1
|
Xing Y, Lin X. Challenges and advances in the management of inflammation in atherosclerosis. J Adv Res 2025; 71:317-335. [PMID: 38909884 DOI: 10.1016/j.jare.2024.06.016] [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: 03/07/2024] [Revised: 06/14/2024] [Accepted: 06/15/2024] [Indexed: 06/25/2024] Open
Abstract
INTRODUCTION Atherosclerosis, traditionally considered a lipid-related disease, is now understood as a chronic inflammatory condition with significant global health implications. OBJECTIVES This review aims to delve into the complex interactions among immune cells, cytokines, and the inflammatory cascade in atherosclerosis, shedding light on how these elements influence both the initiation and progression of the disease. METHODS This review draws on recent clinical research to elucidate the roles of key immune cells, macrophages, T cells, endothelial cells, and clonal hematopoiesis in atherosclerosis development. It focuses on how these cells and process contribute to disease initiation and progression, particularly through inflammation-driven processes that lead to plaque formation and stabilization. Macrophages ingest oxidized low-density lipoprotein (oxLDL), which partially converts to high-density lipoprotein (HDL) or accumulates as lipid droplets, forming foam cells crucial for plaque stability. Additionally, macrophages exhibit diverse phenotypes within plaques, with pro-inflammatory types predominating and others specializing in debris clearance at rupture sites. The involvement of CD4+ T and CD8+ T cells in these processes promotes inflammatory macrophage states, suppresses vascular smooth muscle cell proliferation, and enhances plaque instability. RESULTS The nuanced roles of macrophages, T cells, and the related immune cells within the atherosclerotic microenvironment are explored, revealing insights into the cellular and molecular pathways that fuel inflammation. This review also addresses recent advancements in imaging and biomarker technology that enhance our understanding of disease progression. Moreover, it points out the limitations of current treatment and highlights the potential of emerging anti-inflammatory strategies, including clinical trials for agents such as p38MAPK, tumor necrosis factor α (TNF-α), and IL-1β, their preliminary outcomes, and the promising effects of canakinumab, colchicine, and IL-6R antagonists. CONCLUSION This review explores cutting-edge anti-inflammatory interventions, their potential efficacy in preventing and alleviating atherosclerosis, and the role of nanotechnology in delivering drugs more effectively and safely.
Collapse
Affiliation(s)
- Yiming Xing
- Cardiology Department, The First Affiliated Hospital of Anhui Medical University, Hefei City, Anhui Province, 230022, China
| | - Xianhe Lin
- Cardiology Department, The First Affiliated Hospital of Anhui Medical University, Hefei City, Anhui Province, 230022, China.
| |
Collapse
|
2
|
Stopa V, Dafou D, Karagianni K, Nossent AY, Farrugia R, Devaux Y, Sopic M. Epitranscriptomics in atherosclerosis: Unraveling RNA modifications, editing and splicing and their implications in vascular disease. Vascul Pharmacol 2025; 159:107496. [PMID: 40239855 DOI: 10.1016/j.vph.2025.107496] [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: 01/31/2025] [Revised: 04/08/2025] [Accepted: 04/12/2025] [Indexed: 04/18/2025]
Abstract
Atherosclerosis remains a leading cause of morbidity and mortality worldwide, driven by complex molecular mechanisms involving gene regulation and post-transcriptional processes. Emerging evidence highlights the critical role of epitranscriptomics, the study of chemical modifications occurring on RNA molecules, in atherosclerosis development. Epitranscriptomics provides a new layer of regulation in vascular health, influencing cellular functions in endothelial cells, smooth muscle cells, and macrophages, thereby shedding light on the pathogenesis of atherosclerosis and presenting new opportunities for novel therapeutic targets. This review provides a comprehensive overview of the epitranscriptomic landscape, focusing on key RNA modifications such as N6-methyladenosine (m6A), 5-methylcytosine (m5C), pseudouridine (Ψ), RNA editing mechanisms including A-to-I and C-to-U editing and RNA isoforms. The functional implications of these modifications in RNA stability, alternative splicing, and microRNA biology are discussed, with a focus on their roles in inflammatory signaling, lipid metabolism, and vascular cell adaptation within atherosclerotic plaques. We also highlight how these modifications influence the generation of RNA isoforms, potentially altering cellular phenotypes and contributing to disease progression. Despite the promise of epitranscriptomics, significant challenges remain, including the technical limitations in detecting RNA modifications in complex tissues and the need for deeper mechanistic insights into their causal roles in atherosclerotic pathogenesis. Integrating epitranscriptomics with other omics approaches, such as genomics, proteomics, and metabolomics, holds the potential to provide a more holistic understanding of the disease.
Collapse
Affiliation(s)
- Victoria Stopa
- Cardiovascular Research Unit, Department of Precision Health, Luxembourg Institute of Health, Strassen, Luxembourg
| | - Dimitra Dafou
- Department of Genetics, Development and Molecular Biology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Korina Karagianni
- Department of Genetics, Development and Molecular Biology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - A Yaël Nossent
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Denmark
| | - Rosienne Farrugia
- Department of Applied Biomedical Science, Faculty of Health Sciences, University of Malta, Msida, Malta
| | - Yvan Devaux
- Cardiovascular Research Unit, Department of Precision Health, Luxembourg Institute of Health, Strassen, Luxembourg.
| | - Miron Sopic
- Cardiovascular Research Unit, Department of Precision Health, Luxembourg Institute of Health, Strassen, Luxembourg; Department of Medical Biochemistry, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
3
|
Wu M, Nie Q, Zhang Y, Qin J, Ye L, Zhao R, Dai M, Wu M. METTL3 Plays Regulatory Roles in Acute Pneumonia during Staphylococcus aureus Infection. ACS Infect Dis 2025; 11:905-916. [PMID: 40105125 DOI: 10.1021/acsinfecdis.4c00938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
Pneumonia caused by Staphylococcus aureus infection has consistently been a significant cause of morbidity and mortality worldwide. Extensive research to date indicates that N6-methyladenosine (m6A) modification plays a crucial role in the development and progression of various diseases. However, it remains unknown whether the m6A modification affects the progression of bacterial pneumonia. To explore this question, we assessed the levels of m6A as well as the expression of methyltransferases (METTL3 and METTL14), demethylase fat mass and obesity-related protein (FTO), and methylation reader proteins YTHDF1 and YTHDF2 in mice and MH-S cells during S. aureus infection. The levels of m6A and METTL3 were significantly upregulated in S. aureus-infected mice and MH-S cells. siMETTL3 knockdown resulted in more severe bacterial colonization, lung damage, increased inflammatory cytokines (IL-6, IL-1β, TNF-α), and mortality rates in mice as well as MH-S cells following the bacterial infection. Regulation of lung inflammation levels by METTL3 was associated with the activation of the MAPK/NF-κB/JAK2-STAT3 signaling pathway. Moreover, siMETTL3 mice exhibited an increased release of superoxides and exacerbated oxidative stress in the lungs following S. aureus infection, which was correlated with impaired mitochondrial autophagy mediated by the Pink1/Parkin pathway. Our findings provide previously unrecognized evidence of the protective role of METTL3 in S. aureus-induced acute pneumonia, indicating a potential therapeutic target for S. aureus infections.
Collapse
Affiliation(s)
- Menghui Wu
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan 430070, China
| | - Qihang Nie
- Key Laboratory of Resources Biology and Biotechnology in Western China, Ministry of Education, College of Life Science, Northwest University, Xi'an 710000, China
| | - Yanyan Zhang
- School of Medicine, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Jiaoxia Qin
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Liumei Ye
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Ruoyang Zhao
- Wenzhou Institute, University of the Chinese Academy of Sciences, Wenzhou 325024, China
| | - Menghong Dai
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan 430070, China
| | - Min Wu
- Wenzhou Institute, University of the Chinese Academy of Sciences, Wenzhou 325024, China
| |
Collapse
|
4
|
Diao MN, Lv YJ, Xin H, Zhang YF, Zhang R. A comprehensive review of m6 A methylation in coronary heart disease. J Mol Med (Berl) 2025:10.1007/s00109-025-02540-1. [PMID: 40208302 DOI: 10.1007/s00109-025-02540-1] [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: 06/06/2024] [Revised: 03/31/2025] [Accepted: 04/01/2025] [Indexed: 04/11/2025]
Abstract
The morbidity and mortality rates of coronary heart disease (CHD) are high worldwide. The primary pathological changes in CHD involve stenosis and ischemia caused by coronary atherosclerosis (AS). Extensive research on the pathogenesis of AS has revealed chronic immunoinflammatory processes and cell proliferation in all layers of coronary vessels, including endothelial cells (ECs), vascular smooth muscle cells, and macrophages. m6 A methylation is a common posttranscriptional modification of RNA that is coordinated by a variety of regulators (writers, readers, erasers) to maintain the functional stability of modified mRNAs and ncRNAs. In recent years, there has been increasing focus on the involvement of m6 A methylation in the incidence and progression of CHD, which starts with atherosclerotic plaque formation, leads to myocardial ischemia, and ultimately results in the occurrence of myocardial infarction (MI). m6 A regulators modulate relevant signaling pathways to participate in the inflammatory response, programmed death of cardiomyocytes, and fibrosis. Therefore, diagnostic models based on m6 A profiling are helpful for the early detection of CHD, and m6 A methylation shows promise as a sensitive target for new drugs to treat CHD in the future.
Collapse
Affiliation(s)
- Mei-Ning Diao
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266000, Shandong, P. R. China
- Institute for Translational Medicine, College of Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266021, China
| | - Yi-Jv Lv
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266000, Shandong, P. R. China
- Institute for Translational Medicine, College of Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266021, China
| | - Hui Xin
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266000, Shandong, P. R. China
| | - Yin-Feng Zhang
- Institute for Translational Medicine, College of Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266021, China.
| | - Rui Zhang
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266000, Shandong, P. R. China.
| |
Collapse
|
5
|
Luo X, He C, Yang B, Yin S, Li K. WTAP Promotes Atherosclerosis by Inducing Macrophage Pyroptosis and M1 Polarization through Upregulating NLRP3. Appl Biochem Biotechnol 2025; 197:2397-2416. [PMID: 39747738 DOI: 10.1007/s12010-024-05106-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] [Accepted: 11/19/2024] [Indexed: 01/04/2025]
Abstract
The study was designed to investigate the impact of N6-methyladenosine (m6A) writer Wilms tumor 1-associated protein (WTAP) on the progression of atherosclerosis (AS) and to further elucidate its possible regulatory mechanism. The m6A levels and WTAP expressions were initially assessed through RIP, qRT-PCR, and western blotting. An in vitro model of AS was constructed by ox-LDL treatment in RAW264.7 cells. Next, the impact of WTAP on macrophage pyroptosis and M1 polarization was evaluated. The relationship between WTAP and NLRP3 was then investigated using m6A modification quantification and RIP-qPCR assay. To investigate the effect of WTAP on AS development in vivo, we created an ApoE-/-mouse model of AS by feeding high-fat diet (HFD). Furthermore, the influence of WTAP on macrophage pyroptosis and M1 polarization through NLRP3 was explored by NLRP3 overexpression AAV injection. Here, we found that WTAP was significantly upregulated in peripheral blood mononuclear cells (PBMCs) from AS patients, accompanied by increased total m6A methylation levels. The silencing of WTAP suppressed macrophage pyroptosis and M1 polarization induced by ox-LDL and also ameliorated aortic root lesion damage in AS mice. Mechanistically, m6A modification mediated by WTAP enhanced NLRP3 mRNA stabilization, thereby upregulating NLRP3 expression. Overexpression of NLRP3 was found to enhance macrophage pyroptosis and M1 polarization, contributing to the progression of AS. In conclusion, our findings suggest that WTAP knockdown mitigated AS progression by modulating NLRP3 in an m6A-dependent manner. Our study proposes that targeting WTAP could be a potential preventive and therapeutic strategy for AS patients.
Collapse
Affiliation(s)
- Xing Luo
- Department of Neurology, The Third Hospital of Changsha, Changsha, Hunan, China
| | - Chaogui He
- Department of Vascular Surgery, The Affiliated Changsha Hospital of Xiangya School of Medicine, Central South University (The First Hospital of Changsha), Changsha, Hunan, China
| | - Bo Yang
- Department of Vascular Surgery, The Affiliated Changsha Hospital of Xiangya School of Medicine, Central South University (The First Hospital of Changsha), Changsha, Hunan, China
| | - Shuheng Yin
- University of South China, Hengyang, Hunan, China
| | - Ke Li
- University of South China, Hengyang, Hunan, China.
| |
Collapse
|
6
|
Liu Q, Yan L, Wu T, Wu Q, Ke B, Shen W. Peli1, regulated by m 6A modification, suppresses NLRP3 inflammasome activation in atherosclerosis by inhibiting YB-1. Commun Biol 2025; 8:457. [PMID: 40102597 PMCID: PMC11920095 DOI: 10.1038/s42003-025-07839-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 02/27/2025] [Indexed: 03/20/2025] Open
Abstract
The activation of pyrin domain-containing-3 (NLRP3) inflammasome in macrophages is a risk factor accelerating the progression of atherosclerosis (AS). Here, the function of pellino 1 (Peli1) in regulating the activation of NLRP3 inflammasome during the development of AS was investigated. Our results showed that Y-box binding protein 1 (YB-1) knockdown could inhibit the progression of AS in vivo, and YB-1 silencing repressed oxidized low-density lipoprotein (ox-LDL)-mediated lipid accumulation and inflammation in macrophages by inactivating NLRP3 inflammasome. E3 ubiquitination ligase Peli1 mediated ubiquitination-dependent degradation of YB-1 during AS progression. Moreover, it was found that YTH domain-containing 2 (YTHDC2) recognized methyltransferase-like 3 (METTL3)-mediated Peli1 N6-methyladenosine (m6A) modification and mediated Peli1 mRNA degradation. Rescue studies revealed that YB-1 upregulation abrogated the repressive effect of Peli1 upregulation on AS progression both in vitro and in vivo. Taken together, Peli1, regulated by m6A modification, inhibited YB-1-mediated activation of NLRP3 inflammasome in macrophages by promoting YB-1 ubiquitination to suppress the progression of AS.
Collapse
Affiliation(s)
- Qiang Liu
- Department of Cardiovascular Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Department of Cardiovascular Medicine, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Lu Yan
- Department of Cardiovascular Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Tao Wu
- Department of Cardiovascular Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Qinghua Wu
- Department of Cardiovascular Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Ben Ke
- Department of Nephrology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China.
| | - Wen Shen
- Department of Cardiovascular Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China.
| |
Collapse
|
7
|
Chen J, Chen G, Li J, Wang D, Liang W, Zhao S. NLRC5 in Macrophages Promotes Atherosclerosis in Acute Coronary Syndrome by Regulating STAT3 Expression. Cardiovasc Toxicol 2025; 25:365-378. [PMID: 39833596 DOI: 10.1007/s12012-024-09957-z] [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: 12/18/2023] [Accepted: 12/30/2024] [Indexed: 01/22/2025]
Abstract
The mortality rate of cardiovascular and cerebrovascular diseases ranks first among all causes. This study elucidated the role and potential mechanism of the NLRC5 gene in atherosclerosis (AS). We enrolled patients (number = 30) diagnosed with AS and healthy volunteers (number = 30) as controls from our hospital. In patients with AS, the levels of serum NLRC5 were up-regulated (Fig. 1A) and positively correlated with CIMT/CRP. In a mouse model of AS, the expression of serum NLRC5 mRNA was increased at 6 or 12 weeks after inducing AS. The expression of NLRC5 protein was found to be elevated in a mouse model of AS. The inhibition of NLRC5 reduced development of AS in ApoE-/- Mice. Reducing NLRC5 inhibited the polarization of M2 macrophages and shifted macrophages towards proinflammatory M1 phenotype. STAT3 was identified as a target of NLRC5, with NLRC5 protein expression shown to reduce STAT3 ubiquitination. Methylation promoted NLRC5 DNA stability in vitro model of AS. Sh-NLRC5 increased M1/M2 macrophage ratio, foam cell formation and ox-LDL uptake. STAT3 reduced the effects of sh-NLRC5-mediated M1/M2 macrophage ratio in model of AS. These data confirmed that NLRC5 in macrophages promotes atherosclerosis in acute coronary syndrome by regulating STAT3 expression. This suggests that NLRC5 could be a potential target for the treatment of premature AS.
Collapse
Affiliation(s)
- Jun Chen
- Department of Cardiovascular Medicine, The Affiliated Panyu Central Hospital of Guangzhou Medical University (Cardiovascular Diseases Research Institute of Panyu District), No. 8, Fuyu East Road, Qiaonan Street, Panyu District, Guangzhou, 511400, China.
| | - Guoqin Chen
- Department of Cardiovascular Medicine, The Affiliated Panyu Central Hospital of Guangzhou Medical University (Cardiovascular Diseases Research Institute of Panyu District), No. 8, Fuyu East Road, Qiaonan Street, Panyu District, Guangzhou, 511400, China
| | - Jianhao Li
- Department of Cardiovascular Medicine, The Affiliated Panyu Central Hospital of Guangzhou Medical University (Cardiovascular Diseases Research Institute of Panyu District), No. 8, Fuyu East Road, Qiaonan Street, Panyu District, Guangzhou, 511400, China
| | - Dayu Wang
- Department of Cardiovascular Medicine, The Affiliated Panyu Central Hospital of Guangzhou Medical University (Cardiovascular Diseases Research Institute of Panyu District), No. 8, Fuyu East Road, Qiaonan Street, Panyu District, Guangzhou, 511400, China
| | - Weijie Liang
- Department of Cardiovascular Medicine, The Affiliated Panyu Central Hospital of Guangzhou Medical University (Cardiovascular Diseases Research Institute of Panyu District), No. 8, Fuyu East Road, Qiaonan Street, Panyu District, Guangzhou, 511400, China
| | - Shanjun Zhao
- Department of Cardiovascular Medicine, The Affiliated Panyu Central Hospital of Guangzhou Medical University (Cardiovascular Diseases Research Institute of Panyu District), No. 8, Fuyu East Road, Qiaonan Street, Panyu District, Guangzhou, 511400, China
| |
Collapse
|
8
|
Tao M, Shan L, Zhang W, Wei L, Guo M, Fang Z, Zhao J, Gao J. METTL3-mediated m6A modification of EGR1 mRNA promotes T2DM vasculopathy. Cell Signal 2025; 127:111564. [PMID: 39674391 DOI: 10.1016/j.cellsig.2024.111564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 11/11/2024] [Accepted: 12/09/2024] [Indexed: 12/16/2024]
Abstract
Vascular endothelial dysfunction is one of the leading causes of developing vascular lesions in Type 2 diabetes mellitus (T2DM). In the development of vascular lesions, when endothelial cells are stimulated by hyperglycemia, inflammation and other external conditions, endothelial cell dysfunction will occur, which promotes endothelial cells to lose its typical phenotype and gain mesenchymal characteristics, with the occurrence of endothelial-to-mesenchymal transition (EndMT). At the same time promote endothelial cell proliferation and migration, induce vascular injury. m6A methylation modification enzyme METTL3 is involved in the development of vascular lesions in T2DM. However, the mechanisms by which METTL3 is involved in T2DM vascular lesions are unclear. In this study, we induced T2DM vascular lesions in human umbilical vein endothelial cells (HUVECs) mimicking high glucose and TNF-α (H + T) levels. The effects of METTL3 on HUVECs EndMT, proliferation and migration have been revealed. Protein expression of endothelial calmodulin (VE-Cadherin) and smooth muscle actin (α-SMA) was visualised by western blot and immunofluorescence techniques to evaluate the occurrence of EndMT. In addition, MeRIP-seq revealed a METTL3-mediated m6A modification profile. MeRIP-qPCR combined with m6A site prediction verified the methylation levels of downstream targets and identified EGR1 as a target of METTL3. Activation of EGR1 successfully rescued EndMT in METTL3-deficient HUVECs. In summary, targeting METTL3 may become an important molecular target for intervention in diabetic vascular lesions.
Collapse
Affiliation(s)
- Meng Tao
- Department of Pharmacy, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230031, Anhui, China; College of Pharmacy, Anhui University of Chinese Medicine, Hefei 230011, Anhui, China
| | - Li Shan
- Department of Pharmacy, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230031, Anhui, China
| | - Wei Zhang
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei 230011, Anhui, China
| | - LiangBing Wei
- Department of Pharmacy, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230031, Anhui, China
| | - MingFei Guo
- Department of Scientific Research, The First Affiliated Hospital of Anhui Medical University, Hefei 230012, Anhui, China; Anhui Public Health Clinical Center, Hefei 230012, Anhui, China
| | - ZhaoHui Fang
- Department of Pharmacy, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230031, Anhui, China; Diabetes Prevention Institute of Traditional Chinese Medicine, Anhui Academy of Traditional Chinese Medicine, Hefei 230031, Anhui, China
| | - JinDong Zhao
- Department of Pharmacy, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230031, Anhui, China; Diabetes Prevention Institute of Traditional Chinese Medicine, Anhui Academy of Traditional Chinese Medicine, Hefei 230031, Anhui, China
| | - JiaRong Gao
- Department of Pharmacy, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230031, Anhui, China.
| |
Collapse
|
9
|
Wang Y, Ling S, Feng H, Hua J, Han Z, Chai R. Recent Advances in the Mutual Regulation of m6A Modification and Non-Coding RNAs in Atherosclerosis. Int J Gen Med 2025; 18:1047-1073. [PMID: 40026815 PMCID: PMC11871936 DOI: 10.2147/ijgm.s508197] [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: 11/27/2024] [Accepted: 02/20/2025] [Indexed: 03/05/2025] Open
Abstract
Atherosclerosis, a progressive inflammatory disease of the arteries, remains a leading cause of cardiovascular morbidity and mortality worldwide. Recent years have witnessed the pivotal role of N6-methyladenosine (m6A) RNA methylation in regulating various biological processes, including those implicated in atherosclerosis. Current evidence suggested that m6A regulators (writers, erasers, and readers) participated in the modification of multiple non-coding RNAs (ncRNAs), including microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs), thereby affecting their metabolism and functions. Meanwhile, ncRNAs have also emerged as key modulator of m6A regulators expression in turn. Therefore, understanding the mutual regulation between m6A modifications and ncRNAs is of great significance to identify novel therapeutic targets for atherosclerosis and has great clinical application prospects. This review aims to summarize the recent advances in the reciprocal regulation and provide insights into the interaction between m6A modification and ncRNAs in the context of atherosclerosis.
Collapse
Affiliation(s)
- Yanlu Wang
- Department of Cardiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province, 510260, People’s Republic of China
| | - Sisi Ling
- Department of Cardiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province, 510260, People’s Republic of China
| | - Hao Feng
- Department of Cardiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province, 510260, People’s Republic of China
| | - Junkai Hua
- Department of Cardiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province, 510260, People’s Republic of China
| | - Zhiyu Han
- Department of Cardiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province, 510260, People’s Republic of China
| | - Renjie Chai
- Department of Cardiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province, 510260, People’s Republic of China
| |
Collapse
|
10
|
Li Z, Lao Y, Yan R, Li F, Guan X, Dong Z. N6-methyladenosine in inflammatory diseases: Important actors and regulatory targets. Gene 2025; 936:149125. [PMID: 39613051 DOI: 10.1016/j.gene.2024.149125] [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: 08/21/2024] [Revised: 11/17/2024] [Accepted: 11/25/2024] [Indexed: 12/01/2024]
Abstract
N6-methyladenosine (m6A) is one of the most prevalent epigenetic modifications in eukaryotic cells. It regulates RNA function and stability by modifying RNA methylation through writers, erasers, and readers. As a result, m6A plays a critical role in a wide range of biological processes. Inflammation is a common and fundamental pathological process. Numerous studies have investigated the role of m6A modifications in inflammatory diseases. This review highlights the mechanisms by which m6A contributes to inflammation, focusing on pathogen-induced infectious diseases, autoimmune disorders, allergic conditions, and metabolic disorder-related inflammatory diseases.
Collapse
Affiliation(s)
- Zewen Li
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China; Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, China
| | - Yongfeng Lao
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China; Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, China
| | - Rui Yan
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China; Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, China
| | - Fuhan Li
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China; Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, China
| | - Xin Guan
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China; Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, China
| | - Zhilong Dong
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China; Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, China.
| |
Collapse
|
11
|
Xu Y, Sha W, Lu J, Yu S, Jin X, Chen C, Ge G, Lei T. Danggui Liuhuang Decoction ameliorates endothelial dysfunction by inhibiting the JAK2/STAT3 mediated inflammation. JOURNAL OF ETHNOPHARMACOLOGY 2025; 340:119170. [PMID: 39615770 DOI: 10.1016/j.jep.2024.119170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 11/24/2024] [Accepted: 11/27/2024] [Indexed: 12/10/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Vascular endothelial dysfunction (VED) is recognized as a key triggering diabetic vascular complications. Danggui Liuhuang Decoction (DGLHD) has shown potential in mitigating these complications. However, the clinical efficacy of DGLHD in enhancing endothelial function, as well as the molecular mechanisms underlying its alleviation of Type 2 Diabetes-Related Vascular Endothelial Dysfunction (T2DM-VED), remains insufficiently understood. AIM OF THE STUDY This study aims to validate the therapeutic efficacy of DGLHD in ameliorating T2DM-VED through clinical research. Furthermore it seeks to analyze the pharmacodynamic basis and molecular mechanisms of DGLHD, elucidating the biological processes through which DGLHD alleviates VED in type 2 diabetes mellitus (T2DM). MATERIALS AND METHODS Patients diagnosed with "Yin deficiency with hyperactive fire syndrome", who are at a high risk for atherosclerotic cardiovascular disease (ASCVD) associated with T2DM, were recruited for this study. The effect of DGLHD on vascular endothelial function in T2DM was assessed by measuring the levels of pro-inflammatory factors through enzyme-linked immunosorbent assay (ELISA) and flow-mediated dilation (FMD). The primary components of DGLHD were analyzed using the UHPLC-Q-Exactive Orbitrap system. Potential therapeutic targets of DGLHD were predicted using network pharmacology and molecular docking analysis. To validate the mechanism of DGLHD on T2DM-VED, endothelial injury and inflammation cell models were established using human umbilical vein endothelial cells (HUVECs). A mouse model of diabetic endothelial injury was also developed to observe the effects of DGLHD on pro-inflammatory factors and vascular endothelial factors were observed through immunohistochemistry. Additionally, the effects on the janus kinase 2 (JAK2) / signal transducer and activator of transcription 3 (STAT3) signaling pathway were observed through Western blot experiments. RESULTS DGLHD was found to contain 201 active components. Network pharmacology analysis indicated that the treatment of T2DM-VED with DGLHD is associated with modulation of the JAK2/STAT3 signaling pathway. Molecular docking analysis demonstrated that small molecules in DGLHD interact with JAK2 and STAT3. Our clinical study demonstrated that DGLHD significantly reduces the levels of pro-inflammatory factors and improves FMD readings in diabetic patients, thereby alleviating T2DM-VED. DGLHD was shown to inhibit the phosphorylation of JAK2 and STAT3, which blocks the JAK2/STAT3 signaling pathway transmission, reducing the release of pro-inflammatory and vascular endothelial growth factors, and preventing the inflammatory response in vivo and in vitro. CONCLUSION This study demonstrates the potential efficacy of DGLHD in improving endothelial function in T2DM patients at high risk for ASCVD. By inhibiting the JAK2/STAT3 signaling pathway, DGLHD effectively reduces the release of pro-inflammatory factors and vascular endothelial growth factors, alleviating VED.
Collapse
Affiliation(s)
- Yuanying Xu
- Department of Endocrinology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200333, China.
| | - Wenjun Sha
- Department of Endocrinology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200333, China.
| | - Jun Lu
- Department of Endocrinology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200333, China.
| | - Shanshan Yu
- Department of Endocrinology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200333, China.
| | - Xinyan Jin
- Department of Endocrinology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200333, China.
| | - Cheng Chen
- Department of Endocrinology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200333, China.
| | - Guangbo Ge
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Tao Lei
- Department of Endocrinology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200333, China.
| |
Collapse
|
12
|
Zhang H, Lu W, Tang H, Chen A, Gao X, Zhu C, Zhang J. Novel Insight of N6-Methyladenosine in Cardiovascular System. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:222. [PMID: 40005339 PMCID: PMC11857502 DOI: 10.3390/medicina61020222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 01/19/2025] [Accepted: 01/24/2025] [Indexed: 02/27/2025]
Abstract
N6-methyladenosine (m6A) is the most common and abundant internal co-transcriptional modification in eukaryotic RNAs. This modification is catalyzed by m6A methyltransferases, known as "writers", including METTL3/14 and WTAP, and removed by demethylases, or "erasers", such as FTO and ALKBH5. It is recognized by m6A-binding proteins, or "readers", such as YTHDF1/2/3, YTHDC1/2, IGF2BP1/2/3, and HNRNPA2B1. Cardiovascular diseases (CVDs) are the leading cause of morbidity and mortality worldwide. Recent studies indicate that m6A RNA modification plays a critical role in both the physiological and pathological processes involved in the initiation and progression of CVDs. In this review, we will explore how m6A RNA methylation impacts both the normal and disease states of the cardiovascular system. Our focus will be on recent advancements in understanding the biological functions, molecular mechanisms, and regulatory factors of m6A RNA methylation, along with its downstream target genes in various CVDs, such as atherosclerosis, ischemic diseases, metabolic disorders, and heart failure. We propose that the m6A RNA methylation pathway holds promise as a potential therapeutic target in cardiovascular disease.
Collapse
Affiliation(s)
- Huan Zhang
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China; (H.Z.); (W.L.); (H.T.); (A.C.); (X.G.)
| | - Wei Lu
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China; (H.Z.); (W.L.); (H.T.); (A.C.); (X.G.)
| | - Haoyue Tang
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China; (H.Z.); (W.L.); (H.T.); (A.C.); (X.G.)
| | - Aiqun Chen
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China; (H.Z.); (W.L.); (H.T.); (A.C.); (X.G.)
| | - Xiaofei Gao
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China; (H.Z.); (W.L.); (H.T.); (A.C.); (X.G.)
| | - Congfei Zhu
- Department of Cardiology, Lianshui County People’s Hospital, Affiliated Hospital of Kangda College, Nanjing Medical University, Huaian 223400, China
| | - Junjie Zhang
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China; (H.Z.); (W.L.); (H.T.); (A.C.); (X.G.)
| |
Collapse
|
13
|
Zhang L, Thalakiriyawa DS, Liu J, Yang S, Wang Y, Dissanayaka WL. Semaphorin-4D signaling in recruiting dental stem cells for vascular stabilization. Stem Cell Res Ther 2025; 16:25. [PMID: 39865283 PMCID: PMC11770943 DOI: 10.1186/s13287-025-04149-0] [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: 10/26/2024] [Accepted: 01/14/2025] [Indexed: 01/28/2025] Open
Abstract
BACKGROUND Achieving a stable vasculature is crucial for tissue regeneration. Endothelial cells initiate vascular morphogenesis, followed by mural cells that stabilize new vessels. This study investigated the in vivo effects of Sema4D-Plexin-B1 signaling on stem cells from human exfoliated deciduous teeth (SHED)-supported angiogenesis, focusing on its mechanism in PDGF-BB secretion. We also explored macrophages as an endogenous source of Sema4D for vascular stabilization. METHODS The in vivo Matrigel plug angiogenesis assay was conducted to examine the impact of Sema4D on vessel formation and stabilization supported by SHED. Knockdown of Plexin-B1 in human umbilical vein endothelial cells (HUVECs) and PDGFR-β inhibitors were utilized to explore the fundamental regulatory mechanisms. Furthermore, the m6A methylation levels of total RNA and the expression of Methyltransferase-like 3 (METTL3) were assessed under conditions of Sema4D treatment in vitro. An ELISA was employed to measure the levels of Sema4D in the supernatants derived from THP-1 cell-mediated macrophages. Additionally, a three-dimensional vasculature-on-a-chip microfluidic device was used to investigate the role of M2c macrophage-derived Sema4D in the stabilization of vascular structures. RESULTS Sema4D induced the formation of a greater number of perfused vessels by HUVECs and enhanced the coverage of these vessels by SM22α-positive SHED (SM22α+SHED). Conversely, the knockdown of the Plexin-B1 receptor in HUVECs or inhibition of PDGFR-β reversed the Sema4D-induced vascular stabilization, thereby confirming the regulatory role of the Plexin-B1/PDGF-BB axis in the recruitment of mural cells mediated by Sema4D. Mechanistically, Sema4D was found to upregulate the expression of methyltransferases, specifically METTL3, and to elevate the level of m6A modification in HUVECs. This modification was determined to be critical for enhancing PDGF-BB secretion, suggesting that Sema4D activates an epigenetic regulatory mechanism. Additionally, we investigated the secretion of Sema4D by various macrophage phenotypes, identifying that M2c macrophages secrete significant levels of Sema4D. This secretion recruited SM22α+SHED as mural cells by inducing endothelial PDGF production on a vasculature-on-a-chip platform, indicating a potential role for macrophages in facilitating vascular stabilization. CONCLUSIONS Sema4D acts on Plexin-B1, inducing METTL3-mediated PDGF-BB secretion to recruit SHED to stabilize vessels. Macrophages could be a key source of Sema4D for vascular stabilization.
Collapse
Affiliation(s)
- Lili Zhang
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510055, Guangdong, China
- Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, Prince Philip Dental Hospital, The University of Hong Kong, 34 Hospital Road, Sai Ying Pun, Hong Kong, Hong Kong SAR
| | - Dineshi Sewvandi Thalakiriyawa
- Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, Prince Philip Dental Hospital, The University of Hong Kong, 34 Hospital Road, Sai Ying Pun, Hong Kong, Hong Kong SAR
| | - Jiawei Liu
- Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, Prince Philip Dental Hospital, The University of Hong Kong, 34 Hospital Road, Sai Ying Pun, Hong Kong, Hong Kong SAR
| | - Shengyan Yang
- Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, Prince Philip Dental Hospital, The University of Hong Kong, 34 Hospital Road, Sai Ying Pun, Hong Kong, Hong Kong SAR
| | - Yan Wang
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510055, Guangdong, China
| | - Waruna Lakmal Dissanayaka
- Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, Prince Philip Dental Hospital, The University of Hong Kong, 34 Hospital Road, Sai Ying Pun, Hong Kong, Hong Kong SAR.
| |
Collapse
|
14
|
Guan Z, Wang X, Xu C, Lu R. ALKBH5 alleviates lower extremity arteriosclerosis by regulating ITGB1 demethylation and influencing macrophage polarization. Heliyon 2025; 11:e41495. [PMID: 39866442 PMCID: PMC11757785 DOI: 10.1016/j.heliyon.2024.e41495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 12/17/2024] [Accepted: 12/24/2024] [Indexed: 01/28/2025] Open
Abstract
Objective M6A methylation-regulated macrophages play an important role in the occurrence and development of arteriosclerosis. However, their role in lower extremity arteriosclerosis remains unclear. Therefore, this study aims to explore the key factors that regulate arteriosclerosis methylation in the lower extremities and the mechanism by which they affect arteriosclerosis by influencing macrophage polarization. Methods The m6A methylation levels in peripheral blood mononuclear cells (PBMCs) of patients with lower extremity atherosclerosis was investigated using the Dot blot method. Additionally, the expression levels of RNA methyltransferases and demethylases were examined using ELISA and Western blotting. Inflammatory macrophages were established using RAW264.7 cells stimulated with LPS (100 ng/mL), and the expression of ALKBH5 and ITGB1 was evaluated using Western blotting. Immunofluorescence staining was conducted to assess the expression of M1 macrophage markers (F4/80+CD86) and M2 macrophage markers (F4/80+CD206) in renal tissue. ELISA was employed to measure the levels of cytokines (IL-6, IL-1β, TNF-a, IL-10, and TGF-β) and plasma lipid levels in mice. An atherosclerosis model was established in ApoE-/- mice through balloon pull surgery and high-fat feeding. Oil Red O staining and hematoxylin and eosin (HE) staining were performed to measure the area of atherosclerotic plaques and the size of the necrotic core in mouse femoral artery, respectively. Additionally, Starbase2.0 was used for downstream target gene prediction of ALKBH5. The half-life of ITGB1 was evaluated using RT-qPCR. Results The m6A methylation levels were significantly increased in PBMCs of patients with lower extremity atherosclerosis. Among them, the expression of the RNA demethylase ALKBH5 was the lowest in PBMCs of patients with lower extremity atherosclerosis. Further analysis revealed that ALKBH5 can alleviate the progression of lower extremity atherosclerosis and promote the polarization of M2 macrophages. ALKBH5 can reduce the stability of ITGB1 through demethylation. Mechanistically, ALKBH5 influences macrophage polarization and mitigates lower extremity atherosclerosis by affecting the demethylation of ITGB1. Conclusion ALKBH5 promotes M2 macrophage polarization and alleviates lower extremity atherosclerosis by regulating the demethylation of ITGB1. A deeper understanding of this process not only helps elucidate the molecular mechanisms of atherosclerosis but also provides possibilities for the development of new therapeutic strategies.
Collapse
Affiliation(s)
- Zeyu Guan
- The First Affiliated Hospital of Bengbu Medical University, Department of Vascular Surgery, 287 Changhuai Road, Bengbu, 233004, China
| | - Xiaogao Wang
- The First Affiliated Hospital of Bengbu Medical University, Department of Vascular Surgery, 287 Changhuai Road, Bengbu, 233004, China
| | - Chao Xu
- The First Affiliated Hospital of Bengbu Medical University, Department of Vascular Surgery, 287 Changhuai Road, Bengbu, 233004, China
| | - Ran Lu
- The First Affiliated Hospital of Bengbu Medical University, Department of Vascular Surgery, 287 Changhuai Road, Bengbu, 233004, China
| |
Collapse
|
15
|
Vageli DP, Doukas PG, Georgiou D, Prokopiou MP, Ladaki NE, Papadopoulou A, Doukas SG, Zacharouli K, Makaritsis KP, Ioannou M. HIF-1α and VEGF Immunophenotypes as Potential Biomarkers in the Prognosis and Evaluation of Treatment Efficacy of Atherosclerosis: A Systematic Review of the Literature. FRONT BIOSCI-LANDMRK 2025; 30:27004. [PMID: 39862086 DOI: 10.31083/fbl27004] [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: 10/16/2024] [Revised: 11/09/2024] [Accepted: 11/18/2024] [Indexed: 01/27/2025]
Abstract
BACKGROUND Hypoxia-inducible factor 1 alpha (HIF-1α) and its related vascular endothelial growth factor (VEGF) may play a significant role in atherosclerosis and their targeting is a strategic approach that may affect multiple pathways influencing disease progression. This study aimed to perform a systematic review to reveal current evidence on the role of HIF-1α and VEGF immunophenotypes with other prognostic markers as potential biomarkers of atherosclerosis prognosis and treatment efficacy. METHODS We performed a systematic review of the current literature to explore the role of HIF-1α and VEGF protein expression along with the relation to the prognosis and therapeutic strategies of atherosclerosis. We used the terms {"Atherosclerosis" [OR] "Atheroma" [OR] "atheromatous plaque" [OR] "plaque atherosclerotic"} [AND] {"HIF-1α"} [AND] {"VEGF"} from 2009 up to May 2024 and the Medline/Embase/PubMed database. We used methodological approaches to assess unbiased data [ROBIS (Risk of Bias in Systematic) tool]. We used study eligibility criteria, and data were collected and evaluated from original articles by two independent teams, judged by an independent reviewer, and reported by PRISMA (Preferred Reporting Items for Systematic reviews and Meta-Analyses) 2020. RESULTS We included 34 original studies investigating 650 human specimens, 21 different cell lines, and 9 animal models. Increased HIF-1α in vascular smooth muscle cells, macrophages, or endothelial cells, under hypoxia, chronic loss of nitric oxide (NO), or reduced micro ribonucleic acid (miRNA)-17 and miR-20, is associated with the upregulation of pro-inflammatory molecules, such as interleukin-1 beta (IL-1β) or tumor necrosis factor-alpha (TNF-α), increased migration inhibitory factor of macrophages, glycolytic flux, lipid accumulation, necroptosis via miR-383, and adverse effects in atherosclerosis and plaque vulnerability. However, increased HIF-1α in lymphocytes is associated with decreased interferon-gamma (IFN-γ) and a favorable prognosis. Increased VEGF in a coronary artery, activated macrophages, or chronic exposure to methamphetamine is associated with elevated levels of serum inflammatory cells (interleukin-18; IL18), p38 mitogen-activated protein kinase (MAPK) phosphorylation, lipopolysaccharide-induced tumor necrosis factor-alpha factor (LITAF), and signal transducer and activator of transcription 6 isoform B (STAT6B) overexpression, leading to atherosclerosis progression and plaque break. However, VEGF overexpression in serum is marginally associated with an elevated risk for atherosclerosis. In contrast, stable overexpression of VEGF in macrophages correlates with reduced hyperplasia after arterial injury, reduced foam cell formation, and attenuation of atherosclerosis progression. HIF-1α/VEGF immunophenotypes reflect atherosclerosis treatment efficacy using, among others, HIF-inhibitors, statins, polyphenols, miR-497-5p, methylation modification, adenosine receptor antagonists, natural products, or glycosides. CONCLUSION We present an overview of HIF-1α/VEGF expression in chronic inflammatory-related atherosclerosis disease. Exploring pathogenetic mechanisms and therapeutic options, we included several studies using variable methods to evaluate HIF-1α/VEGF immunophenotypes with controversial and innovative results. Data limitations may include the use of different survival methods. Our data support HIF-1α/VEGF immunophenotypes as potential biomarkers of atherosclerosis prognosis and treatment efficacy.
Collapse
Affiliation(s)
- Dimitra P Vageli
- Department of Neurology, Neuroscience and Regeneration Research Center Yale University School of Medicine & VA-CT, West Haven, CT 06516, USA
- Department of Pathology, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41500 Larissa, Greece
| | - Panagiotis G Doukas
- Department of Medicine, Rutgers-Robert Wood Johnson Medical School/Saint Peter's University Hospital, New Brunswick, NJ 08901, USA
| | - Dimitrios Georgiou
- Department of Pathology, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41500 Larissa, Greece
| | - Michailangelos P Prokopiou
- Department of Pathology, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41500 Larissa, Greece
| | - Nefeli E Ladaki
- Department of Pathology, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41500 Larissa, Greece
| | - Androniki Papadopoulou
- Department of Anesthesiology, G. Gennimatas General Hospital, 54635 Thessaloniki, Greece
| | - Sotirios G Doukas
- Department of Medicine, Section of Gastroenterology and Hepatology, Rutgers-Robert Wood Johnson Medical School/Saint Peter's University Hospital, New Brunswick, NJ 08901, USA
| | - Konstantina Zacharouli
- Department of Pathology, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41500 Larissa, Greece
| | - Konstantinos P Makaritsis
- Department of Medicine & Research Laboratory of Internal Medicine, Faculty of Medicine, University of Thessaly/National Expertise Center of Greece in Autoimmune Liver Diseases, General University Hospital of Larissa, 41110 Larissa, Greece
| | - Maria Ioannou
- Department of Pathology, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41500 Larissa, Greece
| |
Collapse
|
16
|
Feng XM, Zhang Y, Chen N, Ma LL, Gong M, Yan YX. The role of m 6A modification in cardiovascular disease: A systematic review and integrative analysis. Int Immunopharmacol 2024; 143:113603. [PMID: 39536485 DOI: 10.1016/j.intimp.2024.113603] [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: 04/30/2024] [Revised: 09/25/2024] [Accepted: 11/06/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND AND AIMS This study focused on the recent advancements in understanding the association between N6-methyladenosine (m6A) modification and cardiovascular disease (CVD). METHODS The potential mechanisms of m6A related to CVD were summarized by literature review. Associations between m6A levels and CVD were explored across 8 electronic databases: PubMed, Embase, Web of Science, Cochrane Library, Sinomed, Wan Fang, CNKI, and Vip. Standard mean difference (SMD) and 95 % confidence interval (95 % CI) were calculated to assess the total effect in integrated analysis. RESULTS The systematic review summarized previous studies on the association between m6A modification and CVD, highlighting the potential role of m6A in CVD progression. A total of 11 studies were included for integrative analysis. The mean m6A levels were significantly higher in CVD than those in normal controls (SMD = 1.86, 95 % CI: 0.16-3.56, P < 0.01). CONCLUSIONS This systematic review provided new targets for early detection and treatment for CVD. And the integrated analysis showed that increased level of m6A was associated with CVD.
Collapse
Affiliation(s)
- Xu-Man Feng
- Department of Epidemiology and Biostatistics, School of Public Health, Capital Medical University, Beijing, China; Municipal Key Laboratory of Clinical Epidemiology, Beijing, China
| | - Yu Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Capital Medical University, Beijing, China; Municipal Key Laboratory of Clinical Epidemiology, Beijing, China
| | - Ning Chen
- Department of Epidemiology and Biostatistics, School of Public Health, Capital Medical University, Beijing, China; Municipal Key Laboratory of Clinical Epidemiology, Beijing, China
| | - Lin-Lin Ma
- Department of Epidemiology and Biostatistics, School of Public Health, Capital Medical University, Beijing, China; Municipal Key Laboratory of Clinical Epidemiology, Beijing, China
| | - Miao Gong
- Department of Epidemiology and Biostatistics, School of Public Health, Capital Medical University, Beijing, China; Municipal Key Laboratory of Clinical Epidemiology, Beijing, China
| | - Yu-Xiang Yan
- Department of Epidemiology and Biostatistics, School of Public Health, Capital Medical University, Beijing, China; Municipal Key Laboratory of Clinical Epidemiology, Beijing, China
| |
Collapse
|
17
|
Xiong Q, Luo Z, Xie X, Zhou W. KLF7 reverses ox-LDL-induced ferroptosis in HMEC-1 cells through transcriptionally activating ALKBH5 to inhibit the m6A modification of ACSL4. Cytotechnology 2024; 76:653-666. [PMID: 39435423 PMCID: PMC11490622 DOI: 10.1007/s10616-024-00641-2] [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: 02/17/2024] [Accepted: 06/24/2024] [Indexed: 10/23/2024] Open
Abstract
Atherosclerosis is a chronic inflammatory vascular disease. It was confirmed that activation of ferroptosis could induce the development of AS. Meanwhile, Krüppel-like factor 7 was reported to be involved in AS. Nevertheless, the detailed function of KLF7 in ferroptosis during AS has not been not explored. To mimic AS in vitro, human microvascular endothelial cells (HMEC-1) were exposed to 100 μg/mL ox-LDL. Cell viability was tested using MTT assay, and commercial kits were applied to examine the ferroptosis. Flow cytometry was applied for testing lipid ROS level. The relation between KLF7 and AlkB homolog 5 (ALKBH5) was explored using dual luciferase and ChIP assays. Furthermore, MeRIP was used to test the m6A modification level of ACSL4. KLF7 and ALKBH5 overexpression reversed ox-LDL-induced ferroptosis (characterized by up-regulated MDA, iron, Fe2+, lipid ROS and ACSL4, and down-regulated GSH and GPX4) in HMEC-1 cells. In addition, KLF7 transcriptionally activated ALKBH5. ALKBH5 decreased the level of ACSL4 by inhibiting the m6A modification of ACSL4. Furthermore, upregulation of KLF7 restored ox-LDL-induced ferroptosis in HMEC-1 cells via upregulating ALKBH5. KLF7 repressed ox-LDL-induced ferroptosis in HMEC-1 cells through promoting ALKBH5 mediated m6A demethylation of ACSL4. Our study might supply a new therapeutic strategy for AS treatment.
Collapse
Affiliation(s)
- Qinggen Xiong
- Intervention Department (Vascularsurgery Department) of The Second Affiliated Hospital of Nanchang University, No.566 Xuefu Avenue, Honggutan District, Nanchang, 330008 Jiangxi Province China
| | - Zhijian Luo
- Intervention Department (Vascularsurgery Department) of The Second Affiliated Hospital of Nanchang University, No.566 Xuefu Avenue, Honggutan District, Nanchang, 330008 Jiangxi Province China
| | - Xiaoming Xie
- Intervention Department (Vascularsurgery Department) of The Second Affiliated Hospital of Nanchang University, No.566 Xuefu Avenue, Honggutan District, Nanchang, 330008 Jiangxi Province China
| | - Wei Zhou
- Intervention Department (Vascularsurgery Department) of The Second Affiliated Hospital of Nanchang University, No.566 Xuefu Avenue, Honggutan District, Nanchang, 330008 Jiangxi Province China
| |
Collapse
|
18
|
Luan Y, Jia R, Chai P, Fan X. m 6A and beyond: RNA modifications shaping angiogenesis. Trends Mol Med 2024:S1471-4914(24)00302-2. [PMID: 39609142 DOI: 10.1016/j.molmed.2024.11.001] [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: 08/28/2024] [Revised: 10/22/2024] [Accepted: 11/04/2024] [Indexed: 11/30/2024]
Abstract
RNA modifications are crucial post-transcriptional processes that significantly influence gene expression, RNA stability, nuclear transport, and translational capacity. Angiogenesis, the formation of new blood vessels, is a physiological process that is dysregulated in many pathological conditions, including ocular diseases, immune disorders, and cancer. In this review, we compile the current understanding of the intricate relationship between various RNA modifications and angiogenic mechanisms, spotlighting emerging evidence that underscore their pivotal regulatory roles in both physiological and pathological angiogenesis. Furthermore, we delve into recent advances in innovative therapeutic approaches that target RNA modifications to modulate angiogenesis, offering insights into their potential as novel treatment modalities.
Collapse
Affiliation(s)
- Yu Luan
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Renbing Jia
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China.
| | - Peiwei Chai
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China.
| | - Xianqun Fan
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China.
| |
Collapse
|
19
|
Cao Y, Yang Y, Guo C, Zong J, Li M, Li X, Yu T. Role of RNA-binding Proteins in Regulating Cell Adhesion and Progression of the Atherosclerotic Plaque and Plaque Erosion. Curr Atheroscler Rep 2024; 27:8. [PMID: 39576410 DOI: 10.1007/s11883-024-01250-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/15/2024] [Indexed: 11/24/2024]
Abstract
PURPOSE OF REVIEW RNA-binding proteins (RBPs) have emerged as crucial regulators of post-transcriptional processes, influencing the fate of RNA. This review delves into the biological functions of RBPs and their role in alternative splicing concerning atherosclerosis (AS), highlighting their participation in essential cellular processes. Our goal is to offer new insights for cardiovascular disease research and treatment. RECENT FINDING Dysregulation of RBPs is associated with various human diseases, including autoimmune and neurological disorders. The role of RBPs in the pathogenesis of AS is progressively being elucidated, as they influence plaque formation and disease progression by regulating cell function and gene expression. RBPs play intricate biological roles in regulating pre-mRNA, including editing, splicing, stability and translation. Alternative splicing has been demonstrated to enhance biological complexity and diversity. Our findings indicate that alternative splicing is extensively involved in the pathogenesis of AS. The dysregulated expression of specific RBPs in AS is linked to the production of adhesion molecules and vascular endothelium damage. Further research on RBPs could pave the way for the development of novel therapeutic targets.
Collapse
Affiliation(s)
- Ying Cao
- Clinical Laboratory, Central Laboratory, Qingdao Hiser Hospital Affiliated of Qingdao University (Qingdao Traditional Chinese Medicine Hospital), Qingdao, 266000, People's Republic of China
| | - Yanyan Yang
- Department of Immunology, School of Basic Medicine, Qingdao University, No. 308 Ningxia Road, Qingdao, 266000, People's Republic of China
| | - Chuan Guo
- Industrial Synergy Innovation Center, Linyi Vocational University of Science and Technology, Linyi, 276000, People's Republic of China
| | - Jinbao Zong
- Clinical Laboratory, Central Laboratory, Qingdao Hiser Hospital Affiliated of Qingdao University (Qingdao Traditional Chinese Medicine Hospital), Qingdao, 266000, People's Republic of China
| | - Min Li
- Clinical Laboratory, Central Laboratory, Qingdao Hiser Hospital Affiliated of Qingdao University (Qingdao Traditional Chinese Medicine Hospital), Qingdao, 266000, People's Republic of China
| | - Xiaolu Li
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao, 266000, People's Republic of China
| | - Tao Yu
- Clinical Laboratory, Central Laboratory, Qingdao Hiser Hospital Affiliated of Qingdao University (Qingdao Traditional Chinese Medicine Hospital), Qingdao, 266000, People's Republic of China.
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao, 266000, People's Republic of China.
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, No. 38 Dengzhou Road, Qingdao, 266021, People's Republic of China.
| |
Collapse
|
20
|
Chen D, Xu W, Zheng H, Zhang Y, Lin Y, Han Y, Yao F, Shen H. The methyltransferase METTL3 regulates endothelial cell proliferation and inflammation via m 6A RNA methylation-mediated TRAF1 expression. Biochem Biophys Res Commun 2024; 732:150399. [PMID: 39033551 DOI: 10.1016/j.bbrc.2024.150399] [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: 07/10/2024] [Accepted: 07/11/2024] [Indexed: 07/23/2024]
Abstract
The imbalance of vascular endothelial cell homeostasis is the key mechanism for the progression of many vascular diseases. RNA modification, particularly N6-Methyladenosine (m6A), plays important function in numerous biological processes. Nevertheless, the regulatory function of m6A RNA methylation in endothelial dysfunction remains insufficiently characterized. In this study, we established that the m6A methyltransferase METTL3 is critical for regulating endothelial function. Functionally, depletion of METTL3 results in decreased endothelial cells proliferation, survival and inflammatory response. Conversely, overexpression of METTL3 elicited the opposite effects. Mechanistically, MeRIP-seq identified that METTL3 catalyzed m6A modification of TRAF1 mRNA and enhanced TRAF1 translation, thereby up-regulation of TRAF1 protein. Over-expression of TRAF1 successfully rescued the inhibition of proliferation and adhesion of endothelial cells due to METTL3 knockdown. Additionally, m6A methylation-mediated TRAF1 expression can be reversed by the demethylase ALKBH5. Knockdown of ALKBH5 upregulated the level of m6A and protein level of TRAF1, and also increased endothelial cells adhesion and inflammatory response. Collectively, our findings suggest that METTL3 regulates vascular endothelium homeostasis through TRAF1 m6A modification, suggesting that targeting the METTL3-m6A-TRAF1 axis may hold therapeutic potential for patients with vascular diseases.
Collapse
Affiliation(s)
- Duchu Chen
- The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China.
| | - Wentao Xu
- The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Huaxian Zheng
- The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Yuxuan Zhang
- The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Yongzhi Lin
- The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Yulin Han
- The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Fenfen Yao
- The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Haohan Shen
- The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| |
Collapse
|
21
|
Zhen Y, Yang J, Song J, Xing Z, Zheng J. Silencing ARL11 relieved atherosclerotic inflammation and lipid deposition via retraining JAK2/STAT1 pathway. Atherosclerosis 2024; 398:118564. [PMID: 39312826 DOI: 10.1016/j.atherosclerosis.2024.118564] [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: 01/23/2024] [Revised: 08/05/2024] [Accepted: 08/07/2024] [Indexed: 09/25/2024]
Abstract
BACKGROUND AND AIMS Atherosclerosis (AS), an arterial vasculature disease, is characterized by abnormal lipid accumulation and inflammatory response. ADP ribosylation factor like GTPase 11 (ARL11) is linked to multifarious processes in cells. This study aims to clarify the underlying mechanism of ARL11 in AS. METHODS ApoE-/- mice fed with high-fat diet were used as mouse model of AS. Gene expression in AS was determined by mRNA-sequencing. ARL11 expression was detected by real-time PCR, Western blot and immunofluorescence. M1 polarization of macrophages was indicated by TNF-α and IL-6 levels as detected with ELISA, and iNOS expression determined by real-time PCR and Western blot. The role of ARL11 during AS was explored through loss-of-function analysis. RESULTS There were 1301 upregulated and 1110 downregulated genes during AS. These differentially expressed genes (DEGs) were mainly enriched in pathways and terms which are involved in inflammation. Moreover, Arl11 was highly expressed in AS models. Downregulation of Arl11 decreased lipid deposition and atherosclerotic plaques in the aortas of AS mice, and declined inflammatory cytokines and M1 polarization of macrophages induced by IFN-γ. Furthermore, ARL11 interacted with JAK2 and p-JAK2 and modulated their degradation, thus inhibiting the activation of JAK2/STAT1 pathway. CONCLUSIONS ARL11 promoted the development of AS via interacting with JAK2 and activating JAK2/STAT1 pathway. Thus, silencing ARL11 may prevent the process of AS and be a novel way to treat AS.
Collapse
Affiliation(s)
- Yanhua Zhen
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, 110022, China
| | - Jiaqi Yang
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, 110022, China
| | - Ji Song
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Zeyu Xing
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, 110022, China
| | - Jiahe Zheng
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, 110022, China.
| |
Collapse
|
22
|
Jiang M, Zhao W, Wu L, Zhu G. Screening of m6A-associated ferroptosis-related genes in atherosclerosis based on WGCNA. Front Cardiovasc Med 2024; 11:1469805. [PMID: 39529974 PMCID: PMC11550986 DOI: 10.3389/fcvm.2024.1469805] [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: 07/24/2024] [Accepted: 10/10/2024] [Indexed: 11/16/2024] Open
Abstract
Background N6-methyladenosine (m6A) has been shown to mediate ferroptosis but its role in atherosclerosis (AS) is unclear. Methods Differentially expressed m6A-associated ferroptosis-related genes (DE-m6A-Ferr-RGs) were obtained using differential expression analysis and Pearson correlation analysis. Weighted gene co-expression network analysis (WGCNA) was also performed. The intersection of the module genes and the DE-m6A-Ferr-RGs were recorded as candidate m6A-Ferr-related signature genes. Finally, the m6A-Ferr-related signature genes were screened using least absolute shrinkage and selection operator (LASSO) analysis. Expression validation, receiver operating characteristic ( mapping, and immune correlation analysis were also performed based on the m6A-Ferr-related signature genes. The expression of m6A-Ferr-related signature genes was further validated using a real-time polymerase chain reaction (RT-qPCR). Results In total, 6,167 differentially expressed genes were intersected with 24 m6A- and 259 ferroptosis-related genes, respectively, resulting in 113 DE-m6A-Ferr-RGs obtained using Pearson's correlation analysis. The module genes obtained from the WGCNA and the 113 DE-m6A-Ferr-RGs were intersected to obtain 48 candidate m6A-Ferr-related signature genes. LASSO analysis was performed and six m6A-Ferr-related signature genes were screened. In addition, the area under the curve values of all six m6A-Ferr-related signature genes were greater than 0.7, indicating that they had potential diagnostic value. Furthermore, the RT-qPCR results revealed that the expression of SLC3A2, NOX4, and CDO1 was consistent with the transcriptome level. Moreover, there was a significant difference in two types of immune cells between the AS and control groups. Naive B cells, CD8+ T cells, regulatory T cells, and activated natural killer cells were positively correlated with CDO1 and NOX4 but negatively correlated with ATG7, CYBB, and SLC3A2. Conclusion In total, three m6A-Ferr-related signature genes (NOX4, CDO1, and SLC3A2) were obtained through a series of bioinformatics analyses and an RT-qPCR.
Collapse
Affiliation(s)
| | | | | | - Guofu Zhu
- Cardiology Department, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| |
Collapse
|
23
|
Bhattarai PY, Kim G, Lim SC, Choi HS. METTL3-STAT5B interaction facilitates the co-transcriptional m 6A modification of mRNA to promote breast tumorigenesis. Cancer Lett 2024; 603:217215. [PMID: 39218290 DOI: 10.1016/j.canlet.2024.217215] [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: 05/20/2024] [Revised: 08/07/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024]
Abstract
Enhanced expression of methyltransferase-like 3 (METTL3) promotes the m6A modification of specific mRNAs, contributing to breast tumorigenesis. While the mRNA substrates targeted by METTL3 are well characterized, the factors dictating the selection of these specific mRNA remain elusive. This study aimed to examine the regulatory role of the transcription factor STAT5B in METTL3-induced m6A modification. METTL3 specifically interacts with STAT5B in response to mitogenic stimulation by epidermal growth factor (EGF). Chromatin immunoprecipitation and CRISPR/Cas9 mutagenesis showed that STAT5B recruits METTL3 to gene promoters like CCND1, where METTL3 interacts with RPB1, dependent on CDK9-mediated RPB1 (Ser2) phosphorylation during transcription elongation. Inhibition and depletion of either STAT5B or CDK9 prevented the EGF-induced m6A modification of CCND1. The translation efficiency of CCND1 was increased following m6A modification, thereby increasing cell proliferation. STAT5B facilitated METTL3-induced tumor formation by increasing CCND1 expression in an orthotopic mouse model. In clinical context, a positive correlation was observed between p-STAT5B and METTL3 expression in high-grade breast tumors. This study elucidates a novel mechanism that underlies the specificity of m6A modification in breast cancer cells, thereby underscoring its potential therapeutic value.
Collapse
Affiliation(s)
- Poshan Yugal Bhattarai
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Chosun University, Gwangju, 61452, Republic of Korea
| | - Garam Kim
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Chosun University, Gwangju, 61452, Republic of Korea
| | - Sung-Chul Lim
- Department of Pathology, School of Medicine, Chosun University, Gwangju, 61452, Republic of Korea
| | - Hong Seok Choi
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Chosun University, Gwangju, 61452, Republic of Korea.
| |
Collapse
|
24
|
Khan FA, Nsengimana B, Awan UA, Ji XY, Ji S, Dong J. Regulatory roles of N6-methyladenosine (m 6A) methylation in RNA processing and non-communicable diseases. Cancer Gene Ther 2024; 31:1439-1453. [PMID: 38839892 DOI: 10.1038/s41417-024-00789-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 05/12/2024] [Accepted: 05/21/2024] [Indexed: 06/07/2024]
Abstract
Post-transcriptional RNA modification is an emerging epigenetic control mechanism in cells that is important in many different cellular and organismal processes. N6-methyladenosine (m6A) is one of the most prevalent, prolific, and ubiquitous internal transcriptional alterations in eukaryotic mRNAs, making it an important topic in the field of Epigenetics. m6A methylation acts as a dynamical regulatory process that regulates the activity of genes and participates in multiple physiological processes, by supporting multiple aspects of essential mRNA metabolic processes, including pre-mRNA splicing, nuclear export, translation, miRNA synthesis, and stability. Extensive research has linked aberrations in m6A modification and m6A-associated proteins to a wide range of human diseases. However, the impact of m6A on mRNA metabolism and its pathological connection between m6A and other non-communicable diseases, including cardiovascular disease, neurodegenerative disorders, liver diseases, and cancer remains in fragmentation. Here, we review the existing understanding of the overall role of mechanisms by which m6A exerts its activities and address new discoveries that highlight m6A's diverse involvement in gene expression regulation. We discuss m6A deposition on mRNA and its consequences on degradation, translation, and transcription, as well as m6A methylation of non-coding chromosomal-associated RNA species. This study could give new information about the molecular process, early detection, tailored treatment, and predictive evaluation of human non-communicable diseases like cancer. We also explore more about new data that suggests targeting m6A regulators in diseases may have therapeutic advantages.
Collapse
Affiliation(s)
- Faiz Ali Khan
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China.
- Institute of Integrative Medicine, Fudan University, Shanghai, China.
- Department of Basic Sciences Research, Shaukat Khanum Memorial Cancer Hospital and Research Centre (SKMCH&RC), Lahore, Pakistan.
| | - Bernard Nsengimana
- Department of Hepatobiliary Surgery, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Usman Ayub Awan
- Division of Epidemiology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Xin-Ying Ji
- Center for Molecular Medicine, Faculty of Basic Medical Subjects, Shu-Qing Medical College of Zhengzhou, Zhengzhou, Henan, China.
| | - Shaoping Ji
- Center for Molecular Medicine, Faculty of Basic Medical Subjects, Shu-Qing Medical College of Zhengzhou, Zhengzhou, Henan, China.
- Laboratory of Cell Signal Transduction, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China.
| | - Jingcheng Dong
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China.
- Institute of Integrative Medicine, Fudan University, Shanghai, China.
| |
Collapse
|
25
|
Ma S, Qin Y, Ren W. Insulin-like growth factor 2 mRNA-binding protein 1 (IGF2BP1) in hematological diseases. Mol Med 2024; 30:165. [PMID: 39342091 PMCID: PMC11439276 DOI: 10.1186/s10020-024-00936-2] [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: 12/01/2023] [Accepted: 09/13/2024] [Indexed: 10/01/2024] Open
Abstract
The oncofetal mRNA-binding protein IGF2BP1 belongs to a conserved family of RNA-binding proteins. It primarily promotes RNA stability, regulates translation and RNA localization, and mediates gene expression through its downstream effectors. Numerous studies have demonstrated that IGF2BP1 plays crucial roles in embryogenesis and carcinogenesis. IGF2BP1-modulated cell proliferation, invasion, and chemo-resistance in solid tumors have attracted researchers' attention. Additionally, several studies have highlighted the importance of IGF2BP1 in hematologic malignancies and hematological genetic diseases, positioning it as a promising therapeutic target for hematological disorders. However, there is a lack of systematic summaries regarding the IGF2BP1 gene within the hematological field. In this review, we provide a comprehensive overview of the discovery and molecular structure of IGF2BP1, along with recent studies on its role in regulating embryogenesis. We also focus on the mechanisms by which IGF2BP1 regulates hematological malignancies through its interactions with its targeted mRNAs. Furthermore, we systematically elucidate the function and mechanism of IGF2BP1 in promoting fetal hemoglobin expression in adult hematopoietic stem/progenitor cells. Finally, we discuss the limitations and challenges of IGF2BP1 as a therapeutic target, offering insights into its prospects.
Collapse
Affiliation(s)
- Shuangping Ma
- Institutes of Health Central Plains, Xinxiang Medical University, Xinxiang, 453003, China.
| | - Yiran Qin
- Institutes of Health Central Plains, Xinxiang Medical University, Xinxiang, 453003, China
| | - Wenjie Ren
- Institutes of Health Central Plains, Xinxiang Medical University, Xinxiang, 453003, China.
| |
Collapse
|
26
|
Long H, Yu Y, Ouyang J, Lu H, Zhao G. Insights into RNA N6-methyladenosine and programmed cell death in atherosclerosis. Mol Med 2024; 30:137. [PMID: 39227813 PMCID: PMC11373444 DOI: 10.1186/s10020-024-00901-z] [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: 07/07/2024] [Accepted: 08/18/2024] [Indexed: 09/05/2024] Open
Abstract
N6-methyladenosine (m6A) modification stands out among various RNA modifications as the predominant form within eukaryotic cells, influencing numerous cellular processes implicated in disease development. m6A modification has gained increasing attention in the development of atherosclerosis and has become a research hotspot in recent years. Programmed cell death (PCD), encompassing apoptosis, autophagy, pyroptosis, ferroptosis, and necroptosis, plays a pivotal role in atherosclerosis pathogenesis. In this review, we delve into the intricate interplay between m6A modification and diverse PCD pathways, shedding light on their complex association during the onset and progression of atherosclerosis. Clarifying the relationship between m6A and PCD in atherosclerosis is of great significance to provide novel strategies for cardiovascular disease treatment.
Collapse
Affiliation(s)
- Haijiao Long
- Health Management Center, The Third Xiangya Hospital of Central South University, Changsha, 410013, China
- Department of Cardiology, The Third Xiangya Hospital of Central South University, Changsha, 410013, China
| | - Yulu Yu
- Afliated Qingyuan Hospital, Guangzhou Medical University (Qingyuan People's Hospital), Qingyuan, Guangdong, China
| | - Jie Ouyang
- Health Management Center, The Third Xiangya Hospital of Central South University, Changsha, 410013, China
- Department of Cardiology, The Third Xiangya Hospital of Central South University, Changsha, 410013, China
| | - Hongwei Lu
- Health Management Center, The Third Xiangya Hospital of Central South University, Changsha, 410013, China.
- Department of Cardiology, The Third Xiangya Hospital of Central South University, Changsha, 410013, China.
| | - Guojun Zhao
- Afliated Qingyuan Hospital, Guangzhou Medical University (Qingyuan People's Hospital), Qingyuan, Guangdong, China.
| |
Collapse
|
27
|
Wang T, Yu Y, Ding Y, Yang Z, Jiang S, Gao F, Liu S, Shao L, Shen Z. miR-3529-3p/ABCA1 axis regulates smooth muscle cell homeostasis by enhancing inflammation via JAK2/STAT3 pathway. Front Cardiovasc Med 2024; 11:1441123. [PMID: 39257845 PMCID: PMC11384995 DOI: 10.3389/fcvm.2024.1441123] [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: 06/11/2024] [Accepted: 08/12/2024] [Indexed: 09/12/2024] Open
Abstract
Background Thoracic Aortic Dissection (TAD) is a life-threatening disease without effective drug treatments. The disruption of HASMCs homeostasis is one direct histopathologic alteration in TAD pathological process. Several miRNAs have been shown abnormally expressed in TAD and to regulate HASMCs homeostasis. The primary goal of this study is to identify the miRNAs and the specific mechanisms that lead to HASMCs homeostasis disruption. Methods Bulk miRNA sequencing was performed to explore the aberrantly expressed miRNA profile in TAD, and differentially expressed miRNAs were verified with qRT-PCR. To explore the role of the key miRNAs (miR-3529) in HASMCs homeostasis, we overexpressed this miRNA with lentivirus in HASMCs. Integrative transcriptomics and metabolomics analysis were used to uncover the functional roles of this miRNA in regulating HASMCs homeostasis. Further, the target gene of miR-3529 was predicted by bioinformatics and verified through a dual-luciferase reporter assay. Results Bulk miRNA sequencing showed miR-3529 was elevated in TAD tissues and confirmed by qRT-PCR. Further experimental assay revealed miR-3529 upregulation induced HASMCs homeostasis disruption, accompanied by reducing contractile markers and increasing pro-inflammatory cytokines. Integrative transcriptomics and metabolomics analysis showed that miR-3529 overexpression altered the metabolic profile of HASMC, particularly lipid metabolism. ABCA1 was found to be a direct target of miR-3529. Mechanistically, the miR-3529/ABCA1 axis disrupted HASMCs homeostasis through the JAK2/STAT3 signaling pathway. Conclusions miR-3529 is elevated in TAD patients and disrupts HASMCs homeostasis by reprogramming metabolism through the JAK2/STAT3 signaling pathway. These findings favor a role for miR-3529 as a novel target for TAD therapy.
Collapse
Affiliation(s)
- Tingyu Wang
- Department of Cardiovascular Surgery of the First Affiliated Hospital of Soochow University & Institute for Cardiovascular Science, Soochow University, Suzhou, China
| | - You Yu
- Department of Cardiovascular Surgery of the First Affiliated Hospital of Soochow University & Institute for Cardiovascular Science, Soochow University, Suzhou, China
| | - Yinglong Ding
- Department of Cardiovascular Surgery of the First Affiliated Hospital of Soochow University & Institute for Cardiovascular Science, Soochow University, Suzhou, China
| | - Ziying Yang
- Department of Cardiovascular Surgery of the First Affiliated Hospital of Soochow University & Institute for Cardiovascular Science, Soochow University, Suzhou, China
| | - Shumin Jiang
- Department of Cardiovascular Surgery of the First Affiliated Hospital of Soochow University & Institute for Cardiovascular Science, Soochow University, Suzhou, China
| | - Faxiong Gao
- Department of Cardiovascular Surgery of the First Affiliated Hospital of Soochow University & Institute for Cardiovascular Science, Soochow University, Suzhou, China
| | - Shan Liu
- Department of Cardiovascular Surgery of the First Affiliated Hospital of Soochow University & Institute for Cardiovascular Science, Soochow University, Suzhou, China
| | - Lianbo Shao
- Department of Cardiovascular Surgery of the First Affiliated Hospital of Soochow University & Institute for Cardiovascular Science, Soochow University, Suzhou, China
| | - Zhenya Shen
- Department of Cardiovascular Surgery of the First Affiliated Hospital of Soochow University & Institute for Cardiovascular Science, Soochow University, Suzhou, China
| |
Collapse
|
28
|
An W, Zhou J, Qiu Z, Wang P, Han X, Cheng Y, He Z, An Y, Li S. Identification of crosstalk genes and immune characteristics between Alzheimer's disease and atherosclerosis. Front Immunol 2024; 15:1443464. [PMID: 39188714 PMCID: PMC11345154 DOI: 10.3389/fimmu.2024.1443464] [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: 06/04/2024] [Accepted: 07/25/2024] [Indexed: 08/28/2024] Open
Abstract
Background Advancements in modern medicine have extended human lifespan, but they have also led to an increase in age-related diseases such as Alzheimer's disease (AD) and atherosclerosis (AS). Growing research evidence indicates a close connection between these two conditions. Methods We downloaded four gene expression datasets related to AD and AS from the Gene Expression Omnibus (GEO) database (GSE33000, GSE100927, GSE44770, and GSE43292) and performed differential gene expression (DEGs) analysis using the R package "limma". Through Weighted gene correlation network analysis (WGCNA), we selected the gene modules most relevant to the diseases and intersected them with the DEGs to identify crosstalk genes (CGs) between AD and AS. Subsequently, we conducted functional enrichment analysis of the CGs using DAVID. To screen for potential diagnostic genes, we applied the least absolute shrinkage and selection operator (LASSO) regression and constructed a logistic regression model for disease prediction. We established a protein-protein interaction (PPI) network using STRING (https://cn.string-db.org/) and Cytoscape and analyzed immune cell infiltration using the CIBERSORT algorithm. Additionally, NetworkAnalyst (http://www.networkanalyst.ca) was utilized for gene regulation and interaction analysis, and consensus clustering was employed to determine disease subtypes. All statistical analyses and visualizations were performed using various R packages, with a significance level set at p<0.05. Results Through intersection analysis of disease-associated gene modules identified by DEGs and WGCNA, we identified a total of 31 CGs co-existing between AD and AS, with their biological functions primarily associated with immune pathways. LASSO analysis helped us identify three genes (C1QA, MT1M, and RAMP1) as optimal diagnostic CGs for AD and AS. Based on this, we constructed predictive models for both diseases, whose accuracy was validated by external databases. By establishing a PPI network and employing four topological algorithms, we identified four hub genes (C1QB, CSF1R, TYROBP, and FCER1G) within the CGs, closely related to immune cell infiltration. NetworkAnalyst further revealed the regulatory networks of these hub genes. Finally, defining C1 and C2 subtypes for AD and AS respectively based on the expression profiles of CGs, we found the C2 subtype exhibited immune overactivation. Conclusion This study utilized gene expression matrices and various algorithms to explore the potential links between AD and AS. The identification of CGs revealed interactions between these two diseases, with immune and inflammatory imbalances playing crucial roles in their onset and progression. We hope these findings will provide valuable insights for future research on AD and AS.
Collapse
Affiliation(s)
- Wenhao An
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Jiajun Zhou
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Zhiqiang Qiu
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Peishen Wang
- Department of Research and Development, Beijing Yihua Biotechnology Co., Ltd, Beijing, China
| | - Xinye Han
- Department of Research and Development, Beijing Yihua Biotechnology Co., Ltd, Beijing, China
| | - Yanwen Cheng
- Department of Research and Development, Beijing Yihua Biotechnology Co., Ltd, Beijing, China
| | - Zi He
- Department of Research and Development, Beijing Yihua Biotechnology Co., Ltd, Beijing, China
| | - Yihua An
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Shouwei Li
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
29
|
Li C, Liu L, Li S, Liu YS. N 6-Methyladenosine in Vascular Aging and Related Diseases: Clinical Perspectives. Aging Dis 2024; 15:1447-1473. [PMID: 37815911 PMCID: PMC11272212 DOI: 10.14336/ad.2023.0924-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 09/24/2023] [Indexed: 10/12/2023] Open
Abstract
Aging leads to progressive deterioration of the structure and function of arteries, which eventually contributes to the development of vascular aging-related diseases. N6-methyladenosine (m6A) is the most prevalent modification in eukaryotic RNAs. This reversible m6A RNA modification is dynamically regulated by writers, erasers, and readers, playing a critical role in various physiological and pathological conditions by affecting almost all stages of the RNA life cycle. Recent studies have highlighted the involvement of m6A in vascular aging and related diseases, shedding light on its potential clinical significance. In this paper, we comprehensively discuss the current understanding of m6A in vascular aging and its clinical implications. We discuss the molecular insights into m6A and its association with clinical realities, emphasizing its significance in unraveling the mechanisms underlying vascular aging. Furthermore, we explore the possibility of m6A and its regulators as clinical indicators for early diagnosis and prognosis prediction and investigate the therapeutic potential of m6A-associated anti-aging approaches. We also examine the challenges and future directions in this field and highlight the necessity of integrating m6A knowledge into patient-centered care. Finally, we emphasize the need for multidisciplinary collaboration to advance the field of m6A research and its clinical application.
Collapse
Affiliation(s)
- Chen Li
- Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.
- Institute of Aging and Age-related Disease Research, Central South University, Changsha, Hunan, China
| | - Le Liu
- Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.
- Institute of Aging and Age-related Disease Research, Central South University, Changsha, Hunan, China
| | - Shuang Li
- Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.
- Institute of Aging and Age-related Disease Research, Central South University, Changsha, Hunan, China
| | - You-Shuo Liu
- Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.
- Institute of Aging and Age-related Disease Research, Central South University, Changsha, Hunan, China
| |
Collapse
|
30
|
Zheng Y, Lin S, Chen M, Xu L, Huang H. Regulation of N 6-methyladenosine modification in erythropoiesis and thalassemia. Clin Genet 2024; 106:3-12. [PMID: 38488342 DOI: 10.1111/cge.14518] [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: 11/23/2023] [Revised: 02/21/2024] [Accepted: 02/23/2024] [Indexed: 06/04/2024]
Abstract
In eukaryotic RNA, N6-methyladenosine (m6A) is a prevalent form of methylation modification. The m6A modification process is reversible and dynamic, written by m6A methyltransferase complex, erased by m6A demethylase, and recognized by m6A binding proteins. Through mediating RNA stability, decay, alternative splicing, and translation processes, m6A modification regulates gene expression at the post-transcriptional level. Erythropoiesis is the process of hematopoietic stem cells undergoing proliferation, a series of differentiation and maturation to form red blood cells (RBCs). Thalassemia is a common monogenic disease characterized by excessive production of ineffective RBCs in the peripheral circulation, resulting in hemolytic anemia. Increasing evidence suggests that m6A modification plays a crucial role in erythropoiesis. In this review, we comprehensively summarize the function of m6A modification in erythropoiesis and further generalize the mechanism of m6A modification regulating ineffective erythropoiesis and fetal hemoglobin expression. The purpose is to improve the understanding of the pathogenesis of erythroid dysplasia and offer new perspectives for the diagnosis and treatment of thalassemia.
Collapse
Affiliation(s)
- Yanping Zheng
- Medical Genetic Diagnosis and Therapy Center of Fujian Maternity and Child Health Hospital College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Provincial Key Laboratory of Prenatal Diagnosis and Birth Defect, Fujian Medical University, Fuzhou, China
| | - Siyang Lin
- Medical Genetic Diagnosis and Therapy Center of Fujian Maternity and Child Health Hospital College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Provincial Key Laboratory of Prenatal Diagnosis and Birth Defect, Fujian Medical University, Fuzhou, China
- The School of Medical Technology and Engineering, Fujian Medical University, Fuzhou, China
| | - Meihuan Chen
- Medical Genetic Diagnosis and Therapy Center of Fujian Maternity and Child Health Hospital College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Provincial Key Laboratory of Prenatal Diagnosis and Birth Defect, Fujian Medical University, Fuzhou, China
- The School of Medical Technology and Engineering, Fujian Medical University, Fuzhou, China
- Fujian Clinical Research Center for Maternal-Fetal Medicine, Fuzhou, China
- National Key Obstetric Clinical Specialty Construction Institution of China, Fuzhou, China
| | - Liangpu Xu
- Medical Genetic Diagnosis and Therapy Center of Fujian Maternity and Child Health Hospital College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Provincial Key Laboratory of Prenatal Diagnosis and Birth Defect, Fujian Medical University, Fuzhou, China
- The School of Medical Technology and Engineering, Fujian Medical University, Fuzhou, China
- Fujian Clinical Research Center for Maternal-Fetal Medicine, Fuzhou, China
- National Key Obstetric Clinical Specialty Construction Institution of China, Fuzhou, China
| | - Hailong Huang
- Medical Genetic Diagnosis and Therapy Center of Fujian Maternity and Child Health Hospital College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Provincial Key Laboratory of Prenatal Diagnosis and Birth Defect, Fujian Medical University, Fuzhou, China
- The School of Medical Technology and Engineering, Fujian Medical University, Fuzhou, China
- Fujian Clinical Research Center for Maternal-Fetal Medicine, Fuzhou, China
- National Key Obstetric Clinical Specialty Construction Institution of China, Fuzhou, China
| |
Collapse
|
31
|
Li J, Zou C, Zhang Z, Xue F. N 6-methyladenosine (m 6A) reader YTHDF2 accelerates endothelial cells ferroptosis in cerebrovascular atherosclerosis. Mol Cell Biochem 2024; 479:1853-1861. [PMID: 37792239 DOI: 10.1007/s11010-023-04858-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 09/09/2023] [Indexed: 10/05/2023]
Abstract
Cerebrovascular diseases have extreme high mortality and disability rate worldwide, and endothelial cells injury-induced atherosclerosis acts as the main cause of cerebrovascular disease. Ferroptosis is a novel type of programmed cell death depending on iron-lipid peroxidation. Recent studies have revealed that ferroptosis might promote the progression of atherosclerosis (AS). Here, this research aimed to investigate the function and its profound mechanism on vascular endothelial cells in atherosclerosis. Research results revealed that YTHDF2 expression up-regulated in ox-LDL treated human umbilical vein endothelial cells (HUVECs). Gain/loss functional assays indicated that YTHDF2 overexpression inhibited HUVECs' proliferation and accelerated the ferroptosis in ox-LDL-administered HUVECs. Meanwhile, YTHDF2 silencing promoted cell proliferation and reduced the ferroptosis in ox-LDL-administered HUVECs. Mechanistically, in silico analysis suggested that there were potential m6A-modified sites on SLC7A11 mRNA, and YTHDF2 could bind with SLC7A11 mRNA via m6A-dependent manner. YTHDF2 promoted the degradation of SLC7A11 mRNA, thereby reducing its mRNA stability. Taken together, these findings suggest that YTHDF2 accelerates endothelial cells ferroptosis in cerebrovascular atherosclerosis, helping us enhance our comprehension on cerebrovascular disease pathological physiology.
Collapse
Affiliation(s)
- Jia Li
- Department of Surgery, Tianjin Nankai Hospital, Tianjin Medical University, No. 6 Changjiang Road, Tianjin, 300100, China
| | - Changlin Zou
- Department of Surgery, Tianjin Nankai Hospital, Tianjin Medical University, No. 6 Changjiang Road, Tianjin, 300100, China.
| | - Zhiming Zhang
- Department of Surgery, Tianjin Nankai Hospital, Tianjin Medical University, No. 6 Changjiang Road, Tianjin, 300100, China
| | - Feng Xue
- Department of Surgery, Tianjin Nankai Hospital, Tianjin Medical University, No. 6 Changjiang Road, Tianjin, 300100, China
| |
Collapse
|
32
|
Li R, Zhu C, Wang Y, Wang X, Wang Y, Wang J, Wang K. The relationship between the network of non-coding RNAs-molecular targets and N6-methyladenosine modification in tumors of urinary system. Cell Death Dis 2024; 15:275. [PMID: 38632251 PMCID: PMC11024199 DOI: 10.1038/s41419-024-06664-z] [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: 11/02/2023] [Revised: 04/06/2024] [Accepted: 04/10/2024] [Indexed: 04/19/2024]
Abstract
N6-methyladenosine (m6A) methylation, a prevalent eukaryotic post-transcriptional modification, is involved in multiple biological functions, including mediating variable splicing, RNA maturation, transcription, and nuclear export, and also is vital for regulating RNA translation, stability, and cytoplasmic degradation. For example, m6A methylation can regulate pre-miRNA expression by affecting both splicing and maturation. Non-coding RNA (ncRNA), which includes microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs), does not encode proteins but has powerful impacts on transcription and translation. Conversely, ncRNAs may impact m6A methylation by affecting the expression of m6A regulators, including miRNAs targeting mRNA of m6A regulators, or lncRNAs, and circRNAs, acting as scaffolds to regulate transcription of m6A regulatory factors. Dysregulation of m6A methylation is common in urinary tumors, and the regulatory role of ncRNAs is also important for these malignancies. This article provides a systematic review of the role and mechanisms of action of m6A methylation and ncRNAs in urinary tumors.
Collapse
Affiliation(s)
- Ruiming Li
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Chunming Zhu
- Department of Family Medicine, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Yuan Wang
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Xia Wang
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Yibing Wang
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, 110004, China.
| | - Jiahe Wang
- Department of Family Medicine, Shengjing Hospital of China Medical University, Shenyang, 110004, China.
| | - Kefeng Wang
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, 110004, China.
| |
Collapse
|
33
|
Cai Y, Wang Y, Mao B, You Q, Guo X. Targeting insulin-like growth factor 2 mRNA-binding proteins (IGF2BPs) for the treatment of cancer. Eur J Med Chem 2024; 268:116241. [PMID: 38382391 DOI: 10.1016/j.ejmech.2024.116241] [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: 12/14/2023] [Revised: 02/05/2024] [Accepted: 02/14/2024] [Indexed: 02/23/2024]
Abstract
Insulin-like growth factor 2 mRNA-binding proteins (IMPs, IGF2BPs) are RNA-binding proteins that regulate a variety of biological processes. In recent years, several studies have found that IGF2BPs play multiple roles in various biological processes, especially in cancer, and speculated on their mechanism of anticancer effect. In addition, targeting IGF2BPs or their downstream target gene has also received extensive attention as an effective treatment for different types of cancer. In this review, we summarized the recent progress on the role of IGF2BPs in cancers and their structural characteristics. We focused on describing the development of inhibitors targeting IGF2BPs and the prospects for further applications.
Collapse
Affiliation(s)
- Yuanqian Cai
- State Key Laboratory of Natural Medicines, and Jiang Su Key Laboratory of Drug, Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Yingzhe Wang
- State Key Laboratory of Natural Medicines, and Jiang Su Key Laboratory of Drug, Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Bingjie Mao
- State Key Laboratory of Natural Medicines, and Jiang Su Key Laboratory of Drug, Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Qidong You
- State Key Laboratory of Natural Medicines, and Jiang Su Key Laboratory of Drug, Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| | - Xiaoke Guo
- State Key Laboratory of Natural Medicines, and Jiang Su Key Laboratory of Drug, Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
34
|
Guo B, Yu Y, Wang M, Li R, He X, Tang S, Liu Q, Mao Y. Targeting the JAK2/STAT3 signaling pathway with natural plants and phytochemical ingredients: A novel therapeutic method for combatting cardiovascular diseases. Biomed Pharmacother 2024; 172:116313. [PMID: 38377736 DOI: 10.1016/j.biopha.2024.116313] [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: 01/06/2024] [Revised: 02/13/2024] [Accepted: 02/17/2024] [Indexed: 02/22/2024] Open
Abstract
The aim of this article is to introduce the roles and mechanisms of the JAK2/STAT3 pathway in various cardiovascular diseases, such as myocardial fibrosis, cardiac hypertrophy, atherosclerosis, myocardial infarction, and myocardial ischemiareperfusion. In addition, the effects of phytochemical ingredients and different natural plants, mainly traditional Chinese medicines, on the regulation of different cardiovascular diseases via the JAK2/STAT3 pathway are discussed. Surprisingly, the JAK2 pathway has dual roles in different cardiovascular diseases. Future research should focus on the dual regulatory effects of different phytochemical ingredients and natural plants on JAK2 to pave the way for their use in clinical trials.
Collapse
Affiliation(s)
- Bing Guo
- The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha 410005, China
| | - Yunfeng Yu
- The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha 410007, China
| | - Min Wang
- The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha 410007, China
| | - Ronghui Li
- College of Chinese Medicine, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Xuan He
- The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha 410005, China
| | - Siqin Tang
- The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha 410005, China
| | - Qili Liu
- The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha 410007, China
| | - Yilin Mao
- The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha 410005, China.
| |
Collapse
|
35
|
Li B, Xuan H, Yin Y, Wu S, Du L. The N 6-methyladenosine modification in pathologic angiogenesis. Life Sci 2024; 339:122417. [PMID: 38244915 DOI: 10.1016/j.lfs.2024.122417] [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: 11/08/2023] [Revised: 01/03/2024] [Accepted: 01/07/2024] [Indexed: 01/22/2024]
Abstract
The vascular system is a vital circulatory network in the human body that plays a critical role in almost all physiological processes. The production of blood vessels in the body is a significant area of interest for researchers seeking to improve their understanding of vascular function and maintain normal vascular operation. However, an excessive or insufficient vascular regeneration process may lead to the development of various ailments such as cancer, eye diseases, and ischemic diseases. Recent preclinical and clinical studies have revealed new molecular targets and principles that may enhance the therapeutic effect of anti-angiogenic strategies. A thorough comprehension of the mechanism responsible for the abnormal vascular growth in disease processes can enable researchers to better target and effectively suppress or treat the disease. N6-methyladenosine (m6A), a common RNA methylation modification method, has emerged as a crucial regulator of various diseases by modulating vascular development. In this review, we will cover how m6A regulates various vascular-related diseases, such as cancer, ocular diseases, neurological diseases, ischemic diseases, emphasizing the mechanism of m6A methylation regulators on angiogenesis during pathological process.
Collapse
Affiliation(s)
- Bin Li
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| | - Hanqin Xuan
- Department of Pathology, the First Affiliated Hospital of Soochow University, Jiangsu, China
| | - Yuye Yin
- College of Bioscience and Biotechnology, Yangzhou University, Yangzhou, Jiangsu 225009, China
| | - Shusheng Wu
- Department of Neurology, Affiliated Hospital of Yangzhou University, Jiangsu, China.
| | - Longfei Du
- Department of Laboratory Medicine, Affiliated Hospital of Yangzhou University, Yangzhou, Jiangsu, China.
| |
Collapse
|
36
|
Cheng C, Yu F, Yuan G, Jia J. Update on N6-methyladenosine methylation in obesity-related diseases. Obesity (Silver Spring) 2024; 32:240-251. [PMID: 37989724 DOI: 10.1002/oby.23932] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 08/28/2023] [Accepted: 08/28/2023] [Indexed: 11/23/2023]
Abstract
Obesity is a chronic metabolic disease that is closely related to type 2 diabetes mellitus, cardiovascular diseases, nonalcoholic fatty liver disease, obstructive sleep apnea, and osteoarthritis. The prevalence of obesity is increasing rapidly every year and is recognized as a global public health problem. In recent years, the role of epigenetics in the development of obesity and related diseases has been recognized and is currently a research hotspot. N6-methyladenosine (m6A) methylation is the most abundant epigenetic modification in the eukaryotic RNA, including mRNA and noncoding RNA. Several studies have shown that the m6A modifications in the target mRNA and the corresponding m6A regulators play a significant role in lipid metabolism and are strongly associated with the pathogenesis of obesity-related diseases. In this review, the latest research findings regarding the role of m6A methylation in obesity and related metabolic diseases are summarized. The authors' aim is to highlight evidence that suggests the clinical utility of m6A modifications and the m6A regulators as novel early prediction biomarkers and precision therapeutics for obesity and obesity-related diseases.
Collapse
Affiliation(s)
- Caiqin Cheng
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Jiangsu University; Institute of Endocrine and Metabolic Diseases, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Fan Yu
- Department of Endocrinology and Metabolism, Jurong Hospital Affiliated to Jiangsu University, Zhenjiang, Jiangsu, China
| | - Guoyue Yuan
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Jiangsu University; Institute of Endocrine and Metabolic Diseases, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Jue Jia
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Jiangsu University; Institute of Endocrine and Metabolic Diseases, Jiangsu University, Zhenjiang, Jiangsu, China
| |
Collapse
|
37
|
Ma L, Chang X, Gao J, Zhang Y, Chen Y, Zhou H, Zhou N, Du N, Li J, Bi J, Chen Z, Chen X, He Q. METTL3 boosts mitochondrial fission and induces cardiac fibrosis after ischemia/reperfusion injury. Int J Biol Sci 2024; 20:433-445. [PMID: 38169612 PMCID: PMC10758110 DOI: 10.7150/ijbs.87535] [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: 06/27/2023] [Accepted: 10/30/2023] [Indexed: 01/05/2024] Open
Abstract
METTL3, an RNA methyltransferase enzyme, exerts therapeutic effects on various cardiovascular diseases. Myocardial ischemia-reperfusion injury (MIRI) and subsequently cardiac fibrosis is linked to acute cardiomyocyte death or dysfunction induced by mitochondrial damage, particularly mitochondrial fission. Our research aims to elucidate the potential mechanisms underlying the therapeutic actions of METTL3 in MIRI, with focus on mitochondrial fission. When compared with Mettl3flox mice subjected to MIRI, Mettl3 cardiomyocyte knockout (Mettl3Cko) mice have reduced infarct size, decreased serum levels of myocardial injury-related factors, limited cardiac fibrosis, and preserved myocardial ultrastructure and contractile/relaxation capacity. The cardioprotective actions of Mettl3 knockout were associated with reduced inflammatory responses, decreased myocardial neutrophil infiltration, and suppression of cardiomyocyte death. Through signaling pathway validation experiments and assays in cultured HL-1 cardiomyocytes exposed to hypoxia/reoxygenation, we confirmed that Mettl3 deficiency interfere with DNA-PKcs phosphorylation, thereby blocking the downstream activation of Fis1 and preventing pathological mitochondrial fission. In conclusion, this study confirms that inhibition of METTL3 can alleviate myocardial cardiac fibrosis inflammation and prevent cardiomyocyte death under reperfusion injury conditions by disrupting DNA-PKcs/Fis1-dependent mitochondrial fission, ultimately improving cardiac function. These findings suggest new approaches for clinical intervention in patients with MIRI.
Collapse
Affiliation(s)
- Li Ma
- Heart Center, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Xing Chang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Jing Gao
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Ying Zhang
- Senior Department of Cardiology, The Sixth Medical Center of People's Liberation Army General Hospital, Beijing 100048, China
| | - Ye Chen
- Senior Department of Cardiology, The Sixth Medical Center of People's Liberation Army General Hospital, Beijing 100048, China
| | - Hao Zhou
- Senior Department of Cardiology, The Sixth Medical Center of People's Liberation Army General Hospital, Beijing 100048, China
| | - Na Zhou
- Heart Center, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Na Du
- Heart Center, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Jiamin Li
- Heart Center, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Jiachen Bi
- Heart Center, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Ziyue Chen
- Heart Center, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Xinxin Chen
- Heart Center, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Qingyong He
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| |
Collapse
|
38
|
Tian Q, Mu Q, Liu S, Huang K, Tang Y, Zhang P, Zhao J, Shu C. m6A-modified circASXL1 promotes proliferation and migration of ovarian cancer through the miR-320d/RACGAP1 axis. Carcinogenesis 2023; 44:859-870. [PMID: 37738681 DOI: 10.1093/carcin/bgad066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 05/18/2023] [Accepted: 09/21/2023] [Indexed: 09/24/2023] Open
Abstract
Ovarian cancer (OC) is one of the most common malignant tumors in women. Circular RNAs (circRNAs) can potentially regulate the development of OC. Therefore, this study investigated the role of circASXL1 in OC progression. Cell functions were assessed by MTT, colony formation, wound healing, and transwell assays. RIP and dual luciferase reporter assays confirmed the relationship between miR-320d and circASXL1 or RACGAP1. MeRIP was utilized to detect m6A levels. Xenograft tumor was established for in vivo experiments. CircASXL1 and RACGAP1 levels were increased in OC tissues and cells, whereas miR-320d expression was decreased. Upregulation of circASXL1 was associated with poor prognosis in OC patients. CircASXL1 silencing suppressed OC cell proliferation, migration and invasion in vitro and in vivo. Mechanistically, METTL3/IGF2BP1-mediated m6A modification maintained circASXL1 stability and upregulated its expression. CircASXL1 was a ceRNA that sequestrated miR-320d from RACGAP1, leading to increased RACGAP1 expression. CircASXL1 promoted OC cell proliferation, migration and invasion via the miR-320d/RACGAP1 axis. Therefore, m6A-modified circASXL1 acts as an oncogene in OC by targeting miR-320d and activating RACGAP1/PI3K/Akt pathway, which provides novel promising biomarkers for OC diagnosis.
Collapse
Affiliation(s)
- Qi Tian
- Department of Obstetrics and Gynecology, Hunan Provincial Maternal and Child Health Care Hospital (Reproductive Medicine Institute of Hunan Province), Changsha 410008, Hunan Province, P.R. China
- National Health Commission Key Laboratory of Birth Defects Research, Prevention and Treatment, Hunan Provincial Maternal and Child Health Care Hospital, Changsha 410008, Hunan Province, P.R. China
| | - Qingling Mu
- Department of Obstetrics and Gynecology, Qingdao Municipal Hospital, Qingdao 266000, Shandong Province, P.R. China
| | - Shuang Liu
- Department of Obstetrics and Gynecology, Hunan Provincial Maternal and Child Health Care Hospital (Reproductive Medicine Institute of Hunan Province), Changsha 410008, Hunan Province, P.R. China
| | - Kui Huang
- Department of Obstetrics and Gynecology, Hunan Provincial Maternal and Child Health Care Hospital (Reproductive Medicine Institute of Hunan Province), Changsha 410008, Hunan Province, P.R. China
| | - Yi Tang
- Department of Obstetrics and Gynecology, Hunan Provincial Maternal and Child Health Care Hospital (Reproductive Medicine Institute of Hunan Province), Changsha 410008, Hunan Province, P.R. China
| | - Pu Zhang
- Department of Obstetrics and Gynecology, Hunan Provincial Maternal and Child Health Care Hospital (Reproductive Medicine Institute of Hunan Province), Changsha 410008, Hunan Province, P.R. China
| | - Jing Zhao
- Department of Obstetrics and Gynecology, Hunan Provincial Maternal and Child Health Care Hospital (Reproductive Medicine Institute of Hunan Province), Changsha 410008, Hunan Province, P.R. China
| | - Chuqiang Shu
- Department of Obstetrics and Gynecology, Hunan Provincial Maternal and Child Health Care Hospital (Reproductive Medicine Institute of Hunan Province), Changsha 410008, Hunan Province, P.R. China
- National Health Commission Key Laboratory of Birth Defects Research, Prevention and Treatment, Hunan Provincial Maternal and Child Health Care Hospital, Changsha 410008, Hunan Province, P.R. China
| |
Collapse
|
39
|
Zheng X, Zhou B, Li Y, Zhong H, Huang Z, Gu M. Transcriptome-wide N 6-methyladenosine methylation profile of atherosclerosis in mice. BMC Genomics 2023; 24:774. [PMID: 38097926 PMCID: PMC10720251 DOI: 10.1186/s12864-023-09878-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 12/06/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Atherosclerosis (AS) is a critical pathological event during the progression of cardiovascular diseases. It exhibits fibrofatty lesions on the arterial wall and lacks effective treatment. N6-methyladenosine (m6A) is the most common modification of eukaryotic RNA and plays an important role in regulating the development and progression of cardiovascular diseases. However, the role of m6A modification in AS remains largely unknown. Therefore, in this study, we explored the transcriptome distribution of m6A modification in AS and its potential mechanism. METHODS Methylation Quantification Kit was used to detect the global m6A levels in the aorta of AS mice. Western blot was used to analyze the protein level of methyltransferases. Methylated RNA immunoprecipitation with next-generation sequencing (MeRIP-seq) and RNA sequencing (RNA-seq) were used to obtain the first transcriptome range analysis of the m6A methylene map in the aorta of AS mice, followed by bioinformatics analysis. qRT-PCR and MeRIP-qRT-PCR were used to measure the mRNA and m6A levels in target genes. RESULTS The global m6A and protein levels of methyltransferase METTL3 were significantly increased in the aorta of AS mice. However, the protein level of demethylase ALKBH5 was significantly decreased. Through MeRIP-seq, we obtained m6A methylation maps in AS and control mice. In total, 26,918 m6A peaks associated with 13,744 genes were detected in AS group, whereas 26,157 m6A peaks associated with 13,283 genes were detected in the control group. Peaks mainly appeared in the coding sequence (CDS) regions close to the stop codon with the RRACH motif. Moreover, functional enrichment analysis demonstrated that m6A-containing genes were significantly enriched in AS-relevant pathways. Interestingly, a negative correlation between m6A methylation abundance and gene expression level was found through the integrated analysis of MeRIP-seq and RNA-seq data. Among the m6A-modified genes, a hypo-methylated but up-regulated (hypo-up) gene Fabp5 may be a potential biomarker of AS. CONCLUSIONS Our study provides transcriptome-wide m6A methylation for the first time to determine the association between m6A modification and AS progression. Our study lays a foundation for further exploring the pathogenesis of AS and provides a new direction for the treatment of AS.
Collapse
Affiliation(s)
- Xinbin Zheng
- Clinical Research Center, Hainan Provincial Hospital of Traditional Chinese Medicine, 570203, Haikou, Hainan, P. R. China
- Hainan Clinical Research Center for Preventive Treatment of Diseases, 570203, Haikou, Hainan, P. R. China
- Hospital of Chinese Medicine affiliated by Hainan Medical University, 570203, Haikou, Hainan, P. R. China
| | - Bo Zhou
- Department of Cardiology, Hainan Provincial Hospital of Traditional Chinese Medicine, 570203, Haikou, Hainan, P. R. China
- Hospital of Chinese Medicine affiliated by Hainan Medical University, 570203, Haikou, Hainan, P. R. China
| | - Yuzhen Li
- Clinical Research Center, Hainan Provincial Hospital of Traditional Chinese Medicine, 570203, Haikou, Hainan, P. R. China
- Hospital of Chinese Medicine affiliated by Hainan Medical University, 570203, Haikou, Hainan, P. R. China
| | - Hengren Zhong
- Clinical Research Center, Hainan Provincial Hospital of Traditional Chinese Medicine, 570203, Haikou, Hainan, P. R. China
- Hospital of Chinese Medicine affiliated by Hainan Medical University, 570203, Haikou, Hainan, P. R. China
| | - Zhengxin Huang
- Department of Cardiology, Hainan Provincial Hospital of Traditional Chinese Medicine, 570203, Haikou, Hainan, P. R. China.
- Hospital of Chinese Medicine affiliated by Hainan Medical University, 570203, Haikou, Hainan, P. R. China.
| | - Minhua Gu
- Department of Cardiology, Hainan Provincial Hospital of Traditional Chinese Medicine, 570203, Haikou, Hainan, P. R. China.
- Hospital of Chinese Medicine affiliated by Hainan Medical University, 570203, Haikou, Hainan, P. R. China.
| |
Collapse
|
40
|
Feng M, Zhou X, Hu Y, Zhang J, Yang T, Chen Z, Yuan W. Comprehensive Transcriptomic Profiling of m6A Modification in Age-Related Hearing Loss. Biomolecules 2023; 13:1537. [PMID: 37892219 PMCID: PMC10605720 DOI: 10.3390/biom13101537] [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: 08/09/2023] [Revised: 09/27/2023] [Accepted: 10/07/2023] [Indexed: 10/29/2023] Open
Abstract
Age-related hearing loss (ARHL), also known as presbycusis, is one of the most common neurodegenerative disorders in elderly individuals and has a prevalence of approximately 70-80% among individuals aged 65 and older. As ARHL is an intricate and multifactorial disease, the exact pathogenesis of ARHL is not fully understood. There is evidence that transcriptional dysregulation mediated by epigenetic modifications is widespread in ARHL. However, the potential role of N6-methyladenosine (m6A) modification, as a crucial component of epigenetics, in ARHL progression remains unclear. In this study, we confirmed that the downregulation of m6A modification in cochlear tissues is related to ARHL and found that the expression of the m6A methylation regulators Wilms tumour suppressor-1-associated protein (WTAP), methyltransferase-like 3 (METTL3), ALKB homologous protein 5 (ALKBH5) and fat mass and obesity-associated protein (FTO) is decreased significantly at the mRNA and protein levels in ARHL mice. Then, we used methylated RNA immunoprecipitation sequencing (MeRIP-Seq) and RNA sequencing (RNA-Seq) to identify the differentially m6A-methylated genes in the cochlear tissues of ARHL mice. A total of 3438 genes with differential m6A methylation were identified, of which 1332 genes were m6A-hypermethylated and 2106 genes were m6A-hypomethylated in the ARHL group compared to the control group according to MeRIP-seq. Further joint analysis of RNA-Seq and MeRIP-Seq data showed that 262 genes had significant differences in both mRNA expression and m6A methylation. GO and KEGG analyses indicated that 262 unique genes were enriched mainly in the PI3K-AKT signalling pathway. In conclusion, the results of this study reveal differential m6A methylation patterns in the cochlear tissues of ARHL mice, providing a theoretical basis for further study of the pathogenesis of ARHL and potential therapeutic strategies.
Collapse
Affiliation(s)
- Menglong Feng
- Chongqing Medical University, Chongqing 400016, China
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing 400714, China
- Chongqing School, University of Chinese Academy of Sciences, Chongqing 400714, China
- Department of Otolaryngology & Head and Neck, Chongqing General Hospital, Chongqing 401147, China
| | - Xiaoqing Zhou
- Department of Otolaryngology & Head and Neck, Chongqing General Hospital, Chongqing 401147, China
| | - Yaqin Hu
- Department of Otolaryngology & Head and Neck, Chongqing General Hospital, Chongqing 401147, China
| | - Juhong Zhang
- Department of Otolaryngology & Head and Neck, Chongqing General Hospital, Chongqing 401147, China
| | - Ting Yang
- Chongqing Medical University, Chongqing 400016, China
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing 400714, China
- Chongqing School, University of Chinese Academy of Sciences, Chongqing 400714, China
- Department of Otolaryngology & Head and Neck, Chongqing General Hospital, Chongqing 401147, China
| | - Zhiji Chen
- Department of Otolaryngology & Head and Neck, Chongqing General Hospital, Chongqing 401147, China
| | - Wei Yuan
- Chongqing Medical University, Chongqing 400016, China
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing 400714, China
- Chongqing School, University of Chinese Academy of Sciences, Chongqing 400714, China
- Department of Otolaryngology & Head and Neck, Chongqing General Hospital, Chongqing 401147, China
| |
Collapse
|
41
|
Liu H, Zhang J, Yan X, An D, Lei H. The Anti-atherosclerosis Mechanism of Ziziphora clinopodioides Lam. Based On Network Pharmacology. Cell Biochem Biophys 2023; 81:515-532. [PMID: 37523140 DOI: 10.1007/s12013-023-01151-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/11/2023] [Indexed: 08/01/2023]
Abstract
We investigated the mechanisms underlying the effects of Ziziphora clinopodioides Lam. (ZCL) on atherosclerosis (AS) using network pharmacology and in vitro validation.We collected the active components of ZCL and predicted their targets in AS. We constructed the protein-protein interaction, compound-target, and target-compound-pathway networks, and performed GO and KEGG analyses. Molecular docking of the active components and key targets was constructed with Autodock and Pymol software. Validation was performed with qRT-PCR, ELISA, and Western blot.We obtained 80 components of ZCL. The network analysis identified that 14 components and 37 genes were involved in AS. Then, 10 key nodes in the PPI network were identified as the key targets of ZCL because of their importance in network topology. The binding energy of 8 components (Cynaroside, α-Spinasterol, Linarin, Kaempferide, Acacetin, Genkwanin, Chrysin, and Apiin) to 4 targets (MMP9, TP53, AKT1, SRC) was strong and <-1 kJ/mol. In addition, 13 of the 14 components were flavonoids and thus total flavonoids of Ziziphora clinopodioides Lam. (ZCF) were used for in vitro validation. We found that ZCF reduced eNOS, P22phox, gp91phox, and PCSK9 at mRNA and protein levels, as well as the levels of IL-1β, TNF-α, and IL-6 proteins in vitro (P < 0.05).We successfully predicted the active components, targets, and mechanisms of ZCL in treating AS using network pharmacology. We confirmed that ZCF may play a role in AS by modulating oxidative stress, lipid metabolism, and inflammatory response via Cynaroside, Linarin, Kaempferide, Acacetin, Genkwanin, Chrysin, and Apiin.
Collapse
Affiliation(s)
- Hongbing Liu
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, 102488, Beijing, China
- College of Traditional Chinese Medicine, Xinjiang Medical University, 830011, Urumqi, China
- Xinjiang Key Laboratory of Famous Prescription and Science of Formulas, 830011, Urumqi, China
| | - Jianxin Zhang
- College of Traditional Chinese Medicine, Xinjiang Medical University, 830011, Urumqi, China
- Xinjiang Key Laboratory of Famous Prescription and Science of Formulas, 830011, Urumqi, China
| | - Xuehua Yan
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, 102488, Beijing, China
- College of Traditional Chinese Medicine, Xinjiang Medical University, 830011, Urumqi, China
- Xinjiang Key Laboratory of Famous Prescription and Science of Formulas, 830011, Urumqi, China
| | - Dongqing An
- College of Traditional Chinese Medicine, Xinjiang Medical University, 830011, Urumqi, China.
- Xinjiang Key Laboratory of Famous Prescription and Science of Formulas, 830011, Urumqi, China.
| | - Haimin Lei
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, 102488, Beijing, China.
| |
Collapse
|
42
|
Song B, Zeng Y, Cao Y, Zhang J, Xu C, Pan Y, Zhao X, Liu J. Emerging role of METTL3 in inflammatory diseases: mechanisms and therapeutic applications. Front Immunol 2023; 14:1221609. [PMID: 37671161 PMCID: PMC10475571 DOI: 10.3389/fimmu.2023.1221609] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 07/27/2023] [Indexed: 09/07/2023] Open
Abstract
Despite improvements in modern medical therapies, inflammatory diseases, such as atherosclerosis, diabetes, non-alcoholic fatty liver, chronic kidney diseases, and autoimmune diseases have high incidence rates, still threaten human health, and represent a huge financial burden. N6-methyladenosine (m6A) modification of RNA contributes to the pathogenesis of various diseases. As the most widely discussed m6A methyltransferase, the pathogenic role of METTL3 in inflammatory diseases has become a research hotspot, but there has been no comprehensive review of the topic. Here, we summarize the expression changes, modified target genes, and pathogenesis related to METTL3 in cardiovascular, metabolic, degenerative, immune, and infectious diseases, as well as tumors. In addition to epithelial cells, endothelial cells, and fibroblasts, METTL3 also regulates the function of inflammation-related immune cells, including macrophages, neutrophils, dendritic cells, Th17 cells, and NK cells. Regarding therapeutic applications, METTL3 serves as a target for the treatment of inflammatory diseases with natural plant drug components, such as emodin, cinnamaldehyde, total flavonoids of Abelmoschus manihot, and resveratrol. This review focuses on recent advances in the initiation, development, and therapeutic application of METTL3 in inflammatory diseases. Knowledge of the specific regulatory mechanisms involving METTL3 can help to deepen understanding of inflammatory diseases and lay the foundation for the development of precisely targeted drugs to address inflammatory processes.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Jingbo Liu
- Department of Periodontics, School of Stomatology, China Medical University, Shenyang, China
| |
Collapse
|
43
|
Gao P, Yao F, Pang J, Yin K, Zhu X. m 6A methylation in cellular senescence of age-associated diseases. Acta Biochim Biophys Sin (Shanghai) 2023; 55:1168-1183. [PMID: 37394885 PMCID: PMC10449638 DOI: 10.3724/abbs.2023107] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 04/14/2023] [Indexed: 07/04/2023] Open
Abstract
Cellular senescence is a state of irreversible cellular growth arrest that occurs in response to various stresses. In addition to exiting the cell cycle, senescent cells undergo many phenotypic alterations, including metabolic reprogramming, chromatin rearrangement, and senescence-associated secretory phenotype (SASP) development. Furthermore, senescent cells can affect most physiological and pathological processes, such as physiological development; tissue homeostasis; tumour regression; and age-associated disease progression, including diabetes, atherosclerosis, Alzheimer's disease, and hypertension. Although corresponding anti-senescence therapies are actively being explored for the treatment of age-associated diseases, the specific regulatory mechanisms of senescence remain unclear. N 6-methyladenosine (m 6A), a chemical modification commonly distributed in eukaryotic RNA, plays an important role in biological processes such as translation, shearing, and RNA transcription. Numerous studies have shown that m 6A plays an important regulatory role in cellular senescence and aging-related disease. In this review, we systematically summarize the role of m 6A modifications in cellular senescence with regard to oxidative stress, DNA damage, telomere alterations, and SASP development. Additionally, diabetes, atherosclerosis, and Alzheimer's disease regulation via m 6A-mediated cellular senescence is discussed. We further discuss the challenges and prospects of m 6A in cellular senescence and age-associated diseases with the aim of providing rational strategies for the treatment of these age-associated diseases.
Collapse
Affiliation(s)
- Pan Gao
- Guangxi Key Laboratory of Diabetic Systems MedicineGuilin Medical UniversityGuilin541100China
| | - Feng Yao
- Guangxi Key Laboratory of Diabetic Systems MedicineGuilin Medical UniversityGuilin541100China
| | - Jin Pang
- Guangxi Key Laboratory of Diabetic Systems MedicineGuilin Medical UniversityGuilin541100China
| | - Kai Yin
- The Fifth Affiliated Hospital of Southern Medical UniversityGuangzhou510900China
| | - Xiao Zhu
- Guangxi Key Laboratory of Diabetic Systems MedicineGuilin Medical UniversityGuilin541100China
| |
Collapse
|
44
|
Chen ZB, He M, Li JYS, Shyy JYJ, Chien S. Epitranscriptional Regulation: From the Perspectives of Cardiovascular Bioengineering. Annu Rev Biomed Eng 2023; 25:157-184. [PMID: 36913673 DOI: 10.1146/annurev-bioeng-081922-021233] [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] [Indexed: 03/11/2023]
Abstract
The central dogma of gene expression involves DNA transcription to RNA and RNA translation into protein. As key intermediaries and modifiers, RNAs undergo various forms of modifications such as methylation, pseudouridylation, deamination, and hydroxylation. These modifications, termed epitranscriptional regulations, lead to functional changes in RNAs. Recent studies have demonstrated crucial roles for RNA modifications in gene translation, DNA damage response, and cell fate regulation. Epitranscriptional modifications play an essential role in development, mechanosensing, atherogenesis, and regeneration in the cardiovascular (CV) system, and their elucidation is critically important to understanding the molecular mechanisms underlying CV physiology and pathophysiology. This review aims at providing biomedical engineers with an overview of the epitranscriptome landscape, related key concepts, recent findings in epitranscriptional regulations, and tools for epitranscriptome analysis. The potential applications of this important field in biomedical engineering research are discussed.
Collapse
Affiliation(s)
- Zhen Bouman Chen
- Department of Diabetes Complications and Metabolism, Beckman Research Institute, City of Hope, Duarte, California, USA
| | - Ming He
- Department of Medicine, University of California, San Diego, La Jolla, California, USA;
| | - Julie Yi-Shuan Li
- Department of Bioengineering and Institute of Engineering in Medicine, University of California, San Diego, La Jolla, California, USA;
| | - John Y-J Shyy
- Department of Medicine, University of California, San Diego, La Jolla, California, USA;
| | - Shu Chien
- Department of Medicine, University of California, San Diego, La Jolla, California, USA;
- Department of Bioengineering and Institute of Engineering in Medicine, University of California, San Diego, La Jolla, California, USA;
| |
Collapse
|
45
|
Fang Z, Zhang S, Luo H, Jiang D, Huo B, Zhong X, Feng X, Cheng W, Chen Y, Feng G, Wu X, Zhao F, Yi X. Methyltransferase-like 3 suppresses phenotypic switching of vascular smooth muscle cells by activating autophagosome formation. Cell Prolif 2023; 56:e13386. [PMID: 36564367 PMCID: PMC10068948 DOI: 10.1111/cpr.13386] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 11/20/2022] [Accepted: 12/06/2022] [Indexed: 12/25/2022] Open
Abstract
Prevention of neointima formation is the key to improving long-term outcomes after stenting or coronary artery bypass grafting. RNA N6 -methyladenosine (m6 A) methylation has been reported to be involved in the development of various cardiovascular diseases, but whether it has a regulatory effect on neointima formation is unknown. Herein, we revealed that methyltransferase-like 3 (METTL3), the major methyltransferase of m6 A methylation, was downregulated during vascular smooth muscle cell (VSMC) proliferation and neointima formation. Knockdown of METTL3 facilitated, while overexpression of METTL3 suppressed the proliferation of human aortic smooth muscle cells (HASMCs) by arresting HASMCs at G2/M checkpoint and the phosphorylation of CDC2 (p-CDC2) was inactivated by METTL3. On the other hand, the migration and synthetic phenotype of HASMCs were enhanced by METTL3 knockdown, but inhibited by METTL3 overexpression. The protein levels of matrix metalloproteinase 2 (MMP2), MMP7 and MMP9 were reduced, while the expression level of tissue inhibitor of metalloproteinase 3 was increased in HASMCs with METTL3 overexpression. Moreover, METTL3 promoted the autophagosome formation by upregulating the expression of ATG5 (autophagy-related 5) and ATG7. Knockdown of either ATG5 or ATG7 largely reversed the regulatory effects of METTL3 overexpression on phenotypic switching of HASMCs, as evidenced by increased proliferation and migration, and predisposed to synthetic phenotype. These results indicate that METTL3 inhibits the phenotypic switching of VSMCs by positively regulating ATG5-mediated and ATG7-mediated autophagosome formation. Thus, enhancing the level of RNA m6 A or the formation of autophagosomes is the promising strategy to delay neointima formation.
Collapse
Affiliation(s)
- Ze‐Min Fang
- Division of Cardiothoracic and Vascular SurgerySino‐Swiss Heart‐Lung Transplantation Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiChina
| | - Shu‐Min Zhang
- Cardiac Rehabilitation CenterFuwai Hospital CAMS&PUMCBeijingChina
| | - Hanshen Luo
- Division of Cardiothoracic and Vascular SurgerySino‐Swiss Heart‐Lung Transplantation Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiChina
| | - Ding‐Sheng Jiang
- Division of Cardiothoracic and Vascular SurgerySino‐Swiss Heart‐Lung Transplantation Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiChina
- Key Laboratory of Organ Transplantation, Ministry of Education; NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ TransplantationChinese Academy of Medical SciencesWuhanHubeiChina
| | - Bo Huo
- Division of Cardiothoracic and Vascular SurgerySino‐Swiss Heart‐Lung Transplantation Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiChina
| | - Xiaoxuan Zhong
- Division of Cardiothoracic and Vascular SurgerySino‐Swiss Heart‐Lung Transplantation Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiChina
| | - Xin Feng
- Division of Cardiothoracic and Vascular SurgerySino‐Swiss Heart‐Lung Transplantation Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiChina
| | - Wenlin Cheng
- Department of CardiologyZhongnan Hospital of Wuhan UniversityWuhanHubeiChina
- Institute of Myocardial Injury and RepairWuhan UniversityWuhanChina
| | - Yue Chen
- Division of Cardiothoracic and Vascular SurgerySino‐Swiss Heart‐Lung Transplantation Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiChina
| | - Gaoke Feng
- Department of CardiologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Xingliang Wu
- Department of CardiologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Fang Zhao
- Department of CardiologyZhongnan Hospital of Wuhan UniversityWuhanHubeiChina
- Institute of Myocardial Injury and RepairWuhan UniversityWuhanChina
| | - Xin Yi
- Department of CardiologyRenmin Hospital of Wuhan UniversityWuhanChina
| |
Collapse
|
46
|
Liang A, Liu J, Wei Y, Liao Y, Wu F, Ruan J, Li J. m 6A reader IGF2BP1 accelerates apoptosis of high glucose-induced vascular endothelial cells in a m 6A-HMGB1 dependent manner. PeerJ 2023; 11:e14954. [PMID: 37009154 PMCID: PMC10062336 DOI: 10.7717/peerj.14954] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 02/03/2023] [Indexed: 03/29/2023] Open
Abstract
Emerging evidence indicates that N6-methyladenosine (m6A) plays a critical role in vascular biological characteristic. In diabetes mellitus pathophysiology, high glucose (HG)-induced vascular endothelial dysfunction is associated with diabetes vascular complications. Nevertheless, the underlying mechanism of high glucose (HG)-related m6A regulation on vascular endothelial cells is still unclear. Results indicated that m6A reader insulin-like growth factor 2 mRNA-binding protein 1 (IGF2BP1) was up-regulated in HG-treated human umbilical vascular endothelium cells (HUVECs) comparing to normal group. Functionally, results indicated that IGF2BP1 knockdown recovered the proliferation of HUVECs inhibited by HG-administration. Besides, IGF2BP1 knockdown reduced the apoptosis induced by HG-administration. Mechanistically, IGF2BP1 interacted with HMGB1 mRNA and stabilized its expression of m6A-modified RNA. Therefore, these findings provided compelling evidence demonstrating that m6A reader IGF2BP1 contributes to the proliferation and apoptosis of vascular endothelial cells in hyperglycaemia, serving as a target for development of diabetic angiopathy therapeutics.
Collapse
Affiliation(s)
- Anru Liang
- Department of Burns and Plastic Surgery, The Third Affiliated Hospital of Guangxi Medical University and The Second People’s Hospital of Nanning, Nanning, China
| | - Jianyu Liu
- Department of Clinical Laboratory, Guiping People’s Hospital, Guigping, China
| | - Yanlin Wei
- Department of Emergency, The People’s Hospital of Guangxi Zhuang Autonomous Region & Guangxi Academy of Medical Sciences, Nanning, China
| | - Yuan Liao
- Department of Clinical Laboratory, Guiping People’s Hospital, Guigping, China
| | - Fangxiao Wu
- Department of Burns and Plastic Surgery, The Third Affiliated Hospital of Guangxi Medical University and The Second People’s Hospital of Nanning, Nanning, China
| | - Jiang Ruan
- Department of Burns and Plastic Surgery, The Third Affiliated Hospital of Guangxi Medical University and The Second People’s Hospital of Nanning, Nanning, China
| | - Junjun Li
- Research Center of Medical Sciences, The People’s Hospital of Guangxi Zhuang Autonomous Region & Guangxi Academy of Medical Sciences, Nanning, China
| |
Collapse
|
47
|
Li Z, Meng X, Chen Y, Xu X, Guo J. N 6-methyladenosine (m 6A) writer METTL3 accelerates the apoptosis of vascular endothelial cells in high glucose. Heliyon 2023; 9:e13721. [PMID: 36873555 PMCID: PMC9976308 DOI: 10.1016/j.heliyon.2023.e13721] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 02/05/2023] [Accepted: 02/08/2023] [Indexed: 02/15/2023] Open
Abstract
Recent studies have shown that N6-methyladenosine (m6A) methylation, one of the most prevalent epigenetic modifications, is involved in diabetes mellitus. However, whether m6A regulates diabetic vascular endothelium injury is still elusive. Present research aimed to investigate the regulation and mechanism of m6A on vascular endothelium injury. Upregulation of METTL3 was observed in the high glucose (HG)-induced human umbilical vein endothelial cells (HUVECs), following with the upregulation of m6A methylation level. Functionally, METTL3 silencing repressed the apoptosis and recovered the proliferation of HUVECs disposed by HG. Moreover, HG exposure upregulated the expression of suppressor of cytokine signaling3 (SOCS3). Mechanistically, METTL3 targeted the m6A site on SOCS3 mRNA, which positively regulated the mRNA stability of SOCS3. In conclusion, METTL3 silencing attenuated the HG-induced vascular endothelium cells injury via promoting SOCS3 stability. In conclusion, this research expands the understanding of m6A on vasculopathy in diabetes mellitus and provides a potential strategy for the protection of vascular endothelial injury.
Collapse
Affiliation(s)
- Zhenjin Li
- Department of Endocrinology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Xuying Meng
- Department of Endocrinology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Yu Chen
- Department of Endocrinology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Xiaona Xu
- Department of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Jianchao Guo
- Department of Endocrinology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| |
Collapse
|
48
|
Zhang G, Li X, Huang X. m6A-related bioinformatics analysis and functional characterization reveals that METTL3-mediated NPC1L1 mRNA hypermethylation facilitates progression of atherosclerosis via inactivation of the MAPK pathway. Inflamm Res 2023; 72:429-442. [PMID: 36583755 DOI: 10.1007/s00011-022-01681-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 12/10/2022] [Accepted: 12/16/2022] [Indexed: 12/31/2022] Open
Abstract
OBJECTIVE Accumulating evidence has demonstrated that N6-methyladenosine (m6A) plays important roles in many major diseases, including atherosclerosis (AS). In the present study, we aimed to explore the transcriptomic m6A landscape of endothelial function-associated genes and identify potential regulators in AS progression. METHODS The GEO data (GSE142386) from MeRIP-seq in human umbilical vein endothelial cells (HUVECs) with METTL3 knocked down or not were analyzed. RNA-seq was performed to identify differences in gene expression. Gene ontology (GO) functional and Kyoto encyclopedia of genes and genomes (KEGG) pathway analyses were conducted to evaluate the potential functions of the differentially expressed genes. MeRIP-qPCR was used to measure the m6A and mRNA levels of the top 8 downregulated genes, and NPC1L1 was selected as the candidate gene. Oxidized low-density lipoprotein (ox-LDL) was used to stimulate HUVECs, and METTL3 or NPC1L1 was silenced in ox-LDL-treated cells. And Transwell, ELISA, and cell apoptosis assays were performed to assess cell functional injury. ApoE-/- mice were fed with high-fat diet for 8 weeks to establish an AS model, and adenovirus-mediated NPC1L1 shRNA or NC shRNA was injected into the mice through the tail vein. Mouse aortic tissue damage and plaque deposition were evaluated by H&E, Oil Red O, and TUNEL staining. RESULTS One hundred and ninety-four hypermethylated m6A peaks and 222 hypomethylated peaks were detected in response to knockdown of METTL3. Genes with altered m6A peaks were significantly involved in the histone modification, enzyme activity, and formation of multiple complexes and were predominantly enriched in the MAPK pathway. NPC1L1 was a most significantly downregulated transcript in response to knockdown of METTL3. Moreover, knockdown of NPC1L1 or de-m6A (METTL3 knockdown)-mediated downregulation of NPC1L1 could improve ox-LDL-induced dysfunction of HUVECs in vitro and high-fat diet-induced atherosclerotic plaque in vivo, which was associated with the inactivation of the MAPK pathway. CONCLUSION METTL3-mediated NPC1L1 mRNA hypermethylation facilitates AS progression by regulating the MAPK pathway, and NPC1L1 may be a novel target for the treatment of AS.
Collapse
Affiliation(s)
- Guoan Zhang
- Department of Cardiovascular Surgery, Shaanxi Provincial People's Hospital, Xi'an, 710068, Shaanxi, China
| | - Xuewen Li
- Department of Cardiovascular Surgery, Shaanxi Provincial People's Hospital, Xi'an, 710068, Shaanxi, China
| | - Xiaoyan Huang
- Shaanxi Provincial Key Laboratory of Infection and Immune Diseases, Shaanxi Provincial People's Hospital, 256 West Youyi Road, Xi'an, 710068, Shaanxi, China.
| |
Collapse
|
49
|
The Epigenetic Regulation of RNA N6-Methyladenosine Methylation in Glycolipid Metabolism. Biomolecules 2023; 13:biom13020273. [PMID: 36830642 PMCID: PMC9953413 DOI: 10.3390/biom13020273] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/06/2023] [Accepted: 01/30/2023] [Indexed: 02/04/2023] Open
Abstract
The highly conserved and dynamically reversible N6-methyladenine (m6A) modification has emerged as a critical gene expression regulator by affecting RNA splicing, translation efficiency, and stability at the post-transcriptional level, which has been established to be involved in various physiological and pathological processes, including glycolipid metabolism and the development of glycolipid metabolic disease (GLMD). Hence, accumulating studies have focused on the effects and regulatory mechanisms of m6A modification on glucose metabolism, lipid metabolism, and GLMD. This review summarizes the underlying mechanism of how m6A modification regulates glucose and lipid metabolism-related enzymes, transcription factors, and signaling pathways and the advances of m6A regulatory mechanisms in GLMD in order to deepen the understanding of the association of m6A modification with glycolipid metabolism and GLMD.
Collapse
|
50
|
Identification of a Novel Angiogenesis Signalling circSCRG1/miR-1268b/NR4A1 Pathway in Atherosclerosis and the Regulatory Effects of TMP-PF In Vitro. Molecules 2023; 28:molecules28031271. [PMID: 36770940 PMCID: PMC9919304 DOI: 10.3390/molecules28031271] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 01/14/2023] [Accepted: 01/25/2023] [Indexed: 01/31/2023] Open
Abstract
Angiogenesis contributes to plaque instability in atherosclerosis and further increases cardio-cerebrovascular risk. Circular RNAs (circRNAs) are promising biomarkers and potential therapeutic targets for atherosclerosis. Previous studies have demonstrated that tetramethylpyrazine (TMP) and paeoniflorin (PF) combination treatment (TMP-PF) inhibited oxidized low-density lipoprotein (ox-LDL)-induced angiogenesis in vitro. However, whether circRNAs regulate angiogenesis in atherosclerosis and whether TMP-PF can regulate angiogenesis-related target circRNAs in atherosclerosis are unknown. In this study, human RNA sequencing (RNA-seq) data were analysed to identify differentially expressed (DE) circRNAs in atherosclerosis and to obtain angiogenesis-associated circRNA-microRNA (miRNA)-messenger RNA (mRNA) networks. Target circRNA-related mechanisms in angiogenesis in atherosclerosis and the regulatory effects of TMP-PF on target circRNA signalling were studied in ox-LDL-induced human umbilical vein endothelial cells (HUVECs) by cell proliferation, migration, tube formation, and luciferase reporter assays, real-time quantitative polymerase chain reaction (RT-qPCR) and Western blotting. A novel circRNA (circular stimulator of chondrogenesis 1, circSCRG1) was initially identified associated with angiogenesis in atherosclerosis, and circSCRG1 silencing up-regulated miR-1268b expression, increased nuclear receptor subfamily 4 group A member 1 (NR4A1) expression and then promoted ox-LDL-induced angiogenesis. TMP-PF (1 μmol/L TMP combined with 10 μmol/L PF) up-regulated circSCRG1 expression, mediated miR-1268b to suppress NR4A1 expression and then inhibited ox-LDL-induced angiogenesis. However, circSCRG1 silencing abolished the inhibitory effects of TMP-PF on ox-LDL-induced angiogenesis, which were rescued by the miR-1268b inhibitor. In conclusion, circSCRG1 might serve as a new target regulating angiogenesis in atherosclerosis via the circSCRG1/miR-1268b/NR4A1 axis and TMP-PF could regulate the circSCRG1/miR-1268b/NR4A1 axis to inhibit angiogenesis in atherosclerosis in vitro, indicating a novel angiogenesis signalling circSCRG1/miR-1268b/NR4A1 pathway in atherosclerosis and the regulatory effects of TMP-PF, which might provide a new pharmaceutical strategy to combat atherosclerotic plaque instability.
Collapse
|