1
|
Zhang X, Chang X, Chai R, Zhang X, Li J, Guo Z, Qiu Z, Song Y, Shi S, Hu Y, Du B. Xin-Fu-Kang oral liquid mitigates chronic heart failure through NR4A1-Dependent regulation of endoplasmic reticulum-mitochondrial crosstalk in Cardiomyocytes. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 140:156467. [PMID: 40036990 DOI: 10.1016/j.phymed.2025.156467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 01/16/2025] [Accepted: 02/03/2025] [Indexed: 03/06/2025]
Abstract
BACKGROUND Chronic heart failure (CHF) is the terminus of a variety of cardiovascular diseases. Xin-Fu-Kang oral liquid (XFK), a natural herbal compound, has been used in CHF treatment for decades. However, further investigation is required to elucidate the fundamental mechanisms. STUDY DESIGN AND METHODS Transverse aortic constriction (TAC) was performed in mouse models. The pharmacological efficacy of XFK was confirmed by assessing cardiac function and the observation of pathological alterations in myocardial tissue. Following this, single-cell sequencing (scRNA-seq) was implemented. With the identification of XFK metabolites in rat serum via UPLC-QE MS, molecular docking was utilized to conduct preliminary validation of putative therapeutic targets. Subsequently, the phenylephrine-induced model of cardiac pressure overload was established for conducting additional verification and rescue experiments by silencing NR4A1 in vitro. RESULTS XFK intervention significantly ameliorated cardiac function in the TAC-induced CHF model. Based on scRNA-seq, cardiomyocytes exhibited the most notable alterations following XFK intervention, with NR4A1 identified as a significantly differentially expressed gene after both TAC induction and XFK intervention. In vitro experiments demonstrated that XFK enhanced mitochondrial function, mitigated oxidative stress, and restored mitophagy in a NR4A1-dependent manner, consequently decreasing apoptosis in PE-induced H9C2. Furthermore, the upstream mechanism was associated with capacity of XFK to mitigate endoplasmic reticulum stress and regulate crosstalk between the two organelles. CONCLUSION XFK counteracts cardiac chronic pressure overload through regulating NR4A1-mediated functional interaction between endoplasmic reticulum and mitochondria in cardiomyocytes, further preserves mitochondria function and prevents apoptosis. This finding indicates a novel pharmacological therapy for CHF.
Collapse
Affiliation(s)
- Xiaohan Zhang
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xing Chang
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ruoning Chai
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xuesong Zhang
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jiaran Li
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zezhen Guo
- Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - Zhiling Qiu
- Department of Internal Medicine, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuguo Song
- Beijing University of Chinese Medicine, Beijing, China
| | - Shuqing Shi
- Department of Internal Medicine, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Yuanhui Hu
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Bai Du
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
2
|
Hu H, Zhong Z, Meng L, Chen J, Yu Z, Lu K. Knockdown of NR4A1 alleviates doxorubicin-induced cardiotoxicity through inhibiting the activation of the NLRP3 inflammasome. Biochem Biophys Res Commun 2024; 700:149582. [PMID: 38306930 DOI: 10.1016/j.bbrc.2024.149582] [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: 12/15/2023] [Revised: 01/10/2024] [Accepted: 01/24/2024] [Indexed: 02/04/2024]
Abstract
Doxorubicin (DOX) is a widely used antitumor drug, but its clinical applicability is hampered by the unfortunate side effect of DOX-induced cardiotoxicity (DIC). In our current study, we retrieved three high-throughput sequencing datasets related to DIC from the Gene Expression Omnibus (GEO) datasets. We conducted differential analysis using R (DESeq2) to pinpoint differentially expressed genes (DEGs, and identified 11 genes that were consistently altered in both the control and DOX-treated groups. Notably, our Random Forest analysis of these three GEO datasets highlighted the significance of nuclear receptor subfamily 4 group A member 1 (NR4A1) in the context of DIC. The DOX-induced mouse model and cell model were used for the in vivo and in vitro studies to reveal the role of NR4A1 in DIC. We found that silencing NR4A1 by adeno-associated virus serotype 9 (AAV9) contained shRNA in vivo alleviated the DOX-induced cardiac dysfunction, cardiomyocyte injury and fibrosis. Mechanistically, we found NR4A1 silencing was able to inhibit DOX-induced the cleavage of NLRP3, IL-1β and GSDMD in vivo. Further in vitro studies have shown that inhibition of NR4A1 suppressed DOX-induced cytotoxicity and oxidative stress through the same molecular mechanism. We prove that NR4A1 plays a critical role in DOX-induced cardiotoxicity by inducing pyroptosis via activation of the NLRP3 inflammasome, and it might be a promising therapeutic target for DIC.
Collapse
Affiliation(s)
- Huanhuan Hu
- Department of Cardiology, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou Central Hospital, Zhejiang, 313000, China
| | - Zuoquan Zhong
- The First Clinical Medical College, Wenzhou Medical University, Zhejiang, 325000, China
| | - Liping Meng
- Department of Cardiology, Shaoxing People's Hospital, Zhejiang, 312000, China
| | - Jiming Chen
- Department of Cardiology, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou Central Hospital, Zhejiang, 313000, China
| | - Ziheng Yu
- Department of Cardiology, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou Central Hospital, Zhejiang, 313000, China
| | - Kongjie Lu
- Department of Cardiology, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou Central Hospital, Zhejiang, 313000, China.
| |
Collapse
|
3
|
Zhang M, Zhou N, Cao F, Liu W, Yuan H, Huang G. The role and regulatory mechanism of HIF-1α in myocardial injury in rats undergoing cardiopulmonary bypass. Acta Cardiol 2023; 78:1070-1080. [PMID: 37470433 DOI: 10.1080/00015385.2023.2229584] [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: 05/24/2023] [Accepted: 06/19/2023] [Indexed: 07/21/2023]
Abstract
BACKGROUND Hypoxia-inducible factor-1alpha (HIF-1α) is a transcription factor implicated in physiological and pathological responses to hypoxia. The present study aims to investigate the effect and mechanism of HIF-1α on cardiopulmonary bypass (CPB)-related myocardial injury, thereby conferring a theoretical basis for the clinical treatment of myocardial injury in CPB. METHODS An experimental model of CPB was established in rats by surgery. Adenovirus-packaged overexpression vectors and antiagomiRNA were used to overexpress HIF-1α and NR4A1 or inhibit miR-124-3p expression in rat myocardial tissues, respectively. qRT-PCR and Western blot detected HIF-1α, miR-124-3p, and NR4A1 expression in myocardial tissues. The rat cardiac function was monitored through an echocardiogram. The rat plasma at different stages of CPB was collected, followed by the detection of IL-6, cTnT, CK-MB, and IL-1β. TUNEL staining measured apoptosis in myocardial tissues. ChIP assay analysed the enrichment of HIF-1α on the miR-124-3p promoter. The binding relationships between HIF-1α and miR-124-3p promoter sequence and between miR-124-3p and NR4A1 3'UTR sequence were confirmed by dual-luciferase reporter assay. RESULTS HIF-1α expression had no significant change after CPB modelling. Overexpression of HIF-1α improved the cardiac function of CPB rats, decreased plasma IL-6, cTnT, CK-MB, and IL-1β levels, and reduced TUNEL-positive myocardial cells. HIF-1α was enriched on the miR-124-3p promoter and promoted miR-124-3p expression. miR-124-3p bound to NR4A1 3'UTR sequence and targeted NR4A1 expression. Inhibition of miR-124-3p or overexpression of NR4A1 partially reversed the ameliorative effect of HIF-1α overexpression on myocardial injury in CPB rats. CONCLUSION Overexpression of HIF-1α can improve myocardial injury in CPB rats via the miR-124-3p/NR4A1 axis.
Collapse
Affiliation(s)
- Mingxia Zhang
- Guangzhou Women and Children Medical Center, Guangzhou, China
| | - Na Zhou
- Guangzhou Women and Children Medical Center, Guangzhou, China
| | - Fan Cao
- Guangzhou Women and Children Medical Center, Guangzhou, China
| | - Wenhua Liu
- Guangzhou Women and Children Medical Center, Guangzhou, China
| | - Huili Yuan
- Guangzhou Women and Children Medical Center, Guangzhou, China
| | - Guodong Huang
- Guangzhou Women and Children Medical Center, Guangzhou, China
| |
Collapse
|
4
|
Li M, Hu Y, Zhou H, Chen Y. NR4A1 Aggravates Myocardial Ischaemia-Reperfusion Injury by Inhibiting OPA1-Mediated Mitochondrial Fusion. J Cardiovasc Transl Res 2023; 16:1050-1063. [PMID: 37249897 DOI: 10.1007/s12265-023-10396-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Accepted: 05/03/2023] [Indexed: 05/31/2023]
Abstract
Mitochondrial fusion is an important process that protects the myocardium. However, mitochondrial fusion is often inhibited in myocardial ischaemia-reperfusion injury (IR). The upstream mechanism of this effect is unclear. Nuclear receptor subfamily 4 group A member 1 (NR4A1) can aggravate myocardial IR and increase the level of oxidative stress, thereby affecting mitochondrial function and morphology. Inhibiting NR4A1 can improve oxidative stress levels and mitochondrial function and morphology, thereby reducing IR. Downregulating NR4A1 increases the expression level of the mitochondrial fusion-related protein optic atrophy 1 (OPA1), which is associated with these benefits. Inhibiting OPA1 expression with MYLS22 abrogates the effects of NR4A1 downregulation on IR. Furthermore, NR4A1 disrupts mitochondrial dynamics and activates the STING and NF-κB pathways. Insufficient mitochondrial fusion and increased apoptosis and inflammatory reactions worsen irreversible damage to cardiomyocytes. In conclusion, NR4A1 can exacerbate IR by inhibiting OPA1, causing mitochondrial damage.
Collapse
Affiliation(s)
- Muding Li
- Chinese PLA Medical School, Chinese PLA General Hospital, Beijing, China
| | - Yingyun Hu
- School of Medicine, Nankai University, Tianjin, China
| | - Hao Zhou
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Yundai Chen
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China.
| |
Collapse
|
5
|
Kang Q, Chai W, Min J, Qu X. Yin Yang 1 suppresses apoptosis and oxidative stress injury in SH-SY5Y cells by facilitating NR4A1 expression. J Neurogenet 2023; 37:115-123. [PMID: 37922205 DOI: 10.1080/01677063.2023.2270745] [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: 03/12/2023] [Accepted: 09/06/2023] [Indexed: 11/05/2023]
Abstract
Oxidative stress plays a significant role in the development of Parkinson's disease (PD). Previous studies implicate nuclear receptor subfamily 4 group A member 1 (NR4A1) in oxidative stress associated with PD. However, the molecular mechanism underlying the regulation of NR4A1 expression remains incompletely understood. In the present study, a PD cell model was established by using 1-methyl-4-phenylpyridinium (MPP+) in SH-SY5Y cells. Cell viability and apoptosis were assessed by using CCK-8 assay and flow cytometry, respectively. The activities of LDH and SOD, and ROS generation were used as an indicators of oxidative stress. ChIP-PCR was performed to detect the interaction between Yin Yang 1 (YY1) and the NR4A1 promoter. MPP+ treatment inhibited SH-SY5Y cell viability in a dose- and time-dependent manner. NR4A1 and YY1 expression were decreased in MPP+-treated SH-SY5Y cells. Increasing NR4A1 or YY1 alleviated MPP+-induced apoptosis and oxidative stress in SH-SY5Y cells, whereas reduction of NR4A1 aggravated MPP+-induced cell injury. Transcription factor YY1 facilitated NR4A1 expression by binding with NR4A1 promoter. In addition, in MPP+-treated SH-SY5Y cells, the inhibition of NR4A1 to apoptosis and oxidative stress was further enhanced by overexpression of YY1. The reduction of NR4A1 led to an elevation of apoptosis and oxidative stress in MPP+-induced SH-SY5Y cells, and this effect was partially reversed by the overexpression of YY1. In conclusion, YY1 suppresses MPP+-induced apoptosis and oxidative stress in SH-SY5Y cells by binding with NR4A1 promoter and boosting NR4A1 expression. Our findings suggest that NR4A1 may be a candidate target for PD treatment.HIGHLIGHTSNR4A1 and YY1 are decreased in MPP+-treated SH-SY5Y cells.NR4A1 prevents oxidative stress and apoptosis in MPP+-treated SH-SY5Y cells.YY1 binds with NR4A1 promoter and increases NR4A1 expression.YY1 enhances the inhibition of NR4A1 to SH-SY5Y cell apoptosis and oxidative stress.
Collapse
Affiliation(s)
- Qin Kang
- Department of Neurology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi Province, P.R. China
| | - Wen Chai
- Department of Neurology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi Province, P.R. China
| | - Jun Min
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, P.R. China
| | - Xinhui Qu
- Department of Neurology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi Province, P.R. China
| |
Collapse
|
6
|
Wang H, Zhang M, Fang F, Xu C, Liu J, Gao L, Zhao C, Wang Z, Zhong Y, Wang X. The nuclear receptor subfamily 4 group A1 in human disease. Biochem Cell Biol 2023; 101:148-159. [PMID: 36861809 DOI: 10.1139/bcb-2022-0331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2023] Open
Abstract
Nuclear receptor 4A1 (NR4A1), a member of the NR4A subfamily, acts as a gene regulator in a wide range of signaling pathways and responses to human diseases. Here, we provide a brief overview of the current functions of NR4A1 in human diseases and the factors involved in its function. A deeper understanding of these mechanisms can potentially improve drug development and disease therapy.
Collapse
Affiliation(s)
- Hongshuang Wang
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050091, China
| | - Mengjuan Zhang
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050091, China
| | - Fang Fang
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050091, China
| | - Chang Xu
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050091, China
| | - Jiazhi Liu
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050091, China
| | - Lanjun Gao
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050091, China
| | - Chenchen Zhao
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050091, China
| | - Zheng Wang
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns Research, Shijiazhuang 050091, China.,Institute of Integrative Medicine, College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Yan Zhong
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns Research, Shijiazhuang 050091, China.,Institute of Integrative Medicine, College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Xiangting Wang
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns Research, Shijiazhuang 050091, China
| |
Collapse
|
7
|
Zhang Y, Lu J, Ma Y, Sun L, Wang S, Yue X, Yu J, Xue P. Establishment of fingerprint and mechanism of anti-myocardial ischemic effect of Syringa pinnatifolia. Biomed Chromatogr 2022; 36:e5475. [PMID: 35947036 DOI: 10.1002/bmc.5475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 07/19/2022] [Accepted: 07/26/2022] [Indexed: 11/10/2022]
Abstract
OBJECTIVE To establish the fingerprint of Syringa pinnatifolia Hemsl. (SP), analyze the blood components of SP, and explore the possible mechanism of SP's anti-myocardial ischemia, so as to provide scientific basis for the follow-up development and research of SP and lay a foundation for its clinical application. METHODS The fingerprint of SP was established by UPLC-QE-MS and GC-MS. A rat Myocardial infarction (MI) was constructed by ligating the left anterior descending branch (LAD) of the rat coronary artery, and SP alcohol extract was administered to evaluate its anti-myocardial ischemic effect. We analyzed the blood components of SP, screened the active compounds, established a database of SP anti-myocardial ischemic targets, and explored the possible mechanism of SP in treating MI by bioinformatics. The rats were examined by echocardiography, serum biomarkers were determined, and pathological changes were observed by histopathological examination. TUNEL staining was performed to detect the apoptotic level of cells, and western blot and qRT-PCR were performed to detect the expression levels of Bcl-2, Bax and caspase-3 in heart tissues. RESULTS In the fingerprint of SP, 24 common peaks were established, and the similarity evaluation results of 10 batches of SP were all > 0.9. UPLC-QE-MS and GC-MS detected a total of 17 active ingredients in the drug-containing serum, including terpenoids, flavonoids, phenols, phenylpropanoids and phenolic acids, the most abundant of which was resveratrol. Enrichment analysis of SP targets against myocardial ischemia revealed that key candidate targets of SP were significantly enriched in multiple pathways associated with apoptosis. Resveratrol was administered to the successfully modeled rats, and the results showed that the resveratrol group significantly reduced LVEDd and LVEDs and significantly increased EF and FS in all groups compared with the model group. Resveratrol significantly reduced the levels of CK-MB and LDH in serum compared to the model group (p < 0.001). Hematoxylin-eosin (HE) staining of rat myocardial tissue showed that all lesions were reduced under microscopic observation in the resveratrol group compared with the model group. RT-PCR and western blot results showed that resveratrol group down-regulated the expression of the pro-apoptotic factor Bax, up-regulated the expression of the anti-apoptotic factor Bcl-2, and decreased the expression of Caspase-3. CONCLUSION The established fingerprints are accurate, reliable and reproducible, and can be used as an effective method for the quality control of the herbs. The anti-myocardial ischemia effect of SP may be that resveratrol can improve cardiac function and inhibit cardiomyocyte apoptosis to protect cardiomyocytes. The present study provides ample evidence for the clinical use of SP, suggesting that this drug has great potential in the treatment of ischemic heart disease.
Collapse
Affiliation(s)
- Ye Zhang
- College of Pharmacy, Inner Mongolia Medical University, Hohhot, Inner Mongolia, PR China
| | - Jingkun Lu
- College of Basic Medicine, Inner Mongolia Medical University, Hohhot, Inner Mongolia, PR China
| | - Yuheng Ma
- College of Pharmacy, Inner Mongolia Medical University, Hohhot, Inner Mongolia, PR China
| | - Lijun Sun
- College of Pharmacy, Inner Mongolia Medical University, Hohhot, Inner Mongolia, PR China
| | - Suwei Wang
- College of Pharmacy, Inner Mongolia Medical University, Hohhot, Inner Mongolia, PR China
| | - Xin Yue
- College of Pharmacy, Inner Mongolia Medical University, Hohhot, Inner Mongolia, PR China
| | - Jiuwang Yu
- College of Pharmacy, Inner Mongolia Medical University, Hohhot, Inner Mongolia, PR China
| | - Peifeng Xue
- College of Pharmacy, Inner Mongolia Medical University, Hohhot, Inner Mongolia, PR China
| |
Collapse
|