1
|
Chen X, Lai J, Wu Z, Chen J, Yang B, Chen C, Ding C. Fat mass and obesity-mediated N 6 -methyladenosine modification modulates neuroinflammatory responses after traumatic brain injury. Neural Regen Res 2026; 21:730-741. [PMID: 39248160 DOI: 10.4103/nrr.nrr-d-23-01854] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Accepted: 05/24/2024] [Indexed: 09/10/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202602000-00042/figure1/v/2025-05-05T160104Z/r/image-tiff The neuroinflammatory response mediated by microglial activation plays an important role in the secondary nerve injury of traumatic brain injury. The post-transcriptional modification of N 6 -methyladenosine is ubiquitous in the immune response of the central nervous system. The fat mass and obesity-related protein catalyzes the demethylation of N 6 -methyladenosine modifications on mRNA and is widely expressed in various tissues, participating in the regulation of multiple diseases' biological processes. However, the role of fat mass and obesity in microglial activation and the subsequent neuroinflammatory response after traumatic brain injury is unclear. In this study, we found that the expression of fat mass and obesity was significantly down-regulated in both lipopolysaccharide-treated BV2 cells and a traumatic brain injury mouse model. After fat mass and obesity interference, BV2 cells exhibited a pro-inflammatory phenotype as shown by the increased proportion of CD11b + /CD86 + cells and the secretion of pro-inflammatory cytokines. Fat mass and obesity-mediated N 6 -methyladenosine demethylation accelerated the degradation of ADAM17 mRNA, while silencing of fat mass and obesity enhanced the stability of ADAM17 mRNA. Therefore, down-regulation of fat mass and obesity expression leads to the abnormally high expression of ADAM17 in microglia. These results indicate that the activation of microglia and neuroinflammatory response regulated by fat mass and obesity-related N 6 -methyladenosine modification plays an important role in the pro-inflammatory process of secondary injury following traumatic brain injury.
Collapse
Affiliation(s)
- Xiangrong Chen
- Department of Neurosurgery, Second Clinical Medical College, Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China
| | - Jinqing Lai
- Department of Neurosurgery, Second Clinical Medical College, Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China
| | - Zhe Wu
- Department of Neurosurgery, Second Clinical Medical College, Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China
| | - Jianlong Chen
- Department of Neurosurgery, Second Clinical Medical College, Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China
| | - Baoya Yang
- Department of Neurosurgery, Second Clinical Medical College, Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China
| | - Chunnuan Chen
- Department of Neurology, Second Clinical Medical College, Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China
| | - Chenyu Ding
- Department of Neurosurgery, Neurosurgery Research Institute, National Regional Medical Center, Binhai Campus, First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian Province, China
| |
Collapse
|
2
|
Li D, Qian L, Du Y, Liu L, Sun Z, Han Y, Guo X, Shen C, Zhang Z, Liu X. METTL14-mediated m 6A modification of DDIT4 promotes its mRNA stability in aging-related idiopathic pulmonary fibrosis. Epigenetics 2025; 20:2462898. [PMID: 39916577 PMCID: PMC11810098 DOI: 10.1080/15592294.2025.2462898] [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: 06/23/2024] [Revised: 01/01/2025] [Accepted: 01/29/2025] [Indexed: 02/12/2025] Open
Abstract
Although N6-methyladenosine (m6A) may be related to the pathogenesis of fibrotic process, the mechanism of m6A modification in aging-related idiopathic pulmonary fibrosis (IPF) remains unclear. Three-milliliter venous blood was collected from IPF patients and healthy controls. MeRIP-seq and RNA-seq were utilized to investigate differential m6A modification. The expressions of identified m6A regulator and target gene were validated using MeRIP-qPCR and real-time PCR. Moreover, we established an animal model and a senescent model of A549 cells to explore the associated molecular mechanism. Our study provided a panorama of m6A methylation in IPF. Increased peaks (3756) and decreased peaks (4712) were observed in the IPF group. The association analysis showed that 749 DEGs were affected by m6A methylation in IPF. Among the m6A regulators, the expression of METTL14 decreased in IPF. The m6A level of our interested gene DDIT4 decreased significantly, but the mRNA level of DDIT4 was higher in IPF. This was further verified in bleomycin-induced pulmonary fibrosis. At the cellular level, it was further confirmed that METTL14 and DDIT4 might participate in the senescence of alveolar epithelial cells. The downregulation of METTL14 might inhibit the decay of DDIT4 mRNA by reducing the m6A modification level of DDIT4 mRNA, leading to high expression of DDIT4 mRNA and protein. Our study provided a panorama of m6A alterations in IPF and discovered METTL14 as a potential intervention target for epigenetic modification in IPF. These results pave the way for future investigations regarding m6A modifications in aging-related IPF.
Collapse
Affiliation(s)
- Dan Li
- First Clinical Medical College, Shanxi Medical University, Taiyuan, China
- Department of Geriatrics, the First Hospital of Shanxi Medical University, Taiyuan, China
| | - Li Qian
- Department of Geriatrics, the First Hospital of Shanxi Medical University, Taiyuan, China
| | - Yufeng Du
- Department of Geriatrics, the First Hospital of Shanxi Medical University, Taiyuan, China
| | - Lifang Liu
- Department of Geriatrics, the First Hospital of Shanxi Medical University, Taiyuan, China
| | - Ziyue Sun
- First Clinical Medical College, Shanxi Medical University, Taiyuan, China
| | - Yongkang Han
- First Clinical Medical College, Shanxi Medical University, Taiyuan, China
| | - Xiangrui Guo
- First Clinical Medical College, Shanxi Medical University, Taiyuan, China
| | - Chao Shen
- First Clinical Medical College, Shanxi Medical University, Taiyuan, China
| | - Zheng Zhang
- First Clinical Medical College, Shanxi Medical University, Taiyuan, China
| | - Xuejun Liu
- Department of Geriatrics, the First Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
3
|
Li D, Chu X, Liu W, Ma Y, Tian X, Yang Y. The regulatory roles of RNA-binding proteins in the tumour immune microenvironment of gastrointestinal malignancies. RNA Biol 2025; 22:1-14. [PMID: 39718205 DOI: 10.1080/15476286.2024.2440683] [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] [Revised: 11/03/2024] [Accepted: 12/04/2024] [Indexed: 12/25/2024] Open
Abstract
The crosstalk between the tumour immune microenvironment (TIME) and tumour cells promote immune evasion and resistance to immunotherapy in gastrointestinal (GI) tumours. Post-transcriptional regulation of genes is pivotal to GI tumours progression, and RNA-binding proteins (RBPs) serve as key regulators via their RNA-binding domains. RBPs may exhibit either anti-tumour or pro-tumour functions by influencing the TIME through the modulation of mRNAs and non-coding RNAs expression, as well as post-transcriptional modifications, primarily N6-methyladenosine (m6A). Aberrant regulation of RBPs, such as HuR and YBX1, typically enhances tumour immune escape and impacts prognosis of GI tumour patients. Further, while targeting RBPs offers a promising strategy for improving immunotherapy in GI cancers, the mechanisms by which RBPs regulate the TIME in these tumours remain poorly understood, and the therapeutic application is still in its early stages. This review summarizes current advances in exploring the roles of RBPs in regulating genes expression and their effect on the TIME of GI tumours, then providing theoretical insights for RBP-targeted cancer therapies.
Collapse
Affiliation(s)
- Dongqi Li
- Department of Hepatobiliary and Pancreatic Surgery, Peking University First Hospital, Beijing, China
| | - Xiangyu Chu
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Weikang Liu
- Department of Hepatobiliary and Pancreatic Surgery, Peking University First Hospital, Beijing, China
| | - Yongsu Ma
- Department of Hepatobiliary and Pancreatic Surgery, Peking University First Hospital, Beijing, China
| | - Xiaodong Tian
- Department of Hepatobiliary and Pancreatic Surgery, Peking University First Hospital, Beijing, China
| | - Yinmo Yang
- Department of Hepatobiliary and Pancreatic Surgery, Peking University First Hospital, Beijing, China
| |
Collapse
|
4
|
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
|
5
|
Wang H, Han J, Zhang XA. Interplay of m6A RNA methylation and gut microbiota in modulating gut injury. Gut Microbes 2025; 17:2467213. [PMID: 39960310 PMCID: PMC11834532 DOI: 10.1080/19490976.2025.2467213] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Revised: 12/12/2024] [Accepted: 02/10/2025] [Indexed: 02/20/2025] Open
Abstract
The gut microbiota undergoes continuous variations among individuals and across their lifespan, shaped by diverse factors encompassing diet, age, lifestyle choices, medication intake, and disease states. These microbial inhabitants play a pivotal role in orchestrating physiological metabolic pathways through the production of metabolites like bile acids, choline, short-chain fatty acids, and neurotransmitters, thereby establishing a dynamic "gut-organ axis" with the host. The intricate interplay between the gut microbiota and the host is indispensable for gut health, and RNA N6-methyladenosine modification, a pivotal epigenetic mark on RNA, emerges as a key player in this process. M6A modification, the most prevalent internal modification of eukaryotic RNA, has garnered significant attention in the realm of RNA epigenetics. Recent findings underscore its potential to influence gut microbiota diversity and intestinal barrier function by modulating host gene expression patterns. Conversely, the gut microbiota, through its impact on the epigenetic landscape of host cells, may indirectly regulate the recruitment and activity of RNA m6A-modifying enzymes. This review endeavors to delve into the biological functions of m6A modification and its consequences on intestinal injury and disease pathogenesis, elucidating the partial possible mechanisms by which the gut microbiota and its metabolites maintain host intestinal health and homeostasis. Furthermore, it also explores the intricate crosstalk between them in intestinal injury, offering a novel perspective that deepens our understanding of the mechanisms underlying intestinal diseases.
Collapse
Affiliation(s)
- Haixia Wang
- College of Exercise and Health, Shenyang Sport University, Shenyang, China
| | - Juanjuan Han
- College of Exercise and Health, Shenyang Sport University, Shenyang, China
| | - Xin-An Zhang
- College of Exercise and Health, Shenyang Sport University, Shenyang, China
| |
Collapse
|
6
|
Zhang J, Li G, Wu R, Shi L, Tian C, Jiang H, Che H, Jiang Y, Jin Z, Yu R, Liu X, Zhang X. The m6A RNA demethylase FTO promotes radioresistance and stemness maintenance of glioma stem cells. Cell Signal 2025; 132:111782. [PMID: 40185350 DOI: 10.1016/j.cellsig.2025.111782] [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: 09/20/2024] [Revised: 03/09/2025] [Accepted: 03/30/2025] [Indexed: 04/07/2025]
Abstract
Glioblastoma (GBM) was the most common and deadliest malignant brain tumor in adults, with a poor prognosis. Effective targeted drugs are still lacking, and the presence of glioblastoma stem cells (GSC) is a major factor contributing to radiotherapy resistance. Screening for targeted drugs that can sensitize GBM to radiotherapy is crucial. FTO is considered an attractive potential target for tumor therapy, as it mediates m6A demethylation to regulate the stability of target genes. In this study, we evaluated the role of FTO inhibition in promoting the sensitivity of GSC cells to radiotherapy through tumor sphere formation assays, cell apoptosis assays, and in situ GSC tumor models. We preliminarily explored the molecular mechanisms by transcriptome sequencing and m6A methylation sequencing to investigate how inhibiting FTO increases radiotherapy sensitivity. The results showed that downregulation of FTO expression or FTO inhibitor FB23-2 combined with radiotherapy significantly inhibited GSC cell proliferation and self-renewal and increased apoptosis. FB23-2 combined with radiotherapy effectively inhibited intracranial tumor growth in mice and prolonged the survival of tumor-bearing mice. Furthermore, FTO inhibition sustained the increase of γH2AX expression induced by radiotherapy while decreasing Rad51 expression. Importantly, we found that inhibiting FTO could increase m6A methylation modification of VEGFA, thereby downregulating both mRNA and protein expression of VEGFA. Our findings provide a new therapeutic strategy for enhancing GBM radiotherapy sensitivity and lay the theoretical and experimental groundwork for clinical trials targeting FTO.
Collapse
Affiliation(s)
- Junhao Zhang
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China; Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Department of Neurosurgery, Xinxiang Central Hospital, Xinxiang 453003, Henan, China
| | - Guoxi Li
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China; Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Runqiu Wu
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China; Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Lin Shi
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China; Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Department of General Surgery, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Cong Tian
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Hongyan Jiang
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Hongyu Che
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yongang Jiang
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Zhiyong Jin
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Rutong Yu
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China; Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.
| | - Xuejiao Liu
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China; Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.
| | - Xu Zhang
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China; Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.
| |
Collapse
|
7
|
Yang M, Zhang Z, Qin H, Lin X, Liu X, Zhang H. The emerging significance of the METTL family as m6A-modified RNA methyltransferases in head and neck cancer. Cell Signal 2025; 132:111798. [PMID: 40239728 DOI: 10.1016/j.cellsig.2025.111798] [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: 01/14/2025] [Revised: 04/06/2025] [Accepted: 04/07/2025] [Indexed: 04/18/2025]
Abstract
RNA epigenetic modifications are crucial in tumor development, with N6-methyladenosine (m6A) being the most prevalent epigenetic modification found in all eukaryotic messenger RNAs. Accumulating evidence indicates that m6A modifications significantly influence the progression of various malignancies, including head and neck cancer (HNC). The Methyltransferase-like (METTL) family proteins, a group of methyltransferases identified in recent years, function as the "writers" of m6A modifications. These proteins affect RNA stability, translation efficiency, splicing, and localization, thereby regulating diverse cellular functions and promoting tumorigenesis in multiple cancers through their methylation domains. This review aims to summarize existing literature on the METTL family of m6A-modified RNA to elucidate their roles in HNC, providing a theoretical foundation for their potential use as therapeutic targets.
Collapse
Affiliation(s)
- Ming Yang
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, China; Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai, Shandong, China; Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong, China; Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai, Shandong, China; The 2nd Medical College of Binzhou Medical University, Yantai, Shandong, China.
| | - Zile Zhang
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, China; Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai, Shandong, China; Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong, China; Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai, Shandong, China; The 2nd Medical College of Binzhou Medical University, Yantai, Shandong, China
| | - Hanbin Qin
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, China; Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai, Shandong, China; Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong, China; Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai, Shandong, China
| | - Xinhua Lin
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, China; Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai, Shandong, China; Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong, China; Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai, Shandong, China
| | - Xuexia Liu
- Shandong Stem Cell Engineering Technology Research Center, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, China.
| | - Hua Zhang
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, China; Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai, Shandong, China; Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong, China; Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai, Shandong, China.
| |
Collapse
|
8
|
Shi Y, Liu J, Cheng Q, Wu S, Song W, Wang K, Chen Z, Li X, Wei Q, Tayier D, Liao B, Yang Z. METTL3/IGF2BP3 mediates ORC6 via N6-methyladenosine modification to promote the progression of pancreatic ductal adenocarcinoma. Gene 2025; 955:149468. [PMID: 40185346 DOI: 10.1016/j.gene.2025.149468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 03/19/2025] [Accepted: 03/31/2025] [Indexed: 04/07/2025]
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is recognized globally as one of the most lethal tumours, and effective biomarkers to diagnose PDAC early are needed. ORC6, a subunit of the origin recognition complex (ORC), initiates DNA replication and ensures genomic stability. Previous studies have indicated that ORC6 is procarcinogenic in various cancers, yet its role in PDAC remains uninvestigated. METHODS We evaluated the relationships between ORC6 expression and the clinical features of patients with PDAC with the TCGA, GTEx, and GEO databases. The role of ORC6 in PDAC cells was explored by RNA interference in vitro and in vivo. Next, we verified the effect of the METTL3/IGF2BP3/ORC6 axis on PDAC progression by western blotting, RT-qPCR, RNA immunoprecipitation, and methylated RNA immunoprecipitation. Finally, transcriptome analysis was performed to explore the influence of ORC6 on p53 in PDAC cells. RESULTS Elevated ORC6 levels were observed in PDAC cells, which correlated with poorer clinical outcomes. Both in vivo and in vitro experiments demonstrated that ORC6 knockdown suppressed proliferation and promoted apoptosis. Additionally, we demonstrated that METTL3/IGF2BP3 interacted with ORC6 mRNA via N6-methyladenosine modification to improve ORC6 mRNA stability. Transcriptomic analysis and experiments indicated that ORC6 promoted PDAC progression by inhibiting serine-15 phosphorylation in p53. CONCLUSION Our findings validate the role of ORC6 in PDAC and support the hypothesis that the METTL3/IGF2BP3/ORC6/p53 axis may be a novel therapeutic target for PDAC, and inhibiting this axis may be an advantageous therapeutic strategy for curing PDAC.
Collapse
Affiliation(s)
- Yang Shi
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430061 Hubei Province, China; Pancreatic Surgery Center, Zhongnan Hospital of Wuhan University, Wuhan 430061 Hubei Province, China; Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan 430061 Hubei Province, China
| | - Junwei Liu
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430061 Hubei Province, China
| | - Qian Cheng
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430061 Hubei Province, China; Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan 430061 Hubei Province, China
| | - Shuaihui Wu
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430061 Hubei Province, China; Pancreatic Surgery Center, Zhongnan Hospital of Wuhan University, Wuhan 430061 Hubei Province, China; Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan 430061 Hubei Province, China
| | - Wenjing Song
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430061 Hubei Province, China; Pancreatic Surgery Center, Zhongnan Hospital of Wuhan University, Wuhan 430061 Hubei Province, China; Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan 430061 Hubei Province, China
| | - Kunlei Wang
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430061 Hubei Province, China; Pancreatic Surgery Center, Zhongnan Hospital of Wuhan University, Wuhan 430061 Hubei Province, China; Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan 430061 Hubei Province, China
| | - Zhinan Chen
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430061 Hubei Province, China; Pancreatic Surgery Center, Zhongnan Hospital of Wuhan University, Wuhan 430061 Hubei Province, China; Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan 430061 Hubei Province, China
| | - Xinyin Li
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430061 Hubei Province, China; Pancreatic Surgery Center, Zhongnan Hospital of Wuhan University, Wuhan 430061 Hubei Province, China; Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan 430061 Hubei Province, China
| | - Qifeng Wei
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430061 Hubei Province, China; Pancreatic Surgery Center, Zhongnan Hospital of Wuhan University, Wuhan 430061 Hubei Province, China; Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan 430061 Hubei Province, China
| | - Dilinigeer Tayier
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430061 Hubei Province, China; Pancreatic Surgery Center, Zhongnan Hospital of Wuhan University, Wuhan 430061 Hubei Province, China; Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan 430061 Hubei Province, China
| | - Bo Liao
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430061 Hubei Province, China; Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan 430061 Hubei Province, China.
| | - Zhiyong Yang
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430061 Hubei Province, China; Pancreatic Surgery Center, Zhongnan Hospital of Wuhan University, Wuhan 430061 Hubei Province, China; Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan 430061 Hubei Province, China.
| |
Collapse
|
9
|
Wang K, Sun Z. The role of m6A methylation in abdominal aortic aneurysms: Mechanisms, progress and future perspectives (Review). Mol Med Rep 2025; 32:199. [PMID: 40376996 DOI: 10.3892/mmr.2025.13564] [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: 01/25/2025] [Accepted: 04/28/2025] [Indexed: 05/18/2025] Open
Abstract
Abdominal aortic aneurysm (AAA) is a type of cardiovascular disease. Sudden aortic rupture and subsequent bleeding are the main causes of mortality due to AAA. N6‑methyladenosine (m6A) methylation, the most common epitranscriptomic modification in eukaryotic mRNAs, has a key role in the regulation of gene expression. m6A methylation markedly influences the development and progression of AAA. The present review highlights the mechanism of m6A methylation in AAA, including current research progress and future prospects. From a mechanistic perspective, m6A methylation exerts its influence on AAA‑related genes by modulating the post‑transcriptional levels of RNA, thereby impacting the pathological process of AAA. In terms of clinical applications, the mechanisms by which m6A methylation regulators influence their development and progression in AAA involve multiple target genes and signaling pathways. These regulatory factors affect inflammatory immunomodulation, cell proliferation, apoptosis and endogenous processes by modulating the m6A modification status of target genes and the activity of immune‑related signaling pathways. Therefore, for the prevention and treatment of AAA, current therapeutic strategies should comprehensively consider the interactions and synergistic regulation among m6A methylation regulators to reveal the integrated effects of the entire regulatory network in AAA development. Consequently, a more comprehensive understanding of the precise mechanisms of m6A methylation in AAA should be attained, which will support the development of innovative therapeutic strategies aimed at m6A methylation and establish a basis for the early diagnosis and treatment of AAA.
Collapse
Affiliation(s)
- Keyu Wang
- Department of Hepatobiliary and Vascular Surgery, Jining Third People's Hospital, Jining, Shandong 272100, P.R. China
| | - Ziqiang Sun
- Department of Vascular Surgery, Affiliated Hospital of Jining Medical University, Jining, Shandong 272029, P.R. China
| |
Collapse
|
10
|
Li J, Lv L, Hu M, Liu Z, Zhou S. Inhibition of N6-methyladenosine methylation of ASC by berberine ameliorates pyroptosis of renal tubular epithelial cells in acute kidney injury. Cell Signal 2025; 131:111732. [PMID: 40074191 DOI: 10.1016/j.cellsig.2025.111732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Revised: 03/04/2025] [Accepted: 03/09/2025] [Indexed: 03/14/2025]
Abstract
Acute kidney injury (AKI) lacks a definitive therapeutic approach beyond supportive care. One significant pathological mechanism involves the regulated death of tubular epithelial cells; however, the regulatory mechanisms underlying this cell death pathway require further investigation. The N6-methyladenosine (m6A) modification, recognized as the most prevalent modification in eukaryotes, plays a critical role in the regulatory processes associated with AKI. Here, this study investigates the association between methyltransferase-like 3 (METTL3) and pyroptosis in mice with folic acid (FA)-induced AKI. Both in vitro and in vivo experiments have confirmed that METTL3 plays a role in AKI progression, correlating with renal epithelial cell pyroptosis and inflammation. Moreover, RNA immunoprecipitation quantitative PCR (RIP-qPCR) analysis demonstrated that METTL3-mediated m6A methylation occurred in the mRNA of Apoptosis-associated speck-like protein containing a CARD (ASC) in H2O2-induced renal tubular epithelial (TCMK-1) cells. Notably, METTL3 knockdown resulted in reduced ASC protein expression, decreased release of inflammatory factors, and reduced pyroptosis. In addition, we verified the inhibitory effect of berberine hydrochloride, a monomer used in traditional Chinese medicine, on METTL3 expression. We also demonstrated that berberine ameliorated FA-induced AKI and H2O2-induced pyroptosis in TCMK-1 cells by inhibiting METTL3 and modulating the ASC/caspase-1/Gasdermin D axis. These findings provide insights into targeted therapies and drug development for AKI.
Collapse
Affiliation(s)
- Jiacheng Li
- Traditional Chinese Medicine Integrated Department of Nephrology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China; Research Institute of Nephrology, Zhengzhou University, Zhengzhou 450052, PR China; Henan Province Research Center for Kidney Disease, Zhengzhou 450052, PR China; Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou 450052, PR China; Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University Zhengzhou, Henan, China; Innovation Center of Basic Research for Metabolic-Associated Fatty Liver Disease, Ministry of Education of, China
| | - Linxiao Lv
- Traditional Chinese Medicine Integrated Department of Nephrology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China; Research Institute of Nephrology, Zhengzhou University, Zhengzhou 450052, PR China; Henan Province Research Center for Kidney Disease, Zhengzhou 450052, PR China; Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou 450052, PR China; Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University Zhengzhou, Henan, China; Innovation Center of Basic Research for Metabolic-Associated Fatty Liver Disease, Ministry of Education of, China
| | - Mingyang Hu
- Traditional Chinese Medicine Integrated Department of Nephrology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China; Research Institute of Nephrology, Zhengzhou University, Zhengzhou 450052, PR China; Henan Province Research Center for Kidney Disease, Zhengzhou 450052, PR China; Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou 450052, PR China; Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University Zhengzhou, Henan, China; Innovation Center of Basic Research for Metabolic-Associated Fatty Liver Disease, Ministry of Education of, China
| | - Zhangsuo Liu
- Traditional Chinese Medicine Integrated Department of Nephrology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China; Research Institute of Nephrology, Zhengzhou University, Zhengzhou 450052, PR China; Henan Province Research Center for Kidney Disease, Zhengzhou 450052, PR China; Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou 450052, PR China; Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University Zhengzhou, Henan, China; Innovation Center of Basic Research for Metabolic-Associated Fatty Liver Disease, Ministry of Education of, China.
| | - Sijie Zhou
- Traditional Chinese Medicine Integrated Department of Nephrology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China; Research Institute of Nephrology, Zhengzhou University, Zhengzhou 450052, PR China; Henan Province Research Center for Kidney Disease, Zhengzhou 450052, PR China; Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou 450052, PR China; Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University Zhengzhou, Henan, China; Innovation Center of Basic Research for Metabolic-Associated Fatty Liver Disease, Ministry of Education of, China.
| |
Collapse
|
11
|
Chen T, Ye W, Gao S, Li Y, Luan J, Lv X, Wang S. Emerging importance of m6A modification in liver cancer and its potential therapeutic role. Biochim Biophys Acta Rev Cancer 2025; 1880:189299. [PMID: 40088993 DOI: 10.1016/j.bbcan.2025.189299] [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: 10/24/2024] [Revised: 03/04/2025] [Accepted: 03/09/2025] [Indexed: 03/17/2025]
Abstract
Liver cancer refers to malignant tumors that form in the liver and is usually divided into several types, the most common of which is hepatocellular carcinoma (HCC), which originates in liver cells. Other rare types of liver cancer include intrahepatic cholangiocarcinoma (iCCA). m6A modification is a chemical modification of RNA that usually manifests as the addition of a methyl group to adenine in the RNA molecule to form N6-methyladenosine. This modification exerts a critical role in various biological processes by regulating the metabolism of RNA, affecting gene expression. Recent studies have shown that m6A modification is closely related to the occurrence and development of liver cancer, and m6A regulators can further participate in the pathogenesis of liver cancer by regulating the expression of key genes and the function of specific cells. In this review, we provided an overview of the latest advances in m6A modification in liver cancer research and explored in detail the specific functions of different m6A regulators. Meanwhile, we deeply analyzed the mechanisms and roles of m6A modification in liver cancer, aiming to provide novel insights and references for the search for potential therapeutic targets. Finally, we discussed the prospects and challenges of targeting m6A regulators in liver cancer therapy.
Collapse
Affiliation(s)
- Tao Chen
- Department of Pharmacy, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, Anhui Province 241001, China
| | - Wufei Ye
- Department of Pharmacy, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, Anhui Province 241001, China
| | - Songsen Gao
- Department of Orthopedics (Spinal Surgery), The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province 230022, China
| | - Yueran Li
- Department of Pharmacy, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, Anhui Province 241001, China
| | - Jiajie Luan
- Department of Pharmacy, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, Anhui Province 241001, China
| | - Xiongwen Lv
- The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Anhui Province Key Laboratory of Major Autoimmune Diseases, School of Pharmacy, Institute for Liver Disease of Anhui Medical University, Hefei, Anhui Province 230032, China.
| | - Sheng Wang
- Department of Pharmacy, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, Anhui Province 241001, China; The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Anhui Province Key Laboratory of Major Autoimmune Diseases, School of Pharmacy, Institute for Liver Disease of Anhui Medical University, Hefei, Anhui Province 230032, China.
| |
Collapse
|
12
|
Liu M, Ke M, Lu H, Feng Z, Wang K, Wang D, Wang K, Bai Y, Yang S, Miao L, Chen Q, Sun M, Shan C, Hu J, Jiang L, Jin H, Hu J, Huang C, Wang R, Zhao W, Yu F. A novel cinnamic acid derivative for hepatocellular carcinoma therapy by degrading METTL16 protein. Bioorg Med Chem 2025; 124:118178. [PMID: 40186923 DOI: 10.1016/j.bmc.2025.118178] [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: 02/11/2025] [Revised: 03/26/2025] [Accepted: 03/28/2025] [Indexed: 04/07/2025]
Abstract
The RNA methyltransferase methyltransferaselike protein 16 (METTL16) is upregulated in a large proportion of hepatocellular carcinoma (HCC), and its high expression is associated with poor clinical outcomes. METTL16 deletion inhibits HCC growth in vitro and in vivo. Referencing the structure of cinnamic acid, here we designed and synthesized a novel series of small molecular compounds, and found through bioactivity screening that compound 15a effectively reduced METTL16 level and modulated oncogenic PI3K/AKT pathway signaling. Compound 15a inhibited the proliferation and migration of HepG2 cells, and induced apoptosis in vitro. Furthermore, compound 15a significantly inhibited the growth of patient-derived HCC xenografts in nude mice with greater efficacy than the multi-kinase inhibitor lenvatinib. The promising efficacy and good biosafety profile of compound 15a enables us to further develop this compound for treating patients with HCC and possibly other cancers in clinic.
Collapse
Affiliation(s)
- Mingyang Liu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Key Laboratory of Molecular Drug Research and KLMDASR of Tianjin, Nankai University, Tongyan Road, Haihe Education Park, Tianjin 300350, China
| | - Muyan Ke
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Key Laboratory of Molecular Drug Research and KLMDASR of Tianjin, Nankai University, Tongyan Road, Haihe Education Park, Tianjin 300350, China
| | - Hongchen Lu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Key Laboratory of Molecular Drug Research and KLMDASR of Tianjin, Nankai University, Tongyan Road, Haihe Education Park, Tianjin 300350, China
| | - Ziyu Feng
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Key Laboratory of Molecular Drug Research and KLMDASR of Tianjin, Nankai University, Tongyan Road, Haihe Education Park, Tianjin 300350, China
| | - Kaixuan Wang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Key Laboratory of Molecular Drug Research and KLMDASR of Tianjin, Nankai University, Tongyan Road, Haihe Education Park, Tianjin 300350, China
| | - Danyang Wang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Key Laboratory of Molecular Drug Research and KLMDASR of Tianjin, Nankai University, Tongyan Road, Haihe Education Park, Tianjin 300350, China
| | - Kun Wang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Key Laboratory of Molecular Drug Research and KLMDASR of Tianjin, Nankai University, Tongyan Road, Haihe Education Park, Tianjin 300350, China
| | - Yueping Bai
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Key Laboratory of Molecular Drug Research and KLMDASR of Tianjin, Nankai University, Tongyan Road, Haihe Education Park, Tianjin 300350, China; Qingdao Central Hospital, School of Health and Life Sciences, University of Health and Rehabilitation Sciences, No. 369 Dengyun Road, Qingdao 266113, China
| | - Song Yang
- Qingdao Central Hospital, School of Health and Life Sciences, University of Health and Rehabilitation Sciences, No. 369 Dengyun Road, Qingdao 266113, China
| | - Lu Miao
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Key Laboratory of Molecular Drug Research and KLMDASR of Tianjin, Nankai University, Tongyan Road, Haihe Education Park, Tianjin 300350, China
| | - Qiang Chen
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Key Laboratory of Molecular Drug Research and KLMDASR of Tianjin, Nankai University, Tongyan Road, Haihe Education Park, Tianjin 300350, China
| | - Mingming Sun
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Key Laboratory of Molecular Drug Research and KLMDASR of Tianjin, Nankai University, Tongyan Road, Haihe Education Park, Tianjin 300350, China
| | - Changliang Shan
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Key Laboratory of Molecular Drug Research and KLMDASR of Tianjin, Nankai University, Tongyan Road, Haihe Education Park, Tianjin 300350, China.
| | - Jiancheng Hu
- Cancer and Stem Cell Program, Duke-NUS Medical School, 8 College Road, 169857 Singapore, Singapore.
| | - Lingyu Jiang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Key Laboratory of Molecular Drug Research and KLMDASR of Tianjin, Nankai University, Tongyan Road, Haihe Education Park, Tianjin 300350, China
| | - Hongzhen Jin
- Qingdao Central Hospital, School of Health and Life Sciences, University of Health and Rehabilitation Sciences, No. 369 Dengyun Road, Qingdao 266113, China
| | - Jinfang Hu
- State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Tiancheng Drug Assessment Co., Ltd, Tianjin 300193, Chinaa.
| | - Changjiang Huang
- State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Tiancheng Drug Assessment Co., Ltd, Tianjin 300193, Chinaa.
| | - Rui Wang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Key Laboratory of Molecular Drug Research and KLMDASR of Tianjin, Nankai University, Tongyan Road, Haihe Education Park, Tianjin 300350, China.
| | - Wei Zhao
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Key Laboratory of Molecular Drug Research and KLMDASR of Tianjin, Nankai University, Tongyan Road, Haihe Education Park, Tianjin 300350, China; Qingdao Central Hospital, School of Health and Life Sciences, University of Health and Rehabilitation Sciences, No. 369 Dengyun Road, Qingdao 266113, China; Frontiers Science Center for New Organic Matter, Nankai University, Tongyan Road, Haihe Education Park, Tianjin 300350, China.
| | - Fan Yu
- Qingdao Central Hospital, School of Health and Life Sciences, University of Health and Rehabilitation Sciences, No. 369 Dengyun Road, Qingdao 266113, China.
| |
Collapse
|
13
|
Liu X, Zhang L, Chen J, Shao W. Decoding intricate interactions between m6A modification with mRNAs and non-coding RNAs in cervical cancer: Molecular mechanisms and clinical implications. Cell Signal 2025; 131:111745. [PMID: 40107480 DOI: 10.1016/j.cellsig.2025.111745] [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: 10/10/2024] [Revised: 03/11/2025] [Accepted: 03/12/2025] [Indexed: 03/22/2025]
Abstract
N6-methyladenosine (m6A) methylation is the most prevalent RNA modification that is regulated by three regulatory factors: "writers", "erasers" and "readers". m6A modification regulates RNA stability and other mechanisms, including translation, cleavage, and degradation. Current research has demonstrated that m6A methylation is involved in the regulation of occurrence and development of cancers by controlling the expression of cancer-related genes. This review summarizes the role of m6A modification on messenger RNAs (mRNAs) and non-coding RNAs (ncRNAs) in cervical cancer (CC). We highlight the dual role of m6A regulatory factors, which act as oncogenes or tumor suppressors depending on the cellular context and downstream targets. Additionally, we examine how ncRNAs reciprocally regulate m6A modification in two ways: by guiding the deposition or removal of m6A modifications on RNA targets, and by modulating the expression of m6A regulatory factors. These interactions further contribute to tumor progression. Furthermore, the therapeutic potential of targeting m6A modification has been emphasized in CC. Moreover, recent advances in small-molecule inhibitors targeting m6A regulators and RNA-based therapies which may offer new treatment strategies have been summarized. Finally, we discuss the current challenges in m6A modification research and provide suggestions for future research directions. This review aims to deepen the understanding of m6A modification in CC and contribute to the development of targeted and personalized treatment strategies.
Collapse
Affiliation(s)
- Xuefei Liu
- Department of Microbiology and Parasitology, Anhui Provincial Laboratory of Pathogen Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, Anhui, China; First Clinical Medical College, Anhui Medical University, Hefei, Anhui, China
| | - Lizhi Zhang
- First Clinical Medical College, Anhui Medical University, Hefei, Anhui, China
| | - Ji Chen
- Department of Obstetrics, The Third Affiliated Hospital of Anhui Medical University, Hefei 230061, Anhui, China
| | - Wei Shao
- Department of Microbiology and Parasitology, Anhui Provincial Laboratory of Pathogen Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, Anhui, China.
| |
Collapse
|
14
|
Qiu X, Gao Q, Wang J, Zhang Z, Tao L. The microbiota-m 6A-metabolism axis: Implications for therapeutic strategies in gastrointestinal cancers. Biochim Biophys Acta Rev Cancer 2025; 1880:189317. [PMID: 40222422 DOI: 10.1016/j.bbcan.2025.189317] [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: 09/11/2024] [Revised: 04/06/2025] [Accepted: 04/06/2025] [Indexed: 04/15/2025]
Abstract
Gastrointestinal (GI) cancers remain a leading cause of cancer-related mortality worldwide, with metabolic reprogramming recognized as a central driver of tumor progression and therapeutic resistance. Among the key regulatory layers, N6-methyladenosine (m6A) RNA modification-mediated by methyltransferases (writers such as METTL3/14), RNA-binding proteins (readers like YTHDFs and IGF2BPs), and demethylases (erasers including FTO and ALKBH5), plays a pivotal role in controlling gene expression and metabolic flux in the tumor context. Concurrently, the gut microbiota profoundly influences GI tumorigenesis and immune evasion by modulating metabolite availability and remodeling the tumor microenvironment. Recent evidence has uncovered a bidirectional crosstalk between microbial metabolites and m6A methylation: microbiota-derived signals dynamically regulate m6A deposition on metabolic and immune transcripts, while m6A modifications, in turn, regulate the stability and translation of key mRNAs such as PD-L1 and FOXP3. This reciprocal interaction forms self-reinforcing epigenetic circuits that drive tumor plasticity, immune escape, and metabolic adaptation. In this review, we dissect the molecular underpinnings of the microbiota-m6A-metabolism axis in GI cancers and explore its potential to inform novel strategies in immunotherapy, metabolic intervention, and microbiome-guided precision oncology.
Collapse
Affiliation(s)
- Xiuxiu Qiu
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Qi Gao
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Jiahui Wang
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Zhanxia Zhang
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Li Tao
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| |
Collapse
|
15
|
Yao Z, Sun L, Gao Y, Su Y, He B, Ge Y, Yang C, Jia X, Jiao A, Sun C, Zhang B. The m 6A demethylase FTO controls Th1 differentiation and immunity against infections. Mol Immunol 2025; 183:172-181. [PMID: 40378511 DOI: 10.1016/j.molimm.2025.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2025] [Revised: 05/05/2025] [Accepted: 05/05/2025] [Indexed: 05/19/2025]
Abstract
Antigen-specific effector CD4+ T cells are critical for defense against exogenous pathogens. However, the epigenetic mechanisms underlying CD4+ T cell immune responses, particularly RNA modifications, remain incompletely understood. In this study, we employed a T cell-specific deletion of the fat mass and obesity-associated protein (FTO), a key N6-methyladenosine (m6A) demethylase, to elucidate its role in CD4+ T cell mediated immunity. Our findings demonstrate that FTO is essential for maintaining CD4+ T cell immune responses and protective functions. Specifically, FTO deficiency restricts the expansion of CD4+ T helper (Th)1 effector cells following antigen challenge and results in decreased expression of T-bet and IFN-γ in Th1 cells. Additionally, FTO deficient CD4+ T cells exhibit impaired pathogen elimination. Collectively, our study reveals a novel epigenetic regulatory mechanism in supporting CD4+ T cell differentiation, providing new insights into the post-transcriptional regulation of CD4+ T cell immunity and highlighting the potential for therapeutic strategies.
Collapse
Affiliation(s)
- Zhihong Yao
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Faculty of Clinical Medicine, Hanzhong Vocational and Technical College, Hanzhong 723002, China
| | - Lina Sun
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi 710061, China; Key Laboratory for ImmunoHealth of Shaanxi Province, Xi'an, Shannxi 710061, China
| | - Yang Gao
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Department of Kidney Transplantation, Hospital of Nephropathy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China; Institute of Organ Transplantation, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Yanhong Su
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi 710061, China; Key Laboratory for ImmunoHealth of Shaanxi Province, Xi'an, Shannxi 710061, China
| | - Boxiao He
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi 710061, China; Key Laboratory for ImmunoHealth of Shaanxi Province, Xi'an, Shannxi 710061, China
| | - Yao Ge
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Chen Yang
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Xiaoxuan Jia
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Anjun Jiao
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi 710061, China; Key Laboratory for ImmunoHealth of Shaanxi Province, Xi'an, Shannxi 710061, China
| | - Chenming Sun
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi 710061, China; Key Laboratory for ImmunoHealth of Shaanxi Province, Xi'an, Shannxi 710061, China
| | - Baojun Zhang
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi 710061, China; Key Laboratory for ImmunoHealth of Shaanxi Province, Xi'an, Shannxi 710061, China.
| |
Collapse
|
16
|
Jeong H, Yeo N, Hwang H, Park J, Baek D, Ahn K. N6-methyladenosine modification of HCMV IE1 transcript promotes the repressive state of viral genome to achieve latent infection. Proc Natl Acad Sci U S A 2025; 122:e2508475122. [PMID: 40493198 DOI: 10.1073/pnas.2508475122] [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: 04/16/2025] [Accepted: 04/29/2025] [Indexed: 06/12/2025] Open
Abstract
Human cytomegalovirus (HCMV) is a prevalent pathogen that chronically infects the majority of human population. Among the many features that allow such widespread HCMV infection, one is its ability to maintain a transcriptionally dormant immune-evasive state called latency by suppressing its own major immediate early promoter (MIEP) via epigenetic alterations. In this study, we show a mechanism of MIEP regulation in which the major immediate early (MIE) gene product, immediate early 1 (IE1) transcript, downregulates its own promoter activity in an m6A modification-dependent manner. We found that the loss of the m6A writer, METTL3, in host cells impedes latency establishment in these cells. Through transcriptome-wide m6A profiling of latently infected monocytes, we identified that the major immediate early gene product IE1 transcript is m6A-modified during latent infection. Using IE1-specific m6A-abolished mutants, we found that m6A modification of the IE1 transcript was necessary for the efficient repression of MIEP, and these mutant viruses exhibited a significant defect in establishing latency and progressed toward lytic-like infection in the human monocytic cell line (THP-1) and primary CD14+ monocytes. Our findings demonstrate that HCMV exploits the host m6A machinery to suppress its own lytic program to establish latency and uncover an unexpected role of immediate early gene messenger RNA (mRNA) in regulating its own expression.
Collapse
Affiliation(s)
- Heena Jeong
- School of Biological Sciences, Seoul National University, Seoul 08826, Republic of Korea
- Center for RNA Research, Institute for Basic Science, Seoul 08826, Republic of Korea
| | - Nagyeong Yeo
- School of Biological Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Hyeonseo Hwang
- School of Biological Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Junhyun Park
- School of Biological Sciences, Seoul National University, Seoul 08826, Republic of Korea
- Center for RNA Research, Institute for Basic Science, Seoul 08826, Republic of Korea
| | - Daehyun Baek
- School of Biological Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Kwangseog Ahn
- School of Biological Sciences, Seoul National University, Seoul 08826, Republic of Korea
- Center for RNA Research, Institute for Basic Science, Seoul 08826, Republic of Korea
- Institute for Virus Research, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
17
|
Shi K, Zhang Y, Tao Y, Wang Y, Yang J, Deng R, Yang H. Preamplification-Free Detection of RNA N6-Methyladenosine Modification at Single-Base Resolution Using the CRISPR Tandem Assay. Anal Chem 2025; 97:11454-11461. [PMID: 40411798 DOI: 10.1021/acs.analchem.4c06782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/26/2025]
Abstract
N6-Methyladenosine (m6A) ranks among the most prevalent modifications in RNA, which serves as a biomarker for diseases, such as lung cancer. Herein, we developed a CRISPR/Cas13a-Csm6 tandem assay (termed CRISPRm6A assay) allowing for preamplification-free, sensitive, and rapid detection of RNA m6A modifications. The coupling of Cas13a-Csm6 tandem with MazF endoribonuclease enables the assay to identify m6A RNA with single-base resolution. Compared to the CRISPRm6A assay using Cas13a alone, the tandem CRISPRm6A assay yielded an improved sensitivity for RNA detection by ∼22 times, thus enabling preamplification-free detection of RNA m6A. Particularly, the proposed assay enabled quantification of m6A abundance down to 0.5% at the picomole level in lncRNA MALAT1 and demonstrated a 100% correlation in diagnosing nonsmall cell lung cancer. In summary, the CRISPRm6A assay supports two key applications in biological samples: (1) precise determination of m6A sites and (2) quantitative measurement of m6A fractions. Therefore, the CRISPR tandem method presents a promising tool for RNA epigenetics-based diagnostics.
Collapse
Affiliation(s)
- Kunyu Shi
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yong Zhang
- College of Biomass Science and Engineering, Sichuan University, Chengdu 610065, China
| | - Yiran Tao
- West China-California Research Center for Predictive Intervention Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yuxi Wang
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, West China Hospital, Sichuan University, Chengdu 610041, China
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu 610212, China
| | - Jinliang Yang
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ruijie Deng
- College of Biomass Science and Engineering, Sichuan University, Chengdu 610065, China
| | - Hao Yang
- College of Biomass Science and Engineering, Sichuan University, Chengdu 610065, China
- School of Chemical Engineering, Sichuan University, Chengdu 610065, China
| |
Collapse
|
18
|
Zhang J, Zhou X, Wang B, Yin Y, Wei D, Li K. METTL3/YTHDF3 m 6A axis promotes ferroptosis in diabetic kidney disease by stabilizing TfR1. J Diabetes Investig 2025. [PMID: 40492436 DOI: 10.1111/jdi.70094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 05/13/2025] [Accepted: 05/22/2025] [Indexed: 06/12/2025] Open
Abstract
OBJECTIVE Diabetic kidney disease (DKD) is a common complication of diabetes. N6-Methyladenosine (m6A) modification is a widely studied epigenetic mechanism. Methyltransferase-like (METTL) 3 is a well-studied methyltransferase. This study aimed to investigate the role of METTL3 in DKD and the underlying mechanism. METHODS Thirty-five DKD patients and 28 control volunteers were recruited. Animal and cell DKD models were established. QRT-PCR and Western blot were performed to analyze the expression of METTL3 and fibrosis-related indicators. Cell viability and proliferation were assessed via a cell counting kit-8 and colony formation assays. Ferrous iron (Fe2+), malonaldehyde (MDA), and glutathione (GSH) contents were measured by commercial kits. The interaction between METTL3/YTH N6-methyladenosine RNA binding protein (YTHDF)3 and transferrin receptor-1 (TfR1) was examined through RNA immunoprecipitation and dual-luciferase reporter assays. RESULTS Results showed that METTL3-mediated m6A modification was elevated in kidney tissues of DKD patients and in high glucose (HG)-treated human renal mesangial cells (HRMCs). Besides, HG-treated HRMCs showed increased ferroptosis. In addition, METTL3 inhibition increased cell proliferation and inhibited ferroptosis in HRMCs. Mechanically, the METTL3/YTHDF3 m6A axis enhanced the stability of TfR1 mRNA. Moreover, YTHDF3 inhibition increased cell proliferation and inhibited ferroptosis in HRMCs. Finally, in vivo study results indicated that METTL3 deficiency inhibited ferroptosis and improved pathological damages. CONCLUSIONS In summary, METTL3/YTHDF3 m6A axis promoted ferroptosis in DKD by stabilizing TfR1, which could provide a reference for DKD treatment.
Collapse
Affiliation(s)
- Jinmei Zhang
- Department of Endocrine, Weifang Hospital of Traditional Chinese Medicine, Weifang, Shandong, China
| | - Xiaoping Zhou
- Department of Endocrine, Changyi Traditional Chinese Medicine Hospital, Weifang, Shandong, China
| | - Bin Wang
- Department of Endocrine, Weifang High Tech Rehabilitation Hospital, Weifang, Shandong, China
| | - Yan Yin
- Department of Endocrine, Weifang Hospital of Traditional Chinese Medicine, Weifang, Shandong, China
| | - Daihao Wei
- Department of Endocrine, Weifang Hospital of Traditional Chinese Medicine, Weifang, Shandong, China
| | - Kun Li
- Department of Nephrology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, Shandong, China
| |
Collapse
|
19
|
Huang K, Zhao Y, Lei W, Ge H, Zou T, Li W. Low-Dose Lipopolysaccharide Alleviates Neuronal Apoptosis and Oxidative Stress in Rats with Spinal Cord Injury by Inducing Nrf2 m6A Methylation Modification via Suppressing ALKBH5. Neurochem Res 2025; 50:188. [PMID: 40481942 PMCID: PMC12145315 DOI: 10.1007/s11064-025-04442-7] [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: 09/16/2024] [Revised: 05/07/2025] [Accepted: 05/28/2025] [Indexed: 06/11/2025]
Abstract
This work reported the neuronal protection of low-dose lipopolysaccharide (LD-LPS) after spinal cord injury (SCI). SCI rat model was constructed, after adenovirus-mediated ALKBH5 vectors and shRNA transfection and LD-LPS pre-treatment. Hematoxylin and eosin, Nissl, TUNEL staining of spinal cord tissues were adopted to monitor pathological changes, neuronal survival and apoptosis. PC12 cells transfected with ALKBH5 vectors and ALKBH5/Nrf2 siRNAs were treated by LD-LPS, followed by oxygen and glucose deprivation/reoxygenation (OGD/R). Cell viability and apoptosis were assessed by cell counting kit-8 and TUNEL assays. Neuronal oxidative stress was evaluated by appraising MDA and SOD levels. ALKBH5 and Nrf2 expression was monitored through immunohistochemistry, Western blot and qRT-PCR. Methylated RNA immunoprecipitation assay and Dot-blot experiment were for Nrf2 m6A modification detection, while RNA pull-down assay was for the binding validation between ALKBH5 and Nrf2. In rats with SCI, LD-LPS relieved spinal cord tissue damage and neuronal apoptosis; enhanced neuronal survival; decreased MDA content; elevated SOD activity; down-regulated ALKBH5; up-regulated Nrf2; and facilitated Nrf2 m6A methylation. These above influences by LD-LPS were eliminated by ALKBH5. Similar results were found in the OGD/R-induced PC12 cells after LD-LPS treatment. ALKBH5 significantly blocked Nrf2 m6A methylation, and pulled down Nrf2 protein. In the OGD/R-induced PC12 cells, the repressed oxidative stress and apoptosis by ALKBH5 silencing was abrogated by Nrf2 knockdown. LD-LPS might alleviate neuronal apoptosis and oxidative stress after SCI by facilitating Nrf2 m6A methylation via reducing ALKBH5. It was proposed to be a novel strategy for SCI treatment.
Collapse
Affiliation(s)
- Kun Huang
- Department of Orthopedic Surgery, The First People's Hospital of Yunnan province, Affiliated Hospital of Kunming University of Science and Technology, No. 157, Jinbi Road, Kunming, 650032, China
| | - Yayu Zhao
- Faculty of Medical Science, Kunming University of Science and Technology, Kunming, 650500, China
- Yunnan Key Laboratory of Digital Orthopaedics, Kunming, 650032, China
| | - Wen Lei
- Faculty of Medical Science, Kunming University of Science and Technology, Kunming, 650500, China
- Yunnan Key Laboratory of Digital Orthopaedics, Kunming, 650032, China
| | - Hongran Ge
- Faculty of Medical Science, Kunming University of Science and Technology, Kunming, 650500, China
- Yunnan Key Laboratory of Digital Orthopaedics, Kunming, 650032, China
| | - Tiannan Zou
- Department of Orthopedic Surgery, The First People's Hospital of Yunnan province, Affiliated Hospital of Kunming University of Science and Technology, No. 157, Jinbi Road, Kunming, 650032, China
- Yunnan Key Laboratory of Digital Orthopaedics, Kunming, 650032, China
| | - Weichao Li
- Department of Orthopedic Surgery, The First People's Hospital of Yunnan province, Affiliated Hospital of Kunming University of Science and Technology, No. 157, Jinbi Road, Kunming, 650032, China.
- Faculty of Medical Science, Kunming University of Science and Technology, Kunming, 650500, China.
- Yunnan Key Laboratory of Digital Orthopaedics, Kunming, 650032, China.
| |
Collapse
|
20
|
Zhang S, Wang X, Zhao B, Dai T, Wang L, Tong L, Zeng Q, He Q, Zhang H, Du D. m6A reader YTHDF3 elicits hypertensive effects by degrading XRCC1 mRNA in the rostral ventrolateral medulla. Free Radic Biol Med 2025; 237:S0891-5849(25)00760-9. [PMID: 40490202 DOI: 10.1016/j.freeradbiomed.2025.06.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2025] [Revised: 06/06/2025] [Accepted: 06/07/2025] [Indexed: 06/11/2025]
Abstract
N6-methyladenosine (m6A) modification, mediated by its associated regulatory proteins, has been increasingly recognized for its involvement in diverse pathological conditions. The rostral ventrolateral medulla (RVLM), a key vasomotor center, plays a crucial role in regulating hypertension. However, alterations in m6A and associated regulators including YTHDF3 within the RVLM, along with their functional contributions to hypertension development, remain to be fully elucidated. Here, we identified that YTHDF3 levels were significantly higher in the RVLM of SHRs than in WKY rats. YTHDF3 knockdown in the RVLM of SHRs reduced neuronal excitability, sympathetic tone, and blood pressure (BP). Mechanistically, YTHDF3 promoted the degradation of XRCC1 mRNA in an m6A-dependent manner. YTHDF3 silencing increased XRCC1 expression, facilitating the repair of neuronal DNA oxidative damage and suppressing neuronal apoptosis in vitro and in vivo. These beneficial effects were abrogated by XRCC1 inhibition. Notably, XRCC1 downregulation significantly reversed the suppressive effects on RVLM neuronal excitability, sympathetic tone, and BP in SHRs caused by YTHDF3 repression. The study established, for the first time, the significance of YTHDF3 as a key regulatory factor in the neural regulation of hypertension. Targeting the YTHDF3-XRCC1 axis in the RVLM represents a promising therapeutic strategy for hypertension.
Collapse
Affiliation(s)
- Shuai Zhang
- International Cooperation Laboratory of Molecular Medicine, Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China; College of Agriculture and Bioengineering, Heze University, Heze, Shandong, 274015, China
| | - Xueping Wang
- School of Life Sciences, Shanghai University, Shanghai, 200444, China
| | - Bingjie Zhao
- School of Life Sciences, Shanghai University, Shanghai, 200444, China
| | - Tengteng Dai
- School of Life Sciences, Shanghai University, Shanghai, 200444, China
| | - Linping Wang
- School of Life Sciences, Shanghai University, Shanghai, 200444, China
| | - Lei Tong
- School of Life Sciences, Shanghai University, Shanghai, 200444, China
| | - Qiangcheng Zeng
- College of Life Sciences, Dezhou University, Dezhou, Shandong, 253023, China
| | - Qing He
- College of Life Sciences, Dezhou University, Dezhou, Shandong, 253023, China
| | - Haili Zhang
- College of Agriculture and Bioengineering, Heze University, Heze, Shandong, 274015, China
| | - Dongshu Du
- School of Life Sciences, Shanghai University, Shanghai, 200444, China; College of Life Sciences, Dezhou University, Dezhou, Shandong, 253023, China; College of Agriculture and Bioengineering, Heze University, Heze, Shandong, 274015, China.
| |
Collapse
|
21
|
Zhao Y, Chen X, Zhang X, Liu H. RNA epigenetic modifications as dynamic biomarkers in cancer: from mechanisms to clinical translation. Biomark Res 2025; 13:81. [PMID: 40483535 PMCID: PMC12145623 DOI: 10.1186/s40364-025-00794-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2025] [Accepted: 05/26/2025] [Indexed: 06/11/2025] Open
Abstract
RNA modifications are crucial for post-transcriptional gene regulation. Research on RNA modifications has become a novel frontier of epitranscriptomics. Up to now, over 170 kinds of modifications have been identified on mRNA and diverse non-coding RNA. Three classes of proteins (writers, erasers, and readers) regulate the addition, removal, and identification of epigenetic marks, thus affecting RNA biological functions. Increasing evidence identifies the dysregulation of RNA modifications in different cancer types and the therapeutic potential of targeting RNA-modifying enzymes. The ability of RNA modifications to improve mRNA stability and translation efficacy and decrease immunogenicity has been exploited for the clinical use of mRNA cancer vaccines. This review aims to shed light on several vital cap, tail, and internal modifications of RNA with a focus on the connection between RNA epigenetic pathways and cancer pathogenesis. We further explore the clinical potential of RNA modifications as dynamic biomarkers for cancer diagnosis, prognosis, and therapeutic response prediction, addressing both technological challenges and translational opportunities. Finally, we analyze the limitations of current studies and discuss the research focus in the future.
Collapse
Affiliation(s)
- Yingchao Zhao
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, Hunan, China
- Hunan Engineering Research Center of Skin Health and Disease, Changsha, Hunan, China
| | - Xiang Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, Hunan, China
- Hunan Engineering Research Center of Skin Health and Disease, Changsha, Hunan, China
| | - Xingli Zhang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan, China.
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, Hunan, China.
- Hunan Engineering Research Center of Skin Health and Disease, Changsha, Hunan, China.
| | - Hong Liu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan, China.
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, Hunan, China.
- Hunan Engineering Research Center of Skin Health and Disease, Changsha, Hunan, China.
| |
Collapse
|
22
|
Li G, Chen W, Liu D, Tang S. Recent advances in medicinal chemistry strategies for the development of METTL3 inhibitors. Eur J Med Chem 2025; 290:117560. [PMID: 40147343 DOI: 10.1016/j.ejmech.2025.117560] [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: 02/17/2025] [Revised: 03/20/2025] [Accepted: 03/21/2025] [Indexed: 03/29/2025]
Abstract
N6-methyladenosine (m6A), the most abundant RNA modification in eukaryotic cells, exerts a critical influence on RNA function and gene expression. It has attracted considerable attention within the rapidly evolving field of epitranscriptomics. METTL3 is a key enzyme for m6A modification and is essential for maintaining normal m6A levels. High expression of METTL3 is closely associated with various cancers, including gastric cancer, liver cancer, and leukemia. Inhibiting METTL3 has shown potential in slowing cancer progression, thereby driving the development of METTL3 inhibitors. In this work, we summarize recent advancements in the development of METTL3 inhibitor, with a focus on medicinal chemistry strategies employed during discovery and optimization phases. We explore the application of structure-activity relationship (SAR) studies and protein-targeted degradation techniques, while addressing key challenges associated with their characterization and clinical translation. This review underscores the therapeutic potential of METTL3 inhibitors in modulating epitranscriptomic pathways and aims to offer perspectives for future research in this rapidly evolving field.
Collapse
Affiliation(s)
- Gengwu Li
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China; State Key Laboratory of Respiratory Disease, Institute of Drug Discovery, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Wei Chen
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China; State Key Laboratory of Respiratory Disease, Institute of Drug Discovery, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Dan Liu
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| | - Shibing Tang
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China; State Key Laboratory of Respiratory Disease, Institute of Drug Discovery, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China.
| |
Collapse
|
23
|
Wang J, Yu Y, Xiong Z, Wang P, Wang Z, Du A, Han S, Wang X, Zhang L. METTL3 regulates rifampicin-induced CYP3A4 expression by activating PXR translation and nuclear import and stabilizing CYP3A4 mRNA. Biochem Pharmacol 2025; 239:117016. [PMID: 40466953 DOI: 10.1016/j.bcp.2025.117016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 04/11/2025] [Accepted: 06/02/2025] [Indexed: 06/11/2025]
Abstract
The pregnane X receptor (PXR) activator rifampicin (RIF) plays a critical role in drug-drug interactions (DDIs) by inducing cytochrome P450 (CYP) 3A4 expression. Increasing evidence indicates that n6-methyladenosine (m6A) modification is involved in the regulation of CYP basal expression. Here, we showed its effect on RIF-induced CYP3A4 expression. This study found that m6A levels and methyltransferase-like 3 (METTL3) expression were upregulated in HepG2 and LS174T cells treated with RIF, as well as in mice treated with pregnenolone-16α-carbonitrile (PCN, a typical PXR activator in mice), associated with the expression of CYP3A4 and Cyp3a11 (mouse homolog of human CYP3A4). Specifically, knockdown or overexpression of METTL3 downregulated and upregulated the basal and RIF-induced expression of CYP3A4, respectively. Similar results were obtained in a mouse model with liver-specific knockdown of Mettl3 (homolog of human METTL3) treated with PCN, indicating the involvement of m6A methylation in RIF-induced CYP3A4 expression. Mechanistically, METTL3 promotes PXR nuclear translocation and protein translation, while potentially affecting CYP3A4 mRNA stability through its binding to CYP3A4 mRNA (enhanced by RIF), thereby contributing to RIF-induced upregulation of CYP3A4 expression. Functionally, we observed that METTL3 knockdown or treatment with the METTL3 inhibitor STM2457 reduced the cytotoxicity induced by RIF combined with ritonavir. In conclusion, this study identified a novel mechanism of m6A modification in the regulation of RIF-induced CYP3A4 expression, providing valuable insights into CYP3A4-mediated DDIs.
Collapse
Affiliation(s)
- Jingya Wang
- Department of Pharmacology, School of Basic Medical Sciences, Open and Key Laboratory for Pharmacogenomics at Henan Universities, Zhengzhou University, Zhengzhou 450001, China
| | - Yihang Yu
- Department of Pharmacology, School of Basic Medical Sciences, Open and Key Laboratory for Pharmacogenomics at Henan Universities, Zhengzhou University, Zhengzhou 450001, China
| | - Zaihuan Xiong
- Department of Pharmacology, School of Basic Medical Sciences, Open and Key Laboratory for Pharmacogenomics at Henan Universities, Zhengzhou University, Zhengzhou 450001, China
| | - Pei Wang
- Department of Pharmacology, School of Basic Medical Sciences, Open and Key Laboratory for Pharmacogenomics at Henan Universities, Zhengzhou University, Zhengzhou 450001, China
| | - Zijing Wang
- Department of Pharmacology, School of Basic Medical Sciences, Open and Key Laboratory for Pharmacogenomics at Henan Universities, Zhengzhou University, Zhengzhou 450001, China
| | - Anqi Du
- Department of Pharmacology, School of Basic Medical Sciences, Open and Key Laboratory for Pharmacogenomics at Henan Universities, Zhengzhou University, Zhengzhou 450001, China
| | - Shengna Han
- Department of Pharmacology, School of Basic Medical Sciences, Open and Key Laboratory for Pharmacogenomics at Henan Universities, Zhengzhou University, Zhengzhou 450001, China
| | - Xiaofei Wang
- Academy of Medical Sciences, Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou 450052, China.
| | - Lirong Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Open and Key Laboratory for Pharmacogenomics at Henan Universities, Zhengzhou University, Zhengzhou 450001, China.
| |
Collapse
|
24
|
Li N, Wei X, Dai J, Yang J, Xiong S. METTL3: a multifunctional regulator in diseases. Mol Cell Biochem 2025; 480:3429-3454. [PMID: 39853661 DOI: 10.1007/s11010-025-05208-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: 10/11/2024] [Accepted: 01/04/2025] [Indexed: 01/26/2025]
Abstract
N6-methyladenosine (m6A) methylation is the most prevalent and abundant internal modification of mRNAs and is catalyzed by the methyltransferase complex. Methyltransferase-like 3 (METTL3), the best-known m6A methyltransferase, has been confirmed to function as a multifunctional regulator in the reversible epitranscriptome modulation of m6A modification according to follow-up studies. Accumulating evidence in recent years has shown that METTL3 can regulate a variety of functional genes, that aberrant expression of METTL3 is usually associated with many pathological conditions, and that its expression regulatory mechanism is related mainly to its methyltransferase activity or mRNA posttranslational modification. In this review, we discuss the regulatory functions of METTL3 in various diseases, including metabolic diseases, cardiovascular diseases, and cancer. We focus mainly on recent progress in identifying the downstream target genes of METTL3 and its underlying molecular mechanisms and regulators in the above systems. Studies have revealed that the use of METTL3 as a therapeutic target and a new diagnostic biomarker has broad prospects. We hope that this review can serve as a reference for further studies.
Collapse
Affiliation(s)
- Na Li
- Division of Cardiothoracic and Vascular Surgery, Sino-Swiss Heart-Lung Transplantation Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiang Wei
- Division of Cardiothoracic and Vascular Surgery, Sino-Swiss Heart-Lung Transplantation Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jian Dai
- Department of Critical Care Medicine, Wuhan Wuchang Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Jinfeng Yang
- Department of Medical Affairs, Wuhan Wuchang Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, Hubei, China.
| | - Sizheng Xiong
- Department of Vascular Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.
| |
Collapse
|
25
|
Zheng X, Deng S, Li Y, Luo Z, Gan Z, Zheng Z, Xu R, Xiao S, Cai Y, Meng J, Li L, Li C, Xue X, Dai W, Qin S, Wang M, Zeng K, Xiao Z, Xia L. Targeting m 6A demethylase FTO to heal diabetic wounds with ROS-scavenging nanocolloidal hydrogels. Biomaterials 2025; 317:123065. [PMID: 39756272 DOI: 10.1016/j.biomaterials.2024.123065] [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: 07/24/2024] [Revised: 12/26/2024] [Accepted: 12/27/2024] [Indexed: 01/07/2025]
Abstract
Chronic diabetic wounds are a prevalent and severe complication of diabetes, contributing to higher rates of limb amputations and mortality. N6-methyladenosine (m6A) is a common RNA modification that has been shown to regulate tissue repair and regeneration. However, whether targeting m6A could effectively improve chronic diabetic wound healing remains largely unknown. Here, we found a significant reduction in mRNA m6A methylation levels within human diabetic foot ulcers, and the expression level of fat mass and obesity-associated protein (FTO) was significantly increased. We identified that m6A modifies the RNA of matrix Metalloproteinase 9 (MMP9), a key factor in diabetic wound healing, to regulate its expression. Importantly, we developed a ROS-scavenging nanocolloidal hydrogel loaded with an FTO inhibitor to increase the m6A level of MMP9 RNA in wounds. The hydrogel can effectively accelerate wound healing and skin appendage regeneration in streptozotocin-induced type I diabetic rats at day 14 (approximately 98 % compared to 76.98 % in the control group) and type II diabetic db/db mice at day 20 (approximately 93 % compared to 60 % in the control group). Overall, our findings indicate that targeting m6A with ROS-scavenging hydrogel loaded with FTO inhibitor may be an effective therapeutic strategy for diabetic wound healing.
Collapse
Affiliation(s)
- Xinyao Zheng
- Department of Dermatology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, PR China
| | - Shaohui Deng
- The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Southern Medical University, Dongguan, 523018, PR China
| | - Yuan Li
- Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, PR China
| | - Zhipeng Luo
- Department of Dermatology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, PR China
| | - Ziqi Gan
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, 510055, PR China
| | - Zhaoping Zheng
- Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, PR China
| | - Rui Xu
- Department of Dermatology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, PR China
| | - Shan Xiao
- Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, PR China
| | - Yuxiong Cai
- Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, PR China
| | - Jianfu Meng
- Department of Endocrinology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, PR China
| | - Li Li
- Department of Dermatology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, PR China
| | - Changxing Li
- Department of Dermatology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, PR China
| | - Xiaowen Xue
- Department of Dermatology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, PR China
| | - Wei Dai
- Department of Dermatology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, PR China
| | - Si Qin
- Department of Dermatology, Guangdong Second Provincial General Hospital, Guangzhou, 510317, PR China
| | - Mengying Wang
- Department of Biological Products, Chongqing Institute for Food and Drug Control, Chongqing, 401121, PR China
| | - Kang Zeng
- Department of Dermatology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, PR China.
| | - Zecong Xiao
- Nanomedicine Research Center, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, PR China.
| | - Laixin Xia
- Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, PR China; State Key Laboratory of Organ Failure Research, Southern Medical University, Guangzhou, 510515, PR China.
| |
Collapse
|
26
|
Li L, Chai Q, Guo C, Wei J, Qin Y, Liu H, Lu Z. METTL3-mediated N6-methyladenosine modification contributes to vascular calcification. J Mol Cell Cardiol 2025; 203:22-34. [PMID: 40222552 DOI: 10.1016/j.yjmcc.2025.04.006] [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: 10/29/2024] [Revised: 04/09/2025] [Accepted: 04/10/2025] [Indexed: 04/15/2025]
Abstract
AIM Vascular calcification (VC) is a major adverse cardiovascular event in chronic kidney disease (CKD) patients. N6-methyladenosine (m6A) modification is vital for many biological processes, but its function and possible molecular mechanisms in VC are poorly understood. This study aimed to clarify the function and molecular mechanisms of N6-adenosine-methyltransferase-like 3 (METTL3) in VC. METHODS AND RESULTS The results of the bioinformatic analysis showed that METTL3 expression was significantly upregulated in calcified VSMCs. This finding was corroborated by phosphate-induced VSMCs calcification models and 5/6 nephrectomy-induced CKD mouse VC models. Afterward, Alizarin Red S staining and m6A dot blot analysis demonstrated METTL3 overexpression elevated m6A levels and encouraged calcification in VSMCs and mouse aortic rings, while METTL3 knockdown decreased m6A levels and inhibited calcium deposition in these experimental models. Furthermore, METTL3 promoted VC via the PTEN/AKT pathway, and MeRIP verified that METTL3 induced PTEN mRNA degradation by modifying it with m6A. In addition, molecular docking simulations and DARTS assays revealed that quercetin is a natural small-molecule inhibitor of METTL3. The current investigation demonstrated that quercetin mitigated VC by reducing METTL3-dependent m6A levels in vivo and in vitro. CONCLUSION In conclusion, this study unraveled the pathogenic mechanism of METTL3-mediated m6A modification in VC and provided new insights to establish METTL3 as a therapeutic target for VC.
Collapse
MESH Headings
- Adenosine/analogs & derivatives
- Adenosine/metabolism
- Adenosine/chemistry
- Methyltransferases/metabolism
- Methyltransferases/genetics
- Methyltransferases/chemistry
- Animals
- Vascular Calcification/metabolism
- Vascular Calcification/pathology
- Vascular Calcification/genetics
- Vascular Calcification/etiology
- Mice
- Humans
- PTEN Phosphohydrolase/metabolism
- PTEN Phosphohydrolase/genetics
- Male
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Disease Models, Animal
- Renal Insufficiency, Chronic/metabolism
- Renal Insufficiency, Chronic/pathology
- Mice, Inbred C57BL
- Signal Transduction
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Molecular Docking Simulation
- Proto-Oncogene Proteins c-akt/metabolism
Collapse
Affiliation(s)
- Long Li
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine, Hangzhou, China; Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Hangzhou, China; Department of Cardiology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Quanyou Chai
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine, Hangzhou, China; Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Hangzhou, China; Department of Cardiology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Chunling Guo
- Department of Cardiology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Junyi Wei
- Department of Hematology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Yuqiao Qin
- Department of Hematology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Huimin Liu
- Department of Hematology, The Second Hospital of Shanxi Medical University, Taiyuan, China; Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, China.
| | - Zhaoyang Lu
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine, Hangzhou, China; Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Hangzhou, China; Department of Cardiology, The Second Hospital of Shanxi Medical University, Taiyuan, China.
| |
Collapse
|
27
|
Zhao L, Chen G, Li D, Wang K, Schaefer M, Herr I, Yan B. Baicalein disrupts TGF-β-induced EMT in pancreatic cancer by FTO-dependent m6A demethylation of ZEB1. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119969. [PMID: 40262723 DOI: 10.1016/j.bbamcr.2025.119969] [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: 10/31/2024] [Revised: 04/08/2025] [Accepted: 04/18/2025] [Indexed: 04/24/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive malignancy associated with poor prognosis. Baicalein, a flavonoid extracted from the roots of Scutellaria baicalensis, traditionally used in Chinese medicine, has demonstrated potential in inhibiting cancer development and progression. However, its mechanism of action remains poorly understood, particularly regarding epigenetic gene regulation through m6A RNA methylation. In this study, three human PDAC cell lines and one nonmalignant cell line were employed. The effects of baicalein were examined using multiple assays, including RT-qPCR, MeRIP-qPCR, Western blotting, spheroid formation, RNA stability, and MTT, to evaluate cellular functions and m6A regulation. Baicalein significantly reduced cell viability, migration, invasion, and colony formation. It also downregulated FTO, an enzyme critical for m6A RNA demethylation. Knockdown of FTO replicated the effects of baicalein, underscoring its oncogenic role in PDAC. Bioinformatic analysis identified ZEB1-a key transcription factor in epithelial-to-mesenchymal transition-as an m6A-modified target regulated by FTO. Both baicalein treatment and FTO knockdown enhanced m6A modification and decreased ZEB1 mRNA stability, thereby suppressing stemness-related features. Rescue experiments further confirmed that baicalein disrupts the TGF-β/FTO/ZEB1 signaling axis, highlighting its therapeutic potential in PDAC. This study offers fundamental insights for the development of novel therapeutic strategies targeting PDAC.
Collapse
Affiliation(s)
- Lian Zhao
- Department of General, Visceral & Transplant Surgery, Section Surgical Research, University of Heidelberg, Im Neuenheimer Feld 365, Heidelberg 69120, Germany.
| | - Gong Chen
- Department of General, Visceral & Transplant Surgery, Section Surgical Research, University of Heidelberg, Im Neuenheimer Feld 365, Heidelberg 69120, Germany
| | - Dan Li
- Department of General, Visceral & Transplant Surgery, Section Surgical Research, University of Heidelberg, Im Neuenheimer Feld 365, Heidelberg 69120, Germany
| | - Kangtao Wang
- Department of General, Visceral & Transplant Surgery, Section Surgical Research, University of Heidelberg, Im Neuenheimer Feld 365, Heidelberg 69120, Germany
| | - Michael Schaefer
- Department of General, Visceral & Transplant Surgery, Section Surgical Research, University of Heidelberg, Im Neuenheimer Feld 365, Heidelberg 69120, Germany
| | - Ingrid Herr
- Department of General, Visceral & Transplant Surgery, Section Surgical Research, University of Heidelberg, Im Neuenheimer Feld 365, Heidelberg 69120, Germany
| | - Bin Yan
- Department of General, Visceral & Transplant Surgery, Section Surgical Research, University of Heidelberg, Im Neuenheimer Feld 365, Heidelberg 69120, Germany.
| |
Collapse
|
28
|
Shi JL, Lin CS, Gong MH, Cai ZQ. The emerging roles and mechanisms of FAM83H‑AS1 in cancer: Pathophysiology and therapeutic implications (Review). Oncol Lett 2025; 29:270. [PMID: 40235683 PMCID: PMC11998063 DOI: 10.3892/ol.2025.15016] [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: 11/20/2024] [Accepted: 03/21/2025] [Indexed: 04/17/2025] Open
Abstract
Long non-coding RNAs (lncRNAs) are key regulators of gene expression at transcriptional and post-transcriptional levels and serve roles in tumour progression, cancer diagnosis and prognosis. Among these, family with sequence similarity 83 member H-antisense RNA 1 (FAM83H-AS1) is an oncogenic lncRNA with elevated expression in several malignancies. FAM83H-AS1 promotes cancer cell proliferation, inhibits apoptosis, enhances migration and contributes to chemoresistance through interactions with microRNA (miR)-136-5p, miR-545-3p, miR-15a miR-10a-5p and signalling pathways such as Wnt/β-catenin and Notch receptor. FAM83H-AS1 may be a promising biomarker for cancer diagnosis and prognosis. The present review summarises the expression, mechanism and potential clinical application of FAM83H-AS1 in cancer diagnosis and prognosis.
Collapse
Affiliation(s)
- Jin-Long Shi
- Department of Cardio-Thoracic Surgery, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434000, P.R. China
| | - Chen-Shi Lin
- Department of Oncology, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434000, P.R. China
| | - Ming-Hui Gong
- Intensive Care Unit, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434000, P.R. China
| | - Zhi-Qiang Cai
- Department of Oncology, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434000, P.R. China
| |
Collapse
|
29
|
Han L, Wang R, He M, Chen Z, Wang F. METTL3/YTDHF1 Stabilizes CSRP1 mRNA to Regulate Glycolysis and Promote Acute Myeloid Leukemia Progression. Cell Biochem Biophys 2025; 83:1993-2007. [PMID: 39565517 DOI: 10.1007/s12013-024-01610-4] [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: 10/29/2024] [Indexed: 11/21/2024]
Abstract
CSRP1 (Cysteine and Glycine-Rich Protein 1) is a protein often overactivated in various cancers, promoting cell proliferation and survival, making it a key factor in cancer development. However, it is worth noting that the effect of this protein on the glycolysis process in Acute Myeloid Leukemia (AML) has not yet been studied. This study aims to investigate the role of the METTL3/YTHDF1 axis in regulating Glycolysis and its impact on AML progression by stabilizing CSRP1 mRNA. We analyzed CSRP1 expression in AML tissues and cell lines using quantitative real-time PCR (qRT-PCR) and Western blotting. Functional assays, including cell viability, colony formation, glycolysis related indicators, were performed to assess the impact of CSRP1 knockdown or overexpression on AML cells. RNA immunoprecipitation (RIP) and RNA stability assays were conducted to elucidate the mechanism of METTL3/YTHDF1-mediated regulation of CSRP1 mRNA. CSRP1 was significantly upregulated in AML tissues and cell lines. Knockdown of CSRP1 inhibited AML cell proliferation and glycolysis. Overexpression of CSRP1 promoted AML cell survival. Mechanistically, METTL3 enhanced CSRP1 mRNA stability via m6A modification, recognized and bound by YTHDF1, preventing mRNA degradation. The METTL3/YTHDF1/ CSRP1 axis plays a critical role in AML progression by regulating glycolysis. Targeting this pathway may provide a novel therapeutic strategy for AML treatment.
Collapse
Affiliation(s)
- Lili Han
- Department of Hematology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Ruiyan Wang
- Nursing College, Bengbu Medical College, Bengbu, China
| | - Mengyu He
- Department of Cardiology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Zhenyue Chen
- Nursing College, Bengbu Medical College, Bengbu, China
| | - Feng Wang
- Department of Cardiology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China.
| |
Collapse
|
30
|
Qin T, Jin Y, Qin Y, Yuan F, Lu H, Hu J, Cao Y, Li C. Enhancing m6A modification in the motor cortex facilitates corticospinal tract remodeling after spinal cord injury. Neural Regen Res 2025; 20:1749-1763. [PMID: 39104113 PMCID: PMC11688564 DOI: 10.4103/nrr.nrr-d-23-01477] [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: 09/02/2023] [Revised: 12/26/2023] [Accepted: 02/06/2024] [Indexed: 08/07/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202506000-00026/figure1/v/2024-08-05T133530Z/r/image-tiff Spinal cord injury typically causes corticospinal tract disruption. Although the disrupted corticospinal tract can self-regenerate to a certain degree, the underlying mechanism of this process is still unclear. N6-methyladenosine (m6A) modifications are the most common form of epigenetic regulation at the RNA level and play an essential role in biological processes. However, whether m6A modifications participate in corticospinal tract regeneration after spinal cord injury remains unknown. We found that expression of methyltransferase 14 protein (METTL14) in the locomotor cortex was high after spinal cord injury and accompanied by elevated m6A levels. Knockdown of Mettl14 in the locomotor cortex was not favorable for corticospinal tract regeneration and neurological recovery after spinal cord injury. Through bioinformatics analysis and methylated RNA immunoprecipitation-quantitative polymerase chain reaction, we found that METTL14 regulated Trib2 expression in an m6A-regulated manner, thereby activating the mitogen-activated protein kinase pathway and promoting corticospinal tract regeneration. Finally, we administered syringin, a stabilizer of METTL14, using molecular docking. Results confirmed that syringin can promote corticospinal tract regeneration and facilitate neurological recovery by stabilizing METTL14. Findings from this study reveal that m6A modification is involved in the regulation of corticospinal tract regeneration after spinal cord injury.
Collapse
Affiliation(s)
- Tian Qin
- Department of Spine Surgery and Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Yuxin Jin
- Department of Spine Surgery and Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Yiming Qin
- Department of Spine Surgery and Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Feifei Yuan
- Department of Spine Surgery and Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Hongbin Lu
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Jianzhong Hu
- Department of Spine Surgery and Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Yong Cao
- Department of Spine Surgery and Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Chengjun Li
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| |
Collapse
|
31
|
Lu Z, Lyu Z, Dong P, Liu Y, Huang L. N6-methyladenosine RNA modification in stomach carcinoma: Novel insights into mechanisms and implications for diagnosis and treatment. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167793. [PMID: 40088577 DOI: 10.1016/j.bbadis.2025.167793] [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: 11/21/2024] [Revised: 02/16/2025] [Accepted: 03/03/2025] [Indexed: 03/17/2025]
Abstract
N6-methyladenosine (m6A) RNA methylation is crucially involved in the genesis and advancement of gastric cancer (GC) by controlling various pathobiological aspects including gene expression, signal transduction, metabolism, cell death, epithelial-mesenchymal transition, angiogenesis, and exosome function. Despite its importance, the exact mechanisms by which m6A modification influences GC biology remain inadequately explored. This review consolidates the latest advances in uncovering the mechanisms and diverse roles of m6A in GC and proposes new research and translational directions. Key regulators (writers, readers, and erasers) of m6A, such as METTL3/14/16 and WTAP, significantly affect cancer progression, anticancer immune response, and treatment outcomes. m6A modification also impacts immune cell infiltration and the tumor microenvironment, highlighting its potential as a diagnostic and prognostic marker. Interactions between m6A methylation and non-coding RNAs offer further novel insights into GC development and therapeutic targets. Targeting m6A regulators could enhance immunotherapy response, overcome treatment resistance, and improve oncological and clinical outcomes. Models based on m6A can precisely predict treatment response and prognosis in GC. Additional investigation is needed to fully understand the mechanisms of m6A methylation and its potential clinical applications and relevance (e.g., as precise markers for early detection, prediction of outcome, and response to therapy and as therapeutic targets) in GC. Future research should focus on in vivo studies, potential clinical trials, and the examination of m6A modification in other types of cancers.
Collapse
Affiliation(s)
- Zhengmao Lu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
| | - Zhaojie Lyu
- Department of Urology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Peixin Dong
- Department of Obstetrics and Gynecology, Hokkaido University School of Medicine, Hokkaido University, Sapporo, Japan.
| | - Yunmei Liu
- School of Cultural Heritage and Information Management, Shanghai University, Shanghai, China.
| | - Lei Huang
- Department of Gastroenterology, National Clinical Research Center for Digestive Diseases, Shanghai Institute of Pancreatic Diseases, The First Affiliated Hospital of Naval Medical University/Changhai Hospital, Naval Medical University, Shanghai 200433, China; National Key Laboratory of Immunity and Inflammation, Changhai Clinical Research Unit, The First Affiliated Hospital of Naval Medical University/Changhai Hospital, Naval Medical University, Shanghai 200433, China.
| |
Collapse
|
32
|
Chokkalla AK, Arruri V, Mehta SL, Vemuganti R. Loss of Epitranscriptomic Modification N 6-Methyladenosine (m 6A) Reader YTHDF1 Exacerbates Ischemic Brain Injury in a Sexually Dimorphic Manner. Transl Stroke Res 2025; 16:831-847. [PMID: 38869772 PMCID: PMC12066170 DOI: 10.1007/s12975-024-01267-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 05/28/2024] [Accepted: 06/06/2024] [Indexed: 06/14/2024]
Abstract
N6-Methyladenosine (m6A) is a neuronal-enriched, reversible post-transcriptional modification that regulates RNA metabolism. The m6A-modified RNAs recruit various m6A-binding proteins that act as readers. Differential m6A methylation patterns are implicated in ischemic brain damage, yet the precise role of m6A readers in propagating post-stroke m6A signaling remains unclear. We presently evaluated the functional significance of the brain-enriched m6A reader YTHDF1, in post-stroke pathophysiology. Focal cerebral ischemia significantly increased YTHDF1 mRNA and protein expression in adult mice of both sexes. YTHDF1-/- male, but not female, mice subjected to transient middle cerebral artery occlusion (MCAO) showed worsened motor function recovery and increased infarction compared to sex-matched YTHDF1+/+ mice. YTHDF1-/- male, but not female, mice subjected to transient MCAO also showed significantly perturbed expression of genes related to inflammation, and increased infiltration of peripheral immune cells into the peri-infarct cortex, compared with sex-matched YTHDF1+/+ mice. Thus, this study demonstrates a sexual dimorphism of YTHDF1 in regulating post-ischemic inflammation and pathophysiology. Hence, post-stroke epitranscriptomic regulation might be sex-dependent.
Collapse
Affiliation(s)
- Anil K Chokkalla
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, 53792, USA
- Cellular and Molecular Pathology Graduate Program, University of Wisconsin, Madison, WI, USA
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
| | - Vijay Arruri
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, 53792, USA
| | - Suresh L Mehta
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, 53792, USA
| | - Raghu Vemuganti
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, 53792, USA.
- Cellular and Molecular Pathology Graduate Program, University of Wisconsin, Madison, WI, USA.
- William S. Middleton Memorial Veteran Administration Hospital, Madison, WI, USA.
| |
Collapse
|
33
|
Zhou J, Wang J, Yang L, Fu T, Li H, Shan Y, Gao H, Xie C, Zhang L, Zhang M, Ma J, Liu L, Fang H, Jiang D, Xu M, Pan Q, Gu S. N6-methyadenosine-modified YWHAE mRNA promotes proliferation and inhibits ferroptosis in hepatoblastoma by mediating SLC7A11 expression. Oncogene 2025; 44:1634-1645. [PMID: 40074884 DOI: 10.1038/s41388-025-03334-y] [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: 05/21/2024] [Revised: 02/03/2025] [Accepted: 02/26/2025] [Indexed: 03/14/2025]
Abstract
Hepatoblastoma (HB) is a rare but predominant liver cancer in children, with few treatment choices in advanced stages. YWHAE is closely related to several human diseases and acts as a molecular scaffold for malignant transformation. However, whether YWHAE promotes HB development remains unknown. Conducting RNA and m6A sequencing on HB tissues, we found that YWHAE was upregulated and modified by N6-methyadenosine. Functionally, YWHAE promoted proliferation and inhibited cell death in HB by in vitro and in vivo studies. Mechanistically, METTL3-dependent m6A modification activated YWHAE mRNA expression, and the m6A reader IGF2BP2 recognized and bound to the m6A site on YWHAE mRNA, thereby enhancing the mRNA stability of YWHAE. Interestingly, RNA sequencing revealed that YWHAE knockdown was involved in regulating ferroptosis of HB cells by mediating SLC7A11 expression. Moreover, knockdown of YWHAE significantly increased the levels of lipid ROS and peroxides in HB cells, promoting the susceptibility of HB cells to ferroptosis. In summary, these findings illuminated the role of YWHAE in HB progression and uncovered its relevance to ferroptosis as a new therapeutic target for HB.
Collapse
Affiliation(s)
- Jiquan Zhou
- Department of General Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Road, Shanghai, 200127, China
| | - Jing Wang
- Department of General Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Road, Shanghai, 200127, China
| | - Liyuan Yang
- Department of General Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Road, Shanghai, 200127, China
| | - Tingyi Fu
- Department of General Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Road, Shanghai, 200127, China
| | - Hui Li
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Road, Shanghai, 200127, China
| | - Yuhua Shan
- Department of General Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Road, Shanghai, 200127, China
| | - Hongxiang Gao
- Department of General Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Road, Shanghai, 200127, China
| | - Chenjie Xie
- Department of General Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Road, Shanghai, 200127, China
| | - Lei Zhang
- Department of General Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Road, Shanghai, 200127, China
| | - Min Zhang
- Pediatric Translational Medicine Institute and Pediatric Congenital Heart Disease Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Road, Shanghai, 200127, China
| | - Ji Ma
- Department of Laboratory Medicine, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Road, Shanghai, 200127, China
| | - Li Liu
- Department of Laboratory Medicine, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Road, Shanghai, 200127, China
| | - Houshun Fang
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Road, Shanghai, 200127, China
| | - Dapeng Jiang
- Department of General Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Road, Shanghai, 200127, China
| | - Min Xu
- Department of General Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Road, Shanghai, 200127, China
| | - Qiuhui Pan
- Department of Laboratory Medicine, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Road, Shanghai, 200127, China.
| | - Song Gu
- Department of General Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Road, Shanghai, 200127, China.
- Department of International Medical, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Road, Shanghai, 200127, China.
| |
Collapse
|
34
|
Qiu C, Zheng X, Zhou X, Wang B, Chen T, Xu Y, Yu X, Lu W, Wu Z. The IGF2BP2-circ-DAPK1 axis promotes high-glucose-induced ferroptosis of HUVECs by decreasing NQO1 expression. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167797. [PMID: 40086519 DOI: 10.1016/j.bbadis.2025.167797] [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: 10/08/2024] [Revised: 01/25/2025] [Accepted: 03/07/2025] [Indexed: 03/16/2025]
Abstract
Circular RNAs (circRNAs) are non-coding RNAs with covalently closed loop structures that participate in various biological processes. However, the functions of many circRNAs remain unclear. Endothelial cell dysfunction, which involves abnormal ferroptosis, a unique form of regulated cell death, is a characteristic of various diseases. However, the mechanisms governing ferroptosis in endothelial cells are not fully understood. Here, we investigated the impact of a novel circRNA, circ-DAPK1, on ferroptosis in human umbilical vein endothelial cells (HUVECs) under high-glucose conditions. Our data showed that high-glucose conditions upregulate circ-DAPK1 expression in HUVECs. Overexpression of circ-DAPK1 induced ferroptosis in HUVECs, whereas depletion of circ-DAPK1 mitigated the ferroptosis triggered by high-glucose treatment. Inhibition of ferroptosis reversed the decrease in cell viability induced by high glucose or circ-DAPK1 overexpression. Using RNA immunoprecipitation analyses, we identified several ferroptosis-regulating proteins, including NAD(P)H dehydrogenase [quinone] 1 (NQO1) and insulin-like growth factor 2 mRNA binding protein 2 (IGF2BP2). Mechanistically, circ-DAPK1 interacts with NQO1, enhancing its ubiquitination and accelerating its degradation. NQO1 overexpression partially rescues HUVECs from high-glucose-induced ferroptosis. We also found that IGF2BP2 binds to the m6A site on circ-DAPK1. Depletion of IGF2BP2 in HUVECs reduced circ-DAPK1 expression and inhibited high-glucose-induced ferroptosis. These findings reveal the effects of the IGF2BP2-circ-DAPK1 axis in regulating ferroptosis in HUVECs under high-glucose conditions and extend our understanding of the mechanisms controlling ferroptosis in endothelial cells.
Collapse
Affiliation(s)
- Chenyang Qiu
- Department of Vascular Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiangtao Zheng
- Department of Vascular Surgery, The Second Affiliated Hospital of Wenzhou Medical University, Xueyuan Western Road, Lucheng District, Wenzhou, China
| | - Xiaoxiang Zhou
- Department of Vascular Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Bing Wang
- Department of Vascular Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Tianchi Chen
- Department of Vascular Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yiting Xu
- Department of Vascular Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xinyu Yu
- Department of Vascular Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Wei Lu
- Department of Cardiovascular Surgery, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, China.
| | - Ziheng Wu
- Department of Vascular Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
35
|
Wang C, Liang W, Zhong J, Liu J, Zhou C, Ma C, Liao Y, Gao Y, Zhao J, He Y. m6A-mediated regulation of CPSF6 by METTL3 promotes oxaliplatin resistance in colorectal cancer through enhanced glycolysis. Cell Signal 2025; 130:111676. [PMID: 40010558 DOI: 10.1016/j.cellsig.2025.111676] [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: 10/02/2024] [Revised: 12/16/2024] [Accepted: 02/14/2025] [Indexed: 02/28/2025]
Abstract
Oxaliplatin resistance poses a significant challenge in colorectal cancer (CRC) treatment. Recent studies have implicated CPSF6 in cancer progression and drug resistance, although its role in chemotherapy resistance and regulatory mechanisms is unclear. This study investigates CPSF6's involvement in oxaliplatin resistance in CRC and its regulation via m6A methylation by METTL3. We assessed CPSF6 expression in oxaliplatin-resistant (OxR) CRC cell lines (HT29-OxR and HCT116-OxR) compared to sensitive counterparts using qRT-PCR and Western blotting. CPSF6 was significantly upregulated in OxR cells, and its knockdown reduced cell viability, colony formation, and glycolytic activity while increasing apoptosis. m6A modification of CPSF6 mRNA was elevated in OxR cells, correlating with increased METTL3 expression. METTL3 knockdown decreased CPSF6 levels and m6A enrichment, enhancing mRNA degradation, while its overexpression stabilized CPSF6 mRNA, promoting resistance. Xenograft experiments showed that CPSF6 knockdown suppressed tumor growth and glycolysis. Thus, CPSF6 is identified as a mediator of oxaliplatin resistance in CRC, regulated by the METTL3/m6A axis, suggesting potential therapeutic targets to overcome resistance.
Collapse
Affiliation(s)
- Chengxing Wang
- Department of Gastrointestinal Surgery, Jiangmen Central Hospital, Guangdong 529000, China; Digestive Disease Research Center, Jiangmen Central Hospital, Guangdong 529000, China
| | - Weijun Liang
- Department of Gastrointestinal Surgery, Jiangmen Central Hospital, Guangdong 529000, China; Digestive Disease Research Center, Jiangmen Central Hospital, Guangdong 529000, China
| | - Jietao Zhong
- Digestive Disease Research Center, Jiangmen Central Hospital, Guangdong 529000, China; Department of Gastroenterology, Jiangmen Central Hospital, Guangdong 529000, China
| | - Jiachen Liu
- Department of Nuclear Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangdong 510000, China
| | - Chaorong Zhou
- Department of Gastrointestinal Surgery, Jiangmen Central Hospital, Guangdong 529000, China; Digestive Disease Research Center, Jiangmen Central Hospital, Guangdong 529000, China
| | - Changyi Ma
- Department of Radiology, Jiangmen Central Hospital, Guangdong 529000, China
| | - Yuehua Liao
- Department of Pathology, Jiangmen Central Hospital, Guangdong 529000, China
| | - Yuan Gao
- Department of Gastrointestinal Surgery, Jiangmen Central Hospital, Guangdong 529000, China; Digestive Disease Research Center, Jiangmen Central Hospital, Guangdong 529000, China
| | - Jinglin Zhao
- Department of Gastrointestinal Surgery, Jiangmen Central Hospital, Guangdong 529000, China; Digestive Disease Research Center, Jiangmen Central Hospital, Guangdong 529000, China.
| | - Yaoming He
- Department of Gastrointestinal Surgery, Jiangmen Central Hospital, Guangdong 529000, China; Digestive Disease Research Center, Jiangmen Central Hospital, Guangdong 529000, China.
| |
Collapse
|
36
|
Liang X, Yin S, Hu C, Tang D, Luo G, Liu Z. METTL14 Promotes Ischemic Stroke-induced Brain Injury by Stabilizing HDAC3 Expression in an m6A-IGF2BP3 Mechanism. Cell Biochem Biophys 2025; 83:1897-1907. [PMID: 39448421 DOI: 10.1007/s12013-024-01596-z] [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: 10/09/2024] [Indexed: 10/26/2024]
Abstract
N6-methyladenosine (m6A) modification is the most abundant post-transcriptional modification of mRNAs and has been identified to play critical roles in ischemic stroke (IS). Herein, this study aimed to investigate the function and mechanism of Methyltransferase-like 14 (METTL14) methylase in cerebral IS. Murine BV-2 microglial cell OGD/R models and rat middle cerebral artery occlusion (MCAO) models were established to mimic IS-induced neuronal damage in vitro and brain injury in vivo. Levels of METTL14, Histone Deacetylase 3 (HDAC3) and cGAS-STING axis-related proteins were detected using qRT-PCR or western blotting. Cell proliferation and inflammation were assessed by CCK-8 assay, EdU assay and ELISA. Flow cytometry detected microglia polarization. Cell pyroptosis was analyzed by detecting related-protein markers by western blotting. The m6A modification was determined by methylated RNA immunoprecipitation assay. Brain injury was analyzed by evaluating infarct volume and neurologic score. METTL14 levels were higher in OGD/R-induced microglial cells, primary microglia and infarct brain tissues of rat MCAO models. Functionally, METTL14 silencing reversed OGD/R-induced proliferation inhibition, inflammation and pyroptosis in microglial cells and primary microglia in vitro, and ameliorated cerebral ischemic injury in rat MCAO models. Mechanistically, METTL14 induced HDAC3 m6A modification in an IGF2BP3-dependent manner, and could activate cGAS-STING pathway through HDAC3. Moreover, HDAC3 overexpression reversed the neuroprotective effects of METTL14 silencing. METTL14 silencing reversed ischemic stroke-induced brain injury by inducing HDAC3 m6A modification in an IGF2BP3-dependent mechanism, recommending a novel insight for ameliorating cerebral ischemic stroke.
Collapse
Affiliation(s)
- Xuelin Liang
- The Medical Department of Neurology, Jinshan branch of Shanghai Sixth People's Hospital, Shanghai, China
| | - Songhe Yin
- The Medical Department of Neurology, Jinshan branch of Shanghai Sixth People's Hospital, Shanghai, China
| | - Canfang Hu
- The Medical Department of Neurology, Jinshan branch of Shanghai Sixth People's Hospital, Shanghai, China
| | - Dingzhong Tang
- The Medical Department of Neurology, Jinshan branch of Shanghai Sixth People's Hospital, Shanghai, China
| | - Guojun Luo
- The Medical Department of Neurology, Jinshan branch of Shanghai Sixth People's Hospital, Shanghai, China
| | - Zhen Liu
- The Medical Department of Neurology, Jinshan branch of Shanghai Sixth People's Hospital, Shanghai, China.
| |
Collapse
|
37
|
Li W, Alimujiang A. METTL3 Promotes Osteogenic Differentiation of Human Periodontal Ligament Stem Cells Under the Inflammatory Microenvironment Through the miR-141-3p/ZEB1 Axis. Cell Biochem Biophys 2025; 83:1771-1783. [PMID: 39681812 PMCID: PMC12089236 DOI: 10.1007/s12013-024-01586-1] [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] [Accepted: 10/01/2024] [Indexed: 12/18/2024]
Abstract
Periodontitis, a chronic inflammatory condition, often results in gum tissue damage and can lead to tooth loss. This study explores the role of methyltransferase-like 3 (METTL3) in promoting osteogenic differentiation of human periodontal ligament stem cells (hPDLSCs) within an inflammatory microenvironment. An inflammatory environment was simulated in hPDLSCs using lipopolysaccharide (LPS). Both adipogenic and osteogenic differentiation capacities of hPDLSCs were assessed. In LPS-treated hPDLSCs, METTL3 was overexpressed, and alkaline phosphatase (ALP) staining was performed alongside measurements of ALP activity, pro-inflammatory cytokines, METTL3, miR-141-3p, pri-miR-141, Zinc finger E-box binding homeobox 1 (ZEB1), runt-related transcription factor 2 (RUNX2), osteocalcin (OCN). N6-methyladenosine (m6A) and pri-miR-141 levels were quantified, and the binding of miR-141-3p to ZEB1 was analyzed. The results demonstrated that osteogenic differentiation in hPDLSCs was diminished under inflammatory conditions, coinciding with downregulated METTL3 expression. However, METTL3 overexpression enhanced osteogenic differentiation. METTL3 facilitated the conversion of pri-miR-141 into miR-141-3p via m6A modification, resulting in increased miR-141-3p levels, which in turn suppressed ZEB1 expression. Inhibition of miR-141-3p or overexpression of ZEB1 partially counteracted the positive effects of METTL3 on osteogenic differentiation. In conclusion, these findings suggest that METTL3-mediated m6A modification promotes osteogenic differentiation of hPDLSCs within an inflammatory microenvironment through the miR-141-3p/ZEB1 axis.
Collapse
Affiliation(s)
- Weijia Li
- School of Stomatology, Jinan University, Guangzhou, China
| | | |
Collapse
|
38
|
Han Y, Sun J, Yao M, Miao L, Li M. Biological roles of enhancer RNA m6A modification and its implications in cancer. Cell Commun Signal 2025; 23:254. [PMID: 40448182 DOI: 10.1186/s12964-025-02254-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2025] [Accepted: 05/17/2025] [Indexed: 06/02/2025] Open
Abstract
Enhancers, as distal cis-regulatory elements in the genome, have a pivotal influence on orchestrating precise gene expression. Enhancer RNAs (eRNAs), transcribed from active enhancer regions, are increasingly recognized as key regulators of transcription. N6-methyladenosine (m6A), the most plentiful internal modification in eukaryotic mRNAs, has garnered significant research interest in recent years. With advancements in high-throughput sequencing technologies, it has been established that m6A modifications are also present on eRNAs. An accumulative body of evidence demonstrates that aberrant enhancers, eRNAs, and m6A modifications are intimately connected with carcinoma onset, progression, invasion, metastasis, treatment response, drug resistance, and prognosis. However, the underlying molecular mechanisms governing m6A modification of eRNAs in cancer remain elusive. Here, we review and synthesize current understanding of the regulatory roles of enhancers, eRNAs, and m6A modifications in cancer. Furthermore, we investigate the possible roles of eRNAs m6A modification in tumorigenesis based on existing literature, offering novel perspectives and directions for future research on epigenetic regulatory mechanisms in cancer cells.
Collapse
Affiliation(s)
- Yangyang Han
- Department of Biology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi, 830017, China
- Xinjiang Key Laboratory of Molecular Biology for Endemic Diseases, Xinjiang Medical University, Urumqi, 830017, China
| | - Jingqi Sun
- Department of Biology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi, 830017, China
| | - Minghui Yao
- Department of Biology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi, 830017, China
| | - Liying Miao
- Department of Biology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi, 830017, China
| | - Mengjia Li
- Department of Biology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi, 830017, China.
- Xinjiang Key Laboratory of Molecular Biology for Endemic Diseases, Xinjiang Medical University, Urumqi, 830017, China.
| |
Collapse
|
39
|
Chun H, Baima K. METTL3-Driven m 6A Modifications in Esophageal Squamous Cell Carcinoma: Emerging Mechanisms, Biomarker Potential, and Therapeutic Innovations. Eur J Pharmacol 2025:177785. [PMID: 40449646 DOI: 10.1016/j.ejphar.2025.177785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2025] [Revised: 05/20/2025] [Accepted: 05/28/2025] [Indexed: 06/03/2025]
Abstract
Esophageal squamous cell carcinoma (ESCC) remains a highly aggressive malignancy with limited diagnostic and therapeutic advancements, underscoring the urgent need for novel biomarkers and targeted therapies. Methyltransferase-like 3 (METTL3), a pivotal regulator of N6-methyladenosine (m6A) RNA modification, has emerged as a critical player in cancer pathogenesis. This review synthesizes current evidence to elucidate METTL3's multifaceted roles in ESCC progression, metastasis, and therapeutic resistance. Mechanistically, METTL3 promotes tumorigenesis by orchestrating m6A-dependent regulation of oncogenic pathways, including EGR1/Snail, Notch, and c-Myc signaling, while suppressing tumor suppressors like APC. Clinically, METTL3 overexpression correlates with advanced tumor stage, lymph node metastasis, and poor prognosis, highlighting its diagnostic and prognostic utility. Furthermore, METTL3 enhances radioresistance via DNA repair modulation and drives metabolic reprogramming through targets such as GLUT1 and GLS2. Emerging therapeutic strategies targeting METTL3, including small-molecule inhibitors [e.g., Elvitegravir] and RNA-based interventions, demonstrate preclinical efficacy in suppressing ESCC proliferation and metastasis. This review also identifies critical knowledge gaps, such as the interplay between METTL3 and tumor microenvironment dynamics, and advocates for multicenter studies to validate its clinical applicability. Collectively, our findings position METTL3 as a promising biomarker and a tractable therapeutic target, offering actionable insights to advance ESCC management.
Collapse
Affiliation(s)
- Hua Chun
- College of Medicine, Tibet University, Tibet, 850000, China
| | - Kangzhuo Baima
- College of Medicine, Tibet University, Tibet, 850000, China.
| |
Collapse
|
40
|
Tian P, Zhao J, Guo J, Guo G, Zeng L, Lei Q, Chen W, Fu X, Shi X, Xu Z, Zhao D, Zhang Z, Zhang H. Lnc-HZ06 down-regulates HIF1α protein levels in CoCl 2-exposed hypoxic trophoblast cells and villous tissues of miscarriage patients. THE SCIENCE OF THE TOTAL ENVIRONMENT 2025; 978:179477. [PMID: 40262219 DOI: 10.1016/j.scitotenv.2025.179477] [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: 01/01/2025] [Revised: 04/15/2025] [Accepted: 04/16/2025] [Indexed: 04/24/2025]
Abstract
Hypoxia plays significant roles in various biological processes. In recent study, we have found that a novel lnc-HZ06 promotes the SUMOylation of HIF1α in hypoxic human trophoblast cells. Since environmental cobalt (Co) exposure causes trophoblast cell hypoxia, whether and how lnc-HZ06 might regulate the protein levels of HIF1α, an important biomarker of hypoxia, in CoCl2-exposed hypoxic trophoblast cells is still unexplored. In this study, we find that lnc-HZ06 is highly expressed in CoCl2-exposed trophoblast cells; and lnc-HZ06 further down-regulates HIF1α protein levels. In details, (1) lnc-HZ06 up-regulates METTL14 (methyltransferase-like 14) and increases m6A (N6-methyladenosine) RNA modification levels on VHL (a ubiquitin E3 ligase of HIF1α) mRNA, and thus enhances its mRNA stability and up-regulates VHL mRNA levels. (2) VHL interacts with the SUMOylated HIF1α and promotes the ubiquitination of HIF1α, and finally lnc-HZ06 promotes the ubiquitination degradation of HIF1α protein in CoCl2-exposed hypoxic trophoblast cells. Therefore, lnc-HZ06 promotes VHL-mediated HIF1α protein degradation and down-regulates HIF1α protein levels. The cellular mechanisms in hypoxic trophoblast cells were partially consistent to those in villous tissues of patients with unexplained miscarriage (UM), expect for no significantly different Co content in UM and healthy control (HC) villous tissues. Collectively, this study discovers novel regulatory roles of lnc-HZ06 and m6A modification and post-translational modification (SUMO/Ubiquitin) in HIF1α protein levels in hypoxic human trophoblast cells.
Collapse
Affiliation(s)
- Peng Tian
- Department of Pathology, The Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi 563003, China
| | - Jingsong Zhao
- Research Center for Environment and Female Reproductive Health, the Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, China
| | - Jiarong Guo
- Research Center for Environment and Female Reproductive Health, the Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, China
| | - Geng Guo
- Department of Emergency, Cerebrovascular Disease Center, First Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Liqin Zeng
- Department of Obstetrics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong Province, China
| | - Qiong Lei
- Department of Obstetrics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong Province, China
| | - Weina Chen
- Research Center for Environment and Female Reproductive Health, the Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, China
| | - Xia Fu
- Department of Nursing, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen 518033, China
| | - Xianjie Shi
- Department of Hepatobiliary and Pancreatic Surgery, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Zhongyan Xu
- Clinical Laboratory, Bethune International Peace Hospital, 398 Zhongshan Road, Shijiazhuang, Hebei 050082, China.
| | - Depeng Zhao
- Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, Guangdong, China.
| | - Zhihong Zhang
- MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Shanxi Medical University, Taiyuan 030001, China.
| | - Huidong Zhang
- Research Center for Environment and Female Reproductive Health, the Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, China; MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Shanxi Medical University, Taiyuan 030001, China.
| |
Collapse
|
41
|
Guo J, Zhang T, Xie H, Hu H, Shi C, Zhao Y, Yin J, Xu G, Wu Z, Wang P, Liu J, Liu P, Zhong K, Chen F, Chen J, Yang J. An m 6A methyltransferase confers host resistance by degrading viral proteins through ubiquitination. Nat Commun 2025; 16:4821. [PMID: 40410199 PMCID: PMC12102378 DOI: 10.1038/s41467-025-60199-1] [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: 09/19/2024] [Accepted: 05/19/2025] [Indexed: 05/25/2025] Open
Abstract
Posttranscriptional and posttranslational modifications play crucial roles in plant immunity. However, how plants fine-tune such modifications to activate antiviral immunity remains unknown. Here, we report that the m6A methyltransferase TaHAKAI is utilized by wheat yellow mosaic virus (WYMV) to increase viral genomic m6A modification and promote viral replication. However, TaHAKAI also functions as an E3 ligase that targets the viral RNA silencing suppressor P2 for degradation and inhibits viral infection. A major allele of TaHAKAI in a susceptible cultivar exhibited reduced E3 ligase activity but not m6A methyltransferase activity, promoting viral infection. Interestingly, TaHAKAIR attenuates the stability of TaWPS1 (Wheat paired spikelets 1, WPS1) mRNA, the negative regulator of spike development, which might increase panicle length and spikelet number by modulating its m6A modification. Our study reveals a mechanism for balancing disease resistance and yield by fine-tuning m6A modification and ubiquitination.
Collapse
Affiliation(s)
- Jun Guo
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Biotechnology in Plant Protection of MARA, Key Laboratory of Green Plant Protection of Zhejiang Province, Institute of Plant Virology, Ningbo University, Ningbo, China
| | - Tianye Zhang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Biotechnology in Plant Protection of MARA, Key Laboratory of Green Plant Protection of Zhejiang Province, Institute of Plant Virology, Ningbo University, Ningbo, China
| | - Haoxin Xie
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Biotechnology in Plant Protection of MARA, Key Laboratory of Green Plant Protection of Zhejiang Province, Institute of Plant Virology, Ningbo University, Ningbo, China
| | - Haichao Hu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Biotechnology in Plant Protection of MARA, Key Laboratory of Green Plant Protection of Zhejiang Province, Institute of Plant Virology, Ningbo University, Ningbo, China
| | - Chaonan Shi
- State Key Laboratory of High-Efficiency Production of Wheat-Maize Double Cropping /Agronomy College, Henan Agricultural University, Zhengzhou, China
| | - Yingjie Zhao
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Biotechnology in Plant Protection of MARA, Key Laboratory of Green Plant Protection of Zhejiang Province, Institute of Plant Virology, Ningbo University, Ningbo, China
| | - Jingliang Yin
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Biotechnology in Plant Protection of MARA, Key Laboratory of Green Plant Protection of Zhejiang Province, Institute of Plant Virology, Ningbo University, Ningbo, China
| | - Gecheng Xu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Biotechnology in Plant Protection of MARA, Key Laboratory of Green Plant Protection of Zhejiang Province, Institute of Plant Virology, Ningbo University, Ningbo, China
| | - Zechi Wu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Biotechnology in Plant Protection of MARA, Key Laboratory of Green Plant Protection of Zhejiang Province, Institute of Plant Virology, Ningbo University, Ningbo, China
| | - Pengkun Wang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Biotechnology in Plant Protection of MARA, Key Laboratory of Green Plant Protection of Zhejiang Province, Institute of Plant Virology, Ningbo University, Ningbo, China
| | - Jiaqian Liu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Biotechnology in Plant Protection of MARA, Key Laboratory of Green Plant Protection of Zhejiang Province, Institute of Plant Virology, Ningbo University, Ningbo, China
| | - Peng Liu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Biotechnology in Plant Protection of MARA, Key Laboratory of Green Plant Protection of Zhejiang Province, Institute of Plant Virology, Ningbo University, Ningbo, China
| | - Kaili Zhong
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Biotechnology in Plant Protection of MARA, Key Laboratory of Green Plant Protection of Zhejiang Province, Institute of Plant Virology, Ningbo University, Ningbo, China
| | - Feng Chen
- State Key Laboratory of High-Efficiency Production of Wheat-Maize Double Cropping /Agronomy College, Henan Agricultural University, Zhengzhou, China
| | - Jianping Chen
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Biotechnology in Plant Protection of MARA, Key Laboratory of Green Plant Protection of Zhejiang Province, Institute of Plant Virology, Ningbo University, Ningbo, China.
| | - Jian Yang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Biotechnology in Plant Protection of MARA, Key Laboratory of Green Plant Protection of Zhejiang Province, Institute of Plant Virology, Ningbo University, Ningbo, China.
| |
Collapse
|
42
|
Jin C, Ye Y, Gao L, Zhong Z, Zhou C, Wu X, Li X, Zhou G, Chen S, Wei Y, Cai L, Liu S, Xu J. Biological function of RNA-binding proteins in myocardial infarction: a potential emerging therapeutic limelight. Cell Biosci 2025; 15:65. [PMID: 40413549 PMCID: PMC12102849 DOI: 10.1186/s13578-025-01408-8] [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: 12/16/2024] [Accepted: 05/08/2025] [Indexed: 05/27/2025] Open
Abstract
Myocardial infarction (MI) is currently one of the most fatal cardiovascular diseases worldwide. The screening, treatment, and prognosis of MI are top priorities for cardiovascular centers globally due to its characteristic occult onset, high lethality, and poor prognosis. MI is caused by coronary artery occlusion induced by coronary atherosclerotic plaque blockage or other factors, leading to ischemic necrosis and apoptosis of cardiomyocytes. Although significant advancements have been made in the study of cardiomyocytes at the cellular and molecular levels, RNA-binding proteins (RBPs) have not been extensively explored in the context of MI. RBPs, as key regulators coordinating cell differentiation and tissue homeostasis, exhibit specific functions in gene transcription, RNA modification and processing, and post-transcriptional gene expression. By binding to their target RNA, RBPs coordinate various RNA dynamics, including cellular metabolism, subcellular localization, and translation efficiency, thereby controlling the expression of encoded proteins. Classical RBPs, including HuR, hnRNPs, and RBM family molecules, have been identified as critical regulators in myocardial hypoxia, oxidative stress, pro-inflammatory responses, and fibrotic repair. These RBPs exert their effects by modulating key pathophysiological pathways in MI, thereby influencing specific cardiac outcomes. Additionally, specific RBPs, such as QKI and fused in sarcoma (FUS), are implicated in the apoptotic pathways activated during MI. This apoptotic pathway represents a significant molecular phenotype in MI, offering novel perspectives and insights for mitigating cardiomyocyte apoptosis and attenuating the progression of MI. Therefore, this review systematically summarizes the role of RBPs in the main pathophysiological stages of MI and explores their potential therapeutic prospects.
Collapse
Affiliation(s)
- Chenyang Jin
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yutong Ye
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Longzhe Gao
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zikan Zhong
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Changzuan Zhou
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoyu Wu
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xudong Li
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Genqing Zhou
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Songwen Chen
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yong Wei
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lidong Cai
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shaowen Liu
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Juan Xu
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
43
|
Jiang L, Xiao J, Xie L, Zheng F, Ge F, Zhao X, Qiang R, Fang J, Liu Z, Xu Z, Chen R, Wang D, Liu Y, Xia Q. The emerging roles of N6-methyladenosine (m6A) deregulation in polycystic ovary syndrome. J Ovarian Res 2025; 18:107. [PMID: 40410881 PMCID: PMC12100877 DOI: 10.1186/s13048-025-01690-7] [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: 03/05/2025] [Accepted: 05/08/2025] [Indexed: 05/25/2025] Open
Abstract
Polycystic ovary syndrome (PCOS) is an endocrine metabolic syndrome characterized by ovulation disorders, hyperandrogenemia, and polycystic ovaries, which seriously affect the psychological and physical health of childbearing women. N6-methyladenosine (m6A), as the most common mRNA epigenetic modification in eukaryotes, is vital for developing the female reproductive system and reproductive diseases. In recent years, an increasing number of studies have revealed the mechanisms by which m6A modifications and their related proteins are promoting the development of PCOS, including writers, erasers and readers. In this work, we reviewed the research progress of m6A in the pathophysiological development of PCOS from the starting point of PCOS clinical features, included the recent studies or those with significant findings related to m6A and PCOS, summarized the current commonly used therapeutic methods in PCOS and the possible targeted therapies against the m6A mechanism, and looked forward to future research directions of m6A in PCOS. With the gradual revelation of the m6A mechanism, m6A and its related proteins are expected to become a great field for PCOS treatment.
Collapse
Affiliation(s)
- Leyi Jiang
- Department of Gynecology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Department of Neurosurgery, the Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310000, China
- Department of Neurosurgery, Ningbo Hospital, Zhejiang University School of Medicine, Ningbo, 315010, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, China
| | - Jiaying Xiao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, China
| | - Liangzhen Xie
- Department of Gynecology, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Feifei Zheng
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, China
| | - Fangliang Ge
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, China
| | - Xue Zhao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, China
| | - Ruonan Qiang
- Department of Gynecology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Jie Fang
- Department of Gynecology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Zhinan Liu
- Department of Gynecology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Zihan Xu
- Department of Gynecology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Ran Chen
- Department of Gynecology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Dayong Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, China.
| | - Yanfeng Liu
- Department of Gynecology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China.
| | - Qing Xia
- Department of Gynecology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
44
|
Shen W, Ma Y, Yang C, Yan S, Ye K. Role of N6-methyladenosine methyltransferase component RBM15 in cancer progression and its therapeutic potential. Discov Oncol 2025; 16:855. [PMID: 40402374 PMCID: PMC12098234 DOI: 10.1007/s12672-025-02644-7] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 05/09/2025] [Indexed: 05/23/2025] Open
Abstract
Cancer ranks as a primary cause of mortality globally, and the study of its molecular markers and regulatory mechanisms holds paramount importance. N6-methyladenosine (m⁶A) represents the predominant modification in messenger RNA (mRNA), influencing key biological processes including RNA stability, splicing, and translation. The dynamic modulation of m⁶A modification is mediated by an array of enzymes comprising methyltransferases ("writers"), demethylases ("erasers"), and m⁶A-binding proteins ("readers").As a pivotal member of the m⁶A "writer" family, RNA binding motif protein 15 (RBM15) facilitates the recruitment of the methyltransferase complex (MTC) to mRNA, thus orchestrating the addition of m⁶A modifications. Although prior research has underscored the critical role of m⁶A in oncogenesis, the precise mechanisms through which RBM15 operates in cancer are yet to be elucidated. This study endeavors to elucidate the structural characteristics and functional roles of RBM15, investigate its potential regulatory mechanisms across diverse tumors, uncover its distinct functions in tumor genesis, progression, and metastasis, and evaluate the therapeutic potential of targeting RBM15 in cancer treatment.
Collapse
Affiliation(s)
- Wenxiang Shen
- Department of Orthopedics, Second Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory of Bone and Joint Diseases of Gansu Province, Second Hospital of Lanzhou University, Lanzhou, China
| | - Yulong Ma
- Department of Orthopedics, Second Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory of Bone and Joint Diseases of Gansu Province, Second Hospital of Lanzhou University, Lanzhou, China
| | - Chunwang Yang
- Department of Orthopedics, Second Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory of Bone and Joint Diseases of Gansu Province, Second Hospital of Lanzhou University, Lanzhou, China
| | - Shishun Yan
- Department of Orthopedics, Second Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory of Bone and Joint Diseases of Gansu Province, Second Hospital of Lanzhou University, Lanzhou, China
| | - Kaishan Ye
- Department of Orthopedics, Second Hospital of Lanzhou University, Lanzhou, China.
- Key Laboratory of Bone and Joint Diseases of Gansu Province, Second Hospital of Lanzhou University, Lanzhou, China.
| |
Collapse
|
45
|
Li X, Chen WW, Wu JJ, Yuan ZD, Yuan FL, Chen J. METTL3-dependent epigenetic regulation of ULK2 autophagy in hypertrophic scarring. Int J Biol Macromol 2025; 315:144507. [PMID: 40409645 DOI: 10.1016/j.ijbiomac.2025.144507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2025] [Revised: 05/08/2025] [Accepted: 05/20/2025] [Indexed: 05/25/2025]
Abstract
Increased autophagy in fibroblasts drives their differentiation into myofibroblasts, a key process in dermal fibrosis during hypertrophic scar (HS) progression. While N6-methyladenosine (m6A) modification is implicated in fibrosis and autophagy, its mechanistic role in HS remains unclear. In this study, we investigated the involvement of fibroblast autophagy in HS progression and the regulatory mechanisms underlying this process. Our findings demonstrated that HS development is associated with significant autophagy in both human patients and rabbit models, as evidenced by the activation of fibroblast-associated alpha-smooth muscle actin (α-SMA) and type I collagen. Pharmacological inhibition of autophagy using 3-methyladenine effectively suppressed fibroblast-to-myofibroblast differentiation. We further discovered that excessive m6A modifications enhanced autophagy in fibroblasts derived from HS tissues. Mechanistically, we elucidated that methyltransferase-like 3 (METTL3)-mediated m6A modification upregulated unc-51-like kinase 2 (ULK2), a key regulator of autophagy initiation, through techniques such as m6A RNA immunoprecipitation sequencing (MeRIP-seq), qRT-PCR, and Western blotting. Silencing METTL3 impaired autophagic flux, as confirmed by transmission electron microscopy and LC3-II/I ratio analysis, thereby inhibiting fibroblast-to-myofibroblast differentiation. Notably, subcutaneous injection of METTL3 small interfering RNA (siRNA) attenuated cellular autophagy in HS tissues and mitigated HS formation in rabbit ears. These results clarify the causal relationship between METTL3-mediated m6A modification, autophagy, and fibroblast-to-myofibroblast differentiation, providing a mechanistic basis for the therapeutic potential of targeting METTL3 in HS treatment.
Collapse
Affiliation(s)
- Xia Li
- Institute of Integrated Chinese and Western Medicine, The Hospital Affiliated to Jiangnan University, Wuxi, Jiangsu 214041, China
| | - Wei-Wei Chen
- Institute of Integrated Chinese and Western Medicine, The Hospital Affiliated to Jiangnan University, Wuxi, Jiangsu 214041, China
| | - Jun-Jie Wu
- Institute of Integrated Chinese and Western Medicine, The Hospital Affiliated to Jiangnan University, Wuxi, Jiangsu 214041, China
| | - Zheng-Dong Yuan
- Institute of Integrated Chinese and Western Medicine, The Hospital Affiliated to Jiangnan University, Wuxi, Jiangsu 214041, China
| | - Feng-Lai Yuan
- Institute of Integrated Chinese and Western Medicine, The Hospital Affiliated to Jiangnan University, Wuxi, Jiangsu 214041, China.
| | - Jinghua Chen
- Institute of Integrated Chinese and Western Medicine, The Hospital Affiliated to Jiangnan University, Wuxi, Jiangsu 214041, China; Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Pharmaceutical Sciences, Jiangnan University, Wuxi 214122, China.
| |
Collapse
|
46
|
Yang Y, Tan B, Xiao L, Huang H, Xu W, Su J, Zhao Y, Hong L, Cai G, Li Z, Dai L, Gu T. GPCPD1 is regulated by METTL3/IGF2BP3 dependent m 6A modification to affect pig muscle fiber type conversion. Gene 2025; 962:149578. [PMID: 40404071 DOI: 10.1016/j.gene.2025.149578] [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: 01/25/2025] [Revised: 04/18/2025] [Accepted: 05/18/2025] [Indexed: 05/24/2025]
Abstract
The N6-methyladenosine (m6A) modification is the most prevalent and abundant RNA modification in eukaryotes. In our previous study, we identified that the glycerophosphocholine phosphodiesterase 1 (GPCPD1) gene was differentially expressed and diverse m6A modificated in the pig soleus and extensor digitorum longus muscles. In this study, we further investigated the function of GPCPD1 gene in pig muscle development. We found that GPCPD1 inhibited myogenic differentiation and promoted the conversion of fast-twitch to slow-twitch fibers in both porcine muscle satellite cells (PSCs) and in mouse model. We also found that the expression of GPCPD1 was affected by m6A methyltransferase METTL3. The methylated GPCPD1 gene was recognized by the m6A reader protein IGF2BP3, which further modulated the stability of GPCPD1 mRNA. This study provided novel evidence into m6A regulation in pig muscle development and illustrated the new post-transcriptional regulation way in GPCPD1 expression.
Collapse
Affiliation(s)
- Yuming Yang
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, Guangzhou, China
| | - Baohua Tan
- The Key Laboratory of Advanced Interdisciplinary Studies, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Liyao Xiao
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, Guangzhou, China
| | - Huijun Huang
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, Guangzhou, China
| | - Weihua Xu
- College of Life Science, Longyan University, Longyan, China
| | - Jiawei Su
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, Guangzhou, China
| | - Yaolu Zhao
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, China
| | - Linjun Hong
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, Guangzhou, China
| | - Gengyuan Cai
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, Guangzhou, China
| | - Zicong Li
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, Guangzhou, China
| | - Lihe Dai
- Key Laboratory of Animal Genetics and Breeding of Zhejiang Province, Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, Hangzhou, China.
| | - Ting Gu
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, Guangzhou, China.
| |
Collapse
|
47
|
Ding Z, Wang X, Guo S, Kang Y, Cao M, Hu L, Zhang B, Xiong L, Pei J, Yang T, Guo X. Characteristic analysis of N 6-methyladenine in different parts of yak epididymis. BMC Genomics 2025; 26:500. [PMID: 40389816 PMCID: PMC12087211 DOI: 10.1186/s12864-025-11684-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: 06/11/2024] [Accepted: 05/07/2025] [Indexed: 05/21/2025] Open
Abstract
BACKGROUND The epididymis is essential for sperm maturation. During sperm maturation, markable alterations of the payload of small noncoding RNAs are observed in the epididymis, which indicated the role of epigenetic alterations in sperm maturation. However, the N6-Methyladenosine (m6A) modification profile of the epididymis remains unelucidated. Therefore, in this study, we assessed the m6A modification levels in the caput, corpus, and cauda of the yak epididymis using a combination of methylated RNA immunoprecipitation and RNA sequencing. RESULTS The m6A levels were significantly increased in the corpus of the epididymis. Functional enrichment analysis of differentially methylated RNA (DMR) between the corpus and caput group revealed the significant enrichment of DMRs in the gap junction, ErbB signaling pathway, and mTOR signaling pathway, which participate in cell communication and sperm maturation. In addition, the DMRs of cauda-vs-corpus group were enriched in apoptosis, the FoxO signaling pathway, the PI3K-Akt signaling pathway, and the tumor necrosis factor signaling pathway that were associated with sperm autophagy, oxidative stress, and sperm maturation. Furthermore, we identified the key genes exhibiting significant changes in m6A levels but with no differences in RNA levels, including YY1-associated factor 2, forkhead box J2, and forkhead box O1. This finding indicated that m6A modifications affect these genes during translation, thereby participating in sperm maturation. CONCLUSIONS In summary, we generated the m6A profile of the yak epididymis, which will aid in further elucidating the maturation process of sperm and reveal more information related to male infertility.
Collapse
Affiliation(s)
- Ziqiang Ding
- Key Laboratory of Yak Breeding of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, 730050, China
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou, 730050, China
| | - Xingdong Wang
- Key Laboratory of Yak Breeding of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, 730050, China
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou, 730050, China
| | - Shaoke Guo
- Key Laboratory of Yak Breeding of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, 730050, China
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou, 730050, China
| | - Yandong Kang
- Key Laboratory of Yak Breeding of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, 730050, China
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou, 730050, China
| | - Mengli Cao
- Key Laboratory of Yak Breeding of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, 730050, China
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou, 730050, China
| | - Liyan Hu
- Key Laboratory of Yak Breeding of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, 730050, China
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou, 730050, China
| | - Ben Zhang
- Key Laboratory of Yak Breeding of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, 730050, China
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou, 730050, China
| | - Lin Xiong
- Key Laboratory of Yak Breeding of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, 730050, China
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou, 730050, China
| | - Jie Pei
- Key Laboratory of Yak Breeding of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, 730050, China
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou, 730050, China
| | - Tao Yang
- Haibei Animal Husbandry Science and Technology Demonstration Park Management Committee, Haibei, 810299, China.
| | - Xian Guo
- Key Laboratory of Yak Breeding of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, 730050, China.
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou, 730050, China.
| |
Collapse
|
48
|
Elsabbagh RA, Abdelhady G, Urlaub D, Sandusky M, Khorshid O, Gad MZ, Abou-Aisha K, Watzl C, Rady M. N 6-methyladenosine RNA base modification regulates NKG2D-dependent and cytotoxic genes expression in natural killer cells. BMC Med Genomics 2025; 18:91. [PMID: 40389988 PMCID: PMC12090489 DOI: 10.1186/s12920-025-02147-y] [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: 01/02/2024] [Accepted: 04/17/2025] [Indexed: 05/21/2025] Open
Abstract
BACKGROUND Breast cancer (BC) is the most commonly diagnosed cancer in women. N6-methyladenosine (m6A) is the most prevalent internal modification in mammalian mRNAs and plays a crucial role in various biological processes. However, its function in Natural killer (NK) cells in BC remains unclear. NK cells are essential for cancer immunosurveillance. This study aims to assess m6A levels in transcripts involved in the NKG2D cytotoxicity signaling pathway in NK cells of BC patients compared to controls and find out its impact on mRNA levels. Additionally, it evaluates how deliberately altering m6A levels in NK cells affects mRNA and protein expression of NKG2D pathway genes and NK cell functionality. METHODS m6A methylation in transcripts of NKG2D-pathway-related genes in BC patients and controls was determined using methylated RNA immunoprecipitation-reverse transcription-PCR (MERIP-RT-PCR). To deliberately alter m6A levels in primary cultured human NK cells, the m6A demethylases, FTO and ALKBH5, were knocked out using the CRISPR-CAS9 system, and FTO was inhibited using Meclofenamic acid (MA). The impact of m6A alteration on corresponding mRNA and protein levels was assessed using RT-qPCR and Western blot analysis or flow cytometry, respectively. Additionally, NK cell functionality was evaluated through degranulation and 51Cr release cytotoxicity assays. RESULTS Transcripts of NKG2D, an activating receptor that detects stressed non-self tumour cells, had significantly higher m6A levels in the 3' untranslated region (3'UTR) accompanied by a marked reduction in their corresponding mRNA levels in BC patients compared to controls. Conversely, transcripts of ERK2 and PRF1 exhibited significantly lower m6A levels escorted with higher mRNA expression in BC patients relative to controls. The mRNA levels of PI3K, PAK1 and GZMH were also significantly elevated in BC patients. Furthermore, artificially increasing transcripts' m6A levels via MA in cultured primary NK cells reduced mRNA levels of NKG2D pathway genes and death receptor ligands but did not affect protein expression or NK cell functionality. CONCLUSION Transcripts with higher m6A levels in the 3'UTR region were less abundant, and vice versa. However, changes in mRNA levels of the target genes didn't impact their corresponding protein levels or NK cell functionality.
Collapse
Affiliation(s)
- Raghda A Elsabbagh
- Biochemistry Department, Faculty of Pharmacy and Biotechnology, the German University in Cairo, Cairo, Egypt
| | - Ghada Abdelhady
- Microbiology, Immunology and Biotechnology Department, Faculty of Pharmacy and Biotechnology, the German University in Cairo, Cairo, Egypt
| | - Doris Urlaub
- Leibniz Research Centre for Working Environment and Human Factors (IfADo), TU Dortmund, Dortmund, Germany
| | - Mina Sandusky
- Leibniz Research Centre for Working Environment and Human Factors (IfADo), TU Dortmund, Dortmund, Germany
| | - Ola Khorshid
- Medical Oncology Department, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Mohamed Z Gad
- Biochemistry Department, Faculty of Pharmacy and Biotechnology, the German University in Cairo, Cairo, Egypt
| | - Khaled Abou-Aisha
- Microbiology, Immunology and Biotechnology Department, Faculty of Pharmacy and Biotechnology, the German University in Cairo, Cairo, Egypt
| | - Carsten Watzl
- Leibniz Research Centre for Working Environment and Human Factors (IfADo), TU Dortmund, Dortmund, Germany.
| | - Mona Rady
- Microbiology, Immunology and Biotechnology Department, Faculty of Pharmacy and Biotechnology, the German University in Cairo, Cairo, Egypt.
- Faculty of Biotechnology, German International University, New Administrative Capital, Egypt.
| |
Collapse
|
49
|
Zhai Y, Zhang H, Hu C, Wang Q, Wang S, Ge RS, Li X. Bisphenol Z inhibits the function of Leydig cells via upregulation of METTL3 expression in adult male rats. J Steroid Biochem Mol Biol 2025; 252:106786. [PMID: 40398521 DOI: 10.1016/j.jsbmb.2025.106786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 05/06/2025] [Accepted: 05/19/2025] [Indexed: 05/23/2025]
Abstract
The use of bisphenol A has been restricted due to its toxicity. However, the impact of its substitute, bisphenol Z (BPZ), on Leydig cell function remains uncertain. We aimed to examine the associations between BPZ exposure and the disruption of Leydig cell function via upregulating Mettl3 and inducing oxidative stress. To address this, in vivo, male adult Sprague-Dawley rats received BPZ (0, 1, 10, or 100mg/kg/d orally) for 7 days, and in vitro, purified Leydig cells were treated with BPZ (0-20μM, 24h). Leydig cell morphology and function were assessed. The results showed that BPZ did not alter Leydig cell quantity but notably decreased serum testosterone levels. Furthermore, it significantly downregulated the expression levels of genes and proteins (SCARB1, STAR, CYP17A1, HSD17B3, and INSL3) in Leydig cells. Concurrently, BPZ treatment led to diminished expression of antioxidant genes (Gpx1 and Cat), an upregulation in m6A related gene (Mettl3) subsequent to the enrichment of RNA methylation fragments in the testis. In vitro analysis of primary Leydig cells demonstrated that BPZ heightened oxidative stress and diminished testosterone production. In conclusion, BPZ reduces rat testosterone by downregulating steroidogenic genes (Star, Scarb1, Cyp17a1, and Hsd17b3) via METTL3-m6A-Camkk2 pathway, impairing Leydig cell function.
Collapse
Affiliation(s)
- Yingna Zhai
- Department of Anesthesiology and Perioperative Medicine, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China; Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Wenzhou, Zhejiang, 325027, China; Key Laboratory of Precision Anesthesiology of Zhejiang Province, Wenzhou Medical University,Wenzhou, Zhejiang, 325027, China; Key Laboratory of Environment and Male Reproductive Medicine of Wenzhou, Wenzhou, Zhejiang, 325027, China
| | - Huiqian Zhang
- Department of Anesthesiology and Perioperative Medicine, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China; Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Wenzhou, Zhejiang, 325027, China; Key Laboratory of Precision Anesthesiology of Zhejiang Province, Wenzhou Medical University,Wenzhou, Zhejiang, 325027, China; Key Laboratory of Environment and Male Reproductive Medicine of Wenzhou, Wenzhou, Zhejiang, 325027, China
| | - Chunnan Hu
- Department of Anesthesiology and Perioperative Medicine, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China; Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Wenzhou, Zhejiang, 325027, China; Key Laboratory of Precision Anesthesiology of Zhejiang Province, Wenzhou Medical University,Wenzhou, Zhejiang, 325027, China; Key Laboratory of Environment and Male Reproductive Medicine of Wenzhou, Wenzhou, Zhejiang, 325027, China
| | - Qingyuan Wang
- Department of Anesthesiology and Perioperative Medicine, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China; Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Wenzhou, Zhejiang, 325027, China; Key Laboratory of Precision Anesthesiology of Zhejiang Province, Wenzhou Medical University,Wenzhou, Zhejiang, 325027, China; Key Laboratory of Environment and Male Reproductive Medicine of Wenzhou, Wenzhou, Zhejiang, 325027, China
| | - Shaowei Wang
- Department of Anesthesiology and Perioperative Medicine, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China; Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Wenzhou, Zhejiang, 325027, China; Key Laboratory of Precision Anesthesiology of Zhejiang Province, Wenzhou Medical University,Wenzhou, Zhejiang, 325027, China; Key Laboratory of Environment and Male Reproductive Medicine of Wenzhou, Wenzhou, Zhejiang, 325027, China
| | - Ren-Shan Ge
- Department of Anesthesiology and Perioperative Medicine, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China; Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Wenzhou, Zhejiang, 325027, China; Key Laboratory of Precision Anesthesiology of Zhejiang Province, Wenzhou Medical University,Wenzhou, Zhejiang, 325027, China; Key Laboratory of Environment and Male Reproductive Medicine of Wenzhou, Wenzhou, Zhejiang, 325027, China.
| | - Xiaoheng Li
- Department of Anesthesiology and Perioperative Medicine, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China; Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Wenzhou, Zhejiang, 325027, China; Key Laboratory of Precision Anesthesiology of Zhejiang Province, Wenzhou Medical University,Wenzhou, Zhejiang, 325027, China; Key Laboratory of Environment and Male Reproductive Medicine of Wenzhou, Wenzhou, Zhejiang, 325027, China.
| |
Collapse
|
50
|
Feng A, Liang Y, Fu P, Dong Y, Black SM, Wang T. Endotoxin-induced m6A RNA methylation landscape in lung endothelial cells: role of METTL3 in regulating inflammation and injury during acute lung injury. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167907. [PMID: 40379220 DOI: 10.1016/j.bbadis.2025.167907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 03/31/2025] [Accepted: 05/13/2025] [Indexed: 05/19/2025]
Abstract
Acute Lung Injury (ALI) involves diffuse alveolar damage, neutrophil infiltration, and pulmonary edema, with unacceptable mortality. Bacterial lipopolysaccharide (LPS) activates inflammatory pathways in ALI, which are then regulated by transcriptional and post-transcriptional pathways to affect gene expression. RNA methylation, N6-methyladenosine, is the main m6A mRNA modification that controls the expression of various genes in different environments. There are very few facts about LPS's effect on m6A RNA methylation. This study will explore the m6A RNA methylation landscape in lung endothelial cells (ECs) to understand its role in lung inflammation. In this study, lung endothelial cells were treated with LPS, and the dynamics of mRNA m6A methylation were examined through m6A-methylated RNA sequencing. RNA abundance was measured with RNA-seq, and global protein expression and m6A-binding proteins were identified using mass spectrometry (MS). Following LPS treatment, global m6A methylation levels increased along with the upregulation and nuclear translocation of METTL3 protein, while demethylase activity remained unchanged. METTL3 drove LPS-induced m6A methylation and endothelial injury, as shown by selective METTL3 siRNA and the inhibitor STM2457. MeRIP-seq analyses revealed increased m6A sites near the 5' UTR in LPS-treated cells, with m6A methylation correlating positively with gene expression. The metabolic and apoptosis pathways were shown to be more enriched in different types of methylated exons. METTL3-mediated m6A methylation targeted inflammatory genes, enhancing protein expression in chemokine signaling and MAPK pathways. STM2457 effectively mitigated LPS- or CLP-induced experimental ALI. According to this paper, LPS-mediated m6A RNA methylation is described in terms of genomic structure. Modulation of m6A methylation exerts influence over LPS-mediated endothelial gene expression and the ensuing inflammatory response.
Collapse
Affiliation(s)
- Anlin Feng
- Center for Translational Science, Florida International University, Port Saint Lucie, FL 34987, USA; Department of Environmental Health Sciences, Florida International University, Miami, FL 33199, USA
| | - Ying Liang
- Center for Translational Science, Florida International University, Port Saint Lucie, FL 34987, USA; Department of Environmental Health Sciences, Florida International University, Miami, FL 33199, USA
| | - Panfeng Fu
- Center for Translational Science, Florida International University, Port Saint Lucie, FL 34987, USA; Department of Environmental Health Sciences, Florida International University, Miami, FL 33199, USA
| | - Yishu Dong
- Center for Translational Science, Florida International University, Port Saint Lucie, FL 34987, USA; Department of Cellular and Molecular Medicine, Florida International University, Miami, FL 33199, USA
| | - Stephen M Black
- Center for Translational Science, Florida International University, Port Saint Lucie, FL 34987, USA; Department of Environmental Health Sciences, Florida International University, Miami, FL 33199, USA; Department of Cellular and Molecular Medicine, Florida International University, Miami, FL 33199, USA
| | - Ting Wang
- Center for Translational Science, Florida International University, Port Saint Lucie, FL 34987, USA; Department of Environmental Health Sciences, Florida International University, Miami, FL 33199, USA; Department of Cellular and Molecular Medicine, Florida International University, Miami, FL 33199, USA.
| |
Collapse
|