Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros











Intervalo de año de publicación
1.
Am J Physiol Gastrointest Liver Physiol ; 314(5): G610-G622, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29420068

RESUMEN

We investigated whether vasoactive intestinal peptide (VIP) and/or prostaglandins contribute to peripheral corticotropin-releasing factor (CRF)-induced CRF1 receptor-mediated stimulation of colonic motor function and diarrhea in rats. The VIP antagonist, [4Cl-D-Phe6, Leu17]VIP injected intraperitoneally completely prevented CRF (10 µg/kg ip)-induced fecal output and diarrhea occurring within the first hour after injection, whereas pretreatment with the prostaglandins synthesis inhibitor, indomethacin, had no effect. In submucosal plexus neurons, CRF induced significant c-Fos expression most prominently in the terminal ileum compared with duodenum and jejunum, whereas no c-Fos was observed in the proximal colon. c-Fos expression in ileal submucosa was colocalized in 93.4% of VIP-positive neurons and 31.1% of non-VIP-labeled neurons. CRF1 receptor immunoreactivity was found on the VIP neurons. In myenteric neurons, CRF induced only a few c-Fos-positive neurons in the ileum and a robust expression in the proximal colon (17.5 ± 2.4 vs. 0.4 ± 0.3 cells/ganglion in vehicle). The VIP antagonist prevented intraperitoneal CRF-induced c-Fos induction in the ileal submucosal plexus and proximal colon myenteric plexus. At 60 min after injection, CRF decreased VIP levels in the terminal ileum compared with saline (0.8 ± 0.3 vs. 2.5 ± 0.7 ng/g), whereas VIP mRNA level detected by qPCR was not changed. These data indicate that intraperitoneal CRF activates intestinal submucosal VIP neurons most prominently in the ileum and myenteric neurons in the colon. It also implicates VIP signaling as part of underlying mechanisms driving the acute colonic secretomotor response to a peripheral injection of CRF, whereas prostaglandins do not play a role. NEW & NOTEWORTHY Corticotropin-releasing factor (CRF) in the gut plays a physiological role in the stimulation of lower gut secretomotor function induced by stress. We showed that vasoactive intestinal peptide (VIP)-immunoreactive neurons in the ileal submucosal plexus expressed CRF1 receptor and were prominently activated by CRF, unlike colonic submucosal neurons. VIP antagonist abrogated CRF-induced ileal submucosal and colonic myenteric activation along with functional responses (defecation and diarrhea). These data point to VIP signaling in ileum and colon as downstream effectors of CRF.


Asunto(s)
Hormona Liberadora de Corticotropina/metabolismo , Diarrea , Motilidad Gastrointestinal , Plexo Mientérico , Péptido Intestinal Vasoactivo , Animales , Colon/metabolismo , Colon/fisiopatología , Defecación/efectos de los fármacos , Defecación/fisiología , Diarrea/metabolismo , Diarrea/fisiopatología , Motilidad Gastrointestinal/efectos de los fármacos , Motilidad Gastrointestinal/fisiología , Genes fos/fisiología , Íleon/metabolismo , Íleon/fisiopatología , Mucosa Intestinal/metabolismo , Masculino , Plexo Mientérico/efectos de los fármacos , Plexo Mientérico/metabolismo , Fármacos Neuroprotectores/metabolismo , Ratas , Péptido Intestinal Vasoactivo/antagonistas & inhibidores , Péptido Intestinal Vasoactivo/metabolismo
2.
J Med Chem ; 59(3): 854-66, 2016 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-26789203

RESUMEN

CRF mediates numerous stress-related endocrine, autonomic, metabolic, and behavioral responses. We present the synthesis and chemical and biological properties of astressin B analogues {cyclo(30-33)[D-Phe(12),Nle(21,38),C(α)MeLeu(27,40),Glu(30),Lys(33)]-acetyl-h/r-CRF(9-41)}. Out of 37 novel peptides, 17 (2, 4, 6-8, 10, 11, 16, 17, 27, 29, 30, 32-36) and 16 (3, 5, 9, 12-15, 18, 19, 22-26, 28, 31) had k(i) to CRF receptors in the high picomolar and low nanomole ranges, respectively. Peptides 1, 2, and 11 inhibited h/rCRF and urocortin 1-induced cAMP release from AtT20 and A7r5 cells. When Astressin C 2 was administered to adrenalectomized rats at 1.0 mg subcutaneously, it inhibited ACTH release for >7 d. Additional rat data based on the inhibitory effect of (2) on h/rCRF-induced stimulation of colonic secretory motor activity and urocortin 2-induced delayed gastric emptying also indicate a safe and long-lasting antagonistic effect. The overall properties of selected analogues may fulfill the criteria expected from clinical candidates.


Asunto(s)
Hormona Liberadora de Corticotropina/farmacología , Fragmentos de Péptidos/farmacología , Receptores de Hormona Liberadora de Corticotropina/antagonistas & inhibidores , Animales , Hormona Liberadora de Corticotropina/administración & dosificación , Hormona Liberadora de Corticotropina/química , AMP Cíclico/antagonistas & inhibidores , Relación Dosis-Respuesta a Droga , Humanos , Estructura Molecular , Fragmentos de Péptidos/administración & dosificación , Fragmentos de Péptidos/química , Ratas , Relación Estructura-Actividad , Urocortinas/antagonistas & inhibidores
3.
PLoS One ; 10(9): e0139325, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26421719

RESUMEN

Abdominal surgery inhibits food intake and induces c-Fos expression in the hypothalamic and medullary nuclei in rats. Rikkunshito (RKT), a Kampo medicine improves anorexia. We assessed the alterations in meal microstructure and c-Fos expression in brain nuclei induced by abdominal surgery and the modulation by RKT in mice. RKT or vehicle was gavaged daily for 1 week. On day 8 mice had no access to food for 6-7 h and were treated twice with RKT or vehicle. Abdominal surgery (laparotomy-cecum palpation) was performed 1-2 h before the dark phase. The food intake and meal structures were monitored using an automated monitoring system for mice. Brain sections were processed for c-Fos immunoreactivity (ir) 2-h after abdominal surgery. Abdominal surgery significantly reduced bouts, meal frequency, size and duration, and time spent on meals, and increased inter-meal interval and satiety ratio resulting in 92-86% suppression of food intake at 2-24 h post-surgery compared with control group (no surgery). RKT significantly increased bouts, meal duration and the cumulative 12-h food intake by 11%. Abdominal surgery increased c-Fos in the prelimbic, cingulate and insular cortexes, and autonomic nuclei, such as the bed nucleus of the stria terminalis, central amygdala, hypothalamic supraoptic (SON), paraventricular and arcuate nuclei, Edinger-Westphal nucleus (E-W), lateral periaqueduct gray (PAG), lateral parabrachial nucleus, locus coeruleus, ventrolateral medulla and nucleus tractus solitarius (NTS). RKT induced a small increase in c-Fos-ir neurons in the SON and E-W of control mice, and in mice with surgery there was an increase in the lateral PAG and a decrease in the NTS. These findings indicate that abdominal surgery inhibits food intake by increasing both satiation (meal duration) and satiety (meal interval) and activates brain circuits involved in pain, feeding behavior and stress that may underlie the alterations of meal pattern and food intake inhibition. RKT improves food consumption post-surgically that may involve modulation of pain pathway.


Asunto(s)
Abdomen/cirugía , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Medicamentos Herbarios Chinos/administración & dosificación , Ingestión de Alimentos/efectos de los fármacos , Extractos Vegetales/administración & dosificación , Abdomen/patología , Administración Oral , Animales , Anorexia/tratamiento farmacológico , Encéfalo/citología , Ingestión de Alimentos/fisiología , Ingestión de Alimentos/psicología , Ghrelina/metabolismo , Masculino , Ratones Endogámicos C57BL , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Dolor/tratamiento farmacológico , Plantas Medicinales , Complicaciones Posoperatorias/tratamiento farmacológico , Complicaciones Posoperatorias/patología , Proteínas Proto-Oncogénicas c-fos/inmunología , Proteínas Proto-Oncogénicas c-fos/metabolismo , Saciedad/efectos de los fármacos , Respuesta de Saciedad/efectos de los fármacos , Tirosina 3-Monooxigenasa/metabolismo
4.
Neurosci Lett ; 576: 88-92, 2014 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-24915296

RESUMEN

Intracerebroventricular (icv) injection of the stable somatostatin pan-agonist, ODT8-SST induces a somatostatin 2 receptor (sst2) mediated robust feeding response that involves neuropeptide Y and opioid systems in rats. We investigated whether the orexigenic system driven by orexin also plays a role. Food and water intake after icv injection was measured concomitantly in non-fasted and non-water deprived rats during the light phase. In vehicle treated rats (100% DMSO, icv), ODT8-SST (1µg/rat, icv) significantly increased the 2-h food and water intake compared to icv vehicle plus saline (5.1±1.0g vs. 1.2±0.4g and 11.3±1.9mL vs. 2.5±1.2mL, respectively). The orexin-1 receptor antagonist, SB-334867 (16µg/rat, icv) completely inhibited the 2-h food and water intake induced by icv ODT8-SST. In contrast, the icv pretreatment with the selective somatostatin sst2 antagonist, S-406-028, established to block the orexigenic effect of icv ODT8-SST, did not modify the increased food and water intake induced by icv orexin-A (10.7µg/rat). These data indicate that orexin-1 receptor signaling system is part of the brain neurocircuitry contributing to the orexigenic and dipsogenic responses induced by icv ODT8-SST and that orexin-A stimulates food intake independently from brain sst2 activation.


Asunto(s)
Ingestión de Líquidos/efectos de los fármacos , Ingestión de Alimentos/efectos de los fármacos , Fragmentos de Péptidos/farmacología , Receptores de Somatostatina/agonistas , Somatostatina/análogos & derivados , Animales , Benzoxazoles/farmacología , Inyecciones Intraventriculares , Péptidos y Proteínas de Señalización Intracelular/farmacología , Masculino , Naftiridinas , Neuropéptidos/farmacología , Octreótido/farmacología , Antagonistas de los Receptores de Orexina , Receptores de Orexina , Orexinas , Ratas Sprague-Dawley , Receptores de Somatostatina/antagonistas & inhibidores , Somatostatina/farmacología , Urea/análogos & derivados , Urea/farmacología
5.
Am J Physiol Regul Integr Comp Physiol ; 305(6): R582-91, 2013 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-23883680

RESUMEN

Obesity is an increasing health problem. Because drug treatments are limited, diets remain popular. High-protein diets (HPD) reduce body weight (BW), although the mechanisms are unclear. We investigated physiological mechanisms altered by switching diet induced obesity (DIO) rats from Western-type diet (WTD) to HPD. Male rats were fed standard (SD) or WTD (45% calories from fat). After developing DIO (50% of rats), they were switched to SD (15% calories from protein) or HPD (52% calories from protein) for up to 4 weeks. Food intake (FI), BW, body composition, glucose tolerance, insulin sensitivity, and intestinal hormone plasma levels were monitored. Rats fed WTD showed an increased FI and had a 25% greater BW gain after 9 wk compared with SD (P < 0.05). Diet-induced obese rats switched from WTD to HPD reduced daily FI by 30% on day 1, which lasted to day 9 (-9%) and decreased BW during the 2-wk period compared with SD/SD (P < 0.05). During these 2 wk, WTD/HPD rats lost 72% more fat mass than WTD/SD (P < 0.05), whereas lean mass was unaltered. WTD/HPD rats had lower blood glucose than WTD/SD at 30 min postglucose gavage (P < 0.05). The increase of pancreatic polypeptide and peptide YY during the 2-h dark-phase feeding was higher in WTD/HPD compared with WTD/SD (P < 0.05). These data indicate that HPD reduces BW in WTD rats, which may be related to decreased FI and the selective reduction of fat mass accompanied by improved glucose tolerance, suggesting relevant benefits of HPD in the treatment of obesity.


Asunto(s)
Adiposidad/efectos de los fármacos , Proteínas en la Dieta/administración & dosificación , Proteínas en la Dieta/metabolismo , Prueba de Tolerancia a la Glucosa , Glucosa/metabolismo , Obesidad/dietoterapia , Obesidad/metabolismo , Animales , Peso Corporal , Masculino , Ratas , Ratas Sprague-Dawley , Resultado del Tratamiento
6.
Obesity (Silver Spring) ; 19(3): 514-21, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20706204

RESUMEN

An obesity-induced diabetes model using genetically normal mouse strains would be invaluable but remains to be established. One reason is that several normal mouse strains are resistant to high-fat diet-induced obesity. In the present study, we show the effectiveness of gold thioglucose (GTG) in inducing hyperphagia and severe obesity in mice, and demonstrate the development of obesity-induced diabetes in genetically normal mouse strains. GTG treated DBA/2, C57BLKs, and BDF1 mice gained weight rapidly and exhibited significant increases in nonfasting plasma glucose levels 8-12 weeks after GTG treatment. These mice showed significantly impaired insulin secretion, particularly in the early phase after glucose load, and reduced insulin content in pancreatic islets. Interestingly, GTG treated C57BL/6 mice did not become diabetic and retained normal early insulin secretion and islet insulin content despite being as severely obese and insulin resistant as the other mice. These results suggest that the pathogenesis of obesity-induced diabetes in GTG-treated mice is attributable to the inability of their pancreatic ß-cells to secrete enough insulin to compensate for insulin resistance. Mice developing obesity-induced diabetes after GTG treatment might be a valuable tool for investigating obesity-induced diabetes. Furthermore, comparing the genetic backgrounds of mice with different susceptibilities to diabetes may lead to the identification of novel genetic factors influencing the ability of pancreatic ß-cells to secrete insulin.


Asunto(s)
Diabetes Mellitus Experimental/etiología , Modelos Animales de Enfermedad , Hiperfagia/inducido químicamente , Células Secretoras de Insulina/fisiología , Insulina/metabolismo , Obesidad Mórbida/complicaciones , Animales , Aurotioglucosa , Glucemia/metabolismo , Diabetes Mellitus Experimental/fisiopatología , Predisposición Genética a la Enfermedad , Intolerancia a la Glucosa , Resistencia a la Insulina/genética , Resistencia a la Insulina/fisiología , Secreción de Insulina , Masculino , Ratones , Ratones Endogámicos , Obesidad Mórbida/inducido químicamente , Obesidad Mórbida/fisiopatología , Especificidad de la Especie
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA