Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Am J Physiol Gastrointest Liver Physiol ; 310(9): G696-704, 2016 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-26939869

RESUMEN

Stromal cells influence epithelial function in both health and disease. Myofibroblasts are abundant stromal cells that influence the cellular microenvironment by release of extracellular matrix (ECM) proteins, growth factors, proteases, cytokines, and chemokines. Cancer-associated myofibroblasts (CAMs) differ from adjacent tissue (ATMs) and normal tissue myofibroblasts (NTMs), but the basis of this is incompletely understood. We report now the differential expression of miRNAs in gastric cancer CAMs. MicroRNA arrays identified differences in the miRNA profile in gastric and esophageal NTMs and in CAMs from stomach compared with NTMs. miR-181d was upregulated in gastric CAMs. Analysis of differentially regulated miRNAs indicated an involvement in Wnt signaling. Examination of a microarray data set then identified Wnt5a as the only consistently upregulated Wnt ligand in gastric CAMs. Wnt5a stimulated miR-181d expression, and knockdown of miR-181d inhibited Wnt5a stimulation of CAM proliferation and migration. Analysis of miR-181d targets suggested a role in chemotaxis. Conditioned medium from CAMs stimulated gastric cancer cell (AGS) migration more than that from ATMs, and miR-181d knockdown reduced the effect of CAM-CM on AGS cell migration but had no effect on AGS cell responses to ATM conditioned media. The data suggest that dysregulation of miRNA expression in gastric CAMs, secondary to Wnt5a signaling, accounts at least in part for the effect of CAMs in promoting cancer cell migration.


Asunto(s)
MicroARNs/genética , Miofibroblastos/metabolismo , Neoplasias Gástricas/metabolismo , Vía de Señalización Wnt , Proliferación Celular , Células Cultivadas , Quimiotaxis , Humanos , Miofibroblastos/fisiología , Neoplasias Gástricas/genética , Proteína Wnt-5a/genética , Proteína Wnt-5a/metabolismo
2.
J Pathol ; 236(3): 326-36, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25727407

RESUMEN

NF-κB signalling is an important factor in the development of inflammation-associated cancers. Mouse models of Helicobacter-induced gastric cancer and colitis-associated colorectal cancer have demonstrated that classical NF-κB signalling is an important regulator of these processes. In the stomach, it has also been demonstrated that signalling involving specific NF-κB proteins, including NF-κB1/p50, NF-κB2/p52, and c-Rel, differentially regulate the development of gastric pre-neoplasia. To investigate the effect of NF-κB subunit loss on colitis-associated carcinogenesis, we administered azoxymethane followed by pulsed dextran sodium sulphate to C57BL/6, Nfkb1(-/-), Nfkb2(-/-), and c-Rel(-/-) mice. Animals lacking the c-Rel subunit were more susceptible to colitis-associated cancer than wild-type mice, developing 3.5 times more colonic polyps per animal than wild-type mice. Nfkb2(-/-) mice were resistant to colitis-associated cancer, developing fewer polyps per colon than wild-type mice (median 1 compared to 4). To investigate the mechanisms underlying these trends, azoxymethane and dextran sodium sulphate were administered separately to mice of each genotype. Nfkb2(-/-) mice developed fewer clinical signs of colitis and exhibited less severe colitis and an attenuated cytokine response compared with all other groups following DSS administration. Azoxymethane administration did not fully suppress colonic epithelial mitosis in c-Rel(-/-) mice and less colonic epithelial apoptosis was also observed in this genotype compared to wild-type counterparts. These observations demonstrate different functions of specific NF-κB subunits in this model of colitis-associated carcinogenesis. NF-κB2/p52 is necessary for the development of colitis, whilst c-Rel-mediated signalling regulates colonic epithelial cell turnover following DNA damage.


Asunto(s)
Adenoma/metabolismo , Colitis/complicaciones , Neoplasias del Colon/metabolismo , Subunidad p50 de NF-kappa B/metabolismo , Subunidad p52 de NF-kappa B/metabolismo , Proteínas Proto-Oncogénicas c-rel/metabolismo , Adenoma/inducido químicamente , Adenoma/etiología , Animales , Azoximetano/toxicidad , Transformación Celular Neoplásica/metabolismo , Colitis/inducido químicamente , Neoplasias del Colon/inducido químicamente , Neoplasias del Colon/etiología , Citocinas/metabolismo , Sulfato de Dextran/toxicidad , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Células Epiteliales/metabolismo , Femenino , Inflamación , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal
3.
Carcinogenesis ; 35(8): 1798-806, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24710625

RESUMEN

Stromal cells influence cancer progression. Myofibroblasts are an important stromal cell type, which influence the tumour microenvironment by release of extracellular matrix (ECM) proteins, proteases, cytokines and chemokines. The mechanisms of secretion are poorly understood. Here, we describe the secretion of marker proteins in gastric cancer and control myofibroblasts in response to insulin-like growth factor (IGF) stimulation and, using functional genomic approaches, we identify proteins influencing the secretory response. IGF rapidly increased myofibroblast secretion of an ECM protein, TGFßig-h3. The secretory response was not blocked by inhibition of protein synthesis and was partially mediated by increased intracellular calcium (Ca(2+)). The capacity for evoked secretion was associated with the presence of dense-core secretory vesicles and was lost in cells from patients with advanced gastric cancer. In cells responding to IGF-II, the expression of neuroendocrine marker proteins, including secretogranin-II and proenkephalin, was identified by gene array and LC-MS/MS respectively, and verified experimentally. The expression of proenkephalin was decreased in cancers from patients with advanced disease. Inhibition of secretogranin-II expression decreased the secretory response to IGF, and its over-expression recovered the secretory response consistent with a role in secretory vesicle biogenesis. We conclude that normal and some gastric cancer myofibroblasts have a neuroendocrine-like phenotype characterized by Ca(2+)-dependent regulated secretion, dense-core secretory vesicles and expression of neuroendocrine marker proteins; loss of the phenotype is associated with advanced cancer. A failure to regulate myofibroblast protein secretion may contribute to cancer progression.


Asunto(s)
Factor II del Crecimiento Similar a la Insulina/metabolismo , Miofibroblastos/patología , Sistemas Neurosecretores/patología , Secretogranina II/metabolismo , Neoplasias Gástricas/patología , Western Blotting , Estudios de Casos y Controles , Células Cultivadas , Progresión de la Enfermedad , Exocitosis/fisiología , Mucosa Gástrica/metabolismo , Humanos , Técnicas para Inmunoenzimas , Marcaje Isotópico , Miofibroblastos/metabolismo , Sistemas Neurosecretores/metabolismo , Fenotipo , ARN Interferente Pequeño/genética , Secretogranina II/antagonistas & inhibidores , Secretogranina II/genética , Neoplasias Gástricas/metabolismo , Espectrometría de Masas en Tándem
4.
J Physiol ; 592(14): 2951-8, 2014 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-24665102

RESUMEN

The existence of the hormone gastrin in the distal stomach (antrum) has been known for almost 110 years, and the physiological function of this amidated peptide in regulating gastric acid secretion via the CCK2 receptor is now well established. In this brief review we consider important additional roles of gastrin, including regulation of genes encoding proteins such as plasminogen activator inhibitors and matrix metalloproteinases that have important actions on extracellular matrix remodelling. These actions are, at least in part, effected by paracrine signalling pathways and make important contributions to maintaining functional integrity of the gastric epithelium. Recent studies also provide support for the idea that gastrin, in concert with other hormones, could potentially contribute a post-prandial incretin effect. We also review recent developments in the biology of other gastrin gene products, including the precursor progastrin, which causes proliferation of the colonic epithelium and in certain circumstances may induce cancer formation. Glycine-extended biosynthetic processing intermediates also have proliferative effects in colonic mucosa and in some oesophageal cancer cell lines. Whether these additional gene products exert their effects through the CCK2 receptor or a separate entity is currently a matter of debate.


Asunto(s)
Gastrinas/fisiología , Animales , Mucosa Gástrica/metabolismo , Humanos , Incretinas/metabolismo , Receptor de Colecistoquinina B/metabolismo , Neoplasias Gástricas/metabolismo
5.
Am J Physiol Gastrointest Liver Physiol ; 304(9): G814-22, 2013 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-23494120

RESUMEN

Gastric mucosal health is maintained in response to potentially damaging luminal factors. Aspirin and nonsteroidal anti-inflammatory drugs (NSAIDs) disrupt protective mechanisms leading to bleeding and ulceration. The plasminogen activator system has been implicated in fibrinolysis following gastric ulceration, and an inhibitor of this system, plasminogen activator inhibitor (PAI)-1, is expressed in gastric epithelial cells. In Helicobacter pylori-negative patients with normal gastric histology taking aspirin or NSAIDs, we found elevated gastric PAI-1 mRNA abundance compared with controls; the increase in patients on aspirin was independent of whether they were also taking proton pump inhibitors. In the same patients, aspirin tended to lower urokinase plasminogen activator mRNA. Immunohistochemistry indicated PAI-1 localization to epithelial cells. In a model system using MKN45 or AGS-GR cells transfected with a PAI-1 promoter-luciferase reporter construct, we found no evidence for upregulation of PAI-1 expression by indomethacin, and, in fact, cyclooxygenase products such as PGE2 and PGI2 weakly stimulated expression. Increased gastric PAI-1 mRNA was also found in mice following gavage with ethanol or indomethacin, but plasma PAI-1 was unaffected. In PAI-1(-/-) mice, gastric hemorrhagic lesions in response to ethanol or indomethacin were increased compared with C57BL/6 mice. In contrast, in PAI-1-H/Kß mice in which PAI-1 is overexpressed in parietal cells, there were decreased lesions in response to ethanol and indomethacin. Thus, PAI-1 expression is increased in gastric epithelial cells in response to mucosal irritants such as aspirin and NSAIDs probably via an indirect mechanism, and PAI-1 acts as a local autoregulator to minimize mucosal damage.


Asunto(s)
Mucosa Gástrica/efectos de los fármacos , Inhibidor 1 de Activador Plasminogénico/fisiología , Animales , Aspirina/farmacología , Dinoprostona , Etanol/toxicidad , Femenino , Humanos , Indometacina/toxicidad , Masculino , Ratones , Inhibidor 1 de Activador Plasminogénico/biosíntesis , ARN Mensajero/metabolismo , Activador de Plasminógeno de Tipo Uroquinasa/biosíntesis
6.
Pflugers Arch ; 463(3): 459-75, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22138972

RESUMEN

Myofibroblasts play central roles in wound healing, deposition of the extracellular matrix and epithelial function. Their functions depend on migration and proliferation within the subepithelial matrix, which results in accelerated cellular metabolism. Upregulated metabolic pathways generate protons which need to be excreted to maintain intracellular pH (pH(i)). We isolated human gastric myofibroblasts (HGMs) from surgical specimens of five patients. Then we characterized, for the first time, the expression and functional activities of the Na(+)/H(+) exchanger (NHE) isoforms 1, 2 and 3, and the functional activities of the Na(+)/HCO(3)(-) cotransporter (NBC) and the anion exchanger (AE) in cultured HGMs using microfluorimetry, immunocytochemistry, reverse transcription polymerase chain reaction and immunoblot analysis. We showed that NHE1-3, NBC and AE activities are present in HGMs and that NHE1 is the most active of the NHEs. In scratch wound assays we also demonstrated (using the selective NHE inhibitor HOE-642) that carbachol and insulin like growth factor II (IGF-II) partly stimulate migration of HGMs in a NHE1-dependent manner. EdU incorporation assays revealed that IGF-II induces proliferation of HGMs which is inhibited by HOE-642. The results indicate that NHE1 is necessary for IGF-II-induced proliferation response of HGMs. Overall, we have characterized the pH(i) regulatory mechanisms of HGMs. In addition, we demonstrated that NHE1 activity contributes to both IGF-II- and carbachol-stimulated migration and that it is obligatory for IGF-II-induced proliferation of HGMs.


Asunto(s)
Proteínas de Transporte de Catión/fisiología , Miofibroblastos/fisiología , Intercambiadores de Sodio-Hidrógeno/fisiología , Adulto , Anciano , Antiportadores/biosíntesis , Carbacol/farmacología , Proteínas de Transporte de Catión/antagonistas & inhibidores , Movimiento Celular , Proliferación Celular , Femenino , Guanidinas/farmacología , Humanos , Concentración de Iones de Hidrógeno , Factor II del Crecimiento Similar a la Insulina/fisiología , Masculino , Simportadores de Sodio-Bicarbonato/fisiología , Intercambiador 1 de Sodio-Hidrógeno , Intercambiador 3 de Sodio-Hidrógeno , Intercambiadores de Sodio-Hidrógeno/antagonistas & inhibidores , Intercambiadores de Sodio-Hidrógeno/biosíntesis , Estómago/citología , Sulfonas/farmacología
7.
Am J Physiol Gastrointest Liver Physiol ; 301(3): G446-53, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21193525

RESUMEN

Plasminogen activator inhibitor (PAI)-1 is associated with cancer progression, fibrosis and thrombosis. It is expressed in the stomach but the mechanisms controlling its expression there, and its biological role, are uncertain. We sought to define the role of gastrin in regulating PAI-1 expression and to determine the relevance for gastrin-stimulated cell migration and invasion. In gastric biopsies from subjects with elevated plasma gastrin, the abundances of PAI-1, urokinase plasminogen activator (uPA), and uPA receptor (uPAR) mRNAs measured by quantitative PCR were increased compared with subjects with plasma concentrations in the reference range. In patients with hypergastrinemia due to autoimmune chronic atrophic gastritis, there was increased abundance of PAI-1, uPA, and uPAR mRNAs that was reduced by octreotide or antrectomy. Immunohistochemistry revealed localization of PAI-1 to parietal cells and enterochromaffin-like cells in micronodular neuroendocrine tumors in hypergastrinemic subjects. Transcriptional mechanisms were studied by using a PAI-1-luciferase promoter-reporter construct transfected into AGS-G(R) cells. There was time- and concentration-dependent increase of PAI-1-luciferase expression in response to gastrin that was reversed by inhibitors of the PKC and MAPK pathways. In Boyden chamber assays, recombinant PAI-1 inhibited gastrin-stimulated AGS-G(R) cell migration and invasion, and small interfering RNA treatment increased responses to gastrin. We conclude that elevated plasma gastrin concentrations are associated with increased expression of gastric PAI-1, which may act to restrain gastrin-stimulated cell migration and invasion.


Asunto(s)
Células Epiteliales/metabolismo , Gastrinas/farmacología , Inhibidor 1 de Activador Plasminogénico/biosíntesis , Células Similares a las Enterocromafines/metabolismo , Gastrinas/sangre , Humanos , Octreótido , ARN Mensajero/metabolismo , Receptores del Activador de Plasminógeno Tipo Uroquinasa/biosíntesis , Estómago/citología , Activador de Plasminógeno de Tipo Uroquinasa/biosíntesis
8.
Gastroenterology ; 138(4): 1479-90, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19854189

RESUMEN

BACKGROUND & AIMS: Cholecystokinin (CCK) acts on vagal afferent neurons to inhibit food intake and gastric emptying; it also increases expression of the neuropeptide cocaine- and amphetamine-regulated transcript (CART), but the significance of this is unknown. We investigated the role of CARTp in vagal afferent neurons. METHODS: Release of CART peptide (CARTp) from cultured vagal afferent neurons was determined by enzyme-linked immunosorbent assay. Expression of receptors and neuropeptides in rat vagal afferent neurons in response to CARTp was studied using immunohistochemistry and luciferase promoter reporter constructs. Effects of CARTp and CCK were studied on food intake. RESULTS: CCK stimulated CARTp release from cultured nodose neurons. CARTp replicated the effect of CCK in stimulating expression of Y2R and of CART itself in these neurons in vivo and in vitro, but not in inhibiting cannabinoid-1, melanin-concentrating hormone, and melanin-concentrating hormone-1 receptor expression. Effects of CCK on Y2R and CART expression were reduced by CART small interfering RNA or brefeldin A. Exposure of rats to CARTp increased the inhibitory action of CCK on food intake after short-, but not long-duration, fasting. CONCLUSIONS: The actions of CCK in stimulating CART and Y2R expression in vagal afferent neurons and in inhibiting food intake are augmented by CARTp; CARTp is released by CCK from these neurons, indicating that it acts as an autocrine excitatory mediator.


Asunto(s)
Colecistoquinina/farmacología , Proteínas del Tejido Nervioso/fisiología , Neuronas Aferentes/efectos de los fármacos , Nervio Vago/efectos de los fármacos , Animales , Células Cultivadas , Hormonas Hipotalámicas/genética , Masculino , Melaninas/genética , Proteínas del Tejido Nervioso/genética , Ganglio Nudoso/efectos de los fármacos , Hormonas Hipofisarias/genética , Regiones Promotoras Genéticas , Ratas , Ratas Wistar , Receptores de Neuropéptido Y/genética
9.
Artículo en Inglés | MEDLINE | ID: mdl-20430875

RESUMEN

The intestinal hormone cholecystokinin (CCK) inhibits food intake via stimulation of vagal afferent neurons (VAN). Recent studies suggest that CCK also regulates the expression of some G protein-coupled receptors and neuropeptide transmitters in these neurons. The aim of the present study was to characterize the expression of cannabinoid (CB)1 receptors in VAN and to determine whether stimulation of these receptors plays a role in regulating neurochemical phenotype. Expression of CB1 in rat VAN was detectable by in situ hybridization or immunohistochemistry after 6 h of fasting and increased to a maximum after 24 h when approximately 50% of neurons in the mid and caudal regions expressed the receptor. Melanin-concentrating hormone (MCH)1 receptors also increased with fasting, but the changes were delayed compared with CB1; in contrast Y2 receptors (Y2R) exhibited reciprocal changes in expression to CB1. Administration of CCK8s (10 nmol ip) to fasted rats decreased expression of CB1 with a t(1/2) of approximately 1 h compared with 3 h for MCH1. The action of CCK8s was inhibited by ghrelin and orexin-A. The CB1 agonist anandamide (intraperitoneally) reversed the effect of CCK8s on CB1, MCH1, and Y2 receptor expression. In contrast, in rats fasted for 18 h, administration of a CB1 antagonist/inverse agonist (AM281 ip) downregulated CB1 expression and increased Y2 receptor expression. Activation of vagal CB1 receptors therefore influences the neurochemical phenotype of these neurons, indicating a new and hitherto unrecognized role for endocannabinoids in gut-brain signaling.


Asunto(s)
Ingestión de Alimentos , Privación de Alimentos , Neuronas Aferentes/metabolismo , Ganglio Nudoso/metabolismo , Receptor Cannabinoide CB1/metabolismo , Sincalida/metabolismo , Animales , Ácidos Araquidónicos/metabolismo , Agonismo Inverso de Drogas , Endocannabinoides , Ghrelina/metabolismo , Semivida , Inmunohistoquímica , Hibridación in Situ , Inyecciones Intraperitoneales , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Cinética , Masculino , Morfolinas/farmacología , Neuropéptidos/metabolismo , Orexinas , Fenotipo , Alcamidas Poliinsaturadas/metabolismo , Pirazoles/farmacología , Ratas , Ratas Wistar , Receptor Cannabinoide CB1/agonistas , Receptor Cannabinoide CB1/antagonistas & inhibidores , Receptor Cannabinoide CB1/genética , Receptores de Neuropéptido Y/metabolismo , Receptores de Somatostatina/metabolismo , Sincalida/administración & dosificación
10.
J Neurosci ; 28(45): 11583-92, 2008 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-18987194

RESUMEN

The intestinal hormones CCK and PYY3-36 inhibit gastric emptying and food intake via vagal afferent neurons. Here we report that CCK regulates the expression of Y2R, at which PYY3-36 acts. In nodose ganglia from rats fasted up to 48 h, there was a fivefold decrease of Y2R mRNA compared with rats fed ad libitum; Y2R mRNA in fasted rats was increased by administration of CCK, and by refeeding through a mechanism sensitive to the CCK1R antagonist lorglumide. Antibodies to Y2R revealed expression in both neurons and satellite cells; most of the former (89 +/- 4%) also expressed CCK1R. With fasting there was loss of Y2R immunoreactivity in CCK1R-expressing neurons many of which projected to the stomach, but not in satellite cells or neurons projecting to the ileum or proximal colon. Expression of a Y2R promoter-luciferase reporter (Y2R-luc) in cultured vagal afferent neurons was increased in response to CCK by 12.3 +/- 0.1-fold and by phorbol ester (16.2 +/- 0.4-fold); the response to both was abolished by the protein kinase C inhibitor Ro-32,0432. PYY3-36 stimulated CREB phosphorylation in rat nodose neurons after priming with CCK; in wild-type mice PYY3-36 increased Fos labeling in brainstem neurons but in mice null for CCK1R this response was abolished. Thus Y2R is expressed by functionally distinct subsets of nodose ganglion neurons projecting to the stomach and ileum/colon; in the former expression is dependent on stimulation by CCK, and there is evidence that PYY3-36 effects on vagal afferent neurons are CCK dependent.


Asunto(s)
Colagogos y Coleréticos/farmacología , Colecistoquinina/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Neuronas Aferentes/efectos de los fármacos , Receptores de Neuropéptido Y/metabolismo , Estómago/inervación , Nervio Vago/citología , Animales , Conducta Animal/efectos de los fármacos , Conducta Animal/fisiología , Células Cultivadas , Ayuno/fisiología , Antagonistas de Hormonas/farmacología , Humanos , Masculino , Ratones , Ratones Noqueados , Ganglio Nudoso/citología , Proteínas Oncogénicas v-fos/metabolismo , Proglumida/análogos & derivados , Proglumida/farmacología , ARN Mensajero/metabolismo , Ratas , Receptor de Colecistoquinina A/deficiencia , Receptores de Neuropéptido Y/genética , Respuesta de Saciedad/efectos de los fármacos , Respuesta de Saciedad/fisiología
11.
J Neurosci ; 27(11): 2876-82, 2007 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-17360909

RESUMEN

The neuropeptide transmitter cocaine- and amphetamine-regulated transcript (CART) inhibits food intake and is expressed by both vagal afferent and hypothalamic neurons. Here we report that cholecystokinin (CCK) regulates CART expression in rat vagal afferent neurons. Thus, CART was virtually undetectable after energy restriction for 24 h, but administration of CCK to fasted rats increased CART immunoreactivity, and refeeding of fasted animals promptly increased CART by a mechanism sensitive to a CCK-1 receptor antagonist. In vagal afferent neurons incubated in serum-free medium, CART was virtually undetectable, whereas the orexigenic peptide melanin-concentrating hormone (MCH) was readily detected. The addition of CCK rapidly induced CART expression and downregulated MCH. Using a CART promoter-luciferase reporter vector transfected into cultured vagal afferent neurons, we showed that CCK stimulation of CART transcription was mediated by activation of protein kinase C and cAMP response element-binding protein (CREB). The action of CCK on CART expression was inhibited by the orexigenic peptide ghrelin, through a mechanism that involved exclusion of phosphorylated CREB from the nucleus. Thus, CCK reciprocally regulates expression of CART and MCH within the same vagal afferent neuron; ghrelin inhibits the effect of CCK at least in part through control of the nuclear localization of phosphoCREB, revealing previously unsuspected modulation of gut-brain signals implicated in control of food intake.


Asunto(s)
Colecistoquinina/farmacología , Regulación hacia Abajo/fisiología , Proteínas del Tejido Nervioso/biosíntesis , Neuronas Aferentes/metabolismo , Hormonas Peptídicas/farmacología , Regulación hacia Arriba/fisiología , Nervio Vago/metabolismo , Animales , Células Cultivadas , Colecistoquinina/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Ghrelina , Masculino , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/fisiología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas Aferentes/efectos de los fármacos , Ganglio Nudoso/efectos de los fármacos , Ganglio Nudoso/metabolismo , Hormonas Peptídicas/metabolismo , Ratas , Ratas Wistar , Regulación hacia Arriba/efectos de los fármacos , Nervio Vago/efectos de los fármacos
12.
Cancer Res ; 64(5): 1695-702, 2004 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-14996729

RESUMEN

The gastric pathogen Helicobacter pylori is associated with a progression to gastric cancer. The specific targets of H. pylori that might influence this progression are still unclear. Previous studies indicated that the gastric hormone gastrin, which may be increased in H. pylori infection, stimulates gastric expression of plasminogen activator inhibitor (PAI)-2, which is an inhibitor of the urokinase plasminogen activator and has previously been shown to be increased in gastric adenocarcinoma. Here, we report that H. pylori also increases PAI-2 expression. In gastric biopsies of H. pylori-positive subjects there was increased PAI-2, including subjects with plasma gastrin concentrations in the normal range. PAI-2 was expressed mainly in chief and mucous cells. In a gastric cancer cell line (AGS), H. pylori increased PAI-2 expression, which was associated with inhibition of H. pylori-stimulated cell invasion and apoptosis. The induction of PAI-2 by H. pylori was mediated by release of interleukin-8 and activation of cyclooxygenase-2, and interestingly, gastrin stimulated PAI-2 expression by similar paracrine pathways. The activation of NFkappaB was required for interleukin-8 and cyclooxygenase-2 activation but did not occur in cells responding to these paracrine mediators. The data suggest that induction of PAI-2 is a specific target in H. pylori infection, mediated at least partly by paracrine factors; induction of PAI-2 inhibits cell invasion and apoptosis and is a candidate for influencing the progression to gastric cancer.


Asunto(s)
Apoptosis , Mucosa Gástrica/metabolismo , Helicobacter pylori/patogenicidad , FN-kappa B/fisiología , Inhibidor 2 de Activador Plasminogénico/biosíntesis , Neoplasias Gástricas/etiología , Anciano , Ciclooxigenasa 2 , Femenino , Humanos , Interleucina-8/fisiología , Isoenzimas/fisiología , Masculino , Proteínas de la Membrana , Persona de Mediana Edad , Invasividad Neoplásica , Inhibidor 2 de Activador Plasminogénico/análisis , Prostaglandina-Endoperóxido Sintasas/fisiología , Transporte de Proteínas
13.
J Neurosci ; 24(11): 2708-15, 2004 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-15028763

RESUMEN

Both inhibitory (satiety) and stimulatory (orexigenic) factors from the gastrointestinal tract regulate food intake. In the case of the satiety hormone cholecystokinin (CCK), these effects are mediated via vagal afferent neurons. We now report that vagal afferent neurons expressing the CCK-1 receptor also express cannabinoid CB1 receptors. Retrograde tracing established that these neurons project to the stomach and duodenum. The expression of CB1 receptors determined by RT-PCR, immunohistochemistry and in situ hybridization in rat nodose ganglia was increased by withdrawal of food for > or =12 hr. After refeeding of fasted rats there was a rapid loss of CB1 receptor expression identified by immunohistochemistry and in situ hybridization. These effects were blocked by administration of the CCK-1 receptor antagonist lorglumide and mimicked by administration of CCK to fasted rats. Because CCK is a satiety factor that acts via the vagus nerve and CB1 agonists stimulate food intake, the data suggest a new mechanism modulating the effect on food intake of satiety signals from the gastrointestinal tract.


Asunto(s)
Colecistoquinina/fisiología , Neuronas Aferentes/metabolismo , Proglumida/análogos & derivados , Receptor Cannabinoide CB1/metabolismo , Receptor de Colecistoquinina A/metabolismo , Nervio Vago/metabolismo , Animales , Colecistoquinina/farmacología , Técnicas de Cultivo , Duodeno/inervación , Privación de Alimentos/fisiología , Antagonistas de Hormonas/farmacología , Humanos , Inmunohistoquímica , Hibridación in Situ , Masculino , Compresión Nerviosa , Ganglio Nudoso/citología , Ganglio Nudoso/metabolismo , Proglumida/farmacología , Ratas , Ratas Wistar , Receptor Cannabinoide CB1/antagonistas & inhibidores , Receptor Cannabinoide CB1/genética , Receptor Cannabinoide CB2/biosíntesis , Receptor Cannabinoide CB2/genética , Receptor de Colecistoquinina A/antagonistas & inhibidores , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Respuesta de Saciedad/fisiología , Estómago/inervación , Nervio Vago/citología
14.
Biochim Biophys Acta ; 1576(1-2): 198-202, 2002 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-12031502

RESUMEN

In this study, we used the gastric epithelial cell line AGS-G(R) to investigate the role of GATA transcription factors in the regulation of both basal and gastrin-stimulated L-histidine decarboxylase (HDC) gene transcription. Using reporter gene technology, we compared the transcriptional activity of a construct, hHDC503, which contained the 5'-flanking region of the human HDC gene with that of similar constructs lacking selected GATA consensus sequences. We demonstrated the expression of GATA-4 and GATA-6 proteins within the AGS-G(R) cells and found evidence that these transcription factors can negatively regulate HDC gene expression.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Células Epiteliales/metabolismo , Histidina Descarboxilasa/biosíntesis , Factores de Transcripción/metabolismo , Línea Celular , Secuencia de Consenso , Proteínas de Unión al ADN/genética , Ensayo de Cambio de Movilidad Electroforética , Células Epiteliales/efectos de los fármacos , Células Epiteliales/enzimología , Factor de Transcripción GATA4 , Factor de Transcripción GATA6 , Gastrinas/antagonistas & inhibidores , Gastrinas/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Genes Reporteros , Humanos , Factores de Transcripción/genética
15.
Biochem J ; 381(Pt 2): 397-403, 2004 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-15109306

RESUMEN

The expression of members of the Reg family of secreted lectin-like proteins is increased in response to stress, inflammation and damage in many tissues. In the stomach, Reg is located in enterochromaffin-like cells, where its expression is stimulated by the gastric hormone gastrin. We have examined the mechanisms by which gastrin stimulates expression of Reg-1. Deletional mutations of 2.1 to 0.1 kb of the rat Reg-1 promoter in a luciferase reporter vector were transiently transfected into gastric cancer AGS-G(R) cells. All promoter fragments tested showed similar relative increases in luciferase expression in response to gastrin (1 nM). The response to gastrin of the smallest (104 bp) construct was 4.2+/-0.4-fold over basal. These responses were reduced by Ro-32-0432, a protein kinase C inhibitor, by C3-transferase, a Clostridium botulinum toxin and a selective inhibitor of the Rho family GTPase RhoA, and by co-transfection with a dominant negative form of RhoA. Co-transfection with a constitutively active form of RhoA stimulated expression 11.6+/-1.7-fold over basal. Mutations through the 104 bp construct identified a C-rich element (C-79CCCTCCC-72) required for responses to gastrin, PKC (protein kinase C) and L63RhoA (the constitutively active form of human RhoA protein containing a glutamine-to-leucine substitution at position 63). EMSAs (electrophoretic-mobility-shift assays) using nuclear extracts of control and gastrin-stimulated AGS-G(R) cells and a probe spanning -86 to -64 bp revealed multiple binding proteins. There was no effect of gastrin on the pattern of binding. Supershift assays indicated that transcription factors Sp1 and Sp3 bound the C-rich sequence. We conclude that gastrin stimulates Reg expression via activation of PKC and RhoA, that a C-rich region (-79 to -72) is critical for the response and that Sp-family transcription factors bind to this region of the promoter.


Asunto(s)
Citosina/metabolismo , Gastrinas/fisiología , Regulación de la Expresión Génica/fisiología , Proteínas de la Membrana/genética , Proteínas del Tejido Nervioso/genética , Regiones Promotoras Genéticas/fisiología , Proteína de Unión al GTP rhoA/fisiología , Animales , Secuencia de Bases/genética , Secuencia de Bases/fisiología , Sitios de Unión/genética , Sitios de Unión/fisiología , Citocinas/metabolismo , Proteínas de Unión al ADN/fisiología , Activación Enzimática/fisiología , Humanos , Luciferasas/genética , Regiones Promotoras Genéticas/genética , Proteína Quinasa C/fisiología , Ratas , Proteínas Recombinantes de Fusión/genética , Eliminación de Secuencia/genética , Eliminación de Secuencia/fisiología , Factor de Transcripción Sp1/fisiología , Factor de Transcripción Sp3 , Neoplasias Gástricas , Factores de Transcripción/fisiología
16.
PLoS One ; 8(3): e59913, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23544109

RESUMEN

The hormone gastrin physiologically regulates gastric acid secretion and also contributes to maintaining gastric epithelial architecture by regulating expression of genes such as plasminogen activator inhibitor 2 (PAI-2) and regenerating protein 1 (Reg1). Here we examine the role of proteasome subunit PSMB1 in the transcriptional regulation of PAI-2 and Reg1 by gastrin, and its subcellular distribution during gastrin stimulation. We used the gastric cancer cell line AGS, permanently transfected with the CCK2 receptor (AGS-GR) to study gastrin stimulated expression of PAI-2 and Reg1 reporter constructs when PSMB1 was knocked down by siRNA. Binding of PSMB1 to the PAI-2 and Reg1 promoters was assessed by chromatin immunoprecipitation (ChIP) assay. Subcellular distribution of PSMB1 was determined by immunocytochemistry and Western Blot. Gastrin robustly increased expression of PAI-2 and Reg1 in AGS-GR cells, but when PSMB1 was knocked down the responses were dramatically reduced. In ChIP assays, following immunoprecipitation of chromatin with a PSMB1 antibody there was a substantial enrichment of DNA from the gastrin responsive regions of the PAI-2 and Reg1 promoters compared with chromatin precipitated with control IgG. In AGS-GR cells stimulated with gastrin there was a significant increase in the ratio of nuclear:cytoplasmic PSMB1 over the same timescale as recruitment of PSMB1 to the PAI-2 and Reg1 promoters seen in ChIP assays. We conclude that PSMB1 is part of the transcriptional machinery required for gastrin stimulated expression of PAI-2 and Reg1, and that its change in subcellular distribution in response to gastrin is consistent with this role.


Asunto(s)
Gastrinas/farmacología , Litostatina/genética , Inhibidor 2 de Activador Plasminogénico/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Subunidades de Proteína/metabolismo , Transcripción Genética/efectos de los fármacos , Animales , Línea Celular Tumoral , Núcleo Celular/efectos de los fármacos , Núcleo Celular/enzimología , Técnicas de Silenciamiento del Gen , Humanos , Litostatina/metabolismo , Inhibidor 2 de Activador Plasminogénico/metabolismo , Regiones Promotoras Genéticas , Transporte de Proteínas/efectos de los fármacos , Ratas
17.
Endocrinology ; 154(2): 718-26, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23254194

RESUMEN

The adipokine plasminogen activator inhibitor (PAI)-1 is increased in plasma of obese individuals and exhibits increased expression in the stomachs of individuals infected with Helicobacter. To investigate the relevance of gastric PAI-1, we used 1.1 kb of the H(+)/K(+)ß subunit promoter to overexpress PAI-1 specifically in mouse gastric parietal cells (PAI-1-H/Kß mice). We studied the physiological, biochemical, and behavioral characteristics of these and mice null for PAI-1 or a putative receptor, urokinase plasminogen activator receptor (uPAR). PAI-1-H/Kß mice had increased plasma concentrations of PAI-1 and increased body mass, adiposity, and hyperphagia compared with wild-type mice. In the latter, food intake was inhibited by cholecystokinin (CCK)8s, but PAI-1-H/Kß mice were insensitive to the satiating effects of CCK8s. PAI-1-H/Kß mice also had significantly reduced expression of c-fos in the nucleus tractus solitarius in response to CCK8s and refeeding compared with wild-type mice. Exogenous PAI-1 reversed the effects of CCK8s on food intake and c-fos levels in the nucleus tractus solitarius of wild-type mice, but not uPAR-null mice. Infection of C57BL/6 mice with Helicobacter felis increased gastric abundance of PAI-1 and reduced the satiating effects of CCK8s, whereas the response to CCK8s was maintained in infected PAI-1-null mice. In cultured vagal afferent neurons, PAI-1 inhibited stimulation of neuropeptide Y type 2 receptor (Y2R) expression by CCK8s. Thus, gastric expression of PAI-1 is associated with hyperphagia, moderate obesity, and resistance to the satiating effects of CCK indicating a new role in suppressing signals from the upper gut that inhibit food intake.


Asunto(s)
Mucosa Gástrica/metabolismo , Hiperfagia/metabolismo , Obesidad/metabolismo , Inhibidor 1 de Activador Plasminogénico/biosíntesis , Animales , Colecistoquinina/farmacología , Infecciones por Helicobacter/fisiopatología , Helicobacter felis , Ratones , Inhibidor 1 de Activador Plasminogénico/genética , Receptores del Activador de Plasminógeno Tipo Uroquinasa/fisiología , Saciedad/efectos de los fármacos
18.
Eur J Gastroenterol Hepatol ; 25(4): 474-81, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23249603

RESUMEN

BACKGROUND: Type-1 gastric neuroendocrine tumours (NETs) arise in some patients with chronic hypergastrinaemia secondary to autoimmune atrophic gastritis. Patients with small tumours are usually managed conservatively, because their prognosis is very good. However, larger tumours may require surgical intervention. Many type-1 gastric NETs regress following antrectomy because this removes the source of hypergastrinaemia. However, some tumours do not regress following antrectomy and additional surgery may be required. An octreotide suppression test has been previously suggested as a means to assess whether type-1 gastric NETs are likely to regress following antrectomy. AIM: To prospectively examine the role of a short-term intravenous octreotide suppression test in predicting type-1 gastric NET regression in five patients who subsequently underwent antrectomy. MATERIALS AND METHODS: Serum gastrin concentrations and gastric corpus and tumour histidine decarboxylase mRNA abundances were assessed in patients with type-1 gastric NETs before and 72 h after the administration of 25 µg/h intravenous octreotide. Gastric tumour response was assessed endoscopically following subsequent antrectomy. RESULTS: All patients showed significant decreases in serum gastrin concentrations as well as corpus and tumour biopsy histidine decarboxylase mRNA abundance following octreotide infusion. All patients also showed resolution of hypergastrinaemia following subsequent antrectomy. However, tumour regression was only observed in four of the five patients. One patient had a persistent tumour 3 years after antrectomy and required additional surgical resection. CONCLUSION: A positive octreotide suppression test result does not always predict response to antrectomy in patients with type-1 gastric NETs. Assessment of gastric mucosal responses to a gastrin/CCK-2 receptor antagonist may therefore also be helpful.


Asunto(s)
Antineoplásicos Hormonales , Tumores Neuroendocrinos/diagnóstico , Octreótido , Antro Pilórico/cirugía , Neoplasias Gástricas/diagnóstico , Adulto , Anciano , Antineoplásicos Hormonales/administración & dosificación , Femenino , Gastrectomía/métodos , Gastrinas/sangre , Gastroscopía/métodos , Histidina Descarboxilasa/biosíntesis , Histidina Descarboxilasa/genética , Humanos , Infusiones Intravenosas , Masculino , Tumores Neuroendocrinos/cirugía , Octreótido/administración & dosificación , Selección de Paciente , Pronóstico , Estudios Prospectivos , ARN Mensajero/genética , Neoplasias Gástricas/cirugía , Resultado del Tratamiento
19.
Endocrinology ; 151(8): 3589-99, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20534729

RESUMEN

Food intake is regulated by signals from peripheral organs, but the way these are integrated remains uncertain. Cholecystokinin (CCK) from the intestine and leptin from adipocytes interact to inhibit food intake. Our aim was to examine the hypothesis that these interactions occur at the level of vagal afferent neurons via control of the immediate early gene EGR1. We now report that CCK stimulates redistribution to the nucleus of early growth response factor-1 (EGR1) in these neurons in vivo and in culture, and these effects are not dependent on EGR1 synthesis. Leptin stimulates EGR1 expression; leptin alone does not stimulate nuclear translocation, but it strongly potentiates the action of CCK. Ghrelin inhibits CCK-stimulated nuclear translocation of EGR1 and leptin-stimulated EGR1 expression. Expression of the gene encoding the satiety peptide cocaine- and amphetamine-regulated transcript (CARTp) is stimulated by CCK via an EGR1-dependent mechanism, and this is strongly potentiated by leptin. Leptin potentiated inhibition of food intake by endogenous CCK in the rat in conditions reflecting changes in EGR1 activation. The data indicate that by separately regulating EGR1 activation and synthesis, CCK and leptin interact cooperatively to define the capacity for satiety signaling by vagal afferent neurons; manipulation of these interactions may be therapeutically beneficial.


Asunto(s)
Colecistoquinina/farmacología , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Ghrelina/fisiología , Leptina/farmacología , Neuronas Aferentes/efectos de los fármacos , Nervio Vago/efectos de los fármacos , Animales , Regulación del Apetito/efectos de los fármacos , Regulación del Apetito/genética , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Células Cultivadas , Colecistoquinina/metabolismo , Colecistoquinina/fisiología , Regulación hacia Abajo/efectos de los fármacos , Antagonismo de Drogas , Sinergismo Farmacológico , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Proteína 1 de la Respuesta de Crecimiento Precoz/fisiología , Ghrelina/metabolismo , Ghrelina/farmacología , Leptina/metabolismo , Leptina/fisiología , Masculino , Neuronas Aferentes/metabolismo , Transporte de Proteínas/efectos de los fármacos , Ratas , Ratas Wistar , Activación Transcripcional/efectos de los fármacos , Nervio Vago/metabolismo
20.
Am J Physiol Gastrointest Liver Physiol ; 296(2): G414-23, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19074642

RESUMEN

The gastric hormone gastrin regulates the expression of a variety of genes involved in control of acid secretion and also in the growth and organization of the gastric mucosa. One putative target is plasminogen activator inhibitor-2 (PAI-2), which is a component of the urokinase activator system that acts extracellularly to inhibit urokinase plasminogen activator (uPA) and intracellularly to suppress apoptosis. Previous studies have demonstrated that gastrin induces PAI-2 both in gastric epithelial cells expressing the gastrin (CCK-2) receptor and, via activation of paracrine networks, in adjacent cells that do not express the receptor. We have now sought to identify the response element(s) in the PAI-2 promoter targeted by paracrine mediators initiated by gastrin. Mutational analysis identified two putative response elements in the PAI-2 promoter that were downstream of gastrin-activated paracrine signals. One was identified as a putative MAZ site, mutation of which dramatically reduced both basal and gastrin-stimulated responses of the PAI-2 promoter by a mechanism involving PGE(2) and the small GTPase RhoA. Yeast one-hybrid screening identified the other as binding the activating signal cointegrator-1 (ASC-1) complex, which was shown to be the target of IL-8 released by gastrin. RNA interference (RNAi) knockdown of two subunits of the ASC-1 complex (p50 and p65) inhibited induction of PAI-2 expression by gastrin. The data reveal previously unsuspected transcriptional mechanisms activated as a consequence of gastrin-triggered paracrine networks and emphasize the elaborate and complex cellular control mechanisms required for a key component of tissue responses to damage and infection.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Mucosa Gástrica/metabolismo , Gastrinas/metabolismo , Comunicación Paracrina , Inhibidor 2 de Activador Plasminogénico/metabolismo , Elementos de Respuesta , Transducción de Señal , Factores de Transcripción/metabolismo , Secuencia de Bases , Sitios de Unión , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Proteínas de Unión al ADN/genética , Dinoprostona/metabolismo , Humanos , Interleucina-8/metabolismo , Datos de Secuencia Molecular , Mutación , Inhibidor 2 de Activador Plasminogénico/genética , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Factores de Transcripción/genética , Activación Transcripcional , Transfección , Técnicas del Sistema de Dos Híbridos , Regulación hacia Arriba , Proteína de Unión al GTP rhoA/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA