Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
J Biol Chem ; 289(28): 19341-50, 2014 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-24872412

RESUMEN

The chemical uncoupler 2,4-dinitrophenol (DNP) was an effective and widely used weight loss drug in the early 1930s. However, the physiology of DNP has not been studied in detail because toxicity, including hyperthermia and death, reduced interest in the clinical use of chemical uncouplers. To investigate DNP action, mice fed a high fat diet and housed at 30 °C (to minimize facultative thermogenesis) were treated with 800 mg/liter DNP in drinking water. DNP treatment increased energy expenditure by ∼ 17%, but did not change food intake. DNP-treated mice weighed 26% less than controls after 2 months of treatment due to decreased fat mass, without a change in lean mass. DNP improved glucose tolerance and reduced hepatic steatosis without observed toxicity. DNP treatment also reduced circulating T3 and T4 levels, Ucp1 expression, and brown adipose tissue activity, demonstrating that DNP-mediated heat generation substituted for brown adipose tissue thermogenesis. At 22 °C, a typical vivarium temperature that is below thermoneutrality, DNP treatment had no effect on body weight, adiposity, or glucose homeostasis. Thus, environmental temperature should be considered when assessing an anti-obesity drug in mice, particularly agents acting on energy expenditure. Furthermore, the beneficial effects of DNP suggest that chemical uncouplers deserve further investigation for the treatment of obesity and its comorbidities.


Asunto(s)
2,4-Dinitrofenol/farmacología , Adiposidad/efectos de los fármacos , Regulación de la Temperatura Corporal/efectos de los fármacos , Dieta/efectos adversos , Metabolismo Energético/efectos de los fármacos , Obesidad/tratamiento farmacológico , Desacopladores/farmacología , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Pardo/patología , Animales , Ingestión de Alimentos/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Canales Iónicos/biosíntesis , Ratones , Proteínas Mitocondriales/biosíntesis , Obesidad/inducido químicamente , Obesidad/metabolismo , Obesidad/patología , Proteína Desacopladora 1
2.
FASEB J ; 26(10): 4275-86, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22730439

RESUMEN

Spinophilin (SPL), a multidomain scaffolding protein known to modulate the activity of different G-protein-coupled receptors, regulates various central nervous system (CNS) functions. However, little is known about the role of SPL expressed in peripheral cell types including pancreatic ß cells. In this study, we examined the ability of SPL to modulate the activity of ß-cell M(3) muscarinic acetylcholine receptors (M3Rs), which play an important role in facilitating insulin release and maintaining normal blood glucose levels. We demonstrated, by using both in vitro and in vivo approaches (mouse insulinoma cells and SPL-deficient mice), that SPL is a potent negative regulator of M3R-mediated signaling and insulin release. Additional biochemical and biophysical studies, including the use of bioluminescence resonance energy transfer technology, suggested that SPL is able to recruit regulator of G-protein signaling 4 (RGS4) to the M3R signaling complex in an agonist-dependent fashion. Since RGS4 is a member of the RGS family of proteins that act to reduce the lifetime of activated G proteins, these findings support the concept that the inhibitory effects of SPL on M3R activity are mediated by RGS4. These data suggest that SPL or other G-protein-coupled receptor-associated proteins may serve as novel targets for drug therapy aimed at improving ß-cell function for the treatment of type 2 diabetes.


Asunto(s)
Resistencia a la Insulina/fisiología , Proteínas de Microfilamentos/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Receptor Muscarínico M3/metabolismo , Animales , Western Blotting , Células COS , Línea Celular Tumoral , Chlorocebus aethiops , Resistencia a la Insulina/genética , Ratones , Ratones Noqueados , Proteínas de Microfilamentos/genética , Proteínas del Tejido Nervioso/genética , Receptores Acoplados a Proteínas G , Transducción de Señal/genética , Transducción de Señal/fisiología
3.
Proc Natl Acad Sci U S A ; 107(17): 7999-8004, 2010 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-20385802

RESUMEN

Therapeutic strategies that augment insulin release from pancreatic beta-cells are considered beneficial in the treatment of type 2 diabetes. We previously demonstrated that activation of beta-cell M(3) muscarinic receptors (M3Rs) greatly promotes glucose-stimulated insulin secretion (GSIS), suggesting that strategies aimed at enhancing signaling through beta-cell M3Rs may become therapeutically useful. M3R activation leads to the stimulation of G proteins of the G(q) family, which are under the inhibitory control of proteins known as regulators of G protein signaling (RGS proteins). At present, it remains unknown whether RGS proteins play a role in regulating insulin release. To address this issue, we initially demonstrated that MIN6 insulinoma cells express functional M3Rs and that RGS4 was by far the most abundant RGS protein expressed by these cells. Strikingly, siRNA-mediated knockdown of RGS4 expression in MIN6 cells greatly enhanced M3R-mediated augmentation of GSIS and calcium release. We obtained similar findings using pancreatic islets prepared from RGS4-deficient mice. Interestingly, RGS4 deficiency had little effect on insulin release caused by activation of other beta-cell GPCRs. Finally, treatment of mutant mice selectively lacking RGS4 in pancreatic beta-cells with a muscarinic agonist (bethanechol) led to significantly increased plasma insulin and reduced blood glucose levels, as compared to control littermates. Studies with beta-cell-specific M3R knockout mice showed that these responses were mediated by beta-cell M3Rs. These findings indicate that RGS4 is a potent negative regulator of M3R function in pancreatic beta-cells, suggesting that RGS4 may represent a potential target to promote insulin release for therapeutic purposes.


Asunto(s)
Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Proteínas RGS/metabolismo , Receptor Muscarínico M3/metabolismo , Análisis de Varianza , Animales , Betanecol , Glucemia/metabolismo , Western Blotting , Células COS , Calcio/metabolismo , Línea Celular Tumoral , Chlorocebus aethiops , Electroporación , Inmunoprecipitación , Secreción de Insulina , Ratones , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
4.
Cell Metab ; 4(5): 363-75, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17084710

RESUMEN

Most animal models of obesity and hyperinsulinemia are associated with increased vagal cholinergic activity. The M3 muscarinic acetylcholine receptor subtype is widely expressed in the brain and peripheral tissues and plays a key role in mediating the physiological effects of vagal activation. Here, we tested the hypothesis that the absence of M3 receptors in mice might protect against various forms of experimentally or genetically induced obesity and obesity-associated metabolic deficits. In all cases, the lack of M3 receptors greatly ameliorated impairments in glucose homeostasis and insulin sensitivity but had less robust effects on overall adiposity. Under all experimental conditions tested, M3 receptor-deficient mice showed a significant elevation in basal and total energy expenditure, most likely due to enhanced central sympathetic outflow and increased rate of fatty-acid oxidation. These findings suggest that the M3 receptor may represent a potential pharmacologic target for the treatment of obesity and associated metabolic disorders.


Asunto(s)
Cuerpos Aórticos/metabolismo , Enfermedades Metabólicas/etiología , Obesidad , Receptor Muscarínico M3/genética , Receptor Muscarínico M3/fisiología , Animales , Epinefrina/orina , Canales Iónicos/metabolismo , Ratones , Ratones Noqueados , Proteínas Mitocondriales/metabolismo , Norepinefrina/orina , Receptor Muscarínico M3/deficiencia , Proteína Desacopladora 3
5.
J Biol Chem ; 285(47): 36776-84, 2010 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-20847051

RESUMEN

Glucose homeostasis in mammals is mainly regulated by insulin signaling. It was previously shown that SIRT6 mutant mice die before 4 weeks of age, displaying profound abnormalities, including low insulin, hypoglycemia, and premature aging. To investigate mechanisms underlying the pleiotropic phenotypes associated with SIRT6 deficiency, we generated mice carrying targeted disruption of SIRT6. We found that 60% of SIRT6(-/-) animals had very low levels of blood glucose and died shortly after weaning. The remaining animals, which have relatively higher concentrations of glucose, survived the early post-weaning lethality, but most died within one year of age. Significantly, feeding the mice with glucose-containing water increased blood glucose and rescued 83% of mutant mice, suggesting that the hypoglycemia is a major cause for the lethality. We showed that SIRT6 deficiency results in more abundant membrane association of glucose transporters 1 and 4, which enhances glucose uptake. We further demonstrated that SIRT6 negatively regulates AKT phosphorylation at Ser-473 and Thr-308 through inhibition of multiple upstream molecules, including insulin receptor, IRS1, and IRS2. The absence of SIRT6, consequently, enhances insulin signaling and activation of AKT, leading to hypoglycemia. These data uncover an essential role of SIRT6 in modulating glucose metabolism through mediating insulin sensitivity.


Asunto(s)
Glucosa/farmacocinética , Hipoglucemia/etiología , Insulina/farmacología , Sirtuinas/fisiología , Animales , Western Blotting , Diferenciación Celular , Células Cultivadas , Femenino , Técnica del Anticuerpo Fluorescente , Genes Letales , Transportador de Glucosa de Tipo 1/genética , Transportador de Glucosa de Tipo 1/metabolismo , Transportador de Glucosa de Tipo 2/genética , Transportador de Glucosa de Tipo 2/metabolismo , Hipoglucemia/metabolismo , Hipoglucemia/patología , Técnicas para Inmunoenzimas , Resistencia a la Insulina , Masculino , Ratones , Ratones Noqueados , Fibras Musculares Esqueléticas/citología , Fibras Musculares Esqueléticas/metabolismo , Fenotipo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Distribución Tisular
6.
PLoS Comput Biol ; 5(3): e1000324, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19325873

RESUMEN

Adipose tissue grows by two mechanisms: hyperplasia (cell number increase) and hypertrophy (cell size increase). Genetics and diet affect the relative contributions of these two mechanisms to the growth of adipose tissue in obesity. In this study, the size distributions of epididymal adipose cells from two mouse strains, obesity-resistant FVB/N and obesity-prone C57BL/6, were measured after 2, 4, and 12 weeks under regular and high-fat feeding conditions. The total cell number in the epididymal fat pad was estimated from the fat pad mass and the normalized cell-size distribution. The cell number and volume-weighted mean cell size increase as a function of fat pad mass. To address adipose tissue growth precisely, we developed a mathematical model describing the evolution of the adipose cell-size distributions as a function of the increasing fat pad mass, instead of the increasing chronological time. Our model describes the recruitment of new adipose cells and their subsequent development in different strains, and with different diet regimens, with common mechanisms, but with diet- and genetics-dependent model parameters. Compared to the FVB/N strain, the C57BL/6 strain has greater recruitment of small adipose cells. Hyperplasia is enhanced by high-fat diet in a strain-dependent way, suggesting a synergistic interaction between genetics and diet. Moreover, high-fat feeding increases the rate of adipose cell size growth, independent of strain, reflecting the increase in calories requiring storage. Additionally, high-fat diet leads to a dramatic spreading of the size distribution of adipose cells in both strains; this implies an increase in size fluctuations of adipose cells through lipid turnover.


Asunto(s)
Adipocitos/patología , Tejido Adiposo/crecimiento & desarrollo , Tejido Adiposo/patología , Grasas de la Dieta/metabolismo , Modelos Biológicos , Obesidad/patología , Obesidad/fisiopatología , Animales , Aumento de la Célula , Proliferación Celular , Tamaño de la Célula , Simulación por Computador , Hiperplasia/patología , Hiperplasia/fisiopatología , Hipertrofia/patología , Hipertrofia/fisiopatología , Ratones , Ratones Endogámicos C57BL
7.
Virol J ; 6: 61, 2009 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-19450275

RESUMEN

BACKGROUND: Pancreatic islet transplantation is a promising treatment for type I diabetes mellitus, but current immunosuppressive strategies do not consistently provide long-term survival of transplanted islets. We are therefore investigating the use of adeno-associated viruses (AAVs) as gene therapy vectors to transduce rat islets with immunosuppressive genes prior to transplantation into diabetic mice. RESULTS: We compared the transduction efficiency of AAV2 vectors with an AAV2 capsid (AAV2/2) to AAV2 vectors pseudotyped with AAV5 (AAV2/5), AAV8 (AAV2/8) or bovine adeno-associated virus (BAAV) capsids, or an AAV2 capsid with an insertion of the low density lipoprotein receptor ligand from apolipoprotein E (AAV2apoE), on cultured islets, in the presence of helper adenovirus infection to speed expression of a GFP transgene. Confocal microscopy and flow cytometry were used. The AAV2/5 vector was superior to AAV2/2 and AAV2/8 in rat islets. Flow cytometry indicated AAV2/5-mediated gene expression in approximately 9% of rat islet cells and almost 12% of insulin-positive cells. The AAV2/8 vector had a higher dependence on the helper virus multiplicity of infection than the AAV 2/5 vector. In addition, the BAAV and AAV2apoE vectors were superior to AAV2/2 for transducing rat islets. Rat islets (300 per mouse) transduced with an AAV2/5 vector harboring the immunosuppressive transgene, tgf beta 1, retain the ability to correct hyperglycemia when transplanted into immune-deficient diabetic mice. CONCLUSION: AAV2/5 vectors may therefore be useful for pre-treating donor islets prior to transplantation.


Asunto(s)
Dependovirus/genética , Diabetes Mellitus Tipo 1/terapia , Terapia Genética/métodos , Vectores Genéticos/genética , Islotes Pancreáticos/virología , Transducción Genética/métodos , Animales , Línea Celular , Dependovirus/metabolismo , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/cirugía , Femenino , Vectores Genéticos/metabolismo , Humanos , Técnicas In Vitro , Islotes Pancreáticos/metabolismo , Trasplante de Islotes Pancreáticos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Ratas , Ratas Wistar
8.
J Clin Invest ; 115(11): 3217-27, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16239968

RESUMEN

The G protein G(s)alpha is essential for hormone-stimulated cAMP generation and is an important metabolic regulator. We investigated the role of liver G(s)-signaling pathways by developing mice with liver-specific G(s)alpha deficiency (LGsKO mice). LGsKO mice had increased liver weight and glycogen content and reduced adiposity, whereas survival, body weight, food intake, and metabolic rates at ambient temperature were unaffected. LGsKO mice had increased glucose tolerance with both increased glucose-stimulated insulin secretion and increased insulin sensitivity in liver and muscle. Fed LGsKO mice were hypoglycemic and hypoinsulinemic, with low expression of hepatic gluconeogenic enzymes and PPARgamma coactivator-1. However, LGsKO mice maintained normal fasting glucose and insulin levels, probably due to prolonged breakdown of glycogen stores and possibly increased extrahepatic gluconeogenesis. Lipid metabolism was unaffected in fed LGsKO mice, but fasted LGsKO mice had increased lipogenic and reduced lipid oxidation gene expression in liver and increased serum triglyceride and FFA levels. LGsKO mice had very high serum glucagon and glucagon-like peptide-1 levels and pancreatic alpha cell hyperplasia, probably secondary to hepatic glucagon resistance and/or chronic hypoglycemia. Our results define novel roles for hepatic G(s)-signaling pathways in glucose and lipid regulation, which may prove useful in designing new therapeutic targets for diabetes and obesity.


Asunto(s)
Adiposidad/genética , Ayuno/fisiología , Subunidades alfa de la Proteína de Unión al GTP Gs/deficiencia , Subunidades alfa de la Proteína de Unión al GTP Gs/genética , Glucosa/metabolismo , Hígado/metabolismo , Animales , Ingestión de Alimentos/genética , Ayuno/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gs/fisiología , Glucagón/sangre , Péptido 1 Similar al Glucagón/sangre , Prueba de Tolerancia a la Glucosa , Hipoglucemia/genética , Hipoglucemia/metabolismo , Hipoglucemia/fisiopatología , Insulina/metabolismo , Resistencia a la Insulina/genética , Secreción de Insulina , Islotes Pancreáticos/metabolismo , Metabolismo de los Lípidos/genética , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Consumo de Oxígeno/genética , Transducción de Señal/genética
9.
Endocrinology ; 147(6): 2619-30, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16513827

RESUMEN

IGF-I and insulin are structurally related polypeptides that mediate a similar pattern of biological effects via receptors that display considerably homology. Administration of recombinant human IGF-I (rhIGF-I) has been proven to improve glucose control and liver and muscle insulin sensitivity in patients with type 2 diabetes mellitus (DM). The effect of rhIGF-I treatment was evaluated in a mouse model of type 2 DM (MKR mouse), which expresses a dominant-negative form of the human IGF-I receptor under the control of the muscle creatine kinase promoter specifically in skeletal muscle. MKR mice have impaired IGF-I and insulin signaling in skeletal muscle, leading to severe insulin resistance in muscle, liver, and fat, developing type 2 DM at 5 wk of age. Six-week-old MKR mice were treated with either saline or rhIGF-I for 3 wk. Blood glucose levels were decreased in response to rhIGF-I treatment in MKR mice. rhIGF-I treatment also increased body weight in MKR with concomitant changes in body composition such as a decrease in fat mass and an increase in lean body mass. Insulin, fatty acid, and triglyceride levels were not affected by rhIGF-I, nor were insulin or glucose tolerance in MKR mice. Hyperinsulinemic-euglycemic clamp analysis demonstrated no improvement in overall insulin sensitivity. Pyruvate and glutamine tolerance tests proved that there was a decrease in the rate of glucose appearance in MKR mice treated with rhIGF-I, suggesting a reduction in the gluconeogenic capacity of liver, kidney, and small intestine. Taken together these results demonstrate that the improvement of the hyperglycemia was achieved by inhibition of gluconeogenesis rather than an improvement in insulin sensitivity. Also, these results suggest that a functional IGF-I receptor in skeletal muscle is required for IGF-I to improve insulin sensitivity in this mouse model of type 2 DM.


Asunto(s)
Diabetes Mellitus Tipo 2/tratamiento farmacológico , Gluconeogénesis/efectos de los fármacos , Factor I del Crecimiento Similar a la Insulina/uso terapéutico , Animales , Glucemia/análisis , Composición Corporal/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Diabetes Mellitus Tipo 2/metabolismo , Ingestión de Líquidos/efectos de los fármacos , Técnica de Clampeo de la Glucosa , Glucosa-6-Fosfatasa/genética , Hormona del Crecimiento/sangre , Resistencia a la Insulina , Factor I del Crecimiento Similar a la Insulina/farmacología , Intestino Delgado/metabolismo , Riñón/metabolismo , Hígado/metabolismo , Masculino , Ratones , Fosfoenolpiruvato Carboxiquinasa (GTP)/genética , Proteínas Recombinantes/uso terapéutico
10.
Endocrinology ; 146(9): 4024-35, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15947005

RESUMEN

Leptin has metabolic effects on peripheral tissues including muscle, liver, and pancreas, and it has been successfully used to treat lipodystrophic diabetes, a leptin-deficient state. To study whether leptin therapy can be used for treatment of more common cases of type 2 diabetes, we used a mouse model of type 2 diabetes (MKR mice) that show normal leptin levels and are diabetic due to a primary defect in both IGF-I and insulin receptors signaling in skeletal muscle. Here we show that leptin administration to the MKR mice resulted in improvement of diabetes, an effect that was independent of the reduced food intake. The main effect of leptin therapy was enhanced hepatic insulin responsiveness possibly through decreasing gluconeogenesis. In addition, the reduction of lipid stores in liver and muscle induced by enhancing fatty acid oxidation and inhibiting lipogenesis led to an improvement of the lipotoxic condition. Our data suggest that leptin could be a potent antidiabetic drug in cases of type 2 diabetes that are not leptin resistant.


Asunto(s)
Diabetes Mellitus Tipo 2/tratamiento farmacológico , Hiperglucemia/tratamiento farmacológico , Resistencia a la Insulina , Leptina/farmacología , Proteínas Quinasas Activadas por AMP , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/metabolismo , Animales , Glucemia/efectos de los fármacos , Glucemia/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Modelos Animales de Enfermedad , Ingestión de Alimentos , Metabolismo Energético/efectos de los fármacos , Hiperglucemia/metabolismo , Hiperinsulinismo/tratamiento farmacológico , Hiperinsulinismo/metabolismo , Insulina/metabolismo , Secreción de Insulina , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos , Ratones Mutantes , Complejos Multienzimáticos/metabolismo , Músculo Esquelético/enzimología , Proteínas Serina-Treonina Quinasas/metabolismo , Triglicéridos/metabolismo
11.
Diabetes ; 53(11): 2901-9, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15504971

RESUMEN

The chronic hyperglycemia that occurs in type 2 diabetes may cause deterioration of beta-cell function and insulin resistance in peripheral tissues. Mice that express a dominant-negative IGF-1 receptor, specifically in skeletal muscle (MKR mice), exhibit severe insulin resistance, hyperinsulinemia, dyslipidemia, and hyper-glycemia. To determine the role of hyperglycemia in the worsening of the diabetes state in these animals, MKR mice were treated with phloridzin (PHZ), which inhibits intestinal glucose uptake and renal glucose reabsorption. Blood glucose levels were decreased and urine glucose levels were increased in response to PHZ treatment in MKR mice. PHZ treatment also increased food intake in MKR mice; however, the fat mass was decreased and lean body mass did not change. Serum insulin, fatty acid, and triglyceride levels were not affected by PHZ treatment in MKR mice. Hyperinsulinemic-euglycemic clamp analysis demonstrated that glucose uptake in white adipose tissue was significantly increased in response to PHZ treatment. Despite the reduction in blood glucose following PHZ treatment, there was no improvement in insulin-stimulated whole-body glucose uptake in MKR mice and neither was there suppression of endogenous glucose production by insulin. These results suggest that glucotoxicity plays little or no role in the worsening of insulin resistance that occurs in the MKR mouse model of type 2 diabetes.


Asunto(s)
Diabetes Mellitus Tipo 2/fisiopatología , Hiperglucemia/tratamiento farmacológico , Resistencia a la Insulina/fisiología , Hígado/fisiopatología , Florizina/uso terapéutico , Tejido Adiposo/anatomía & histología , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/patología , Animales , Glucemia/efectos de los fármacos , Glucemia/metabolismo , Composición Corporal/efectos de los fármacos , Modelos Animales de Enfermedad , Ingestión de Energía , Hígado/efectos de los fármacos , Hígado/fisiología , Masculino , Ratones , Ratones Transgénicos
12.
Endocrinology ; 145(10): 4667-76, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15231693

RESUMEN

Insulin resistance is one of the primary characteristics of type 2 diabetes. Mice overexpressing a dominant-negative IGF-I receptor specifically in muscle (MKR mice) demonstrate severe insulin resistance with high levels of serum and tissue lipids and eventually develop type 2 diabetes at 5-6 wk of age. To determine whether lipotoxicity plays a role in the progression of the disease, we crossed MKR mice with mice overexpressing a fatty acid translocase, CD36, in skeletal muscle. The double-transgenic MKR/CD36 mice showed normalization of the hyperglycemia and the hyperinsulinemia as well as a marked improvement in liver insulin sensitivity. The MKR/CD36 mice also exhibited normal rates of fatty acid oxidation in skeletal muscle when compared with the decreased rate of fatty acid oxidation in MKR. With the reduction in insulin resistance, beta-cell function returned to normal. These and other results suggest that the insulin resistance in the MKR mice is associated with increased muscle triglycerides levels and that whole-body insulin resistance can be, at least partially, reversed in association with a reduction in muscle triglycerides levels, although the mechanisms are yet to be determined.


Asunto(s)
Antígenos CD36/metabolismo , Diabetes Mellitus Tipo 2/fisiopatología , Resistencia a la Insulina , Músculo Esquelético/metabolismo , Animales , Diabetes Mellitus Tipo 2/patología , Ácidos Grasos/sangre , Glucosa/farmacocinética , Técnica de Clampeo de la Glucosa , Glucógeno/metabolismo , Hiperglucemia/sangre , Hiperinsulinismo/sangre , Técnicas In Vitro , Insulina/metabolismo , Secreción de Insulina , Islotes Pancreáticos/patología , Hígado/metabolismo , Masculino , Ratones , Ratones Transgénicos , Oxidación-Reducción , Triglicéridos/sangre , Triglicéridos/metabolismo
13.
Cell Metab ; 20(2): 333-45, 2014 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-24981835

RESUMEN

The melanocortin system regulates metabolic homeostasis and inflammation. Melanocortin agonists have contradictorily been reported to both increase and decrease metabolic rate and body temperature. We find two distinct physiologic responses occurring at similar doses. Intraperitoneal administration of the nonselective melanocortin agonist MTII causes a melanocortin-4 receptor (Mc4r)-mediated hypermetabolism/hyperthermia. This is preceded by a profound, transient hypometabolism/hypothermia that is preserved in mice lacking any one of Mc1r, Mc3r, Mc4r, or Mc5r. Three other melanocortin agonists also caused hypothermia, which is actively achieved via seeking a cool environment, vasodilation, and inhibition of brown adipose tissue thermogenesis. These results suggest that the hypometabolic/hypothermic effect of MTII is not due to a failure of thermoregulation. The hypometabolism/hypothermia was prevented by dopamine antagonists, and MTII selectively activated arcuate nucleus dopaminergic neurons, suggesting that these neurons may contribute to the hypometabolism/hypothermia. We propose that the hypometabolism/hypothermia is a regulated response, potentially beneficial during extreme physiologic stress.


Asunto(s)
Temperatura Corporal/efectos de los fármacos , Receptores de Melanocortina/agonistas , alfa-MSH/análogos & derivados , Animales , Núcleo Arqueado del Hipotálamo/metabolismo , Antagonistas de Dopamina/farmacología , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/metabolismo , Metabolismo Energético/efectos de los fármacos , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor de Melanocortina Tipo 1/agonistas , Receptor de Melanocortina Tipo 1/genética , Receptor de Melanocortina Tipo 1/metabolismo , Receptor de Melanocortina Tipo 3/agonistas , Receptor de Melanocortina Tipo 3/genética , Receptor de Melanocortina Tipo 3/metabolismo , Receptor de Melanocortina Tipo 4/agonistas , Receptor de Melanocortina Tipo 4/genética , Receptor de Melanocortina Tipo 4/metabolismo , Receptores de Melanocortina/genética , Receptores de Melanocortina/metabolismo , alfa-MSH/farmacología
14.
Endocrinology ; 154(10): 3539-51, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23861369

RESUMEN

Increased hepatic glucose production is a key pathophysiological feature of type 2 diabetes. Like all other cell types, hepatocytes express many G protein-coupled receptors (GPCRs) that are linked to different functional classes of heterotrimeric G proteins. The important physiological functions mediated by G(s)-coupled hepatic glucagon receptors are well-documented. In contrast, little is known about the in vivo physiological roles of hepatocyte GPCRs that are linked to G proteins of the G(q) family. To address this issue, we established a transgenic mouse line (Hep-Rq mice) that expressed a G(q)-linked designer receptor (Rq) in a hepatocyte-selective fashion. Importantly, Rq could no longer bind endogenous ligands but could be selectively activated by a synthetic drug, clozapine-N-oxide. Clozapine-N-oxide treatment of Hep-Rq mice enabled us to determine the metabolic consequences caused by selective activation of a G(q)-coupled GPCR in hepatocytes in vivo. We found that acute Rq activation in vivo led to pronounced increases in blood glucose levels, resulting from increased rates of glycogen breakdown and gluconeogenesis. We also demonstrated that the expression of the V(1b) vasopressin receptor, a G(q)-coupled receptor expressed by hepatocytes, was drastically increased in livers of ob/ob mice, a mouse model of diabetes. Strikingly, treatment of ob/ob mice with a selective V(1b) receptor antagonist led to reduced glucose excursions in a pyruvate challenge test. Taken together, these findings underscore the importance of G(q)-coupled receptors in regulating hepatic glucose fluxes and suggest novel receptor targets for the treatment of type 2 diabetes.


Asunto(s)
Diabetes Mellitus Tipo 2/inducido químicamente , Activadores de Enzimas/efectos adversos , Quinasas de Receptores Acoplados a Proteína-G/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Gluconeogénesis/efectos de los fármacos , Glucogenólisis/efectos de los fármacos , Hepatocitos/efectos de los fármacos , Animales , Antagonistas de los Receptores de Hormonas Antidiuréticas , Células Cultivadas , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Activadores de Enzimas/farmacología , Femenino , Quinasas de Receptores Acoplados a Proteína-G/química , Quinasas de Receptores Acoplados a Proteína-G/genética , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/antagonistas & inhibidores , Hepatocitos/citología , Hepatocitos/metabolismo , Humanos , Hipoglucemiantes/uso terapéutico , Masculino , Ratones , Ratones Obesos , Ratones Transgénicos , Ingeniería de Proteínas , Dominios y Motivos de Interacción de Proteínas , Receptor Muscarínico M3/agonistas , Receptor Muscarínico M3/química , Receptor Muscarínico M3/genética , Receptor Muscarínico M3/metabolismo , Receptores de Vasopresinas/metabolismo , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Organismos Libres de Patógenos Específicos
15.
Diabetes ; 61(10): 2414-23, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22596054

RESUMEN

Lipodystrophies are characterized by a loss of white adipose tissue, which causes ectopic lipid deposition, peripheral insulin resistance, reduced adipokine levels, and increased food intake (hyperphagia). The growth factor myostatin (MSTN) negatively regulates skeletal muscle growth, and mice with MSTN inhibition have reduced adiposity and improved insulin sensitivity. MSTN inhibition may therefore be efficacious in ameliorating diabetes. To test this hypothesis, we inhibited MSTN signaling in a diabetic model of generalized lipodystrophy to analyze its effects on glucose metabolism separate from effects on adipose mass. A-ZIP/F1 lipodystrophic mice were crossed to mice expressing a dominant-negative MSTN receptor (activin receptor type IIB) in muscle. MSTN inhibition in A-ZIP/F1 mice reduced blood glucose, serum insulin, triglyceride levels, and the rate of triglyceride synthesis, and improved insulin sensitivity. Unexpectedly, hyperphagia was normalized by MSTN inhibition in muscle. Blood glucose and hyperphagia were reduced in double mutants independent of the adipokine leptin. These results show that the effect of MSTN inhibition on insulin sensitivity is not secondary to an effect on adipose mass and that MSTN inhibition may be an effective treatment for diabetes. These results further suggest that muscle may play a heretofore unappreciated role in regulating food intake.


Asunto(s)
Diabetes Mellitus/genética , Hiperfagia/genética , Resistencia a la Insulina/genética , Lipodistrofia/complicaciones , Músculo Esquelético/metabolismo , Miostatina/genética , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/metabolismo , Animales , Glucemia/metabolismo , Diabetes Mellitus/etiología , Diabetes Mellitus/metabolismo , Modelos Animales de Enfermedad , Glucosa/farmacología , Hiperfagia/etiología , Hiperfagia/metabolismo , Insulina/farmacología , Peroxidación de Lípido/efectos de los fármacos , Peroxidación de Lípido/genética , Lipodistrofia/genética , Lipodistrofia/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones , Ratones Transgénicos , Músculo Esquelético/efectos de los fármacos , Miostatina/metabolismo
16.
Diabetes ; 60(10): 2484-95, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21873554

RESUMEN

OBJECTIVE: Obesity, insulin resistance, and type 2 diabetes form a tightly correlated cluster of metabolic disorders in which adipose is one of the first affected tissues. The role of hypoxia and hypoxia-inducible factor 1 (HIF1) in the development of high-fat diet (HFD)-induced obesity and insulin resistance was investigated using animal models. RESEARCH DESIGN AND METHODS: Mice with adipocyte-specific targeted disruption of the genes encoding the HIF1 obligatory subunits Hif1α or Arnt (Hif1ß) were generated using an aP2-Cre transgene with the Cre/LoxP system. The mice were fed an HFD for 12 weeks and their metabolic phenotypes were determined. Gene expression patterns in adipose tissues were also determined by microarray and quantitative PCR. RESULTS: On an HFD, adipocyte-specific ARNT knockout mice and adipocyte-specific HIF1α knockout mice exhibit similar metabolic phenotypes, including reduced fat formation, protection from HFD-induced obesity, and insulin resistance compared with similarly fed wild-type controls. The cumulative food intake remained similar; however, the metabolic efficiency was lower in adipocyte-specific HIF1α knockout mice. Moreover, indirect calorimetry revealed respiratory exchange ratios were reduced in adipocyte-specific HIF1α knockout mice. Hyperinsulinemic-euglycemic clamp studies demonstrated that targeted disruption of HIF1α in adipocytes enhanced whole-body insulin sensitivity. The improvement of insulin resistance is associated with decreased expression of Socs3 and induction of adiponectin. CONCLUSIONS: Inhibition of HIF1 in adipose tissue ameliorates obesity and insulin resistance. This study reveals that HIF1 could provide a novel potential therapeutic target for obesity and type 2 diabetes.


Asunto(s)
Adipocitos/metabolismo , Adiposidad/fisiología , Grasas de la Dieta/efectos adversos , Regulación de la Expresión Génica/fisiología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Resistencia a la Insulina , Tejido Adiposo Pardo , Tejido Adiposo Blanco , Envejecimiento , Animales , Translocador Nuclear del Receptor de Aril Hidrocarburo/genética , Translocador Nuclear del Receptor de Aril Hidrocarburo/metabolismo , Grasas de la Dieta/administración & dosificación , Técnica de Clampeo de la Glucosa , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Hígado/metabolismo , Ratones , Ratones Noqueados , Músculo Esquelético/metabolismo , Consumo de Oxígeno
17.
J Mol Endocrinol ; 46(3): 175-92, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21321096

RESUMEN

Adipose-specific inactivation of both AP-1 and CCAAT-enhancer-binding protein (C/EBP) families of B-ZIP transcription factors in transgenic mice causes severe lipoatrophy. To evaluate whether inactivation of only C/EBP members was critical for lipoatrophy, A-C/EBP, a dominant-negative protein that specifically inhibits the DNA binding of the C/EBP members, was expressed in adipose tissue. For the first 2 weeks after birth, aP2-A-C/EBP mice had no white adipose tissue (WAT), drastically reduced brown adipose tissue (BAT), and exhibited marked hepatic steatosis, hyperinsulinemia, and hyperlipidemia. However, WAT appeared during the third week, coinciding with significantly improved metabolic functioning. In adults, BAT remained reduced, causing cold intolerance. At 30 weeks, the aP2-A-C/EBP mice had only 35% reduced WAT, with clear morphological signs of lipodystrophy in subcutaneous fat. Circulating leptin and adiponectin levels were less than the wild-type levels, and these mice exhibited impaired triglyceride clearance. Insulin resistance, glucose intolerance, and reduced free fatty acid release in response to ß3-adrenergic agonist suggest improper functioning of the residual WAT. Gene expression analysis of inguinal WAT identified reduced mRNA levels of several enzymes involved in fatty acid synthesis and glucose metabolism that are known C/EBPα transcriptional targets. There were increased levels for genes involved in inflammation and muscle differentiation. However, when dermal fibroblasts from aP2-A-C/EBP mice were differentiated into adipocytes in tissue culture, muscle markers were elevated more than the inflammatory markers. These results demonstrate that the C/EBP family is essential for adipose tissue development during the early postnatal period, the regulation of glucose and lipid homeostasis in adults, and the suppression of the muscle lineage.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Lipodistrofia/etiología , Lipodistrofia/metabolismo , Proteínas/metabolismo , Tejido Adiposo/citología , Tejido Adiposo/metabolismo , Tejido Adiposo Pardo/citología , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/citología , Tejido Adiposo Blanco/metabolismo , Animales , Western Blotting , Composición Corporal/genética , Composición Corporal/fisiología , Proteínas Potenciadoras de Unión a CCAAT/genética , Calorimetría Indirecta , Células Cultivadas , Ingestión de Alimentos/genética , Ingestión de Alimentos/fisiología , Proteínas de Unión a Ácidos Grasos/genética , Fibroblastos/citología , Fibroblastos/metabolismo , Lipodistrofia/genética , Ratones , Ratones Transgénicos , Análisis de Secuencia por Matrices de Oligonucleótidos , Regiones Promotoras Genéticas/genética , Proteínas/genética , Triglicéridos/metabolismo
18.
Diabetes ; 60(10): 2578-87, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21831968

RESUMEN

OBJECTIVE: The role of adenosine (ADO) in the regulation of glucose homeostasis is not clear. In the current study, we used A1-ADO receptor (A1AR)-deficient mice to investigate the role of ADO/A1AR signaling for glucose homeostasis. RESEARCH DESIGN AND METHODS: After weaning, A1AR(-/-) and wild-type mice received either a standard diet (12 kcal% fat) or high-fat diet (HFD; 45 kcal% fat). Body weight, fasting plasma glucose, plasma insulin, and intraperitoneal glucose tolerance tests were performed in 8-week-old mice and again after 12-20 weeks of subsequent observation. Body composition was quantified by magnetic resonance imaging and epididymal fat-pad weights. Glucose metabolism was investigated by hyperinsulinemic-euglycemic clamp studies. To describe pathophysiological mechanisms, adipokines and Akt phosphorylation were measured. RESULTS: A1AR(-/-) mice were significantly heavier than wild-type mice because of an increased fat mass. Fasting plasma glucose and insulin were significantly higher in A1AR(-/-) mice after weaning and remained higher in adulthood. An intraperitoneal glucose challenge disclosed a significantly slower glucose clearance in A1AR(-/-) mice. An HFD enhanced this phenotype in A1AR(-/-) mice and unmasked a dysfunctional insulin secretory mechanism. Insulin sensitivity was significantly impaired in A1AR(-/-) mice on the standard diet shortly after weaning. Clamp studies detected a significant decrease of net glucose uptake in A1AR(-/-) mice and a reduced glucose uptake in muscle and white adipose tissue. Effects were not triggered by leptin deficiency but involved a decreased Akt phosphorylation. CONCLUSIONS: ADO/A1AR signaling contributes importantly to insulin-controlled glucose homeostasis and insulin sensitivity in C57BL/6 mice and is involved in the metabolic regulation of adipose tissue.


Asunto(s)
Intolerancia a la Glucosa/genética , Intolerancia a la Glucosa/metabolismo , Receptor de Adenosina A1/genética , Receptor de Adenosina A1/metabolismo , Transducción de Señal/fisiología , Adiposidad , Animales , Glucemia/metabolismo , Peso Corporal , Grasas de la Dieta/administración & dosificación , Grasas de la Dieta/efectos adversos , Ingestión de Alimentos , Femenino , Insulina/sangre , Insulina/metabolismo , Masculino , Ratones , Ratones Noqueados , Actividad Motora , Transducción de Señal/genética
19.
Endocrinology ; 151(11): 5185-94, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20843999

RESUMEN

Previous studies have shown that ß-cell M(3) muscarinic acetylcholine receptors (M3Rs) play a key role in maintaining blood glucose homeostasis by enhancing glucose-dependent insulin release. In this study, we tested the hypothesis that long-term, persistent activation of ß-cell M3Rs can improve glucose tolerance and ameliorate the metabolic deficits associated with the consumption of a high-fat diet. To achieve the selective and persistent activation of ß-cell M3Rs in vivo, we generated transgenic mice that expressed the Q490L mutant M3R in their pancreatic ß-cells (ß-M3-Q490L Tg mice). The Q490L point mutation is known to render the M3R constitutively active. The metabolic phenotypes of the transgenic mice were examined in several in vitro and in vivo metabolic tests. In the presence of 15 mm glucose and the absence of M3R ligands, isolated perifused islets prepared from ß-M3-Q490L Tg mice released considerably more insulin than wild-type control islets. This effect could be completely blocked by incubation of the transgenic islets with atropine (10 µm), an inverse muscarinic agonist, indicating that the Q490L mutant M3R exhibited ligand-independent signaling (constitutive activity) in mouse ß-cells. In vivo studies showed that ß-M3-Q490L Tg mice displayed greatly improved glucose tolerance and increased serum insulin levels as well as resistance to diet-induced glucose intolerance and hyperglycemia. These results suggest that chronic activation of ß-cell M3Rs may represent a useful approach to boost insulin output in the long-term treatment of type 2 diabetes.


Asunto(s)
Glucemia/metabolismo , Células Secretoras de Insulina/metabolismo , Receptor Muscarínico M3/metabolismo , Análisis de Varianza , Animales , Atropina/farmacología , Glucemia/efectos de los fármacos , Genotipo , Homeostasis/efectos de los fármacos , Insulina/sangre , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/efectos de los fármacos , Ratones , Ratones Transgénicos , Antagonistas Muscarínicos/farmacología , Fenotipo , Receptor Muscarínico M3/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
20.
PLoS One ; 4(4): e5370, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19401758

RESUMEN

BACKGROUND: Does diet-induced obesity persist after an obesigenic diet is removed? We investigated this question by providing male C57BL/6 mice with free access to two different obesigenic diets followed by a switch to chow to determine if obesity was reversible. METHODOLOGY/PRINCIPAL FINDINGS: Male C57BL/6 mice were randomly assigned to five weight-matched groups: 1) C group that continuously received a chow diet; 2) HF group on a 60% high fat diet; 3) EN group on the high fat diet plus liquid Ensure; 4) HF-C group switched from high fat to chow after 7 weeks; 5) EN-C group switched from high fat plus Ensure to chow after 7 weeks. All food intake was ad libitum. Body weight was increased after 7 weeks on both obesigenic diets (44.6+/-0.65, 39.8+/-0.63, and 28.6+/-0.63 g for EN, HF, and C groups, respectively) and resulted in elevated concentrations of serum insulin, glucose, and leptin and lower serum triglycerides. Development of obesity in HF and EN mice was caused by increased energy intake and a relative decrease of average energy output along with decreased ambulatory activity. After the switch to chow, the HF-C and EN-C groups lost weight but subsequently maintained a state of persistent obesity in comparison to the C group (34.8+/-1.2, 34.1+/-1.2 vs. 30.8+/-0.8 g respectively; P<0.05) with a 40-50% increase of body fat. All serum hormones and metabolites returned to control levels with the exception of a trend for increased leptin. The HF-C and EN-C groups had an average energy output in line with the C group and the persistent obesity was maintained despite a non-significant increase of energy intake of less than 1 kcal/d at the end of the study. CONCLUSION: Our results illustrate the importance of considering the history of energy imbalance in determining body weight and that a persistent elevation of body weight after removal of obesigenic diets can result from very small increases of energy intake.


Asunto(s)
Dieta/efectos adversos , Obesidad/etiología , Tejido Adiposo/patología , Animales , Glucemia/metabolismo , Distribución de la Grasa Corporal , Peso Corporal , Modelos Animales de Enfermedad , Ingestión de Energía , Metabolismo Energético , Glicerol/sangre , Leptina/sangre , Masculino , Ratones , Ratones Endogámicos C57BL , Actividad Motora , Obesidad/patología , Obesidad/fisiopatología , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA