast, overexpression of BRG1 dramatically reduced the expression of NRF2/HO-1 signaling pathway and microglial activation. Our research reveals that BRG1 reduces postischemic oxidative damage via the KEAP1-NRF2/HO-1 signaling pathway, protecting against brain ischemia/reperfusion injury. Using BRG1 as a pharmaceutical target to inhibit inflammatory responses to reduce oxidative damage may be a unique way to explore techniques for the treatment of ischemic stroke and other cerebrovascular illnesses.
Collapse
Affiliation(s)
- Kongwei Guo
- Medical Research Center, Affiliated Hospital 2 of Nantong University, Nantong 226001, People's Republic of China; Department of Clinical Medicine, Medical College, Nantong University, Nantong 226001, People's Republic of China; Nantong Key Laboratory of Molecular Immunology, Affiliated Hospital 2 of Nantong University, Nantong 226001, People's Republic of China
| | - Yanxing Shang
- Medical Research Center, Affiliated Hospital 2 of Nantong University, Nantong 226001, People's Republic of China; Nantong Key Laboratory of Molecular Immunology, Affiliated Hospital 2 of Nantong University, Nantong 226001, People's Republic of China
| | - Zhao Wang
- Medical Research Center, Affiliated Hospital 2 of Nantong University, Nantong 226001, People's Republic of China; Department of Clinical Medicine, Medical College, Nantong University, Nantong 226001, People's Republic of China
| | - Yu Li
- Medical Research Center, Affiliated Hospital 2 of Nantong University, Nantong 226001, People's Republic of China; Department of Pathogen Biology, Medical College, Nantong University, Nantong 226001, People's Republic of China
| | - Jinliang Chen
- Medical Research Center, Affiliated Hospital 2 of Nantong University, Nantong 226001, People's Republic of China; Department of Respiratory Medicine, Affiliated Hospital 2 of Nantong University, Nantong 226001, People's Republic of China
| | - Baofeng Zhu
- Medical Research Center, Affiliated Hospital 2 of Nantong University, Nantong 226001, People's Republic of China; Department of Emergency, Affiliated Hospital 2 of Nantong University, Nantong 226001, People's Republic of China
| | - Dongmei Zhang
- Medical Research Center, Affiliated Hospital 2 of Nantong University, Nantong 226001, People's Republic of China; Nantong Key Laboratory of Molecular Immunology, Affiliated Hospital 2 of Nantong University, Nantong 226001, People's Republic of China.
| | - Jianrong Chen
- Medical Research Center, Affiliated Hospital 2 of Nantong University, Nantong 226001, People's Republic of China; Department of Respiratory Medicine, Affiliated Hospital 2 of Nantong University, Nantong 226001, People's Republic of China; Department of Emergency, Affiliated Hospital 2 of Nantong University, Nantong 226001, People's Republic of China; Nantong Key Laboratory of Molecular Immunology, Affiliated Hospital 2 of Nantong University, Nantong 226001, People's Republic of China.
| |
Collapse
|
14
|
Song YQ, Yan XD, Wang Y, Wang ZZ, Mao XL, Ye LP, Li SW. Role of ferroptosis in colorectal cancer. World J Gastrointest Oncol 2023; 15:225-239. [PMID: 36908317 PMCID: PMC9994046 DOI: 10.4251/wjgo.v15.i2.225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 12/15/2022] [Accepted: 01/10/2023] [Indexed: 02/14/2023] Open
Abstract
Colorectal cancer (CRC) is the second deadliest cancer and the third-most common malignancy in the world. Surgery, chemotherapy, and targeted therapy have been widely used to treat CRC, but some patients still develop resistance to these treatments. Ferroptosis is a novel non-apoptotic form of cell death. It is an iron-dependent non-apoptotic cell death characterized by the accumulation of lipid reactive oxygen species and has been suggested to play a role in reversing resistance to anticancer drugs. This review summarizes recent advances in the prognostic role of ferroptosis in CRC and the mechanism of action in CRC.
Collapse
Affiliation(s)
- Ya-Qi Song
- Taizhou Hospital of Zhejiang Province, Zhejiang University, Linhai 317000, Zhejiang Province, China
| | - Xiao-Dan Yan
- Department of Gastroenterology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai 317000, Zhejiang Province, China
| | - Yi Wang
- Department of Gastroenterology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai 317000, Zhejiang Province, China
| | - Zhen-Zhen Wang
- Department of Gastroenterology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai 317000, Zhejiang Province, China
| | - Xin-Li Mao
- Department of Gastroenterology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai 317000, Zhejiang Province, China
| | - Li-Ping Ye
- Taizhou Hospital of Zhejiang Province, Zhejiang University, Linhai 317000, Zhejiang Province, China
- Department of Gastroenterology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai 317000, Zhejiang Province, China
- Institute of Digestive Disease, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai 317000, Zhejiang Province, China
| | - Shao-Wei Li
- Department of Gastroenterology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai 317000, Zhejiang Province, China
- Institute of Digestive Disease, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai 317000, Zhejiang Province, China
- Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai 317000, Zhejiang Province, China
| |
Collapse
|
15
|
Wang XJ, Zhang D, Yang YT, Li XY, Li HN, Zhang XP, Long JY, Lu YQ, Liu L, Yang G, Liu J, Hong J, Wu HG, Ma XP. Suppression of microRNA-222-3p ameliorates ulcerative colitis and colitis-associated colorectal cancer to protect against oxidative stress via targeting BRG1 to activate Nrf2/HO-1 signaling pathway. Front Immunol 2023; 14:1089809. [PMID: 36776858 PMCID: PMC9911687 DOI: 10.3389/fimmu.2023.1089809] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 01/16/2023] [Indexed: 01/28/2023] Open
Abstract
Oxidative stress is an important pathogenic factor in ulcerative colitis (UC) and colitis-associated colorectal cancer (CAC), further impairing the entire colon. Intestinal epithelial cells (IECs) are crucial components of innate immunity and play an important role in maintaining intestinal barrier function. Recent studies have indicated that microRNA-222-3p (miR-222-3p) is increased in colon of UC and colorectal cancer (CRC) patients, and miR-222-3p is a crucial regulator of oxidative stress. However, whether miR-222-3p influences IEC oxidative stress in UC and CAC remains unknown. This study investigated the effect of miR-222-3p on the regulation of IEC oxidative stress in UC and CAC. An in vitro inflammation model was established in NCM460 colonic cells, mouse UC and CAC models were established in vivo, and IECs were isolated. The biological role and mechanism of miR-222-3p-mediated oxidative stress in UC and CAC were determined. We demonstrated that miR-222-3p expression was notably increased in dextran sulfate sodium (DSS)-induced NCM460 cells and IECs from UC and CAC mice. In vitro, these results showed that the downregulation of miR-222-3p reduced oxidative stress, caspase-3 activity, IL-1β and TNF-α in DSS-induced NCM460 cells. We further identified BRG1 as the target gene of miR-222-3p, and downregulating miR-222-3p alleviated DSS-induced oxidative injury via promoting BRG1-mediated activation Nrf2/HO-1 signaling in NCM460 cells. The in vivo results demonstrated that inhibiting miR-222-3p in IECs significantly relieved oxidative stress and inflammation in the damaged colons of UC and CAC mice, as evidenced by decreases in ROS, MDA, IL-1β and TNF-α levels and increases in GSH-Px levels. Our study further demonstrated that inhibiting miR-222-3p in IECs attenuated oxidative damage by targeting BRG1 to activate the Nrf2/HO-1 signaling. In summary, inhibiting miR-222-3p in IECs attenuates oxidative stress by targeting BRG1 to activate the Nrf2/HO-1 signaling, thereby reducing colonic inflammation and tumorigenesis.
Collapse
Affiliation(s)
- Xue-Jun Wang
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
| | - Dan Zhang
- Shanghai Research Institute of Acupuncture and Meridian, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yan-Ting Yang
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiao-Ying Li
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hong-Na Li
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiao-Peng Zhang
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jun-Yi Long
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yun-Qiong Lu
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Li Liu
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Guang Yang
- Shanghai Research Institute of Acupuncture and Meridian, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jie Liu
- Shanghai Research Institute of Acupuncture and Meridian, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jue Hong
- Shanghai Research Institute of Acupuncture and Meridian, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Huan-Gan Wu
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Shanghai Research Institute of Acupuncture and Meridian, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiao-Peng Ma
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Shanghai Research Institute of Acupuncture and Meridian, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
16
|
Chen X, Li Y, Yuan X, Yuan W, Li C, Zeng Y, Lian Y, Qiu X, Qin Y, Zhang G, Liu X, Luo C, Luo JD, Hou N. Methazolamide Attenuates the Development of Diabetic Cardiomyopathy by Promoting β-Catenin Degradation in Type 1 Diabetic Mice. Diabetes 2022; 71:795-811. [PMID: 35043173 DOI: 10.2337/db21-0506] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 12/21/2021] [Indexed: 11/13/2022]
Abstract
Methazolamide (MTZ), a carbonic anhydrase inhibitor, has been shown to inhibit cardiomyocyte hypertrophy and exert a hypoglycemic effect in patients with type 2 diabetes and diabetic db/db mice. However, whether MTZ has a cardioprotective effect in the setting of diabetic cardiomyopathy is not clear. We investigated the effects of MTZ in a mouse model of streptozotocin-induced type 1 diabetes mellitus (T1DM). Diabetic mice received MTZ by intragastric gavage (10, 25, or 50 mg/kg, daily for 16 weeks). In the diabetic group, MTZ significantly reduced both random and fasting blood glucose levels and improved glucose tolerance in a dose-dependent manner. MTZ ameliorated T1DM-induced changes in cardiac morphology and dysfunction. Mechanistic analysis revealed that MTZ blunted T1DM-induced enhanced expression of β-catenin. Similar results were observed in neonatal rat cardiomyocytes (NRCMs) and adult mouse cardiomyocytes treated with high glucose or Wnt3a (a β-catenin activator). There was no significant change in β-catenin mRNA levels in cardiac tissues or NRCMs. MTZ-mediated β-catenin downregulation was recovered by MG132, a proteasome inhibitor. Immunoprecipitation and immunofluorescence analyses showed augmentation of AXIN1-β-catenin interaction by MTZ in T1DM hearts and in NRCMs treated with Wnt3a; thus, MTZ may potentiate AXIN1-β-catenin linkage to increase β-catenin degradation. Overall, MTZ may alleviate cardiac hypertrophy by mediating AXIN1-β-catenin interaction to promote degradation and inhibition of β-catenin activity. These findings may help inform novel therapeutic strategy to prevent heart failure in patients with diabetes.
Collapse
Affiliation(s)
- Xiaoqing Chen
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State and NMPA Key Laboratory, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Yilang Li
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State and NMPA Key Laboratory, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Xun Yuan
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State and NMPA Key Laboratory, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Wenchang Yuan
- Guangzhou Key Laboratory for Clinical Rapid Diagnosis and Early Warning of Infectious Diseases, KingMed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Conglin Li
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State and NMPA Key Laboratory, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Yue Zeng
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State and NMPA Key Laboratory, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Yuling Lian
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State and NMPA Key Laboratory, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Xiaoxia Qiu
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State and NMPA Key Laboratory, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China
- Zhujiang Hospital of Southern Medical University, Guangzhou, People's Republic of China
| | - Yuan Qin
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State and NMPA Key Laboratory, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Guiping Zhang
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State and NMPA Key Laboratory, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Xiawen Liu
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State and NMPA Key Laboratory, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Chengfeng Luo
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Jian-Dong Luo
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State and NMPA Key Laboratory, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Ning Hou
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State and NMPA Key Laboratory, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China
| |
Collapse
|
17
|
Liu S, Cao X, Wang D, Zhu H. Iron metabolism: State of the art in hypoxic cancer cell biology. Arch Biochem Biophys 2022; 723:109199. [DOI: 10.1016/j.abb.2022.109199] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 03/25/2022] [Accepted: 03/25/2022] [Indexed: 02/08/2023]
|
18
|
Zhang X, Xia Y, Wang B. Brg1 in heart disease: Friend or foe? Int J Cardiol 2021; 348:116. [PMID: 34856298 DOI: 10.1016/j.ijcard.2021.11.071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 11/05/2021] [Accepted: 11/26/2021] [Indexed: 11/24/2022]
Affiliation(s)
- Xuesong Zhang
- Department of Cardiology, Liaocheng People's Hospital, Liaocheng 252000, PR China
| | - Ying Xia
- Department of Nursing, Liaocheng Vocational & Technical College, Liaocheng 252000, PR China
| | - Bin Wang
- Department of General Practice, The Second Hospital of Jiaxing, Jiaxing 314000, PR China.
| |
Collapse
|
19
|
Yang J, Mo J, Dai J, Ye C, Cen W, Zheng X, Jiang L, Ye L. Cetuximab promotes RSL3-induced ferroptosis by suppressing the Nrf2/HO-1 signalling pathway in KRAS mutant colorectal cancer. Cell Death Dis 2021; 12:1079. [PMID: 34775496 PMCID: PMC8590697 DOI: 10.1038/s41419-021-04367-3] [Citation(s) in RCA: 167] [Impact Index Per Article: 41.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 10/22/2021] [Accepted: 10/27/2021] [Indexed: 12/13/2022]
Abstract
Cetuximab is approved for the treatment of metastatic colorectal cancer (mCRC) with RAS wild-type. Nevertheless, the prognosis remains poor and the effectiveness of cetuximab is limited in KRAS mutant mCRC. Recently, emerging evidence has shown that ferroptosis, a newly discovered form of nonapoptotic cell death, is closely related to KRAS mutant cells. Here, we further investigated whether cetuximab-mediated regulation of p38/Nrf2/HO-1 promotes RSL3-induced ferroptosis and plays a pivotal role in overcoming drug resistance in KRAS mutant colorectal cancer (CRC). In our research, we used two KRAS mutant CRC cell lines, HCT116 and DLD-1, as models of intrinsic resistance to cetuximab. The viability of cells treated with the combination of RSL3 and cetuximab was assessed by the CCK-8 and colony formation assays. The effective of cetuximab to promote RSL3-induced ferroptosis was investigated by evaluating lipid reactive oxygen species accumulation and the expression of the malondialdehyde and the intracellular iron assay. Cetuximab therapy contributed to regulating the p38/Nrf2/HO-1 axis, as determined by western blotting and transfection with small interfering RNAs. Cetuximab promoted RSL3-induced ferroptosis by inhibiting the Nrf2/HO-1 in KRAS mutant CRC cells, and this was further demonstrated in a xenograft nude mouse model. Our work reveals that cetuximab enhances the cytotoxic effect of RSL3 on KRAS mutant CRC cells and that cetuximab enhances RSL3-induced ferroptosis by inhibiting the Nrf2/HO-1 axis through the activation of p38 MAPK.
Collapse
Affiliation(s)
- Jiawen Yang
- Department of Colorectal and Anal Surgery, The First Affiliated Hospital of Wenzhou Medical University, 325000, Wenzhou, People's Republic of China
| | - Jiajie Mo
- Department of Colorectal and Anal Surgery, The First Affiliated Hospital of Wenzhou Medical University, 325000, Wenzhou, People's Republic of China
| | - Juji Dai
- Department of Colorectal and Anal Surgery, The First Affiliated Hospital of Wenzhou Medical University, 325000, Wenzhou, People's Republic of China
| | - Chenqiao Ye
- Department of Colorectal and Anal Surgery, The First Affiliated Hospital of Wenzhou Medical University, 325000, Wenzhou, People's Republic of China
| | - Wei Cen
- Department of Colorectal and Anal Surgery, The First Affiliated Hospital of Wenzhou Medical University, 325000, Wenzhou, People's Republic of China
| | - Xuzhi Zheng
- Department of Colorectal and Anal Surgery, The First Affiliated Hospital of Wenzhou Medical University, 325000, Wenzhou, People's Republic of China
| | - Lei Jiang
- Central Laboratory, The First Affiliated Hospital of Wenzhou Medical University, 325000, Wenzhou, People's Republic of China.
| | - Lechi Ye
- Department of Colorectal and Anal Surgery, The First Affiliated Hospital of Wenzhou Medical University, 325000, Wenzhou, People's Republic of China.
| |
Collapse
|
20
|
Kim Y, Zharkinbekov Z, Sarsenova M, Yeltay G, Saparov A. Recent Advances in Gene Therapy for Cardiac Tissue Regeneration. Int J Mol Sci 2021; 22:9206. [PMID: 34502115 PMCID: PMC8431496 DOI: 10.3390/ijms22179206] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 08/16/2021] [Accepted: 08/16/2021] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular diseases (CVDs) are responsible for enormous socio-economic impact and the highest mortality globally. The standard of care for CVDs, which includes medications and surgical interventions, in most cases, can delay but not prevent the progression of disease. Gene therapy has been considered as a potential therapy to improve the outcomes of CVDs as it targets the molecular mechanisms implicated in heart failure. Cardiac reprogramming, therapeutic angiogenesis using growth factors, antioxidant, and anti-apoptotic therapies are the modalities of cardiac gene therapy that have led to promising results in preclinical studies. Despite the benefits observed in animal studies, the attempts to translate them to humans have been inconsistent so far. Low concentration of the gene product at the target site, incomplete understanding of the molecular pathways of the disease, selected gene delivery method, difference between animal models and humans among others are probable causes of the inconsistent results in clinics. In this review, we discuss the most recent applications of the aforementioned gene therapy strategies to improve cardiac tissue regeneration in preclinical and clinical studies as well as the challenges associated with them. In addition, we consider ongoing gene therapy clinical trials focused on cardiac regeneration in CVDs.
Collapse
Affiliation(s)
| | | | | | | | - Arman Saparov
- Department of Medicine, School of Medicine, Nazarbayev University, Nur-Sultan 010000, Kazakhstan; (Y.K.); (Z.Z.); (M.S.); (G.Y.)
| |
Collapse
|
21
|
Zhou F, Zhang X, Jiang L, Li S, Chen Y, Wu J. Pseudoginsenoside F11 Enhances the Viability of Random-Pattern Skin Flaps by Promoting TFEB Nuclear Translocation Through AMPK-mTOR Signal Pathway. Front Pharmacol 2021; 12:667524. [PMID: 33995096 PMCID: PMC8116945 DOI: 10.3389/fphar.2021.667524] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Accepted: 04/12/2021] [Indexed: 12/11/2022] Open
Abstract
Random-pattern skin flap is widely used in tissue reconstruction. However, necrosis occurring in the distal part of the flap limits its clinical application to some extent. Activation of autophagy has been considered as an effective approach to enhance the survival of skin flaps. Pseudoginsenoside F11 (PF11), an ocotillol-type saponin, is an important component of Panax quinquefolium which has been shown to confer protection against cerebral ischemia and alleviate oxidative stress. However, it is currently unknown whether PF11 induces autophagy to improve the survival of skin flaps. In this study, we investigated the effects of PF11 on blood flow and tissue edema. The results of histological examination and western blotting showed that PF11 enhanced angiogenesis, alleviated apoptosis and oxidative stress, thereby improving the survival of the flap. Further experiments showed that PF11 promoted nuclear translocation of TFEB and by regulating the phosphorylation of AMPK. In summary, this study demonstrates that PF11 activates autophagy through the AMPK-TFEB signal pathway in skin flaps and it could be a promising strategy for enhancing flap viability.
Collapse
Affiliation(s)
- Feiya Zhou
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| | - Xian Zhang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| | - Liangfu Jiang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| | - Shi Li
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| | - Yiheng Chen
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| | - Jianbin Wu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|