Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
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.
Am J Physiol Endocrinol Metab ; 324(2): E154-E166, 2023 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-36598900

RESUMEN

Maternal obesity is an important risk factor for obesity, cardiovascular, and metabolic diseases in the offspring. Studies have shown that it leads to hypothalamic inflammation in the progeny, affecting the function of neurons regulating food intake and energy expenditure. In adult mice fed a high-fat diet, one of the hypothalamic abnormalities that contribute to the development of obesity is the damage of the blood-brain barrier (BBB) at the median eminence-arcuate nucleus (ME-ARC) interface; however, how the hypothalamic BBB is affected in the offspring of obese mothers requires further investigation. Here, we used confocal and transmission electron microscopy, transcript expression analysis, glucose tolerance testing, and a cross-fostering intervention to determine the impact of maternal obesity and breastfeeding on BBB integrity at the ME-ARC interface. The offspring of obese mothers were born smaller; conversely, at weaning, they presented larger body mass and glucose intolerance. In addition, maternal obesity-induced structural and functional damage of the offspring's ME-ARC BBB. By a cross-fostering intervention, some of the defects in barrier integrity and metabolism seen during development in an obesogenic diet were recovered. The offspring of obese dams breastfed by lean dams presented a reduction of body mass and glucose intolerance as compared to the offspring continuously exposed to an obesogenic environment during intrauterine and perinatal life; this was accompanied by partial recovery of the anatomical structure of the ME-ARC interface, and by the normalization of transcript expression of genes coding for hypothalamic neurotransmitters involved in energy balance and BBB integrity. Thus, maternal obesity promotes structural and functional damage of the hypothalamic BBB, which is, in part, reverted by lactation by lean mothers.NEW & NOTEWORTHY Maternal dietary habits directly influence offspring health. In this study, we aimed at determining the impact of maternal obesity on BBB integrity. We show that DIO offspring presented a leakier ME-BBB, accompanied by changes in the expression of transcripts encoding for endothelial and tanycytic proteins, as well as of hypothalamic neuropeptides. Breastfeeding in lean dams was sufficient to protect the offspring from ME-BBB disruption, providing a preventive strategy of nutritional intervention during early life.


Asunto(s)
Intolerancia a la Glucosa , Obesidad Materna , Humanos , Femenino , Animales , Ratones , Embarazo , Barrera Hematoencefálica/metabolismo , Eminencia Media/metabolismo , Obesidad Materna/metabolismo , Madres , Intolerancia a la Glucosa/metabolismo , Obesidad/metabolismo , Hipotálamo/metabolismo , Dieta Alta en Grasa/efectos adversos , Fenómenos Fisiologicos Nutricionales Maternos
3.
Clin Sci (Lond) ; 133(22): 2345-2360, 2019 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-31722009

RESUMEN

There is no consensus on the effects of omega-3 (ω-3) fatty acids (FA) on cutaneous repair. To solve this problem, we used 2 different approaches: (1) FAT-1 transgenic mice, capable of producing endogenous ω-3 FA; (2) wild-type (WT) mice orally supplemented with DHA-enriched fish oil. FAT-1 mice had higher systemic (serum) and local (skin tissue) ω-3 FA levels, mainly docosahexaenoic acid (DHA), in comparison with WT mice. FAT-1 mice had increased myeloperoxidase (MPO) activity and content of CXCL-1 and CXCL-2, and reduced IL-10 in the skin wound tissue three days after the wound induction. Inflammation was maintained by an elevated TNF-α concentration and presence of inflammatory cells and edema. Neutrophils and macrophages, isolated from FAT-1 mice, also produced increased TNF-α and reduced IL-10 levels. In these mice, the wound closure was delayed, with a wound area 6-fold bigger in relation with WT group, on the last day of analysis (14 days post-wounding). This was associated with poor orientation of collagen fibers and structural aspects in repaired tissue. Similarly, DHA group had a delay during late inflammatory phase. This group had increased TNF-α content and CD45+F4/80+ cells at the third day after skin wounding and increased concentrations of important metabolites derived from ω-3, like 18-HEPE, and reduced concentrations of those from ω-6 FA. In conclusion, elevated DHA content, achieved in both FAT-1 and DHA groups, slowed inflammation resolution and impaired the quality of healed skin tissue.


Asunto(s)
Ácidos Docosahexaenoicos/fisiología , Cicatrización de Heridas , Animales , Caenorhabditis elegans , Proteínas de Caenorhabditis elegans/genética , Suplementos Dietéticos , Ácido Graso Desaturasas/genética , Inflamación , Macrófagos/fisiología , Masculino , Ratones Transgénicos , Neutrófilos/fisiología , Piel/metabolismo
4.
Endocrinology ; 157(12): 4803-4816, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27732087

RESUMEN

A temporary and reversible inhibition of the hypothalamo-pituitary-gonadal axis is adaptive when energy reserves are diminished, allowing individual survival and energy accumulation for eventual reproduction. The AMP-activated protein kinase (AMPK) works as a cellular sensor of the AMP to ATP ratio and ultimately of energy availability. Activation of AMPK suppresses ATP-consuming processes and stimulates ATP-producing pathways. The AMPK α2 catalytic subunit is expressed in multiple hypothalamic nuclei including those associated with reproductive control, ie, the anteroventral periventricular nucleus and the arcuate nucleus. Subsets of kisspeptin neurons in the anteroventral periventricular nucleus (20% in females) and arcuate nucleus (45% in males and 65% in females) coexpress AMPKα2 mRNA. Using the Cre-loxP approach, we assessed whether AMPKα2 in Kiss1 cells is required for body weight and reproductive function. The AMPKα2-deleted mice show no difference in body weight and time for sexual maturation compared with controls. Males and females are fertile and have normal litter size. The AMPKα2-deleted and control females have similar estradiol feedback responses and show no difference in Kiss1 mRNA expression after ovariectomy or ovariectomy plus estradiol replacement. In males, acute fasting decreased Kiss1 mRNA expression in both groups, but no effect was observed in females. However, after an acute fasting, control mice displayed prolonged diestrous phase, but AMPKα2-deleted females showed no disruption of estrous cycles. Our findings demonstrate that the AMPKα2 catalytic subunit in Kiss1 cells is dispensable for body weight and reproductive function in mice but is necessary for the reproductive adaptations to conditions of acute metabolic distress.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Ayuno/metabolismo , Hipotálamo/metabolismo , Kisspeptinas/metabolismo , Neuronas/metabolismo , Maduración Sexual/fisiología , Animales , Peso Corporal/fisiología , Ciclo Estral/metabolismo , Femenino , Masculino , Ratones , Ratones Noqueados
5.
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
6.
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
7.
PLoS One ; 8(4): e62669, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23626844

RESUMEN

BACKGROUND: Hypothalamic AMPK acts as a cell energy sensor and can modulate food intake, glucose homeostasis, and fatty acid biosynthesis. Intrahypothalamic fatty acid injection is known to suppress liver glucose production, mainly by activation of hypothalamic ATP-sensitive potassium (K(ATP)) channels. Since all models employed seem to involve malonyl-CoA biosynthesis, we hypothesized that acetyl-CoA carboxylase can modulate the counter-regulatory response independent of nutrient availability. METHODOLOGY/PRINCIPAL FINDINGS: In this study employing immunoblot, real-time PCR, ELISA, and biochemical measurements, we showed that reduction of the hypothalamic expression of acetyl-CoA carboxylase by antisense oligonucleotide after intraventricular injection increased food intake and NPY mRNA, and diminished the expression of CART, CRH, and TRH mRNA. Additionally, as in fasted rats, in antisense oligonucleotide-treated rats, serum glucagon and ketone bodies increased, while the levels of serum insulin and hepatic glycogen diminished. The reduction of hypothalamic acetyl-CoA carboxylase also increased PEPCK expression, AMPK phosphorylation, and glucose production in the liver. Interestingly, these effects were observed without modification of hypothalamic AMPK phosphorylation. CONCLUSION/SIGNIFICANCE: Hypothalamic ACC inhibition can activate hepatic counter-regulatory response independent of hypothalamic AMPK activation.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Acetil-CoA Carboxilasa/genética , Hipotálamo/metabolismo , Hígado/metabolismo , Acetil-CoA Carboxilasa/metabolismo , Animales , Peso Corporal , Dieta , Regulación de la Expresión Génica , Gluconeogénesis/fisiología , Hormonas/sangre , Masculino , Oligonucleótidos/administración & dosificación , Fosforilación , Ratas
8.
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
9.
Exp Biol Med (Maywood) ; 236(10): 1147-55, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21844120

RESUMEN

Hydroalcoholic extract of Solidago chilensis (Sc) is employed in popular medicine to treat inflammatory disease. The low-grade proinflammatory state and the activation of serine/threonine kinases in adipose tissue, like c-jun kinase (JNK) and IKK, and transcription factors, have an important role in obesity-associated insulin resistance. The aim of this study was to further investigate the effects of the Sc extract on glucose homeostasis in diet-induced obesity mice. Male Swiss mice were randomized to three groups: a control group (C) fed with standard laboratory chow; a group with an experimental high-fat diet (HFD); and a group fed with a high-fat (45% kcal from fat) diet + extract of Sc (via intraperitoneal, 3 mg/kg) (ScHFD). The dietary treatment lasted for eight weeks. Subsequently, the expression and phosphorylation of proteins of interest in the liver, hypothalamus and skeletal muscle were evaluated by Western blot analysis. Body weight, epididymal fat pad mass and liver triglycerides were higher in HFD than in control mice, but these parameters were reduced by intraperitoneal administration of the extracts (3 mg/kg) to the HFD group. AKT phosphorylation stimulated by insulin in the liver, hypothalamus and skeletal muscle was higher in ScHFD as compared with HFD mice. Additionally, liver expression of phosphoenolpyruvate carboxykinase (PEPCK) and fatty acid synthase were lower in ScHFD as compared with HFD mice. Nuclear factor κB, p-IκB and p-JNK levels were higher in HFD when compared with control mice, but they were lowered by treatment with extract (ScHFD). In addition, in db/db mice, Sc extract also improved liver AKT phosphorylation stimulated by insulin and reduced PEPCK expression. The data presented herein show that Sc improves AKT activation. This effect may be promoted by reduction of the proinflammatory pathway in the liver and hypothalamus. Therefore, systemic action of the Sc components may contribute to improve obesity-associated pathophysiology.


Asunto(s)
Proteínas I-kappa B/metabolismo , Resistencia a la Insulina , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Obesidad/metabolismo , Extractos Vegetales/uso terapéutico , Solidago , Animales , Glucosa/análisis , Prueba de Tolerancia a la Glucosa , Proteínas I-kappa B/efectos de los fármacos , Proteínas Quinasas JNK Activadas por Mitógenos/efectos de los fármacos , Hígado/química , Hígado/metabolismo , Glucógeno Hepático/análisis , Masculino , Ratones , Transducción de Señal/efectos de los fármacos , Triglicéridos/análisis
10.
Vitam Horm ; 82: 129-43, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20472136

RESUMEN

Obesity is one of the most prevalent diseases in the modern world. It results from the progressive loss of balance between food intake and whole body energy expenditure. Recent studies have shown that consumption of fat-rich diets induces hypothalamic inflammation and dysfunction which is characterized by defective response to anorexygenic and thermogenic hormones, such as leptin and insulin, leading to anomalous neurotransmitter production and favoring body mass gain. In this chapter, we present the main recent advances in this rapidly evolving field, focusing on the role of hypothalamic inflammation on the genesis of obesity.


Asunto(s)
Hipotálamo/fisiopatología , Inflamación/fisiopatología , Obesidad/fisiopatología , Ingestión de Energía , Metabolismo Energético , Conducta Alimentaria , Humanos , Inflamación/complicaciones , Obesidad/complicaciones
11.
Mol Cell Endocrinol ; 314(1): 62-9, 2010 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-19698760

RESUMEN

Central leptin action requires PI3K activity to modulate glucose homeostasis and peripheral metabolism. However, the mechanism behind this phenomenon is not clearly understood. We hypothesize that hypothalamic PI3K activity is important for the modulation of the AMP-activated protein kinase (AMPK)/acetyl-CoA carboxylase (ACC) pathway, PGC1 alpha, and AKT in skeletal muscle (SM). To address this issue, we injected leptin into the lateral ventricle of rats. Hypothalamic JAK2 and AKT were activated by intracerebroventricular (ICV) injection of leptin in a time-dependent manner. Central leptin improved tolerance to glucose (GTT), increased PGC1 alpha expression, and AKT, AMPK, ACC and JAK2 phosphorylation in the soleus muscle. Previous ICV administration of either LY294002 or propranolol (IP) blocked these effects. We concluded that the activation of the hypothalamic PI3K pathway is important for leptin-induced AKT phosphorylation, as well as for active catabolic pathway through AMPK and PGC1 alpha in SM. Thus, a defective leptin signalling PI3K pathway in the hypothalamus may contribute to peripheral resistance to insulin associated to diet-induced obesity.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Hipotálamo , Leptina/metabolismo , Músculo Esquelético/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas de Unión al ARN/metabolismo , Factores de Transcripción/metabolismo , Proteínas Quinasas Activadas por AMP/genética , Antagonistas Adrenérgicos beta/metabolismo , Animales , Cromonas/metabolismo , Metabolismo Energético , Glucosa/metabolismo , Homeostasis , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Insulina/metabolismo , Janus Quinasa 2/genética , Janus Quinasa 2/metabolismo , Leptina/farmacología , Masculino , Morfolinas/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Fosfatidilinositol 3-Quinasas/genética , Inhibidores de las Quinasa Fosfoinosítidos-3 , Propranolol/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas de Unión al ARN/genética , Ratas , Ratas Wistar , Transducción de Señal/fisiología , Factores de Transcripción/genética
12.
Rev Neurosci ; 20(5-6): 441-9, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-20397624

RESUMEN

The prevalence of obesity has grown to an alarming magnitude, affecting more than 300 million humans worldwide. Although in most instances obesity is caused by excessive caloric consumption, only recently have we begun to understand the mechanisms involved in the loss of balance between caloric intake and energy expenditure. In the hypothalamus, groups of specialized neurons provide the signals that, under physiological conditions, determine the stability of body mass. Recent studies have shown that under certain environmental and genetic conditions, this equilibrium is lost and body adiposity may increase. Here, we review the work that provided the basis for the current understanding of hypothalamic dysfunction and the genesis of obesity.


Asunto(s)
Hipotálamo/fisiopatología , Obesidad/patología , Animales , Humanos , Hipotálamo/metabolismo , Insulina/metabolismo , Leptina/metabolismo
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.
J Endocrinol ; 198(1): 157-68, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18469022

RESUMEN

We hypothesized that citrate might modulate the AMP-activated protein kinase/acetyl-CoA carboxylase (AMPK)/(ACC) pathway and participate in neuronal feeding control and glucose homeostasis. To address this issue, we injected citrate into the lateral ventricle of rats. Intracerebroventricular (ICV) injection of citrate diminished the phosphorylation of hypothalamic AMPK/ACC, increased the expression of anorexigenic neuropeptide (pro-opiomelanocortin and corticotropin-releasing hormone), elevated the level of malonyl-CoA in the hypothalamus, and reduced food intake. No change was observed in the concentration of blood insulin after the injection of citrate. With a euglycemic-hyperinsulinemic clamp, the glucose infusion rate was higher in the citrate group than in the control group (28.6+/-0.8 vs 19.3+/-0.2 mU/kg body weight/min respectively), and so was glucose uptake in skeletal muscle and the epididymal fat pad. Concordantly, insulin receptor (IR), IR substrate type 1 (IRS1), IRS2, and protein kinase B (AKT) phosphorylation in adipose tissue and skeletal muscle was improved by citrate ICV treatment. Moreover, the treatment with citrate for 7 days promoted body weight loss and decreased the adipose tissue. Our results suggest that citrate and glucose may serve as signals of energy and nutrient availability to hypothalamic cells.


Asunto(s)
Proteínas Quinasas Activadas por AMP/antagonistas & inhibidores , Ácido Cítrico/farmacología , Conducta Alimentaria/efectos de los fármacos , Hipotálamo/enzimología , Insulina/farmacología , Transducción de Señal/efectos de los fármacos , Proteínas Quinasas Activadas por AMP/fisiología , Acetil-CoA Carboxilasa/fisiología , Animales , Ácido Cítrico/administración & dosificación , Inyecciones Intraventriculares , Masculino , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Wistar
15.
Peptides ; 28(5): 1050-8, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17459524

RESUMEN

Acting in the hypothalamus, tumor necrosis factor-alpha (TNF-alpha) produces a potent anorexigenic effect. However, the molecular mechanisms involved in this phenomenon are poorly characterized. In this study, we investigate the capacity of TNF-alpha to activate signal transduction in the hypothalamus through elements of the pathways employed by the anorexigenic hormones insulin and leptin. High dose TNF-alpha promotes a reduction of 25% in 12h food intake, which is an inhibitory effect that is marginally inferior to that produced by insulin and leptin. In addition, high dose TNF-alpha increases body temperature and respiratory quotient, effects not reproduced by insulin or leptin. TNF-alpha, predominantly at the high dose, is also capable of activating canonical pro-inflammatory signal transduction in the hypothalamus, inducing JNK, p38, and NFkappaB, which results in the transcription of early responsive genes and expression of proteins of the SOCS family. Also, TNF-alpha activates signal transduction through JAK-2 and STAT-3, but does not activate signal transduction through early and intermediary elements of the insulin/leptin signaling pathways such as IRS-2, Akt, ERK and FOXO1. When co-injected with insulin or leptin, TNF-alpha, at both high and low doses, partially impairs signal transduction through IRS-2, Akt, ERK and FOXO1 but not through JAK-2 and STAT-3. This effect is accompanied by the partial inhibition of the anorexigenic effects of insulin and leptin, when the low, but not the high dose of TNF-alpha is employed. In conclusion, TNF-alpha, on a dose-dependent way, modulates insulin and leptin signaling and action in the hypothalamus.


Asunto(s)
Respiración de la Célula/efectos de los fármacos , Ingestión de Alimentos/efectos de los fármacos , Hipotálamo/efectos de los fármacos , Insulina/metabolismo , Leptina/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Animales , Relación Dosis-Respuesta a Droga , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Factores de Transcripción Forkhead/metabolismo , Hipotálamo/metabolismo , Immunoblotting , Inmunoprecipitación , Insulina/administración & dosificación , Insulina/farmacología , Janus Quinasa 2/metabolismo , Leptina/administración & dosificación , Leptina/farmacología , Masculino , Proteínas del Tejido Nervioso/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Wistar , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Factor de Necrosis Tumoral alfa/administración & dosificación
16.
J Physiol ; 568(Pt 3): 993-1001, 2005 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-16141267

RESUMEN

The exposure of homeothermic animals to a cold environment leads to a powerful activation of orexigenic signalling which is accompanied by molecular and functional resistance to insulin-induced inhibition of feeding. Recent evidence suggests that AMPK participates in nutrient-dependent control of satiety and adiposity. The objective of the present study was to evaluate the effect of cold exposure upon the molecular activation of AMPK signalling in the hypothalamus of rats. Immunoblotting demonstrated that cold exposure per se is sufficient for inducing, on a time-dependent basis, the molecular activation of the serine/threonine kinase AMP-activated protein kinase (AMPK) and inactivation of the acetyl-CoA carboxylase (ACC). These molecular phenomena were accompanied by resistance to nutrient-induced inactivation of AMPK and activation of ACC. Moreover, cold-exposure led to a partial inhibition of a feeding-induced anorexigenic response, which was paralleled by resistance to insulin-induced suppression of feeding. Finally, cold exposure significantly impaired insulin-induced inhibition of AMPK through a mechanism dependent on the molecular cross-talk between phosphatidylinositol-3(PI3)-kinase/Akt and AMPK. In conclusion, increased feeding during cold exposure results, at least in part, from resistance to insulin- and nutrient-dependent anorexigenic signalling in the hypothalamus.


Asunto(s)
Anorexia/fisiopatología , Frío , Conducta Alimentaria/fisiología , Hipotálamo/fisiopatología , Inmunidad Innata , Complejos Multienzimáticos/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal , Proteínas Quinasas Activadas por AMP , Adaptación Fisiológica , Animales , Activación Enzimática , Resistencia a la Insulina , Masculino , Fenómenos Fisiológicos de la Nutrición , Ratas , Ratas Wistar
17.
Obes Res ; 13(1): 48-57, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15761162

RESUMEN

OBJECTIVE: To investigate whether insulin and leptin share common intracellular signal transduction pathways and to determine whether these hormonal signaling systems modulate each other's action in rat hypothalamus. RESEARCH METHODS AND PROCEDURES: Male Wistar rats were studied after chronic implantation of an intracerebroventricular catheter into the third ventricle. Immunoprecipitation and immunoblotting were used to examine the activation of insulin and leptin signaling molecules in the rat hypothalamus. RESULTS: Insulin alone is able to produce molecular activation of insulin receptor substrates (IRSs)/phosphatidylinositol 3-kinase (PI 3-kinase)/Akt and mitogen-activated protein (MAP) kinase signaling pathways in hypothalamus, whereas leptin alone activates MAP kinase and IRSs/PI 3-kinase signaling with no effect on Akt. Combined infusion of leptin and insulin provokes a dual action. There was no quantitative potentialization of any single hormone's action on the elements of the insulin signaling pathway, IRSs/PI 3-kinase/Akt, and MAP kinase. Conversely, leptin plus insulin leads to quantitative potentialization of molecular signaling through the Janus kinase/signal transducer and activator of transcription pathway. DISCUSSION: We provide evidence for a convergence of leptin and insulin signaling at the level of IRSs-PI 3-kinase and a divergence at the level of Akt. Moreover, our results indicate a direct and positive cross-talk between insulin and leptin at the level of Janus kinase 2 and signal transducer and activator of transcription 3 tyrosine phosphorylation. This mechanism may serve to potentiate the activity of both insulin and leptin pathways and to increase stimulation in physiological processes such as the control of food intake and body weight, which are under the combined control of insulin and leptin.


Asunto(s)
Hipotálamo/fisiología , Insulina/fisiología , Leptina/fisiología , Animales , Western Blotting , Proteínas de Unión al ADN/fisiología , Ingestión de Alimentos/fisiología , Inyecciones Intraventriculares , Proteínas Sustrato del Receptor de Insulina , Péptidos y Proteínas de Señalización Intracelular , Masculino , Proteínas Quinasas Activadas por Mitógenos/fisiología , Fosfatidilinositol 3-Quinasas/fisiología , Fosfoproteínas/fisiología , Fosforilación , Proteínas Serina-Treonina Quinasas/fisiología , Proteínas Proto-Oncogénicas/fisiología , Proteínas Proto-Oncogénicas c-akt , Ratas , Ratas Wistar , Factor de Transcripción STAT3 , Transducción de Señal/fisiología , Transactivadores/fisiología
18.
J Endocrinol ; 181(1): 117-28, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15072572

RESUMEN

Angiotensin II exerts a potent dypsogenic stimulus on the hypothalamus, which contributes to its centrally mediated participation in the control of water balance and blood pressure. Repetitive intracerebroventricular (i.c.v.) injections of angiotensin II lead to a loss of effect characterized as physiological desensitization to the peptide's action. In the present study, we demonstrate that angiotensin II induces the expression of suppressor of cytokine signaling (SOCS)-3 via angiotensin receptor 1 (AT1) and JAK-2, mostly located at the median preoptic lateral and anterodorsal preoptic nuclei. SOCS-3 produces an inhibitory effect upon the signal transduction pathways of several cytokines and hormones that employ members of the JAK/STAT families as intermediaries. The partial inhibition of SOCS-3 translation by antisense oligonucleotide was sufficient to significantly reduce the refractoriness of repetitive i.c.v. angiotensin II injections, as evaluated by water ingestion. Thus, by acting through AT1 on the hypothalamus, angiotensin II induces the expression of SOCS-3 which, in turn, blocks further activation of the pathway and consequently leads to desensitization to angiotensin II stimuli concerning its dypsogenic effect.


Asunto(s)
Angiotensina II/farmacología , Conducta de Ingestión de Líquido/efectos de los fármacos , Hipotálamo/metabolismo , Proteínas Proto-Oncogénicas , Proteínas Represoras/metabolismo , Transducción de Señal/efectos de los fármacos , Factores de Transcripción/metabolismo , Animales , Depresión Química , Hipotálamo/química , Hipotálamo/efectos de los fármacos , Inmunohistoquímica , Inyecciones Intraventriculares , Janus Quinasa 2 , Masculino , Oligonucleótidos Antisentido/farmacología , Proteínas Tirosina Quinasas/metabolismo , ARN Mensajero/análisis , Ratas , Ratas Wistar , Receptor de Angiotensina Tipo 1/metabolismo , Proteínas Represoras/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína 3 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas , Factores de Transcripción/genética
19.
Endocrinology ; 144(11): 4831-40, 2003 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-12960043

RESUMEN

Short-term cold exposure of homeothermic animals leads to higher thermogenesis and food consumption accompanied by weight loss. An analysis of cDNA-macroarray was employed to identify candidate mRNA species that encode proteins involved in thermogenic adaptation to cold. A cDNA-macroarray analysis, confirmed by RT-PCR, immunoblot, and RIA, revealed that the hypothalamic expression of melanin-concentrating hormone (MCH) is enhanced by exposure of rats to cold environment. The blockade of hypothalamic MCH expression by antisense MCH oligonucleotide in cold-exposed rats promoted no changes in feeding behavior and body temperature. However, MCH blockade led to a significant drop in body weight, which was accompanied by decreased liver glycogen, increased relative body fat, increased absolute and relative interscapular brown adipose tissue mass, increased uncoupling protein 1 expression in brown adipose tissue, and increased consumption of lean body mass. Thus, increased hypothalamic MCH expression in rats exposed to cold may participate in the process that allows for efficient use of energy for heat production during thermogenic adaptation to cold.


Asunto(s)
Frío , Metabolismo Energético/fisiología , Hormonas Hipotalámicas/fisiología , Hipotálamo/metabolismo , Melaninas/fisiología , Hormonas Hipofisarias/fisiología , Adaptación Fisiológica , Tejido Adiposo Pardo/metabolismo , Animales , Composición Corporal , Regulación de la Temperatura Corporal , Peso Corporal/fisiología , Proteínas Portadoras/metabolismo , Ingestión de Alimentos/fisiología , Perfilación de la Expresión Génica , Glucógeno/metabolismo , Hormonas Hipotalámicas/metabolismo , Canales Iónicos , Hígado/metabolismo , Masculino , Melaninas/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Mitocondriales , Músculo Esquelético/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Consumo de Oxígeno/fisiología , Hormonas Hipofisarias/metabolismo , Ratas , Ratas Wistar , Proteína Desacopladora 1
20.
Am J Physiol Endocrinol Metab ; 285(1): E216-23, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12644444

RESUMEN

Insulin and leptin act in the hypothalamus, providing robust anorexigenic signals. The exposure of homeothermic animals to a cold environment leads to increased feeding, accompanied by sustained low levels of insulin and leptin. In the present study, the initial and intermediate steps of the insulin-signaling cascade were evaluated in the hypothalamus of cold-exposed Wistar rats. By immunohistochemistry, most insulin receptor (IR) and insulin receptor substrate-2 (IRS-2) immunoreactivity localized to the arcuate nucleus. Basal levels of tyrosine phosphorylation of IR and IRS-2 were increased in cold-exposed rats compared with rats maintained at room temperature. However, after an acute, peripheral infusion of exogenous insulin, significantly lower increases of IR and IRS-2 tyrosine phosphorylation were detected in the hypothalamus of cold-exposed rats. Insulin-induced association of p85/phosphatidylinositol 3-kinase with IRS-2, Ser473 phosphorylation of Akt, and tyrosine phosphorylation of ERK was significantly reduced in the hypothalamus of cold-exposed rats. To test the hypothesis of functional impairment of insulin signaling in the hypothalamus, intracerebroventricularly cannulated rats were acutely treated with insulin, and food ingestion was measured over a period of 12 h. Cold-exposed animals presented a significantly lower insulin-induced reduction in food consumption compared with animals maintained at room temperature. Hence, the present studies reveal that animals exposed to cold are resistant, both at the molecular and the functional level, to the actions of insulin in the hypothalamus.


Asunto(s)
Frío , Hipotálamo/fisiología , Resistencia a la Insulina/fisiología , Animales , Glucemia/metabolismo , Ingestión de Alimentos/efectos de los fármacos , Ácidos Grasos no Esterificados/sangre , Hormonas/sangre , Hipoglucemiantes/administración & dosificación , Hipoglucemiantes/farmacología , Immunoblotting , Inyecciones Intraventriculares , Insulina/administración & dosificación , Insulina/farmacología , Masculino , Proteínas Quinasas Activadas por Mitógenos/biosíntesis , Fosforilación , Pruebas de Precipitina , Ratas , Ratas Wistar , Respuesta de Saciedad/fisiología , Serina/metabolismo , Transducción de Señal/fisiología , Tirosina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA