1
|
Li LN, Wu JM, Zheng ZJ, Li SX, Cai MY, Zou MC. N6-methyladenosine modification of THBS1 induced by affluent WTAP promotes Graves' ophthalmopathy progression through glycolysis to affect Th17/Treg balance. Autoimmunity 2025; 58:2433628. [PMID: 39689341 DOI: 10.1080/08916934.2024.2433628] [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: 07/26/2024] [Revised: 10/15/2024] [Accepted: 11/17/2024] [Indexed: 12/19/2024]
Abstract
Graves' ophthalmopathy (GO) obvious manifestation is the imbalance of Th17/Treg. N6-methyladenosine (m6A) methylation is an important regulator of Th17/Treg balance. However, few reports narrate how m6A regulators mediate the role of genes in GO progression. We explored the m6A modification of THBS1 mediated by WTAP, and the mechanism by which THBS1 regulated glycolysis and Th17/Treg balance. A total of 12 peripheral blood (4 GO samples, 4 GH samples, and 4 health samples) were collected to measure the percentage of Th17/Treg in monocytes by flow cytometry. RNA sequencing (RNA-seq) combined with MeRIP sequencing (MeRIP-seq) was used to screen differentially expressed and methylated genes. MeRIP-qPCR was performed to evaluate the m6A abundance of THBS1 after WTAP silencing. Glycolysis of CD4+ T cells was reflected by the lactate content and glucose uptake. The number of Th17 cells was increased in GO peripheral blood, whereas the Treg cells decreased. RNA-seq acquired 679 differentially expressed genes (308 up-regulated, and 371 down-regulated) in the CD4+ T cells of GO compared to healthy control. MeRIP-seq identified 3277 m6A peaks between the GO group and the healthy control group, corresponding with 2744 genes (1143 hypermethylated and 1601 hypomethylated). Combined analysis of RNA-seq and MeRIP-seq showed 81 hypermethylated and up-regulated genes. Among the six candidate genes in the PI3K-signaling pathway, THBS1 was the most significantly differentially expressed and hypermethylated. THBS1 silencing resulted in decreased lactate content and glucose uptake in CD4+ T cells. WTAP was significantly upregulated in CD4+ T cells of GO, and WTAP silencing significantly reduced m6A abundance and expression of THBS1. Upregulated and hypermethylated THBS1 mediated by WTAP promoted glycolysis of CD4+ T cells, affected Th17/Treg balance, and facilitated GO progression. We provided a novel potential target for GO treatment and revealed the molecular mechanism of WTAP and THBS1 in GO under the m6A perspective.
Collapse
Affiliation(s)
- Lin-Na Li
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jie-Man Wu
- Department of Health Management, Nanfang Hospital Zengcheng Campus, Guangzhou, China
| | - Zong-Ji Zheng
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Shu-Xian Li
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Meng-Yi Cai
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Meng-Chen Zou
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
2
|
Yang Y, Zhang Q, Liu S, Yuan H, Wu X, Zou Y, Zhang Y, Guo J. Suv39h1 Regulates Phenotypic Modulation of Smooth Muscle Cells and Contributes to Vascular Injury by Repressing HIC1 Transcription. Arterioscler Thromb Vasc Biol 2025; 45:965-978. [PMID: 40308197 PMCID: PMC12094260 DOI: 10.1161/atvbaha.124.322048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 04/10/2025] [Indexed: 05/02/2025]
Abstract
BACKGROUND Vascular smooth muscle cells (VSMCs), in response to a myriad of injurious stimuli, switch from a contractile state to a proliferative/migratory state in a process known as phenotypic modulation. Phenotypic modulation of VSMCs contributes to neointima formation and underscores a host of vascular pathologies, including atherosclerosis. In the present study, we investigated the involvement of Suv39h1 (suppressor of variegation 3-9 homolog 1), a lysine methyltransferase, in this process. METHODS Suv39h1f/f mice were crossbred to the Myh11-CreERT2 mice to generate VSMC-restricted Suv39h1 knockout mice (conditional knockout). Vascular injury was created by carotid artery ligation. Cellular transcriptome was evaluated by RNA sequencing and cleavage under targets and tagmentation with deep sequencing. RESULTS Suv39h1 upregulation was observed in animal and cell models of phenotypic modulation. Consistently, Suv39h1 silencing restored expression of contractile genes and attenuated proliferation/migration in VSMCs exposed to PDGF (platelet-derived growth factor)-BB. Importantly, Suv39h1 deletion significantly ameliorated neointima formation in mice in both the carotid artery injury model and the femoral artery injury model. Importantly, a small-molecule Suv39h1 inhibitor F5446 suppressed phenotypic modulation in vitro and mitigated vascular injury in mice. RNA sequencing identified HIC1 (hypermethylated in cancer 1) as a novel target for Suv39h1. HIC1 expression was repressed by Suv39h1 during VSMC phenotypic modulation, whereas HIC1 overexpression antagonized neointima formation in mice. Integrated transcriptomic analysis indicated that HIC1 might regulate VSMC phenotypic modulation by activating Jag1 (Jagged 1) transcription. CONCLUSIONS Our data suggest that Suv39h1 is a novel regulator of vascular injury and can be targeted for intervention of restenosis.
Collapse
MESH Headings
- Animals
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/enzymology
- Muscle, Smooth, Vascular/drug effects
- Phenotype
- Methyltransferases/genetics
- Methyltransferases/metabolism
- Methyltransferases/deficiency
- Kruppel-Like Transcription Factors/genetics
- Kruppel-Like Transcription Factors/metabolism
- Myocytes, Smooth Muscle/pathology
- Myocytes, Smooth Muscle/enzymology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Mice, Knockout
- Carotid Artery Injuries/genetics
- Carotid Artery Injuries/pathology
- Carotid Artery Injuries/enzymology
- Disease Models, Animal
- Cell Proliferation
- Neointima
- Cell Movement
- Cells, Cultured
- Transcription, Genetic
- Male
- Repressor Proteins/genetics
- Repressor Proteins/metabolism
- Repressor Proteins/deficiency
- Mice
- Mice, Inbred C57BL
- Signal Transduction
Collapse
Affiliation(s)
- Yuyu Yang
- Jiangsu Key Laboratory of Medical Biotechnology, College of Life Sciences, Nanjing Normal University, China (Y.Y., H.Y.)
| | - Qiumei Zhang
- State Key Laboratory of Natural Medicines, Department of Pharmacology, China Pharmaceutical University, Nanjing (Q.Z., Y. Zou)
| | - Shuai Liu
- Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, Medical Research Center of the First Affiliated Hospital, Hainan Women and Children Medical Center, Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Medical University, Haikou, China (S.L., X.W., Y. Zhang)
| | - Haihang Yuan
- Jiangsu Key Laboratory of Medical Biotechnology, College of Life Sciences, Nanjing Normal University, China (Y.Y., H.Y.)
| | - Xiaoping Wu
- Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, Medical Research Center of the First Affiliated Hospital, Hainan Women and Children Medical Center, Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Medical University, Haikou, China (S.L., X.W., Y. Zhang)
| | - Yi Zou
- State Key Laboratory of Natural Medicines, Department of Pharmacology, China Pharmaceutical University, Nanjing (Q.Z., Y. Zou)
| | - Yuanyuan Zhang
- Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, Medical Research Center of the First Affiliated Hospital, Hainan Women and Children Medical Center, Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Medical University, Haikou, China (S.L., X.W., Y. Zhang)
| | - Junli Guo
- Key Laboratory of Tropical Translational Medicine of Ministry of Education & Hainan Provincial Key Laboratory for Tropical Cardiovascular Diseases Research, School of Public Health, Hainan Medical University, Haikou, China (J.G.)
| |
Collapse
|
3
|
Zhou C, Zhang Y, Shi SM, Yin D, Li XD, Shi YH, Zhou J, Wang Z, Chen Q. FTO downregulation-mediated m6A modification resulting in enhanced hepatocellular carcinoma invasion. Cell Biosci 2025; 15:58. [PMID: 40316995 PMCID: PMC12049069 DOI: 10.1186/s13578-025-01395-w] [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: 07/12/2024] [Accepted: 12/09/2024] [Indexed: 05/04/2025] Open
Abstract
BACKGROUND Dysregulation of N6-methyladenosine (m6A) modifications has been implicated in various cancers, including hepatocellular carcinoma (HCC). This study aimed to elucidate the role of m6A modifications in HCC prognosis and the molecular mechanisms involved, particularly focusing on the demethylase FTO. METHODS We analyzed m6A expression in a cohort of 323 HCC patients using immunohistochemical (IHC) staining. The expression of m6A-related genes (FTO, ALKBH5, METTL3, METTL14) was evaluated by qRT-PCR in 120 paired HCC tissues. Further, we established HCC cell lines with altered FTO expression to assess its impact on cell proliferation, invasion, and metastasis through various in vitro assays and in vivo orthotopic HCC mouse models. Statistical analyses included Pearson chi-square test, Kaplan-Meier survival analysis, and both univariate and multivariate Cox regression analyses. RESULTS IHC staining revealed elevated m6A levels in HCC tissues compared to adjacent non-tumorous tissues, with 57.3% of HCC patients showing increased m6A expression. High m6A levels were correlated with poorer overall survival (OS) and recurrence-free survival (RFS) rates. FTO, a demethylase, was significantly downregulated in HCC tissues and cell lines, particularly in highly metastatic lines. Overexpression of FTO in HCC cells reduced proliferation, migration, and invasion, whereas FTO knockdown had the opposite effect. In vivo, FTO overexpression decreased tumor growth and metastasis. RNA-Seq analysis identified VEGFA as a key gene downregulated by FTO, implicating its role in angiogenesis and tumor progression. CONCLUSIONS Our findings suggest that elevated m6A levels are associated with poor prognosis in HCC patients. FTO downregulation contributes to aberrant m6A modifications, promoting HCC progression and metastasis. FTO acts as a tumor suppressor by negatively regulating VEGFA expression, highlighting its potential as a therapeutic target for HCC treatment. These results highlight the significance of m6A modifications in HCC and provide a foundation for future research on targeted therapies.
Collapse
Affiliation(s)
- Cheng Zhou
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai, 200032, China
| | - Yong Zhang
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Shi-Ming Shi
- Department of Vascular, Thyroid, and Breast Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong Province, 524000, China
| | - Dan Yin
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai, 200032, China
- Institute of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Xue-Dong Li
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai, 200032, China
| | - Ying-Hong Shi
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai, 200032, China
| | - Jian Zhou
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai, 200032, China
- Institute of Biomedical Sciences, Fudan University, Shanghai, 200032, China
- State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, 200032, China
| | - Zheng Wang
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai, 200032, China.
| | - Qing Chen
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai, 200032, China.
- Department of Liver Oncology, Zhongshan Hospital (Minhang Meilong), Fudan University (Shanghai Geriatric Medical Center), Shanghai, 201104, China.
| |
Collapse
|
4
|
Weng J, Shan Y, Chang Q, Cao C, Liu X. Research progress on N 6-Methyladenosine modification in angiogenesis, vasculogenic mimicry, and therapeutic implications in breast cancer. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2025; 195:57-70. [PMID: 39710080 DOI: 10.1016/j.pbiomolbio.2024.12.003] [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: 10/07/2024] [Revised: 12/19/2024] [Accepted: 12/19/2024] [Indexed: 12/24/2024]
Abstract
N6-methyladenosine (m6A) modification is the most common epitranscriptomic modification in eukaryotic RNA and has garnered extensive attention in the context of breast cancer research. The m6A modification significantly impacts tumorigenesis and tumor progression by regulating RNA stability, splicing, translation, and degradation. In this review we summarize recent advances in understanding the roles of m6A modification in the mechanisms underlying angiogenesis and vasculogenic mimicry in breast cancer. We review how m6A modification and associated transcripts influence relevant factors by affecting key factors and signaling pathways, highlighting the interactions among m6A "writers," "erasers," and "readers," and their overall impact on tumor angiogenesis and vasculogenic mimicry, as well as potential new therapeutic targets.
Collapse
Affiliation(s)
- Jiachen Weng
- Translational Medical Innovation Center, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou City, Jiangsu, 215600, China
| | - Yisi Shan
- Translational Medical Innovation Center, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou City, Jiangsu, 215600, China
| | - Qingyu Chang
- Translational Medical Innovation Center, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou City, Jiangsu, 215600, China
| | - Chenyan Cao
- Translational Medical Innovation Center, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou City, Jiangsu, 215600, China
| | - Xuemin Liu
- Translational Medical Innovation Center, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou City, Jiangsu, 215600, China.
| |
Collapse
|
5
|
Wang H, Xu P, Yin K, Wang S. The role of m 6A modification during macrophage metabolic reprogramming in human diseases and animal models. Front Immunol 2025; 16:1521196. [PMID: 40066451 PMCID: PMC11891544 DOI: 10.3389/fimmu.2025.1521196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 01/28/2025] [Indexed: 05/13/2025] Open
Abstract
Macrophage metabolic reprogramming refers to the process by which macrophages adjust their physiological pathways to meet survival and functional demands in different immune microenvironments. This involves a range of metabolic pathways, including glycolysis, the tricarboxylic acid cycle, oxidative phosphorylation, fatty acid oxidation, and cholesterol transport. By modulating the expression and activity of key enzymes and molecules within these pathways, macrophages can make the transition between pro- and anti-inflammatory phenotypes, thereby linking metabolic reprogramming to inflammatory responses and the progression of several diseases, such as atherosclerosis, inflammatory bowel disease (IBD), and acute lung injury (ALI). N6-methyladenosine (m6A) modification has emerged as a critical regulatory mechanism during macrophage metabolic reprogramming, broadly affecting RNA stability, translation, and degradation. Therapeutic strategies targeting m6A modification can regulate the onset of metabolic diseases by influencing macrophage metabolic changes, for instance, small molecule inhibitors of methyltransferase-like 3 (METTL3) can affect glucose metabolism and inhibit IBD. This review systematically explores recent findings on the role and molecular mechanisms of m6A modification during macrophage metabolic reprogramming in human diseases and animal models, underscoring its potential as a therapeutic target for metabolic diseases.
Collapse
Affiliation(s)
- Huiling Wang
- Department of Laboratory Medicine, Jiangsu Province Engineering Research Center for Precise Diagnosis and Treatment of Inflammatory Diseases, The Affiliated Hospital of Jiangsu University, Zhenjiang, China
- Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Peiqi Xu
- Department of Laboratory Medicine, Jiangsu Province Engineering Research Center for Precise Diagnosis and Treatment of Inflammatory Diseases, The Affiliated Hospital of Jiangsu University, Zhenjiang, China
- Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Kai Yin
- Department of General Surgery, Affiliated Hospital of Jiangsu University, Institute of Digestive Diseases, Jiangsu University, Zhenjiang, China
| | - Shengjun Wang
- Department of Laboratory Medicine, Jiangsu Province Engineering Research Center for Precise Diagnosis and Treatment of Inflammatory Diseases, The Affiliated Hospital of Jiangsu University, Zhenjiang, China
- Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| |
Collapse
|
6
|
Zhang Z, Zhao R, Wu X, Ma Y, He Y. Research progress on the correlation between corneal neovascularization and lymphangiogenesis (Review). Mol Med Rep 2025; 31:47. [PMID: 39635819 PMCID: PMC11638739 DOI: 10.3892/mmr.2024.13412] [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: 07/30/2024] [Accepted: 10/23/2024] [Indexed: 12/07/2024] Open
Abstract
The cornea is a clear connective tissue membrane at the front of the outer layer of the eyeball wall. It plays a crucial role in the refractive system of the eyeball, making it essential to maintain its transparency. Neovascularization and lymphangiogenesis in the cornea significantly impact corneal transparency and immune privilege. The growth of corneal neovascularization (CNV) and corneal lymphangiogenesis (CL) vessels is interconnected yet independent. Currently, there is a substantial amount of clinical and experimental research on CNV and CL vessels. However, due to the relatively recent focus on CL vessel research compared with CNV research, most scholars tend to concentrate on CNV, with few articles offering a comprehensive comparison and discussion of the two processes. The present review emphasizes the similarities and differences between CNV and CL and summarizes recent research progress on their correlation in animal models, growth characteristics, cytokine effects and related diseases.
Collapse
Affiliation(s)
- Zhaochen Zhang
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| | - Rongxuan Zhao
- Department of Rehabilitation, School of Nursing, Jilin University, Changchun, Jilin 130041, P.R. China
| | - Xuhui Wu
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| | - Yunkun Ma
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| | - Yuxi He
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| |
Collapse
|
7
|
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
|
8
|
Gong Q, Hu L, Liu G, Yin X, Zhao X, Li Q, Li Y, Sun Y, Zhou Y, Guo C, Du Z. WTAP-mediated N6-methyladenosine mRNA methylation regulates laser-induced macular neovascularization. Mol Vis 2024; 30:336-347. [PMID: 40330498 PMCID: PMC12055036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 10/05/2024] [Indexed: 05/08/2025] Open
Abstract
Purpose Neovascular age-related macular degeneration (nAMD) is now a major cause of central vision loss in older adults worldwide. The primary characteristic of nAMD is the formation of macular neovascularization (MNV), which is a pathologic form of angiogenesis. Epigenetics plays a role in multiple pathological physiologic processes. N6-methyladenosine (m6A) modification is the most common, abundant, and reversible modification in eukaryotic mRNAs, and it plays a role in various pathological angiogenesis processes. This study intends to reveal the expression and functions of m6A during the macular neovascularization (MNV) process. Methods A laser-induced MNV mouse model was used in this study. m6A quantitative analysis was performed to detect the expression of m6A. Subsequently, the expression of various m6A writers and erasers was detected using quantitative real-time polymerase chain reaction (qRT-PCR) and western blot. Immunohistochemistry was used to detect Wilms' tumor 1-associating protein (WTAP) expression in the MNV lesions. Intravitreal injection of WTAP siRNA in MNV mice to silence the WTAP gene. Hematoxylin and eosin (H&E) were used to determine the thickness and length of the MNV. Fundus fluorescein angiography (FFA) and indocyanine green angiography (ICGA) were examined to measure the leakage area of the MNV. Proliferating cell nuclear antigen (PCNA) expression was detected with a western blot. The mRNA and protein levels of β-catenin were tested with qRT-PCR and western blot. Results We found increased m6A modification levels after laser induction compared with the normal control group. Subsequently, the expression of various m6A writers and erasers was detected. The results showed that WTAP increased in the MNV model in mice. After the injection of WTAP siRNA into the vitreous body, the expression of WTAP significantly decreased, subsequently decreasing the m6A modification levels. The width, breadth, and leakage area of MNV damage markedly decreased, and endothelial cell proliferation was inhibited. After laser-induced MNV, the expression of β-catenin increased, and that of β-catenin significantly decreased after WTAP knockout. Conclusions In conclusion, this study suggests that WTAP-mediated m6A methylation can regulate pathological angiogenesis during MNV and that WTAP may participate in the formation of MNV through the wingless-related integration site (Wnt) pathway. WTAP may be a potential target for MNV treatment.
Collapse
Affiliation(s)
- Qingyun Gong
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Liting Hu
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Guibo Liu
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaoni Yin
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaoran Zhao
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Qinghua Li
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ying Li
- Department of Ophthalmology, Linyi People's Hospital, Shandong, China
| | - Yibin Sun
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yuzheng Zhou
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Chunyan Guo
- Department of Ophthalmology, Gansu Province, China
| | - Zhaodong Du
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
9
|
Gao W, Miao X, Xu T. Wilms tumor 1-associated protein mediated m6A modification promotes osteogenic differentiation of stem cells from human exfoliated deciduous teeth. J Dent Sci 2024; 19:2305-2314. [PMID: 39347097 PMCID: PMC11437296 DOI: 10.1016/j.jds.2024.02.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 02/20/2024] [Indexed: 10/01/2024] Open
Affiliation(s)
- Weiheng Gao
- Department of Emergency, The Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- National Clinical Research Center for Child Health, Hangzhou, China
| | - Xixi Miao
- National Clinical Research Center for Child Health, Hangzhou, China
- Department of Respiratory Medicine, The Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Tao Xu
- Department of Stomatology, Nanjing Geriatric Hospital, Nanjing, China
| |
Collapse
|
10
|
Wang W, Li H, Qian Y, Li M, Deng M, Bi D, Zou J. ALKBH5 Regulates Corneal Neovascularization by Mediating FOXM1 M6A Demethylation. Invest Ophthalmol Vis Sci 2024; 65:34. [PMID: 39441582 PMCID: PMC11512564 DOI: 10.1167/iovs.65.12.34] [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: 08/14/2024] [Accepted: 09/30/2024] [Indexed: 10/25/2024] Open
Abstract
Purpose This study aims to explore the regulatory role and potential mechanisms of ALKBH5-mediated N6-methyladenosine (m6A) demethylation modification in corneal neovascularization (CNV). Methods A mouse CNV model was established through corneal alkali burns. Total m6A levels were measured using an m6A RNA methylation quantification kit. The mRNA expression of candidate m6A-related enzymes was quantified by quantitative RT-PCR. Small interfering RNA targeting ALKBH5 was injected subconjunctivally into alkali-burned mice. The CNV area, corneal epithelial thickness, and pathological changes were evaluated. Protein expression was detected by western blot and immunofluorescence. Human umbilical vein endothelial cells (HUVECs) were treated with IL-6. Plasmid transfection knocked down ALKBH5 or overexpressed FOXM1 in IL-6-induced HUVECs. The assays of CCK8, wound healing, and tube formation evaluated the cell proliferation, migration, and tube formation abilities, respectively. The dual-luciferase assay examined the binding between ALKBH5 and FOXM1. Methylated RNA immunoprecipitation-qPCR detected the m6A levels of FOXM1. Results Significant CNV was observed on the seventh day. Total m6A levels were reduced, and ALKBH5 expression was increased in CNV corneas and IL-6-induced HUVECs. ALKBH5 knockdown alleviated corneal neovascularization and inflammation and countered IL-6-induced promotion of cell proliferation, migration, and tube formation in HUVECs. ALKBH5 depletion increased m6A levels and decreased VEGFA and CD31 expression both in vivo and in vitro. This knockdown in HUVECs elevated m6A levels on FOXM1 mRNA while reducing its mRNA and protein expression. Notably, FOXM1 overexpression can reverse ALKBH5 depletion effects. Conclusions ALKBH5 modulates FOXM1 m6A demethylation, influencing CNV progression and highlighting its potential as a therapeutic target.
Collapse
Affiliation(s)
- Wei Wang
- Department of Ophthalmology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Hua Li
- Department of Ophthalmology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yiyong Qian
- Department of Ophthalmology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Min Li
- Department of Ophthalmology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Manli Deng
- Department of Ophthalmology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Dexi Bi
- Department of Pathology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jun Zou
- Department of Ophthalmology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
11
|
Xie X, Cheng S, Chen X, Wang X. WTAP promotes the progression of ulcerative colitis by silencing the expression of CES2 through m 6A modification. J Autoimmun 2024; 148:103295. [PMID: 39141984 DOI: 10.1016/j.jaut.2024.103295] [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: 12/05/2023] [Revised: 07/08/2024] [Accepted: 07/22/2024] [Indexed: 08/16/2024]
Abstract
OBJECTIVE This study will explore the function of WTAP, the critical segment of m6A methyltransferase complex, in UC and its regulation on immune response. METHODS The expression levels of key proteins were detected in colon tissues which were derived from UC patients and mice. Macrophage polarization and CD4+ T cell infiltration were detected by flow cytometry and IF staining. ELISA assay was utilized to analyze the level of the inflammatory cytokines. m6A-RIP-PCR, actinomycin D test, and RIP assays were utilized to detect the m6A level, stability, and bound proteins of CES2 mRNA. A dual luciferase reporter assay was conducted to confirm the transcriptional interactions between genes. A co-culture system of intestinal epithelium-like organs was constructed to detect the primary mouse intestinal epithelial cells (PMIEC) differentiation. The interaction between proteins was detected via Co-IP assay. RESULTS The expression of WTAP and CES2 in UC tissues was increased and decreased, respectively. Knockdown of WTAP inhibited the progression of UC in mice by inhibiting M1 macrophage polarization and CD4+ T cell infiltration. WTAP combined YTHDF2 to promote the m6A modification of CES2 mRNA and inhibited its expression. CES2 co-expressed with EPHX2 and overexpression of CES2 promoted the differentiation of PMIEC. The inhibitory effect of WTAP knockdown on the progress of UC was partially abrogated by CES2 knockdown. CONCLUSION WTAP/YTHDF2 silences CES2 by promoting its m6A modification and then promotes the progression of UC. WTAP could be a promoting therapy target of UC.
Collapse
Affiliation(s)
- Xiaoran Xie
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, Shandong, China; Laboratory of Translational Gastroenterology, Qilu Hospital of Shandong University, Jinan, Shandong, China; Robot Engineering Laboratory for Precise Diagnosis and Therapy of GI Tumor, Qilu Hospital of Shandong University, Jinan, Shandong, China.
| | - Sha Cheng
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Xiong Chen
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China.
| | - Xia Wang
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, Shandong, China; Laboratory of Translational Gastroenterology, Qilu Hospital of Shandong University, Jinan, Shandong, China; Robot Engineering Laboratory for Precise Diagnosis and Therapy of GI Tumor, Qilu Hospital of Shandong University, Jinan, Shandong, China.
| |
Collapse
|
12
|
An TY, Hu QM, Ni P, Hua YQ, Wang D, Duan GC, Chen SY, Jia B. N6-methyladenosine modification of hypoxia-inducible factor-1α regulates Helicobacter pylori-associated gastric cancer via the PI3K/AKT pathway. World J Gastrointest Oncol 2024; 16:3270-3283. [PMID: 39072157 PMCID: PMC11271789 DOI: 10.4251/wjgo.v16.i7.3270] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 05/09/2024] [Accepted: 05/20/2024] [Indexed: 07/12/2024] Open
Abstract
BACKGROUND Helicobacter pylori (H. pylori) colonizes the human gastric mucosa and is implicated in the development of gastric cancer (GC). The tumor microenvironment is characterized by hypoxia, where hypoxia-inducible factor-1α (HIF-1α) plays a key role as a transcription factor, but the mechanisms underlying H. pylori-induced HIF-1α expression and carcinogenesis remain unclear. AIM To explore the underlying mechanism of H. pylori-induced HIF-1α expression in promoting the malignant biological behavior of gastric epithelial cells (GES-1). METHODS The study was conducted with human GES-1 cells in vitro. Relative protein levels of methyltransferase-like protein 14 (METTL14), HIF-1α, main proteins of the PI3K/AKT pathway, epithelial-mesenchymal transition (EMT) biomarkers, and invasion indicators were detected by Western blot. Relative mRNA levels of METTL14 and HIF-1α were detected by quantitative reverse transcription-polymerase chain reaction. mRNA stability was evaluated using actinomycin D, and the interaction between METTL14 and HIF-1α was confirmed by immunofluorescence staining. Cell proliferation and migration were evaluated by cell counting kit-8 assay and wound healing assay, respectively. RESULTS H. pylori promoted HIF-1α expression and activated the PI3K/AKT pathway. Notably, METTL14 was downregulated in H. pylori-infected gastric mucosal epithelial cells and positively regulated HIF-1α expression. Functional experiments showed that the overexpression of HIF-1α or knockdown of METTL14 enhanced the activity of the PI3K/AKT pathway, thereby driving a series of malignant transformation, such as EMT and cell proliferation, migration, and invasion. By contrast, the knockdown of HIF-1α or overexpression of METTL14 had an opposite effect. CONCLUSION H. pylori-induced underexpression of METTL14 promotes the translation of HIF-1α and accelerates tumor progression by activating the PI3K/AKT pathway. These results provide novel insights into the carcinogenesis of GC.
Collapse
Affiliation(s)
- Tong-Yan An
- Department of Epidemiology, College of Public Health, Zhengzhou University, Zhengzhou 450001, Henan Province, China
| | - Quan-Man Hu
- Department of Epidemiology, College of Public Health, Zhengzhou University, Zhengzhou 450001, Henan Province, China
| | - Peng Ni
- Department of Epidemiology, College of Public Health, Zhengzhou University, Zhengzhou 450001, Henan Province, China
| | - Yan-Qiao Hua
- Department of Epidemiology, College of Public Health, Zhengzhou University, Zhengzhou 450001, Henan Province, China
| | - Di Wang
- Department of Epidemiology, College of Public Health, Zhengzhou University, Zhengzhou 450001, Henan Province, China
| | - Guang-Cai Duan
- Department of Epidemiology, College of Public Health, Zhengzhou University, Zhengzhou 450001, Henan Province, China
| | - Shuai-Yin Chen
- Department of Epidemiology, College of Public Health, Zhengzhou University, Zhengzhou 450001, Henan Province, China
| | - Bin Jia
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| |
Collapse
|
13
|
Long F, Zheng P, Su Q, Zhang Y, Wang D, Xiao Z, Wu M, Li J. LncRNA SNHG12 regulated by WTAP aggravated the oxygen-glucose deprivation/reperfusion-induced injury in bEnd.3 cell. J Stroke Cerebrovasc Dis 2024; 33:107613. [PMID: 38301749 DOI: 10.1016/j.jstrokecerebrovasdis.2024.107613] [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: 09/22/2023] [Revised: 01/25/2024] [Accepted: 01/29/2024] [Indexed: 02/03/2024] Open
Abstract
OBJECTIVES Previous studies have identified abnormal expression of lncRNA SNHG12 in ischemic stroke, but the underlying molecular mechanism remains unclear. MATERIALS AND METHODS Through database predictions, m6A methylation sites were found on SNHG12, suggesting post-transcriptional modification. To further elucidate the role of SNHG12 and m6A methyltransferase WTAP in oxygen-glucose deprivation/reperfusion (OGD/R)-induced damage in cerebral microvascular endothelial cells, we conducted investigations. Additionally, we examined the impact of m6A methyltransferase WTAP on SNHG12 expression. RESULTS Overexpressing SNHG12 in bEnd.3 cells was found to inhibit cell proliferation and promote apoptosis, as well as activate the production of reactive oxygen species and inflammatory cytokines (E-selectin, IL-6 and MCP-1), along with angiogenic proteins (VEGFA and FGFb). Conversely, SNHG12 knockdown alleviated OGD/R-induced damage to BEnd.3 cells, resulting in improved cell proliferation, reduced apoptosis, decreased ROS and LDH production, as well as diminished expression of inflammatory cytokines (E-selectin, IL-6 and MCP-1) and angiogenic proteins (VEGFA and FGFb). Furthermore, WTAP was found to positively regulate SNHG12 expression, and WTAP knockdown in bEnd.3 cells under the OGD/R conditions inhibited cell proliferation, promoted apoptosis, and increased ROS and LDH production. CONCLUSION These findings suggest that WTAP may play a crucial role in SNHG12-mediated OGD/R-induced damage in bEnd.3 cells. More molecular experiments are needed to further analyze its mechanism. Overall, our study helps to enrich our understanding of the dysregulation of SNHG12 in ischemic stroke.
Collapse
Affiliation(s)
- Faqing Long
- Department of Neurology, The Second Affiliated Hospital of Hainan Medical University, #368 Yehai Avenue, Longhua District, Haikou 570311, Hainan, China
| | - Pisi Zheng
- Hainan Medical University, Haikou, China
| | - Qingjie Su
- Department of Neurology, The Second Affiliated Hospital of Hainan Medical University, #368 Yehai Avenue, Longhua District, Haikou 570311, Hainan, China
| | - Yuhui Zhang
- Department of Neurology, The Second Affiliated Hospital of Hainan Medical University, #368 Yehai Avenue, Longhua District, Haikou 570311, Hainan, China
| | - Desheng Wang
- Department of Neurology, The Second Affiliated Hospital of Hainan Medical University, #368 Yehai Avenue, Longhua District, Haikou 570311, Hainan, China
| | - Zhixiang Xiao
- Department of Neurology, The Second Affiliated Hospital of Hainan Medical University, #368 Yehai Avenue, Longhua District, Haikou 570311, Hainan, China
| | - Mingchang Wu
- Department of Neurology, The Second Affiliated Hospital of Hainan Medical University, #368 Yehai Avenue, Longhua District, Haikou 570311, Hainan, China
| | - Jianhong Li
- Department of Neurology, The Second Affiliated Hospital of Hainan Medical University, #368 Yehai Avenue, Longhua District, Haikou 570311, Hainan, China.
| |
Collapse
|
14
|
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
|