#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Adult Onset Global Loss of the Gene Alters Body Composition and Metabolism in the Mouse


The strongest BMI–associated GWAS locus in humans is the FTO gene. Rodent studies demonstrate a role for FTO in energy homeostasis and body composition. The phenotypes observed in loss of expression studies are complex with perinatal lethality, stunted growth from weaning, and significant alterations in body composition. Thus understanding how and where Fto regulates food intake, energy expenditure, and body composition is a challenge. To address this we generated a series of mice with distinct temporal and spatial loss of Fto expression. Global germline loss of Fto resulted in high perinatal lethality and a reduction in body length, fat mass, and lean mass. When ratio corrected for lean mass, mice had a significant increase in energy expenditure, but more appropriate multiple linear regression normalisation showed no difference in energy expenditure. Global deletion of Fto after the in utero and perinatal period, at 6 weeks of age, removed the high lethality of germline loss. However, there was a reduction in weight by 9 weeks, primarily as loss of lean mass. Over the subsequent 10 weeks, weight converged, driven by an increase in fat mass. There was a switch to a lower RER with no overall change in food intake or energy expenditure. To test if the phenotype can be explained by loss of Fto in the mediobasal hypothalamus, we sterotactically injected adeno-associated viral vectors encoding Cre recombinase to cause regional deletion. We observed a small reduction in food intake and weight gain with no effect on energy expenditure or body composition. Thus, although hypothalamic Fto can impact feeding, the effect of loss of Fto on body composition is brought about by its actions at sites elsewhere. Our data suggest that Fto may have a critical role in the control of lean mass, independent of its effect on food intake.


Vyšlo v časopise: Adult Onset Global Loss of the Gene Alters Body Composition and Metabolism in the Mouse. PLoS Genet 9(1): e32767. doi:10.1371/journal.pgen.1003166
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1003166

Souhrn

The strongest BMI–associated GWAS locus in humans is the FTO gene. Rodent studies demonstrate a role for FTO in energy homeostasis and body composition. The phenotypes observed in loss of expression studies are complex with perinatal lethality, stunted growth from weaning, and significant alterations in body composition. Thus understanding how and where Fto regulates food intake, energy expenditure, and body composition is a challenge. To address this we generated a series of mice with distinct temporal and spatial loss of Fto expression. Global germline loss of Fto resulted in high perinatal lethality and a reduction in body length, fat mass, and lean mass. When ratio corrected for lean mass, mice had a significant increase in energy expenditure, but more appropriate multiple linear regression normalisation showed no difference in energy expenditure. Global deletion of Fto after the in utero and perinatal period, at 6 weeks of age, removed the high lethality of germline loss. However, there was a reduction in weight by 9 weeks, primarily as loss of lean mass. Over the subsequent 10 weeks, weight converged, driven by an increase in fat mass. There was a switch to a lower RER with no overall change in food intake or energy expenditure. To test if the phenotype can be explained by loss of Fto in the mediobasal hypothalamus, we sterotactically injected adeno-associated viral vectors encoding Cre recombinase to cause regional deletion. We observed a small reduction in food intake and weight gain with no effect on energy expenditure or body composition. Thus, although hypothalamic Fto can impact feeding, the effect of loss of Fto on body composition is brought about by its actions at sites elsewhere. Our data suggest that Fto may have a critical role in the control of lean mass, independent of its effect on food intake.


Zdroje

1. GerkenT, GirardCA, TungYCL, WebbyCJ, SaudekV, et al. (2007) The obesity-associated FTO gene encodes a 2-oxoglutarate-dependent nucleic acid demethylase. Science 318: 1469–1472.

2. HanZF, NiuTH, ChangJB, LeiXG, ZhaoMY, et al. (2010) Crystal structure of the FTO protein reveals basis for its substrate specificity. Nature 464: 1205–U1129.

3. JiaGF, FuY, ZhaoX, DaiQ, ZhengGQ, et al. (2011) N6-Methyladenosine in nuclear RNA is a major substrate of the obesity-associated FTO. Nature Chemical Biology 7: 885–887.

4. ScottLJ, MohlkeKL, BonnycastleLL, WillerCJ, LiY, et al. (2007) A genome-wide association study of type 2 diabetes in Finns detects multiple susceptibility variants. Science 316: 1341–1345.

5. FraylingTM, TimpsonNJ, WeedonMN, ZegginiE, FreathyRM, et al. (2007) A common variant in the FTO gene is associated with body mass index and predisposes to childhood and adult obesity. Science 316: 889–894.

6. DinaC, MeyreD, GallinaS, DurandE, KornerA, et al. (2007) Variation in FTO contributes to childhood obesity and severe adult obesity. Nature genetics 39: 724–726.

7. ScuteriA, SannaS, ChenWM, UdaM, AlbaiG, et al. (2007) Genome-wide association scan shows genetic variants in the FTO gene are associated with obesity-related traits. PLoS Genet 3: e115 doi:10.1371/journal.pgen.0030115.

8. FawcettKA, BarrosoI (2010) The genetics of obesity: FTO leads the way. Trends in genetics : TIG 26: 266–274.

9. TungYC, YeoGS (2011) From GWAS to biology: lessons from FTO. Annals of the New York Academy of Sciences 1220: 162–171.

10. SovioU, Mook-KanamoriDO, WarringtonNM, LawrenceR, BriollaisL, et al. (2011) Association between Common Variation at the FTO Locus and Changes in Body Mass Index from Infancy to Late Childhood: The Complex Nature of Genetic Association through Growth and Development. PLoS Genet 7: e1001307 doi:10.1371/journal.pgen.1001307.

11. FraylingTM, OngK (2011) Piecing together the FTO jigsaw. Genome biology 12: 104.

12. HardyR, WillsAK, WongA, ElksCE, WarehamNJ, et al. (2010) Life course variations in the associations between FTO and MC4R gene variants and body size. Human Molecular Genetics 19: 545–552.

13. BoisselS, ReishO, ProulxK, Kawagoe-TakakiH, SedgwickB, et al. (2009) Loss-of-function mutation in the dioxygenase-encoding FTO gene causes severe growth retardation and multiple malformations. American journal of human genetics 85: 106–111.

14. MeyreD, ProulxK, Kawagoe-TakakiH, VatinV, Gutierrez-AguilarR, et al. (2010) Prevalence of Loss-of-Function FTO Mutations in Lean and Obese Individuals. Diabetes 59: 311–318.

15. TimpsonNJ, EmmettPM, FraylingTM, RogersI, HattersleyAT, et al. (2008) The fat mass- and obesity-associated locus and dietary intake in children. American Journal of Clinical Nutrition 88: 971–978.

16. CecilJE, TavendaleR, WattP, HetheringtonMM, PalmerCN (2008) An obesity-associated FTO gene variant and increased energy intake in children. The New England journal of medicine 359: 2558–2566.

17. Tanofsky-KraffM, HanJC, AnandalingamK, ShomakerLB, ColumboKM, et al. (2009) The FTO gene rs9939609 obesity-risk allele and loss of control over eating. American Journal of Clinical Nutrition 90: 1483–1488.

18. WardleJ, CarnellS, HaworthCMA, FarooqiIS, O'RahillyS, et al. (2008) Obesity associated genetic variation in FTO is associated with diminished satiety. Journal of Clinical Endocrinology & Metabolism 93: 3640–3643.

19. den HoedM, Westerterp-PlantengaMS, BouwmanFG, MarimanECM, WesterterpKR (2009) Postprandial responses in hunger and satiety are associated with the rs9939609 single nucleotide polymorphism in FTO. American Journal of Clinical Nutrition 90: 1426–1432.

20. SpeakmanJR, RanceKA, JohnstoneAM (2008) Polymorphisms of the FTO gene are associated with variation in energy intake, but not energy expenditure. Obesity 16: 1961–1965.

21. HauptA, ThamerC, StaigerH, TschritterO, KirchhoffK, et al. (2009) Variation in the FTO Gene Influences Food Intake but not Energy Expenditure. Experimental and Clinical Endocrinology & Diabetes 117: 194–197.

22. WardleJ, LlewellynC, SandersonS, PlominR (2009) The FTO gene and measured food intake in children. International Journal of Obesity 33: 42–45.

23. SonestedtE, RoosC, GullbergB, EricsonU, WirfaltE, et al. (2009) Fat and carbohydrate intake modify the association between genetic variation in the FTO genotype and obesity. American Journal of Clinical Nutrition 90: 1418–1425.

24. LappalainenT, LindstromJ, PaananenJ, ErikssonJG, KarhunenL, et al. (2012) Association of the fat mass and obesity-associated (FTO) gene variant (rs9939609) with dietary intake in the Finnish Diabetes Prevention Study. The British journal of nutrition 1–7 DOI: http://dx.doi.org/10.1017/S0007114511007410, Published online: 23 January 2012

25. AhmadT, LeeIM, PareG, ChasmanDI, RoseL, et al. (2011) Lifestyle interaction with fat mass and obesity-associated (FTO) genotype and risk of obesity in apparently healthy U.S. women. Diabetes care 34: 675–680.

26. FischerJ, KochL, EmmerlingC, VierkottenJ, PetersT, et al. (2009) Inactivation of the Fto gene protects from obesity. Nature 458: 894–U810.

27. GaoX, ShinYH, LiM, WangF, TongQA, et al. (2010) The Fat Mass and Obesity Associated Gene FTO Functions in the Brain to Regulate Postnatal Growth in Mice. PLoS ONE 5: e14005 doi:10.1371/journal.pone.0014005.

28. SpeakmanJR (2010) FTO effect on energy demand versus food intake. Nature 464: E1; discussion E2.

29. TschopMH, SpeakmanJR, ArchJRS, AuwerxJ, BruningJC, et al. (2012) A guide to analysis of mouse energy metabolism. Nature Methods 9: 57–63.

30. KaiyalaKJ, SchwartzMW (2011) Toward a more complete (and less controversial) understanding of energy expenditure and its role in obesity pathogenesis. Diabetes 60: 17–23.

31. ChurchC, LeeS, BaggEAL, McTaggartJS, DeaconR, et al. (2009) A Mouse Model for the Metabolic Effects of the Human Fat Mass and Obesity Associated FTO Gene. PLoS Genet 5: e1000599 doi:10.1371/journal.pgen.1000599.

32. ChurchC, MoirL, McMurrayF, GirardC, BanksGT, et al. (2010) Overexpression of Fto leads to increased food intake and results in obesity. Nature genetics 42: 1086–U1147.

33. TungYC, AyusoE, ShanX, BoschF, O'RahillyS, et al. (2010) Hypothalamic-specific manipulation of Fto, the ortholog of the human obesity gene FTO, affects food intake in rats. PLoS ONE 5: e8771 doi:10.1371/journal.pone.0008771.

34. ButlerAA, KozakLP (2010) A recurring problem with the analysis of energy expenditure in genetic models expressing lean and obese phenotypes. Diabetes 59: 323–329.

35. ChoiSJ, Yablonka-ReuveniZ, KaiyalaKJ, OgimotoK, SchwartzMW, et al. (2011) Increased energy expenditure and leptin sensitivity account for low fat mass in myostatin-deficient mice. American journal of physiology Endocrinology and metabolism 300: E1031–1037.

36. CheungMK, GulatiP, O'RahillyS, YeoGS (2012) FTO expression is regulated by availability of essential amino acids. International Journal of Obesity doi:10.1038/ijo.2012.77.

37. DahlstrandJ, LardelliM, LendahlU (1995) Nestin mRNA expression correlates with the central nervous system progenitor cell state in many, but not all, regions of developing central nervous system. Brain research Developmental brain research 84: 109–129.

38. McTaggartJS, LeeS, IberlM, ChurchC, CoxRD, et al. (2011) FTO Is Expressed in Neurones throughout the Brain and Its Expression Is Unaltered by Fasting. PLoS ONE 6: e27968 doi:10.1371/journal.pone.0027968.

39. WangP, YangFJ, DuH, GuanYF, XuTY, et al. (2011) Involvement of leptin receptor long isoform (LepRb)-STAT3 signaling pathway in brain fat mass- and obesity-associated (FTO) downregulation during energy restriction. Molecular medicine 17: 523–532.

40. StratigopoulosG, PadillaSL, LeDucCA, WatsonE, HattersleyAT, et al. (2008) Regulation of Fto/Ftm gene expression in mice and humans. American Journal of Physiology-Regulatory Integrative and Comparative Physiology 295: R1360–R1363.

41. FredrikssonR, HagglundM, OlszewskiPK, StephanssonO, JacobssonJA, et al. (2008) 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 149: 2062–2071.

42. OlszewskiPK, FredrikssonR, OlszewskaAM, StephanssonO, AlsioJ, et al. (2009) Hypothalamic FTO is associated with the regulation of energy intake not feeding reward. BMC neuroscience 10: 129 doi:10.1186/1471-2202-10-129.

43. WanES, ChoMH, BoutaouiN, KlandermanBJ, SylviaJS, et al. (2011) Genome-wide association analysis of body mass in chronic obstructive pulmonary disease. American journal of respiratory cell and molecular biology 45: 304–310.

44. RodriguezCI, BuchholzF, GallowayJ, SequerraR, KasperJ, et al. (2000) High-efficiency deleter mice show that FLPe is an alternative to Cre-loxP. Nat Genet 25: 139–140.

45. Paxinos G, Franklin KBJ (2001) The Mouse Brain in Stereotaxic Coordinates. New York: Academic Press.

46. Diggle P, Heagerty P, Liang K-Y, Zeger SL (2002) Analysis of Longitudinal Data. Oxford: Oxford University press.

47. Pinheiro JC, Bates DM (2000) Mixed-Effects Models in S and S-Plus; Chambers J, Eddy W, Hardle W, Sheather S, Tierney L, editors. New York: Springer Verlag.

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2013 Číslo 1
Najčítanejšie tento týždeň
Najčítanejšie v tomto čísle
Kurzy

Zvýšte si kvalifikáciu online z pohodlia domova

Získaná hemofilie - Povědomí o nemoci a její diagnostika
nový kurz

Eozinofilní granulomatóza s polyangiitidou
Autori: doc. MUDr. Martina Doubková, Ph.D.

Všetky kurzy
Prihlásenie
Zabudnuté heslo

Zadajte e-mailovú adresu, s ktorou ste vytvárali účet. Budú Vám na ňu zasielané informácie k nastaveniu nového hesla.

Prihlásenie

Nemáte účet?  Registrujte sa

#ADS_BOTTOM_SCRIPTS#