Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros










Base de datos
Tipo de estudio
Intervalo de año de publicación
1.
Gene Ther ; 22(9): 750-7, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25965396

RESUMEN

Ghrelin is a stomach-derived peptide hormone that stimulates appetite and promotes adiposity through binding to the growth hormone secretagogue receptor (GHS-R1a). Administration of ghrelin in rodents increases weight gain due to stimulating food intake and reducing fat utilization. Therefore, reducing circulating ghrelin levels holds the potential to reduce weight gain. We developed a GHS-R1a-fusion constructs of a decoy protein containing the ligand-binding domains of the ghrelin receptor. Intramuscular injection of the GHSR/Fc plasmid decreased circulating levels of acylated-ghrelin. When challenged with the high fat diet, treated mice displayed reduced weight gain compared with controls, which was associated with reduced fat accumulation in the peritoneum but not lean mass. Quantitative PCR with reverse transcription showed increased PPARγ and hormone sensitive lipase transcripts levels in adipose tissue of treated animals, illustrating a preference for increased fat utilization. Intra-peritoneal glucose tolerance and insulin tolerance tests showed improved glucose clearance and insulin sensitivity in GHSR/Fc treated animals. We suggest that in vivo expression of the GHSR-based fusion protein prevents diet-induced weight gain, altering adipose gene expression and improving glucose tolerance. These findings, while confirming the role of ghrelin in peripheral energy metabolism, suggest that a strategy involving neutralization of the circulation ghrelin by intramuscular injection of the GHSR1/Fc fusion construct may find clinical application in the treatment of obesity.


Asunto(s)
Ghrelina/metabolismo , Obesidad/metabolismo , Receptores de Ghrelina/metabolismo , Tejido Adiposo/metabolismo , Animales , Dieta Alta en Grasa , Vectores Genéticos , Ghrelina/antagonistas & inhibidores , Masculino , Ratones Endogámicos C57BL , Receptores de Ghrelina/genética
2.
Diabetes Obes Metab ; 15(3): 276-9, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23066988

RESUMEN

The antidiabetic drug Metformin causes weight loss in both diabetic and non-diabetic individuals. Metformin treatment is also associated with lower circulating levels of the orexigenic hormone ghrelin. To test whether Metformin directly affects ghrelin cells, rat primary stomach cells were treated with Metformin and the levels of ghrelin secretion, proghrelin gene expression and activation of adenosine monophosphate-activated protein kinase (AMPK) were examined. Metformin significantly reduced ghrelin secretion and proghrelin mRNA production and both these effects were blocked by co-incubation with the AMPK inhibitor compound C. Furthermore, the AMPK activator 5-amino-1-ß-D-ribofuranosyl-imidazole-4-carboxamide (AICAR) significantly inhibited ghrelin secretion. Additionally, ghrelin cells were shown to express AMPK. Finally, Metformin treatment caused a significant increase in the level of phosphorylated (active) AMPK. Our results show that Metformin directly inhibits stomach ghrelin production and secretion through AMPK. This reduction in ghrelin secretion may be one of the key components in Metformin's mechanism of weight loss.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Células Principales Gástricas/efectos de los fármacos , Células Principales Gástricas/metabolismo , Ghrelina/metabolismo , Metformina/farmacología , Proteínas Quinasas Activadas por AMP/efectos de los fármacos , Animales , Animales Recién Nacidos , Femenino , Mucosa Gástrica/metabolismo , Regulación de la Expresión Génica , Ghrelina/efectos de los fármacos , Embarazo , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Estómago/efectos de los fármacos , Pérdida de Peso/efectos de los fármacos
3.
Int J Obes (Lond) ; 34(11): 1599-607, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20498660

RESUMEN

BACKGROUND: Proprotein convertase subtilisin/xexin type 2 (PCSK2) is an endoproteinase responsible for proteolytic activation of a number of precursors to active neuropeptides and peptide hormones, known to influence glucose homeostasis, food intake and ultimately body mass. In this study, we examined the consequences of PCSK2 deficiency on these phenotypic traits. STUDY DESIGN: Weight gain with age under diets of different fat contents was monitored. White adipose tissue (WAT) and muscle masses were evaluated. Plasma levels of triglycerides, leptin, ghrelin, insulin and proglucagon-derived peptides were measured as well as leptin and acetyl coenzyme-α carboxylase (ACCα) mRNA levels in adipose tissue. RESULTS: Compared with their Pcsk2 (+/+) littermates, Pcsk2 (-/-) mice weighed significantly less as weanlings and as adults. As adults, they carried noticeably less fat mass, with similar lean muscle mass: their plasma leptin level and adipose tissue leptin mRNA level were accordingly lower. PCSK2 deficiency did not affect food intake or the level of the orexigenic hormone ghrelin. However, PCSK2 deficiency resulted in decreased plasma triglycerides and reduced ACCα mRNA levels in WAT. Interestingly, unlike their Pcsk2 (+/+) littermates, Pcsk2 (-/-) were resistant to enhanced body weight gain when fed a high-fat diet. Consistent with a role of PCSK2 in body mass gain, diet-induced or genetically obese mice were found to contain significantly higher levels of PCSK2 mRNA in their brain and stomach than their lean counterparts. CONCLUSION: Collectively, these results suggest that PCSK2 contributes to increase in body mass through the various regulatory peptides generated through its action. It represents a potential target in the prevention and treatment of obesity.


Asunto(s)
Adiposidad/fisiología , Peso Corporal/fisiología , Proproteína Convertasa 2/deficiencia , Adiposidad/genética , Animales , Peso Corporal/genética , Grasas de la Dieta/administración & dosificación , Ingestión de Alimentos , Masculino , Ratones , Ratones Endogámicos C57BL , Proproteína Convertasa 2/genética , ARN Mensajero/metabolismo , Aumento de Peso/genética , Aumento de Peso/fisiología
4.
Pancreas ; 20(4): 348-60, 2000 May.
Artículo en Inglés | MEDLINE | ID: mdl-10824688

RESUMEN

Glicentin (GLIC), oxyntomodulin (OXM), and peptide YY (PYY) released in blood by ileocolonic L-cells after meals may inhibit pancreatic secretion. Whereas OXM interacts with glucagon and tGLP-1 receptors, OXM 19-37, a biologically active fragment, does not. The purpose of this study was to measure the effect of OXM, OXM 19-37, GLIC, tGLP-1, and PYY on pancreatic secretion stimulated by 2 deoxyglucose (2DG), electrical stimulation of the vagus nerves (VES), acetylcholine and cholecystokinin octapeptide (CCK8) in anesthetized rats. The effect of OXM was also studied in dispersed pancreatic acini. Plasma oxyntomodulin-like immunoreactivity (OLI) was measured by radioimmunoassay after the exogenous infusion of OXM and after an intraduodenal meal. OXM 19-37, infused at doses mimicking postprandial plasma levels of OLI, decreased pancreatic secretion stimulated by 2DG, VES, or CCK8. Similar effects were found with OXM and GLIC. OXM 19-37 did not change the pancreatic stimulation induced by acetylcholine in vivo, or CCK-induced amylase release in isolated acini. Vagotomy completely suppressed the inhibitory effect of OXM 19-37 on CCK8-stimulated pancreatic secretion. PYY inhibited the effect of 2DG, but not that of CCK8, whereas tGLP-1, even in pharmacologic doses, had no effect on stimulated pancreatic secretion. OXM, OXM 19-37, but not tGLP-1, inhibit pancreatic secretion at physiologic doses, through a vagal neural indirect mechanism, different from that used by PYY, and probably through a GLIC-related peptide-specific receptor.


Asunto(s)
Péptidos Similares al Glucagón/farmacología , Páncreas/inervación , Páncreas/metabolismo , Animales , Desoxiglucosa/farmacología , Interacciones Farmacológicas , Glicentina , Glucagón/farmacología , Péptido 1 Similar al Glucagón , Péptidos Similares al Glucagón/sangre , Cinética , Masculino , Oxintomodulina , Páncreas/efectos de los fármacos , Fragmentos de Péptidos/sangre , Fragmentos de Péptidos/farmacología , Péptido YY/sangre , Péptido YY/farmacología , Precursores de Proteínas/farmacología , Ratas , Ratas Wistar , Sincalida/farmacología , Nervio Vago/efectos de los fármacos , Nervio Vago/fisiología
5.
Am J Physiol Gastrointest Liver Physiol ; 278(4): G578-84, 2000 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-10762612

RESUMEN

Colonic fermentation of carbohydrate has been shown to influence gastric and intestinal motility. Our aim was to investigate the effects of colonic infusion of lactose and short-chain fatty acids (SCFAs) on lower esophageal sphincter (LES) function in humans. LES pressure (LESP), transient relaxations of LES (TLESRs), and esophageal pH were monitored over 6 h on 4 different days in 7 healthy volunteers. After 1 h of baseline recording, the effects of different colonic infusions (270 ml of isotonic or hypertonic saline, 30 g lactose, or 135 mmol SCFAs) were tested in fasting conditions and after a standard meal. Peptide YY (PYY) and oxyntomodulin (OLI) were also measured in plasma. Both lactose and SCFA infusions increased the number of TLESRs as well as the proportion of TLESRs associated with acid reflux episodes, but saline solutions did not. The postprandial fall of LESP was enhanced by previous SCFA infusion. Plasma PYY and OLI increased similarly after all colonic infusions. Colonic fermentation of lactose markedly affected LES function, and this effect was reproduced by SCFA infusion. Whether the mechanisms of this feedback phenomenon are of hormonal nature, neural nature, or both remains to be determined.


Asunto(s)
Colon/metabolismo , Unión Esofagogástrica/fisiología , Ácidos Grasos Volátiles/metabolismo , Lactosa/metabolismo , Adulto , Ácidos Grasos Volátiles/administración & dosificación , Femenino , Fermentación , Péptidos Similares al Glucagón/sangre , Humanos , Lactosa/administración & dosificación , Masculino , Manometría , Relajación Muscular/fisiología , Oxintomodulina , Péptido YY/sangre , Peristaltismo/fisiología , Presión , Valores de Referencia
6.
Pflugers Arch ; 438(3): 299-306, 1999 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-10398859

RESUMEN

Endocrine L-cells of the distal intestine synthesize both peptide YY (PYY) and proglucagon-derived peptides (PGDPs), whose release has been reported to be either parallel or selective. Here we compare the release mechanisms of PYY, glucagon-like peptide-1 (GLP-1), and oxyntomodulin-like immunoreactivity (OLI) in vivo. Anaesthetized rats were intraduodenally (ID) given either a mixed semi-liquid meal or oleic acid, or they received oleic acid or short chain fatty acids (SCFA) intracolonically (IC). The ID meal released the three peptides with a similar time-course (peak at 30 min); ID oleic acid produced a progressive release of PYY and OLI, while GLP-1 release was less. IC oleic acid or SCFA released smaller (but significant) amounts of PYY but no OLI or GLP-1. Hexamethonium inhibited most of the response to the ID meal and ID oleic acid, but did not change the PYY response to IC oleic acid. NG-nitro-l-arginine methyl ester (l-NAME, a nitric oxide synthase inhibitor) inhibited meal-induced PYY release and left OLI and GLP-1 unaffected. BW10 (a gastrin-releasing peptide antagonist) had no effect on the meal-induced release of either peptide. These results suggest a parallel initial release of PYY, OLI and GLP-1 after the ID meal, or oleic acid, by an indirect mechanism triggered in the proximal bowel, using nicotinic synapses, and involving nitric oxide release for PYY and an unknown mediator for PGDPs. For PYY there is a later phase of peptide release, probably induced by direct contact between nutrients and colonic L-cells.


Asunto(s)
Alimentos , Glucagón/metabolismo , Péptido YY/metabolismo , Precursores de Proteínas/metabolismo , Animales , Ácidos Grasos/administración & dosificación , Péptido 1 Similar al Glucagón , Péptidos Similares al Glucagón/metabolismo , Hexametonio/farmacología , Cinética , Masculino , Ácido Oléico/administración & dosificación , Oxintomodulina , Fragmentos de Péptidos/metabolismo , Proglucagón , Ratas , Ratas Wistar
7.
Int J Cancer ; 80(3): 448-54, 1999 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-9935189

RESUMEN

The intestine is a large endocrine organ, but the dependence of colon cancer on hormones remains unknown. We show here that neurotensin, a paracrine/endocrine peptide in the gut, and the neurotensin receptor antagonist SR 48692 control colon cancer cell growth in vitro and in vivo by interacting with receptors that are ectopically expressed in colon cancers. In cell culture, neurotensin stimulates the growth of human colon cancer cell lines (SW480, SW620, HT29, HCT116 and Cl.19A) expressing the neurotensin receptor NTR1 but does not change the growth of Caco2 cells, which do not express NTR1. In SW480 cells, neurotensin is active in the 10(-10) to 10(-6) M concentration range (ED50 = 0.47 nM) while the neurotensin fragment (I-II) is inactive. Neurotensin also enhances the cellular cloning efficiency of SW480 cells in soft agar by inducing a 50% increase of colony formation. This effect is blocked by SR 48692, which alone does not alter colony formation. Subcutaneous delivery of neurotensin (0.54 micromol/kg every 24 hr) by osmotic pumps to nude mice that have been xenografted with SW480 cells results in a significant increase of tumor volume, i.e., up to 255% of control at day 20 of treatment. SR 48692 administered alone (1.7 micromol/kg every 24 hr) by daily i.p. injections reduces the development of tumors formed by xenografting SW480 cells in nude mice. A significant mean reduction of tumor volume of 38% is observed during the 22-day period of treatment. SR 48692 alone is also active at reducing tumor volume after xenografting HCT116 cells in nude mice. Our results support the notion that colon cancer growth may be dependent on blood-borne neurotensin and suggest that non-peptide neurotensin antagonists, such as SR 48692, may be useful for the development of novel therapeutic strategies of colon cancer.


Asunto(s)
División Celular/efectos de los fármacos , Neoplasias del Colon/patología , Neurotensina/farmacología , Pirazoles/farmacología , Quinolinas/farmacología , Receptores de Neurotensina/antagonistas & inhibidores , Agar , Animales , Neoplasias del Colon/metabolismo , Medios de Cultivo , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neurotensina/metabolismo , Reacción en Cadena de la Polimerasa , Receptores de Neurotensina/metabolismo , Trasplante Heterólogo , Células Tumorales Cultivadas/efectos de los fármacos
8.
Peptides ; 19(9): 1503-9, 1998.
Artículo en Inglés | MEDLINE | ID: mdl-9864056

RESUMEN

The interaction of PYY and VIP was studied in stripped and intact rat jejunum preparations mounted in Ussing chambers. PYY decreased basal Isc in intact as well as in stripped jejunum. Stripping was necessary to evidence a stimulation of basal Isc by VIP. When PYY and VIP were administered at the same time in the serosal bath, their effects seemed additive; VIP stimulation took over when VIP was present in ten times larger amounts than PYY, while PYY inhibition predominated at isomolar concentrations (10(-7) M) of both peptides. However, when PYY was administered three to six minutes before isomolar amounts of VIP, the VIP stimulation developed without being notably hampered. At this time, however, the amount of radioimmunoassayable PYY in the serosal compartment represented still 60% of the added amount. In conclusion, the experimental conditions can significantly change the results: stripping the longitudinal muscle/myenteric plexus impairs the effect of PYY and VIP in a different fashion, while the timing and order of administration of the peptides may change the apparent interaction between VIP stimulation and PYY inhibition.


Asunto(s)
Secreciones Intestinales/efectos de los fármacos , Yeyuno/efectos de los fármacos , Péptido YY/farmacología , Péptido Intestinal Vasoactivo/farmacología , Animales , Transporte Biológico/efectos de los fármacos , Interacciones Farmacológicas , Conductividad Eléctrica , Técnicas In Vitro , Masculino , Músculo Liso , Plexo Mientérico , Ratas , Ratas Wistar
9.
Am J Physiol ; 275(6): G1415-22, 1998 12.
Artículo en Inglés | MEDLINE | ID: mdl-9843779

RESUMEN

Short-chain fatty acids (SCFAs) are recognized as the major anions of the large intestinal content in humans, but their effect on colonic motility is controversial. This study explores the colonic motor effect of SCFAs and their mechanisms in the rat. Colonic motility (electromyography) and transit time (plastic markers) were measured in conscious rats while SCFAs were infused into the colon, either alone or after administration of neural antagonists or immunoneutralization of circulating polypeptide YY (PYY). SCFA-induced PYY release was measured by RIA and then simulated by infusing exogenous PYY. Intracolonic infusion of 0.4 mmol/h SCFAs had no effect, whereas 2 mmol/h SCFAs reduced colonic motility (36 +/- 3 vs. 57 +/- 4 spike bursts/h with saline, P < 0.05) by decreasing the ratio of nonpropulsive to propulsive activity. This resulted in an increased transit rate (P < 0.01). Neither alpha-adrenoceptor blockade nor nitric oxide synthase inhibition prevented SCFA-induced motility reduction. Intraluminal procaine infusion suppressed the SCFA effect, indicating that a local neural mechanism was involved. SCFA colonic infusion stimulated PYY release in blood. Immunoneutralization of circulating PYY abolished the effect of SCFAs on colonic motility, whereas exogenous PYY infusion partly reproduced this effect. SCFAs modify colonic motor patterns in the rat and increase transit rate; local nerve fibers and PYY are involved in this effect.


Asunto(s)
Colon/efectos de los fármacos , Colon/inervación , Ácidos Grasos/farmacología , Motilidad Gastrointestinal/efectos de los fármacos , Péptido YY/metabolismo , Animales , Colon/química , Relación Dosis-Respuesta a Droga , Electromiografía , Ácidos Grasos/análisis , Tránsito Gastrointestinal/efectos de los fármacos , Masculino , Complejo Mioeléctrico Migratorio/efectos de los fármacos , Complejo Mioeléctrico Migratorio/fisiología , Fenómenos Fisiológicos del Sistema Nervioso , Concentración Osmolar , Péptido YY/antagonistas & inhibidores , Péptido YY/farmacología , Ratas , Ratas Wistar
10.
Pflugers Arch ; 433(5): 571-9, 1997 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-9049141

RESUMEN

Peptide YY (PYY) release in anaesthetized rats was studied during the 2 h following the intraduodenal administration of a semi-liquid meal of 21 kJ. Surgical and pharmacological manipulations were performed in order to analyse the mechanisms of PYY release. Postprandial PYY release was suppressed or strongly decreased by caecocolonectomy, truncal vagotomy, tetrodotoxin, hexamethonium, sensory denervation by perivagal capsaicin, and by the NO-synthase inhibitor L-N-arginine methyl ester, while atropine, adrenergic blockers, antagonists of type-A or type-B cholecystokinin (CCK) receptors or bombesin receptors had no effect. Comparing the digestive transit of the semi-liquid meal with the amount of PYY contained in the small bowel wall showed that nutrients had not reached the area rich in cells containing PYY by 30 min, the time at which there was a large PYY release in plasma. By 120 min, the meal front had travelled 72% of the small intestine length, just beginning to reach the PYY-rich part of the ileum. We conclude that the main postprandial PYY release studied in this model comes from ileal and colonic L-cells indirectly stimulated through a neural mechanism originating in the proximal gut and involving sensory vagal fibres, nicotinic synapses and NO release, while CCK and bombesin do not seem to be physiologically involved.


Asunto(s)
Duodeno/inervación , Péptidos/metabolismo , Animales , Bombesina/antagonistas & inhibidores , Bombesina/farmacología , Colecistoquinina/antagonistas & inhibidores , Duodeno/metabolismo , Nutrición Enteral , Alimentos Formulados , Masculino , Óxido Nítrico Sintasa/antagonistas & inhibidores , Péptido YY , Ratas , Ratas Wistar , Sincalida/farmacología , Nervio Vago/fisiología
11.
Peptides ; 18(8): 1249-55, 1997.
Artículo en Inglés | MEDLINE | ID: mdl-9396069

RESUMEN

It is intriguing that the antisecretory peptide YY is present in plasma in two forms: PYY1-36 and PYY3-36. PYY3-36 has been found in human and rabbit blood within 30 min of the beginning of the meal, when the peak of water and electrolyte secretion occurs in the duodeno-jejunum. The aim of this study was therefore to compare the antisecretory effect of PYY1-36 and PYY3-36 in fed and fasted rat jejunum. The variations in electrolyte secretion were assessed by measuring the variations in short-circuit current (delta Isc) and transepithelial isotopic chloride fluxes in jejunal mucosa isolated from fed and fasted animal, and mounted in Ussing Chambers. In fasted animals, 2 x 10(-7) M PYY3-36 induced a reduction in Isc of -0.50 +/- 0.01 microEq/hr.cm2, which was not statistically different from that induced by 2 x 10(-7) M PYY1-36 (-0.60 +/- 0.01 microEq/h cm2). In contrast, in fed animals, 2 x 10(-7) M PYY3-36 did not trigger a significant response on Isc and net chloride flux, while the response to PYY1-36 was present but blunted. The absence of response was probably not related to the presence of secretory peptides because PYY3-36 was still able to induce a reduction in Isc after stimulation by a series of 10 different secretory peptides. After 10(-8) M PYY3-36 addition to an epithelium from the fasted animal, response to 10(-7) M PYY3-36 was blunted for 30 min and returned to control value after 60 min. Plasma concentration of PYY was higher in the fed rats compared to fasted (213.78 +/- 38 vs. 53.62 +/- 11.47 pg/ml p < 0.01). After incubation of crypt cells with or without 0.1 microM of unlabeled PYY for 60 min, Scatchard analysis of equilibrium binding data show that binding capacity (Bmax) of receptors was reduced when crypt cells were previously incubated with unlabeled PYY without significant modification of dissociation constants. Bmax were 183 +/- 27 in control vs. 56 +/- 11 fmol/mg protein. These results confirm the antisecretory activity of PYY1-36 in the jejunum of fasted and fed rats. They further indicate that PYY3-36 displays similar activity to PYY1-36 in fasted animals, but lack of activity in fed animals. These results suggest that the two circulating forms of PYY act as antisecretory peptides by two different mechanisms, implying a C-terminal specificity.


Asunto(s)
Mucosa Intestinal/metabolismo , Yeyuno/metabolismo , Péptido YY/fisiología , Animales , Cloruros/metabolismo , Conductividad Eléctrica , Ayuno , Hormonas Gastrointestinales/metabolismo , Técnicas In Vitro , Masculino , Fragmentos de Péptidos , Péptido YY/sangre , Péptido YY/metabolismo , Unión Proteica , Ratas , Ratas Sprague-Dawley
12.
Pflugers Arch ; 431(1): 66-75, 1995 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-8584419

RESUMEN

Peptide YY (PYY) release was studied by measuring radioimmunoassayable PYY in the arterial plasma of anaesthetized rats receiving into the duodenum, ileum or colon either a complete semi-liquid meal (3ml, 21kJ) or elemental nutrients as isocaloric or isoosmolar solutions. PYY release induced by the intraduodenal meal peaked at 60min and lasted more than 120min. The integrated response of PYY over 120min was larger when the meal was administered into the duodenum than into the ileum. The undigested meal induced no release of PYY over a 120-min period when administered into the colon. When injected into the duodenum in isocaloric amounts to the meal, glucose and amino acids led to the release of as much PYY as did the meal, whereas oleic acid led to the release of less PYY. Part of these responses were due to osmolarity, since administration of intraduodenal hyperosmolar saline led to the release of about half as much PYY as did hyperosmolar glucose. In moderate amounts, and injected as a solution isoosmolar to plasma, oleic acid was a major PYY releaser; the amounts released were at least two times larger when oleic acid was administered into the duodenum than into the ileum and colon. Isoosmolar glucose and amino acids led to the release of no PYY when injected into the duodenum, but were nearly as active as oleic acid in the colon. Short-chain fatty acids induced the release of PYY when injected into the colon, but not into the ileum. Hexamethonium suppressed PYY release induced by the intraduodenal meal, but did not change PYY release induced by glucose or oleic acid in the colon. Urethane anaesthesia did not reduce PYY release induced by the intraduodenal meal. These results suggest that two mechanisms at least contribute to PYY release in the rat. An indirect, neural mechanism, involving nicotinic synapses, is prominent in the proximal small intestine; the stimulation is transmitted to ileal and colonic L-cells by undetermined pathways, but contact of nutrients with L-cells is not needed. Another mechanism, probably direct and quantitatively smaller, occurs in the distal intestine when nutrients come into contact with the mucosa containing L-cells. Glucose, fatty acids and amino acids stimulate differentially the proximal and distal mechanisms.


Asunto(s)
Nutrición Enteral , Hormonas Gastrointestinales/metabolismo , Intestinos/fisiología , Péptidos/metabolismo , Anestesia , Animales , Colon/fisiología , Duodeno/fisiología , Ingestión de Energía , Alimentos Formulados , Hormonas Gastrointestinales/sangre , Tránsito Gastrointestinal/fisiología , Íleon/fisiología , Intubación Gastrointestinal , Masculino , Ácido Oléico , Ácidos Oléicos/farmacología , Concentración Osmolar , Péptido YY , Péptidos/sangre , Radioinmunoensayo , Ratas , Ratas Wistar
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...