Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 278
Filtrar
1.
Acta Histochem ; 122(8): 151650, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33161374

RESUMEN

Huntingtin-associated protein 1 (HAP1) is a neuronal cytoplasmic protein that is predominantly expressed in the brain and spinal cord. In addition to the central nervous system, HAP1 is also expressed in the peripheral organs including endocrine system. Different types of enteroendocrine cells (EEC) are present in the digestive organs. To date, the characterization of HAP1-immunoreactive (ir) cells remains unreported there. In the present study, the expression of HAP1 in pyloric stomach in adult male rats and its relationships with different chemical markers for EEC [gastrin, marker of gastrin (G) cells; somatostatin, marker of delta (D) cells; 5-HT, marker of enterochromaffin (EC) cells; histamine, marker of enterochromaffin-like (ECL) cells] were examined employing single- or double-labelled immunohistochemistry and with light-, fluorescence- or electron-microscopy. HAP1-ir cells were abundantly expressed in the glandular mucosa but were very few or none in the surface epithelium. Double-labelled immunofluorescence staining for HAP1 and markers for EECs showed that almost all the G-cells expressed HAP1. In contrast, HAP1 was completely lacking in D-cells, EC-cells or ECL-cells. Our current study is the first to clarify that HAP1 is selectively expressed in G-cells in rat pyloric stomach, which probably reflects HAP1's involvement in regulation of the secretion of gastrin.


Asunto(s)
Células Enterocromafines/metabolismo , Células Similares a las Enterocromafines/metabolismo , Mucosa Gástrica/metabolismo , Proteínas del Tejido Nervioso/genética , Píloro/metabolismo , Células Secretoras de Somatostatina/metabolismo , Animales , Biomarcadores/metabolismo , Células Enterocromafines/citología , Células Similares a las Enterocromafines/citología , Mucosa Gástrica/citología , Gastrinas/biosíntesis , Expresión Génica , Histamina/biosíntesis , Inmunohistoquímica , Masculino , Proteínas del Tejido Nervioso/metabolismo , Especificidad de Órganos , Píloro/citología , Ratas , Ratas Wistar , Somatostatina/biosíntesis , Células Secretoras de Somatostatina/citología
2.
Int J Mol Sci ; 20(20)2019 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-31623145

RESUMEN

The use of proton pump inhibitors (PPIs) over the last 30 years has rapidly increased both in the United States and worldwide. PPIs are not only very widely used both for approved indications (peptic ulcer disease, gastroesophageal reflux disease (GERD), Helicobacter pylori eradication regimens, stress ulcer prevention), but are also one of the most frequently off-label used drugs (25-70% of total). An increasing number of patients with moderate to advanced gastroesophageal reflux disease are remaining on PPI indefinitely. Whereas numerous studies show PPIs remain effective and safe, most of these studies are <5 years of duration and little data exist for >10 years of treatment. Recently, based primarily on observational/epidemiological studies, there have been an increasing number of reports raising issues about safety and side-effects with very long-term chronic treatment. Some of these safety issues are related to the possible long-term effects of chronic hypergastrinemia, which occurs in all patients taking chronic PPIs, others are related to the hypo-/achlorhydria that frequently occurs with chronic PPI treatment, and in others the mechanisms are unclear. These issues have raised considerable controversy in large part because of lack of long-term PPI treatment data (>10-20 years). Zollinger-Ellison syndrome (ZES) is caused by ectopic secretion of gastrin from a neuroendocrine tumor resulting in severe acid hypersecretion requiring life-long antisecretory treatment with PPIs, which are the drugs of choice. Because in <30% of patients with ZES, a long-term cure is not possible, these patients have life-long hypergastrinemia and require life-long treatment with PPIs. Therefore, ZES patients have been proposed as a good model of the long-term effects of hypergastrinemia in man as well as the effects/side-effects of very long-term PPI treatment. In this article, the insights from studies on ZES into these controversial issues with pertinence to chronic PPI use in non-ZES patients is reviewed, primarily concentrating on data from the prospective long-term studies of ZES patients at NIH.


Asunto(s)
Gastrinas/biosíntesis , Inhibidores de la Bomba de Protones/uso terapéutico , Gastropatías/tratamiento farmacológico , Gastropatías/etiología , Síndrome de Zollinger-Ellison/complicaciones , Síndrome de Zollinger-Ellison/metabolismo , Animales , Carcinoma Neuroendocrino , Enfermedad Crónica , Gastrinoma/metabolismo , Humanos , Inhibidores de la Bomba de Protones/efectos adversos , Factores de Riesgo , Gastropatías/metabolismo , Factores de Tiempo , Resultado del Tratamiento
3.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 33(11): 1557-1561, 2017 Nov.
Artículo en Chino | MEDLINE | ID: mdl-29268861

RESUMEN

Objective To investigate the expressions and correlations of gastrin and apoptosis-associated proteins involved in mitochondrial apoptotic pathway in gastric cancer tissues, and explore their clinicopathological characteristics. Methods The tissue chip technology and immunohistochemistry (IHC) were used to detect the expressions of gastrin and apoptosis-associated proteins (Bcl2, caspase-9 and caspase-3) in human gastric cancer tissues and their paracancerous tissues. The correlations of these markers and their clinicopathological characteristics were analyzed using Spearman rank correlation analysis and Chi-square test. Results The expressions of gastrin and Bcl2 in gastric cancer tissues were significantly higher than those in the corresponding paracancerous tissues, whereas the expressions of caspase-9 and caspase-3 in gastric cancer tissues was significantly lower than those in the corresponding tissues. There was a significant positive correlation between the expressions of gastrin and Bcl2 in gastric cancer tissues (r=0.237). The expression of gastrin was associated with tumor position, and the expression of Bcl2 was associated with tumor size, TNM stage and lymph node metastasis. Conclusion Gastrin and Bcl2 are highly expressed in gastric cancer tissues, and they are correlated with the clinicopathologic features.


Asunto(s)
Gastrinas/biosíntesis , Mitocondrias/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Transducción de Señal , Neoplasias Gástricas/metabolismo , Adulto , Anciano , Biomarcadores de Tumor/biosíntesis , Caspasa 3/biosíntesis , Caspasa 9/biosíntesis , Femenino , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Neoplasias Gástricas/patología
4.
Pancreas ; 45(7): 947-52, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-26684862

RESUMEN

OBJECTIVES: Pancreatic neuroendocrine tumors (PNETs) are known to have heterogeneity in terms of their ability to produce multiple hormones. The aim of this study was to evaluate the heterogeneity of PNETs from the viewpoint of hormonal expression. METHODS: The expressions of 4 representative hormones, gastrin, insulin, glucagon, and somatostatin, in both primary and metastatic lesions, were analyzed by immunohistochemical staining in 20 patients with metastatic PNETs (6 gastrinomas, 1 insulinoma, 1 glucagonoma, and 12 nonfunctioning PNETs [NF-PNETs]). Metastatic sites included lymph nodes in all 20 patients and liver metastasis in 7 patients (2 gastrinomas and 5 NF-PNETs). RESULTS: There were 6 PNETs with multiple hormone secretion (30%), and positive expression of 1 or more hormones was found in 9 of 12 patients whose primary tumors were diagnosed as NF-PNETs. The positive concordance rate of the hormonal expression pattern between primary tumors and metastatic lymph nodes and between primary tumors and hepatic metastasis were 50% and 11%, respectively. Three patients had metastatic lesions with positive hormonal expression, whereas their primary tumors were negative. CONCLUSIONS: Hormonal expressions are often different between the primary tumors and metastatic sites of PNETs.


Asunto(s)
Gastrinas/biosíntesis , Glucagón/biosíntesis , Insulina/biosíntesis , Tumores Neuroendocrinos/metabolismo , Neoplasias Pancreáticas/metabolismo , Somatostatina/biosíntesis , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Metástasis de la Neoplasia , Tumores Neuroendocrinos/patología , Neoplasias Pancreáticas/patología
5.
Aliment Pharmacol Ther ; 42(6): 649-63, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26177572

RESUMEN

BACKGROUND: Proton pump inhibitors (PPIs) have a well-established safety profile. However, concerns have been raised about a potential relationship between PPI-induced hypergastrinaemia and the development of enterochromaffin-like (ECL) cell hyperplasia, neuroendocrine tumours and gastric cancer during long-term therapy. AIM: To review the effects of long-term PPI use on serum gastrin levels and gastric histopathology. METHODS: A systematic literature search was conducted in PubMed on 21 April 2015 to identify studies reporting the effects of long-term (defined as >3 years) PPI use on gastrin levels and gastric histopathology. RESULTS: A total of 16 studies (1920 patients) met the inclusion criteria. During long-term PPI therapy, mean gastrin levels rose to one to three times the upper limit of the normal range (~100 pg/mL), and an increased prevalence of ECL cell hyperplasia was observed (+7.8-52.0%). Helicobacter pylori-positive patients had a significantly increased risk of developing ECL linear/micronodular hyperplasia compared with H. pylori-negative patients [OR: 2.45 (95% CI: 1.47-4.10), P = 0.0006]; however, no evidence of neoplastic changes was found. The risk of corpus atrophy was markedly higher in H. pylori-positive patients than in H. pylori-negative patients [OR: 11.45 (95% CI: 6.25-20.99), P < 0.00001]. Not a single case of gastric adenocarcinoma was found. CONCLUSIONS: Long-term PPI therapy induced moderate hypergastrinaemia in most patients and an increased prevalence of ECL cell hyperplasia. H. pylori-positive patients receiving long-term PPI therapy were exposed to a higher risk of corpus atrophy than H. pylori-negative patients. No neuroendocrine tumours or gastric cancers were found.


Asunto(s)
Células Similares a las Enterocromafines/patología , Gastrinas/sangre , Hiperplasia/inducido químicamente , Inhibidores de la Bomba de Protones/administración & dosificación , Inhibidores de la Bomba de Protones/efectos adversos , Esquema de Medicación , Gastrinas/biosíntesis , Infecciones por Helicobacter/tratamiento farmacológico , Helicobacter pylori/aislamiento & purificación , Humanos , Tumores Neuroendocrinos/inducido químicamente , Tumores Neuroendocrinos/patología , Factores de Riesgo , Neoplasias Gástricas/inducido químicamente , Neoplasias Gástricas/patología
6.
J Pharm Pharmacol ; 67(1): 133-41, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25213103

RESUMEN

OBJECTIVES: The aim of this study was to extend our knowledge about the mechanism involved in the gastroprotective effect of P1G10, a proteolytic fraction rich in cysteine proteinases from Vasconcellea cundinamarcensis (syn. Carica candamarcensis) latex, which demonstrated gastric healing and protection activities in rats. METHODS: Wistar rats were submitted to gastric lesions by indomethacin and treated with P1G10 (10 mg/kg). Free thiol groups and prostaglandin E2 content were measured in gastric mucosal and gastrin levels in blood samples. To evaluate the participation of nitric oxide (NO) or proteolytic activity of P1G10 on its gastroprotective effect, animals were treated with an inhibitor of NO production (L-NAME) or the fraction inhibited by iodoacetamide, respectively. Gastric secretion study (acidity and pepsin activity) was also performed. KEY FINDINGS: P1G10 (10 mg/kg) inhibited the occurrence of gastric lesions by indomethacin, restored the free thiol groups content on gastric mucosa and increased moderately prostaglandin E2 levels (34%). Furthermore, the treatment decreased the gastrin levels (95%), suggesting a possible modulation of secretory activity. This effect was accordant with attenuation of gastric acidity (42%) and pepsin activity (69%) seen in animals subjected to pyloric ligation. The inhibition of NO production or the proteolytic activity of P1G10 does not affect the gastroprotective effect. CONCLUSIONS: These results can explain the gastroprotective activity of P1G10 and serve a basis for further studies of this active principle.


Asunto(s)
Carica , Proteasas de Cisteína/farmacología , Dinoprostona/metabolismo , Ácido Gástrico/metabolismo , Extractos Vegetales/farmacología , Compuestos de Sulfhidrilo/metabolismo , Animales , Femenino , Ácido Gástrico/química , Ácido Gástrico/fisiología , Mucosa Gástrica , Gastrinas/biosíntesis , Gastrinas/sangre , Indometacina/farmacología , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico/antagonistas & inhibidores , Ratas , Ratas Wistar
7.
Environ Toxicol ; 30(2): 129-36, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23765435

RESUMEN

Divalent lead ions (Pb(2+) ) are toxic environmental pollutants known to cause serious health problems in humans and animals. Absorption of Pb(2+) from air, water, and food takes place in the respiratory and digestive tracts. The ways in which absorbed Pb(2+) affects cell physiology are just beginning to be understood at the molecular level. Here, we used reverse transcription PCR and Western blotting to analyze cultures of human gastric carcinoma cells exposed to 10 µM lead nitrate. We found that Pb(2+) induces gastrin hormone gene transcription and translation in a time-dependent manner. Promoter deletion analysis revealed that activator protein 1 (AP1) was necessary for gastrin gene transcription in cells exposed to Pb(2+) . MitogIen-activated protein kinase (MAPK)/ERK kinase inhibitor PD98059 suppressed the Pb(2+) -induced increase in messenger RNA. Epidermal growth factor receptor (EGFR) inhibitors AG1478 and PD153035 reduced both transcription and phosphorylation by extracellular signal-regulated kinase (ERK1/2). Cells exposed to Pb(2+) also increased production of c-Jun protein, a component of AP1, and over-expression of c-Jun enhanced activation of the gastrin promoter. In sum, the findings suggest the EGFR-ERK1/2-AP1 pathway mediates the effects of Pb(2+) on gastrin gene activity in cell culture.


Asunto(s)
Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Gastrinas/biosíntesis , Gastrinas/genética , Regulación de la Expresión Génica/efectos de los fármacos , Plomo/toxicidad , Factor de Transcripción AP-1/efectos de los fármacos , Línea Celular Tumoral , Represión Epigenética/efectos de los fármacos , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/efectos de los fármacos , Humanos , Sistema de Señalización de MAP Quinasas , Fosforilación , Proteínas Proto-Oncogénicas c-jun/farmacología
8.
Invest Ophthalmol Vis Sci ; 55(3): 1965-75, 2014 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-24576871

RESUMEN

PURPOSE: Cholecystokinin (CCK) is a neuropeptide that has been identified in trigeminal ganglion neurons. Gastrin (GAST) is a related peptide never explored in the cornea. The presence and role of both gastrointestinal peptides in the cornea and corneal sensory neurons remain to be established. We explored here in mice whether CCK, GAST, and their receptors CCK1R and CCK2R are expressed in the corneal epithelium and trigeminal ganglion neurons innervating the cornea. METHODS: We used RT-PCR analysis to detect mRNAs of CCK, GAST, CCK1R, and CCK2R in mouse cornea epithelium, trigeminal ganglia, and primary cultured corneal epithelial cells. Immunofluorescence microscopy was used to localize these peptides and their receptors in the cornea, cultured corneal epithelial cells, and corneal nerves, as well as in the cell bodies of corneal trigeminal ganglion neurons identified by retrograde labeling with Fast Blue. RESULTS: Mouse corneal epithelial cells in the cornea in situ and in cell cultures expressed CCK and GAST. Only the receptor CCK2R was found in the corneal epithelium. In addition, mouse corneal afferent sensory neurons expressed CCK and GAST, and the CCK1R receptors. CONCLUSIONS: The presence of CCK, GAST, and their receptors in the mouse corneal epithelium, and in trigeminal ganglion neurons supplying sensory innervation to the cornea, opens the possibility that these neuropeptides are involved in corneal neurogenic inflammation and in the modulation of repairing/remodeling processes following corneal injury.


Asunto(s)
Colecistoquinina/genética , Córnea/metabolismo , Gastrinas/genética , Nervio Trigémino/metabolismo , Animales , Células Cultivadas , Colecistoquinina/biosíntesis , Córnea/inervación , Epitelio Corneal/citología , Epitelio Corneal/metabolismo , Gastrinas/biosíntesis , Regulación de la Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , ARN/genética , Receptor de Colecistoquinina B/biosíntesis , Receptor de Colecistoquinina B/genética , Receptores de Colecistoquinina/biosíntesis , Receptores de Colecistoquinina/genética
9.
Pathol Res Pract ; 209(10): 670-3, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23891137

RESUMEN

We report a rare case of an ovarian mucinous cystadenoma in which there were peculiar neuroendocrine micronests composed of gastrin-immunoreactive cells. There was no clinical evidence of hypergastrinemia. The mucinous component of the neoplasm was represented by columnar cells mostly expressing a gastric phenotype with MUC5AC and claudin 18 positivity, which was consistent with the presence of interspersed gastrin cells. The tumoral stroma displayed areas of luteinization with cells intensely positive for α-inhibin, MART-1 and calretinin.


Asunto(s)
Cistoadenoma Mucinoso/patología , Células Neuroendocrinas/patología , Neoplasias Ováricas/patología , Biomarcadores de Tumor/análisis , Cistoadenoma Mucinoso/metabolismo , Femenino , Gastrinas/biosíntesis , Humanos , Hiperplasia , Inmunohistoquímica , Persona de Mediana Edad , Células Neuroendocrinas/metabolismo , Neoplasias Ováricas/metabolismo
10.
Endocrinology ; 153(7): 3006-16, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22593272

RESUMEN

Gastrin and its precursors have been shown to promote mitogenesis and angiogenesis in gastrointestinal tumors. Hypoxia stimulates tumor growth, but its effect on gastrin gene regulation has not been examined in detail. Here we have investigated the effect of hypoxia on the transcription of the gastrin gene in human gastric cancer (AGS) cells. Gastrin mRNA was measured by real-time PCR, gastrin peptides were measured by RIA, and gastrin promoter activity was measured by dual-luciferase reporter assay. Exposure to a low oxygen concentration (1%) increased gastrin mRNA concentrations in wild-type AGS cells (AGS) and in AGS cells overexpressing the gastrin receptor (AGS-cholecystokinin receptor 2) by 2.1 ± 0.4- and 4.1 ± 0.3-fold (P < 0.05), respectively. The hypoxia mimetic, cobalt chloride (300 µM), increased gastrin promoter activity in AGS cells by 2.4 ± 0.3-fold (P < 0.05), and in AGS-cholecystokinin receptor 2 cells by 4.0 ± 0.3-fold (P < 0.05), respectively. The observations that either deletion from the gastrin promoter of the putative binding sites for the transcription factor hypoxia-inducible factor 1 (HIF-1) or knockdown of either the HIF-1α or HIF-1ß subunit did not affect gastrin promoter inducibility under hypoxia indicated that the hypoxic activation of the gastrin gene is likely HIF independent. Mutational analysis of previously identified Sp1 regulatory elements in the gastrin promoter also failed to abrogate the induction of promoter activity by hypoxia. The observations that hypoxia up-regulates the gastrin gene in AGS cells by HIF-independent mechanisms, and that this effect is enhanced by the presence of gastrin receptors, provide potential targets for gastrointestinal cancer therapy.


Asunto(s)
Cobalto/farmacología , Gastrinas/biosíntesis , Tracto Gastrointestinal/citología , Factor 1 Inducible por Hipoxia/fisiología , Hipoxia , Translocador Nuclear del Receptor de Aril Hidrocarburo/metabolismo , Línea Celular Tumoral , Neoplasias Colorrectales/metabolismo , Gastrinas/genética , Gastrinas/metabolismo , Neoplasias Gastrointestinales/metabolismo , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Modelos Biológicos , Regiones Promotoras Genéticas , ARN Mensajero/metabolismo , Factor de Transcripción Sp1/metabolismo
11.
Biochem Biophys Res Commun ; 420(1): 210-5, 2012 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-22426477

RESUMEN

Nonsteroidal anti-inflammatory drugs (NSAIDs) are valuable agents; however, their use has been limited by their association with mucosal damage in the upper gastrointestinal tract. NSAIDs inhibit cyclooxygenase and consequently block the synthesis of prostaglandins, which have cytoprotective effects in gastric mucosa; these effects on prostaglandins have been thought to be major cause of NSAID-induced ulceration. However, studies indicate that additional NSAID-related mechanisms are involved in formation of gastric lesions. Here, we used a toxicoproteomic approach to understand cellular processes that are affected by NSAIDs in mouse stomach tissue during ulcer formation. We used fluorogenic derivatization-liquid chromatography-tandem mass spectrometry (FD-LC-MS/MS)-which consists of fluorogenic derivatization, separation and fluorescence detection by LC, and identification by LC-tandem mass spectrometry-in this proteomic analysis of pyrolic stomach from control and diclofenac (Dic)-treated mice. FD-LC-MS/MS results were highly sensitive; 10 differentially expressed proteins were identified, and all 10 were more highly expressed in Dic-treated mice than in control mice. Specifically, expression levels of 78 kDa glucose-regulated protein (GRP78), heat shock protein beta-1 (HSP27), and gastrin were more than 3-fold higher in Dic-treated mice than in control mice. This study represents a first step to ascertain the precise actors of early NSAID-induced ulceration.


Asunto(s)
Antiinflamatorios no Esteroideos/efectos adversos , Inhibidores de la Ciclooxigenasa/efectos adversos , Mucosa Gástrica/efectos de los fármacos , Mucosa Gástrica/metabolismo , Proteómica/métodos , Úlcera Gástrica/inducido químicamente , Úlcera Gástrica/metabolismo , Animales , Antiinflamatorios no Esteroideos/administración & dosificación , Cromatografía Liquida/métodos , Inhibidores de la Ciclooxigenasa/administración & dosificación , Diclofenaco/administración & dosificación , Diclofenaco/efectos adversos , Modelos Animales de Enfermedad , Chaperón BiP del Retículo Endoplásmico , Colorantes Fluorescentes/análisis , Mucosa Gástrica/patología , Gastrinas/análisis , Gastrinas/biosíntesis , Proteínas de Choque Térmico HSP27/análisis , Proteínas de Choque Térmico HSP27/biosíntesis , Proteínas de Choque Térmico/análisis , Proteínas de Choque Térmico/biosíntesis , Masculino , Ratones , Ratones Endogámicos C57BL , Úlcera Gástrica/patología , Espectrometría de Masas en Tándem/métodos
12.
Cancer Prev Res (Phila) ; 5(4): 675-84, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22366915

RESUMEN

The most frequently occurring lesions in the colon are the hyperplastic polyps. Hyperplastic polyps have long been considered as lesions with no malignant potential and colonoscopy for these patients is not recommended. However, recent works suggest that hyperplastic polyps may represent precursor lesions of some sporadic colorectal cancers. Until now, no biomarker allows to identify the subset of hyperplastic polyps that may have a malignant potential. Because the hormone precursor progastrin has been involved in colon carcinogenesis, we investigated whether its expression in hyperplastic polyps predicts the occurrence of colonic neoplasm after resection of hyperplastic polyps. We retrospectively analyzed progastrin expression in hyperplastic polyps from 74 patients without history of colorectal pathology. In our study, 41% of patients presenting an initial hyperplastic polyp subsequently developed adenomatous polyps, recognized as precursor lesions for colorectal adenocarcinomas. Progastrin was overexpressed in the hyperplastic polyps in 40% of the patients. We showed a significant association between progastrin overexpression and shortened neoplasm-free survival (P = 0.001). Patients with high overexpression of progastrin had a 5-year neoplasm-free survival rate of 38% as compared with 100% for the patients with low progastrin expression. In addition, we established a predictive test on the basis of progastrin staining and patients' age that predicts occurrence of neoplasm after developing a first hyperplastic polyp with a sensitivity of 100% [95% confidence interval (CI), 79%-100%] and a specificity of 74% (51%-90%). We show that progastrin expression evaluation in hyperplastic polyps is an efficient prognostic tool to determine patients with higher risk of metachronous neoplasms who could benefit from an adapted follow-up.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias del Colon/genética , Neoplasias del Colon/metabolismo , Pólipos del Colon/metabolismo , Gastrinas/biosíntesis , Precursores de Proteínas/biosíntesis , Adulto , Anciano , Anciano de 80 o más Años , Estudios de Cohortes , Supervivencia sin Enfermedad , Femenino , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Curva ROC , Reproducibilidad de los Resultados
13.
Virchows Arch ; 459(2): 147-54, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21739268

RESUMEN

We recently identified the transcription factor (TF) islet 1 gene product (ISL1) as a marker for well-differentiated pancreatic neuroendocrine tumors (P-NETs). In order to better understand the expression of the four TFs, ISL1, pancreatico-duodenal homeobox 1 gene product (PDX1), neurogenin 3 gene product (NGN3), and CDX-2 homeobox gene product (CDX2), that mainly govern the development and differentiation of the pancreas and duodenum, we studied their expression in hormonally defined P-NETs and duodenal (D-) NETs. Thirty-six P-NETs and 14 D-NETs were immunostained with antibodies against the four pancreatic hormones, gastrin, serotonin, calcitonin, ISL1, PDX1, NGN3, and CDX2. The TF expression pattern of each case was correlated with the tumor's hormonal profile. Insulin-positive NETs expressed only ISL1 (10/10) and PDX1 (9/10). Glucagon-positive tumors expressed ISL1 (7/7) and were almost negative for the other TFs. Gastrin-positive NETs, whether of duodenal or pancreatic origin, frequently expressed PDX1 (17/18), ISL1 (14/18), and NGN3 (14/18). CDX2 was mainly found in the gastrin-positive P-NETs (5/8) and rarely in the D-NETs (1/10). Somatostatin-positive NETs, whether duodenal or pancreatic in origin, expressed ISL1 (9/9), PDX1 (3/9), and NGN3 (3/9). The remaining tumors showed labeling for ISL1 in addition to NGN3. There was no association between a particular TF pattern and NET features such as grade, size, location, presence of metastases, and functional activity. We conclude from our data that there is a correlation between TF expression patterns and certain hormonally defined P-NET and D-NET types, suggesting that most of the tumor types originate from embryologically determined precursor cells. The observed TF signatures do not allow us to distinguish P-NETs from D-NETs.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/biosíntesis , Neoplasias Duodenales/metabolismo , Proteínas de Homeodominio/biosíntesis , Proteínas del Tejido Nervioso/biosíntesis , Tumores Neuroendocrinos/metabolismo , Neoplasias Pancreáticas/metabolismo , Transactivadores/biosíntesis , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor/análisis , Factor de Transcripción CDX2 , Calcitonina/biosíntesis , Neoplasias Duodenales/patología , Femenino , Gastrinas/biosíntesis , Glucagón/biosíntesis , Humanos , Inmunohistoquímica , Insulina/biosíntesis , Proteínas con Homeodominio LIM , Masculino , Persona de Mediana Edad , Tumores Neuroendocrinos/patología , Neoplasias Pancreáticas/patología , Polipéptido Pancreático/biosíntesis , Somatostatina/biosíntesis , Factores de Transcripción , Adulto Joven
14.
Immunopharmacol Immunotoxicol ; 33(1): 84-9, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20370597

RESUMEN

Gastric mucosa is one of the most vulnerable tissues in human and animal. However, little is known about the effects of calcitonin gene-related peptide (CGRP) on gastric mucosa injuries induced by gastric ischemia reperfusion. The purpose of the present study was to investigate the protective effects and mechanism of CGRP on gastric mucosa injury after gastric ischemia reperfusion in rats. Thirty-six healthy Wistar rats were randomly divided into CGRP-treated, sham-operated, and control groups. Twelve rats were involved in each group. These groups were further divided into 24-h and 48-h subgroups. Gastric ischemia reperfusion injury (GI-RI) rat model was established by a 30-min celiac artery occlusion by an artery clamp, followed by 24 h or 48 h of reperfusion. CGRP (1 µg/ml) at the dose of 3 µg/kg was given intraperiloneally (IP) at the beginning of reperfusion for rats in CGRP-treated group. Saline as vehicle (3 ml/kg body weight), IP, was administered at the beginning of reperfusion for rats in control group. Sham-operated animals were subjected to an operation without GI-RI. Twenty-four hours or 48 h after operation, the samples were taken out and processed for calculating stomach mucous membrane damage index according to Guth method, detecting pathological changes of gastric mucosa tissue by light microscopy and observing the expression of gastrin (Gas) and somatostatin (SST) by immunohistochemical staining. The results showed the following: (i) gastric mucosa with diffuse edema, splinter hemorrhage and erosion, numerous endothelial cells necrosis, mucosa dissociation, and infiltration of inflammatory cells were observed in both control and CGRP-treated animals, especially in the earlier period (24 h) and then gradually healing. CGRP administration could reduce the damage of gastric mucosa. The injury index of gastric mucosa was lower in CGRP-treated group as compared with that in control group (P < 0.01). (ii) Gas expression in gastric antrum mucosa was lower in CGRP-treated group than that in control group (P < 0.01). SST expression in gastric antrum mucosa was higher in CGRP-treated group than that in control group (P < 0.01). It is concluded that CGRP regulated the secretion of Gas and SST and thus alleviated the damage of gastric mucosa induced by ischemia and reperfusion. CGRP might be a potential candidate for clinical therapy on modulating gastric mucosal protection and maintaining gastric mucosal integrity after ischemia and reperfusion of the stomach.


Asunto(s)
Péptido Relacionado con Gen de Calcitonina/uso terapéutico , Mucosa Gástrica/efectos de los fármacos , Daño por Reperfusión/prevención & control , Vasodilatadores/uso terapéutico , Animales , Modelos Animales de Enfermedad , Mucosa Gástrica/irrigación sanguínea , Mucosa Gástrica/patología , Gastrinas/biosíntesis , Gastrinas/metabolismo , Inmunohistoquímica , Masculino , Ratas , Ratas Wistar , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , Somatostatina/biosíntesis , Somatostatina/metabolismo
15.
Helicobacter ; 15(5): 438-48, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21083750

RESUMEN

BACKGROUND: Helicobacter pylori (H. pylori) infection has been linked to the development of chronic gastritis, duodenal ulcer disease, and gastric cancer. Helicobacter pylori- infected patients and animal models develop hypergastrinemia, chronic gastritis, and gastric atrophy. Since gastrin is an important regulator of gastric acid secretion and cell growth, H. pylori regulation of this hormone has been implicated in its pathogenesis. OBJECTIVES: To investigate the effect of H. pylori on gastrin gene expression in mice and of human bacterial isolates on gastrin mRNA expressed in a human cell line. METHODS: Gastrin mRNA was measured by qRT-PCR in H. pylori-infected mice. H. pylori were co-cultured with AGS cells to study regulation of human gastrin gene expression. Various MAP kinases were implicated in signal transduction from the bacteria using specific inhibitors. Gastrin reporter constructs and gel shift assays were used to map DNA responsive elements. RESULTS: In addition to an increase in gastrin mRNA in H. pylori-infected mice, H. pylori induced the endogenous human gastrin gene through MAP kinase-dependent signaling but not NFκB-dependent signaling. Activation of gastrin through MAPK signaling did not require CagA or VacA virulence factors. Transfection studies demonstrated that a GC-rich motif mediated H. pylori-induction of the gastrin promoter and that the motif inducibly binds Sp1 and Sp3 transcription factors. CONCLUSIONS: Direct contact of live H. pylori bacteria with human cells is sufficient to induce gastrin gene expression.


Asunto(s)
Gastrinas/biosíntesis , Expresión Génica , Infecciones por Helicobacter/patología , Helicobacter pylori/patogenicidad , Regiones Promotoras Genéticas , Animales , Fusión Artificial Génica , Línea Celular Tumoral , Modelos Animales de Enfermedad , Ensayo de Cambio de Movilidad Electroforética , Perfilación de la Expresión Génica , Genes Reporteros , Infecciones por Helicobacter/microbiología , Humanos , Ratones , Ratones Endogámicos C57BL , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
16.
Zhonghua Zhong Liu Za Zhi ; 32(7): 501-6, 2010 Jul.
Artículo en Chino | MEDLINE | ID: mdl-21029692

RESUMEN

OBJECTIVE: To study the regulatory effect of Helicobacter pylori CagA protein on gastrin promoter and the related signaling pathways as to further elucidate the mechanism of the development and progression of human gastric carcinoma. METHODS: After pcDNA3.1ZEO(-)/CagAand PGL/GP were identified by double restriction enzyme digestion, PCR and sequencing, the gastric cancer cell lines AGS and SGC-7901 cells were co-transfected with pcDNA3.1ZEO(-)/CagA and PGL/GP for 48 h. Alternatively, AGS and SGC-7901 cells were transfected by PGL/GP for 36 h later, and infected with Helicobacter pylori for additional 12 h. Meanwhile, the transfected and infected cells were treated using the JAK2 signaling pathway inhibitor AG490 and the ERK signaling pathway inhibitor U0126. The untreated cells and empty-vector-transfected cells were used as the control. Finally, luciferase activity was detected using the luciferase reporter assay system in transfected and infected cells. The levels of gastrin mRNA was determined by TaqMan® real-time quantitative PCR. RESULTS: After co-transfection with pcDNA3.1ZEO(-)/CagA and PGL/GP, the activities of luciferase were increased by 251.3, 106.1 and 2.4 times in AGS cells and 35.8, 22.7 and 13.4 times in SGC-7901 cells, respectively, as compared with that of the control, pcDNA3.1 ZEO(-)/CagA + PGL3/Basic and pcDNA3.1 ZEO(-) + PGL/GP groups. The activities of luciferase in PGL/GP transfection and HP infection group were also increased by 1673.2, 33.5, 1.4 times in AGS cells and 1180.2, 72.2 and 1.5 times in SGC-7901 cells, respectively, as compared with that of the control, PGL3/Basic + HP and PGL/GP groups. There were statistically significant differences between them (P < 0.05), which suggested that the transcription activity of gastrin promoter increased significantly. But after adding the inhibitor AG490 and U0126, respectively, the activities of luciferase were significantly decreased by 95.7% (U0126) and 33.0% (AG490) in co-transfected AGS cells and 94.8% (U0126) and 86.2% (AG490) in co-transfected SGC-7901 cells with pcDNA3.1ZEO(-)/CagA and PGL/GP (P < 0.05). In the PGL/GP transfection and HP infection group, the activities of luciferase were significantly decreased by 24.6% (U0126) and 25.8% (AG490) in AGS cells and 57.3% (U0126) and 14.1% (AG490) after adding the inhibitor AG490 and U0126, respectively (P < 0.05). The results showed that the gastrin promoter activities were significantly inhibited. The gastrin mRNA levels were 3.0 and 4.5 times higher in HP-infected AGS and SGC-7901 cells, respectively, than that in the control groups. In the cells transfected with pcDNA3.1ZEO(-)/CagA, the gastrin mRNA levels were raised 10.8 and 2.3 times (AGS cells) and 10.9 and 16.2 times (SGC-7901 cells), respectively, as compared with that of control and pcDNA3.1ZEO(-) groups. All of the differences were statistically significant (P < 0.05). CONCLUSION: These results suggest that CagA may activate the gastrin promoter and up-regulate the expression of gastrin gene, and CagA is one of the important proteins in regulating gastrin gene expression. The ERK/MAPK and JAK/STAT signaling pathways may be involved in the controlling of gastrin gene expression by CagA.


Asunto(s)
Antígenos Bacterianos/metabolismo , Proteínas Bacterianas/metabolismo , Gastrinas/biosíntesis , Infecciones por Helicobacter , Neoplasias Gástricas/metabolismo , Antígenos Bacterianos/genética , Antineoplásicos/farmacología , Proteínas Bacterianas/genética , Butadienos/farmacología , Línea Celular Tumoral , Inhibidores Enzimáticos/farmacología , Gastrinas/genética , Regulación Neoplásica de la Expresión Génica , Vectores Genéticos , Helicobacter pylori/aislamiento & purificación , Humanos , Nitrilos/farmacología , Regiones Promotoras Genéticas , ARN Mensajero/metabolismo , Neoplasias Gástricas/patología , Neoplasias Gástricas/virología , Transfección , Tirfostinos/farmacología , Regulación hacia Arriba
17.
FEBS Lett ; 584(21): 4413-8, 2010 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-20932834

RESUMEN

The involvement of the gastrointestinal hormone gastrin in the development of gastrointestinal cancer is highly controversial. Here we demonstrate a positive-feedback loop whereby gastrin, acting via the CCK2 receptor, increases its own expression. Such an autocrine loop has not previously been reported for any other gastrointestinal hormone. Gastrin promoter activation was dependent on the MAP kinase pathway and did not involve Sp1 binding sites or epidermal growth factor receptor transactivation. As the treatment of gastrointestinal cancer cells with amidated gastrin led to increased expression of non-amidated gastrins, the positive-feedback loop may contribute to the sustained increase in circulating gastrins observed in colorectal cancer patients.


Asunto(s)
Gastrinas/biosíntesis , Gastrinas/metabolismo , Neoplasias Gastrointestinales/patología , Receptor de Colecistoquinina B/metabolismo , Secuencia de Bases , Línea Celular Tumoral , Neoplasias Colorrectales/sangre , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Retroalimentación Fisiológica , Gastrinas/sangre , Gastrinas/genética , Neoplasias Gastrointestinales/genética , Neoplasias Gastrointestinales/metabolismo , Regulación Neoplásica de la Expresión Génica , Genes Reporteros/genética , Humanos , Luciferasas/genética , Sistema de Señalización de MAP Quinasas , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa , Regiones Promotoras Genéticas/genética , Factores de Tiempo , Transcripción Genética , Activación Transcripcional
19.
Gut ; 59(8): 1037-45, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20584780

RESUMEN

BACKGROUND AND AIMS: Helicobacter pylori (H pylori) infection is a major risk factor in the development of distal gastric adenocarcinoma. Development of the invasive phenotype is associated with the phenomenon of epithelial:mesenchymal transition (EMT). Soluble heparin-binding epidermal growth factor (HB-EGF) has been implicated in this process. A study was undertaken to investigate the possibility that matrix metalloproteinase (MMP)-7 is upregulated in H pylori infection as a result of hypergastrinaemia, which may enhance shedding of HB-EGF and contribute towards EMT in gastric adenocarcinoma cell lines. METHODS: Three gastric epithelial cell lines (AGS, MGLVA1 and ST16) were co-cultured with the pathogenic H pylori strain 60190 and non-pathogenic strain Tx30a in an in vitro infection model. Gene expression was quantified by real-time PCR, HB-EGF shedding by ELISA and protein expression by immunofluorescence or immunohistochemistry. The INS-GAS mouse, a transgenic mouse model of gastric carcinogenesis which overexpresses amidated gastrin, was used to investigate the in vivo relationship between HB-EGF, MMP-7, gastrin and EMT. RESULTS: The pathogenic strain of H pylori significantly upregulated EMT-associated genes Snail, Slug and vimentin in all three gastric cell lines to a greater degree than the non-pathogenic strain. Pathogenic H pylori also upregulated HB-EGF shedding, a factor implicated in EMT, which was partially dependent on both gastrin and MMP-7 expression. Gastrin and MMP-7 siRNAs and MMP-7 neutralising antibody significantly reduced upregulation of HB-EGF shedding in H pylori infected gastric cell lines and reduced EMT gene expression. The effect of H pylori on EMT was also reversed by gastrin siRNA. Neutralisation of gastrin in the INS-GAS mouse model reduced expression of MMP-7, HB-EGF and key EMT proteins. CONCLUSION: The upregulation of MMP-7 by pathogenic H pylori is partially dependent on gastrin and may have a role in the development of gastric cancer, potentially through EMT, by indirectly increasing levels of soluble HB-EGF.


Asunto(s)
Infecciones por Helicobacter/metabolismo , Helicobacter pylori/patogenicidad , Proteínas de Neoplasias/biosíntesis , Neoplasias Gástricas/microbiología , Animales , Transformación Celular Neoplásica/genética , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Células Epiteliales/patología , Gastrinas/biosíntesis , Gastrinas/genética , Gastrinas/fisiología , Regulación Neoplásica de la Expresión Génica , Infecciones por Helicobacter/patología , Factor de Crecimiento Similar a EGF de Unión a Heparina , Humanos , Péptidos y Proteínas de Señalización Intercelular/biosíntesis , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/fisiología , Metaloproteinasa 7 de la Matriz/biosíntesis , Metaloproteinasa 7 de la Matriz/genética , Metaloproteinasa 7 de la Matriz/fisiología , Células Madre Mesenquimatosas/patología , Ratones , Ratones Transgénicos , Proteínas de Neoplasias/genética , ARN Interferente Pequeño/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología , Células Tumorales Cultivadas , Regulación hacia Arriba , Virulencia
20.
Surg Today ; 40(3): 267-71, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20180083

RESUMEN

We report the case of a patient demonstrating multiple gastric carcinoids with hypergastrinemia. A 50-year-old Japanese woman was admitted to our hospital for the further examination of multiple carcinoids of the stomach with hypergastrinemia, although she was asymptomatic. However, based on our clinical examination, this case seemed to be neither type I nor II carcinoid. We performed a total gastrectomy with D1 lymph node dissection. A pathological examination showed numerous endocrine micronests, hyperplasia of the parietal cells extending to the foveolar neck region, and numerous dilated oxyntic glands filled with eosinophilic secretions. Many parietal cells exhibited vacuolated cytoplasms and apical snouts. Furthermore, the dilated glands at the base of the mucosa had hyperchromatic nuclei and ciliated surfaces. The postoperative serum gastrin level was soon normalized to 47 pg/ml. This is only the third reported case of multiple gastric carcinoids with hypergastrinemia due to an intrinsic abnormality in the acid secretion of the parietal cells.


Asunto(s)
Tumor Carcinoide/metabolismo , Gastrinas/biosíntesis , Neoplasias Gástricas/metabolismo , Tumor Carcinoide/diagnóstico , Tumor Carcinoide/patología , Femenino , Gastrectomía , Ácido Gástrico/metabolismo , Gastrinas/sangre , Humanos , Escisión del Ganglio Linfático , Persona de Mediana Edad , Células Parietales Gástricas/patología , Neoplasias Gástricas/diagnóstico , Neoplasias Gástricas/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA