Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
Cell Metab ; 35(8): 1327-1340.e5, 2023 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-37473755

RESUMEN

Growth differentiation factor 15 (GDF15) induces weight loss and increases insulin action in obese rodents. Whether and how GDF15 improves insulin action without weight loss is unknown. Obese rats were treated with GDF15 and displayed increased insulin tolerance 5 h later. Lean and obese female and male mice were treated with GDF15 on days 1, 3, and 5 without weight loss and displayed increased insulin sensitivity during a euglycemic hyperinsulinemic clamp on day 6 due to enhanced suppression of endogenous glucose production and increased glucose uptake in WAT and BAT. GDF15 also reduced glucagon levels during clamp independently of the GFRAL receptor. The insulin-sensitizing effect of GDF15 was completely abrogated in GFRAL KO mice and also by treatment with the ß-adrenergic antagonist propranolol and in ß1,ß2-adrenergic receptor KO mice. GDF15 activation of the GFRAL receptor increases ß-adrenergic signaling, in turn, improving insulin action in the liver and white and brown adipose tissue.


Asunto(s)
Resistencia a la Insulina , Receptores Adrenérgicos beta , Ratones , Ratas , Masculino , Femenino , Animales , Factor 15 de Diferenciación de Crecimiento/farmacología , Obesidad , Tejido Adiposo , Pérdida de Peso , Insulina , Tejido Adiposo Pardo , Hígado
2.
Nature ; 619(7968): 143-150, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37380764

RESUMEN

Caloric restriction that promotes weight loss is an effective strategy for treating non-alcoholic fatty liver disease and improving insulin sensitivity in people with type 2 diabetes1. Despite its effectiveness, in most individuals, weight loss is usually not maintained partly due to physiological adaptations that suppress energy expenditure, a process known as adaptive thermogenesis, the mechanistic underpinnings of which are unclear2,3. Treatment of rodents fed a high-fat diet with recombinant growth differentiating factor 15 (GDF15) reduces obesity and improves glycaemic control through glial-cell-derived neurotrophic factor family receptor α-like (GFRAL)-dependent suppression of food intake4-7. Here we find that, in addition to suppressing appetite, GDF15 counteracts compensatory reductions in energy expenditure, eliciting greater weight loss and reductions in non-alcoholic fatty liver disease (NAFLD) compared to caloric restriction alone. This effect of GDF15 to maintain energy expenditure during calorie restriction requires a GFRAL-ß-adrenergic-dependent signalling axis that increases fatty acid oxidation and calcium futile cycling in the skeletal muscle of mice. These data indicate that therapeutic targeting of the GDF15-GFRAL pathway may be useful for maintaining energy expenditure in skeletal muscle during caloric restriction.


Asunto(s)
Metabolismo Energético , Factor 15 de Diferenciación de Crecimiento , Músculo Esquelético , Pérdida de Peso , Animales , Humanos , Ratones , Depresores del Apetito/metabolismo , Depresores del Apetito/farmacología , Depresores del Apetito/uso terapéutico , Restricción Calórica , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/metabolismo , Dieta Alta en Grasa , Ingestión de Alimentos/efectos de los fármacos , Metabolismo Energético/efectos de los fármacos , Factor 15 de Diferenciación de Crecimiento/metabolismo , Factor 15 de Diferenciación de Crecimiento/farmacología , Factor 15 de Diferenciación de Crecimiento/uso terapéutico , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Enfermedad del Hígado Graso no Alcohólico/complicaciones , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/terapia , Receptores Adrenérgicos beta/metabolismo , Pérdida de Peso/efectos de los fármacos
3.
Nat Metab ; 4(2): 157-158, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35177852
4.
Am J Physiol Endocrinol Metab ; 321(4): E443-E452, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34370594

RESUMEN

Growth differentiating factor 15 (GDF15) is expressed in the intestine and is one of the most recently identified satiety peptides. The mechanisms controlling its secretion are unclear. The present study investigated whether plasma GDF15 concentrations are meal-related and if potential responses depend on macronutrient type or are affected by previous bariatric surgery. The study included 1) volunteers ingesting rapidly vs. slowly digested carbohydrates (sucrose vs. isomaltose; n = 10), 2) volunteers who had undergone Roux-en-Y gastric bypass (RYGB) or sleeve gastrectomy (SG) surgery and unoperated matched controls ingesting a liquid mixed meal (n = 9-10 in each group), and 3) individuals with previous RYGB compared with unoperated controls ingesting isocaloric glucose, fat, or protein (n = 6 in each group). Plasma was collected after an overnight fast and up to 6 h after ingestion (≥12 time points). In cohort 1, fasting GDF15 concentrations were ∼480 pg/mL. Concentrations after sucrose or isomaltose intake did not differ from baseline (P = 0.26 to P > 0.99) and total area under the curves (tAUCs were similar between groups (P = 0.77). In cohort 2, fasting GDF15 concentrations were as follows (pg/mL): RYGB = 540 ± 41.4, SG = 477 ± 36.4, and controls = 590 ± 41.8, with no between-group differences (P = 0.73). Concentrations did not increase at any postprandial time point (over all time factor: P = 0.10) and tAUCs were similar between groups (P = 0.73). In cohort 3, fasting plasma GDF15 was similar among the groups (P > 0.99) and neither glucose, fat, nor protein intake consistently increased the concentrations. In conclusion, we find that plasma GDF15 was not stimulated by meal intake and that fasting concentrations did not differ between RYGB-, SG-, and body mass index (BMI)-matched controls when investigated during the weight stable phase after RYGB and SG.NEW & NOTEWORTHY Our combined data show that GDF15 does not increase in response to a liquid meal. Moreover, we show for the first time that ingestion of sucrose, isomaltose, glucose, fat, or protein also does not increase plasma GDF15 concentrations, questioning the role of GDF15 in regulation of food source preference. Finally, we find that neither fasting nor postprandial plasma GDF15 concentrations are increased in individuals with previous bariatric surgery compared with unoperated body mass index (BMI)-matched controls.


Asunto(s)
Cirugía Bariátrica/métodos , Biomarcadores/sangre , Tracto Gastrointestinal/metabolismo , Factor 15 de Diferenciación de Crecimiento/sangre , Comidas , Obesidad Mórbida/sangre , Adulto , Glucemia/análisis , Índice de Masa Corporal , Estudios de Casos y Controles , Estudios Cruzados , Femenino , Estudios de Seguimiento , Humanos , Insulina/sangre , Masculino , Persona de Mediana Edad , Obesidad Mórbida/patología , Obesidad Mórbida/cirugía , Periodo Posprandial , Pronóstico , Ensayos Clínicos Controlados Aleatorios como Asunto , Pérdida de Peso
5.
Nat Rev Endocrinol ; 17(10): 592-607, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34381196

RESUMEN

Growth differentiation factor 15 (GDF15) is a member of the TGFß superfamily whose expression is increased in response to cellular stress and disease as well as by metformin. Elevations in GDF15 reduce food intake and body mass in animal models through binding to glial cell-derived neurotrophic factor family receptor alpha-like (GFRAL) and the recruitment of the receptor tyrosine kinase RET in the hindbrain. This effect is largely independent of other appetite-regulating hormones (for example, leptin, ghrelin or glucagon-like peptide 1). Consistent with an important role for the GDF15-GFRAL signalling axis, some human genetic studies support an interrelationship with human obesity. Furthermore, findings in both mice and humans have shown that metformin and exercise increase circulating levels of GDF15. GDF15 might also exert anti-inflammatory effects through mechanisms that are not fully understood. These unique and distinct mechanisms for suppressing food intake and inflammation makes GDF15 an appealing candidate to treat many metabolic diseases, including obesity, type 2 diabetes mellitus, non-alcoholic fatty liver disease, cardiovascular disease and cancer cachexia. Here, we review the mechanisms regulating GDF15 production and secretion, GDF15 signalling in different cell types, and how GDF15-targeted pharmaceutical approaches might be effective in the treatment of metabolic diseases.


Asunto(s)
Enfermedades Cardiovasculares/tratamiento farmacológico , Factor 15 de Diferenciación de Crecimiento/antagonistas & inhibidores , Factor 15 de Diferenciación de Crecimiento/metabolismo , Enfermedades Metabólicas/tratamiento farmacológico , Terapia Molecular Dirigida , Obesidad/tratamiento farmacológico , Animales , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Humanos , Metformina/farmacología , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico
6.
Proc Natl Acad Sci U S A ; 118(27)2021 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-34187898

RESUMEN

An acute increase in the circulating concentration of glucocorticoid hormones is essential for the survival of severe somatic stresses. Circulating concentrations of GDF15, a hormone that acts in the brain to reduce food intake, are frequently elevated in stressful states. We now report that GDF15 potently activates the hypothalamic-pituitary-adrenal (HPA) axis in mice and rats. A blocking antibody to the GDNF-family receptor α-like receptor completely prevented the corticosterone response to GDF15 administration. In wild-type mice exposed to a range of stressful stimuli, circulating levels of both corticosterone and GDF15 rose acutely. In the case of Escherichia coli or lipopolysaccharide injections, the vigorous proinflammatory cytokine response elicited was sufficient to produce a near-maximal HPA response, regardless of the presence or absence of GDF15. In contrast, the activation of the HPA axis seen in wild-type mice in response to the administration of genotoxic or endoplasmic reticulum toxins, which do not provoke a marked rise in cytokines, was absent in Gdf15-/- mice. In conclusion, consistent with its proposed role as a sentinel hormone, endogenous GDF15 is required for the activation of the protective HPA response to toxins that do not induce a substantial cytokine response. In the context of efforts to develop GDF15 as an antiobesity therapeutic, these findings identify a biomarker of target engagement and a previously unrecognized pharmacodynamic effect, which will require monitoring in human studies.


Asunto(s)
Factor 15 de Diferenciación de Crecimiento/metabolismo , Sistema Hipotálamo-Hipofisario/metabolismo , Sistema Hipófiso-Suprarrenal/metabolismo , Animales , Cisplatino/administración & dosificación , Cisplatino/farmacología , Estrés del Retículo Endoplásmico/efectos de los fármacos , Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Glucocorticoides/metabolismo , Factor 15 de Diferenciación de Crecimiento/administración & dosificación , Humanos , Lipopolisacáridos , Ratones , Ratas , Tunicamicina/farmacología
7.
Proc Natl Acad Sci U S A ; 118(8)2021 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-33593916

RESUMEN

The TGFß cytokine family member, GDF-15, reduces food intake and body weight and represents a potential treatment for obesity. Because the brainstem-restricted expression pattern of its receptor, GDNF Family Receptor α-like (GFRAL), presents an exciting opportunity to understand mechanisms of action for area postrema neurons in food intake; we generated GfralCre and conditional GfralCreERT mice to visualize and manipulate GFRAL neurons. We found infection or pathophysiologic states (rather than meal ingestion) stimulate GFRAL neurons. TRAP-Seq analysis of GFRAL neurons revealed their expression of a wide range of neurotransmitters and neuropeptides. Artificially activating GfralCre -expressing neurons inhibited feeding, decreased gastric emptying, and promoted a conditioned taste aversion (CTA). GFRAL neurons most strongly innervate the parabrachial nucleus (PBN), where they target CGRP-expressing (CGRPPBN) neurons. Silencing CGRPPBN neurons abrogated the aversive and anorexic effects of GDF-15. These findings suggest that GFRAL neurons link non-meal-associated pathophysiologic signals to suppress nutrient uptake and absorption.


Asunto(s)
Reacción de Prevención/efectos de los fármacos , Ingestión de Alimentos/efectos de los fármacos , Conducta Alimentaria/efectos de los fármacos , Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Factor 15 de Diferenciación de Crecimiento/farmacología , Neuronas/fisiología , Núcleos Parabraquiales/fisiología , Animales , Peso Corporal , Femenino , Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Masculino , Ratones , Neuronas/efectos de los fármacos , Núcleos Parabraquiales/efectos de los fármacos , Ratas , Ratas Long-Evans
8.
Nat Commun ; 12(1): 1041, 2021 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-33589633

RESUMEN

Growing evidence supports that pharmacological application of growth differentiation factor 15 (GDF15) suppresses appetite but also promotes sickness-like behaviors in rodents via GDNF family receptor α-like (GFRAL)-dependent mechanisms. Conversely, the endogenous regulation of GDF15 and its physiological effects on energy homeostasis and behavior remain elusive. Here we show, in four independent human studies that prolonged endurance exercise increases circulating GDF15 to levels otherwise only observed in pathophysiological conditions. This exercise-induced increase can be recapitulated in mice and is accompanied by increased Gdf15 expression in the liver, skeletal muscle, and heart muscle. However, whereas pharmacological GDF15 inhibits appetite and suppresses voluntary running activity via GFRAL, the physiological induction of GDF15 by exercise does not. In summary, exercise-induced circulating GDF15 correlates with the duration of endurance exercise. Yet, higher GDF15 levels after exercise are not sufficient to evoke canonical pharmacological GDF15 effects on appetite or responsible for diminishing exercise motivation.


Asunto(s)
Regulación del Apetito/fisiología , Ejercicio Físico/fisiología , Conducta Alimentaria/fisiología , Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Factor 15 de Diferenciación de Crecimiento/genética , Resistencia Física/fisiología , Adulto , Animales , Creatina Quinasa/sangre , Creatina Quinasa/genética , Regulación de la Expresión Génica , Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial/deficiencia , Factor 15 de Diferenciación de Crecimiento/sangre , Factor 15 de Diferenciación de Crecimiento/metabolismo , Humanos , Interleucina-10/sangre , Interleucina-10/genética , Interleucina-6/administración & dosificación , Leptina/sangre , Leptina/genética , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones , Ratones Noqueados , Motivación/fisiología , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Miocardio/metabolismo , Condicionamiento Físico Animal , Factores de Tiempo
9.
Mol Metab ; 21: 13-21, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30685336

RESUMEN

OBJECTIVE: Analogues of GDF15 (Growth Differentiation Factor 15) are promising new anti-obesity therapies as pharmacological treatment with GDF15 results in dramatic reductions of food intake and body weight. GDF15 exerts its central anorexic effects by binding to the GFRAL receptor exclusively expressed in the Area Postrema (AP) and the Nucleus of the Solitary Tract (NTS) of the hindbrain. We sought to determine if GDF15 is an indispensable factor for other interventions that cause weight loss and which are also known to act via these hindbrain regions. METHODS: To explore the role of GDF15 on food choice we performed macronutrient intake studies in mice treated pharmacologically with GDF15 and in mice having either GDF15 or GFRAL deleted. Next we performed vertical sleeve gastrectomy (VSG) surgeries in a cohort of diet-induced obese Gdf15-null and control mice. To explore the anatomical co-localization of neurons in the hindbrain responding to GLP-1 and/or GDF15 we used GLP-1R reporter mice treated with GDF15, as well as naïve mouse brain and human brain stained by ISH and IHC, respectively, for GLP-1R and GFRAL. Lastly we performed a series of food intake experiments where we treated mice with targeted genetic disruption of either Gdf15 or Gfral with liraglutide; Glp1r-null mice with GDF15; or combined liraglutide and GDF15 treatment in wild-type mice. RESULTS: We found that GDF15 treatment significantly lowered the preference for fat intake in mice, whereas no changes in fat intake were observed after genetic deletion of Gdf15 or Gfral. In addition, deletion of Gdf15 did not alter the food intake or bodyweight after sleeve gastrectomy. Lack of GDF15 or GFRAL signaling did not alter the ability of the GLP-1R agonist liraglutide to reduce food intake. Similarly lack of GLP-1R signaling did not reduce GDF15's anorexic effect. Interestingly, there was a significant synergistic effect on weight loss when treating wild-type mice with both GDF15 and liraglutide. CONCLUSION: These data suggest that while GDF15 does not play a role in the potent effects of VSG in mice there seems to be a potential therapeutic benefit of activating GFRAL and GLP-1R systems simultaneously.


Asunto(s)
Cirugía Bariátrica , Factor 15 de Diferenciación de Crecimiento/metabolismo , Factor 15 de Diferenciación de Crecimiento/uso terapéutico , Hipoglucemiantes/uso terapéutico , Liraglutida/uso terapéutico , Obesidad/tratamiento farmacológico , Pérdida de Peso/efectos de los fármacos , Animales , Área Postrema/metabolismo , Peso Corporal/efectos de los fármacos , Dieta Alta en Grasa/efectos adversos , Sinergismo Farmacológico , Ingestión de Alimentos/efectos de los fármacos , Gastrectomía , Eliminación de Gen , Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Factor 15 de Diferenciación de Crecimiento/genética , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/etiología , Núcleo Solitario/metabolismo
10.
Nat Med ; 23(10): 1158-1166, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28846099

RESUMEN

Growth differentiation factor 15 (GDF15; also known as MIC-1) is a divergent member of the TGF-ß superfamily and is associated with body-weight regulation in humans and rodents. However, the cognate receptor of GDF15 is unknown. Here we show that GDF15 binds specifically to GDNF family receptor α-like (GFRAL) with high affinity, and that GFRAL requires association with the coreceptor RET to elicit intracellular signaling in response to GDF15 stimulation. We also found that GDF15-mediated reductions in food intake and body weight of mice with obesity were abolished in GFRAL-knockout mice. We further found that GFRAL expression was limited to hindbrain neurons and not present in peripheral tissues, which suggests that GDF15-GFRAL-mediated regulation of food intake is by a central mechanism. Lastly, given that GDF15 did not increase energy expenditure in treated mice with obesity, the anti-obesity actions of the cytokine are likely driven primarily by a reduction in food intake.


Asunto(s)
Ingestión de Alimentos/efectos de los fármacos , Metabolismo Energético/efectos de los fármacos , Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial/efectos de los fármacos , Factor 15 de Diferenciación de Crecimiento/farmacología , Obesidad/metabolismo , Pérdida de Peso/efectos de los fármacos , Animales , Ingestión de Alimentos/genética , Metabolismo Energético/genética , Citometría de Flujo , Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Células HEK293 , Humanos , Técnicas In Vitro , Ratones , Ratones Noqueados , Ratas , Ratas Sprague-Dawley , Resonancia por Plasmón de Superficie , Pérdida de Peso/genética
11.
J Steroid Biochem Mol Biol ; 170: 39-48, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-27353462

RESUMEN

We investigated the effects of estrogens on glucose homeostasis using the Aromatase Knockout (ArKO) mouse, which is unable to convert androgens into estrogens. The ArKO mouse is a model of total estrogen ablation which develops symptoms of metabolic syndrome. To determine the development and progression of whole body state of insulin resistance of ArKO mice, comprehensive whole body tolerance tests were performed on WT, ArKO and estrogen administrated mice at 3 and 12 months of age. The absence of estrogens in the male ArKO mice leads to hepatic insulin resistance, glucose and pyruvate intolerance from 3 to 12 months with consistent improvement upon estrogen treatment. Estrogen absence in the female ArKO mice leads to glucose intolerance without pyruvate intolerance or insulin resistance. The replacement of estrogens in the female WT and ArKO mice exhibited both insulin sensitizing and resistance effects depending on age and dosage. In conclusion, this study presents information on the sexually dimorphic roles of estrogens on glucose homeostasis regulation.


Asunto(s)
Aromatasa/deficiencia , Aromatasa/genética , Estrógenos/metabolismo , Glucosa/metabolismo , Homeostasis , Animales , Aromatasa/metabolismo , Índice de Masa Corporal , Femenino , Prueba de Tolerancia a la Glucosa , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Caracteres Sexuales
12.
PLoS One ; 10(8): e0136143, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26317527

RESUMEN

The maintenance of glucose homeostasis within the body is crucial for constant and precise performance of energy balance and is sustained by a number of peripheral organs. Estrogens are known to play a role in the maintenance of glucose homeostasis. Aromatase knockout (ArKO) mice are estrogen-deficient and display symptoms of dysregulated glucose metabolism. We aim to investigate the effects of estrogen ablation and exogenous estrogen administration on glucose homeostasis regulation. Six month-old female wildtype, ArKO, and 17ß-estradiol (E2) treated ArKO mice were subjected to whole body tolerance tests, serum examination of estrogen, glucose and insulin, ex-vivo muscle glucose uptake, and insulin signaling pathway analyses. Female ArKO mice display increased body weight, gonadal (omental) adiposity, hyperinsulinemia, and liver triglycerides, which were ameliorated upon estrogen treatment. Tolerance tests revealed that estrogen-deficient ArKO mice were pyruvate intolerant hence reflecting dysregulated hepatic gluconeogenesis. Analyses of skeletal muscle, liver, and adipose tissues supported a hepatic-based glucose dysregulation, with a down-regulation of Akt phosphorylation (a key insulin signaling pathway molecule) in the ArKO liver, which was improved with E2 treatment. Concurrently, estrogen treatment lowered ArKO serum leptin and adiponectin levels and increased inflammatory adipokines such as tumour necrosis factor alpha (TNFα) and interleukin 6 (IL6). Furthermore, estrogen deficiency resulted in the infiltration of CD45 macrophages into gonadal adipose tissues, which cannot be reversed by E2 treatment. This study describes the effects of estrogens on glucose homeostasis in female ArKO mice and highlights a primary phenotype of hepatic glucose dysregulation and a parallel estrogen modified adipokine profile.


Asunto(s)
Adipoquinas/sangre , Aromatasa/genética , Estradiol/sangre , Estrógenos/sangre , Gluconeogénesis , Glucosa/metabolismo , Homeostasis/efectos de los fármacos , Tejido Adiposo/metabolismo , Animales , Estradiol/farmacología , Estrógenos/farmacología , Femenino , Interleucina-6/sangre , Leptina/sangre , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Músculo Esquelético/metabolismo , Triglicéridos/metabolismo , Factor de Necrosis Tumoral alfa/sangre
13.
Am J Physiol Endocrinol Metab ; 309(2): E142-53, 2015 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-25968579

RESUMEN

Members of the IL-6 family, IL-6 and ciliary neurotrophic factor (CNTF), have been shown to increase glucose uptake and fatty acid oxidation in skeletal muscle. However, the metabolic effects of another family member, leukemia inhibitory factor (LIF), are not well characterized. Effects of LIF on skeletal muscle glucose uptake and palmitate oxidation and signaling were investigated in ex vivo incubated mouse soleus and EDL muscles from muscle-specific AMPKα2 kinase-dead, muscle-specific SOCS3 knockout, and lean and high-fat-fed mice. Inhibitors were used to investigate involvement of specific signaling pathways. LIF increased muscle glucose uptake in dose (50-5,000 pM/l) and time-dependent manners with maximal effects at the 30-min time point. LIF increased Akt Ser(473) phosphorylation (P) in soleus and EDL, whereas AMPK Thr(172) P was unaffected. Incubation with parthenolide abolished LIF-induced glucose uptake and STAT3 Tyr(705) P, whereas incubation with LY-294002 and wortmannin suppressed both basal and LIF-induced glucose uptake and Akt Ser(473) P, indicating that JAK and PI 3-kinase signaling is required for LIF-stimulated glucose uptake. Incubation with rapamycin and AZD8055 indicated that mammalian target of rapamycin complex (mTORC)2, but not mTORC1, also is required for LIF-stimulated glucose uptake. In contrast to CNTF, LIF stimulation did not alter palmitate oxidation. LIF-stimulated glucose uptake was maintained in EDL from obese insulin-resistant mice, whereas soleus developed LIF resistance. Lack of SOCS3 and AMPKα2 did not affect LIF-stimulated glucose uptake. In conclusion, LIF acutely increased muscle glucose uptake by a mechanism potentially involving the PI 3-kinase/mTORC2/Akt pathway and is not impaired in EDL muscle from obese insulin-resistant mice.


Asunto(s)
Glucosa/metabolismo , Factor Inhibidor de Leucemia/farmacología , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Proteínas Recombinantes/farmacología , Animales , Transporte Biológico/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Transducción de Señal/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos
14.
Diabetologia ; 57(8): 1693-702, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24913514

RESUMEN

AIMS/HYPOTHESIS: Obesity is characterised by lipid accumulation in skeletal muscle, which increases the risk of developing insulin resistance and type 2 diabetes. AMP-activated protein kinase (AMPK) is a sensor of cellular energy status and is activated in skeletal muscle by exercise, hormones (leptin, adiponectin, IL-6) and pharmacological agents (5-amino-4-imidazolecarboxamide ribonucleoside [AICAR] and metformin). Phosphorylation of acetyl-CoA carboxylase 2 (ACC2) at S221 (S212 in mice) by AMPK reduces ACC activity and malonyl-CoA content but the importance of the AMPK-ACC2-malonyl-CoA pathway in controlling fatty acid metabolism and insulin sensitivity is not understood; therefore, we characterised Acc2 S212A knock-in (ACC2 KI) mice. METHODS: Whole-body and skeletal muscle fatty acid oxidation and insulin sensitivity were assessed in ACC2 KI mice and wild-type littermates. RESULTS: ACC2 KI mice were resistant to increases in skeletal muscle fatty acid oxidation elicited by AICAR. These mice had normal adiposity and liver lipids but elevated contents of triacylglycerol and ceramide in skeletal muscle, which were associated with hyperinsulinaemia, glucose intolerance and skeletal muscle insulin resistance. CONCLUSIONS/INTERPRETATION: These findings indicate that the phosphorylation of ACC2 S212 is required for the maintenance of skeletal muscle lipid and glucose homeostasis.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Acetil-CoA Carboxilasa/metabolismo , Resistencia a la Insulina/fisiología , Insulina/farmacología , Músculo Esquelético/metabolismo , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacología , Animales , Hipoglucemiantes/farmacología , Leptina/metabolismo , Metabolismo de los Lípidos/efectos de los fármacos , Metabolismo de los Lípidos/fisiología , Malonil Coenzima A/metabolismo , Ratones , Músculo Esquelético/efectos de los fármacos , Obesidad/metabolismo , Oxidación-Reducción , Fosforilación/efectos de los fármacos , Ribonucleótidos/farmacología
15.
PLoS One ; 9(6): e100370, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24971956

RESUMEN

Macrophage inhibitory cytokine-1 (MIC-1/GDF15) modulates food intake and body weight under physiological and pathological conditions by acting on the hypothalamus and brainstem. When overexpressed in disease, such as in advanced cancer, elevated serum MIC-1/GDF15 levels lead to an anorexia/cachexia syndrome. To gain a better understanding of its actions in the brainstem we studied MIC-1/GDF15 induced neuronal activation identified by induction of Fos protein. Intraperitoneal injection of human MIC-1/GDF15 in mice activated brainstem neurons in the area postrema (AP) and the medial (m) portion of the nucleus of the solitary tract (NTS), which did not stain with tyrosine hydroxylase (TH). To determine the importance of these brainstem nuclei in the anorexigenic effect of MIC-1/GDF15, we ablated the AP alone or the AP and the NTS. The latter combined lesion completely reversed the anorexigenic effects of MIC-1/GDF15. Altogether, this study identified neurons in the AP and/or NTS, as being critical for the regulation of food intake and body weight by MIC-1/GDF15.


Asunto(s)
Depresores del Apetito/farmacología , Área Postrema/efectos de los fármacos , Área Postrema/fisiología , Factor 15 de Diferenciación de Crecimiento/farmacología , Núcleo Solitario/efectos de los fármacos , Núcleo Solitario/fisiología , Animales , Anorexia/inducido químicamente , Depresores del Apetito/administración & dosificación , Factor 15 de Diferenciación de Crecimiento/administración & dosificación , Infusiones Intraventriculares , Masculino , Ratones , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Tirosina 3-Monooxigenasa/metabolismo , Pérdida de Peso/efectos de los fármacos
16.
FASEB J ; 28(8): 3384-95, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24732133

RESUMEN

Some gene deletions or mutations have little effect on metabolism and metabolic adaptation because of redundancy and/or compensation in metabolic pathways. The mechanisms for redundancy and/or compensation in metabolic adaptation in mammalian cells are unidentified. Here, we show that in mouse muscle and myogenic cells, compensatory regulation of the histone deacetylase (HDAC5) transcriptional repressor maintains metabolic integrity. HDAC5 phosphorylation regulated the expression of diverse metabolic genes and glucose metabolism in mouse C2C12 myogenic cells. However, loss of AMP-activated protein kinase (AMPK), a HDAC5 kinase, in muscle did not affect HDAC5 phosphorylation in mouse skeletal muscle during exercise, but resulted in a compensatory increase (32.6%) in the activation of protein kinase D (PKD), an alternate HDAC5 kinase. Constitutive PKD activation in mouse C2C12 myogenic cells regulated metabolic genes and glucose metabolism. Although aspects of this response were HDAC5 phosphorylation dependent, blocking HDAC5 phosphorylation when PKD was active engaged an alternative compensatory adaptive mechanism, which involved post-transcriptional reductions in HDAC5 mRNA (-93.1%) and protein. This enhanced the expression of a specific subset of metabolic genes and mitochondrial metabolism. These data show that compensatory regulation of HDAC5 maintains metabolic integrity in mammalian cells and reinforces the importance of preserving the cellular metabolic adaptive response.


Asunto(s)
Adaptación Fisiológica/fisiología , Metabolismo Energético/fisiología , Regulación Enzimológica de la Expresión Génica/fisiología , Histona Desacetilasas/fisiología , Músculo Esquelético/enzimología , Mioblastos/metabolismo , Condicionamiento Físico Animal/fisiología , Proteína Quinasa C/fisiología , Proteínas Quinasas Activadas por AMP/genética , Proteínas Quinasas Activadas por AMP/fisiología , Acetilación , Animales , Línea Celular , Activación Enzimática , Glucosa/metabolismo , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/biosíntesis , Histona Desacetilasas/genética , Ratones , Ratones Endogámicos C57BL , Contracción Muscular , Fosforilación , Mutación Puntual , Proteína Quinasa C/genética , Procesamiento Proteico-Postraduccional , ARN Mensajero/biosíntesis , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/genética , Transducción de Señal/fisiología , Transcripción Genética/fisiología , Transgenes
17.
PLoS One ; 9(2): e87230, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24520329

RESUMEN

Estrogens are known to play a role in modulating metabolic processes within the body. The Aromatase knockout (ArKO) mice have been shown to harbor factors of Metabolic syndrome with central adiposity, hyperinsulinemia and male-specific hepatic steatosis. To determine the effects of estrogen ablation and subsequent replacement in males on whole body glucose metabolism, three- and six-month-old male ArKO mice were subjected to whole body glucose, insulin and pyruvate tolerance tests and analyzed for ensuing metabolic changes in liver, adipose tissue, and skeletal muscle. Estrogen-deficient male ArKO mice showed increased gonadal adiposity which was significantly reduced upon 17ß-estradiol (E2) treatment. Concurrently, elevated ArKO serum leptin levels were significantly reduced upon E2 treatment and lowered serum adiponectin levels were restored to wild type levels. Three-month-old male ArKO mice were hyperglycemic, and both glucose and pyruvate intolerant. These phenotypes continued through to 6 months of age, highlighting a loss of glycemic control. ArKO livers displayed changes in gluconeogenic enzyme expression, and in insulin signaling pathways upon E2 treatment. Liver triglycerides were increased in the ArKO males only after 6 months of age, which could be reversed by E2 treatment. No differences were observed in insulin-stimulated ex vivo muscle glucose uptake nor changes in ArKO adipose tissue and muscle insulin signaling pathways. Therefore, we conclude that male ArKO mice develop hepatic glucose intolerance by the age of 3 months which precedes the sex-specific development of hepatic steatosis. This can be reversed upon the administration of exogenous E2.


Asunto(s)
Aromatasa/deficiencia , Aromatasa/metabolismo , Intolerancia a la Glucosa/enzimología , Hígado/metabolismo , Hígado/patología , Adiponectina/sangre , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/patología , Animales , Glucemia/metabolismo , Peso Corporal/efectos de los fármacos , Estrógenos/farmacología , Gluconeogénesis/efectos de los fármacos , Gluconeogénesis/genética , Intolerancia a la Glucosa/sangre , Intolerancia a la Glucosa/patología , Insulina/sangre , Resistencia a la Insulina , Leptina/sangre , Hígado/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculos/metabolismo , Tamaño de los Órganos/efectos de los fármacos , Fosforilación/efectos de los fármacos , Ácido Pirúvico/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Triglicéridos/metabolismo
18.
J Physiol ; 591(20): 5207-20, 2013 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-23918774

RESUMEN

Deacetylases such as sirtuins (SIRTs) convert NAD to nicotinamide (NAM). Nicotinamide phosphoribosyl transferase (Nampt) is the rate-limiting enzyme in the NAD salvage pathway responsible for converting NAM to NAD to maintain cellular redox state. Activation of AMP-activated protein kinase (AMPK) increases SIRT activity by elevating NAD levels. As NAM directly inhibits SIRTs, increased Nampt activation or expression could be a metabolic stress response. Evidence suggests that AMPK regulates Nampt mRNA content, but whether repeated AMPK activation is necessary for increasing Nampt protein levels is unknown. To this end, we assessed whether exercise training- or 5-amino-1-ß-D-ribofuranosyl-imidazole-4-carboxamide (AICAR)-mediated increases in skeletal muscle Nampt abundance are AMPK dependent. One-legged knee-extensor exercise training in humans increased Nampt protein by 16% (P < 0.05) in the trained, but not the untrained leg. Moreover, increases in Nampt mRNA following acute exercise or AICAR treatment (P < 0.05 for both) were maintained in mouse skeletal muscle lacking a functional AMPK α2 subunit. Nampt protein was reduced in skeletal muscle of sedentary AMPK α2 kinase dead (KD), but 6.5 weeks of endurance exercise training increased skeletal muscle Nampt protein to a similar extent in both wild-type (WT) (24%) and AMPK α2 KD (18%) mice. In contrast, 4 weeks of daily AICAR treatment increased Nampt protein in skeletal muscle in WT mice (27%), but this effect did not occur in AMPK α2 KD mice. In conclusion, functional α2-containing AMPK heterotrimers are required for elevation of skeletal muscle Nampt protein, but not mRNA induction. These findings suggest AMPK plays a post-translational role in the regulation of skeletal muscle Nampt protein abundance, and further indicate that the regulation of cellular energy charge and nutrient sensing is mechanistically related.


Asunto(s)
Músculo Esquelético/metabolismo , Nicotinamida Fosforribosiltransferasa/metabolismo , Adenilato Quinasa/genética , Adenilato Quinasa/metabolismo , Adulto , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacología , Animales , Ejercicio Físico , Células HEK293 , Humanos , Masculino , Ratones , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/fisiología , Nicotinamida Fosforribosiltransferasa/genética , Esfuerzo Físico , Ribonucleótidos/farmacología
19.
PLoS One ; 8(2): e55174, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23468844

RESUMEN

The TGF-b superfamily cytokine MIC-1/GDF15 circulates in all humans and when overproduced in cancer leads to anorexia/cachexia, by direct action on brain feeding centres. In these studies we have examined the role of physiologically relevant levels of MIC-1/GDF15 in the regulation of appetite, body weight and basal metabolic rate. MIC-1/GDF15 gene knockout mice (MIC-1(-/-)) weighed more and had increased adiposity, which was associated with increased spontaneous food intake. Female MIC-1(-/-) mice exhibited some additional alterations in reduced basal energy expenditure and physical activity, possibly owing to the associated decrease in total lean mass. Further, infusion of human recombinant MIC-1/GDF15 sufficient to raise serum levels in MIC-1(-/-) mice to within the normal human range reduced body weight and food intake. Taken together, our findings suggest that MIC-1/GDF15 is involved in the physiological regulation of appetite and energy storage.


Asunto(s)
Apetito/genética , Peso Corporal/genética , Factor 15 de Diferenciación de Crecimiento/genética , Tejido Adiposo/crecimiento & desarrollo , Animales , Apetito/fisiología , Peso Corporal/fisiología , Ingestión de Alimentos , Metabolismo Energético/genética , Femenino , Genotipo , Factor 15 de Diferenciación de Crecimiento/metabolismo , Humanos , Masculino , Ratones , Ratones Noqueados , Tamaño de los Órganos , Factores Sexuales , Transducción de Señal , Aumento de Peso/genética
20.
Diabetes ; 62(1): 56-64, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22961088

RESUMEN

Obesity is associated with chronic low-grade inflammation that contributes to defects in energy metabolism and insulin resistance. Suppressor of cytokine signaling (SOCS)-3 expression is increased in skeletal muscle of obese humans. SOCS3 inhibits leptin signaling in the hypothalamus and insulin signal transduction in adipose tissue and the liver. Skeletal muscle is an important tissue for controlling energy expenditure and whole-body insulin sensitivity; however, the physiological importance of SOCS3 in this tissue has not been examined. Therefore, we generated mice that had SOCS3 specifically deleted in skeletal muscle (SOCS MKO). The SOCS3 MKO mice had normal muscle development, body mass, adiposity, appetite, and energy expenditure compared with wild-type (WT) littermates. Despite similar degrees of obesity when fed a high-fat diet, SOCS3 MKO mice were protected against the development of hyperinsulinemia and insulin resistance because of enhanced skeletal muscle insulin receptor substrate 1 (IRS1) and Akt phosphorylation that resulted in increased skeletal muscle glucose uptake. These data indicate that skeletal muscle SOCS3 does not play a critical role in regulating muscle development or energy expenditure, but it is an important contributing factor for inhibiting insulin sensitivity in obesity. Therapies aimed at inhibiting SOCS3 in skeletal muscle may be effective in reversing obesity-related glucose intolerance and insulin resistance.


Asunto(s)
Resistencia a la Insulina , Músculo Esquelético/metabolismo , Obesidad/metabolismo , Proteínas Supresoras de la Señalización de Citocinas/fisiología , Animales , Proteínas Sustrato del Receptor de Insulina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación , Proteína 3 Supresora de la Señalización de Citocinas , Triglicéridos/sangre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...