Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Proc Natl Acad Sci U S A ; 120(4): e2211933120, 2023 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-36656866

RESUMEN

Metformin is the most prescribed drug for DM2, but its site and mechanism of action are still not well established. Here, we investigated the effects of metformin on basolateral intestinal glucose uptake (BIGU), and its consequences on hepatic glucose production (HGP). In diabetic patients and mice, the primary site of metformin action was the gut, increasing BIGU, evaluated through PET-CT. In mice and CaCo2 cells, this increase in BIGU resulted from an increase in GLUT1 and GLUT2, secondary to ATF4 and AMPK. In hyperglycemia, metformin increased the lactate (reducing pH and bicarbonate in portal vein) and acetate production in the gut, modulating liver pyruvate carboxylase, MPC1/2, and FBP1, establishing a gut-liver crosstalk that reduces HGP. In normoglycemia, metformin-induced increases in BIGU is accompanied by hypoglycemia in the portal vein, generating a counter-regulatory mechanism that avoids reductions or even increases HGP. In summary, metformin increases BIGU and through gut-liver crosstalk influences HGP.


Asunto(s)
Tracto Gastrointestinal , Glucosa , Hígado , Metformina , Animales , Humanos , Ratones , Células CACO-2 , Diabetes Mellitus Tipo 2 , Glucosa/metabolismo , Hipoglucemiantes/farmacología , Hígado/metabolismo , Metformina/farmacología , Tomografía Computarizada por Tomografía de Emisión de Positrones , Tracto Gastrointestinal/metabolismo
2.
FASEB J ; 33(11): 11909-11924, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31366244

RESUMEN

Growth hormone (GH) is secreted during hypoglycemia, and GH-responsive neurons are found in brain areas containing glucose-sensing neurons that regulate the counter-regulatory response (CRR). However, whether GH modulates the CRR to hypoglycemia via specific neuronal populations is currently unknown. Mice carrying ablation of GH receptor (GHR) either in leptin receptor (LepR)- or steroidogenic factor-1 (SF1)-expressing cells were studied. We also investigated the importance of signal transducer and activator of transcription 5 (STAT5) signaling in SF1 cells for the CRR. GHR ablation in LepR cells led to impaired capacity to recover from insulin-induced hypoglycemia and to a blunted CRR caused by 2-deoxy-d-glucose (2DG) administration. GHR inactivation in SF1 cells, which include ventromedial hypothalamic neurons, also attenuated the CRR. The reduced CRR was prevented by parasympathetic blockers. Additionally, infusion of 2DG produced an abnormal hyperactivity of parasympathetic preganglionic neurons, whereas the 2DG-induced activation of anterior bed nucleus of the stria terminalis neurons was reduced in mice without GHR in SF1 cells. Mice carrying ablation of Stat5a/b genes in SF1 cells showed no defects in the CRR. In summary, GHR expression in SF1 cells is required for a normal CRR, and these effects are largely independent of STAT5 pathway.-Furigo, I. C., de Souza, G. O., Teixeira, P. D. S., Guadagnini, D., Frazão, R., List, E. O., Kopchick, J. J., Prada, P. O., Donato, J., Jr. Growth hormone enhances the recovery of hypoglycemia via ventromedial hypothalamic neurons.


Asunto(s)
Hormona del Crecimiento/farmacología , Hipoglucemia/tratamiento farmacológico , Hipotálamo/efectos de los fármacos , Neuronas/efectos de los fármacos , Recuperación de la Función/efectos de los fármacos , Animales , Desoxiglucosa/farmacología , Hipoglucemia/fisiopatología , Hipotálamo/citología , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/metabolismo , Neuronas/fisiología , Receptores de Leptina/genética , Receptores de Leptina/metabolismo , Receptores de Somatotropina/genética , Receptores de Somatotropina/metabolismo , Factor de Transcripción STAT5/genética , Factor de Transcripción STAT5/metabolismo , Transducción de Señal/efectos de los fármacos , Factor Esteroidogénico 1/genética , Factor Esteroidogénico 1/metabolismo
3.
Clin Sci (Lond) ; 130(8): 601-12, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26785675

RESUMEN

Metabolic dysfunction is associated with adipose tissue inflammation and macrophage infiltration. PAFR (platelet-activating factor receptor) is expressed in several cell types and binds to PAF (platelet-activating factor) and oxidized phospholipids. Engagement of PAFR in macrophages drives them towards the anti-inflammatory phenotype. In the present study, we investigated whether genetic deficiency of PAFR affects the phenotype of ATMs (adipose tissue macrophages) and its effect on glucose and insulin metabolism. PARFKO (PAFR-knockout) and WT (wild-type) mice were fed on an SD (standard diet) or an HFD (high-fat diet). Glucose and insulin tolerance tests were performed by blood monitoring. ATMs were evaluated by FACS for phenotypic markers. Gene and protein expression was investigated by real-time reverse transcription-quantitative PCR and Western blotting respectively. Results showed that the epididymal adipose tissue of PAFRKO mice had increased gene expression of Ccr7, Nos2, Il6 and Il12, associated with pro-inflammatory mediators, and reduced expression of the anti-inflammatory Il10. Moreover, the adipose tissue of PAFRKO mice presented more pro-inflammatory macrophages, characterized by an increased frequency of F4/80(+)CD11c(+) cells. Blood monocytes of PAFRKO mice also exhibited a pro-inflammatory phenotype (increased frequency of Ly6C(+) cells) and PAFR ligands were detected in the serum of both PAFRKO and WT mice. Regarding metabolic parameters, compared with WT, PAFRKO mice had: (i) higher weight gain and serum glucose concentration levels; (ii) decreased insulin-stimulated glucose disappearance; (iii) insulin resistance in the liver; (iv) increased expression of Ldlr in the liver. In mice fed on an HFD, some of these changes were potentiated, particularly in the liver. Thus it seems that endogenous ligands of PAFR are responsible for maintaining the anti-inflammatory profile of blood monocytes and ATMs under physiological conditions. In the absence of PAFR signalling, monocytes and macrophages acquire a pro-inflammatory phenotype, resulting in adipose tissue inflammation and metabolic dysfunction.


Asunto(s)
Tejido Adiposo/metabolismo , Metabolismo Energético , Inflamación/prevención & control , Macrófagos/metabolismo , Glicoproteínas de Membrana Plaquetaria/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Glucemia/metabolismo , Células Cultivadas , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Genotipo , Homeostasis , Inflamación/genética , Inflamación/metabolismo , Mediadores de Inflamación/metabolismo , Insulina/sangre , Resistencia a la Insulina , Ligandos , Ratones Endogámicos BALB C , Ratones Noqueados , Fenotipo , Glicoproteínas de Membrana Plaquetaria/deficiencia , Glicoproteínas de Membrana Plaquetaria/genética , Receptores Acoplados a Proteínas G/deficiencia , Receptores Acoplados a Proteínas G/genética , Transducción de Señal , Factores de Tiempo , Aumento de Peso
5.
PLoS Biol ; 9(12): e1001212, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22162948

RESUMEN

Environmental factors and host genetics interact to control the gut microbiota, which may have a role in the development of obesity and insulin resistance. TLR2-deficient mice, under germ-free conditions, are protected from diet-induced insulin resistance. It is possible that the presence of gut microbiota could reverse the phenotype of an animal, inducing insulin resistance in an animal genetically determined to have increased insulin sensitivity, such as the TLR2 KO mice. In the present study, we investigated the influence of gut microbiota on metabolic parameters, glucose tolerance, insulin sensitivity, and signaling of TLR2-deficient mice. We investigated the gut microbiota (by metagenomics), the metabolic characteristics, and insulin signaling in TLR2 knockout (KO) mice in a non-germ free facility. Results showed that the loss of TLR2 in conventionalized mice results in a phenotype reminiscent of metabolic syndrome, characterized by differences in the gut microbiota, with a 3-fold increase in Firmicutes and a slight increase in Bacteroidetes compared with controls. These changes in gut microbiota were accompanied by an increase in LPS absorption, subclinical inflammation, insulin resistance, glucose intolerance, and later, obesity. In addition, this sequence of events was reproduced in WT mice by microbiota transplantation and was also reversed by antibiotics. At the molecular level the mechanism was unique, with activation of TLR4 associated with ER stress and JNK activation, but no activation of the IKKß-IκB-NFκB pathway. Our data also showed that in TLR2 KO mice there was a reduction in regulatory T cell in visceral fat, suggesting that this modulation may also contribute to the insulin resistance of these animals. Our results emphasize the role of microbiota in the complex network of molecular and cellular interactions that link genotype to phenotype and have potential implications for common human disorders involving obesity, diabetes, and even other immunological disorders.


Asunto(s)
Resistencia a la Insulina , Intestinos/microbiología , Síndrome Metabólico/metabolismo , Receptor Toll-Like 2/metabolismo , Animales , Antibacterianos/uso terapéutico , Cruzamientos Genéticos , Dieta Alta en Grasa/efectos adversos , Estrés del Retículo Endoplásmico/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Intestinos/efectos de los fármacos , Intestinos/inmunología , Grasa Intraabdominal/inmunología , Grasa Intraabdominal/patología , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Masculino , Síndrome Metabólico/tratamiento farmacológico , Metagenómica/métodos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Distribución Aleatoria , Organismos Libres de Patógenos Específicos , Linfocitos T Reguladores/inmunología , Receptor Toll-Like 2/genética
6.
Am J Physiol Endocrinol Metab ; 305(5): E649-59, 2013 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-23880311

RESUMEN

Hypothalamic inflammation is associated with insulin and leptin resistance, hyperphagia, and obesity. In this scenario, hypothalamic protein tyrosine phosphatase 1B (PTP1B) has emerged as the key phosphatase induced by inflammation that is responsible for the central insulin and leptin resistance. Here, we demonstrated that acute exercise reduced inflammation and PTP1B protein level/activity in the hypothalamus of obese rodents. Exercise disrupted the interaction between PTP1B with proteins involved in the early steps of insulin (IRß and IRS-1) and leptin (JAK2) signaling, increased the tyrosine phosphorylation of these molecules, and restored the anorexigenic effects of insulin and leptin in obese rats. Interestingly, the anti-inflammatory action and the reduction of PTP1B activity mediated by exercise occurred in an interleukin-6 (IL-6)-dependent manner because exercise failed to reduce inflammation and PTP1B protein level after the disruption of hypothalamic-specific IL-6 action in obese rats. Conversely, intracerebroventricular administration of recombinant IL-6 reproduced the effects of exercise, improving hypothalamic insulin and leptin action by reducing the inflammatory signaling and PTP1B activity in obese rats at rest. Taken together, our study reports that physical exercise restores insulin and leptin signaling, at least in part, by reducing hypothalamic PTP1B protein level through the central anti-inflammatory response.


Asunto(s)
Hipotálamo/metabolismo , Inflamación/metabolismo , Insulina/metabolismo , Leptina/metabolismo , Obesidad/metabolismo , Condicionamiento Físico Animal/fisiología , Proteína Tirosina Fosfatasa no Receptora Tipo 1/metabolismo , Animales , Western Blotting , Corticosterona/orina , Hipotálamo/enzimología , Inmunohistoquímica , Inflamación/enzimología , Insulina/sangre , Interleucina-6/sangre , Interleucina-6/metabolismo , Leptina/sangre , Masculino , Ratones , Ratones Obesos , Obesidad/enzimología , Distribución Aleatoria , Ratas , Ratas Wistar , Transducción de Señal , Organismos Libres de Patógenos Específicos
7.
PLoS Biol ; 8(8)2010 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-20808781

RESUMEN

Overnutrition caused by overeating is associated with insulin and leptin resistance through IKKbeta activation and endoplasmic reticulum (ER) stress in the hypothalamus. Here we show that physical exercise suppresses hyperphagia and associated hypothalamic IKKbeta/NF-kappaB activation by a mechanism dependent upon the pro-inflammatory cytokine interleukin (IL)-6. The disruption of hypothalamic-specific IL-6 action blocked the beneficial effects of exercise on the re-balance of food intake and insulin and leptin resistance. This molecular mechanism, mediated by physical activity, involves the anti-inflammatory protein IL-10, a core inhibitor of IKKbeta/NF-kappaB signaling and ER stress. We report that exercise and recombinant IL-6 requires IL-10 expression to suppress hyperphagia-related obesity. Moreover, in contrast to control mice, exercise failed to reverse the pharmacological activation of IKKbeta and ER stress in C3H/HeJ mice deficient in hypothalamic IL-6 and IL-10 signaling. Hence, inflammatory signaling in the hypothalamus links beneficial physiological effects of exercise to the central action of insulin and leptin.


Asunto(s)
Antiinflamatorios/metabolismo , Retículo Endoplásmico/patología , Proteínas I-kappa B/metabolismo , Interleucina-10/metabolismo , Interleucina-6/metabolismo , Condicionamiento Físico Animal/fisiología , Animales , Antiinflamatorios/farmacología , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/metabolismo , Metabolismo Energético , Hiperfagia , Hipotálamo/fisiopatología , Insulina/fisiología , Interleucina-10/farmacología , Interleucina-6/farmacología , Leptina/fisiología , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Obesidad/metabolismo , Ratas , Ratas Wistar
8.
Proteomics ; 12(17): 2668-81, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22740317

RESUMEN

Intracellular peptides generated by the proteasome and oligopeptidases have been suggested to function in signal transduction and to improve insulin resistance in mice fed a high-caloric diet. The aim of this study was to identify specific intracellular peptides in the adipose tissue of Wistar rats that could be associated with the physiological and therapeutic control of glucose uptake. Using semiquantitative mass spectrometry and LC/MS/MS analyses, we identified ten peptides in the epididymal adipose tissue of the Wistar rats; three of these peptides were present at increased levels in rats that were fed a high-caloric Western diet (WD) compared with rats fed a control diet (CD). The results of affinity chromatography suggested that in the cytoplasm of epididymal adipose tissue from either WD or CD rats, distinctive proteins bind to these peptides. However, despite the observed increase in the WD animals, the evaluated peptides increased insulin-stimulated glucose uptake in 3T3-L1 adipocytes treated with palmitate. Thus, intracellular peptides from the adipose tissue of Wistar rats can bind to specific proteins and facilitate insulin-induced glucose uptake in 3T3-L1 adipocytes.


Asunto(s)
Tejido Adiposo/citología , Tejido Adiposo/metabolismo , Glucosa/metabolismo , Resistencia a la Insulina , Péptidos/análisis , Péptidos/metabolismo , Células 3T3 , Adipocitos/citología , Adipocitos/metabolismo , Secuencia de Aminoácidos , Animales , Cromatografía de Afinidad , Cromatografía Liquida , Ingestión de Energía , Insulina/metabolismo , Masculino , Ratones , Datos de Secuencia Molecular , Ácido Palmítico/metabolismo , Unión Proteica , Proteínas/metabolismo , Ratas , Ratas Wistar , Espectrometría de Masas en Tándem
9.
Sci Rep ; 11(1): 3813, 2021 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-33589652

RESUMEN

The maintenance of mitochondrial activity in hypothalamic neurons is determinant to the control of energy homeostasis in mammals. Disturbs in the mitochondrial proteostasis can trigger the mitonuclear imbalance and mitochondrial unfolded protein response (UPRmt) to guarantee the mitochondrial integrity and function. However, the role of mitonuclear imbalance and UPRmt in hypothalamic cells are unclear. Combining the transcriptomic analyses from BXD mice database and in vivo experiments, we demonstrated that physical training alters the mitochondrial proteostasis in the hypothalamus of C57BL/6J mice. This physical training elicited the mitonuclear protein imbalance, increasing the mtCO-1/Atp5a ratio, which was accompanied by high levels of UPRmt markers in the hypothalamus. Also, physical training increased the maximum mitochondrial respiratory capacity in the brain. Interestingly, the transcriptomic analysis across several strains of the isogenic BXD mice revealed that hypothalamic mitochondrial DNA-encoded genes were negatively correlated with body weight and several genes related to the orexigenic response. As expected, physical training reduced body weight and food intake. Interestingly, we found an abundance of mt-CO1, a mitochondrial DNA-encoded protein, in NPY-producing neurons in the lateral hypothalamus nucleus of exercised mice. Collectively, our data demonstrated that physical training altered the mitochondrial proteostasis and induced the mitonuclear protein imbalance and UPRmt in hypothalamic cells.


Asunto(s)
Metabolismo Energético/genética , Mitocondrias/genética , Proteostasis/genética , Respuesta de Proteína Desplegada/genética , Animales , Humanos , Hipotálamo/metabolismo , Ratones , Mitocondrias/metabolismo , Neuronas/metabolismo , Condicionamiento Físico Animal/fisiología
10.
Life Sci ; 234: 116793, 2019 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-31465735

RESUMEN

INTRODUCTION: Environmental factors have a key role in the control of gut microbiota and obesity. TLR2 knockout (TLR2-/-) mice in some housing conditions are protected from diet-induced insulin resistance. However, in our housing conditions these animals are not protected from diet-induced insulin-resistance. AIM: The aim of the present study was to investigate the influence of our animal housing conditions on the gut microbiota, glucose tolerance and insulin sensitivity in TLR2-/- mice. MATERIAL AND METHODS: The microbiota was investigated by metagenomics, associated with hyperinsulinemic euglycemic clamp and GTT associated with insulin signaling through immunoblotting. RESULTS: The results showed that TLR2-/- mice in our housing conditions presented a phenotype of metabolic syndrome characterized by insulin resistance, glucose intolerance and increase in body weight. This phenotype was associated with differences in microbiota in TLR2-/- mice that showed a decrease in the Proteobacteria and Bacteroidetes phyla and an increase in the Firmicutesphylum, associated with and in increase in the Oscillospira and Ruminococcus genera. Furthermore there is also an increase in circulating LPS and subclinical inflammation in TLR2-/-. The molecular mechanism that account for insulin resistance was an activation of TLR4, associated with ER stress and JNK activation. The phenotype and metabolic behavior was reversed by antibiotic treatment and reproduced in WT mice by microbiota transplantation. CONCLUSIONS: Our data show, for the first time, that the intestinal microbiota can induce insulin resistance and obesity in an animal model that is genetically protected from these processes.


Asunto(s)
Microbioma Gastrointestinal , Resistencia a la Insulina , Insulina/metabolismo , Receptor Toll-Like 2/genética , Animales , Estrés del Retículo Endoplásmico , Eliminación de Gen , Intolerancia a la Glucosa/genética , Intolerancia a la Glucosa/metabolismo , Intolerancia a la Glucosa/microbiología , Vivienda para Animales , Resistencia a la Insulina/genética , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor Toll-Like 2/metabolismo
11.
Endocrinology ; 160(1): 193-204, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-30462197

RESUMEN

Previous studies have shown that bromocriptine mesylate (Bromo) lowers blood glucose levels in adults with type 2 diabetes mellitus; however, the mechanism of action of the antidiabetic effects of Bromo is unclear. As a dopamine receptor agonist, Bromo can alter brain dopamine activity affecting glucose control, but it also suppresses prolactin (Prl) secretion, and Prl levels modulate glucose homeostasis. Thus, the objective of the current study was to investigate whether Bromo improves insulin sensitivity via inhibition of Prl secretion. Male and female ob/ob animals (a mouse model of obesity and insulin resistance) were treated with Bromo and/or Prl. Bromo-treated ob/ob mice exhibited lower serum Prl concentration, improved glucose and insulin tolerance, and increased insulin sensitivity in the liver and skeletal muscle compared with vehicle-treated mice. Prl replacement in Bromo-treated mice normalized serum Prl concentration without inducing hyperprolactinemia. Importantly, Prl replacement partially reversed the improvements in glucose homeostasis caused by Bromo treatment. The effects of the Prl receptor antagonist G129R-hPrl on glucose homeostasis were also investigated. We found that central G129R-hPrl infusion increased insulin tolerance of male ob/ob mice. In summary, our findings indicate that part of Bromo effects on glucose homeostasis are associated with decrease in serum Prl levels. Because G129R-hPrl treatment also improved the insulin sensitivity of ob/ob mice, pharmacological compounds that inhibit Prl signaling may represent a promising therapeutic approach to control blood glucose levels in individuals with insulin resistance.


Asunto(s)
Bromocriptina/administración & dosificación , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Hipoglucemiantes/administración & dosificación , Prolactina/metabolismo , Animales , Glucemia/metabolismo , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Femenino , Humanos , Insulina/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones , Ratones Obesos , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo
12.
Diabetes ; 56(4): 1014-24, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17229938

RESUMEN

The actions of acetylcholine (ACh) on endothelium mainly are mediated through muscarinic receptors, which are members of the G protein-coupled receptor family. In the present study, we show that ACh induces rapid tyrosine phosphorylation and activation of Janus kinase 2 (JAK2) in rat aorta. Upon JAK2 activation, tyrosine phosphorylation of insulin receptor substrate (IRS)-1 is detected. In addition, ACh induces JAK2/IRS-1 and IRS-1/phosphatidylinositol (PI) 3-kinase associations, downstream activation of Akt/protein kinase B, endothelial cell-nitric oxide synthase (eNOS), and extracellular signal-regulated kinase (ERK)-1/2. The pharmacological blockade of JAK2 or PI 3-kinase reduced ACh-stimulated eNOS phosphorylation, NOS activity, and aorta relaxation. These data indicate a new signal transduction pathway for IRS-1/PI 3-kinase/Akt/eNOS activation and ERK1/2 by means of JAK2 tyrosine phosphorylation stimulated by ACh in vessels. Moreover, we demonstrate that in aorta of obese rats (high-fat diet), there is an impairment in the insulin- and ACh-stimulated IRS-1/PI 3-kinase pathway, leading to reduced activation with lower protein levels of eNOS associated with a hyperactivated ERK/mitogen-activated protein kinase pathway. These results suggest that in aorta of obese rats, there not only is insulin resistance but also ACh resistance, probably mediated by a common signaling pathway that controls the activity and the protein levels of eNOS.


Asunto(s)
Acetilcolina/farmacología , Endotelio Vascular/enzimología , Insulina/farmacología , Óxido Nítrico Sintasa de Tipo III/biosíntesis , Obesidad/fisiopatología , Proteínas Proto-Oncogénicas c-akt/fisiología , Receptor de Insulina/fisiología , Animales , Grasas de la Dieta , Endotelio Vascular/efectos de los fármacos , Ingestión de Energía , Inducción Enzimática , Janus Quinasa 2/metabolismo , Masculino , Obesidad/enzimología , Ratas , Ratas Mutantes , Ratas Wistar , Transducción de Señal
13.
Life Sci ; 82(25-26): 1262-71, 2008 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-18534630

RESUMEN

The hypothalamic AMP-activated protein kinase (AMPK)/acetyl-CoA carboxylase (ACC) pathway is known to play an important role in the control of food intake and energy expenditure. Here, we hypothesize that citrate, an intermediate metabolite, activates hypothalamic ACC and is involved in the control of energy mobilization. Initially, we showed that ICV citrate injection decreased food intake and diminished weight gain significantly when compared to control and pair-fed group results. In addition, we showed that intracerebroventricular (ICV) injection of citrate diminished (80% of control) the phosphorylation of ACC, an important AMPK substrate. Furthermore, citrate treatment inhibited (75% of control) hypothalamic AMPK phosphorylation during fasting. In addition to its central effect, ICV citrate injection led to low blood glucose levels during glucose tolerance test (GTT) and high glucose uptake during hyperglycemic-euglycemic clamp. Accordingly, liver glycogen content was higher in animals given citrate (ICV) than in the control group (23.3+/-2.5 vs. 2.7+/-0.5 microg mL(-1) mg(-1), respectively). Interestingly, liver AMPK phosphorylation was reduced (80%) by the citrate treatment. The pharmacological blockade of beta3-adrenergic receptor (SR 59230A) blocked the effect of ICV citrate and citrate plus insulin on liver AMPK phosphorylation. Consistently with these results, rats treated with citrate (ICV) presented improved insulin signal transduction in liver, skeletal muscle, and epididymal fat pad. Similar results were obtained by hypothalamic administration of ARA-A, a competitive inhibitor of AMPK. Our results suggest that the citrate produced by mitochondria may modulate ACC phosphorylation in the hypothalamus, controlling food intake and coordinating a multiorgan network that controls glucose homeostasis and energy uptake through the adrenergic system.


Asunto(s)
Acetil-CoA Carboxilasa/metabolismo , Ácido Cítrico/farmacología , Glucosa/metabolismo , Homeostasis/efectos de los fármacos , Hipotálamo/enzimología , Hígado/metabolismo , Respuesta de Saciedad/efectos de los fármacos , Proteínas Quinasas Activadas por AMP , Animales , Peso Corporal/efectos de los fármacos , Ácido Cítrico/administración & dosificación , Corticosterona/sangre , Conducta Alimentaria/efectos de los fármacos , Prueba de Tolerancia a la Glucosa , Glucógeno/metabolismo , Hipotálamo/efectos de los fármacos , Inyecciones Intraventriculares , Insulina/sangre , Hígado/enzimología , Masculino , Complejos Multienzimáticos/metabolismo , Fosforilación/efectos de los fármacos , Propanolaminas , Proteínas Serina-Treonina Quinasas/metabolismo , Ratas , Ratas Wistar , Transducción de Señal/efectos de los fármacos
14.
Mol Metab ; 6(2): 206-218, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28180062

RESUMEN

OBJECTIVE: Recent data show that iNOS has an essential role in ER stress in obesity. However, whether iNOS is sufficient to account for obesity-induced ER stress and Unfolded Protein Response (UPR) has not yet been investigated. In the present study, we used iNOS knockout mice to investigate whether high-fat diet (HFD) can still induce residual ER stress-associated insulin resistance. METHODS: For this purpose, we used the intraperitoneal glucose tolerance test (GTT), euglycemic-hyperinsulinemic clamp, western blotting and qPCR in liver, muscle, and adipose tissue of iNOS KO and control mice on HFD. RESULTS: The results of the present study demonstrated that, in HFD fed mice, iNOS-induced alteration in insulin signaling is an essential mechanism of insulin resistance in muscle, suggesting that iNOS may represent an important target that could be blocked in order to improve insulin sensitivity in this tissue. However, in liver and adipose tissue, the insulin resistance induced by HFD was only partially dependent on iNOS, and, even in the presence of genetic or pharmacological blockade of iNOS, a clear ER stress associated with altered insulin signaling remained evident in these tissues. When this ER stress was blocked pharmacologically, insulin signaling was improved, and a complete recovery of glucose tolerance was achieved. CONCLUSIONS: Taken together, these results reinforce the tissue-specific regulation of insulin signaling in obesity, with iNOS being sufficient to account for insulin resistance in muscle, but in liver and adipose tissue ER stress and insulin resistance can be induced by both iNOS-dependent and iNOS-independent mechanisms.


Asunto(s)
Estrés del Retículo Endoplásmico/fisiología , Óxido Nítrico Sintasa de Tipo II/antagonistas & inhibidores , Tejido Adiposo/metabolismo , Animales , Glucemia/metabolismo , Dieta Alta en Grasa , Grasas de la Dieta/metabolismo , Insulina/genética , Resistencia a la Insulina/fisiología , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Óxido Nítrico Sintasa de Tipo II/deficiencia , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Obesidad/genética , Transducción de Señal/fisiología , Respuesta de Proteína Desplegada
15.
J Nutr Biochem ; 50: 16-25, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28968517

RESUMEN

Obesity and type 2 diabetes are characterized by subclinical inflammatory process. Changes in composition or modulation of the gut microbiota may play an important role in the obesity-associated inflammatory process. In the current study, we evaluated the effects of probiotics (Lactobacillus rhamnosus, L. acidophilus and Bifidobacterium bifidumi) on gut microbiota, changes in permeability, and insulin sensitivity and signaling in high-fat diet and control animals. More importantly, we investigated the effects of these gut modulations on hypothalamic control of food intake, and insulin and leptin signaling. Swiss mice were submitted to a high-fat diet (HFD) with probiotics or pair-feeding for 5 weeks. Metagenome analyses were performed on DNA samples from mouse feces. Blood was drawn to determine levels of glucose, insulin, LPS, cytokines and GLP-1. Liver, muscle, ileum and hypothalamus tissue proteins were analyzed by Western blotting and real-time polymerase chain reaction. In addition, liver and adipose tissues were analyzed using histology and immunohistochemistry. The HFD induced huge alterations in gut microbiota accompanied by increased intestinal permeability, LPS translocation and systemic low-grade inflammation, resulting in decreased glucose tolerance and hyperphagic behavior. All these obesity-related features were reversed by changes in the gut microbiota profile induced by probiotics. Probiotics also induced an improvement in hypothalamic insulin and leptin resistance. Our data demonstrate that the intestinal microbiome is a key modulator of inflammatory and metabolic pathways in both peripheral and central tissues. These findings shed light on probiotics as an important tool to prevent and treat patients with obesity and insulin resistance.


Asunto(s)
Diabetes Mellitus Tipo 2/prevención & control , Disbiosis/prevención & control , Microbioma Gastrointestinal , Resistencia a la Insulina , Mucosa Intestinal/fisiopatología , Obesidad/dietoterapia , Probióticos/uso terapéutico , Tejido Adiposo Blanco/inmunología , Tejido Adiposo Blanco/metabolismo , Tejido Adiposo Blanco/patología , Animales , Regulación del Apetito , Bifidobacterium bifidum/clasificación , Bifidobacterium bifidum/crecimiento & desarrollo , Bifidobacterium bifidum/inmunología , Bifidobacterium bifidum/aislamiento & purificación , Permeabilidad de la Membrana Celular , Diabetes Mellitus Tipo 2/etiología , Diabetes Mellitus Tipo 2/inmunología , Diabetes Mellitus Tipo 2/microbiología , Dieta Alta en Grasa/efectos adversos , Disbiosis/etiología , Disbiosis/inmunología , Disbiosis/microbiología , Heces/microbiología , Microbioma Gastrointestinal/inmunología , Técnica de Clampeo de la Glucosa , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Lactobacillus acidophilus/clasificación , Lactobacillus acidophilus/crecimiento & desarrollo , Lactobacillus acidophilus/inmunología , Lactobacillus acidophilus/aislamiento & purificación , Lacticaseibacillus rhamnosus/clasificación , Lacticaseibacillus rhamnosus/crecimiento & desarrollo , Lacticaseibacillus rhamnosus/inmunología , Lacticaseibacillus rhamnosus/aislamiento & purificación , Hígado/inmunología , Hígado/metabolismo , Hígado/patología , Masculino , Ratones , Tipificación Molecular , Obesidad/metabolismo , Obesidad/patología , Obesidad/fisiopatología , Distribución Aleatoria
16.
FEBS Lett ; 579(14): 3152-8, 2005 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-15922338

RESUMEN

In this study, we demonstrate that pretreatment with aspirin inhibits GH-induced insulin resistance. GH was observed to lead to serine phosphorylation of IRS-1, a phenomenon which was reversed by aspirin in liver, muscle and WAT in parallel with a reduction in JNK activity. In addition, our data show an impairment of insulin activation in the IR/IRS/PI(3)kinase pathway and a reduction in IRS-1 protein levels in rats treated with GH, which was also reversed in the animals pretreated with aspirin. Overall, these results provide new insights into the mechanism of GH-induced insulin resistance.


Asunto(s)
Aspirina/farmacología , Hormona del Crecimiento/farmacología , Fosfoproteínas/metabolismo , Fosfoserina/metabolismo , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/metabolismo , Animales , Insulina/farmacología , Proteínas Sustrato del Receptor de Insulina , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Músculos/efectos de los fármacos , Músculos/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-jun/metabolismo , Ratas , Ratas Wistar , Receptor de Insulina/metabolismo
17.
Diabetes ; 63(10): 3334-45, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24812431

RESUMEN

IKK epsilon (IKKε) is induced by the activation of nuclear factor-κB (NF-κB). Whole-body IKKε knockout mice on a high-fat diet (HFD) were protected from insulin resistance and showed altered energy balance. We demonstrate that IKKε is expressed in neurons and is upregulated in the hypothalamus of obese mice, contributing to insulin and leptin resistance. Blocking IKKε in the hypothalamus of obese mice with CAYMAN10576 or small interfering RNA decreased NF-κB activation in this tissue, relieving the inflammatory environment. Inhibition of IKKε activity, but not TBK1, reduced IRS-1(Ser307) phosphorylation and insulin and leptin resistance by an improvement of the IR/IRS-1/Akt and JAK2/STAT3 pathways in the hypothalamus. These improvements were independent of body weight and food intake. Increased insulin and leptin action/signaling in the hypothalamus may contribute to a decrease in adiposity and hypophagia and an enhancement of energy expenditure accompanied by lower NPY and increased POMC mRNA levels. Improvement of hypothalamic insulin action decreases fasting glycemia, glycemia after pyruvate injection, and PEPCK protein expression in the liver of HFD-fed and db/db mice, suggesting a reduction in hepatic glucose production. We suggest that IKKε may be a key inflammatory mediator in the hypothalamus of obese mice, and its hypothalamic inhibition improves energy and glucose metabolism.


Asunto(s)
Hipotálamo/metabolismo , Quinasa I-kappa B/metabolismo , Resistencia a la Insulina/fisiología , Neuronas/metabolismo , Obesidad/metabolismo , Animales , Peso Corporal/fisiología , Dieta Alta en Grasa , Quinasa I-kappa B/antagonistas & inhibidores , Insulina/metabolismo , Leptina/metabolismo , Hígado/metabolismo , Masculino , Ratones , Ratones Obesos , Fosforilación , Transducción de Señal/fisiología , Regulación hacia Arriba
18.
Nat Commun ; 5: 4859, 2014 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-25255053

RESUMEN

Sphingosine 1-phosphate receptor 1 (S1PR1) is a G-protein-coupled receptor for sphingosine-1-phosphate (S1P) that has a role in many physiological and pathophysiological processes. Here we show that the S1P/S1PR1 signalling pathway in hypothalamic neurons regulates energy homeostasis in rodents. We demonstrate that S1PR1 protein is highly enriched in hypothalamic POMC neurons of rats. Intracerebroventricular injections of the bioactive lipid, S1P, reduce food consumption and increase rat energy expenditure through persistent activation of STAT3 and the melanocortin system. Similarly, the selective disruption of hypothalamic S1PR1 increases food intake and reduces the respiratory exchange ratio. We further show that STAT3 controls S1PR1 expression in neurons via a positive feedback mechanism. Interestingly, several models of obesity and cancer anorexia display an imbalance of hypothalamic S1P/S1PR1/STAT3 axis, whereas pharmacological intervention ameliorates these phenotypes. Taken together, our data demonstrate that the neuronal S1P/S1PR1/STAT3 signalling axis plays a critical role in the control of energy homeostasis in rats.


Asunto(s)
Metabolismo Energético , Hipotálamo/metabolismo , Lisofosfolípidos/metabolismo , Receptores de Lisoesfingolípidos/metabolismo , Esfingosina/análogos & derivados , Animales , Homeostasis , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/metabolismo , Proopiomelanocortina/metabolismo , Ratas , Ratas Wistar , Receptores de Lisoesfingolípidos/genética , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Esfingosina/metabolismo , Receptores de Esfingosina-1-Fosfato
19.
Expert Opin Investig Drugs ; 22(6): 751-63, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23705634

RESUMEN

INTRODUCTION: Some inhibitors of tyrosine kinase, as imatinib, erlotinib and sunitinib have antihyperglycemic effects but the mechanisms are not totally clear. AREAS COVERED: It is well established that insulin resistance and beta-cell failure are hallmarks of type 2 diabetes mellitus (DM2). The present review will discuss the molecular mechanisms that account for insulin resistance and beta-cell failure in DM2, and also the effect of tyrosine kinase inhibitors in these processes. EXPERT OPINION: A better understanding of how these drugs improve the two most important mechanisms of DM2 associated with suggestions of clinical studies will lead to improve the treatment of this disease.


Asunto(s)
Diabetes Mellitus Tipo 2/tratamiento farmacológico , Hipoglucemiantes/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Animales , Ensayos Clínicos como Asunto , Diabetes Mellitus Tipo 2/enzimología , Diseño de Fármacos , Humanos , Resistencia a la Insulina , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Proteínas Tirosina Quinasas/antagonistas & inhibidores
20.
FEBS Open Bio ; 3: 443-9, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24251109

RESUMEN

Insulin acts in the hypothalamus, decreasing food intake (FI) by the IR/PI3K/Akt pathway. This pathway is impaired in obese animals and endoplasmic reticulum (ER) stress and low-grade inflammation are possible mechanisms involved in this impairment. Here, we highlighted the amygdala as an important brain region for FI regulation in response to insulin. This regulation was dependent on PI3K/AKT pathway similar to the hypothalamus. Insulin was able to decrease neuropeptide Y (NPY) and increase oxytocin mRNA levels in the amygdala via PI3K, which may contribute to hypophagia. Additionally, obese rats did not reduce FI in response to insulin and AKT phosphorylation was decreased in the amygdala, suggesting insulin resistance. Insulin resistance was associated with ER stress and low-grade inflammation in this brain region. The inhibition of ER stress with PBA reverses insulin action/signaling, decreases NPY and increases oxytocin mRNA levels in the amygdala from obese rats, suggesting that ER stress is probably one of the mechanisms that induce insulin resistance in the amygdala.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA