1
|
Xu X, Huang X, Zhang C, Mi X, Zhang C, Hua F, Zhang L. METTL3 promotes podocyte pyroptosis in diabetic nephropathy through N 6-methyladenosine modification of TRIM29 mRNA. Ren Fail 2025; 47:2497492. [PMID: 40325507 PMCID: PMC12054578 DOI: 10.1080/0886022x.2025.2497492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 04/07/2025] [Accepted: 04/17/2025] [Indexed: 05/07/2025] Open
Abstract
Multiple studies have revealed the critical roles of epigenetic modifications in the development of diabetic nephropathy (DN). Methyltransferase-like 3 (METTL3)-mediated N6-methyladenosine (m6A) RNA modification in podocytes represents a new disease mechanism in DN. The tripartite motif-containing (TRIM) family member TRIM29 was reported to promote podocyte pyroptosis by activating the nuclear factor-κB/NLR family pyrin domain containing 3 (NLRP3) inflammasome pathway. However, whether METTL3-mediated m6A modification of TRIM29 mRNA is involved in podocyte injury remain unknown. Here, we found that METTL3 upregulated the m6A content in mRNA from kidney tissues of mice with streptozotocin-induced DN and in hyperglycemia-induced MPC-5 murine podocytes. METTL3 expression in high glucose-treated MPC-5 cells resulted in elevated release of interleukin (IL)-1β, IL-18, and lactate dehydrogenase and upregulated expression of pyroptosis-associated molecules. Mechanistically, METTL3 was found to directly target TRIM29 for m6A modification and activate TRIM29 transcription. Moreover, the m6A reader YT521-B homology (YTH) domain family member YTHDF1 was recruited by METTL3 to maintain the stability of TRIM29 mRNA, which contributed significantly to increased podocyte pyroptosis. Furthermore, the potent METTL3-specific inhibitor STM2457 prominently alleviated podocyte injury through attenuating activation of the NLRP3 inflammasome/pyroptosis pathway in the DN mouse model. Our results suggest that METTL3 plays a critical role in hyperglycemia-induced podocyte injury through m6A modification of TRIM29 mRNA, which provides new insight for the development of METTL3- and pyroptosis-targeted strategies to treat DN and other diabetic kidney diseases.
Collapse
Affiliation(s)
- Xiaohong Xu
- Department of Nephrology, The Affiliated Suqian First People’s Hospital of Nanjing Medical University, Suqian, China
- Department of Nephrology, Jiangsu Province (Suqian) Hospital, Suqian, China
| | - Xiaolin Huang
- Department of Endocrinology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Ce Zhang
- Department of Nephrology, Nanjing Drum Tower Hospital Group Suqian Hospital, Suqian, China
| | - Xia Mi
- Department of Nephrology, Nanjing Drum Tower Hospital Group Suqian Hospital, Suqian, China
| | - Chi Zhang
- Department of Nephrology, Nanjing Drum Tower Hospital Group Suqian Hospital, Suqian, China
| | - Fei Hua
- Department of Endocrinology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Liexiang Zhang
- Department of Neurosurgery, The Affiliated Suqian First People’s Hospital of Nanjing Medical University, Suqian, China
| |
Collapse
|
2
|
Duan W, Peng Z, Yang K, Hu M, Yu X, Zhang B, Liu L. Transcriptome-wide N6-methyladenosine methylation dynamic profile in type 1 diabetes progression. Epigenomics 2025:1-12. [PMID: 40421929 DOI: 10.1080/17501911.2025.2510196] [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: 01/19/2025] [Accepted: 05/19/2025] [Indexed: 05/28/2025] Open
Abstract
AIMS To map transcriptome-wide N6-methyladenosine (m6A) profile at different stages of type 1 diabetes (T1D). MATERIALS & METHODS RNA extracted from islet tissues of non-obese diabetic mice at different ages (4-week-old control group, 8-week-old pre-diabetes group, and 14-week-old diabetes group) was analyzed by MeRIP-seq and mRNA-seq. Bioinformatics analyses, including m6A motif enrichment, differential methylation, gene ontology and pathway analysis. RESULTS Bioinformatic analysis revealed a progressive increase in m6A methylation sites and unique peaks throughout diabetes progression. The predominant m6A motif was "GGACU" in the control and pre-diabetes stages, shifting to "GGACU/A" in the diabetes stage. m6A modifications were primarily enriched at the start codon, coding region, and stop codon. Integrated analysis found pre-diabetes, diabetes groups showed distinct m6A and mRNA changes versus control. Pathway analysis indicated that differentially expressed mRNAs with m6A methylation were predominantly enriched in insulin/IGF-MAPKK/MAPK cascades, apoptosis, T-cell activation, interleukins, CCKR, and inflammation-related pathways in the pre-diabetes stage. As diabetes progressed, additional pathways, including Wnt, EGF receptor, PDGF, GnRH receptor, and angiogenesis, became involved in disease development. In vitro, cytokine stimulation of INS-1 cells increased m6A methylation, upregulated METTL3, downregulated ALKBH5. CONCLUSIONS m6A methyl group dynamics in T1D disease progression, potentially influencing mRNA expression and signal transduction pathways.
Collapse
Affiliation(s)
- Wu Duan
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Clinical Medical Research Center for Endocrinology and Metabolic Diseases, Hubei, China
- Branch of National Clinical Research Center for Metabolic Diseases, Hubei, China
- Department of Endocrinology, Qilu Hospital of Shandong University, Jinan, China
| | - Ziyi Peng
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Clinical Medical Research Center for Endocrinology and Metabolic Diseases, Hubei, China
- Branch of National Clinical Research Center for Metabolic Diseases, Hubei, China
| | - Kun Yang
- Department of Dermatology and Venereology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mengyuan Hu
- The Second School of Clinical Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xuefeng Yu
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Clinical Medical Research Center for Endocrinology and Metabolic Diseases, Hubei, China
- Branch of National Clinical Research Center for Metabolic Diseases, Hubei, China
| | - Benping Zhang
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Clinical Medical Research Center for Endocrinology and Metabolic Diseases, Hubei, China
- Branch of National Clinical Research Center for Metabolic Diseases, Hubei, China
| | - Li Liu
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Clinical Medical Research Center for Endocrinology and Metabolic Diseases, Hubei, China
- Branch of National Clinical Research Center for Metabolic Diseases, Hubei, China
| |
Collapse
|
3
|
Luo Y, Li JE, Xu S, Zeng H, Zhang Y, Yang S, He X, Liu J. METTL3 promotes human amniotic epithelial stem cells differentiation into insulin-producing cells by regulation of MaFA expression. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167904. [PMID: 40374016 DOI: 10.1016/j.bbadis.2025.167904] [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: 10/31/2024] [Revised: 04/23/2025] [Accepted: 05/12/2025] [Indexed: 05/17/2025]
Abstract
OBJECTIVE Generating mature β-cells from stem cells remains a significant challenge in diabetes cell therapy. Human amniotic epithelial stem cells (hAESCs) have made their mark in regenerative medicine, and provide several advantages compared to other stem cells. Methyltransferase-like 3 (METTL3), an essential RNA methyltransferase participating in N6-methyladenosine (m6A) mRNA methylation, plays a critical role in the normal development of β-cells, yet its deletion in β-cells leads to β-cell dysfunction and hyperglycemia. METHODS In this study, we isolated and characterized hAESCs from human amniotic membranes, differentiated these hAESCs into insulin-producing cells (IPCs), and explored the role of METTL3 in such differentiation. We examined the expression of METTL3 and insulin like growth factor 2 mRNA binding protein 2 (IGF2BP2, a decodes m6A methylation "reader") in the generated IPCs. Subsequently, we suppressed METTL3 using an inhibitor (STM2457) and overexpressed METTL3 via plasmid transfection (METTL3-OE). The differentiated STM2457 and METTL3-OE IPCs were compared to normal induction (WT) IPCs regarding the expression of β-cell markers by RT-qPCR and western blotting, immunofluorescence, C-peptide release, and glucose-stimulated insulin secretion (GSIS). Methylated RNA immunoprecipitation (MeRIP)-qPCR was used to examine the molecular mechanism underlying METTL3/m6A signaling axis in MaFA (endocrine pancreatic β-cells marker) expression. We examined the potential therapeutic uses and efficacy of IPCs through streptozotocin (STZ)-induced C57BL/6 DM. RESULTS Isolated hAESCs displayed all characteristics of ESCs and could generate IPCs. METTL3 and IGF2BP2 were elevated during differentiation. Overexpressing METTL3 improved the expression of β-cell markers in the final differentiated IPCs, improved C-peptide release, and demonstrated increased insulin secretion upon challenging with high glucose conditions, whereas inhibiting METTL3 attenuated these effects. Moreover, METTL3 modulated the MaFA expression in an m6A-dependent manner. CONCLUSIONS These findings suggest METTL3 as a promoting factor of IPCs generation, with its up-regulation potentially generating more mature IPCs for hAESCs therapy of diabetes mellitus.
Collapse
Affiliation(s)
- Yunfei Luo
- Department of Endocrinology and Metabolism of the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang City, Jiangxi Province, China; School of basic medicine, Nanchang Medical College, Nanchang City, Jiangxi Province, China
| | - Jin-E Li
- Department of Endocrinology and Metabolism of the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang City, Jiangxi Province, China
| | - Shan Xu
- Department of Endocrinology and Metabolism of the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang City, Jiangxi Province, China
| | - Haixia Zeng
- Department of Endocrinology and Metabolism of the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang City, Jiangxi Province, China
| | - Yuying Zhang
- Department of Endocrinology and Metabolism of the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang City, Jiangxi Province, China
| | - Shiqi Yang
- Department of Endocrinology and Metabolism of the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang City, Jiangxi Province, China
| | - Xiaoju He
- Department of Obstetrics and Gynecology of the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang City, Jiangxi Province, China
| | - Jianping Liu
- Department of Endocrinology and Metabolism of the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang City, Jiangxi Province, China; Institute for the Study of Endocrinology and Metabolism in Jiangxi Province, Nanchang City, Jiangxi Province, China; Jiangxi Key Laboratory of Molecular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang City, Jiangxi Province, China.
| |
Collapse
|
4
|
Zhang X, Shan A, Chen J, Cao Y, Jiang X. Mettl3 deficiency leads to impaired insulin secretion via regulating Ire1a of mature β-cells in mice. Sci Rep 2025; 15:10835. [PMID: 40155600 PMCID: PMC11953400 DOI: 10.1038/s41598-025-93799-4] [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: 09/12/2024] [Accepted: 03/10/2025] [Indexed: 04/01/2025] Open
Abstract
The modification of N6-methyladenosine (m6A) influences the translation and stability of transcripts, allowing for the coordination of gene regulation during cell state maintenance and transition. Deregulation of components in the m6A regulatory network is associated with glucose homeostasis and development of diabetes. In this study, we investigated the functional role of Mettl3, which is the key component of the m6A methyltransferase complex, in regulating β-cell identity and function in two pancreatic β-cell-specific Mettl3 knockout mouse models. The glucose metabolic phenotype, β-cell proliferation, islet architecture and insulin secretion were analyzed in vivo. We next analyzed the expression levels of genes associated with endoplasmic reticulum (ER) stress in the Mettl3 ablated islets. MeRIP-qPCR was applied to detect the m6A modification enrichment of Ire1α mRNA. Adenovirus-mediated Mettl3 infection was performed on islets to explore the effect of Mettl3 overexpression on ER stress and insulin secretion. Our results showed that Mettl3 deficiency led to loss of β-cell identity and impaired insulin secretion in mice. Depletion of Mettl3 verified the m6A modification in Ire1α and consequently induced ER stress in islet cells. Mettl3 overexpression in islets could alleviate ER stress and improve the insulin secretion capacity. Our findings demonstrated that Mettl3 was an important regulator of ER stress and insulin secretion in mouse pancreatic β-cells.
Collapse
Affiliation(s)
- Xu Zhang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, National Clinical Research Centre for Metabolic Diseases, State Key Laboratory of Medical Genomics, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission, Research Unit of Clinical and Basic Research on Metabolic Diseases of Chinese Academy of Medical Sciences, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Aijing Shan
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, National Clinical Research Centre for Metabolic Diseases, State Key Laboratory of Medical Genomics, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission, Research Unit of Clinical and Basic Research on Metabolic Diseases of Chinese Academy of Medical Sciences, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jie Chen
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, National Clinical Research Centre for Metabolic Diseases, State Key Laboratory of Medical Genomics, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission, Research Unit of Clinical and Basic Research on Metabolic Diseases of Chinese Academy of Medical Sciences, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yanan Cao
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, National Clinical Research Centre for Metabolic Diseases, State Key Laboratory of Medical Genomics, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission, Research Unit of Clinical and Basic Research on Metabolic Diseases of Chinese Academy of Medical Sciences, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Ruijin Yangtze River Delta Health Institute, Wuxi Branch of Ruijin Hospital, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xiuli Jiang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, National Clinical Research Centre for Metabolic Diseases, State Key Laboratory of Medical Genomics, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission, Research Unit of Clinical and Basic Research on Metabolic Diseases of Chinese Academy of Medical Sciences, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
5
|
Luo Y, Li JE, Zeng H, Zhang Y, Yang S, Liu J. Semaglutide alleviates the pancreatic β cell function via the METTL14 signaling and modulating gut microbiota in type 2 diabetes mellitus mice. Life Sci 2025; 361:123328. [PMID: 39719165 DOI: 10.1016/j.lfs.2024.123328] [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: 10/25/2024] [Revised: 12/09/2024] [Accepted: 12/19/2024] [Indexed: 12/26/2024]
Abstract
AIMS Semaglutide, a novel long-acting GLP-1RA, stimulates insulin and suppresses islet-secreted glucagon to reduce glucose levels. It has been unveiled that m6A mRNA modification plays a pivotal role in regulating β cell function. However, it remains unclear whether semaglutide can elicit protective effects through manipulating m6A modification and the underlying mechanism. We aimed to elucidate the role played by semaglutide in m6A modification, and to explore its specific regulatory targets. Furthermore, we also delve into its effects on gut microbiota. MAIN METHODS Five-week-old male C57BL/6 mice were assigned to two dietary groups and fed a control or high-fat diet for 4 weeks. Then T2DM was induced in high-fat diet-fed mice via streptozotocin (STZ), the main groups were resampled to include treatment with semaglutide (SEM, 40 μg/kg) for another 4 weeks, totaling three groups: Control, Model (T2DM), T2DM + SEM. Additionally, we elucidated specific regulatory targets and signaling pathways in palmitic acid (PA)-stimulated beta-TC-6 cells. Immunofluorescence, Western blot, and RT-qPCR were used in the study. KEY FINDINGS Semaglutide mitigated pancreatic damage, enhanced islet cell proliferation, and restored islet size and alpha- and beta-cell masses. It also improved the expression of METTL14, pancreatic duodenal homeobox 1 (PDX-1), and protecting mitochondria, and modulated the PDX1 expression in an m6A-dependent manner. Concurrently, semaglutide significantly decreases the abundance of Firmicutes, Actinobacteriota, and Lactobacillus, while increasing the Bacteroides and norank_f_Muribaculaceae content, and the production of short-chain fatty acids (SCFA). SIGNIFICANCE Semaglutide positively influences by regulating m6A modifications to alleviate pancreatic beta cell dysfunction and modulate the gut microbiome.
Collapse
Affiliation(s)
- Yunfei Luo
- Department of Endocrinology and Metabolism of the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang City, Jiangxi Province, China
| | - Jin-E Li
- Department of Endocrinology and Metabolism of the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang City, Jiangxi Province, China
| | - Haixia Zeng
- Department of Endocrinology and Metabolism of the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang City, Jiangxi Province, China
| | - Yuying Zhang
- Department of Endocrinology and Metabolism of the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang City, Jiangxi Province, China
| | - Shiqi Yang
- Department of Endocrinology and Metabolism of the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang City, Jiangxi Province, China
| | - Jianping Liu
- Department of Endocrinology and Metabolism of the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang City, Jiangxi Province, China; Institute for the Study of Endocrinology and Metabolism in Jiangxi Province, Nanchang City, Jiangxi Province, China; Branch of National Clinical Research Center for Metabolic Diseases, Nanchang City, Jiangxi Province, China.
| |
Collapse
|
6
|
Qiu T, Zhang J, Song J, Wu C, Yao X, Wang N, Yang G, Bai J, Lv L, Sun X. Arsenic inducible islet β-cell dysfunction and ferroptosis through m 6A-YTHDF2-dependent CHAC1 enhancement. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 289:117479. [PMID: 39667319 DOI: 10.1016/j.ecoenv.2024.117479] [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: 07/14/2024] [Revised: 09/21/2024] [Accepted: 12/03/2024] [Indexed: 12/14/2024]
Abstract
Arsenic, recognized as an environmental and food contaminant, has been linked to the dysfunction of islet β-cells, the primary lesions in type 2 diabetes (T2D). Ferroptosis, a regulated cell death pathway dependent on GPX4, has been implicated in arsenic-induced β-cell dysfunction. However, the underlying molecular mechanisms remain unclear. GPX4 activity is significantly modulated by glutathione levels. In this study, we demonstrate that arsenic inhibits GPX4 expression by upregulating the expression of glutathione-specific γ-glutamylcyclotransferase 1 (CHAC1) (>2-fold in vivo and 1.5-fold in vitro). Conversely, arsenic does not affect the expression of the glutathione-cysteine ligase catalytic subunit (GCLC), which is crucial for glutathione synthesis. Notably, CHAC1 knockdown significantly ameliorated arsenic-induced β-cell dysfunction and ferroptosis. N6-methyladenosine (m6A) plays a crucial role in the post-transcriptional modification of mRNA. Arsenic treatment downregulated the expression of methyltransferases METTL3/14 (approximately 0.5-fold), and overexpression of METTL3 alleviated arsenic-induced β-cell dysfunction and ferroptosis. The m6A modification site on CHAC1 was identified, and RIP assays confirmed that arsenic treatment inhibited the interaction between METTL3/YTHDF2 and CHAC1. Furthermore, METTL3 overexpression reduced the half-life of CHAC1 mRNA (almost 0.5-fold). This study uncovers a novel mechanism by which arsenic modulates CHAC1 and ferroptosis through m6A in β-cell dysfunction, highlighting potential therapeutic targets for arsenic-related T2D.
Collapse
Affiliation(s)
- Tianming Qiu
- Department of Occupational and Environmental Health, School of Public Health, Dalian Medical University, No. 9 West Section Lvshun South Road, Dalian 116044, PR China
| | - Jingyuan Zhang
- Department of Occupational and Environmental Health, School of Public Health, Dalian Medical University, No. 9 West Section Lvshun South Road, Dalian 116044, PR China
| | - Jinwei Song
- Department of Occupational and Environmental Health, School of Public Health, Dalian Medical University, No. 9 West Section Lvshun South Road, Dalian 116044, PR China
| | - Chenbing Wu
- Preventive Medicine Laboratory, School of Public Health, Dalian Medical University, No. 9 West Section Lvshun South Road, 116044, PR China
| | - Xiaofeng Yao
- Department of Occupational and Environmental Health, School of Public Health, Dalian Medical University, No. 9 West Section Lvshun South Road, Dalian 116044, PR China
| | - Ningning Wang
- Department of Nutrition and Food Safety, School of Public Health, Dalian Medical University, No. 9 West Section Lvshun South Road, Dalian 116044, PR China
| | - Guang Yang
- Department of Nutrition and Food Safety, School of Public Health, Dalian Medical University, No. 9 West Section Lvshun South Road, Dalian 116044, PR China
| | - Jie Bai
- Preventive Medicine Laboratory, School of Public Health, Dalian Medical University, No. 9 West Section Lvshun South Road, 116044, PR China
| | - Li Lv
- Department of Pathology, the Second Affiliated Hospital of Dalian Medical University, No. 467 Zhongshan Road, Dalian 116023, PR China.
| | - Xiance Sun
- Department of Occupational and Environmental Health, School of Public Health, Dalian Medical University, No. 9 West Section Lvshun South Road, Dalian 116044, PR China; Global Health Research Center, Dalian Medical University, No. 9 West Section Lvshun South Road, Dalian 116044, PR China.
| |
Collapse
|
7
|
Kahraman S, De Jesus DF, Wei J, Brown NK, Zou Z, Hu J, Pirouz M, Gregory RI, He C, Kulkarni RN. m 6A mRNA methylation by METTL14 regulates early pancreatic cell differentiation. EMBO J 2024; 43:5445-5468. [PMID: 39322760 PMCID: PMC11574190 DOI: 10.1038/s44318-024-00213-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 08/05/2024] [Accepted: 08/13/2024] [Indexed: 09/27/2024] Open
Abstract
N6-methyladenosine (m6A) is the most abundant chemical modification in mRNA and plays important roles in human and mouse embryonic stem cell pluripotency, maintenance, and differentiation. We have recently reported that m6A is involved in the postnatal control of β-cell function in physiological states and in type 1 and 2 diabetes. However, the precise mechanisms by which m6A acts to regulate the development of human and mouse pancreas are unexplored. Here, we show that the m6A landscape is dynamic during human pancreas development, and that METTL14, one of the m6A writer complex proteins, is essential for the early differentiation of both human and mouse pancreatic cells.
Collapse
Affiliation(s)
- Sevim Kahraman
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA, USA
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, USA
| | - Dario F De Jesus
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA, USA
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, USA
| | - Jiangbo Wei
- Department of Chemistry, Department of Biochemistry and Molecular Biology, and Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL, 60637, USA
- Howard Hughes Medical Institute, The University of Chicago, Chicago, IL, 60637, USA
- Department of Chemistry and Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - Natalie K Brown
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA, USA
| | - Zhongyu Zou
- Department of Chemistry, Department of Biochemistry and Molecular Biology, and Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL, 60637, USA
- Howard Hughes Medical Institute, The University of Chicago, Chicago, IL, 60637, USA
| | - Jiang Hu
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA, USA
| | - Mehdi Pirouz
- Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, USA
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA
| | - Richard I Gregory
- Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, USA
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA
| | - Chuan He
- Department of Chemistry, Department of Biochemistry and Molecular Biology, and Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL, 60637, USA
- Howard Hughes Medical Institute, The University of Chicago, Chicago, IL, 60637, USA
| | - Rohit N Kulkarni
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA, USA.
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
- Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
8
|
Li Y, Yang Y, Sun Y, He L, Zhao L, Sun H, Chang X, Liang R, Wang S, Han X, Zhu Y. The miR-203/ZBTB20/MAFA Axis Orchestrates Pancreatic β-Cell Maturation and Identity During Weaning and Diabetes. Diabetes 2024; 73:1673-1686. [PMID: 39058664 DOI: 10.2337/db23-0604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 07/23/2024] [Indexed: 07/28/2024]
Abstract
Maturation of postnatal β-cells is regulated in a cell-autonomous manner, and metabolically stressed β-cells regress to an immature state, ensuring defective β-cell function and the onset of type 2 diabetes. The molecular mechanisms connecting the nutritional transition to β-cell maturation remain largely unknown. Here, we report a mature form of miRNA (miR-203)/ZBTB20/MAFA regulatory axis that mediates the β-cell maturation process. We show that the level of the mature form of miRNA (miR-203) in β-cells changes during the nutritional transition and that miR-203 inhibits β-cell maturation at the neonatal stage and under high-fat diet conditions. Using single-cell RNA sequencing, we demonstrated that miR-203 elevation promoted the transition of immature β-cells into CgBHi endocrine cells while suppressing gene expressions associated with β-cell maturation in a ZBTB20/MAFA-dependent manner. ZBTB20 is an authentic target of miR-203 and transcriptionally upregulates MAFA expression. Manipulating the miR-203/ZBTB20/MAFA axis may therefore offer a novel strategy for boosting functional β-cell numbers to alleviate diabetes. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Yating Li
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yuqian Yang
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yi Sun
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Lu He
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Lin Zhao
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Haoran Sun
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiaoai Chang
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Rui Liang
- Organ Transplant Center, Tianjin First Central Hospital, Nankai University, Tianjin, China
| | - Shusen Wang
- Organ Transplant Center, Tianjin First Central Hospital, Nankai University, Tianjin, China
| | - Xiao Han
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yunxia Zhu
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
9
|
Wang Y, Zou J, Zhou H. N6-methyladenine RNA methylation epigenetic modification and diabetic microvascular complications. Front Endocrinol (Lausanne) 2024; 15:1462146. [PMID: 39296713 PMCID: PMC11408340 DOI: 10.3389/fendo.2024.1462146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 08/20/2024] [Indexed: 09/21/2024] Open
Abstract
N6-methyladensine (m6A) has been identified as the best-characterized and the most abundant mRNA modification in eukaryotes. It can be dynamically regulated, removed, and recognized by its specific cellular components (respectively called "writers," "erasers," "readers") and have become a hot research field in a variety of biological processes and diseases. Currently, the underlying molecular mechanisms of m6A epigenetic modification in diabetes mellitus (DM) and diabetic microvascular complications have not been extensively clarified. In this review, we focus on the effects and possible mechanisms of m6A as possible potential biomarkers and therapeutic targets in the treatment of DM and diabetic microvascular complications.
Collapse
Affiliation(s)
- Yuanyuan Wang
- Department of Nephrology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jiayun Zou
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Hua Zhou
- Department of Nephrology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
10
|
Liu X, Wang N, Gu S, He Z. Changes of RNA m 6A/m 5C Modification Regulatory Molecules in Ferroptosis of T2DM Rat Pancreas. Cell Biochem Biophys 2024; 82:1279-1289. [PMID: 38709441 DOI: 10.1007/s12013-024-01282-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/17/2024] [Indexed: 05/07/2024]
Abstract
N6-methyladenine (m6A) and 5-methylcytosine (m5C) are two common forms of RNA methylation that play an important role in the epigenetics of type 2 diabetes mellitus (T2DM). One type of cell death, ferroptosis, has been implicated in islet β-cell damage in T2DM. Notably, RNA methylation, an upstream regulatory mechanism of mRNAs, can regulate the expression of ferroptosis signaling molecules, thereby affecting cell proliferation and death. Here, we found that the ferroptosis signaling pathway was activated in pancreas of T2DM rats, followed by significant changes in m6A/m5C modification regulatory molecules. These detection data together with the prediction results that m6A and m5C exist in the mRNAs of ferroptosis molecules, we speculate that m6A and m5C are probably involved in pancreatic cell damage by modifying of ferroptosis signaling molecules. In short, our findings provide a new research idea for future studies on the molecular mechanisms of pancreatic cell damage and point to a new direction for exploring the mechanisms of ferroptosis from the perspective of RNA methylation modification.
Collapse
Affiliation(s)
- Xiaoyu Liu
- Institute of Preventive Medicine, School of Public Health, Dali University, Dali, Yunnan, China
| | - Nan Wang
- Institute of Preventive Medicine, School of Public Health, Dali University, Dali, Yunnan, China
| | - Shiyan Gu
- Institute of Preventive Medicine, School of Public Health, Dali University, Dali, Yunnan, China.
| | - Zuoshun He
- Institute of Preventive Medicine, School of Public Health, Dali University, Dali, Yunnan, China.
| |
Collapse
|
11
|
Wang X, Gan M, Wang Y, Wang S, Lei Y, Wang K, Zhang X, Chen L, Zhao Y, Niu L, Zhang S, Zhu L, Shen L. Comprehensive review on lipid metabolism and RNA methylation: Biological mechanisms, perspectives and challenges. Int J Biol Macromol 2024; 270:132057. [PMID: 38710243 DOI: 10.1016/j.ijbiomac.2024.132057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 04/26/2024] [Accepted: 05/01/2024] [Indexed: 05/08/2024]
Abstract
Adipose tissue plays a crucial role in maintaining energy balance, regulating hormones, and promoting metabolic health. To address disorders related to obesity and develop effective therapies, it is essential to have a deep understanding of adipose tissue biology. In recent years, RNA methylation has emerged as a significant epigenetic modification involved in various cellular functions and metabolic pathways. Particularly in the realm of adipogenesis and lipid metabolism, extensive research is ongoing to uncover the mechanisms and functional importance of RNA methylation. Increasing evidence suggests that RNA methylation plays a regulatory role in adipocyte development, metabolism, and lipid utilization across different organs. This comprehensive review aims to provide an overview of common RNA methylation modifications, their occurrences, and regulatory mechanisms, focusing specifically on their intricate connections to fat metabolism. Additionally, we discuss the research methodologies used in studying RNA methylation and highlight relevant databases that can aid researchers in this rapidly advancing field.
Collapse
Affiliation(s)
- Xingyu Wang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Mailin Gan
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Yan Wang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Saihao Wang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Yuhang Lei
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Kai Wang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Xin Zhang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Lei Chen
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Ye Zhao
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Lili Niu
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Shunhua Zhang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Li Zhu
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China.
| | - Linyuan Shen
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China.
| |
Collapse
|
12
|
Pang J, Kuang TD, Yu XY, Novák P, Long Y, Liu M, Deng WQ, Zhu X, Yin K. N6-methyladenosine in myeloid cells: a novel regulatory factor for inflammation-related diseases. J Physiol Biochem 2024; 80:249-260. [PMID: 38158555 DOI: 10.1007/s13105-023-01002-x] [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: 04/20/2023] [Accepted: 12/04/2023] [Indexed: 01/03/2024]
Abstract
N6-methyladenosine (m6A) is one of the most abundant epitranscriptomic modifications on eukaryotic mRNA. Evidence has highlighted that m6A is altered in response to inflammation-related factors and it is closely associated with various inflammation-related diseases. Multiple subpopulations of myeloid cells, such as macrophages, dendritic cells, and granulocytes, are crucial for the regulating of immune process in inflammation-related diseases. Recent studies have revealed that m6A plays an important regulatory role in the functional of multiple myeloid cells. In this review, we comprehensively summarize the function of m6A modification in myeloid cells from the perspective of myeloid cell production, activation, polarization, and migration. Furthermore, we discuss how m6A-mediated myeloid cell function affects the progression of inflammation-related diseases, including autoimmune diseases, chronic metabolic diseases, and malignant tumors. Finally, we discuss the challenges encountered in the study of m6A in myeloid cells, intended to provide a new direction for the study of the pathogenesis of inflammation-related diseases.
Collapse
Affiliation(s)
- Jin Pang
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin, Guangxi, China
| | - Tong-Dong Kuang
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin, Guangxi, China
| | - Xin-Yuan Yu
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin, Guangxi, China
| | - Petr Novák
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin, Guangxi, China
| | - Yuan Long
- Department of General Practice, The Fifth Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Min Liu
- Department of General Practice, The Fifth Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Wei-Qian Deng
- Department of General Practice, The Fifth Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Xiao Zhu
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin, Guangxi, China.
| | - Kai Yin
- Department of General Practice, The Fifth Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
13
|
Yao Y, Liu P, Li Y, Wang W, Jia H, Bai Y, Yuan Z, Yang Z. Regulatory role of m 6A epitranscriptomic modifications in normal development and congenital malformations during embryogenesis. Biomed Pharmacother 2024; 173:116171. [PMID: 38394844 DOI: 10.1016/j.biopha.2024.116171] [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: 10/18/2023] [Revised: 01/08/2024] [Accepted: 01/13/2024] [Indexed: 02/25/2024] Open
Abstract
The discovery of N6-methyladenosine (m6A) methylation and its role in translation has led to the emergence of a new field of research. Despite accumulating evidence suggesting that m6A methylation is essential for the pathogenesis of cancers and aging diseases by influencing RNA stability, localization, transformation, and translation efficiency, its role in normal and abnormal embryonic development remains unclear. An increasing number of studies are addressing the development of the nervous and gonadal systems during embryonic development, but only few are assessing that of the immune, hematopoietic, urinary, and respiratory systems. Additionally, these studies are limited by the requirement for reliable embryonic animal models and the difficulty in collecting tissue samples of fetuses during development. Multiple studies on the function of m6A methylation have used suitable cell lines to mimic the complex biological processes of fetal development or the early postnatal phase; hence, the research is still in the primary stage. Herein, we discuss current advances in the extensive biological functions of m6A methylation in the development and maldevelopment of embryos/fetuses and conclude that m6A modification occurs extensively during fetal development. Aberrant expression of m6A regulators is probably correlated with single or multiple defects in organogenesis during the intrauterine life. This comprehensive review will enhance our understanding of the pivotal role of m6A modifications involved in fetal development and examine future research directions in embryogenesis.
Collapse
Affiliation(s)
- Yifan Yao
- Department of Pediatric Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China; Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Peiqi Liu
- Department of Pediatric Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yue Li
- Department of Pediatric Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Weilin Wang
- Department of Pediatric Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Huimin Jia
- Department of Pediatric Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yuzuo Bai
- Department of Pediatric Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China.
| | - Zhengwei Yuan
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China.
| | - Zhonghua Yang
- Department of Pediatric Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China; Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China.
| |
Collapse
|
14
|
Liu WW, Zheng SQ, Li T, Fei YF, Wang C, Zhang S, Wang F, Jiang GM, Wang H. RNA modifications in cellular metabolism: implications for metabolism-targeted therapy and immunotherapy. Signal Transduct Target Ther 2024; 9:70. [PMID: 38531882 DOI: 10.1038/s41392-024-01777-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 02/08/2024] [Accepted: 02/19/2024] [Indexed: 03/28/2024] Open
Abstract
Cellular metabolism is an intricate network satisfying bioenergetic and biosynthesis requirements of cells. Relevant studies have been constantly making inroads in our understanding of pathophysiology, and inspiring development of therapeutics. As a crucial component of epigenetics at post-transcription level, RNA modification significantly determines RNA fates, further affecting various biological processes and cellular phenotypes. To be noted, immunometabolism defines the metabolic alterations occur on immune cells in different stages and immunological contexts. In this review, we characterize the distribution features, modifying mechanisms and biological functions of 8 RNA modifications, including N6-methyladenosine (m6A), N6,2'-O-dimethyladenosine (m6Am), N1-methyladenosine (m1A), 5-methylcytosine (m5C), N4-acetylcytosine (ac4C), N7-methylguanosine (m7G), Pseudouridine (Ψ), adenosine-to-inosine (A-to-I) editing, which are relatively the most studied types. Then regulatory roles of these RNA modification on metabolism in diverse health and disease contexts are comprehensively described, categorized as glucose, lipid, amino acid, and mitochondrial metabolism. And we highlight the regulation of RNA modifications on immunometabolism, further influencing immune responses. Above all, we provide a thorough discussion about clinical implications of RNA modification in metabolism-targeted therapy and immunotherapy, progression of RNA modification-targeted agents, and its potential in RNA-targeted therapeutics. Eventually, we give legitimate perspectives for future researches in this field from methodological requirements, mechanistic insights, to therapeutic applications.
Collapse
Affiliation(s)
- Wei-Wei Liu
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- School of Clinical Medicine, Shandong University, Jinan, China
| | - Si-Qing Zheng
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Core Unit of National Clinical Research Center for Laboratory Medicine, Hefei, China
| | - Tian Li
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Core Unit of National Clinical Research Center for Laboratory Medicine, Hefei, China
| | - Yun-Fei Fei
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Core Unit of National Clinical Research Center for Laboratory Medicine, Hefei, China
| | - Chen Wang
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Core Unit of National Clinical Research Center for Laboratory Medicine, Hefei, China
| | - Shuang Zhang
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Core Unit of National Clinical Research Center for Laboratory Medicine, Hefei, China
| | - Fei Wang
- Neurosurgical Department, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| | - Guan-Min Jiang
- Department of Clinical Laboratory, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China.
| | - Hao Wang
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
- Core Unit of National Clinical Research Center for Laboratory Medicine, Hefei, China.
| |
Collapse
|
15
|
Li YL, Zhang Y, Chen N, Yan YX. The role of m 6A modification in type 2 diabetes: A systematic review and integrative analysis. Gene 2024; 898:148130. [PMID: 38181926 DOI: 10.1016/j.gene.2024.148130] [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: 11/02/2023] [Revised: 12/14/2023] [Accepted: 01/02/2024] [Indexed: 01/07/2024]
Abstract
This study focuses on the latest developments in the studies of m6A modification and provides an up-to-date summary of the association between m6A modification and type 2 diabetes (T2D). The possible mechanisms of m6A related to T2D were summarized by literature review. The differentially expressed genes (DEGs) of m6A methylase in T2D were analyzed from 12 datasets in Gene Expression Omnibus (GEO). The associations between m6A level and T2D were explored in four electronic databases, including PubMed, EmBase, Web of Science and CNKI. Standard mean difference (SMD) and 95 % confidence interval (95 %CI) was calculated to assess the total effect in integrative analysis. Differential expression genes detected in at least three of six tissues were ZC3H13, YTHDC1/2, and IGF2BP2. LRPPRC were differentially expressed in five tissues except in arterial tissue. A total of 6 studies were included for integrative analysis. The mean m6A levels were significantly lower in T2D than those in normal controls (SMD = -1.35, 95 %CI: -2.58 to -0.11). This systematic review and integrative analysis summarize the previous studies on the association between m6A modification and T2D and the possible role of m6A modification in the progression of T2D, such as abnormal blood glucose, abnormal pancreatic β-cell function, insulin resistance, and abnormal lipid metabolism. The integrative analysis showed that decreased level of m6A was associated with T2D. These findings provide new targets for early detection and treatment for T2D.
Collapse
Affiliation(s)
- Yan-Ling Li
- Department of Epidemiology and Biostatistics, School of Public Health, Capital Medical University, Beijing, China; Municipal Key Laboratory of Clinical Epidemiology, Beijing, China.
| | - Yu Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Capital Medical University, Beijing, China; Municipal Key Laboratory of Clinical Epidemiology, Beijing, China.
| | - Ning Chen
- Department of Epidemiology and Biostatistics, School of Public Health, Capital Medical University, Beijing, China; Municipal Key Laboratory of Clinical Epidemiology, Beijing, China.
| | - Yu-Xiang Yan
- Department of Epidemiology and Biostatistics, School of Public Health, Capital Medical University, Beijing, China; Municipal Key Laboratory of Clinical Epidemiology, Beijing, China.
| |
Collapse
|
16
|
Su X, Qu Y, Mu D. Methyltransferase-like 3 modifications of RNAs: Implications for the pathology in the endocrine system. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167010. [PMID: 38176459 DOI: 10.1016/j.bbadis.2023.167010] [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: 09/26/2023] [Revised: 12/18/2023] [Accepted: 12/27/2023] [Indexed: 01/06/2024]
Abstract
Methyltransferase-like 3 (METTL3) is the most well-known element of N6-methyladenosine modification on RNAs. METTL3 deposits a methyl group onto target RNAs to modify their expression, ultimately regulating various physiological and pathological events. Numerous studies have suggested the significant role of METTL3 in endocrine dysfunction and related disorders. However, reviews that summarize and interpret these studies are lacking. In this review, we systematically analyze such studies, including obesity, type 2 diabetes mellitus (T2DM), T2DM-induced diseases, pancreatic cancer, and thyroid carcinoma. This review indicates that METTL3 contributes remarkably to the endocrine dysfunction and progression of obesity, T2DM, T2DM-induced diseases, pancreatic cancer, and thyroid carcinoma. In conclusion, this review provides a comprehensive interpretation of the mechanism via which METTL3 functions on RNAs and regulates various endocrine dysfunction events and suggest potential associated correlations. Our review, thus, provides a valuable reference for further fundamental studies and clinical applications.
Collapse
Affiliation(s)
- Xiaojuan Su
- Department of Pediatrics/Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Yi Qu
- Department of Pediatrics/Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Dezhi Mu
- Department of Pediatrics/Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), West China Second University Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
17
|
Han F. N6-methyladenosine modification in ischemic stroke: Functions, regulation, and therapeutic potential. Heliyon 2024; 10:e25192. [PMID: 38317953 PMCID: PMC10840115 DOI: 10.1016/j.heliyon.2024.e25192] [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: 06/29/2023] [Revised: 12/09/2023] [Accepted: 01/22/2024] [Indexed: 02/07/2024] Open
Abstract
N6-methyladenosine (m6A) modification is the most frequently occurring internal modification in eukaryotic RNAs. By modulating various aspects of the RNA life cycle, it has been implicated in a wide range of pathological and physiological processes associated with human diseases. Ischemic stroke is a major cause of death and disability worldwide with few treatment options and a narrow therapeutic window, and accumulating evidence has indicated the involvement of m6A modifications in the development and progression of this type of stroke. In this review, which provides insights for the prevention and clinical treatment of stroke, we present an overview of the roles played by m6A modification in ischemic stroke from three main perspectives: (1) the association of m6A modification with established risk factors for stroke, including hypertension, diabetes mellitus, hyperlipidemia, obesity, and heart disease; (2) the roles of m6A modification regulators and their functional regulation in the pathophysiological injury mechanisms of stroke, namely oxidative stress, mitochondrial dysfunction, endothelial dysfunction, neuroinflammation, and cell death processes; and (3) the diagnostic and therapeutic potential of m6A regulators in the treatment of stroke.
Collapse
Affiliation(s)
- Fei Han
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, 100730, China
| |
Collapse
|
18
|
Sun J, Wang Y, Fu H, Kang F, Song J, Xu M, Ning G, Wang J, Wang W, Wang Q. Mettl3-Mediated m6A Methylation Controls Pancreatic Bipotent Progenitor Fate and Islet Formation. Diabetes 2024; 73:237-249. [PMID: 37963393 DOI: 10.2337/db23-0360] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Accepted: 11/06/2023] [Indexed: 11/16/2023]
Abstract
The important role of m6A RNA modification in β-cell function has been established; however, how it regulates pancreatic development and endocrine differentiation remains unknown. Here, we generated transgenic mice lacking RNA methyltransferase-like 3 (Mettl3) specifically in Pdx1+ pancreatic progenitor cells and found the mice with the mutation developed hyperglycemia and hypoinsulinemia at age 2 weeks, along with an atrophic pancreas, reduced islet mass, and abnormal increase in ductal formation. At embryonic day 15.5, Mettl3 deletion had caused a significant loss of Ngn3+ endocrine progenitor cells, which was accompanied by increased Sox9+ ductal precursor cells. We identified histone deacetylase 1 (Hdac1) as the critical direct m6A target in bipotent progenitors, the degeneration of which caused abnormal activation of the Wnt/Notch signaling pathway and blocked endocrine differentiation. This transformation could be manipulated in embryonic pancreatic culture in vitro through regulation of the Mettl3-Hdac1-Wnt/Notch signaling axis. Our finding that Mettl3 determines endocrine lineage by modulating Hdac1 activity during the transition of bipotent progenitors might help in the development of targeted endocrine cell protocols for diabetes treatment. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Jiajun Sun
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the People's Republic of China, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanqiu Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the People's Republic of China, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hui Fu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the People's Republic of China, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fuyun Kang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the People's Republic of China, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiaxi Song
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the People's Republic of China, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Min Xu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the People's Republic of China, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guang Ning
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the People's Republic of China, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jian Wang
- International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weiqing Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the People's Republic of China, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qidi Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the People's Republic of China, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Sino-French Research Center for Life Sciences and Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
19
|
Huang J, Yang F, Liu Y, Wang Y. N6-methyladenosine RNA methylation in diabetic kidney disease. Biomed Pharmacother 2024; 171:116185. [PMID: 38237350 DOI: 10.1016/j.biopha.2024.116185] [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: 11/06/2023] [Revised: 01/13/2024] [Accepted: 01/16/2024] [Indexed: 02/08/2024] Open
Abstract
Diabetic kidney disease (DKD) is a major microvascular complication of diabetes, and hyperglycemic memory associated with diabetes carries the risk of disease occurrence, even after the termination of blood glucose injury. The existence of hyperglycemic memory supports the concept of an epigenetic mechanism involving n6-methyladenosine (m6A) modification. Several studies have shown that m6A plays a key role in the pathogenesis of DKD. This review addresses the role and mechanism of m6A RNA modification in the progression of DKD, including the regulatory role of m6A modification in pathological processes, such as inflammation, oxidative stress, fibrosis, and non-coding (nc) RNA. This reveals the importance of m6A in the occurrence and development of DKD, suggesting that m6A may play a role in hyperglycemic memory phenomenon. This review also discusses how some gray areas, such as m6A modified multiple enzymes, interact to affect the development of DKD and provides countermeasures. In conclusion, this review enhances our understanding of DKD from the perspective of m6A modifications and provides new targets for future therapeutic strategies. In addition, the insights discussed here support the existence of hyperglycemic memory effects in DKD, which may have far-reaching implications for the development of novel treatments. We hypothesize that m6A RNA modification, as a key factor regulating the development of DKD, provides a new perspective for the in-depth exploration of DKD and provides a novel option for the clinical management of patients with DKD.
Collapse
Affiliation(s)
- Jiaan Huang
- Hebei Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Liver and Kidney Diseases, Shijiazhuang 05000, China; Hebei University of Traditional Chinese Medicine, NO.326, Xinshi South Road, Qiaoxi District, Shijiazhuang 05000, China
| | - Fan Yang
- Hebei Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Liver and Kidney Diseases, Shijiazhuang 05000, China; Hebei University of Traditional Chinese Medicine, NO.326, Xinshi South Road, Qiaoxi District, Shijiazhuang 05000, China
| | - Yan Liu
- Hebei Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Liver and Kidney Diseases, Shijiazhuang 05000, China; Hebei University of Traditional Chinese Medicine, NO.326, Xinshi South Road, Qiaoxi District, Shijiazhuang 05000, China
| | - Yuehua Wang
- Hebei Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Liver and Kidney Diseases, Shijiazhuang 05000, China; Hebei University of Traditional Chinese Medicine, NO.326, Xinshi South Road, Qiaoxi District, Shijiazhuang 05000, China.
| |
Collapse
|
20
|
Zhou SM, Yao XM, Cheng Y, Xing YJ, Sun Y, Hua Q, Wan SJ, Meng XJ. Metformin enhances METTL14-Mediated m6A methylation to alleviate NIT-1 cells apoptosis induced by hydrogen peroxide. Heliyon 2024; 10:e24432. [PMID: 38312705 PMCID: PMC10835167 DOI: 10.1016/j.heliyon.2024.e24432] [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: 05/31/2023] [Revised: 01/08/2024] [Accepted: 01/09/2024] [Indexed: 02/06/2024] Open
Abstract
Injuries to pancreatic β-cells are intricately linked to the onset of diabetes mellitus (DM). Metformin (Met), one of the most widely prescribed medications for diabetes and metabolic disorders, has been extensively studied for its antioxidant, anti-aging, anti-glycation, and hepatoprotective activities. N6-methyladenosine (m6A) plays a crucial role in the regulation of β-cell growth and development, and its dysregulation is associated with metabolic disorders. This study aimed to elucidate the mechanistic basis of m6A involvement in the protective effects of Met against oxidative damage in pancreatic β-cells. Hydrogen peroxide (H2O2) was employed to induce β-cell damage. Remarkably, Met treatment effectively increased methylation levels and the expression of the methyltransferase METTL14, subsequently reducing H2O2-induced apoptosis. Knocking down METTL14 expression using siRNA significantly compromised cell viability. Conversely, targeted overexpression of METTL14 specifically in β-cells substantially enhanced their capacity to withstand H2O2-induced stress. Molecular evidence suggests that the anti-apoptotic properties of Met may be mediated through Bcl-xL and Bim proteins. In conclusion, our findings indicate that Met induces METTL14-mediated alterations in m6A methylation levels, thereby shielding β-cells from apoptosis and oxidative damage induced by oxidative stress.
Collapse
Affiliation(s)
- Si-Min Zhou
- Department of Endocrinology, The First Affiliated Hospital of Wannan Medical College, Wannan Medical College, Wuhu, 241002, China
- Key Laboratory of Non-coding RNA Transformation Research of Anhui Higher Education Institution, Wannan Medical College, Wuhu, 241002, China
| | - Xin-Ming Yao
- Department of Endocrinology, The First Affiliated Hospital of Wannan Medical College, Wannan Medical College, Wuhu, 241002, China
| | - Yi Cheng
- Department of Endocrinology, The First Affiliated Hospital of Wannan Medical College, Wannan Medical College, Wuhu, 241002, China
| | - Yu-Jie Xing
- Department of Endocrinology, The First Affiliated Hospital of Wannan Medical College, Wannan Medical College, Wuhu, 241002, China
- Key Laboratory of Non-coding RNA Transformation Research of Anhui Higher Education Institution, Wannan Medical College, Wuhu, 241002, China
| | - Yue Sun
- Department of Endocrinology, The First Affiliated Hospital of Wannan Medical College, Wannan Medical College, Wuhu, 241002, China
- Key Laboratory of Non-coding RNA Transformation Research of Anhui Higher Education Institution, Wannan Medical College, Wuhu, 241002, China
| | - Qiang Hua
- Department of Endocrinology, The First Affiliated Hospital of Wannan Medical College, Wannan Medical College, Wuhu, 241002, China
| | - Shu-Jun Wan
- Key Laboratory of Non-coding RNA Transformation Research of Anhui Higher Education Institution, Wannan Medical College, Wuhu, 241002, China
| | - Xiang-Jian Meng
- Department of Endocrinology, The First Affiliated Hospital of Wannan Medical College, Wannan Medical College, Wuhu, 241002, China
| |
Collapse
|
21
|
Sun YH, Zhao TJ, Li LH, Wang Z, Li HB. Emerging role of N6-methyladenosine in the homeostasis of glucose metabolism. Am J Physiol Endocrinol Metab 2024; 326:E1-E13. [PMID: 37938178 DOI: 10.1152/ajpendo.00225.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 09/21/2023] [Accepted: 11/02/2023] [Indexed: 11/09/2023]
Abstract
N6-methyladenosine (m6A) is the most prevalent post-transcriptional internal RNA modification, which is involved in the regulation of diverse physiological processes. Dynamic and reversible m6A modification has been shown to regulate glucose metabolism, and dysregulation of m6A modification contributes to glucose metabolic disorders in multiple organs and tissues including the pancreas, liver, adipose tissue, skeletal muscle, kidney, blood vessels, and so forth. In this review, the role and molecular mechanism of m6A modification in the regulation of glucose metabolism were summarized, the potential therapeutic strategies that improve glucose metabolism by targeting m6A modifiers were outlined, and feasible directions of future research in this field were discussed as well, providing clues for translational research on combating metabolic diseases based on m6A modification in the future.
Collapse
Affiliation(s)
- Yuan-Hai Sun
- Institute of Pharmacology, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, People's Republic of China
| | - Teng-Jiao Zhao
- Institute of Pharmacology, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, People's Republic of China
| | - Ling-Huan Li
- Institute of Pharmacology, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, People's Republic of China
- College of Chemistry and Materials Science, Zhejiang Normal University, Jinhua, People's Republic of China
| | - Zhen Wang
- Center for Laboratory Medicine, Allergy Center, Department of Transfusion Medicine, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, People's Republic of China
| | - Han-Bing Li
- Institute of Pharmacology, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, People's Republic of China
- Section of Endocrinology, School of Medicine, Yale University, New Haven, Connecticut, United States
| |
Collapse
|
22
|
Sun Y, Jin D, Zhang Z, Ji H, An X, Zhang Y, Yang C, Sun W, Zhang Y, Duan Y, Kang X, Jiang L, Zhao X, Lian F. N6-methyladenosine (m6A) methylation in kidney diseases: Mechanisms and therapeutic potential. BIOCHIMICA ET BIOPHYSICA ACTA. GENE REGULATORY MECHANISMS 2023; 1866:194967. [PMID: 37553065 DOI: 10.1016/j.bbagrm.2023.194967] [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: 03/27/2023] [Revised: 07/31/2023] [Accepted: 08/01/2023] [Indexed: 08/10/2023]
Abstract
The N6-methyladenosine (m6A) modification is regulated by methylases, commonly referred to as "writers," and demethylases, known as "erasers," leading to a dynamic and reversible process. Changes in m6A levels have been implicated in a wide range of cellular processes, including nuclear RNA export, mRNA metabolism, protein translation, and RNA splicing, establishing a strong correlation with various diseases. Both physiologically and pathologically, m6A methylation plays a critical role in the initiation and progression of kidney disease. The methylation of m6A may also facilitate the early diagnosis and treatment of kidney diseases, according to accumulating research. This review aims to provide a comprehensive overview of the potential role and mechanism of m6A methylation in kidney diseases, as well as its potential application in the treatment of such diseases. There will be a thorough examination of m6A methylation mechanisms, paying particular attention to the interplay between m6A writers, m6A erasers, and m6A readers. Furthermore, this paper will elucidate the interplay between various kidney diseases and m6A methylation, summarize the expression patterns of m6A in pathological kidney tissues, and discuss the potential therapeutic benefits of targeting m6A in the context of kidney diseases.
Collapse
Affiliation(s)
- Yuting Sun
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - De Jin
- Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou, China
| | - Ziwei Zhang
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Hangyu Ji
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xuedong An
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuehong Zhang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Cunqing Yang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Wenjie Sun
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuqing Zhang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yingying Duan
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaomin Kang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Linlin Jiang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xuefei Zhao
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Fengmei Lian
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
23
|
Huang C, Chen W, Wang X. Studies on the fat mass and obesity-associated (FTO) gene and its impact on obesity-associated diseases. Genes Dis 2023; 10:2351-2365. [PMID: 37554175 PMCID: PMC10404889 DOI: 10.1016/j.gendis.2022.04.014] [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: 12/19/2021] [Revised: 03/29/2022] [Accepted: 04/01/2022] [Indexed: 10/18/2022] Open
Abstract
Obesity has become a major health crisis in the past ∼50 years. The fat mass and obesity-associated (FTO) gene, identified by genome-wide association studies (GWAS), was first reported to be positively associated with obesity in humans. Mice with more copies of the FTO gene were observed to be obese, while loss of the gene in mice was found to protect from obesity. Later, FTO was found to encode an m6A RNA demethylase and has a profound effect on many biological and metabolic processes. In this review, we first summarize recent studies that demonstrate the critical roles and regulatory mechanisms of FTO in obesity and metabolic disease. Second, we discuss the ongoing debates concerning the association between FTO polymorphisms and obesity. Third, since several small molecule drugs and micronutrients have been found to regulate metabolic homeostasis through controlling the expression or activity of FTO, we highlight the broad potential of targeting FTO for obesity treatment. Improving our understanding of FTO and the underlying mechanisms may provide new approaches for treating obesity and metabolic diseases.
Collapse
Affiliation(s)
- Chaoqun Huang
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
- Key Laboratory of Molecular Animal Nutrition, Zhejiang University, Ministry of Education, Hangzhou, Zhejiang 310058, China
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, Zhejiang 310058, China
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou, Zhejiang 310058, China
| | - Wei Chen
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
- Key Laboratory of Molecular Animal Nutrition, Zhejiang University, Ministry of Education, Hangzhou, Zhejiang 310058, China
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, Zhejiang 310058, China
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou, Zhejiang 310058, China
| | - Xinxia Wang
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
- Key Laboratory of Molecular Animal Nutrition, Zhejiang University, Ministry of Education, Hangzhou, Zhejiang 310058, China
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, Zhejiang 310058, China
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou, Zhejiang 310058, China
| |
Collapse
|
24
|
Li X, Liu C, Zhang Z, Li X, Yao Z, Dong Y, Wang X, Chen Z. Hepatocyte-specific Wtap deficiency promotes hepatocellular carcinoma by activating GRB2-ERK depending on downregulation of proteasome-related genes. J Biol Chem 2023; 299:105301. [PMID: 37777158 PMCID: PMC10630636 DOI: 10.1016/j.jbc.2023.105301] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 09/05/2023] [Accepted: 09/18/2023] [Indexed: 10/02/2023] Open
Abstract
Wilm's tumor 1-associating protein (WTAP), a regulatory protein of the m6A methyltransferase complex, has been found to play a role in regulating various physiological and pathological processes. However, the in vivo role of WTAP in the pathogenesis of hepatocellular carcinoma (HCC) is unknown. In this study, we have elucidated the crucial role of WTAP in HCC progression and shown that hepatic deletion of Wtap promotes HCC pathogenesis through activation of multiple signaling pathways. A single dose of diethylnitrosamine injection causes more and larger HCCs in hepatocyte-specific Wtap knockout (Wtap-HKO) mice than Wtapflox/flox mice fed with either normal chow diet or a high-fat diet. Elevated CD36, IGFBP1 (insulin-like growth factor-binding protein 1), and chemokine (C-C motif) ligand 2 (CCL2) expression leads to steatosis and inflammation in the Wtap-HKO livers. The hepatocyte proliferation is dramatically increased in Wtap-HKO mice, which is due to higher activation of extracellular signal-regulated kinase (ERK) and signal transducer and activator of transcription-3 signaling pathways. Hepatic deletion of Wtap activates the ERK signaling pathway by increasing the protein stability of GRB2 and ERK1/2, which is due to the decreased expression of proteasome-related genes. Restoring PSMB4 or PSMB6 (two key components of the proteasome) leads to the downregulation of GRB2 and ERK1/2 in Wtap-HKO hepatocytes. Mechanistically, WTAP interacts with RNA polymerase II and H3K9ac to maintain expression of proteasome-related genes. These results demonstrate that hepatic deletion of Wtap promotes HCC progression through activating GRB2-ERK1/2-mediated signaling pathway depending on the downregulation of proteasome-related genes especially Psmb4 and Psmb6.
Collapse
Affiliation(s)
- Xinzhi Li
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Chunhong Liu
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Zhimin Zhang
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Xueying Li
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Zhicheng Yao
- Department of General surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yanbin Dong
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Xin Wang
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Zheng Chen
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin, China.
| |
Collapse
|
25
|
Kang Q, Zhu X, Ren D, Ky A, MacDougald OA, O'Rourke RW, Rui L. Adipose METTL14-Elicited N 6 -Methyladenosine Promotes Obesity, Insulin Resistance, and NAFLD Through Suppressing β Adrenergic Signaling and Lipolysis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2301645. [PMID: 37526326 PMCID: PMC10558699 DOI: 10.1002/advs.202301645] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 06/08/2023] [Indexed: 08/02/2023]
Abstract
White adipose tissue (WAT) lipolysis releases free fatty acids as a key energy substance to support metabolism in fasting, cold exposure, and exercise. Atgl, in concert with Cgi-58, catalyzes the first lipolytic reaction. The sympathetic nervous system (SNS) stimulates lipolysis via neurotransmitter norepinephrine that activates adipocyte β adrenergic receptors (Adrb1-3). In obesity, adipose Adrb signaling and lipolysis are impaired, contributing to pathogenic WAT expansion; however, the underling mechanism remains poorly understood. Recent studies highlight importance of N6 -methyladenosine (m6A)-based RNA modification in health and disease. METTL14 heterodimerizes with METTL3 to form an RNA methyltransferase complex that installs m6A in transcripts. Here, this work shows that adipose Mettl3 and Mettl14 are influenced by fasting, refeeding, and insulin, and are upregulated in high fat diet (HFD) induced obesity. Adipose Adrb2, Adrb3, Atgl, and Cgi-58 transcript m6A contents are elevated in obesity. Mettl14 ablation decreases these transcripts' m6A contents and increases their translations and protein levels in adipocytes, thereby increasing Adrb signaling and lipolysis. Mice with adipocyte-specific deletion of Mettl14 are resistant to HFD-induced obesity, insulin resistance, glucose intolerance, and nonalcoholic fatty liver disease (NAFLD). These results unravel a METTL14/m6A/translation pathway governing Adrb signaling and lipolysis. METTL14/m6A-based epitranscriptomic reprogramming impairs adipose Adrb signaling and lipolysis, promoting obesity, NAFLD, and metabolic disease.
Collapse
Affiliation(s)
- Qianqian Kang
- Department of Molecular and Integrative PhysiologyUniversity of Michigan Medical SchoolAnn ArborMI48109USA
- Elizabeth Weiser Caswell Diabetes InstituteUniversity of MichiganAnn ArborMI48109USA
| | - Xiaorong Zhu
- Department of Molecular and Integrative PhysiologyUniversity of Michigan Medical SchoolAnn ArborMI48109USA
- Department of EndocrinologyBeijing Tongren HospitalCapital Medical UniversityBeijing Diabetes InstituteBeijing100730China
| | - Decheng Ren
- Department of MedicineUniversity of ChicagoChicagoIL60637USA
| | - Alexander Ky
- Department of SurgeryUniversity of Michigan Medical SchoolAnn ArborMI48109USA
| | - Ormond A. MacDougald
- Department of Molecular and Integrative PhysiologyUniversity of Michigan Medical SchoolAnn ArborMI48109USA
- Elizabeth Weiser Caswell Diabetes InstituteUniversity of MichiganAnn ArborMI48109USA
- Department of Internal MedicineUniversity of Michigan Medical SchoolAnn ArborMI48109USA
| | - Robert W. O'Rourke
- Department of SurgeryUniversity of Michigan Medical SchoolAnn ArborMI48109USA
- Department of SurgeryVeterans Affairs Ann Arbor Healthcare SystemAn ArborMI48105USA
| | - Liangyou Rui
- Department of Molecular and Integrative PhysiologyUniversity of Michigan Medical SchoolAnn ArborMI48109USA
- Elizabeth Weiser Caswell Diabetes InstituteUniversity of MichiganAnn ArborMI48109USA
- Department of Internal MedicineUniversity of Michigan Medical SchoolAnn ArborMI48109USA
| |
Collapse
|
26
|
Kahraman S, De Jesus DF, Wei J, Brown NK, Zou Z, Hu J, He C, Kulkarni RN. m 6 A mRNA Methylation Regulates Early Pancreatic β-Cell Differentiation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.03.551675. [PMID: 37577492 PMCID: PMC10418275 DOI: 10.1101/2023.08.03.551675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
N 6 -methyladenosine (m 6 A) is the most abundant chemical modification in mRNA, and plays important roles in human and mouse embryonic stem cell pluripotency, maintenance, and differentiation. We have recently reported, for the first time, the role of m 6 A in the postnatal control of β-cell function in physiological states and in Type 1 and 2 Diabetes. However, the precise mechanisms by which m 6 A acts to regulate the development of human and mouse β-cells are unexplored. Here, we show that the m 6 A landscape is dynamic during human pancreas development, and that METTL14, one of the m 6 A writer complex proteins, is essential for the early differentiation of both human and mouse β-cells.
Collapse
|
27
|
Liu C, Li X, Gao M, Dong Y, Chen Z. Downregulation of hepatic METTL3 contributes to APAP-induced liver injury in mice. JHEP Rep 2023; 5:100766. [PMID: 37456679 PMCID: PMC10338307 DOI: 10.1016/j.jhepr.2023.100766] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 04/06/2023] [Accepted: 04/12/2023] [Indexed: 07/18/2023] Open
Abstract
Background & Aims Acetaminophen (APAP) overdose is a major cause of acute liver failure in the Western world, but its molecular mechanisms are not fully understood. Methyltransferase-like 3 (METTL3) is a core N6-methyl-adenosine (m6A) RNA methyltransferase that has been shown to regulate many physiological and pathological processes. This study aimed to investigate the role of METTL3 in APAP-induced liver injury in mice. Methods Hepatocyte-specific Mettl3 knockout (Mettl3-HKO) mice and adenovirus-mediated gene overexpression or knockdown were used. We assayed APAP-induced liver injury by measuring serum alanine aminotransferase/aspartate aminotransferase activity, necrotic area, cell death, reactive oxygen species levels and activation of signalling pathways. We also performed mechanistic studies using a variety of assays and molecular techniques. Results Hepatic METTL3 is downregulated in APAP-induced liver injury, and hepatocyte-specific deletion of Mettl3 accelerates APAP-induced liver injury, leading to increased mortality as a result of the dramatic activation of the mitogen-activated protein kinase kinase 4 (MKK4) / c-Jun NH2-terminal kinase (JNK) signalling pathway. Inhibition of JNK by SP600125 largely blocks APAP-induced liver injury in Mettl3-HKO mice. Hepatic deletion of Mettl3 activates the MKK4/JNK signalling pathway by increasing the protein stability of MKK4 and JNK1/2 as a result of decreased proteasome activity. Restoration of proteasome activity by overexpression of proteasome 20S subunit beta 4 (PSMB4) or proteasome 20S subunit beta 6 (PSMB6) leads to the downregulation of MKK4 and JNK in Mettl3-HKO hepatocytes. Mechanistically, METTL3 interacts with RNA polymerase II and active histone modifications such as H3K9ac, H3K27ac, and H3K36me3 to maintain the expression of proteasome-related genes. Conclusions Our study demonstrated that downregulation of METTL3 promotes APAP-induced liver injury by decreasing proteasome activity and thereby enhancing activity of the MKK4/JNK signalling pathway. Impact and Implications Acetaminophen (APAP) overdose is a key cause of acute liver failure in the Western world, but its molecular mechanisms are not fully understood. We demonstrated in this study that methyltransferase-like 3 (METTL3), a core m6A RNA methyltransferase, is downregulated in APAP-induced liver injury, which exacerbates APAP-induced liver injury through enhancing the MKK4/JNK signalling pathway with involvement of the decreased proteasome activity.
Collapse
Affiliation(s)
- Chunhong Liu
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| | - Xinzhi Li
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| | - Ming Gao
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| | - Yanbin Dong
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Zheng Chen
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| |
Collapse
|
28
|
Zhang H, Gu Y, Gang Q, Huang J, Xiao Q, Ha X. N6-methyladenosine RNA modification: an emerging molecule in type 2 diabetes metabolism. Front Endocrinol (Lausanne) 2023; 14:1166756. [PMID: 37484964 PMCID: PMC10360191 DOI: 10.3389/fendo.2023.1166756] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 06/16/2023] [Indexed: 07/25/2023] Open
Abstract
Type 2 diabetes (T2D) is a metabolic disease with an increasing rate of incidence worldwide. Despite the considerable progress in the prevention and intervention, T2D and its complications cannot be reversed easily after diagnosis, thereby necessitating an in-depth investigation of the pathophysiology. In recent years, the role of epigenetics has been increasingly demonstrated in the disease, of which N6-methyladenosine (m6A) is one of the most common post-transcriptional modifications. Interestingly, patients with T2D show a low m6A abundance. Thus, a comprehensive analysis and understanding of this phenomenon would improve our understanding of the pathophysiology, as well as the search for new biomarkers and therapeutic approaches for T2D. In this review, we systematically introduced the metabolic roles of m6A modification in organs, the metabolic signaling pathways involved, and the effects of clinical drugs on T2D.
Collapse
Affiliation(s)
- Haocheng Zhang
- The Second School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, China
- Department of Clinical Laboratory, The 940th Hospital of Joint Logistics Support Force of Chinese People’s Liberation Army, Lanzhou, Gansu, China
- Key Laboratory of Stem Cells and Gene Drugs of Gansu Province, Lanzhou, Gansu, China
| | - Yan Gu
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Qiaojian Gang
- The Second School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, China
| | - Jing Huang
- School of Public Health, Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu, China
| | - Qian Xiao
- School of Public Health, Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu, China
| | - Xiaoqin Ha
- The Second School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, China
- Department of Clinical Laboratory, The 940th Hospital of Joint Logistics Support Force of Chinese People’s Liberation Army, Lanzhou, Gansu, China
- Key Laboratory of Stem Cells and Gene Drugs of Gansu Province, Lanzhou, Gansu, China
| |
Collapse
|
29
|
Benak D, Benakova S, Plecita-Hlavata L, Hlavackova M. The role of m 6A and m 6Am RNA modifications in the pathogenesis of diabetes mellitus. Front Endocrinol (Lausanne) 2023; 14:1223583. [PMID: 37484960 PMCID: PMC10360938 DOI: 10.3389/fendo.2023.1223583] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 06/26/2023] [Indexed: 07/25/2023] Open
Abstract
The rapidly developing research field of epitranscriptomics has recently emerged into the spotlight of researchers due to its vast regulatory effects on gene expression and thereby cellular physiology and pathophysiology. N6-methyladenosine (m6A) and N6,2'-O-dimethyladenosine (m6Am) are among the most prevalent and well-characterized modified nucleosides in eukaryotic RNA. Both of these modifications are dynamically regulated by a complex set of epitranscriptomic regulators called writers, readers, and erasers. Altered levels of m6A and also several regulatory proteins were already associated with diabetic tissues. This review summarizes the current knowledge and gaps about m6A and m6Am modifications and their respective regulators in the pathophysiology of diabetes mellitus. It focuses mainly on the more prevalent type 2 diabetes mellitus (T2DM) and its treatment by metformin, the first-line antidiabetic agent. A better understanding of epitranscriptomic modifications in this highly prevalent disease deserves further investigation and might reveal clinically relevant discoveries in the future.
Collapse
Affiliation(s)
- Daniel Benak
- Laboratory of Developmental Cardiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia
- Department of Physiology, Faculty of Science, Charles University, Prague, Czechia
| | - Stepanka Benakova
- Laboratory of Pancreatic Islet Research, Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia
- First Faculty of Medicine, Charles University, Prague, Czechia
| | - Lydie Plecita-Hlavata
- Laboratory of Pancreatic Islet Research, Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia
| | - Marketa Hlavackova
- Laboratory of Developmental Cardiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia
| |
Collapse
|
30
|
Yan B, Li X, Peng M, Zuo Y, Wang Y, Liu P, Ren W, Jin X. The YTHDC1/GLUT3/RNF183 axis forms a positive feedback loop that modulates glucose metabolism and bladder cancer progression. Exp Mol Med 2023; 55:1145-1158. [PMID: 37258572 PMCID: PMC10318083 DOI: 10.1038/s12276-023-00997-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 02/09/2023] [Accepted: 02/24/2023] [Indexed: 06/02/2023] Open
Abstract
Aberrant glucose metabolism is a characteristic of bladder cancer. Hyperglycemia contributes to the development and progression of bladder cancer. However, the underlying mechanism by which hyperglycemia promotes the aggressiveness of cancers, especially bladder cancer, is still incompletely understood. N6-methyladenosine (m6A) modification is a kind of methylation modification occurring at the N6 position of adenosine that is important for the pathogenesis of urological tumors. Recently, it was found that the m6A reader YTHDC1 is regulated by high-glucose conditions. In our study, we revealed that YTHDC1 is not only regulated by high-glucose conditions but is also downregulated in bladder cancer tissue and associated with the prognosis of cancer. We also showed that YTHDC1 suppresses the malignant progression of and the glycolytic process in bladder cancer cells in an m6A-dependent manner and determined that this effect is partially mediated by GLUT3. Moreover, GLUT3 was found to destabilize YTHDC1 by upregulating RNF183 expression. In summary, we identified a novel YTHDC1/GLUT3/RNF183 feedback loop that regulates disease progression and glucose metabolism in bladder cancer. Collectively, this study provides new insight regarding the pathogenesis of bladder cancer under hyperglycemic conditions and might reveal ideal candidates for the development of drugs for bladder cancer.
Collapse
Affiliation(s)
- Bin Yan
- Department of Urology, The Second Xiangya Hospital, Central South University, 410011, Changsha, Hunan, China
- Uro-Oncology Institute of Central South University, 410011, Changsha, Hunan, China
| | - Xurui Li
- Department of Urology, The Second Xiangya Hospital, Central South University, 410011, Changsha, Hunan, China
- Uro-Oncology Institute of Central South University, 410011, Changsha, Hunan, China
| | - Mou Peng
- Department of Urology, The Second Xiangya Hospital, Central South University, 410011, Changsha, Hunan, China
- Uro-Oncology Institute of Central South University, 410011, Changsha, Hunan, China
| | - Yali Zuo
- Department of Urology, The Second Xiangya Hospital, Central South University, 410011, Changsha, Hunan, China
| | - Yinhuai Wang
- Department of Urology, The Second Xiangya Hospital, Central South University, 410011, Changsha, Hunan, China
| | - Pian Liu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China.
| | - Weigang Ren
- Department of Urology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, 410005, Changsha, Hunan, China.
| | - Xin Jin
- Department of Urology, The Second Xiangya Hospital, Central South University, 410011, Changsha, Hunan, China.
- Uro-Oncology Institute of Central South University, 410011, Changsha, Hunan, China.
| |
Collapse
|
31
|
Zhu X, Zhou C, Zhao S, Zheng Z. Role of m6A methylation in retinal diseases. Exp Eye Res 2023; 231:109489. [PMID: 37084873 DOI: 10.1016/j.exer.2023.109489] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 03/06/2023] [Accepted: 04/19/2023] [Indexed: 04/23/2023]
Abstract
Retinal diseases remain among the leading causes of visual impairment in developed countries, despite great efforts in prevention and early intervention. Due to the limited efficacy of current retinal therapies, novel therapeutic methods are urgently required. Over the past two decades, advances in next-generation sequencing technology have facilitated research on RNA modifications, which can elucidate the relevance of epigenetic mechanisms to disease. N6-methyladenosine (m6A), formed by methylation of adenosine at the N6-position, is the most widely studied RNA modification and plays an important role in RNA metabolism. It is dynamically regulated by writers (methyltransferases) and erasers (demethylases), and recognized by readers (m6A binding proteins). Although the discovery of m6A methylation can be traced back to the 1970s, its regulatory roles in retinal diseases are rarely appreciated. Here, we provide an overview of m6A methylation, and discuss its effects and possible mechanisms on retinal diseases, including diabetic retinopathy, age-related macular degeneration, retinoblastoma, retinitis pigmentosa, and proliferative vitreoretinopathy. Furthermore, we highlight potential agents targeting m6A methylation for retinal disease treatment and discuss the limitations and challenges of research in the field of m6A methylation.
Collapse
Affiliation(s)
- Xinyu Zhu
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China; National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, 200080, China
| | - Chuandi Zhou
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China; National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, 200080, China
| | - Shuzhi Zhao
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China; National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, 200080, China.
| | - Zhi Zheng
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China; National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, 200080, China.
| |
Collapse
|
32
|
Liu J, Lin Y, Peng C, Jiang C, Li J, Wang W, Luo S, Fu P, Lin Z, Liang Y, Shen H, Lin Y, Wei J. Bisphenol F induced hyperglycemia via activation of oxidative stress-responsive miR-200 family in the pancreas. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 255:114769. [PMID: 36924560 DOI: 10.1016/j.ecoenv.2023.114769] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 03/06/2023] [Accepted: 03/09/2023] [Indexed: 06/18/2023]
Abstract
Bisphenol F (BPF), BPS and BPAF are gaining popularity as main substitutes to BPA, but there is no clear evidence that these compounds disrupt glycemic homeostasis in the same way. In this study, four bisphenols were administered to C57BL/6 J mice, and showed that the serum insulin was elevated in the BPA and BPS exposed mice, whereas BPF exposed mice exhibited lower serum insulin and higher blood glucose. BPF decreased oxidized glutathione/reduced glutathione ratio (GSSG/GSH) and N6-methyladenosine (m6A) levels, which was responsible for pancreatic apoptosis in mice. Additionally, the downregulation of Nrf2 and the aberrant regulation of the p53-lncRNA H19 signaling pathway further increased miR-200 family in the BPF-exposed pancreas. The miR-200 family directly suppressed Mettl14 and Xiap by targeting their 3' UTR, leading to islet apoptosis. Antioxidant treatment not only elevated m6A levels and insulin contents but also suppressed the miR-200 family in the pancreas, ultimately improving BPF-induced hyperglycemia. Taken together, miR-200 family could serve as a potential oxidative stress-responsive regulator in the pancreas. And moreover, we demonstrated a novel toxicological mechanism in that BPF disrupted the Keap1-Nrf2 redox system to upregulate miR-141/200b/c which controlled pancreatic insulin production and apoptosis via Mettl14 and Xiap, respectively. As the major surrogates of BPA in various applications, BPF was also diabetogenic, which warrants attention in future research.
Collapse
Affiliation(s)
- Jintao Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Yilong Lin
- Department of Basic Medical Sciences, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Cai Peng
- Department of Basic Medical Sciences, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Chunyang Jiang
- Department of Thoracic Surgery, Tianjin Union Medical Center, Nankai University, 190 Jieyuan Road, Hongqiao District, Tianjin 300121, China
| | - Juan Li
- Department of Basic Medical Sciences, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Wenyu Wang
- Department of Basic Medical Sciences, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Shuyue Luo
- Department of Basic Medical Sciences, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Pengbin Fu
- Department of Basic Medical Sciences, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Zhenxin Lin
- Department of Basic Medical Sciences, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Yujie Liang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Heqing Shen
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen 361102, China.
| | - Yi Lin
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen 361102, China.
| | - Jie Wei
- Department of Basic Medical Sciences, School of Medicine, Xiamen University, Xiamen 361102, China.
| |
Collapse
|
33
|
Yang K, Sun J, Zhang Z, Xiao M, Ren D, Liu SM. Reduction of mRNA m 6A associates with glucose metabolism via YTHDC1 in human and mice. Diabetes Res Clin Pract 2023; 198:110607. [PMID: 36878322 DOI: 10.1016/j.diabres.2023.110607] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 02/23/2023] [Accepted: 03/01/2023] [Indexed: 03/07/2023]
Abstract
AIMS N6-methyladenosine (m6A) in mRNA is involved in glucose metabolism. Our goal is to investigate the relationship of glucose metabolism, m6A and YTH domain-containing protein 1 (YTHDC1), a binding protein to m6A, in the development of type 2 diabetes (T2D). METHODS HPLC-MS/MS and qRT-PCR were used to quantify m6A and YTHDC1 levels in white blood cells from patients with T2D and healthy individuals. MIP-CreERT and tamoxifen treatment were used to create β-cell Ythdc1 knockout mice (βKO). m6A sequencing and RNA sequencing were performed in wildtype/βKO islets and MIN6 cells to identify the differential genes. RESULTS In T2D patients, both of m6A and YTHDC1 levels were reduced and associated with fasting glucose. Deletion of Ythdc1 resulted in glucose intolerance and diabetes due to decreased insulin secretion, even though β-cell mass in βKO mice was comparable to wildtype mice. Moreover, Ythdc1 was shown to bind to SRSF3 (serine/arginine-rich splicing factor 3) and CPSF6 (cleavage and polyadenylation specific factor 6) in β-cells. CONCLUSIONS Our data suggested that YTHDC1 may regulate mRNA splicing and export by interacting with SRSF3 and CPSF6 to modulate glucose metabolism via regulating insulin secretion, implying YTHDC1 might be a novel potential target for lowing glucose.
Collapse
Affiliation(s)
- Kun Yang
- Department of Clinical Laboratory, Center for Gene Diagnosis, and Program of Clinical Laboratory, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province 430071, China
| | - Juan Sun
- Department of Neurobiology, The University of Chicago, 5841, S. Maryland Avenue, MC 1027, Chicago, IL 60637, USA
| | - Zijie Zhang
- Department of Chemistry, The University of Chicago, 5841, S. Maryland Avenue, MC 1027, Chicago, IL 60637, USA
| | - Mengyao Xiao
- Department of Clinical Laboratory, Center for Gene Diagnosis, and Program of Clinical Laboratory, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province 430071, China
| | - Decheng Ren
- Department of Medicine, The University of Chicago, 5841 S. Maryland Avenue, MC 1027, Chicago, IL 60637, USA.
| | - Song-Mei Liu
- Department of Clinical Laboratory, Center for Gene Diagnosis, and Program of Clinical Laboratory, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province 430071, China.
| |
Collapse
|
34
|
Li X, Yang Y, Li Z, Wang Y, Qiao J, Chen Z. Deficiency of WTAP in islet beta cells results in beta cell failure and diabetes in mice. Diabetologia 2023; 66:1084-1096. [PMID: 36920524 DOI: 10.1007/s00125-023-05900-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Accepted: 02/13/2023] [Indexed: 03/16/2023]
Abstract
AIMS/HYPOTHESIS N6-methyladenosine (m6A) mRNA methylation and m6A-related proteins (methyltransferase-like 3 [METTL3], methyltransferase-like 14 [METTL14] and YTH domain containing 1 [YTHDC1]) have been shown to regulate islet beta cell function and the pathogenesis of diabetes. However, whether Wilms' tumour 1-associating protein (WTAP), a key regulator of the m6A RNA methyltransferase complex, regulates islet beta cell failure during pathogenesis of diabetes is largely unknown. The present study aimed to investigate the role of WTAP in the regulation of islet beta cell failure and diabetes. METHODS Islet beta cell-specific Wtap-knockout and beta cell-specific Mettl3-overexpressing mice were generated for this study. Blood glucose, glucose tolerance, serum insulin, glucose-stimulated insulin secretion (both in vivo and in vitro), insulin levels, glucagon levels and beta cell apoptosis were examined. RNA-seq and MeRIP-seq were performed, and the data were well analysed. RESULTS WTAP was downregulated in islet beta cells in type 2 diabetes, due to lipotoxicity and chronic inflammation, and islet beta cell-specific deletion of Wtap (Wtap-betaKO) induced beta cell failure and diabetes. Wtap-betaKO mice showed severe hyperglycaemia (above 20 mmol/l [360 mg/dl]) from 8 weeks of age onwards. Mechanistically, WTAP deficiency decreased m6A mRNA modification and reduced the expression of islet beta cell-specific transcription factors and insulin secretion-related genes by reducing METTL3 protein levels. Islet beta cell-specific overexpression of Mettl3 partially reversed the abnormalities observed in Wtap-betaKO mice. CONCLUSIONS/INTERPRETATION WTAP plays a key role in maintaining beta cell function by regulating m6A mRNA modification depending on METTL3, and the downregulation of WTAP leads to beta cell failure and diabetes. DATA AVAILABILITY The RNA-seq and MeRIP-seq datasets generated during the current study are available in the Gene Expression Omnibus database repository ( https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE215156 ; https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE215360 ).
Collapse
Affiliation(s)
- Xinzhi Li
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Ying Yang
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Zhenzhi Li
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Yuqin Wang
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Jingting Qiao
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China
| | - Zheng Chen
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin, China.
| |
Collapse
|
35
|
Cai Y, Chen T, Wang M, Deng L, Li C, Fu S, Xie K. N6-methylation of RNA-bound adenosine regulator HNRNPC promotes vascular endothelial dysfunction in type 2 diabetes mellitus by activating the PSEN1-mediated Notch pathway. Diabetes Res Clin Pract 2023; 197:110261. [PMID: 36681355 DOI: 10.1016/j.diabres.2023.110261] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 12/26/2022] [Accepted: 01/16/2023] [Indexed: 01/20/2023]
Abstract
AIM The regulatory mechanism of m6A regulators in vascular endothelial function of type 2 diabetes mellitus (T2DM) remains largely unknown. We addressed this issue based on the data retrieved Gene Expression Omnibus (GEO) database and experimental validations. METHODS Expression of m6A methylation regulators was evaluated in T2DM samples of GSE76894 dataset and GSE156341 dataset. Further analysis of candidate m6A methylation regulators was conducted in the thoracic aorta of db/db mice and high glucose (HG)-induced human umbilical vein endothelial cells (HUVECs). Ectopic expression and depletion experiments were conducted to detect effects of m6A methylation regulators on vascular endothelial function in T2DM. RESULTS It emerged that three m6A methylation regulators (HNRNPC, RBM15B, and ZC3H13) were highly expressed in T2DM, which were related to vascular EC function, showing diagnostic values for T2DM. HNRNPC expression in the thoracic aorta of db/db mice was higher than that in heterozygous db mice, and HNRNPC expression in HG-induced HUVECs was upregulated when compared with normal glucose-exposed HUVECs. Furthermore, HNRNPC activated PSEN1-dependent Notch pathway to induce eNOS inactivation and NO production decrease, thereby causing vascular endothelial dysfunction in T2DM. CONCLUSIONS HNRNPC impaired vascular endothelial function to enhance the development of vascular complications in T2DM through PSEN1-mediated Notch signaling pathway.
Collapse
Affiliation(s)
- Ying Cai
- Department of Rehabilitation Medicine, Xiangya Hospital Central South University, Changsha 410008, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University, Changsha 410008, PR China
| | - Tao Chen
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, PR China
| | - Mingzhu Wang
- Department of Rehabilitation Medicine, Xiangya Hospital Central South University, Changsha 410008, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University, Changsha 410008, PR China
| | - Lihua Deng
- Department of Rehabilitation Medicine, Xiangya Hospital Central South University, Changsha 410008, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University, Changsha 410008, PR China
| | - Cui Li
- Department of Rehabilitation Medicine, Xiangya Hospital Central South University, Changsha 410008, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University, Changsha 410008, PR China
| | - Siqian Fu
- Department of Rehabilitation Medicine, Xiangya Hospital Central South University, Changsha 410008, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University, Changsha 410008, PR China
| | - Kangling Xie
- Department of Rehabilitation Medicine, Xiangya Hospital Central South University, Changsha 410008, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University, Changsha 410008, PR China.
| |
Collapse
|
36
|
Jiao Y, Wang S, Wang X, Yin L, Zhang YH, Li YZ, Yu YH. The m 6A reader YTHDC2 promotes SIRT3 expression by reducing the stabilization of KDM5B to improve mitochondrial metabolic reprogramming in diabetic peripheral neuropathy. Acta Diabetol 2023; 60:387-399. [PMID: 36574062 DOI: 10.1007/s00592-022-01990-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 10/03/2022] [Indexed: 12/28/2022]
Abstract
AIMS Diabetic peripheral neuropathy (DPN) is a common diabetic complication. Aberrant mitochondrial function causes neurodegeneration under hyperglycemia-induced metabolic stress, which in turn results in DPN progression. m6A and m6A reader (YTHDC2) are closely related to diabetes and diabetes complications, while the role of YTHDC2 in regulating mitochondrial metabolism in DPN needs to be further probed. METHODS For HG treatment, Schwann cells (RSC96) were subjected to D-glucose for 72 h. db/db mice were used as the diabetic mouse model. Me-RIP assay was performed to evaluate KDM5B m6A level. RNA degradation assay was conducted to examine KDM5B mRNA stability. In addition, OCR and ECAR were examined by XF96 Analyzer. Moreover, the content of ATP and PDH activity in RSC96 cells were detected using kits, and the level of ROS was detected using MitoSOX staining. RIP, RNA pull-down and dual-luciferase reporter gene assays were carried out to verify the binding relationships between YTHDC2, KDM5B and SIRT3. RESULTS We first observed that KDM5B expression and KDM5B mRNA stabilization were significantly increased in DPN. The m6A reader YTHDC2 was lowly expressed in DPN. Meanwhile, YTHDC2 over expression decreased KDM5B mRNA stability in an m6A-dependent manner. Our results also revealed that YTHDC2 overexpression resulted in reduced ROS level and increased ATP level, PDH activity, OCR and ECAR in HG-treated Schwann cells, while these effects were reversed by KDM5B overexpression. Additionally, SIRT3 served as the target of YTHDC2/KDM5B axis in regulating mitochondrial metabolism in DPN. CONCLUSIONS Taken together, YTHDC2 promoted SIRT3 expression by reducing the stabilization of KDM5B to improve mitochondrial metabolic reprogramming in DPN.
Collapse
Affiliation(s)
- Yang Jiao
- Department of Anesthesiology, Tianjin Medical University General Hospital, No. 154 Anshan Road, Tianjin, 300052, Tianjin, People's Republic of China
- Tianjin Research Institute of Anesthesiology, Tianjin, 300052, Tianjin, People's Republic of China
| | - Shu Wang
- Department of Extracorporeal Circulation, Tianjin Chest Hospital, Tianjin, 300222, Tianjin, People's Republic of China
| | - Xin Wang
- Department of Anesthesiology, Tianjin Medical University General Hospital, No. 154 Anshan Road, Tianjin, 300052, Tianjin, People's Republic of China
- Tianjin Research Institute of Anesthesiology, Tianjin, 300052, Tianjin, People's Republic of China
| | - Ling Yin
- Department of Anesthesiology, Tianjin Medical University General Hospital, No. 154 Anshan Road, Tianjin, 300052, Tianjin, People's Republic of China
- Tianjin Research Institute of Anesthesiology, Tianjin, 300052, Tianjin, People's Republic of China
| | - Yue-Hua Zhang
- Department of Anesthesiology, Tianjin Medical University General Hospital, No. 154 Anshan Road, Tianjin, 300052, Tianjin, People's Republic of China
- Tianjin Research Institute of Anesthesiology, Tianjin, 300052, Tianjin, People's Republic of China
| | - Yi-Ze Li
- Department of Anesthesiology, Tianjin Medical University General Hospital, No. 154 Anshan Road, Tianjin, 300052, Tianjin, People's Republic of China.
- Tianjin Research Institute of Anesthesiology, Tianjin, 300052, Tianjin, People's Republic of China.
| | - Yong-Hao Yu
- Department of Anesthesiology, Tianjin Medical University General Hospital, No. 154 Anshan Road, Tianjin, 300052, Tianjin, People's Republic of China.
- Tianjin Research Institute of Anesthesiology, Tianjin, 300052, Tianjin, People's Republic of China.
| |
Collapse
|
37
|
Fang J, Wu X, He J, Zhang H, Chen X, Zhang H, Novakovic B, Qi H, Yu X. RBM15 suppresses hepatic insulin sensitivity of offspring of gestational diabetes mellitus mice via m6A-mediated regulation of CLDN4. Mol Med 2023; 29:23. [PMID: 36803098 PMCID: PMC9942341 DOI: 10.1186/s10020-023-00615-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Accepted: 01/29/2023] [Indexed: 02/22/2023] Open
Abstract
BACKGROUND Gestational diabetes Mellitus (GDM) is a common pregnancy-specific disease with high morbidity, which is linked to a high risk of obesity and diabetes in offspring. N6-methyladenosine modification of RNA is emerging as an important epigenetic mechanism that is widely manifested in many diseases. This study aimed to investigate the mechanism of m6A methylation in metabolic syndrome in offspring result from intrauterine hyperglycemia. METHODS GDM mice were established by feeding a high-fat diet 1 weeks before pregnancy. The m6A RNA methylation quantification kit was used to detect liver tissue methylation levels. PCR array was used to determine the expression of the m6A methylation modification enzyme. Immunohistochemistry, qRT-PCR, and western blot were used to examine the expression of RBM15, METTL13, IGF2BP1, and IGF2BP2. Subsequently, methylated RNA immunoprecipitation sequencing combined with mRNA sequencing, followed by dot blot and glucose uptake tests, were performed. RESULTS In this study, we found that offspring from a GDM mother were more vulnerable to glucose intolerance and insulin resistance. GC-MS revealed significant metabolic changes including saturated fatty acids and unsaturated fatty acids in liver of GDM offspring. We also demonstrated that global mRNA m6A methylation level was significantly increased in the fetal liver of GDM mice, indicating epigenetic change may have a strong relationship with the mechanism of metabolism syndrome. Concordantly, RBM15, the RNA binding methyltransferase, was upregulated in the liver. In vitro, RBM15 suppressed insulin sensitivity and increased insulin resistance through m6A-regulated epigenetic inhabitation of CLDN4. Moreover, MeRIP-sequencing and mRNA-sequencing revealed that differently regulated genes with differential m6A peaks were enriched in metabolic pathways. CONCLUSION Our study revealed the essential role of RBM15 in insulin resistance and the effect of RBM15-regulated m6A modification in the metabolic syndrome of offspring of GDM mice.
Collapse
Affiliation(s)
- Jie Fang
- The Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, 1 Youyi Rd., Chongqing, 400016, China.,Chongqing Key Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Xiafei Wu
- The Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, 1 Youyi Rd., Chongqing, 400016, China.,Chongqing Key Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Jie He
- The Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, 1 Youyi Rd., Chongqing, 400016, China.,Chongqing Key Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Hanwen Zhang
- The Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, 1 Youyi Rd., Chongqing, 400016, China.,Chongqing Key Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Xuyang Chen
- The Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, 1 Youyi Rd., Chongqing, 400016, China.,Chongqing Key Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Hua Zhang
- The Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, 1 Youyi Rd., Chongqing, 400016, China.,Chongqing Key Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Boris Novakovic
- Department of Paediatrics, University of Melbourne, Royal Children's Hospital, Parkville, VIC, Australia
| | - Hongbo Qi
- The Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, 1 Youyi Rd., Chongqing, 400016, China. .,Chongqing Key Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing, 400016, China. .,Chongqing Health Center for Women and Children, Chongqing, 401120, China.
| | - Xinyang Yu
- The Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, 1 Youyi Rd., Chongqing, 400016, China. .,Chongqing Key Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
38
|
The Epigenetic Regulation of RNA N6-Methyladenosine Methylation in Glycolipid Metabolism. Biomolecules 2023; 13:biom13020273. [PMID: 36830642 PMCID: PMC9953413 DOI: 10.3390/biom13020273] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/06/2023] [Accepted: 01/30/2023] [Indexed: 02/04/2023] Open
Abstract
The highly conserved and dynamically reversible N6-methyladenine (m6A) modification has emerged as a critical gene expression regulator by affecting RNA splicing, translation efficiency, and stability at the post-transcriptional level, which has been established to be involved in various physiological and pathological processes, including glycolipid metabolism and the development of glycolipid metabolic disease (GLMD). Hence, accumulating studies have focused on the effects and regulatory mechanisms of m6A modification on glucose metabolism, lipid metabolism, and GLMD. This review summarizes the underlying mechanism of how m6A modification regulates glucose and lipid metabolism-related enzymes, transcription factors, and signaling pathways and the advances of m6A regulatory mechanisms in GLMD in order to deepen the understanding of the association of m6A modification with glycolipid metabolism and GLMD.
Collapse
|
39
|
Zhu X, Tang H, Yang M, Yin K. N6-methyladenosine in macrophage function: a novel target for metabolic diseases. Trends Endocrinol Metab 2023; 34:66-84. [PMID: 36586778 DOI: 10.1016/j.tem.2022.12.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/26/2022] [Accepted: 12/06/2022] [Indexed: 12/30/2022]
Abstract
N6-methyladenosine (m6A) is one of the most prevalent internal transcriptional modifications. Evidence has highlighted changes in m6A in metabolic disorders and various metabolic diseases. However, the precise mechanisms of these m6A changes in such conditions are not understood. Macrophages are crucial for the innate immune system and exert either beneficial or harmful roles in metabolic disease. Notably, m6A was found to be closely related to macrophage phenotype and dysfunction. In this review, we summarize m6A in macrophage function from the perspective of macrophage development, activation, and polarization, pyroptosis, and metabolic disorders. Furthermore, we discuss how m6A-mediated macrophage function affects metabolic diseases, including atherosclerosis and nonalcoholic fatty liver disease (NAFLD). Finally, we discuss challenges and prospects for m6A in macrophage and metabolic diseases with the aim of providing guidance for the treatment of metabolic diseases.
Collapse
Affiliation(s)
- Xiao Zhu
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin, Guangxi 541100, China; Department of Cardiology, The Second Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin, Guangxi 541199, China
| | - HaoJun Tang
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin, Guangxi 541100, China
| | - Min Yang
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin, Guangxi 541100, China
| | - Kai Yin
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin, Guangxi 541100, China; Department of Cardiology, The Second Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin, Guangxi 541199, China; Guangxi Health Commission Key Laboratory of Glucose and Lipid Metabolism Disorders, The Second Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541199, China.
| |
Collapse
|
40
|
Li X, Yang Y, Chen Z. Downregulation of the m 6A reader protein YTHDC1 leads to islet β-cell failure and diabetes. Metabolism 2023; 138:155339. [PMID: 36302453 DOI: 10.1016/j.metabol.2022.155339] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 10/17/2022] [Accepted: 10/19/2022] [Indexed: 11/06/2022]
Abstract
N6-methyladenosine (m6A) methyltransferase writer proteins (METTL3/METTL14) have been shown to regulate β-cell function and diabetes. However, whether and which m6A reader proteins regulate β-cell function and the pathogenesis of diabetes are largely unknown. In this study, we showed that YTHDC1 (YTH domain-containing protein 1), a key m6A nuclear reader protein, plays an essential role in maintaining β-cell function. YTHDC1 is downregulated in islet β cells in type 2 diabetes, which is due to lipotoxicity and chronic inflammation. β-Cell specific deletion of Ythdc1 results in β-cell failure and diabetes, which is likely due to the decreased expression of β-cell specific transcription factors and insulin secretion-related genes. Taken together, YTHDC1 is required for maintaining β-cell function, and the downregulation of YTHDC1 leads to β-cell failure and diabetes.
Collapse
Affiliation(s)
- Xinzhi Li
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| | - Ying Yang
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| | - Zheng Chen
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China.
| |
Collapse
|
41
|
Zhou B, Luo Y, Ji N, Mao F, Xiang L, Bian H, Zheng MH, Hu C, Li Y, Lu Y. Promotion of nonalcoholic steatohepatitis by RNA N 6-methyladenosine reader IGF2BP2 in mice. LIFE METABOLISM 2022; 1:161-174. [PMID: 39872354 PMCID: PMC11749640 DOI: 10.1093/lifemeta/loac006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 04/20/2022] [Accepted: 06/07/2022] [Indexed: 01/30/2025]
Abstract
Nonalcoholic steatohepatitis (NASH) has emerged as the major cause of end-stage liver diseases. However, an incomplete understanding of its molecular mechanisms severely dampens the development of pharmacotherapies. In the present study, through systematic screening of genome-wide mRNA expression from three mouse models of hepatic inflammation and fibrosis, we identified IGF2BP2, an N6-methyladenosine modification reader, as a key regulator that promotes NASH progression in mice. Adenovirus or adeno-associated virus-mediated overexpression of IGF2BP2 could induce liver steatosis, inflammation, and fibrosis in mice, at least in part, by increasing Tab2 mRNA stability. Besides, hepatic overexpression of IGF2BP2 mimicked gene expression profiles and molecular pathways of human NASH livers. Of potential clinical significance, IGF2BP2 expression is significantly upregulated in the livers of NASH patients. Moreover, knockdown of IGF2BP2 substantially alleviated liver injury, inflammation, and fibrosis in diet-induced NASH mice. Taken together, our findings reveal an important role of IGF2BP2 in NASH, which may provide a new therapeutic target for the treatment of NASH.
Collapse
Affiliation(s)
- Bing Zhou
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Centre for Diabetes, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Endocrinology and Metabolism of Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yunchen Luo
- Department of Endocrinology and Metabolism, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Nana Ji
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Endocrinology and Metabolism of Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Endocrinology and Metabolism, Qingpu Branch of Zhongshan Hospital, Fudan University, Wenzhou, China
| | - Fei Mao
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Endocrinology and Metabolism of Zhongshan Hospital, Fudan University, Shanghai, China
| | - Liping Xiang
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Centre for Diabetes, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Hua Bian
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Endocrinology and Metabolism of Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ming-Hua Zheng
- NAFLD Research Center, Department of Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Diagnosis and Treatment for The Development of Chronic Liver Disease in Zhejiang Province, Wenzhou, China
| | - Cheng Hu
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Centre for Diabetes, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
- Department of Endocrinology and Metabolism, Fengxian Central Hospital Affiliated to the Southern Medical University, Shanghai, China
| | - Yao Li
- Department of Laboratory Animal Science, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Lu
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Endocrinology and Metabolism of Zhongshan Hospital, Fudan University, Shanghai, China
- Institute of Metabolism and Regenerative Medicine, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| |
Collapse
|
42
|
Melnik BC, Schmitz G. Milk Exosomal microRNAs: Postnatal Promoters of β Cell Proliferation but Potential Inducers of β Cell De-Differentiation in Adult Life. Int J Mol Sci 2022; 23:ijms231911503. [PMID: 36232796 PMCID: PMC9569743 DOI: 10.3390/ijms231911503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/15/2022] [Accepted: 09/20/2022] [Indexed: 11/16/2022] Open
Abstract
Pancreatic β cell expansion and functional maturation during the birth-to-weaning period is driven by epigenetic programs primarily triggered by growth factors, hormones, and nutrients provided by human milk. As shown recently, exosomes derived from various origins interact with β cells. This review elucidates the potential role of milk-derived exosomes (MEX) and their microRNAs (miRs) on pancreatic β cell programming during the postnatal period of lactation as well as during continuous cow milk exposure of adult humans to bovine MEX. Mechanistic evidence suggests that MEX miRs stimulate mTORC1/c-MYC-dependent postnatal β cell proliferation and glycolysis, but attenuate β cell differentiation, mitochondrial function, and insulin synthesis and secretion. MEX miR content is negatively affected by maternal obesity, gestational diabetes, psychological stress, caesarean delivery, and is completely absent in infant formula. Weaning-related disappearance of MEX miRs may be the critical event switching β cells from proliferation to TGF-β/AMPK-mediated cell differentiation, whereas continued exposure of adult humans to bovine MEX miRs via intake of pasteurized cow milk may reverse β cell differentiation, promoting β cell de-differentiation. Whereas MEX miR signaling supports postnatal β cell proliferation (diabetes prevention), persistent bovine MEX exposure after the lactation period may de-differentiate β cells back to the postnatal phenotype (diabetes induction).
Collapse
Affiliation(s)
- Bodo C. Melnik
- Department of Dermatology, Environmental Medicine and Health Theory, University of Osnabrück, D-49076 Osnabrück, Germany
- Correspondence: ; Tel.: +49-52-4198-8060
| | - Gerd Schmitz
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital of Regensburg, University of Regensburg, D-93053 Regensburg, Germany
| |
Collapse
|
43
|
Zhou S, Sun Y, Xing Y, Wang Z, Wan S, Yao X, Hua Q, Meng X, Cheng J, Zhong M, Lv K, Kong X. Exenatide ameliorates hydrogen peroxide-induced pancreatic β-cell apoptosis through regulation of METTL3-mediated m 6A methylation. Eur J Pharmacol 2022; 924:174960. [PMID: 35436474 DOI: 10.1016/j.ejphar.2022.174960] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 04/09/2022] [Accepted: 04/12/2022] [Indexed: 01/05/2023]
Abstract
Exenatide, a glucagon-like peptide-1 (GLP-1) receptor agonist, is a commonly used hypoglycemic agent in clinical practice; it inhibits reactive oxygen species-induced pancreatic β-cell apoptosis. N6-methyladenosine (m6A) is produced by the methylation of RNA N6 residues and has recently been shown to play a crucial role in the regulation of islet β-cell growth and development. However, the involvement of m6A methylation in the β-cell protective effects of exenatide has not been clarified. In this study, the m6A-methylated RNA content and methyltransferase-like 3 (METTL3) expression levels in NIT-1 cells and primary mouse islets were found to significantly decrease following treatment with hydrogen peroxide (H2O2). Treatment with exenatide induced an increase in m6A content and METTL3 expression in the H2O2-treated NIT-1 cells and islets. Moreover, METTL3 silencing resulted in NIT-1 cell apoptosis under normal culture conditions. METTL3 upregulation significantly ameliorated H2O2-induced apoptosis in NIT-1 cells and primary islets. Furthermore, the anti-apoptotic effects of exenatide were obviously reversed by METTL3 knockdown. In conclusion, these findings suggest that exenatide elicits its anti-apoptotic effects in pancreatic β-cells by promoting m6A methylation through the upregulation METTL3 expression.
Collapse
Affiliation(s)
- Simin Zhou
- Department of Endocrinology, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital, Wuhu, 241001, China; Key Laboratory of Non-coding RNA Transformation Research of Anhui Higher Education Institution, Wannan Medical College, Wuhu, 241002, China
| | - Yue Sun
- Department of Endocrinology, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital, Wuhu, 241001, China; Key Laboratory of Non-coding RNA Transformation Research of Anhui Higher Education Institution, Wannan Medical College, Wuhu, 241002, China
| | - Yujie Xing
- Department of Endocrinology, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital, Wuhu, 241001, China; Key Laboratory of Non-coding RNA Transformation Research of Anhui Higher Education Institution, Wannan Medical College, Wuhu, 241002, China
| | - Zhi Wang
- Key Laboratory of Non-coding RNA Transformation Research of Anhui Higher Education Institution, Wannan Medical College, Wuhu, 241002, China; Department of Cardiology, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital, Wuhu, 241001, China
| | - Shujun Wan
- Key Laboratory of Non-coding RNA Transformation Research of Anhui Higher Education Institution, Wannan Medical College, Wuhu, 241002, China; Central Laboratory of Yijishan Hospital, Wuhu, 241001, China
| | - Xinming Yao
- Department of Endocrinology, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital, Wuhu, 241001, China
| | - Qiang Hua
- Department of Endocrinology, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital, Wuhu, 241001, China
| | - Xiangjian Meng
- Department of Endocrinology, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital, Wuhu, 241001, China
| | - Jinhan Cheng
- Department of Endocrinology, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital, Wuhu, 241001, China
| | - Min Zhong
- Key Laboratory of Non-coding RNA Transformation Research of Anhui Higher Education Institution, Wannan Medical College, Wuhu, 241002, China; Central Laboratory of Yijishan Hospital, Wuhu, 241001, China; Clinical Research Center for Critical Respiratory Medicine of Anhui Province, Wannan Medical College, Wuhu, 241002, China
| | - Kun Lv
- Key Laboratory of Non-coding RNA Transformation Research of Anhui Higher Education Institution, Wannan Medical College, Wuhu, 241002, China; Central Laboratory of Yijishan Hospital, Wuhu, 241001, China; Clinical Research Center for Critical Respiratory Medicine of Anhui Province, Wannan Medical College, Wuhu, 241002, China.
| | - Xiang Kong
- Department of Endocrinology, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital, Wuhu, 241001, China; Key Laboratory of Non-coding RNA Transformation Research of Anhui Higher Education Institution, Wannan Medical College, Wuhu, 241002, China; Central Laboratory of Yijishan Hospital, Wuhu, 241001, China; Clinical Research Center for Critical Respiratory Medicine of Anhui Province, Wannan Medical College, Wuhu, 241002, China.
| |
Collapse
|
44
|
Abderrahmani A, Jacovetti C, Regazzi R. Lessons from neonatal β-cell epigenomic for diabetes prevention and treatment. Trends Endocrinol Metab 2022; 33:378-389. [PMID: 35382967 DOI: 10.1016/j.tem.2022.03.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 03/08/2022] [Accepted: 03/10/2022] [Indexed: 12/30/2022]
Abstract
Pancreatic β-cell expansion and functional maturation during the birth-to-weaning period plays an essential role in the adaptation of plasma insulin levels to metabolic needs. These events are driven by epigenetic programs triggered by growth factors, hormones, and nutrients. These mechanisms operating in the neonatal period can be at least in part reactivated in adult life to increase the functional β-cell mass and face conditions of increased insulin demand such as obesity or pregnancy. In this review, we will highlight the importance of studying these signaling pathways and epigenetic programs to understand the causes of different forms of diabetes and to permit the design of novel therapeutic strategies to prevent and treat this metabolic disorder affecting hundreds of millions of people worldwide.
Collapse
Affiliation(s)
- Amar Abderrahmani
- Universitéde Lille, CNRS, Centrale Lille, Université Polytechnique Hauts-de-France, UMR 8520 - IEMN, F-59000 Lille, France.
| | - Cécile Jacovetti
- Department of Fundamental Neuroscience, University of Lausanne, 1005 Lausanne, Switzerland
| | - Romano Regazzi
- Department of Fundamental Neuroscience, University of Lausanne, 1005 Lausanne, Switzerland; Department of Biomedical Science, University of Lausanne, 1005 Lausanne, Switzerland.
| |
Collapse
|
45
|
Sun J, Cheng B, Su Y, Li M, Ma S, Zhang Y, Zhang A, Cai S, Bao Q, Wang S, Zhu P. The Potential Role of m6A RNA Methylation in the Aging Process and Aging-Associated Diseases. Front Genet 2022; 13:869950. [PMID: 35518355 PMCID: PMC9065606 DOI: 10.3389/fgene.2022.869950] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Accepted: 03/31/2022] [Indexed: 12/15/2022] Open
Abstract
N6-methyladenosine (m6A) is the most common and conserved internal eukaryotic mRNA modification. m6A modification is a dynamic and reversible post-transcriptional regulatory modification, initiated by methylase and removed by RNA demethylase. m6A-binding proteins recognise the m6A modification to regulate gene expression. Recent studies have shown that altered m6A levels and abnormal regulator expression are crucial in the ageing process and the occurrence of age-related diseases. In this review, we summarise some key findings in the field of m6A modification in the ageing process and age-related diseases, including cell senescence, autophagy, inflammation, oxidative stress, DNA damage, tumours, neurodegenerative diseases, diabetes, and cardiovascular diseases (CVDs). We focused on the biological function and potential molecular mechanisms of m6A RNA methylation in ageing and age-related disease progression. We believe that m6A modification may provide a new target for anti-ageing therapies.
Collapse
Affiliation(s)
- Jin Sun
- Department of Geriatrics, The Second Medical Center and National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China.,Medical School of Chinese PLA, Beijing, China
| | - Bokai Cheng
- Department of Geriatrics, The Second Medical Center and National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China.,Medical School of Chinese PLA, Beijing, China
| | - Yongkang Su
- Department of Geriatrics, The Second Medical Center and National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China.,Medical School of Chinese PLA, Beijing, China
| | - Man Li
- Department of Geriatrics, The Second Medical Center and National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China.,Medical School of Chinese PLA, Beijing, China
| | - Shouyuan Ma
- Department of Geriatric Cardiology, The Second Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yan Zhang
- Department of Outpatient, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Anhang Zhang
- Department of Geriatrics, The Second Medical Center and National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China.,Medical School of Chinese PLA, Beijing, China
| | - Shuang Cai
- Department of Geriatrics, The Second Medical Center and National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China.,Medical School of Chinese PLA, Beijing, China
| | - Qiligeer Bao
- Department of Geriatrics, The Second Medical Center and National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China.,Medical School of Chinese PLA, Beijing, China
| | - Shuxia Wang
- Department of Geriatrics, The Second Medical Center and National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China.,Medical School of Chinese PLA, Beijing, China
| | - Ping Zhu
- Department of Geriatrics, The Second Medical Center and National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China.,Medical School of Chinese PLA, Beijing, China
| |
Collapse
|
46
|
METTL3 enhances NSD2 mRNA stability to reduce renal impairment and interstitial fibrosis in mice with diabetic nephropathy. BMC Nephrol 2022; 23:124. [PMID: 35354439 PMCID: PMC8969340 DOI: 10.1186/s12882-022-02753-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 03/24/2022] [Indexed: 02/06/2023] Open
Abstract
Background Nuclear receptor-binding SET domain protein 2 (NSD2) is a histone methyltransferase that has been demonstrated to regulate insulin secretion and glucose concentration. This study focused on the role of NSD2 in the renal impairment during diabetic nephropathy (DN). Methods Serum NSD2 level in patients with DN was examined, and its correlations with the renal impairment-related indicators were examined. A murine model of DN was established, and mouse mesangial cells (SV40-MES-13) were treated with high-glucose (HG) to mimic a DN-like condition in vitro. Overexpression of NSD2 was introduced into mice or cells for in vivo and in vitro studies. The m6A level in HG-treated SV40-MES-13 cells was analyzed. METTL3 expression and its correlation with NSD2 were determined. Results NSD2 was poorly expressed in the serum of patients with DN and was negatively correlated with the levels of fasting blood sugar (FBG), serum creatinine (SCr), serum cystatin C (S-Cys-C), the 24-h urine protein (24-h U-protein) and the urine cystatin C (U-Cys-C). NSD2 overexpression reduced the kidney weight and reduced renal impairment in mice. It also suppressed interstitial fibrosis in mouse kidney tissues and reduced fibrosis-related markers in HG-treated SV40-MES-13 cells. HG treatment reduced the m6A level in the cells. METTL3 promoted m6A modification of NDS2 mRNA and enhanced its stability by YTHDF1. METTL3 overexpression alleviated renal impairment and fibrosis in vivo and in vitro. But the protective role was blocked upon NSD2 silencing. Conclusion This study demonstrates that METTL3 promotes NSD2 mRNA stability by YTHDF1 to alleviate progression of DN. Supplementary Information The online version contains supplementary material available at 10.1186/s12882-022-02753-3.
Collapse
|
47
|
Mechanisms Underlying the Expansion and Functional Maturation of β-Cells in Newborns: Impact of the Nutritional Environment. Int J Mol Sci 2022; 23:ijms23042096. [PMID: 35216239 PMCID: PMC8877060 DOI: 10.3390/ijms23042096] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 02/04/2022] [Accepted: 02/09/2022] [Indexed: 12/24/2022] Open
Abstract
The functional maturation of insulin-secreting β-cells is initiated before birth and is completed in early postnatal life. This process has a critical impact on the acquisition of an adequate functional β-cell mass and on the capacity to meet and adapt to insulin needs later in life. Many cellular pathways playing a role in postnatal β-cell development have already been identified. However, single-cell transcriptomic and proteomic analyses continue to reveal new players contributing to the acquisition of β-cell identity. In this review, we provide an updated picture of the mechanisms governing postnatal β-cell mass expansion and the transition of insulin-secreting cells from an immature to a mature state. We then highlight the contribution of the environment to β-cell maturation and discuss the adverse impact of an in utero and neonatal environment characterized by calorie and fat overload or by protein deficiency and undernutrition. Inappropriate nutrition early in life constitutes a risk factor for developing diabetes in adulthood and can affect the β-cells of the offspring over two generations. A better understanding of these events occurring in the neonatal period will help developing better strategies to produce functional β-cells and to design novel therapeutic approaches for the prevention and treatment of diabetes.
Collapse
|
48
|
Glucose Regulates m6A Methylation of RNA in Pancreatic Islets. Cells 2022; 11:cells11020291. [PMID: 35053407 PMCID: PMC8773766 DOI: 10.3390/cells11020291] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 01/10/2022] [Accepted: 01/12/2022] [Indexed: 02/04/2023] Open
Abstract
Type 2 diabetes is characterized by chronic hyperglycemia associated with impaired insulin action and secretion. Although the heritability of type 2 diabetes is high, the environment, including blood components, could play a major role in the development of the disease. Amongst environmental effects, epitranscriptomic modifications have been recently shown to affect gene expression and glucose homeostasis. The epitranscriptome is characterized by reversible chemical changes in RNA, with one of the most prevalent being the m6A methylation of RNA. Since pancreatic β cells fine tune glucose levels and play a major role in type 2 diabetes physiopathology, we hypothesized that the environment, through variations in blood glucose or blood free fatty acid concentrations, could induce changes in m6A methylation of RNAs in pancreatic β cells. Here we observe a significant decrease in m6A methylation upon high glucose concentration, both in mice and human islets, associated with altered expression levels of m6A demethylases. In addition, the use of siRNA and/or specific inhibitors against selected m6A enzymes demonstrate that these enzymes modulate the expression of genes involved in pancreatic β-cell identity and glucose-stimulated insulin secretion. Our data suggest that environmental variations, such as glucose, control m6A methylation in pancreatic β cells, playing a key role in the control of gene expression and pancreatic β-cell functions. Our results highlight novel causes and new mechanisms potentially involved in type 2 diabetes physiopathology and may contribute to a better understanding of the etiology of this disease.
Collapse
|
49
|
Cheng Y, Yao XM, Zhou SM, Sun Y, Meng XJ, Wang Y, Xing YJ, Wan SJ, Hua Q. The m 6A Methyltransferase METTL3 Ameliorates Methylglyoxal-Induced Impairment of Insulin Secretion in Pancreatic β Cells by Regulating MafA Expression. Front Endocrinol (Lausanne) 2022; 13:910868. [PMID: 35872977 PMCID: PMC9304699 DOI: 10.3389/fendo.2022.910868] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 06/06/2022] [Indexed: 01/13/2023] Open
Abstract
Methylglyoxal, a major precursor of advanced glycation end products, is elevated in the plasma of patients with type 2 diabetes mellitus. Islet β-cell function was recently shown to be regulated by N6-methyladenosine (m6A), an RNA modification consisting of methylation at the N6 position of adenosine. However, the role of m6A methylation modification in methylglyoxal-induced impairment of insulin secretion in pancreatic β cells has not been clarified. In this study, we showed that treatment of two β-cell lines, NIT-1 and β-TC-6, with methylglyoxal reduced m6A RNA content and methyltransferase-like 3 (METTL3) expression levels. We also showed that silencing of METTL3 inhibited glucose-stimulated insulin secretion (GSIS) from NIT-1 cells, whereas upregulation of METTL3 significantly reversed the methylglyoxal-induced decrease in GSIS. The methylglyoxal-induced decreases in m6A RNA levels and METTL3 expression were not altered by knockdown of the receptor for the advanced glycation end product but were further decreased by silencing of glyoxalase 1. Mechanistic investigations revealed that silencing of METTL3 reduced m6A levels, mRNA stability, and the mRNA and protein expression levels of musculoaponeurotic fibrosarcoma oncogene family A (MafA). Overexpression of MafA greatly improved the decrease in GSIS induced by METTL3 silencing; silencing of MafA blocked the reversal of the MG-induced decrease in GSIS caused by METTL3 overexpression. The current study demonstrated that METTL3 ameliorates MG-induced impairment of insulin secretion in pancreatic β cells by regulating MafA.
Collapse
Affiliation(s)
- Yi Cheng
- Department of Endocrinology, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital, Wuhu, China
| | - Xin-Ming Yao
- Department of Endocrinology, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital, Wuhu, China
| | - Si-Min Zhou
- Department of Endocrinology, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital, Wuhu, China
| | - Yue Sun
- Department of Endocrinology, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital, Wuhu, China
| | - Xiang-Jian Meng
- Department of Endocrinology, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital, Wuhu, China
| | - Yong Wang
- Department of Endocrinology, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital, Wuhu, China
| | - Yu-Jie Xing
- Department of Endocrinology, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital, Wuhu, China
- Key Laboratory of Non-coding RNA Transformation Research of Anhui Higher Education Institution, Wannan Medical College, Wuhu, China
- *Correspondence: Qiang Hua, ; Shu-Jun Wan, ; Yu-Jie Xing,
| | - Shu-Jun Wan
- Key Laboratory of Non-coding RNA Transformation Research of Anhui Higher Education Institution, Wannan Medical College, Wuhu, China
- Central Laboratory of Yijishan Hospital, The First Affiliated Hospital of Wannan Medical College, Wuhu, China
- Clinical Research Center for Critical Respiratory Medicine of Anhui Province, Wannan Medical College, Wuhu, China
- *Correspondence: Qiang Hua, ; Shu-Jun Wan, ; Yu-Jie Xing,
| | - Qiang Hua
- Department of Endocrinology, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital, Wuhu, China
- Clinical Research Center for Critical Respiratory Medicine of Anhui Province, Wannan Medical College, Wuhu, China
- *Correspondence: Qiang Hua, ; Shu-Jun Wan, ; Yu-Jie Xing,
| |
Collapse
|
50
|
Zhang W, Zhang S, Dong C, Guo S, Jia W, Jiang Y, Wang C, Zhou M, Gong Y. A bibliometric analysis of RNA methylation in diabetes mellitus and its complications from 2002 to 2022. Front Endocrinol (Lausanne) 2022; 13:997034. [PMID: 36157472 PMCID: PMC9492860 DOI: 10.3389/fendo.2022.997034] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 08/18/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND RNA methylation has emerged as an active research field in diabetes mellitus (DM) and its complications, while few bibliometric analyses have been performed. We aimed to visualize the hotspots and trends using bibliometric analysis to provide a comprehensive and objective overview of the current search state in this field. METHODS The articles and reviews regarding RNA methylation in DM and its complications were from the Web of Science Core Collection. A retrospective bibliometric analysis and science mapping was performed using the CiteSpace software to plot the knowledge maps and predict the hotspots and trends. RESULTS Three hundred seventy-five qualified records were retrieved. The annual publications gradually increased over the past 20 years. These publications mainly came from 66 countries led by Canada and 423 institutions. Leiter and Sievenpiper were the most productive authors, and Jenkins ranked first in the cited authors. Diabetes Care was the most co-cited journal. The most common keywords were "Type 2 diabetes", "cardiovascular disease", "diabetes mellitus", and "n 6 methyladenosine". The extracted keywords mainly clustered in "beta-cell function", "type 2 diabetes", "diabetic nephropathy", "aging", and "n6-methyladenosine". N6-methyladenosine (m6A) in DM and its complications were the developing areas of study. CONCLUSION Studies on RNA methylation, especially m6A modification, are the current hotspots and the future trends in type 2 diabetes (T2D) and diabetic nephropathy (DN), as well as a frontier field for other complications of DM. Strengthening future cooperation and exchange between countries and institutions is strongly advisable to promote research developments in this field.
Collapse
Affiliation(s)
- Wenhua Zhang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Shuwen Zhang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Chenlu Dong
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Shuaijie Guo
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
- Cardiovascular Disease Research Department, Beijing Institute of Chinese Medicine, Beijing, China
| | - Weiyu Jia
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yijia Jiang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Churan Wang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Mingxue Zhou
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
- Cardiovascular Disease Research Department, Beijing Institute of Chinese Medicine, Beijing, China
- *Correspondence: Mingxue Zhou, ; Yanbing Gong,
| | - Yanbing Gong
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- *Correspondence: Mingxue Zhou, ; Yanbing Gong,
| |
Collapse
|