1
|
Liao J, Wu X, Zeng Q, Huo Q, Nie G. STM2457 decreases m6A methylation to reduce cisplatin-induced ototoxicity via MAPK signaling. Biochem Pharmacol 2025; 235:116820. [PMID: 39983847 DOI: 10.1016/j.bcp.2025.116820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 02/04/2025] [Accepted: 02/18/2025] [Indexed: 02/23/2025]
Abstract
Cisplatin, a chemotherapeutic drug used to treat cancerous solid tumors, can result in ototoxicity due to serious toxic side effects resulting in irreversible hearing loss. Here, we investigated the effects of N6-methyladenosine (m6A) methylation on cisplatin-induced ototoxicity by using in vitro cochlear explants as a model system to explore the effect of the Methyltransferase-like 3 (METTL3) inhibitor STM2457 in ameliorating cisplatin-induced ototoxicity. STM2457 pretreatment was shown to significantly reduce reactive oxygen species (ROS) accumulation and the loss of hair cells (HCs) in different regions of the organ of Corti. STM2457 pretreatment led to significant reductions in TUNEL labeling, signifying a reduction in apoptosis. Additionally, expression of the apoptosis-related protein BAX was significantly decreased, while the ratio of BCL-XL was markedly increased. Transcriptomic measurements of the STM2457 + cisplatin group revealed significant enrichment of the Mitogen-Activated Protein Kinases (MAPK) signaling pathway, which when stimulated, could block the protective effect of STM2457 in cisplatin-treated HCs. Thus, we describe a mechanism by which STM2457 decreases cisplatin-related HC death in cochlear explants in vitro through activation of the MAPK pathway. This study reports for the first time that reducing RNA m6A methylation might protects against cisplatin-induced ototoxicity. Our data indicate that STM2457 can serve as an effect anti-apoptotic drug to decrease ototoxicity caused by cisplatin-induced ROS accumulation, effectively preventing cisplatin-induced hair cells loss.
Collapse
Affiliation(s)
- Jiahao Liao
- Department of Otolaryngology, Shenzhen Institute of Translational Medicine, Shenzhen Key Laboratory of Nanozymes and Translational Cancer Research, Medical Innovation Technology Transformation Center, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518035, China; School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong 518055, China
| | - Xingxing Wu
- Department of Otolaryngology, Shenzhen Institute of Translational Medicine, Shenzhen Key Laboratory of Nanozymes and Translational Cancer Research, Medical Innovation Technology Transformation Center, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518035, China; School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong 518055, China
| | - Qingdong Zeng
- Department of Otolaryngology, Shenzhen Institute of Translational Medicine, Shenzhen Key Laboratory of Nanozymes and Translational Cancer Research, Medical Innovation Technology Transformation Center, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518035, China
| | - Qin Huo
- Department of Otolaryngology, Shenzhen Institute of Translational Medicine, Shenzhen Key Laboratory of Nanozymes and Translational Cancer Research, Medical Innovation Technology Transformation Center, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518035, China; School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong 518055, China
| | - Guohui Nie
- Department of Otolaryngology, Shenzhen Institute of Translational Medicine, Shenzhen Key Laboratory of Nanozymes and Translational Cancer Research, Medical Innovation Technology Transformation Center, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518035, China.
| |
Collapse
|
2
|
Yan W, Saqirile, Li K, Li K, Wang C. The Role of N6-Methyladenosine in Mitochondrial Dysfunction and Pathology. Int J Mol Sci 2025; 26:3624. [PMID: 40332101 PMCID: PMC12026702 DOI: 10.3390/ijms26083624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 04/01/2025] [Accepted: 04/09/2025] [Indexed: 05/08/2025] Open
Abstract
Mitochondria are indispensable in cells and play crucial roles in maintaining cellular homeostasis, energy production, and regulating cell death. Mitochondrial dysfunction has various manifestations, causing different diseases by affecting the diverse functions of mitochondria in the body. Previous studies have mainly focused on mitochondrial-related diseases caused by nuclear gene mutations or mitochondrial gene mutations, or mitochondrial dysfunction resulting from epigenetic regulation, such as DNA and histone modification. In recent years, as a popular research area, m6A has been involved in a variety of important processes under physiological and pathological conditions. However, there are few summaries on how RNA methylation, especially m6A RNA methylation, affects mitochondrial function. Additionally, the role of m6A in pathology through influencing mitochondrial function may provide us with a new perspective on disease treatment. In this review, we summarize several manifestations of mitochondrial dysfunction and compile examples from recent years of how m6A affects mitochondrial function and its role in some diseases.
Collapse
Affiliation(s)
| | | | | | | | - Changshan Wang
- School of Life Science, Inner Mongolia University, Hohhot 010020, China; (W.Y.); (S.); (K.L.); (K.L.)
| |
Collapse
|
3
|
Xia Y, Wang Y, Chen K, Zhang M, Jiang Q, Xu T. Quercetin attenuated necroptosis and apoptosis caused by LPS-induced mitochondrial function dysfunction through the METTL3-mediated PTEN m 6A methylation/PI3K/AKT signaling in broiler livers. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 139:156551. [PMID: 40020631 DOI: 10.1016/j.phymed.2025.156551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 02/10/2025] [Accepted: 02/19/2025] [Indexed: 03/03/2025]
Abstract
BACKGROUND Quercetin (QUE), a natural flavonoid, offered an efficient protection against organism injury. N6-methyladenosine (m6A) methylation is considered to be the most prevalent and abundant modifications involved in various diseases. PURPOSE We sought to explore protective roles of QUE in mitigating necroptosis and apoptosis triggered by LPS-induced imbalances in mitochondria dynamic and energy metabolism in broiler livers, with a focus on m6A methylation modulation. STUDY DESIGN/METHODS We used LPS as a stimulus and treated with QUE to establish this in vivo and in vitro. In addition, we treated LMH cells with siMETTL3 (80 nM) to determine its detailed mechanism. RESULTS Our findings revealed QUE significantly decreased METTL3 expression, leading to a decrease in PTEN m6A methylation and factors related to mitochondria fission, necroptosis, and apoptosis in the QUE+LPS group. In contrast, QUE treatment promoted the expression levels of marker factors for mitochondria fusion, energy metabolism, anti-apoptosis, and PI3K/AKT compared with the LPS group. Additionally, an increase of ΔΨm, ATP content, and ATPase activity was observed. AO/EB staining, Flow cytometry and TUNEL assays confirmed QUE inhibited LPS-induced apoptosis and necroptosis. Molecular docking analysis and cellular thermal shift assay supported an interaction between QUE and METTL3. CONCLUSION In summary, QUE mitigated necroptosis and apoptosis triggered by LPS-induced disorders of mitochondrial kinetic and metabolic processes in broiler livers through its interaction with METTL3, regulating PTEN m6A methylation/PI3K/AKT signaling pathway. This study enhances our understanding of biological functions for QUE and lays a theoretical foundation for developing new therapeutic interventions, highlighting its potential value.
Collapse
Affiliation(s)
- Yu Xia
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China; Key Laboratory of Preventive Veterinary Medicine, Department of Veterinary Medicine, Animal Science College, Hebei North University, Zhangjiakou 075000, PR China
| | - Yidan Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Kai Chen
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Muyue Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Qihang Jiang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Tong Xu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China.
| |
Collapse
|
4
|
Yi R, Liu Y, Zhang X, Sun X, Wang N, Zhang C, Deng H, Yao X, Wang S, Yang G. Unraveling Quercetin's Potential: A Comprehensive Review of Its Properties and Mechanisms of Action, in Diabetes and Obesity Complications. Phytother Res 2024; 38:5641-5656. [PMID: 39307545 DOI: 10.1002/ptr.8332] [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: 05/05/2024] [Revised: 07/29/2024] [Accepted: 08/18/2024] [Indexed: 12/13/2024]
Abstract
The prevalence of diabetes is escalating alarmingly, placing a significant economic burden on the global healthcare system. The use of chemical substances extracted from plants has been demonstrated to be an effective method for the treatment and control of insulin resistance and Type 2 diabetes mellitus (T2DM). New research indicates that natural phytochemicals present in fruits and vegetables are expected to become drugs for the treatment of diabetes and the prevention of related complications. Quercetin, a widely distributed flavonoid, is well-known for its antioxidant, anti-inflammatory, anticancer, and antidiabetic properties. This article provides a comprehensive account of the mechanism of action of quercetin on diabetes and obesity complications in vivo and in vitro. It elucidates the impact of quercetin on various cells. These include hepatocytes, renal cells, skeletal muscle cells, and adipocytes. Furthermore, this article discusses the mechanism of quercetin on organ damage in diabetic mice induced by STZ, alloxan, diet, and spontaneous Type 2 diabetic mice caused by genetic defects. Additionally, it addresses the pharmacokinetics of quercetin and its potential for synergistic effects with existing diabetic drugs.
Collapse
Affiliation(s)
- Ruhan Yi
- Department of Food Nutrition and Safety, Dalian Medical University, Dalian, China
| | - Yun Liu
- Department of Food Nutrition and Safety, Dalian Medical University, Dalian, China
| | - Xu Zhang
- Department of Food Nutrition and Safety, Dalian Medical University, Dalian, China
| | - Xiance Sun
- Department of Occupational & Environmental Health, Dalian Medical University, Dalian, China
| | - Ningning Wang
- Department of Food Nutrition and Safety, Dalian Medical University, Dalian, China
| | - Cong Zhang
- Department of Food Nutrition and Safety, Dalian Medical University, Dalian, China
| | - Haoyuan Deng
- Department of Food Nutrition and Safety, Dalian Medical University, Dalian, China
| | - Xiaofeng Yao
- Department of Occupational & Environmental Health, Dalian Medical University, Dalian, China
| | - Shaopeng Wang
- Department of Cardiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Guang Yang
- Department of Food Nutrition and Safety, Dalian Medical University, Dalian, China
| |
Collapse
|
5
|
Shen J, Zhang Q, Lan Y, Peng Q, Ji Z, Wu Y, Liu H. Identification and Characterisation of Potential Targets for N6-methyladenosine (m6A) Modification during Intervertebral Disc Degeneration. FRONT BIOSCI-LANDMRK 2024; 29:405. [PMID: 39735982 DOI: 10.31083/j.fbl2912405] [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: 06/27/2024] [Revised: 08/26/2024] [Accepted: 09/10/2024] [Indexed: 12/31/2024]
Abstract
BACKGROUND The mechanism for RNA methylation during disc degeneration is unclear. The aim of this study was to identify N6-methyladenosine (m6A) markers and therapeutic targets for the prevention and treatment of intervertebral disc degeneration (IDD). METHODS Methylated RNA immunoprecipitation sequencing (MeRIP-seq) and quantitative reverse transcription PCR (RT-qPCR) were employed to analyze m6A modifications of IDD-related gene expression. Bioinformatics was used to identify enriched gene pathways in IDD. m6A-RIP-qPCR was used to validate potential targets and markers. RESULTS AND CONCLUSION Human IDD samples exhibited a distinct m6A modification pattern that allowed associated genes and pathways to be identified. These genes had functions such as "nuclear factor kappa-B (NF-κB) binding" and "extracellular matrix components", which are crucial for IDD pathogenesis. ANXA2 showed increased m6A modification in IDD, while SLC3A2 and PBX3 showed decreased m6A methylation. The results of this study offer novel insights for the prevention and treatment of IDD.
Collapse
Affiliation(s)
- Jianlin Shen
- Department of Orthopaedics, Affiliated Hospital of Putian University, 351100 Putian, Fujian, China
- Central Laboratory, Affiliated Hospital of Putian University, 351100 Putian, Fujian, China
| | - Qiang Zhang
- Central Laboratory, Affiliated Hospital of Putian University, 351100 Putian, Fujian, China
| | - Yujian Lan
- School of Integrated Traditional Chinese and Western Medicine, Southwest Medical University, 646000 Luzhou, Sichuan, China
| | - Qingping Peng
- School of Integrated Traditional Chinese and Western Medicine, Southwest Medical University, 646000 Luzhou, Sichuan, China
| | - Ziyu Ji
- School of Integrated Traditional Chinese and Western Medicine, Southwest Medical University, 646000 Luzhou, Sichuan, China
| | - Yanjiao Wu
- Department of Orthopaedics, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), 528308 Foshan, Guangdong, China
| | - Huan Liu
- Department of Orthopaedics, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, 646000 Luzhou, Sichuan, China
| |
Collapse
|
6
|
Dai W, Qiao X, Fang Y, Guo R, Bai P, Liu S, Li T, Jiang Y, Wei S, Na Z, Xiao X, Li D. Epigenetics-targeted drugs: current paradigms and future challenges. Signal Transduct Target Ther 2024; 9:332. [PMID: 39592582 PMCID: PMC11627502 DOI: 10.1038/s41392-024-02039-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 10/14/2024] [Accepted: 10/29/2024] [Indexed: 11/28/2024] Open
Abstract
Epigenetics governs a chromatin state regulatory system through five key mechanisms: DNA modification, histone modification, RNA modification, chromatin remodeling, and non-coding RNA regulation. These mechanisms and their associated enzymes convey genetic information independently of DNA base sequences, playing essential roles in organismal development and homeostasis. Conversely, disruptions in epigenetic landscapes critically influence the pathogenesis of various human diseases. This understanding has laid a robust theoretical groundwork for developing drugs that target epigenetics-modifying enzymes in pathological conditions. Over the past two decades, a growing array of small molecule drugs targeting epigenetic enzymes such as DNA methyltransferase, histone deacetylase, isocitrate dehydrogenase, and enhancer of zeste homolog 2, have been thoroughly investigated and implemented as therapeutic options, particularly in oncology. Additionally, numerous epigenetics-targeted drugs are undergoing clinical trials, offering promising prospects for clinical benefits. This review delineates the roles of epigenetics in physiological and pathological contexts and underscores pioneering studies on the discovery and clinical implementation of epigenetics-targeted drugs. These include inhibitors, agonists, degraders, and multitarget agents, aiming to identify practical challenges and promising avenues for future research. Ultimately, this review aims to deepen the understanding of epigenetics-oriented therapeutic strategies and their further application in clinical settings.
Collapse
Affiliation(s)
- Wanlin Dai
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xinbo Qiao
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yuanyuan Fang
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Renhao Guo
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Peng Bai
- Department of Forensic Genetics, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Shuang Liu
- Shenyang Maternity and Child Health Hospital, Shenyang, China
| | - Tingting Li
- Department of General Internal Medicine VIP Ward, Liaoning Cancer Hospital & Institute, Shenyang, China
| | - Yutao Jiang
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Shuang Wei
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Zhijing Na
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China.
- NHC Key Laboratory of Advanced Reproductive Medicine and Fertility (China Medical University), National Health Commission, Shenyang, China.
| | - Xue Xiao
- Department of Gynecology and Obstetrics, West China Second Hospital, Sichuan University, Chengdu, China.
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, China.
| | - Da Li
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China.
- NHC Key Laboratory of Advanced Reproductive Medicine and Fertility (China Medical University), National Health Commission, Shenyang, China.
- Key Laboratory of Reproductive Dysfunction Diseases and Fertility Remodeling of Liaoning Province, Shenyang, China.
| |
Collapse
|
7
|
Rada P, Carceller-López E, Hitos AB, Gómez-Santos B, Fernández-Hernández C, Rey E, Pose-Utrilla J, García-Monzón C, González-Rodríguez Á, Sabio G, García A, Aspichueta P, Iglesias T, Valverde ÁM. Protein kinase D2 modulates hepatic insulin sensitivity in male mice. Mol Metab 2024; 90:102045. [PMID: 39401614 PMCID: PMC11535753 DOI: 10.1016/j.molmet.2024.102045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/30/2024] [Accepted: 10/07/2024] [Indexed: 10/21/2024] Open
Abstract
OBJECTIVES Protein kinase D (PKD) family is emerging as relevant regulator of metabolic homeostasis. However, the precise role of PKD2 in modulating hepatic insulin signaling has not been fully elucidated and it is the aim of this study. METHODS PKD inhibition was analyzed for insulin signaling in mouse and human hepatocytes. PKD2 was overexpressed in Huh7 hepatocytes and mouse liver, and insulin responses were evaluated. Mice with hepatocyte-specific PKD2 depletion (PKD2ΔHep) and PKD2fl/fl mice were fed a chow (CHD) or high fat diet (HFD) and glucose homeostasis and lipid metabolism were investigated. RESULTS PKD2 silencing enhanced insulin signaling in hepatocytes, an effect also found in primary hepatocytes from PKD2ΔHep mice. Conversely, a constitutively active PKD2 mutant reduced insulin-stimulated AKT phosphorylation. A more in-depth analysis revealed reduced IRS1 serine phosphorylation under basal conditions and increased IRS1 tyrosine phosphorylation in PKD2ΔHep primary hepatocytes upon insulin stimulation and, importantly PKD co-immunoprecipitates with IRS1. In vivo constitutively active PKD2 overexpression resulted in a moderate impairment of glucose homeostasis and reduced insulin signaling in the liver. On the contrary, HFD-fed PKD2ΔHep male mice displayed improved glucose and pyruvate tolerance, as well as higher peripheral insulin tolerance and enhanced hepatic insulin signaling compared to control PKD2fl/fl mice. Despite of a remodeling of hepatic lipid metabolism in HFD-fed PKD2ΔHep mice, similar steatosis grade was found in both genotypes. CONCLUSIONS Results herein have unveiled an unknown role of PKD2 in the control of insulin signaling in the liver at the level of IRS1 and point PKD2 as a therapeutic target for hepatic insulin resistance.
Collapse
Affiliation(s)
- Patricia Rada
- Instituto de Investigaciones Biomédicas Sols-Morreale (IIBM), Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain.
| | - Elena Carceller-López
- Instituto de Investigaciones Biomédicas Sols-Morreale (IIBM), Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| | - Ana B Hitos
- Instituto de Investigaciones Biomédicas Sols-Morreale (IIBM), Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| | - Beatriz Gómez-Santos
- Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, Leioa, Spain; BioBizkaia Health Research Institute, Barakaldo, Spain
| | - Constanza Fernández-Hernández
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - Esther Rey
- Liver Research Unit, Santa Cristina University Hospital, Instituto de Investigación Sanitaria Princesa, Madrid, Spain
| | - Julia Pose-Utrilla
- Instituto de Investigaciones Biomédicas Sols-Morreale (IIBM), Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Carmelo García-Monzón
- Liver Research Unit, Santa Cristina University Hospital, Instituto de Investigación Sanitaria Princesa, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
| | - Águeda González-Rodríguez
- Instituto de Investigaciones Biomédicas Sols-Morreale (IIBM), Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain; Liver Research Unit, Santa Cristina University Hospital, Instituto de Investigación Sanitaria Princesa, Madrid, Spain
| | - Guadalupe Sabio
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain; Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Antonia García
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - Patricia Aspichueta
- Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, Leioa, Spain; BioBizkaia Health Research Institute, Barakaldo, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
| | - Teresa Iglesias
- Instituto de Investigaciones Biomédicas Sols-Morreale (IIBM), Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Ángela M Valverde
- Instituto de Investigaciones Biomédicas Sols-Morreale (IIBM), Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain.
| |
Collapse
|
8
|
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
|
9
|
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
|
10
|
Mao-Mao, Zhang JJ, Xu YP, Shao MM, Wang MC. Regulatory effects of natural products on N6-methyladenosine modification: A novel therapeutic strategy for cancer. Drug Discov Today 2024; 29:103875. [PMID: 38176674 DOI: 10.1016/j.drudis.2023.103875] [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: 10/12/2023] [Revised: 12/17/2023] [Accepted: 12/28/2023] [Indexed: 01/06/2024]
Abstract
N6-methyladenosine (m6A) is considered to be the most common and abundant epigenetics modification in messenger RNA (mRNA) and noncoding RNA. Abnormal modification of m6A is closely related to the occurrence, development, progression, and prognosis of cancer. m6A regulators have been identified as novel targets for anticancer drugs. Natural products, a rich source of traditional anticancer drugs, have been utilized for the development of m6A-targeting drugs. Here, we review the key role of m6A modification in cancer progression and explore the prospects and structural modification mechanisms of natural products as potential drugs targeting m6A modification for cancer treatment.
Collapse
Affiliation(s)
- Mao-Mao
- Affiliated Cixi Hospital, Wenzhou Medical University, Cixi, China
| | - Jin-Jing Zhang
- Affiliated Cixi Hospital, Wenzhou Medical University, Cixi, China
| | - Yue-Ping Xu
- Affiliated Cixi Hospital, Wenzhou Medical University, Cixi, China
| | - Min-Min Shao
- Affiliated Cixi Hospital, Wenzhou Medical University, Cixi, China
| | - Meng-Chuan Wang
- Affiliated Cixi Hospital, Wenzhou Medical University, Cixi, China.
| |
Collapse
|
11
|
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
|
12
|
Farhadi F, Sharififar F, Jafari M, Rahimi VB, Askari N, Askari VR. Hallmarks of Quercetin Benefits as a Functional Supplementary in the Management of Diabetes Mellitus-Related Maladies: From Basic to Clinical Applications. Curr Drug Metab 2024; 25:653-669. [PMID: 39878112 DOI: 10.2174/0113892002339410250108031621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 11/27/2024] [Accepted: 12/05/2024] [Indexed: 01/31/2025]
Abstract
Quercetin (QE), a particular flavonoid, is well known for its medicinal effects, including anti-oxidant, hypoglycemic, and anti-inflammatory effects. In this review, the findings of QE effects on diabetes STZinduced, alloxan-induced, and its complications have been summarized with a particular focus on in vitro, in vivo, and clinical trials. Consequently, QE mediates several mechanisms, including ameliorating tumor necrosis factor (TNF)-α, nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), interleukin (IL)-1β, IL-8, and IL-10 expression, increasing insulin glucose uptake to inhibit insulin resistance. Moreover, QE stimulates insulin secretion and attenuates insulin resistance through various pathways, namely transient KATP channel, motivating peroxisome proliferator-activated receptor expression, increasing glucose transporter-4, and decreasing inducible nitric oxide synthase in skeletal muscle. QE has protective effects on the complications caused by diabetes, such as polycystic ovary syndrome, high-fat diet-induced obesity, diabetic-induced hepatic damage, vascular inflammation, nephropathy, and neuropathy.
Collapse
Affiliation(s)
- Faegheh Farhadi
- Herbal and Traditional Medicines Research Center, Department of Pharmacognosy, School of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran
| | - Fariba Sharififar
- Herbal and Traditional Medicines Research Center, Department of Pharmacognosy, School of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran
| | - Mandana Jafari
- Herbal and Traditional Medicines Research Center, Department of Pharmacognosy, School of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran
| | - Vafa Baradaran Rahimi
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Cardiovascular Diseases, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Nafiseh Askari
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Vahid Reza Askari
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
13
|
Zhu J, Cheng X, Naumovski N, Hu L, Wang K. Epigenetic regulation by quercetin: a comprehensive review focused on its biological mechanisms. Crit Rev Food Sci Nutr 2023; 65:627-646. [PMID: 38062765 DOI: 10.1080/10408398.2023.2278760] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2025]
Abstract
Epigenetics regulates gene expression and play significant roles across diverse disease states. Epigenetics mechanisms, including DNA methylation, histone modifications, microRNAs/lncRNA, and N6-methyladenosine (m6A) RNA methylation, elicit heritable but reversible modifications in gene expression without modifying the DNA sequence. Recent research suggests that certain natural phytochemicals with chemopreventive properties have the potential to function as epigenetic regulators. Quercetin, a derivative of natural flavonoid glycosides and a constituent of the human diet, is linked to a variety of health benefits including anti-inflammatory, anticancer activity, antiapoptotic, antihypertensive, and neuroprotective effects. Recent findings suggest that quercetin possesses the ability to modulate canonical biochemical signaling pathways and exert an impact on epigenetic networks. This review aims to synthesize the most recent research findings that elucidate the potential biological effects of quercetin and its influence on in vitro and in vivo models via epigenetic mechanisms. In light of our findings, it is evident that quercetin possesses the potential to function as an exemplary instance of naturally derived phytochemicals, which can be effectively employed as a pivotal constituent in functional foods and dietary supplements aimed at the amelioration of various ailments. More specifically, its mechanism of action involves the alteration of diverse epigenetic targets.
Collapse
Affiliation(s)
- Jinfeng Zhu
- School for Radiological and interdisciplinary Sciences (RAD-X) and Collaborative Innovation Centre of Radiation Medicine of Jiangsu Higher Education Institutions. Soochow University, Suzhou, China
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, Roma, Italy
| | - Xiaju Cheng
- School for Radiological and interdisciplinary Sciences (RAD-X) and Collaborative Innovation Centre of Radiation Medicine of Jiangsu Higher Education Institutions. Soochow University, Suzhou, China
| | - Nenad Naumovski
- Discipline of Nutrition and Dietetics, Faculty of Health, University of Canberra, Bruce, Canberra, ACT, Australia
- Functional Foods and Nutrition Research (FFNR) Laboratory, University of Canberra, Ngunnawal Country, Canberra, ACT, Australia
- University of Canberra Research Institute for Sport and Exercise (UCRISE), University of Canberra, Canberra, ACT, Australia
- Department of Nutrition-Dietetics, Harokopio University, Athens, Greece
| | - Lin Hu
- School for Radiological and interdisciplinary Sciences (RAD-X) and Collaborative Innovation Centre of Radiation Medicine of Jiangsu Higher Education Institutions. Soochow University, Suzhou, China
| | - Kai Wang
- State Key Laboratory of Resource Insects, Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing, China
| |
Collapse
|
14
|
Ren Y, Li Z, Li J, Liang R, Wang Z, Bai Y, Yang Y, Tang Q, Fu Y, Zhang X, Zhang Y, Yu Y, Xiong Y. m 6 A mRNA methylation: Biological features, mechanisms, and therapeutic potentials in type 2 diabetes mellitus. Obes Rev 2023; 24:e13639. [PMID: 37732463 DOI: 10.1111/obr.13639] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 08/10/2023] [Accepted: 08/27/2023] [Indexed: 09/22/2023]
Abstract
As the most common internal post-transcriptional RNA modification in eukaryotic cells, N6-methyladenosine (m6 A) performs a dynamic and reversible role in a variety of biological processes mediated by methyltransferases (writers), demethylases (erasers), and m6 A binding proteins (readers). M6 A methylation enables transcriptome conversion in different signals that regulate various physiological activities and organ development. Over the past few years, emerging studies have identified that mRNA m6 A regulators defect in β-cell leads to abnormal regulation of the target mRNAs, thereby resulting in β-cell dysfunction and loss of β-cell identity and mass, which are strongly associated with type 2 diabetes mellitus (T2DM) pathogenesis. Also, mRNA m6 A modification has been implicated with insulin resistance in muscles, fat, and liver cells/tissues. In this review, we elaborate on the biological features of m6 A methylation; provide a comprehensive overview of the underlying mechanisms that how it controls β-cell function, identity, and mass as well as insulin resistance; highlight its connections to glucose metabolism and lipid metabolism linking to T2DM; and further discuss its role in diabetes complications and its therapeutic potentials for T2DM diagnosis and treatment.
Collapse
Affiliation(s)
- Yuanyuan Ren
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, College of Life Sciences, Northwest University, Xi'an, Shaanxi, China
| | - Zi Li
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, College of Life Sciences, Northwest University, Xi'an, Shaanxi, China
| | - Jiaoyu Li
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, College of Life Sciences, Northwest University, Xi'an, Shaanxi, China
| | - Rui Liang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, College of Life Sciences, Northwest University, Xi'an, Shaanxi, China
| | - Zhen Wang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, College of Life Sciences, Northwest University, Xi'an, Shaanxi, China
| | - Yiduo Bai
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, College of Life Sciences, Northwest University, Xi'an, Shaanxi, China
| | - Yafang Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, College of Life Sciences, Northwest University, Xi'an, Shaanxi, China
| | - Qian Tang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, College of Life Sciences, Northwest University, Xi'an, Shaanxi, China
| | - Yaolei Fu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, College of Life Sciences, Northwest University, Xi'an, Shaanxi, China
| | - Xiaobo Zhang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, College of Life Sciences, Northwest University, Xi'an, Shaanxi, China
| | - Yu Zhang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, College of Life Sciences, Northwest University, Xi'an, Shaanxi, China
| | - Yi Yu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, College of Life Sciences, Northwest University, Xi'an, Shaanxi, China
- School of Medicine, Northwest University, Xi'an, Shaanxi, China
| | - Yuyan Xiong
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, College of Life Sciences, Northwest University, Xi'an, Shaanxi, China
- School of Medicine, Northwest University, Xi'an, Shaanxi, China
| |
Collapse
|
15
|
Wu Y, Zeng Y, Ren Y, Yu J, Zhang Q, Xiao X. Insights into RNA N6-methyladenosine in Glucose and Lipid Metabolic Diseases and Their Therapeutic Strategies. Endocrinology 2023; 165:bqad170. [PMID: 37950364 DOI: 10.1210/endocr/bqad170] [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: 09/24/2023] [Revised: 10/20/2023] [Accepted: 11/07/2023] [Indexed: 11/12/2023]
Abstract
The incidence of glucose and lipid metabolism diseases, including type 2 diabetes, obesity, metabolic syndrome, and nonalcoholic fatty liver disease, is rising, which places an enormous burden on people around the world. However, the mechanism behind these disorders remains incompletely understood. N6-methyladenosine (m6A) is 1 type of posttranscriptional RNA modification, and research has shown that it plays a crucial role in several metabolic diseases. m6A methylation is reversibly and dynamically regulated by methyltransferases (writers), demethylases (erasers), and m6A binding proteins (readers). Dysregulation of RNA m6A modification is related to different metabolic processes. Targeting RNA m6A methylation is a potential treatment strategy for these chronic metabolic diseases. This review discusses studies on RNA m6A modification in metabolic diseases and existing therapeutic drugs, with the aim of providing a concise perspective on its potential applications in managing metabolic disorders.
Collapse
Affiliation(s)
- Yifan Wu
- Key Laboratory of Endocrinology, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Yuan Zeng
- Key Laboratory of Endocrinology, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Yaolin Ren
- Key Laboratory of Endocrinology, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Jie Yu
- Key Laboratory of Endocrinology, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Qian Zhang
- Key Laboratory of Endocrinology, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Xinhua Xiao
- Key Laboratory of Endocrinology, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China
| |
Collapse
|
16
|
El Midaoui A, Khallouki F, Couture R, Moldovan F, Ismael MA, Ongali B, Akoume MY, Alem C, Ait Boughrous A, Zennouhi W, Roqai MC, Hajji L, Ghzaiel I, Vejux A, Lizard G. Thymus atlanticus: A Source of Nutrients with Numerous Health Benefits and Important Therapeutic Potential for Age-Related Diseases. Nutrients 2023; 15:4077. [PMID: 37764861 PMCID: PMC10534698 DOI: 10.3390/nu15184077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 09/19/2023] [Accepted: 09/20/2023] [Indexed: 09/29/2023] Open
Abstract
Thymus atlanticus (Lamiaceae) is a plant endemic to the Mediterranean basin that is found in significant quantities in the arid regions of Morocco. Thymus atlanticus is used in traditional medicine to treat infectious and non-infectious diseases. It is also used for the isolation of essential oils and for the seasoning of many dishes in the Mediterranean diet. The major constituents of Thymus atlanticus are saponins, flavonoids, tannins, alkaloids, various simple and hydroxycinnamic phenolic compounds, and terpene compounds. Several of these compounds act on signaling pathways of oxidative stress, inflammation, and blood sugar, which are parameters often dysregulated during aging. Due to its physiochemical characteristics and biological activities, Thymus atlanticus could be used for the prevention and/or treatment of age-related diseases. These different aspects are treated in the present review, and we focused on phytochemistry and major age-related diseases: dyslipidemia, cardiovascular diseases, and type 2 diabetes.
Collapse
Affiliation(s)
- Adil El Midaoui
- Department of Pharmacology and Physiology, Faculty of Medicine, University of Montreal, Montreal, QC H3C 3J7, Canada; (R.C.); (B.O.)
- Department of Biology, Faculty of Sciences and Techniques, Errachidia, Moulay Ismail University of Meknes, Meknes 50050, Morocco; (F.K.); (A.A.B.); (W.Z.)
| | - Farid Khallouki
- Department of Biology, Faculty of Sciences and Techniques, Errachidia, Moulay Ismail University of Meknes, Meknes 50050, Morocco; (F.K.); (A.A.B.); (W.Z.)
| | - Réjean Couture
- Department of Pharmacology and Physiology, Faculty of Medicine, University of Montreal, Montreal, QC H3C 3J7, Canada; (R.C.); (B.O.)
| | - Florina Moldovan
- Research Center of CHU Sainte Justine, Faculty of Dentistry, Université de Montréal, Montreal, QC H3T 1J4, Canada; (F.M.); (M.Y.A.)
| | | | - Brice Ongali
- Department of Pharmacology and Physiology, Faculty of Medicine, University of Montreal, Montreal, QC H3C 3J7, Canada; (R.C.); (B.O.)
| | - Marie Yvonne Akoume
- Research Center of CHU Sainte Justine, Faculty of Dentistry, Université de Montréal, Montreal, QC H3T 1J4, Canada; (F.M.); (M.Y.A.)
| | - Chakib Alem
- Research Team in Biochemistry and Natural Resources, Faculty of Sciences and Techniques, Moulay Ismail University of Meknes, Meknes 20250, Morocco;
| | - Ali Ait Boughrous
- Department of Biology, Faculty of Sciences and Techniques, Errachidia, Moulay Ismail University of Meknes, Meknes 50050, Morocco; (F.K.); (A.A.B.); (W.Z.)
| | - Wafa Zennouhi
- Department of Biology, Faculty of Sciences and Techniques, Errachidia, Moulay Ismail University of Meknes, Meknes 50050, Morocco; (F.K.); (A.A.B.); (W.Z.)
| | - Mhammed Chaoui Roqai
- Ecole des Hautes Etudes de Biotechnologie et de Santé (EHEB), 183 Bd de la Résistance, Casablanca 20250, Morocco;
| | - Lhoussain Hajji
- Laboratory of Bioactives and Environmental Health, Faculty of Sciences, Moulay Ismail University, Meknes 50050, Morocco;
| | - Imen Ghzaiel
- Laboratory “Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism”, Bio-peroxIL/EA7270, Université de Bourgogne/Inserm, 21000 Dijon, France; (I.G.); (A.V.)
| | - Anne Vejux
- Laboratory “Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism”, Bio-peroxIL/EA7270, Université de Bourgogne/Inserm, 21000 Dijon, France; (I.G.); (A.V.)
| | - Gérard Lizard
- Laboratory “Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism”, Bio-peroxIL/EA7270, Université de Bourgogne/Inserm, 21000 Dijon, France; (I.G.); (A.V.)
| |
Collapse
|
17
|
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
|
18
|
Bellavite P, Fazio S, Affuso F. A Descriptive Review of the Action Mechanisms of Berberine, Quercetin and Silymarin on Insulin Resistance/Hyperinsulinemia and Cardiovascular Prevention. Molecules 2023; 28:4491. [PMID: 37298967 PMCID: PMC10254920 DOI: 10.3390/molecules28114491] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 05/26/2023] [Accepted: 05/30/2023] [Indexed: 06/12/2023] Open
Abstract
Insulin resistance (IR) and the associated hyperinsulinemia are early pathophysiological changes which, if not well treated, can lead to type 2 diabetes, endothelial dysfunction and cardiovascular disease. While diabetes care is fairly well standardized, the prevention and treatment of IR lacks a single pharmaceutical approach and many lifestyle and dietary interventions have been proposed, including a wide range of food supplements. Among the most interesting and well-known natural remedies, alkaloid berberine and the flavonol quercetin have particular relevance in the literature, while silymarin-the active principle of the Silybum marianum thistle-was traditionally used for lipid metabolism disorders and to sustain liver function. This review describes the major defects of insulin signaling leading to IR and the main properties of the three mentioned natural substances, their molecular targets and synergistic action mechanisms. The actions of berberine, quercetin and silymarin are partially superimposable as remedies against reactive oxygen intermediates generated by a high-lipid diet and by NADPH oxidase, which is triggered by phagocyte activation. Furthermore, these compounds inhibit the secretion of a battery of pro-inflammatory cytokines, modulate intestinal microbiota and are especially able to control the various disorders of the insulin receptor and post-receptor signaling systems. Although most of the evidence on the effects of berberine, quercetin and silymarin in modulating insulin resistance and preventing cardiovascular disease derive from experimental studies on animals, the amount of pre-clinical knowledge strongly suggests the need to investigate the therapeutic potential of these substances in human pathology.
Collapse
Affiliation(s)
- Paolo Bellavite
- Pathophysiology Chair, Homeopathic Medical School of Verona, 37121 Verona, Italy
| | - Serafino Fazio
- Department of Internal Medicine, University of Naples Federico II, 80138 Naples, Italy;
| | | |
Collapse
|
19
|
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
|