Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Sci Rep ; 9(1): 17123, 2019 11 20.
Artigo em Inglês | MEDLINE | ID: mdl-31748580

RESUMO

We sequenced coding regions of the cluster of differentiation 36 (CD36) gene in 184 French individuals of European ancestry presenting simultaneously with type 2 diabetes (T2D), arterial hypertension, dyslipidemia, and coronary heart disease. We identified rare missense mutations (p.Pro191Leu/rs143150225 and p.Ala252Val/rs147624636) in two heterozygous cases. The two CD36 mutation carriers had no family history of T2D and no clustering of cardio-metabolic complications. While the p.Pro191Leu mutation was found in 84 heterozygous carriers from five ethnic groups from the genome aggregation database (global frequency: 0.0297%, N = 141,321), only one European carrier of the p.Ala252Val mutation was identified (global frequency: 0.00040%, N = 125,523). The Pro191 and Ala252 amino acids were not conserved (74.8% and 68.9% across 131 animal species, respectively). In vitro experiments showed that the two CD36 mutant proteins are expressed and trafficked to the plasma membrane where they bind modified low-density-lipoprotein (LDL) cholesterol as normal. However, molecular modelling of the recent CD36 crystal structure showed that Pro191 was located at the exit/entrance gate of the lipid binding chamber and Ala252 was in line with the chamber. Overall, our data do not support a major contribution of CD36 rare coding mutations to T2D and its cardio-metabolic complications in the French population.


Assuntos
Antígenos CD36/genética , Doença das Coronárias/genética , Diabetes Mellitus Tipo 2/genética , Dislipidemias/genética , Doenças Metabólicas/genética , Mutação de Sentido Incorreto/genética , Hipertensão Arterial Pulmonar/genética , Membrana Celular/genética , Genótipo , Heterozigoto , Humanos , Lipoproteínas LDL/genética
2.
PLoS Genet ; 10(4): e1004235, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24699409

RESUMO

Variants in the growth factor receptor-bound protein 10 (GRB10) gene were in a GWAS meta-analysis associated with reduced glucose-stimulated insulin secretion and increased risk of type 2 diabetes (T2D) if inherited from the father, but inexplicably reduced fasting glucose when inherited from the mother. GRB10 is a negative regulator of insulin signaling and imprinted in a parent-of-origin fashion in different tissues. GRB10 knock-down in human pancreatic islets showed reduced insulin and glucagon secretion, which together with changes in insulin sensitivity may explain the paradoxical reduction of glucose despite a decrease in insulin secretion. Together, these findings suggest that tissue-specific methylation and possibly imprinting of GRB10 can influence glucose metabolism and contribute to T2D pathogenesis. The data also emphasize the need in genetic studies to consider whether risk alleles are inherited from the mother or the father.


Assuntos
Proteína Adaptadora GRB10/genética , Proteína Adaptadora GRB10/metabolismo , Ilhotas Pancreáticas/metabolismo , Alelos , Diabetes Mellitus Tipo 2 , Jejum/metabolismo , Estudo de Associação Genômica Ampla/métodos , Glucose/genética , Glucose/metabolismo , Humanos , Insulina/genética , Insulina/metabolismo , Resistência à Insulina/genética , Masculino , Pessoa de Meia-Idade , Polimorfismo de Nucleotídeo Único/genética , Receptor de Insulina/genética , Receptor de Insulina/metabolismo , Transdução de Sinais/genética
3.
J Clin Invest ; 123(7): 3037-41, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23778136

RESUMO

Sim1 haploinsufficiency in mice induces hyperphagic obesity and developmental abnormalities of the brain. In humans, abnormalities in chromosome 6q16, a region that includes SIM1, were reported in obese children with a Prader-Willi-like syndrome; however, SIM1 involvement in obesity has never been conclusively demonstrated. Here, SIM1 was sequenced in 44 children with Prader-Willi-like syndrome features, 198 children with severe early-onset obesity, 568 morbidly obese adults, and 383 controls. We identified 4 rare variants (p.I128T, p.Q152E, p.R581G, and p.T714A) in 4 children with Prader-Willi-like syndrome features (including severe obesity) and 4 other rare variants (p.T46R, p.E62K, p.H323Y, and p.D740H) in 7 morbidly obese adults. By assessing the carriers' relatives, we found a significant contribution of SIM1 rare variants to intra-family risk for obesity. We then assessed functional effects of the 8 substitutions on SIM1 transcriptional activities in stable cell lines using luciferase gene reporter assays. Three mutations showed strong loss-of-function effects (p.T46R, p.H323Y, and p.T714A) and were associated with high intra-family risk for obesity, while the variants with mild or no effects on SIM1 activity were not associated with obesity within families. Our genetic and functional studies demonstrate a firm link between SIM1 loss of function and severe obesity associated with, or independent of, Prader-Willi-like features.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Mutação de Sentido Incorreto , Obesidade Mórbida/genética , Síndrome de Prader-Willi/genética , Proteínas Repressoras/genética , Adulto , Estudos de Casos e Controles , Pré-Escolar , Análise Mutacional de DNA , Feminino , Expressão Gênica , Frequência do Gene , Genes Reporter , Estudos de Associação Genética , Células HEK293 , Humanos , Lactente , Luciferases/biossíntese , Luciferases/genética , Masculino , Pessoa de Meia-Idade , Modelos Moleculares , Ativação Transcricional , Adulto Jovem
4.
PLoS One ; 7(11): e49919, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23209618

RESUMO

OBJECTIVES: Prevention of obesity should start as early as possible after birth. We aimed to build clinically useful equations estimating the risk of later obesity in newborns, as a first step towards focused early prevention against the global obesity epidemic. METHODS: We analyzed the lifetime Northern Finland Birth Cohort 1986 (NFBC1986) (N = 4,032) to draw predictive equations for childhood and adolescent obesity from traditional risk factors (parental BMI, birth weight, maternal gestational weight gain, behaviour and social indicators), and a genetic score built from 39 BMI/obesity-associated polymorphisms. We performed validation analyses in a retrospective cohort of 1,503 Italian children and in a prospective cohort of 1,032 U.S. children. RESULTS: In the NFBC1986, the cumulative accuracy of traditional risk factors predicting childhood obesity, adolescent obesity, and childhood obesity persistent into adolescence was good: AUROC = 0·78[0·74-0.82], 0·75[0·71-0·79] and 0·85[0·80-0·90] respectively (all p<0·001). Adding the genetic score produced discrimination improvements ≤1%. The NFBC1986 equation for childhood obesity remained acceptably accurate when applied to the Italian and the U.S. cohort (AUROC = 0·70[0·63-0·77] and 0·73[0·67-0·80] respectively) and the two additional equations for childhood obesity newly drawn from the Italian and the U.S. datasets showed good accuracy in respective cohorts (AUROC = 0·74[0·69-0·79] and 0·79[0·73-0·84]) (all p<0·001). The three equations for childhood obesity were converted into simple Excel risk calculators for potential clinical use. CONCLUSION: This study provides the first example of handy tools for predicting childhood obesity in newborns by means of easily recorded information, while it shows that currently known genetic variants have very little usefulness for such prediction.


Assuntos
Obesidade/epidemiologia , Risco , Adolescente , Adulto , Peso ao Nascer , Índice de Massa Corporal , Criança , Estudos de Coortes , Feminino , Finlândia/epidemiologia , Humanos , Modelos Logísticos , Masculino , Pessoa de Meia-Idade , Obesidade/prevenção & controle , População Branca , Adulto Jovem
5.
PLoS One ; 7(6): e37423, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22701567

RESUMO

BACKGROUND: Maturity-onset of the young (MODY) is a clinically heterogeneous form of diabetes characterized by an autosomal-dominant mode of inheritance, an onset before the age of 25 years, and a primary defect in the pancreatic beta-cell function. Approximately 30% of MODY families remain genetically unexplained (MODY-X). Here, we aimed to use whole-exome sequencing (WES) in a four-generation MODY-X family to identify a new susceptibility gene for MODY. METHODOLOGY: WES (Agilent-SureSelect capture/Illumina-GAIIx sequencing) was performed in three affected and one non-affected relatives in the MODY-X family. We then performed a high-throughput multiplex genotyping (Illumina-GoldenGate assay) of the putative causal mutations in the whole family and in 406 controls. A linkage analysis was also carried out. PRINCIPAL FINDINGS: By focusing on variants of interest (i.e. gains of stop codon, frameshift, non-synonymous and splice-site variants not reported in dbSNP130) present in the three affected relatives and not present in the control, we found 69 mutations. However, as WES was not uniform between samples, a total of 324 mutations had to be assessed in the whole family and in controls. Only one mutation (p.Glu227Lys in KCNJ11) co-segregated with diabetes in the family (with a LOD-score of 3.68). No KCNJ11 mutation was found in 25 other MODY-X unrelated subjects. CONCLUSIONS/SIGNIFICANCE: Beyond neonatal diabetes mellitus (NDM), KCNJ11 is also a MODY gene ('MODY13'), confirming the wide spectrum of diabetes related phenotypes due to mutations in NDM genes (i.e. KCNJ11, ABCC8 and INS). Therefore, the molecular diagnosis of MODY should include KCNJ11 as affected carriers can be ideally treated with oral sulfonylureas.


Assuntos
Diabetes Mellitus Tipo 2/genética , Exoma/genética , Predisposição Genética para Doença/genética , Canais de Potássio Corretores do Fluxo de Internalização/genética , Sequência de Bases , Mapeamento Cromossômico , Análise Mutacional de DNA , França , Genótipo , Humanos , Escore Lod , Dados de Sequência Molecular , Linhagem
6.
Obesity (Silver Spring) ; 20(11): 2278-82, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22447289

RESUMO

Melanocortin-4 receptor (MC4R) loss-of-function mutations are the commonest genetic cause of human monogenic obesity, so far. The contribution of MC4R coding mutations to severe obesity in the high-obesity prone Greek population has not been investigated to date. We determined the MC4R coding sequence of 510 obese and 469 lean control subjects of Greek origin, and we estimated the prevalence and the penetrance on obesity of MC4R loss-of-function mutations. The functional impact of novel nonsynonymous variants detected was investigated in vitro. We found two novel synonymous mutations (L23L and I102I), four nonsynonymous mutations (T112M, S127L, N274S, and S295L), and two polymorphisms (V103I and I251L) previously described in literature. We also detected a novel mutation (L207V) in a severely obese 69-year-old female patient, although the mutation did not cosegregate with obesity in the corresponding pedigree and had no functional consequences on MC4R protein function. Loss-of-function mutations represented 75% of all nonsynonymous rare mutations identified among lean carriers and only 25% among obese subjects (P = 0.0001). The prevalence of loss-of-function mutations was lower in the obese group than in lean control subjects (0.20 vs. 0.64%) but this difference was not significant. Therefore, the estimated penetrance of deleterious MC4R mutations was very low (6.3%) in heterozygous Greek carriers of MC4R loss-of-function mutations. Our data suggest that MC4R loss-of-function mutations are rare in the Greek population. MC4R genetic deficiency is unlikely to explain the high propensity to develop severe obesity in this specific population.


Assuntos
Mutação de Sentido Incorreto , Obesidade Mórbida/genética , Receptor Tipo 4 de Melanocortina/genética , Idoso , Distribuição da Gordura Corporal , Estudos de Casos e Controles , Feminino , Grécia/epidemiologia , Humanos , Masculino , Pessoa de Meia-Idade , Obesidade Mórbida/epidemiologia , Linhagem , Fenótipo , Receptor Tipo 4 de Melanocortina/deficiência , Análise de Sequência , Transdução de Sinais
7.
Diabetes ; 61(2): 383-90, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22210313

RESUMO

Null mutations in the PCSK1 gene, encoding the proprotein convertase 1/3 (PC1/3), cause recessive monogenic early onset obesity. Frequent coding variants that modestly impair PC1/3 function mildly increase the risk for common obesity. The aim of this study was to determine the contribution of rare functional PCSK1 mutations to obesity. PCSK1 exons were sequenced in 845 nonconsanguineous extremely obese Europeans. Eight novel nonsynonymous PCSK1 mutations were identified, all heterozygous. Seven mutations had a deleterious effect on either the maturation or the enzymatic activity of PC1/3 in cell lines. Of interest, five of these novel mutations, one of the previously described frequent variants (N221D), and the mutation found in an obese mouse model (N222D), affect residues at or near the structural calcium binding site Ca-1. The prevalence of the newly identified mutations was assessed in 6,233 obese and 6,274 lean European adults and children, which showed that carriers of any of these mutations causing partial PCSK1 deficiency had an 8.7-fold higher risk to be obese than wild-type carriers. These results provide the first evidence of an increased risk of obesity in heterozygous carriers of mutations in the PCSK1 gene. Furthermore, mutations causing partial PCSK1 deficiency are present in 0.83% of extreme obesity phenotypes.


Assuntos
Mutação , Obesidade/genética , Pró-Proteína Convertase 1/genética , Adulto , Feminino , Genótipo , Glicosilação , Heterozigoto , Humanos , Masculino , Pessoa de Meia-Idade , Pró-Proteína Convertase 1/química , Pró-Proteína Convertase 1/deficiência
8.
Obesity (Silver Spring) ; 20(2): 389-95, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21720444

RESUMO

Twenty-four single-nucleotide polymorphisms (SNPs) have been reproducibly associated with obesity. We performed a follow-up study for obesity in the Greek adult population. A total of 510 obese and 469 lean adults were genotyped for 24 SNPs. We tested the association with obesity status using logistic regression and we evaluated the combined genetic risk of 24 SNPs by calculating the area under the receiver-operating characteristic (ROC) curves. We nominally replicated the association with obesity (BMI ≥30 kg/m(2)) of six SNPs in or near the FTO, MC4R, TMEM18, PRL, AIF1, and PCSK1 loci (1.28 ≤ odds ratio (OR) ≤ 1.35; 0.004 ≤ P ≤ 0.043). The discrimination ability for obesity was slightly stronger (P = 9.59 × 10(-6)) when the genetic information of the 24 SNPs was added to nongenetic risk factors (area under the curve (AUC) = 0.722) in comparison with nongenetic factors analyzed alone (AUC = 0.685). Our data suggest that SNPs in or near the FTO, MC4R, TMEM18, PRL, AIF1, and PCSK1 loci contribute to obesity risk in the Greek population.


Assuntos
Proteínas de Ciclo Celular/genética , Proteínas de Ligação a DNA/genética , Proteínas de Membrana/genética , Neuropeptídeos/genética , Obesidade/genética , Polimorfismo de Nucleotídeo Único , Proteínas Tirosina Fosfatases/genética , Proteínas/genética , Receptor Tipo 4 de Melanocortina/genética , Dioxigenase FTO Dependente de alfa-Cetoglutarato , Proteínas de Ligação ao Cálcio , Estudos de Casos e Controles , Feminino , Seguimentos , Predisposição Genética para Doença , Estudo de Associação Genômica Ampla , Genótipo , Grécia/epidemiologia , Humanos , Masculino , Proteínas dos Microfilamentos , Pessoa de Meia-Idade , Obesidade/epidemiologia , Fatores de Risco
9.
PLoS Genet ; 6(4): e1000916, 2010 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-20421936

RESUMO

Meta-analyses of population-based genome-wide association studies (GWAS) in adults have recently led to the detection of new genetic loci for obesity. Here we aimed to discover additional obesity loci in extremely obese children and adolescents. We also investigated if these results generalize by estimating the effects of these obesity loci in adults and in population-based samples including both children and adults. We jointly analysed two GWAS of 2,258 individuals and followed-up the best, according to lowest p-values, 44 single nucleotide polymorphisms (SNP) from 21 genomic regions in 3,141 individuals. After this DISCOVERY step, we explored if the findings derived from the extremely obese children and adolescents (10 SNPs from 5 genomic regions) generalized to (i) the population level and (ii) to adults by genotyping another 31,182 individuals (GENERALIZATION step). Apart from previously identified FTO, MC4R, and TMEM18, we detected two new loci for obesity: one in SDCCAG8 (serologically defined colon cancer antigen 8 gene; p = 1.85x10(-8) in the DISCOVERY step) and one between TNKS (tankyrase, TRF1-interacting ankyrin-related ADP-ribose polymerase gene) and MSRA (methionine sulfoxide reductase A gene; p = 4.84x10(-7)), the latter finding being limited to children and adolescents as demonstrated in the GENERALIZATION step. The odds ratios for early-onset obesity were estimated at approximately 1.10 per risk allele for both loci. Interestingly, the TNKS/MSRA locus has recently been found to be associated with adult waist circumference. In summary, we have completed a meta-analysis of two GWAS which both focus on extremely obese children and adolescents and replicated our findings in a large followed-up data set. We observed that genetic variants in or near FTO, MC4R, TMEM18, SDCCAG8, and TNKS/MSRA were robustly associated with early-onset obesity. We conclude that the currently known major common variants related to obesity overlap to a substantial degree between children and adults.


Assuntos
Peso Corporal/genética , Loci Gênicos , Genoma Humano , Obesidade/genética , Adolescente , Adulto , Idade de Início , Alelos , Índice de Massa Corporal , Criança , França/epidemiologia , Predisposição Genética para Doença , Estudo de Associação Genômica Ampla , Alemanha/epidemiologia , Humanos , Obesidade/epidemiologia , Polimorfismo de Nucleotídeo Único
10.
Clin Biochem ; 43(6): 549-52, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20132806

RESUMO

OBJECTIVES: We investigated two genetic markers in pro inflammatory molecules : TNFalpha -308G/A and IL6 -174G/C in order to assess their effect on type 2 diabetes (T2D) and obesity in the Tunisian population. DESIGN AND METHODS: The study sample includes 228 patients with T2D and 300 healthy controls. Genotyping of IL6 -174G/C (rs1800795) was performed using Automated Dye Terminator Sequencing and of TNFalpha -308G/A (rs1800629) using the LightTyper technology. RESULTS: SNPs IL6 -174G/C and TNFalpha -308G/A are associated neither with T2D (p=0.89, p=0.34 respectively) nor with risk for overweight (p=0.86, p=0.12 respectively) in Tunisian population. Bonferroni correction showed that the founded association of IL6 -174G/C SNP with T2D susceptibility restricted to overweight patients (p(nominal)=0.03, p(corrected)=0.0033) is likely to be a random result. CONCLUSION: SNPs IL6 -174G/C and TNFalpha -308G/A are not major contributors to T2D or obesity risk in our Tunisian population.


Assuntos
Diabetes Mellitus Tipo 2/genética , Interleucina-6/genética , Obesidade/genética , Polimorfismo de Nucleotídeo Único , Fator de Necrose Tumoral alfa/genética , Adulto , Idoso , Estudos de Casos e Controles , Diabetes Mellitus Tipo 2/epidemiologia , Feminino , Ligação Genética , Predisposição Genética para Doença , Genética Populacional , Genótipo , Humanos , Masculino , Pessoa de Meia-Idade , Obesidade/epidemiologia , Polimorfismo de Nucleotídeo Único/fisiologia , Regiões Promotoras Genéticas/genética , Fatores de Risco , Tunísia/epidemiologia
11.
Obesity (Silver Spring) ; 18(8): 1670-5, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20075856

RESUMO

SIM1 (single-minded 1) haploinsufficiency is responsible for obesity in both humans and mice, but the contribution of frequent DNA variation to polygenic obesity is unknown. Sequencing of all exons, exon/intron boundaries, 870 base pairs (bp) of the putative promoter, and 1,095 bp of the 3'UTR of SIM1 gene in 143 obese children and 24 control adults identified 13 common variants. After analysis of the linkage disequilibrium (LD) structure, association study of eight variants was performed in 1,275 obese children and severely obese adults, in 1,395 control subjects, and in 578 obesity-selected pedigrees. A nominal evidence of association was found for the nonsynonymous variant P352T C/A (rs3734354) (P = 0.01, OR = 0.81 (0.70-0.95)), the +2,004 TGA -/insT SNP (rs35180395) (P = 0.02, OR = 1.21 (1.02-1.43)), the +2,215A/G TGA SNP (rs9386126) (P = 0.002, OR = 0.81 (0.71-0.93)), and pooled childhood/adult obesity. Even though transmission disequilibrium test (TDT) further supported the association of P352T and +2,004 -/inst T with obesity, none of these nominal associations remained significant after a multiple testing Bonferroni correction. Therefore, our study excludes a major contribution of SIM1 common variants in exons, 5' and 3' UTR regions in polygenic obesity susceptibility in French Europeans.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Herança Multifatorial , Obesidade/genética , Polimorfismo de Nucleotídeo Único , Proteínas Repressoras/genética , População Branca/genética , Adolescente , Adulto , Sequência de Bases , Criança , França , Predisposição Genética para Doença , Humanos , Desequilíbrio de Ligação , Pessoa de Meia-Idade , Linhagem
12.
Diabetes ; 59(1): 311-8, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19833892

RESUMO

OBJECTIVE: Single nucleotide polymorphisms (SNPs) in intron 1 of fat mass- and obesity-associated gene (FTO) are strongly associated with human adiposity, whereas Fto(-/-) mice are lean and Fto(+/-) mice are resistant to diet-induced obesity. We aimed to determine whether FTO mutations are disproportionately represented in lean or obese humans and to use these mutations to understand structure-function relationships within FTO. RESEARCH DESIGN AND METHODS: We sequenced all coding exons of FTO in 1,433 severely obese and 1,433 lean individuals. We studied the enzymatic activity of selected nonsynonymous variants. RESULTS: We identified 33 heterozygous nonsynonymous variants in lean (2.3%) and 35 in obese (2.4%) individuals, with 8 mutations unique to the obese and 11 unique to the lean. Two novel mutations replace absolutely conserved residues: R322Q in the catalytic domain and R96H in the predicted substrate recognition lid. R322Q was unable to catalyze the conversion of 2-oxoglutarate to succinate in the presence or absence of 3-methylthymidine. R96H retained some basal activity, which was not enhanced by 3-methylthymidine. However, both were found in lean and obese individuals. CONCLUSIONS: Heterozygous, loss-of-function mutations in FTO exist but are found in both lean and obese subjects. Although intron 1 SNPs are unequivocally associated with obesity in multiple populations and murine studies strongly suggest that FTO has a role in energy balance, it appears that loss of one functional copy of FTO in humans is compatible with being either lean or obese. Functional analyses of FTO mutations have given novel insights into structure-function relationships in this enzyme.


Assuntos
Mutação , Obesidade/genética , Polimorfismo de Nucleotídeo Único , Proteínas/genética , Magreza/genética , Dioxigenase FTO Dependente de alfa-Cetoglutarato , Substituição de Aminoácidos , Éxons , Humanos , Íntrons , Valores de Referência
13.
Obesity (Silver Spring) ; 18(7): 1289-96, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19851307

RESUMO

Using restriction fragment differential display (RFDD) technology, we have identified the imprinted gene neuronatin (Nnat) as a hypothalamic target under the influence of leptin. Nnat mRNA expression is decreased in several key appetite regulatory hypothalamic nuclei in rodents with impaired leptin signaling and during fasting conditions. Furthermore, peripheral administration of leptin to ob/ob mice normalizes hypothalamic Nnat expression. Comparative immunohistochemical analysis of human and rat hypothalami demonstrates that NNAT protein is present in anatomically equivalent nuclei, suggesting human physiological relevance of the gene product(s). A putative role of Nnat in human energy homeostasis is further emphasized by a consistent association between single nucleotide polymorphisms (SNPs) in the human Nnat gene and severe childhood and adult obesity.


Assuntos
Impressão Genômica/fisiologia , Leptina/metabolismo , Proteínas de Membrana/genética , Proteínas do Tecido Nervoso/genética , Obesidade/genética , Obesidade/fisiopatologia , Tecido Adiposo/fisiologia , Animais , Metabolismo Energético/genética , Perfilação da Expressão Gênica , Genótipo , Homeostase/genética , Hipotálamo/fisiologia , Leptina/genética , Masculino , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Proteínas do Tecido Nervoso/metabolismo , Células PC12 , Pâncreas/fisiologia , Hipófise/fisiologia , Polimorfismo de Nucleotídeo Único , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Transdução de Sinais/fisiologia
14.
Obesity (Silver Spring) ; 17(8): 1549-53, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19300429

RESUMO

Visfatin was recently reported as a novel adipokine encoded by the NAMPT (PBEF1) gene. This study was aimed at investigation of the possibility that single-nucleotide polymorphisms (SNPs) in the visfatin gene are associated with either obesity or type 2 diabetes (T2D). A set of eight "tag-SNPs" were selected and ABI SNPlex assays designed for genotyping purposes. A total of 1,709 severely obese subjects were typed (896 class III obese adults and 813 children) together with 2,367 T2D individuals and 2,850 controls. For quantitative trait analysis, an additional 2,362 subjects were typed for rs10487818 from a general population sample. One rare SNP, rs10487818, located in intron 4 of NAMPT was associated with severe obesity, with a minor allele frequency of 1.6% in controls, 0.4% in the class III obese adults and, remarkably, 0% in the severely obese children. A highly significant association was observed for the presence or absence of the rare allele, i.e., (A,A) vs. (A,T + T,T) genotypes, in children (P = 6 x 10(-9)) and in adults (P = 8 x 10(-5)). No other significant (P < 0.05) association was observed with obesity or T2D for this or any other SNP. No association with BMI or waist-to-hip ratio was observed in a general population sample (n = 5,212). This is one of the first rare SNPs shown to be protective against a common polygenic disease and provides further evidence that rare alleles of strong effect can contribute to complex diseases such as severe obesity.


Assuntos
Diabetes Mellitus Tipo 2/genética , Predisposição Genética para Doença , Nicotinamida Fosforribosiltransferase/genética , Nicotinamida Fosforribosiltransferase/fisiologia , Obesidade/genética , Adulto , Índice de Massa Corporal , Criança , Estudos de Coortes , Feminino , Genótipo , Haplótipos , Humanos , Íntrons , Desequilíbrio de Ligação , Masculino , Obesidade/prevenção & controle , Razão de Chances , Polimorfismo de Nucleotídeo Único
15.
Obesity (Silver Spring) ; 17(4): 745-54, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19165163

RESUMO

Ghrelin exerts a stimulatory effect on appetite and regulates energy homeostasis. Ghrelin gene variants have been shown to be associated with metabolic traits, although there is evidence suggesting linkage and association with obesity and the ghrelin receptor (GHSR). We hypothesized that these genes are good candidates for susceptibility to obesity. Direct sequencing identified 12 ghrelin single-nucleotide polymorphisms (SNPs) and 8 GHSR SNPs. The 10 common SNPs were genotyped in 1,275 obese subjects and in 1,059 subjects from a general population cohort of European origin. In the obesity case-control study, the GHSR SNP rs572169 was found to be associated with obesity (P = 0.007 in additive model, P = 0.001 in dominant model, odds ratio (OR) 1.73, 95% confidence interval (1.23-2.44)). The ghrelin variant, g.A265T (rs4684677), showed an association with obesity (P = 0.009, BMI adjusted for age and sex) in obese families. The ghrelin variant, g.A-604G (rs27647), showed an association with insulin levels at 2-h post-oral glucose tolerance test (OGTT) (P = 0.009) in obese families. We found an association between the eating behavior "overeating" and the GHSR SNP rs2232169 (P = 0.02) in obese subjects. However, none of these associations remained significant when corrected for multiple comparisons. Replication of the nominal associations with obesity could not be confirmed in a German genome-wide association (GWA) study for rs4684677 and rs572169 polymorphisms. Our data suggest that common polymorphisms in ghrelin and its receptor genes are not major contributors to the development of polygenic obesity, although common variants may alter body weight and eating behavior and contribute to insulin resistance, in particular in the context of early-onset obesity.


Assuntos
Predisposição Genética para Doença/genética , Grelina/genética , Obesidade/genética , Polimorfismo de Nucleotídeo Único/genética , Receptores de Grelina/genética , Adolescente , Adulto , Glicemia/metabolismo , Peso Corporal/genética , Estudos de Casos e Controles , Criança , Comportamento Alimentar , Feminino , França , Ligação Genética/genética , Predisposição Genética para Doença/etnologia , Alemanha , Haplótipos/genética , Humanos , Insulina/sangue , Desequilíbrio de Ligação/genética , Masculino , Pessoa de Meia-Idade , Obesidade/etnologia
16.
Nat Genet ; 41(2): 157-9, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19151714

RESUMO

We analyzed genome-wide association data from 1,380 Europeans with early-onset and morbid adult obesity and 1,416 age-matched normal-weight controls. Thirty-eight markers showing strong association were further evaluated in 14,186 European subjects. In addition to FTO and MC4R, we detected significant association of obesity with three new risk loci in NPC1 (endosomal/lysosomal Niemann-Pick C1 gene, P = 2.9 x 10(-7)), near MAF (encoding the transcription factor c-MAF, P = 3.8 x 10(-13)) and near PTER (phosphotriesterase-related gene, P = 2.1 x 10(-7)).


Assuntos
Estudo de Associação Genômica Ampla , Obesidade Mórbida/genética , Locos de Características Quantitativas , Adulto , Idade de Início , Dioxigenase FTO Dependente de alfa-Cetoglutarato , Proteínas de Transporte/genética , Estudos de Casos e Controles , Criança , Europa (Continente)/epidemiologia , Frequência do Gene , Predisposição Genética para Doença , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Glicoproteínas de Membrana/genética , Proteína C1 de Niemann-Pick , Obesidade Mórbida/epidemiologia , Polimorfismo de Nucleotídeo Único , População , Proteínas/genética , Proteínas Proto-Oncogênicas c-maf/genética , Receptor Tipo 4 de Melanocortina/genética , Fatores de Risco
17.
Obesity (Silver Spring) ; 17(1): 202-6, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18948963

RESUMO

Ectonucleotide Pyrophosphatase Phosphodiesterase 1 (ENPP1) downregulates insulin signaling by inhibiting the insulin receptor's tyrosine-kinase. K121Q and other ENPP1 single-nucleotide polymorphisms (SNPs), IVS20delT-11 and A/G+1044TGA, have been previously associated with obesity in French children, and the risk haplotype QdelTG has also been associated with this condition in both French and German children. Our aim was to perform a case-control replication study in order to assess the possible association of childhood obesity and overweight with the above-mentioned ENPP1 SNPs, and with the QdelTG haplotype, in the Italian population. A total of 865 healthy Italian children were studied: 453 normal-weight, 243 overweight and 169 obese subjects. Genotyping was performed by Taq-Man or Light-Cycler Technology. The Q variant of K121Q showed a negative association with overweight-obesity under both additive (odds ratio (OR) = 0.74, 95% confidence interval (CI) = 0.57-0.97, P = 0.030) and recessive (OR = 0.32, 95% CI = 0.10-0.97, P = 0.035) modes of inheritance. The Z-score of BMI showed a significant decreasing trend from children K/K homozygous to K/Q heterozygous, and to Q/Q homozygous (0.45 vs. 0.28 vs. -0.19; P = 0.009), according to the additive model. The two other SNPs and the QdelTG haplotype did not exhibit any association with overweight/obesity. This is the first child-based study showing a protective role of the 121Q variant of ENPP1 against overweight/obesity.


Assuntos
Variação Genética , Obesidade/genética , Sobrepeso/genética , Sobrepeso/prevenção & controle , Diester Fosfórico Hidrolases/genética , Polimorfismo de Nucleotídeo Único , Pirofosfatases/genética , Substituição de Aminoácidos , Glicemia/análise , Criança , Feminino , Genótipo , Humanos , Insulina/sangue , Itália/epidemiologia , Masculino , Obesidade/epidemiologia , Obesidade/prevenção & controle , Sobrepeso/epidemiologia
18.
Diabetes ; 57(9): 2511-8, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18559663

RESUMO

OBJECTIVE: Melanocortin-4 receptor (MC4R) deficiency is the most frequent genetic cause of obesity. However, there is uncertainty regarding the degree of penetrance of this condition, and the putative impact of the environment on the development of obesity in MC4R mutation carriers is unknown. RESEARCH DESIGN AND METHODS: We determined the MC4R sequence in 2,257 obese individuals and 2,677 nonobese control subjects of European origin and established the likely functional impact of all variants detected. We then included relatives of probands carriers and studied 25 pedigrees, including 97 carriers and 94 noncarriers from three generations. RESULTS: Of the MC4R nonsynonymous mutations found in obese subjects, 68% resulted in a loss of function in vitro. They were found in 1.72% of obese versus 0.15% of nonobesed subjects (P = 6.9 x 10(-10)). Among the families, abnormal eating behavior was more frequent in both MC4R-deficient children and adults than in noncarriers. Although BMI was inversely associated with educational status in noncarrier adults, no such relationship was seen in MC4R mutation carriers. We observed a generational effect, with a penetrance of 40% in MC4R-deficient adults aged >52 years, 60% in 18- to 52-year-old adults, and 79% in children. The longitudinal study of adult carriers showed an increasing age-dependent penetrance (37% at 20 years versus 60% at >40 years). CONCLUSIONS: We have established a robust estimate of age-related penetrance for MC4R deficiency and demonstrated a generational effect on penetrance, which may relate to the development of an "obesogenic" environment. It remains to be seen whether appropriate manipulation of environmental factors may contribute to preventing the development of obesity even in those strongly genetically predisposed to it.


Assuntos
Obesidade/etnologia , Obesidade/genética , Penetrância , Receptor Tipo 4 de Melanocortina/genética , População Branca/genética , População Branca/estatística & dados numéricos , Adolescente , Adulto , Distribuição por Idade , Criança , Comportamento Alimentar , Feminino , Humanos , Hiperfagia/etnologia , Hiperfagia/genética , Masculino , Pessoa de Meia-Idade , Mutação , Linhagem , Fenótipo , Prevalência , Receptor Tipo 4 de Melanocortina/deficiência
19.
Obesity (Silver Spring) ; 16(6): 1471-5, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18388898

RESUMO

Previous studies have described genetic associations of the insulin gene variable number tandem repeat (INS VNTR) variant with childhood obesity and associated phenotypes. We aimed to assess the contribution of INS VNTR genotypes to childhood obesity and variance of insulin resistance, insulin secretion, and birth weight using family-based design. Participants were either French or German whites. We used transmission disequilibrium tests (TDTs) for assessing binary traits and quantitative pedigree disequilibrium tests for assessing continuous traits. In contrast to previous findings, we did not observe any familial association with childhood obesity (T = 50%, P = 0.77) in the 1,023 families tested. In French obese children, INS VNTR did not associate with fasting insulin levels (P = 0.23) and class I allele showed only borderline association with increased insulin secretion index at 30 min (P = 0.03). INS VNTR did not associate with birth weight in obese children (P = 0.98) and TDT analyses in 350 French families with history of low birth weight (LBW) showed no association with this condition (P = 0.92). In summary, our study, the largest performed so far, does not support the previously reported associations between INS VNTR and childhood obesity, insulin resistance, or birth weight, and does not suggest any major role for this variant in modulating these traits.


Assuntos
Insulina/genética , Repetições Minissatélites/genética , Obesidade/genética , Adolescente , Alelos , Peso ao Nascer/genética , Criança , Feminino , França , Humanos , Resistência à Insulina/etnologia , Resistência à Insulina/genética , Masculino , Obesidade/etnologia
20.
J Clin Endocrinol Metab ; 92(11): 4403-9, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17698913

RESUMO

CONTEXT: The melanin-concentrating hormone receptor 2 (MCHR2) is a G protein-coupled receptor for melanin-concentrating hormone, a neuropeptide that plays an important role in feeding behaviors. MCHR2 maps on chromosome 6q16.3, in a susceptibility locus for childhood obesity. OBJECTIVE: The aim of this study was to investigate the association between MCHR2 variation and human obesity. DESIGN: Case control and family-based studies were performed. PARTICIPANTS: A total of 141 obese children and 24 nonobese adult subjects was sequenced, and case-control analyses were conducted using 628 severely obese children and 1,401 controls. RESULTS: There were 11 single nucleotide polymorphisms (SNPs) identified. We showed nominal association among -38,245 ATG A/G SNP (P = 0.03; 95% confidence interval 1.02-1.34; odds ratio 1.17), A76A T/C SNP (P = 0.03; 95% confidence interval 0.58-0.97; odds ratio 0.75), and childhood obesity. Analysis of 645 trios with childhood obesity supported further the A76A T/C association, showing an overtransmission to obese children of the at risk T allele (59.0%; P = 0.01), especially in children with most severe forms of obesity (Z score of body mass index > 4) (67.0%; P = 0.003). The A76A at risk T allele was also associated with overeating during meals (P = 0.02) in an additional group of 102 nonobese children. None of the MCHR2 variants, including the A76A SNP, showed association with adult severe obesity, although a trend for association of the T allele of this variant with food disinhibition (P = 0.06) and higher hunger (P = 0.09) was found. This variant was not associated with childhood obesity in an independent case-control study, including 1,573 subjects (P = 0.98). Moreover, the A76A SNP did not explain the linkage on the 6q locus. CONCLUSION: Our results altogether suggest that MCHR2 is not a major contributor to polygenic obesity and support a modest effect of the A76A SNP on food intake abnormalities in childhood.


Assuntos
Obesidade/genética , Receptores Acoplados a Proteínas G/genética , Receptores do Hormônio Hipofisário/genética , Adulto , Alelos , Sequência de Aminoácidos , Apetite/fisiologia , Índice de Massa Corporal , Criança , Estudos de Coortes , Replicação do DNA/fisiologia , Éxons/genética , Comportamento Alimentar , Transtornos da Alimentação e da Ingestão de Alimentos/genética , Feminino , Ligação Genética/genética , Haplótipos , Humanos , Masculino , Dados de Sequência Molecular , Obesidade/psicologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...