Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
FASEB J ; 33(12): 13515-13526, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31581846

RESUMO

In utero overnutrition can predispose offspring to metabolic disease. Although the mechanisms are unclear, increased oxidative stress accelerating cellular aging has been shown to play a role. Mitochondria are the main site of reactive oxygen species (ROS) production in most cell types. Levels of ROS and the risk for oxidative damage are dictated by the balance between ROS production and antioxidant defense mechanisms. Originally considered as toxic species, physiologic levels of ROS are now known to be essential cell signaling molecules. Using a model of maternal overnutrition in C57BL6N mice, we investigate the mechanisms involved in the development of insulin resistance (IR) in muscle. In red and white gastrocnemius muscles of offspring, we are the first to report characteristics of oxidative phosphorylation, H2O2 production, activity of mitoflashes, and electron transport chain supercomplex formation. Results demonstrate altered mitochondrial function with reduced response to glucose in offspring of mice fed a high-fat and high-sucrose diet, increases in mitochondrial leak respiration, and a reduction in ROS production in red gastrocnemius in response to palmitoyl carnitine. We also demonstrate differences in supercomplex formation between red and white gastrocnemius, which may be integral to fiber-type specialization. We conclude that in this model of maternal overnutrition, mitochondrial alterations occur before the development of IR.-McMurray, F., MacFarlane, M., Kim, K., Patten, D. A., Wei-LaPierre, L., Fullerton, M. D., Harper, M. E. Maternal diet-induced obesity alters muscle mitochondrial function in offspring without changing insulin sensitivity.


Assuntos
Dieta Hiperlipídica/efeitos adversos , Intolerância à Glucose/patologia , Resistência à Insulina , Mitocôndrias Musculares/patologia , Obesidade/fisiopatologia , Estresse Oxidativo , Animais , Feminino , Intolerância à Glucose/metabolismo , Masculino , Fenômenos Fisiológicos da Nutrição Materna , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias Musculares/metabolismo , Fosforilação Oxidativa , Espécies Reativas de Oxigênio/metabolismo
2.
J Med Genet ; 53(3): 200-7, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26378117

RESUMO

BACKGROUND: A homozygous loss-of-function mutation p.(Arg316Gln) in the fat mass and obesity-associated (FTO) gene, which encodes for an iron and 2-oxoglutarate-dependent oxygenase, was previously identified in a large family in which nine affected individuals present with a lethal syndrome characterised by growth retardation and multiple malformations. To date, no other pathogenic mutation in FTO has been identified as a cause of multiple congenital malformations. METHODS: We investigated a 21-month-old girl who presented distinctive facial features, failure to thrive, global developmental delay, left ventricular cardiac hypertrophy, reduced vision and bilateral hearing loss. We performed targeted next-generation sequencing of 4813 clinically relevant genes in the patient and her parents. RESULTS: We identified a novel FTO homozygous missense mutation (c.956C>T; p.(Ser319Phe)) in the affected individual. This mutation affects a highly conserved residue located in the same functional domain as the previously characterised mutation p.(Arg316Gln). Biochemical studies reveal that p.(Ser319Phe) FTO has reduced 2-oxoglutarate turnover and N-methyl-nucleoside demethylase activity. CONCLUSION: Our findings are consistent with previous reports that homozygous mutations in FTO can lead to rare growth retardation and developmental delay syndrome, and further support the proposal that FTO plays an important role in early development of human central nervous and cardiovascular systems.


Assuntos
Deficiências do Desenvolvimento/genética , Mutação de Sentido Incorreto , Proteínas/genética , Dioxigenase FTO Dependente de alfa-Cetoglutarato , Feminino , Predisposição Genética para Doença , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Lactente
3.
PLoS Genet ; 9(1): e1003166, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23300482

RESUMO

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.


Assuntos
Composição Corporal/genética , Ingestão de Alimentos/genética , Metabolismo Energético/genética , Oxigenases de Função Mista/genética , Obesidade , Oxo-Ácido-Liases/genética , Dioxigenase FTO Dependente de alfa-Cetoglutarato , Animais , Peso Corporal/genética , Desenvolvimento Embrionário/genética , Regulação da Expressão Gênica no Desenvolvimento , Células Germinativas/metabolismo , Homeostase , Humanos , Masculino , Camundongos , Obesidade/genética , Obesidade/metabolismo
4.
Diabetologia ; 58(12): 2861-6, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26404066

RESUMO

AIMS/HYPOTHESIS: Skeletal muscle mitochondrial dysfunction has been documented in patients with type 2 diabetes mellitus; however, specific respiratory defects and their mechanisms are poorly understood. The aim of the current study was to examine oxidative phosphorylation and electron transport chain (ETC) supercomplex assembly in rectus abdominis muscles of 10 obese diabetic and 10 obese non-diabetic individuals. METHODS: Twenty obese women undergoing Roux-en-Y gastric bypass surgery were recruited for this study. Muscle samples were obtained intraoperatively and subdivided for multiple analyses, including high-resolution respirometry and assessment of supercomplex assembly. Clinical data obtained from referring physicians were correlated with laboratory findings. RESULTS: Participants in both groups were of a similar age, weight and BMI. Mitochondrial respiration rates were markedly reduced in diabetic vs non-diabetic patients. This defect was observed during maximal ADP-stimulated respiration in the presence of complex I-linked substrates and complex I- and II-linked substrates, and during maximal uncoupled respiration. There were no differences in fatty acid (octanoyl carnitine) supported respiration, leak respiration or isolated activity of cytochrome c oxidase. Intriguingly, significant correlations were found between glycated haemoglobin (HbA1c) levels and maximal respiration or respiration supported by complex I, complex I and II or fatty acid. In the muscle of diabetic patients, blue native gel electrophoresis revealed a striking decrease in complex I, III and IV containing ETC supercomplexes. CONCLUSIONS/INTERPRETATION: These findings support the hypothesis that ETC supercomplex assembly may be an important underlying mechanism of muscle mitochondrial dysfunction in type 2 diabetes mellitus.


Assuntos
Diabetes Mellitus Tipo 2/metabolismo , Complexo de Proteínas da Cadeia de Transporte de Elétrons/metabolismo , Complexo I de Transporte de Elétrons/metabolismo , Mitocôndrias/metabolismo , Obesidade/metabolismo , Fosforilação Oxidativa , Reto do Abdome/metabolismo , Difosfato de Adenosina/farmacologia , Adulto , Diabetes Mellitus Tipo 2/tratamento farmacológico , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Ácidos Graxos/metabolismo , Feminino , Hemoglobinas Glicadas/análise , Humanos , Músculo Esquelético/metabolismo
5.
Curr Diab Rep ; 12(6): 651-8, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22996130

RESUMO

The genomes of many species have now been completely sequenced including human and mouse. Great progress has been made in understanding the complex genetics that underlie diabetes and obesity in human populations. One of the current challenges is the functional identification and characterization of the genes within loci that are being mapped. There are many approaches to this problem and this review outlines the valuable role that the mouse can play. We outline the mouse resources that are available to the research community, including knockouts with conditional potential for every gene, and the efforts of the International Mouse Phenotyping Consortium to attach phenotype information to these genes. We also briefly consider the potential of TALEN technology to tailor-make new mouse models of specific mutations discovered in humans. Finally, we consider the recent progress in characterizing the GWAS genes FTO, TCF7L2, CDKAL1, and SLC30A8 in engineered mouse models.


Assuntos
Diabetes Mellitus Tipo 2/genética , Modelos Animais de Doenças , Estudo de Associação Genômica Ampla , Obesidade/genética , Dioxigenase FTO Dependente de alfa-Cetoglutarato , Animais , Quinase 5 Dependente de Ciclina/genética , Predisposição Genética para Doença , Humanos , Camundongos , Camundongos Knockout , Oxigenases de Função Mista/genética , Proteínas do Tecido Nervoso/genética , Oxo-Ácido-Liases/genética , Proteínas/genética , Proteína 2 Semelhante ao Fator 7 de Transcrição/genética , tRNA Metiltransferases
7.
Mamm Genome ; 22(7-8): 390-400, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21713584

RESUMO

Type 2 diabetes prevalence is increasing worldwide. Treatments are available, but glycaemic control is not always effective in many patients. Better models are needed to create new and improved therapies and to expand our understanding of how type 2 diabetes begins and progresses. Translational research involves the transformation of knowledge from basic scientific discoveries to impacting on public health. This can allow identification of novel molecular mechanisms underlying the disease which can lead to preventative measures, biomarkers for diagnosis, or future therapies. Generation of genetically modified mice has allowed us to investigate the function of genes and develop reproducible models in which the phenotype of the animal can be tested. Mouse models have already given us insight into glucose metabolism and insulin secretion, identified novel pathways, and have been used to confirm genome-wide association studies. In this review we discuss the use of the mouse to clarify human genome-wide association study loci, understand genes and pathways involved in type 2 diabetes, and uncover novel targets for drug discovery.


Assuntos
Diabetes Mellitus Tipo 2/genética , Modelos Animais de Doenças , Camundongos , Pesquisa Translacional Biomédica , Animais , Diabetes Mellitus Tipo 2/tratamento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Camundongos/genética , Camundongos/metabolismo
8.
Biochem Soc Trans ; 37(Pt 4): 838-42, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19614604

RESUMO

ENU (N-ethyl-N-nitrosourea) is a chemical mutagen that randomly induces point mutations in DNA. Since the 1990s ENU has been successfully used as a means to obtain mouse mutants using both gene-driven (reverse genetics) and phenotype-driven (forward genetics) approaches. A high-efficiency ENU approach results in approx. 25 functional mutations per genome; most of these will result in hypomorphic alleles. Our group has recently begun using ENU mutagenesis as a tool for understanding lung development and disease. In collaboration with other groups at MRC Harwell, we have undertaken a screen for recessive mutations affecting mouse lung development. We are currently pursuing two lines identified from this screen, Hel (head, eye and lung) and RecBA17. Both these lines exhibit lung defects and we believe that by studying the phenotypes and identifying the causative mutations, we may also shed light on lung disease pathogenesis. In collaboration with Bill Cookson and Miriam Moffatt, we are also taking a gene-driven approach for understanding asthma. Using the Harwell ENU sperm archive, we have recovered mouse lines harbouring mutations in the asthma-susceptibility genes Phf11 (PHD finger protein 11) and Dpp10 (dipeptidylpeptidase 10). Functional analyses of these alleles are currently under way.


Assuntos
Alquilantes/farmacologia , Etilnitrosoureia/farmacologia , Pulmão/patologia , Mutagênese/efeitos dos fármacos , Animais , Asma/metabolismo , Asma/patologia , Humanos , Pulmão/crescimento & desenvolvimento , Pulmão/metabolismo , Camundongos , Mutação Puntual/efeitos dos fármacos
9.
Cancer Res ; 77(20): 5491-5502, 2017 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-28830860

RESUMO

Lymphangioleiomyomatosis (LAM) is a progressive destructive neoplasm of the lung associated with inactivating mutations in the TSC1 or TSC2 tumor suppressor genes. Cell or animal models that accurately reflect the pathology of LAM have been challenging to develop. Here, we generated a robust human cell model of LAM by reprogramming TSC2 mutation-bearing fibroblasts from a patient with both tuberous sclerosis complex (TSC) and LAM (TSC-LAM) into induced pluripotent stem cells (iPSC), followed by selection of cells that resemble those found in LAM tumors by unbiased in vivo differentiation. We established expandable cell lines under smooth muscle cell (SMC) growth conditions that retained a patient-specific genomic TSC2+/- mutation and recapitulated the molecular and functional characteristics of pulmonary LAM cells. These include multiple indicators of hyperactive mTORC1 signaling, presence of specific neural crest and SMC markers, expression of VEGF-D and female sex hormone receptors, reduced autophagy, and metabolic reprogramming. Intriguingly, the LAM-like features of these cells suggest that haploinsufficiency at the TSC2 locus contributes to LAM pathology, and demonstrated that iPSC reprogramming and SMC lineage differentiation of somatic patient cells with germline mutations was a viable approach to generate LAM-like cells. The patient-derived SMC lines we have developed thus represent a novel cellular model of LAM that can advance our understanding of disease pathogenesis and develop therapeutic strategies against LAM. Cancer Res; 77(20); 5491-502. ©2017 AACR.


Assuntos
Linfangioleiomiomatose/genética , Linfangioleiomiomatose/patologia , Miócitos de Músculo Liso/fisiologia , Células-Tronco Pluripotentes/fisiologia , Animais , Proliferação de Células/fisiologia , Feminino , Haploinsuficiência , Humanos , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Células-Tronco Pluripotentes/metabolismo , Células-Tronco Pluripotentes/patologia
10.
Obesity (Silver Spring) ; 24(11): 2301-2310, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27804267

RESUMO

OBJECTIVE: High levels of reactive oxygen species (ROS) are intricately linked to obesity and associated pathologies, notably insulin resistance and type 2 diabetes. However, ROS are also thought to be important in intracellular signaling, which may paradoxically be required for insulin sensitivity. Many theories have been developed to explain this apparent paradox, which have broadened our understanding of these important small molecules. While many sites for intracellular ROS production have been described, mitochondrial generated ROS remain a major contributor in most cell types. Mitochondrial ROS generation is controlled by a number of factors described in this review. Moreover, these studies have established both a demand for novel sensitive approaches to measure ROS, as well as a need to standardize and review their suitability for different applications. METHODS: To properly assess levels of ROS and mitochondrial ROS in the development of obesity and its complications, a growing number of tools have been developed. This paper reviews many of the common methods for the investigation of ROS in mitochondria, cell, animal, and human models. RESULTS: Available approaches can be generally divided into those that measure ROS-induced damage (e.g., DNA, lipid, and protein damage); those that measure antioxidant levels and redox ratios; and those that use novel biosensors and probes for a more direct measure of different forms of ROS (e.g., 2',7'-di-chlorofluorescein (DCF), dihydroethidium (DHE) and its mitochondrial targeted form (MitoSOX), Amplex Red, roGFP, HyPer, mt-cpYFP, ratiometric H2 O2 probes, and their derivatives). Moreover, this review provides caveats and strengths for the use of these techniques in different models. CONCLUSIONS: Advances in these techniques will undoubtedly advance the understanding of ROS in obesity and may help resolve unanswered questions in the field.


Assuntos
Obesidade/diagnóstico , Estresse Oxidativo , Espécies Reativas de Oxigênio/metabolismo , Animais , Antioxidantes/análise , Modelos Animais de Doenças , Pesquisa Empírica , Humanos , Mitocôndrias/metabolismo , Obesidade/metabolismo
11.
PLoS One ; 11(10): e0163214, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27788147

RESUMO

BACKGROUND: Although reduced glutathione (rGSH) is decreased in obese mice and humans, block of GSH synthesis by buthionine sulfoximine (BSO) results in a lean, insulin-sensitive phenotype. Data is lacking about the effect of BSO on GSH precursors, cysteine and glutamate. Plasma total cysteine (tCys) is positively associated with stearoyl-coenzyme A desaturase (SCD) activity and adiposity in humans and animal models. OBJECTIVE: To explore the phenotype, amino acid and fatty acid profiles in BSO-treated mice. DESIGN: Male C3H/HeH mice aged 11 weeks were fed a high-fat diet with or without BSO in drinking water (30 mmol/L) for 8 weeks. Amino acid and fatty acid changes were assessed, as well as food consumption, energy expenditure, locomotor activity, body composition and liver vacuolation (steatosis). RESULTS: Despite higher food intake, BSO decreased particularly fat mass but also lean mass (both P<0.001), and prevented fatty liver vacuolation. Physical activity increased during the dark phase. BSO decreased plasma free fatty acids and enhanced insulin sensitivity. BSO did not alter liver rGSH, but decreased plasma total GSH (tGSH) and rGSH (by ~70%), and liver tGSH (by 82%). Glutamate accumulated in plasma and liver. Urine excretion of cysteine and its precursors was increased by BSO. tCys, rCys and cystine decreased in plasma (by 23-45%, P<0.001 for all), but were maintained in liver, at the expense of decreased taurine. Free and total plasma concentrations of the SCD products, oleic and palmitoleic acids were decreased (by 27-38%, P <0.001 for all). CONCLUSION: Counterintuitively, block of GSH synthesis decreases circulating tCys, raising the question of whether the BSO-induced obesity-resistance is linked to cysteine depletion. Cysteine-supplementation of BSO-treated mice is warranted to dissect the effects of cysteine and GSH depletion on energy metabolism.


Assuntos
Aminoácidos/metabolismo , Peso Corporal , Ácidos Graxos/metabolismo , Glutationa/deficiência , Fenótipo , Compostos de Sulfidrila/metabolismo , Tecido Adiposo/citologia , Aminoácidos/sangue , Animais , Composição Corporal , Butionina Sulfoximina/metabolismo , Ingestão de Alimentos , Metabolismo Energético , Ácidos Graxos/sangue , Glutationa/urina , Insulina/metabolismo , Fígado/citologia , Fígado/metabolismo , Locomoção , Masculino , Camundongos , Compostos de Sulfidrila/sangue
12.
Nat Commun ; 6: 6792, 2015 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-25881961

RESUMO

The fat mass and obesity-associated (FTO) gene plays a pivotal role in regulating body weight and fat mass; however, the underlying mechanisms are poorly understood. Here we show that primary adipocytes and mouse embryonic fibroblasts (MEFs) derived from FTO overexpression (FTO-4) mice exhibit increased potential for adipogenic differentiation, while MEFs derived from FTO knockout (FTO-KO) mice show reduced adipogenesis. As predicted from these findings, fat pads from FTO-4 mice fed a high-fat diet show more numerous adipocytes. FTO influences adipogenesis by regulating events early in adipogenesis, during the process of mitotic clonal expansion. The effect of FTO on adipogenesis appears to be mediated via enhanced expression of the pro-adipogenic short isoform of RUNX1T1, which enhanced adipocyte proliferation, and is increased in FTO-4 MEFs and reduced in FTO-KO MEFs. Our findings provide novel mechanistic insight into how upregulation of FTO leads to obesity.


Assuntos
Adipócitos/citologia , Adipogenia/genética , Tecido Adiposo/citologia , Fibroblastos/citologia , Mitose/genética , Proteínas/genética , Adipócitos/metabolismo , Dioxigenase FTO Dependente de alfa-Cetoglutarato , Animais , Diferenciação Celular , Proteínas de Ligação a DNA/metabolismo , Dieta Hiperlipídica , Fibroblastos/metabolismo , Camundongos , Camundongos Knockout , Obesidade/genética , Obesidade/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Fatores de Transcrição/metabolismo
13.
PLoS One ; 10(4): e0121829, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25830347

RESUMO

In 2007, a genome wide association study identified a SNP in intron one of the gene encoding human FTO that was associated with increased body mass index. Homozygous risk allele carriers are on average three kg heavier than those homozygous for the protective allele. FTO is a DNA/RNA demethylase, however, how this function affects body weight, if at all, is unknown. Here we aimed to pharmacologically inhibit FTO to examine the effect of its demethylase function in vitro and in vivo as a first step in evaluating the therapeutic potential of FTO. We showed that IOX3, a known inhibitor of the HIF prolyl hydroxylases, decreased protein expression of FTO (in C2C12 cells) and reduced maximal respiration rate in vitro. However, FTO protein levels were not significantly altered by treatment of mice with IOX3 at 60 mg/kg every two days. This treatment did not affect body weight, or RER, but did significantly reduce bone mineral density and content and alter adipose tissue distribution. Future compounds designed to selectively inhibit FTO's demethylase activity could be therapeutically useful for the treatment of obesity.


Assuntos
Fármacos Antiobesidade/farmacologia , Glicina/análogos & derivados , Isoquinolinas/farmacologia , Oxigenases de Função Mista/antagonistas & inibidores , Obesidade/tratamento farmacológico , Oxo-Ácido-Liases/antagonistas & inibidores , Dioxigenase FTO Dependente de alfa-Cetoglutarato , Animais , Linhagem Celular , Avaliação Pré-Clínica de Medicamentos , Glicina/farmacologia , Concentração Inibidora 50 , Masculino , Camundongos Endogâmicos C57BL , Oxigenases de Função Mista/metabolismo , Obesidade/metabolismo , Oxo-Ácido-Liases/metabolismo
14.
PLoS One ; 9(5): e97162, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24842286

RESUMO

Single nucleotide polymorphisms in the first intron of the fat-mass-and-obesity-related gene FTO are associated with increased body weight and adiposity. Increased expression of FTO is likely underlying this obesity phenotype, as mice with two additional copies of Fto (FTO-4 mice) exhibit increased adiposity and are hyperphagic. FTO is a demethylase of single stranded DNA and RNA, and one of its targets is the m6A modification in RNA, which might play a role in the regulation of gene expression. In this study, we aimed to examine the changes in gene expression that occur in FTO-4 mice in order to gain more insight into the underlying mechanisms by which FTO influences body weight and adiposity. Our results indicate an upregulation of anabolic pathways and a downregulation of catabolic pathways in FTO-4 mice. Interestingly, although genes involved in methylation were differentially regulated in skeletal muscle of FTO-4 mice, no effect of FTO overexpression on m6A methylation of total mRNA was detected.


Assuntos
Oxigenases de Função Mista/metabolismo , Oxo-Ácido-Liases/metabolismo , Adiposidade/genética , Adiposidade/fisiologia , Dioxigenase FTO Dependente de alfa-Cetoglutarato , Animais , Células Cultivadas , Expressão Gênica , Camundongos , Camundongos Endogâmicos C57BL , Oxigenases de Função Mista/genética , Músculo Esquelético/metabolismo , Obesidade/genética , Obesidade/metabolismo , Oxo-Ácido-Liases/genética , Polimorfismo de Nucleotídeo Único/genética
15.
PLoS One ; 9(2): e87662, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24503721

RESUMO

Common intronic variants in the Human fat mass and obesity-associated gene (FTO) are found to be associated with an increased risk of obesity. Overexpression of FTO correlates with increased food intake and obesity, whilst loss-of-function results in lethality and severe developmental defects. Despite intense scientific discussions around the role of FTO in energy metabolism, the function of FTO during development remains undefined. Here, we show that loss of Fto leads to developmental defects such as growth retardation, craniofacial dysmorphism and aberrant neural crest cells migration in Zebrafish. We find that the important developmental pathway, Wnt, is compromised in the absence of FTO, both in vivo (zebrafish) and in vitro (Fto(-/-) MEFs and HEK293T). Canonical Wnt signalling is down regulated by abrogated ß-Catenin translocation to the nucleus whilst non-canonical Wnt/Ca(2+) pathway is activated via its key signal mediators CaMKII and PKCδ. Moreover, we demonstrate that loss of Fto results in short, absent or disorganised cilia leading to situs inversus, renal cystogenesis, neural crest cell defects and microcephaly in Zebrafish. Congruently, Fto knockout mice display aberrant tissue specific cilia. These data identify FTO as a protein-regulator of the balanced activation between canonical and non-canonical branches of the Wnt pathway. Furthermore, we present the first evidence that FTO plays a role in development and cilia formation/function.


Assuntos
Cílios/genética , Cílios/metabolismo , Anormalidades Congênitas/genética , Anormalidades Congênitas/metabolismo , Proteínas/genética , Via de Sinalização Wnt , Dioxigenase FTO Dependente de alfa-Cetoglutarato , Animais , Sinalização do Cálcio , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Linhagem Celular , Cílios/patologia , Ativação Enzimática , Feminino , Técnicas de Inativação de Genes , Humanos , Camundongos , Camundongos Knockout , Morfogênese/genética , Especificidade de Órgãos/genética , Fenótipo , Peixe-Zebra , beta Catenina/metabolismo
16.
Nat Genet ; 42(12): 1086-92, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21076408

RESUMO

Genome-wide association studies have identified SNPs within FTO, the human fat mass and obesity-associated gene, that are strongly associated with obesity. Individuals homozygous for the at-risk rs9939609 A allele weigh, on average, ~3 kg more than individuals with the low-risk T allele. Mice that lack FTO function and/or Fto expression display increased energy expenditure and a lean phenotype. We show here that ubiquitous overexpression of Fto leads to a dose-dependent increase in body and fat mass, irrespective of whether mice are fed a standard or a high-fat diet. Our results suggest that increased body mass results primarily from increased food intake. Mice with increased Fto expression on a high-fat diet develop glucose intolerance. This study provides the first direct evidence that increased Fto expression causes obesity in mice.


Assuntos
Comportamento Alimentar/fisiologia , Obesidade/genética , Oxo-Ácido-Liases/metabolismo , Adiposidade/efeitos dos fármacos , Adiposidade/genética , Dioxigenase FTO Dependente de alfa-Cetoglutarato , Animais , Área Sob a Curva , Temperatura Corporal , Ritmo Circadiano/efeitos dos fármacos , Ritmo Circadiano/genética , Gorduras na Dieta/administração & dosagem , Gorduras na Dieta/farmacologia , Metabolismo Energético/efeitos dos fármacos , Metabolismo Energético/genética , Comportamento Alimentar/efeitos dos fármacos , Feminino , Glucose/metabolismo , Teste de Tolerância a Glucose , Homeostase/genética , Masculino , Camundongos , Oxigenases de Função Mista , Modelos Animais , Atividade Motora/efeitos dos fármacos , Obesidade/sangue , Oxo-Ácido-Liases/genética
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa