Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 464
Filtrar
1.
Am J Physiol Endocrinol Metab ; 322(1): E10-E23, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34779255

RESUMEN

Cholecystokinin (CCK) increases core body temperature via CCK2 receptors when administered intracerebroventricularly (icv). The mechanisms of CCK-induced hyperthermia are unknown, and it is also unknown whether CCK contributes to the fever response to systemic inflammation. We studied the interaction between central CCK signaling and the cyclooxygenase (COX) pathway. Body temperature was measured in adult male Wistar rats pretreated with intraperitoneal infusion of the nonselective COX enzyme inhibitor metamizol (120 mg/kg) or a selective COX-2 inhibitor, meloxicam, or etoricoxib (10 mg/kg for both) and, 30 min later, treated with intracerebroventricular CCK (1.7 µg/kg). In separate experiments, CCK-induced neuronal activation (with and without COX inhibition) was studied in thermoregulation- and feeding-related nuclei with c-Fos immunohistochemistry. CCK increased body temperature by ∼0.4°C from 10 min postinfusion, which was attenuated by metamizol. CCK reduced the number of c-Fos-positive cells in the median preoptic area (by ∼70%) but increased it in the dorsal hypothalamic area and in the rostral raphe pallidus (by ∼50% in both); all these changes were completely blocked with metamizol. In contrast, CCK-induced satiety and neuronal activation in the ventromedial hypothalamus were not influenced by metamizol. CCK-induced hyperthermia was also completely blocked with both selective COX-2 inhibitors studied. Finally, the CCK2 receptor antagonist YM022 (10 µg/kg icv) attenuated the late phases of fever induced by bacterial lipopolysaccharide (10 µg/kg; intravenously). We conclude that centrally administered CCK causes hyperthermia through changes in the activity of "classical" thermoeffector pathways and that the activation of COX-2 is required for the development of this response.NEW & NOTEWORTHY An association between central cholecystokinin signaling and the cyclooxygenase-prostaglandin E pathway has been proposed but remained poorly understood. We show that the hyperthermic response to the central administration of cholecystokinin alters the neuronal activity within efferent thermoeffector pathways and that these effects are fully blocked by the inhibition of cyclooxygenase. We also show that the activation of cyclooxygenase-2 is required for the hyperthermic effect of cholecystokinin and that cholecystokinin is a modulator of endotoxin-induced fever.


Asunto(s)
Temperatura Corporal/efectos de los fármacos , Colecistoquinina/administración & dosificación , Ciclooxigenasa 2/metabolismo , Hipertermia/inducido químicamente , Hipertermia/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Anorexia/inducido químicamente , Benzodiazepinas/administración & dosificación , Regulación de la Temperatura Corporal/efectos de los fármacos , Colecistoquinina/efectos adversos , Inhibidores de la Ciclooxigenasa 2/administración & dosificación , Modelos Animales de Enfermedad , Ingestión de Alimentos/efectos de los fármacos , Fiebre/inducido químicamente , Fiebre/tratamiento farmacológico , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Inyecciones Intraventriculares , Lipopolisacáridos/efectos adversos , Masculino , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas , Ratas Wistar , Receptor de Colecistoquinina B/antagonistas & inhibidores , Resultado del Tratamiento
2.
Endocr Res ; 46(2): 57-65, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33426974

RESUMEN

The vagus nerve and the celiaco-mesenteric ganglia (CMG) are required for reduction of meal size (MS) and prolongation of the intermeal interval (IMI) by intraperitoneal (ip) sulfated cholecystokinin-8 (CCK-8). However, recently we have shown that the gut regulates these responses. Therefore, reevaluating the role of the vagus and the CMG in the feeding responses evoked by CCK is necessary because the gut contains the highest concentration of enteric, vagal and splanchnic afferents and CCK-A receptors, which are required for reduction of food intake by this peptide, compared to other abdominal organs. To address this necessity, we injected sulfated CCK-8 (0, 0.1, 0.5, 1 and 3 nmol/kg) in the aorta, near the gastrointestinal sites of action of the peptide, in three groups of free-feeding rats (n = 10 rats per group), subdiaphragmatic vagotomy (VGX), celiaco-mesenteric ganglionectomy (CMGX) and sham-operated, and recorded seven feeding responses. In the sham group, CCK-8 reduced MS (normal chow), prolonged the intermeal interval (IMI, time between first and second meals), increased satiety ratio (SR, IMI/MS), shortened duration of first meal, reduced total (24 hrs) food intake and reduced number of meals relative to saline vehicle. Vagotomy attenuated all of the previous responses except IMI length and SR, and CMGX attenuated all of those responses. In conclusion, the feeding responses evoked by sulfated CCK-8 require, independently, the vagus nerve and the CMG.


Asunto(s)
Conducta Animal/fisiología , Colecistoquinina/farmacología , Conducta Alimentaria/fisiología , Ganglios Simpáticos/fisiología , Fragmentos de Péptidos/farmacología , Saciedad/fisiología , Simpatectomía , Vagotomía , Nervio Vago/fisiología , Animales , Arteria Celíaca , Colecistoquinina/administración & dosificación , Conducta Alimentaria/efectos de los fármacos , Ganglios Simpáticos/cirugía , Masculino , Fragmentos de Péptidos/administración & dosificación , Ratas , Ratas Sprague-Dawley , Saciedad/efectos de los fármacos , Factores de Tiempo , Nervio Vago/cirugía
3.
Am J Physiol Regul Integr Comp Physiol ; 318(5): R1014-R1023, 2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-32292065

RESUMEN

In rats, overnight fasting reduces the ability of systemic cholecystokinin-8 (CCK) to suppress food intake and to activate cFos in the caudal nucleus of the solitary tract (cNTS), specifically within glucagon-like peptide-1 (GLP-1) and noradrenergic (NA) neurons of the A2 cell group. Systemic CCK increases vagal sensory signaling to the cNTS, an effect that is amplified by leptin and reduced by ghrelin. Since fasting reduces plasma leptin and increases plasma ghrelin levels, we hypothesized that peripheral leptin administration and/or antagonism of ghrelin receptors in fasted rats would rescue the ability of CCK to activate GLP-1 neurons and a caudal subset of A2 neurons that coexpress prolactin-releasing peptide (PrRP). To test this, cFos expression was examined in ad libitum-fed and overnight food-deprived (DEP) rats after intraperitoneal CCK, after coadministration of leptin and CCK, or after intraperitoneal injection of a ghrelin receptor antagonist (GRA) before CCK. In fed rats, CCK activated cFos in ~60% of GLP-1 and PrRP neurons. Few or no GLP-1 or PrRP neurons expressed cFos in DEP rats treated with CCK alone, CCK combined with leptin, or GRA alone. However, GRA pretreatment increased the ability of CCK to activate GLP-1 and PrRP neurons and also enhanced the hypophagic effect of CCK in DEP rats. Considered together, these new findings suggest that reduced behavioral sensitivity to CCK in fasted rats is at least partially due to ghrelin-mediated suppression of hindbrain GLP-1 and PrRP neural responsiveness to CCK.


Asunto(s)
Regulación del Apetito/efectos de los fármacos , Colecistoquinina/administración & dosificación , Ingestión de Alimentos/efectos de los fármacos , Ayuno/metabolismo , Conducta Alimentaria/efectos de los fármacos , Ghrelina/sangre , Neuronas/efectos de los fármacos , Rombencéfalo/efectos de los fármacos , Animales , Péptido 1 Similar al Glucagón/metabolismo , Leptina/sangre , Masculino , Neuronas/metabolismo , Hormona Liberadora de Prolactina/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas Sprague-Dawley , Receptores de Ghrelina/metabolismo , Rombencéfalo/metabolismo , Transducción de Señal
4.
Endocr Res ; 45(2): 73-83, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31573821

RESUMEN

We have shown that non-sulfated cholecystokinin-8 (NS CCK-8) reduces food intake in adult male Sprague Dawley rats by activating cholecystokinin-B receptor (CCK-BR). Here, we tested the hypothesis that the vagus nerve and the celiaco-mesenteric ganglia may play a role in this reduction. The hypothesis stems from the following facts. The vagus and the celiaco-mesenteric ganglia contain NS CCK-8, they express and have binding sites for CCK-BR, NS CCK-8 activates CCK-BR on afferent vagal and sympathetic fibers and the two structures link the gastrointestinal tract to central feeding nuclei in the brain, which also contain the peptide and CCK-BR. To test this hypothesis, three groups of free-feeding rats, vagotomy (VGX), celiaco-mesenteric ganglionectomy (CMGX) and sham-operated, received NS CCK-8 (0, 0.5 and 1 nmol/kg) intraperitoneally prior to the onset of the dark cycle and various feeding behaviors were recorded. We found that in sham-operated rats both doses of NS CCK-8 reduced meal size (MS), prolonged the intermeal interval (IMI, time between first and second meal), increased satiety ratio (SR = IMI/MS), reduced 24-h food intake and reduced the number of meals relative to saline control. In the VGX and the CMGX groups, all of the previous responses were attenuated. Consistent with our hypothesis, the findings of the current work suggest a role for the vagus nerve and the celiaco-mesenteric ganglia in the feeding responses evoked by NS CCK-8.


Asunto(s)
Conducta Animal/fisiología , Colecistoquinina/metabolismo , Conducta Alimentaria/fisiología , Ganglios Simpáticos/metabolismo , Fragmentos de Péptidos/metabolismo , Nervio Vago/metabolismo , Animales , Conducta Animal/efectos de los fármacos , Colecistoquinina/administración & dosificación , Conducta Alimentaria/efectos de los fármacos , Masculino , Fragmentos de Péptidos/administración & dosificación , Ratas , Ratas Sprague-Dawley
5.
Horm Behav ; 106: 105-111, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30342011

RESUMEN

Cholecystokinin (CCK) is one of the main neurohormone peptide systems in the brain, and a major anxiogenic mediator. The periaqueductal gray (PAG) is a key midbrain structure for defensive behaviors, which could include anxiety, fear, or even panic. The CCK system has wide distribution in the PAG, where the dorsolateral region (DL) participates in active defensive behavior and the ventrolateral region (VL) in passive defensive behavior. The aim of this study was to assess the effect of CCK-8 microinjection into DL-PAG or VL-PAG on anxiety-like behavior through two tests: elevated plus maze (EPM) and defensive burying behavior (DBB). CCK-8 (0.5 and 1.0 µg/0.5 µL) presently microinjected into the DL-PAG produced an anxiogenic-like effect on the EPM evidenced by decreasing the time spent/number of entries in open arms compared to vehicle group. Additionally, the latency to burying decreased and burying time increased on the DBB test. Contrarily, CCK-8 microinjected into the VL-PAG resulted in greater open-arm time and more open-arm entries compared to the vehicle-microinjected group. The results on the DBB test confirmed an anxiolytic-like response of CCK-8 into the VL-PAG. In conclusion, CCK-8 microinjected into DL-PAG produced anxiety-like behavior on EPM, and for first time reported on DBB. Contrarily, CCK-8 microinjected into the VL-PAG reduced anxiety-like behavior also for first time reported using both behavioral models EPM and DBB.


Asunto(s)
Ansiedad/patología , Colecistoquinina/farmacología , Sustancia Gris Periacueductal/efectos de los fármacos , Animales , Ansiolíticos/administración & dosificación , Ansiolíticos/farmacología , Ansiedad/metabolismo , Conducta Animal/efectos de los fármacos , Colecistoquinina/administración & dosificación , Modelos Animales de Enfermedad , Reacción de Fuga/efectos de los fármacos , Miedo/efectos de los fármacos , Miedo/fisiología , Inyecciones Intraventriculares , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Microinyecciones , Ratas , Ratas Wistar , Sincalida/farmacología
6.
Appetite ; 127: 334-340, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29782892

RESUMEN

Combination approaches for the treatment of metabolic diseases such as obesity and diabetes are becoming increasingly relevant. Co-administration of a glucagon-like peptide-1 receptor (GLP-1R) agonist with a cholecystokinin receptor-1 (CCKR1) agonist exert synergistic effects on weight loss in obese rodents. Here, we report on the effects of a novel fusion peptide (C2816) comprised of a stabilized GLP-1R agonist, AC3174, and a CCKR1-selective agonist, AC170222. C2816 was constructed such that AC3174 was linked to the N-terminus of AC170222, thus preserving the C-terminal amide of the CCK moiety. In functional in vitro assays C2816 retained full agonism at GLP-1R and CCKR1 at lower potency compared to parent molecules, whereas a previously reported fusion peptide in the opposite orientation, (pGlu-Gln)-CCK-8/exendin-4, exhibited no activity at either receptor. Acutely, in vivo, C2816 increased cFos in key central nuclei relevant to feeding behavior, and reduced food intake in wildtype (WT), but less so in GLP-1R-deficient (GLP-1RKO), mice. In sub-chronic studies in diet-induced obese (DIO) mice, C2816 exerted superior reduction in body weight compared to co-administration of AC3174 and AC170222 albeit at a higher molar dose. These data suggest that the synergistic pharmacological effects of GLP-1 and CCK pathways can be harnessed in a single therapeutic peptide.


Asunto(s)
Fármacos Antiobesidad/química , Colecistoquinina/química , Péptido 1 Similar al Glucagón/química , Receptor del Péptido 1 Similar al Glucagón/agonistas , Receptor de Colecistoquinina A/agonistas , Animales , Fármacos Antiobesidad/administración & dosificación , Fármacos Antiobesidad/farmacología , Encéfalo/efectos de los fármacos , Colecistoquinina/administración & dosificación , Sinergismo Farmacológico , Ingestión de Alimentos/efectos de los fármacos , Péptido 1 Similar al Glucagón/administración & dosificación , Receptor del Péptido 1 Similar al Glucagón/deficiencia , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/tratamiento farmacológico , Péptidos/administración & dosificación , Péptidos/química , Péptidos/farmacología , Ratas Sprague-Dawley , Pérdida de Peso
7.
Ecotoxicol Environ Saf ; 153: 181-187, 2018 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-29433086

RESUMEN

Anorexia is a hallmark of animal and human exposed to T-2 toxin, a most poisonous trichothecene mycotoxins contaminating various cereal grains including wheat, corn and barley. Although this adverse effect has been well characterized in several animal species, the underlying mechanisms are unclear. The goal for this study was to elucidate the roles of two gut satiety hormones, glucagon-like peptide-17-36 amide (GLP-1) and cholecystokinin (CCK) in T-2 toxin-evoked anorectic response using a mouse anorexia bioassay. Elevations of plasma GLP-1 and CCK significantly corresponded to anorexia induction by T-2 toxin. Direct administration of exogenous GLP-1 and CCK markedly evoked anorectic responses similar to T-2 toxin. The GLP-1 receptor (GLP-1R) antagonist Exendin9-39 dose-dependently cause attenuation of both GLP-1- and T-2 toxin-induced anorectic responses. Pretreatment with the CCK1 receptor (CCK1R) antagonist SR 27897 and CCK2 receptor (CCK2R) antagonist L-365,260 attenuated anorexia induction by both CCK- and T-2 toxin in a dose dependent manner. Taken together, our findings suggest that both GLP-1 and CCK play contributory roles in T-2 toxin-induced anorexia.


Asunto(s)
Anorexia/inducido químicamente , Colecistoquinina/sangre , Péptido 1 Similar al Glucagón/sangre , Fragmentos de Péptidos/sangre , Toxina T-2/toxicidad , Animales , Anorexia/sangre , Colecistoquinina/administración & dosificación , Relación Dosis-Respuesta a Droga , Ingestión de Alimentos/efectos de los fármacos , Femenino , Péptido 1 Similar al Glucagón/administración & dosificación , Ratones , Ratones Endogámicos , Fragmentos de Péptidos/administración & dosificación
8.
Appetite ; 109: 172-181, 2017 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-27916474

RESUMEN

We hypothesized that exogenous gastrin releasing peptide-29 (GRP-29), cholecystokinin-8 (CCK-8) and their combination reduce body weight (BW). To test this hypothesis, BW was measured in four groups of diet-induced obese (DIO) male rats infused in the aorta (close to the junctions of the celiac and cranial mesenteric arteries) with saline, CCK-8 (0.5 nmol/kg), GRP-29 (0.5 nmol/kg) and CCK-8+GRP-29 (0.5 nmol/kg each) once daily for a total of 23 days. We found that CCK-8, GRP-29 and CCK-8+GRP-29 reduce BW relative to saline control. In conclusion, CCK-8, GRP-29 and their combination reduce BW in the DIO rat model. If infused near their gastrointestinal sites of action CCK-8, GRP-29 and their combination may have a role in regulating BW.


Asunto(s)
Peso Corporal/efectos de los fármacos , Colecistoquinina/administración & dosificación , Péptido Liberador de Gastrina/administración & dosificación , Fármacos Gastrointestinales/administración & dosificación , Obesidad/tratamiento farmacológico , Fragmentos de Péptidos/administración & dosificación , Pérdida de Peso , Animales , Dieta/efectos adversos , Quimioterapia Combinada , Infusiones Parenterales , Masculino , Obesidad/etiología , Ratas
9.
Artículo en Inglés | MEDLINE | ID: mdl-28377124

RESUMEN

The regulation of energy in fish, like most vertebrates, is a complex process that involves a number of brain and peripheral hormones. These signals include anorexigenic (e.g. cholecystokinin (CCK) and cocaine- and amphetamine-regulated transcript (CART)) as well as orexigenic (e.g. orexin and neuropeptide Y (NPY)) peptides. Platyfish, Xiphophorus maculatus, are freshwater viviparous fish for which little is known about the endocrine mechanisms regulating feeding. In order to elucidate the role of these peptides in the regulation of feeding of platyfish, we examined the effects of peripheral injections of CCK and orexin on feeding behavior and food intake. Injections of CCK decreased both food intake and searching behavior, while injections of orexin increased searching behavior but did not affect food consumption. In order to better characterize these peptides, we examined their mRNA tissue distribution and assessed the effects of a 10-day fast on their brain and intestine expressions in both males and females. CCK, CART, NPY and orexin all show widespread distributions in brain and several peripheral tissues, including intestine and gonads. Fasting induced decreases in both CCK and CART and an increase in orexin mRNA expressions in the brain and a decrease in CCK expression in the intestine, but did not affect either expressions of NPY. There were no significant sex-specific differences in either the behavioral responses to injections or the expression responses to fasting. The widespread distribution and the fasting-induced changes in expression of these peptides suggest that they might have several physiological roles in platyfish, including the regulation of feeding.


Asunto(s)
Apetito/genética , Metabolismo Energético/genética , Conducta Alimentaria/fisiología , Neuropéptido Y/genética , Animales , Apetito/efectos de los fármacos , Colecistoquinina/administración & dosificación , Colecistoquinina/metabolismo , Ciprinodontiformes/fisiología , Ingestión de Alimentos/efectos de los fármacos , Ingestión de Alimentos/genética , Metabolismo Energético/efectos de los fármacos , Ayuno , Conducta Alimentaria/efectos de los fármacos , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Masculino , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuropéptido Y/metabolismo , Orexinas/administración & dosificación , Orexinas/metabolismo , ARN Mensajero
10.
Hepatobiliary Pancreat Dis Int ; 16(2): 197-201, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28381385

RESUMEN

BACKGROUND: Despite the increasing use of fatty meal (FM) as a substitute for cholecystokinin (CCK) in pain reproduction during hepato-imino-diacetic acid (HIDA) scan in functional gallbladder disorder, there are no studies comparing the differences between CCK and FM. The present study was to compare the efficacy of FM in comparison of CCK in FGBD application. METHODS: Patients undergoing HIDA scans from August 2013 to May 2014 were divided into two groups: those undergoing CCK-stimulated HIDA scan versus FM-stimulated HIDA scan. These groups were compared according to demographics and HIDA results. RESULTS: Of 153 patients, 70 received CCK and 83 FM. There was no difference regarding age, gender, gallstones, gallbladder ejection fraction and time to visualization. However, significantly more of the patients receiving CCK than FM experienced pain reproduction (61% vs 30%, P<0.01). CONCLUSIONS: Stimulation of gallbladder contractility with a FM during HIDA is less than half as likely to reproduce biliary symptoms compared to CCK, despite similar ejection fractions and other parameters. It is essential that providers account for this difference when counseling patients regarding cholecystectomy for functional gallbladder disorder.


Asunto(s)
Discinesia Biliar/diagnóstico por imagen , Vesícula Biliar/diagnóstico por imagen , Iminoácidos/administración & dosificación , Radiofármacos/administración & dosificación , Dolor Abdominal/etiología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Discinesia Biliar/fisiopatología , Discinesia Biliar/cirugía , Colecistectomía , Colecistoquinina/administración & dosificación , Colecistoquinina/efectos adversos , Grasas de la Dieta/administración & dosificación , Grasas de la Dieta/efectos adversos , Femenino , Vesícula Biliar/fisiopatología , Vesícula Biliar/cirugía , Humanos , Masculino , Persona de Mediana Edad , Valor Predictivo de las Pruebas , Vitamina K/administración & dosificación , Vitamina K/efectos adversos , Adulto Joven
11.
Am J Physiol Regul Integr Comp Physiol ; 311(5): R930-R939, 2016 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-27534875

RESUMEN

Increased neuropeptide Y (NPY) gene expression in the dorsomedial hypothalamus (DMH) has been shown to cause hyperphagia, but the pathway underlying this effect remains less clear. Hypothalamic neural systems play a key role in the control of food intake, in part, by modulating the effects of meal-related signals, such as cholecystokinin (CCK). An increase in DMH NPY gene expression decreases CCK-induced satiety. Since activation of catecholaminergic neurons within the nucleus of solitary tract (NTS) contributes to the feeding effects of CCK, we hypothesized that DMH NPY modulates NTS neural catecholaminergic signaling to affect food intake. We used an adeno-associated virus system to manipulate DMH NPY gene expression in rats to examine this pathway. Viral-mediated hrGFP anterograde tracing revealed that DMH NPY neurons project to the NTS; the projections were in close proximity to catecholaminergic neurons, and some contained NPY. Viral-mediated DMH NPY overexpression resulted in an increase in NPY content in the NTS, a decrease in NTS tyrosine hydroxylase (TH) expression, and reduced exogenous CCK-induced satiety. Knockdown of DMH NPY produced the opposite effects. Direct NPY administration into the fourth ventricle of intact rats limited CCK-induced satiety and overall TH phosphorylation. Taken together, these results demonstrate that DMH NPY descending signals affect CCK-induced satiety, at least in part, via modulation of NTS catecholaminergic neuronal signaling.


Asunto(s)
Tronco Encefálico/fisiología , Catecolaminas/metabolismo , Colecistoquinina/administración & dosificación , Núcleo Hipotalámico Dorsomedial/fisiología , Neuropéptido Y/metabolismo , Respuesta de Saciedad/fisiología , Animales , Tronco Encefálico/efectos de los fármacos , Colagogos y Coleréticos/administración & dosificación , Colagogos y Coleréticos/farmacología , Colecistoquinina/farmacología , Núcleo Hipotalámico Dorsomedial/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ingestión de Alimentos/efectos de los fármacos , Ingestión de Alimentos/fisiología , Masculino , Neuronas/efectos de los fármacos , Neuronas/fisiología , Ratas , Ratas Sprague-Dawley , Respuesta de Saciedad/efectos de los fármacos , Transmisión Sináptica/fisiología
12.
Horm Behav ; 67: 48-53, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25479193

RESUMEN

The site(s) of action that control meal size and intermeal interval (IMI) length by cholecystokinin-58 (CCK-58), the only detectable endocrine form of CCK in the rat, are not known. To test the hypothesis that the gastrointestinal tract may contain such sites, we infused low doses of CCK-58 (0.01, 0.05, 0.15 and 0.25nmol/kg) into the celiac artery (CA, supplying stomach and upper duodenum), the cranial mesenteric artery (CMA, supplying small and most of the large intestines), the femoral artery (FA, control) and the portal vein (PV, draining the gastrointestinal tract) prior to the onset of the dark cycle in freely fed male rats. We measured the first meal size (chow), second meal size, IMI and satiety ratio (SR, IMI/meal size). We found that (1) all doses of CCK-58 given in the CA and the highest dose given in the CMA reduced the first meal size, (2) all doses of CCK-58 given in the CA reduced the second meal size, (3) a CCK-58 dose of 0.15nmol/kg given in the CA and 0.15 and 0.25nmol/kg given in the CMA prolonged the IMI, (4) CCK-58 (0.05, 0.15, 0.25nmol/kg) given in the CA and 0.25nmol/kg given in the CMA increased the SR, and (5) CCK-58 given in the FA and PV had no effect on the meal size or intermeal interval. These results support our hypothesis that the gastrointestinal tract contains sites of action that regulate meal size and IMI length via CCK-58. The stomach and upper duodenum may contain sites regulating meal size, whereas the small intestine and part of the large intestine may contain sites regulating the IMI.


Asunto(s)
Arteria Celíaca/metabolismo , Colagogos y Coleréticos/farmacología , Colecistoquinina/farmacología , Conducta Alimentaria/fisiología , Arteria Mesentérica Superior/metabolismo , Respuesta de Saciedad/fisiología , Animales , Colagogos y Coleréticos/administración & dosificación , Colecistoquinina/administración & dosificación , Arteria Femoral/metabolismo , Masculino , Vena Porta/metabolismo , Ratas , Ratas Sprague-Dawley , Respuesta de Saciedad/efectos de los fármacos
13.
Diabetes Obes Metab ; 17(1): 61-73, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25204356

RESUMEN

AIM: To test the impact of cholecystokinin (CCK) plus either amylin or a glucagon-like peptide-1 receptor (GLP-1R) agonist on metabolic variables in diet-induced obese (DIO) rodents. METHODS: A stabilized acetylated version of CCK-8 (Ac-Y*-CCK-8), selective CCK1 receptor (CCK1R) or CCK2 receptor (CCK2R) agonists, amylin or the GLP-1R agonist and exenatide analogue AC3174 were administered in select combinations via continuous subcutaneous infusion to DIO rats for 14 days, or Lep(ob) /Lep(ob) mice for 28 days, and metabolic variables were assessed. RESULTS: Combined administration of Ac-Y*-CCK-8 with either amylin or AC3174 induced greater than additive weight loss in DIO rats, with the overall magnitude of effect being greater with AC3174 + Ac-Y*-CCK-8 treatment. Co-infusion of AC3174 with a specific CCK1R agonist, but not a CCK2R agonist, recapitulated the weight loss mediated by AC3174 + Ac-Y*-CCK-8 in DIO rats, suggesting that synergy is mediated by CCK1R activation. In a 4 × 4 full-factorial response surface methodology study in DIO rats, a synergistic interaction between AC3174 and the CCK1R-selective agonist on body weight and food intake was noted. Co-administration of AC3174 and the CCK1R-selective agonist to obese diabetic Lep(ob) /Lep(ob) mice elicited a significantly greater reduction in percentage of glycated haemoglobin and food intake relative to the sum effects of monotherapy groups. CONCLUSIONS: The anti-obesity and antidiabetic potential of combined GLP-1R and CCK1R agonism is an approach that warrants further investigation.


Asunto(s)
Fármacos Antiobesidad/uso terapéutico , Colecistoquinina/análogos & derivados , Diabetes Mellitus/tratamiento farmacológico , Hipoglucemiantes/uso terapéutico , Polipéptido Amiloide de los Islotes Pancreáticos/uso terapéutico , Obesidad/tratamiento farmacológico , Péptidos/uso terapéutico , Acetilación , Animales , Fármacos Antiobesidad/administración & dosificación , Fármacos Antiobesidad/efectos adversos , Colecistoquinina/administración & dosificación , Colecistoquinina/efectos adversos , Colecistoquinina/uso terapéutico , Diabetes Mellitus/metabolismo , Dieta Alta en Grasa/efectos adversos , Sinergismo Farmacológico , Quimioterapia Combinada/efectos adversos , Ingestión de Energía/efectos de los fármacos , Receptor del Péptido 1 Similar al Glucagón , Hipoglucemiantes/administración & dosificación , Hipoglucemiantes/efectos adversos , Infusiones Subcutáneas , Polipéptido Amiloide de los Islotes Pancreáticos/administración & dosificación , Polipéptido Amiloide de los Islotes Pancreáticos/efectos adversos , Masculino , Ratones Mutantes , Obesidad/complicaciones , Obesidad/etiología , Obesidad/metabolismo , Péptidos/administración & dosificación , Péptidos/efectos adversos , Distribución Aleatoria , Ratas Sprague-Dawley , Receptor de Colecistoquinina A/agonistas , Receptor de Colecistoquinina A/metabolismo , Receptor de Colecistoquinina B/agonistas , Receptor de Colecistoquinina B/metabolismo , Receptores de Glucagón/agonistas , Receptores de Glucagón/metabolismo , Pérdida de Peso/efectos de los fármacos
14.
Artículo en Inglés | MEDLINE | ID: mdl-26151373

RESUMEN

Both intrinsic and extrinsic factors modulate food intake and glycemia in vertebrates, in part through interactions with hypothalamic neuropeptide Y (NPY) and proopiomelanocortin (POMC) neurons. The objective of this project was to elucidate the effects of ghrelin (GHRL), gastrin-releasing peptide (GRP), cholecystokinin (CCK), glucagon-like peptide (GLP), pancreatic polypeptide (PP), and peptide YY (PYY) on appetite, glycemia, and hypothalamic expression of NPY and POMC in channel catfish. Catfish were injected intraperitoneally with a single peptide at concentrations of either 0 (control), 50, 100, or 200 ng/g body weight (BW), respectively. Fish were allowed to recover for 30 min, and then fed to satiation over 1 h. Feed intake was determined 1h post-feeding. Catfish injected with GHRL at 50 and 100 ng/g BW and GRP at 200 ng/g BW consumed significantly (P<0.05) less feed compared to controls. A tendency (P<0.1) to suppress feed intake was also observed in the 200 ng/g BW GHRL and PP treatments. PYY, CCK, and GLP had no effects on feed intake. Glycemia was not affected by GHRL, GRP, PP, and PYY treatments, but was suppressed by CCK. A tendency toward lower plasma glucose concentrations was observed in fish administered GLP at 50 ng/g BW. Hypothalamic NPY expression was highly variable and not significantly affected by treatment. POMC expression was also variable, but tended to be reduced by the highest concentration of CCK. These results provide new insight into the roles and regulation of gut neuropeptides in catfish appetite and glycemia.


Asunto(s)
Glucemia/metabolismo , Ingestión de Alimentos/efectos de los fármacos , Ictaluridae/fisiología , Neuropéptido Y/genética , Hormonas Peptídicas/farmacología , Proopiomelanocortina/genética , Animales , Colecistoquinina/administración & dosificación , Colecistoquinina/farmacología , Relación Dosis-Respuesta a Droga , Ingestión de Alimentos/fisiología , Péptido Liberador de Gastrina/administración & dosificación , Péptido Liberador de Gastrina/farmacología , Expresión Génica/efectos de los fármacos , Ghrelina/administración & dosificación , Ghrelina/farmacología , Péptidos Similares al Glucagón/administración & dosificación , Péptidos Similares al Glucagón/farmacología , Hipotálamo/metabolismo , Ictaluridae/sangre , Ictaluridae/genética , Inyecciones Intraperitoneales , Polipéptido Pancreático/administración & dosificación , Polipéptido Pancreático/farmacología , Hormonas Peptídicas/administración & dosificación , Péptido YY/administración & dosificación , Péptido YY/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
15.
Am J Physiol Endocrinol Metab ; 307(8): E619-29, 2014 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-25117406

RESUMEN

Cholecystokinin (CCK)-induced suppression of feeding is mediated by vagal sensory neurons that are destroyed by the neurotoxin capsaicin (CAP). Here we determined whether CAP-sensitive neurons mediate anorexic responses to intravenous infusions of gut hormones peptide YY-(3-36) [PYY-(3-36)] and glucagon-like peptide-1 (GLP-1). Rats received three intraperitoneal injections of CAP or vehicle (VEH) in 24 h. After recovery, non-food-deprived rats received at dark onset a 3-h intravenous infusion of CCK-8 (5, 17 pmol·kg⁻¹·min⁻¹), PYY-(3-36) (5, 17, 50 pmol·kg⁻¹·min⁻¹), or GLP-1 (17, 50 pmol·kg⁻¹·min⁻¹). CCK-8 was much less effective in reducing food intake in CAP vs. VEH rats. CCK-8 at 5 and 17 pmol·kg⁻¹·min⁻¹ reduced food intake during the 3-h infusion period by 39 and 71% in VEH rats and 7 and 18% in CAP rats. In contrast, PYY-(3-36) and GLP-1 were similarly effective in reducing food intake in VEH and CAP rats. PYY-(3-36) at 5, 17, and 50 pmol·kg⁻¹·min⁻¹ reduced food intake during the 3-h infusion period by 15, 33, and 70% in VEH rats and 13, 30, and 33% in CAP rats. GLP-1 at 17 and 50 pmol·kg⁻¹·min⁻¹ reduced food intake during the 3-h infusion period by 48 and 60% in VEH rats and 30 and 52% in CAP rats. These results suggest that anorexic responses to PYY-(3-36) and GLP-1 are not primarily mediated by the CAP-sensitive peripheral sensory neurons (presumably vagal) that mediate CCK-8-induced anorexia.


Asunto(s)
Anorexia/fisiopatología , Colecistoquinina/metabolismo , Modelos Animales de Enfermedad , Mucosa Intestinal/inervación , Intestino Delgado/inervación , Neuritis/fisiopatología , Neuronas Aferentes/metabolismo , Fragmentos de Péptidos/metabolismo , Animales , Anorexia/metabolismo , Anorexia/prevención & control , Conducta Animal/efectos de los fármacos , Capsaicina/administración & dosificación , Capsaicina/toxicidad , Colecistoquinina/administración & dosificación , Ingestión de Energía/efectos de los fármacos , Conducta Alimentaria/efectos de los fármacos , Péptido 1 Similar al Glucagón/administración & dosificación , Péptido 1 Similar al Glucagón/metabolismo , Infusiones Intravenosas , Inyecciones Intraperitoneales , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/fisiopatología , Intestino Delgado/efectos de los fármacos , Intestino Delgado/metabolismo , Intestino Delgado/fisiopatología , Masculino , Neuritis/inducido químicamente , Neuritis/metabolismo , Neuronas Aferentes/efectos de los fármacos , Fragmentos de Péptidos/administración & dosificación , Péptido YY/administración & dosificación , Péptido YY/metabolismo , Ratas , Nervio Vago/efectos de los fármacos , Nervio Vago/metabolismo , Nervio Vago/fisiopatología , Enfermedades del Nervio Vago/inducido químicamente , Enfermedades del Nervio Vago/metabolismo , Enfermedades del Nervio Vago/fisiopatología
16.
Gen Comp Endocrinol ; 196: 34-40, 2014 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-24287340

RESUMEN

The effects of intraperitoneal injections of cholecystokinin (CCK), apelin, ghrelin, and orexin on food intake were examined in the blind cavefish Astyanax fasciatus mexicanus. CCK (50ng/g) induced a decrease in food intake whereas apelin (100ng/g), orexin (100ng/g), and ghrelin (100ng/g) induced an increase in food intake as compared to saline-injected control fish. In order to better understand the central mechanism by which these hormones act, we examined the effects of injections on the brain mRNA expression of two metabolic enzymes, tyrosine hydroxylase (TH), and mechanistic target of rapamycin (mTOR), and of appetite-regulating peptides, CCK, orexin, apelin and cocaine and amphetamine regulated transcript (CART). CCK injections induced a decrease in brain apelin injections, apelin injections induced an increase in TH, mTOR, and orexin brain expressions, orexin treatment increased brain TH expression and ghrelin injections induced an increase in mTOR and orexin brain expressions. CART expression was not affected by any of the injection treatments. Our results suggest that the enzymes TH and mTOR and the hormones CCK, apelin, orexin, and ghrelin all regulate food intake in cavefish through a complex network of interactions.


Asunto(s)
Apetito/fisiología , Encéfalo/efectos de los fármacos , Colecistoquinina/administración & dosificación , Ingestión de Alimentos/efectos de los fármacos , Ghrelina/administración & dosificación , Hormonas/farmacología , Péptidos y Proteínas de Señalización Intracelular/administración & dosificación , Neuropéptidos/administración & dosificación , Sirolimus/farmacología , Tirosina 3-Monooxigenasa/metabolismo , Animales , Encéfalo/enzimología , Characidae , Quimiocinas/administración & dosificación , Colagogos y Coleréticos/administración & dosificación , Inmunosupresores/farmacología , Neurotransmisores/administración & dosificación , Orexinas , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Tirosina 3-Monooxigenasa/antagonistas & inhibidores , Tirosina 3-Monooxigenasa/genética
17.
Poult Sci ; 103(7): 103819, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38772088

RESUMEN

Adrenomedullin has various physiological roles including appetite regulation. The objective of present study was to determine the effects of ICV injection of adrenomedullin and its interaction with NPY and CCK receptors on food intake regulation. In experiment 1, chickens received ICV injection of saline and adrenomedullin (1, 2, and 3 nmol). In experiment 2, birds injected with saline, B5063 (NPY1 receptor antagonist, 1.25 µg), adrenomedullin (3 nmol) and co-injection of B5063+adrenomedullin. Experiments 3 to 5 were similar to experiment 2 and only SF22 (NPY2 receptor antagonist, 1.25 µg), SML0891 (NPY5 receptor antagonist, 1.25 µg) and CCK4 (1 nmol) were injected instead of B5063. In experiment 6, ICV injection of saline and CCK8s (0.125, 0.25, and 0.5 nmol) were done. In experiment 7, chickens injected with saline, CCK8s (0.125 nmol), adrenomedullin (3 nmol) and co-injection of CCK8s+adrenomedullin. After ICV injection, birds were returned to their individual cages immediately and cumulative food intake was measured at 30, 60, and 120 min after injection. Adrenomedullin (2 and 3 nmol) decreased food intake compared to control group (P < 0.05). Coinjection of B5063+adrenomedullin amplified hypophagic effect of adrenomedullin (P < 0.05). The ICV injection of the CCK8s (0.25 and 0.5 nmol) reduced food intake (P < 0.05). Co-injection of the CCK8s+adrenomedullin significantly potentiated adrenomedullin-induced hypophagia (P < 0.05). Administration of the SF22, SML0891 and CCK4 had no effect on the anorexigenic response evoked by adrenomedullin (P > 0.05). These results suggested that the hypophagic effect of the adrenomedullin is mediated by NPY1 and CCK8s receptors. However, our novel results should form the basis for future experiments.


Asunto(s)
Adrenomedulina , Pollos , Animales , Adrenomedulina/administración & dosificación , Adrenomedulina/farmacología , Pollos/fisiología , Inyecciones Intraventriculares/veterinaria , Neuropéptido Y/administración & dosificación , Neuropéptido Y/farmacología , Neuropéptido Y/metabolismo , Ingestión de Alimentos/efectos de los fármacos , Femenino , Proteínas Aviares/metabolismo , Regulación del Apetito/efectos de los fármacos , Regulación del Apetito/fisiología , Masculino , Receptores de Colecistoquinina , Colecistoquinina/administración & dosificación , Colecistoquinina/farmacología
18.
Horm Metab Res ; 45(6): 471-3, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23315994

RESUMEN

Cholecystokinin (CCK) is a gastrointestinal hormone with potential therapeutic promise for obesity-diabetes. The present study examined the effects of twice daily administration of the N-terminally modified stable CCK-8 analogue, (pGlu-Gln)-CCK-8, on metabolic control and hypothalamic gene expression in high fat fed mice. Sub-chronic twice daily injection of (pGlu-Gln)-CCK-8 for 16 days significantly decreased body weight (p<0.05), energy intake (p<0.01), circulating blood glucose (p<0.001), and plasma insulin (p<0.001) compared to high fat controls. Furthermore, (pGlu-Gln)-CCK-8 markedly improved glucose tolerance (p<0.05) and insulin sensitivity (p<0.05). Assessment of hypothalamic gene expression on day 16 revealed significantly elevated NPY (p<0.05) and reduced POMC (p<0.05) and MC4R (p<0.05) mRNA expression in (pGlu-Gln)-CCK-8 treated mice. High fat feeding or (pGlu-Gln)-CCK-8 treatment had no significant effects on hypothalamic gene expression of receptors for leptin, CCK1 and GLP-1. These studies underscore the potential of (pGlu-Gln)-CCK-8 for the treatment of obesity-diabetes and suggest modulation of NPY and melanocortin related pathways may be involved in the observed beneficial effects.


Asunto(s)
Colecistoquinina/administración & dosificación , Ingestión de Energía/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Hipotálamo/metabolismo , Obesidad/tratamiento farmacológico , Obesidad/genética , Fragmentos de Péptidos/administración & dosificación , Péptidos/administración & dosificación , Animales , Glucemia/análisis , Dieta Alta en Grasa/efectos adversos , Péptido 1 Similar al Glucagón/genética , Péptido 1 Similar al Glucagón/metabolismo , Humanos , Hipotálamo/efectos de los fármacos , Insulina/genética , Insulina/metabolismo , Leptina/genética , Leptina/metabolismo , Masculino , Ratones , Obesidad/metabolismo , Proopiomelanocortina/genética , Proopiomelanocortina/metabolismo , Receptor de Melanocortina Tipo 4/genética , Receptor de Melanocortina Tipo 4/metabolismo
19.
Hippocampus ; 22(6): 1438-50, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22072552

RESUMEN

Although cholecystokinin (CCK) has long been known to exert anxiogenic effects in both animal anxiety models and humans, the underlying cellular and molecular mechanisms are ill-defined. CCK interacts with CCK-1 and CCK-2 receptors resulting in up-regulation of phospholipase C (PLC) and protein kinase C (PKC). However, the roles of PLC and PKC in CCK-mediated anxiogenic effects have not been determined. We have shown previously that CCK facilitates glutamate release in the hippocampus especially at the synapses formed by the perforant path and dentate gyrus granule cells via activations of PLC and PKC. Here we further demonstrated that CCK enhanced NMDA receptor function in dentate gyrus granule cells via activation of PLC and PKC pathway. At the single-channel level, CCK increased NMDA single-channel open probability and mean open time, reduced the mean close time, and had no effects on the conductance of NMDA channels. Because elevation of glutamatergic functions results in anxiety, we explored the roles of PLC and PKC in CCK-induced anxiogenic actions using the Vogel Conflict Test (VCT). Our results from both pharmacological approach and knockout mice demonstrated that microinjection of CCK into the dentate gyrus concentration-dependently increased anxiety-like behavior via activation of PLC and PKC. Our results provide a novel unidentified signaling mechanism whereby CCK increases anxiety.


Asunto(s)
Ansiedad/inducido químicamente , Ansiedad/enzimología , Colecistoquinina/administración & dosificación , Proteína Quinasa C/fisiología , Receptores de N-Metil-D-Aspartato/fisiología , Fosfolipasas de Tipo C/fisiología , Animales , Colecistoquinina/fisiología , Relación Dosis-Respuesta a Droga , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Femenino , Activación del Canal Iónico/fisiología , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Microinyecciones , Ratas , Ratas Sprague-Dawley
20.
Am J Physiol Regul Integr Comp Physiol ; 303(8): R850-60, 2012 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-22874423

RESUMEN

Various molecular forms of CCK reduce food intake in rats. Although CCK-8 is the most studied form, we reported that CCK-58 is the only detectable endocrine peptide form in rats. We investigated the dark-phase rat chow intake pattern following injection of CCK-8 and CCK-58. Ad libitum-fed male Sprague-Dawley rats were intraperitoneally injected with CCK-8, CCK-58 (0.6, 1.8, and 5.2 nmol/kg), or vehicle. Food intake pattern was assessed during the dark phase using an automated weighing system that allowed continuous undisturbed monitoring of physiological eating behavior. Both CCK-8 and CCK-58 dose dependently reduced 1-h, dark-phase food intake, with an equimolar dose of 1.8 nmol being similarly effective (-49% and -44%). CCK-58 increased the latency to the first meal, whereas CCK-8 did not. The intermeal interval was reduced after CCK-8 (1.8 nmol/kg, -41%) but not after CCK-58. At this dose, CCK-8 increased the satiety ratio by 80% and CCK-58 by 160%, respectively, compared with vehicle. When behavior was assessed manually, CCK-8 reduced locomotor activity (-31%), whereas grooming behavior was increased (+59%). CCK-58 affected neither grooming nor locomotor activity. In conclusion, reduction of food intake by CCK-8 and CCK-58 is achieved by differential modulation of food intake microstructure and behavior. These data highlight the importance of studying the molecular forms of peptides that exist in vivo in tissue and circulation of the animal being studied.


Asunto(s)
Colecistoquinina/administración & dosificación , Ritmo Circadiano/efectos de los fármacos , Ingestión de Alimentos/efectos de los fármacos , Conducta Alimentaria/efectos de los fármacos , Sincalida/administración & dosificación , Animales , Relación Dosis-Respuesta a Droga , Aseo Animal/efectos de los fármacos , Inyecciones Intraperitoneales , Masculino , Actividad Motora/efectos de los fármacos , Fotoperiodo , Ratas , Ratas Sprague-Dawley , Tiempo de Reacción/efectos de los fármacos , Respuesta de Saciedad/efectos de los fármacos , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda