Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
1.
Am J Physiol Endocrinol Metab ; 301(1): E187-95, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21521717

RESUMO

Ingestion of high-fat, high-calorie diets is associated with hyperphagia, increased body fat, and obesity. The mechanisms responsible are currently unclear; however, altered leptin signaling may be an important factor. Vagal afferent neurons (VAN) integrate signals from the gut in response to ingestion of nutrients and express leptin receptors. Therefore, we tested the hypothesis that leptin resistance occurs in VAN in response to a high-fat diet. Sprague-Dawley rats, which exhibit a bimodal distribution of body weight gain, were used after ingestion of a high-fat diet for 8 wk. Body weight, food intake, and plasma leptin levels were measured. Leptin signaling was determined by immunohistochemical localization of phosphorylated STAT3 (pSTAT3) in cultured VAN and by quantifaction of pSTAT3 protein levels by Western blot analysis in nodose ganglia and arcuate nucleus in vivo. To determine the mechanism of leptin resistance in nodose ganglia, cultured VAN were stimulated with leptin alone or with lipopolysaccharide (LPS) and SOCS-3 expression measured. SOCS-3 protein levels in VAN were measured by Western blot following leptin administration in vivo. Leptin resulted in appearance of pSTAT3 in VAN of low-fat-fed rats and rats resistant to diet-induced obesity but not diet-induced obese (DIO) rats. However, leptin signaling was normal in arcuate neurons. SOCS-3 expression was increased in VAN of DIO rats. In cultured VAN, LPS increased SOCS-3 expression and inhibited leptin-induced pSTAT3 in vivo. We conclude that VAN of diet-induced obese rats become leptin resistant; LPS and SOCS-3 may play a role in the development of leptin resistance.


Assuntos
Dieta/efeitos adversos , Resistência a Medicamentos , Leptina/metabolismo , Neurônios Aferentes/metabolismo , Obesidade/complicações , Obesidade/etiologia , Nervo Vago/metabolismo , Animais , Peso Corporal/efeitos dos fármacos , Dieta Aterogênica , Gorduras na Dieta/farmacologia , Resistência a Medicamentos/efeitos dos fármacos , Resistência a Medicamentos/fisiologia , Ingestão de Energia/efeitos dos fármacos , Ingestão de Energia/fisiologia , Leptina/farmacologia , Lipopolissacarídeos/farmacologia , Neurônios Aferentes/efeitos dos fármacos , Neurônios Aferentes/patologia , Obesidade/metabolismo , Obesidade/patologia , Ratos , Ratos Sprague-Dawley , Receptores para Leptina/metabolismo , Proteína 3 Supressora da Sinalização de Citocinas , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Proteínas Supressoras da Sinalização de Citocina/fisiologia , Nervo Vago/efeitos dos fármacos , Nervo Vago/patologia , Doenças do Nervo Vago/etiologia , Doenças do Nervo Vago/metabolismo
2.
Am J Physiol Gastrointest Liver Physiol ; 300(5): G895-902, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21311026

RESUMO

Dietary protein is a major stimulant for cholecystokinin (CCK) secretion by the intestinal I cell, however, the mechanism by which protein is detected is unknown. Indirect functional evidence suggests that PepT1 may play a role in CCK-mediated changes in gastric motor function. However, it is unclear whether this oligopeptide transporter directly or indirectly activates the I cell. Using both the CCK-expressing enteroendocrine STC-1 cell and acutely isolated native I cells from CCK-enhanced green fluorescent protein (eGFP) mice, we aimed to determine whether PepT1 directly activates the enteroendocrine cell to elicit CCK secretion in response to oligopeptides. Both STC-1 cells and isolated CCK-eGFP cells expressed PepT1 transcripts. STC-1 cells were activated, as measured by ERK(1/2) phosphorylation, by both peptone and the PepT1 substrate Cefaclor; however, the PepT1 inhibitor 4-aminomethyl benzoic acid (AMBA) had no effect on STC-1 cell activity. The PepT1-transportable substrate glycyl-sarcosine dose-dependently decreased gastric motility in anesthetized rats but had no affect on activation of STC-1 cells or on CCK secretion by CCK-eGFP cells. CCK secretion was significantly increased in response to peptone but not to Cefaclor, cephalexin, or Phe-Ala in CCK-eGFP cells. Taken together, the data suggest that PepT1 does not directly mediate CCK secretion in response to PepT1 specific substrates. PepT1, instead, may have an indirect role in protein sensing in the intestine.


Assuntos
Colecistocinina/metabolismo , Células Enteroendócrinas/metabolismo , Hidrolisados de Proteína/farmacologia , Simportadores/fisiologia , Animais , Western Blotting , Células CACO-2 , Cefaclor/farmacologia , Linhagem Celular , Separação Celular , Colecistocinina/genética , Eletroforese em Gel de Poliacrilamida , Células Enteroendócrinas/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Motilidade Gastrointestinal/fisiologia , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Transgênicos , Transportador 1 de Peptídeos , Peptonas/farmacologia , Fosforilação , RNA/biossíntese , RNA/isolamento & purificação , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Simportadores/antagonistas & inibidores , Ácido Tranexâmico/metabolismo
3.
PLoS One ; 7(3): e32967, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22412960

RESUMO

BACKGROUND AND AIMS: The gastrointestinal hormone cholecystokinin (CCK) plays an important role in regulating meal size and duration by activating CCK1 receptors on vagal afferent neurons (VAN). Leptin enhances CCK signaling in VAN via an early growth response 1 (EGR1) dependent pathway thereby increasing their sensitivity to CCK. In response to a chronic ingestion of a high fat diet, VAN develop leptin resistance and the satiating effects of CCK are reduced. We tested the hypothesis that leptin resistance in VAN is responsible for reducing CCK signaling and satiation. RESULTS: Lean Zucker rats sensitive to leptin signaling, significantly reduced their food intake following administration of CCK8S (0.22 nmol/kg, i.p.), while obese Zucker rats, insensitive to leptin, did not. CCK signaling in VAN of obese Zucker rats was reduced, preventing CCK-induced up-regulation of Y2 receptor and down-regulation of melanin concentrating hormone 1 receptor (MCH1R) and cannabinoid receptor (CB1). In VAN from diet-induced obese (DIO) Sprague Dawley rats, previously shown to become leptin resistant, we demonstrated that the reduction in EGR1 expression resulted in decreased sensitivity of VAN to CCK and reduced CCK-induced inhibition of food intake. The lowered sensitivity of VAN to CCK in DIO rats resulted in a decrease in Y2 expression and increased CB1 and MCH1R expression. These effects coincided with the onset of hyperphagia in DIO rats. CONCLUSIONS: Leptin signaling in VAN is required for appropriate CCK signaling and satiation. In response to high fat feeding, the onset of leptin resistance reduces the sensitivity of VAN to CCK thus reducing the satiating effects of CCK.


Assuntos
Colecistocinina/metabolismo , Leptina/metabolismo , Neurônios Aferentes/metabolismo , Saciação , Transdução de Sinais , Nervo Vago/metabolismo , Animais , Peso Corporal , Células Cultivadas , Colecistocinina/farmacologia , Dieta Hiperlipídica , Ingestão de Alimentos , Leptina/farmacologia , Masculino , Neurônios Aferentes/efeitos dos fármacos , Obesidade/etiologia , Fenótipo , Ratos , Ratos Sprague-Dawley , Ratos Zucker , Saciação/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Nervo Vago/efeitos dos fármacos
4.
Am J Physiol Regul Integr Comp Physiol ; 296(3): R528-36, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19073908

RESUMO

G protein-coupled receptors that signal bitter taste (T2Rs) are expressed in the mucosal lining of the oral cavity and gastrointestinal (GI) tract. In mice, intragastric infusion of T2R ligands activates Fos expression within the caudal viscerosensory portion of the nucleus of the solitary tract (NTS) through a vagal pathway (Hao S, Sternini C, Raybould HE. Am J Physiol Regul Integr Comp Physiol 294: R33-R38, 2008). The present study was performed in rats to further characterize the distribution and chemical phenotypes of brain stem and forebrain neurons activated to express Fos after intragastric gavage of T2R ligands, and to determine a potential behavioral correlate of this central neural activation. Compared with relatively low brain stem and forebrain Fos expression in control rats gavaged intragastrically with water, rats gavaged intragastrically with T2R ligands displayed significantly increased activation of neurons within the caudal medial (visceral) NTS and caudal ventrolateral medulla, including noradrenergic neurons, and within the lateral parabrachial nucleus, central nucleus of the amygdala, and paraventricular nucleus of the hypothalamus. A behavioral correlate of this Fos activation was evidenced when rats avoided consuming flavors that previously were paired with intragastric gavage of T2R ligands. While unconditioned aversive responses to bitter tastants in the oral cavity are often sufficient to inhibit further consumption, a second line of defense may be provided postingestively by ligand-induced signaling at GI T2Rs that signal the brain via vagal sensory inputs to the caudal medulla.


Assuntos
Aprendizagem da Esquiva/efeitos dos fármacos , Genes fos/efeitos dos fármacos , Receptores Acoplados a Proteínas G/efeitos dos fármacos , Paladar/efeitos dos fármacos , Tonsila do Cerebelo/fisiologia , Animais , Técnicas Imunoenzimáticas , Imuno-Histoquímica , Intubação Gastrointestinal , Ligantes , Masculino , Vias Neurais/citologia , Vias Neurais/efeitos dos fármacos , Vias Neurais/fisiologia , Norepinefrina/fisiologia , Propiltiouracila/farmacologia , Compostos de Amônio Quaternário/farmacologia , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa