Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
FASEB J ; 35(7): e21728, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34110658

RESUMEN

Proliferation and differentiation of preadipocytes, and other cell types, is accompanied by an increase in glucose uptake. Previous work showed that a pulse of high glucose was required during the first 3 days of differentiation in vitro, but was not required after that. The specific glucose metabolism pathways required for adipocyte differentiation are unknown. Herein, we used 3T3-L1 adipocytes as a model system to study glucose metabolism and expansion of the adipocyte metabolome during the first 3 days of differentiation. Our primary outcome measures were GLUT4 and adiponectin, key proteins associated with healthy adipocytes. Using complete media with 0 or 5 mM glucose, we distinguished between developmental features that were dependent on the differentiation cocktail of dexamethasone, insulin, and isobutylmethylxanthine alone or the cocktail plus glucose. Cocktail alone was sufficient to activate the capacity for 2-deoxglucose uptake and glycolysis, but was unable to support the expression of GLUT4 and adiponectin in mature adipocytes. In contrast, 5 mM glucose in the media promoted a transient increase in glucose uptake and glycolysis as well as a significant expansion of the adipocyte metabolome and proteome. Using genetic and pharmacologic approaches, we found that the positive effects of 5 mM glucose on adipocyte differentiation were specifically due to increased expression of 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3 (PFKFB3), a key regulator of glycolysis and the ancillary glucose metabolic pathways. Our data reveal a critical role for PFKFB3 activity in regulating the cellular metabolic remodeling required for adipocyte differentiation and maturation.


Asunto(s)
Adipocitos/metabolismo , Glucosa/metabolismo , Fosfofructoquinasa-2/metabolismo , Células 3T3-L1 , Adipocitos/efectos de los fármacos , Adiponectina/metabolismo , Animales , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Línea Celular , Dexametasona/farmacología , Transportador de Glucosa de Tipo 4/metabolismo , Glucólisis/efectos de los fármacos , Glucólisis/fisiología , Insulina/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Xantinas/farmacología
2.
F1000Res ; 92020.
Artículo en Inglés | MEDLINE | ID: mdl-32595948

RESUMEN

Deficient glucose transport and glucose disposal are key pathologies leading to impaired glucose tolerance and risk of type 2 diabetes.  The cloning and identification of the family of facilitative glucose transporters have helped to identify that underlying mechanisms behind impaired glucose disposal, particularly in muscle and adipose tissue.  There is much more than just transporter protein concentration that is needed to regulate whole body glucose uptake and disposal.  The purpose of this review is to discuss recent findings in whole body glucose disposal.  We hypothesize that impaired glucose uptake and disposal is a consequence of mismatched energy input and energy output.  Decreasing the former while increasing the latter is key to normalizing glucose homeostasis.


Asunto(s)
Glucosa/metabolismo , Tejido Adiposo/metabolismo , Transporte Biológico , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Proteínas de Transporte de Monosacáridos/metabolismo
3.
Obesity (Silver Spring) ; 27(12): 2025-2036, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31746554

RESUMEN

OBJECTIVE: Obesity is a major risk factor for cardiovascular disease, metabolic syndrome, and type 2 diabetes mellitus, whereas weight loss is associated with improved health outcomes. It is therefore important to learn how adipose contraction during weight loss contributes to improved health. It was hypothesized that adipose tissue undergoing weight loss would have a unique transcriptomic profile, expressing specific genes that might improve health. METHODS: This study conducted an RNA-sequencing analysis of the epididymal adipose tissue of mice fed either a high-fat diet (HFD) or a regular rodent chow diet (RD) ad libitum for 10 weeks versus a cohort of mice fed HFD for the first 5 weeks before being swapped to an RD for the remainder of the study (swapped diet [SWAP]). RESULTS: The swapped diet resulted in weight loss, with a parallel improvement in insulin sensitivity. RNA sequencing revealed several transcriptomic signatures distinct to adipose tissue in SWAP mice, distinguished from both RD and HFD adipose tissue. The analysis found a unique upregulated mRNA that encodes a secreted lipopolysaccharide-binding glycoprotein (CRISPLD2) in adipose tissue. Whereas cellular CRISPLD2 protein levels were unchanged, plasma CRIPSLD2 levels increased in SWAP mice following weight loss and could correlate with insulin sensitivity. CONCLUSIONS: Taken together, these data demonstrate that CRISPLD2 is a circulating adipokine that may regulate adipocyte remodeling during weight loss.


Asunto(s)
Tejido Adiposo/efectos de los fármacos , Moléculas de Adhesión Celular/metabolismo , Factores Reguladores del Interferón/metabolismo , Obesidad/tratamiento farmacológico , Pérdida de Peso/efectos de los fármacos , Animales , Masculino , Ratones , Ratones Endogámicos C57BL
5.
Methods Mol Biol ; 1713: 161-173, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29218525

RESUMEN

It is crucial to determine the regulation of GLUT4 translocation and redistribution to the plasma membrane. The HA-GLUT4-GFP dual-reporter construct has become an important tool in the assessment of GLUT4 recycling in cultured adipocytes and myocytes. Through the use of light microscopy, this reporter construct allows for visualization of GLUT4 specifically at the cell surface or GLUT4 that has recycled from the cell surface while simultaneously marking the total GLUT4 pool. Here, we discuss and outline the general application of this reporter construct and its use in evaluating GLUT4 translocation within cultured adipocytes.


Asunto(s)
Expresión Génica , Genes Reporteros , Transportador de Glucosa de Tipo 4/metabolismo , Imagen Molecular , Adipocitos/metabolismo , Animales , Línea Celular , Humanos , Ratones , Transporte de Proteínas , Ratas
6.
J Biol Chem ; 292(45): 18556-18564, 2017 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-28916720

RESUMEN

Expansion of adipose tissue in response to a positive energy balance underlies obesity and occurs through both hypertrophy of existing cells and increased differentiation of adipocyte precursors (hyperplasia). To better understand the nutrient signals that promote adipocyte differentiation, we investigated the role of glucose availability in regulating adipocyte differentiation and maturation. 3T3-L1 preadipocytes were grown and differentiated in medium containing a standard differentiation hormone mixture and either 4 or 25 mm glucose. Adipocyte maturation at day 9 post-differentiation was determined by key adipocyte markers, including glucose transporter 4 (GLUT4) and adiponectin expression and Oil Red O staining of neutral lipids. We found that adipocyte differentiation and maturation required a pulse of 25 mm glucose only during the first 3 days of differentiation. Importantly, fatty acids were unable to substitute for the 25 mm glucose pulse during this period. The 25 mm glucose pulse increased adiponectin and GLUT4 expression and accumulation of neutral lipids via distinct mechanisms. Adiponectin expression and other early markers of differentiation required an increase in the intracellular pool of total NAD/P. In contrast, GLUT4 protein expression was only partially restored by increased NAD/P levels. Furthermore, GLUT4 mRNA expression was mediated by glucose-dependent activation of GLUT4 gene transcription through the cis-acting GLUT4-liver X receptor element (LXRE) promoter element. In summary, this study supports the conclusion that high glucose promotes adipocyte differentiation via distinct metabolic pathways and independently of fatty acids. This may partly explain the mechanism underlying adipocyte hyperplasia that occurs much later than adipocyte hypertrophy in the development of obesity.


Asunto(s)
Adipocitos Blancos/metabolismo , Adipogénesis , Regulación de la Expresión Génica , Transportador de Glucosa de Tipo 4/metabolismo , Glucosa/metabolismo , NADP/metabolismo , NAD/metabolismo , Células 3T3-L1 , Adipocitos Blancos/citología , Adipocitos Blancos/patología , Adiponectina/genética , Adiponectina/metabolismo , Tejido Adiposo Blanco/citología , Tejido Adiposo Blanco/metabolismo , Tejido Adiposo Blanco/patología , Animales , Biomarcadores/metabolismo , Células Cultivadas , Transportador de Glucosa de Tipo 4/genética , Hiperglucemia/metabolismo , Hiperglucemia/patología , Hipoglucemia/metabolismo , Hipoglucemia/patología , Lipogénesis , Receptores X del Hígado/genética , Receptores X del Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Regiones Promotoras Genéticas , Células del Estroma/citología , Células del Estroma/metabolismo , Células del Estroma/patología , Regulación hacia Arriba
8.
Diabetes ; 65(12): 3585-3597, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27679559

RESUMEN

Impaired GLUT4-dependent glucose uptake is a contributing factor in the development of whole-body insulin resistance in obese patients and obese animal models. Previously, we demonstrated that transgenic mice engineered to express the human GLUT4 gene under the control of the human GLUT4 promoter (i.e., transgenic [TG] mice) are resistant to obesity-induced insulin resistance. A likely mechanism underlying increased insulin sensitivity is increased glucose uptake in skeletal muscle. The purpose of this study was to investigate the broader metabolic consequences of enhanced glucose uptake into muscle. We observed that the expression of several nuclear and mitochondrially encoded mitochondrial enzymes was decreased in TG mice but that mitochondrial number, size, and fatty acid respiration rates were unchanged. Interestingly, both pyruvate and glutamate respiration rates were decreased in TG mice. Metabolomics analyses of skeletal muscle samples revealed that increased GLUT4 transgene expression was associated with decreased levels of some tricarboxylic acid intermediates and amino acids, whereas the levels of several glucogenic amino acids were elevated. Furthermore, fasting acyl carnitines in obese TG mice were decreased, indicating that increased GLUT4-dependent glucose flux decreases nutrient stress by altering lipid and amino acid metabolism in skeletal muscle.


Asunto(s)
Aminoácidos/metabolismo , Transportador de Glucosa de Tipo 4/metabolismo , Glucosa/metabolismo , Mitocondrias/metabolismo , Obesidad/metabolismo , Animales , Transporte Biológico/fisiología , Western Blotting , Carnitina O-Palmitoiltransferasa/genética , Carnitina O-Palmitoiltransferasa/metabolismo , Transportador de Glucosa de Tipo 4/genética , Glucógeno/metabolismo , Humanos , Metabolismo de los Lípidos/genética , Metabolismo de los Lípidos/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Obesidad/genética , Triglicéridos/metabolismo
9.
Diabetes ; 65(10): 2911-9, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27411383

RESUMEN

Exercise promotes glucose clearance by increasing skeletal muscle GLUT4-mediated glucose uptake. Importantly, exercise upregulates muscle GLUT4 expression in an insulin-independent manner under conditions of insulin resistance, such as with type 2 diabetes. However, the insulin-independent mechanism responsible for rescued muscle GLUT4 expression is poorly understood. We used voluntary wheel running (VWR) in mice to test the prevailing hypothesis that insulin-independent upregulation of skeletal muscle GLUT4 protein expression with exercise is through increased Glut4 transcription. We demonstrate that 4 weeks of VWR exercise in obese mice rescued high-fat diet-induced decreased muscle GLUT4 protein and improved both fasting plasma insulin and hepatic triacylglyceride levels, but did not rescue muscle Glut4 mRNA. Persistent reduction in Glut4 mRNA suggests that a posttranscriptional mechanism regulated insulin-independent muscle GLUT4 protein expression in response to exercise in lean and obese mice. Reduction of GLUT4 protein in sedentary animals upon treatment with rapamycin revealed mTORC1-dependent GLUT4 regulation. However, no difference in GLUT4 protein expression was observed in VWR-exercised mice treated with either rapamycin or Torin 1, indicating that exercise-dependent regulation on GLUT4 was mTOR independent. The findings provide new insight into the mechanisms responsible for exercise-dependent regulation of GLUT4 in muscle.


Asunto(s)
Regulación de la Expresión Génica , Transportador de Glucosa de Tipo 4/metabolismo , Músculo Esquelético/metabolismo , Condicionamiento Físico Animal/fisiología , Adiposidad/efectos de los fármacos , Animales , Glucemia/efectos de los fármacos , Dieta Alta en Grasa , Prueba de Tolerancia a la Glucosa , Resistencia a la Insulina/fisiología , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Naftiridinas/farmacología , Procesamiento Postranscripcional del ARN , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/metabolismo
10.
Endocrinology ; 155(9): 3315-28, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24932807

RESUMEN

Glucose transporter isoform 4 (GLUT4) is the insulin-responsive glucose transporter mediating glucose uptake in adipose and skeletal muscle. Reduced GLUT4 translocation from intracellular storage compartments to the plasma membrane is a cause of peripheral insulin resistance. Using a chronic hyperinsulinemia (CHI)-induced cell model of insulin resistance and Rab5 mutant overexpression, we determined these manipulations altered endosomal sorting of GLUT4, thus contributing to the development of insulin resistance. We found that CHI induced insulin resistance in 3T3-L1 adipocytes by retaining GLUT4 in a Rab5-activity-dependent compartment that is unable to equilibrate with the cell surface in response to insulin. Furthermore, CHI-mediated retention of GLUT4 in this non-insulin-responsive compartment impaired filling of the transferrin receptor (TfR)-positive and TfR-negative insulin-responsive storage compartments. Our data suggest that hyperinsulinemia may inhibit GLUT4 by chronically maintaining GLUT4 in the Rab5 activity-dependent endosomal pathway and impairing formation of the TfR-negative and TfR-positive insulin-responsive GLUT4 pools. This model suggests that an early event in the development of insulin-resistant glucose transport in adipose tissue is to alter the intracellular localization of GLUT4 to a compartment that does not efficiently equilibrate with the cell surface when insulin levels are elevated for prolonged periods of time.


Asunto(s)
Endosomas/metabolismo , Transportador de Glucosa de Tipo 4/metabolismo , Hiperinsulinismo/metabolismo , Resistencia a la Insulina , Insulina/metabolismo , Proteínas de Unión al GTP rab5/metabolismo , Adipocitos/metabolismo , Tejido Adiposo/metabolismo , Animales , Endosomas/genética , Transportador de Glucosa de Tipo 4/genética , Humanos , Hiperinsulinismo/genética , Ratones , Transporte de Proteínas , Receptores de Transferrina/genética , Receptores de Transferrina/metabolismo , Proteínas de Unión al GTP rab5/genética
11.
Diabetes ; 62(7): 2249-58, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23474483

RESUMEN

The GLUT4 facilitative glucose transporter mediates insulin-dependent glucose uptake. We tested the hypothesis that moderate overexpression of human GLUT4 in mice, under the regulation of the human GLUT4 promoter, can prevent the hyperinsulinemia that results from obesity. Transgenic mice engineered to express the human GLUT4 gene and promoter (hGLUT4 TG) and their nontransgenic counterparts (NT) were fed either a control diet (CD) or a high-fat diet (HFD) for up to 10 weeks. Homeostasis model assessment of insulin resistance scores revealed that hGLUT4 TG mice fed an HFD remained highly insulin sensitive. The presence of the GLUT4 transgene did not completely prevent the metabolic adaptations to HFD. For example, HFD resulted in loss of dynamic regulation of the expression of several metabolic genes in the livers of fasted and refed NT and hGLUT4 TG mice. The hGLUT4 TG mice fed a CD showed no feeding-dependent regulation of SREBP-1c and fatty acid synthase (FAS) mRNA expression in the transition from the fasted to the fed state. Similarly, HFD altered the response of SREBP-1c and FAS mRNA expression to feeding in both strains. These changes in hepatic gene expression were accompanied by increased nuclear phospho-CREB in refed mice. Taken together, a moderate increase in expression of GLUT4 is a good target for treatment of insulin resistance.


Asunto(s)
Dieta Alta en Grasa , Transportador de Glucosa de Tipo 4/metabolismo , Resistencia a la Insulina/genética , Triglicéridos/sangre , Animales , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Ingestión de Alimentos/fisiología , Ácido Graso Sintasas/genética , Ácido Graso Sintasas/metabolismo , Expresión Génica , Glucosa/metabolismo , Transportador de Glucosa de Tipo 4/genética , Insulina/metabolismo , Hígado/metabolismo , Masculino , Ratones , Ratones Transgénicos , Fosforilación , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo
12.
Horm Mol Biol Clin Investig ; 15(3): 115-21, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25436738

RESUMEN

Skeletal muscle and adipose tissue play a major role in the regulation of whole-body glucose homeostasis. Much of the coordinated regulation of whole-body glucose homeostasis results from the regulation of lipid storage and release by adipose tissue and efficient switching between glucose oxidation and fatty acid oxidation in skeletal muscle. A control point for these biochemical actions center around the regulation of the insulin responsive glucose transporter, GLUT4. This review examines the regulation of GLUT4 in adipose tissue and skeletal muscle, in the context of the steroid nuclear hormone receptor signaling.


Asunto(s)
Tejido Adiposo/metabolismo , Resistencia a la Insulina , Músculo Esquelético/metabolismo , Receptores Nucleares Huérfanos/metabolismo , PPAR alfa/metabolismo , Transducción de Señal , Animales , Transportador de Glucosa de Tipo 4/metabolismo , Humanos , Receptores X del Hígado
13.
Diabetes ; 61(6): 1404-14, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22403301

RESUMEN

Insulin-mediated glucose uptake is highly sensitive to the levels of the facilitative glucose transporter protein, GLUT4. Repression of GLUT4 expression is correlated with insulin resistance in adipose tissue. We have shown that differentiation-dependent GLUT4 transcription was under control of class II histone deacetylases (HDACs). We hypothesized that HDACs may regulate gene expression in adipocytes as a result of adrenergic activation. To test this hypothesis, we activated cAMP signaling in 3T3-L1 adipocytes and in mice after an overnight fast. Chromatin immunoprecipitation experiments showed the association of HDAC4/5 with the GLUT4 promoter in vivo and in vitro in response to elevated cAMP. Knockdown of HDACs by small interfering RNA in cultured adipocytes prevented the cAMP-dependent decrease in GLUT4 transcription. HDAC4/5 recruitment to the GLUT4 promoter was dependent on the GLUT4 liver X receptor (LXR) binding site. Treatment of cells with an LXR agonist prevented the cAMP-dependent decrease in GLUT4 transcription. A loss of function mutation in the LXR response element was required for cAMP-dependent downregulation of GLUT4 expression in vitro, in fasted mice, and in mice subjected to diet-induced obesity. This suggests that activation of LXR signaling can prevent loss of GLUT4 expression in diabetes and obesity.


Asunto(s)
Adipocitos/metabolismo , AMP Cíclico/metabolismo , Ayuno/metabolismo , Transportador de Glucosa de Tipo 4/metabolismo , Histona Desacetilasas/metabolismo , Transducción de Señal/fisiología , Células 3T3-L1 , Animales , Células Cultivadas , Regulación hacia Abajo/fisiología , Glucosa/metabolismo , Transportador de Glucosa de Tipo 4/genética , Histona Desacetilasas/genética , Receptores X del Hígado , Ratones , Obesidad/genética , Obesidad/metabolismo , Receptores Nucleares Huérfanos/genética , Receptores Nucleares Huérfanos/metabolismo , Regiones Promotoras Genéticas , Transcripción Genética
14.
ISRN Mol Biol ; 2012: 856987, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-27335671

RESUMEN

GLUT4 has long been known to be an insulin responsive glucose transporter. Regulation of GLUT4 has been a major focus of research on the cause and prevention of type 2 diabetes. Understanding how insulin signaling alters the intracellular trafficking of GLUT4 as well as understanding the fate of glucose transported into the cell by GLUT4 will be critically important for seeking solutions to the current rise in diabetes and metabolic disease.

15.
J Biol Chem ; 286(1): 460-8, 2011 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-21047791

RESUMEN

Insulin-dependent glucose homeostasis is highly sensitive to the levels of insulin-responsive glucose transporter 4 (GLUT4) expression in adipocytes. The level of GLUT4 protein expression is highly dependent on the rate of GLUT4 gene transcription. GLUT4 gene transcription is decreased in a variety of physiologic states of insulin resistance including type 2 diabetes, obesity, and prolonged fasting. GLUT4 gene expression in adipocytes is differentiation-dependent, with full expression delayed until late in the differentiation program. In this paper, we have tested the hypothesis that differentiation-dependent GLUT4 gene expression in 3T3-L1 adipocytes is dependent on the nuclear concentration of a class II histone deacetylase (HDAC) protein, HDAC5. We have tested this hypothesis by reducing the levels of class II HDACs in the nuclear compartment of 3T3-L1 preadipocytes using two experimental approaches. First, preadipocytes were treated with phenylephrine, an α-adrenergic receptor agonist, to drive HDACS out of the nuclear compartment. Also, the class II HDAC concentrations were reduced using siRNA knockdown. In each case, reduction of nuclear class II HDAC concentration resulted in increased expression of endogenous GLUT4 mRNA in preadipocytes. Together, our data indicate that class II HDAC expression is the major regulatory mechanism for inhibiting GLUT4 expression in the predifferentiated state.


Asunto(s)
Adipocitos/citología , Adipocitos/metabolismo , Diferenciación Celular , Regulación de la Expresión Génica , Transportador de Glucosa de Tipo 4/genética , Histona Desacetilasas/metabolismo , Células 3T3-L1 , Animales , Núcleo Celular/metabolismo , Técnicas de Silenciamiento del Gen , Histona Desacetilasas/deficiencia , Histona Desacetilasas/genética , Humanos , Ratones , Regiones Promotoras Genéticas/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/genética , Transcripción Genética
16.
Diabetes ; 59(4): 800-7, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20103707

RESUMEN

OBJECTIVE: Insulin-mediated glucose uptake is highly sensitive to the levels of the facilitative GLUT protein GLUT4. Transcription of the GLUT4 gene is repressed in states of insulin deficiency and insulin resistance and can be induced by states of enhanced energy output, such as exercise. The cellular signals that regulate GLUT4 transcription are not well understood. We hypothesized that changes in energy substrate flux regulate GLUT4 transcription. RESEARCH DESIGN AND METHODS: To test this hypothesis, we used transgenic mice in which expression of the chloramphenicol acetyltransferase (CAT) gene is driven by a functional 895-bp fragment of the human GLUT4 promoter, thereby acting as a reporter for transcriptional activity. Mice were treated with a single dose of etomoxir, which inhibits the transport of long-chain fatty acids into mitochondria and increases basal, but not insulin-mediated, glucose flux. GLUT4 and transgenic CAT mRNA were measured. RESULTS: Etomoxir treatment significantly reduced CAT and GLUT4 mRNA transcription in adipose tissue, but did not change transcription in heart and skeletal muscle. Downregulation of GLUT4 transcription was cell autonomous, since etomoxir treatment of 3T3-L1 adipocytes resulted in a similar downregulation of GLUT4 mRNA. GLUT4 transcriptional downregulation required the putative liver X receptor (LXR) binding site in the human GLUT4 gene promoter in adipose tissue and 3T3-L1 adipocytes. Treatment of 3T3-L1 adipocytes with the LXR agonist, TO901317, partially restored GLUT4 expression in etomoxir-treated cells. CONCLUSIONS: Our data suggest that long-chain fatty acid import into mitochondria in adipose tissue may produce ligands that regulate expression of metabolic genes.


Asunto(s)
Cloranfenicol O-Acetiltransferasa/genética , Transportador de Glucosa de Tipo 4/genética , Tejido Adiposo/metabolismo , Animales , Cloranfenicol O-Acetiltransferasa/efectos de los fármacos , Cloranfenicol O-Acetiltransferasa/metabolismo , Cartilla de ADN , Compuestos Epoxi/farmacología , Transportador de Glucosa de Tipo 4/efectos de los fármacos , Transportador de Glucosa de Tipo 4/metabolismo , Humanos , Hipoglucemiantes/farmacología , Insulina/fisiología , Ratones , Ratones Transgénicos , Mitocondrias Cardíacas/efectos de los fármacos , Mitocondrias Cardíacas/metabolismo , Mitocondrias Hepáticas/efectos de los fármacos , Mitocondrias Hepáticas/metabolismo , Mitocondrias Musculares/efectos de los fármacos , Mitocondrias Musculares/metabolismo , Especificidad de Órganos , Regiones Promotoras Genéticas
17.
Vitam Horm ; 80: 193-215, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19251039

RESUMEN

One of the early events in the development of Type 2 diabetes appears to be an inhibition of insulin-mediated GLUT4 redistribution to the cell surface in tissues that express GLUT4. Understanding this process, and how it begins to breakdown in the development of insulin resistance is quite important as we face treatment and prevention of metabolic diseases. Over the past few years, and increasing number of laboratories have produced compelling data to demonstrate a role for both the actin and microtubule networks in the regulation of insulin-mediated GLUT4 redistribution to the cell surface. In this review, we explore this process from insulin-signal transduction to fusion of GLUT4 membrane vesicles, focusing on studies that have implicated a role for the cytoskeleton. We see from this body of work that both the actin network and the microtubule cytoskeleton play roles as targets of insulin action and effectors of insulin signaling leading to changes in GLUT4 redistribution to the cell surface and insulin-mediated glucose uptake.


Asunto(s)
Citoesqueleto/fisiología , Transportador de Glucosa de Tipo 4/metabolismo , Insulina/metabolismo , Transducción de Señal/fisiología , Animales , Protocolos de Quimioterapia Combinada Antineoplásica , Cisplatino , Humanos , Ifosfamida , Mitomicina
18.
Am J Physiol Regul Integr Comp Physiol ; 296(6): R1820-8, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19321702

RESUMEN

Denervation by sciatic nerve resection causes decreased muscle glucose transporter 4 (GLUT4) expression, but little is known about the signaling events that cause this decrease. Experiments were designed to test the hypothesis that decreased GLUT4 expression in denervated muscle occurs because of decreased calcium/CaMK activity, which would then lead to decreased activation of the transcription factors myocyte enhancer factor 2 (MEF2) and GLUT4 enhancer factor (GEF), which are required for normal GLUT4 expression. GLUT4 mRNA was elevated in mice expressing constitutively active CaMK isoform IV (CaMKIV) and decreased by denervation. Denervation decreased GEF binding to the promoter and the content of GEF in the nucleus, but there was no change in either MEF2 binding or MEF2 protein content. Expression of a MEF2-dependent reporter gene did not change in denervated skeletal muscle. To determine the domains of the GLUT4 promoter that respond to denervation, transgenic mice expressing the chloramphenicol acetyl transferase (CAT) reporter gene driven by different lengths of the human GLUT4 promoter were denervated. Using several different promoter/reporter gene constructs, we found that all areas of the GLUT4 promoter were truncated or missing, except for the MEF2 binding domain and the basal promoter. All of the GLUT4 promoter/CAT reporter constructs evaluated responded normally to denervation. Our data lead us to conclude that decreased CaMK activity is not the reason for decreased GLUT4 content in denervated muscle and that negative control of GLUT4 expression is not mediated through the MEF2 or GEF-binding domains. These findings indicate that withdrawal of a GEF- or MEF2-dependent signal is not likely a major determinant of the denervation effect on GLUT4 expression. Thus, the response to denervation may be mediated by other elements present in the basal promoter of the GLUT4 gene.


Asunto(s)
Transportador de Glucosa de Tipo 4/metabolismo , Músculo Esquelético/metabolismo , Animales , Sitios de Unión , Proteína Quinasa Tipo 4 Dependiente de Calcio Calmodulina/genética , Proteína Quinasa Tipo 4 Dependiente de Calcio Calmodulina/metabolismo , Cloranfenicol O-Acetiltransferasa/genética , Proteínas de Unión al ADN/metabolismo , Regulación hacia Abajo , Genes Reporteros , Transportador de Glucosa de Tipo 4/genética , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Desnervación Muscular , Músculo Esquelético/inervación , Factores Reguladores Miogénicos/genética , Factores Reguladores Miogénicos/metabolismo , Regiones Promotoras Genéticas , ARN Mensajero/metabolismo , Nervio Ciático/cirugía , Transducción de Señal , Factores de Transcripción/metabolismo
19.
J Biol Chem ; 283(12): 7429-37, 2008 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-18216015

RESUMEN

The insulin-responsive glucose transporter, GLUT4, is regulated in various physiologic states at the transcriptional level. When expressed in transgenic mice, the human GLUT4 promoter is governed by two cis-acting sequences: an MEF2 binding domain and Domain I, that function both as positive and negative regulators depending on the physiologic state. MEF2 proteins and GLUT4 enhancer factor (GEF) are known ligands for these cis-acting elements, but their mechanism of action is unclear. To begin to understand this important process, we have characterized GEF structural domains and its interactions with the MEF2A isoform. We find that the C terminus of GEF comprises its DNA-binding domain, but does not contribute to GEF homo-oligomerization. We also have found that GEF dimerizes with increased affinity to a hypophosphorylated form of MEF2A. Furthermore, we demonstrated that MEF2A binding to its cognate binding site can increase the DNA binding activity of GEF to Domain I, suggesting a novel mechanism for MEF2A transcriptional activation. Finally, we have demonstrated that the transcriptional co-repressor HDAC5 can interact with GEF in the absence of MEF2 proteins and specifically inhibit GLUT4 promoter activity. These findings lead to the hypothesis that GEF and the MEF2 proteins form a complex on the GLUT4 promoter that allows for recruitment of transcriptional co-regulators (repressors and/or activators) to control GLUT4 promoter activity.


Asunto(s)
Adipocitos/metabolismo , Proteínas de Unión al ADN/metabolismo , Transportador de Glucosa de Tipo 4/metabolismo , Histona Desacetilasas/metabolismo , Factores Reguladores Miogénicos/metabolismo , Regiones Promotoras Genéticas , Factores de Transcripción/metabolismo , Adipocitos/citología , Animales , Células COS , Chlorocebus aethiops , Proteínas de Unión al ADN/genética , Dimerización , Transportador de Glucosa de Tipo 4/genética , Histona Desacetilasas/genética , Factores de Transcripción MEF2 , Ratones , Ratones Transgénicos , Factores Reguladores Miogénicos/genética , Fosforilación , Unión Proteica/fisiología , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Factores de Transcripción/genética
20.
Am J Physiol Heart Circ Physiol ; 293(1): H402-8, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17369465

RESUMEN

We previously showed that GLUT4 expression is decreased in arterial smooth muscle of deoxycorticosterone acetate (DOCA)-salt hypertensive rats and that GLUT4-knockout mice have enhanced arterial reactivity. Therefore, we hypothesized that increased GLUT4 expression in vascular smooth muscle in vivo would prevent enhanced arterial reactivity and possibly reduce blood pressure in DOCA-salt hypertensive mice. Adult wild-type (WT) and GLUT4 transgenic (TG) mice were subjected to DOCA-salt hypertension with uninephrectomy or underwent uninephrectomy and remained normotensive. GLUT4 expression was increased more than twofold in the aortas of GLUT4 TG mice compared with WT aortas. Eight weeks after implantation of the DOCA pellets, GLUT4 expression decreased by 75% in aortas of WT hypertensive mice, but not in GLUT4 TG hypertensive aortas. Systolic blood pressure was significantly and similarly increased in WT and GLUT4 TG DOCA-salt mice compared with their respective sham-treated controls (159 vs. 111 mmHg). Responsiveness to the contractile agonist 5-HT was significantly increased in aortic rings from WT DOCA-salt mice but remained normal in GLUT4 TG DOCA mice. Phosphorylation of the myosin phosphatase targeting subunit MYPT1 was significantly enhanced in aortas of WT DOCA-salt mice, and this increase was prevented in GLUT4 TG mice. MYPT1 phosphorylation was also increased in nonhypertensive GLUT4-knockout mice. Myosin phosphatase, a major negative regulator of calcium sensitivity, is itself negatively regulated by phosphorylation of MYPT1. Therefore, our results show that preservation of GLUT4 expression prevents enhanced arterial reactivity in hypertension, possibly via effects on myosin phosphatase activity.


Asunto(s)
Aorta/fisiopatología , Presión Sanguínea , Transportador de Glucosa de Tipo 4/metabolismo , Hipertensión/fisiopatología , Quinasa de Cadena Ligera de Miosina/metabolismo , Animales , Masculino , Ratones , Ratones Transgénicos , Fosfatasa de Miosina de Cadena Ligera , Fosforilación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...