Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Sci Rep ; 14(1): 11839, 2024 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-38782973

RESUMEN

The intestinal extracellular matrix (ECM) helps maintain appropriate tissue barrier function and regulate host-microbial interactions. Chondroitin sulfate- and dermatan sulfate-glycosaminoglycans (CS/DS-GAGs) are integral components of the intestinal ECM, and alterations in CS/DS-GAGs have been shown to significantly influence biological functions. Although pathologic ECM remodeling is implicated in inflammatory bowel disease (IBD), it is unknown whether changes in the intestinal CS/DS-GAG composition are also linked to IBD in humans. Our aim was to characterize changes in the intestinal ECM CS/DS-GAG composition in intestinal biopsy samples from patients with IBD using mass spectrometry. We characterized intestinal CS/DS-GAGs in 69 pediatric and young adult patients (n = 13 control, n = 32 active IBD, n = 24 IBD in remission) and 6 adult patients. Here, we report that patients with active IBD exhibit a significant decrease in the relative abundance of CS/DS isomers associated with matrix stability (CS-A and DS) compared to controls, while isomers implicated in matrix instability and inflammation (CS-C and CS-E) were significantly increased. This imbalance of intestinal CS/DS isomers was restored among patients in clinical remission. Moreover, the abundance of pro-stabilizing CS/DS isomers negatively correlated with clinical disease activity scores, whereas both pro-inflammatory CS-C and CS-E content positively correlated with disease activity scores. Thus, pediatric patients with active IBD exhibited increased pro-inflammatory and decreased pro-stabilizing CS/DS isomer composition, and future studies are needed to determine whether changes in the CS/DS-GAG composition play a pathogenic role in IBD.


Asunto(s)
Sulfatos de Condroitina , Glicosaminoglicanos , Enfermedades Inflamatorias del Intestino , Humanos , Enfermedades Inflamatorias del Intestino/metabolismo , Enfermedades Inflamatorias del Intestino/patología , Sulfatos de Condroitina/metabolismo , Masculino , Femenino , Adulto , Adolescente , Niño , Glicosaminoglicanos/metabolismo , Adulto Joven , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Matriz Extracelular/metabolismo , Intestinos/patología
2.
Diabetes ; 72(9): 1207-1213, 2023 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-37347793

RESUMEN

To investigate whether glucoregulatory neurons in the hypothalamus can sense and respond to physiological variation in the blood glucose (BG) level, we combined continuous arterial glucose monitoring with continuous measures of the activity of a specific subset of neurons located in the hypothalamic ventromedial nucleus that express pituitary adenylate cyclase activating peptide (VMNPACAP neurons) obtained using fiber photometry. Data were collected in conscious, free-living mice during a 1-h baseline monitoring period and a subsequent 2-h intervention period during which the BG level was raised either by consuming a chow or a high-sucrose meal or by intraperitoneal glucose injection. Cross-correlation analysis revealed that, following a 60- to 90-s delay, interventions that raise the BG level reliably associate with reduced VMNPACAP neuron activity (P < 0.01). In addition, a strong positive correlation between BG and spontaneous VMNPACAP neuron activity was observed under basal conditions but with a much longer (∼25 min) temporal offset, consistent with published evidence that VMNPACAP neuron activation raises the BG level. Together, these findings are suggestive of a closed-loop system whereby VMNPACAP neuron activation increases the BG level; detection of a rising BG level, in turn, feeds back to inhibit these neurons. To our knowledge, these findings constitute the first evidence of a role in glucose homeostasis for glucoregulatory neurocircuits that, like pancreatic ß-cells, sense and respond to physiological variation in glycemia. ARTICLE HIGHLIGHTS: By combining continuous arterial glucose monitoring with fiber photometry, studies investigated whether neurons in the murine ventromedial nucleus that express pituitary adenylate cyclase activating peptide (VMNPACAP neurons) detect and respond to changes in glycemia in vivo. VMNPACAP neuron activity rapidly decreases (within <2 min) when the blood glucose level is raised by either food consumption or glucose administration. Spontaneous VMNPACAP neuron activity also correlates positively with glycemia, but with a longer temporal offset, consistent with reports that hyperglycemia is induced by experimental activation of these neurons. Like pancreatic ß-cells, neurons in the hypothalamic ventromedial nucleus appear to sense and respond to physiological variation in glycemia.


Asunto(s)
Automonitorización de la Glucosa Sanguínea , Glucemia , Ratones , Animales , Glucemia/análisis , Adenilil Ciclasas , Hipotálamo , Glucosa , Neuronas/fisiología , Péptidos
3.
Elife ; 92020 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-33320088

RESUMEN

To maintain energy homeostasis during cold exposure, the increased energy demands of thermogenesis must be counterbalanced by increased energy intake. To investigate the neurobiological mechanisms underlying this cold-induced hyperphagia, we asked whether agouti-related peptide (AgRP) neurons are activated when animals are placed in a cold environment and, if so, whether this response is required for the associated hyperphagia. We report that AgRP neuron activation occurs rapidly upon acute cold exposure, as do increases of both energy expenditure and energy intake, suggesting the mere perception of cold is sufficient to engage each of these responses. We further report that silencing of AgRP neurons selectively blocks the effect of cold exposure to increase food intake but has no effect on energy expenditure. Together, these findings establish a physiologically important role for AgRP neurons in the hyperphagic response to cold exposure.


Asunto(s)
Proteína Relacionada con Agouti/metabolismo , Frío , Conducta Alimentaria/fisiología , Hiperfagia/fisiopatología , Termogénesis/fisiología , Animales , Ingestión de Alimentos/fisiología , Homeostasis/fisiología , Masculino , Ratones , Neuronas/fisiología
4.
J Vis Exp ; (159)2020 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-32449706

RESUMEN

Stereotactic surgery is an essential tool in the modern neuroscience lab. However, the ability to precisely and accurately target difficult-to-reach brain regions still presents a challenge, particularly when targeting brain structures along the midline. These challenges include avoiding of the superior sagittal sinus and third ventricle and the ability to consistently target selective and discrete brain nuclei. In addition, more advanced neuroscience techniques (e.g., optogenetics, fiber photometry, and two-photon imaging) rely on targeted implantation of significant hardware to the brain, and spatial limitations are a common hindrance. Presented here is a modifiable protocol for stereotactic targeting of rodent brain structures using an angled coronal approach. It can be adapted to 1) mouse or rat models, 2) various neuroscience techniques, and 3) multiple brain regions. As a representative example, it includes the calculation of stereotactic coordinates for targeting of the mouse hypothalamic ventromedial nucleus (VMN) for an optogenetic inhibition experiment. This procedure begins with the bilateral microinjection of an adeno-associated virus (AAV) encoding a light-sensitive chloride channel (SwiChR++) to a Cre-dependent mouse model, followed by the angled bilateral implantation of fiberoptic cannulae. Using this approach, findings show that activation of a subset of VMN neurons is required for intact glucose counterregulatory responses to insulin-induced hypoglycemia.


Asunto(s)
Neurociencias/instrumentación , Técnicas Estereotáxicas/instrumentación , Animales , Modelos Animales de Enfermedad , Ratones , Ratas
5.
PLoS One ; 14(4): e0215601, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31017943

RESUMEN

During periods in which glucose absorption from the gastrointestinal (GI) tract is insufficient to meet body requirements, hepatic gluconeogenesis plays a key role to maintain normal blood glucose levels. The current studies investigated the role in this process played by vasodilatory-associated phosphoprotein (VASP), a protein that is phosphorylated in hepatocytes by cAMP/protein kinase A (PKA), a key mediator of the action of glucagon. We report that following stimulation of hepatocytes with 8Br-cAMP, phosphorylation of VASP preceded induction of genes encoding key gluconeogenic enzymes, glucose-6-phosphatase (G6p) and phosphoenolpyruvate carboxykinase (Pck1), and that VASP overexpression enhanced this gene induction. Conversely, hepatocytes from mice lacking VASP (Vasp-/-) displayed blunted induction of gluconeogenic enzymes in response to cAMP, and Vasp-/- mice exhibited both greater fasting hypoglycemia and blunted hepatic gluconeogenic enzyme gene expression in response to fasting in vivo. These effects of VASP deficiency were associated with reduced phosphorylation of both CREB (a key transcription factor for gluconeogenesis that lies downstream of PKA) and histone deacetylase 4 (HDAC4), a combination of effects that inhibit transcription of gluconeogenic genes. These data support a model in which VASP functions as a molecular bridge linking the two key signal transduction pathways governing hepatic gluconeogenic gene expression.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Gluconeogénesis/genética , Hígado/metabolismo , Proteínas de Microfilamentos/metabolismo , Fosfoproteínas/metabolismo , Animales , Glucemia/metabolismo , Moléculas de Adhesión Celular/deficiencia , Moléculas de Adhesión Celular/genética , Células Cultivadas , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Ayuno/metabolismo , Regulación de la Expresión Génica , Glucosa-6-Fosfatasa/genética , Hepatocitos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Microfilamentos/deficiencia , Proteínas de Microfilamentos/genética , Modelos Biológicos , Fosfoenolpiruvato Carboxiquinasa (GTP)/genética , Fosfoproteínas/deficiencia , Fosfoproteínas/genética , Fosforilación , Transducción de Señal
6.
Asian J Androl ; 20(3): 276-283, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29205180

RESUMEN

Androgen deprivation in men leads to increased adiposity, but the mechanisms underlying androgen regulation of fat mass have not been fully defined. Androgen receptor (AR) is expressed in monocytes/macrophages, which are resident in key metabolic tissues and influence energy metabolism in surrounding cells. Male mice bearing a cell-specific knockout of the AR in monocytes/macrophages (M-ARKO) were generated to determine whether selective loss of androgen signaling in these cells would lead to altered body composition. Wild-type (WT) and M-ARKO mice (12-22 weeks of age, n = 12 per group) were maintained on a regular chow diet for 8 weeks and then switched to a high-fat diet for 8 additional weeks. At baseline and on both the regular chow and high-fat diets, no differences in lean mass or fat mass were observed between groups. Consistent with the absence of differential body weight or adiposity, no differences in food intake (3.0 ± 0.5 g per day for WT mice vs 2.8 ± 0.4 g per day for M-ARKO mice) or total energy expenditure (0.6 ± 0.1 Kcal h-1 for WT mice vs 0.5 ± 0.1 Kcal h-1 for M-ARKO mice) were evident between groups during high-fat feeding. Liver weight was greater in M-ARKO than that in WT mice (1.5 ± 0.1 g vs 1.3 ± 0.0 g, respectively, P = 0.02). Finally, M-ARKO mice did not exhibit impairments in glucose tolerance or insulin sensitivity relative to WT mice at any study time point. In aggregate, these findings suggest that AR signaling specifically in monocytes/macrophages does not contribute to the regulation of systemic energy balance, adiposity, or insulin sensitivity in male mice.


Asunto(s)
Adiposidad/genética , Hígado/anatomía & histología , Macrófagos/metabolismo , Monocitos/metabolismo , Receptores Androgénicos/genética , Animales , Glucemia/genética , Glucemia/metabolismo , Metabolismo Energético/genética , Prueba de Tolerancia a la Glucosa , Homeostasis/genética , Masculino , Ratones , Ratones Noqueados , Tamaño de los Órganos , Receptores Androgénicos/metabolismo , Transducción de Señal
7.
Nat Commun ; 8: 14556, 2017 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-28223698

RESUMEN

Female mice are less susceptible to the negative metabolic consequences of high-fat diet feeding than male mice, for reasons that are incompletely understood. Here we identify sex-specific differences in hypothalamic microglial activation via the CX3CL1-CX3CR1 pathway that mediate the resistance of female mice to diet-induced obesity. Female mice fed a high-fat diet maintain CX3CL1-CX3CR1 levels while male mice show reductions in both ligand and receptor expression. Female Cx3cr1 knockout mice develop 'male-like' hypothalamic microglial accumulation and activation, accompanied by a marked increase in their susceptibility to diet-induced obesity. Conversely, increasing brain CX3CL1 levels in male mice through central pharmacological administration or virally mediated hypothalamic overexpression converts them to a 'female-like' metabolic phenotype with reduced microglial activation and body-weight gain. These data implicate sex differences in microglial activation in the modulation of energy homeostasis and identify CX3CR1 signalling as a potential therapeutic target for the treatment of obesity.


Asunto(s)
Receptor 1 de Quimiocinas CX3C/metabolismo , Microglía/metabolismo , Microglía/patología , Obesidad/metabolismo , Obesidad/patología , Caracteres Sexuales , Transducción de Señal , Animales , Receptor 1 de Quimiocinas CX3C/deficiencia , Proteínas de Unión al Calcio/metabolismo , Dieta Alta en Grasa , Susceptibilidad a Enfermedades , Estrógenos/farmacología , Conducta Alimentaria/efectos de los fármacos , Femenino , Hipotálamo/patología , Inflamación/patología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Microfilamentos/metabolismo , Microglía/efectos de los fármacos , Fenotipo , Aumento de Peso
8.
Nat Med ; 22(7): 800-6, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27213816

RESUMEN

Type 2 diabetes (T2D) is among the most common and costly disorders worldwide. The goal of current medical management for T2D is to transiently ameliorate hyperglycemia through daily dosing of one or more antidiabetic drugs. Hypoglycemia and weight gain are common side effects of therapy, and sustained disease remission is not obtainable with nonsurgical approaches. On the basis of the potent glucose-lowering response elicited by activation of brain fibroblast growth factor (FGF) receptors, we explored the antidiabetic efficacy of centrally administered FGF1, which, unlike other FGF peptides, activates all FGF receptor subtypes. We report that a single intracerebroventricular injection of FGF1 at a dose one-tenth of that needed for antidiabetic efficacy following peripheral injection induces sustained diabetes remission in both mouse and rat models of T2D. This antidiabetic effect is not secondary to weight loss, does not increase the risk of hypoglycemia, and involves a novel and incompletely understood mechanism for increasing glucose clearance from the bloodstream. We conclude that the brain has an inherent potential to induce diabetes remission and that brain FGF receptors are potential pharmacological targets for achieving this goal.


Asunto(s)
Glucemia/efectos de los fármacos , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Factor 1 de Crecimiento de Fibroblastos/farmacología , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/metabolismo , Animales , Glucemia/metabolismo , Western Blotting , Composición Corporal , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Radioisótopos de Carbono , Desoxiglucosa , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Células Ependimogliales/efectos de los fármacos , Células Ependimogliales/metabolismo , Proteína Forkhead Box O1/genética , Prueba de Tolerancia a la Glucosa , Corazón/efectos de los fármacos , Proteínas de Choque Térmico/efectos de los fármacos , Proteínas de Choque Térmico/metabolismo , Hiperglucemia/metabolismo , Hipotálamo/citología , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Inyecciones Intraventriculares , Hígado/metabolismo , Masculino , Ratones , Ratones Noqueados , Ratones Obesos , Chaperonas Moleculares , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Miocardio/metabolismo , Proteínas de Neoplasias/efectos de los fármacos , Proteínas de Neoplasias/metabolismo , Proteínas Proto-Oncogénicas c-fos/efectos de los fármacos , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas , Ratas Zucker , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptor de Insulina/antagonistas & inhibidores , Receptor de Insulina/genética , Inducción de Remisión
9.
Diabetes ; 64(7): 2376-87, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25633417

RESUMEN

Several lines of evidence implicate excess glucagon secretion in the elevated rates of hepatic glucose production (HGP), hyperglycemia, and ketosis characteristic of uncontrolled insulin-deficient diabetes (uDM), but whether hyperglucagonemia is required for hyperglycemia in this setting is unknown. To address this question, adult male Wistar rats received either streptozotocin (STZ) to induce uDM (STZ-DM) or vehicle and remained nondiabetic. Four days later, animals received daily subcutaneous injections of either the synthetic GLP-1 receptor agonist liraglutide in a dose-escalating regimen to reverse hyperglucagonemia or its vehicle for 10 days. As expected, plasma glucagon levels were elevated in STZ-DM rats, and although liraglutide treatment lowered glucagon levels to those of nondiabetic controls, it failed to attenuate diabetic hyperglycemia, elevated rates of glucose appearance (Ra), or increased hepatic gluconeogenic gene expression. In contrast, it markedly reduced levels of both plasma ketone bodies and hepatic expression of the rate-limiting enzyme involved in ketone body production. To independently confirm this finding, in a separate study, treatment of STZ-DM rats with a glucagon-neutralizing antibody was sufficient to potently lower plasma ketone bodies but failed to normalize elevated levels of either blood glucose or Ra. These data suggest that in rats with uDM, hyperglucagonemia is required for ketosis but not for increased HGP or hyperglycemia.


Asunto(s)
Diabetes Mellitus Experimental/complicaciones , Glucagón/sangre , Glucosa/metabolismo , Hiperglucemia/etiología , Cetosis/etiología , Hígado/metabolismo , Animales , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/fisiología , Glucagón/fisiología , Péptido 1 Similar al Glucagón/análogos & derivados , Péptido 1 Similar al Glucagón/farmacología , Hiperglucemia/sangre , Insulina/farmacología , Cuerpos Cetónicos/sangre , Cetosis/sangre , Liraglutida , Masculino , Ratas Wistar , Receptores de Glucagón/fisiología , Estreptozocina
10.
Menopause ; 21(4): 424-32, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24149922

RESUMEN

OBJECTIVE: Leptin, a hormone secreted by adipocytes, plays a crucial role in regulating energy balance. Estrogen, like leptin, reduces food intake and adiposity while increasing energy expenditure in animals and humans of both sexes through its actions on the central nervous system. We reviewed the literature for studies of the effects of exogenously administered estrogen on serum leptin concentrations and adiposity in women. METHODS: Using PubMed/Medline, we searched for studies of hormone therapy that enrolled healthy postmenopausal women. Studies were further evaluated to determine if leptin and adiposity were monitored both at baseline and throughout a treatment period of at least 2 months. RESULTS: Twenty articles met inclusion criteria. We found no consistent effects of exogenous estrogen on serum leptin concentrations, adiposity, or weight gain. CONCLUSIONS: Despite suggestive data from animal studies, the current literature does not provide compelling evidence that estrogen therapy attenuates weight gain, alters circulating leptin levels, or improves leptin action in postmenopausal women.


Asunto(s)
Adiposidad/efectos de los fármacos , Estrógenos/administración & dosificación , Leptina/sangre , Aumento de Peso/efectos de los fármacos , Animales , Índice de Masa Corporal , Terapia de Reemplazo de Estrógeno , Femenino , Humanos , MEDLINE , Obesidad/prevención & control , Ensayos Clínicos Controlados Aleatorios como Asunto
11.
Am J Physiol Endocrinol Metab ; 302(1): E134-44, 2012 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-22008455

RESUMEN

Growing evidence suggests that oxytocin plays an important role in the regulation of energy balance and that central oxytocin administration induces weight loss in diet-induced obese (DIO) animals. To gain a better understanding of how oxytocin mediates these effects, we examined feeding and neuronal responses to oxytocin in animals rendered obese following exposure to either a high-fat (HFD) or low-fat diet (LFD). Our findings demonstrate that peripheral administration of oxytocin dose-dependently reduces food intake and body weight to a similar extent in rats maintained on either diet. Moreover, the effect of oxytocin to induce weight loss remained intact in leptin receptor-deficient Koletsky (fa(k)/fa(k)) rats relative to their lean littermates. To determine whether systemically administered oxytocin activates hindbrain areas that regulate meal size, we measured neuronal c-Fos induction in the nucleus of the solitary tract (NTS) and area postrema (AP). We observed a robust neuronal response to oxytocin in these hindbrain areas that was unexpectedly increased in rats rendered obese on a HFD relative to lean, LFD-fed controls. Finally, we report that repeated daily peripheral administration of oxytocin in DIO animals elicited a sustained reduction of food intake and body weight while preventing the reduction of energy expenditure characteristic of weight-reduced animals. These findings extend recent evidence suggesting that oxytocin circumvents leptin resistance and induces weight-loss in DIO animals through a mechanism involving activation of neurons in the NTS and AP, key hindbrain areas for processing satiety-related inputs.


Asunto(s)
Depresores del Apetito/uso terapéutico , Grasas de la Dieta/efectos adversos , Obesidad/tratamiento farmacológico , Oxitocina/uso terapéutico , Pérdida de Peso/efectos de los fármacos , Animales , Depresores del Apetito/administración & dosificación , Área Postrema/efectos de los fármacos , Área Postrema/metabolismo , Área Postrema/patología , Terapia Combinada , Cruzamientos Genéticos , Relación Dosis-Respuesta a Droga , Inyecciones Intraperitoneales , Leptina/sangre , Masculino , Proteínas del Tejido Nervioso/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Obesidad/sangre , Obesidad/dietoterapia , Oxitocina/administración & dosificación , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas , Ratas Mutantes , Ratas Sprague-Dawley , Receptores de Leptina/genética , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/uso terapéutico , Núcleo Solitario/efectos de los fármacos , Núcleo Solitario/metabolismo , Núcleo Solitario/patología
12.
Hepatology ; 55(4): 1103-11, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21994008

RESUMEN

UNLABELLED: Childhood obesity is associated with type 2 diabetes mellitus and nonalcoholic fatty liver disease (NAFLD). Recent studies have found associations between vitamin D deficiency (VDD), insulin resistance (IR), and NAFLD among overweight children. To further explore mechanisms mediating these effects, we fed young (age 25 days) Sprague-Dawley rats with a low-fat diet (LFD) alone or with vitamin D depletion (LFD+VDD). A second group of rats was exposed to a Westernized diet (WD: high-fat/high-fructose corn syrup) that is more typically consumed by overweight children, and was either replete (WD) or deficient in vitamin D (WD+VDD). Liver histology was assessed using the nonalcoholic steatohepatitis (NASH) Clinical Research Network (CRN) scoring system and expression of genes involved in inflammatory pathways were measured in liver and visceral adipose tissue after 10 weeks. In VDD groups, 25-OH-vitamin D levels were reduced to 29% (95% confidence interval [CI]: 23%-36%) compared to controls. WD+VDD animals exhibited significantly greater hepatic steatosis compared to LFD groups. Lobular inflammation as well as NAFLD Activity Score (NAS) were higher in WD+VDD versus the WD group (NAS: WD+VDD 3.2 ± 0.47 versus WD 1.50 ± 0.48, P < 0.05). Hepatic messenger RNA (mRNA) levels of Toll-like receptors (TLR)2, TLR4, and TLR9, as well as resistin, interleukins (IL)-1ß, IL-4, and IL-6 and oxidative stress marker heme oxygenase (HO)-1, were higher in WD+VDD versus WD animals (P < 0.05). Logistic regression analyses showed significant associations between NAS score and liver mRNA levels of TLRs 2, 4, and 9, endotoxin receptor CD14, as well as peroxisome proliferator activated receptor (PPAR)γ, and HO-1. CONCLUSION: VDD exacerbates NAFLD through TLR-activation, possibly by way of endotoxin exposure in a WD rat model. In addition it causes IR, higher hepatic resistin gene expression, and up-regulation of hepatic inflammatory and oxidative stress genes.


Asunto(s)
Hígado Graso/epidemiología , Hígado Graso/fisiopatología , Hígado/metabolismo , Obesidad/epidemiología , Resistina/metabolismo , Receptores Toll-Like/metabolismo , Deficiencia de Vitamina D/epidemiología , Animales , Comorbilidad , Carbohidratos de la Dieta/efectos adversos , Grasas de la Dieta/efectos adversos , Modelos Animales de Enfermedad , Hígado Graso/metabolismo , Resistencia a la Insulina/fisiología , Hígado/patología , Masculino , Enfermedad del Hígado Graso no Alcohólico , Obesidad/etiología , Obesidad/fisiopatología , Estrés Oxidativo/fisiología , Ratas , Ratas Sprague-Dawley , Índice de Severidad de la Enfermedad , Deficiencia de Vitamina D/fisiopatología
13.
Pediatr Res ; 69(3): 230-6, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21372758

RESUMEN

Patients with craniopharyngioma (CP), a tumor located in the pituitary and/or hypothalamus, are susceptible to developing obesity and many metabolic complications. The study aim was to create a rodent model that mimics the complex neuroanatomical and metabolic disturbances commonly seen in obese CP patients. We compared the metabolic phenotype of animals with three distinct types of hypothalamic lesions: 1) destruction of the arcuate nucleus (ARC) induced by monosodium glutamate (MSG), 2) electrolytic lesion of the adjacent ventromedial nucleus (VMN) alone, 3) both the VMN and dorsomedial nucleus (DMN), or a 4) combined medial hypothalamic lesion (CMHL) affecting the VMN, DMN, and the ARC. Only the CMHL model exhibited all key features observed in patients with hypothalamic obesity induced by CP. These features included excessive weight gain due to increased adiposity, increased food intake, and pronounced hyperinsulinemia and hyperleptinemia. Similar to characteristics of patients with CP, CMHL animals exhibited reduced plasma levels of alpha-melanocyte stimulating hormone and reduced ambulatory activity compared with weight-matched controls. Therefore, the CMHL model best mimics the complex metabolic abnormalities observed in obese CP patients compared with lesions to other hypothalamic areas and provides a foundation for future pharmacological approaches to treat obesity in children with hypothalamic damage.


Asunto(s)
Craneofaringioma/complicaciones , Modelos Animales de Enfermedad , Neoplasias Hipotalámicas/complicaciones , Obesidad/etiología , Neoplasias Hipofisarias/complicaciones , Animales , Núcleo Arqueado del Hipotálamo/efectos de los fármacos , Núcleo Arqueado del Hipotálamo/patología , Peso Corporal , Niño , Craneofaringioma/patología , Núcleo Hipotalámico Dorsomedial/metabolismo , Núcleo Hipotalámico Dorsomedial/patología , Ingestión de Alimentos , Metabolismo Energético , Femenino , Homeostasis , Humanos , Neoplasias Hipotalámicas/patología , Hipotálamo/anatomía & histología , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Hipotálamo/patología , Masculino , Neoplasias Hipofisarias/patología , Embarazo , Ratas , Ratas Sprague-Dawley , Glutamato de Sodio/efectos adversos , Núcleo Hipotalámico Ventromedial/metabolismo , Núcleo Hipotalámico Ventromedial/patología
14.
Am J Physiol Endocrinol Metab ; 300(2): E392-401, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21062956

RESUMEN

Mechanisms regulating spontaneous physical activity remain poorly characterized despite evidence of influential genetic and acquired factors. We evaluated ambulatory activity and wheel running in leptin-deficient ob/ob mice and in wild-type mice rendered hypoleptinemic by fasting in both the presence and absence of subcutaneous leptin administration. In ob/ob mice, leptin treatment to plasma levels characteristic of wild-type mice acutely increased both ambulatory activity (by 4,000 ± 200 beam breaks/dark cycle, P < 0.05) and total energy expenditure (TEE; by 0.11 ± 0.01 kcal/h during the dark cycle, P < 0.05) in a dose-dependent manner and acutely increased wheel running (+350%, P < 0.05). Fasting potently increased ambulatory activity and wheel running in wild-type mice (AA: +25%, P < 0.05; wheel running: +80%, P < 0.05), and the effect of fasting was more pronounced in ob/ob mice (AA: +400%, P < 0.05; wheel running: +1,600%, P < 0.05). However, unlike what occurred in ad libitum-fed ob/ob mice, physiological leptin replacement attenuated or prevented fasting-induced increases of ambulatory activity and wheel running in both wild-type and ob/ob mice. Thus, plasma leptin is a physiological regulator of spontaneous physical activity, but the nature of leptin's effect on activity is dependent on food availability.


Asunto(s)
Leptina/fisiología , Actividad Motora/fisiología , Carrera/fisiología , Animales , Composición Corporal , Calorimetría Indirecta , Oscuridad , Relación Dosis-Respuesta a Droga , Metabolismo Energético , Ensayo de Inmunoadsorción Enzimática , Ayuno/fisiología , Hipotálamo/metabolismo , Leptina/metabolismo , Leptina/farmacología , Luz , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Obesos , Actividad Motora/efectos de los fármacos , Neuropéptidos/biosíntesis , Neuropéptidos/genética , Consumo de Oxígeno/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
15.
Endocrinology ; 151(9): 4207-13, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20610562

RESUMEN

Evidence suggests that release of oxytocin in the nucleus tractus solitarius (NTS) of the hindbrain from descending projections that originate in the paraventricular nucleus can inhibit food intake by amplifying the satiety response to cholecystokinin (CCK). To further evaluate this mechanism in rats, we used a novel cytotoxin, saporin conjugated to oxytocin (OXY-SAP), a compound designed to destroy cells that express oxytocin receptors (OXYr). OXY-SAP was injected directly into the NTS to lesion neurons that express OXYr and that are implicated in potentiating CCK's satiety effects. The control consisted of injection of saporin conjugated to a nonsense peptide. We found that OXY-SAP was cytotoxic to human uterine smooth muscle cells in vitro, demonstrating that OXY-SAP can lesion cells that express OXYr. Using laser capture microdissection and real-time quantitative PCR, we demonstrated that OXYr mRNA levels were reduced in the NTS after OXY-SAP administration. Moreover, we found that OXY-SAP attenuated the efficacy of CCK-8 to reduce food intake and blocked the actions of an OXYr antagonist to stimulate food intake. The findings suggest that OXY-SAP is an effective neurotoxin for in vivo elimination of cells that express OXYr and is potentially useful for studies to analyze central nervous system mechanisms that involve the action of oxytocin on food intake and other physiological processes.


Asunto(s)
Neuronas/efectos de los fármacos , Oxitocina/farmacología , Receptores de Oxitocina/genética , Proteínas Inactivadoras de Ribosomas Tipo 1/farmacología , Animales , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Citotoxinas/química , Citotoxinas/farmacología , Relación Dosis-Respuesta a Droga , Ingestión de Alimentos/efectos de los fármacos , Femenino , Expresión Génica/efectos de los fármacos , Humanos , Interleucina-1beta/genética , Masculino , Miometrio/citología , Neuronas/metabolismo , Neuronas/patología , Oxitocina/química , Ratas , Ratas Wistar , Receptores de Oxitocina/antagonistas & inhibidores , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Rombencéfalo/efectos de los fármacos , Rombencéfalo/metabolismo , Rombencéfalo/patología , Proteínas Inactivadoras de Ribosomas Tipo 1/química , Saporinas , Sincalida/farmacología , Factor de Necrosis Tumoral alfa/genética
16.
Endocrinology ; 151(3): 993-1001, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20056822

RESUMEN

Hepatic lipase (HL)-mediated lipoprotein hydrolysis provides free fatty acids for energy, storage, and nutrient signaling and may play a role in energy homeostasis. Because HL-activity increases with increased visceral fat, we hypothesized that increased HL-activity favors weight gain and obesity and consequently, that HL deficiency would reduce body fat stores and protect against diet-induced obesity. To test this hypothesis, we compared wild-type mice (with endogenous HL) and mice genetically deficient in HL with respect to daily body weight and food intake, body composition, and adipocyte size on both chow and high-fat (HF) diets. Key determinants of energy expenditure, including rate of oxygen consumption, heat production, and locomotor activity, were measured by indirect calorimetry. HL-deficient mice exhibited reduced weight gain on both diets (by 32%, chow; by 50%, HF; both P < 0.0001, n = 6-7 per genotype), effects that were associated with reduced average daily food intake (by 22-30% on both diets, P < 0.0001) and a modest increase in the rate of oxygen consumption (by 25%, P < 0.003) during the light cycle. Moreover, in mice fed the HF diet, HL deficiency reduced both body fat (by 30%, P < 0.0001) and adipocyte size (by 53%, P < 0.01) and fully prevented the development of hepatic steatosis. Also, HL deficiency reduced adipose tissue macrophage content, consistent with reduced inflammation and a lean phenotype. Our results demonstrate that in mice, HL deficiency protects against diet-induced obesity and its hepatic sequelae. Inhibition of HL-activity may therefore have value in the prevention and/or treatment of obesity.


Asunto(s)
Ingestión de Alimentos , Metabolismo Energético , Hígado Graso/enzimología , Lipasa/metabolismo , Obesidad/enzimología , Tejido Adiposo/citología , Tejido Adiposo/inmunología , Adiposidad , Animales , Glucemia/metabolismo , Peso Corporal , Ritmo Circadiano , Grasas de la Dieta/efectos adversos , Hígado Graso/etiología , Femenino , Prueba de Tolerancia a la Glucosa , Metabolismo de los Lípidos , Hígado/metabolismo , Locomoción , Macrófagos/citología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/etiología , Consumo de Oxígeno , Termogénesis , Triglicéridos/metabolismo
17.
Endocrinology ; 150(10): 4502-11, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19574396

RESUMEN

Recent evidence suggests that hormones such as insulin and leptin act in the hypothalamus to regulate energy balance and glucose metabolism. Here we show that in leptin receptor-deficient Koletsky (fa(k)/fa(k)) rats, adenovirally induced expression of leptin receptors in the area of the hypothalamic arcuate nucleus improved peripheral insulin sensitivity via enhanced suppression of hepatic glucose production, with no change of insulin-stimulated glucose uptake or disposal. This effect was associated with increased insulin signal transduction via phosphatidylinositol-3-OH kinase (as measured by pY-insulin receptor substrate-1 and pS-PKB/Akt) in liver, but not skeletal muscle, and with reduced hepatic expression of the gluconeogenic genes, glucose-6-phosphatase and phosphoenolpyruvate kinase. Moreover, the beneficial effects of hypothalamic leptin signaling on hepatic insulin sensitivity were blocked by selective hepatic vagotomy. We conclude that hypothalamic leptin action increases peripheral insulin sensitivity primarily via effects on the liver and that the mechanism underlying this effect is dependent on the hepatic branch of the vagus nerve.


Asunto(s)
Núcleo Arqueado del Hipotálamo/metabolismo , Insulina/metabolismo , Hígado/metabolismo , Receptores de Leptina/metabolismo , Nervio Vago/fisiología , Animales , Expresión Génica , Genes Reporteros , Terapia Genética , Gluconeogénesis/genética , Glucosa/metabolismo , Técnica de Clampeo de la Glucosa , Resistencia a la Insulina , Masculino , Obesidad/terapia , Fosfatidilinositol 3-Quinasas/metabolismo , Ratas , Transducción de Señal , Vagotomía
18.
Endocrinology ; 149(12): 6053-64, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18687778

RESUMEN

Energy homeostasis involves central nervous system integration of afferent inputs that coordinately regulate food intake and energy expenditure. Here, we report that adult homozygous TNFalpha converting enzyme (TACE)-deficient mice exhibit one of the most dramatic examples of hypermetabolism yet reported in a rodent system. Because this effect is not matched by increased food intake, mice lacking TACE exhibit a lean phenotype. In the hypothalamus of these mice, neurons in the arcuate nucleus exhibit intact responses to reduced fat mass and low circulating leptin levels, suggesting that defects in other components of the energy homeostasis system explain the phenotype of Tace(DeltaZn/DeltaZn) mice. Elevated levels of uncoupling protein-1 in brown adipose tissue from Tace(DeltaZn/DeltaZn) mice when compared with weight-matched controls suggest that deficient TACE activity is linked to increased sympathetic outflow. These findings collectively identify a novel and potentially important role for TACE in energy homeostasis.


Asunto(s)
Proteínas ADAM/deficiencia , Metabolismo Energético/fisiología , Delgadez/fisiopatología , Proteínas ADAM/genética , Proteína ADAM17 , Adipocitos/citología , Adipocitos/metabolismo , Animales , Núcleo Arqueado del Hipotálamo/metabolismo , Western Blotting , Diferenciación Celular/fisiología , Ingestión de Alimentos/fisiología , Grasas/metabolismo , Hipotálamo/metabolismo , Inmunohistoquímica , Canales Iónicos/metabolismo , Leptina/sangre , Masculino , Ratones , Ratones Mutantes , Proteínas Mitocondriales/metabolismo , Actividad Motora/fisiología , Fenotipo , Delgadez/sangre , Delgadez/genética , Proteína Desacopladora 1
19.
Brain Res ; 1144: 101-6, 2007 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-17320056

RESUMEN

Hypothalamic IL1 is suggested to be a critical mediator of the central effects of the adipocyte hormone leptin on energy balance. We hypothesized that IL1 receptor signaling is required for exogenously administered leptin to cause anorexia and weight loss, but not for physiological effects of endogenous leptin signaling on energy balance. To test this hypothesis, we investigated whether chronic hypothalamic over-expression of an IL1 receptor antagonist (AdV-IL1ra) alters food intake and weight gain in normal rats. Our findings demonstrate that impaired IL1 signaling in the CNS did not cause excess weight gain over a period of 11 days (AdV-IL1ra +38.1+/-4.1 g vs. VEH +42.2+/-5.6g; p=0.6) and caused a slightly reduced daily food intake (AdV-IL1ra 29.0+/-1.1 g/day vs. VEH 33.0+/-1.6 g/day; p<0.05). Blocking central IL1 signaling also did not alter the re-feeding response to a prolonged fast, yet was entirely effective in preventing the anorexic effect of exogenously administered leptin (2 mg/kg ip, cumulative food intake at 18 h AdV-IL1ra 30.5+/-1.1 g vs. VEH 26.4+/-1.7 g, p<0.05) and prevented leptin-induced weight loss (AdV-IL1ra -0.1+/-1.3 g vs. VEH -2.7+/-1.9 g, p<0.05). Together these findings suggest that hypothalamic IL1 signaling is required for the pharmacological effects of leptin administration, but that impaired hypothalamic IL1 signaling does not alter the physiological regulation of energy balance.


Asunto(s)
Sistema Nervioso Central/metabolismo , Metabolismo Energético/efectos de los fármacos , Interleucina-1/metabolismo , Leptina/farmacología , Transducción de Señal/fisiología , Adenoviridae/fisiología , Análisis de Varianza , Animales , Conducta Animal , Peso Corporal/efectos de los fármacos , Sistema Nervioso Central/anatomía & histología , Sistema Nervioso Central/efectos de los fármacos , Ingestión de Alimentos/efectos de los fármacos , Ayuno/fisiología , Masculino , Ratas , Ratas Wistar , Transducción de Señal/efectos de los fármacos , Factores de Tiempo
20.
Cell Metab ; 2(6): 411-20, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16330326

RESUMEN

To investigate whether phosphatidylinositol-3 kinase (PI3K) signaling mediates the metabolic effects of hypothalamic leptin action, adenoviral gene therapy was used to direct expression of leptin receptors to the area of the hypothalamic arcuate nucleus (ARC). This intervention markedly improved insulin sensitivity in genetically obese, leptin-receptor-deficient Koletsky (fa(k)/fa(k)) rats via a mechanism that was not dependent on reduced food intake but was attenuated by approximately 44% by third-ventricular infusion of the PI3K inhibitor LY294002. Conversely, ARC-directed expression of a constitutively active mutant of protein kinase B (PKB/Akt, an enzyme activated by PI3K) mimicked the insulin-sensitizing effect of restored hypothalamic leptin signaling in these animals, despite having no effect on food intake or body weight. These findings suggest that hypothalamic leptin signaling is an important determinant of glucose metabolism and that the underlying neuronal mechanism involves PI3K.


Asunto(s)
Hipotálamo/patología , Insulina/metabolismo , Leptina/biosíntesis , Neuronas/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Adenoviridae/genética , Animales , Animales Modificados Genéticamente , Glucemia/metabolismo , Peso Corporal , Cromonas/farmacología , Inhibidores Enzimáticos/farmacología , Terapia Genética , Glucosa/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Leptina/fisiología , Masculino , Morfolinas/farmacología , Ratas , Receptores de Superficie Celular/genética , Receptores de Leptina , Transducción de Señal , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA