Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Am J Physiol Gastrointest Liver Physiol ; 319(1): G23-G35, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32421358

RESUMEN

Glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) are released from enteroendocrine cells (EECs) in response to nutrient ingestion and lower blood glucose levels by stimulation of insulin secretion and thus are defined as incretins. GLP-1 receptor (GLP-1R) expression has been identified on enteric neurons that include intrinsic afferent neurons, extrinsic spinal, and vagal sensory afferents but has not been shown to have an incretin effect through these nerves. GLP-1 and GIP enter the mesenteric lymphatic fluid (MLF) after a meal via the interstitial fluid (IF) from local tissue secretion and/or blood capillaries. We tested if MLF could induce diet-dependent intransient increases in intracellular calcium ([Ca2+]i) in cultured sensory neurons. Postprandial rat MLF, collected from the superior mesenteric lymphatic duct, induced a significant twofold higher intransient increase in [Ca2+]i in primary-cultured sensory neurons than MLF from fasted rats. Inhibition of transient receptor potential vanilloid 1 (TRPV1) and TRPV1 and ankyrin 1 cation channels (TRPA1) with ruthenium red eliminated the difference. Substance P (SP) (a peptide that stimulates insulin secretion) sensor cells cocultured with sensory neurons showed both the GLP-1R agonist exendin-4 (Ex-4) and GIP induced transient increases in [Ca2+]i directly coupled to SP secretion in the sensory nerves. Ex-4-induced release of SP required expression of either TRPA1 or TRPV1. These data identify unrecognized actions of GLP-1 and GIP as incretins by acting as neurolymphocrines and suggest a mechanism for sensory nerves to respond to the postprandial state through MLF.NEW & NOTEWORTHY Glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) are secreted upon eating to lower blood sugar. GLP-1 and GIP were found to induce the secretion of substance P (SP) from cultured sensory nerves. SP enhances insulin secretion. Mesenteric lymphatic fluid (MLF) also stimulates sensory neurons in a diet-dependent manner. These studies identify new actions of GLP-1 and GIP as incretins and suggest a mechanism for sensory nerves to respond to diet through MLF.


Asunto(s)
Polipéptido Inhibidor Gástrico/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , Glucosa/metabolismo , Sustancia P/metabolismo , Canal Catiónico TRPA1/metabolismo , Animales , Glucemia/metabolismo , Células Enteroendocrinas/metabolismo , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Incretinas/metabolismo , Periodo Posprandial , Ratas , Receptores de la Hormona Gastrointestinal
2.
Am J Physiol Gastrointest Liver Physiol ; 306(8): G686-98, 2014 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-24578341

RESUMEN

Lymphatic fluid is a plasma filtrate that can be viewed as having biological activity through the passive accumulation of molecules from the interstitial fluid. The possibility that lymphatic fluid is part of an active self-contained signaling process that parallels the endocrine system, through the activation of G-protein coupled receptors (GPCR), has remained unexplored. We show that the GPCR lysophosphatidic acid 5 (LPA5) is found in sensory nerve fibers expressing calcitonin gene-related peptide (CGRP) that innervate the lumen of lymphatic lacteals and enteric nerves. Using LPA5 as a model for nutrient-responsive GPCRs present on sensory nerves, we demonstrate that dietary protein hydrolysate (peptone) can induce c-Fos expression in enterocytes and nerves that express LPA5. Mesenteric lymphatic fluid (MLF) mobilizes intracellular calcium in cell models expressing LPA5 upon feeding in a time- and dose-dependent manner. Primary cultured neurons of the dorsal root ganglia expressing CGRP are activated by MLF, which is enhanced upon LPA5 overexpression. Activation is independent of the known LPA5 agonists, lysophosphatidic acid and farnesyl pyrophosphate. These data bring forth a pathway for the direct stimulation of sensory nerves by luminal contents and interstitial fluid. Thus, by activating LPA5 on sensory nerves, MLF provides a means for known and yet to be identified constituents of the interstitial fluid to act as signals to comprise a "neurolymphocrine" system.


Asunto(s)
Enterocitos/fisiología , Linfa/fisiología , Receptores del Ácido Lisofosfatídico/metabolismo , Células Receptoras Sensoriales/fisiología , Animales , Péptido Relacionado con Gen de Calcitonina/metabolismo , Fenómenos Fisiológicos Celulares , Proteínas en la Dieta/metabolismo , Ratones , Ratones Endogámicos C57BL , Peptonas/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores Acoplados a Proteínas G/metabolismo
3.
Am J Physiol Gastrointest Liver Physiol ; 297(4): G641-54, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19679818

RESUMEN

P2Y5 is a G protein-coupled receptor that binds and is activated by lysophosphatidic acid (LPA). We determined that P2Y5 transcript is expressed along the intestinal mucosa and investigated the intracellular pathways induced by P2Y5 activation, which could contribute to LPA effects on intestinal cell adhesion. P2Y5 heterologously expressed in CHO and small intestinal hBRIE 380i cells was activated by LPA resulting in an increase in intracellular calcium ([Ca(2+)](i)) when the cells concurrently expressed G(alpha)(Delta6qi5myr). P2Y5 activation also increased the phosphorylation of ERK1/2 that was sensitive to pertussis toxin. Together these indicate that P2Y5 activation by LPA induces an increase in [Ca(2+)](i) and ERK1/2 phosphorylation through G(alpha)(i). We discovered that P2Y5 was activated by farnesyl pyrophosphate (FPP) without a detectable change in [Ca(2+)](i). The activation of P2Y5 by LPA or FPP induced the activity of a serum response element (SRE)-linked luciferase reporter that was inhibited by the RGS domain of p115RhoGEF, C3 exotoxin, and Y-27632, suggesting the involvement of G(alpha)(12/13), Rho GTPase, and ROCK, respectively. However, only LPA-mediated induction of SRE reporter activity was sensitive to inhibitors targeting p38 MAPK, PI3K, PLC, and PKC. In addition, only LPA transactivated the epidermal growth factor receptor, leading to an induction of ERK1/2 phosphorylation. These observations correlate with our subsequent finding that P2Y5 activation by LPA, and not FPP, reduced intestinal cell adhesion. This study elucidates a mechanism whereby LPA can act as a luminal and/or serosal cue to alter mucosal integrity.


Asunto(s)
Adhesión Celular , Células Epiteliales/metabolismo , Subunidades alfa de la Proteína de Unión al GTP G12-G13/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Mucosa Intestinal/metabolismo , Lisofosfolípidos/metabolismo , Receptores Purinérgicos P2/metabolismo , Animales , Células CHO , Señalización del Calcio , Adhesión Celular/efectos de los fármacos , Cricetinae , Cricetulus , Regulación hacia Abajo , Inhibidores Enzimáticos/farmacología , Células Epiteliales/efectos de los fármacos , Receptores ErbB/metabolismo , Quinasa 2 del Receptor Acoplado a Proteína-G/genética , Quinasa 2 del Receptor Acoplado a Proteína-G/metabolismo , Subunidades alfa de la Proteína de Unión al GTP G12-G13/antagonistas & inhibidores , Subunidades alfa de la Proteína de Unión al GTP G12-G13/genética , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/antagonistas & inhibidores , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/genética , Factores de Intercambio de Guanina Nucleótido/genética , Factores de Intercambio de Guanina Nucleótido/metabolismo , Células HeLa , Humanos , Mucosa Intestinal/efectos de los fármacos , MAP Quinasa Quinasa 1/metabolismo , MAP Quinasa Quinasa 2/metabolismo , Masculino , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fosforilación , Fosfatos de Poliisoprenilo/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Receptor Cross-Talk , Receptores Purinérgicos P2/genética , Proteínas Recombinantes de Fusión/metabolismo , Factores de Intercambio de Guanina Nucleótido Rho , Elemento de Respuesta al Suero , Sesquiterpenos/metabolismo , Transfección , Proteínas de Unión al GTP rho/metabolismo , Quinasas Asociadas a rho/metabolismo
4.
Am J Physiol Gastrointest Liver Physiol ; 292(5): G1366-75, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17290006

RESUMEN

In the intestinal lumen, protein hydrolysate increases the transcription and release of cholecystokinin (CCK) from enteroendocrine cells of the duodenal-jejunal mucosa. Our recent discovery that a G protein-coupled receptor, GPR93, is activated by dietary protein hydrolysate causing induced intracellular calcium-mediated signaling events in intestinal epithelial cells raises a possibility that GPR93 might be involved in the protein hydrolysate induction of CCK expression and/or secretion. Using the enteroendocrine STC-1 cells as a model, the present study demonstrates that increasing expression of GPR93 amplifies the peptone induction of endogenous CCK mRNA levels. A similar increase in CCK transcription, indicated by the luciferase reporter activity driven by an 820-bp CCK promoter, is also observed in response to peptone at a dose as little as 6.25 mg/ml, but not to lysophosphatidic acid (LPA), an agonist of GPR93. We discovered that the upregulation of CCK transcription involves ERK1/2, PKA, and calmodulin-dependent protein kinase-mediated pathways. Additionally, GPR93 activation by peptone induces a response in CCK release at 15 min, which continues over a 2-h period. The cAMP level in STC-1 cells overexpressing GPR93 is induced at a greater extent by peptone than by LPA, suggesting a possible explanation of the different effects of peptone and LPA on CCK transcription and secretion. Our data indicate that GPR93 can contribute to the observed induction of CCK expression and secretion by peptone and provide evidence that G protein-coupled receptors can transduce dietary luminal signals.


Asunto(s)
Colecistoquinina/metabolismo , Células Enteroendocrinas/metabolismo , Peptonas/farmacología , Receptores del Ácido Lisofosfatídico/fisiología , Animales , Línea Celular Tumoral , Colecistoquinina/biosíntesis , AMP Cíclico/metabolismo , Células Enteroendocrinas/efectos de los fármacos , Lisofosfolípidos/farmacología , Ratones , ARN Mensajero/metabolismo , Transcripción Genética/efectos de los fármacos
5.
Am J Physiol Gastrointest Liver Physiol ; 292(1): G98-G112, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16935853

RESUMEN

G protein-coupled receptors (GPCRs) have the potential to play a role as molecular sensors responsive to luminal dietary contents. Although such a role for GPCRs has been implicated in the intestinal response to protein hydrolysate, no GPCR directly involved in this process has been previously identified. In the present study, for the first time, we identified GPR93 expression in enterocytes and demonstrated its activation in these cells by protein hydrolysate with EC50 of 10.6 mg/ml as determined by the induction of intracellular free Ca2+. In enterocytes, GPR93 was synergistically activated by protein hydrolysate in combination with an agonist, oleoyl-l-alpha-lysophosphatidic acid (LPA), which activated the receptor in these enterocytes with EC50 of 7.9 nM. The increased intracellular Ca2+ by GPR93 activation was observed without the addition of a promiscuous Galpha protein and was pertussis toxin sensitive, which suggests Galpha(q)- and Galpha(i)-mediated pathways. Activated GPR93 also induced pertussis toxin-sensitive ERK1/2 phosphorylation. Both nuclear factor of activated T cells and 12-O-tetradecanoylphorbol 13-acetate responsive elements reporter activities were induced by protein hydrolysate in cells exogenously expressing GPR93. The peptidomimetic cefaclor by itself did not activate GPR93 but potentiated the protein hydrolysate response and further amplified the synergistic enhancement of GPR93 activation by protein hydrolysate and LPA. These data suggest that, physiologically, the composition of stimuli might determine GPR93 activity or its sensitivity toward a given activator and suggest a new mechanism of the regulation of mucosal cell proliferation and differentiation and hormonal secretion by dietary products in the lumen.


Asunto(s)
Hidrolisados de Proteína/farmacología , Receptores Acoplados a Proteínas G/fisiología , Animales , Células CHO , Línea Celular , Cricetinae , Cricetulus , Humanos , Sistemas de Lectura Abierta , Plásmidos , Receptores Acoplados a Proteínas G/efectos de los fármacos , Receptores Acoplados a Proteínas G/genética , Receptores de Neuropéptido Y/genética , Receptores de Neuropéptido Y/fisiología , Transfección
6.
J Biol Chem ; 280(1): 125-36, 2005 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-15504731

RESUMEN

Peptide YY (PYY) and neuropeptide Y (NPY) are regulatory peptides synthesized in the intestine and brain, respectively, that modify physiological functions affecting nutrient assimilation and feeding behavior. Because PYY and NPY also alter the expression of intestine-specific differentiation marker proteins and the tetraspanin CD63, which is involved in cell adhesion, we investigated whether intestinal cell differentiation could be linked to mucosal cell adhesion and migration through these peptides. PYY and NPY significantly decreased cell adhesion and increased cell migration in a dose-dependent manner prior to cell confluency in our model system, non-tumorigenic small intestinal hBRIE 380i cells. Both peptides reduced CD63 expression and CD63-dependent cell adhesion. CD63 overexpression increased and antisense CD63 cDNA decreased intestinal cell adhesion. In parallel, both PYY and NPY increased expression of matrix metalloproteinase-3 (MMP-3) to a level sufficient to induce cell migration by activating the Rho GTPase Cdc42. The effects of both peptides on cell migration were blocked in cells constitutively overexpressing dominant-negative Cdc42. PYY and NPY also significantly induced the expression of the differentiation marker villin, which could be eliminated by an MMP inhibitor at a concentration that inhibits cell migration. Increased MMP-3 activity, which enhanced cell migration, also induced villin mRNA levels. Therefore, these data indicate that the alteration of adhesion and migration by PYY and NPY occurs in part by synchronous modulation of three proteins that are involved in extracellular matrix-basolateral membrane interactions, CD63, MMP-3 and Cdc42, and that PYY/NPY regulation of expression of mucosal proteins such as villin is linked to the process of cell migration and adhesion.


Asunto(s)
Movimiento Celular/fisiología , Intestino Delgado/fisiología , Proteínas de Microfilamentos/biosíntesis , Proteínas de Microfilamentos/fisiología , Neuropéptido Y/fisiología , Péptido YY/fisiología , Animales , Antígenos CD/fisiología , Adhesión Celular/efectos de los fármacos , Adhesión Celular/fisiología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Línea Celular , Movimiento Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Regulación de la Expresión Génica/fisiología , Intestino Delgado/citología , Metaloproteinasa 3 de la Matriz/fisiología , Proteínas de la Membrana/fisiología , Neuropéptido Y/farmacología , Péptido YY/farmacología , Glicoproteínas de Membrana Plaquetaria , Ratas , Transducción de Señal , Tetraspanina 30 , Proteína de Unión al GTP cdc42/fisiología
7.
Peptides ; 23(2): 367-76, 2002 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11825651

RESUMEN

An essential process for fatty acid digestion, absorption and assimilation is the constant replacement of mature intestinal epithelial cells by differentiating stem cells. Free fatty acids (FFA) and PYY may act in concert to alter mucosal cell differentiation through the cytoskeletal-extracellular matrix interactions. PYY induced expression of tetraspanins and intestinal fatty acid binding protein (I-FABP) may be part of a mechanism whereby FFA modulate expression of differentiation dependent proteins in the mucosa. This modulation provides a means for FFA to act as signal molecules in the feedback regulation of their own assimilation.


Asunto(s)
Ácidos Grasos/metabolismo , Mucosa Intestinal/metabolismo , Proteínas de Neoplasias , Péptido YY/metabolismo , Proteínas Supresoras de Tumor , Animales , Proteínas Portadoras/metabolismo , Adhesión Celular , Diferenciación Celular , Movimiento Celular , Citoesqueleto/metabolismo , Células Epiteliales/metabolismo , Proteína de Unión a los Ácidos Grasos 7 , Proteínas de Unión a Ácidos Grasos , Humanos , Intestinos/citología , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...