Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Diabetes Care ; 45(2): 469-476, 2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-34857533

RESUMO

OBJECTIVE: The role of the gut in diabetes remission after Roux-en-Y gastric bypass (RYGB) is incompletely understood. We assessed the temporal change in insulin secretory capacity after RYGB, using oral and intravenous (IV) glucose, in individuals with type 2 diabetes. RESEARCH DESIGN AND METHODS: Longitudinal, prospective measures of ß-cell function were assessed after oral glucose intake and graded glucose infusion in individuals with severe obesity and diabetes studied at 0, 3 (n = 29), 12 (n = 24), and 24 (n = 20) months after RYGB. Data were collected between 2015 and 2019 in an academic clinical research center. RESULTS: The decreases in body weight, fat mass, waist circumference, and insulin resistance after surgery (all P < 0.001 at 12 and 24 months) did not differ according to diabetes remission status. In contrast, both the magnitude and temporal changes in ß-cell glucose sensitivity after oral glucose intake differed by remission status (P = 0.04): greater (6.5-fold; P < 0.01) and sustained in those in full remission, moderate and not sustained past 12 months in those with partial remission (3.3-fold; P < 0.001), and minimal in those not experiencing remission (2.7-fold; P = not significant). The improvement in ß-cell function after IV glucose administration was not apparent until 12 months, significant only in those in full remission, and only ∼33% of that observed after oral glucose intake. Preintervention ß-cell function and its change after surgery predicted remission; weight loss and insulin sensitivity did not. CONCLUSIONS: Our data show the time course of changes in ß-cell function after RYGB. The improvement in ß-cell function after RYGB, but not changes in weight loss or insulin sensitivity, drives diabetes remission.


Assuntos
Diabetes Mellitus Tipo 2 , Derivação Gástrica , Resistência à Insulina , Obesidade Mórbida , Glicemia , Diabetes Mellitus Tipo 2/cirurgia , Humanos , Insulina , Resistência à Insulina/fisiologia , Obesidade Mórbida/cirurgia , Estudos Prospectivos
2.
Surg Obes Relat Dis ; 15(8): 1367-1373, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31296445

RESUMO

BACKGROUND: Some of the metabolic effects of bariatric surgery may be mediated by the gut microbiome. OBJECTIVES: To study the effect of bariatric surgery on changes to gut microbiota composition and bacterial pathways, and their relation to metabolic parameters after bariatric surgery. SETTINGS: University hospitals in the United States and Spain. METHODS: Microbial diversity and composition by 16 S rRNA sequencing, putative bacterial pathways, and targeted circulating metabolites were studied in 26 individuals with severe obesity, with and without type 2 diabetes, before and at 3, 6, and 12 months after either gastric bypass or sleeve gastrectomy. RESULTS: Bariatric surgery tended to increase alpha diversity, and significantly altered beta diversity, microbiota composition, and function up to 6 months after surgery, but these changes tend to regress to presurgery levels by 12 months. Twelve of 15 bacterial pathways enriched after surgery also regressed to presurgery levels at 12 months. Network analysis identified groups of bacteria significantly correlated with levels of circulating metabolites over time. There were no differences between study sites, surgery type, or diabetes status in terms of microbial diversity and composition at baseline and after surgery. CONCLUSIONS: The association among changes in microbiome with decreased circulating biomarkers of inflammation, increased bile acids, and products of choline metabolism and other bacterial pathways suggest that the microbiome partially mediates improvement of metabolism during the first year after bariatric surgery.


Assuntos
Cirurgia Bariátrica , Microbioma Gastrointestinal/genética , Obesidade/cirurgia , Redução de Peso/fisiologia , Estudos de Coortes , DNA Bacteriano/genética , Humanos , Metaboloma/genética , Metagenoma/genética
3.
Diabetes Care ; 42(2): 311-317, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30523032

RESUMO

OBJECTIVE: Hepatic insulin clearance is a significant regulator of glucose homestasis. We hypothesized that the improvement in insulin clearance rates (ICRs) under fasting conditions and in response to oral and intravenous (IV) glucose would improve similarly after Roux-en-Y gastric bypass (RYGB) and adjustable gastric banding (AGB) as a function of weight loss; the difference in ICR after oral and IV glucose stimulation will be enhanced after RYGB compared with AGB, an effect mediated by glucagon-like peptide 1 (GLP-1). RESEARCH DESIGN AND METHODS: In study 1, the ICR was calculated under fasting condition (F-ICR), after oral glucose (O-ICR), and after an isoglycemic IV glucose clamp (IV-ICR) in individuals from an established cohort with type 2 diabetes mellitus (T2DM) before, after 10% matched weight loss, and 1 year after either RYGB (n = 22) or AGB (n = 12). In study 2, O-ICR was studied in a separate cohort of individuals with T2DM (n = 22), before and 3 months after RYGB, with and without exendin(9-39) infusion. RESULTS: In study 1, age, BMI, T2DM duration and control, and ICR did not differ between RYGB and AGB preintervention. Weight loss at 1 year was two times greater after RYGB than after AGB (31.6 ± 5.9% vs. 16.6 ± 9.8%; P < 0.05). RYGB and AGB both significantly increased F-ICR, O-ICR, and IV-ICR at 1 year. ICR was inversely associated with insulinemia. The difference between IV-ICR and O-ICR was significantly greater after RYGB versus AGB. GLP-1 antagonism with exendin(9-39) led to an increase in O-ICR in subjects post-RYGB. CONCLUSIONS: Weight loss increased ICR, an effect more pronounced after RYGB compared with AGB. Our data support a potential role for endogenous GLP-1 in the control of postprandial ICR after RYGB.


Assuntos
Diabetes Mellitus Tipo 2/cirurgia , Derivação Gástrica , Gastroplastia , Glucose/administração & dosagem , Insulina/metabolismo , Redução de Peso/fisiologia , Administração Intravenosa , Administração Oral , Adulto , Cirurgia Bariátrica/reabilitação , Estudos de Coortes , Diabetes Mellitus Tipo 2/metabolismo , Feminino , Derivação Gástrica/reabilitação , Gastroplastia/reabilitação , Teste de Tolerância a Glucose , Humanos , Resistência à Insulina/fisiologia , Fígado/metabolismo , Estudos Longitudinais , Masculino , Taxa de Depuração Metabólica , Pessoa de Meia-Idade , Período Pós-Prandial
4.
Diabetes Obes Metab ; 20(4): 1018-1023, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29072800

RESUMO

The present study was a 4-week randomized trial to assess the efficacy and safety of sitagliptin, a dipeptidyl-peptidase-4 inhibitor, in persistent or recurring type 2 diabetes after Roux-en-Y gastric bypass surgery (RYGB). Participants (n = 32) completed a mixed meal test (MMT) and self-monitoring of plasma glucose (SMPG) before and 4 weeks after randomization to either sitagliptin 100 mg daily or placebo daily. Questionnaires were administered to assess gastrointestinal discomfort. Outcome variables were glucose, active glucagon-like peptide-1 and ß-cell function during the MMT, and glucose levels during SMPG. Age (56.3 ± 8.2 years), body mass index (34.4 ± 6.7 kg/m2 ), glycated haemoglobin (7.21 ± 0.77%), diabetes duration (12.9 ± 10.0 years), years since RYGB (5.6 ± 3.3 years) and ß-cell function did not differ between the placebo and sitagliptin groups at pre-intervention. Sitagliptin was well tolerated, decreased postprandial glucose levels during the MMT (from 8.31 ± 1.92 mmol/L to 7.67 ± 1.59 mmol/L, P = 0.03) and mean SMPG levels, but had no effect on ß-cell function. In patients with diabetes and mild hyperglycemia after RYGB, a short course of sitagliptin provided a small but significant glucose-lowering effect, with no identified improvement in ß-cell function.


Assuntos
Glicemia/efeitos dos fármacos , Diabetes Mellitus Tipo 2/tratamento farmacológico , Diabetes Mellitus Tipo 2/cirurgia , Derivação Gástrica , Fosfato de Sitagliptina/farmacologia , Adulto , Idoso , Glicemia/metabolismo , Terapia Combinada , Diabetes Mellitus Tipo 2/sangue , Feminino , Hemoglobinas Glicadas/efeitos dos fármacos , Hemoglobinas Glicadas/metabolismo , Humanos , Estudos Longitudinais , Masculino , Pessoa de Meia-Idade , Fosfato de Sitagliptina/uso terapêutico
5.
Surg Obes Relat Dis ; 13(12): 1975-1983, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29055668

RESUMO

BACKGROUND: Roux-en-Y gastric bypass (RYGB) accelerates gastric pouch emptying, enhances postprandial glucagon-like peptide 1 (GLP-1) and insulin, and lowers glucose concentrations. To prevent discomfort and reactive hypoglycemia, it is recommended that post-RYGB patients eat small, frequent meals and avoid caloric drinks. However, the effect of meal size and texture on GLP-1 and metabolic response has not been studied. OBJECTIVES: To demonstrate that frequent minimeals and solid meals (S) elicit less GLP-1 and insulin release and less reactive hypoglycemia and are better tolerated compared with a single isocaloric liquid meal (L). SETTING: A university hospital. METHODS: In this prospective study, 32 RYGB candidates were enrolled and randomized to L or S groups before gastric bypass. Each subject received an L or S 600-kcal single meal (SM) administered at hour 0 or three 200-kcal minimeals administered at hours 0, 2, and 4 on 2 separate days. Twenty-one patients were retested 1 year after RYGB. Blood and visual analogue scale measurements were collected up to 6 hours postprandially. Outcome measures included gastric pouch emptying, glucose, insulin, and GLP-1; hunger, fullness, and stomach discomfort were measured by visual analogue scale. RESULTS: Twenty-one were patients retested after RYGB (L: n = 12; S: n = 9). Meal texture had a significant effect on peak GLP-1 (L-SM: 106.1 ± 67.2 versus S-SM: 45.3 ± 25.2 pM, P = .004), peak insulin, and postprandial glucose. Hypoglycemia was more frequent after the L-SM meal compared with the S-SM. Gastric pouch emptying was 2.4 times faster after RYGB but was not affected by texture. CONCLUSIONS: Meal texture and size have significant impact on tolerance and metabolic response after RYGB.


Assuntos
Dieta , Derivação Gástrica , Esvaziamento Gástrico/fisiologia , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Obesidade Mórbida/metabolismo , Obesidade Mórbida/cirurgia , Adulto , Glicemia , Feminino , Humanos , Insulina/sangue , Masculino , Refeições , Obesidade Mórbida/fisiopatologia , Estudos Prospectivos , Adulto Jovem
6.
Diabetes Care ; 40(1): 7-15, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27999001

RESUMO

OBJECTIVE: The superior effect of Roux-en-Y gastric bypass (RYGB) on glucose control compared with laparoscopic adjustable gastric banding (LAGB) is confounded by the greater weight loss after RYGB. We therefore examined the effect of these two surgeries on metabolic parameters matched on small and large amounts of weight loss. RESEARCH DESIGN AND METHODS: Severely obese individuals with type 2 diabetes were tested for glucose metabolism, ß-cell function, and insulin sensitivity after oral and intravenous glucose stimuli, before and 1 year after RYGB and LAGB, and at 10% and 20% weight loss after each surgery. RESULTS: RYGB resulted in greater glucagon-like peptide 1 release and incretin effect, compared with LAGB, at any level of weight loss. RYGB decreased glucose levels (120 min and area under the curve for glucose) more than LAGB at 10% weight loss. However, the improvement in glucose metabolism, the rate of diabetes remission and use of diabetes medications, insulin sensitivity, and ß-cell function were similar after the two types of surgery after 20% equivalent weight loss. CONCLUSIONS: Although RYGB retained its unique effect on incretins, the superiority of the effect of RYGB over that of LAGB on glucose metabolism, which is apparent after 10% weight loss, was attenuated after larger weight loss.


Assuntos
Cirurgia Bariátrica/métodos , Diabetes Mellitus Tipo 2/metabolismo , Derivação Gástrica/métodos , Glucose/metabolismo , Obesidade/cirurgia , Edulcorantes/metabolismo , Adulto , Diabetes Mellitus Tipo 2/etiologia , Diabetes Mellitus Tipo 2/cirurgia , Feminino , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Glucose/administração & dosagem , Humanos , Incretinas/metabolismo , Resistência à Insulina/fisiologia , Estudos Longitudinais , Masculino , Pessoa de Meia-Idade , Obesidade/complicações , Obesidade/metabolismo , Período Pós-Operatório , Estudos Prospectivos , Edulcorantes/administração & dosagem , Redução de Peso/fisiologia
8.
Diabetes ; 63(4): 1214-23, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24296713

RESUMO

The mechanisms responsible for the remarkable remission of type 2 diabetes after Roux-en-Y gastric bypass (RYGBP) are still puzzling. To elucidate the role of the gut, we compared ß-cell function assessed during an oral glucose tolerance test (OGTT) and an isoglycemic intravenous glucose clamp (iso-IVGC) in: 1) 16 severely obese patients with type 2 diabetes, up to 3 years post-RYGBP; 2) 11 severely obese normal glucose-tolerant control subjects; and 3) 7 lean control subjects. Diabetes remission was observed after RYGBP. ß-Cell function during the OGTT, significantly blunted prior to RYGBP, normalized to levels of both control groups after RYGBP. In contrast, during the iso-IVGC, ß-cell function improved minimally and remained significantly impaired compared with lean control subjects up to 3 years post-RYGBP. Presurgery, ß-cell function, weight loss, and glucagon-like peptide 1 response were all predictors of postsurgery ß-cell function, although weight loss appeared to be the strongest predictor. These data show that ß-cell dysfunction persists after RYGBP, even in patients in clinical diabetes remission. This impairment can be rescued by oral glucose stimulation, suggesting that RYGBP leads to an important gastrointestinal effect, critical for improved ß-cell function after surgery.


Assuntos
Diabetes Mellitus Tipo 2/cirurgia , Derivação Gástrica , Trato Gastrointestinal/fisiologia , Células Secretoras de Insulina/fisiologia , Adulto , Glicemia/metabolismo , Diabetes Mellitus Tipo 2/fisiopatologia , Técnica Clamp de Glucose , Teste de Tolerância a Glucose , Humanos , Pessoa de Meia-Idade , Obesidade Mórbida/fisiopatologia , Obesidade Mórbida/cirurgia , Indução de Remissão , Redução de Peso
9.
PLoS One ; 8(6): e65317, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23762342

RESUMO

Activation of brain melanocortin-4 receptors (MC4-R) by α-melanocyte-stimulating hormone (MSH) or inhibition by agouti-related protein (AgRP) regulates food intake and energy expenditure and can modulate neuroendocrine responses to changes in energy balance. To examine the effects of AgRP inhibition on energy balance, a small molecule, non-peptide compound, TTP2515, developed by TransTech Pharma, Inc., was studied in vitro and in rodent models in vivo. TTP2515 prevented AgRP from antagonizing α-MSH-induced increases in cAMP in HEK 293 cells overexpressing the human MC4-R. When administered to rats by oral gavage TTP2515 blocked icv AgRP-induced increases in food intake, weight gain and adiposity and suppression of T4 levels. In both diet-induced obese (DIO) and leptin-deficient mice, TTP2515 decreased food intake, weight gain, adiposity and respiratory quotient. TTP2515 potently suppressed food intake and weight gain in lean mice immediately after initiation of a high fat diet (HFD) but had no effect on these parameters in lean chow-fed mice. However, when tested in AgRP KO mice, TTP2515 also suppressed food intake and weight gain during HFD feeding. In several studies TTP2515 increased T4 but not T3 levels, however this was also observed in AgRP KO mice. TTP2515 also attenuated refeeding and weight gain after fasting, an effect not evident in AgRP KO mice when administered at moderate doses. This study shows that TTP2515 exerts many effects consistent with AgRP inhibition however experiments in AgRP KO mice indicate some off-target effects of this drug. TTP2515 was particularly effective during fasting and in mice with leptin deficiency, conditions in which AgRP is elevated, as well as during acute and chronic HFD feeding. Thus the usefulness of this drug in treating obesity deserves further exploration, to define the AgRP dependent and independent mechanisms by which TTP2515 exerts its effects on energy balance.


Assuntos
Proteína Relacionada com Agouti/antagonistas & inibidores , Fármacos Antiobesidade/farmacologia , Ingestão de Alimentos/efeitos dos fármacos , Metabolismo Energético/efeitos dos fármacos , Receptor Tipo 4 de Melanocortina/agonistas , alfa-MSH/genética , Adiposidade/efeitos dos fármacos , Administração Oral , Proteína Relacionada com Agouti/genética , Proteína Relacionada com Agouti/metabolismo , Animais , Dieta Hiperlipídica , Humanos , Leptina/deficiência , Leptina/genética , Masculino , Camundongos , Camundongos Knockout , Ratos , Receptor Tipo 4 de Melanocortina/genética , Receptor Tipo 4 de Melanocortina/metabolismo , Tiroxina/sangue , Tri-Iodotironina/sangue , Aumento de Peso/efeitos dos fármacos , alfa-MSH/metabolismo
10.
Obes Surg ; 23(8): 1294-301, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23549962

RESUMO

BACKGROUND: Weight regain after gastric bypass (GBP) can be associated with a gastrogastric fistula (GGF), in which a channel forms between the gastric pouch and gastric remnant, allowing nutrients to pass through the "old route" rather than bypassing the duodenum. To further understand the mechanisms by which GGF may lead to weight regain, we investigated gut hormone levels in GBP patients with a GGF, before and after repair. MATERIALS AND METHODS: Seven post-GBP subjects diagnosed with GGF were studied before and 4 months after GGF repair. Another cohort of 22 GBP control subjects without GGF complication were studied before and 1 year post-GBP. All subjects underwent a 50-g oral glucose tolerance test and blood was collected from 0-120 min for glucose, insulin, ghrelin, PYY3-36, GIP, and GLP-1 levels. RESULTS: Four months after GGF repair subjects lost 6.0 ± 3.9 kg and had significantly increased postprandial PYY3-36 levels. After GGF repair, fasting and postprandial ghrelin levels decreased and were strongly correlated with weight loss. The insulin response to glucose also tended to be increased after GGF repair, however no concomitant increase in GLP-1 was observed. Compared to the post-GBP group, GLP-1 and PYY3-36 levels were significantly lower before GGF repair; however, after GGF repair, PYY3-36 levels were no longer lower than the post-GBP group. CONCLUSIONS: These data utilize the GGF model to highlight the possible role of duodenal shunting as a mechanism of sustained weight loss after GBP, and lend support to the potential link between blunted satiety peptide release and weight regain.


Assuntos
Derivação Gástrica , Fístula Gástrica/cirurgia , Hormônios Gastrointestinais/sangue , Obesidade Mórbida/sangue , Complicações Pós-Operatórias/sangue , Adulto , Glicemia/metabolismo , Índice de Massa Corporal , Endoscopia Gastrointestinal/métodos , Feminino , Fístula Gástrica/diagnóstico , Fístula Gástrica/etiologia , Gastroscopia/métodos , Grelina/sangue , Peptídeo 1 Semelhante ao Glucagon/sangue , Humanos , Insulina/sangue , Laparoscopia/métodos , Masculino , Obesidade Mórbida/complicações , Obesidade Mórbida/cirurgia , Fragmentos de Peptídeos/sangue , Peptídeo YY/sangue , Complicações Pós-Operatórias/etiologia , Complicações Pós-Operatórias/cirurgia , Período Pós-Operatório , Período Pré-Operatório , Técnicas de Sutura , Fatores de Tempo , Resultado do Tratamento , Estados Unidos/epidemiologia , Redução de Peso
11.
Endocrinology ; 153(9): 4246-55, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22778225

RESUMO

Proopiomelanocortin (POMC) is posttranslationally processed to several peptides including α-MSH, a primary regulator of energy balance that inhibits food intake and stimulates energy expenditure. However, another POMC-derived peptide, ß-endorphin (ß-EP), has been shown to stimulate food intake. In this study we examined the effects of intracerebroventricular (icv) ß-EP on food intake and its ability to antagonize the negative effects of α-MSH on energy balance in male rats. A single icv injection of ß-EP stimulated food intake over a 2- to 6-h period during both the light and dark cycles. This effect was, however, not sustained with chronic icv ß-EP infusion. In the next study, a subthreshold dose of ß-EP was injected together with Nle(4), d-Phe(7) (NDP)-MSH after a 16-h fast, and the negative effects of NDP-MSH on refeeding and body weight gain were partially reversed. Finally, peptide interactions were studied in a chronic icv infusion model. Weight gain and food intake were significantly suppressed in the NDP-MSH group during the entire study. A subthreshold dose of ß-EP antagonized these suppressive effects on food intake and weight gain for the first 3 d. However on d 4-7, ß-EP no longer blocked these effects. Of note, the stimulatory effect of ß-EP on feeding and its ability to antagonize MSH were specific for ß-EP(1-31) and were not observed with ß-EP(1-27). This study highlights the importance of understanding how the balance between α-MSH and ß-EP is maintained and the potential role of differential POMC processing in regulating energy balance.


Assuntos
Peso Corporal/efeitos dos fármacos , Ingestão de Alimentos/efeitos dos fármacos , alfa-MSH/farmacologia , beta-Endorfina/farmacologia , Animais , Masculino , Ratos , Ratos Sprague-Dawley
12.
Peptides ; 37(1): 6-12, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22800691

RESUMO

This study used two mouse models with genetic manipulation of the melanocortin system to investigate prolactin regulation. Mice with overexpression of the melanocortin receptor (MC-R) agonist, α-melanocyte-stimulating hormone (Tg-MSH) or deletion of the MC-R antagonist agouti-related protein (AgRP KO) were studied. Male Tg-MSH mice had lower blood prolactin levels at baseline (2.9±0.3 vs. 4.7±0.7ng/ml) and after restraint stress (68±6.5 vs. 117±22ng/ml) vs. WT (p<0.05); however, pituitary prolactin content was not different. Blood prolactin was also decreased in male AgRP KO mice at baseline (4.2±0.5 vs. 7.6±1.3ng/ml) and after stress (60±4.5 vs. 86.1±5.7ng/ml) vs. WT (p<0.001). Pituitary prolactin content was lower in male AgRP KO mice (4.3±0.3 vs. 6.7±0.5µg/pituitary, p<0.001) vs. WT. No differences in blood or pituitary prolactin levels were observed in female AgRP KO mice vs. WT. Hypothalamic dopamine activity was assessed as the potential mechanism responsible for changes in prolactin levels. Hypothalamic tyrosine hydroxylase mRNA was measured in both genetic models vs. WT mice and hypothalamic dopamine and 3,4-dihydroxyphenylacetic acid (DOPAC) content were measured in male AgRP KO and WT mice but neither were significantly different. However, these results do not preclude changes in dopamine activity as dopamine turnover was not directly investigated. This is the first study to show that baseline and stress-induced prolactin release and pituitary prolactin content are reduced in mice with genetic alterations of the melanocortin system and suggests that changes in hypothalamic melanocortin activity may be reflected in measurements of serum prolactin levels.


Assuntos
Hipotálamo/metabolismo , Melanocortinas/fisiologia , Prolactina/sangue , Ácido 3,4-Di-Hidroxifenilacético/metabolismo , Proteína Relacionada com Agouti/genética , Proteína Relacionada com Agouti/metabolismo , Animais , Corticosterona/sangue , Dopamina/metabolismo , Antagonistas de Dopamina/farmacologia , Feminino , Expressão Gênica , Técnicas de Inativação de Genes , Hipotálamo/enzimologia , Masculino , Melanocortinas/metabolismo , Metoclopramida/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Hipófise/metabolismo , Tirosina 3-Mono-Oxigenase/genética , Tirosina 3-Mono-Oxigenase/metabolismo , alfa-MSH/genética , alfa-MSH/metabolismo
13.
Endocrinology ; 151(3): 1002-9, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20056830

RESUMO

Hypothalamic proopiomelanocortin (POMC)-derived MSH peptides and the melanocortin receptor antagonist, agouti-related protein (AgRP), interact to regulate energy balance. Both POMC and AgRP neurons express estrogen receptors, but little is known about estrogen regulation of the melanocortin system in the primate. We have therefore examined the effects of physiological doses of estradiol (E2) on POMC and AgRP in lumbar cerebrospinal fluid (CSF) of ovariectomized monkeys. POMC prohormone was measured by ELISA. AgRP was measured by RIA (sensitive for the more biologically active C-terminal AgRP(83-132) but also detects full-length AgRP) and by ELISA (measures primarily full length AgRP). In the first experiment, 14 animals were studied before and after 3 wk of E2. CSF POMC did not change, but AgRP(RIA) decreased from 7.9 +/- 1.2 to 4.7 +/- 1.2 fmol/ml after E2 (P = 0.03) and the POMC/AgRP(RIA) ratio increased from 4.2 +/- 0.89 to 6.8 +/- 1.04 (P = 0.04). AgRP(ELISA) did not change, but the ratio of AgRP(RIA) compared with AgRP(ELISA) was reduced after E2 (P = 0.02). In the second experiment, 11 animals were studied after 6 wk of E2, and similar changes were noted. The degree of AgRP(RIA) suppression with E2 was inversely related to body mass index (r = 0.569; P = 0.03). These results show for the first time that E2 suppresses AgRP(C-terminal) in CSF, increases the POMC to AgRP ratio, and may decrease AgRP processing, thus leading to increased melanocortin signaling. Furthermore, obesity was associated with resistance to the suppressive effects of E2 on AgRP, analogous to what is seen with obesity and leptin resistance.


Assuntos
Proteína Relacionada com Agouti/líquido cefalorraquidiano , Estradiol/metabolismo , Hipotálamo/metabolismo , Pró-Opiomelanocortina/líquido cefalorraquidiano , Animais , Estradiol/administração & dosagem , Feminino , Macaca mulatta , Ovariectomia
14.
Nat Med ; 15(10): 1195-201, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19767734

RESUMO

Reduced food intake brings about an adaptive decrease in energy expenditure that contributes to the recidivism of obesity after weight loss. Insulin and leptin inhibit food intake through actions in the central nervous system that are partly mediated by the transcription factor FoxO1. We show that FoxO1 ablation in pro-opiomelanocortin (Pomc)-expressing neurons in mice (here called Pomc-Foxo1(-/-) mice) increases Carboxypeptidase E (Cpe) expression, resulting in selective increases of alpha-melanocyte-stimulating hormone (alpha-Msh) and carboxy-cleaved beta-endorphin, the products of Cpe-dependent processing of Pomc. This neuropeptide profile is associated with decreased food intake and normal energy expenditure in Pomc-Foxo1(-/-) mice. We show that Cpe expression is downregulated by diet-induced obesity and that FoxO1 deletion offsets the decrease, protecting against weight gain. Moreover, moderate Cpe overexpression in the arcuate nucleus phenocopies features of the FoxO1 mutation. The dissociation of food intake from energy expenditure in Pomc-Foxo1(-/-) mice represents a model for therapeutic intervention in obesity and raises the possibility of targeting Cpe to develop weight loss medications.


Assuntos
Carboxipeptidase H/genética , Ingestão de Alimentos/fisiologia , Fatores de Transcrição Forkhead/metabolismo , Neurônios/fisiologia , Obesidade/genética , Animais , Núcleo Arqueado do Hipotálamo/metabolismo , Carboxipeptidase H/metabolismo , Feminino , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead/genética , Hipotálamo/metabolismo , Masculino , Camundongos , Camundongos Knockout , Pró-Opiomelanocortina/genética , Pró-Opiomelanocortina/metabolismo , Transdução de Sinais/genética , alfa-MSH/genética , alfa-MSH/metabolismo , beta-Endorfina/genética , beta-Endorfina/metabolismo
15.
J Biol Chem ; 281(42): 31594-604, 2006 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-16606609

RESUMO

Cells acquire cholesterol either by de novo synthesis in the endoplasmic reticulum or by internalization of cholesterol-containing lipoproteins, particularly low density lipoprotein (LDL), via receptor-mediated endocytosis. The inherited disorder Niemann-Pick type C (NPC), in which abnormal LDL-cholesterol trafficking from the endo/lysosomal compartment leads to substantial cholesterol and glycolipid accumulation in lysosomes, is caused by defects in either of two genes that encode for proteins designated as NPC1 and NPC2. NPC2 is a small intralysosomal protein that has been characterized biochemically as a cholesterol binding protein. We determined the rate and mechanism by which NPC2 delivers cholesterol to model phospholipid membranes. A fluorescence dequenching assay was used to monitor the kinetics of cholesterol transfer from the protein to membranes. The endogenous tryptophan fluorescence of the NPC2 was quenched upon binding of cholesterol, and the subsequent addition of acceptor vesicles resulted in dequenching of the tryptophan signal, enabling the monitoring of cholesterol transfer to membranes. The rates of cholesterol transfer were evaluated as a function of acceptor vesicle concentration, acceptor vesicle phospholipid headgroup composition, and aqueous phase properties. The results suggest that NPC2 rapidly transports cholesterol to phospholipid vesicles via a collisional mechanism which involves a direct interaction with the acceptor membrane. Transfer of cholesterol to membranes is faster in an acidic environment and is greatly enhanced by the presence of the unique lysosomal/late endosomal phospholipid lyso-bisphosphatidic acid (LBPA) (also known as bismonoacylglycerol phosphate). Finally, we found that the rate of transfer of cholesterol from vesicles to NPC2 was dramatically increased by the presence of lyso-bisphosphatidic acid in the donor vesicles. These results support a role for the NPC2 protein in the egress of LDL derived cholesterol out of the endosomal/lysosomal compartment.


Assuntos
Proteínas de Transporte/fisiologia , Colesterol/metabolismo , Endossomos/metabolismo , Glicoproteínas/fisiologia , Lisossomos/metabolismo , Animais , Transporte Biológico , Células CHO , Membrana Celular/metabolismo , Colesterol/química , Cricetinae , Retículo Endoplasmático/metabolismo , Humanos , Lipoproteínas LDL/química , Lisofosfolipídeos/química , Monoglicerídeos/química , Ligação Proteica , Proteínas de Transporte Vesicular
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...