Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Epigenetics ; 17(10): 1219-1233, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-34818986

RESUMO

Vitamin B12 has multiple biochemical functions including in the one-carbon cycle generating a methyl group for DNA methylation, and metabolism of fatty acids and amino acids to generate energy via the citric acid cycle. The aim of our study was to use a combined epigenomic and transcriptomic approach to identify novel genes mediating the effect of B12 on adipogenesis.Human pre-adipocytes (CHUB-S7) were treated with a range of B12 (0-500 nM) concentrations from the day of cell seeding until harvesting in discovery and validation experiments prior to genome-wide methylation analysis using the Illumina HumanMethylation 450Beadchip. For transcriptomic analysis, RNA-seq libraries were run on the Illumina HiSeq 2500. To further investigate the expression of any genes on human adipogenesis, a second human preadipocyte strain was studied (SGBS) by real-time quantitative PCR (qRT-PCR).A combined epigenetic and transcriptomic approach in differentiated human pre-adipocyte cell line, CHUB-S7, identified that the Human cartilage chitinase 3-like protein 2 (CHI3L2) gene was hypo-methylated and had increased expression in low B12 conditions. Furthermore, there was an approximately 1000-fold increase in CHI3L2 expression in the early days of adipocyte differentiation, which paralleled an increase of lipid droplets in differentiated SGBS cells and an increased expression level of markers of mature adipocytes.In summary, we have identified a potential role of the human cartilage chitinase 3-like protein 2 (CHI3L2) in adipocyte function in the presence of low B12 levels.


Assuntos
Quitinases , Adipócitos/metabolismo , Adipogenia/genética , Aminoácidos/genética , Aminoácidos/metabolismo , Aminoácidos/farmacologia , Cartilagem/metabolismo , Diferenciação Celular/genética , Quitinases/genética , Quitinases/metabolismo , Quitinases/farmacologia , Metilação de DNA , Ácidos Graxos/metabolismo , Ácidos Graxos/farmacologia , Humanos , Transcriptoma , Vitamina B 12/metabolismo , Vitamina B 12/farmacologia
2.
Handb Clin Neurol ; 181: 369-379, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34238471

RESUMO

Prader-Willi syndrome (PWS) is a complex neurodevelopmental disorder, arising from a loss of paternity expressed genetic material on the imprinted chromosome locus 15q11-q13. Despite increasing clarity on the underlying genetic defects, the molecular basis of the condition remains poorly understood. Hypothalamic dysfunction is widely recognized as the basis of the core symptoms of PWS, which include a deficiency in growth hormone and reproductive hormones, circadian rhythm abnormalities, and a lack of satiety, leading to an extreme obesity, among others. Genome-wide gene expression analysis (transcriptomics) offers an unbiased interrogation of complex disease pathogenesis and a potential window into the dysregulated pathways involved in disease. In this chapter, we review the findings from recent work investigating the PWS hypothalamic transcriptome, discuss the significance of the findings in relation to the clinical presentation and molecular underpinnings of PWS, and highlight future research directions.


Assuntos
Síndrome de Prader-Willi , Transcriptoma , Genoma , Humanos , Hipotálamo , Obesidade , Síndrome de Prader-Willi/genética
3.
Cell Metab ; 31(6): 1107-1119.e12, 2020 06 02.
Artigo em Inglês | MEDLINE | ID: mdl-32492392

RESUMO

Obesity is genetically heterogeneous with monogenic and complex polygenic forms. Using exome and targeted sequencing in 2,737 severely obese cases and 6,704 controls, we identified three genes (PHIP, DGKI, and ZMYM4) with an excess burden of very rare predicted deleterious variants in cases. In cells, we found that nuclear PHIP (pleckstrin homology domain interacting protein) directly enhances transcription of pro-opiomelanocortin (POMC), a neuropeptide that suppresses appetite. Obesity-associated PHIP variants repressed POMC transcription. Our demonstration that PHIP is involved in human energy homeostasis through transcriptional regulation of central melanocortin signaling has potential diagnostic and therapeutic implications for patients with obesity and developmental delay. Additionally, we found an excess burden of predicted deleterious variants involving genes nearest to loci from obesity genome-wide association studies. Genes and gene sets influencing obesity with variable penetrance provide compelling evidence for a continuum of causality in the genetic architecture of obesity, and explain some of its missing heritability.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/genética , Obesidade Infantil/genética , Pró-Opiomelanocortina/genética , Adulto , Animais , Células Cultivadas , Criança , Chlorocebus aethiops , Exoma , Feminino , Variação Genética/genética , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade
4.
Nat Commun ; 10(1): 1718, 2019 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-30979869

RESUMO

Hypothalamic neurons expressing the anorectic peptide Pro-opiomelanocortin (Pomc) regulate food intake and body weight. Here, we show that Steroid Receptor Coactivator-1 (SRC-1) interacts with a target of leptin receptor activation, phosphorylated STAT3, to potentiate Pomc transcription. Deletion of SRC-1 in Pomc neurons in mice attenuates their depolarization by leptin, decreases Pomc expression and increases food intake leading to high-fat diet-induced obesity. In humans, fifteen rare heterozygous variants in SRC-1 found in severely obese individuals impair leptin-mediated Pomc reporter activity in cells, whilst four variants found in non-obese controls do not. In a knock-in mouse model of a loss of function human variant (SRC-1L1376P), leptin-induced depolarization of Pomc neurons and Pomc expression are significantly reduced, and food intake and body weight are increased. In summary, we demonstrate that SRC-1 modulates the function of hypothalamic Pomc neurons, and suggest that targeting SRC-1 may represent a useful therapeutic strategy for weight loss.


Assuntos
Hipotálamo/metabolismo , Neurônios/metabolismo , Coativador 1 de Receptor Nuclear/genética , Coativador 1 de Receptor Nuclear/metabolismo , Obesidade/genética , Alelos , Animais , Peso Corporal , Linhagem Celular Tumoral , Cruzamentos Genéticos , Deleção de Genes , Técnicas de Introdução de Genes , Variação Genética , Células HEK293 , Heterozigoto , Homeostase , Humanos , Leptina/metabolismo , Masculino , Potenciais da Membrana , Camundongos , Camundongos Transgênicos , Mutação de Sentido Incorreto , Obesidade/metabolismo , Fenótipo
5.
Cell ; 176(4): 729-742.e18, 2019 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-30661757

RESUMO

Hypothalamic melanocortin neurons play a pivotal role in weight regulation. Here, we examined the contribution of Semaphorin 3 (SEMA3) signaling to the development of these circuits. In genetic studies, we found 40 rare variants in SEMA3A-G and their receptors (PLXNA1-4; NRP1-2) in 573 severely obese individuals; variants disrupted secretion and/or signaling through multiple molecular mechanisms. Rare variants in this set of genes were significantly enriched in 982 severely obese cases compared to 4,449 controls. In a zebrafish mutagenesis screen, deletion of 7 genes in this pathway led to increased somatic growth and/or adiposity demonstrating that disruption of Semaphorin 3 signaling perturbs energy homeostasis. In mice, deletion of the Neuropilin-2 receptor in Pro-opiomelanocortin neurons disrupted their projections from the arcuate to the paraventricular nucleus, reduced energy expenditure, and caused weight gain. Cumulatively, these studies demonstrate that SEMA3-mediated signaling drives the development of hypothalamic melanocortin circuits involved in energy homeostasis.


Assuntos
Metabolismo Energético/genética , Melanocortinas/metabolismo , Semaforinas/genética , Adolescente , Adulto , Animais , Peso Corporal , Linhagem Celular , Criança , Pré-Escolar , Modelos Animais de Doenças , Ingestão de Alimentos , Feminino , Variação Genética/genética , Homeostase , Humanos , Hipotálamo/metabolismo , Leptina/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Obesidade/genética , Obesidade/metabolismo , Receptores de Superfície Celular/metabolismo , Semaforinas/metabolismo , Adulto Jovem , Peixe-Zebra
6.
Cell Rep ; 22(13): 3401-3408, 2018 03 27.
Artigo em Inglês | MEDLINE | ID: mdl-29590610

RESUMO

Transcriptional analysis of brain tissue from people with molecularly defined causes of obesity may highlight disease mechanisms and therapeutic targets. We performed RNA sequencing of hypothalamus from individuals with Prader-Willi syndrome (PWS), a genetic obesity syndrome characterized by severe hyperphagia. We found that upregulated genes overlap with the transcriptome of mouse Agrp neurons that signal hunger, while downregulated genes overlap with the expression profile of Pomc neurons activated by feeding. Downregulated genes are expressed mainly in neuronal cells and contribute to neurogenesis, neurotransmitter release, and synaptic plasticity, while upregulated, predominantly microglial genes are involved in inflammatory responses. This transcriptional signature may be mediated by reduced brain-derived neurotrophic factor expression. Additionally, we implicate disruption of alternative splicing as a potential molecular mechanism underlying neuronal dysfunction in PWS. Transcriptomic analysis of the human hypothalamus may identify neural mechanisms involved in energy homeostasis and potential therapeutic targets for weight loss.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/deficiência , Jejum/fisiologia , Hipotálamo/metabolismo , Síndrome de Prader-Willi/genética , Síndrome de Prader-Willi/metabolismo , Animais , Fator Neurotrófico Derivado do Encéfalo/genética , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Humanos , Camundongos , Obesidade/metabolismo , Síndrome de Prader-Willi/patologia , Transcriptoma
7.
Mol Metab ; 6(11): 1419-1428, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-29107289

RESUMO

OBJECTIVE: Genetic studies in obese rodents and humans can provide novel insights into the mechanisms involved in energy homeostasis. METHODS: In this study, we genetically mapped the chromosomal region underlying the development of severe obesity in a mouse line identified as part of a dominant N-ethyl-N-nitrosourea (ENU) mutagenesis screen. We characterized the metabolic and behavioral phenotype of obese mutant mice and examined changes in hypothalamic gene expression. In humans, we examined genetic data from people with severe early onset obesity. RESULTS: We identified an obese mouse heterozygous for a missense mutation (pR108W) in orthopedia homeobox (Otp), a homeodomain containing transcription factor required for the development of neuroendocrine cell lineages in the hypothalamus, a region of the brain important in the regulation of energy homeostasis. OtpR108W/+ mice exhibit increased food intake, weight gain, and anxiety when in novel environments or singly housed, phenotypes that may be partially explained by reduced hypothalamic expression of oxytocin and arginine vasopressin. R108W affects the highly conserved homeodomain, impairs DNA binding, and alters transcriptional activity in cells. We sequenced OTP in 2548 people with severe early-onset obesity and found a rare heterozygous loss of function variant in the homeodomain (Q153R) in a patient who also had features of attention deficit disorder. CONCLUSIONS: OTP is involved in mammalian energy homeostasis and behavior and appears to be necessary for the development of hypothalamic neural circuits. Further studies will be needed to investigate the contribution of rare variants in OTP to human energy homeostasis.


Assuntos
Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Sequência de Aminoácidos , Animais , Ansiedade/metabolismo , Sequência de Bases , Encéfalo/metabolismo , Mapeamento Cromossômico , Bases de Dados Genéticas , Feminino , Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento/genética , Genes Homeobox , Proteínas de Homeodomínio/fisiologia , Humanos , Hipotálamo/metabolismo , Masculino , Camundongos , Proteínas do Tecido Nervoso/fisiologia , Sistemas Neurossecretores/metabolismo , Obesidade/metabolismo , Fatores de Transcrição/genética , Transcriptoma/genética
8.
Sci Rep ; 7(1): 4394, 2017 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-28663568

RESUMO

Obesity is a genetically heterogeneous disorder. Using targeted and whole-exome sequencing, we studied 32 human and 87 rodent obesity genes in 2,548 severely obese children and 1,117 controls. We identified 52 variants contributing to obesity in 2% of cases including multiple novel variants in GNAS, which were sometimes found with accelerated growth rather than short stature as described previously. Nominally significant associations were found for rare functional variants in BBS1, BBS9, GNAS, MKKS, CLOCK and ANGPTL6. The p.S284X variant in ANGPTL6 drives the association signal (rs201622589, MAF~0.1%, odds ratio = 10.13, p-value = 0.042) and results in complete loss of secretion in cells. Further analysis including additional case-control studies and population controls (N = 260,642) did not support association of this variant with obesity (odds ratio = 2.34, p-value = 2.59 × 10-3), highlighting the challenges of testing rare variant associations and the need for very large sample sizes. Further validation in cohorts with severe obesity and engineering the variants in model organisms will be needed to explore whether human variants in ANGPTL6 and other genes that lead to obesity when deleted in mice, do contribute to obesity. Such studies may yield druggable targets for weight loss therapies.


Assuntos
Estudos de Associação Genética , Predisposição Genética para Doença , Variação Genética , Obesidade Mórbida/genética , Obesidade Infantil/genética , Animais , Estudos de Casos e Controles , Cromograninas/química , Cromograninas/genética , Cromograninas/metabolismo , Feminino , Subunidades alfa Gs de Proteínas de Ligação ao GTP/química , Subunidades alfa Gs de Proteínas de Ligação ao GTP/genética , Subunidades alfa Gs de Proteínas de Ligação ao GTP/metabolismo , Humanos , Masculino , Camundongos , Modelos Moleculares , Mutação , Obesidade Mórbida/diagnóstico , Razão de Chances , Obesidade Infantil/diagnóstico , Linhagem , Conformação Proteica , Roedores
9.
J Clin Invest ; 126(3): 992-6, 2016 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-26854926

RESUMO

Selenium is a trace element that is essential for human health and is incorporated into more than 25 human selenocysteine-containing (Sec-containing) proteins via unique Sec-insertion machinery that includes a specific, nuclear genome-encoded, transfer RNA (tRNA[Ser]Sec). Here, we have identified a human tRNA[Ser]Sec mutation in a proband who presented with a variety of symptoms, including abdominal pain, fatigue, muscle weakness, and low plasma levels of selenium. This mutation resulted in a marked reduction in expression of stress-related, but not housekeeping, selenoproteins. Evaluation of primary cells from the homozygous proband and a heterozygous parent indicated that the observed deficit in stress-related selenoprotein production is likely mediated by reduced expression and diminished 2'-O-methylribosylation at uridine 34 in mutant tRNA[Ser]Sec. Moreover, this methylribosylation defect was restored by cellular complementation with normal tRNA[Ser]Sec. This study identifies a tRNA mutation that selectively impairs synthesis of stress-related selenoproteins and demonstrates the importance of tRNA modification for normal selenoprotein synthesis.


Assuntos
Doenças Genéticas Inatas/diagnóstico , RNA de Transferência Aminoácido-Específico/genética , Selenoproteínas/genética , Sequência de Bases , Criança , Análise Mutacional de DNA , Estudos de Associação Genética , Doenças Genéticas Inatas/genética , Humanos , Masculino , Dados de Sequência Molecular , Mutação Puntual , Polimorfismo de Nucleotídeo Único , Biossíntese de Proteínas , Selenoproteínas/sangue , Selenoproteínas/deficiência
10.
Endocrinology ; 155(9): 3219-26, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24971614

RESUMO

We have previously reported rare variants in sarcoma (Src) homology 2 (SH2) B adaptor protein 1 (SH2B1) in individuals with obesity, insulin resistance, and maladaptive behavior. Here, we identify 4 additional SH2B1 variants by sequencing 500 individuals with severe early-onset obesity. SH2B1 has 4 alternatively spliced isoforms. One variant (T546A) lies within the N-terminal region common to all isoforms. As shown for past variants in this region, T546A impairs SH2B1ß enhancement of nerve growth factor-induced neurite outgrowth, and the individual with the T546A variant exhibits mild developmental delay. The other 3 variants (A663V, V695M, and A723V) lie in the C-terminal tail of SH2B1α. SH2B1α variant carriers were hyperinsulinemic but did not exhibit the behavioral phenotype observed in individuals with SH2B1 variants that disrupt all isoforms. In in vitro assays, SH2B1α, like SH2B1ß, enhances insulin- and leptin-induced insulin receptor substrate 2 (IRS2) phosphorylation and GH-induced cell motility. None of the variants affect SH2B1α enhancement of insulin- and leptin-induced IRS2 phosphorylation. However, T546A, A663V, and A723V all impair the ability of SH2B1α to enhance GH-induced cell motility. In contrast to SH2B1ß, SH2B1α does not enhance nerve growth factor-induced neurite outgrowth. These studies suggest that genetic variants that disrupt isoforms other than SH2B1ß may be functionally significant. Further studies are needed to understand the mechanism by which the individual isoforms regulate energy homeostasis and behavior.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Obesidade/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Adolescente , Adulto , Processamento Alternativo , Criança , Feminino , Humanos , Insulina/metabolismo , Leptina/metabolismo , Masculino , Mutação de Sentido Incorreto , Obesidade/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Transdução de Sinais , Adulto Jovem
11.
Cell ; 155(4): 765-77, 2013 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-24209692

RESUMO

Kinase suppressor of Ras 2 (KSR2) is an intracellular scaffolding protein involved in multiple signaling pathways. Targeted deletion of Ksr2 leads to obesity in mice, suggesting a role in energy homeostasis. We explored the role of KSR2 in humans by sequencing 2,101 individuals with severe early-onset obesity and 1,536 controls. We identified multiple rare variants in KSR2 that disrupt signaling through the Raf-MEKERK pathway and impair cellular fatty acid oxidation and glucose oxidation in transfected cells; effects that can be ameliorated by the commonly prescribed antidiabetic drug, metformin. Mutation carriers exhibit hyperphagia in childhood, low heart rate, reduced basal metabolic rate and severe insulin resistance. These data establish KSR2 as an important regulator of energy intake, energy expenditure, and substrate utilization in humans. Modulation of KSR2-mediated effects may represent a novel therapeutic strategy for obesity and type 2 diabetes.


Assuntos
Resistência à Insulina , Obesidade/genética , Proteínas Serina-Treonina Quinases/genética , Fatores Etários , Idade de Início , Sequência de Aminoácidos , Animais , Criança , Metabolismo Energético , Ácidos Graxos/metabolismo , Feminino , Glucose/metabolismo , Humanos , Hiperfagia/genética , Hiperfagia/metabolismo , Sistema de Sinalização das MAP Quinases , Masculino , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Obesidade/epidemiologia , Obesidade/metabolismo , Oxirredução , Proteínas Serina-Treonina Quinases/química , Proteínas Serina-Treonina Quinases/metabolismo , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas B-raf/química , Proteínas Proto-Oncogênicas B-raf/metabolismo , Alinhamento de Sequência
12.
Nat Genet ; 45(9): 1055-60, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23913004

RESUMO

At least 5% of individuals with hypertension have adrenal aldosterone-producing adenomas (APAs). Gain-of-function mutations in KCNJ5 and apparent loss-of-function mutations in ATP1A1 and ATP2A3 were reported to occur in APAs. We find that KCNJ5 mutations are common in APAs resembling cortisol-secreting cells of the adrenal zona fasciculata but are absent in a subset of APAs resembling the aldosterone-secreting cells of the adrenal zona glomerulosa. We performed exome sequencing of ten zona glomerulosa-like APAs and identified nine with somatic mutations in either ATP1A1, encoding the Na(+)/K(+) ATPase α1 subunit, or CACNA1D, encoding Cav1.3. The ATP1A1 mutations all caused inward leak currents under physiological conditions, and the CACNA1D mutations induced a shift of voltage-dependent gating to more negative voltages, suppressed inactivation or increased currents. Many APAs with these mutations were <1 cm in diameter and had been overlooked on conventional adrenal imaging. Recognition of the distinct genotype and phenotype for this subset of APAs could facilitate diagnosis.


Assuntos
Doenças do Córtex Suprarrenal/genética , Canais de Cálcio Tipo L/genética , Hipertensão/genética , Mutação , ATPase Trocadora de Sódio-Potássio/genética , Doenças do Córtex Suprarrenal/complicações , Doenças do Córtex Suprarrenal/diagnóstico , Substituição de Aminoácidos , Canais de Cálcio Tipo L/química , Canais de Cálcio Tipo L/metabolismo , Análise por Conglomerados , Feminino , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/genética , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/metabolismo , Perfilação da Expressão Gênica , Humanos , Hipertensão/diagnóstico , Hipertensão/etiologia , Masculino , Conformação Proteica , ATPase Trocadora de Sódio-Potássio/química , ATPase Trocadora de Sódio-Potássio/metabolismo
13.
J Clin Invest ; 123(7): 3042-50, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23778139

RESUMO

Single-minded 1 (SIM1) is a basic helix-loop-helix transcription factor involved in the development and function of the paraventricular nucleus of the hypothalamus. Obesity has been reported in Sim1 haploinsufficient mice and in a patient with a balanced translocation disrupting SIM1. We sequenced the coding region of SIM1 in 2,100 patients with severe, early onset obesity and in 1,680 controls. Thirteen different heterozygous variants in SIM1 were identified in 28 unrelated severely obese patients. Nine of the 13 variants significantly reduced the ability of SIM1 to activate a SIM1-responsive reporter gene when studied in stably transfected cells coexpressing the heterodimeric partners of SIM1 (ARNT or ARNT2). SIM1 variants with reduced activity cosegregated with obesity in extended family studies with variable penetrance. We studied the phenotype of patients carrying variants that exhibited reduced activity in vitro. Variant carriers exhibited increased ad libitum food intake at a test meal, normal basal metabolic rate, and evidence of autonomic dysfunction. Eleven of the 13 probands had evidence of a neurobehavioral phenotype. The phenotypic similarities between patients with SIM1 deficiency and melanocortin 4 receptor (MC4R) deficiency suggest that some of the effects of SIM1 deficiency on energy homeostasis are mediated by altered melanocortin signaling.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Mutação de Sentido Incorreto , Obesidade/genética , Proteínas Repressoras/genética , Adolescente , Estatura/genética , Estudos de Casos e Controles , Criança , Pré-Escolar , Análise Mutacional de DNA , Feminino , Expressão Gênica , Genes Reporter , Estudos de Associação Genética , Células HEK293 , Heterozigoto , Humanos , Lactente , Luciferases de Renilla/biossíntese , Luciferases de Renilla/genética , Masculino , Modelos Moleculares , Obesidade/patologia , Linhagem , Receptor Tipo 4 de Melanocortina/deficiência , Ativação Transcricional
14.
Mol Genet Metab ; 110(1-2): 191-4, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23800642

RESUMO

Non-synonymous mutations affecting both alleles of PCSK1 (proprotein convertase 1/3) are associated with obesity and impaired prohormone processing. We report a proband who was compound heterozygous for a maternally inherited frameshift mutation and a paternally inherited 474kb deletion that encompasses PCSK1, representing a novel genetic mechanism underlying this phenotype. Although pro-vasopressin is not a known physiological substrate of PCSK1, the development of central diabetes insipidus in this proband suggests that PCSK1 deficiency can be associated with impaired osmoregulation.


Assuntos
Diabetes Insípido/genética , Doenças do Sistema Endócrino/genética , Obesidade Mórbida/genética , Obesidade/genética , Pró-Proteína Convertase 1/deficiência , Pró-Proteína Convertases/genética , Alelos , Pré-Escolar , Diabetes Insípido/complicações , Diabetes Insípido/patologia , Doenças do Sistema Endócrino/complicações , Doenças do Sistema Endócrino/patologia , Heterozigoto , Humanos , Lactente , Mutação , Obesidade/complicações , Obesidade/patologia , Obesidade Mórbida/complicações , Obesidade Mórbida/patologia , Osmorregulação/genética , Fenótipo , Polimorfismo de Nucleotídeo Único , Pró-Proteína Convertase 1/genética
15.
Nat Genet ; 45(5): 513-7, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23563609

RESUMO

Common and rare variants associated with body mass index (BMI) and obesity account for <5% of the variance in BMI. We performed SNP and copy number variation (CNV) association analyses in 1,509 children with obesity at the extreme tail (>3 s.d. from the mean) of the BMI distribution and 5,380 controls. Evaluation of 29 SNPs (P < 1 × 10(-5)) in an additional 971 severely obese children and 1,990 controls identified 4 new loci associated with severe obesity (LEPR, PRKCH, PACS1 and RMST). A previously reported 43-kb deletion at the NEGR1 locus was significantly associated with severe obesity (P = 6.6 × 10(-7)). However, this signal was entirely driven by a flanking 8-kb deletion; absence of this deletion increased risk for obesity (P = 6.1 × 10(-11)). We found a significant burden of rare, single CNVs in severely obese cases (P < 0.0001). Integrative gene network pathway analysis of rare deletions indicated enrichment of genes affecting G protein-coupled receptors (GPCRs) involved in the neuronal regulation of energy homeostasis.


Assuntos
Variações do Número de Cópias de DNA/genética , Predisposição Genética para Doença , Genoma Humano , Estudo de Associação Genômica Ampla , Obesidade/genética , Polimorfismo de Nucleotídeo Único/genética , Locos de Características Quantitativas , Índice de Massa Corporal , Estudos de Casos e Controles , Criança , Genótipo , Humanos , Fenótipo
16.
PLoS One ; 8(3): e59061, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23533600

RESUMO

Tourette syndrome (TS) is a neuropsychiatric disorder with a strong genetic component. However, the genetic architecture of TS remains uncertain. Copy number variation (CNV) has been shown to contribute to the genetic make-up of several neurodevelopmental conditions, including schizophrenia and autism. Here we describe CNV calls using SNP chip genotype data from an initial sample of 210 TS cases and 285 controls ascertained in two Latin American populations. After extensive quality control, we found that cases (N = 179) have a significant excess (P = 0.006) of large CNV (>500 kb) calls compared to controls (N = 234). Amongst 24 large CNVs seen only in the cases, we observed four duplications of the COL8A1 gene region. We also found two cases with ∼400 kb deletions involving NRXN1, a gene previously implicated in neurodevelopmental disorders, including TS. Follow-up using multiplex ligation-dependent probe amplification (and including 53 more TS cases) validated the CNV calls and identified additional patients with rearrangements in COL8A1 and NRXN1, but none in controls. Examination of available parents indicates that two out of three NRXN1 deletions detected in the TS cases are de-novo mutations. Our results are consistent with the proposal that rare CNVs play a role in TS aetiology and suggest a possible role for rearrangements in the COL8A1 and NRXN1 gene regions.


Assuntos
Moléculas de Adesão Celular Neuronais/genética , Colágeno Tipo IX/genética , Variações do Número de Cópias de DNA/genética , Proteínas do Tecido Nervoso/genética , Síndrome de Tourette/genética , Adolescente , Proteínas de Ligação ao Cálcio , Criança , Feminino , Predisposição Genética para Doença/genética , Genótipo , Humanos , Masculino , Moléculas de Adesão de Célula Nervosa , Polimorfismo de Nucleotídeo Único/genética , Síndrome de Tourette/etiologia
17.
J Clin Invest ; 122(12): 4732-6, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23160192

RESUMO

Src homology 2 B adapter protein 1 (SH2B1) modulates signaling by a variety of ligands that bind to receptor tyrosine kinases or JAK-associated cytokine receptors, including leptin, insulin, growth hormone (GH), and nerve growth factor (NGF). Targeted deletion of Sh2b1 in mice results in increased food intake, obesity, and insulin resistance, with an intermediate phenotype seen in heterozygous null mice on a high-fat diet. We identified SH2B1 loss-of-function mutations in a large cohort of patients with severe early-onset obesity. Mutation carriers exhibited hyperphagia, childhood-onset obesity, disproportionate insulin resistance, and reduced final height as adults. Unexpectedly, mutation carriers exhibited a spectrum of behavioral abnormalities that were not reported in controls, including social isolation and aggression. We conclude that SH2B1 plays a critical role in the control of human food intake and body weight and is implicated in maladaptive human behavior.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Mutação da Fase de Leitura , Mutação de Sentido Incorreto , Obesidade/genética , Adolescente , Adulto , Agressão , Sequência de Bases , Estudos de Casos e Controles , Movimento Celular , Criança , Pré-Escolar , Análise Mutacional de DNA , Ingestão de Energia/genética , Feminino , Estudos de Associação Genética , Células HEK293 , Humanos , Masculino , Pessoa de Meia-Idade , Fenótipo , Transporte Proteico , Isolamento Social , Adulto Jovem
18.
N Engl J Med ; 366(3): 243-9, 2012 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-22168587

RESUMO

Thyroid hormones exert their effects through alpha (TRα1) and beta (TRß1 and TRß2) receptors. Here we describe a child with classic features of hypothyroidism (growth retardation, developmental retardation, skeletal dysplasia, and severe constipation) but only borderline-abnormal thyroid hormone levels. Using whole-exome sequencing, we identified a de novo heterozygous nonsense mutation in a gene encoding thyroid hormone receptor alpha (THRA) and generating a mutant protein that inhibits wild-type receptor action in a dominant negative manner. Our observations are consistent with defective human TRα-mediated thyroid hormone resistance and substantiate the concept of hormone action through distinct receptor subtypes in different target tissues.


Assuntos
Códon sem Sentido , Transtornos do Crescimento/genética , Hipotireoidismo/genética , Receptores alfa dos Hormônios Tireóideos/genética , Tiroxina/sangue , Tiroxina/uso terapêutico , Tri-Iodotironina/sangue , Criança , Feminino , Transtornos do Crescimento/tratamento farmacológico , Heterozigoto , Humanos , Hipotireoidismo/tratamento farmacológico , Modelos Moleculares , Conformação Proteica , Receptores alfa dos Hormônios Tireóideos/química , Hormônios Tireóideos/sangue
19.
J Med Genet ; 47(12): 803-8, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19755431

RESUMO

BACKGROUND: Craniosynostosis can be caused by both genetic and environmental factors, the relative contributions of which vary between patients. Genetic testing identifies a pathogenic mutation or chromosomal abnormality in ∼ 21% of cases, but it is likely that further causative mutations remain to be discovered. OBJECTIVE: To identify a shared signature of genetically determined craniosynostosis by comparing the expression patterns in three monogenic syndromes with a control group of patients with non-syndromic sagittal synostosis. METHODS: Fibroblasts from 10 individuals each with Apert syndrome (FGFR2 substitution S252W), Muenke syndrome (FGFR3 substitution P250R), Saethre-Chotzen syndrome (various mutations in TWIST1) and non-syndromic sagittal synostosis (no mutation detected) were cultured. The relative expression of ∼ 47,000 transcripts was quantified on Affymetrix arrays. RESULTS: 435, 45 and 46 transcripts were identified in the Apert, Muenke and Saethre-Chotzen groups, respectively, that differed significantly from the controls. Forty-six of these transcripts were shared between two or more syndromes and, in all but one instance, showed the same direction of altered expression level compared with controls. Pathway analysis showed over-representation of the shared transcripts in core modules involving cell-to-cell communication and signal transduction. Individual samples from the Apert syndrome cases could be reliably distinguished from non-syndromic samples based on the gene expression profile, but this was not possible for samples from patients with Muenke and Saethre-Chotzen syndromes. CONCLUSIONS: Common modules of altered gene expression shared by genetically distinct forms of craniosynostosis were identified. Although the expression profiles cannot currently be used to classify individual patients, this may be overcome by using more sensitive assays and sampling additional tissues.


Assuntos
Craniossinostoses/genética , Fibroblastos/metabolismo , Perfilação da Expressão Gênica , Couro Cabeludo/patologia , Acrocefalossindactilia/genética , Estudos de Casos e Controles , Pré-Escolar , Análise por Conglomerados , Regulação da Expressão Gênica , Humanos , Lactente , Análise de Sequência com Séries de Oligonucleotídeos , Reprodutibilidade dos Testes , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Síndrome
20.
Nature ; 463(7281): 666-70, 2010 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-19966786

RESUMO

Obesity is a highly heritable and genetically heterogeneous disorder. Here we investigated the contribution of copy number variation to obesity in 300 Caucasian patients with severe early-onset obesity, 143 of whom also had developmental delay. Large (>500 kilobases), rare (<1%) deletions were significantly enriched in patients compared to 7,366 controls (P < 0.001). We identified several rare copy number variants that were recurrent in patients but absent or at much lower prevalence in controls. We identified five patients with overlapping deletions on chromosome 16p11.2 that were found in 2 out of 7,366 controls (P < 5 x 10(-5)). In three patients the deletion co-segregated with severe obesity. Two patients harboured a larger de novo 16p11.2 deletion, extending through a 593-kilobase region previously associated with autism and mental retardation; both of these patients had mild developmental delay in addition to severe obesity. In an independent sample of 1,062 patients with severe obesity alone, the smaller 16p11.2 deletion was found in an additional two patients. All 16p11.2 deletions encompass several genes but include SH2B1, which is known to be involved in leptin and insulin signalling. Deletion carriers exhibited hyperphagia and severe insulin resistance disproportionate for the degree of obesity. We show that copy number variation contributes significantly to the genetic architecture of human obesity.


Assuntos
Deleção Cromossômica , Cromossomos Humanos Par 16/genética , Obesidade/genética , Obesidade/fisiopatologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Idade de Início , Criança , Estudos de Coortes , Variações do Número de Cópias de DNA/genética , Deficiências do Desenvolvimento/complicações , Deficiências do Desenvolvimento/genética , Estudo de Associação Genômica Ampla , Heterozigoto , Humanos , Hiperfagia/genética , Padrões de Herança/genética , Resistência à Insulina/genética , Mutação/genética , Obesidade/complicações , Obesidade/epidemiologia , Reino Unido/epidemiologia , População Branca
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...