Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 83
Filtrar
1.
Aliment Pharmacol Ther ; 47(12): 1639-1651, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29696666

RESUMEN

BACKGROUND: Sirtuin 1 (Sirt1) is suppressed in non-alcoholic fatty liver disease (NAFLD), while its' stimulation or overexpression results in reduced disease severity in pre-clinical NAFLD models. Leucine allosterically activates Sirt1 and synergise with other Sirt/AMPK/NO pathway activators. We developed a triple combination of leucine, metformin and sildenafil (NS-0200), which was effective in a mouse model of non-alcoholic steatohepatitis (NASH). AIM: To report the results from a Phase 2, randomised clinical trial of of NS-0200 in 91 subjects with NAFLD (liver fat ≥15% by magnetic resonance imaging-proton-density fat fraction (MRI-PDFF)). METHODS: Subjects were randomised to placebo, low-dose (1.1 g leucine/0.5 g metformin/0.5 mg sildenafil) or high-dose NS-0200 (1.1 g leucine/0.5 g metformin/1.0 mg sildenafil) b.d. for 16 weeks; change in hepatic fat was assessed via MRI-PDFF, and lipid metabolism was assessed via changes in the lipidomic signature. Seventy subjects completed the trial and met a priori compliance criteria. Analyses were conducted on the full cohort and on those with alanine aminotransferase (ALT) values above median (50 U/L; n = 35). RESULTS: In the full cohort, active treatments did not separate from placebo. High dose NS-0200 reduced hepatic fat by 15.7% (relative change from baseline) in the high ALT group (P < 0.005) while low dose NS-0200 and placebo did not significantly change hepatic fat. Lipidomic analysis showed dose-responsive treatment effects in both overall and high ALT cohorts, with significant decreases in metabolically active lipids and up-regulation of fatty acid oxidation. CONCLUSION: These data support further evaluation of high-dose NS-0200 for treating NASH, especially in those with elevated ALT (NCT 02546609).


Asunto(s)
Leucina/administración & dosificación , Metformina/administración & dosificación , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Citrato de Sildenafil/administración & dosificación , Adulto , Alanina Transaminasa/metabolismo , Estudios de Cohortes , Método Doble Ciego , Combinación de Medicamentos , Femenino , Humanos , Lípidos/química , Imagen por Resonancia Magnética/métodos , Masculino , Metformina/uso terapéutico , Enfermedad del Hígado Graso no Alcohólico/patología
2.
Int J Obes (Lond) ; 32(8): 1305-11, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18427562

RESUMEN

OBJECTIVE: We previously demonstrated that high calcium diets inhibit obesity in both mice and humans; this effect is mediated, in part, by dietary calcium suppression of 1alpha, 25-dihydroxyvitamin D, which exerts both non-genomic and genomic effects on adipocyte metabolism. However, the presence of the nuclear vitamin D receptor (nVDR) and its role in this regulation in adipocytes have not been investigated. METHODS AND RESULTS: The nVDR is expressed at low levels in preadipocytes, and differentiation induced a rapid 1150% increase within 3 h (P<0.001), which then declined rapidly to preadipocyte levels at differentiation day 3. However, this was not a differentiation event, as the components of the differentiation mixture (isobutylmethylxanthine and dexamethasone) also increased nVDR expression markedly by 1620 and 284%, respectively, in fully differentiated adipocytes (P<0.05). 1alpha, 25-Dihydroxyvitamin D and 11beta-dehydrocorticosterone each stimulated nVDR expression at 48 h, but not 3 h, by approximately 650% in fully differentiated adipocytes, whereas the combination augmented this effect (P<0.005). Knockdown of 11beta-hydroxysteroid dehydrogenase I (11beta-HSD I) markedly decreased adipocyte nVDR expression and attenuated 1alpha, 25-dihydroxyvitamin D stimulation of nVDR. Thus, 1alpha, 25-dihydroxyvitamin D stimulation of corticosteroid synthesis via the 11beta-HSD appears to provide an indirect positive feedback on the nVDR. 1alpha, 25-Dihydroxyvitamin D also regulated short-term corticosterone release independently of either 11beta-HSD I or nVDR. Knockdown 11beta-HSD I did not affect short-term corticosterone release, whereas modulating calcium influx by KCl, BAYK8644 and the membrane 1alpha, 25-dihydroxyvitamin D receptor agonist lumisterol markedly increased corticosterone release. CONCLUSION: These data suggest a potential role of nVDR and corticosterone in the regulation in adipocyte responses to 1alpha, 25-dihydroxyvitamin D and dietary calcium.


Asunto(s)
Adipocitos/metabolismo , Glucocorticoides/farmacología , Receptores de Calcitriol/metabolismo , Vitamina D/análogos & derivados , 1-Metil-3-Isobutilxantina/farmacología , Adipocitos/efectos de los fármacos , Adipogénesis/efectos de los fármacos , Animales , Diferenciación Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Células Cultivadas , Dexametasona/farmacología , Ratones , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Vitamina D/farmacología
3.
Obes Rev ; 9(5): 428-45, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18282178

RESUMEN

The influence of calcium and dairy food intake on energy balance is the object of a growing scientific literature. This manuscript presents the information discussed by subject experts during a symposium on calcium and obesity, initially planned to document in a comprehensive manner the role of calcium and dairy food on energy balance and body composition. This manuscript is organized into 13 propositions statements which either resume the presentation of an invited speaker or integrate recent developments in calcium-related obesity research. More specifically, the effects of calcium and dairy consumption on body weight and adiposity level, appetite, weight loss intervention outcome, lipid-lipoprotein profile and the risk to develop metabolic syndrome are discussed together with the metabolic mechanisms proposed to explain these effects. Taken together, the observations presented in this manuscript suggest that calcium and dairy food intake can influence many components of energy and fat balance, indicating that inadequate calcium/dairy intake may increase the risk of positive energy balance and of other health problems.


Asunto(s)
Calcio de la Dieta/administración & dosificación , Metabolismo Energético/efectos de los fármacos , Obesidad/etiología , Obesidad/prevención & control , Composición Corporal/efectos de los fármacos , Composición Corporal/fisiología , Calcio/fisiología , Productos Lácteos , Ingestión de Energía/efectos de los fármacos , Ingestión de Energía/fisiología , Metabolismo Energético/fisiología , Humanos
4.
Int J Obes (Lond) ; 30(9): 1341-6, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16520808

RESUMEN

OBJECTIVE: We have previously demonstrated that 1alpha, 25(OH)2D3 promotes adipocyte reactive oxygen species (ROS) production. We have now evaluated whether decreasing 1alpha, 25(OH)2D3 levels by increasing dietary calcium will decrease oxidative stress in vivo. METHODS: We fed low-calcium (0.4% Ca) and high-calcium (1.2% Ca from CaCO3) obesity-promoting (high sucrose/high fat) diets to aP2-agouti transgenic mice and assessed regulation of ROS production in adipose tissue and skeletal muscle. RESULTS: Mice on the high-calcium diet gained 50% of the body weight (P=0.04) and fat (P<0.001) as mice on the low-calcium diet (0.4% Ca). The high-calcium diet significantly reduced adipose intracellular ROS production by 64 and 18% (P<0.001) and inhibited adipose tissue nicotinamide adenine dinucleotide phosphate oxidase expression by 49% (P=0.012) and 63% (P=0.05) in visceral and subcutaneous adipose tissue, respectively. Adipocyte intracellular calcium ([Ca2+]i) levels were suppressed in mice on the high-calcium diet by 73-80% (P<0.001). The high-calcium diet also induced 367 and 191% increases in adipose mitochondrial uncoupling protein 2 (UCP2) expression (P<0.001) in visceral and subcutaneous adipose tissue, respectively. The pattern of UCP3 expression and indices of ROS production in skeletal muscle were consistent with those in adipose tissue. The high-calcium diet also suppressed 11beta-hydroxysteroid dehydrogenase (11beta-HSD) expression in visceral adipose tissue by 39% (P=0.034). 11beta-HSD expression was markedly higher in visceral vs subcutaneous adipose tissue in mice on the low-calcium diet (P=0.034), whereas no difference was observed between the fat depots in mice on the high-calcium diet. CONCLUSION: These data support a potential role for dietary calcium in the regulation of obesity-induced oxidative stress.


Asunto(s)
Calcio de la Dieta/farmacología , Especies Reactivas de Oxígeno/metabolismo , Adipocitos/metabolismo , Tejido Adiposo/metabolismo , Animales , Grasas de la Dieta/administración & dosificación , Sacarosa en la Dieta/administración & dosificación , Masculino , Ratones , Ratones Transgénicos , Músculo Esquelético/metabolismo , Obesidad/metabolismo , Reacción en Cadena de la Polimerasa/métodos , ARN/metabolismo , Distribución Aleatoria
5.
Int J Obes (Lond) ; 29(4): 391-7, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15672113

RESUMEN

BACKGROUND AND OBJECTIVE: We have previously demonstrated an antiobesity effect of dietary Ca; this is largely mediated by Ca suppression of calcitriol levels, resulting in reduced adipocyte intracellular Ca2+ and, consequently, a coordinated increase in lipid utilization and decrease in lipogenesis. Notably, dairy Ca is markedly more effective than other Ca sources. DESIGN: Obese subjects were placed on balanced deficit (-500 kcal/day) diets and randomized to control (400-500 mg Ca/day; n = 16) or yogurt (1100 mg Ca/day; n = 18) treatments for 12 weeks. Dietary macronutrients and fiber were held constant at the US average. MEASUREMENTS: Body weight, body fat and fat distribution (by dual-energy X-ray absorptiometry), blood pressure and circulating lipids were measured at baseline and after 12 weeks of intervention. RESULTS: Fat loss was markedly increased on the yogurt diet (-4.43+/-0.47 vs -2.75+/-0.73 kg in yogurt and control groups; P<0.005) while lean tissue loss was reduced by 31% on the yogurt diet. Trunk fat loss was augmented by 81% on the yogurt vs control diet (P<0.001), and this was reflected in a markedly greater reduction in waist circumference (-3.99+/-0.48 vs -0.58+/-1.04 cm, P<0.001). Further, the fraction of fat lost from the trunk was higher on the yogurt diet vs control (P<0.005). CONCLUSION: Isocaloric substitution of yogurt for other foods significantly augments fat loss and reduces central adiposity during energy restriction..


Asunto(s)
Calcio de la Dieta/administración & dosificación , Dieta Reductora , Obesidad/dietoterapia , Yogur , Adolescente , Adulto , Análisis de Varianza , Animales , Glucemia/análisis , Presión Sanguínea , Composición Corporal , Colesterol/sangre , HDL-Colesterol/sangre , LDL-Colesterol/sangre , Diástole , Femenino , Glicerol/sangre , Humanos , Insulina/análisis , Masculino , Persona de Mediana Edad , Obesidad/sangre , Obesidad/fisiopatología , Triglicéridos/sangre , Pérdida de Peso
6.
J Am Coll Nutr ; 20(5 Suppl): 428S-435S; discussion 440S-442S, 2001 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-11603653

RESUMEN

Regulation of intracellular calcium plays a key role in hypertension and obesity. Dysregulation of calcium homeostasis appears to be a fundamental factor linking these conditions. Regulation of intracellular calcium in key disease-related target tissues by calcitrophic hormones provides the opportunity to modulate disease risk with dietary calcium. Overall, sub-optimal calcium intakes contribute to the etiology of salt-sensitivity and hypertension. High salt diets exert a calciuretic effect, serving to exacerbate the physiological consequences of sub-optimal calcium diets. Among these are increases in 1,25-dihydroxyvitamin D, which increases vascular smooth muscle intracellular calcium, thereby increasing peripheral vascular resistance and blood pressure. Dietary calcium reduces blood pressure in large part via suppression of 1,25-dihydroxyvitamin D, thereby normalizing intracellular calcium. The practical relevance of this approach has been confirmed in the DASH (Dietary Approaches to Stop Hypertension) trial, which demonstrated that increasing low-fat dairy product and fruit and vegetable consumption exerted profound blood pressure-lowering effects. The magnitude of this effect among hypertensives was comparable to that typically found in pharmacological trials of mild hypertension. 1,25-dihydroxyvitamin D also stimulates calcium influx in human adipocytes, resulting in stimulation of lipogenesis, inhibition of lipolysis and expansion of triglyceride stores. Accordingly, suppression of 1,25-dihydroxyvitamin D by dietary calcium has been identified as a target, which may contribute to the prevention and management of obesity. Indeed, laboratory, clinical and population data all indicate a significant anti-obesity effect of dietary calcium, although large-scale prospective clinical trials have not yet been conducted to definitively demonstrate the scope of this effect. Thus, available evidence indicates that increasing dietary calcium intakes may result in reductions in fat mass as well as in blood pressure.


Asunto(s)
Calcio de la Dieta/administración & dosificación , Calcio/metabolismo , Hipertensión/prevención & control , Obesidad/prevención & control , Vitamina D/análogos & derivados , Vitamina D/metabolismo , Tejido Adiposo/efectos de los fármacos , Presión Sanguínea/efectos de los fármacos , Calcio de la Dieta/uso terapéutico , Homeostasis , Humanos , Hipertensión/tratamiento farmacológico , Obesidad/tratamiento farmacológico , Obesidad/metabolismo , Cloruro de Sodio Dietético/efectos adversos
7.
FASEB J ; 15(13): 2527-9, 2001 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11641262

RESUMEN

We investigated the mechanisms responsible for the anti-lipolytic effect of intracellular Ca2+ ([Ca2+]i) in human adipocytes. Increasing [Ca2+]i inhibited lipolysis induced by b-adrenergic receptor activation, A1 adenosine receptor inhibition, adenylate cyclase activation, and phosphodiesterase (PDE) inhibition, as well as by a hydrolyzable cAMP analog, but not by a nonhydrolyzable cAMP analog. This finding indicates that the anti-lipolytic effect of [Ca2+]i may be mediated by the activation of adipocyte PDE. Consistent with this theory, [Ca2+]i inhibition of isoproterenol-stimulated lipolysis was reversed completely by the nonselective PDE inhibitor isobutyl methylxanthine and also by the selective PDE 3B inhibitor cilostamide, but not by selective PDE 1 and 4 inhibitors. In addition, phosphatidylinositol-3 kinase inhibition with wortmannin completely prevented insulin's anti-lipolytic effect but only minimally blocked [Ca2+]i's effect, which suggests that [Ca2+]i and insulin may activate PDE 3B via different mechanisms. In contrast, the antilipolytic effect of [Ca2+]i was not affected by inhibitors of calmodulin, Ca2+/calmodulin-dependent kinase, protein phosphatase 2B, and protein kinase C. Finally, [Ca2+]i inhibited significantly isoproterenol-stimulated increases in cAMP levels and hormone-sensitive lipase phosphorylation in human adipocytes. In conclusion, increasing [Ca2+]i exerts an antilipolytic effect mainly by activation of PDE, leading to a decrease in cAMP and HSL phosphorylation and, consequently, inhibition of lipolysis.


Asunto(s)
Adipocitos/metabolismo , Calcio/metabolismo , Lipólisis/fisiología , 1-Metil-3-Isobutilxantina/farmacología , 3',5'-AMP Cíclico Fosfodiesterasas/metabolismo , 8-Bromo Monofosfato de Adenosina Cíclica/farmacología , Adipocitos/citología , Adipocitos/efectos de los fármacos , Androstadienos/farmacología , Bucladesina/farmacología , Colforsina/farmacología , AMP Cíclico/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3 , Relación Dosis-Respuesta a Droga , Humanos , Insulina/farmacología , Isoproterenol/farmacología , Lipólisis/efectos de los fármacos , Inhibidores de Fosfodiesterasa/farmacología , Fosforilación/efectos de los fármacos , Cloruro de Potasio/farmacología , Quinolonas/farmacología , Esterol Esterasa/efectos de los fármacos , Esterol Esterasa/metabolismo , Wortmanina , Xantinas/farmacología
8.
FASEB J ; 15(14): 2751-3, 2001 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11606486

RESUMEN

We reported recently that suppression of the renal 1alpha,25-dihyroxyvitamin D3 (1lpha,25-(OH)2-D3) production in aP2-agouti transgenic mice by increasing dietary calcium decreases adipocyte intracellular Ca2+ ([Ca2+]i), stimulates lipolysis, inhibits lipogenesis, and reduces adiposity. However, it was not clear whether this modulation of adipocyte metabolism by dietary calcium is a direct effect of inhibition of 1alpha,25-(OH)2-D3-induced [Ca2+]i. Accordingly, we have now evaluated the direct role of 1alpha,25-(OH)2-D3. Human adipocytes exhibited a 1alpha,25-(OH)2-D3 dose-responsive (1-50 nM) increase in [Ca2+]i (P<0.01). This action was mimicked by 1alpha,25-dihyroxylumisterol3 (1alpha,25-(OH)2-lumisterol3) (P<0.001), a specific agonist for a putative membrane vitamin D receptor (mVDR), and completely prevented by 1b,25-dihydroxyvitamin D3 (1beta,25-(OH)2-D3), a specific antagonist for the mVDR. Similarly, 1alpha,25-(OH)2-D3 (5 nM) caused 50%-100% increases in adipocyte fatty acid synthase (FAS) expression and activity (P<0.02), a 61% increase in glycerol-3-phosphate dehydrogenase (GPDH) activity (P<0.01), and an 80% inhibition of isoproterenol-stimulated lipolysis (P<0.001), whereas 1beta,25-(OH)2-D3 completely blocked all these effects. Notably, 1alpha,25-(OH)2-lumisterol3 exerted more potent effects in modulating adipocyte lipid metabolism, with 2.5- to 3.0-fold increases in FAS expression and activity (P<0.001) and a threefold increase in GPDH activity (P<0.001). Also 1alpha,25-(OH)2-lumisterol3 was approximately twice as potent in inhibiting basal lipolysis (P<0.025), whereas 1beta,25-(OH)2-D3 completely blocked all these effects. These data suggest that 1alpha,25-(OH)2-D3 modulates adipocyte Ca2+ signaling and, consequently, exerts a coordinated control over lipogenesis and lipolysis. Thus, a direct inhibition of 1alpha,25-(OH)2-D3-induced [Ca2+]i may contribute to an anti-obesity effect of dietary calcium, and the mVDR may represent an important target for obesity.


Asunto(s)
Adipocitos/efectos de los fármacos , Calcitriol/farmacología , Adipocitos/metabolismo , Calcio/metabolismo , Dihidroxicolecalciferoles/farmacología , Relación Dosis-Respuesta a Droga , Ácido Graso Sintasas/efectos de los fármacos , Ácido Graso Sintasas/genética , Ácido Graso Sintasas/metabolismo , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Glicerolfosfato Deshidrogenasa/efectos de los fármacos , Glicerolfosfato Deshidrogenasa/metabolismo , Humanos , Isoproterenol/farmacología , Lípidos/biosíntesis , Lipólisis/efectos de los fármacos , ARN Mensajero/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo
9.
Exp Biol Med (Maywood) ; 226(6): 565-9, 2001 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-11395927

RESUMEN

Ectopic overexpression of the murine agouti gene results in yellow coat color, obesity, hyperinsulinemia, and type II diabetes. We have shown the human homologue of agouti (agouti signaling protein; ASP) to regulate human adipocyte metabolism and lipid storage via a Ca(2+)-dependent mechanism. We have also demonstrated agouti expression in human pancreas, and that ASP stimulates insulin release via a similar Ca(2+)-dependent mechanism. Plasma amylin is also elevated in agouti mutant mice. Amylin is cosecreted with insulin from beta-cells, and overexpression of human amylin in beta-cells in yellow agouti mutant mice resulted in accelerated pancreatic amyloid deposition, severely impaired beta-cell function, and a diabetic phenotype. We report here that ASP stimulates amylin release in both the HIT-T15 beta-cell line and human pancreatic islets in the presence of a wide range of glucose concentrations (0-16.7 mmol/L), similar to its effect on insulin release; this effect was blocked by 30 mumol/L nitrendipine, confirming a Ca(2+)-dependent mechanism. Accordingly, ASP stimulation of amylin release may serve as a compensatory system to regulate blood glucose in yellow agouti mutants.


Asunto(s)
Amiloide/metabolismo , Péptidos y Proteínas de Señalización Intercelular , Islotes Pancreáticos/metabolismo , Proteínas/metabolismo , Proteína de Señalización Agouti , Calcio/metabolismo , Línea Celular , Humanos , Polipéptido Amiloide de los Islotes Pancreáticos , Islotes Pancreáticos/efectos de los fármacos , Proteínas/farmacología , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes de Fusión/farmacología , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología
10.
FASEB J ; 15(2): 291-3, 2001 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11156940

RESUMEN

We have demonstrated previously a regulatory role for intracellular Ca2+ ([Ca2+]i) in adipocyte lipogenesis and lipolysis and have recently reported that 1,25-(OH)2-D increases adipocyte [Ca2+]i, which causes increased lipogenesis and decreased lipolysis. We have now tested the hypothesis that suppressing 1,25-(OH)2-D by increasing dietary calcium will suppress adipocyte [Ca2+]i, thereby facilitating weight loss by stimulating lipolysis and inhibiting lipogenesis in calorically (Kcal)-restricted (70% of ad lib) aP2-agouti transgenic (aP2-a) mice. Mice (aP2-a) exhibiting a pattern of obesity gene expression similar to humans were fed a low-Ca (0.4%)/high-fat/high-sucrose diet for six weeks, resulting in a 27% and twofold increase in body weight and total fat pad mass, respectively, with a twofold increase in adipocyte [Ca2+]i pad lib or Kcal-restricted (70% of ad lib) on this diet either unsupplemented (basal) or with 25% or 50% of the protein replaced by non-fat dry milk (medium or high) dairy or supplemented with CaCO3 to 1.2% Ca for six weeks. Adipocyte [Ca2+]i was unaffected by Kcal restriction but was reduced markedly by all three high Ca diets (290 vs. 130 nM, p2+]i and thereby reduce energy storage and increase thermogenesis during Kcal restriction.


Asunto(s)
Adipocitos/metabolismo , Tejido Adiposo/fisiología , Peso Corporal , Calcio de la Dieta/farmacología , Metabolismo Energético , Metabolismo de los Lípidos , Proteínas de Transporte de Membrana , Proteínas Mitocondriales , Obesidad/fisiopatología , Adipocitos/efectos de los fármacos , Tejido Adiposo/anatomía & histología , Tejido Adiposo/citología , Animales , Regulación de la Temperatura Corporal , Peso Corporal/genética , Calcitriol/farmacología , Dieta Reductora , Metabolismo Energético/genética , Canales Iónicos , Lipólisis , Ratones , Ratones Endogámicos , Ratones Transgénicos , Modelos Biológicos , Obesidad/genética , Proteínas/genética , Proteínas/metabolismo , Proteína Desacopladora 2
11.
J Nutr ; 130(10): 2478-81, 2000 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-11015476

RESUMEN

The human homologue of the murine obesity gene, agouti, is expressed in adipose tissue. We have shown that recombinant agouti protein regulates adipocyte lipogenesis and lipolysis coordinately and promotes lipid storage via a Ca(2+)-dependent mechanism in vitro, which may contribute to agouti-induced obesity. However, little is known about agouti's physiologic function in humans. We first studied the agouti content in human mature adipocytes vs. preadipocytes. The agouti content of human mature adipocytes was five times as abundant as in preadipocytes (19.18 +/- 2.46 vs. 4.07 +/- 0.51 pg/microg protein, P: < 0.005), suggesting that agouti is up-regulated during adipocyte differentiation. We next studied the relationship of agouti mRNA and protein to fatty acid synthase (FAS) mRNA and activity in adipose tissue obtained from nonobese and mildly obese patients (body mass index range, 21-31 kg/m(2)). Agouti protein was correlated with FAS activity (r = 0.782, P: < 0.005). Similarly, human adipose tissue agouti mRNA level was also correlated with FAS mRNA level (r = 0.846, P: < 0.001). These data suggest that agouti may be another adipocyte-produced factor that modulates adipocyte lipid metabolism via a paracrine/autocrine mechanism.


Asunto(s)
Adipocitos/metabolismo , Ácido Graso Sintasas/metabolismo , Péptidos y Proteínas de Señalización Intercelular , Obesidad/metabolismo , Proteínas/metabolismo , Adipocitos/citología , Tejido Adiposo/metabolismo , Proteína de Señalización Agouti , Diferenciación Celular , Ácido Graso Sintasas/genética , Expresión Génica , Humanos , Proteínas/genética , ARN Mensajero/metabolismo
12.
Physiol Genomics ; 2(3): 101-5, 2000 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-11015588

RESUMEN

Dominant mutations at the mouse Agouti locus lead to ectopic expression of the Agouti gene and exhibit diabetes, obesity, and yellow coat color. Obese yellow mice are hyperinsulinemic and hyperleptinemic, and we hypothesized that Agouti directly induces leptin secretion. Accordingly, we used transgenic mice expressing agouti in adipocytes (under the control of aP2 promoter, aP212) to examine changes in leptin levels. Agouti expression in adipose tissue did not significantly alter food intake, weight gain, fat pad weight, or insulinemia; however, the transgenic mice were hyperglycemic. We demonstrated that plasma leptin levels are approximately twofold higher in aP212 transgenic mice compared with their respective controls, whereas ubiquitous expression of agouti (under the control of beta-actin promoter, BAP20) led to a sixfold increase in leptin. Insulin treatment of aP212 mice increased adipocyte leptin content without affecting plasma leptin levels. These findings were further confirmed in vitro in 3T3-L1 adipocytes treated with recombinant Agouti protein and/or insulin. Agouti but not insulin significantly increased leptin secretion, indicating that insulin enhances leptin synthesis but not secretion while Agouti increases both leptin synthesis and secretion. This increased leptin synthesis and secretion was due to increased leptin mRNA levels by Agouti. Interestingly, agouti regulation of leptin was not mediated by melanocortin receptor 4, previously implicated in agouti regulation of food intake. These results suggest that increased leptin secretion by agouti may serve to limit agouti-induced obesity, independent of melanocortin receptor antagonism, and indicate that interaction between obesity genes may play a key role in obesity.


Asunto(s)
Adipocitos/metabolismo , Péptidos y Proteínas de Señalización Intercelular , Leptina/sangre , Proteínas de Neoplasias , Proteínas del Tejido Nervioso , Proteínas/metabolismo , Adipocitos/citología , Adipocitos/efectos de los fármacos , Tejido Adiposo/química , Tejido Adiposo/citología , Tejido Adiposo/efectos de los fármacos , Proteína de Señalización Agouti , Animales , Proteínas Portadoras , Células Cultivadas , Proteínas de Unión al ADN , Diabetes Mellitus/genética , Esquema de Medicación , Proteína de Unión a los Ácidos Grasos 7 , Proteínas de Unión a Ácidos Grasos , Homocigoto , Inyecciones Subcutáneas , Insulina/administración & dosificación , Leptina/análisis , Leptina/genética , Masculino , Hormonas Estimuladoras de los Melanocitos/farmacología , Ratones , Ratones Transgénicos , Obesidad , Regiones Promotoras Genéticas/genética , Proteínas/genética , Proteínas/farmacología , ARN Mensajero/metabolismo , Receptor de Melanocortina Tipo 4 , Receptores de Corticotropina/agonistas , Receptores de Leptina , Receptores de Melanocortina , Receptores de Péptidos/antagonistas & inhibidores
13.
Physiol Genomics ; 3(2): 75-82, 2000 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-11015602

RESUMEN

Intracellular calcium ([Ca(2+)](i)) modulates adipocyte lipid metabolism and inhibits the early stages of murine adipogenesis. Consequently, we evaluated effects of increasing [Ca(2+)](i) in early and late stages of human adipocyte differentiation. Increasing [Ca(2+)](i) with either thapsigargin or A23187 at 0-1 h of differentiation markedly suppressed differentiation, with a 40-70% decrease in triglyceride accumulation and glycerol-3 phosphate dehydrogenase (GPDH) activity (P < 0.005). However, a 1-h pulse of either agent at 47-48 h only modestly inhibited differentiation. Sustained, mild stimulation of Ca(2+) influx with either agouti protein or 10 mM KCl-induced depolarization during 0-48 h of differentiation inhibited triglyceride accumulation and GPDH activity by 20-70% (P < 0.05) and markedly suppressed peroxisome proliferator-activated receptor gamma (PPARgamma) expression. These effects were reversed by Ca(2+) channel antagonism. In contrast, Ca(2+) pulses late in differentiation (71-72 h or 48-72 h) markedly increased these markers of differentiation. Thus increasing [Ca(2+)](i) appears to exert a biphasic regulatory role in human adipocyte differentiation, inhibiting the early stages while promoting the late stage of differentiation and lipid filling.


Asunto(s)
Adipocitos/citología , Adipocitos/metabolismo , Señalización del Calcio , Calcio/metabolismo , Diferenciación Celular , Péptidos y Proteínas de Señalización Intercelular , Adipocitos/efectos de los fármacos , Adipocitos/enzimología , Proteína de Señalización Agouti , Calcimicina/farmacología , Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio/metabolismo , Señalización del Calcio/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Glicerolfosfato Deshidrogenasa/metabolismo , Humanos , Ionóforos/farmacología , Nitrendipino/farmacología , Cloruro de Potasio/farmacología , Proteínas/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores Citoplasmáticos y Nucleares/genética , Células Madre/citología , Células Madre/efectos de los fármacos , Células Madre/enzimología , Células Madre/metabolismo , Tapsigargina/farmacología , Factores de Tiempo , Factores de Transcripción/genética , Triglicéridos/metabolismo
14.
Physiol Genomics ; 3(3): 157-62, 2000 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-11015611

RESUMEN

Mice carrying dominant mutations at the agouti locus exhibit ectopic expression of agouti gene transcripts, obesity, and type II diabetes through unknown mechanisms. To gain insight into the role of agouti protein in modulating adiposity, we investigated regulation of a key lipogenic gene, fatty acid synthase (FAS) by agouti alone and in combination with insulin. Both agouti and insulin increase FAS activity in 3T3-L1 and in human adipocytes. Agouti and insulin independently and additively increase FAS activity in 3T3-L1 adipocytes. We further investigated the mechanism responsible for the agouti-induced FAS expression in these cells and demonstrated that both insulin (3-fold increase) and agouti (2-fold) increased FAS gene expression at the transcriptional level. Furthermore, insulin and agouti together exerted additive effects (5-fold increase) on FAS gene transcription. Transfection assays of FAS promoter-luciferase fusion gene constructs into 3T3-L1 adipocytes indicated that the agouti response element(s) is (are) located in the -435 to -415 region (-435/-415) of the FAS promoter. Nuclear proteins binding to this novel sequence are adipocyte specific. Thus the agouti response sequences mapped to a region upstream of the insulin-responsive element (which we previously reported to be located at -67/-52), consistent with additive effects of these two factors on FAS gene transcription.


Asunto(s)
Adipocitos/enzimología , Ácido Graso Sintasas/genética , Péptidos y Proteínas de Señalización Intercelular , Proteínas/farmacología , Células 3T3 , Adipocitos/citología , Proteína de Señalización Agouti , Animales , ADN/genética , Interacciones Farmacológicas , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Humanos , Insulina/farmacología , Luciferasas/genética , Luciferasas/metabolismo , Ratones , Proteínas Nucleares/metabolismo , Oligonucleótidos/genética , Oligonucleótidos/metabolismo , Regiones Promotoras Genéticas/genética , Unión Proteica , Proteínas/metabolismo , ARN Mensajero/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Elementos de Respuesta/genética , Transcripción Genética/efectos de los fármacos , Células Tumorales Cultivadas
15.
Nutr Rev ; 58(6): 177-80, 2000 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10885325

RESUMEN

Melanocortin peptides, derived from pro-opiomelanocortin (POMC), appear to play a significant role in appetite and body weight regulation. Expression of the Pomc gene in the central nervous system results in the production of melanocortin peptides, which bind to the melanocortin-4 receptor (MC4-R) and inhibit food intake. MC4-R knockout mice exhibit adult-onset obesity, whereas MC4-R agonists suppress food intake in several models of obesity. Recently, Pomc knockout mice were generated and shown to develop hyperphagia and obesity with a time-course and severity comparable to MC4-R knockout mice, whereas daily administration of a stable alpha-melanocyte stimulating hormone analogue reversed this effect. These data clearly implicate POMC peptides and melanocortin receptors in the pathophysiology of obesity and provide important new tools for their development as therapeutic targets in obesity.


Asunto(s)
Obesidad/genética , Proopiomelanocortina/deficiencia , Receptores de Corticotropina/fisiología , Animales , Ingestión de Alimentos/fisiología , Humanos , Ratones , Ratones Noqueados , Proopiomelanocortina/genética , Receptor de Melanocortina Tipo 4 , Receptores de Corticotropina/genética , Receptores de Melanocortina , alfa-MSH/farmacología
16.
FASEB J ; 14(9): 1132-8, 2000 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10834935

RESUMEN

Recent data from this laboratory demonstrate that increasing adipocyte intracellular Ca(2+) results in a coordinated stimulation of lipogenesis and inhibition of lipolysis. We have also noted that increasing dietary calcium of obese patients for 1 year resulted in a 4.9 kg loss of body fat (P<0.01). Accordingly, we tested the possibility that calcitrophic hormones may act on adipocytes to increase Ca(2+) and lipid metabolism by measuring the effects of 1, 25-(OH)(2)-D in primary cultures of human adipocytes, and found significant, sustained increases in intracellular Ca(2+) and a corresponding marked inhibition of lipolysis (EC(50) approximately 50 pM; P<0.001), suggesting that dietary calcium could reduce adipocyte mass by suppressing 1,25-(OH)(2)-D. To test this hypothesis, we placed transgenic mice expressing the agouti gene specifically in adipocytes on a low (0.4%) Ca/high fat/high sucrose diet either unsupplemented or with 25 or 50% of the protein replaced by non-fat dry milk or supplemented to 1.2% Ca with CaCO(3) for 6 wk. Weight gain and fat pad mass were reduced by 26-39% by the three high calcium diets (P<0.001). The high calcium diets exerted a corresponding 51% inhibition of adipocyte fatty acid synthase expression and activity (P<0.002) and stimulation of lipolysis by 3. 4- to 5.2-fold (P<0.015). This concept of calcium modulation of adiposity was further evaluated epidemiologically in the NHANES III data set. After controlling for energy intake, relative risk of being in the highest quartile of body fat was set to 1.00 for the lowest quartile of Ca intake and was reduced to 0.75, 0.40, and 0.16 for the second, third, and fourth quartiles, respectively, of calcium intake for women (n=380;P<0.0009); a similar inverse relationship was also noted in men (n=7114; P<0.0006). Thus, increasing dietary calcium suppresses adipocyte intracellular Ca(2+) and thereby modulates energy metabolism and attenuates obesity risk.


Asunto(s)
Adipocitos/efectos de los fármacos , Calcio de la Dieta/farmacología , Calcio/metabolismo , Péptidos y Proteínas de Señalización Intercelular , Lipólisis/efectos de los fármacos , Adipocitos/citología , Adipocitos/enzimología , Adipocitos/metabolismo , Adulto , Proteína de Señalización Agouti , Animales , Glucemia/análisis , Temperatura Corporal/efectos de los fármacos , Calcio de la Dieta/administración & dosificación , Calcio de la Dieta/uso terapéutico , Células Cultivadas , Productos Lácteos , Ingestión de Energía , Metabolismo Energético/efectos de los fármacos , Ácido Graso Sintasas/genética , Ácido Graso Sintasas/metabolismo , Femenino , Humanos , Insulina/sangre , Masculino , Ratones , Ratones Transgénicos , Obesidad/sangre , Obesidad/dietoterapia , Obesidad/metabolismo , Hormona Paratiroidea/antagonistas & inhibidores , Hormona Paratiroidea/farmacología , Proteínas/genética , Proteínas/metabolismo , Vitamina D/antagonistas & inhibidores , Vitamina D/farmacología , Aumento de Peso/efectos de los fármacos
17.
Nutr Rev ; 57(9 Pt 1): 273-6, 1999 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-10568336

RESUMEN

Although Americans have decreased the percent of energy they consume from fat, obesity and obesity-related comorbidities have progressively increased. Less attention has been paid to the role of carbohydrates, especially carbohydrate source, in these metabolic diseases. However, recent epidemiologic studies demonstrate consistently higher rates of cardiovascular disease and type II diabetes in individuals deriving a greater percentage of energy from refined grains and simple carbohydrates than from whole grains. Differences in the metabolic response to carbohydrates can be classified by glycemic index (GI), the blood glucose response to a given food compared with a standard (typically white bread or glucose). Classification of carbohydrates as "simple" or "complex" is of little use in predicting GI, because GI is influenced by starch structure (amylose versus amylopectin), fiber content, food processing, physical structure of the food, and other macronutrients in the meal. Low-GI diets have been reported to lower postprandial glucose and insulin responses, improve lipid profiles, and increase insulin sensitivity. Moreover, high-GI diets stimulate de novo lipogenesis and result in increased adipocyte size, whereas low-GI diets have been reported to inhibit these responses. Thus, the GI of dietary carbohydrates appears to play an important role in the metabolic fate of carbohydrates and, consequently, may significantly affect the risk of cardiovascular disease, diabetes, and obesity.


Asunto(s)
Glucemia/metabolismo , Enfermedades Cardiovasculares/inducido químicamente , Carbohidratos de la Dieta/metabolismo , Obesidad/inducido químicamente , Comorbilidad , Carbohidratos de la Dieta/efectos adversos , Carbohidratos de la Dieta/clasificación , Humanos , Insulina/metabolismo , Encuestas Nutricionales , Obesidad/metabolismo , Periodo Posprandial
18.
FASEB J ; 13(13): 1833-8, 1999 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-10506587

RESUMEN

A regulatory role for intracellular Ca2+ ([Ca2+]i) in adipocyte lipogenesis, lipolysis and triglyceride accumulation has been demonstrated. Compounds acting on the pancreatic sulfonylurea receptor (SUR) to increase (e.g., glibenclamide) or decrease (e.g., diazoxide) [Ca2+]i cause corresponding increases and decreases in weight gain. However, these weight gain and loss effects have been attributed to insulin release rather than to the primary effects of these compounds on the adipocyte SUR and its associated K(ATP) channel. Accordingly, we have evaluated the direct role of the human adipocyte SUR in regulating adipocyte metabolism. We used RT-PCR with primers for a highly conserved region of SUR1 to demonstrate that human adipocytes express SUR1. The PCR product was confirmed by sequence analysis and used as a probe to demonstrate adipocyte SUR1 expression by Northern blot analysis. Adipocytes exhibited glibenclamide dose-responsive (0-20 microM) increases in [Ca2+]i (P<0.05). Similarly, glibenclamide (10 microM) caused a 67% increase in adipocyte fatty acid synthase activity (P<0.001), a 48% increase in glycerol-3-phosphate dehydrogenase activity (P<0.01) and a 68% inhibition in lipolysis (P<0.01), whereas diazoxide (10 microM) completely prevented each of these effects. These data demonstrate that human adipocytes express a SUR that regulates [Ca2+]i and, consequently, exerts coordinate control over lipogenesis and lipolysis. Accordingly, the adipocyte SUR1 may represent an important target for the development of therapeutic interventions in obesity.


Asunto(s)
Transportadoras de Casetes de Unión a ATP , Adipocitos/metabolismo , Calcio/metabolismo , Metabolismo de los Lípidos , Canales de Potasio de Rectificación Interna , Canales de Potasio/metabolismo , Receptores de Droga/metabolismo , Tejido Adiposo/citología , Tejido Adiposo/metabolismo , Diazóxido/farmacología , Relación Dosis-Respuesta a Droga , Ácido Graso Sintasas/efectos de los fármacos , Gliburida/farmacología , Glicerolfosfato Deshidrogenasa/efectos de los fármacos , Humanos , Lipólisis/efectos de los fármacos , Receptores de Sulfonilureas
19.
Physiol Genomics ; 1(1): 11-9, 1999 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-11015556

RESUMEN

Ubiquitous expression of the mouse agouti gene results in obesity and hyperinsulinemia. Human agouti is expressed in adipose tissue, and we found recombinant agouti protein to stimulate lipogenesis in adipocytes in a Ca(2+)-dependent fashion. However, adipocyte-specific agouti transgenic mice only became obese in the presence of hyperinsulinemia. Because intracellular Ca(2+) concentration ([Ca(2+)](i)) is a primary signal for insulin release, and we have shown agouti protein to increase [Ca(2+)](i) in several cell types, we examined the effects of agouti on [Ca(2+)](i) and insulin release. We demonstrated the expression of agouti in human pancreas and generated recombinant agouti to study its effects on Ca(2+) signaling and insulin release. Agouti (100 nM) stimulated Ca(2+) influx, [Ca(2+)](i) increase, and a marked stimulation of insulin release in two beta-cell lines (RIN-5F and HIT-T15; P < 0. 05). Agouti exerted comparable effects in isolated human pancreatic islets and beta-cells, with a 5-fold increase in Ca(2+) influx (P < 0.001) and a 2.2-fold increase in insulin release (P < 0.01). These data suggest a potential role for agouti in the development of hyperinsulinemia in humans.


Asunto(s)
Señalización del Calcio/fisiología , Fura-2/análogos & derivados , Expresión Génica , Hiperinsulinismo/fisiopatología , Insulina/metabolismo , Péptidos y Proteínas de Señalización Intercelular , Islotes Pancreáticos/metabolismo , Proteínas/metabolismo , Proteína de Señalización Agouti , Animales , Calcio/metabolismo , Señalización del Calcio/efectos de los fármacos , Células Cultivadas , Cricetinae , Relación Dosis-Respuesta a Droga , Glucosa/metabolismo , Glucosa/farmacología , Humanos , Hiperinsulinismo/genética , Secreción de Insulina , Islotes Pancreáticos/citología , Islotes Pancreáticos/efectos de los fármacos , Manganeso/farmacología , Comunicación Paracrina/efectos de los fármacos , Cloruro de Potasio/metabolismo , Cloruro de Potasio/farmacología , Proteínas/genética , Proteínas/farmacología , ARN Mensajero/metabolismo , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
20.
Mol Cell Biochem ; 188(1-2): 129-36, 1998 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-9823018

RESUMEN

Regulation of intracellular Ca2+ ([Ca2+]i) plays a key role in obesity, insulin resistance and hypertension, and [Ca2+]i disorders may represent a fundamental factor linking these three conditions. We have shown insulin to be a direct vasodilator, attenuating voltage-gated Ca2+ influx and stimulating Ca(2+)-ATPase transcription via a glucose-6-phosphate response element. These result in a net decrease in [Ca2+]i and thereby decrease vascular resistance, while these effects are blunted in insulin resistance, leading to increased vascular resistance. Consistent with this concept, pharmacological amplification of peripheral insulin sensitivity results in reduced arterial pressure. While insulin regulates [Ca2+]i, Ca2+ also regulates insulin signaling, as increasing [Ca2+]i impairs insulin signaling in some systems, possibly due to Ca2+ inhibition of insulin-regulated dephosphorylation. Finally, in recent studies of the mouse agouti gene, we have also demonstrated increased [Ca2+]i to play a key role in adipocyte lipogenesis, as follows. We have found dominant agouti mutants to exhibit increased [Ca2+]i in most tissues, leading to increased vascular reactivity and insulin resistance in vascular smooth muscle and skeletal muscle cells, respectively. Further, we have found recombinant agouti protein to directly increase [Ca2+]i in a variety of cells, including murine and human adipocytes, and to stimulate both the expression and activity of adipocyte fatty acid synthase and increase triglyceride accumulation in a Ca(2+)-dependent manner. These effects can be mimicked by stimulation of Ca2+ influx and blocked by Ca2+ channel inhibition, while treatment of mice with a Ca2+ antagonist attenuates agouti-induced obesity. Since humans express agouti in adipose tissue, it may similarly exert paracrine effects on [Ca2+]i and thereby stimulate de novo lipogenesis and promote obesity. Thus, Ca2+ signaling represents a target for therapeutic intervention in obesity as well as hypertension and insulin resistance.


Asunto(s)
Calcio/metabolismo , Glándulas Endocrinas/metabolismo , Hipertensión/metabolismo , Resistencia a la Insulina/fisiología , Líquido Intracelular/metabolismo , Obesidad/metabolismo , Animales , Calcio/fisiología , Calcio de la Dieta/metabolismo , Glándulas Endocrinas/fisiología , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...