Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Am J Physiol Endocrinol Metab ; 306(3): E332-43, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24326423

RESUMEN

Rho kinase (ROCK) isoforms regulate insulin signaling and glucose metabolism negatively or positively in cultured cell lines and skeletal muscle. However, the in vivo function of the ROCK1 isoform in adipose tissue has not been addressed. To determine the specific role of the adipose ROCK1 isoform in the development of insulin resistance and obesity, mice lacking ROCK1 in adipose tissue globally or selectively were studied. Here, we show that insulin's ability to activate IRS-1/PI3K/Akt signaling was greatly enhanced in adipose tissue of ROCK1(-/-) mice compared with wild-type mice. These effects resulted from the inhibitory effect of ROCK1 on insulin receptor action, as evidenced by the fact that IR tyrosine phosphorylation was abolished in ROCK1(-/-) MEF cells when ROCK1 was reexpressed. Consistently, adipose-specific disruption of ROCK1 increased IR tyrosine phosphorylation in adipose tissue and modestly improved sensitivity to insulin in obese mice induced by high-fat feeding. This effect is independent of any changes in adiposity, number or size of adipocytes, and metabolic parameters, including glucose, insulin, leptin, and triglyceride levels, demonstrating a minimal effect of adipose ROCK1 on whole body metabolism. Enzymatic activity of ROCK1 in adipose tissue remained ∼50%, which likely originated from the fraction of stromal vascular cells, suggesting involvement of these cells for adipose metabolic regulation. Moreover, ROCK isoform activities were increased in adipose tissue of diet-induced or genetically obese mice. These data suggest that adipose ROCK1 isoform plays an inhibtory role for the regulation of insulin sensitivity in diet-induced obesity in vivo.


Asunto(s)
Dieta/efectos adversos , Eliminación de Gen , Resistencia a la Insulina/genética , Quinasas Asociadas a rho/genética , Tejido Adiposo/metabolismo , Animales , Células Cultivadas , Femenino , Isoenzimas/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/genética , Obesidad/metabolismo , Especificidad de Órganos/genética
2.
Cell Metab ; 6(4): 247-9, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17908551

RESUMEN

Insulin resistance is an important risk factor for the development of type 2 diabetes and the metabolic syndrome. A new study in this issue of Cell Metabolism (Sun et al., 2007) shows that SIRT1, a mammalian sirtuin homolog and histone deacetylase, can ameliorate insulin resistance by silencing expression of protein tyrosine phosphatase 1B, a major negative regulator of insulin action.


Asunto(s)
Silenciador del Gen , Resistencia a la Insulina/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 1/genética , Sirtuinas/metabolismo , Regulación hacia Abajo , Humanos , Sirtuina 1
3.
Nature ; 436(7049): 356-62, 2005 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-16034410

RESUMEN

In obesity and type 2 diabetes, expression of the GLUT4 glucose transporter is decreased selectively in adipocytes. Adipose-specific Glut4 (also known as Slc2a4) knockout (adipose-Glut4(-/-)) mice show insulin resistance secondarily in muscle and liver. Here we show, using DNA arrays, that expression of retinol binding protein-4 (RBP4) is elevated in adipose tissue of adipose-Glut4(-/-) mice. We show that serum RBP4 levels are elevated in insulin-resistant mice and humans with obesity and type 2 diabetes. RBP4 levels are normalized by rosiglitazone, an insulin-sensitizing drug. Transgenic overexpression of human RBP4 or injection of recombinant RBP4 in normal mice causes insulin resistance. Conversely, genetic deletion of Rbp4 enhances insulin sensitivity. Fenretinide, a synthetic retinoid that increases urinary excretion of RBP4, normalizes serum RBP4 levels and improves insulin resistance and glucose intolerance in mice with obesity induced by a high-fat diet. Increasing serum RBP4 induces hepatic expression of the gluconeogenic enzyme phosphoenolpyruvate carboxykinase (PEPCK) and impairs insulin signalling in muscle. Thus, RBP4 is an adipocyte-derived 'signal' that may contribute to the pathogenesis of type 2 diabetes. Lowering RBP4 could be a new strategy for treating type 2 diabetes.


Asunto(s)
Diabetes Mellitus Tipo 2/sangre , Resistencia a la Insulina/fisiología , Obesidad/sangre , Proteínas de Unión al Retinol/metabolismo , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/metabolismo , Animales , Diabetes Mellitus Tipo 2/metabolismo , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Transportador de Glucosa de Tipo 4 , Hepatocitos/efectos de los fármacos , Hepatocitos/enzimología , Hepatocitos/metabolismo , Insulina/farmacología , Hígado/citología , Hígado/efectos de los fármacos , Hígado/enzimología , Ratones , Ratones Noqueados , Proteínas de Transporte de Monosacáridos/deficiencia , Proteínas de Transporte de Monosacáridos/genética , Proteínas de Transporte de Monosacáridos/metabolismo , Proteínas Musculares/deficiencia , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Músculos/efectos de los fármacos , Músculos/metabolismo , Obesidad/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Fosfoenolpiruvato Carboxiquinasa (GTP)/metabolismo , Proteínas de Unión al Retinol/genética , Proteínas Plasmáticas de Unión al Retinol , Rosiglitazona , Transducción de Señal/efectos de los fármacos , Tiazolidinedionas/farmacología
4.
Dev Cell ; 2(4): 489-95, 2002 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11970898

RESUMEN

Mice lacking the protein-tyrosine phosphatase PTP1B are hypersensitive to insulin and resistant to obesity. However, the molecular basis for resistance to obesity has been unclear. Here we show that PTP1B regulates leptin signaling. In transfection studies, PTP1B dephosphorylates the leptin receptor-associated kinase, Jak2. PTP1B is expressed in hypothalamic regions harboring leptin-responsive neurons. Compared to wild-type littermates, PTP1B(-/-) mice have decreased leptin/body fat ratios, leptin hypersensitivity, and enhanced leptin-induced hypothalamic Stat3 tyrosyl phosphorylation. Gold thioglucose treatment, which ablates leptin-responsive hypothalamic neurons, partially overcomes resistance to obesity in PTP1B(-/-) mice. Our data indicate that PTP1B regulates leptin signaling in vivo, likely by targeting Jak2. PTP1B may be a novel target to treat leptin resistance in obesity.


Asunto(s)
Leptina/metabolismo , Obesidad/metabolismo , Proteínas Tirosina Fosfatasas/metabolismo , Proteínas Proto-Oncogénicas , Transducción de Señal/fisiología , Animales , Células COS , Regulación Enzimológica de la Expresión Génica , Hipocampo/fisiología , Hipotálamo/fisiología , Janus Quinasa 2 , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Proteína Tirosina Fosfatasa no Receptora Tipo 1 , Proteínas Tirosina Fosfatasas/genética , Proteínas Tirosina Quinasas/metabolismo , Transfección , Aumento de Peso
5.
Mol Metab ; 5(8): 589-601, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27656397

RESUMEN

BACKGROUND: Insulin, a pleotrophic hormone, has diverse effects in the body. Recent work has highlighted the important role of insulin's action in the nervous system on glucose and energy homeostasis, memory, and mood. SCOPE OF REVIEW: Here we review experimental and clinical work that has broadened the understanding of insulin's diverse functions in the central and peripheral nervous systems, including glucose and body weight homeostasis, memory and mood, with particular emphasis on intranasal insulin. MAJOR CONCLUSIONS: Implications for the treatment of obesity, type 2 diabetes, dementia, and mood disorders are discussed in the context of brain insulin action. Intranasal insulin may have potential in the treatment of central nervous system-related metabolic disorders.

6.
Diabetes ; 65(7): 2006-19, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27207516

RESUMEN

Diabetic foot ulceration is a severe complication of diabetes that lacks effective treatment. Mast cells (MCs) contribute to wound healing, but their role in diabetes skin complications is poorly understood. Here we show that the number of degranulated MCs is increased in unwounded forearm and foot skin of patients with diabetes and in unwounded dorsal skin of diabetic mice (P < 0.05). Conversely, postwounding MC degranulation increases in nondiabetic mice, but not in diabetic mice. Pretreatment with the MC degranulation inhibitor disodium cromoglycate rescues diabetes-associated wound-healing impairment in mice and shifts macrophages to the regenerative M2 phenotype (P < 0.05). Nevertheless, nondiabetic and diabetic mice deficient in MCs have delayed wound healing compared with their wild-type (WT) controls, implying that some MC mediator is needed for proper healing. MCs are a major source of vascular endothelial growth factor (VEGF) in mouse skin, but the level of VEGF is reduced in diabetic mouse skin, and its release from human MCs is reduced in hyperglycemic conditions. Topical treatment with the MC trigger substance P does not affect wound healing in MC-deficient mice, but improves it in WT mice. In conclusion, the presence of nondegranulated MCs in unwounded skin is required for proper wound healing, and therapies inhibiting MC degranulation could improve wound healing in diabetes.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Neuropatías Diabéticas/metabolismo , Mastocitos/metabolismo , Piel/metabolismo , Cicatrización de Heridas/fisiología , Anciano , Animales , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Tipo 1/patología , Neuropatías Diabéticas/patología , Femenino , Humanos , Masculino , Mastocitos/patología , Ratones , Persona de Mediana Edad , Piel/patología
7.
Sci Rep ; 4: 3664, 2014 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-24441171

RESUMEN

Experimental and bioinformatic studies of transcription initiation by RNA polymerase II (RNAP2) have revealed a mechanism of RNAP2 transcription initiation less uniform across gene promoters than initially thought. However, the general transcription factor TFIIB is presumed to be universally required for RNAP2 transcription initiation. Based on bioinformatic analysis of data and effects of TFIIB knockdown in primary and transformed cell lines on cellular functionality and global gene expression, we report that TFIIB is dispensable for transcription of many human promoters, but is essential for herpes simplex virus-1 (HSV-1) gene transcription and replication. We report a novel cell cycle TFIIB regulation and localization of the acetylated TFIIB variant on the transcriptionally silent mitotic chromatids. Taken together, these results establish a new paradigm for TFIIB functionality in human gene expression, which when downregulated has potent anti-viral effects.


Asunto(s)
Factor de Transcripción TFIIB/metabolismo , Acetilación , Animales , Sitios de Unión , Ciclo Celular/genética , Línea Celular , Conjuntos de Datos como Asunto , Expresión Génica , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Regulación Viral de la Expresión Génica , Técnicas de Silenciamiento del Gen , Silenciador del Gen , Genes Letales , Genoma Humano , Herpesvirus Humano 1/genética , Humanos , Especificidad de Órganos/genética , Unión Proteica , ARN Polimerasa II/química , ARN Polimerasa II/metabolismo , Factor de Transcripción TFIIB/deficiencia , Factor de Transcripción TFIIB/genética , Sitio de Iniciación de la Transcripción , Transcripción Genética , Transcriptoma
8.
Trends Endocrinol Metab ; 24(10): 506-14, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23938132

RESUMEN

Obesity has increased at an alarming rate in recent years and is now a worldwide public health problem. Elucidating the mechanisms behind the metabolic dysfunctions associated with obesity is of high priority. The metabolic function of Rho-kinase (Rho-associated coiled-coil-containing kinase; ROCK) has been the subject of a great deal of investigation in metabolic-related diseases. It appears that inhibition of ROCK activity is beneficial for the treatment of a wide range of cardiovascular-related diseases. However, recent studies with genetic models of ROCK demonstrate that ROCK plays a positive role in insulin and leptin signaling. Here we discuss the newly identified functions of ROCK in regulating glucose and energy metabolism, with particular emphasis on metabolic actions of insulin and leptin.


Asunto(s)
Encéfalo/enzimología , Sistema Nervioso Periférico/enzimología , Quinasas Asociadas a rho/metabolismo , Animales , Encéfalo/metabolismo , Metabolismo Energético/fisiología , Humanos , Sistema Nervioso Periférico/metabolismo , Transducción de Señal/fisiología , Quinasas Asociadas a rho/genética
9.
Endocrinology ; 154(10): 3660-70, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23885017

RESUMEN

Normal leptin signaling is essential for the maintenance of body weight homeostasis. Proopiomelanocortin- and agouti-related peptide (AgRP)-producing neurons play critical roles in regulating energy metabolism. Our recent work demonstrates that deletion of Rho-kinase 1 (ROCK1) in the AgRP neurons of mice increased body weight and adiposity. Here, we report that selective loss of ROCK1 in AgRP neurons caused a significant decrease in energy expenditure and locomotor activity of mice. These effects were independent of any change in food intake. Furthermore, AgRP neuron-specific ROCK1-deficient mice displayed central leptin resistance, as evidenced by impaired Signal Transducer and Activator of Transcription 3 activation in response to leptin administration. Leptin's ability to hyperpolarize and decrease firing rate of AgRP neurons was also abolished in the absence of ROCK1. Moreover, diet-induced and genetic forms of obesity resulted in reduced ROCK1 activity in murine arcuate nucleus. Of note, high-fat diet also impaired leptin-stimulated ROCK1 activity in arcuate nucleus, suggesting that a defect in hypothalamic ROCK1 activity may contribute to the pathogenesis of central leptin resistance in obesity. Together, these data demonstrate that ROCK1 activation in hypothalamic AgRP neurons is required for the homeostatic regulation of energy expenditure and adiposity. These results further support previous work identifying ROCK1 as a key regulator of energy balance and suggest that targeting ROCK1 in the hypothalamus may lead to development of antiobesity therapeutics.


Asunto(s)
Proteína Relacionada con Agouti/metabolismo , Núcleo Arqueado del Hipotálamo/metabolismo , Metabolismo Energético , Actividad Motora , Neuronas/metabolismo , Obesidad/metabolismo , Fragmentos de Péptidos/metabolismo , Quinasas Asociadas a rho/metabolismo , Proteína Relacionada con Agouti/genética , Animales , Núcleo Arqueado del Hipotálamo/patología , Conducta Animal , Cruzamientos Genéticos , Ingestión de Energía , Leptina/sangre , Leptina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuronas/patología , Obesidad/sangre , Obesidad/etiología , Obesidad/patología , Fragmentos de Péptidos/genética , Proteínas Recombinantes de Fusión/metabolismo , Factor de Transcripción STAT3/metabolismo , Transmisión Sináptica , Quinasas Asociadas a rho/genética
10.
Endocrinology ; 153(4): 1649-62, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22355071

RESUMEN

A role of Rho-associated coiled-coil-containing protein kinase (ROCK)1 in regulating whole-body glucose homeostasis has been reported. However, cell-autonomous effects of ROCK1 on insulin-dependent glucose transport in adipocytes and muscle cells have not been elucidated. To determine the specific role of ROCK1 in glucose transport directly, ROCK1 expression in 3T3-L1 adipocytes and L6 myoblasts was biologically modulated. Here, we show that small interfering RNA-mediated ROCK1 depletion decreased insulin-induced glucose transport in adipocytes and myoblasts, whereas adenovirus-mediated ROCK1 expression increased this in a dose-dependent manner, indicating that ROCK1 is permissive for glucose transport. Inhibition of ROCK1 also impaired glucose transporter 4 translocation in 3T3-L1 adipocytes. Importantly, the ED50 of insulin for adipocyte glucose transport was reduced when ROCK1 was expressed, leading to hypersensitivity to insulin. These effects are dependent on actin cytoskeleton remodeling, because inhibitors of actin polymerization significantly decreased ROCK1's effect to promote insulin-stimulated glucose transport. Unlike ROCK2, ROCK1 binding to insulin receptor substrate (IRS)-1 was not detected by immunoprecipitation, although cell fractionation demonstrated both ROCK isoforms localize with IRS-1 in low-density microsomes. Moreover, insulin's ability to increase IRS-1 tyrosine 612 and serine 632/635 phosphorylation was attenuated by ROCK1 suppression. Replacing IRS-1 serine 632/635 with alanine reduced insulin-stimulated phosphatidylinositol 3-kinase activation and glucose transport in 3T3-L1 adipocytes, indicating that phosphorylation of these serine residues of IRS-1, which are substrates of the ROCK2 isoform in vitro, are crucial for maximal stimulation of glucose transport by insulin. Our studies identify ROCK1 as an important positive regulator of insulin action on glucose transport in adipocytes and muscle cells.


Asunto(s)
Actinas/metabolismo , Adipocitos/metabolismo , Glucosa/metabolismo , Polimerizacion , Quinasas Asociadas a rho/metabolismo , Células 3T3-L1 , Adipocitos/citología , Adipocitos/efectos de los fármacos , Animales , Transporte Biológico/fisiología , Células Cultivadas , Transportador de Glucosa de Tipo 4/metabolismo , Técnicas In Vitro , Insulina/farmacología , Proteínas Sustrato del Receptor de Insulina/metabolismo , Ratones , Modelos Animales , Mioblastos Esqueléticos/citología , Mioblastos Esqueléticos/efectos de los fármacos , Mioblastos Esqueléticos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo
11.
Nat Neurosci ; 15(10): 1391-8, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22941110

RESUMEN

Leptin regulates energy balance. However, knowledge of the critical intracellular transducers of leptin signaling remains incomplete. We found that Rho-kinase 1 (ROCK1) regulates leptin action on body weight homeostasis by activating JAK2, an initial trigger of leptin receptor signaling. Leptin promoted the physical interaction of JAK2 and ROCK1, thereby increasing phosphorylation of JAK2 and downstream activation of Stat3 and FOXO1. Mice lacking ROCK1 in either pro-opiomelanocortin (POMC) or agouti-related protein neurons, mediators of leptin action, displayed obesity and impaired leptin sensitivity. In addition, deletion of ROCK1 in the arcuate nucleus markedly enhanced food intake, resulting in severe obesity. Notably, ROCK1 was a specific mediator of leptin, but not insulin, regulation of POMC neuronal activity. Our data identify ROCK1 as a key regulator of leptin action on energy homeostasis.


Asunto(s)
Metabolismo Energético/fisiología , Hipotálamo/metabolismo , Leptina/fisiología , Receptores de Leptina/fisiología , Quinasas Asociadas a rho/fisiología , Potenciales de Acción/genética , Potenciales de Acción/fisiología , Proteína Relacionada con Agouti/fisiología , Animales , Regulación del Apetito/genética , Regulación del Apetito/fisiología , Núcleo Arqueado del Hipotálamo/metabolismo , Células Cultivadas , Ingestión de Alimentos , Janus Quinasa 2/metabolismo , Leptina/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Ratones Transgénicos , Neuronas/metabolismo , Obesidad/genética , Fosforilación , Proopiomelanocortina/metabolismo , Receptores de Leptina/agonistas , Receptores de Leptina/antagonistas & inhibidores , Factor de Transcripción STAT3/metabolismo , Quinasas Asociadas a rho/genética
12.
J Biol Chem ; 284(18): 11776-80, 2009 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-19276091

RESUMEN

Insulin signaling is essential for normal glucose homeostasis. Rho-kinase (ROCK) isoforms have been shown to participate in insulin signaling and glucose metabolism in cultured cell lines. To investigate the physiological role of ROCK1 in the regulation of whole body glucose homeostasis and insulin sensitivity in vivo, we studied mice with global disruption of ROCK1. Here we show that, at 16-18 weeks of age, ROCK1-deficient mice exhibited insulin resistance, as revealed by the failure of blood glucose levels to decrease after insulin injection. However, glucose tolerance was normal in the absence of ROCK1. These effects were independent of changes in adiposity. Interestingly, ROCK1 gene ablation caused a significant increase in glucose-induced insulin secretion, leading to hyperinsulinemia. To determine the mechanism(s) by which deletion of ROCK1 causes insulin resistance, we measured the ability of insulin to activate phosphatidylinositol 3-kinase and multiple distal pathways in skeletal muscle. Insulin-stimulated phosphatidylinositol 3-kinase activity associated with IRS-1 or phospho-tyrosine was also reduced approximately 40% without any alteration in tyrosine phosphorylation of insulin receptor in skeletal muscle. Concurrently, serine phosphorylation of IRS-1 at serine 632/635, which is phosphorylated by ROCK in vitro, was also impaired in these mice. Insulin-induced phosphorylation of Akt, AS160, S6K, and S6 was also decreased in skeletal muscle. These data suggest that ROCK1 deficiency causes systemic insulin resistance by impairing insulin signaling in skeletal muscle. Thus, our results identify ROCK1 as a novel regulator of glucose homeostasis and insulin sensitivity in vivo, which could lead to new treatment approaches for obesity and type 2 diabetes.


Asunto(s)
Glucosa/metabolismo , Resistencia a la Insulina , Insulina/metabolismo , Transducción de Señal , Adiposidad/genética , Animales , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/terapia , Proteínas Activadoras de GTPasa/genética , Proteínas Activadoras de GTPasa/metabolismo , Glucosa/genética , Insulina/genética , Proteínas Sustrato del Receptor de Insulina/genética , Proteínas Sustrato del Receptor de Insulina/metabolismo , Ratones , Ratones Noqueados , Obesidad/genética , Obesidad/metabolismo , Obesidad/terapia , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación/genética , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Quinasas S6 Ribosómicas/genética , Proteínas Quinasas S6 Ribosómicas/metabolismo , Quinasas Asociadas a rho
13.
J Biol Chem ; 283(21): 14230-41, 2008 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-18281274

RESUMEN

Protein-tyrosine phosphatase 1B (PTP1B) is a major negative regulator of insulin and leptin sensitivity. PTP1B overexpression in adipose tissue and skeletal muscle of humans and rodents may contribute to insulin resistance and obesity. The mechanisms mediating PTP1B overexpression in obese and diabetic states have been unclear. We find that adipose tissue inflammation and the pro-inflammatory cytokine tumor necrosis factor alpha (TNFalpha) regulate PTP1B expression in vivo. High fat feeding of mice increased PTP1B expression 1.5- to 7-fold in adipose tissue, liver, skeletal muscle, and arcuate nucleus of hypothalamus. PTP1B overexpression in high fat-fed mice coincided with increased adipose tissue expression of the macrophage marker CD68 and TNFalpha, which is implicated in causing obesity-induced insulin resistance. TNFalpha increased PTP1B mRNA and protein levels by 2- to 5-fold in a dose- and time-dependent manner in adipocyte and hepatocyte cell lines. TNFalpha administration in mice increased PTP1B mRNA 1.4- to 4-fold in adipose tissue, liver, skeletal muscle, and hypothalamic arcuate nucleus and PTP1B protein 2-fold in liver. Actinomycin D treatment blocked, and high dose salicylate treatment inhibited by 80%, TNFalpha-induced PTP1B expression in adipocyte cell lines, suggesting TNFalpha may induce PTP1B transcription via nuclear factor kappaB (NFkappaB) activation. Chromatin immunoprecipitation from adipocyte cell lines and liver of mice demonstrated TNFalpha-induced recruitment of NFkappaB subunit p65 to the PTP1B promoter in vitro and in vivo. In mice with diet-induced obesity, TNFalpha deficiency also partly blocked PTP1B overexpression in adipose tissue. Our data suggest that PTP1B overexpression in multiple tissues in obesity is regulated by inflammation and that PTP1B may be a target of anti-inflammatory therapies.


Asunto(s)
Regulación Enzimológica de la Expresión Génica , Proteína Tirosina Fosfatasa no Receptora Tipo 1/metabolismo , Alimentación Animal , Animales , Secuencia de Bases , Células Cultivadas , Diabetes Mellitus/enzimología , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Inflamación/enzimología , Insulina/metabolismo , Resistencia a la Insulina , Leptina/metabolismo , Ratones , Datos de Secuencia Molecular , Obesidad/enzimología , Proteína Tirosina Fosfatasa no Receptora Tipo 1/genética , Ratas , Transcripción Genética/efectos de los fármacos , Transcripción Genética/genética , Factor de Necrosis Tumoral alfa/deficiencia , Factor de Necrosis Tumoral alfa/farmacología
14.
J Biol Chem ; 280(15): 15038-46, 2005 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-15699041

RESUMEN

Protein-tyrosine phosphatase 1B (PTP1B) is an important negative regulator of insulin and leptin signaling in vivo. Mice lacking PTP1B (PTP1B-/- mice) are hyper-responsive to insulin and leptin and resistant to diet-induced obesity. The tissue(s) that mediate these effects of global PTP1B deficiency remain controversial. We exploited the high degree of hepatotropism of adenoviruses to assess the role of PTP1B in the liver. Liver-specific re-expression of PTP1B in PTP1B-/- mice led to marked attenuation of their enhanced insulin sensitivity. This correlated with, and was probably caused by, decreased insulin-stimulated tyrosyl phosphorylation of the insulin receptor (IR) and IR substrate 2-associated phosphatidylinositide 3-kinase activity. Analysis using phospho-specific antibodies for the IR revealed preferential dephosphorylation of Tyr-1162/1163 compared with Tyr-972 by PTP1B in vivo. Our findings show that the liver is a major site of the peripheral action of PTP1B in regulating glucose homeostasis.


Asunto(s)
Glucosa/metabolismo , Hígado/enzimología , Proteínas Tirosina Fosfatasas/biosíntesis , Proteínas Tirosina Fosfatasas/fisiología , Adenoviridae/genética , Animales , Sitios de Unión , Glucemia/metabolismo , Peso Corporal , Técnicas de Transferencia de Gen , Prueba de Tolerancia a la Glucosa , Humanos , Immunoblotting , Inmunohistoquímica , Inmunoprecipitación , Insulina/metabolismo , Operón Lac , Leptina/metabolismo , Hígado/metabolismo , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Proteína Tirosina Fosfatasa no Receptora Tipo 1 , Proteínas Tirosina Fosfatasas/genética , Receptor de Insulina/metabolismo , Factores de Tiempo , Tirosina/química
15.
J Virol ; 76(2): 532-40, 2002 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11752144

RESUMEN

The herpes simplex virus type 1 (HSV-1) 2-kb latency-associated transcript (LAT) is a stable intron, which accumulates in cells both lytically and latently infected with HSV-1. We have used a tetracycline-repressible expression system to determine the half-life of the 2-kb LAT RNA intron in the human neuroblastoma cell line SY5Y. Using Northern hybridization analyses of RNA isolated from transiently transfected SY5Y cells over time after repression of LAT expression, we measured the half-life of the 2-kb LAT to be approximately 24 h. Thus, unlike typical introns that are rapidly degraded in a matter of seconds following excision, the 2-kb LAT intron has a half-life similar to those of some of the more stable cellular mRNAs. Furthermore, a similar half-life was measured for the 2-kb LAT in transiently transfected nonneuronal monkey COS-1 cells, suggesting that the stability of the 2-kb LAT is neither cell type nor species specific. Previously, we found that the determinant responsible for the unusual stability of the 2-kb LAT maps to the 3' terminus of the intron. At this site is a nonconsensus intron branch point located adjacent to a predicted stem-loop structure that is hypothesized to prevent debranching by cellular enzymes. Here we show that mutations which alter the predicted stem-loop structure, such that branching is redirected, either reduce or abolish the stability of the 2-kb LAT intron.


Asunto(s)
Herpesvirus Humano 1/genética , Intrones/genética , Estabilidad del ARN , ARN no Traducido/metabolismo , ARN Viral/metabolismo , Latencia del Virus/genética , Animales , Secuencia de Bases , Células COS , Regulación de la Expresión Génica/efectos de los fármacos , Semivida , Humanos , Mutación/genética , Especificidad de Órganos , Estabilidad del ARN/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN no Traducido/genética , ARN Viral/genética , Especificidad de la Especie , Tetraciclina/farmacología , Factores de Tiempo , Transfección , Células Tumorales Cultivadas
16.
Am J Physiol Endocrinol Metab ; 283(2): E233-40, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12110527

RESUMEN

Elevation of plasma lactate levels induces peripheral insulin resistance, but the underlying mechanisms are unclear. We examined whether lactate infusion in rats suppresses glycolysis preceding insulin resistance and whether lactate-induced insulin resistance is accompanied by altered insulin signaling and/or insulin-stimulated glucose transport in skeletal muscle. Hyperinsulinemic euglycemic clamps were conducted for 6 h in conscious, overnight-fasted rats with or without lactate infusion (120 micromol x kg(-1) x min(-1)) during the final 3.5 h. Lactate infusion increased plasma lactate levels about fourfold. The elevation of plasma lactate had rapid effects to suppress insulin-stimulated glycolysis, which clearly preceded its effect to decrease insulin-stimulated glucose uptake. Both submaximal and maximal insulin-stimulated glucose transport decreased 25-30% (P < 0.05) in soleus but not in epitrochlearis muscles of lactate-infused rats. Lactate infusion did not alter insulin's ability to phosphorylate the insulin receptor, the insulin receptor substrate (IRS)-1, or IRS-2 but decreased insulin's ability to stimulate IRS-1- and IRS-2-associated phosphatidylinositol 3-kinase activities and Akt/protein kinase B activity by 47, 75, and 55%, respectively (P < 0.05 for all). In conclusion, elevation of plasma lactate suppressed glycolysis before its effect on insulin-stimulated glucose uptake, consistent with the hypothesis that suppression of glucose metabolism could precede and cause insulin resistance. In addition, lactate-induced insulin resistance was associated with impaired insulin signaling and decreased insulin-stimulated glucose transport in skeletal muscle.


Asunto(s)
Glucólisis/efectos de los fármacos , Resistencia a la Insulina/fisiología , Insulina/fisiología , Ácido Láctico/farmacología , Proteínas Musculares , Músculo Esquelético/fisiología , Transducción de Señal/efectos de los fármacos , Animales , Glucemia/análisis , Transportador de Glucosa de Tipo 4 , Insulina/sangre , Insulina/farmacología , Ácido Láctico/sangre , Masculino , Proteínas de Transporte de Monosacáridos/metabolismo , Ratas , Ratas Wistar
17.
J Biol Chem ; 279(23): 24844-51, 2004 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-15031294

RESUMEN

Previous studies implicate protein-tyrosine phosphatase 1B (PTP1B) and leukocyte antigen-related phosphatase (LAR) as negative regulators of insulin signaling. The expression and/or activity of PTP1B and LAR are increased in muscle of insulin-resistant rodents and humans. Overexpression of LAR selectively in muscle of transgenic mice causes whole body insulin resistance. To determine whether overexpression of PTP1B also causes insulin resistance, we generated transgenic mice overexpressing human PTP1B selectively in muscle at levels similar to those observed in insulin-resistant humans. Insulin-stimulated insulin receptor (IR) tyrosyl phosphorylation and phosphatidylinositol 3'-kinase activity were impaired by 35% and 40-60% in muscle of PTP1B-overexpressing mice compared with controls. Insulin stimulation of protein kinase C (PKC)lambda/zeta activity, which is required for glucose transport, was impaired in muscle of PTP1B-overexpressing mice compared with controls, showing that PTP1B overexpression impairs activation of these PKC isoforms. Furthermore, hyperinsulinemic-euglycemic clamp studies revealed that whole body glucose disposal and muscle glucose uptake were decreased by 40-50% in PTP1B-overexpressing mice. Overexpression of PTP1B or LAR alone in muscle caused similar impairments in insulin action; however, compound overexpression achieved by crossing PTP1B- and LAR-overexpressing mice was not additive. Antibodies against specific IR phosphotyrosines indicated overlapping sites of action of PTP1B and LAR. Thus, overexpression of PTP1B in vivo impairs insulin sensitivity, suggesting that overexpression of PTP1B in muscle of obese humans and rodents may contribute to their insulin resistance. Lack of additive impairment of insulin signaling by PTP1B and LAR suggests that these PTPs have overlapping actions in causing insulin resistance in vivo.


Asunto(s)
Antígenos HLA/metabolismo , Insulina/metabolismo , Músculos/metabolismo , Proteínas Tirosina Fosfatasas/genética , Proteínas Tirosina Fosfatasas/fisiología , Transgenes , Animales , Ácidos Grasos/metabolismo , Genotipo , Glucosa/metabolismo , Humanos , Resistencia a la Insulina , Isoenzimas , Masculino , Ratones , Ratones Transgénicos , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Fosfotirosina/química , Pruebas de Precipitina , Isoformas de Proteínas , Proteína Quinasa C/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 1 , Receptor de Insulina/metabolismo , Transducción de Señal , Tirosina/química
18.
J Biol Chem ; 279(20): 21598-605, 2004 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-14985357

RESUMEN

The majority of GLUT4 is sequestered in unique intracellular vesicles in the absence of insulin. Upon insulin stimulation GLUT4 vesicles translocate to, and fuse with, the plasma membrane. To determine the effect of GLUT4 content on the distribution and subcellular trafficking of GLUT4 and other vesicle proteins, adipocytes of adipose-specific, GLUT4-deficient (aP2-GLUT4-/-) mice and adipose-specific, GLUT4-overexpressing (aP2-GLUT4-Tg) mice were studied. GLUT4 amount was reduced by 80-95% in aP2-GLUT4-/- adipocytes and increased approximately 10-fold in aP2-GLUT4-Tg adipocytes compared with controls. Insulin-responsive aminopeptidase (IRAP) protein amount was decreased 35% in aP2-GLUT4-/- adipocytes and increased 45% in aP2-GLUT4-Tg adipocytes. VAMP2 protein was also decreased by 60% in aP2-GLUT4-/- adipocytes and increased 2-fold in aP2-GLUT4-Tg adipocytes. IRAP and VAMP2 mRNA levels were unaffected in aP2-GLUT4-Tg, suggesting that overexpression of GLUT4 affects IRAP and VAMP2 protein stability. The amount and subcellular distribution of syntaxin4, SNAP23, Munc-18c, and GLUT1 were unchanged in either aP2-GLUT4-/- or aP2-GLUT4-Tg adipocytes, but transferrin receptor was partially redistributed to the plasma membrane in aP2-GLUT4-Tg adipocytes. Immunogold electron microscopy revealed that overexpression of GLUT4 in adipocytes increased the number of GLUT4 molecules per vesicle nearly 2-fold and the number of GLUT4 and IRAP-containing vesicles per cell 3-fold. In addition, the proportion of cellular GLUT4 and IRAP at the plasma membrane in unstimulated aP2-GLUT4-Tg adipocytes was increased 4- and 2-fold, respectively, suggesting that sequestration of GLUT4 and IRAP is saturable. Our results show that GLUT4 overexpression or deficiency affects the amount of other GLUT4-vesicle proteins including IRAP and VAMP2 and that GLUT4 sequestration is saturable.


Asunto(s)
Adipocitos/fisiología , Aminopeptidasas/metabolismo , Proteínas de Transporte de Monosacáridos/genética , Proteínas Musculares , Adipocitos/efectos de los fármacos , Animales , Transporte Biológico , Cistinil Aminopeptidasa , Glucosa/metabolismo , Transportador de Glucosa de Tipo 4 , Insulina/farmacología , Cinética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Ratones Transgénicos , Proteínas de Transporte de Monosacáridos/deficiencia , Proteínas de Transporte de Monosacáridos/metabolismo , Proteínas R-SNARE , Fracciones Subcelulares/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA