Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
FASEB J ; 37(9): e23120, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37527279

RESUMEN

The α7nAChR is crucial to the anti-inflammatory reflex, and to the expression of neuropeptides that control food intake, but its expression can be decreased by environmental factors. We aimed to investigate whether microRNA modulation could be an underlying mechanism in the α7nAchR downregulation in mouse hypothalamus following a short-term exposure to an obesogenic diet. Bioinformatic analysis revealed Let-7 microRNAs as candidates to regulate Chrna7, which was confirmed by the luciferase assay. Mice exposed to an obesogenic diet for 3 days had increased Let-7a and decreased α7nAChR levels, accompanied by hypothalamic fatty acids and TNFα content. Hypothalamic neuronal cells exposed to fatty acids presented higher Let-7a and TNFα levels and lower Chrna7 expression, but when the cells were pre-treated with TLR4 inhibitor, Let-7a, TNFα, and Chrna7 were rescued to normal levels. Thus, the fatty acids overload trigger TNFα-induced Let-7 overexpression in hypothalamic neuronal cells, which negatively regulates α7nAChR, an event that can be related to hyperphagia and obesity predisposition in mice.


Asunto(s)
Factor de Necrosis Tumoral alfa , Receptor Nicotínico de Acetilcolina alfa 7 , Animales , Ratones , Receptor Nicotínico de Acetilcolina alfa 7/genética , Receptor Nicotínico de Acetilcolina alfa 7/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Ácidos Grasos , Regulación hacia Abajo , Hipotálamo/metabolismo
2.
Front Immunol ; 14: 1273556, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38193079

RESUMEN

Introduction: High-fat diet (HFD) consumption is associated with various metabolic disorders and diseases. Both pre-pregnancy and maternal obesity can have long-term consequences on offspring health. Furthermore, consuming an HFD in adulthood significantly increases the risk of obesity and metabolic disorders. However, an intriguing phenomenon known as the obesity paradox suggests that obesity may confer a protective effect on mortality outcomes in sepsis. In sepsis, activation of the cholinergic anti-inflammatory pathway (CAP) can help mitigate systemic inflammation. We employed a metabolic programming model to explore the relationship between maternal HFD consumption and offspring response to sepsis. Methods: We fed female mice either a standard diet (SC) or an HFD during the pre-pregnancy, pregnancy, and lactation periods. Subsequently, we evaluated 28-day-old male offspring. Results: Notably, we discovered that offspring from HFD-fed dams (HFD-O) exhibited a higher survival rate compared with offspring from SC-fed dams (SC-O). Importantly, inhibition of the m1 muscarinic acetylcholine receptor (m1mAChR), involved in the CAP, in the hypothalamus abolished this protection. The expression of m1mAChR in the hypothalamus was higher in HFD-O at different ages, peaking on day 28. Treatment with an m1mAChR agonist could modulate the inflammatory response in peripheral tissues. Specifically, CAP activation was greater in the liver of HFD-O following agonist treatment. Interestingly, lipopolysaccharide (LPS) challenge failed to induce a more inflammatory state in HFD-O, in contrast to SC-O, and agonist treatment had no additional effect. Analysis of spleen immune cells revealed a distinct phenotype in HFD-O, characterized by elevated levels of CD4+ lymphocytes rather than CD8+ lymphocytes. Moreover, basal Il17 messenger RNA (mRNA) levels were lower while Il22 mRNA levels were higher in HFD-O, and we observed the same pattern after LPS challenge. Discussion: Further examination of myeloid cells isolated from bone marrow and allowed to differentiate showed that HFD-O macrophages displayed an anti-inflammatory phenotype. Additionally, treatment with the m1mAChR agonist contributed to reducing inflammatory marker levels in both groups. In summary, our findings demonstrate that HFD-O are protected against LPS-induced sepsis, and this protection is mediated by the central m1mAChR. Moreover, the inflammatory response in the liver, spleen, and bone marrow-differentiated macrophages is diminished. However, more extensive analysis is necessary to elucidate the specific mechanisms by which m1mAChR modulates the immune response during sepsis.


Asunto(s)
Enfermedades Metabólicas , Sepsis , Humanos , Embarazo , Femenino , Masculino , Animales , Ratones , Receptor Muscarínico M1 , Dieta Alta en Grasa/efectos adversos , Lipopolisacáridos , Acetilcolina , Obesidad/etiología , ARN Mensajero
3.
Sci Rep ; 11(1): 8980, 2021 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-33903707

RESUMEN

Nutritional status during gestation may lead to a phenomenon known as metabolic programming, which can be triggered by epigenetic mechanisms. The Let-7 family of microRNAs were one of the first to be discovered, and are closely related to metabolic processes. Bioinformatic analysis revealed that Prkaa2, the gene that encodes AMPK α2, is a predicted target of Let-7. Here we aimed to investigate whether Let-7 has a role in AMPKα2 levels in the NAFLD development in the offspring programmed by maternal obesity. Let-7 levels were upregulated in the liver of newborn mice from obese dams, while the levels of Prkaa2 were downregulated. Let-7 levels strongly correlated with serum glucose, insulin and NEFA, and in vitro treatment of AML12 with glucose and NEFA lead to higher Let-7 expression. Transfection of Let-7a mimic lead to downregulation of AMPKα2 levels, while the transfection with Let-7a inhibitor impaired both NEFA-mediated reduction of Prkaa2 levels and the fat accumulation driven by NEFA. The transfection of Let-7a inhibitor in ex-vivo liver slices from the offspring of obese dams restored phospho-AMPKα2 levels. In summary, Let-7a appears to regulate hepatic AMPKα2 protein levels and lead to the early hepatic metabolic disturbances in the offspring of obese dams.


Asunto(s)
Proteínas Quinasas Activadas por AMP/biosíntesis , Regulación de la Expresión Génica , Metabolismo de los Lípidos , Hígado/metabolismo , MicroARNs/biosíntesis , Obesidad Materna/metabolismo , Efectos Tardíos de la Exposición Prenatal/metabolismo , Animales , Femenino , Hígado/patología , Ratones , Embarazo
4.
Lipids Health Dis ; 17(1): 90, 2018 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-29678181

RESUMEN

BACKGROUND: Peroxisome proliferator-activated receptor γ coactivator- 1alpha (PGC-1α) plays an important role in whole body metabolism and, particularly in glucose homeostasis. Its expression is highly regulated and, small variations in tissue levels can have a major impact in a number of physiological and pathological conditions. Recent studies have shown that the ubiquitin/proteasome system plays a role in the control of PGC-1α degradation. METHODS: Here we evaluated the interaction of PGC-1α with the protein A20, which plays a dual-role in the control of the ubiquitin/proteasome system acting as a deubiquitinase and as an E3 ligase. We employed immunoprecipitation, quantitative real-time PCR and immunofluorescence staining to evaluate PGC-1α, A20, PPARγ and ubiquitin in the adipose tissue of humans and mice. RESULTS: In distinct sites of the adipose tissue, A20 binds to PGC-1α. At least in the subcutaneous fat of humans and mice the levels of PGC-1α decrease during obesity, while its physical association with A20 increases. The inhibition of A20 leads to a reduction of PGC-1α and PPARγ expression, suggesting that A20 acts as a protective factor against PGC-1α disposal. CONCLUSION: We provide evidence that mechanisms regulating PGC-1α ubiquitination are potentially involved in the control of the function of this transcriptional co-activator.


Asunto(s)
Tejido Adiposo/metabolismo , Obesidad/genética , PPAR gamma/genética , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/genética , Tejido Adiposo/patología , Adulto , Animales , Estudios de Casos y Controles , Metabolismo Energético/genética , Femenino , Regulación de la Expresión Génica , Glucosa/metabolismo , Homeostasis/genética , Humanos , Masculino , Ratones , Obesidad/metabolismo , Obesidad/patología , PPAR gamma/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Unión Proteica , Transducción de Señal , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/metabolismo , Ubiquitina/genética , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
6.
J Nutr Biochem ; 34: 30-41, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27180121

RESUMEN

Nutritional excess during pregnancy and lactation has a negative impact on offspring phenotype. In adulthood, obesity and lipid overload represent factors that compromise autophagy, a process of lysosomal degradation. Despite knowledge of the impact of obesity on autophagy, changes in offspring of obese dams have yet to be investigated. In this study, we tested the hypothesis that maternal obesity induced by a high fat diet (HFD) modulates autophagy proteins in the hypothalamus and liver of the offspring of mice. At birth (d0), offspring of obese dams (HFD-O) showed an increase in p62 protein and a decrease in LC3-II, but only in the liver. After weaning (d18), the offspring of HFD-O animals showed impairment of autophagy markers in both tissues compared to control offspring (SC-O). Between day 18 and day 42, both groups received a control diet and we observed that the protein content of p62 remained increased in the livers of the HFD-O offspring. However, after 82days, we did not find any modulation in offspring autophagy proteins. On the other hand, when the offspring of obese dams that received an HFD from day 42 until day 82 (OH-H) were compared with the offspring from the controls that only received an HFD in adulthood (OC-H), we saw impairment in autophagy proteins in both tissues. In conclusion, this study describes that HFD-O offspring showed early impairment of autophagy proteins. Although the molecular mechanisms have not been explored, it is possible that changes in autophagy markers could be associated with metabolic disturbances of offspring.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Hipotálamo/metabolismo , Lactancia , Hígado/metabolismo , Fenómenos Fisiologicos Nutricionales Maternos , Proteínas Asociadas a Microtúbulos/metabolismo , Proteína Sequestosoma-1/metabolismo , Animales , Animales Recién Nacidos , Dieta Alta en Grasa/efectos adversos , Femenino , Desarrollo Fetal , Masculino , Ratones , Proteínas Asociadas a Microtúbulos/genética , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Obesidad/etiología , Obesidad/fisiopatología , Especificidad de Órganos , Obesidad Infantil/etiología , Obesidad Infantil/metabolismo , Obesidad Infantil/patología , Embarazo , Complicaciones del Embarazo/etiología , Complicaciones del Embarazo/fisiopatología , Distribución Aleatoria , Proteína Sequestosoma-1/genética , Destete
7.
Diabetes ; 65(3): 673-86, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26512023

RESUMEN

Apoptosis of hypothalamic neurons is believed to play an important role in the development and perpetuation of obesity. Similar to the hippocampus, the hypothalamus presents constitutive and stimulated neurogenesis, suggesting that obesity-associated hypothalamic dysfunction can be repaired. Here, we explored the hypothesis that n-3 polyunsaturated fatty acids (PUFAs) induce hypothalamic neurogenesis. Both in the diet and injected directly into the hypothalamus, PUFAs were capable of increasing hypothalamic neurogenesis to levels similar or superior to the effect of brain-derived neurotrophic factor (BDNF). Most of the neurogenic activity induced by PUFAs resulted in increased numbers of proopiomelanocortin but not NPY neurons and was accompanied by increased expression of BDNF and G-protein-coupled receptor 40 (GPR40). The inhibition of GPR40 was capable of reducing the neurogenic effect of a PUFA, while the inhibition of BDNF resulted in the reduction of global hypothalamic cell. Thus, PUFAs emerge as a potential dietary approach to correct obesity-associated hypothalamic neuronal loss.


Asunto(s)
Glucemia/efectos de los fármacos , Ácidos Grasos Omega-3/farmacología , Hipotálamo/efectos de los fármacos , Neurogénesis/efectos de los fármacos , Neuronas/efectos de los fármacos , ARN Mensajero/efectos de los fármacos , Animales , Glucemia/metabolismo , Factor Neurotrófico Derivado del Encéfalo/efectos de los fármacos , Factor Neurotrófico Derivado del Encéfalo/genética , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Prueba de Tolerancia a la Glucosa , Hipotálamo/citología , Hipotálamo/metabolismo , Ratones , Neuronas/metabolismo , Neuropéptido Y , Proopiomelanocortina/metabolismo , ARN Mensajero/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores Acoplados a Proteínas G/efectos de los fármacos , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
8.
PLoS One ; 10(3): e0119850, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25786112

RESUMEN

Autophagy is an important process that regulates cellular homeostasis by degrading dysfunctional proteins, organelles and lipids. In this study, the hypothesis that obesity could lead to impairment in hypothalamic autophagy in mice was evaluated by examining the hypothalamic distribution and content of autophagic proteins in animal with obesity induced by 8 or 16 weeks high fat diet to induce obesity and in response to intracerebroventricular injections of palmitic acid. The results showed that chronic exposure to a high fat diet leads to an increased expression of inflammatory markers and downregulation of autophagic proteins. In obese mice, autophagic induction leads to the downregulation of proteins, such as JNK and Bax, which are involved in the stress pathways. In neuron cell-line, palmitate has a direct effect on autophagy even without inflammatory activity. Understanding the cellular and molecular bases of overnutrition is essential for identifying new diagnostic and therapeutic targets for obesity.


Asunto(s)
Fenómenos Fisiológicos Nutricionales de los Animales , Autofagia/fisiología , Ácidos Grasos/metabolismo , Hipotálamo/fisiología , Obesidad/fisiopatología , Análisis de Varianza , Animales , Línea Celular , Técnica del Anticuerpo Fluorescente , Prueba de Tolerancia a la Glucosa , Hipotálamo/metabolismo , Immunoblotting , MAP Quinasa Quinasa 4/metabolismo , Masculino , Ratones , Ratones Obesos , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína X Asociada a bcl-2/metabolismo
9.
Endocrinology ; 155(8): 2831-44, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24892821

RESUMEN

In both human and experimental obesity, inflammatory damage to the hypothalamus plays an important role in the loss of the coordinated control of food intake and energy expenditure. Upon prolonged maintenance of increased body mass, the brain changes the defended set point of adiposity, and returning to normal weight becomes extremely difficult. Here we show that in prolonged but not in short-term obesity, the ubiquitin/proteasome system in the hypothalamus fails to maintain an adequate rate of protein recycling, leading to the accumulation of ubiquitinated proteins. This is accompanied by an increased colocalization of ubiquitin and p62 in the arcuate nucleus and reduced expression of autophagy markers in the hypothalamus. Genetic protection from obesity is accompanied by the normal regulation of the ubiquitin/proteasome system in the hypothalamus, whereas the inhibition of proteasome or p62 results in the acceleration of body mass gain in mice exposed for a short period to a high-fat diet. Thus, the defective regulation of the ubiquitin/proteasome system in the hypothalamus may be an important mechanism involved in the progression and autoperpetuation of obesity.


Asunto(s)
Hipotálamo/metabolismo , Obesidad/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Ubiquitina/metabolismo , Animales , Autofagia , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Humanos , Masculino , Ratones , Ratones Endogámicos C3H , Microglía/metabolismo , Neuronas/metabolismo , Fenotipo , Factor de Transcripción TFIIH , Factores de Transcripción/metabolismo , Aumento de Peso , Pérdida de Peso
10.
Metabolism ; 63(5): 682-92, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24636055

RESUMEN

OBJECTIVE: The goal of this study was to determine the presence early of markers of endoplasmic reticulum stress (ERS) and insulin resistance in the offspring from dams fed HFD (HFD-O) or standard chow diet (SC-O) during pregnancy and lactation. MATERIALS/METHODS: To address this question, we evaluated the hypothalamic and hepatic tissues in recently weaned mice (d28) and the hypothalamus of newborn mice (d0) from dams fed HFD or SC during pregnancy and lactation. RESULTS: Body weight, adipose tissue mass, and food intake were more accentuated in HFD-O mice than in SC-O mice. In addition, intolerance to glucose and insulin was higher in HFD-O mice than in SC-O mice. Compared with SC-O mice, levels of hypothalamic IL1-ß mRNA, NFκB protein, and p-JNK were increased in HFD-O mice. Furthermore, compared with SC-O mice, hypothalamic AKT phosphorylation after insulin challenge was reduced, while markers of ERS (p-PERK, p-eIF2α, XBP1s, GRP78, and GRP94) and p-AMPK were increased in the hypothalamic tissue of HFD-O at d28 but not at d0. These damages to hypothalamic signaling were accompanied by increased triglyceride deposits, activation of NFκB, p-JNK, p-PERK and p-eIF2α. CONCLUSION: These point out lactation period as maternal trigger for metabolic changes in the offspring. These changes may occur early and quietly contribute to obesity and associated pathologies in adulthood. Although in rodents the establishment of ARC neuronal projections occurs during the lactation period, in humans it occurs during the third trimester. Gestational diabetes and obesity in this period may contribute to impairment of energy homeostasis.


Asunto(s)
Dieta Alta en Grasa , Estrés del Retículo Endoplásmico , Hipotálamo/metabolismo , Resistencia a la Insulina , Lactancia , Fenómenos Fisiologicos Nutricionales Maternos , Animales , Animales Lactantes , Grasas de la Dieta/farmacología , Chaperón BiP del Retículo Endoplásmico , Estrés del Retículo Endoplásmico/efectos de los fármacos , Femenino , Hipotálamo/efectos de los fármacos , Hipotálamo/embriología , Lactancia/efectos de los fármacos , Lactancia/fisiología , Masculino , Fenómenos Fisiologicos Nutricionales Maternos/efectos de los fármacos , Ratones , Embarazo , Efectos Tardíos de la Exposición Prenatal/etiología , Efectos Tardíos de la Exposición Prenatal/metabolismo
11.
Am J Physiol Endocrinol Metab ; 305(2): E230-42, 2013 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-23695212

RESUMEN

Melatonin can contribute to glucose homeostasis either by decreasing gluconeogenesis or by counteracting insulin resistance in distinct models of obesity. However, the precise mechanism through which melatonin controls glucose homeostasis is not completely understood. Male Wistar rats were administered an intracerebroventricular (icv) injection of melatonin and one of following: an icv injection of a phosphatidylinositol 3-kinase (PI3K) inhibitor, an icv injection of a melatonin receptor (MT) antagonist, or an intraperitoneal (ip) injection of a muscarinic receptor antagonist. Anesthetized rats were subjected to pyruvate tolerance test to estimate in vivo glucose clearance after pyruvate load and in situ liver perfusion to assess hepatic gluconeogenesis. The hypothalamus was removed to determine Akt phosphorylation. Melatonin injections in the central nervous system suppressed hepatic gluconeogenesis and increased hypothalamic Akt phosphorylation. These effects of melatonin were suppressed either by icv injections of PI3K inhibitors and MT antagonists and by ip injection of a muscarinic receptor antagonist. We conclude that melatonin activates hypothalamus-liver communication that may contribute to circadian adjustments of gluconeogenesis. These data further suggest a physiopathological relationship between the circadian disruptions in metabolism and reduced levels of melatonin found in type 2 diabetes patients.


Asunto(s)
Antioxidantes/farmacología , Gluconeogénesis/efectos de los fármacos , Hipotálamo/metabolismo , Hígado/metabolismo , Melatonina/farmacología , Proteína Oncogénica v-akt/metabolismo , Receptor de Melatonina MT1/efectos de los fármacos , Receptor de Melatonina MT2/efectos de los fármacos , Animales , Western Blotting , Técnica del Anticuerpo Fluorescente , Prueba de Tolerancia a la Glucosa , Hipotálamo/efectos de los fármacos , Inyecciones Intraventriculares , Hígado/efectos de los fármacos , Masculino , Fosfatidilinositol 3-Quinasas/metabolismo , Ácido Pirúvico/metabolismo , Ratas , Ratas Wistar , Receptores Muscarínicos/efectos de los fármacos
12.
Endocrinology ; 153(8): 3633-45, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22585831

RESUMEN

Fructose consumption causes insulin resistance and favors hepatic gluconeogenesis through mechanisms that are not completely understood. Recent studies demonstrated that the activation of hypothalamic 5'-AMP-activated protein kinase (AMPK) controls dynamic fluctuations in hepatic glucose production. Thus, the present study was designed to investigate whether hypothalamic AMPK activation by fructose would mediate increased gluconeogenesis. Both ip and intracerebroventricular (icv) fructose treatment stimulated hypothalamic AMPK and acetyl-CoA carboxylase phosphorylation, in parallel with increased hepatic phosphoenolpyruvate carboxy kinase (PEPCK) and gluconeogenesis. An increase in AMPK phosphorylation by icv fructose was observed in the lateral hypothalamus as well as in the paraventricular nucleus and the arcuate nucleus. These effects were mimicked by icv 5-amino-imidazole-4-carboxamide-1-ß-d-ribofuranoside treatment. Hypothalamic AMPK inhibition with icv injection of compound C or with injection of a small interfering RNA targeted to AMPKα2 in the mediobasal hypothalamus (MBH) suppressed the hepatic effects of ip fructose. We also found that fructose increased corticosterone levels through a mechanism that is dependent on hypothalamic AMPK activation. Concomitantly, fructose-stimulated gluconeogenesis, hepatic PEPCK expression, and glucocorticoid receptor binding to the PEPCK gene were suppressed by pharmacological glucocorticoid receptor blockage. Altogether the data presented herein support the hypothesis that fructose-induced hypothalamic AMPK activation stimulates hepatic gluconeogenesis by increasing corticosterone levels.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Corticosterona/metabolismo , Fructosa/farmacología , Gluconeogénesis/efectos de los fármacos , Hipotálamo/metabolismo , Hígado/metabolismo , Animales , Inmunoprecipitación de Cromatina , Activación Enzimática/efectos de los fármacos , Técnica del Anticuerpo Fluorescente , Hipotálamo/efectos de los fármacos , Immunoblotting , Hígado/efectos de los fármacos , Masculino , Fosfoenolpiruvato Carboxiquinasa (ATP) , Fosforilación/efectos de los fármacos , Ratas , Ratas Wistar
13.
Diabetes ; 61(6): 1455-62, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22522614

RESUMEN

Defective liver gluconeogenesis is the main mechanism leading to fasting hyperglycemia in type 2 diabetes, and, in concert with steatosis, it is the hallmark of hepatic insulin resistance. Experimental obesity results, at least in part, from hypothalamic inflammation, which leads to leptin resistance and defective regulation of energy homeostasis. Pharmacological or genetic disruption of hypothalamic inflammation restores leptin sensitivity and reduces adiposity. Here, we evaluate the effect of a hypothalamic anti-inflammatory approach to regulating hepatic responsiveness to insulin. Obese rodents were treated by intracerebroventricular injections, with immunoneutralizing antibodies against Toll-like receptor (TLR)4 or tumor necrosis factor (TNF)α, and insulin signal transduction, hepatic steatosis, and gluconeogenesis were evaluated. The inhibition of either TLR4 or TNFα reduced hypothalamic inflammation, which was accompanied by the reduction of hypothalamic resistance to leptin and improved insulin signal transduction in the liver. This was accompanied by reduced liver steatosis and reduced hepatic expression of markers of steatosis. Furthermore, the inhibition of hypothalamic inflammation restored defective liver glucose production. All these beneficial effects were abrogated by vagotomy. Thus, the inhibition of hypothalamic inflammation in obesity results in improved hepatic insulin signal transduction, leading to reduced steatosis and reduced gluconeogenesis. All these effects are mediated by parasympathetic signals delivered by the vagus nerve.


Asunto(s)
Anticuerpos Neutralizantes/administración & dosificación , Hipotálamo/metabolismo , Inflamación/metabolismo , Resistencia a la Insulina/fisiología , Hígado/metabolismo , Receptor Toll-Like 4/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Animales , Hígado Graso/tratamiento farmacológico , Hígado Graso/metabolismo , Gluconeogénesis/efectos de los fármacos , Gluconeogénesis/fisiología , Homeostasis/efectos de los fármacos , Hipotálamo/efectos de los fármacos , Inflamación/tratamiento farmacológico , Insulina/metabolismo , Leptina/metabolismo , Hígado/efectos de los fármacos , Masculino , Ratones , Obesidad/tratamiento farmacológico , Obesidad/metabolismo , Ratas , Ratas Wistar , Transducción de Señal/efectos de los fármacos
14.
J Biol Chem ; 287(19): 15580-9, 2012 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-22427667

RESUMEN

Inflammation plays an important pathogenic role in a number of metabolic diseases such as obesity, type 2 diabetes, and atherosclerosis. The activation of inflammation in these diseases depends at least in part on the combined actions of TLR4 signaling and endoplasmic reticulum stress, which by acting in concert can boost the inflammatory response. Defining the mechanisms involved in this phenomenon may unveil potential targets for the treatment of metabolic/inflammatory diseases. Here we used LPS to induce endoplasmic reticulum stress in the human monocyte cell-line, THP-1. The unfolded protein response, produced after LPS, was dependent on CD14 activity but not on RNA-dependent protein kinase and could be inhibited by an exogenous chemical chaperone. The induction of the endoplasmic reticulum resident chaperones, GRP94 and GRP78, by LPS was of a much lower magnitude than the effect of LPS on TLR4 and MD-2 expression. In face of this apparent insufficiency of chaperone expression, we induced the expression of GRP94 and GRP78 by glucose deprivation. This approach completely reverted endoplasmic reticulum stress. The inhibition of either GRP94 or GRP78 with siRNA was sufficient to rescue the protective effect of glucose deprivation on LPS-induced endoplasmic reticulum stress. Thus, insufficient LPS-induced chaperone expression links TLR4 signaling to endoplasmic reticulum stress.


Asunto(s)
Estrés del Retículo Endoplásmico/fisiología , Chaperonas Moleculares/metabolismo , Transducción de Señal/fisiología , Receptor Toll-Like 4/metabolismo , Animales , Línea Celular , Proteínas de Unión al ADN/metabolismo , Chaperón BiP del Retículo Endoplásmico , Estrés del Retículo Endoplásmico/efectos de los fármacos , Glucosa/farmacología , Proteínas HSP70 de Choque Térmico/genética , Proteínas HSP70 de Choque Térmico/metabolismo , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Humanos , Immunoblotting , Receptores de Lipopolisacáridos/metabolismo , Lipopolisacáridos/farmacología , Antígeno 96 de los Linfocitos/metabolismo , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Chaperonas Moleculares/genética , Monocitos/citología , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Fosforilación/efectos de los fármacos , Interferencia de ARN , Transducción de Señal/efectos de los fármacos , Factores de Transcripción/metabolismo , Respuesta de Proteína Desplegada/efectos de los fármacos , Respuesta de Proteína Desplegada/fisiología , eIF-2 Quinasa/deficiencia , eIF-2 Quinasa/genética , eIF-2 Quinasa/metabolismo
15.
J Cell Biochem ; 113(4): 1182-9, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22095528

RESUMEN

Glutamate acts in the hypothalamus promoting region-, and cell-dependent effects on feeding. Part of these effects are mediated by NMDA receptors, which are up regulated in conditions known to promote increased food intake and thermogenesis, such as exposure to cold and consumption of highly caloric diets. Here, we hypothesized that at least part of the effect of glutamate on hypothalamic control of energy homeostasis would depend on the control of neurotransmitter expression and JAK2 signaling. The expression of NMDA receptors was co-localized to NPY/AgRP, POMC, CRH, and MCH but not to TRH and orexin neurons of the hypothalamus. The acute intracerebroventricular injection of glutamate promoted a dose-dependent increase in JAK2 tyrosine phosphorylation. In obese rats, 5 days intracerebroventricular treatment with glutamate resulted in the reduction of food intake, accompanied by a reduction of spontaneous motility and reduction of body mass, without affecting oxygen consumption. The reduction of food intake and body mass were partially restrained by the inhibition of JAK2. In addition, glutamate produced an increased hypothalamic expression of NPY, POMC, CART, MCH, orexin, CRH, and TRH, and the reduction of AgRP. All these effects on neurotransmitters were hindered by the inhibition of JAK2. Thus, the intracerebroventricular injection of glutamate results in the reduction of body mass through a mechanism, at least in part, dependent on JAK2, and on the broad regulation of neurotransmitter expression. These effects are not impaired by obesity, which suggest that glutamate actions in the hypothalamus may be pharmacologically explored to treat this disease.


Asunto(s)
Glutamatos/farmacología , Hipotálamo/efectos de los fármacos , Janus Quinasa 2/metabolismo , Pérdida de Peso/efectos de los fármacos , Animales , Western Blotting , Conducta Alimentaria/efectos de los fármacos , Técnica del Anticuerpo Fluorescente , Janus Quinasa 2/química , Leptina/sangre , Masculino , Fosforilación , Ratas , Ratas Wistar , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal , Tirosina/metabolismo
16.
Amino Acids ; 42(6): 2403-10, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21818562

RESUMEN

Taurine is known to modulate a number of metabolic parameters such as insulin secretion and action and blood cholesterol levels. Recent data have suggested that taurine can also reduce body adiposity in C. elegans and in rodents. Since body adiposity is mostly regulated by insulin-responsive hypothalamic neurons involved in the control of feeding and thermogenesis, we hypothesized that some of the activity of taurine in the control of body fat would be exerted through a direct action in the hypothalamus. Here, we show that the intracerebroventricular injection of an acute dose of taurine reduces food intake and locomotor activity, and activates signal transduction through the Akt/FOXO1, JAK2/STAT3 and mTOR/AMPK/ACC signaling pathways. These effects are accompanied by the modulation of expression of NPY. In addition, taurine can enhance the anorexigenic action of insulin. Thus, the aminoacid, taurine, exerts a potent anorexigenic action in the hypothalamus and enhances the effect of insulin on the control of food intake.


Asunto(s)
Ingestión de Alimentos/efectos de los fármacos , Expresión Génica/efectos de los fármacos , Hipotálamo/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Taurina/administración & dosificación , Quinasas de la Proteína-Quinasa Activada por el AMP , Acetil-CoA Carboxilasa/genética , Acetil-CoA Carboxilasa/metabolismo , Animales , Peso Corporal/efectos de los fármacos , Sinergismo Farmacológico , Ingestión de Alimentos/fisiología , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Expresión Génica/fisiología , Hipotálamo/metabolismo , Inyecciones Intraventriculares , Insulina/administración & dosificación , Janus Quinasa 2/genética , Janus Quinasa 2/metabolismo , Masculino , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuropéptido Y/genética , Neuropéptido Y/metabolismo , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Wistar , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/fisiología , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo
17.
Exp Physiol ; 97(4): 525-33, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22198013

RESUMEN

Congenitally analbuminaemic individuals and rats (NARs) exhibit several metabolic abnormalities, including hypertriglyceridaemia and plasma free fatty acid deficiency. Our aim was to study glucose homeostasis and insulin secretion in NARs. Plasma concentrations of lipids, glucose and insulin and secretion of insulin from the pancreatic islets were measured in female NARs and control animals (Sprague-Dawley rats; SDRs). Glucose homeostasis tests were also performed. Plasma glucose levels were similar between NARs and SDRs, irrespective of feeding status. However, fed insulinaemia was ∼37% higher (P 0.05) in NARs than in SDRs. The NARs displayed a markedly increased glucose tolerance, i.e. the integrated glycaemic response was one-third that of the control animals. Enhanced glucose tolerance was associated with threefold higher insulinaemia at peak glycaemia after a glucose load than in the control animals. Similar peripheral insulin sensitivity was observed between groups. Isolated pancreatic islets from NARs secreted significantly more insulin than islets from SDRs in response to a wide range of glucose concentrations (2.8-33.3 mm). Despite having similar liver glycogen contents in the fully fed state, NARs had ∼40% (P 0.05) lower glycogen contents than SDRs after 6 h fasting. The injection of a gluconeogenic substrate, pyruvate, elicited a faster rise in glycaemia in NARs compared with SDRs. Overall, NARs displayed enhanced glucose tolerance, insulin secretion and gluconeogenic flux. The higher glucose tolerance in NARs compared with SDRs is attributed to enhanced islet responsiveness to secretagogues, while peripheral insulin sensitivity seems not to be involved in this alteration. We propose that the enhanced glucose metabolism is a chronic compensatory adaptation to decreased free fatty acid availability in NARs.


Asunto(s)
Glucemia/metabolismo , Ácidos Grasos no Esterificados/sangre , Hipertrigliceridemia/sangre , Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Albúmina Sérica/deficiencia , Animales , Enfermedades Carenciales/sangre , Enfermedades Carenciales/complicaciones , Enfermedades Carenciales/diagnóstico , Femenino , Prueba de Tolerancia a la Glucosa/métodos , Hipertrigliceridemia/etiología , Secreción de Insulina , Ratas , Ratas Sprague-Dawley
18.
Cell Biochem Funct ; 30(2): 114-21, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22034157

RESUMEN

In pancreatic islets, glucose metabolism is a key process for insulin secretion, and pregnancy requires an increase in insulin secretion to compensate for the typical insulin resistance at the end of this period. Because a low-protein diet decreases insulin secretion, this type of diet could impair glucose homeostasis, leading to gestational diabetes. In pancreatic islets, we investigated GLUT2, glucokinase and hexokinase expression patterns as well as glucose uptake, utilization and oxidation rates. Adult control non-pregnant (CNP) and control pregnant (CP) rats were fed a normal protein diet (17%), whereas low-protein non-pregnant (LPNP) and low-protein pregnant (LPP) rats were fed a low-protein diet (6%) from days 1 to 15 of pregnancy. The insulin secretion in 2.8 mmol l(-1) of glucose was higher in islets from LPP rats than that in islets from CP, CNP and LPNP rats. Maximal insulin release was obtained at 8.3 and 16.7 mmol l(-1) of glucose in LPP and CP groups, respectively. The glucose dose-response curve from LPNP group was shifted to the right in relation to the CNP group. In the CP group, the concentration-response curve to glucose was shifted to the left compared with the CNP group. The LPP groups exhibited an "inverted U-shape" dose-response curve. The alterations in the GLUT2, glucokinase and hexokinase expression patterns neither impaired glucose metabolism nor correlated with glucose islet sensitivity, suggesting that ß-cell sensitivity to glucose requires secondary events other than the observed metabolic/molecular events.


Asunto(s)
Diabetes Gestacional/metabolismo , Dieta con Restricción de Proteínas/efectos adversos , Glucosa/metabolismo , Insulina/metabolismo , Animales , Diabetes Gestacional/enzimología , Diabetes Gestacional/etiología , Diabetes Gestacional/genética , Femenino , Glucoquinasa/genética , Glucoquinasa/metabolismo , Transportador de Glucosa de Tipo 2/genética , Transportador de Glucosa de Tipo 2/metabolismo , Hexoquinasa/genética , Hexoquinasa/metabolismo , Humanos , Secreción de Insulina , Islotes Pancreáticos/metabolismo , Embarazo , Ratas , Ratas Wistar
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...