We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×

Structural characteristics of small-molecule inhibitors targeting FTO demethylase

    Shuting Gao

    Green Catalysis Center, College of Chemistry, Zhengzhou University, Zhengzhou, China

    ,
    Xitong Li

    Green Catalysis Center, College of Chemistry, Zhengzhou University, Zhengzhou, China

    ,
    Miao Zhang

    Green Catalysis Center, College of Chemistry, Zhengzhou University, Zhengzhou, China

    ,
    Ning Zhang

    Green Catalysis Center, College of Chemistry, Zhengzhou University, Zhengzhou, China

    ,
    Ruiyong Wang

    *Author for correspondence:

    E-mail Address: wangry@zzu.edu.cn

    Green Catalysis Center, College of Chemistry, Zhengzhou University, Zhengzhou, China

    &
    Junbiao Chang

    **Author for correspondence:

    E-mail Address: changjunbiao@zzu.edu.cn

    Green Catalysis Center, College of Chemistry, Zhengzhou University, Zhengzhou, China

    Published Online:https://doi.org/10.4155/fmc-2021-0132

    Studies have shown that the FTO gene is closely related to obesity and weight gain in humans. FTO is an N6-methyladenosine demethylase and is linked to an increased risk of obesity and a variety of diseases, such as acute myeloid leukemia, type 2 diabetes, breast cancer, glioblastoma and cervical squamous cell carcinoma. In light of the significant role of FTO, the development of small-molecule inhibitors targeting the FTO protein provides not only a powerful tool for grasping the active site of FTO but also a theoretical basis for the design and synthesis of drugs targeting the FTO protein. This review focuses on the structural characteristics of FTO inhibitors and discusses the occurrence of obesity and cancer caused by FTO gene overexpression.

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    References

    • 1. Frayling TM, Timpson NJ, Weedon MN. A common variant in the FTO gene is associated with body mass index and predisposes to childhood and adult obesity. Science 316(5826), 889–894 (2007). • Demonstrates that the FTO gene is associated with obesity.
    • 2. Dina C, Meyre D, Gallina S et al. Variation in FTO contributes to childhood obesity and severe adult obesity. Nat. Genet. 39(6), 724–726 (2007).
    • 3. Cecil JE, Tavendale R, Watt P, Hetherington MM, Palmer CNA. An obesity-associated FTO gene variant and increased energy intake in children. N. Engl. J. Med. 359(24), 2558–2566 (2008).
    • 4. Claussnitzer M, Dankel SN, Kim KH et al. FTO obesity variant circuitry and adipocyte browning in humans. N. Engl. J. Med. 373(10), 895–907 (2015).
    • 5. Villalobos-Comparan M, Teresa Flores-Dorantes M, Teresa Villarreal-Molina M et al. The FTO gene is associated with adulthood obesity in the Mexican population. Obesity (Silver Spring) 16(10), 2296–2301 (2008).
    • 6. Livingstone KM, Celis-Morales C, Papandonatos GD et al. FTO genotype and weight loss: systematic review and meta-analysis of 9563 individual participant data from eight randomised controlled trials. BMJ 354, i4707 (2016).
    • 7. Scuteri A, Sanna S, Chen WM et al. Genome-wide association scan shows genetic variants in the FTO gene are associated with obesity-related traits. PLoS Genet. 3(7), 1200–1210 (2007).
    • 8. Cho YS, Go MJ, Kim YJ et al. A large-scale genome-wide association study of Asian populations uncovers genetic factors influencing eight quantitative traits. Nat. Genet. 41(5), 527–534 (2009).
    • 9. Dong CH, Beecham A, Slifer S et al. Genome-wide linkage and peak-wide association study of obesity-related quantitative traits in Caribbean Hispanics. Hum. Genet. 129(2), 209–219 (2011).
    • 10. Berulava T, Horsthemke B. The obesity-associated SNPs in intron 1 of the FTO gene affect primary transcript levels. Eur. J. Hum. Genet. 18(9), 1054–1056 (2010).
    • 11. Loos RJF, Yeo GSH. The bigger picture of FTO – the first GWAS-identified obesity gene. Nat. Rev. Endocrinol. 10(1), 51–61 (2014).
    • 12. Kalantari N, Mohammadi NK, Izadi P et al. A complete linkage disequilibrium in a haplotype of three SNPs in fat mass and obesity associated (FTO) gene was strongly associated with anthropometric indices after controlling for calorie intake and physical activity. BMC Med. Genet. 19(1), 146–152 (2018).
    • 13. Church C, Lee S, Bagg EA et al. A mouse model for the metabolic effects of the human fat mass and obesity associated FTO gene. PLoS Genet. 5(8), e1000599 (2009).
    • 14. Gao X, Shin YH, Li M, Wang F, Tong Q, Zhang P. The fat mass and obesity associated gene FTO functions in the brain to regulate postnatal growth in mice. PLoS ONE 5(11), e14005 (2010).
    • 15. Ronkainen J, Mondini E, Cinti F et al. FTO-deficiency affects the gene and microRNA expression involved in brown adipogenesis and browning of white adipose tissue in mice. Int. J. Mol. Sci. 17(11), 1851 (2016).
    • 16. Eissing L. FTO-associated obesity risk is linked to brain food responses via modulation of ghrelin levels. Nat. Rev. Endocrinol. 9(10), 564–564 (2013).
    • 17. Karra E, O'Daly OG, Choudhury AI et al. A link between FTO, ghrelin, and impaired brain food-cue responsivity. J. Clin. Invest. 123(8), 3539–3551 (2013).
    • 18. Merkestein M, Sellayah D. Role of FTO in adipocyte development and function: recent insights. Int. J. Endocrinol. 2015, 521381 (2015).
    • 19. Fischer J, Koch L, Emmerling C et al. Inactivation of the FTO gene protects from obesity. Nature 458(7240), 894–898 (2009).
    • 20. McMurray F, Church CD, Larder R et al. Adult onset global loss of the fto gene alters body composition and metabolism in the mouse. PLoS Genet. 9(1), e1003166 (2013).
    • 21. Stratigopoulos G, Martin Carli JF, O'Day DR et al. Hypomorphism for RPGRIP1L, a ciliary gene vicinal to the FTO locus, causes increased adiposity in mice. Cell Metab. 19(5), 767–779 (2014).
    • 22. Anselme I, Laclef C, Lanaud M, Ruther U, Schneider-Maunoury S. Defects in brain patterning and head morphogenesis in the mouse mutant fused toes. Dev. Biol. 304(1), 208–220 (2007).
    • 23. Rong R, Hanson RL, Ortiz D et al. Association analysis of variation in/near FTO, CDKAL1, SLC30A8, HHEX, EXT2, IGF2BP2, LOC387761, and CDKN2B with type 2 diabetes and related quantitative traits in Pima Indians. Diabetes 58(2), 478–488 (2009).
    • 24. Han Z, Niu T, Chang J et al. Crystal structure of the FTO protein reveals basis for its substrate specificity. Nature 464(7292), 1205–1209 (2010). •• Reveals the FTO protein crystal structure.
    • 25. Sanchez-Pulido L, Andrade-Navarro MA. The FTO (fat mass and obesity associated) gene codes for a novel member of the non-heme dioxygenase superfamily. BMC Biochem. 8(1), 23 (2007).
    • 26. Gerken T, Girard CA, Tung YCL et al. The obesity-associated FTO gene encodes a 2-oxoglutarate-dependent nucleic acid demethylase. Science 318(5855), 1469–1472 (2007).
    • 27. Hausinger RP. FeII/alpha-ketoglutarate-dependent hydroxylases and related enzymes. Crit. Rev. Biochem. Mol. Biol. 39(1), 21–68 (2004).
    • 28. Jia G, Yang C-G, Yang S et al. Oxidative demethylation of 3-methylthymine and 3-methyluracil in single-stranded DNA and RNA by mouse and human FTO. FEBS Lett. 582(23–24), 3313–3319 (2008).
    • 29. Jia G, Fu Y, Zhao X et al. N6-methyladenosine in nuclear RNA is a major substrate of the obesity-associated FTO. Nat. Chem. Biol. 7(12), 885–887 (2011).
    • 30. Fu Y, Jia G, Pang X et al. FTO-mediated formation of N6-hydroxymethyladenosine and N6-formyladenosine in mammalian RNA. Nat. Commun. 4(1), 1798–1806 (2013).
    • 31. Wei J, Liu F, Lu Z et al. Differential m(6)A, m(6)Am, and m(1)A demethylation mediated by FTO in the cell nucleus and cytoplasm. Mol. Cell 71(6), 973–985 (2018).
    • 32. Zhang C, Samanta D, Lu H et al. Hypoxia induces the breast cancer stem cell phenotype by HIF-dependent and ALKBH5-mediated m(6)A-demethylation of NANOG mRNA. Proc. Natl Acad. Sci. USA 113(14), e2047–e2056 (2016).
    • 33. Lin S, Choe J, Du P, Triboulet R, Gregory RI. The m(6)A methyltransferase METTL3 promotes translation in human cancer cells. Mol. Cell 62(3), 335–345 (2016).
    • 34. Vu LP, Pickering BF, Cheng Y et al. The N(6)-methyladenosine (m(6)A)-forming enzyme METTL3 controls myeloid differentiation of normal hematopoietic and leukemia cells. Nat. Med. 23(11), 1369–1376 (2017).
    • 35. Barbieri I, Tzelepis K, Pandolfini L et al. Promoter-bound METTL3 maintains myeloid leukaemia by m(6)A-dependent translation control. Nature 552(7683), 126–131 (2017).
    • 36. Zhang S, Zhao BS, Zhou A et al. m(6)A demethylase ALKBH5 maintains tumorigenicity of glioblastoma stem-like cells by sustaining FOXM1 expression and cell proliferation program. Cancer Cell 31(4), 591–606 (2017).
    • 37. Deng X, Su R, Feng X, Wei M, Chen J. Role of N(6)-methyladenosine modification in cancer. Curr. Opin. Genet. Dev. 48, 1–7 (2018).
    • 38. Deng X, Su R, Weng H, Huang H, Li Z, Chen J. RNA N(6)-methyladenosine modification in cancers: current status and perspectives. Cell Res. 28(5), 507–517 (2018).
    • 39. Muthusamy S. m(6)A mRNA methylation: a pleiotropic regulator of cancer. Gene 736, 144415 (2020).
    • 40. Li J, Chen F, Peng Y et al. N6-methyladenosine regulates the expression and secretion of TGFbeta1 to affect the epithelial–mesenchymal transition of cancer cells. Cells 9(2), 196 (2020).
    • 41. Hernandez-Caballero ME, Sierra-Ramirez JA. Single nucleotide polymorphisms of the FTO gene and cancer risk: an overview. Mol. Biol. Rep. 42(3), 699–704 (2015).
    • 42. Iles MM, Law MH, Stacey SN et al. A variant in FTO shows association with melanoma risk not due to BMI. Nat. Genet. 45(4), 428–432 (2013).
    • 43. Huang X, Zhao J, Yang M, Li M, Zheng J. Association between FTO gene polymorphism (rs9939609 T/A) and cancer risk: a meta-analysis. Eur. J. Cancer Care (Engl.) 26(5), e12464 (2017).
    • 44. Li Z, Weng H, Su R et al. FTO plays an oncogenic role in acute myeloid leukemia as a N(6)-methyladenosine RNA demethylase. Cancer Cell 31(1), 127–141 (2017).
    • 45. Su R, Dong L, Li C et al. R-2HG exhibits anti-tumor activity by targeting FTO/m(6)A/MYC/CEBPA signaling. Cell 172(1–2), 90–105 (2018).
    • 46. Chen B, Ye F, Yu L et al. Development of cell-active N6-methyladenosine RNA demethylase FTO inhibitor. J. Am. Chem. Soc. 134(43), 17963–17971 (2012). •• Discusses the first FTO inhibitor.
    • 47. Huang Y, Yan J, Li Q et al. Meclofenamic acid selectively inhibits FTO demethylation of m6A over ALKBH5. Nucleic Acids Res. 43(1), 373–384 (2015). • Investigates meclofenamic acid (MA) inhibitors.
    • 48. Wang T, Hong T, Huang Y et al. Fluorescein derivatives as bifunctional molecules for the simultaneous inhibiting and labeling of FTO protein. J. Am. Chem. Soc. 137(43), 13736–13739 (2015).
    • 49. Huang Y, Su R, Sheng Y et al. Small-molecule targeting of oncogenic FTO demethylase in acute myeloid leukemia. Cancer Cell 35(4), 677–691 (2019).
    • 50. He W, Zhou B, Liu W et al. Identification of a novel small-molecule binding site of the fat mass and obesity associated protein (FTO). J. Med. Chem. 58(18), 7341–7348 (2015).
    • 51. Qiao Y, Zhou B, Zhang M et al. A novel inhibitor of the obesity-related protein FTO. Biochemistry 55(10), 1516–1522 (2016).
    • 52. Padariya M, Kalathiya U. Structure-based design and evaluation of novel N-phenyl-1H-indol-2-amine derivatives for fat mass and obesity-associated (FTO) protein inhibition. Comput. Biol. Chem. 64, 414–425 (2016).
    • 53. Zhang L, Wang Z, Ren T et al. Investigation of the interaction between FTO and 3-substituted 2-aminochromones by spectroscopy and molecular modeling. Med. Chem. Res. 26(7), 1349–1358 (2017).
    • 54. Wang N, Han X, Li J et al. Comparative study of the bindings between 3-phenyl-1H-indazole and five proteins by isothermal titration calorimetry, spectroscopy and docking methods. J. Biomol. Struct. Dyn. 37(17), 4580–4589 (2019).
    • 55. Zhang L, Ren T, Tian X et al. Investigation of the interaction between 1,3-diazaheterocyclic compounds and the fat mass and obesity-associated protein by fluorescence spectroscopy and molecular modeling. J. Fluoresc. 27(1), 369–378 (2017).
    • 56. Li J, Wang Y, Han X et al. The role of chlorine atom on the binding between 2-phenyl-1H-benzimidazole analogues and fat mass and obesity-associated protein. J. Mol. Recognit. 32(6), e2774 (2019).
    • 57. Bai N, Gan Y, Li X et al. The role of chlorine atom on the binding between acrylonitrile derivatives and fat mass and obesity-associated protein. J. Mol. Recognit. 34(4), e2880 (2021).
    • 58. 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. 16(2), 324–333 (2021).
    • 59. Zheng G, Cox T, Tribbey L et al. Synthesis of a FTO inhibitor with anticonvulsant activity. ACS Chem. Neurosci. 5(8), 658–665 (2014).
    • 60. Ren T, Wang Z, Zhang L et al. Study of the binding between camptothecin analogs and FTO by spectroscopy and molecular docking. J. Fluoresc. 27(4), 1467–1477 (2017).
    • 61. Wang R, Han Z, Liu B et al. Identification of natural compound radicicol as a potent FTO inhibitor. Mol. Pharm. 15(9), 4092–4098 (2018).
    • 62. Han X, Wang N, Li J, Wang Y, Wang R, Chang J. Identification of nafamostat mesilate as an inhibitor of the fat mass and obesity-associated protein (FTO) demethylase activity. Chem. Biol. Interact. 297, 80–84 (2019).
    • 63. Peng SM, Xiao W, Ju DP et al. Identification of entacapone as a chemical inhibitor of FTO mediating metabolic regulation through FOXO1. Sci. Transl. Med. 11(488), eaau7116 (2019).
    • 64. Su R, Dong L, Li Y et al. Targeting FTO suppresses cancer stem cell maintenance and immune evasion. Cancer Cell 38(1), 79–96 (2020). •• Discusses the antileukocyte activity of FTO inhibitors.
    • 65. Li X, Gao S, Zhang N, Zhang M, Wang R, Chang J. Identification of tectoridin as the inhibitor of FTO by isothermal titration calorimetric and spectroscopic methods. New J. Chem. 45(20), 8993–9001 (2021).
    • 66. Aik W, Demetriades M, Hamdan MK et al. Structural basis for inhibition of the fat mass and obesity associated protein (FTO). J. Med. Chem. 56(9), 3680–3688 (2013).
    • 67. Toh JDW, Sun L, Lau LZM et al. A strategy based on nucleotide specificity leads to a subfamily-selective and cell-active inhibitor of N(6)-methyladenosine demethylase FTO. Chem. Sci. 6(1), 112–122 (2015).
    • 68. Wang Z, Wang N, Han X, Wang R, Chang J. Fluorescence quenching and molecular docking study on the binding of four hydroxyanthraquinones to FTO. Phys. Chem. Liq. 56(4), 482–495 (2017).
    • 69. A Shisheva, Y Shechter. Quercetin selectively inhibits insulin receptor function in vitro and the bioresponses of insulin and insulinomimetic agents in rat adipocytes. Biochemistry 31(34), 8059–8063 (1992).
    • 70. Wu R, Yao Y, Jiang Q et al. Epigallocatechin gallate targets FTO and inhibits adipogenesis in an mRNA m(6)A-YTHDF2-dependent manner. Int. J. Obes. (Lond.) 42(7), 1378–1388 (2018).
    • 71. Sumaryada T, Simamora R, Ambarsari L. Docking evaluation of catechin and its derivatives on fat mass and obesity-associated (FTO) protein for anti-obesity agent. J. Appl. Pharm. Sci. 8(8), 63–68 (2018).
    • 72. Wang Z, Wang N, Han X, Wang R, Chang J. Interaction of two flavonols with fat mass and obesity-associated protein investigated by fluorescence quenching and molecular docking. J. Biomol. Struct. Dyn. 36(13), 3388–3397 (2018).
    • 73. Zhang Y, Li Q-N, Zhou K, Xu Q, Zhang C-Y. Identification of specific N6-methyladenosine RNA demethylase FTO inhibitors by single-quantum-dot-based FRET nanosensors. Anal. Chem. 92(20), 13936–13944 (2020).
    • 74. Yang L, Yu W, Li Z et al. Comparative study of the binding between FTO protein and five pyrazole derivatives by spectrofluorimetry. J. Mol. Liq. 218, 156–165 (2016).
    • 75. Li Z, Wang Z, Wang N et al. Identification of the binding between three fluoronucleoside analogues and fat mass and obesity-associated protein by isothermal titration calorimetry and spectroscopic techniques. J. Pharm. Biomed. Anal. 149, 290–295 (2018).
    • 76. Wang Y, Li J, Han X et al. Identification of clausine E as an inhibitor of fat mass and obesity-associated protein (FTO) demethylase activity. J. Mol. Recognit. 32(10), e2800 (2019).
    • 77. Afshin A, Forouzanfar MH, Reitsma MB et al. Health effects of overweight and obesity in 195 countries over 25 years. N. Engl. J. Med. 377(1), 13–27 (2017).
    • 78. World Health Organization. Obesity and overweight (2021). https://www.who.int/news-room/fact-sheets/detail/obesity-and-overweight
    • 79. Wardle J, Carnell S, Haworth CM, Farooqi IS, O'Rahilly S, Plomin R. Obesity associated genetic variation in FTO is associated with diminished satiety. J. Clin. Endocrinol. Metab. 93(9), 3640–3643 (2008).
    • 80. Haupt A, Thamer C, Staiger H et al. Variation in the FTO gene influences food intake but not energy expenditure. Exp. Clin. Endocrinol. Diabetes 117(4), 194–197 (2009).
    • 81. Graff M, Scott RA, Justice AE et al. Genome-wide physical activity interactions in adiposity – a meta-analysis of 200,452 adults. PLoS Genet. 13(4), e1006528 (2017).
    • 82. Graff M, Scott RA, Justice AE et al. Correction: genome-wide physical activity interactions in adiposity – a meta-analysis of 200,452 adults. PLoS Genet. 13(8), e1006972 (2017).
    • 83. Han Y, Wang W, Jia J et al. WGCNA analysis of the subcutaneous fat transcriptome in a novel tree shrew model. Exp. Biol. Med. (Maywood) 245(11), 945–955 (2020).
    • 84. Doaei S, Kalantari N, Izadi P et al. Interactions between macro-nutrients' intake, FTO and IRX3 gene expression, and FTO genotype in obese and overweight male adolescents. Adipocyte 8(1), 386–391 (2019).
    • 85. Doaei S, Kalantari N, Izadi P et al. Changes in FTO and IRX3 gene expression in obese and overweight male adolescents undergoing an intensive lifestyle intervention and the role of FTO genotype in this interaction. J. Transl. Med. 17(1), 176 (2019).
    • 86. Doaei S, Kalantari N, Mohammadi NK et al. Up-regulation of FTO gene expression was associated with increase in skeletal muscle mass in overweight male adolescents. Arch. Med. Sci. 15(5), 1133–1137 (2019).
    • 87. Yu K, Fang L, Yao LJO. Is FTO gene variant related to cancer risk independently of adiposity? An updated meta-analysis of 129,467 cases and 290,633 controls. Oncotarget 8(31), 50987–50996 (2017).
    • 88. Tews D, Fischer-Posovszky P, Wabitsch M. Regulation of FTO and FTM expression during human preadipocyte differentiation. Horm Metab Res 43(1), 17–21 (2011).
    • 89. Lappalainen T, Kolehmainen M, Schwab U et al. Gene expression of FTO in human subcutaneous adipose tissue, peripheral blood mononuclear cells and adipocyte cell line. J. Nutrigenet. Nutrigenomics 3(1), 37–45 (2010).
    • 90. Ben-Haim MS, Moshitch-Moshkovitz S, Rechavi G. FTO: linking m6A demethylation to adipogenesis. Cell Res. 25(1), 3–4 (2015).
    • 91. Zhang M, Zhang Y, Ma J et al. The demethylase activity of FTO (fat mass and obesity associated protein) is required for preadipocyte differentiation. PLoS ONE 10(7), e0133788 (2015).
    • 92. Eghbalsaied S, Khorasgani FR, Ebrahimi YB, Ostadsharif M. Gene polymorphisms of human FTO and obesity in part of Iranian population. Rec. Adv. Biol. Med. 5, 881123 (2019).
    • 93. Lopez-Rodriguez G, Estrada-Neria A, Suarez-Dieguez T, Tejero ME, Fernandez JC, Galvan M. Common polymorphisms in MC4R and FTO genes are associated with BMI and metabolic indicators in Mexican children: differences by sex and genetic ancestry. Gene 754, 144840 (2020).
    • 94. Deng T, Lyon CJ, Bergin S, Caligiuri MA, Hsueh WA. Obesity, inflammation, and cancer. Annu. Rev. Pathol. 11, 421–449 (2016).
    • 95. Gu Z, Bi Y, Yuan F et al. FTO polymorphisms are associated with metabolic dysfunction-associated fatty liver disease (MAFLD) susceptibility in the older Chinese Han population. Clin. Int. Aging 15, 1333–1341 (2020).
    • 96. Korek E, Krauss H. Novel adipokines: their potential role in the pathogenesis of obesity and metabolic disorders. Postepy. Hig. Med. Dosw. 69, 799–810 (2015).
    • 97. Kaklamani V, Yi NJ, Sadim M et al. The role of the fat mass and obesity associated gene (FTO) in breast cancer risk. BMC Med. Genet. 12(1), 52 (2011).
    • 98. Chen J, Du B. Novel positioning from obesity to cancer: FTO, an m(6)A RNA demethylase, regulates tumour progression. J. Cancer Res. Clin. Oncol. 145(1), 19–29 (2019).
    • 99. Singh B, Kinne HE, Milligan RD, Washburn LJ, Olsen M, Lucci A. Important role of FTO in the survival of rare panresistant triple-negative inflammatory breast cancer cells facing a severe metabolic challenge. PLoS ONE 11(7), e0159072 (2016).
    • 100. Khan S, Verma AK, Khan V et al. Role of FTO and MC4R polymorphisms in escalating obesity and their indirect association with risk of T2D in Indian population. Diabetes Ther. 11(9), 2145–2157 (2020).
    • 101. Li H, Ren Y, Mao K et al. FTO is involved in Alzheimer's disease by targeting TSC1-mTOR-Tau signaling. Biochem. Biophys. Res. Commun. 498(1), 234–239 (2018).
    • 102. Zhang Z, Zhou D, Lai Y et al. Estrogen induces endometrial cancer cell proliferation and invasion by regulating the fat mass and obesity-associated gene via PI3K/AKT and MAPK signaling pathways. Cancer Lett. 319(1), 89–97 (2012).
    • 103. Xu D, Shao W, Jiang Y, Wang X, Liu Y, Liu X. FTO expression is associated with the occurrence of gastric cancer and prognosis. Oncol. Rep. 38(4), 2285–2292 (2017).
    • 104. Fustin JM, Doi M, Yamaguchi Y et al. RNA-methylation-dependent RNA processing controls the speed of the circadian clock. Cell 155(4), 793–806 (2013).
    • 105. Lim A, Zhou J, Sinha RA et al. Hepatic FTO expression is increased in NASH and its silencing attenuates palmitic acid-induced lipotoxicity. Biochem. Biophys. Res. Commun. 479(3), 476–481 (2016).
    • 106. Kang H, Zhang Z, Yu L, Li Y, Liang M, Zhou L. FTO reduces mitochondria and promotes hepatic fat accumulation through RNA demethylation. J. Cell. Biochem. 119(7), 5676–5685 (2018).
    • 107. Zhuang C, Zhuang C, Luo X et al. N6-methyladenosine demethylase FTO suppresses clear cell renal cell carcinoma through a novel FTO-PGC-1alpha signalling axis. J. Cell. Mol. Med. 23(3), 2163–2173 (2019).
    • 108. Xu W, Yang H, Liu Y et al. Oncometabolite 2-hydroxyglutarate is a competitive inhibitor of alpha-ketoglutarate-dependent dioxygenases. Cancer Cell 19(1), 17–30 (2011).
    • 109. Feliciano P. PAK inhibitor in fragile X. Nat. Genet. 45(5), 477–477 (2013).
    • 110. Zhou S, Bai ZL, Xia D et al. FTO regulates the chemo-radiotherapy resistance of cervical squamous cell carcinoma (CSCC) by targeting beta-catenin through mRNA demethylation. Mol. Carcinog. 57(5), 590–597 (2018).
    • 111. Rose NR, McDonough MA, King ONF, Kawamura A, Schofield CJ. Inhibition of 2-oxoglutarate dependent oxygenases. Chem. Soc. Rev. 40(8), 4364–4397 (2011).
    • 112. Rodrigues AS, Pérez-Gregorio MR, García-Falcón MS, Simal-Gándara J, Almeida DPF. Effect of meteorological conditions on antioxidant flavonoids in Portuguese cultivars of white and red onions. Food Chem. 124(1), 303–308 (2011).
    • 113. Kobori M, Masumoto S, Akimoto Y, Takahashi Y. Dietary quercetin alleviates diabetic symptoms and reduces streptozotocin-induced disturbance of hepatic gene expression in mice. Mol. Nutr. Food Res. 53(7), 859–868 (2009).
    • 114. Sharma B, Balomajumder C, Roy P. Hypoglycemic and hypolipidemic effects of flavonoid rich extract from Eugenia jambolana seeds on streptozotocin induced diabetic rats. Food Chem. Toxicol. 46(7), 2376–2383 (2008).
    • 115. Majumdar D, Jung KH, Zhang H et al. Luteolin nanoparticle in chemoprevention: in vitro and in vivo anticancer activity. Cancer Prev. Res. (Phila.) 7(1), 65–73 (2014).
    • 116. Grazul M, Budzisz E. Biological activity of metal ions complexes of chromones, coumarins and flavones. Coord. Chem. Rev. 253(21–22), 2588–2598 (2009).
    • 117. Kostyuk VA, Potapovich AI, Strigunova EN, Kostyuk TV, Afanas'ev IB. Experimental evidence that flavonoid metal complexes may act as mimics of superoxide dismutase. Arch. Biochem. Biophys. 428(2), 204–208 (2004).
    • 118. Ni Y, Du S, Kokot S. Interaction between quercetin-copper(II) complex and DNA with the use of the neutral red dye fluorophor probe. Anal. Chim. Acta 584(1), 19–27 (2007).
    • 119. Shisheva A, Shechter Y. Quercetin selectively inhibits insulin receptor function in vitro and the bioresponses of insulin and insulinomimetic agents in rat adipocytes. Biochemistry 31(34), 8059–8063 (1992).
    • 120. Hollander PA, Elbein SC, Hirsch IB et al. Role of orlistat in the treatment of obese patients with type 2 diabetes: a 1-year randomized double-blind study. Diabetes Care 21(8), 1288–1294 (1998).
    • 121. Mohammed A, Al-Numair KS, Balakrishnan A. Docking studies on the interaction of flavonoids with fat mass and obesity associated protein. Pak. J. Pharm. Sci. 28(5), 1647–1653 (2015).
    • 122. Zhou P, Wu M, Ye C, Xu Q, Wang L. Meclofenamic acid promotes cisplatin-induced acute kidney injury by inhibiting fat mass and obesity-associated protein-mediated m(6)A abrogation in RNA. J. Biol. Chem. 294(45), 16908–16917 (2019).