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

Bases de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 114(10): 2747-2752, 2017 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-28143927

RESUMEN

Genetic disruption or pharmacologic inhibition of glucagon signaling effectively lowers blood glucose but results in compensatory glucagon hypersecretion involving expansion of pancreatic α-cell mass. Ben-Zvi et al. recently reported that angiopoietin-like protein 4 (Angptl4) links glucagon receptor inhibition to hyperglucagonemia and α-cell proliferation [Ben-Zvi et al. (2015) Proc Natl Acad Sci USA 112:15498-15503]. Angptl4 is a secreted protein and inhibitor of lipoprotein lipase-mediated plasma triglyceride clearance. We report that Angptl4-/- mice treated with an anti-glucagon receptor monoclonal antibody undergo elevation of plasma glucagon levels and α-cell expansion similar to wild-type mice. Overexpression of Angptl4 in liver of mice caused a 8.6-fold elevation in plasma triglyceride levels, but did not alter plasma glucagon levels or α-cell mass. Furthermore, administration of glucagon receptor-blocking antibody to healthy individuals increased plasma glucagon and amino acid levels, but did not change circulating Angptl4 concentration. These data show that Angptl4 does not link glucagon receptor inhibition to compensatory hyperglucagonemia or expansion of α-cell mass, and that it cannot be given to induce such secretion and growth. The reduction of plasma triglyceride levels in Angptl4-/- mice and increase following Angptl4 overexpression suggest that changes in plasma triglyceride metabolism do not regulate α-cells in the pancreas. Our findings corroborate recent data showing that increased plasma amino acids and their transport into α-cells link glucagon receptor blockage to α-cell hyperplasia.


Asunto(s)
Proteína 4 Similar a la Angiopoyetina/genética , Glucagón/metabolismo , Hiperplasia/genética , Metabolismo de los Lípidos/genética , Animales , Glucemia/genética , Proliferación Celular/genética , Glucagón/genética , Células Secretoras de Glucagón/metabolismo , Células Secretoras de Glucagón/patología , Humanos , Hiperplasia/sangre , Hiperplasia/patología , Lipoproteínas/sangre , Hígado/metabolismo , Hígado/patología , Ratones , Ratones Noqueados , Receptores de Glucagón/genética , Transducción de Señal/genética , Triglicéridos/sangre
2.
Diabetes Obes Metab ; 20(2): 283-291, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-28755409

RESUMEN

AIMS: Glucagon receptor (GCGR) blockers are being investigated as potential therapeutics for type 1 and type 2 diabetes. Here we report the safety, tolerability, pharmacokinetics (PK) and pharmacodynamics (PD) of REGN1193, a fully human glucagon receptor blocking monoclonal antibody from a first-in-human healthy volunteer randomized double-blinded trial. METHODS: Healthy men and women received single ascending doses of REGN1193 ranging from 0.05 to 0.6 mg/kg (n = 42) or placebo (n = 14) intravenously. Safety, tolerability and PK were assessed over 106 days. The glucose-lowering effect of REGN1193 was assessed after induction of hyperglycaemia by serial glucagon challenges. RESULTS: REGN1193 was generally well tolerated. There were small (<3× the upper limit of normal) and transient dose-dependent increases in hepatic aminotransferases. No increase in LDL-C was observed. Hypoglycaemia, assessed as laboratory blood glucose ≤70 mg/dL, occurred in 6/14 (43%) subjects on placebo and 27/42 (57%) on REGN1193 across all dose groups. All episodes of hypoglycaemia were asymptomatic, >50 mg/dL, and did not require treatment or medical assistance. Concentration-time profiles suggest a 2-compartment disposition and marked nonlinearity, consistent with target-mediated clearance. REGN1193 inhibited the glucagon-stimulated glucose increase in a dose-dependent manner. The 0.6 mg/kg dose inhibited the glucagon-induced glucose area under the curve for 0 to 90 minutes (AUC0-90 minutes ) by 80% to 90% on days 3 and 15, while blunting the increase in C-peptide. REGN1193 dose-dependently increased total GLP-1, GLP-2 and glucagon, with plasma levels returning to baseline by day 29 in all dose groups. CONCLUSION: REGN1193, a GCGR-blocking monoclonal antibody, produced a safety, tolerability and PK/PD profile suitable for further clinical development. The occurrence of transient elevations in serum hepatic aminotransferases observed here and reported with several small molecule glucagon receptor antagonists suggests an on-target effect of glucagon receptor blockade. The underlying mechanism is unknown.


Asunto(s)
Anticuerpos Monoclonales/farmacocinética , Hipoglucemiantes/farmacocinética , Receptores de Glucagón/antagonistas & inhibidores , Adulto , Anticuerpos Bloqueadores/administración & dosificación , Anticuerpos Bloqueadores/efectos adversos , Anticuerpos Bloqueadores/sangre , Anticuerpos Bloqueadores/farmacología , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/efectos adversos , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales Humanizados , Biomarcadores/sangre , Glucemia/análisis , Estudios de Cohortes , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Femenino , Estudios de Seguimiento , Semivida , Humanos , Hipoglucemiantes/administración & dosificación , Hipoglucemiantes/efectos adversos , Hipoglucemiantes/farmacología , Infusiones Intravenosas , Perdida de Seguimiento , Masculino , Tasa de Depuración Metabólica , Persona de Mediana Edad , Pacientes Desistentes del Tratamiento , Receptores de Glucagón/metabolismo , Adulto Joven
3.
Sci Transl Med ; 15(723): eadd4897, 2023 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-37992152

RESUMEN

Deficiency in the adipose-derived hormone leptin or leptin receptor signaling causes class 3 obesity in individuals with genetic loss-of-function mutations in leptin or its receptor LEPR and metabolic and liver disease in individuals with hypoleptinemia secondary to lipoatrophy such as in individuals with generalized lipodystrophy. Therapies that restore leptin-LEPR signaling may resolve these metabolic sequelae. We developed a fully human monoclonal antibody (mAb), REGN4461 (mibavademab), that activates the human LEPR in the absence or presence of leptin. In obese leptin knockout mice, REGN4461 normalized body weight, food intake, blood glucose, and insulin sensitivity. In a mouse model of generalized lipodystrophy, REGN4461 alleviated hyperphagia, hyperglycemia, insulin resistance, dyslipidemia, and hepatic steatosis. In a phase 1, randomized, double-blind, placebo-controlled two-part study, REGN4461 was well tolerated with an acceptable safety profile. Treatment of individuals with overweight or obesity with REGN4461 decreased body weight over 12 weeks in those with low circulating leptin concentrations (<8 ng/ml) but had no effect on body weight in individuals with higher baseline leptin. Furthermore, compassionate-use treatment of a single patient with atypical partial lipodystrophy and a history of undetectable leptin concentrations associated with neutralizing antibodies to metreleptin was associated with noteable improvements in circulating triglycerides and hepatic steatosis. Collectively, these translational data unveil an agonist LEPR mAb that may provide clinical benefit in disorders associated with relatively low leptin concentrations.


Asunto(s)
Resistencia a la Insulina , Lipodistrofia Generalizada Congénita , Animales , Ratones , Humanos , Leptina/uso terapéutico , Ensayos de Uso Compasivo , Receptores de Leptina/metabolismo , Lipodistrofia Generalizada Congénita/tratamiento farmacológico , Obesidad/tratamiento farmacológico , Anticuerpos/uso terapéutico , Peso Corporal
4.
Cell Metab ; 6(6): 506-12, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18054319

RESUMEN

An overactive renin-angiotensin system is associated with obesity and the metabolic syndrome. However, the mechanisms behind it are unclear. Cleaving angiotensinogen to angiotensin I by renin is a rate-limiting step of angiotensin II production, but renin is suggested to have angiotensin-independent effects. We generated mice lacking renin (Ren1c) using embryonic stem cells from C57BL/6 mice, a strain prone to diet-induced obesity. Ren1c(-/-) mice are lean, insulin sensitive, and resistant to diet-induced obesity without changes in food intake and physical activity. The lean phenotype is likely due to a higher metabolic rate and gastrointestinal loss of dietary fat. Most of the metabolic changes in Ren1c(-/-) mice were reversed by angiotensin II administration. These results support a role for angiotensin II in the pathogenesis of diet-induced obesity and insulin resistance.


Asunto(s)
Grasas de la Dieta/metabolismo , Metabolismo Energético , Obesidad/prevención & control , Renina/deficiencia , Tejido Adiposo/metabolismo , Angiotensina II/deficiencia , Angiotensina II/farmacología , Animales , Metabolismo Basal , Diacilglicerol O-Acetiltransferasa/genética , Diacilglicerol O-Acetiltransferasa/metabolismo , Grasas de la Dieta/administración & dosificación , Insulina/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/genética , Obesidad/metabolismo , Fenotipo , Renina/genética , Delgadez/genética , Delgadez/metabolismo
5.
Endocrinology ; 149(4): 1581-90, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18096662

RESUMEN

To determine the role of STAT3 in adipose tissue, we used Cre-loxP DNA recombination to create mice with an adipocyte-specific disruption of the STAT3 gene (ASKO mice). aP2-Cre-driven disappearance of STAT3 expression occurred on d 6 of adipogenesis, a time point when preadipocytes have already undergone conversion to adipocytes. Thus, this knockout model examined the role of STAT3 in mature but not differentiating adipocytes. Beginning at 9 wk of age, ASKO mice weighed more than their littermate controls and had increased adipose tissue mass, associated with adipocyte hypertrophy, but not adipocyte hyperplasia, hyperphagia, or reduced energy expenditure. Leptin-induced, but not isoproterenol-induced, lipolysis was impaired in ASKO adipocytes, which may partially explain the increased cell size. Despite reduced adiponectin and increased liver triacylglycerol, ASKO mice displayed normal glucose tolerance. Overall, these findings demonstrate that adipocyte STAT3 regulates body weight homeostasis in part through direct effects of leptin on adipocytes.


Asunto(s)
Adipocitos/fisiología , Adiposidad , Peso Corporal , Factor de Transcripción STAT3/fisiología , Animales , Ingestión de Alimentos , Metabolismo Energético , Femenino , Leptina/farmacología , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal , Triglicéridos/biosíntesis
6.
Schizophr Res ; 101(1-3): 287-94, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18255275

RESUMEN

OBJECTIVE: To examine prospective changes in cardiovascular disease (CVD) and type-2 diabetes risk factors in young adult first episode psychotic (FEP) patients treated with second generation antipsychotic medications. METHODS: At baseline, fasting serum and anthropometric measures were obtained from 45 FEP patients and 41 healthy adults (controls) of similar age, ethnicity and sex; sixteen of the FEP patients remained on the same antipsychotic medication and were available for a second blood draw at 24 weeks of treatment. Serum was assayed for glucose, insulin, triglycerides, total cholesterol and high and low density lipoproteins (HDL, LDL), adiponectin, leptin, interleukin 6, E-selectin and VCAM-1. Wilcoxon nonparametric tests were used to compare risk markers between the FEP and control group at baseline and to evaluate pre-post treatment changes within the FEP group. RESULTS: At baseline, the distributions of risk marker values were similar between the two groups and the percentages of FEP patients and healthy controls who were overweight/obese, dyslipidemic, hyperglycemic, and hyperinsulinemic did not differ. At 24 weeks, compared to baseline, FEP patients showed significant increases in BMI (p=0.0002), glucose (p=0.0449), insulin (p=0.0161), cholesterol (p=0.0129), leptin (p=0.0215), and E-selectin (p=0.0195), and a decrease in adiponectin (p=0.0371). CONCLUSIONS: Among patients with first episode psychosis, 6-month treatment with second generation antipsychotics is associated with the exacerbation of pre-existing and emergence of new CVD and diabetes risk factors.


Asunto(s)
Antipsicóticos/efectos adversos , Enfermedades Cardiovasculares/inducido químicamente , Enfermedades Cardiovasculares/diagnóstico , Diabetes Mellitus/inducido químicamente , Diabetes Mellitus/diagnóstico , Adolescente , Adulto , Glucemia/efectos de los fármacos , Índice de Masa Corporal , HDL-Colesterol/sangre , LDL-Colesterol/sangre , Femenino , Estudios de Seguimiento , Humanos , Insulina/sangre , Leptina/sangre , Masculino , Trastornos Psicóticos/sangre , Trastornos Psicóticos/tratamiento farmacológico , Estudios Retrospectivos , Factores de Riesgo , Estadísticas no Paramétricas , Triglicéridos/metabolismo
7.
Endocrinology ; 148(4): 1598-604, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17204554

RESUMEN

Mitotic clonal expansion is believed to be necessary for 3T3-L1 adipocyte formation. Signal transducer and activator of transcription 3 (STAT3), a mitogenic signaling protein, is activated through tyrosine phosphorylation during the proliferative phases of adipogenesis. We hypothesize that this signaling protein plays a key role in mitotic clonal expansion and differentiation. Here we determined that the adipocyte differentiation cocktail containing isobutylmethylxanthine, dexamethasone, and insulin (MDI) induced STAT3 tyrosine phosphorylation indirectly through the synthesis of an autocrine/paracrine factor. We further determined that the factor has heparin binding properties and identified the factor as midkine, a pleiotrophic growth factor previously associated with neuronal development and oncogenesis. Recombinant midkine induced STAT3 tyrosine phosphorylation in a time- and dose-dependent manner and stimulated the proliferation of postconfluent 3T3-L1 cells. Midkine neutralizing antibodies inhibited differentiation-induced STAT3 tyrosine phosphorylation as well as adipogenesis. These results show that MDI-induced synthesis and release of midkine explains the delayed activation of STAT3 during adipogenesis and that the midkine-STAT3 signaling pathway plays a necessary role in mitotic clonal expansion and differentiation.


Asunto(s)
Comunicación Autocrina , Citocinas/fisiología , Factor de Transcripción STAT3/metabolismo , Células 3T3-L1 , Adipogénesis/efectos de los fármacos , Animales , Proliferación Celular/efectos de los fármacos , Citocinas/metabolismo , Citocinas/farmacología , Ratones , Midkina , Comunicación Paracrina , Biosíntesis de Proteínas , Proteínas Recombinantes/farmacología , Transcripción Genética
8.
Am J Clin Nutr ; 85(2): 346-54, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17284728

RESUMEN

Maximal weight loss observed in low-calorie diet (LCD) studies tends to be small, and the mechanisms leading to this low treatment efficacy have not been clarified. Less-than-expected weight loss with LCDs can arise from an increase in fractional energy absorption (FEA), adaptations in energy expenditure, or incomplete patient diet adherence. We systematically reviewed studies of FEA and total energy expenditure (TEE) in obese patients undergoing weight loss with LCDs and in patients with reduced obesity (RO), respectively. This information was used to support an energy balance model that was then applied to examine patient adherence to prescribed LCD treatment programs. In the limited available literature, FEA was unchanged from baseline in short-term (<12 wk) treatment studies with LCDs; no long-term (>or=26 wk) studies were found. Review of doubly labeled water and respiratory chamber studies identified 10 reports of TEE in RO patients (n = 150) with long-term weight loss. These patients, who were weight stable, had a TEE almost identical to measured or predicted values in never-obese subjects (weighted mean difference: 1.3%; range: -1.7-8.5%). Modeling of energy balance, as supported by reviewed FEA and TEE studies, suggests that obese subjects participating in LCD programs have a weight loss less than half of that predicted. The small maximal weight loss observed with LCD treatments thus is likely not due to gastrointestinal adaptations but may be attributed, by deduction, to difficulties with patient adherence or, to a lesser degree, to metabolic adaptations induced by negative energy balance that are not captured by the current models.


Asunto(s)
Ingestión de Energía/fisiología , Metabolismo Energético/fisiología , Conducta Alimentaria/fisiología , Obesidad/metabolismo , Pérdida de Peso/fisiología , Humanos , Modelos Biológicos
9.
Am J Psychiatry ; 162(9): 1744-6, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16135638

RESUMEN

OBJECTIVE: This study sought to determine if amantadine affects weight gain in psychiatric patients taking olanzapine. METHOD: Twenty-one adults who had gained at least 5 lb with olanzapine were randomly assigned to receive amantadine (N=12) or placebo (N=9) in addition to olanzapine. The length of time taking olanzapine ranged from 1 to 44 months. Body mass index, psychiatric status, and fasting blood levels were assessed at baseline and 12 weeks. RESULTS: Significantly fewer subjects taking amantadine gained weight, with a mean change in body mass index of -0.07 kg/m2 for the amantadine group and 1.24 kg/m2 for the placebo group. This effect remained significant when the authors controlled for baseline body mass index and length of olanzapine treatment. No changes in fasting glucose, insulin, leptin, prolactin, and lipid levels were seen. Positive and Negative Syndrome Scale scores remained stable. CONCLUSIONS: Amantadine induced weight stabilization in subjects taking olanzapine and was well tolerated.


Asunto(s)
Amantadina/uso terapéutico , Antipsicóticos/efectos adversos , Dopaminérgicos/uso terapéutico , Obesidad/inducido químicamente , Obesidad/tratamiento farmacológico , Pérdida de Peso/efectos de los fármacos , Amantadina/farmacología , Antipsicóticos/uso terapéutico , Benzodiazepinas/efectos adversos , Benzodiazepinas/uso terapéutico , Índice de Masa Corporal , Dopaminérgicos/farmacología , Método Doble Ciego , Femenino , Humanos , Masculino , Olanzapina , Placebos , Esquizofrenia/tratamiento farmacológico , Resultado del Tratamiento
10.
Am J Psychiatry ; 162(1): 118-23, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15625209

RESUMEN

OBJECTIVE: Weight gain is a commonly observed adverse effect of atypical antipsychotic medications, but associated changes in energy balance and body composition are not well defined. The authors report here the effect of olanzapine on body weight, body composition, resting energy expenditure, and substrate oxidation as well as leptin, insulin, glucose, and lipid levels in a group of outpatient volunteers with first-episode psychosis. METHOD: Nine adults (six men and three women) experiencing their first psychotic episode who had no previous history of antipsychotic drug therapy began a regimen of olanzapine and were studied within 7 weeks and approximately 12 weeks after olanzapine initiation. RESULTS: After approximately 12 weeks of olanzapine therapy, the median increase in body weight was 4.7 kg, a significant increase of 7.3% from first observation. Body fat, measured by dual-energy x-ray absorptiometry, increased significantly, with a propensity for central fat deposition. Lean body mass and bone mineral content did not change. Resting energy expenditure, measured by indirect calorimetry, did not change. Respiratory quotient significantly increased 0.12 with olanzapine and was greatest in those who gained >5% of their initial weight. Fasting insulin, C-peptide, and triglyceride levels significantly increased, but there were no changes in glucose levels; total, high density lipoprotein, or low density lipoprotein cholesterol levels; or leptin levels. CONCLUSIONS: Olanzapine appears to have induced an increase in central body fat deposition, insulin, and triglyceride levels, suggesting the possible development of insulin resistance. The decrease in fat oxidation may be secondary or predispose patients to olanzapine-induced weight gain.


Asunto(s)
Antipsicóticos/farmacología , Antipsicóticos/uso terapéutico , Benzodiazepinas/farmacología , Benzodiazepinas/uso terapéutico , Composición Corporal/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Metabolismo Energético/efectos de los fármacos , Trastornos Psicóticos/tratamiento farmacológico , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/metabolismo , Adulto , Benzodiazepinas/efectos adversos , Péptido C/sangre , Femenino , Humanos , Insulina/sangre , Masculino , Obesidad/inducido químicamente , Olanzapina , Oxidación-Reducción/efectos de los fármacos , Trastornos Psicóticos/metabolismo , Trastornos Psicóticos/psicología , Fenómenos Fisiológicos Respiratorios/efectos de los fármacos , Esquizofrenia/tratamiento farmacológico , Esquizofrenia/metabolismo , Triglicéridos/sangre
11.
Endocrinology ; 156(8): 2781-94, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26020795

RESUMEN

Antagonizing glucagon action represents an attractive therapeutic option for reducing hepatic glucose production in settings of hyperglycemia where glucagon excess plays a key pathophysiological role. We therefore generated REGN1193, a fully human monoclonal antibody that binds and inhibits glucagon receptor (GCGR) signaling in vitro. REGN1193 administration to diabetic ob/ob and diet-induced obese mice lowered blood glucose to levels observed in GCGR-deficient mice. In diet-induced obese mice, REGN1193 reduced food intake, adipose tissue mass, and body weight. REGN1193 increased circulating levels of glucagon and glucagon-like peptide 1 and was associated with reversible expansion of pancreatic α-cell area. Hyperglucagonemia and α-cell hyperplasia was observed in fibroblast growth factor 21-deficient mice treated with REGN1193. Single administration of REGN1193 to diabetic cynomolgus monkeys normalized fasting blood glucose and glucose tolerance and increased circulating levels of glucagon and amino acids. Finally, administration of REGN1193 for 8 weeks to normoglycemic cynomolgus monkeys did not cause hypoglycemia or increase pancreatic α-cell area. In summary, the GCGR-blocking antibody REGN1193 normalizes blood glucose in diabetic mice and monkeys but does not produce hypoglycemia in normoglycemic monkeys. Thus, REGN1193 provides a potential therapeutic modality for diabetes mellitus and acute hyperglycemic conditions.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Glucemia/efectos de los fármacos , Diabetes Mellitus Experimental/sangre , Diabetes Mellitus Experimental/tratamiento farmacológico , Hipoglucemiantes/uso terapéutico , Receptores de Glucagón/inmunología , Animales , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales Humanizados , Glucemia/metabolismo , Diabetes Mellitus Experimental/complicaciones , Femenino , Factores de Crecimiento de Fibroblastos/genética , Humanos , Hipoglucemiantes/farmacología , Macaca fascicularis , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Ratones Transgénicos , Obesidad/complicaciones , Obesidad/tratamiento farmacológico , Obesidad/patología , Receptores de Glucagón/antagonistas & inhibidores , Receptores de Glucagón/genética
12.
Diabetes Care ; 35(3): 495-7, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22279033

RESUMEN

OBJECTIVE: To identify the characteristics associated with glycemic response to newly initiated insulin therapy. RESEARCH DESIGN AND METHODS: We identified 1,139 type 2 diabetic patients who initiated insulin therapy between 1 January 2009 and 30 June 2010. Outcomes of interest were the proportion of patients achieving A1C <7% and mean change in A1C within 3-9 months. RESULTS: Mean A1C at insulin initiation was 8.2 vs. 9.2% among those who did and did not attain A1C <7% (P < 0.001). Within a mean of 5 months, 464 (40.7%) patients attained A1C <7%. In multivariable analyses controlling for insulin regimen, dose, and oral agent use, preinsulin A1C was responsible for nearly all the explained variance in A1C change. Each one percentage point of preinsulin A1C reduced the probability of attaining <7% by 26% (odds ratio 0.74 [95% CI 0.68-0.80]). CONCLUSIONS: Insulin initiation at lower levels of A1C improves goal attainment and independently increases glycemic response.


Asunto(s)
Glucemia/efectos de los fármacos , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Hipoglucemiantes/uso terapéutico , Insulina/uso terapéutico , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/embriología , Hemoglobina Glucada/metabolismo , Humanos , Persona de Mediana Edad , Análisis Multivariante
14.
J Nutr ; 137(5): 1236-43, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17449587

RESUMEN

Obesity is associated with an impaired immune response, an increased susceptibility to bacterial infection, and a chronic increase in proinflammatory cytokines such as IL-6 and TNFalpha. However, few studies have examined the effect of obesity on the immune response to viral infections. Because infection with influenza is a leading cause of morbidity and mortality worldwide, we investigated the effect of obesity on early immune responses to influenza virus exposure. Diet-induced obese and lean control C57BL/6 mice were infected with influenza A/PR8/34, and lung pathology and immune responses were examined at d 0 (uninfected), 3, and 6, postinfection. Following infection, diet-induced obese mice had a significantly higher mortality rate than the lean controls and elevated lung pathology. Antiviral and proinflammatory cytokine mRNA production in the lungs of the infected mice was markedly different between obese and lean mice. IFNalpha and beta were only minimally expressed in the infected lungs of obese mice and there was a notable delay in expression of the proinflammatory cytokines IL-6 and TNFalpha. Additionally, obese mice had a substantial reduction in NK cell cytotoxicity. These data indicate that obesity inhibits the ability of the immune system to appropriately respond to influenza infection and suggests that obesity may lead to increased morbidity and mortality from viral infections.


Asunto(s)
Dieta , Obesidad/complicaciones , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/mortalidad , Orthomyxoviridae , Animales , Quimiocina CCL2/genética , Quimiocina CCL5/genética , Citocinas/sangre , Citocinas/inmunología , Citotoxicidad Inmunológica , Insulina/sangre , Células Asesinas Naturales/inmunología , Leptina/sangre , Pulmón/metabolismo , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Obesidad/etiología , Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/sangre , Infecciones por Orthomyxoviridae/complicaciones , Infecciones por Orthomyxoviridae/patología , ARN Mensajero/metabolismo , Carga Viral
15.
Biochem Biophys Res Commun ; 339(3): 923-31, 2006 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-16329991

RESUMEN

Protein inhibitor of activated STAT3 (PIAS3), a cytokine-induced repressor of signal transducer and activator of transcription 3 (STAT3) and a modulator of a broad array of nuclear proteins, is expressed in white adipose tissue, but its role in adipogenesis is not known. Here, we determined that PIAS3 was constitutively expressed in 3T3-L1 cells at all stages of adipogenesis. However, it translocated from the nucleus to the cytoplasm 4 days after induction of differentiation by isobutylmethylxanthine, dexamethasone, and insulin (MDI). In ob/ob mice, PIAS3 expression was increased in white adipose tissue depots compared to lean mice and was found in the cytoplasm of adipocytes. Overexpression of PIAS3 in differentiating preadipocytes, which localized primarily to the nucleus, inhibited mRNA level gene expression of adipogenic transcription factors C/EBPalpha and PPARgamma, as well as their downstream target genes aP2 and adiponectin. PIAS3 also inhibited C/EBPalpha promoter activation mediated specifically by insulin, but not dexamethasone or isobutylmethylxanthine. Taken together, these data suggest that PIAS3 may play an inhibitory role in adipogenesis by modulating insulin-activated transcriptional activation events. Increased PIAS3 expression in adipose tissue may play a role in the metabolic disturbances of obesity.


Asunto(s)
Adipocitos/citología , Adipocitos/metabolismo , Adipogénesis/fisiología , Proteína alfa Potenciadora de Unión a CCAAT/metabolismo , Insulina/metabolismo , Proteínas Inhibidoras de STAT Activados/metabolismo , Células 3T3-L1 , Animales , Diferenciación Celular , Regulación del Desarrollo de la Expresión Génica/fisiología , Ratones , PPAR gamma/metabolismo , Proteínas Inhibidoras de STAT Activados/genética , Proteínas Recombinantes/metabolismo
16.
Curr Opin Lipidol ; 15(3): 303-7, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15166786

RESUMEN

PURPOSE OF REVIEW: Adipose tissue is a dynamic organ that changes mass throughout life in response to the metabolic needs of the animal. In the past three decades, significant advances have been made in delineating key extracellular and intracellular stimulators of fat cell formation or adipogenesis. In this review, the author focuses on new findings of specific inhibitors of adipogenesis. Understanding the balance between positive and negative regulators of adipogenesis has important health-related implications for anti-obesity medical therapy and lipodystrophy. RECENT FINDINGS: Adipogenesis is a highly regulated process requiring coordinated expression and activation of two main groups of adipogenic transcription factors, CCAAT/enhancer binding proteins and peroxisome proliferators activated receptor gamma. In response to hormonal and nutrient stimuli, the increased expression and activation of these transcription factors induce the expression of adipocyte-specific genes. More recently, several groups have identified extracellular inhibitors of adipocyte formation, including cytokines, lipid molecules, genistein, and protease inhibitors. Intracellular signaling molecules, which negatively regulate adipogenesis, include Pref-1, Foxo1, Foxa2, SMAD-3, WNT-10b, GATA-2 and GATA-3. SUMMARY: The prevalence of obesity is increasing in the United States and in other westernized societies. Understanding the mechanisms of excessive energy storage in adipose tissue is necessary to develop a comprehensive strategy to prevent and treat obesity. One potential, but unrealized, approach to obesity treatment is to target excessive adipose tissue enlargement. A number of promising extra- and intracellular inhibitors of fat cell formation have been identified, but the modulation of adipose tissue mass may have both advantageous and deleterious health effects.


Asunto(s)
Tejido Adiposo/citología , Tejido Adiposo/metabolismo , Tejido Adiposo/efectos de los fármacos , Animales , Fármacos Anti-VIH/efectos adversos , Fármacos Anti-VIH/farmacología , Diferenciación Celular/efectos de los fármacos , Citocinas/metabolismo , Depresión Química , Ácidos Grasos/farmacología , Genisteína/farmacología , Síndrome de Lipodistrofia Asociada a VIH/inducido químicamente , Síndrome de Lipodistrofia Asociada a VIH/metabolismo , Síndrome de Lipodistrofia Asociada a VIH/patología , Humanos , Obesidad/tratamiento farmacológico , Obesidad/metabolismo , Obesidad/patología , Inhibidores de Proteasas/efectos adversos , Inhibidores de Proteasas/farmacología , Factores de Transcripción/metabolismo
17.
Cytokine ; 25(2): 61-7, 2004 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-14693161

RESUMEN

Interleukin-4 (IL-4) activates STAT6 in 3T3-L1 preadipocytes but its functional role is not known. In this report, we first assessed interleukin-4 receptor alpha (IL-4Ralpha) expression during adipogenesis. IL-4Ralpha was highly expressed in proliferating 3T3-L1 preadipocytes. Receptor expression was down-regulated in post-confluent growth arrested preadipocytes. Induction of differentiation led to a transient 36-h increase in expression, but then levels decreased to undetectable amounts 3-8 days after induction of differentiation. Depending on the cell type, IL-4 either increases or decreases cell proliferation. In growth arrested preconfluent 3T3-L1 preadipocytes, IL-4 alone had no effect on preadipocyte proliferation. In contrast, IL-4 inhibited platelet-derived growth factor (PDGF-BB) induced preadipocyte proliferation. PDGF-BB, but not IL-4, induced STAT3 tyrosine and AKT serine phosphorylation. Both PDGF-BB and IL-4 induced STAT6 tyrosine phosphorylation, but the bands showed distinct electrophoretic migration patterns. IL-4 alone and IL-4 added to the differentiation cocktail had no effect on adipocyte formation or PPARgamma expression. Collectively, these studies demonstrate that IL-4 inhibits PDGF-BB-induced preadipocyte proliferation, possibly through STAT6 activation. The pattern of IL-4 receptor expression suggests that the effects of IL-4 are targeted primarily towards preadipocytes.


Asunto(s)
Adipocitos/citología , Interleucina-4/farmacología , Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Células 3T3-L1 , Animales , Becaplermina , División Celular/efectos de los fármacos , Regulación hacia Abajo , Interleucina-4/fisiología , Ratones , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Proteínas Proto-Oncogénicas c-sis , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores de Interleucina-4/metabolismo , Factor de Transcripción STAT6 , Transducción de Señal , Células Madre/citología , Células Madre/efectos de los fármacos , Transactivadores/metabolismo , Factores de Transcripción/metabolismo
18.
Obes Res ; 10(10): 985-90, 2002 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12376578

RESUMEN

OBJECTIVE: To study the effect of dietary weight loss, postural change, and an oral glucose load on serum angiotensin-converting enzyme (ACE) activity in obese adults. RESEARCH METHODS AND PROCEDURES: Sixteen obese adult men and women with a mean body mass index of 35.7 +/- 4.3 kg/m(2) were studied after 1 week on a maintenance energy lead-in diet and after 5 weeks on an identical but 40% reduced-energy diet provided by the General Clinical Research Center (GCRC). ACE activity was measured spectrophotometrically. Plasma renin activity and serum aldosterone were measured by radioimmunoassay. RESULTS: All subjects lost weight, with a mean decrease in body weight of 7.0 +/- 2.1 kg or 6 +/- 3% of initial body weight (p < 0.00001). Systolic and diastolic blood pressure, supine plasma renin activity, and serum aldosterone levels decreased with weight loss (p < 0.05). Supine ACE activity decreased 23 +/- 12% with weight loss (p < 0.00001). Standing ACE activity, which was significantly higher than supine ACE activity before and after weight loss (p < 0.05), also decreased 18 +/- 17% with weight loss (p = 0.0007). A 75-g oral glucose load had no effect on serum ACE activity over a 3-hour period. DISCUSSION: In obese adults, serum ACE activity declines with modest weight loss, increases with postural change, and is unaffected by an oral glucose load.


Asunto(s)
Obesidad/enzimología , Peptidil-Dipeptidasa A/sangre , Pérdida de Peso/fisiología , Adulto , Aldosterona/sangre , Metabolismo Basal/fisiología , Glucemia/metabolismo , Presión Sanguínea/fisiología , Índice de Masa Corporal , Calorimetría , Dieta Reductora , Femenino , Humanos , Insulina/sangre , Masculino , Persona de Mediana Edad , Postura , Renina/sangre , Estadísticas no Paramétricas
19.
Biochem J ; 367(Pt 1): 203-8, 2002 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-12095417

RESUMEN

The tyrosine kinase inhibitor genistein inhibits 3T3-L1 adipogenesis when present during the first 72 h of differentiation. In this report, we investigated the underlying mechanisms involved in the anti-adipogenic effects of genistein. We found that genistein blocked the DNA binding and transcriptional activity of CCAAT/enhancer-binding protein beta (C/EBPbeta) during differentiation by promoting the expression of C/EBP homologous protein, a dominant-negative member of the C/EBP family. Loss of C/EBPbeta activity was manifested as a loss of differentiation-induced C/EBPalpha and peroxisome-proliferator-activated receptor gamma protein expression and a dramatic reduction in lipid accumulation. Further, we documented for the first time that C/EBPbeta was tyrosine-phosphorylated in vivo during differentiation and in vitro by activated epidermal growth factor receptor. Genistein inhibited both of these events. Collectively, these results indicate that genistein blocks adipogenesis and C/EBPbeta activity by increasing the level of C/EBP homologous protein and possibly by inhibiting the tyrosine phosphorylation of C/EBPbeta.


Asunto(s)
Adipocitos/citología , Proteína beta Potenciadora de Unión a CCAAT/antagonistas & inhibidores , Genisteína/farmacología , Células 3T3 , Adipocitos/metabolismo , Animales , Anticarcinógenos/farmacología , Compuestos Azo/farmacología , Diferenciación Celular , Línea Celular , Núcleo Celular/metabolismo , Colorantes/farmacología , ADN/metabolismo , Inhibidores Enzimáticos/farmacología , Immunoblotting , Isoflavonas/farmacología , Metabolismo de los Lípidos , Ratones , Fosforilación , Pruebas de Precipitina , Unión Proteica , Receptores Citoplasmáticos y Nucleares/metabolismo , Factores de Tiempo , Factores de Transcripción/metabolismo , Transcripción Genética , Transfección , Tirosina/metabolismo
20.
Am J Physiol Cell Physiol ; 286(2): C349-54, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-14576085

RESUMEN

Two pathways are initiated upon 3T3-L1 preadipocyte differentiation: the reentry of cells into the cell cycle and the initiation of a cascade of transcriptional events that "prime" the cell for differentiation. The "priming" event involves the synthesis of members of the CCAAT/enhancer binding protein (C/EBP) family of transcription factors. However, the relationship between these two pathways is unknown. Here we report that in the 3T3-L1 preadipocytes induced to differentiate, cell cycle progression and the initiation of differentiation are linked by a cell cycle-dependent Rb-C/EBPbeta interaction. Cell cycle arrest in G1 by l-mimosine inhibited differentiation-induced C/EBPbeta-DNA-binding activity and Rb phosphorylation. However, cell cycle arrest after the G1/S transition by aphidicolin or nocodazole did not prevent C/EBPbeta-DNA-binding activity or Rb phosphorylation. Furthermore, hypophosphorylated Rb and C/EBPbeta coimmunoprecipitated, whereas phosphorylated Rb and C/EBPbeta did not. Electrophoretic mobility shift assays demonstrated that recombinant hypophosphorylated Rb decreased C/EBPbeta-DNA-binding activity and that Rb overexpression inhibited C/EBPbeta-induced transcriptional activation of a C/EBPalpha-promoter-luciferase reporter gene. We conclude that C/EBPbeta-DNA-binding activity is regulated by its interaction with hypophosphorylated Rb, thereby linking the progression of the cell cycle to the initiation of differentiation during 3T3-L1 adipogenesis.


Asunto(s)
Adipocitos/citología , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteína de Retinoblastoma/fisiología , Células 3T3 , Animales , Ciclo Celular/fisiología , División Celular/fisiología , Proteínas de Unión al ADN/antagonistas & inhibidores , Ratones , Fosforilación , Proteína de Retinoblastoma/metabolismo , Activación Transcripcional/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA