AI Chat Paper
Note: Please note that the following content is generated by AMiner AI. SciOpen does not take any responsibility related to this content.
{{lang === 'zh_CN' ? '文章概述' : 'Summary'}}
{{lang === 'en_US' ? '中' : 'Eng'}}
Chat more with AI
PDF (1.4 MB)
Collect
Submit Manuscript AI Chat Paper
Show Outline
Outline
Show full outline
Hide outline
Outline
Show full outline
Hide outline
Review Article | Open Access

Studies on the fat mass and obesity-associated (FTO) gene and its impact on obesity-associated diseases

Chaoqun Huanga,b,c,d,Wei Chena,b,c,dXinxia Wanga,b,c,d( )
College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
Key Laboratory of Molecular Animal Nutrition, Zhejiang University, Ministry of Education, Hangzhou, Zhejiang 310058, China
Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, Zhejiang 310058, China
Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou, Zhejiang 310058, China

Peer review under responsibility of Chongqing Medical University.

Show Author Information

Abstract

Obesity has become a major health crisis in the past ~50 years. The fat mass and obesity-associated (FTO) gene, identified by genome-wide association studies (GWAS), was first reported to be positively associated with obesity in humans. Mice with more copies of the FTO gene were observed to be obese, while loss of the gene in mice was found to protect from obesity. Later, FTO was found to encode an m6A RNA demethylase and has a profound effect on many biological and metabolic processes. In this review, we first summarize recent studies that demonstrate the critical roles and regulatory mechanisms of FTO in obesity and metabolic disease. Second, we discuss the ongoing debates concerning the association between FTO polymorphisms and obesity. Third, since several small molecule drugs and micronutrients have been found to regulate metabolic homeostasis through controlling the expression or activity of FTO, we highlight the broad potential of targeting FTO for obesity treatment. Improving our understanding of FTO and the underlying mechanisms may provide new approaches for treating obesity and metabolic diseases.

References

1

Blüher M. Obesity: global epidemiology and pathogenesis. Nat Rev Endocrinol. 2019;15(5):288–298.

2

Lempradl A, Pospisilik JA, Penninger JM. Exploring the emerging complexity in transcriptional regulation of energy homeostasis. Nat Rev Genet. 2015;16(11):665–681.

3

Sturm R, An R. Obesity and economic environments. CA Cancer J Clin. 2014;64(5):337–350.

4

Swinburn BA, Kraak VI, Allender S, et al. The global syndemic of obesity, undernutrition, and climate change: the lancet commission report. Lancet. 2019;393(10173):791–846.

5

González-Muniesa P, Mártinez-González MA, Hu FB, et al. Obesity. Nat Rev Dis Prim. 2017;3:17034.

6

Gadde KM, Martin CK, Berthoud HR, Heymsfield SB. Obesity: pathophysiology and management. J Am Coll Cardiol. 2018;71(1):69–84.

7

Heymsfield SB, Wadden TA. Mechanisms, pathophysiology, and management of obesity. N Engl J Med. 2017;376(3):254–266.

8

Wang HN, Xiang JZ, Qi Z, Du M. Plant extracts in prevention of obesity. Crit Rev Food Sci Nutr. 2022;62(8):2221–2234.

9

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. 2007;3(7):e115.

10

Hinney A, Nguyen TT, Scherag A, et al. Genome wide association (GWA) study for early onset extreme obesity supports the role of fat mass and obesity associated gene (FTO) variants. PLoS One. 2007;2(12):e1361.

11

Frayling TM, Timpson NJ, Weedon MN, et al. A common variant in the FTO gene is associated with body mass index and predisposes to childhood and adult obesity. Science. 2007;316(5826):889–894.

12

Dina C, Meyre D, Gallina S, et al. Variation in FTO contributes to childhood obesity and severe adult obesity. Nat Genet. 2007;39(6):724–726.

13

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. 2011;7(12):885–887.

14

Zhao X, Yang Y, Sun BF, et al. FTO-dependent demethylation of N6-methyladenosine regulates mRNA splicing and is required for adipogenesis. Cell Res. 2014;24(12):1403–1419.

15

Tung YCL, Yeo GSH, O’Rahilly S, Coll AP. Obesity and FTO: changing focus at a complex locus. Cell Metab. 2014;20(5):710–718.

16

Speakman JR. The ‘fat mass and obesity related’ (FTO) gene: mechanisms of impact on obesity and energy balance. Curr Obes Rep. 2015;4(1):73–91.

17

Church C, Moir L, McMurray F, et al. Overexpression of Fto leads to increased food intake and results in obesity. Nat Genet. 2010;42(12):1086–1092.

18

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. 2010;5(11):e14005.

19

Fischer J, Koch L, Emmerling C, et al. Inactivation of the Fto gene protects from obesity. Nature. 2009;458(7240):894–898.

20

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.

21

Meyer KD, Saletore Y, Zumbo P, Elemento O, Mason CE, Jaffrey SR. Comprehensive analysis of mRNA methylation reveals enrichment in 3′ UTRs and near stop codons. Cell. 2012;149(7):1635–1646.

22

McTaggart JS, Lee S, Iberl M, Church C, Cox RD, Ashcroft FM. FTO is expressed in neurones throughout the brain and its expression is unaltered by fasting. PLoS One. 2011;6(11):e27968.

23

Wang DD, Hu FB. Precision nutrition for prevention and management of type 2 diabetes. Lancet Diabetes Endocrinol. 2018;6(5):416–426.

24

Ling C, Rönn T. Epigenetics in human obesity and type 2 diabetes. Cell Metab. 2019;29(5):1028–1044.

25

Wu R, Wang X. Epigenetic regulation of adipose tissue expansion and adipogenesis by N6-methyladenosine. Obes Rev. 2021;22(2):e13124.

26

Peters T, Ausmeier K, Dildrop R, Rüther U. The mouse Fused toes (Ft) mutation is the result of a 1.6-Mb deletion including the entire Iroquois B gene cluster. Mamm Genome. 2002;13(4):186–188.

27

van der Hoeven F, Schimmang T, Volkmann A, Mattei MG, Kyewski B, Rüther U. Programmed cell death is affected in the novel mouse mutant Fused toes (Ft). Development. 1994;120(9):2601–2607.

28

Lesche R, Peetz A, van der Hoeven F, Rüther U. Ft1, a novel gene related to ubiquitin-conjugating enzymes, is deleted in the Fused toes mouse mutation. Mamm Genome. 1997;8(12):879–883.

29

Peters T, Ausmeier K, Rüther U. Cloning of Fatso (Fto), a novel gene deleted by the Fused toes (Ft) mouse mutation. Mamm Genome. 1999;10(10):983–986.

30

Delous M, Baala L, Salomon R, et al. The ciliary gene RPGRIP1L is mutated in cerebello-oculo-renal syndrome (Joubert syndrome type B) and Meckel syndrome. Nat Genet. 2007;39(7):875–881.

31

Trewick SC, Henshaw TF, Hausinger RP, Lindahl T, Sedgwick B. Oxidative demethylation by Escherichia coli AlkB directly reverts DNA base damage. Nature. 2002;419(6903):174–178.

32

Falnes PØ, Johansen RF, Seeberg E. AlkB-mediated oxidative demethylation reverses DNA damage in Escherichia coli. Nature. 2002;419(6903):178–182.

33

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. 2008;359(24):2558–2566.

34

Speakman JR. FTO effect on energy demand versus food intake. Nature. 2010;464(7289):E1. discussion E2.

35

Tews D, Fischer-Posovszky P, Wabitsch M. FTO – friend or foe? Horm Metab Res. 2010;42(2):75–80.

36

Fredriksson R, Hägglund M, Olszewski PK, et al. The obesity gene, FTO, is of ancient origin, up-regulated during food deprivation and expressed in neurons of feeding-related nuclei of the brain. Endocrinology. 2008;149(5):2062–2071.

37

Robbens S, Rouzé P, Cock JM, Spring J, Worden AZ, Van de Peer Y. The FTO gene, implicated in human obesity, is found only in vertebrates and marine algae. J Mol Evol. 2008;66(1):80–84.

38

Zhang GW, Gao L, Chen SY, et al. Single nucleotide polymorphisms in the FTO gene and their association with growth and meat quality traits in rabbits. Gene. 2013;527(2):553–557.

39

Fan B, Du ZQ, Rothschild MF. The fat mass and obesity-associated (FTO) gene is associated with intramuscular fat content and growth rate in the pig. Anim Biotechnol. 2009;20(2):58–70.

40

Fontanesi L, Scotti E, Buttazzoni L, Davoli R, Russo V. The porcine fat mass and obesity associated (FTO) gene is associated with fat deposition in Italian Duroc pigs. Anim Genet. 2009;40(1):90–93.

41

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. 2007;8:23.

42

Sebert S, Salonurmi T, Keinänen-Kiukaanniemi S, et al. Programming effects of FTO in the development of obesity. Acta Physiol. 2014;210(1):58–69.

43

Han Z, Niu T, Chang J, et al. Crystal structure of the FTO protein reveals basis for its substrate specificity. Nature. 2010;464(7292):1205–1209.

44

Xiao MZ, Liu JM, Xian CL, Chen KY, Liu ZQ, Cheng YY. Therapeutic potential of ALKB homologs for cardiovascular disease. Biomed Pharmacother. 2020;131:110645.

45

Zhang X, Wei LH, Wang Y, et al. Structural insights into FTO’s catalytic mechanism for the demethylation of multiple RNA substrates. Proc Natl Acad Sci U S A. 2019;116(8):2919–2924.

46

Bayoumi M, Munir M. Structural insights into m6A-erasers: a step toward understanding molecule specificity and potential antiviral targeting. Front Cell Dev Biol. 2021;8:587108.

47

Chang JY, Park JH, Park SE, Shon J, Park YJ. The fat mass- and obesity-associated (FTO) gene to obesity: lessons from mouse models. Obesity. 2018;26(11):1674–1686.

48

Boissel S, Reish O, Proulx K, et al. Loss-of-function mutation in the dioxygenase-encoding FTO gene causes severe growth retardation and multiple malformations. Am J Hum Genet. 2009;85(1):106–111.

49

Aas A, Isakson P, Bindesbøll C, Alemu EA, Klungland A, Simonsen A. Nucleocytoplasmic shuttling of FTO does not affect starvation-induced autophagy. PLoS One. 2017;12(3):e0168182.

50

Gulati P, Avezov E, Ma M, et al. Fat mass and obesity-related (FTO) shuttles between the nucleus and cytoplasm. Biosci Rep. 2014;34(5):e00144.

51

Wei J, Liu F, Lu Z, et al. Differential m6A, m6Am, and m1A demethylation mediated by FTO in the cell nucleus and cytoplasm. Mol Cell. 2018;71(6):973–985.

52

Ferenc K, Pilžys T, Garbicz D, et al. Intracellular and tissue specific expression of FTO protein in pig: changes with age, energy intake and metabolic status. Sci Rep. 2020;10(1):13029.

53

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. 2015;10(7):e0133788.

54

Merkestein M, Laber S, McMurray F, et al. FTO influences adipogenesis by regulating mitotic clonal expansion. Nat Commun. 2015;6:6792.

55

Chen X, Luo Y, Jia G, Liu G, Zhao H, Huang Z. FTO promotes adipogenesis through inhibition of the Wnt/β-catenin signaling pathway in porcine intramuscular preadipocytes. Anim Biotechnol. 2017;28(4):268–274.

56

Wu R, Guo G, Bi Z, et al. m6A methylation modulates adipogenesis through JAK2-STAT3-C/EBPβ signaling. Biochim Biophys Acta Gene Regul Mech. 2019;1862(8):796–806.

57

Wu R, Liu Y, Yao Y, et al. FTO regulates adipogenesis by controlling cell cycle progression via m6A-YTHDF2 dependent mechanism. Biochim Biophys Acta Mol Cell Biol Lipids. 2018;1863(10):1323–1330.

58

Chen X, Zhou B, Luo Y, et al. Tissue distribution of porcine FTO and its effect on porcine intramuscular preadipocytes proliferation and differentiation. PLoS One. 2016;11(3):e0151056.

59

Wang X, Zhu L, Chen J, Wang Y. mRNA m6 A methylation downregulates adipogenesis in porcine adipocytes. Biochem Biophys Res Commun. 2015;459(2):201–207.

60

Church C, Lee S, Bagg EAL, et al. A mouse model for the metabolic effects of the human fat mass and obesity associated FTO gene. PLoS Genet. 2009;5(8):e1000599.

61

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. 2013;9(1):e1003166.

62

Wang X, Sun B, Jiang Q, et al. mRNA m6A plays opposite role in regulating UCP2 and PNPLA2 protein expression in adipocytes. Int J Obes. 2018;42(11):1912–1924.

63

Bird A. Perceptions of epigenetics. Nature. 2007;447(7143):396–398.

64

Bi Z, Liu Y, Zhao Y, et al. A dynamic reversible RNA N6-methyladenosine modification: current status and perspectives. J Cell Physiol. 2019;234(6):7948–7956.

65

Lee JE, Schmidt H, Lai B, Ge K. Transcriptional and epigenomic regulation of adipogenesis. Mol Cell Biol. 2019;39(11):e00601–18.

66

Wang X, Wang Y. From histones to RNA: role of methylation in signal proteins involved in adipogenesis. Curr Protein Pept Sci. 2017;18(6):589–598.

67

Dominissini D, Moshitch-Moshkovitz S, Schwartz S, et al. Topology of the human and mouse m6A RNA methylomes revealed by m6A-seq. Nature. 2012;485(7397):201–206.

68

Li X, Kim JW, Grønborg M, Urlaub H, Lane MD, Tang QQ. Role of cdk2 in the sequential phosphorylation/activation of C/EBPbeta during adipocyte differentiation. Proc Natl Acad Sci U S A. 2007;104(28):11597–11602.

69

Hochegger H, Takeda S, Hunt T. Cyclin-dependent kinases and cell-cycle transitions: does one fit all? Nat Rev Mol Cell Biol. 2008;9(11):910–916.

70

Jiang Q, Sun B, Liu Q, et al. MTCH2 promotes adipogenesis in intramuscular preadipocytes via an m6A-YTHDF1-dependent mechanism. FASEB J. 2019;33(2):2971–2981.

71

Lee Y, Choe J, Park OH, Kim YK. Molecular mechanisms driving mRNA degradation by m6A modification. Trends Genet. 2020;36(3):177–188.

72

Zhao YL, Liu YH, Wu RF, et al. Understanding m6A function through uncovering the diversity roles of YTH domain-containing proteins. Mol Biotechnol. 2019;61(5):355–364.

73

Cai M, Liu Q, Jiang Q, Wu R, Wang X, Wang Y. Loss of m6A on FAM134B promotes adipogenesis in porcine adipocytes through m6A-YTHDF2-dependent way. IUBMB Life. 2019;71(5):580–586.

74

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

75

Wang X, Wu R, Liu Y, et al. m6A mRNA methylation controls autophagy and adipogenesis by targeting Atg5 and Atg7. Autophagy. 2020;16(7):1221–1235.

76

Ho AJ, Stein JL, Hua X, et al. A commonly carried allele of the obesity-related FTO gene is associated with reduced brain volume in the healthy elderly. Proc Natl Acad Sci U S A. 2010;107(18):8404–8409.

77

Melhorn SJ, Askren MK, Chung WK, et al. FTO genotype impacts food intake and corticolimbic activation. Am J Clin Nutr. 2018;107(2):145–154.

78

Ranzenhofer LM, Mayer LES, Davis HA, et al. The FTO gene and measured food intake in 5- to 10-year-old children without obesity. Obesity. 2019;27(6):1023–1029.

79

Abdella HM, El Farssi HO, Broom DR, Hadden DA, Dalton CF. Eating behaviours and food cravings; influence of age, sex, BMI and FTO genotype. Nutrients. 2019;11(2):377.

80

Tung YC, Ayuso E, Shan X, et al. Hypothalamic-specific manipulation of Fto, the ortholog of the human obesity gene FTO, affects food intake in rats. PLoS One. 2010;5(1):e8771.

81

Hess ME, Hess S, Meyer KD, et al. The fat mass and obesity associated gene (Fto) regulates activity of the dopaminergic midbrain circuitry. Nat Neurosci. 2013;16(8):1042–1048.

82

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

83

Wu R, Chen Y, Liu Y, et al. m6A methylation promotes white-to-beige fat transition by facilitating Hif1a translation. EMBO Rep. 2021;22(11):e52348.

84

Graff M, Ngwa JS, Workalemahu T, et al. Genome-wide analysis of BMI in adolescents and young adults reveals additional insight into the effects of genetic loci over the life course. Hum Mol Genet. 2013;22(17):3597–3607.

85

Bradfield JP, Taal HR, Timpson NJ, et al. A genome-wide association meta-analysis identifies new childhood obesity loci. Nat Genet. 2012;44(5):526–531.

86

Meyre D, Delplanque J, Chèvre JC, et al. Genome-wide association study for early-onset and morbid adult obesity identifies three new risk loci in European populations. Nat Genet. 2009;41(2):157–159.

87

Scherag A, Dina C, Hinney A, et al. Two new Loci for body-weight regulation identified in a joint analysis of genome-wide association studies for early-onset extreme obesity in French and German study groups. PLoS Genet. 2010;6(4):e1000916.

88

Wheeler E, Huang N, Bochukova EG, et al. Genome-wide SNP and CNV analysis identifies common and low-frequency variants associated with severe early-onset obesity. Nat Genet. 2013;45(5):513–517.

89

Willer CJ, Speliotes EK, Loos RJF, et al. Six new loci associated with body mass index highlight a neuronal influence on body weight regulation. Nat Genet. 2009;41(1):25–34.

90

Yajnik CS, Janipalli CS, Bhaskar S, et al. FTO gene variants are strongly associated with type 2 diabetes in South Asian Indians. Diabetologia. 2009;52(2):247–252.

91

Ramya K, Radha V, Ghosh S, Majumder PP, Mohan V. Genetic variations in the FTO gene are associated with type 2 diabetes and obesity in south Indians (CURES-79). Diabetes Technol Ther. 2011;13(1):33–42.

92

Vasan SK, Fall T, Neville MJ, et al. Associations of variants in FTO and near MC4R with obesity traits in South Asian Indians. Obesity. 2012;20(11):2268–2277.

93

Li H, Kilpeläinen TO, Liu C, et al. Association of genetic variation in FTO with risk of obesity and type 2 diabetes with data from 96,551 East and South Asians. Diabetologia. 2012;55(4):981–995.

94

Monda KL, Chen GK, Taylor KC, et al. A meta-analysis identifies new loci associated with body mass index in individuals of African ancestry. Nat Genet. 2013;45(6):690–696.

95

Nock NL, Plummer SJ, Thompson CL, Casey G, Li L. FTO polymorphisms are associated with adult body mass index (BMI) and colorectal adenomas in African-Americans. Carcinogenesis. 2011;32(5):748–756.

96

Xiang L, Wu H, Pan A, et al. FTO genotype and weight loss in diet and lifestyle interventions: a systematic review and meta-analysis. Am J Clin Nutr. 2016;103(4):1162–1170.

97

Yang Q, Xiao T, Guo J, Su Z. Complex relationship between obesity and the fat mass and obesity locus. Int J Biol Sci. 2017;13(5):615–629.

98

Sonestedt E, Roos C, Gullberg B, Ericson U, Wirfält E, Orho-Melander M. Fat and carbohydrate intake modify the association between genetic variation in the FTO genotype and obesity. Am J Clin Nutr. 2009;90(5):1418–1425.

99

Scott RA, Bailey MES, Moran CN, et al. FTO genotype and adiposity in children: physical activity levels influence the effect of the risk genotype in adolescent males. Eur J Hum Genet. 2010;18(12):1339–1343.

100

Young AI, Wauthier F, Donnelly P. Multiple novel gene-by-environment interactions modify the effect of FTO variants on body mass index. Nat Commun. 2016;7:12724.

101

Kilpeläinen TO, Qi L, Brage S, et al. Physical activity attenuates the influence of FTO variants on obesity risk: a meta-analysis of 218,166 adults and 19,268 children. PLoS Med. 2011;8(11):e1001116.

102

Ruiz JR, Labayen I, Ortega FB, et al. Attenuation of the effect of the FTO rs9939609 polymorphism on total and central body fat by physical activity in adolescents: the HELENA study. Arch Pediatr Adolesc Med. 2010;164(4):328–333.

103

Cho HW, Jin HS, Eom YB. The interaction between FTO rs9939609 and physical activity is associated with a 2-fold reduction in the risk of obesity in Korean population. Am J Hum Biol. 2021;33(3):e23489.

104

Graff M, Scott RA, Justice AE, et al. Genome-wide physical activity interactions in adiposity – a meta-analysis of 200,452 adults. PLoS Genet. 2017;13(4):e1006528.

105

Reddon H, Gerstein HC, Engert JC, et al. Physical activity and genetic predisposition to obesity in a multiethnic longitudinal study. Sci Rep. 2016;6:18672.

106

Cauchi S, Stutzmann F, Cavalcanti-Proença C, et al. Combined effects of MC4R and FTO common genetic variants on obesity in European general populations. J Mol Med. 2009;87(5):537–546.

107

Andreasen CH, Stender-Petersen KL, Mogensen MS, et al. Low physical activity accentuates the effect of the FTO rs9939609 polymorphism on body fat accumulation. Diabetes. 2008;57(1):95–101.

108

Hall KD, Guo J. Obesity energetics: body weight regulation and the effects of diet composition. Gastroenterology. 2017;152(7):1718–1727.

109

Puhl RM, Peterson JL, DePierre JA, Luedicke J. Headless, hungry, and unhealthy: a video content analysis of obese persons portrayed in online news. J Health Commun. 2013;18(6):686–702.

110

Dulloo AG, Miles-Chan J, Schutz Y, Montani JP. Targeting lifestyle energy expenditure in the management of obesity and health: from biology to built environment. Obes Rev. 2018;19(suppl 1):3–7.

111

Qi Q, Downer MK, Kilpeläinen TO, et al. Dietary intake, FTO genetic variants, and adiposity: a combined analysis of over 16,000 children and adolescents. Diabetes. 2015;64(7):2467–2476.

112

Speakman JR, Rance KA, Johnstone AM. Polymorphisms of the FTO gene are associated with variation in energy intake, but not energy expenditure. Obesity. 2008;16(8):1961–1965.

113

Brunkwall L, Ericson U, Hellstrand S, Gullberg B, Orho-Melander M, Sonestedt E. Genetic variation in the fat mass and obesity-associated gene (FTO) in association with food preferences in healthy adults. Food Nutr Res. 2013;57.

114

Cameron JD, Tasca GA, Little J, et al. Effects of fat mass and obesity-associated (FTO) gene polymorphisms on binge eating in women with binge-eating disorder: the moderating influence of attachment style. Nutrition. 2019;61:208–212.

115

Mehrdad M, Doaei S, Gholamalizadeh M, Eftekhari MH. The association between FTO genotype with macronutrients and calorie intake in overweight adults. Lipids Health Dis. 2020;19(1):197.

116

Tanaka T, Ngwa JS, van Rooij FJ, et al. Genome-wide meta-analysis of observational studies shows common genetic variants associated with macronutrient intake. Am J Clin Nutr. 2013;97(6):1395–1402.

117

Chuang YF, Tanaka T, Beason-Held LL, et al. FTO genotype and aging: pleiotropic longitudinal effects on adiposity, brain function, impulsivity and diet. Mol Psychiatry. 2015;20(1):133–139.

118

Saber-Ayad M, Manzoor S, Radwan H, et al. The FTO genetic variants are associated with dietary intake and body mass index amongst Emirati population. PLoS One. 2019;14(10):e0223808.

119

Magno FCCM, Guaraná HC, Fonseca ACP, et al. Influence of FTO rs9939609 polymorphism on appetite, ghrelin, leptin, IL6, TNFα levels, and food intake of women with morbid obesity. Diabetes Metab Syndr Obes. 2018;11:199–207.

120

Benedict C, Axelsson T, Söderberg S, et al. Fat mass and obesity-associated gene (FTO) is linked to higher plasma levels of the hunger hormone ghrelin and lower serum levels of the satiety hormone leptin in older adults. Diabetes. 2014;63(11):3955–3959.

121

Dixon JR, Selvaraj S, Yue F, et al. Topological domains in mammalian genomes identified by analysis of chromatin interactions. Nature. 2012;485(7398):376–380.

122

Hunt LE, Noyvert B, Bhaw-Rosun L, et al. Complete re-sequencing of a 2Mb topological domain encompassing the FTO/IRXB genes identifies a novel obesity-associated region upstream of IRX5. Genome Med. 2015;7:126.

123

Williams CL, Li C, Kida K, et al. MKS and NPHP modules cooperate to establish basal body/transition zone membrane associations and ciliary gate function during ciliogenesis. J Cell Biol. 2011;192(6):1023–1041.

124

Liu L, Zhang M, Xia Z, Xu P, Chen L, Xu T. Caenorhabditis elegans ciliary protein NPHP-8, the homologue of human RPGRIP1L, is required for ciliogenesis and chemosensation. Biochem Biophys Res Commun. 2011;410(3):626–631.

125

Stratigopoulos G, LeDuc CA, Cremona ML, Chung WK, Leibel RL. Cut-like homeobox 1 (CUX1) regulates expression of the fat mass and obesity-associated and retinitis pigmentosa GTPase regulator-interacting protein-1-like (RPGRIP1L) genes and coordinates leptin receptor signaling. J Biol Chem. 2011;286(3):2155–2170.

126

Vierkotten J, Dildrop R, Peters T, Wang B, Rüther U. Ftm is a novel basal body protein of cilia involved in Shh signalling. Development. 2007;134(14):2569–2577.

127

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. 2014;19(5):767–779.

128

Smemo S, Tena JJ, Kim KH, et al. Obesity-associated variants within FTO form long-range functional connections with IRX3. Nature. 2014;507(7492):371–375.

129

Laber S, Forcisi S, Bentley L, et al. Linking the FTO obesity rs1421085 variant circuitry to cellular, metabolic, and organismal phenotypes in vivo. Sci Adv. 2021;7(30):eabg0108.

130

Zhang Z, Chen N, Liu R, et al. The rs1421085 variant within FTO promotes but not inhibits thermogenesis and is potentially associated with human migration. bioRxiv. 2021. https://doi.org/10.1101/2021.08.13.456245.

131

Claussnitzer M, Dankel SN, Kim KH, et al. FTO obesity variant circuitry and adipocyte browning in humans. N Engl J Med. 2015;373(10):895–907.

132

Sobreira DR, Joslin AC, Zhang Q, et al. Extensive pleiotropism and allelic heterogeneity mediate metabolic effects of IRX3 and IRX5. Science. 2021;372(6546):1085–1091.

133

DeFronzo RA, Ferrannini E, Groop L, et al. Type 2 diabetes mellitus. Nat Rev Dis Prim. 2015;1:15019.

134

Zeggini E, Weedon MN, Lindgren CM, et al. Replication of genome-wide association signals in UK samples reveals risk loci for type 2 diabetes. Science. 2007;316(5829):1336–1341.

135

Scott LJ, Mohlke KL, Bonnycastle LL, et al. A genome-wide association study of type 2 diabetes in Finns detects multiple susceptibility variants. Science. 2007;316(5829):1341–1345.

136

Samaras K, Botelho NK, Chisholm DJ, Lord RV. Subcutaneous and visceral adipose tissue FTO gene expression and adiposity, insulin action, glucose metabolism, and inflammatory adipokines in type 2 diabetes mellitus and in health. Obes Surg. 2010;20(1):108–113.

137

Bravard A, Lefai E, Meugnier E, et al. FTO is increased in muscle during type 2 diabetes, and its overexpression in myotubes alters insulin signaling, enhances lipogenesis and ROS production, and induces mitochondrial dysfunction. Diabetes. 2011;60(1):258–268.

138

Bravard A, Veilleux A, Disse E, et al. The expression of FTO in human adipose tissue is influenced by fat depot, adiposity, and insulin sensitivity. Obesity. 2013;21(6):1165–1173.

139

Yang Y, Shen F, Huang W, et al. Glucose is involved in the dynamic regulation of m6A in patients with type 2 diabetes. J Clin Endocrinol Metab. 2019;104(3):665–673.

140

Souness JE, Stouffer JE, de Sanchez VC. Effect of N6-methyladenosine on fat-cell glucose metabolism: evidence for two modes of action. Biochem Pharmacol. 1982;31(24):3961–3971.

141

Shen F, Huang W, Huang JT, et al. Decreased N(6)-methyladenosine in peripheral blood RNA from diabetic patients is associated with FTO expression rather than ALKBH5. J Clin Endocrinol Metab. 2015;100(1):E148–E154.

142

Poritsanos NJ, Lew PS, Mizuno TM. Relationship between blood glucose levels and hepatic Fto mRNA expression in mice. Biochem Biophys Res Commun. 2010;400(4):713–717.

143

Mizuno TM, Lew PS, Luo Y, Leckstrom A. Negative regulation of hepatic fat mass and obesity associated (Fto) gene expression by insulin. Life Sci. 2017;170:50–55.

144

Guo J, Ren W, Li X, et al. Altering of FTO in the serum and livers of NAFLD patients: a correlation analysis. Int J Clin Exp Med. 2018;11(6):6046–6053.

145

Lei KJ, Shelly LL, Pan CJ, Sidbury JB, Chou JY. Mutations in the glucose-6-phosphatase gene that cause glycogen storage disease type 1a. Science. 1993;262(5133):580–583.

146

Menendez JA, Vazquez-Martin A, Ortega FJ, Fernandez-Real JM. Fatty acid synthase: association with insulin resistance, type 2 diabetes, and cancer. Clin Chem. 2009;55(3):425–438.

147

Buhman KK, Chen HC, Farese Jr RV. The enzymes of neutral lipid synthesis. J Biol Chem. 2001;276(44):40369–40372.

148

Li Y, Ma Z, Jiang S, et al. A global perspective on FOXO1 in lipid metabolism and lipid-related diseases. Prog Lipid Res. 2017;66:42–49.

149

Dong XC, Copps KD, Guo S, et al. Inactivation of hepatic Foxo1 by insulin signaling is required for adaptive nutrient homeostasis and endocrine growth regulation. Cell Metab. 2008;8(1):65–76.

150

Chou JY, Jun HS, Mansfield BC. Glycogen storage disease type I and G6Pase-β deficiency: etiology and therapy. Nat Rev Endocrinol. 2010;6(12):676–688.

151

De Jesus DF, Kulkarni RN. Epigenetic modifiers of islet function and mass. Trends Endocrinol Metab. 2014;25(12):628–636.

152

De Jesus DF, Zhang Z, Kahraman S, et al. m6A mRNA methylation regulates human β-cell biology in physiological states and in type 2 diabetes. Nat Metab. 2019;1(8):765–774.

153

Wang Y, Sun J, Lin Z, et al. m6A mRNA methylation controls functional maturation in neonatal murine β-cells. Diabetes. 2020;69(8):1708–1722.

154

Liu J, Luo G, Sun J, et al. METTL14 is essential for β-cell survival and insulin secretion. Biochim Biophys Acta Mol Basis Dis. 2019;1865(9):2138–2148.

155

Men L, Sun J, Luo G, Ren D. Acute deletion of METTL14 in β-cells of adult mice results in glucose intolerance. Endocrinology. 2019;160(10):2388–2394.

156

Russell MA, Morgan NG. Conditional expression of the FTO gene product in rat INS-1 cells reveals its rapid turnover and a role in the profile of glucose-induced insulin secretion. Clin Sci. 2011;120(9):403–413.

157

Fan HQ, He W, Xu KF, Wang ZX, Xu XY, Chen H. FTO inhibits insulin secretion and promotes NF-κB activation through positively regulating ROS production in pancreatic β cells. PLoS One. 2015;10(5):e0127705.

158

Taneera J, Prasad RB, Dhaiban S, et al. Silencing of the FTO gene inhibits insulin secretion: an in vitro study using GRINCH cells. Mol Cell Endocrinol. 2018;472:10–17.

159

Fan JG, Kim SU, Wong VW. New trends on obesity and NAFLD in Asia. J Hepatol. 2017;67(4):862–873.

160

Guo J, Ren W, Li A, et al. Fat mass and obesity-associated gene enhances oxidative stress and lipogenesis in nonalcoholic fatty liver disease. Dig Dis Sci. 2013;58(4):1004–1009.

161

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. 2016;479(3):476–481.

162

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. 2018;119(7):5676–5685.

163

Chen A, Chen X, Cheng S, et al. FTO promotes SREBP1c maturation and enhances CIDEC transcription during lipid accumulation in HepG2 cells. Biochim Biophys Acta Mol Cell Biol Lipids. 2018;1863(5):538–548.

164

Candia R, Riquelme A, Baudrand R, et al. Overexpression of 11β-hydroxysteroid dehydrogenase type 1 in visceral adipose tissue and portal hypercortisolism in non-alcoholic fatty liver disease. Liver Int. 2012;32(3):392–399.

165

Hu Y, Feng Y, Zhang L, et al. GR-mediated FTO transactivation induces lipid accumulation in hepatocytes via demethylation of m6A on lipogenic mRNAs. RNA Biol. 2020;17(7):930–942.

166

Roth GA, Mensah GA, Fuster V. The global burden of cardiovascular diseases and risks: a compass for global action. J Am Coll Cardiol. 2020;76(25):2980–2981.

167

Dorn LE, Lasman L, Chen J, et al. The N6-methyladenosine mRNA methylase METTL3 controls cardiac homeostasis and hypertrophy. Circulation. 2019;139(4):533–545.

168

Mathiyalagan P, Liang Y, Sassi Y, et al. Abstract 12:M6A modification in RNA regulates cardiomyocyte and cardiac function in heart failure. Circ Res. 2017;121(suppl 1):A12.

169

Berulava T, Buchholz E, Elerdashvili V, et al. Changes in m6A RNA methylation contribute to heart failure progression by modulating translation. Eur J Heart Fail. 2020;22(1):54–66.

170

Carnevali L, Graiani G, Rossi S, et al. Signs of cardiac autonomic imbalance and proarrhythmic remodeling in FTO deficient mice. PLoS One. 2014;9(4):e95499.

171

Gustavsson J, Mehlig K, Leander K, et al. FTO genotype, physical activity, and coronary heart disease risk in Swedish men and women. Circ Cardiovasc Genet. 2014;7(2):171–177.

172

Mathiyalagan P, Adamiak M, Mayourian J, et al. FTO-dependent N6-methyladenosine regulates cardiac function during remodeling and repair. Circulation. 2019;139(4):518–532.

173

Shen W, Li H, Su H, Chen K, Yan J. FTO overexpression inhibits apoptosis of hypoxia/reoxygenation-treated myocardial cells by regulating m6A modification of Mhrt. Mol Cell Biochem. 2021;476(5):2171–2179.

174

Lavie CJ, Arena R, Alpert MA, Milani RV, Ventura HO. Management of cardiovascular diseases in patients with obesity. Nat Rev Cardiol. 2018;15(1):45–56.

175

Li X, Yang Y, Chen S, Zhou J, Li J, Cheng Y. Epigenetics-based therapeutics for myocardial fibrosis. Life Sci. 2021;271:119186.

176

Baur JA, Pearson KJ, Price NL, et al. Resveratrol improves health and survival of mice on a high-calorie diet. Nature. 2006;444(7117):337–342.

177

Gan Z, Wei W, Wu J, et al. Resveratrol and curcumin improve intestinal mucosal integrity and decrease m6A RNA methylation in the intestine of weaning piglets. ACS Omega. 2019;4(17):17438–17446.

178

Wu J, Li Y, Yu J, et al. Resveratrol attenuates high-fat diet induced hepatic lipid homeostasis disorder and decreases m6A RNA methylation. Front Pharmacol. 2020;11:568006.

179

Meydani M, Hasan ST. Dietary polyphenols and obesity. Nutrients. 2010;2(7):737–751.

180

Lu N, Li X, Yu J, et al. Curcumin attenuates lipopolysaccharide-induced hepatic lipid metabolism disorder by modification of m6A RNA methylation in piglets. Lipids. 2018;53(1):53–63.

181

Chen Y, Wu R, Chen W, et al. Curcumin prevents obesity by targeting TRAF4-induced ubiquitylation in m6A-dependent manner. EMBO Rep. 2021;22(5):e52146.

182

Patra SK, Rizzi F, Silva A, Rugina DO, Bettuzzi S. Molecular targets of (-)-epigallocatechin-3-gallate (EGCG): specificity and interaction with membrane lipid rafts. J Physiol Pharmacol. 2008;59(suppl 9):217–235.

183

Wu R, Yao Y, Jiang Q, et al. Epigallocatechin gallate targets FTO and inhibits adipogenesis in an mRNA m6A-YTHDF2-dependent manner. Int J Obes. 2018;42(7):1378–1388.

184

Wei CM, Gershowitz A, Moss B. Methylated nucleotides block 5′ terminus of HeLa cell messenger RNA. Cell. 1975;4(4):379–386.

185

Wei CM, Gershowitz A, Moss B. 5′-Terminal and internal methylated nucleotide sequences in HeLa cell mRNA. Biochemistry. 1976;15(2):397–401.

186

Lee M, Kim B, Kim VN. Emerging roles of RNA modification: m(6)A and U-tail. Cell. 2014;158(5):980–987.

187

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.

188

Zheng G, Dahl JA, Niu Y, et al. ALKBH5 is a mammalian RNA demethylase that impacts RNA metabolism and mouse fertility. Mol Cell. 2013;49(1):18–29.

189

Boles NC, Temple S. Epimetronomics: m6A marks the tempo of corticogenesis. Neuron. 2017;96(4):718–720.

190

Yoon KJ, Ringeling FR, Vissers C, et al. Temporal control of mammalian cortical neurogenesis by m6A methylation. Cell. 2017;171(4):877–889.

191

Su R, Dong L, Li Y, et al. Targeting FTO suppresses cancer stem cell maintenance and immune evasion. Cancer Cell. 2020;38(1):79–96.

192

Huang Y, Su R, Sheng Y, et al. Small-molecule targeting of oncogenic FTO demethylase in acute myeloid leukemia. Cancer Cell. 2019;35(4):677–691.

193

Liu Y, Liang G, Xu H, et al. Tumors exploit FTO-mediated regulation of glycolytic metabolism to evade immune surveillance. Cell Metab. 2021;33(6):1221–1233.

194

Deng X, Su R, Stanford S, Chen J. Critical enzymatic functions of FTO in obesity and cancer. Front Endocrinol. 2018;9:396.

195

Peng S, Xiao W, Ju D, et al. Identification of entacapone as a chemical inhibitor of FTO mediating metabolic regulation through FOXO1. Sci Transl Med. 2019;11(488):eaau7116.

196

Wang X, Huang N, Yang M, et al. FTO is required for myogenesis by positively regulating mTOR-PGC-1α pathway-mediated mitochondria biogenesis. Cell Death Dis. 2017;8(3):e2702.

Genes & Diseases
Pages 2351-2365
Cite this article:
Huang C, Chen W, Wang X. Studies on the fat mass and obesity-associated (FTO) gene and its impact on obesity-associated diseases. Genes & Diseases, 2023, 10(6): 2351-2365. https://doi.org/10.1016/j.gendis.2022.04.014

263

Views

3

Downloads

10

Crossref

7

Web of Science

10

Scopus

0

CSCD

Altmetrics

Received: 19 December 2021
Revised: 29 March 2022
Accepted: 01 April 2022
Published: 06 May 2022
© 2022 The Authors.

This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

Return