Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Am J Physiol Endocrinol Metab ; 310(10): E855-61, 2016 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-27026085

RESUMO

Roux-en-Y gastric bypass (RYGB) causes profound weight loss and remission of diabetes by influencing metabolic physiology, yet the mechanisms behind these clinical improvements remain undefined. After RYGB, levels of glucagon-like peptide-1 (GLP-1), a hormone that enhances insulin secretion and promotes satiation, are substantially elevated. Because GLP-1 signals in both the periphery and the brain to influence energy balance and glucose regulation, we aimed to determine the relative requirements of these systems to weight loss and improved glucose tolerance following RYGB surgery in mice. By pharmacologically blocking peripheral or central GLP-1R signaling, we examined whether GLP-1 action is necessary for the metabolic improvements observed after RYGB. Diet-induced obese mice underwent RYGB or sham operation and were implanted with osmotic pumps delivering the GLP-1R antagonist exendin-(9-39) (2 pmol·kg(-1)·min(-1) peripherally; 0.5 pmol·kg(-1)·min(-1) centrally) for up to 10 wk. Blockade of peripheral GLP-1R signaling partially reversed the improvement in glucose tolerance after RYGB. In contrast, fasting glucose and insulin sensitivity, as well as body weight, were unaffected by GLP-1R antagonism. Central GLP-1R signaling did not appear to be required for any of the metabolic improvements seen after this operation. Collectively, these results suggest a detectable but only modest role for GLP-1 in mediating the effects of RYGB and that this role is limited to its well-described action on glucose regulation.


Assuntos
Glicemia/metabolismo , Derivação Gástrica , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Receptor do Peptídeo Semelhante ao Glucagon 1/metabolismo , Resistência à Insulina , Insulina/metabolismo , Obesidade/metabolismo , Animais , Peso Corporal , Dieta Hiperlipídica , Metabolismo Energético , Receptor do Peptídeo Semelhante ao Glucagon 1/antagonistas & inibidores , Teste de Tolerância a Glucose , Masculino , Camundongos , Obesidade/cirurgia , Fragmentos de Peptídeos/farmacologia , Transdução de Sinais
2.
Bone ; 85: 23-8, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26806052

RESUMO

Roux-en-Y gastric bypass (RYGB) is a profoundly effective treatment for severe obesity, but results in significant bone loss in patients. Developing a murine model that recapitulates this skeletal phenotype will provide a robust tool with which to study the physiologic mechanisms of this bone loss. We studied adult male C57BL/6J mice who underwent either RYGB or sham operation. Twelve weeks after surgery, we characterized biochemical bone markers (parathyroid hormone, PTH; C-telopeptide, CTX; and type 1 procollagen, P1NP) and bone microarchitectural parameters as measured by microcomputed tomography. RYGB-treated mice had significant trabecular and cortical bone deficits compared with sham-operated controls. Although adjustment for final body weight eliminated observed cortical differences, the trabecular bone volume fraction remained significantly lower in RYGB mice even after weight adjustment. PTH levels were similar between groups, but RYGB mice had significantly higher indices of bone turnover than sham controls. These data demonstrate that murine models of RYGB recapitulate patterns of bone loss and turnover that have been observed in human clinical studies. Future studies that exploit this murine model will help delineate the alterations in bone metabolism and mechanisms of bone loss after RYGB.


Assuntos
Osso Esponjoso/patologia , Osso Cortical/patologia , Derivação Gástrica , Animais , Biomarcadores/metabolismo , Peso Corporal , Remodelação Óssea , Osso Esponjoso/metabolismo , Osso Cortical/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Modelos Animais , Hormônio Paratireóideo/metabolismo
3.
Endocrinology ; 156(9): 3183-91, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26066076

RESUMO

Roux-en-Y gastric bypass (RYGB) typically leads to substantial, long-term weight loss (WL) and diabetes remission, although there is a wide variation in response to RYGB among individual patients. Defining the pathways through which RYGB works should aid in the development of less invasive anti-obesity treatments, whereas identifying weight-regulatory pathways unengaged by RYGB could facilitate the development of therapies that complement the beneficial effects of surgery. Activation of serotonin 2C receptors (5-HT2CR) by serotonergic drugs causes WL in humans and animal models. 5-HT2CR are located on neurons that activate the melanocortin-4 receptors, which are essential for WL after RYGB. We therefore sought to determine whether 5-HT2CR signaling is also essential for metabolic effects of RYGB or whether it is a potentially complementary pathway, the activation of which could extend the benefits of RYGB. Diet-induced obese male mice deficient for the 5-HT2CR and their wild-type littermates underwent RYGB or sham operation. Both groups lost similar amounts of weight after RYGB, demonstrating that the improved metabolic phenotype after RYGB is 5-HT2CR independent. Consistent with this hypothesis, wild-type RYGB-treated mice lost additional weight after the administration of the serotonergic drugs fenfluramine and meta-chlorophenylpiperazine but not the nonserotonergic agent topiramate. The fact that RYGB does not depend on 5-HT2CR signaling suggests that there are important WL mechanisms not fully engaged by surgery that could potentially be harnessed for medical treatment. These results suggest a rational basis for designing medical-surgical combination therapies to optimize clinical outcomes by exploiting complementary physiological mechanisms of action.


Assuntos
Derivação Gástrica , Receptor 5-HT2C de Serotonina/metabolismo , Redução de Peso , Animais , Glicemia , Peso Corporal , Comportamento Alimentar , Fenfluramina , Masculino , Camundongos Endogâmicos C57BL , Agonistas do Receptor 5-HT2 de Serotonina
4.
Am J Physiol Regul Integr Comp Physiol ; 303(10): R985-93, 2012 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-22972837

RESUMO

Roux-en-Y gastric bypass (RYGB) in rodent models reduces food intake (FI), increases resting energy expenditure (EE), and improves glycemic control. We have shown that mimicking the duodenal component of RYGB by implantation of a 10-cm endoluminal sleeve device (ELS-10) induces weight loss and improves glycemic control in diet-induced obese (DIO) rats. We sought to determine the mechanisms and structural requirements of these effects. We examined the effects of ELS-10 devices implanted in male DIO rats on body weight, food intake (FI), meal patterns, total and resting EE, and multiple parameters of glucose homeostasis, comparing them with sham-operated (SO) rats and with SO rats weight matched to the ELS-10-treated group. To determine the extent of duodenal exclusion required to influence metabolic outcomes, we compared the effects of implanting 10-, 4-, or 1-cm ELS devices. ELS-10 rats exhibited 13% higher total and 9% higher resting EE than SO controls. ELS-10 rats also exhibited enhanced postprandial GLP-1 secretion and improved glucose tolerance and insulin sensitivity out of proportion to the effects of weight loss alone. Implantation of 4- or 1-cm ELS devices had no effect on EE and limited effects on glucose homeostasis. Complete duodenal exclusion with ELS-10 induces weight loss by decreasing FI and increasing EE and improves glycemic control through weight loss-independent mechanisms. Thus signals originating in the proximal small intestine appear to exert a direct influence on the physiological regulation of EE and glucose homeostasis. Their selective manipulation could provide effective new therapies for obesity and diabetes that mimic the benefits of RYGB.


Assuntos
Duodeno/cirurgia , Metabolismo Energético/fisiologia , Glucose/metabolismo , Homeostase/fisiologia , Obesidade/metabolismo , Animais , Glicemia , Dieta , Ingestão de Energia , Implantes Experimentais , Absorção Intestinal , Masculino , Ratos , Ratos Sprague-Dawley , Redução de Peso
5.
Obesity (Silver Spring) ; 19(3): 492-9, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20948526

RESUMO

Maternal obesity can influence susceptibility to obesity and type 2 diabetes in progeny. We examined the relationship of maternal insulin resistance (IR), a metabolically important consequence of increased adiposity, to adverse consequences of obesity for fetal development. We used mice heterozygous for a null allele of the insulin receptor (Insr) to study the contributions of maternal IR to offspring phenotype without the potential confound of obesity per se, and how maternal consumption of high-fat diet (HFD) may, independently and interactively, affect progeny. In progeny fed a 60% HFD, body weight and adiposity were transiently (5-7 weeks) increased in wild-type (+/+) offspring of Insr(+/-) HFD-fed dams compared to offspring of wild-type HFD-fed dams. Offspring of HFD-fed wild-type dams had increased body weight, blood glucose, and plasma insulin concentrations compared to offspring of chow-fed wild-type dams. Quantification of proopiomelanocortin (POMC) and neuropeptide-Y (NPY) populations in the arcuate nucleus of the hypothalamus (ARH) of offspring of wild-type vs. Insr(+/-) dams was performed to determine whether maternal IR affects the formation of central feeding circuits. We found a 20% increase in the number of Pomc-expressing cells at postnatal day 9 in offspring of Insr(+/-) dams. In conclusion, maternal HFD consumption-distinct from overt obesity per se-was a major contributor to increased body weight, adiposity, IR, and liver triglyceride (TG) phenotypes in progeny. Maternal IR played a minor role in predisposing progeny to obesity and IR, though it acted synergistically with maternal HFD to exacerbate early obesity in progeny.


Assuntos
Gorduras na Dieta/efeitos adversos , Hipotálamo/metabolismo , Resistência à Insulina/fisiologia , Obesidade/etiologia , Complicações na Gravidez/metabolismo , Fenômenos Fisiológicos da Nutrição Pré-Natal , Adiposidade/efeitos dos fármacos , Animais , Glicemia/metabolismo , Feminino , Hipotálamo/citologia , Insulina/sangue , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neuropeptídeo Y/metabolismo , Obesidade/fisiopatologia , Fenótipo , Gravidez , Pró-Opiomelanocortina/metabolismo , Receptor de Insulina/genética , Triglicerídeos/metabolismo , Aumento de Peso/efeitos dos fármacos
6.
Nat Med ; 16(4): 403-5, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20348924

RESUMO

Hypothalamic neuron circuits regulating energy balance are highly plastic and develop in response to nutrient and hormonal cues. To identify processes that might be susceptible to gestational influences in mice, we characterized the ontogeny of proopiomelanocortin (POMC) and neuropeptide Y (NPY) cell populations, which exert opposing influences on food intake and body weight. These analyses revealed that Pomc is broadly expressed in immature hypothalamic neurons and that half of embryonic Pomc-expressing precursors subsequently adopt a non-POMC fate in adult mice. Moreover, nearly one quarter of the mature NPY+ cell population shares a common progenitor with POMC+ cells.


Assuntos
Ingestão de Alimentos/fisiologia , Células-Tronco Embrionárias/fisiologia , Hipotálamo/fisiologia , Pró-Opiomelanocortina/fisiologia , Animais , Peso Corporal/fisiologia , Imunofluorescência , Hipotálamo/embriologia , Camundongos , Neurônios/fisiologia , Neuropeptídeo Y/fisiologia
7.
Am J Physiol Regul Integr Comp Physiol ; 287(5): R1190-3, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15231492

RESUMO

The central glucagon-like peptide-1 (GLP-1) system has been implicated in the control of feeding behavior. Here we explore GLP-1 mediation of the anorexic response to administration of systemic LPS and address the relative importance of caudal brain stem and forebrain GLP-1 receptor (GLP-1-R) for the mediation of the response. Fourth-intracerebroventricular delivery of the GLP-1-R antagonist exendin-(9-39) (10 microg) did not itself affect food intake in the 24 h after injection but significantly attenuated the otherwise robust (approximately 60%) reduction in food intake obtained after LPS (100 microg/kg) treatment. This result highlights a role for caudal brain stem GLP-1-R in the mediation of LPS anorexia but does not rule out the possibility that forebrain receptors also contribute to the response. Forebrain contribution was addressed by delivery of the GLP-1-R antagonist to the third ventricle with the caudal flow of cerebrospinal fluid blocked by occlusion of the cerebral aqueduct. Exendin-(9-39) delivery thus limited to forebrain did not attenuate the anorexic response to LPS. These data suggest that LPS anorexia is mediated, in part, by release of the native peptide acting on GLP-1-R within the caudal brain stem.


Assuntos
Anorexia/prevenção & controle , Tronco Encefálico/fisiologia , Lipopolissacarídeos/antagonistas & inibidores , Lipopolissacarídeos/farmacologia , Prosencéfalo/fisiologia , Receptores de Glucagon/antagonistas & inibidores , Animais , Anorexia/induzido quimicamente , Tronco Encefálico/efeitos dos fármacos , Aqueduto do Mesencéfalo/fisiologia , Receptor do Peptídeo Semelhante ao Glucagon 1 , Habituação Psicofisiológica/fisiologia , Injeções Intraventriculares , Masculino , Fragmentos de Peptídeos/farmacologia , Prosencéfalo/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Terceiro Ventrículo/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...