Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Cell Rep Med ; 4(8): 101155, 2023 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-37586323

RESUMO

New approaches are needed to treat people whose obesity and type 2 diabetes (T2D) are driven by specific mechanisms. We investigate a deletion on chromosome 16p11.2 (breakpoint 2-3 [BP2-3]) encompassing SH2B1, a mediator of leptin and insulin signaling. Phenome-wide association scans in the UK (N = 502,399) and Estonian (N = 208,360) biobanks show that deletion carriers have increased body mass index (BMI; p = 1.3 × 10-10) and increased rates of T2D. Compared with BMI-matched controls, deletion carriers have an earlier onset of T2D, with poorer glycemic control despite higher medication usage. Cystatin C, a biomarker of kidney function, is significantly elevated in deletion carriers, suggesting increased risk of renal impairment. In a Mendelian randomization study, decreased SH2B1 expression increases T2D risk (p = 8.1 × 10-6). We conclude that people with 16p11.2 BP2-3 deletions have early, complex obesity and T2D and may benefit from therapies that enhance leptin and insulin signaling.


Assuntos
Diabetes Mellitus Tipo 2 , Insulinas , Doenças Metabólicas , Humanos , Leptina , Diabetes Mellitus Tipo 2/genética , Obesidade/genética , Proteínas Adaptadoras de Transdução de Sinal
2.
Cell Rep ; 34(12): 108862, 2021 03 23.
Artigo em Inglês | MEDLINE | ID: mdl-33761344

RESUMO

The Melanocortin-4 Receptor (MC4R) plays a pivotal role in energy homeostasis. We used human MC4R mutations associated with an increased or decreased risk of obesity to dissect mechanisms that regulate MC4R function. Most obesity-associated mutations impair trafficking to the plasma membrane (PM), whereas obesity-protecting mutations either accelerate recycling to the PM or decrease internalization, resulting in enhanced signaling. MC4R mutations that do not affect canonical Gαs protein-mediated signaling, previously considered to be non-pathogenic, nonetheless disrupt agonist-induced internalization, ß-arrestin recruitment, and/or coupling to Gαs, establishing their causal role in severe obesity. Structural mapping reveals ligand-accessible sites by which MC4R couples to effectors and residues involved in the homodimerization of MC4R, which is disrupted by multiple obesity-associated mutations. Human genetic studies reveal that endocytosis, intracellular trafficking, and homodimerization regulate MC4R function to a level that is physiologically relevant, supporting the development of chaperones, agonists, and allosteric modulators of MC4R for weight loss therapy.


Assuntos
Peso Corporal/genética , Endocitose , Variação Genética , Multimerização Proteica , Receptor Tipo 4 de Melanocortina/genética , Animais , Células COS , Membrana Celular/metabolismo , Chlorocebus aethiops , AMP Cíclico/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Subunidades alfa Gs de Proteínas de Ligação ao GTP , Células HEK293 , Humanos , Modelos Biológicos , Proteínas Mutantes/metabolismo , Mutação/genética , Fosforilação , Receptor Tipo 4 de Melanocortina/química , Transdução de Sinais , beta-Arrestinas/metabolismo
3.
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
4.
PLoS Genet ; 15(1): e1007603, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30677029

RESUMO

The variation in weight within a shared environment is largely attributable to genetic factors. Whilst many genes/loci confer susceptibility to obesity, little is known about the genetic architecture of healthy thinness. Here, we characterise the heritability of thinness which we found was comparable to that of severe obesity (h2 = 28.07 vs 32.33% respectively), although with incomplete genetic overlap (r = -0.49, 95% CI [-0.17, -0.82], p = 0.003). In a genome-wide association analysis of thinness (n = 1,471) vs severe obesity (n = 1,456), we identified 10 loci previously associated with obesity, and demonstrate enrichment for established BMI-associated loci (pbinomial = 3.05x10-5). Simulation analyses showed that different association results between the extremes were likely in agreement with additive effects across the BMI distribution, suggesting different effects on thinness and obesity could be due to their different degrees of extremeness. In further analyses, we detected a novel obesity and BMI-associated locus at PKHD1 (rs2784243, obese vs. thin p = 5.99x10-6, obese vs. controls p = 2.13x10-6 pBMI = 2.3x10-13), associations at loci recently discovered with much larger sample sizes (e.g. FAM150B and PRDM6-CEP120), and novel variants driving associations at previously established signals (e.g. rs205262 at the SNRPC/C6orf106 locus and rs112446794 at the PRDM6-CEP120 locus). Our ability to replicate loci found with much larger sample sizes demonstrates the value of clinical extremes and suggest that characterisation of the genetics of thinness may provide a more nuanced understanding of the genetic architecture of body weight regulation and may inform the identification of potential anti-obesity targets.


Assuntos
Proteínas Musculares/genética , Proteínas de Neoplasias/genética , Obesidade Mórbida/genética , Receptores de Superfície Celular/genética , Magreza/genética , Fatores de Transcrição/genética , Adulto , Alelos , Índice de Massa Corporal , Feminino , Predisposição Genética para Doença , Estudo de Associação Genômica Ampla , Humanos , Masculino , Pessoa de Meia-Idade , Obesidade Mórbida/fisiopatologia , Polimorfismo de Nucleotídeo Único , Magreza/fisiopatologia
5.
FASEB J ; 32(4): 1830-1840, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29180441

RESUMO

Psychiatric disorders are associated with aberrant brain development and/or aggressive behavior and are influenced by genetic factors; however, genes that affect brain aggression circuits remain elusive. Here, we show that neuronal Src-homology-2 (SH2)B adaptor protein-1 ( Sh2b1) is indispensable for both brain growth and protection against aggression. Global and brain-specific deletion of Sh2b1 decreased brain weight and increased aggressive behavior. Global and brain-specific Sh2b1 knockout (KO) mice exhibited fatal, intermale aggression. In a resident-intruder paradigm, latency to attack was markedly reduced, whereas the number and the duration of attacks was significantly increased in global and brain-specific Sh2b1 KO mice compared with wild-type littermates. Consistently, core aggression circuits were activated to a higher level in global and brain-specific Sh2b1 KO males, based on c-fos immunoreactivity in the amygdala and periaqueductal gray. Brain-specific restoration of Sh2b1 normalized brain size and reversed pathologic aggression and aberrant activation of core aggression circuits in Sh2b1 KO males. SH2B1 mutations in humans were linked to aberrant brain development and behavior. At the molecular level, Sh2b1 enhanced neurotrophin-stimulated neuronal differentiation and protected against oxidative stress-induced neuronal death. Our data suggest that neuronal Sh2b1 promotes brain development and the integrity of core aggression circuits, likely through enhancing neurotrophin signaling.-Jiang, L., Su, H., Keogh, J. M., Chen, Z., Henning, E., Wilkinson, P., Goodyer, I., Farooqi, I. S., Rui, L. Neural deletion of Sh2b1 results in brain growth retardation and reactive aggression.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Agressão , Encéfalo/crescimento & desenvolvimento , Adulto , Animais , Encéfalo/metabolismo , Encéfalo/fisiologia , Criança , Feminino , Deleção de Genes , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Mutação , Células PC12 , Ratos
6.
J Clin Endocrinol Metab ; 99(10): E2101-6, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25062455

RESUMO

CONTEXT: Mutations in the melanocortin-4 receptor (MC4R) represent the commonest genetic form of obesity and are associated with hyperphagia. OBJECTIVE: The aim of this study was to investigate whether melanocortin signaling modulates anticipatory food reward by studying the brain activation response to food cues in individuals with MC4R mutations. Design/Setting/Participants/Main Outcome Measure: We used functional magnetic resonance imaging to measure blood oxygen level-dependent responses to images of highly palatable, appetizing foods, bland foods, and non-food objects in eight obese individuals with MC4R mutations, 10 equally obese controls, and eight lean controls with normal MC4R genotypes. Based on previous evidence, we performed a region-of-interest analysis centered on the caudate/putamen (dorsal striatum) and ventral striatum. RESULTS: Compared to non-foods, appetizing foods were associated with activation in the dorsal and ventral striatum in lean controls and in MC4R-deficient individuals. Surprisingly, we observed reduced activation of the dorsal and ventral striatum in obese controls relative to MC4R-deficient patients and lean controls. There were no group differences for the contrast of disgusting foods with bland foods or non-foods, suggesting that the effects observed in response to appetizing foods were not related to arousal. CONCLUSION: We identified differences in the striatal response to food cues between two groups of obese individuals, those with and those without MC4R mutations. These findings are consistent with a role for central melanocortinergic circuits in the neural response to visual food cues.


Assuntos
Apetite/fisiologia , Hiperfagia/genética , Neostriado/metabolismo , Obesidade/genética , Receptor Tipo 4 de Melanocortina/genética , Transdução de Sinais/fisiologia , Adolescente , Adulto , Comportamento Alimentar/fisiologia , Feminino , Alimentos , Humanos , Hiperfagia/metabolismo , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Mutação , Obesidade/metabolismo , Oxigênio/sangue , Receptor Tipo 4 de Melanocortina/metabolismo , Aumento de Peso/fisiologia , Adulto Jovem
7.
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
8.
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
9.
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
10.
J Clin Invest ; 120(4): 1345-54, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20237409

RESUMO

Resistance to thyroid hormone (RTH), a dominantly inherited disorder usually associated with mutations in thyroid hormone receptor beta (THRB), is characterized by elevated levels of circulating thyroid hormones (including thyroxine), failure of feedback suppression of thyrotropin, and variable tissue refractoriness to thyroid hormone action. Raised energy expenditure and hyperphagia are recognized features of hyperthyroidism, but the effects of comparable hyperthyroxinemia in RTH patients are unknown. Here, we show that resting energy expenditure (REE) was substantially increased in adults and children with THRB mutations. Energy intake in RTH subjects was increased by 40%, with marked hyperphagia particularly evident in children. Rates of muscle TCA cycle flux were increased by 75% in adults with RTH, whereas rates of ATP synthesis were unchanged, as determined by 13C/31P magnetic resonance spectroscopy. Mitochondrial coupling index between ATP synthesis and mitochondrial rates of oxidation (as estimated by the ratio of ATP synthesis to TCA cycle flux) was significantly decreased in RTH patients. These data demonstrate that basal mitochondrial substrate oxidation is increased and energy production in the form of ATP synthesis is decreased in the muscle of RTH patients and that resting oxidative phosphorylation is uncoupled in this disorder. Furthermore, these observations suggest that mitochondrial uncoupling in skeletal muscle is a major contributor to increased REE in patients with RTH, due to tissue selective retention of thyroid hormone receptor alpha sensitivity to elevated thyroid hormone levels.


Assuntos
Metabolismo Energético , Hiperfagia/etiologia , Mitocôndrias Musculares/metabolismo , Síndrome da Resistência aos Hormônios Tireóideos/metabolismo , Trifosfato de Adenosina/biossíntese , Adulto , Criança , Ciclo do Ácido Cítrico , Ingestão de Alimentos , Humanos , Hiperfagia/metabolismo , Resistência à Insulina , Pessoa de Meia-Idade , Músculo Esquelético/metabolismo , Receptores beta dos Hormônios Tireóideos/genética
11.
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
12.
Hum Mol Genet ; 18(17): 3257-65, 2009 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-19498035

RESUMO

Genetic studies in patients with severe early-onset obesity have provided insights into the molecular and physiological pathways that regulate body weight in humans. We report a 19-year-old male with hyperphagia and severe obesity, mild learning difficulties and hypogonadism, in whom diagnostic tests for Prader-Willi syndrome (PWS) had been negative. We carried out detailed clinical and metabolic phenotyping of this patient and investigated the genetic basis of this obesity syndrome using Agilent 185 k array comparative genomic hybridization (aCGH) and Affymetrix 6.0 genotyping arrays. The identified deletion was validated using multiplex ligation-dependent probe amplification and long-range PCR, followed by breakpoint sequencing which enabled precise localization of the deletion. We identified a approximately 187 kb microdeletion at chromosome 15q11-13 that encompasses non-coding small nucleolar RNAs (including HBII-85 snoRNAs) which were not expressed in peripheral lymphocytes from the patient. Characterization of the clinical phenotype revealed increased ad libitum food intake, normal basal metabolic rate when adjusted for fat-free mass, partial hypogonadotropic hypogonadism and growth failure. We have identified a novel deletion on chromosome 15q11-13 in an individual with hyperphagia, obesity, hypogonadism and other features associated with PWS, which is normally caused by deficiency of several paternally expressed imprinted transcripts within chromosome 15q11-13, a region that includes multiple protein-coding genes as well as several non-coding snoRNAs. These findings provide direct evidence for the role of a particular family of non-coding RNAs, the HBII-85 snoRNA cluster, in human energy homeostasis, growth and reproduction.


Assuntos
Hiperfagia/genética , Hipogonadismo/genética , Obesidade/genética , RNA Nucleolar Pequeno/genética , Sequência de Aminoácidos , Mapeamento Cromossômico , Cromossomos Humanos Par 15/genética , Humanos , Hiperfagia/metabolismo , Hipogonadismo/metabolismo , Masculino , Dados de Sequência Molecular , Obesidade/metabolismo , RNA Nucleolar Pequeno/metabolismo , Alinhamento de Sequência , Adulto Jovem
13.
Am J Clin Nutr ; 89(1): 106-113, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19056578

RESUMO

BACKGROUND: Incretin hormones, such as glucagon-like peptide 1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP), play an important role in meal-related insulin secretion. We previously demonstrated that glutamine is a potent stimulus of GLP-1 secretion in vitro. OBJECTIVE: Our objective was to determine whether glutamine increases circulating GLP-1 and GIP concentrations in vivo and, if so, whether this is associated with an increase in plasma insulin. DESIGN: We recruited 8 healthy normal-weight volunteers (LEAN), 8 obese individuals with type 2 diabetes or impaired glucose tolerance (OB-DIAB) and 8 obese nondiabetic control subjects (OB-CON). Oral glucose (75 g), glutamine (30 g), and water were administered on 3 separate days in random order, and plasma concentrations of GLP-1, GIP, insulin, glucagon, and glucose were measured over 120 min. RESULTS: Oral glucose led to increases in circulating GLP-1 concentrations, which peaked at 30 min in LEAN (31.9 +/- 5.7 pmol/L) and OB-CON (24.3 +/- 2.1 pmol/L) subjects and at 45 min in OB-DIAB subjects (19.5 +/- 1.8 pmol/L). Circulating GLP-1 concentrations increased in all study groups after glutamine ingestion, with peak concentrations at 30 min of 22.5 +/- 3.4, 17.9 +/- 1.1, and 17.3 +/- 3.4 pmol/L in LEAN, OB-CON, and OB-DIAB subjects, respectively. Glutamine also increased plasma GIP concentrations but less effectively than glucose. Consistent with the increases in GLP-1 and GIP, glutamine significantly increased circulating plasma insulin concentrations. Glutamine stimulated glucagon secretion in all 3 study groups. CONCLUSION: Glutamine effectively increases circulating GLP-1, GIP, and insulin concentrations in vivo and may represent a novel therapeutic approach to stimulating insulin secretion in obesity and type 2 diabetes.


Assuntos
Diabetes Mellitus Tipo 2/sangue , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Glucagon/metabolismo , Glutamina/farmacologia , Insulina/metabolismo , Administração Oral , Adulto , Área Sob a Curva , Glicemia/metabolismo , Estudos Cross-Over , Diabetes Mellitus Tipo 2/tratamento farmacológico , Feminino , Polipeptídeo Inibidor Gástrico/sangue , Glucagon/sangue , Glucagon/efeitos dos fármacos , Peptídeo 1 Semelhante ao Glucagon/sangue , Peptídeo 1 Semelhante ao Glucagon/efeitos dos fármacos , Teste de Tolerância a Glucose , Humanos , Insulina/sangue , Secreção de Insulina , Masculino , Obesidade/sangue , Magreza/sangue
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA