Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 95
Filtrar
1.
J Clin Pathol ; 61(1): 72-8, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17412866

RESUMEN

BACKGROUND: Matrix metalloproteinases (MMPs) have roles in inflammation and other processes relevant to the architectural disturbances seen in the gastric mucosa in response to Helicobacter pylori infection. Upregulation of MMPs has been reported in H pylori infection, but there are no detailed reports regarding altered production of their inhibitors, the tissue inhibitors of metalloproteinases (TIMPs). AIMS: To investigate changes in the abundance of TIMPs in human gastric corpus mucosa and murine stomach in Helicobacter infection, and to study cellular sources in man. METHODS: Gastric corpus biopsy samples were assessed for abundance of mRNA or protein for TIMP-1 to -4 by real-time quantitative PCR or western blotting, respectively. Antral and corpus biopsies were processed for histology, H pylori status and inflammatory scoring. Cellular sources of TIMP-1, -3 and -4 were examined by indirect immunohistochemistry. Circulating gastrin was measured by radioimmunoassay. Also, abundance of TIMP-1, -3 and -4 mRNA in the stomach of Helicobacter felis infected mice post-infection was compared with that of uninfected control animals. RESULTS: Compared with uninfected patients, mRNA and protein for TIMP-1, -3 and -4 were significantly more abundant in the gastric corpus of H pylori infected subjects. Gastric TIMP expression did not differ significantly between hyper- and normogastrinaemic subjects within the H pylori negative and positive groups. There was no difference in mRNA abundance for MMP-3 or -8. Immunohistochemistry showed TIMP proteins localised to gastric epithelial, stromal cells and inflammatory cells. Murine H felis infection was associated with upregulation of TIMP-1 and -3 mRNA. CONCLUSIONS: Helicobacter infection is associated with upregulation of specific TIMPs (TIMP-1 and -3) in glandular epithelium and stroma. It is suggested that increased expression of specific protease inhibitors in the corpus mucosa may exert important effects on extracellular matrix remodelling and influence the outcome of H pylori infection.


Asunto(s)
Mucosa Gástrica/metabolismo , Gastritis/metabolismo , Infecciones por Helicobacter/metabolismo , Helicobacter pylori , Inhibidores Tisulares de Metaloproteinasas/metabolismo , Animales , Gastrinas/sangre , Gastritis/sangre , Gastritis/microbiología , Gastritis/patología , Infecciones por Helicobacter/sangre , Infecciones por Helicobacter/patología , Humanos , Ratones , Ratones Transgénicos , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Índice de Severidad de la Enfermedad , Inhibidores Tisulares de Metaloproteinasas/genética , Regulación hacia Arriba
2.
Regul Pept ; 146(1-3): 147-56, 2008 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-17900712

RESUMEN

Plasma concentrations of the hormone gastrin are elevated by Helicobacter pylori infection and by gastric atrophy. It has previously been proposed that gastrin acts as a cofactor during gastric carcinogenesis and hypergastrinemic transgenic INS-GAS mice are prone to developing gastric adenocarcinoma, particularly following H. pylori infection. We hypothesised that the increased risk of carcinogenesis in these animals may partly result from altered susceptibility of gastric epithelial cells to undergo apoptosis. Gastric corpus apoptosis was significantly increased 48 h after 12Gy gamma-radiation in mice rendered hypergastrinemic by transgenic (INS-GAS) or pharmacological (omeprazole treatment of FVB/N mice) methods and in both cases the effects were inhibited by the CCK-2 receptor antagonist YM022. However, no alteration in susceptibility to gamma-radiation-induced gastric epithelial apoptosis was observed in mice overexpressing progastrin or glycine-extended gastrin. Apoptosis was also significantly increased in gastric corpus biopsies obtained from H. pylori-infected humans with moderate degrees of hypergastrinemia. We conclude that hypergastrinemia specifically renders cells within the gastric corpus epithelium more susceptible to induction of apoptosis by radiation or H. pylori. Altered susceptibility to apoptosis may therefore be one factor predisposing to gastric carcinogenesis in INS-GAS mice and similar mechanisms may also be involved in humans.


Asunto(s)
Apoptosis , Susceptibilidad a Enfermedades , Mucosa Gástrica/patología , Gastrinas/sangre , Neoplasias Gástricas/etiología , Animales , Antiulcerosos/farmacología , Benzodiazepinas/farmacología , Células Cultivadas , Rayos gamma , Mucosa Gástrica/efectos de los fármacos , Mucosa Gástrica/efectos de la radiación , Helicobacter pylori , Humanos , Ratones , Ratones Transgénicos , Omeprazol/farmacología , Receptor de Colecistoquinina B/antagonistas & inhibidores
3.
Am J Physiol Gastrointest Liver Physiol ; 288(3): G541-9, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15486344

RESUMEN

Transgenic mice (hGAS) that overexpress human progastrin are more susceptible than wild-type mice (FVB/N) to the induction of colonic aberrant crypt foci (ACF) and adenomas by the chemical carcinogen azoxymethane. We have previously shown significantly increased levels of colonic mitosis in hGAS compared with FVB/N mice after gamma-radiation. To investigate whether the effects of progastrin observed in hGAS colon require the presence of other forms of circulating gastrin, we have crossed hGAS (hg(+/+)) with gastrin knockout (G(-/-)) mice to generate mice that express progastrin and no murine gastrin (G(-/-)hg(+/+)). After azoxymethane, G(-/-)hg(+/+) mice developed significantly more ACF than control G(-/-)hg(-/-) mice (which do not express any forms of gastrin). G(-/-)hg(+/+) mice also exhibited significantly increased colonic mitosis both before and after exposure to 8 Gray Gy gamma-radiation or 50 mg/kg azoxymethane compared with G(-/-)hg(-/-). Treatment of G(-/-)hg(-/-) mice with synthetic progastrin (residues 21-101 of human preprogastrin) or G17 extended at its COOH terminus corresponding to the COOH-terminal 26-amino-acid residues of human preprogastrin (residues 76-101, G17-CFP) resulted in continued colonic epithelial mitosis after gamma-radiation, whereas glycine-extended gastrin-17 and the COOH-terminal tryptic fragment of progastrin [human preprogastrin-(96-101)] had no effect. Immunoneutralization with an antibody against G17-CFP before gamma-radiation significantly decreased colonic mitosis in G(-/-)hg(+/+) mice to levels similar to G(-/-)hg(-/-). We conclude that progastrin does not require the presence of other forms of gastrin to exert proliferative effects on colonic epithelia and that the portion of the peptide responsible for these effects is contained within amino acid residues 76-101 of human preprogastrin.


Asunto(s)
Colon/citología , Células Epiteliales/efectos de los fármacos , Gastrinas/farmacología , Mitógenos , Mitosis/efectos de los fármacos , Fragmentos de Péptidos/farmacología , Precursores de Proteínas/farmacología , Secuencia de Aminoácidos , Animales , Antimetabolitos , Apoptosis/efectos de los fármacos , Azoximetano/farmacología , Bromodesoxiuridina , Carcinógenos/farmacología , Colon/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Daño del ADN/efectos de la radiación , Rayos gamma , Gastrinas/química , Gastrinas/genética , Genotipo , Humanos , Inmunohistoquímica , Ratones , Ratones Noqueados , Ratones Transgénicos , Fragmentos de Péptidos/química , Precursores de Proteínas/química
5.
Am J Physiol Gastrointest Liver Physiol ; 283(3): G787-93, 2002 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12181195

RESUMEN

Recent studies suggest that gastrin regulates parietal cell maturation. We asked whether it also regulates parietal cell life span and migration along the gland. Dividing cells were labeled with 5'-bromo-2'-deoxyuridine (BrdU), and parietal cells were identified by staining with Dolichos biflorus lectin. Cells positive for D. biflorus lectin and BrdU were reliably identified 10-30 days after BrdU injection in mice in which the gastrin gene had been deleted by homologous recombination (Gas-KO) and wild-type (C57BL/6) mice. The time course of labeling was similar in the two groups. The distribution of BrdU-labeled parietal cells in wild-type mice was consistent with migration to the base of the gland, but in Gas-KO mice, a higher proportion of BrdU-labeled cells was found more superficially 20 and 30 days after BrdU injection. Conversely, in transgenic mice overexpressing gastrin, BrdU-labeled parietal cells accounted for a higher proportion of the labeled pool in the base of the gland 10 days after BrdU injection. Gastrin, therefore, stimulates movement of parietal cells along the gland axis but does not influence their life span.


Asunto(s)
Gastrinas/fisiología , Células Parietales Gástricas/fisiología , Lectinas de Plantas , Animales , Antibacterianos/farmacología , Bromodesoxiuridina , Movimiento Celular/fisiología , Supervivencia Celular , Gastrinas/sangre , Gastrinas/genética , ATPasa Intercambiadora de Hidrógeno-Potásio/metabolismo , Lectinas/farmacocinética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados/genética , Células Parietales Gástricas/efectos de los fármacos , Distribución Tisular
6.
Am J Physiol Gastrointest Liver Physiol ; 283(2): G292-9, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12121875

RESUMEN

Epithelial organization is maintained by cell proliferation, migration, and differentiation. In the case of the gastric epithelium, at least some of these events are regulated by the hormone gastrin. In addition, gastric epithelial cells are organized into characteristic tubular structures (the gastric glands), but the cellular mechanisms regulating the organization of tubular structures (sometimes called branching morphogenesis) are uncertain. In the present study, we examined the role of the gastrin-cholecystokinin(B) receptor in promoting branching morphogenesis of gastric epithelial cells. When gastric cancer AGS-G(R) cells were cultured on plastic, gastrin and PMA stimulated cell adhesion, formation of lamellipodia, and extension of long processes in part by activation of protein kinase C (PKC) and phosphatidylinositol (PI)-3 kinase. Branching morphogenesis was not observed in these circumstances. However, when cells were cultured on artificial basement membrane, the same stimuli increased the formation of organized multicellular arrays, exhibiting branching morphogenesis. These effects were reversed by inhibitors of PKC but not of PI-3 kinase. We conclude that, in the presence of basement membrane, activation of PKC by gastrin stimulates branching morphogenesis.


Asunto(s)
Receptores de Colecistoquinina/metabolismo , Neoplasias Gástricas/patología , Membrana Basal/patología , Adhesión Celular/efectos de los fármacos , Gastrinas/farmacología , Humanos , Lisofosfolípidos/farmacología , Proteína Quinasa C/fisiología , Receptor de Colecistoquinina B , Neoplasias Gástricas/fisiopatología , Acetato de Tetradecanoilforbol/farmacología , Células Tumorales Cultivadas/efectos de los fármacos
7.
Gut ; 50(6): 827-33, 2002 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12010885

RESUMEN

BACKGROUND: Activation of the gastrin-cholecystokinin(B) (CCK(B)) receptor stimulates cell proliferation and increases production of ligands for the epidermal growth factor receptor (EGF-R). AIMS: To determine the role of gastrin-CCK(B) activation in stimulation of cell proliferation via paracrine activation of EGF-R. METHODS: AGS cells were transfected with the gastrin-CCK(B) receptor (AGS-G(R) cells) or with green fluorescent protein (AGS-GFP cells). Proliferation was determined by [(3)H] thymidine incorporation, flow cytometry, and cell counting. RESULTS: Gastrin inhibited proliferation of AGS-G(R) cells by delaying entry into S phase. However, when AGS-G(R) cells were cocultured with AGS-GFP cells, gastrin stimulated proliferation of the latter. Immunoneutralisation and pharmacological studies using metalloproteinase and kinase inhibitors indicated that the proliferative response was mediated by paracrine stimulation of EGF-R and activation of the mitogen activated protein kinase pathway through release of heparin binding EGF. CONCLUSIONS: Gastrin can directly inhibit, and indirectly stimulate, proliferation of gastric AGS cells.


Asunto(s)
Células Enterocromafines/metabolismo , Receptores ErbB/metabolismo , Receptores de Colecistoquinina/metabolismo , División Celular/fisiología , Citometría de Flujo , Proteínas Fluorescentes Verdes , Humanos , Proteínas Luminiscentes/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Receptor de Colecistoquinina B , Timidina/metabolismo
8.
J Physiol ; 531(Pt 3): 605-17, 2001 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-11251044

RESUMEN

1. The acidic interior of neuroendocrine secretory vesicles provides both an energy gradient for amine-proton exchangers (VMATs) to concentrate small transmitter molecules, for example catecholamines, and an optimal pH for the prohormone convertases which cleave hormone precursors. There is evidence that VMAT activity modulates prohormone cleavage, but in the absence of measurements of pH in secretory vesicles in intact cells, it has not been possible to establish whether these effects are attributable to raised intravesicular pH due to proton transport through VMATs. 2. Clones were generated of the hamster insulinoma cell line HIT-T15 expressing a pH-sensitive form of green fluorescent protein (GFP-F64L/S65T) targeted to secretory vesicles, with and without co-expression of VMAT2. In order to study prohormone cleavage, further clones were generated that expressed preprogastrin with and without co-expression of VMAT2. 3. Confocal microscopy of GFP fluorescence indicated that the pH in the secretory vesicles was 5.6 in control cells, compared with 6.6 in cells expressing VMAT2; the latter was reduced to 5.8 by the VMAT inhibitor reserpine. 4. Using a pulse-chase labelling protocol, cleavage of 34-residue gastrin (G34) was found to be inhibited by co-expression with VMAT2, and this was reversed by reserpine. Similar effects on vesicle pH and G34 cleavage were produced by ammonium chloride. 5. We conclude that VMAT expression confers the linked abilities to store biogenic amines and modulate secretory vesicle pH over a range influencing prohormone cleavage and therefore determining the identity of regulatory peptide secretory products.


Asunto(s)
Álcalis/metabolismo , Hidrógeno/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas de Transporte de Membrana , Neuropéptidos , Sistemas Neurosecretores/metabolismo , Vesículas Secretoras/metabolismo , Cloruro de Amonio/farmacología , Animales , Cricetinae , Gastrinas/antagonistas & inhibidores , Gastrinas/efectos de los fármacos , Gastrinas/metabolismo , Gastrinas/fisiología , Proteínas Fluorescentes Verdes , Concentración de Iones de Hidrógeno , Indicadores y Reactivos , Proteínas Luminiscentes , Glicoproteínas de Membrana/antagonistas & inhibidores , Microscopía Confocal , Sistemas Neurosecretores/citología , Precursores de Proteínas/antagonistas & inhibidores , Precursores de Proteínas/efectos de los fármacos , Precursores de Proteínas/metabolismo , Precursores de Proteínas/fisiología , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Reserpina/farmacología , Células Tumorales Cultivadas , Proteínas de Transporte Vesicular de Aminas Biógenas , Proteínas de Transporte Vesicular de Monoaminas
10.
Annu Rev Physiol ; 63: 119-39, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11181951

RESUMEN

Gastric epithelial organization and function are controlled and maintained by a variety of endocrine and paracrine mediators. Peptides encoded by the gastrin gene are an important part of this system because targeted deletion of the gene, or of the gastrin-CCKB receptor gene, leads to decreased numbers of parietal cells and decreased gastric acid secretion. Recent studies indicate that the gastrin precursor, preprogastrin, gives rise to a variety of products, each with a distinctive spectrum of biological activity. The conversion of progastrin to smaller peptides is regulated by multiple mechanisms including prohormone phosphorylation and secretory vesicle pH. Progastrin itself stimulates colonic epithelial proliferation; biosynthetic intermediates (Gly-gastrins) stimulate colonic epithelial proliferation and gastric epithelial differentiation; and C-terminally amidated gastrins stimulate colonic proliferation, gastric epithelial proliferation and differentiation, and acid secretion. The effects of progastrin-derived peptides on gastric epithelial function are mediated in part by release of paracrine factors that include histamine, epidermal growth factor (EGF)-receptor ligands, and Reg. The importance of the appropriate regulation of this system is shown by the observation that prolonged moderate hypergastrinemia in transgenic mice leads to remodelling of the gastric epithelium, and in the presence of Helicobacter, to gastric cancer.


Asunto(s)
Gastrinas/biosíntesis , Gastrinas/metabolismo , Células Parietales Gástricas/metabolismo , Animales , Humanos
12.
Gastroenterology ; 119(3): 756-65, 2000 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-10982770

RESUMEN

BACKGROUND & AIMS: Studies in gastrin-deficient mice have demonstrated critical roles for gastrin peptides in the regulation of gastric acid secretion, but the relative contributions of amidated (G-17) and glycine-extended (G17-Gly) gastrin remain unclear. We examined the effects of these 2 forms of gastrin on acid secretion in gastrin-deficient mice. METHODS: Sixty gastrin-deficient mice received infusions of saline, or 1, 6, or 14 days of amidated gastrin 17 (G-17), G17-Gly, or both G-17 and G17-Gly at 10 nmol. kg(-1). h(-1). Twenty-four gastrin-deficient mice were then infused for 14 days with 1, 2, or 5 nmol. kg(-1). h(-1) of G-17 or G-17 and G17-Gly. Acid secretion was determined 4 hours after pyloric ligation, and gastric tissue was processed for histology, immunohistochemistry, and electron microscopy. RESULTS: Infusion of G-17 increased acid secretion in a dose-dependent manner with a peak at 5 nmol. kg(-1). h(-1) and a subsequent decrease in acid secretion at higher doses. Infusion of G17-Gly alone had no effect on acid secretion, but coinfusion with G-17 resulted in significantly higher levels of acid secretion at all doses examined than infusion with G-17 alone. The potentiating effect of G17-Gly on G-17-induced acid secretion was associated with increased parietal cell activation but was independent of changes in parietal and enterochromaffin-like cell number, fundic proliferation rates, and H(+),K(+)-adenine triphosphatase expression. G17-Gly also prevented the formation of vacuolar canaliculi and lipofuscin bodies in the parietal cells induced by G-17. CONCLUSIONS: G17-Gly appears to synergize with G-17 to up-regulate acid secretion and prevent parietal cell degradation. These results suggest that G17-Gly plays an important role in parietal cell function.


Asunto(s)
Ácido Gástrico/metabolismo , Gastrinas/deficiencia , Gastrinas/farmacología , Animales , División Celular/efectos de los fármacos , Sinergismo Farmacológico , Mucosa Gástrica/patología , Gastrinas/sangre , Gastrinas/genética , ATPasa Intercambiadora de Hidrógeno-Potásio/metabolismo , Hiperplasia , Ratones , Ratones Noqueados/genética , Células Parietales Gástricas/efectos de los fármacos , Células Parietales Gástricas/patología , Células Parietales Gástricas/fisiología , Estómago/enzimología
13.
Am J Physiol Gastrointest Liver Physiol ; 279(3): G605-12, 2000 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-10960361

RESUMEN

The mechanisms by which neuroendocrine stimulants regulate CCK gene transcription are unclear. We examined promoter activation by pituitary adenylate cyclase-activating polypeptide (PACAP), a known CCK secretagogue, in the enteroendocrine cell line STC-1. The promoter region from -70 to -87 bp, relative to the transcriptional start site, contains a composite calcium/cyclic AMP response element (CRE)/activator protein 1 (AP1) site that may bind CRE binding protein (CREB) and AP1. PACAP (with IBMX) stimulated expression of an 87-bp construct 3.35+/-0.36-fold but had no effect on a -70 construct. The effect was blocked by the protein kinase A inhibitor H-89 and by a dominant-negative CREB plasmid. Mutation of the CRE/AP1 site to a canonical CRE site did not affect the response to PACAP, but mutation to a canonical AP1 site prevented it. CREB phosphorylation was increased after PACAP treatment. Electrophoretic mobility shift assay and supershift analysis revealed that CREB and not AP1 bound to the CRE/AP1 site and that PACAP increased the proportion of phosphorylated CREB that was bound. We conclude that PACAP increases CCK gene expression via a cAMP-mediated pathway involving CREB phosphorylation by protein kinase A and activation of a composite CRE/AP1 site.


Asunto(s)
Colecistoquinina/genética , Neuropéptidos/genética , Sulfonamidas , Transcripción Genética/fisiología , Animales , Calcio/metabolismo , Colforsina/farmacología , AMP Cíclico/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/antagonistas & inhibidores , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Electroforesis , Inhibidores Enzimáticos/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/fisiología , Genes Reporteros , Isoquinolinas/farmacología , Luciferasas/genética , Ratones , Mutagénesis/fisiología , Tumores Neuroendocrinos , Sondas de Oligonucleótidos , Fosforilación , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa , Regiones Promotoras Genéticas/fisiología , Ratas , Factor de Transcripción AP-1/genética , Factor de Transcripción AP-1/metabolismo , Células Tumorales Cultivadas
14.
J Clin Invest ; 106(4): 533-9, 2000 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-10953028

RESUMEN

Mutations in the adenomatous polyposis coli (APC) tumor suppressor gene occur in most colorectal cancers and lead to activation of beta-catenin. Whereas several downstream targets of beta-catenin have been identified (c-myc, cyclin D1, PPARdelta), the precise functional significance of many of these targets has not been examined directly using genetic approaches. Previous studies have shown that the gene encoding the hormone gastrin is activated during colon cancer progression and the less-processed forms of gastrin are important colonic trophic factors. We show here that the gastrin gene is a downstream target of the beta-catenin/TCF-4 signaling pathway and that cotransfection of a constitutively active beta-catenin expression construct causes a threefold increase in gastrin promoter activity. APC(min-/+) mice overexpressing one of the alternatively processed forms of gastrin, glycine-extended gastrin, show a significant increase in polyp number. Gastrin-deficient APC(min-/+) mice, conversely, showed a marked decrease in polyp number and a significantly decreased polyp proliferation rate. Activation of gastrin by beta-catenin may therefore represent an early event in colorectal tumorigenesis and may contribute significantly toward neoplastic progression. The identification of gastrin as a functionally relevant downstream target of the beta-catenin signaling pathway provides a new target for therapeutic modalities in the treatment of colorectal cancer.


Asunto(s)
Poliposis Adenomatosa del Colon/etiología , Proteínas del Citoesqueleto/fisiología , Gastrinas/fisiología , Transactivadores , Factores de Transcripción/fisiología , Poliposis Adenomatosa del Colon/genética , Poliposis Adenomatosa del Colon/fisiopatología , Animales , Secuencia de Bases , Proteínas del Citoesqueleto/genética , Cartilla de ADN/genética , Modelos Animales de Enfermedad , Femenino , Gastrinas/deficiencia , Gastrinas/genética , Expresión Génica , Genes APC , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Mutación , Regiones Promotoras Genéticas , Transducción de Señal , Factores de Transcripción TCF , Proteína 2 Similar al Factor de Transcripción 7 , Factores de Transcripción/genética , Transfección , beta Catenina
15.
Eur J Pharmacol ; 394(1): 27-34, 2000 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-10771030

RESUMEN

Enteroendocrine cells respond to nutrient and non-nutrient stimuli in the gut lumen. The intestinal hormone cholecystokinin (CCK) is secreted in response to luminal fatty acids, amino acids, peptides and proteins. The peptidomimetic cephalosporins have been reported to provide model, stable, compounds with similar secretagogue activity to peptide. Putative luminal stimuli also influence transcriptional activity in enteroendocrine cells, but the mechanisms are uncertain. In the present study we have investigated the control of c-fos expression in STC-1 cells (an enteroendocrine cell line). Peptidomimetics stimulated calcium-dependent release of CCK, and increased intracellular calcium, phosphorylation of p42/44 mitogen-activated protein kinase (MAP kinase) and c-fos mRNA abundance. Hypotonic stress also increased p42/44 MAP kinase phosphorylation and c-fos mRNA, but not CCK release. The increase in c-fos mRNA was strikingly potentiated by peptidomimetics in hypotonic medium. Increased c-fos expression, but not CCK release, was suppressed by the MAP kinase (MEK) inhibitor PD98059, and by the tyrosine kinase inhibitor genistein. We conclude that in STC-1 cells, peptidomimetics act through the p42/44 MAP kinase pathway to increase c-fos expression but not exocytosis. Moreover, a putative non-nutritive stimulus, hypotonic stress, may interact with this pathway to enhance c-fos expression, independently of hormone release.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Genes fos , Proteína Quinasa 1 Activada por Mitógenos/fisiología , Proteínas Quinasas Activadas por Mitógenos/fisiología , Animales , Calcio/fisiología , Cefaclor/farmacología , Línea Celular , Colecistoquinina/metabolismo , Ratones , Proteína Quinasa 3 Activada por Mitógenos , Concentración Osmolar , ARN Mensajero/análisis
16.
Gastroenterology ; 118(1): 36-47, 2000 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-10611152

RESUMEN

BACKGROUND & AIMS: Hypergastrinemia occurs frequently in association with acid suppression and Helicobacter infection, but its role in the progression to gastric atrophy and gastric cancer has not been well defined. METHODS: The effects of hypergastrinemia, and possible synergy with Helicobacter felis infection, were investigated in insulin-gastrin (INS-GAS) transgenic mice. RESULTS: INS-GAS mice initially showed mild hypergastrinemia, increased maximal gastric acid secretion, and increased parietal cell number but later progressed to decreased parietal cell number and hypochlorhydria. Development of gastric atrophy was associated with increased expression of growth factors, heparin-binding epidermal growth factor and transforming growth factor alpha. At 20 months of age, INS-GAS mice showed no evidence of increased enterochromaffin-like cell number, but instead exhibited gastric metaplasia, dysplasia, carcinoma in situ, and gastric cancer with vascular invasion. Invasive gastric carcinoma was observed in 6 of 8 INS-GAS mice that were >20 months old. Helicobacter felis infection of INS-GAS mice led to accelerated (< or = 8 mo) development of intramucosal carcinoma (85%), with submucosal invasion (54%) and intravascular invasion (46%; P < or = 0.05). CONCLUSIONS: These findings support the unexpected conclusion that chronic hypergastrinemia in mice can synergize with Helicobacter infection and contribute to eventual parietal cell loss and progression to gastric cancer.


Asunto(s)
Gastrinas/sangre , Gastritis Atrófica/sangre , Infecciones por Helicobacter/sangre , Neoplasias Gástricas/sangre , Neoplasias Gástricas/microbiología , Animales , Recuento de Células , Factor de Crecimiento Epidérmico/metabolismo , Células Epiteliales/patología , Ácido Gástrico/metabolismo , Gastritis Atrófica/microbiología , Heparina/metabolismo , Factor de Crecimiento Similar a EGF de Unión a Heparina , Hiperplasia , Hipertrofia , Péptidos y Proteínas de Señalización Intercelular , Metaplasia , Ratones , Ratones Transgénicos , Neoplasias Gástricas/patología , Factor de Crecimiento Transformador alfa/metabolismo
17.
J Physiol ; 518 ( Pt 2): 315-24, 1999 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-10381581

RESUMEN

Transepithelial transducing cells, particularly the gastrin (G) cell, co-ordinate gastric acid secretion with the arrival of food in the stomach. Recent work suggests that multiple active products are generated from the gastrin precursor, and that there are multiple control points in gastrin biosynthesis. Biosynthetic precursors and intermediates (progastrin and Gly-gastrins) are putative growth factors; their products, the amidated gastrins, regulate epithelial cell proliferation, the differentiation of acid-producing parietal cells and histamine-secreting enterochromaffin-like (ECL) cells, and the expression of genes associated with histamine synthesis and storage in ECL cells, as well as acutely stimulating acid secretion. Gastrin also stimulates the production of members of the epidermal growth factor (EGF) family, which in turn inhibit parietal cell function but stimulate the growth of surface epithelial cells. Plasma gastrin concentrations are elevated in subjects with Helicobacter pylori, who are known to have increased risk of duodenal ulcer disease and gastric cancer. Studies of the physiology of gastrin may therefore contribute to an understanding of the mechanisms relevant to major upper gastrointestinal tract disease.


Asunto(s)
Mucosa Gástrica/fisiología , Gastrinas/fisiología , Animales , Células Epiteliales/metabolismo , Células Epiteliales/fisiología , Epitelio/fisiología , Mucosa Gástrica/citología , Humanos , Receptores de Colecistoquinina/fisiología
18.
Gastroenterology ; 116(6): 1310-8, 1999 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10348814

RESUMEN

BACKGROUND & AIMS: The RegIalpha gene (Reg) encodes a secretory protein proposed to regulate islet beta-cell and gastric mucous cell growth. Reg is expressed in rat gastric enterochromaffin-like (ECL) cells. The aim of this study was to examine Reg expression in human corpus and to determine the identity of Reg in ECL cell carcinoid tumors in hypergastrinemic patients. METHODS: Reg messenger RNA (mRNA) abundance was quantified by Northern blot in extracts of gastric corpus from patients with and without ECL cell tumors and in AR4-2J cells stimulated by gastrin; cellular origins were determined by immunocytochemistry. Mutations of Reg were determined by reverse-transcription polymerase chain reaction, cloning, and sequencing, and the mutated protein was expressed in HIT-T15 cells. RESULTS: Reg mRNA abundance was increased approximately threefold in the corpus of hypergastrinemic patients compared with controls, and was enriched in 3 of 7 ECL cell carcinoid tumors but not in non-endocrine cell gastric polyps. In AR4-2J cells, gastrin stimulated Reg mRNA abundance; this was eliminated by the gastrin/cholecystokinin B antagonist L-740,093 (10(-9) mol/L). Immunocytochemistry indicated that Reg was located in both chief cells and ECL cells in human corpus. Mutations of Reg were identified in 3 of 5 patients with ECL cell carcinoid tumors; in 2 cases, mutation of the initiator methionine residue led to exclusion of the protein from the secretory pathway. CONCLUSIONS: Gastrin regulates Reg mRNA abundance in human corpus. Mutations of Reg that prevent secretion are associated with ECL cell carcinoids, suggesting a function as an autocrine or paracrine tumor suppressor.


Asunto(s)
Proteínas de Unión al Calcio/genética , Tumor Carcinoide/genética , Tumor Carcinoide/patología , Células Enterocromafines/patología , Gastrinas/sangre , Mutación/fisiología , Proteínas del Tejido Nervioso , Neoplasias Gástricas/genética , Neoplasias Gástricas/patología , Adulto , Anciano , Anciano de 80 o más Años , Proteínas de Unión al Calcio/metabolismo , Tumor Carcinoide/sangre , Tumor Carcinoide/metabolismo , Neoplasias de las Glándulas Endocrinas/sangre , Femenino , Mucosa Gástrica/metabolismo , Gastrinas/fisiología , Humanos , Litostatina , Masculino , Metionina/genética , Persona de Mediana Edad , Biosíntesis de Proteínas/fisiología , ARN Mensajero/sangre , Neoplasias Gástricas/sangre , Neoplasias Gástricas/metabolismo , Distribución Tisular/fisiología
19.
J Clin Invest ; 103(8): 1119-26, 1999 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-10207163

RESUMEN

Gastrin is a peptide hormone involved in the growth of both normal and malignant gastrointestinal tissue. Recent studies suggest that the glycine-extended biosynthetic intermediates mediate many of these trophic effects, but the in vivo relevance of glycine-extended gastrin (G-Gly) has not been tested. We have generated mice (MTI/G-GLY) that overexpress progastrin truncated at glycine-72 to evaluate the trophic effects of G-Gly in an in vivo model. MTI/G-GLY mice have elevated serum and colonic mucosal levels of G-Gly compared with wild-type mice. MTI/G-GLY mice had a 43% increase in colonic mucosal thickness and a 41% increase in the percentage of goblet cells per crypt. MTI/G-GLY mice exhibited increased colonic proliferation compared with wild-type controls, with an expansion of the proliferative zone into the upper third of the colonic crypts. Continuous infusion of G-Gly into gastrin-deficient mice for two weeks also resulted in elevated G-Gly levels, a 10% increase in colonic mucosal thickness, and an 81% increase in colonic proliferation when compared with gastrin-deficient mice that received saline alone. To our knowledge, these studies demonstrate for the first time that G-Gly's contribute to colonic mucosal proliferation in vivo.


Asunto(s)
Colon/patología , Gastrinas/fisiología , Glicina , Precursores de Proteínas/fisiología , Animales , División Celular , Gastrinas/genética , Neoplasias Gastrointestinales/prevención & control , Expresión Génica , Células Caliciformes/patología , Humanos , Hiperplasia/patología , Hipertrofia/patología , Masculino , Ratones , Ratones Transgénicos , Precursores de Proteínas/genética , Estómago/patología , Células Tumorales Cultivadas
20.
Eur J Pharmacol ; 364(2-3): 199-203, 1999 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-9932724

RESUMEN

Amidated forms of gastrin are derived by post-translational processing of a large precursor peptide and stimulate gastric acid secretion via the gastrin/CCK(B) receptor. Non-amidated biosynthetic intermediates may exert biological effects through other mechanisms, but their effect on gastric acid secretion is unclear. Amidated gastrins stimulate acid secretion mainly by releasing histamine from mucosal enterochromaffin-like cells. This study examines the effects on histamine release from the vascularly perfused rat stomach of amidated gastrin-17, COOH-terminal glycine-extended gastrin-17, gastrin-17 extended at the COOH-terminal including the remaining progastrin sequence, and carboxy-terminal progastrin fragments (SAEDEN and GRRSAEDEN). Carboxy-terminal extended gastrins induced histamine release which was inhibited by the gastrin/CCK(B) antagonist L-740,093, but had to be given in concentrations 100-fold higher than amidated gastrin-17 to produce comparable effects. These progastrin-derived peptides are found in high concentrations in some patients with the Zollinger-Ellison syndrome and may contribute to acid hypersecretion and other gastrin/CCK(B) receptor mediated responses.


Asunto(s)
Gastrinas/farmacología , Precursores de Proteínas/farmacología , Secuencia de Aminoácidos , Animales , Benzodiazepinonas/farmacología , Mucosa Gástrica/metabolismo , Gastrinas/química , Liberación de Histamina/efectos de los fármacos , Humanos , Masculino , Oligopéptidos/farmacología , Compuestos de Fenilurea/farmacología , Precursores de Proteínas/química , Ratas , Ratas Wistar , Receptor de Colecistoquinina B , Receptores de Colecistoquinina/antagonistas & inhibidores , Estómago/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA