Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
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.
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
5.
Nat Neurosci ; 20(7): 934-942, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28581479

RESUMEN

Anorexia is a common manifestation of chronic diseases, including cancer. Here we investigate the contribution to cancer anorexia made by calcitonin gene-related peptide (CGRP) neurons in the parabrachial nucleus (PBN) that transmit anorexic signals. We show that CGRPPBN neurons are activated in mice implanted with Lewis lung carcinoma cells. Inactivation of CGRPPBN neurons before tumor implantation prevents anorexia and loss of lean mass, and their inhibition after symptom onset reverses anorexia. CGRPPBN neurons are also activated in Apcmin/+ mice, which develop intestinal cancer and lose weight despite the absence of reduced food intake. Inactivation of CGRPPBN neurons in Apcmin/+ mice permits hyperphagia that counteracts weight loss, revealing a role for these neurons in a 'nonanorexic' cancer model. We also demonstrate that inactivation of CGRPPBN neurons prevents lethargy, anxiety and malaise associated with cancer. These findings establish CGRPPBN neurons as key mediators of cancer-induced appetite suppression and associated behavioral changes.


Asunto(s)
Anorexia/fisiopatología , Péptido Relacionado con Gen de Calcitonina/fisiología , Carcinoma Pulmonar de Lewis/fisiopatología , Conducta de Enfermedad/fisiología , Neoplasias/fisiopatología , Núcleos Parabraquiales/fisiología , Proteína de la Poliposis Adenomatosa del Colon/genética , Animales , Conducta Animal/fisiología , Peso Corporal , Caquexia/fisiopatología , Péptido Relacionado con Gen de Calcitonina/antagonistas & inhibidores , Péptido Relacionado con Gen de Calcitonina/genética , Clozapina/análogos & derivados , Clozapina/farmacología , Metabolismo Energético/fisiología , Femenino , Masculino , Metaloendopeptidasas/farmacología , Ratones , Ratones Transgénicos , Núcleos Parabraquiales/efectos de los fármacos , Toxina Tetánica/farmacología , Células Tumorales Cultivadas/trasplante
6.
Cell Metab ; 23(5): 811-20, 2016 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-27166945

RESUMEN

The lateral parabrachial nucleus is a conduit for visceral signals that cause anorexia. We previously identified a subset of neurons located in the external lateral parabrachial nucleus (PBel) that express calcitonin gene-related peptide (CGRP) and inhibit feeding when activated by illness mimetics. We report here that in otherwise normal mice, functional inactivation of CGRP neurons markedly increases meal size, with meal frequency being reduced in a compensatory manner, and renders mice insensitive to the anorexic effects of meal-related satiety peptides. Furthermore, CGRP neurons are directly innervated by orexigenic hypothalamic AgRP neurons, and photostimulation of AgRP fibers supplying the PBel delays satiation by inhibiting CGRP neurons, thereby contributing to AgRP-driven hyperphagia. By establishing a role for CGRP neurons in the control of meal termination and as a downstream mediator of feeding elicited by AgRP neurons, these findings identify a node in which hunger and satiety circuits interact to control feeding behavior.


Asunto(s)
Péptido Relacionado con Gen de Calcitonina/metabolismo , Conducta Alimentaria , Núcleos Parabraquiales/metabolismo , Proteína Relacionada con Agouti/metabolismo , Animales , Anorexia/metabolismo , Anorexia/patología , Núcleo Amigdalino Central/metabolismo , Colecistoquinina , Receptor del Péptido 1 Similar al Glucagón/agonistas , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Hiperfagia/metabolismo , Hiperfagia/patología , Leptina , Ratones Endogámicos C57BL , Neuronas/metabolismo , Respuesta de Saciedad
7.
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
8.
Nat Commun ; 6: 7079, 2015 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-25963540

RESUMEN

FoxO proteins are major targets of insulin action. To better define the role of FoxO1 in mediating insulin effects in the liver, we generated liver-specific insulin receptor knockout (LIRKO) and IR/FoxO1 double knockout (LIRFKO) mice. Here we show that LIRKO mice are severely insulin resistant based on glucose, insulin and C-peptide levels, and glucose and insulin tolerance tests, and genetic deletion of hepatic FoxO1 reverses these effects. (13)C-glucose and insulin clamp studies indicate that regulation of both hepatic glucose production (HGP) and glucose utilization is impaired in LIRKO mice, and these defects are also restored in LIRFKO mice corresponding to changes in gene expression. We conclude that (1) inhibition of FoxO1 is critical for both direct (hepatic) and indirect effects of insulin on HGP and utilization, and (2) extrahepatic effects of insulin are sufficient to maintain normal whole-body and hepatic glucose metabolism when liver FoxO1 activity is disrupted.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Gluconeogénesis/fisiología , Glucosa/metabolismo , Insulina/metabolismo , Hígado/metabolismo , Animales , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/genética , Regulación de la Expresión Génica/fisiología , Técnica de Clampeo de la Glucosa , Masculino , Ratones
9.
Diabetes ; 64(8): 2836-46, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25845662

RESUMEN

Endothelial nitric oxide (NO) signaling plays a physiological role in limiting obesity-associated insulin resistance and inflammation. This study was undertaken to investigate whether this NO effect involves polarization of macrophages toward an anti-inflammatory M2 phenotype. Mice with transgenic endothelial NO synthase overexpression were protected against high-fat diet (HFD)-induced hepatic inflammation and insulin resistance, and this effect was associated with reduced proinflammatory M1 and increased anti-inflammatory M2 activation of Kupffer cells. In cell culture studies, exposure of macrophages to endothelial NO similarly reduced inflammatory (M1) and increased anti-inflammatory (M2) gene expression. Similar effects were induced by macrophage overexpression of vasodilator-stimulated phosphoprotein (VASP), a key downstream mediator of intracellular NO signaling. Conversely, VASP deficiency induced proinflammatory M1 macrophage activation, and the transplantation of bone marrow from VASP-deficient donor mice into normal recipients caused hepatic inflammation and insulin resistance resembling that induced in normal mice by consumption of an HFD. These data suggest that proinflammatory macrophage M1 activation and macrophage-mediated inflammation are tonically inhibited by NO → VASP signal transduction, and that reduced NO → VASP signaling is involved in the effect of HFD feeding to induce M1 activation of Kupffer cells and associated hepatic inflammation. Our data implicate endothelial NO → VASP signaling as a physiological determinant of macrophage polarization and show that signaling via this pathway is required to prevent hepatic inflammation and insulin resistance.


Asunto(s)
Polaridad Celular/fisiología , Endotelio Vascular/metabolismo , Inflamación/metabolismo , Macrófagos/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Óxido Nítrico/metabolismo , Animales , Inflamación/genética , Mediadores de Inflamación/metabolismo , Resistencia a la Insulina/fisiología , Macrófagos del Hígado/metabolismo , Hígado/metabolismo , Activación de Macrófagos/fisiología , Ratones , Ratones Transgénicos , Óxido Nítrico Sintasa de Tipo III/genética , Transducción de Señal/fisiología , Triglicéridos/metabolismo
10.
Am J Physiol Endocrinol Metab ; 307(7): E571-9, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25117404

RESUMEN

Among the pleotropic effects of endothelial nitric oxide (NO) is protection against vascular inflammation during high-fat diet (HFD) feeding. The current work investigated the role of the enzyme vasodilatory-stimulated phosphoprotein (VASP) as a downstream mediator of the anti-inflammatory effect of NO signaling in vascular tissue. Relative to mice fed a low-fat diet (LFD), levels of VASP Ser(239) phosphorylation, a marker of VASP activation, were dramatically reduced in aortic tissue of mice with obesity induced by consuming a HFD. As reported previously, the effect of the HFD was associated with increased aortic inflammation, as measured by increased NF-κB-dependent gene expression, and reduced vascular insulin sensitivity (including insulin-stimulated phosphorylation of eNOS and Akt). These effects of the HFD were recapitulated by VASP knockout, implying a physiological role for VASP to constrain inflammatory signaling and thereby maintain vascular insulin sensitivity. Conversely, overexpression of VASP in endothelial cells blocked inflammation and insulin resistance induced by palmitate. The finding that transplantation of bone marrow from VASP-deficient donors into normal recipients does not recapitulate the vascular effects of whole body VASP deficiency suggests that the protective effects of this enzyme are not mediated in immune or other bone marrow-derived cells. These studies implicate VASP as a downstream mediator of the NO/cGMP pathway that is both necessary and sufficient to protect against vascular inflammation and insulin resistance. As such, this work identifies VASP as a potential therapeutic target in the treatment of obesity-related vascular dysfunction.


Asunto(s)
Aorta/metabolismo , Moléculas de Adhesión Celular/fisiología , Células Endoteliales/metabolismo , Resistencia a la Insulina , Proteínas de Microfilamentos/fisiología , Óxido Nítrico/metabolismo , Obesidad/metabolismo , Fosfoproteínas/fisiología , Vasculitis/metabolismo , Animales , Aorta/citología , Aorta/inmunología , Trasplante de Médula Ósea , Bovinos , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Células Cultivadas , Dieta Alta en Grasa , Células Endoteliales/inmunología , Perfilación de la Expresión Génica , Humanos , Inflamación/metabolismo , Resistencia a la Insulina/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Microvasos/citología , Óxido Nítrico/inmunología , Óxido Nítrico Sintasa de Tipo III/genética , Obesidad/inmunología , Palmitatos/farmacología , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Fosforilación , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Vasculitis/inmunología
11.
Endocrinology ; 155(8): 2858-67, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24914942

RESUMEN

Gliosis, the activation of astrocyte and microglial cell populations, is a hallmark of central nervous system injury and is detectable using either immunohistochemistry or in vivo magnetic resonance imaging (MRI). Obesity in rodents and humans is associated with gliosis of the arcuate nucleus, a key hypothalamic region for the regulation of energy homeostasis and adiposity, but whether this response is permanent or reversible is unknown. Here we combine terminal immunohistochemistry analysis with serial, noninvasive MRI to characterize the progression and reversibility of hypothalamic gliosis in high-fat diet (HFD)-fed mice. The effects of HFD feeding for 16 weeks to increase body weight and adiposity relative to chow were nearly normalized after the return to chow feeding for an additional 4 weeks in the diet-reversal group. Mice maintained on the HFD for the full 20-week study period experienced continued weight gain associated with the expected increases of astrocyte and microglial activation in the arcuate nucleus, but these changes were not observed in the diet-reversal group. The proopiomelanocortin neuron number did not differ between groups. Although MRI demonstrated a positive correlation between body weight, adiposity, and the gliosis-associated T2 signal in the mediobasal hypothalamus, it did not detect the reversal of gliosis among the HFD-fed mice after the return to chow diet. We conclude that hypothalamic gliosis associated with 16-week HFD feeding is largely reversible in rodents, consistent with the reversal of the HFD-induced obesity phenotype, and extend published evidence regarding the utility of MRI as a tool for studying obesity-associated hypothalamic gliosis in vivo.


Asunto(s)
Núcleo Arqueado del Hipotálamo , Dieta Alta en Grasa/efectos adversos , Gliosis/etiología , Enfermedades Hipotalámicas/etiología , Enfermedades Hipotalámicas/metabolismo , Obesidad/complicaciones , Obesidad/metabolismo , Adiposidad , Animales , Ingestión de Alimentos , Enfermedades Hipotalámicas/prevención & control , Inmunohistoquímica , Imagen por Resonancia Magnética , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/dietoterapia , Pérdida de Peso
12.
Glia ; 62(1): 17-25, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24166765

RESUMEN

The arcuate nucleus (ARC) of the hypothalamus plays a key role in sensing metabolic feedback and regulating energy homeostasis. Recent studies revealed activation of microglia in mice with high-fat diet (HFD)-induced obesity (DIO), suggesting a potential pathophysiological role for inflammatory processes within the hypothalamus. To further investigate the metabolic causes and molecular underpinnings of such glial activation, we analyzed the microglial activity in wild-type (WT), monogenic obese ob/ob (leptin deficient), db/db (leptin-receptor mutation), and Type-4 melanocortin receptor knockout (MC4R KO) mice on either a HFD or on standardized chow (SC) diet. Following HFD exposure, we observed a significant increase in the total number of ARC microglia, immunoreactivity of ionized calcium binding adaptor molecule 1 (iba1-ir), cluster of differentiation 68 (CD68-ir), and ramification of microglial processes. The ob/ob mice had significantly less iba1-ir and ramifications. Leptin replacement rescued these phenomena. The db/db mice had similar iba1-ir comparable with WT mice but had significantly lower CD68-ir and more ramifications than WT mice. After 2 weeks of HFD, ob/ob mice showed an increase of iba1-ir, and db/db mice showed increase of CD68-ir. Obese MC4R KO mice fed a SC diet had comparable iba1-ir and CD68-ir with WT mice but had significantly more ramifications than WT mice. Intriguingly, treatment of DIO mice with glucagon-like peptide-1 receptor agonists reduced microglial activation independent of body weight. Our results show that diet type, adipokines, and gut signals, but not body weight, affect the presence and activity levels of hypothalamic microglia in obesity.


Asunto(s)
Hormonas/farmacología , Microglía/metabolismo , Núcleo Supraóptico/citología , Animales , Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/metabolismo , Peso Corporal/efectos de los fármacos , Peso Corporal/fisiología , Citocininas/metabolismo , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Exenatida , Leptina/deficiencia , Leptina/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía/efectos de los fármacos , Obesidad/inducido químicamente , Obesidad/fisiopatología , Péptidos/farmacología , Receptor de Melanocortina Tipo 4/deficiencia , Receptores de Interleucina-8A/genética , Receptores de Interleucina-8A/metabolismo , Receptores de Leptina/deficiencia , Receptores de Leptina/genética , Transducción de Señal/efectos de los fármacos , Ponzoñas/farmacología
13.
J Biol Chem ; 288(15): 10722-35, 2013 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-23457303

RESUMEN

Sirt1 is a NAD(+)-dependent class III deacetylase that functions as a cellular energy sensor. In addition to its well-characterized effects in peripheral tissues, emerging evidence suggests that neuronal Sirt1 activity plays a role in the central regulation of energy balance and glucose metabolism. To assess this idea, we generated Sirt1 neuron-specific knockout (SINKO) mice. On both standard chow and HFD, SINKO mice were more insulin sensitive than Sirt1(f/f) mice. Thus, SINKO mice had lower fasting insulin levels, improved glucose tolerance and insulin tolerance, and enhanced systemic insulin sensitivity during hyperinsulinemic euglycemic clamp studies. Hypothalamic insulin sensitivity of SINKO mice was also increased over controls, as assessed by hypothalamic activation of PI3K, phosphorylation of Akt and FoxO1 following systemic insulin injection. Intracerebroventricular injection of insulin led to a greater systemic effect to improve glucose tolerance and insulin sensitivity in SINKO mice compared with controls. In line with the in vivo results, insulin-induced AKT and FoxO1 phosphorylation were potentiated by inhibition of Sirt1 in a cultured hypothalamic cell line. Mechanistically, this effect was traced to a reduced effect of Sirt1 to directly deacetylate and repress IRS-1 function. The enhanced central insulin signaling in SINKO mice was accompanied by increased insulin receptor signal transduction in liver, muscle, and adipose tissue. In summary, we conclude that neuronal Sirt1 negatively regulates hypothalamic insulin signaling, leading to systemic insulin resistance. Interventions that reduce neuronal Sirt1 activity have the potential to improve systemic insulin action and limit weight gain on an obesigenic diet.


Asunto(s)
Metabolismo Energético/fisiología , Hipotálamo/metabolismo , Resistencia a la Insulina/fisiología , Insulina/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Sirtuina 1/metabolismo , Animales , Células Cultivadas , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Glucosa/genética , Glucosa/metabolismo , Hipoglucemiantes/metabolismo , Hipoglucemiantes/farmacología , Insulina/genética , Insulina/farmacología , Proteínas Sustrato del Receptor de Insulina/genética , Proteínas Sustrato del Receptor de Insulina/metabolismo , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Especificidad de Órganos , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación/efectos de los fármacos , Fosforilación/fisiología , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Sirtuina 1/genética
14.
Biochem J ; 447(1): 175-84, 2012 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-22849606

RESUMEN

GSK3ß (glycogen synthase kinase 3ß) is a ubiquitous kinase that plays a key role in multiple intracellular signalling pathways, and increased GSK3ß activity is implicated in disorders ranging from cancer to Alzheimer's disease. In the present study, we provide the first evidence of increased hypothalamic signalling via GSK3ß in leptin-deficient Lep(ob/ob) mice and show that intracerebroventricular injection of a GSK3ß inhibitor acutely improves glucose tolerance in these mice. The beneficial effect of the GSK3ß inhibitor was dependent on hypothalamic signalling via PI3K (phosphoinositide 3-kinase), a key intracellular mediator of both leptin and insulin action. Conversely, neuron-specific overexpression of GSK3ß in the mediobasal hypothalamus exacerbated the hyperphagia, obesity and impairment of glucose tolerance induced by a high-fat diet, while having little effect in controls fed standard chow. These results demonstrate that increased hypothalamic GSK3ß signalling contributes to deleterious effects of leptin deficiency and exacerbates high-fat diet-induced weight gain and glucose intolerance.


Asunto(s)
Ingestión de Alimentos/fisiología , Glucosa/metabolismo , Glucógeno Sintasa Quinasa 3/metabolismo , Hipotálamo/enzimología , Animales , Núcleo Arqueado del Hipotálamo/enzimología , Núcleo Arqueado del Hipotálamo/fisiología , Secuencia de Bases , Cartilla de ADN/genética , Dieta Alta en Grasa/efectos adversos , Intolerancia a la Glucosa/enzimología , Intolerancia a la Glucosa/etiología , Glucógeno Sintasa Quinasa 3/deficiencia , Glucógeno Sintasa Quinasa 3/genética , Glucógeno Sintasa Quinasa 3 beta , Humanos , Hipotálamo/fisiología , Leptina/deficiencia , Leptina/genética , Masculino , Ratones , Ratones Noqueados , Obesidad/enzimología , Obesidad/etiología , Transducción de Señal , Aumento de Peso/fisiología
16.
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
17.
J Clin Invest ; 121(6): 2152-5, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21606602

RESUMEN

Rather than arising from the passive accumulation of excess calories, obesity is a state in which the biologically defended level of body fat stores increases due to defects in the homeostatic process that matches food intake and energy expenditure over time. By deleting leptin receptors from distinct brain regions and neuronal subsets, researchers are beginning to identify the neuroanatomical substrates responsible for this regulation. In this issue of the JCI, Scott et al. demonstrate that loss of leptin receptors in a subset of hindbrain neurons increases food intake in mice, but, unlike what is observed when leptin receptors are deleted from hypothalamic neurons, these mice compensate by increasing energy expenditure and hence do not become obese. Although many brain areas can regulate energy intake and/or energy expenditure, it is likely that only a small subset of neurons actively matches the two over time. It is vital to clarify how this works if we are to improve our understanding of obesity pathogenesis and options available for its treatment.


Asunto(s)
Peso Corporal/fisiología , Encéfalo/fisiología , Homeostasis/fisiología , Receptores de Leptina/fisiología , Animales , Estado de Descerebración/fisiopatología , Ingestión de Energía , Metabolismo Energético/genética , Metabolismo Energético/fisiología , Conducta Alimentaria/fisiología , Homeostasis/genética , Humanos , Hiperfagia/fisiopatología , Leptina/fisiología , Ratones , Ratones Noqueados , Neuronas/fisiología , Obesidad/etiología , Obesidad/fisiopatología , Obesidad/terapia , Especificidad de Órganos , Hormonas Peptídicas/fisiología , Ratas , Receptores de Leptina/deficiencia , Receptores de Leptina/genética
18.
Diabetes Care ; 34(6): 1424-30, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21602431

RESUMEN

OBJECTIVE: This report examines what is known about the relationship between obesity and type 2 diabetes and how future research in these areas might be directed to benefit prevention, interventions, and overall patient care. RESEARCH DESIGN AND METHODS: An international working group of 32 experts in the pathophysiology, genetics, clinical trials, and clinical care of obesity and/or type 2 diabetes participated in a conference held on 6-7 January 2011 and cosponsored by The Endocrine Society, the American Diabetes Association, and the European Association for the Study of Diabetes. A writing group comprising eight participants subsequently prepared this summary and recommendations. Participants reviewed and discussed published literature and their own unpublished data. RESULTS: The writing group unanimously supported the summary and recommendations as representing the working group's majority or unanimous opinions. CONCLUSIONS: The major questions linking obesity to type 2 diabetes that need to be addressed by combined basic, clinical, and population-based scientific approaches include the following: 1) Why do not all patients with obesity develop type 2 diabetes? 2) Through what mechanisms do obesity and insulin resistance contribute to ß-cell decompensation, and if/when obesity prevention ensues, how much reduction in type 2 diabetes incidence will follow? 3) How does the duration of type 2 diabetes relate to the benefits of weight reduction by lifestyle, weight-loss drugs, and/or bariatric surgery on ß-cell function and glycemia? 4) What is necessary for regulatory approval of medications and possibly surgical approaches for preventing type 2 diabetes in patients with obesity? Improved understanding of how obesity relates to type 2 diabetes may help advance effective and cost-effective interventions for both conditions, including more tailored therapy. To expedite this process, we recommend further investigation into the pathogenesis of these coexistent conditions and innovative approaches to their pharmacological and surgical management.


Asunto(s)
Diabetes Mellitus Tipo 2/etiología , Resistencia a la Insulina/fisiología , Obesidad/complicaciones , Animales , Fármacos Antiobesidad/uso terapéutico , Cirugía Bariátrica , Terapia Conductista , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/genética , Humanos , Hipoglucemiantes/uso terapéutico , Células Secretoras de Insulina/fisiología , Obesidad/tratamiento farmacológico , Obesidad/genética , Obesidad/fisiopatología , Factores de Riesgo , Pérdida de Peso
19.
J Clin Endocrinol Metab ; 96(6): 1654-63, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21602457

RESUMEN

OBJECTIVE: This report examines what is known about the relationship between obesity and type 2 diabetes and how future research in these areas might be directed to benefit prevention, interventions, and overall patient care. RESEARCH DESIGN AND METHODS: An international working group of 32 experts in the pathophysiology, genetics, clinical trials, and clinical care of obesity and/or type 2 diabetes participated in a conference held on 6-7 January 2011 and cosponsored by The Endocrine Society, the American Diabetes Association, and the European Association for the Study of Diabetes. A writing group comprising eight participants subsequently prepared this summary and recommendations. Participants reviewed and discussed published literature and their own unpublished data. RESULTS: The writing group unanimously supported the summary and recommendations as representing the working group's majority or unanimous opinions. CONCLUSIONS: The major questions linking obesity to type 2 diabetes that need to be addressed by combined basic, clinical, and population-based scientific approaches include the following: 1) Why do not all patients with obesity develop type 2 diabetes? 2) Through what mechanisms do obesity and insulin resistance contribute to ß-cell decompensation, and if/when obesity prevention ensues, how much reduction in type 2 diabetes incidence will follow? 3) How does the duration of type 2 diabetes relate to the benefits of weight reduction by lifestyle, weight-loss drugs, and/or bariatric surgery on ß-cell function and glycemia? 4) What is necessary for regulatory approval of medications and possibly surgical approaches for preventing type 2 diabetes in patients with obesity? Improved understanding of how obesity relates to type 2 diabetes may help advance effective and cost-effective interventions for both conditions, including more tailored therapy. To expedite this process, we recommend further investigation into the pathogenesis of these coexistent conditions and innovative approaches to their pharmacological and surgical management.


Asunto(s)
Diabetes Mellitus Tipo 2/terapia , Obesidad/terapia , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Obesidad/complicaciones , Obesidad/metabolismo , Pérdida de Peso
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA