Epigenetic regulation in cognitive impairment: Focus on N6-methyladenosine modification and its potential role in perioperative neurocognitive disorders

Ting Liu , Xiao-Juan Yang , Lin Zhou , Mi Gan , Ting-Ting He , Sen Hong , Yan-Yan Feng , Gao Su , You-Xiao Zhao , Ying Cao , Qing-Fan Zeng

Ibrain ›› 2025, Vol. 11 ›› Issue (3) : 332 -346.

PDF
Ibrain ›› 2025, Vol. 11 ›› Issue (3) : 332 -346. DOI: 10.1002/ibra.70000
REVIEW

Epigenetic regulation in cognitive impairment: Focus on N6-methyladenosine modification and its potential role in perioperative neurocognitive disorders

Author information +
History +
PDF

Abstract

N6-methyladenosine (m6A), the most abundant internal modification in mammalian mRNA, plays a critical role in cognitive function by dynamically regulating gene expression. This narrative review examines m6A's role in cognitive processes and its potential impact on perioperative neurocognitive disorders (PNDs), which encompass a spectrum including postoperative delirium, delayed neurocognitive recovery, and postoperative cognitive dysfunction. The m6A regulatory machinery—comprising methyltransferases (“writers”), demethylases (“erasers”), and recognition proteins (“readers”)—modulates neuronal development, synaptic plasticity, and cognitive processes by influencing mRNA stability, translation, and degradation. Evidence from animal models indicates that m6A dysregulation contributes to neuroinflammation, neurodegeneration, and neuronal injury, which are pathophysiological mechanisms implicated in PNDs. Notably, anesthetic agents and surgical stress have been shown to alter hippocampal m6A levels, and manipulation of m6A-related proteins may ameliorate cognitive deficits. While these findings suggest potential mechanistic connections, direct evidence specifically linking m6A modifications to PNDs pathogenesis remains preliminary and largely based on preclinical models. Further research is needed to establish causal relationships, identify m6A-modified targets relevant to PNDs pathology, and evaluate m6A as a potential biomarker or therapeutic target. This review provides a foundation for understanding how m6A modification may influence perioperative cognitive outcomes and identifies promising avenues for future investigation.

Keywords

cognitive impairment / epitranscriptomics / N6-methyladenosine / neuroinflammation / perioperative neurocognitive disorders

Cite this article

Download citation ▾
Ting Liu, Xiao-Juan Yang, Lin Zhou, Mi Gan, Ting-Ting He, Sen Hong, Yan-Yan Feng, Gao Su, You-Xiao Zhao, Ying Cao, Qing-Fan Zeng. Epigenetic regulation in cognitive impairment: Focus on N6-methyladenosine modification and its potential role in perioperative neurocognitive disorders. Ibrain, 2025, 11(3): 332-346 DOI:10.1002/ibra.70000

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

Kong H, Xu LM, Wang DX. Perioperative neurocognitive disorders: a narrative review focusing on diagnosis, prevention, and treatment. CNS Neurosci Ther. 2022; 28(8): 1147-1167.

[2]

Evered L, Silbert B, Knopman DS, et al. Recommendations for the nomenclature of cognitive change associated with anaesthesia and surgery—2018. Br J Anaesth. 2018; 121(5): 1005-1012.

[3]

Tropea J, Logiudice D, Liew D, Gorelik A, Brand C. Poorer outcomes and greater healthcare costs for hospitalised older people with dementia and delirium: a retrospective cohort study. Int J Geriatr Psychiatry. 2017; 32(5): 539-547.

[4]

Park D, Kim HS, Kim JH. The effect of all-cause hospitalization on cognitive decline in older adults: a longitudinal study using databases of The National Health Insurance Service and the memory clinics of a self-run hospital. BMC Geriatr. 2023; 23(1): 61.

[5]

Duan S, Liao Y, Tang Y, et al. Short-term perioperative cognitive therapy combined with rehabilitation exercise reduces the incidence of neurocognitive disorder in elderly patients: a randomized controlled trial. Minerva Anestesiol. 2022; 88(3): 145-155.

[6]

Vacas S, Canales C, Deiner SG, Cole DJ. Perioperative brain health in the older adult: a patient safety imperative. Anesth Analg. 2022; 135(2): 316-328.

[7]

Linder B, Sharma P, Wu J, et al. tRNA modifications tune m(6)A-dependent mRNA decay. Cell. 2025; 188(14): 3715-3727.e13.

[8]

Huang H, Camats-Perna J, Medeiros R, Anggono V, Widagdo J. Altered expression of the m6A methyltransferase METTL3 in Alzheimer's disease. eNeuro. 2020; 7(5):ENEURO.0125-20.2020.

[9]

Qiu X, He H, Huang Y, Wang J, Xiao Y. Genome-wide identification of m(6)A-associated single-nucleotide polymorphisms in Parkinson's disease. Neurosci Lett. 2020; 737:135315.

[10]

Chen K, Hu Q, Xie Z, Yang G. Monocyte NLRP3-IL-1β hyperactivation mediates neuronal and synaptic dysfunction in perioperative neurocognitive disorder. Adv Sci. 2022; 9(16):e2104106.

[11]

He B, Wang J. METTL3 regulates hippocampal gene transcription via N6-methyladenosine methylation in sevoflurane-induced postoperative cognitive dysfunction mouse. Aging. 2021; 13(19): 23108-23118.

[12]

Ju LS, Morey TE, Seubert CN, Martynyuk AE. Intergenerational perioperative neurocognitive disorder. Biology. 2023; 12(4):567.

[13]

Zou Y, Shao LJZ, Xue FS. New nomenclature of peri-operative cognitive impairments: possible impacts on further practice and research. Chin Med J. 2019; 132(15): 1859-1861.

[14]

Chen H, Mo L, Hu H, Ou Y, Luo J. Risk factors of postoperative delirium after cardiac surgery: a meta-analysis. J Cardiothorac Surg. 2021; 16(1): 113.

[15]

Pedemonte JC, Sun H, Franco-Garcia E, et al. Postoperative delirium mediates 180-day mortality in orthopaedic trauma patients. Br J Anaesth. 2021; 127(1): 102-109.

[16]

Wang LW, Zhu MJ, Li Y, et al. FKBP51 is associated with early postoperative cognitive dysfunction in elderly patients undergoing hip fracture surgery. Medicine. 2019; 98(5):e14037.

[17]

Monk TG, Weldon BC, Garvan CW, et al. Predictors of cognitive dysfunction after major noncardiac surgery. Anesthesiology. 2008; 108(1): 18-30.

[18]

Joris J, Kehlet H, Slim K. Postoperative cognitive dysfunction: time for enhanced recovery after surgery programmes. Eur J Anaesthesiol. 2022; 39(9): 733-734.

[19]

Newman MF, Kirchner JL, Phillips-Bute B, et al. Longitudinal assessment of neurocognitive function after coronary-artery bypass surgery. N Engl J Med. 2001; 344(6): 395-402.

[20]

Selnes OA, Gottesman RF, Grega MA, Baumgartner WA, Zeger SL, McKhann GM. Cognitive and neurologic outcomes after coronary-artery bypass surgery. N Engl J Med. 2012; 366(3): 250-257.

[21]

Saczynski JS, Marcantonio ER, Quach L, et al. Cognitive trajectories after postoperative delirium. N Engl J Med. 2012; 367(1): 30-39.

[22]

Wang F. Postoperative cognitive dysfunction: current developments in mechanism and prevention. Med Sci Monit. 2014; 20: 1908-1912.

[23]

Moller J, Cluitmans P, Rasmussen L, et al. Long-term postoperative cognitive dysfunction in the elderly: ISPOCD1 study. The Lancet. 1998; 351(9106): 857-861.

[24]

Yang Y, Zhao X, Gao L, Wang Y, Wang J. Incidence and associated factors of delirium after orthopedic surgery in elderly patients: a systematic review and meta-analysis. Aging Clin Exp Res. 2021; 33(6): 1493-1506.

[25]

Liu XH, Zhang QF, Liu Y, et al. Risk factors associated with postoperative delirium in elderly patients undergoing hip surgery. Front Psychiatry. 2023; 14:1288117.

[26]

Harris MJ, Brovman EY, Urman RD. Clinical predictors of postoperative delirium, functional status, and mortality in geriatric patients undergoing non-elective surgery for hip fracture. J Clin Anesth. 2019; 58: 61-71.

[27]

Konishi Y, Evered LA, Scott DA, Silbert BS. Postoperative cognitive dysfunction after sevoflurane or propofol general anaesthesia in combination with spinal anaesthesia for hip arthroplasty. Anaesth Intensive Care. 2018; 46(6): 596-600.

[28]

Lai CC, Liu KH, Tsai CY, et al. Risk factors and effect of postoperative delirium on adverse surgical outcomes in older adults after elective abdominal cancer surgery in Taiwan. Asian J Surg. 2023; 46(3): 1199-1206.

[29]

Janssen TL, Steyerberg EW, Faes MC, et al. Risk factors for postoperative delirium after elective major abdominal surgery in elderly patients: a cohort study. Int J Surg. 2019; 71: 29-35.

[30]

Hovens IB, van Leeuwen BL, Mariani MA, Kraneveld AD, Schoemaker RG. Postoperative cognitive dysfunction and neuroinflammation; cardiac surgery and abdominal surgery are not the same. Brain Behav Immun. 2016; 54: 178-193.

[31]

Deeken F, Sánchez A, Rapp MA, et al. Outcomes of a delirium prevention program in older persons after elective surgery: a stepped-wedge cluster randomized clinical trial. JAMA Surg. 2022; 157(2):e216370.

[32]

Budėnas A, Tamašauskas Š, Šliaužys A, et al. Incidence and clinical significance of postoperative delirium after brain tumor surgery. Acta Neurochir. 2018; 160(12): 2327-2337.

[33]

Oh YS, Kim DW, Chun HJ, Yi HJ. Incidence and risk factors of acute postoperative delirium in geriatric neurosurgical patients. J Korean Neurosurg Soc. 2008; 43(3): 143-148.

[34]

Bronheim RS, Cotter E, Skolasky RL. Cognitive impairment is associated with greater preoperative symptoms, worse health-related quality of life, and reduced likelihood of recovery after cervical and lumbar spine surgery. North American Spine Soc J. 2022; 10:100128.

[35]

Hendriks EJ, Habets EJJ, Taphoorn MJB, et al. Linking late cognitive outcome with glioma surgery location using resection cavity maps. Hum Brain Mapp. 2018; 39(5): 2064-2074.

[36]

Pasqui E, de Donato G, Brancaccio B, et al. The predictive role of inflammatory biochemical markers in post-operative delirium after vascular surgery procedures. Vasc Health Risk Manag. 2022; 18: 747-756.

[37]

Alhamdah Y, Li WY, Nagappa M, et al. Perioperative approaches to prevent delayed neurocognitive recovery and postoperative neurocognitive disorder in older surgical patients: a systematic review and meta-analysis of randomized controlled trials. J Anaesthesiol Clin Pharmacol. 2025; 41(1): 3-14.

[38]

Mansouri N, Nasrollahi K, Shetabi H. Prevention of cognitive dysfunction after cataract surgery with intravenous administration of midazolam and dexmedetomidine in elderly patients undergoing cataract surgery. Adv Biomed Res. 2019; 8: 6.

[39]

Canet J, Raeder J, Rasmussen LS, et al. Cognitive dysfunction after minor surgery in the elderly. Acta Anaesthesiol Scand. 2003; 47(10): 1204-1210.

[40]

Potestio CP, Dibato J, Bolkus K, et al. Post-operative cognitive dysfunction in elderly patients receiving propofol sedation for gastrointestinal endoscopies: an observational study utilizing processed electroencephalography. Cureus. 2023; 15(10):e46588.

[41]

Li X, Lai H, Wang P, et al. Risk factors for postoperative cognitive decline after orthopedic surgery in elderly Chinese patients: a retrospective cohort study. Clin Interv Aging. 2024; 19: 491-502.

[42]

Yang X, Huang X, Li M, Jiang Y, Zhang H. Identification of individuals at risk for postoperative cognitive dysfunction (POCD). Ther Adv Neurol Disord. 2022; 15:17562864221114356.

[43]

Cibelli M, Fidalgo AR, Terrando N, et al. Role of interleukin-1β in postoperative cognitive dysfunction. Ann Neurol. 2010; 68(3): 360-368.

[44]

Wang J, Xin Y, Chu T, Liu C, Xu A. Dexmedetomidine attenuates perioperative neurocognitive disorders by suppressing hippocampal neuroinflammation and HMGB1/RAGE/NF-κB signaling pathway. Biomed Pharmacother. 2022; 150:113006.

[45]

Kwak MS, Kim HS, Lee B, Kim YH, Son M, Shin JS. Immunological significance of HMGB1 post-translational modification and redox biology. Front Immunol. 2020; 11:1189.

[46]

Xu F, Han L, Wang Y, et al. Prolonged anesthesia induces neuroinflammation and complement-mediated microglial synaptic elimination involved in neurocognitive dysfunction and anxiety-like behaviors. BMC Med. 2023; 21(1): 7.

[47]

Terrando N, Yang T, Wang X, et al. Systemic HMGB1 neutralization prevents postoperative neurocognitive dysfunction in aged rats. Front Immunol. 2016; 7:441.

[48]

Xiao MZ, Liu CX, Zhou LG, Yang Y, Wang Y. Postoperative delirium, neuroinflammation, and influencing factors of postoperative delirium: a review. Medicine. 2023; 102(8):e32991.

[49]

Westhoff D, Witlox J, Koenderman L, et al. Preoperative cerebrospinal fluid cytokine levels and the risk of postoperative delirium in elderly hip fracture patients. J Neuroinflammation. 2013; 10: 889.

[50]

Yang S, Gu C, Mandeville ET, et al. Anesthesia and surgery impair blood-brain barrier and cognitive function in mice. Front Immunol. 2017; 8:902.

[51]

Price CC. The new frontier of perioperative cognitive medicine for Alzheimer's disease and related dementias. Neurotherapeutics. 2022; 19(1): 132-142.

[52]

Tang Z, Cao J, Yao J, et al. KDM1A-mediated upregulation of METTL3 ameliorates Alzheimer's disease via enhancing autophagic clearance of p-Tau through m6A-dependent regulation of STUB1. Free Radic Biol Med. 2023; 195: 343-358.

[53]

Mao L, Zeng Q, Su W, Song M, Li J, Xie M. Elevation of miR-146a inhibits BTG2/BAX expression to ameliorate postoperative cognitive dysfunction following probiotics (VSL#3) treatment. Mol Neurobiol. 2021; 58(7): 3457-3470.

[54]

Yu Y, Yang Y, Tan H, et al. Tau contributes to sevoflurane-induced neurocognitive impairment In neonatal mice. Anesthesiology. 2020; 133(3): 595-610.

[55]

Xie Z, Swain CA, Ward SAP, et al. Preoperative cerebrospinal fluid β-Amyloid/Tau ratio and postoperative delirium. Ann Clin Transl Neurol. 2014; 1(5): 319-328.

[56]

Lu B, Nagappan G, Guan X, Nathan PJ, Wren P. BDNF-based synaptic repair as a disease-modifying strategy for neurodegenerative diseases. Nat Rev Neurosci. 2013; 14(6): 401-416.

[57]

Zhao BS, Wang X, Beadell AV, et al. m(6)A-dependent maternal mRNA clearance facilitates zebrafish maternal-to-zygotic transition. Nature. 2017; 542(7642): 475-478.

[58]

Wang X, Lu Z, Gomez A, et al. N6-methyladenosine-dependent regulation of messenger RNA stability. Nature. 2014; 505(7481): 117-120.

[59]

Liu J, Yue Y, Han D, et al. A METTL3-METTL14 complex mediates mammalian nuclear RNA N6-adenosine methylation. Nat Chem Biol. 2014; 10(2): 93-95.

[60]

Linder B, Grozhik AV, Olarerin-George AO, Meydan C, Mason CE, Jaffrey SR. Single-nucleotide-resolution mapping of m6A and m6Am throughout the transcriptome. Nat Methods. 2015; 12(8): 767-772.

[61]

Wang X, Zhao BS, Roundtree IA, et al. N(6)-methyladenosine modulates messenger RNA translation efficiency. Cell. 2015; 161(6): 1388-1399.

[62]

Roundtree IA, Luo GZ, Zhang Z, et al. YTHDC1 mediates nuclear export of N6-methyladenosine methylated mRNAs. eLife. 2017; 6:e31311.

[63]

Xiao W, Adhikari S, Dahal U, et al. Nuclear m(6)A reader YTHDC1 regulates mRNA splicing. Mol Cell. 2016; 61(4): 507-519.

[64]

Chang M, Lv H, Zhang W, et al. Region-specific RNA m6A methylation represents a new layer of control in the gene regulatory network in the mouse brain. Open Biol. 2017; 7(9):170166.

[65]

Merkurjev D, Hong WT, Iida K, et al. Synaptic N(6)-methyladenosine (m(6)A) epitranscriptome reveals functional partitioning of localized transcripts. Nature Neurosci. 2018; 21(7): 1004-1014.

[66]

Batista PJ, Molinie B, Wang J, et al. m(6)A RNA modification controls cell fate transition in mammalian embryonic stem cells. Cell Stem Cell. 2014; 15(6): 707-719.

[67]

Yoon KJ, Ringeling FR, Vissers C, et al. Temporal control of mammalian cortical neurogenesis by m(6)A methylation. Cell. 2017; 171(4): 877-889.e17.

[68]

Meyer KD, Jaffrey SR. Rethinking m(6)A readers, writers, and erasers. Annu Rev Cell Dev Biol. 2017; 33: 319-342.

[69]

Zaccara S, Ries RJ, Jaffrey SR. Reading, writing and erasing mRNA methylation. Nat Rev Mol Cell Biol. 2019; 20(10): 608-624.

[70]

Shi H, Wei J, He C. Where, when, and how: context-dependent functions of RNA methylation writers, readers, and erasers. Mol Cell. 2019; 74(4): 640-650.

[71]

Roundtree IA, Evans ME, Pan T, He C. Dynamic RNA modifications in gene expression regulation. Cell. 2017; 169(7): 1187-1200.

[72]

Wang P, Doxtader KA, Nam Y. Structural basis for cooperative function of Mettl3 and Mettl14 methyltransferases. Mol Cell. 2016; 63(2): 306-317.

[73]

Schöller E, Weichmann F, Treiber T, et al. Interactions, localization, and phosphorylation of the m(6)A generating METTL3-METTL14-WTAP complex. RNA. 2018; 24(4): 499-512.

[74]

Li F, Zhao D, Wu J, Shi Y. Structure of the YTH domain of human YTHDF2 in complex with an m(6)A mononucleotide reveals an aromatic cage for m(6)A recognition. Cell Res. 2014; 24(12): 1490-1492.

[75]

Radbakhsh S, Najar M, Merimi M, et al. RNA modifications in osteoarthritis: epitranscriptomic insights into pathogenesis and therapeutic targets. Int J Mol Sci. 2025; 26(10):4955.

[76]

Li HB, Tong J, Zhu S, et al. m(6)A mRNA methylation controls T cell homeostasis by targeting the IL-7/STAT5/SOCS pathways. Nature. 2017; 548(7667): 338-342.

[77]

Yue Y, Liu J, Cui X, et al. VIRMA mediates preferential m(6)A mRNA methylation in 3'UTR and near stop codon and associates with alternative polyadenylation. Cell Discov. 2018; 4: 10.

[78]

Ping XL, Sun BF, Wang L, et al. Mammalian WTAP is a regulatory subunit of the RNA N6-methyladenosine methyltransferase. Cell Res. 2014; 24(2): 177-189.

[79]

Chen Y, Peng C, Chen J, et al. WTAP facilitates progression of hepatocellular carcinoma via m6A-HuR-dependent epigenetic silencing of ETS1. Mol Cancer. 2019; 18(1): 127.

[80]

Deng G, Xu Y, Li Z, Zeng G. WTAP mediates IL-1β-induced chondrocyte injury by enhancing CA12 mRNA stability depending on m6A modification. J Orthop Surg. 2024; 19(1): 826.

[81]

Warda AS, Kretschmer J, Hackert P, et al. Human METTL16 is a N(6)-methyladenosine (m(6)A) methyltransferase that targets pre-mRNAs and various non-coding RNAs. EMBO Rep. 2017; 18(11): 2004-2014.

[82]

Ma H, Wang X, Cai J, et al. N(6-)Methyladenosine methyltransferase ZCCHC4 mediates ribosomal RNA methylation. Nat Chem Biol. 2019; 15(1): 88-94.

[83]

Zhao BS, Roundtree IA, He C. Post-transcriptional gene regulation by mRNA modifications. Nat Rev Mol Cell Biol. 2017; 18(1): 31-42.

[84]

Ueda Y, Ooshio I, Fusamae Y, et al. AlkB homolog 3-mediated tRNA demethylation promotes protein synthesis in cancer cells. Sci Rep. 2017; 7:42271.

[85]

Fedeles BI, Singh V, Delaney JC, Li D, Essigmann JM. The AlkB family of Fe(II)/α-Ketoglutarate-dependent dioxygenases: repairing nucleic acid alkylation damage and beyond. J Biol Chem. 2015; 290(34): 20734-20742.

[86]

Zhang C, Samanta D, Lu H, et al. Hypoxia induces the breast cancer stem cell phenotype by HIF-dependent and ALKBH5-mediated m6A-demethylation of NANOG mRNA. Proc Natl Acad Sci. 2016; 113(14): E2047-E2056.

[87]

Huang Y, Yan J, Li Q, et al. Meclofenamic acid selectively inhibits FTO demethylation of m6A over ALKBH5. Nucleic Acids Res. 2015; 43(1): 373-384.

[88]

Zhang X, Deng S, Peng Y, Wei H, Tian Z. ALKBH5 inhibits TNF-α-induced apoptosis of HUVECs through Bcl-2 pathway. Open Med. 2022; 17(1): 1092-1099.

[89]

Zhou J, Wan J, Gao X, Zhang X, Jaffrey SR, Qian SB. Dynamic m(6)A mRNA methylation directs translational control of heat shock response. Nature. 2015; 526(7574): 591-594.

[90]

Song H, Feng X, Zhang H, et al. METTL3 and ALKBH5 oppositely regulate m(6)A modification of TFEB mRNA, which dictates the fate of hypoxia/reoxygenation-treated cardiomyocytes. Autophagy. 2019; 15(8): 1419-1437.

[91]

Gerken T, Girard CA, Tung YCL, et al. The obesity-associated FTO gene encodes a 2-oxoglutarate-dependent nucleic acid demethylase. Science. 2007; 318(5855): 1469-1472.

[92]

Li L, Zang L, Zhang F, et al. Fat mass and obesity-associated (FTO) protein regulates adult neurogenesis. Hum Mol Gen. 2017; 26(13): 2398-2411.

[93]

Zhu T, Roundtree IA, Wang P, et al. Crystal structure of the YTH domain of YTHDF2 reveals mechanism for recognition of N6-methyladenosine. Cell Res. 2014; 24(12): 1493-1496.

[94]

Huang H, Weng H, Sun W, et al. Recognition of RNA N(6)-methyladenosine by IGF2BP proteins enhances mRNA stability and translation. Nature Cell Biol. 2018; 20(3): 285-295.

[95]

Liu N, Dai Q, Zheng G, He C, Parisien M, Pan T. N(6)-methyladenosine-dependent RNA structural switches regulate RNA-protein interactions. Nature. 2015; 518(7540): 560-564.

[96]

Alarcón CR, Goodarzi H, Lee H, Liu X, Tavazoie S, Tavazoie SF. HNRNPA2B1 is a mediator of m(6)A-dependent nuclear RNA processing events. Cell. 2015; 162(6): 1299-1308.

[97]

Du H, Zhao Y, He J, et al. YTHDF2 destabilizes m(6)A-containing RNA through direct recruitment of the CCR4-NOT deadenylase complex. Nat Commun. 2016; 7:12626.

[98]

Yu R, Li Q, Feng Z, Cai L, Xu Q. m6A reader YTHDF2 regulates LPS-induced inflammatory response. Int J Mol Sci. 2019; 20(6):1323.

[99]

Shi H, Wang X, Lu Z, et al. YTHDF3 facilitates translation and decay of N(6)-methyladenosine-modified RNA. Cell Res. 2017; 27(3): 315-328.

[100]

Li A, Chen YS, Ping XL, et al. Cytoplasmic m(6)A reader YTHDF3 promotes mRNA translation. Cell Res. 2017; 27(3): 444-447.

[101]

Hsu PJ, Zhu Y, Ma H, et al. Ythdc2 is an N(6)-methyladenosine binding protein that regulates mammalian spermatogenesis. Cell Res. 2017; 27(9): 1115-1127.

[102]

Li Z, Li Y, Shen L, Shen L, Li N. Molecular characterization, clinical relevance and immune feature of m7G regulator genes across 33 cancer types. Front Genet. 2022; 13:981567.

[103]

Ries RJ, Zaccara S, Klein P, et al. m(6)A enhances the phase separation potential of mRNA. Nature. 2019; 571(7765): 424-428.

[104]

Zhang F, Kang Y, Wang M, et al. Fragile X mental retardation protein modulates the stability of its m6A-marked messenger RNA targets. Hum Mol Gen. 2018; 27(22): 3936-3950.

[105]

Wu R, Li A, Sun B, et al. A novel m(6)A reader Prrc2a controls oligodendroglial specification and myelination. Cell Res. 2019; 29(1): 23-41.

[106]

Shi H, Zhang X, Weng YL, et al. m(6)A facilitates hippocampus-dependent learning and memory through YTHDF1. Nature. 2018; 563(7730): 249-253.

[107]

Song B, Zeng Y, Cao Y, et al. Emerging role of METTL3 in inflammatory diseases: mechanisms and therapeutic applications. Front Immunol. 2023; 14:1221609.

[108]

Wu X, Liu H, Wang J, et al. The m(6)A methyltransferase METTL3 drives neuroinflammation and neurotoxicity through stabilizing BATF mRNA in microglia. Cell Death Differ. 2025; 32(1): 100-117.

[109]

Bao T, Liao T, Cai X, et al. METTL3 mediated ferroptosis in chondrocytes and promoted pain in KOA via HMGB1 m6A modification. Cell Biol Int. 2024; 48(11): 1755-1765.

[110]

Zhou J, Zhang X, Hu J, et al. m(6)A demethylase ALKBH5 controls CD4(+) T cell pathogenicity and promotes autoimmunity. Sci Adv. 2021; 7(25):eabg0470.

[111]

Chen X, Lai J, Wu Z, et al. Fat mass and obesity-mediated N 6 -methyladenosine modification modulates neuroinflammatory responses after traumatic brain injury. Neural Regen Res. 2026; 21(2): 730-741.

[112]

Huang P, Liu M, Zhang J, Zhong X, Zhong C. YTHDF1 attenuates TBI-induced brain-gut axis dysfunction in mice. Int J Mol Sci. 2023; 24(4):4240.

[113]

Zeng Z, Lan Y, Zhang L, et al. The m6A reader YTHDF2 alleviates the inflammatory response by inhibiting IL-6R/JAK2/STAT1 pathway-mediated high-mobility group box-1 release. Burns Trauma. 2023; 11:tkad023.

[114]

Galea I. The blood-brain barrier in systemic infection and inflammation. Cell Mol Immunol. 2021; 18(11): 2489-2501.

[115]

Chen J, Zhang YC, Huang C, et al. m(6)A regulates neurogenesis and neuronal development by modulating histone methyltransferase Ezh2. Genomics Insights. 2019; 17(2): 154-168.

[116]

Zhao F, Xu Y, Gao S, et al. METTL3-dependent RNA m(6)A dysregulation contributes to neurodegeneration in Alzheimer's disease through aberrant cell cycle events. Mol Neurodegener. 2021; 16(1): 70.

[117]

Zhao J, Ren J, Liu S, et al. Repeated exposure to sevoflurane in neonatal rats impairs cognition in adulthood via the PKA-CREB-BDNF signaling pathway. Exp Ther Med. 2021; 22(6): 1442.

[118]

Song J, Hao J, Lu Y, Ding X, Li M, Xin Y. Increased m(6)A modification of BDNF mRNA via FTO promotes neuronal apoptosis following aluminum-induced oxidative stress. Environ Pollut. 2024; 349:123848.

[119]

Huang Y, Yang Y, Ye C, Liu Z, Wei F. The m(6)A reader YTHDF1 improves sevoflurane-induced neuronal pyroptosis and cognitive dysfunction through augmenting CREB-BDNF signaling. Neurochem Res. 2023; 48(12): 3625-3638.

[120]

Ma LH, Yan J, Jiao XH, Zhou CH, Wu YQ. The role of epigenetic modifications in neurotoxicity induced by neonatal general anesthesia. Front Mol Neurosci. 2022; 15:877263.

[121]

Wu WF, Lin JT, Qiu YK, et al. The role of epigenetic modification in postoperative cognitive dysfunction. Ageing Res Rev. 2023; 89:101983.

[122]

He PC, He C. m(6) A RNA methylation: from mechanisms to therapeutic potential. EMBO J. 2021; 40(3):e105977.

[123]

Lv J, Xing L, Zhong X, Li K, Liu M, Du K. Role of N6-methyladenosine modification in central nervous system diseases and related therapeutic agents. Biomed Pharmacother. 2023; 162:114583.

[124]

Meng X, Wang Y, Zhao W, et al. Identification of differential m6A RNA methylomes and ALKBH5 as a potential prevention target in the developmental neurotoxicity induced by multiple sevoflurane exposures. FASEB J. 2024; 38(14):e23793.

[125]

Zhao L, Zhu H, Mao W, Zhou X, Xie Y, Li L. Effects of perioperative cognitive function training on postoperative cognitive dysfunction and postoperative delirium: a systematic review and meta-analysis. Front Neurol. 2023; 14:1293153.

[126]

Huff S, Tiwari SK, Gonzalez GM, Wang Y, Rana TM. m(6)A-RNA demethylase FTO inhibitors impair self-renewal in glioblastoma stem cells. ACS Chem Biol. 2021; 16(2): 324-333.

RIGHTS & PERMISSIONS

2025 The Author(s). Ibrain published by Affiliated Hospital of Zunyi Medical University and Wiley-VCH GmbH.

AI Summary AI Mindmap
PDF

50

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/