Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Diabetes Care ; 44(9): 1992-1999, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34116986

RESUMEN

OBJECTIVE: Gestational diabetes mellitus (GDM) is associated with an increased risk of obesity and insulin resistance in offspring later in life, which might be explained by epigenetic changes in response to maternal hyperglycemic exposure. RESEARCH DESIGN AND METHODS: We explored the association between GDM exposure and maternal blood and newborn cord blood methylation in 536 mother-offspring pairs from the prospective FinnGeDi cohort using Illumina MethylationEPIC 850K BeadChip arrays. We assessed two hypotheses. First, we tested for shared maternal and offspring epigenetic effects resulting from GDM exposure. Second, we tested whether GDM exposure and maternal methylation had an epigenetic effect on the offspring. RESULTS: We did not find any epigenetic marks (differentially methylated CpG probes) with shared and consistent effects between mothers and offspring. After including maternal methylation in the model, we identified a single significant (false discovery rate 1.38 × 10-2) CpG at the cg22790973 probe (TFCP2) associated with GDM. We identified seven additional FDR-significant interactions of maternal methylation and GDM status, with the strongest association at the same cg22790973 probe (TFCP2), as well as cg03456133, cg24440941 (H3C6), cg20002843 (LOC127841), cg19107264, and cg11493553 located within the UBE3C gene and cg17065901 in FAM13A, both susceptibility genes for type 2 diabetes and BMI, and cg23355087 within the DLGAP2 gene, known to be involved in insulin resistance during pregnancy. CONCLUSIONS: Our study reveals the potential complexity of the epigenetic transmission between mothers with GDM and their offspring, likely determined by not only GDM exposure but also other factors indicated by maternal epigenetic status, such as maternal metabolic history.


Asunto(s)
Diabetes Mellitus Tipo 2 , Diabetes Gestacional , Metilación de ADN , Proteínas de Unión al ADN/genética , Diabetes Mellitus Tipo 2/genética , Diabetes Gestacional/genética , Epigenoma , Femenino , Proteínas Activadoras de GTPasa , Humanos , Embarazo , Estudios Prospectivos , Factores de Transcripción/genética
2.
Diabetes ; 69(7): 1424-1438, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32349990

RESUMEN

Monogenic forms of obesity have been identified in ≤10% of severely obese European patients. However, the overall spectrum of deleterious variants (point mutations and structural variants) responsible for childhood severe obesity remains elusive. In this study, we genetically screened 225 severely obese children from consanguineous Pakistani families through a combination of techniques, including an in-house-developed augmented whole-exome sequencing method (CoDE-seq) that enables simultaneous detection of whole-exome copy number variations (CNVs) and point mutations in coding regions. We identified 110 (49%) probands carrying 55 different pathogenic point mutations and CNVs in 13 genes/loci responsible for nonsyndromic and syndromic monofactorial obesity. CoDE-seq also identified 28 rare or novel CNVs associated with intellectual disability in 22 additional obese subjects (10%). Additionally, we highlight variants in candidate genes for obesity warranting further investigation. Altogether, 59% of cases in the studied cohort are likely to have a discrete genetic cause, with 13% of these as a result of CNVs, demonstrating a remarkably higher prevalence of monofactorial obesity than hitherto reported and a plausible overlapping of obesity and intellectual disabilities in several cases. Finally, inbred populations with a high prevalence of obesity provide unique, genetically enriched material in the quest of new genes/variants influencing energy balance.


Asunto(s)
Obesidad Mórbida/genética , Obesidad Infantil/genética , Adolescente , Niño , Preescolar , Variaciones en el Número de Copia de ADN , Femenino , Humanos , Lactante , Leptina/genética , Masculino , Mutación , Obesidad Mórbida/epidemiología , Obesidad Mórbida/etiología , Obesidad Infantil/epidemiología , Obesidad Infantil/etiología , Prevalencia , Receptor de Melanocortina Tipo 4/genética , Receptores de Leptina/genética , Adulto Joven
3.
Clin Epigenetics ; 12(1): 68, 2020 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-32410704

RESUMEN

BACKGROUND: Adipogenesis, the process whereby preadipocytes differentiate into mature adipocytes, is crucial for maintaining metabolic homeostasis. Cholesterol-lowering statins increase type 2 diabetes (T2D) risk possibly by affecting adipogenesis and insulin resistance but the (epi)genetic mechanisms involved are unknown. Here, we characterised the effects of statin treatment on adipocyte differentiation using in vitro human preadipocyte cell model to identify putative effective genes. RESULTS: Statin treatment during adipocyte differentiation caused a reduction in key genes involved in adipogenesis, such as ADIPOQ, GLUT4 and ABCG1. Using Illumina's Infinium '850K' Methylation EPIC array, we found a significant hypomethylation of cg14566882, located in the promoter of the histone deacetylase 9 (HDAC9) gene, in response to two types of statins (atorvastatin and mevastatin), which correlates with an increased HDAC9 mRNA expression. We confirmed that HDAC9 is a transcriptional repressor of the cholesterol efflux ABCG1 gene expression, which is epigenetically modified in obesity and prediabetic states. Thus, we assessed the putative impact of ABCG1 knockdown in mimicking the effect of statin in adipogenesis. ABCG1 KD reduced the expression of key genes involved in adipocyte differentiation and decreased insulin signalling and glucose uptake. In human blood cells from two cohorts, ABCG1 expression was impaired in response to statins, confirming that ABCG1 is targeted in vivo by these drugs. CONCLUSIONS: We identified an epigenetic link between adipogenesis and adipose tissue insulin resistance in the context of T2D risk associated with statin use, which has important implications as HDAC9 and ABCG1 are considered potential therapeutic targets for obesity and metabolic diseases.


Asunto(s)
Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 1/genética , Adipogénesis/efectos de los fármacos , Epigénesis Genética , Histona Desacetilasas/genética , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Proteínas Represoras/genética , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 1/sangre , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 1/fisiología , Adipogénesis/genética , Atorvastatina/farmacología , Línea Celular , Metilación de ADN , Histona Desacetilasas/metabolismo , Humanos , Insulina/fisiología , Lovastatina/análogos & derivados , Lovastatina/farmacología , Regiones Promotoras Genéticas , Proteínas Represoras/metabolismo , Células Madre/efectos de los fármacos , Células Madre/metabolismo
4.
Int J Obes (Lond) ; 44(2): 539-543, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31388097

RESUMEN

BACKGROUND: Genome-wide association studies (GWAS) have identified more than 250 loci associated with body mass index (BMI) and obesity. However, post-GWAS functional genomic investigations have been inadequate for understanding how these genetic loci physiologically impact disease development. METHODS: We performed a PCR-free expression assay targeting genes located nearby the GWAS-identified SNPs associated with BMI/obesity in a large panel of human tissues. Furthermore, we analyzed several genetic risk scores (GRS) summing GWAS-identified alleles associated with increased BMI in 4236 individuals. RESULTS: We found that the expression of BMI/obesity susceptibility genes was strongly enriched in the brain, especially in the insula (p = 4.7 × 10-9) and substantia nigra (p = 6.8 × 10-7), which are two brain regions involved in addiction and reward. Inversely, we found that top obesity/BMI-associated loci, including FTO, showed the strongest gene expression enrichment in the two brain regions. CONCLUSIONS: Our data suggest for the first time that the susceptibility genes for common obesity may have an effect on eating addiction and reward behaviors through their high expression in substantia nigra and insula, i.e., a different pattern from monogenic obesity genes that act in the hypothalamus and cause hyperphagia. Further epidemiological studies with relevant food behavior phenotypes are necessary to confirm these findings.


Asunto(s)
Conducta Adictiva/genética , Corteza Cerebral/metabolismo , Obesidad , Recompensa , Sustancia Negra/metabolismo , Adulto , Índice de Masa Corporal , Predisposición Genética a la Enfermedad/genética , Estudio de Asociación del Genoma Completo , Humanos , Hiperfagia , Persona de Mediana Edad , Obesidad/genética , Obesidad/metabolismo , Polimorfismo de Nucleótido Simple
5.
Diabetes ; 67(7): 1310-1321, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29728363

RESUMEN

In type 2 diabetes (T2D), hepatic insulin resistance is strongly associated with nonalcoholic fatty liver disease (NAFLD). In this study, we hypothesized that the DNA methylome of livers from patients with T2D compared with livers of individuals with normal plasma glucose levels can unveil some mechanism of hepatic insulin resistance that could link to NAFLD. Using DNA methylome and transcriptome analyses of livers from obese individuals, we found that hypomethylation at a CpG site in PDGFA (encoding platelet-derived growth factor α) and PDGFA overexpression are both associated with increased T2D risk, hyperinsulinemia, increased insulin resistance, and increased steatohepatitis risk. Genetic risk score studies and human cell modeling pointed to a causative effect of high insulin levels on PDGFA CpG site hypomethylation, PDGFA overexpression, and increased PDGF-AA secretion from the liver. We found that PDGF-AA secretion further stimulates its own expression through protein kinase C activity and contributes to insulin resistance through decreased expression of insulin receptor substrate 1 and of insulin receptor. Importantly, hepatocyte insulin sensitivity can be restored by PDGF-AA-blocking antibodies, PDGF receptor inhibitors, and by metformin, opening therapeutic avenues. Therefore, in the liver of obese patients with T2D, the increased PDGF-AA signaling contributes to insulin resistance, opening new therapeutic avenues against T2D and possibly NAFLD.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Resistencia a la Insulina , Hígado/metabolismo , Obesidad/metabolismo , Factor de Crecimiento Derivado de Plaquetas/genética , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Adulto , Estudios de Casos y Controles , Células Cultivadas , Metilación de ADN , Diabetes Mellitus Tipo 2/etiología , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/patología , Epigénesis Genética/fisiología , Femenino , Predisposición Genética a la Enfermedad , Humanos , Resistencia a la Insulina/genética , Hígado/patología , Masculino , Persona de Mediana Edad , Enfermedad del Hígado Graso no Alcohólico/genética , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/patología , Obesidad/complicaciones , Obesidad/genética , Obesidad/patología , Transducción de Señal/genética , Regulación hacia Arriba/genética
6.
Diabetes ; 66(1): 25-35, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27999105

RESUMEN

Hepatic DPP4 expression is elevated in subjects with ectopic fat accumulation in the liver. However, whether increased dipeptidyl peptidase 4 (DPP4) is involved in the pathogenesis or is rather a consequence of metabolic disease is not known. We therefore studied the transcriptional regulation of hepatic Dpp4 in young mice prone to diet-induced obesity. Already at 6 weeks of age, expression of hepatic Dpp4 was increased in mice with high weight gain, independent of liver fat content. In the same animals, methylation of four intronic CpG sites was decreased, amplifying glucose-induced transcription of hepatic Dpp4 In older mice, hepatic triglyceride content was increased only in animals with elevated Dpp4 expression. Expression and release of DPP4 were markedly higher in the liver compared with adipose depots. Analysis of human liver biopsy specimens revealed a correlation of DPP4 expression and DNA methylation to stages of hepatosteatosis and nonalcoholic steatohepatitis. In summary, our results indicate a crucial role of the liver in participation to systemic DPP4 levels. Furthermore, the data show that glucose-induced expression of Dpp4 in the liver is facilitated by demethylation of the Dpp4 gene early in life. This might contribute to early deteriorations in hepatic function, which in turn result in metabolic disease such as hepatosteatosis later in life.


Asunto(s)
Dipeptidil Peptidasa 4/genética , Dipeptidil Peptidasa 4/metabolismo , Hígado Graso/metabolismo , Hígado/metabolismo , Animales , Western Blotting , Línea Celular , Células Cultivadas , Islas de CpG/genética , Metilación de ADN/genética , Metilación de ADN/fisiología , Regulación de la Expresión Génica , Glucosa/metabolismo , Hepatocitos/metabolismo , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Triglicéridos/metabolismo
7.
J Clin Endocrinol Metab ; 101(11): 4244-4250, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27588439

RESUMEN

CONTEXT: Young-onset obesity is strongly associated with the early development of type 2 diabetes (T2D). Genetic risk scores (GRSs) related to T2D might help predicting the early impairment of glucose homeostasis in obese youths. OBJECTIVE: Our objective was to investigate the contributions of four GRSs (associated with: T2D [GRS-T2D], beta-cell function [GRS-ß], insulin resistance [GRS-IR], and body mass index) to the variation of traits derived from oral glucose tolerance test (OGTT) in obese and normal-weight children and young adults. DESIGN: This was a cross-sectional association study. PATIENTS: A total of 1076 obese children/adolescents (age = 11.4 ± 2.8 years) and 1265 normal-weight young volunteers (age = 21.1 ± 4.4 years) of European ancestry were recruited from pediatric obesity clinics and general population, respectively. INTERVENTION: Standard OGTT was the intervention in this study. MAIN OUTCOME MEASURES: Associations between GRSs and OGTT-derived traits including fasting glucose and insulin, insulinogenic index, insulin sensitivity index, disposition index (DI) and associations between GRSs and pre-diabetic conditions were measured. RESULTS: GRS-ß significantly associated with fasting glucose (ß = 0.019; P = 3.5 × 10-4) and DI (ß = -0.031; P = 8.9 × 10-4, last quartile 18% lower than first) in obese children, and nominally associated with fasting glucose (ß = 0.009; P = 0.017) and DI (ß = -0.030; P = 1.1 × 10-3, last quartile 11% lower than first) in normal-weight youths. GRS-T2D showed weaker contribution to fasting glucose and DI compared to GRS-ß, in both obese and normal-weight youths. GRS associated with insulin resistance and GRS associated with body mass index did not associate with any traits. None of the GRSs associated with prediabetes, which affected only 4% of participants overall. CONCLUSION: Single nucleotide polymorphisms identified by genome-wide association studies to influence beta-cell function were associated with fasting glucose and indices of insulin secretion in youths, especially in obese children.


Asunto(s)
Glucemia/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Predisposición Genética a la Enfermedad/clasificación , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Obesidad Infantil/metabolismo , Adolescente , Adulto , Niño , Estudios de Cohortes , Estudios Transversales , Diabetes Mellitus Tipo 2/epidemiología , Diabetes Mellitus Tipo 2/genética , Francia/epidemiología , Estudio de Asociación del Genoma Completo , Prueba de Tolerancia a la Glucosa , Humanos , Secreción de Insulina , Italia/epidemiología , Masculino , Obesidad Infantil/epidemiología , Obesidad Infantil/genética , Polimorfismo de Nucleótido Simple , Medición de Riesgo/clasificación , Adulto Joven
8.
Am J Med Genet A ; 167A(11): 2720-6, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26109092

RESUMEN

The Prader-Willi syndrome (PWS) is caused by lack of expression of paternal allele of the 15q11.2-q13 region, due to deletions at paternal 15q11.2-q13 (<70%), maternal uniparental disomy of chromosome 15 (mat-UPD 15) (30%) or imprinting defects (1%). Hyperphagia, intellectual disabilities/behavioral disorders, neonatal hypotonia, and hypogonadism are cardinal features for PWS. Methylation sensitive PCR (MS-PCR) of the SNRPN locus, which assesses the presence of both the unmethylated (paternal) and the methylated (maternal) allele of 15q11.2-q13, is considered a sensitive reference technique for PWS diagnosis regardless of genetic subtype. We describe a 17-year-old girl with severe obesity, short stature, and intellectual disability, without hypogonadism and history of neonatal hypotonia, who was suspected to have an incomplete PWS. The MS-PCR showed a normal pattern with similar maternal and paternal electrophoretic bands. Afterwards, a SNP array showed the presence of iso-UPD 15, that is, UPD15 with two copies of the same chromosome 15, in about 50% of cells, suggesting a diagnosis of partial PWS due to mosaic maternal iso-UPD15 arisen as rescue of a post-fertilization error. A quantitative methylation analysis confirmed the presence of mosaic UPD15 in about 50% of cells. We propose that complete clinical criteria for PWS and MS-PCR should not be considered sensitive in suspecting and diagnosing partial PWS due to mosaic UPD15. In contrast, clinical suspicion based on less restrictive criteria followed by SNP array is a more powerful approach to diagnose atypical PWS due to UPD15 mosaicism.


Asunto(s)
Cromosomas Humanos Par 15/genética , Metilación de ADN/genética , Mosaicismo , Reacción en Cadena de la Polimerasa , Polimorfismo de Nucleótido Simple/genética , Síndrome de Prader-Willi/genética , Disomía Uniparental/genética , Adolescente , Adulto , Niño , Preescolar , Electroforesis en Gel de Agar , Femenino , Humanos , Lactante , Recién Nacido , Masculino , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas Nucleares snRNP/genética
9.
Diabetologia ; 57(8): 1601-10, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24893864

RESUMEN

AIMS/HYPOTHESIS: Genome-wide association studies have firmly established 65 independent European-derived loci associated with type 2 diabetes and 36 loci contributing to variations in fasting plasma glucose (FPG). Using individual data from the Data from an Epidemiological Study on the Insulin Resistance Syndrome (DESIR) prospective study, we evaluated the contribution of three genetic risk scores (GRS) to variations in metabolic traits, and to the incidence and prevalence of impaired fasting glycaemia (IFG) and type 2 diabetes. METHODS: Three GRS (GRS-1, 65 type 2 diabetes-associated single nucleotide polymorphisms [SNPs]; GRS-2, GRS-1 combined with 24 FPG-raising SNPs; and GRS-3, FPG-raising SNPs alone) were analysed in 4,075 DESIR study participants. GRS-mediated effects on longitudinal variations in quantitative traits were assessed in 3,927 nondiabetic individuals using multivariate linear mixed models, and on the incidence and prevalence of hyperglycaemia at 9 years using Cox and logistic regression models. The contribution of each GRS to risk prediction was evaluated using the C-statistic and net reclassification improvement (NRI) analysis. RESULTS: The two most inclusive GRS were significantly associated with increased FPG (ß = 0.0011 mmol/l per year per risk allele, p GRS-1 = 8.2 × 10(-5) and p GRS-2 = 6.0 × 10(-6)), increased incidence of IFG and type 2 diabetes (per allele: HR GRS-1 1.03, p = 4.3 × 10(-9) and HR GRS-2 1.04, p = 1.0 × 10(-16)), and the 9 year prevalence (OR GRS-1 1.13 [95% CI 1.10, 1.17], p = 1.9 × 10(-14) for type 2 diabetes only; OR GRS-2 1.07 [95% CI 1.05, 1.08], p = 7.8 × 10(-25), for IFG and type 2 diabetes). No significant interaction was found between GRS-1 or GRS-2 and potential confounding factors. Each GRS yielded a modest, but significant, improvement in overall reclassification rates (NRI GRS-1 17.3%, p = 6.6 × 10(-7); NRI GRS-2 17.6%, p = 4.2 × 10(-7); NRI GRS-3 13.1%, p = 1.7 × 10(-4)). CONCLUSIONS/INTERPRETATION: Polygenic scores based on combined genetic information from type 2 diabetes risk and FPG variation contribute to discriminating middle-aged individuals at risk of developing type 2 diabetes in a general population.


Asunto(s)
Glucemia/análisis , Diabetes Mellitus Tipo 2/genética , Predisposición Genética a la Enfermedad , Homeostasis/genética , Polimorfismo de Nucleótido Simple , Adulto , Anciano , Alelos , Diabetes Mellitus Tipo 2/sangre , Femenino , Estudio de Asociación del Genoma Completo , Genotipo , Humanos , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Población Blanca/genética
10.
PLoS One ; 9(4): e92684, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24710396

RESUMEN

The "mechanistic target of rapamycin" (mTOR) is a central controller of growth, proliferation and/or motility of various cell-types ranging from adipocytes to immune cells, thereby linking metabolism and immunity. mTOR signaling is overactivated in obesity, promoting inflammation and insulin resistance. Therefore, great interest exists in the development of mTOR inhibitors as therapeutic drugs for obesity or diabetes. However, despite a plethora of studies characterizing the metabolic consequences of mTOR inhibition in rodent models, its impact on immune changes associated with the obese condition has never been questioned so far. To address this, we used a mouse model of high-fat diet (HFD)-fed mice with and without pharmacologic mTOR inhibition by rapamycin. Rapamycin was weekly administrated to HFD-fed C57BL/6 mice for 22 weeks. Metabolic effects were determined by glucose and insulin tolerance tests and by indirect calorimetry measures of energy expenditure. Inflammatory response and immune cell populations were characterized in blood, adipose tissue and liver. In parallel, the activities of both mTOR complexes (e. g. mTORC1 and mTORC2) were determined in adipose tissue, muscle and liver. We show that rapamycin-treated mice are leaner, have enhanced energy expenditure and are protected against insulin resistance. These beneficial metabolic effects of rapamycin were associated to significant changes of the inflammatory profiles of both adipose tissue and liver. Importantly, immune cells with regulatory functions such as regulatory T-cells (Tregs) and myeloid-derived suppressor cells (MDSCs) were increased in adipose tissue. These rapamycin-triggered metabolic and immune effects resulted from mTORC1 inhibition whilst mTORC2 activity was intact. Taken together, our results reinforce the notion that controlling immune regulatory cells in metabolic tissues is crucial to maintain a proper metabolic status and, more generally, comfort the need to search for novel pharmacological inhibitors of the mTOR signaling pathway to prevent and/or treat metabolic diseases.


Asunto(s)
Grasas de la Dieta/efectos adversos , Inmunosupresores/farmacología , Células Mieloides/inmunología , Obesidad/inmunología , Sirolimus/farmacología , Linfocitos T Reguladores/inmunología , Tejido Adiposo/inmunología , Tejido Adiposo/patología , Animales , Proliferación Celular/efectos de los fármacos , Grasas de la Dieta/farmacología , Modelos Animales de Enfermedad , Femenino , Resistencia a la Insulina/inmunología , Hígado/inmunología , Hígado/patología , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina , Diana Mecanicista del Complejo 2 de la Rapamicina , Ratones , Complejos Multiproteicos/inmunología , Células Mieloides/patología , Obesidad/inducido químicamente , Obesidad/patología , Transducción de Señal/efectos de los fármacos , Linfocitos T Reguladores/patología , Serina-Treonina Quinasas TOR/inmunología
11.
Nat Genet ; 45(9): 1040-3, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23852171

RESUMEN

Large chromosomal clonal mosaic events (CMEs) have been suggested to be linked to aging and to predict cancer. Type 2 diabetes (T2D) has been conceptualized as an accelerated-aging disease and is associated with higher prevalence of cancers. Here we aimed to assess the association between T2D and CME occurrence in blood. We evaluated the presence of CMEs in 7,659 individuals (including 2,208 with T2D) using DNA arrays. A significant association between CME occurrence and T2D was found (odds ratio (OR) = 5.3; P = 5.1 × 10(-5)) and was stronger when we only considered non-obese individuals with T2D (OR = 5.6; P = 4.9 × 10(-5)). Notably, CME carriers with T2D had higher prevalence of vascular complications than non-carriers with T2D (71.4% versus 37.1%, respectively; P = 7.7 × 10(-4)). In CME carriers, we found an increase in the percentage of abnormal cells over 6 years (P = 8.60 × 10(-3)). In conclusion, given the increased risk of cancer in CME carriers, our results may have profound clinical implications in patients with severe T2D.


Asunto(s)
Diabetes Mellitus Tipo 2/genética , Angiopatías Diabéticas/genética , Estudios de Asociación Genética , Mosaicismo , Anciano , Índice de Masa Corporal , Estudios de Casos y Controles , Aberraciones Cromosómicas , Mapeo Cromosómico , Diabetes Mellitus Tipo 2/complicaciones , Femenino , Humanos , Masculino , Persona de Mediana Edad , Oportunidad Relativa , Estudios Prospectivos
12.
J Mol Med (Berl) ; 91(9): 1109-15, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23640704

RESUMEN

We investigated the effect of 24 obesity-predisposing single nucleotide polymorphisms (SNPs), separately and in combination, on snacking behavior in three European populations. The 24 SNPs were genotyped in 7,502 subjects (1,868 snackers and 5,634 non-snackers). We tested the hypothesis that obesity risk variants or a genetic risk score increases snacking using a logistic regression adjusted for sex, age, and body mass index. The obesity genetic risk score was not associated with snacking (odds ratio (OR) = 1.00 [0.98-1.02], P value = 0.48). The obesity risk variants of two SNPs (rs925946 and rs7498665) close to the BDNF and SH2B1 genes showed nominal evidence of association with increased snacking (OR = 1.09 [1.01-1.17], P value = 0.0348 and OR = 1.11 [1.04-1.19], P value = 0.00703, respectively) but did not survive Bonferroni corrections for multiple testing. The associations of rs925946 and rs7498665 obesity risk variants with increased BMI (ß = 0.180 [0.022-0.339], P value = 0.0258 and ß = 0.166 [0.019-0.313], P value = 0.0271, respectively) were slightly attenuated after adjusting for snacking (ß = 0.151 [-0.006 to 0.309], P value = 0.0591 and ß = 0.152 [0.006-0.297], P value = 0.0413). Our data suggest that genetic predisposition to obesity does not significantly contribute to snacking behavior. The nominal associations of rs925946 and rs7498665 obesity risk variants near the BDNF and SH2B1 genes with increased snacking deserve further investigation.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Factor Neurotrófico Derivado del Encéfalo/genética , Obesidad/genética , Bocadillos/fisiología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Niño , Femenino , Predisposición Genética a la Enfermedad , Variación Genética , Genotipo , Humanos , Masculino , Persona de Mediana Edad , Obesidad/epidemiología , Oportunidad Relativa , Fenotipo , Polimorfismo de Nucleótido Simple , Población Blanca/genética , Adulto Joven
13.
PLoS One ; 7(11): e49919, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23209618

RESUMEN

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.


Asunto(s)
Obesidad/epidemiología , Riesgo , Adolescente , Adulto , Peso al Nacer , Índice de Masa Corporal , Niño , Estudios de Cohortes , Femenino , Finlandia/epidemiología , Humanos , Modelos Logísticos , Masculino , Persona de Mediana Edad , Obesidad/prevención & control , Población Blanca , Adulto Joven
14.
Diabetes ; 61(2): 524-30, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22210315

RESUMEN

It has recently been suggested that the low-frequency c.136-14_136-13insC variant in high-mobility group A1 (HMGA1) may strongly contribute to insulin resistance and type 2 diabetes risk. In our study, we attempted to confirm that HMGA1 is a novel type 2 diabetes locus in French Caucasians. The gene was sequenced in 368 type 2 diabetic case subjects with a family history of type 2 diabetes and 372 normoglycemic control subjects without a family history of type 2 diabetes. None of the 41 genetic variations identified were associated with type 2 diabetes. The lack of association between the c.136-14_136-13insC variant and type 2 diabetes was confirmed in an independent French group of 4,538 case subjects and 4,015 control subjects and in a large meta-analysis of 16,605 case subjects and 46,179 control subjects. Finally, this variant had no effects on metabolic traits and was not involved in variations of HMGA1 and insulin receptor (INSR) expressions. The c.136-14_136-13insC variant was not associated with type 2 diabetes in individuals of European descent. Our study emphasizes the need to analyze a large number of subjects to reliably assess the association of low-frequency variants with the disease.


Asunto(s)
Diabetes Mellitus Tipo 2/genética , Proteína HMGA1a/genética , Diabetes Mellitus Tipo 2/etiología , Estudio de Asociación del Genoma Completo , Genotipo , Humanos , Resistencia a la Insulina , Receptor de Insulina/genética , Riesgo
15.
Obesity (Silver Spring) ; 20(2): 389-95, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21720444

RESUMEN

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.


Asunto(s)
Proteínas de Ciclo Celular/genética , Proteínas de Unión al ADN/genética , Proteínas de la Membrana/genética , Neuropéptidos/genética , Obesidad/genética , Polimorfismo de Nucleótido Simple , Proteínas Tirosina Fosfatasas/genética , Proteínas/genética , Receptor de Melanocortina Tipo 4/genética , Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato , Proteínas de Unión al Calcio , Estudios de Casos y Controles , Femenino , Estudios de Seguimiento , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Genotipo , Grecia/epidemiología , Humanos , Masculino , Proteínas de Microfilamentos , Persona de Mediana Edad , Obesidad/epidemiología , Factores de Riesgo
16.
J Biol Chem ; 286(32): 28414-24, 2011 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-21592955

RESUMEN

Krüppel-like transcription factors (KLFs) have elicited significant attention because of their regulation of essential biochemical pathways and, more recently, because of their fundamental role in the mechanisms of human diseases. Neonatal diabetes mellitus is a monogenic disorder with primary alterations in insulin secretion. We here describe a key biochemical mechanism that underlies neonatal diabetes mellitus insulin biosynthesis impairment, namely a homozygous mutation within the insulin gene (INS) promoter, c.-331C>G, which affects a novel KLF-binding site. The combination of careful expression profiling, electromobility shift assays, reporter experiments, and chromatin immunoprecipitation demonstrates that, among 16 different KLF proteins tested, KLF11 is the most reliable activator of this site. Congruently, the c.-331C>G INS mutation fails to bind KLF11, thus inhibiting activation by this transcription factor. Klf11(-/-) mice recapitulate the disruption in insulin production and blood levels observed in patients. Thus, these data demonstrate an important role for KLF11 in the regulation of INS transcription via the novel c.-331 KLF site. Lastly, our screening data raised the possibility that other members of the KLF family may also regulate this promoter under distinct, yet unidentified, cellular contexts. Collectively, this study underscores a key role for KLF proteins in biochemical mechanisms of human diseases, in particular, early infancy onset diabetes mellitus.


Asunto(s)
Proteínas de Ciclo Celular , Proteínas de Unión al ADN , Diabetes Mellitus , Enfermedades del Recién Nacido , Células Secretoras de Insulina , Insulina , Mutagénesis Insercional , Proteínas Represoras , Elementos de Respuesta/genética , Transactivadores , Factores de Transcripción , Adulto , Animales , Proteínas Reguladoras de la Apoptosis , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Línea Celular , Preescolar , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Diabetes Mellitus/genética , Diabetes Mellitus/metabolismo , Femenino , Humanos , Lactante , Recién Nacido , Enfermedades del Recién Nacido/genética , Enfermedades del Recién Nacido/metabolismo , Enfermedades del Recién Nacido/patología , Insulina/biosíntesis , Insulina/genética , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patología , Masculino , Ratones , Ratones Noqueados , Ratas , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Transactivadores/genética , Transactivadores/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
17.
Proc Natl Acad Sci U S A ; 108(17): 7119-24, 2011 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-21471458

RESUMEN

Alcohol consumption is a moderately heritable trait, but the genetic basis in humans is largely unknown, despite its clinical and societal importance. We report a genome-wide association study meta-analysis of ∼2.5 million directly genotyped or imputed SNPs with alcohol consumption (gram per day per kilogram body weight) among 12 population-based samples of European ancestry, comprising 26,316 individuals, with replication genotyping in an additional 21,185 individuals. SNP rs6943555 in autism susceptibility candidate 2 gene (AUTS2) was associated with alcohol consumption at genome-wide significance (P = 4 × 10(-8) to P = 4 × 10(-9)). We found a genotype-specific expression of AUTS2 in 96 human prefrontal cortex samples (P = 0.026) and significant (P < 0.017) differences in expression of AUTS2 in whole-brain extracts of mice selected for differences in voluntary alcohol consumption. Down-regulation of an AUTS2 homolog caused reduced alcohol sensitivity in Drosophila (P < 0.001). Our finding of a regulator of alcohol consumption adds knowledge to our understanding of genetic mechanisms influencing alcohol drinking behavior.


Asunto(s)
Consumo de Bebidas Alcohólicas/genética , Polimorfismo de Nucleótido Simple , Proteínas/genética , Carácter Cuantitativo Heredable , Población Blanca/genética , Consumo de Bebidas Alcohólicas/metabolismo , Animales , Proteínas del Citoesqueleto , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Femenino , Regulación de la Expresión Génica/genética , Estudio de Asociación del Genoma Completo , Genotipo , Humanos , Masculino , Ratones , Proteínas Nucleares/biosíntesis , Proteínas Nucleares/genética , Proteínas/metabolismo , Factores de Transcripción
18.
PLoS One ; 5(10): e13630, 2010 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-21049026

RESUMEN

BACKGROUND: Accurate molecular diagnosis of monogenic non-autoimmune neonatal diabetes mellitus (NDM) is critical for patient care, as patients carrying a mutation in KCNJ11 or ABCC8 can be treated by oral sulfonylurea drugs instead of insulin therapy. This diagnosis is currently based on Sanger sequencing of at least 42 PCR fragments from the KCNJ11, ABCC8, and INS genes. Here, we assessed the feasibility of using the next-generation whole exome sequencing (WES) for the NDM molecular diagnosis. METHODOLOGY/PRINCIPAL FINDINGS: We carried out WES for a patient presenting with permanent NDM, for whom mutations in KCNJ11, ABCC8 and INS and abnormalities in chromosome 6q24 had been previously excluded. A solution hybridization selection was performed to generate WES in 76 bp paired-end reads, by using two channels of the sequencing instrument. WES quality was assessed using a high-resolution oligonucleotide whole-genome genotyping array. From our WES with high-quality reads, we identified a novel non-synonymous mutation in ABCC8 (c.1455G>C/p.Q485H), despite a previous negative sequencing of this gene. This mutation, confirmed by Sanger sequencing, was not present in 348 controls and in the patient's mother, father and young brother, all of whom are normoglycemic. CONCLUSIONS/SIGNIFICANCE: WES identified a novel de novo ABCC8 mutation in a NDM patient. Compared to the current Sanger protocol, WES is a comprehensive, cost-efficient and rapid method to identify mutations in NDM patients. We suggest WES as a near future tool of choice for further molecular diagnosis of NDM cases, negative for chr6q24, KCNJ11 and INS abnormalities.


Asunto(s)
Diabetes Mellitus/diagnóstico , Exones , Enfermedades del Recién Nacido/diagnóstico , Técnicas de Diagnóstico Molecular , Cromosomas Humanos Par 6 , Diabetes Mellitus/genética , Humanos , Recién Nacido , Enfermedades del Recién Nacido/genética , Mutación , Polimorfismo de Nucleótido Simple
19.
J Hum Genet ; 55(4): 227-31, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20186155

RESUMEN

Genetic and environmental factors are involved in insulin resistance (IR). IR and dyslipidemia associate with increased risk of cardiovascular diseases. Plasma low-density lipoprotein cholesterol (LDL-C) level is a marker of cardiovascular risk. In a Caucasian general population we aimed at determining the multifactorial components of LDL-C levels using 10 genes and 3 phenotypes. In the PPARG, UCP3, ADIPOQ, TNF, LIPC, CARTPT, PCSK9, SCAP, SCARB1 and ENPP1 genes known to be associated with IR or dyslipidemia we genotyped 19 single nucleotide polymorphisms (SNPs) in 846 subjects. When several SNPs were genotyped for a given gene we constructed haplotypes. Including genetic and environmental variables (gender, body mass index (BMI) and adiponectin level) we used (1) the multifactor dimensionality reduction method to explain clusters of high and low LDL-C, and (2) the restricted partition method to explain LDL-C levels. Both methods showed that BMI and haplotypes at the ADIPOQ adiponectin encoding gene but not adiponectin level itself, were discriminant regarding to LDL-C. Subjects bearing an at-risk combination of BMI and ADIPOQ genotypes were prone to have a higher LDL-C (OR=3.13, 95% CI=2.20-4.46, P<0.0001). Our results suggest that in interaction with BMI, ADIPOQ haplotypes capture genetic variation(s) from neighboring gene(s) that would modulate LDL-C level.


Asunto(s)
Adiponectina/genética , Índice de Masa Corporal , LDL-Colesterol/sangre , Haplotipos/genética , Epistasis Genética , Femenino , Francia , Genética de Población , Genotipo , Humanos , Masculino , Herencia Multifactorial , Análisis Multivariante , Fenotipo , Polimorfismo de Nucleótido Simple , Población Blanca/genética
20.
Obesity (Silver Spring) ; 18(7): 1469-73, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19893502

RESUMEN

Minor allele A of single-nucleotide polymorphism (SNP) 11391 G/A of ADIPOQ gene (rs17300539) has been consistently associated with higher adiponectin levels in adults and children. The aim of this study was to investigate the metabolic role of this variant in a large cohort of children of European origin. A total of 1,852 children from two general populations in Verona and in Fleurbaix-Laventie and from the Lille childhood obesity cohort, were genotyped and pooled together after checking for the absence of genetic heterogeneity for rs17300539 between Italian and French children. The genotype of rs17300539 was studied in relation to circulating adiponectin levels, BMI, fasting plasma glucose, fasting serum insulin (FSI), insulin resistance index (homeostasis model assessment of insulin resistance (HOMA(IR))), high-density lipoprotein cholesterol, and triglycerides. After adjustment for known confounders, rs17300539 GA+AA carriers had 1.6 microg/ml higher adiponectin levels (P = 6 x 10(-8)) than GG carriers. They also showed higher BMI (B = 0.97, P = 0.015) and higher prevalence of obesity (OR = 1.35 (1.06-1.85), P = 0.015) than GG carriers. Before adjusting for obesity status, GA+AA carriers had higher FSI (B = 1.10, P = 0.040) and higher HOMA(IR) (B = 0.31, P = 0.020) than GG carriers. After adjustment for obesity status, they did not differ from GG carriers for any metabolic parameter, either among obese or nonobese children. The rs17300539-A variant, though consistently associated with higher adiponectin levels, does not exert any appreciable protective metabolic effect in children, either in the presence or absence of obesity. In contrast, this SNP may increase the risk for childhood obesity and related insulin resistance.


Asunto(s)
Obesidad/etnología , Obesidad/genética , Polimorfismo de Nucleótido Simple , Población Blanca/estadística & datos numéricos , Adiponectina/sangre , Adiponectina/genética , Adolescente , Niño , HDL-Colesterol/sangre , Femenino , Genotipo , Homeostasis/genética , Humanos , Resistencia a la Insulina/genética , Masculino , Obesidad/metabolismo , Factores de Riesgo , Triglicéridos/sangre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA