Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Elife ; 72018 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-30230471

RESUMEN

Hypothalamic neurons respond to nutritional cues by altering gene expression and neuronal excitability. The mechanisms that control such adaptive processes remain unclear. Here we define populations of POMC neurons in mice that are activated or inhibited by insulin and thereby repress or inhibit hepatic glucose production (HGP). The proportion of POMC neurons activated by insulin was dependent on the regulation of insulin receptor signaling by the phosphatase TCPTP, which is increased by fasting, degraded after feeding and elevated in diet-induced obesity. TCPTP-deficiency enhanced insulin signaling and the proportion of POMC neurons activated by insulin to repress HGP. Elevated TCPTP in POMC neurons in obesity and/or after fasting repressed insulin signaling, the activation of POMC neurons by insulin and the insulin-induced and POMC-mediated repression of HGP. Our findings define a molecular mechanism for integrating POMC neural responses with feeding to control glucose metabolism.


Asunto(s)
Glucosa/metabolismo , Insulina/farmacología , Plasticidad Neuronal/efectos de los fármacos , Neuronas/metabolismo , Proopiomelanocortina/metabolismo , Animales , Humanos , Hipoglucemiantes/administración & dosificación , Hipoglucemiantes/farmacología , Hipotálamo/citología , Insulina/administración & dosificación , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Plasticidad Neuronal/genética , Proopiomelanocortina/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 2/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 2/metabolismo , Receptor de Insulina/genética , Receptor de Insulina/metabolismo
2.
Diabetes ; 67(7): 1246-1257, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29712668

RESUMEN

Insulin regulates glucose metabolism by eliciting effects on peripheral tissues as well as the brain. Insulin receptor (IR) signaling inhibits AgRP-expressing neurons in the hypothalamus to contribute to the suppression of hepatic glucose production (HGP) by insulin, whereas AgRP neuronal activation attenuates brown adipose tissue (BAT) glucose uptake. The tyrosine phosphatase TCPTP suppresses IR signaling in AgRP neurons. Hypothalamic TCPTP is induced by fasting and degraded after feeding. Here we assessed the influence of TCPTP in AgRP neurons in the control of glucose metabolism. TCPTP deletion in AgRP neurons (Agrp-Cre;Ptpn2fl/fl ) enhanced insulin sensitivity, as assessed by the increased glucose infusion rates, and reduced HGP during hyperinsulinemic-euglycemic clamps, accompanied by increased [14C]-2-deoxy-d-glucose uptake in BAT and browned white adipose tissue. TCPTP deficiency in AgRP neurons promoted the intracerebroventricular insulin-induced repression of hepatic gluconeogenesis in otherwise unresponsive food-restricted mice, yet had no effect in fed/satiated mice where hypothalamic TCPTP levels are reduced. The improvement in glucose homeostasis in Agrp-Cre;Ptpn2fl/fl mice was corrected by IR heterozygosity (Agrp-Cre;Ptpn2fl/fl ;Insrfl/+ ), causally linking the effects on glucose metabolism with the IR signaling in AgRP neurons. Our findings demonstrate that TCPTP controls IR signaling in AgRP neurons to coordinate HGP and brown/beige adipocyte glucose uptake in response to feeding/fasting.


Asunto(s)
Proteína Relacionada con Agouti/metabolismo , Ingestión de Alimentos/fisiología , Gluconeogénesis/genética , Glucosa/metabolismo , Insulina/metabolismo , Neuronas/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 2/fisiología , Tejido Adiposo Pardo/metabolismo , Animales , Metabolismo de los Hidratos de Carbono/fisiología , Metabolismo Energético/genética , Ayuno , Técnica de Clampeo de la Glucosa , Hígado/metabolismo , Ratones , Ratones Transgénicos , Proteína Tirosina Fosfatasa no Receptora Tipo 2/genética , Receptor de Insulina/metabolismo , Transducción de Señal/genética
3.
Mol Metab ; 5(11): 1083-1091, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27818934

RESUMEN

OBJECTIVE: The development of skeletal muscle insulin resistance is an early physiological defect, yet the intracellular mechanisms accounting for this metabolic defect remained unresolved. Here, we have examined the role of glucose-6-phosphate dehydrogenase (G6PDH) activity in the pathogenesis of insulin resistance in skeletal muscle. METHODS: Multiple mouse disease states exhibiting insulin resistance and glucose intolerance, as well as obese humans defined as insulin-sensitive, insulin-resistant, or pre-diabetic, were examined. RESULTS: We identified increased glucose-6-phosphate dehydrogenase (G6PDH) activity as a common intracellular adaptation that occurs in parallel with the induction of insulin resistance in skeletal muscle and is present across animal and human disease states with an underlying pathology of insulin resistance and glucose intolerance. We observed an inverse association between G6PDH activity and nitric oxide synthase (NOS) activity and show that increasing NOS activity via the skeletal muscle specific neuronal (n)NOSµ partially suppresses G6PDH activity in skeletal muscle cells. Furthermore, attenuation of G6PDH activity in skeletal muscle cells via (a) increased nNOSµ/NOS activity, (b) pharmacological G6PDH inhibition, or (c) genetic G6PDH inhibition increases insulin-independent glucose uptake. CONCLUSIONS: We have identified a novel, previously unrecognized role for G6PDH in the regulation of skeletal muscle glucose metabolism.


Asunto(s)
Glucosa/metabolismo , Glucosafosfato Deshidrogenasa/metabolismo , Músculo Esquelético/metabolismo , Animales , Glucosa-6-Fosfato , Humanos , Insulina , Resistencia a la Insulina , Ratones , Fibras Musculares Esqueléticas , Óxido Nítrico
5.
Am J Physiol Endocrinol Metab ; 310(10): E838-45, 2016 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-27006199

RESUMEN

Nitric oxide influences intramuscular signaling that affects skeletal muscle glucose uptake during exercise. The role of the main NO-producing enzyme isoform activated during skeletal muscle contraction, neuronal nitric oxide synthase-µ (nNOSµ), in modulating glucose uptake has not been investigated in a physiological exercise model. In this study, conscious and unrestrained chronically catheterized nNOSµ(+/+) and nNOSµ(-/-) mice either remained at rest or ran on a treadmill at 17 m/min for 30 min. Both groups of mice demonstrated similar exercise capacity during a maximal exercise test to exhaustion (17.7 ± 0.6 vs. 15.9 ± 0.9 min for nNOSµ(+/+) and nNOSµ(-/-), respectively, P > 0.05). Resting and exercise blood glucose levels were comparable between the genotypes. Very low levels of NOS activity were detected in skeletal muscle from nNOSµ(-/-) mice, and exercise increased NOS activity only in nNOSµ(+/+) mice (4.4 ± 0.3 to 5.2 ± 0.4 pmol·mg(-1)·min(-1), P < 0.05). Exercise significantly increased glucose uptake in gastrocnemius muscle (5- to 7-fold) and, surprisingly, more so in nNOSµ(-/-) than in nNOSµ(+/+) mice (P < 0.05). This is in parallel with a greater increase in AMPK phosphorylation during exercise in nNOSµ(-/-) mice. In conclusion, nNOSµ is not essential for skeletal muscle glucose uptake during exercise, and the higher skeletal muscle glucose uptake during exercise in nNOSµ(-/-) mice may be due to compensatory increases in AMPK activation.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Glucemia/metabolismo , Músculo Esquelético/metabolismo , Óxido Nítrico Sintasa de Tipo I/genética , Condicionamiento Físico Animal , Animales , Femenino , Glucosa/metabolismo , Masculino , Ratones , Ratones Noqueados , Fosforilación
6.
Elife ; 42015 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-26496200

RESUMEN

T follicular helper cells (Tfh) are critical for the longevity and quality of antibody-mediated protection against infection. Yet few signaling pathways have been identified to be unique solely to Tfh development. ROQUIN is a post-transcriptional repressor of T cells, acting through its ROQ domain to destabilize mRNA targets important for Th1, Th17, and Tfh biology. Here, we report that ROQUIN has a paradoxical function on Tfh differentiation mediated by its RING domain: mice with a T cell-specific deletion of the ROQUIN RING domain have unchanged Th1, Th2, Th17, and Tregs during a T-dependent response but show a profoundly defective antigen-specific Tfh compartment. ROQUIN RING signaling directly antagonized the catalytic α1 subunit of adenosine monophosphate-activated protein kinase (AMPK), a central stress-responsive regulator of cellular metabolism and mTOR signaling, which is known to facilitate T-dependent humoral immunity. We therefore unexpectedly uncover a ROQUIN-AMPK metabolic signaling nexus essential for selectively promoting Tfh responses.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Diferenciación Celular , Transducción de Señal , Linfocitos T Colaboradores-Inductores/fisiología , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Ratones , Eliminación de Secuencia , Ubiquitina-Proteína Ligasas/genética
7.
Biochem Biophys Res Commun ; 463(4): 818-24, 2015 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-26086096

RESUMEN

RATIONALE: Cardiac metabolism is thought to be altered in insulin resistance and type 2 diabetes (T2D). Our understanding of the regulation of cardiac substrate metabolism and insulin sensitivity has largely been derived from ex vivo preparations which are not subject to the same metabolic regulation as in the intact heart in vivo. Studies are therefore required to examine in vivo cardiac glucose metabolism under physiologically relevant conditions. OBJECTIVE: To determine the temporal pattern of the development of cardiac insulin resistance and to compare with dynamic approaches to interrogate cardiac glucose and intermediary metabolism in vivo. METHODS AND RESULTS: Studies were conducted to determine the evolution of cardiac insulin resistance in C57Bl/6 mice fed a high-fat diet (HFD) for between 1 and 16 weeks. Dynamic in vivo cardiac glucose metabolism was determined following oral administration of [U-(13)C] glucose. Hearts were collected after 15 and 60 min and flux profiling was determined by measuring (13)C mass isotopomers in glycolytic and tricarboxylic acid (TCA) cycle intermediates. Cardiac insulin resistance, determined by euglycemic-hyperinsulinemic clamp, was evident after 3 weeks of HFD. Despite the presence of insulin resistance, in vivo cardiac glucose metabolism following oral glucose administration was not compromised in HFD mice. This contrasts our recent findings in skeletal muscle, where TCA cycle activity was reduced in mice fed a HFD. Similar to our report in muscle, glucose derived pyruvate entry into the TCA cycle in the heart was almost exclusively via pyruvate dehydrogenase, with pyruvate carboxylase mediated anaplerosis being negligible after oral glucose administration. CONCLUSIONS: Under experimental conditions which closely mimic the postprandial state, the insulin resistant mouse heart retains the ability to stimulate glucose metabolism.


Asunto(s)
Dieta Alta en Grasa , Técnica de Clampeo de la Glucosa , Glucosa/metabolismo , Hiperinsulinismo/metabolismo , Metabolómica , Miocardio/metabolismo , Animales , Cromatografía de Gases y Espectrometría de Masas , Resistencia a la Insulina , Masculino , Ratones , Ratones Endogámicos C57BL
8.
Cell Metab ; 21(5): 718-30, 2015 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-25955207

RESUMEN

Accumulation of diacylglycerol (DG) in muscle is thought to cause insulin resistance. DG is a precursor for phospholipids, thus phospholipid synthesis could be involved in regulating muscle DG. Little is known about the interaction between phospholipid and DG in muscle; therefore, we examined whether disrupting muscle phospholipid synthesis, specifically phosphatidylethanolamine (PtdEtn), would influence muscle DG content and insulin sensitivity. Muscle PtdEtn synthesis was disrupted by deleting CTP:phosphoethanolamine cytidylyltransferase (ECT), the rate-limiting enzyme in the CDP-ethanolamine pathway, a major route for PtdEtn production. While PtdEtn was reduced in muscle-specific ECT knockout mice, intramyocellular and membrane-associated DG was markedly increased. Importantly, however, this was not associated with insulin resistance. Unexpectedly, mitochondrial biogenesis and muscle oxidative capacity were increased in muscle-specific ECT knockout mice and were accompanied by enhanced exercise performance. These findings highlight the importance of the CDP-ethanolamine pathway in regulating muscle DG content and challenge the DG-induced insulin resistance hypothesis.


Asunto(s)
Citidina Difosfato/análogos & derivados , Diglicéridos/metabolismo , Etanolaminas/metabolismo , Resistencia a la Insulina , Músculo Esquelético/metabolismo , Biogénesis de Organelos , Animales , Citidina Difosfato/metabolismo , Glucosa/metabolismo , Metabolismo de los Lípidos , Ratones , Ratones Noqueados , Obesidad/genética , Obesidad/metabolismo , ARN Nucleotidiltransferasas/genética , ARN Nucleotidiltransferasas/metabolismo
9.
PLoS One ; 10(5): e0128398, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26020641

RESUMEN

One serious side effect of statin drugs is skeletal muscle myopathy. Although the mechanism(s) responsible for statin myopathy remains to be fully determined, an increase in muscle atrophy gene expression and changes in mitochondrial content and/or function have been proposed to play a role. In this study, we examined the relationship between statin-induced expression of muscle atrophy genes, regulators of mitochondrial biogenesis, and markers of mitochondrial content in slow- (ST) and fast-twitch (FT) rat skeletal muscles. Male Sprague Dawley rats were treated with simvastatin (60 or 80 mg·kg(-1)·day(-1)) or vehicle control via oral gavage for 14 days. In the absence of overt muscle damage, simvastatin treatment induced an increase in atrogin-1, MuRF1 and myostatin mRNA expression; however, these were not associated with changes in peroxisome proliferator gamma co-activator 1 alpha (PGC-1α) protein or markers of mitochondrial content. Simvastatin did, however, increase neuronal nitric oxide synthase (nNOS), endothelial NOS (eNOS) and AMPK α-subunit protein expression, and tended to increase total NOS activity, in FT but not ST muscles. Furthermore, simvastatin induced a decrease in ß-hydroxyacyl CoA dehydrogenase (ß-HAD) activity only in FT muscles. These findings suggest that the statin-induced activation of muscle atrophy genes occurs independent of changes in PGC-1α protein and mitochondrial content. Moreover, muscle-specific increases in NOS expression and possibly NO production, and decreases in fatty acid oxidation, could contribute to the previously reported development of overt statin-induced muscle damage in FT muscles.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Mitocondrias Musculares/metabolismo , Proteínas Musculares/biosíntesis , Atrofia Muscular/metabolismo , Simvastatina/farmacología , Factores de Transcripción/metabolismo , Animales , Masculino , Mitocondrias Musculares/patología , Atrofia Muscular/patología , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Ratas , Ratas Sprague-Dawley
10.
Cell Metab ; 21(3): 403-16, 2015 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-25738456

RESUMEN

Interleukin-6 (IL-6) plays a paradoxical role in inflammation and metabolism. The pro-inflammatory effects of IL-6 are mediated via IL-6 "trans-signaling," a process where the soluble form of the IL-6 receptor (sIL-6R) binds IL-6 and activates signaling in inflammatory cells that express the gp130 but not the IL-6 receptor. Here we show that trans-signaling recruits macrophages into adipose tissue (ATM). Moreover, blocking trans-signaling with soluble gp130Fc protein prevents high-fat diet (HFD)-induced ATM accumulation, but does not improve insulin action. Importantly, however, blockade of IL-6 trans-signaling, unlike complete ablation of IL-6 signaling, does not exacerbate obesity-induced weight gain, liver steatosis, or insulin resistance. Our data identify the sIL-6R as a critical chemotactic signal for ATM recruitment and suggest that selectively blocking IL-6 trans-signaling may be a more favorable treatment option for inflammatory diseases, compared with current treatments that completely block the action of IL-6 and negatively impact upon metabolic homeostasis.


Asunto(s)
Tejido Adiposo/metabolismo , Dieta Alta en Grasa/efectos adversos , Resistencia a la Insulina/fisiología , Interleucina-6/metabolismo , Macrófagos/metabolismo , Macrófagos/fisiología , Transducción de Señal/fisiología , Tejido Adiposo/fisiología , Animales , Receptor gp130 de Citocinas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptores de Interleucina-6/metabolismo
11.
Biochim Biophys Acta ; 1851(2): 210-9, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25490466

RESUMEN

Hepatic insulin resistance is a major risk factor for the development of type 2 diabetes and is associated with the accumulation of lipids, including diacylglycerol (DAG), triacylglycerols (TAG) and ceramide. There is evidence that enzymes involved in ceramide or sphingolipid metabolism may have a role in regulating concentrations of glycerolipids such as DAG and TAG. Here we have investigated the role of sphingosine kinase (SphK) in regulating hepatic lipid levels. We show that mice on a high-fat high-sucrose diet (HFHS) displayed glucose intolerance, elevated liver TAG and DAG, and a reduction in total hepatic SphK activity. Reduced SphK activity correlated with downregulation of SphK1, but not SphK2 expression, and was not associated with altered ceramide levels. The role of SphK1 was further investigated by overexpressing this isoform in the liver of mice in vivo. On a low-fat diet (LFD) mice overexpressing liver SphK1, displayed reduced hepatic TAG synthesis and total TAG levels, but with no change to DAG or ceramide. These mice also exhibited no change in gluconeogenesis, glycogenolysis or glucose tolerance. Similarly, overexpression of SphK1 had no effect on the pattern of endogenous glucose production determined during a glucose tolerance test. Under HFHS conditions, normalization of liver SphK activity to levels observed in LFD controls did not alter hepatic TAG concentrations. Furthermore, DAG, ceramide and glucose tolerance were also unaffected. In conclusion, our data suggest that SphK1 plays an important role in regulating TAG metabolism under LFD conditions.


Asunto(s)
Dieta con Restricción de Grasas , Dieta Alta en Grasa , Grasas de la Dieta/metabolismo , Hígado/enzimología , Fosfotransferasas (Aceptor de Grupo Alcohol)/biosíntesis , Triglicéridos/metabolismo , Animales , Ceramidas/metabolismo , Sacarosa en la Dieta/metabolismo , Glucosa/metabolismo , Homeostasis , Masculino , Ratones Endogámicos C57BL , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , ARN Mensajero/biosíntesis , Factores de Tiempo , Regulación hacia Arriba
12.
Mamm Genome ; 25(9-10): 522-38, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25074441

RESUMEN

Obesity and type 2 diabetes lessen the quality of life of those afflicted and place considerable burden on the healthcare system. Furthermore, the detrimental impact of these pathologies is expected to persist or even worsen. Diabetes is characterized by impaired insulin action and glucose homeostasis. This has led to a rapid increase in the number of mouse models of metabolic disease being used in the basic sciences to assist in facilitating a greater understanding of the metabolic dysregulation associated with obesity and diabetes, the identification of therapeutic targets, and the discovery of effective treatments. This review briefly describes the most frequently utilized models of metabolic disease. A presentation of standard methods and technologies on the horizon for assessing metabolic phenotypes in mice, with particular emphasis on glucose handling and energy balance, is provided. The article also addresses issues related to study design, selection and execution of metabolic tests of glucose metabolism, the presentation of data, and interpretation of results.


Asunto(s)
Glucosa/metabolismo , Homeostasis , Animales , Estado de Conciencia , Metabolismo Energético , Glucosa/química , Resistencia a la Insulina/genética , Enfermedades Metabólicas/diagnóstico , Enfermedades Metabólicas/genética , Enfermedades Metabólicas/metabolismo , Metaboloma , Metabolómica/métodos , Ratones , Ratones Transgénicos , Modelos Animales , Mutación , Fenotipo
13.
Diabetes ; 63(6): 1881-94, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24430435

RESUMEN

Induction of heat shock protein (HSP)72 protects against obesity-induced insulin resistance, but the underlying mechanisms are unknown. Here, we show that HSP72 plays a pivotal role in increasing skeletal muscle mitochondrial number and oxidative metabolism. Mice overexpressing HSP72 in skeletal muscle (HSP72Tg) and control wild-type (WT) mice were fed either a chow or high-fat diet (HFD). Despite a similar energy intake when HSP72Tg mice were compared with WT mice, the HFD increased body weight, intramuscular lipid accumulation (triacylglycerol and diacylglycerol but not ceramide), and severe glucose intolerance in WT mice alone. Whole-body VO2, fatty acid oxidation, and endurance running capacity were markedly increased in HSP72Tg mice. Moreover, HSP72Tg mice exhibited an increase in mitochondrial number. In addition, the HSP72 coinducer BGP-15, currently in human clinical trials for type 2 diabetes, also increased mitochondrial number and insulin sensitivity in a rat model of type 2 diabetes. Together, these data identify a novel role for activation of HSP72 in skeletal muscle. Thus, the increased oxidative metabolism associated with activation of HSP72 has potential clinical implications not only for type 2 diabetes but also for other disorders where mitochondrial function is compromised.


Asunto(s)
Respiración de la Célula , Diabetes Mellitus Tipo 2/metabolismo , Proteínas del Choque Térmico HSP72/metabolismo , Resistencia a la Insulina , Mitocondrias Musculares/metabolismo , Obesidad/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Glucemia , Western Blotting , Peso Corporal , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/fisiopatología , Dieta Alta en Grasa , Metabolismo Energético , Ácidos Grasos/metabolismo , Leptina/metabolismo , Masculino , Ratones , Músculo Esquelético/metabolismo , Obesidad/genética , Obesidad/fisiopatología , Oxidación-Reducción , Fosforilación Oxidativa , Receptores Activados del Proliferador del Peroxisoma/metabolismo , Ratas , Reacción en Cadena en Tiempo Real de la Polimerasa , Sirtuina 1/metabolismo
14.
Endocrinology ; 154(1): 65-76, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23183172

RESUMEN

FTY720 is a sphingosine-1-phosphate analog that has been shown to inhibit ceramide synthesis in vitro. Because ceramide accumulation in muscle is associated with insulin resistance, we aimed to examine whether FTY720 would prevent muscle ceramide accumulation in high fat-fed mice and subsequently improve glucose homeostasis. Male C57Bl/6 mice were fed either a chow or high fat-diet (HFD) for 6 wk, after which they were treated with vehicle or FTY720 (5 mg/kg) daily for a further 6 wk. The ceramide content of muscle was examined and insulin action was assessed. Whereas the HFD increased muscle ceramide, this was prevented by FTY720 treatment. This was not associated with alterations in the expression of genes involved in sphingolipid metabolism. Interestingly, the effects of FTY720 on lipid metabolism were not limited to ceramide because FTY720 also prevented the HFD-induced increase in diacylglycerol and triacylglycerol in muscle. Furthermore, the increase in CD36 mRNA expression induced by fat feeding was prevented in muscle of FTY720-treated mice. This was associated with an attenuation of the HFD-induced increase in palmitate uptake and esterification. In addition, FTY720 improved glucose homeostasis as demonstrated by a reduction in plasma insulin, an improvement in whole-body glucose tolerance, an increase in insulin-stimulated glucose uptake, and Akt phosphorylation in muscle. In conclusion, FTY720 exerts beneficial effects on muscle lipid metabolism that prevent lipid accumulation and improve glucose tolerance in high fat-fed mice. Thus, FTY720 and other compounds that target sphingosine-1-phosphate signaling may have therapeutic potential in treating insulin resistance.


Asunto(s)
Ceramidas/metabolismo , Grasas de la Dieta/efectos adversos , Glucosa/metabolismo , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Glicoles de Propileno/uso terapéutico , Esfingosina/análogos & derivados , Animales , Clorhidrato de Fingolimod , Resistencia a la Insulina/fisiología , Metabolismo de los Lípidos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/tratamiento farmacológico , Obesidad/metabolismo , Esfingosina/uso terapéutico
15.
Cell ; 151(2): 414-26, 2012 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-23063129

RESUMEN

Diabetes, obesity, and cancer affect upward of 15% of the world's population. Interestingly, all three diseases juxtapose dysregulated intracellular signaling with altered metabolic state. Exactly which genetic factors define stable metabolic set points in vivo remains poorly understood. Here, we show that hedgehog signaling rewires cellular metabolism. We identify a cilium-dependent Smo-Ca(2+)-Ampk axis that triggers rapid Warburg-like metabolic reprogramming within minutes of activation and is required for proper metabolic selectivity and flexibility. We show that Smo modulators can uncouple the Smo-Ampk axis from canonical signaling and identify cyclopamine as one of a new class of "selective partial agonists," capable of concomitant inhibition of canonical and activation of noncanonical hedgehog signaling. Intriguingly, activation of the Smo-Ampk axis in vivo drives robust insulin-independent glucose uptake in muscle and brown adipose tissue. These data identify multiple noncanonical endpoints that are pivotal for rational design of hedgehog modulators and provide a new therapeutic avenue for obesity and diabetes.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Glucólisis , Proteínas Hedgehog/metabolismo , Células Musculares/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Quinasas de la Proteína-Quinasa Activada por el AMP , Adipocitos/metabolismo , Animales , Línea Celular , Células Cultivadas , Cilios/metabolismo , Diabetes Mellitus/metabolismo , Humanos , Ratones , Neoplasias/metabolismo , Obesidad/metabolismo , Proteínas Quinasas/metabolismo , Receptor Smoothened
16.
Diabetes ; 61(12): 3148-55, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22961081

RESUMEN

The sphingolipids sphingosine-1-phosphate (S1P) and ceramide are important bioactive lipids with many cellular effects. Intracellular ceramide accumulation causes insulin resistance, but sphingosine kinase 1 (SphK1) prevents ceramide accumulation, in part, by promoting its metabolism into S1P. Despite this, the role of SphK1 in regulating insulin action has been largely overlooked. Transgenic (Tg) mice that overexpress SphK1 were fed a standard chow or high-fat diet (HFD) for 6 weeks before undergoing several metabolic analyses. SphK1 Tg mice fed an HFD displayed increased SphK activity in skeletal muscle, which was associated with an attenuated intramuscular ceramide accumulation compared with wild-type (WT) littermates. This was associated with a concomitant reduction in the phosphorylation of c-jun amino-terminal kinase, a serine threonine kinase associated with insulin resistance. Accordingly, skeletal muscle and whole-body insulin sensitivity were improved in SphK1 Tg, compared with WT mice, when fed an HFD. We have identified that the enzyme SphK1 is an important regulator of lipid partitioning and insulin action in skeletal muscle under conditions of increased lipid supply.


Asunto(s)
Ceramidas/metabolismo , Dieta Alta en Grasa/efectos adversos , Resistencia a la Insulina/fisiología , Músculo Esquelético/enzimología , Músculo Esquelético/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Animales , Western Blotting , Resistencia a la Insulina/genética , Ratones , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Reacción en Cadena de la Polimerasa
17.
J Appl Physiol (1985) ; 113(8): 1173-83, 2012 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-22653994

RESUMEN

The objective of this study was to test the hypothesis that exercise-stimulated muscle glucose uptake (MGU) is augmented by increasing mitochondrial reactive oxygen species (mtROS) scavenging capacity. This hypothesis was tested in genetically altered mice fed chow or a high-fat (HF) diet that accelerates mtROS formation. Mice overexpressing SOD2 (sod2(Tg)), mitochondria-targeted catalase (mcat(Tg)), and combined SOD2 and mCAT (mtAO) were used to increase mtROS scavenging. mtROS was assessed by the H(2)O(2) emitting potential (JH(2)O(2)) in muscle fibers. sod2(Tg) did not decrease JH(2)O(2) in chow-fed mice, but decreased JH(2)O(2) in HF-fed mice. mcat(Tg) and mtAO decreased JH(2)O(2) in both chow- and HF-fed mice. In parallel, the ratio of reduced to oxidized glutathione (GSH/GSSG) was unaltered in sod2(Tg) in chow-fed mice, but was increased in HF-fed sod2(Tg) and both chow- and HF-fed mcat(Tg) and mtAO. Nitrotyrosine, a marker of NO-dependent, reactive nitrogen species (RNS)-induced nitrative stress, was decreased in both chow- and HF-fed sod2(Tg), mcat(Tg), and mtAO mice. This effect was not changed with exercise. Kg, an index of MGU was assessed using 2-[(14)C]-deoxyglucose during exercise. In chow-fed mice, sod2(Tg), mcat(Tg), and mtAO increased exercise Kg compared with wild types. Exercise Kg was also augmented in HF-fed sod2(Tg) and mcat(Tg) mice but unchanged in HF-fed mtAO mice. In conclusion, mtROS scavenging is a key regulator of exercise-mediated MGU and this regulation depends on nutritional state.


Asunto(s)
Antioxidantes/metabolismo , Glucosa/metabolismo , Mitocondrias/metabolismo , Músculo Esquelético/metabolismo , Potenciales de Acción/fisiología , Alimentación Animal , Animales , Transporte Biológico/fisiología , Catalasa/metabolismo , Dieta Alta en Grasa , Disulfuro de Glutatión/metabolismo , Peróxido de Hidrógeno/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos/metabolismo , Óxido Nítrico/metabolismo , Condicionamiento Físico Animal/fisiología , Especies de Nitrógeno Reactivo/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Superóxido Dismutasa/metabolismo , Tirosina/análogos & derivados , Tirosina/metabolismo
18.
Am J Physiol Endocrinol Metab ; 303(3): E301-7, 2012 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-22550064

RESUMEN

Nitric oxide (NO) is an important vasodilator and regulator in the cardiovascular system, and this link was the subject of a Nobel prize in 1998. However, NO also plays many other regulatory roles, including thrombosis, immune function, neural activity, and gastrointestinal function. Low concentrations of NO are thought to have important signaling effects. In contrast, high concentrations of NO can interact with reactive oxygen species, causing damage to cells and cellular components. A less-recognized site of NO production is within skeletal muscle, where small increases are thought to have beneficial effects such as regulating glucose uptake and possibly blood flow, but higher levels of production are thought to lead to deleterious effects such as an association with insulin resistance. This review will discuss the role of NO in skeletal muscle during and following exercise, including in mitochondrial biogenesis, muscle efficiency, and blood flow with a particular focus on its potential role in regulating skeletal muscle glucose uptake during exercise.


Asunto(s)
Ejercicio Físico/fisiología , Glucosa/metabolismo , Músculo Esquelético/metabolismo , Óxido Nítrico/metabolismo , Animales , Humanos , Modelos Biológicos , Contracción Muscular/fisiología , Músculo Esquelético/química , Óxido Nítrico Sintasa/metabolismo , Transducción de Señal/fisiología
19.
Diabetes ; 60(11): 2720-9, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21885872

RESUMEN

OBJECTIVE: Exercise is an effective intervention to treat fatty liver. However, the mechanism(s) that underlie exercise-induced reductions in fatty liver are unclear. Here we tested the hypothesis that exercise requires hepatic glucagon action to reduce fatty liver. RESEARCH DESIGN AND METHODS: C57BL/6 mice were fed high-fat diet (HFD) and assessed using magnetic resonance, biochemical, and histological techniques to establish a timeline for fatty liver development over 20 weeks. Glucagon receptor null (gcgr(-/-)) and wild-type (gcgr(+/+)) littermate mice were subsequently fed HFD to provoke moderate fatty liver and then performed either 10 or 6 weeks of running wheel or treadmill exercise, respectively. RESULTS: Exercise reverses progression of HFD-induced fatty liver in gcgr(+/+) mice. Remarkably, such changes are absent in gcgr(-/-) mice, thus confirming the hypothesis that exercise-stimulated hepatic glucagon receptor activation is critical to reduce HFD-induced fatty liver. CONCLUSIONS: These findings suggest that therapies that use antagonism of hepatic glucagon action to reduce blood glucose may interfere with the ability of exercise and perhaps other interventions to positively affect fatty liver.


Asunto(s)
Hígado Graso/metabolismo , Hígado Graso/terapia , Glucagón/metabolismo , Hígado/metabolismo , Actividad Motora , Receptores de Glucagón/metabolismo , Animales , Peso Corporal , Grasas de la Dieta/efectos adversos , Progresión de la Enfermedad , Hígado Graso/patología , Metabolismo de los Lípidos , Hígado/patología , Imagen por Resonancia Magnética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Glucagón/genética , Transducción de Señal
20.
Diabetes ; 60(2): 416-26, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21270253

RESUMEN

OBJECTIVE: The hypothesis that high-fat (HF) feeding causes skeletal muscle extracellular matrix (ECM) remodeling in C57BL/6J mice and that this remodeling contributes to diet-induced muscle insulin resistance (IR) through the collagen receptor integrin α(2)ß(1) was tested. RESEARCH DESIGN AND METHODS: The association between IR and ECM remodeling was studied in mice fed chow or HF diet. Specific genetic and pharmacological murine models were used to study effects of HF feeding on ECM in the absence of IR. The role of ECM-integrin interaction in IR was studied using hyperinsulinemic-euglycemic clamps on integrin α(2)ß(1)-null (itga2(-/-)), integrin α(1)ß(1)-null (itga1(-/-)), and wild-type littermate mice fed chow or HF. Integrin α(2)ß(1) and integrin α(1)ß(1) signaling pathways have opposing actions. RESULTS: HF-fed mice had IR and increased muscle collagen (Col) III and ColIV protein; the former was associated with increased transcript, whereas the latter was associated with reduced matrix metalloproteinase 9 activity. Rescue of muscle IR by genetic muscle-specific mitochondria-targeted catalase overexpression or by the phosphodiesterase 5a inhibitor, sildenafil, reversed HF feeding effects on ECM remodeling and increased muscle vascularity. Collagen remained elevated in HF-fed itga2(-/-) mice. Nevertheless, muscle insulin action and vascularity were increased. Muscle IR in HF-fed itga1(-/-) mice was unchanged. Insulin sensitivity in chow-fed itga1(-/-) and itga2(-/-) mice was not different from wild-type littermates. CONCLUSIONS: ECM collagen expansion is tightly associated with muscle IR. Studies with itga2(-/-) mice provide mechanistic insight for this association by showing that the link between muscle IR and increased collagen can be uncoupled by the absence of collagen-integrin α(2)ß(1) interaction.


Asunto(s)
Matriz Extracelular/metabolismo , Resistencia a la Insulina/fisiología , Insulina/metabolismo , Integrina alfa2beta1/metabolismo , Músculo Esquelético/metabolismo , Análisis de Varianza , Animales , Western Blotting , Colágeno Tipo III/metabolismo , Colágeno Tipo V/metabolismo , Dieta , Matriz Extracelular/efectos de los fármacos , Técnica de Clampeo de la Glucosa , Inmunohistoquímica , Integrina alfa2beta1/genética , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Noqueados , Músculo Esquelético/efectos de los fármacos , Inhibidores de Fosfodiesterasa 5/farmacología , Piperazinas/farmacología , Purinas/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Citrato de Sildenafil , Sulfonas/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...