1
|
Zaidi SA, Fan Z, Chauhdari T, Ding Y. MicroRNA regulatory dynamic, emerging diagnostic and therapeutic frontier in atherosclerosis. Microvasc Res 2025; 160:104818. [PMID: 40368159 DOI: 10.1016/j.mvr.2025.104818] [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: 03/01/2025] [Revised: 05/07/2025] [Accepted: 05/09/2025] [Indexed: 05/16/2025]
Abstract
MicroRNAs (miRNAs), a class of non-coding RNAs, are pivotal post-transcriptional regulators of gene expression with profound implications in the pathogenesis of atherosclerosis (AS). AS a progressive arterial disease driven by vascular cells dysfunction, lipid dysregulation and subsequent chronic inflammation, AS remains a leading cause of global morbidity. Recent studies have demonstrated how important miRNAs are in regulating central biological processes in the vascular wall, such as endothelial function, vascular smooth muscle cell (VSMC) phenotypic switching, and macrophage polarization. This review provides comprehensive insight into the role of miRNAs in the development and complexity of atherosclerotic plaques according to their effects on endothelial cells, macrophages, and VSMCs. We also go over the growing prospects of miRNAs as therapeutic targets and diagnostic biomarkers, providing information to be used in the study of vascular diseases. Lastly, we address recent complications and potential applications of miRNA-based approaches in clinical practice.
Collapse
Affiliation(s)
- Syeda Armana Zaidi
- College of Life Sciences, University of Chinese Academy of Sciences, No.1 Yanqihu East Rd, Huairou District, Beijing 101408, PR China.
| | - Zhiyu Fan
- College of Life Sciences, University of Chinese Academy of Sciences, No.1 Yanqihu East Rd, Huairou District, Beijing 101408, PR China.
| | - Talha Chauhdari
- College of Life Sciences, University of Chinese Academy of Sciences, No.1 Yanqihu East Rd, Huairou District, Beijing 101408, PR China.
| | - Yongsheng Ding
- College of Life Sciences, University of Chinese Academy of Sciences, No.1 Yanqihu East Rd, Huairou District, Beijing 101408, PR China.
| |
Collapse
|
2
|
Zhang L, Lin L, Xiao B, Huang K, Liu J, Chen YL, Lu L, Zhang Z, Zhang L, Li J, Ho KT, Luo L, Huang SY, Li G. Eicosapentaenoic acid (EPA) reduced lipopolysaccharide-stimulated inflammatory response of RAW264.7 cells via the miR-125b-5p/CREB axis. Int J Biol Macromol 2025; 310:143511. [PMID: 40286950 DOI: 10.1016/j.ijbiomac.2025.143511] [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: 12/22/2024] [Revised: 04/13/2025] [Accepted: 04/24/2025] [Indexed: 04/29/2025]
Abstract
Inflammation represents an adaptive physiological response of body immune system to infection or tissue damage, which may be regulated by food supplementation. Eicosapentaenoic acid (EPA) has multi-functions and its anti-inflammatory effect has gained great attention. This study aimed to address the exact molecular mechanism underlying its inflammatory control. The results showed that EPA decreased lipopolysaccharide-induced inflammatory response in RAW264.7 cells by modulating the production of cellular cytokines. In addition, EPA downregulated miR-125b-5p, which showed pro-inflammatory effect and its forced expression attenuated EPA's anti-inflammatory activity. Moreover, the cAMP-responsive element-binding protein (CREB) is targeted by miR-125b-5p. CREB overexpression reduced inflammation probably via modulating the PGC-1α/NF-κB pathway, which resembled the effect of EPA pre-treatment. Therefore, EPA exhibited anti-inflammatory activity by targeting the miR-125b-5p/CREB axis, which modulated the production of inflammatory mediators probably via transcription control. This study provides insights into microRNA-mediated action mechanism and facilitates the relief of inflammation-associated diseases by food ingredients.
Collapse
Affiliation(s)
- Liyuan Zhang
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen Fujian 361021, PR China; The National & Local Joint Engineering Research Center of Deep Processing Technology for Aquatic Products, Xiamen, Fujian 361021, PR China; Fujian Marine Functional Food Engineering Technology Research Center, Xiamen, Fujian 361021, PR China; Xiamen Marine Functional Food Key Laboratory, Xiamen, Fujian 361021, PR China
| | - Lingli Lin
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen Fujian 361021, PR China; The National & Local Joint Engineering Research Center of Deep Processing Technology for Aquatic Products, Xiamen, Fujian 361021, PR China; Fujian Marine Functional Food Engineering Technology Research Center, Xiamen, Fujian 361021, PR China; Xiamen Marine Functional Food Key Laboratory, Xiamen, Fujian 361021, PR China
| | - Baoping Xiao
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen Fujian 361021, PR China; The National & Local Joint Engineering Research Center of Deep Processing Technology for Aquatic Products, Xiamen, Fujian 361021, PR China; Fujian Marine Functional Food Engineering Technology Research Center, Xiamen, Fujian 361021, PR China; Xiamen Marine Functional Food Key Laboratory, Xiamen, Fujian 361021, PR China
| | - Kaiyan Huang
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen Fujian 361021, PR China; The National & Local Joint Engineering Research Center of Deep Processing Technology for Aquatic Products, Xiamen, Fujian 361021, PR China; Fujian Marine Functional Food Engineering Technology Research Center, Xiamen, Fujian 361021, PR China; Xiamen Marine Functional Food Key Laboratory, Xiamen, Fujian 361021, PR China
| | - Jingwen Liu
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen Fujian 361021, PR China
| | - Yu-Lei Chen
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen Fujian 361021, PR China; The National & Local Joint Engineering Research Center of Deep Processing Technology for Aquatic Products, Xiamen, Fujian 361021, PR China; Fujian Marine Functional Food Engineering Technology Research Center, Xiamen, Fujian 361021, PR China; Xiamen Marine Functional Food Key Laboratory, Xiamen, Fujian 361021, PR China
| | - Liming Lu
- Shanghai Institute of Immunology, School of Medicine, Shanghai Jiaotong University, Shanghai 200025, PR China
| | - Zhengxiao Zhang
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen Fujian 361021, PR China; The National & Local Joint Engineering Research Center of Deep Processing Technology for Aquatic Products, Xiamen, Fujian 361021, PR China; Fujian Marine Functional Food Engineering Technology Research Center, Xiamen, Fujian 361021, PR China; Xiamen Marine Functional Food Key Laboratory, Xiamen, Fujian 361021, PR China
| | - Lingyu Zhang
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen Fujian 361021, PR China; The National & Local Joint Engineering Research Center of Deep Processing Technology for Aquatic Products, Xiamen, Fujian 361021, PR China; Fujian Marine Functional Food Engineering Technology Research Center, Xiamen, Fujian 361021, PR China; Xiamen Marine Functional Food Key Laboratory, Xiamen, Fujian 361021, PR China
| | - Jian Li
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen Fujian 361021, PR China; The National & Local Joint Engineering Research Center of Deep Processing Technology for Aquatic Products, Xiamen, Fujian 361021, PR China; Fujian Marine Functional Food Engineering Technology Research Center, Xiamen, Fujian 361021, PR China; Xiamen Marine Functional Food Key Laboratory, Xiamen, Fujian 361021, PR China
| | - Kuo-Ting Ho
- Center for Precision Medicine, Quanzhou, Fujian 362123, PR China; HI. Q Biomedical Laboratory, Quanzhou, Fujian 362123, PR China
| | - Lianzhong Luo
- Engineering Research Center of Marine Biopharmaceutical Resource, Fujian Province University, Xiamen Medical College, Xiamen, Fujian 361023, PR China
| | - Shi-Ying Huang
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen Fujian 361021, PR China; The National & Local Joint Engineering Research Center of Deep Processing Technology for Aquatic Products, Xiamen, Fujian 361021, PR China; Fujian Marine Functional Food Engineering Technology Research Center, Xiamen, Fujian 361021, PR China; Xiamen Marine Functional Food Key Laboratory, Xiamen, Fujian 361021, PR China.
| | - Guiling Li
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen Fujian 361021, PR China; The National & Local Joint Engineering Research Center of Deep Processing Technology for Aquatic Products, Xiamen, Fujian 361021, PR China; Fujian Marine Functional Food Engineering Technology Research Center, Xiamen, Fujian 361021, PR China; Xiamen Marine Functional Food Key Laboratory, Xiamen, Fujian 361021, PR China.
| |
Collapse
|
3
|
Heo KS, Phan LP, Le NTT, Jin Y. Mechanistic insights and emerging therapeutic strategies targeting endothelial dysfunction in cardiovascular diseases. Arch Pharm Res 2025; 48:305-332. [PMID: 40301174 DOI: 10.1007/s12272-025-01542-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Accepted: 04/08/2025] [Indexed: 05/01/2025]
Abstract
Endothelial dysfunction plays a pivotal role in the pathogenesis of various cardiovascular diseases (CVDs), including atherosclerosis, hypertension, heart failure, stroke, and peripheral artery disease. It disrupts vascular homeostasis, leading to reduced nitric oxide (NO) bioavailability, increased oxidative stress, and chronic inflammation, all of which collectively drive vascular damage, atherosclerotic plaque formation, and thrombosis. Additionally, shear stress-induced alterations in blood flow patterns, particularly disturbed flow (d-flow), aggravate endothelial dysfunction. Furthermore, the endothelial-to-mesenchymal transition (EndMT), a process in which endothelial cells acquire mesenchymal-like properties, contributes to vascular remodeling and accelerates CVD progression.This review explores the significant role of epigenetic mechanisms, such as DNA methylation, histone modifications, and noncoding RNAs (ncRNAs), which serve as critical regulators of endothelial function in response to shear stress in endothelial dysfunction and the development of atherosclerosis. Furthermore, we discuss the pivotal role of endothelial dysfunction in cardiovascular and metabolic diseases, emphasizing the need for innovative therapeutic strategies beyond conventional treatments. In particular, we highlight the endothelial-protective mechanisms of emerging pharmacological agents, including proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors, glucagon-like peptide-1 receptor agonists (GLP-1RAs), and sodium-glucose cotransporter 2 (SGLT2) inhibitors, along with supporting clinical evidence demonstrating their efficacy in improving endothelial function and reducing cardiovascular risk.
Collapse
Affiliation(s)
- Kyung-Sun Heo
- Department of Pharmacology, Chungnam National University, College of Pharmacy, 99 Daehak-ro, Yuseong-gu, Daejeon, 34134, Republic of Korea.
| | - Lan Phuong Phan
- Department of Pharmacology, Chungnam National University, College of Pharmacy, 99 Daehak-ro, Yuseong-gu, Daejeon, 34134, Republic of Korea
| | - Nhi Thi Thao Le
- Department of Pharmacology, Chungnam National University, College of Pharmacy, 99 Daehak-ro, Yuseong-gu, Daejeon, 34134, Republic of Korea
| | - Yujin Jin
- Department of Pharmacology, Chungnam National University, College of Pharmacy, 99 Daehak-ro, Yuseong-gu, Daejeon, 34134, Republic of Korea
| |
Collapse
|
4
|
Ryszkiewicz P, Schlicker E, Malinowska B. Is Inducible Nitric Oxide Synthase (iNOS) Promising as a New Target Against Pulmonary Hypertension? Antioxidants (Basel) 2025; 14:377. [PMID: 40298665 PMCID: PMC12024173 DOI: 10.3390/antiox14040377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 03/12/2025] [Accepted: 03/19/2025] [Indexed: 04/30/2025] Open
Abstract
Pulmonary hypertension (PH) is a progressive disease characterized by elevated blood pressure in the pulmonary arteries, associated also with inflammation and oxidative stress. Inducible nitric oxide synthase (iNOS) is one of the key mediators of inflammation and immune system activation. Although preclinical studies mostly suggest a detrimental role of iNOS overactivation in PH, there is a lack of exhaustive analyses and summaries. Therefore, this literature overview aims to fill this gap. The involvement of iNOS in the pathogenesis of the four main clinical groups of PH is discussed to assess whether targeting iNOS could be a promising way to treat PH. iNOS expression patterns in the organs primarily affected by PH are analyzed both in animals and in humans. Consequently, the effectiveness of pharmacological iNOS inhibition and/or iNOS gene deletion is discussed and compared, also with reference to the activity of constitutive NOS isoforms, particularly endothelial NOS (eNOS). Overall, our overview suggests that selective iNOS inhibitors could be considered as a novel treatment strategy for PH, as decreases in right ventricular and pulmonary artery pressure, the alleviation of ventricular hypertrophy, and improvements of pulmonary and cardiac function were observed, among others. Nevertheless, further research efforts in this area are needed.
Collapse
Affiliation(s)
- Piotr Ryszkiewicz
- Department of Experimental Physiology and Pathophysiology, Medical University of Bialystok, Mickiewicz Str. 2A, 15-222 Bialystok, Poland
| | - Eberhard Schlicker
- Department of Pharmacology and Toxicology, University of Bonn, Venusberg Campus 1, 53127 Bonn, Germany;
| | - Barbara Malinowska
- Department of Experimental Physiology and Pathophysiology, Medical University of Bialystok, Mickiewicz Str. 2A, 15-222 Bialystok, Poland
| |
Collapse
|
5
|
Zhu M, Tai S. Chrysophanol Mitigates Chronic Heart Failure in Rats by Modulating ROS-Mediated Parthanatos and Pyroptosis. Int Heart J 2025; 66:126-136. [PMID: 39894541 DOI: 10.1536/ihj.24-387] [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] [Indexed: 02/04/2025]
Abstract
Chronic heart failure (CHF) triggers a cascade of events involving parthanatos and pyroptosis, culminating in cellular malfunction, inflammation, and tissue degeneration. This study aims to inquire into the inherent mechanism of chrysophanol (CHR) in the treatment of CHF.In vitro, we cultured the rat embryonic cardiomyocyte cell line H9c2. Parthanatos was initiated through N-methyl-N'-nitro-N'-nitrosoguanidine (MNNG) induction, followed by treatment with varying concentrations of CHR. The evaluation of parthanatos and pyroptosis in cardiomyocytes was assessed by western blotting. In vivo, the transverse aortic constriction (TAC) model was used to simulate CHF. The hemodynamic indices were performed to evaluate cardiac function in rats. The degree of inflammatory cell infiltration and fibrosis within cardiac tissue was assessed using hematoxylin and eosin staining and Masson's trichrome staining, respectively. Cardiac tissues were obtained and subjected to immunohistochemical analysis to assess PARP-1 expression. Subsequently, dual immunofluorescence staining (caspase-1 and NLRP3) was conducted, aiming to comprehensively evaluate the status of parthanatos and pyroptosis in the cardiac tissues of rats.In contrast to the MNNG or TAC group, the groups administered with CHR exhibited an inhibitory effect on Reactive oxygen species (ROS) expression, as well as parthanatos and pyroptosis proved by cell and animal experiments (P < 0.05). The reduced expression of PAR, PARP-1, AIF, NLRP3, IL-1β, caspase-1, and cleaved-GSDMD compared with the MNNG or TAC group proved it (P < 0.05). Moreover, compared with the TAC group, CHR significantly improved the cardiac histology of TAC rats. These findings collectively suggested the potential of CHR in ameliorating CHF.CHR may mitigate CHF in rats by modulating ROS-mediated parthanatos and pyroptosis.
Collapse
Affiliation(s)
- Mengjiao Zhu
- Department of Neurology, Wenzhou City Hospital of Integrated Traditional Chinese and Western Medicine
| | - Sichao Tai
- Department of Cardiology, The Second Affiliated Hospital of Wenzhou Medical University
| |
Collapse
|
6
|
Yibulayin K, Abulizi M. The function of miRNAs in the immune system's inflammatory reaction to heart failure. Front Cardiovasc Med 2024; 11:1506836. [PMID: 39687084 PMCID: PMC11646975 DOI: 10.3389/fcvm.2024.1506836] [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: 10/08/2024] [Accepted: 11/18/2024] [Indexed: 12/18/2024] Open
Abstract
Heart failure is the end stage of cardiovascular disease, with high morbidity and mortality rates worldwide. Heart failure is associated with long-term and insufficient inhibition of inflammatory response. miRNA is a class of endogenous, non-coding, single-stranded small RNA molecules, that can regulate gene expression through translational inhibition or degradation of targeted mRNA, widely regulate myocardial remodeling, inflammatory response, and other pathological processes, and play an important regulatory role in the occurrence and development of cardiovascular diseases. This article reviews the role of miRNA in the inflammatory response in heart failure.
Collapse
|
7
|
Wang J, Huang Y, Wu X, Li D. MicroRNA-939 amplifies Staphylococcus aureus-induced matrix metalloproteinase expression in atopic dermatitis. Front Immunol 2024; 15:1354154. [PMID: 38903509 PMCID: PMC11188349 DOI: 10.3389/fimmu.2024.1354154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 05/21/2024] [Indexed: 06/22/2024] Open
Abstract
Background Atopic dermatitis (AD) is a common chronic inflammatory skin diseases that seriously affects life quality of the patients. Staphylococcus aureus (S. aureus) colonization on the skin plays an important role in the pathogenesis of AD; however, the mechanism of how it modulates skin immunity to exacerbate AD remains unclear. MicroRNAs are short non-coding RNAs that act as post-transcriptional regulators of genes. They are involved in the pathogenesis of various inflammatory skin diseases. Methods In this study, we established miRNA expression profiles for keratinocytes stimulated with heat-killed S. aureus (HKSA). The expression of miR-939 in atopic dermatitis patients was analyzed by fluorescence in situ hybridization (FISH). miR-939 mimic was transfected to human primary keratinocyte to investigate its impact on the expression of matrix metalloproteinase genes (MMPs) in vitro. Subsequently, miR-939, along with Polyplus transfection reagent, was administered to MC903-induced atopic dermatitis skin to assess its function in vivo. Results MiR-939 was highly upregulated in HKSA-stimulated keratinocytes and AD lesions. In vitro studies revealed that miR-939 increased the expression of matrix metalloproteinase genes, including MMP1, MMP3, and MMP9, as well as the cell adhesion molecule ICAM1 in human primary keratinocytes. In vivo studies indicated that miR-939 increased the expression of matrix metalloproteinases to promote the colonization of S. aureus and exacerbated S. aureus-induced AD-like skin inflammation. Conclusions Our work reveals miR-939 is an important regulator of skin inflammation in AD that could be used as a potential therapeutic target for AD.
Collapse
Affiliation(s)
- Jiating Wang
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, China
| | - Yejing Huang
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, China
| | - Xinfeng Wu
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, China
| | - Dongqing Li
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, China
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Nanjing, China
| |
Collapse
|
8
|
Xu W, Huang Y, Lei Z, Zhou J. miR-939-3p induces sarcoma proliferation and poor prognosis via suppressing BATF2. Front Oncol 2024; 14:1346531. [PMID: 38420020 PMCID: PMC10899471 DOI: 10.3389/fonc.2024.1346531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 01/31/2024] [Indexed: 03/02/2024] Open
Abstract
Background Sarcoma is a rare and aggressive malignancy with poor prognosis, in which oncogene activation and tumor suppressor inactivation are involved. Accumulated studies suggested basic leucine zipper transcription factor ATF-like 2 (BATF2) as a candidate tumor suppressor, but its specific role and mechanism in sarcoma remain unclear. Methods The expression levels of BATF2 and miR-939-3p were evaluated by using human sarcoma samples, cell lines and xenograft mouse models. Bioinformatics analysis, qPCR, Western blot, cell proliferation assay, overexpression plasmid construction, point mutation and dual luciferase reporter assay were utilized to investigate the role and mechanism of miR-939-3p in sarcoma. Results In this study, we demonstrated that the expression of BATF2 was downregulated in human sarcoma tissues and cell lines. The downregulation of BATF2 was negatively associated with the prognosis of sarcoma patients. Subsequent bioinformatic prediction and experimental validations showed that BATF2 expression was reduced by microRNA (miR)-939-3p mimic and increased by miR-939-3p inhibitor. Additionally, miR-939-3p was upregulated in sarcoma tissues and cells, correlating with a poor prognosis of sarcoma patients. Moreover, miR-939-3p overexpression suppressed sarcoma cell proliferation, which was significantly attenuated by the restoration of BATF2, while siRNA-mediated knockdown of BATF2 aggravated the miR-939-3p-induced promotion of sarcoma cell proliferation. Further computational algorithms and dual-luciferase reporter assays demonstrated that miR-939-3p repressed BATF2 expression via directly binding to its 3' untranslated region (3' UTR). Conclusion Collectively, these findings identified miR-939-3p as a novel regulator of BATF2, as well as a prognostic biomarker in sarcoma, and revealed that suppressing miR-939-3p or inducing BATF2 expression may serve as a promising therapeutic strategy against sarcoma.
Collapse
Affiliation(s)
- Wanwen Xu
- Wuhan Third Hospital (Tongren Hospital of Wuhan University), Wuhan, Hubei, China
| | - Yinghui Huang
- Department of Nephrology, The Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Zengjie Lei
- Department of Medical Oncology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Jie Zhou
- Department of Oncology and Southwest Cancer Center, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| |
Collapse
|
9
|
Wilmes V, Mildeberger L, Verhoff MA, Kauferstein S. Influence of microRNAs on iNOS expression in postmortem human infarction hearts. Forensic Sci Int 2024; 354:111892. [PMID: 38150896 DOI: 10.1016/j.forsciint.2023.111892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 11/14/2023] [Accepted: 11/21/2023] [Indexed: 12/29/2023]
Abstract
MicroRNAs (miRNAs) are important post-transcriptional regulators in several diseases, including cancer, immunologic and cardiovascular diseases. A growing list of miRNAs are dysregulated in cardiac arrhythmias, contractility diseases, myocardial infarction (MI), sudden cardiac death (SCD), chronic heart failure and hypertrophy. However, the exact regulatory pathways, through which miRNAs exert their effects are often unclear. In this study, we measured the expression patterns of miR-21, miR-939 and miR-30e in postmortem human MI. The aim of the study was to examine the influence of these miRNAs on cardiac inducible nitric oxide synthase (iNOS) mRNA levels. We measured iNOS mRNA and miRNA expression patterns by means of qPCR. Further we used correlation analyses to determine causality between miRNA expression and cardiac iNOS levels. iNOS mRNA, miR-21, miR-939 and miR-30e were significantly upregulated in infarcted and non-infarcted regions of postmortem human MI hearts in comparison to healthy controls. While miR-21 and miR-939 showed their strongest expression in infarcted regions, miR-30e peaked in the non-infarcted myocardium. Further, we found a significant correlation between miR-939 and iNOS expression levels in controls and infarcted regions. The results indicate, that miR-939 is a regulator of cardiac iNOS expression. However, a massive iNOS activation might exceed the capability of miR-939 to keep its expression in balance. miR-21 and miR-30e do not seem to influence cardiac iNOS levels in MI. Further studies are needed to evaluate downstream targets of these miRNAs and their signaling pathways to clarify their role in human MI.
Collapse
Affiliation(s)
- Verena Wilmes
- Institute of Legal Medicine, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany.
| | - Luise Mildeberger
- Institute of Legal Medicine, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Marcel A Verhoff
- Institute of Legal Medicine, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Silke Kauferstein
- Institute of Legal Medicine, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| |
Collapse
|
10
|
Zhang Y, Zhu W, Wang J, Zuo Y. Identification of biomarkers associated with immune scores in diabetic retinopathy. Front Endocrinol (Lausanne) 2023; 14:1228843. [PMID: 37867507 PMCID: PMC10585271 DOI: 10.3389/fendo.2023.1228843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 09/11/2023] [Indexed: 10/24/2023] Open
Abstract
Background Diabetic retinopathy (DR) causes irreversible visual impairment in diabetes mellitus (DM) patients. Immunity played a crucial role in DR. Nevertheless, the triggering mechanism of DR was not yet thorough enough. Herein, we aim to identify the immune-associated genes as biomarkers associated with immune scores that can distinguish early DR from DM without DR. Methods In this study, total RNA of peripheral blood mononuclear cell (PBMC) samples from 15 non-proliferative DR patients and 15 DM patients without DR were collected and the transcriptome sequencing data were extracted. Firstly, the target genes were obtained by intersecting the differentially expressed genes (DEGs), which were screened by "limma", and the module genes (related to immune scores), which were screened by "WGCNA". In order to screen for the crucial genes, three machine learning algorithms were implemented, and a receiver operating characteristic (ROC) curve was used to obtain the diagnostic genes. Moreover, the gene set enrichment analysis (GSEA) was performed to understand the function of diagnostic genes, and analysis of the proportions of immune cells and their association with diagnostic genes was performed to analyze the pathogenesis of DR. Furthermore, the regulatory network of TF-mRNA-miRNA was built to reveal the possible regulation of diagnostic genes. Finally, the quantitative real-time polymerase chain reaction (qRT-PCR) was performed to verify the mRNA level of diagnostic genes. Results A total of three immune-associated diagnostic genes, namely, FAM209B, POM121L1P, and PTGES, were obtained, and their expression was increased in PBMC samples of DR, and qRT-PCR results confirmed these results. Moreover, the functions of these genes were associated with immune response. The expression of POM121L1P and PTGES was significantly negatively associated with naive B cells, and the expression of FAM209B was significantly negatively associated with immature dendritic cells. Moreover, ESR1 could regulate both FAM209B and PTGES. Conclusion This study identified three immune-associated diagnostic genes, FAM209B, POM121L1P, and PTGES, as biomarkers associated with immune scores in DR for the first time. This finding might proffer a novel perspective of the triggering mechanism of DR, and help to understand the role of immune-associated genes in the molecular mechanism of DR more deeply.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Ophthalmology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Weidong Zhu
- Department of Spinal Surgery, No. 215 Hospital of Shaanxi Nuclear Industry, Xianyang, China
| | - Jianming Wang
- Department of Ophthalmology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Yi Zuo
- Department of Neurosurgery, No. 215 Hospital of Shaanxi Nuclear Industry, Xianyang, China
| |
Collapse
|
11
|
Li K, Ma L, Lu Z, Yan L, Chen W, Wang B, Xu H, Asemi Z. Apoptosis and heart failure: The role of non-coding RNAs and exosomal non-coding RNAs. Pathol Res Pract 2023; 248:154669. [PMID: 37422971 DOI: 10.1016/j.prp.2023.154669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 07/01/2023] [Accepted: 07/02/2023] [Indexed: 07/11/2023]
Abstract
Heart failure is a condition that affects the cardio vascular system and occurs if the heart cannot adequately pump the oxygen and blood to the body. Myocardial infarction, reperfusion injury, and this disease is the only a few examples of the numerous cardiovascular illnesses that are impacted by the closely controlled cell deletion process known as apoptosis. Attention has been paid to the creation of alternative diagnostic and treatment modalities for the condition. Recent evidences have shown that some non-coding RNAs (ncRNAs) influence the stability of proteins, control of transcription factors, and HF apoptosis through a variety of methods. Exosomes make a significant paracrine contribution to the regulation of illnesses as well as to the communication between nearby and distant organs. However, it has not yet been determined whether exosomes regulate the cardiomyocyte-tumor cell interaction in ischemia HF to limit the vulnerability of malignancy to ferroptosis. Here, we list the numerous ncRNAs in HF that are connected to apoptosis. In addition, we emphasize the significance of exosomal ncRNAs in the HF.
Collapse
Affiliation(s)
- Ketao Li
- Department of cardiology, Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou, Zhejiang 310022, China
| | - Liping Ma
- Department of cardiology, Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou, Zhejiang 310022, China
| | - Zhiwei Lu
- Hangzhou Heyunjia Hospital, Hangzhou, Zhe'jiang 310000, China
| | - Laixing Yan
- Department of cardiology, Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou, Zhejiang 310022, China
| | - Wan Chen
- Department of Cardiology, Jiulongpo First People's Hospital, Chongqing 400051, China
| | - Bing Wang
- Department of cardiology, Zouping People's Hospital, Zouping, Shandong 256299, China
| | - Huiju Xu
- Department of cardiology, Hangzhou Mingzhou Hospital, Hangzhou, Zhe'jiang 311215, China.
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran.
| |
Collapse
|
12
|
Fan M, Zhang J, Zeng L, Wang D, Chen J, Xi X, Long J, Huang J, Li X. Non-coding RNA mediates endoplasmic reticulum stress-induced apoptosis in heart disease. Heliyon 2023; 9:e16246. [PMID: 37251826 PMCID: PMC10209419 DOI: 10.1016/j.heliyon.2023.e16246] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 05/04/2023] [Accepted: 05/10/2023] [Indexed: 05/31/2023] Open
Abstract
Apoptosis is a complex and highly self-regulating form of cell death, which is an important cause of the continuous decline in ventricular function and is widely involved in the occurrence and development of heart failure, myocardial infarction, and myocarditis. Endoplasmic reticulum stress plays a crucial role in apoptosis-inducing. Accumulation of misfolded or unfolded proteins causes cells to undergo a stress response called unfolded protein response (UPR). UPR initially has a cardioprotective effect. Nevertheless, prolonged and severe ER stress will lead up to apoptosis of stressed cells. Non-coding RNA is a type of RNA that does not code proteins. An ever-increasing number of studies have shown that non-coding RNAs are involved in regulating endoplasmic reticulum stress-induced cardiomyocyte injury and apoptosis. In this study, the effects of miRNA and LncRNA on endoplasmic reticulum stress in various heart diseases were mainly discussed to clarify their protective effects and potential therapeutic strategies for apoptosis.
Collapse
Affiliation(s)
- Mingyuan Fan
- Department of Senile Disease, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
| | - Jing Zhang
- Department of Senile Disease, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
| | - Lei Zeng
- Department of Senile Disease, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
| | - Danpeng Wang
- Department of Senile Disease, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
| | - Jiao Chen
- Department of Senile Disease, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
| | - Xiaorong Xi
- Department of Senile Disease, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
| | - Jing Long
- Department of Senile Disease, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
| | - Jinzhu Huang
- Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Xueping Li
- Department of Senile Disease, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
| |
Collapse
|
13
|
Dong X, Han X, Zhang X, Li S, Li Z, Kang J, Jiang J, Ni S, Lu L, He Z, Huang H, Xian S, Yuan T, Yang Z, Long W, Wan Z. A Simplified Herbal Formula Improves Cardiac Function and Reduces Inflammation in Mice Through the TLR-Mediated NF-κB Signaling Pathway. Front Pharmacol 2022; 13:865614. [PMID: 35734399 PMCID: PMC9207450 DOI: 10.3389/fphar.2022.865614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 05/16/2022] [Indexed: 12/03/2022] Open
Abstract
Nuanxinkang tablet (NXK), a Chinese herbal formula, can improve heart function and quality of life in patients with chronic heart failure (CHF). However, the mechanisms of action of NXK are not fully understood. In this study, we investigated the effects of NXK on inflammation in the CHF mouse model. This model was established by transverse aortic constriction (TAC) and treated with NXK for 8 weeks. Then, the cardiac function and myocardial fibrosis were evaluated. The monocytes/macrophages were evaluated by immunofluorescence. The mRNA levels of IL-1β, IL-6, TNF-α, ICAM-1, and VCAM-1 were measured by quantitative real-time polymerase chain reaction (qRT-PCR), while TLR4, MyD88, NF-κB p65, P-IκBα, TLR2, TLR7 and TLR9 protein levels were evaluated by Western blot. The results showed that NXK improved the left ventricular ejection fraction (LVEF) and left ventricular end-systolic dimension, reversed myocardial fibrosis, and inhibited pro-inflammatory (CD11b + Ly6C+) monocytes/macrophages in the TAC mouse model. NXK also reduced the mRNA and protein levels of the above markers. Taken together, NXK improved heart function and reduced inflammation through the TLR-mediated NF-κB signaling pathway, suggesting that it might be used as an innovative treatment strategy for CHF.
Collapse
Affiliation(s)
- Xiaoming Dong
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaowei Han
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaojiao Zhang
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Sijing Li
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ziru Li
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jinhua Kang
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jialin Jiang
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shihao Ni
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lu Lu
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhiling He
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Cardiology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Haoming Huang
- Department of Radiology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shaoxiang Xian
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Geriatrics, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Tianhui Yuan
- Department of Geriatrics, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhongqi Yang
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Geriatrics, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- *Correspondence: Zhongqi Yang, ; Wenjie Long, ; Zemin Wan,
| | - Wenjie Long
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Geriatrics, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- *Correspondence: Zhongqi Yang, ; Wenjie Long, ; Zemin Wan,
| | - Zemin Wan
- Department of Laboratory Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- *Correspondence: Zhongqi Yang, ; Wenjie Long, ; Zemin Wan,
| |
Collapse
|
14
|
Peng Y, Yang Q, Gao S, Liu Z, Kong W, Bian X, Li Z, Ye J. IL-6 protects cardiomyocytes from oxidative stress at the early stage of LPS-induced sepsis. Biochem Biophys Res Commun 2022; 603:144-152. [DOI: 10.1016/j.bbrc.2022.03.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 03/01/2022] [Indexed: 12/24/2022]
|
15
|
Fang F, Zhang X, Li B, Gan S. miR-182-5p combined with brain-derived neurotrophic factor assists the diagnosis of chronic heart failure and predicts a poor prognosis. J Cardiothorac Surg 2022; 17:88. [PMID: 35501813 PMCID: PMC9063236 DOI: 10.1186/s13019-022-01802-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 03/21/2022] [Indexed: 11/10/2022] Open
Abstract
Objective Chronic heart failure (CHF) is a general progressive disorder with high morbidity and poor prognosis. This study analyzed the serum expression and clinical value of miR-182-5p and brain-derived neurotrophic factor (BDNF) in CHF patients. Methods A total of 82 CHF patients were selected as the study subjects (15 cases in NYHA stage I, 29 cases in stage II, 27 cases in stage III, and 11 cases in stage IV), with another 78 healthy people as the controls. The expression of serum miR-182-5p was detected by RT-qPCR. BDNF expression was measured by ELISA. Furthermore, the Pearson coefficient was used to analyze the correlation of miR-182-5p/BDNF with BNP and LVEF. ROC curve was employed to assess the potential of miR-182-5p or/and BDNF for the diagnosis of CHF. Kaplan–Meier survival curve was implemented to evaluate the prognostic value of miR-182-5p and BDNF. Results Serum miR-182-5p level was elevated and BDNF expression was lowered in CHF patients. Serum miR-182-5p in CHF patients was positively-related with BNP and inversely-correlated with LVEF, while serum BDNF was negatively-linked with BNP and positively-correlated with LVEF. ROC curve indicated the diagnostic value of serum miR-182-5p and BDNF for CHF and the diagnostic accuracy of miR-182-5p combined with BDNF was improved. Kaplan–Meier analysis unveiled that miR-182-5p low expression and BDNF high expression could predict the overall survival in CHF patients. Conclusion miR-182-5p expression is increased and BDNF level is decreased in CHF patients. miR-182-5p combined with BDNF can assist the diagnosis of CHF and predict a poor prognosis.
Collapse
Affiliation(s)
- Fang Fang
- Department of Cardiovascular Medicine, Xianning Central Hospital, No. 228 Jingui Road, Xian'an District, Xianning City, 437000, Hubei Province, China.
| | - Xiaonan Zhang
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Shenyang Medical College, Xianning, 110000, Liaoning Province, China
| | - Bin Li
- Department of Cardiovascular Medicine, Xianning Central Hospital, No. 228 Jingui Road, Xian'an District, Xianning City, 437000, Hubei Province, China
| | - Shouyi Gan
- Department of Cardiovascular Medicine, Xianning Central Hospital, No. 228 Jingui Road, Xian'an District, Xianning City, 437000, Hubei Province, China
| |
Collapse
|
16
|
Santovito D, Weber C. Non-canonical features of microRNAs: paradigms emerging from cardiovascular disease. Nat Rev Cardiol 2022; 19:620-638. [PMID: 35304600 DOI: 10.1038/s41569-022-00680-2] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/14/2022] [Indexed: 02/08/2023]
Abstract
Research showing that microRNAs (miRNAs) are versatile regulators of gene expression has instigated tremendous interest in cardiovascular research. The overwhelming majority of studies are predicated on the dogmatic notion that miRNAs regulate the expression of specific target mRNAs by inhibiting mRNA translation or promoting mRNA decay in the RNA-induced silencing complex (RISC). These efforts mostly identified and dissected contributions of multiple regulatory networks of miRNA-target mRNAs to cardiovascular pathogenesis. However, evidence from studies in the past decade indicates that miRNAs also operate beyond this canonical paradigm, featuring non-conventional regulatory functions and cellular localizations that have a pathophysiological role in cardiovascular disease. In this Review, we highlight the functional relevance of atypical miRNA biogenesis and localization as well as RISC heterogeneity. Moreover, we delineate remarkable non-canonical examples of miRNA functionality, including direct interactions with proteins beyond the Argonaute family and their role in transcriptional regulation in the nucleus and in mitochondria. We scrutinize the relevance of non-conventional biogenesis and non-canonical functions of miRNAs in cardiovascular homeostasis and pathology, and contextualize how uncovering these non-conventional properties can expand the scope of translational research in the cardiovascular field and beyond.
Collapse
Affiliation(s)
- Donato Santovito
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität (LMU), Munich, Germany. .,German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany. .,Institute for Genetic and Biomedical Research (IRGB), Unit of Milan, National Research Council, Milan, Italy.
| | - Christian Weber
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität (LMU), Munich, Germany. .,German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany. .,Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, Netherlands. .,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
| |
Collapse
|
17
|
Peng H, Zhang S, Zhang Z, Wang X, Tian X, Zhang L, Du J, Huang Y, Jin H. Nitric oxide inhibits endothelial cell apoptosis by inhibiting cysteine-dependent SOD1 monomerization. FEBS Open Bio 2022; 12:538-548. [PMID: 34986524 PMCID: PMC8804620 DOI: 10.1002/2211-5463.13362] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 11/07/2021] [Accepted: 01/04/2022] [Indexed: 11/23/2022] Open
Abstract
Endothelial cell apoptosis is an important pathophysiology in many cardiovascular diseases. The gasotransmitter nitric oxide (NO) is known to regulate cell survival and apoptosis. However, the mechanism underlying the effect of NO remains unclear. In this research, by targeting cytosolic copper/zinc superoxide dismutase (SOD1) monomerization, we aimed to explore how NO inhibited endothelial cell apoptosis. We showed that treatment with the NO synthase (NOS) inhibitor nomega‐nitro‐l‐arginine methyl ester hydrochloride (L‐NAME) significantly decreased the endogenous NO content of endothelial cells, facilitated the formation of SOD1 monomers, inhibited dismutase activity, and promoted reactive oxygen species (ROS) accumulation in human umbilical vein endothelial cells (HUVECs); by contrast, supplementation with the NO donor sodium nitroprusside (SNP) upregulated NO content, prevented the formation of SOD1 monomers, enhanced dismutase activity, and reduced ROS accumulation in L‐NAME‐treated HUVECs. Mechanistically, tris(2‐carboxyethyl) phosphine hydrochloride (TCEP), a specific reducer of cysteine thiol, increased SOD1 monomer formation, thus preventing the NO‐induced increase in dismutase activity and the decrease in ROS. Furthermore, SNP inhibited HUVEC apoptosis caused by the decrease in endogenous NO, whereas TCEP abolished this protective effect of SNP. In summary, our data reveal that NO protects endothelial cells against apoptosis by inhibiting cysteine‐dependent SOD1 monomerization to enhance SOD1 activity and inhibit oxidative stress.
Collapse
Affiliation(s)
- Hanlin Peng
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Shangyue Zhang
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Zaifeng Zhang
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Xiuli Wang
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Xiaoyu Tian
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Lulu Zhang
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Junbao Du
- Department of Pediatrics, Peking University First Hospital, Beijing, China.,Key Laboratory of Molecular Cardiology, Ministry of Education, Beijing, China
| | - Yaqian Huang
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Hongfang Jin
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| |
Collapse
|
18
|
Zhu J, Du S, Zhang J, Huang G, Dong L, Ren E, Liu D. microRNA-10a-5p from gastric cancer cell-derived exosomes enhances viability and migration of human umbilical vein endothelial cells by targeting zinc finger MYND-type containing 11. Bioengineered 2022; 13:496-507. [PMID: 34969361 PMCID: PMC8805907 DOI: 10.1080/21655979.2021.2009962] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 11/18/2021] [Indexed: 12/20/2022] Open
Abstract
Tumor-derived exosomes (exo) could modulate the biological behaviors of human umbilical vein endothelial cells (HUVECs). Here, the role of microRNA (miR)-10a-5p-modified gastric cancer (GC) cells-derived exo for HUVECs was studied. GC tissue specimens were collected, and miR-10a-5p and zinc finger MYND-type containing 11 (ZMYND11) levels were determined. HUVECs interfered with ZMYND11 or miR-10a-5p-related oligonucleotides. Exo was extracted from GC cells (HGC-27 exo), and miR-10a-5p mimic-modified HGC-27 exo were co-cultured with HUVECs. HUVECs viability, migration and angiogenesis were evaluated, and miR-10a-5p/ZMYND11 crosstalk was explored. It was observed that GC patients had raised miR-10a-5p and reduced ZMYND11, and miR-10a-5p negatively mediated ZMYND11 expression. Suppression of miR-10a-5p or overexpression of ZMYND11 inhibited viability, migration and tube formation ability of HUVECs. Notably, miR-10a-5p mimic-modified HGC-27 exo enhanced the viability, migration and tube formation ability of HUVECs, but this effect was impaired after up-regulating ZMYND11. In summary, miR-10a-5p from GC cells-derived exo enhances viability and migration of HUVECs by suppressing ZMYND11.
Collapse
Affiliation(s)
- Jiaxin Zhu
- Department of Gastrointestinal Surgery, the First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan, China
| | - Shasha Du
- Department of Nephrology, the First Affiliated Hospital, And College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan, China
| | - Jianfeng Zhang
- Department of Gastrointestinal Surgery, the First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan, China
| | - Guangzhao Huang
- Department of Emergency Medicine, the First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan, China
| | - Lujia Dong
- Department of Gastrointestinal Surgery, the First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan, China
| | - Enbo Ren
- Department of Gastrointestinal Surgery, the First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan, China
| | - Dechun Liu
- Department of Gastrointestinal Surgery, the First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan, China
| |
Collapse
|
19
|
Xiang C, Zhang F, Gao J, Guo F, Zhang M, Zhou R, Wei J, Wang P, Zhang Y, Zhang J, Yang H. Yixin-Shu Capsules Ameliorated Ischemia-Induced Heart Failure by Restoring Trx2 and Inhibiting JNK/p38 Activation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:8049079. [PMID: 33643519 PMCID: PMC7902134 DOI: 10.1155/2021/8049079] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 12/31/2020] [Accepted: 01/20/2021] [Indexed: 12/16/2022]
Abstract
Traditional Chinese medicine has shown great safety and efficacy in the treatment of heart failure (HF), whereas the mechanism remains unclear. In this study, the protective effect of Yixin-shu (YXS) capsules, a conventional medicine for various cardiovascular diseases, against myocardial ischemia-induced HF in rats was systematically investigated by RNA-seq technology. HF rats treated with YXS (0.8 or 1.6 g/kg/d, ig) for 6 weeks had significantly decreased brain natriuretic peptide (BNP) and atrial natriuretic peptide (ANP) and collagen III and attenuated cardiac structure rupture and collagen deposition. Additionally, YXS treatment decreased the levels of interleukin-1β (IL-1β), interleukin 6 (IL-6), tumor necrosis factor-α (TNF-α), and lactate dehydrogenase (LDH) and TUNEL-positive rate and the nitrotyrosine staining, but increased levels of glutathione (GSH), total antioxidant capacity (T-AOC) activity, and mitochondrial membrane potential. Further experiments demonstrated that YXS restored Trx2 and inhibited the phosphorylation of JNK and p38, thereby improving cardiac function in the rats with HF. Silencing Trx2 decreased the protection of YXS in the response to H2O2 as evidenced by the increase of caspase-3 activity and decrease of GSH level. Thus, YXS enhanced heart function and decreased myocardial damage through restoring Trx2 and inhibiting JNK and p38 activation in ischemia-induced HF.
Collapse
Affiliation(s)
- Changpei Xiang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Fangbo Zhang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Jinhuan Gao
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Feifei Guo
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Mao Zhang
- Institute of Molecular Medicine, Peking University, Beijing 100871, China
| | - Rui Zhou
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Junying Wei
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Ping Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Yi Zhang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Jingjing Zhang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Hongjun Yang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| |
Collapse
|
20
|
Chen C, Zong M, Lu Y, Guo Y, Lv H, Xie L, Fu Z, Cheng Y, Si Y, Ye B, Fan L. Differentially expressed lnc-NOS2P3-miR-939-5p axis in chronic heart failure inhibits myocardial and endothelial cells apoptosis via iNOS/TNFα pathway. J Cell Mol Med 2020; 24:11381-11396. [PMID: 32844595 PMCID: PMC7576245 DOI: 10.1111/jcmm.15740] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 07/06/2020] [Accepted: 07/30/2020] [Indexed: 12/11/2022] Open
Abstract
Inflammatory cytokine‐induced cell apoptosis is important for initiation and progression of chronic heart failure (CHF). Non‐coding RNAs, including long non‐coding RNAs and microRNAs, have emerged as critical regulators of this pathological process. The role in regulating inflammation and induction to cell apoptosis in CHF is not well understood. This study found CHF patients had elevated serum miR‐939‐5p, with greater increase in New York Heart Association (NYHA) I‐II patients than in NYHA III‐IV. Moreover, miR‐939‐5p was positively correlated with B‐type natriuretic peptide (BNP) in NYHA III‐IV patients, while not in NYHA I‐II. Further study showed miR‐939‐5p mimics promoted cell proliferation and inhibited inflammatory cytokine‐induced apoptosis of HUVECs and H9C2, while inhibition of endogenous miR‐939‐5p produced the opposite effects. Induced nitric oxide synthase (iNOS) and tumour necrosis factor α (TNFα) were identified as target genes of miR‐939‐5p. Additionally, lncRNA‐NOS2P3 acted as an endogenous sponge RNA to inhibit miR‐939‐5p expression, regulate the expression of iNOS/TNFα and control inflammation‐induced cells apoptosis. These suggest that CHF patients exhibited elevated serum miR‐939‐5p level especially in NYHA I‐II grades. And lnc‐NOS2P3‐miR‐939‐5p‐iNOS/TNFα pathway regulated inflammatory cytokine‐induced endothelial and myocardial cells apoptosis and provided a promising strategy for diagnosis and treatment of CHF.
Collapse
Affiliation(s)
- Cuncun Chen
- Department of Clinical Laboratory, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ming Zong
- Department of Clinical Laboratory, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ying Lu
- Department of Clinical Laboratory, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yide Guo
- Department of Clinical Laboratory, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Honggen Lv
- Department of Clinical Laboratory, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Lihong Xie
- Department of Clinical Laboratory, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zhiyan Fu
- Department of Clinical Laboratory, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yu Cheng
- Department of Clinical Laboratory, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yuying Si
- Department of Clinical Laboratory, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Bei Ye
- Department of Clinical Laboratory, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Lieying Fan
- Department of Clinical Laboratory, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|