Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Gastroenterology ; 140(3): 879-91, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21111741

RESUMEN

BACKGROUND & AIMS: Epigenetic alterations have been correlated with field cancerization in human patients, but evidence from experimental models that specific epigenetic changes can initiate cancer has been lacking. Although hormones have been associated with cancer risk, the mechanisms have not been determined. The peptide hormone gastrin exerts a suppressive effect on antral gastric carcinogenesis. METHODS: N-methyl-N-nitrosourea (MNU)-dependent gastric cancer was investigated in hypergastrinemic (INS-GAS), gastrin-deficient (GAS(-/-)), Tff1-deficient (Tff1(+/-)), and wild-type (WT) mice. Epigenetic alterations of the trefoil factor 1 (TFF1) tumor suppressor gene were evaluated in vitro and in vivo. RESULTS: Human intestinal-type gastric cancers in the antrum exhibited progressive TFF1 repression and promoter hypermethylation. Mice treated with MNU exhibited a field defect characterized by widespread Tff1 repression associated with histone H3 lysine 9 methylation and H3 deacetylation at the Tff1 promoter in epithelial cells. In MNU-induced advanced cancers, DNA methylation at the Tff1 promoter was observed. Tumor induction and Tff1 repression were increased in MNU-treated mice by Helicobacter infection. Hypergastrinemia suppressed MNU-dependent tumor initiation and progression in a manner that correlated with gene silencing and epigenetic alterations of Tff1. In contrast, homozygous gastrin-deficient and heterozygous Tff1-deficient mice showed enhanced MNU-dependent field defects and cancer initiation compared with WT mice. In gastric cancer cells, gastrin stimulation partially reversed the epigenetic silencing in the TFF1 promoter. CONCLUSIONS: Initiation of antral gastric cancer is associated with progressive epigenetic silencing of TFF1, which can be suppressed by the hormone gastrin.


Asunto(s)
Transformación Celular Neoplásica/genética , Gastrinas/metabolismo , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Péptidos/genética , Neoplasias Gástricas/prevención & control , Proteínas Supresoras de Tumor/genética , Adulto , Anciano , Anciano de 80 o más Años , Animales , Apoptosis , Línea Celular Tumoral , Proliferación Celular , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Ensamble y Desensamble de Cromatina , Metilación de ADN , Modelos Animales de Enfermedad , Femenino , Gastrinas/deficiencia , Gastrinas/genética , Infecciones por Helicobacter/genética , Infecciones por Helicobacter/metabolismo , Infecciones por Helicobacter/microbiología , Helicobacter felis/patogenicidad , Histonas/metabolismo , Humanos , Masculino , Metilnitrosourea , Ratones , Ratones Noqueados , Persona de Mediana Edad , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Péptidos/deficiencia , Regiones Promotoras Genéticas , ARN Mensajero/metabolismo , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/microbiología , Neoplasias Gástricas/patología , Factores de Tiempo , Transfección , Factor Trefoil-1 , Proteínas Supresoras de Tumor/metabolismo
2.
J Biol Chem ; 285(39): 29787-96, 2010 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-20639202

RESUMEN

The adenosine A(2A) receptor (A(2A)R) is increasingly recognized as a novel therapeutic target in Parkinson disease. In striatopallidal neurons, the G-protein α(olf) subtype is required to couple this receptor to adenylyl cyclase activation. It is now well established that the ßγ dimer also performs an active role in this signal transduction process. In principal, sixty distinct ßγ dimers could arise from combinatorial association of the five known ß and 12 γ subunit genes. However, key questions regarding which ßγ subunit combinations exist and whether they perform specific signaling roles in the context of the organism remain to be answered. To explore these questions, we used a gene targeting approach to specifically ablate the G-protein γ(7) subtype. Revealing a potentially new signaling paradigm, we show that the level of the γ(7) protein controls the hierarchial assembly of a specific G-protein α(olf)ß(2)γ(7) heterotrimer in the striatum. Providing a probable basis for the selectivity of receptor signaling, we further demonstrate that loss of this specific G-protein heterotrimer leads to reduced A(2A)R activation of adenylyl cyclase. Finally, substantiating an important role for this signaling pathway in pyschostimulant responsiveness, we show that mice lacking the G-protein γ(7) subtype exhibit an attenuated behavioral response to caffeine. Collectively, these results further support the A(2A)R G-protein α(olf)ß(2)γ(7) interface as a possible therapeutic target for Parkinson disease.


Asunto(s)
Cuerpo Estriado/metabolismo , Subunidades gamma de la Proteína de Unión al GTP/metabolismo , Receptor de Adenosina A2A/metabolismo , Transducción de Señal/fisiología , Animales , Conducta Animal/efectos de los fármacos , Conducta Animal/fisiología , Cafeína/farmacología , Estimulantes del Sistema Nervioso Central/farmacología , Subunidades alfa de la Proteína de Unión al GTP/genética , Subunidades alfa de la Proteína de Unión al GTP/metabolismo , Subunidades beta de la Proteína de Unión al GTP/genética , Subunidades beta de la Proteína de Unión al GTP/metabolismo , Subunidades gamma de la Proteína de Unión al GTP/genética , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Complejos Multienzimáticos/genética , Complejos Multienzimáticos/metabolismo , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/terapia , Receptor de Adenosina A2A/genética , Transducción de Señal/efectos de los fármacos
3.
Am J Physiol Gastrointest Liver Physiol ; 300(2): G334-44, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21051525

RESUMEN

Gastrin is secreted from a subset of neuroendocrine cells residing in the gastric antrum known as G cells, but low levels are also expressed in fetal pancreas and intestine and in many solid malignancies. Although past studies have suggested that antral gastrin is transcriptionally regulated by inflammation, gastric pH, somatostatin, and neoplastic transformation, the transcriptional regulation of gastrin has not previously been demonstrated in vivo. Here, we describe the creation of an enhanced green fluorescent protein reporter (mGAS-EGFP) mouse using a bacterial artificial chromosome that contains the entire mouse gastrin gene. Three founder lines expressed GFP signals in the gastric antrum and the transitional zone to the corpus. In addition, GFP(+) cells could be detected in the fetal pancreatic islets and small intestinal villi, but not in these organs of the adult mice. The administration of acid-suppressive reagents such as proton pump inhibitor omeprazole and gastrin/CCK-2 receptor antagonist YF476 significantly increased GFP signal intensity and GFP(+) cell numbers in the antrum, whereas these parameters were decreased by overnight fasting, octreotide (long-lasting somatostatin ortholog) infusion, and Helicobacter felis infection. GFP(+) cells were also detected in the anterior lobe of the pituitary gland and importantly in the colonic tumor cells induced by administration with azoxymethane and dextran sulfate sodium salt. This transgenic mouse provides a useful tool to study the regulation of mouse gastrin gene in vivo, thus contributing to our understanding of the mechanisms involved in transcriptional control of the gastrin gene.


Asunto(s)
Cromosomas Artificiales Bacterianos/genética , Células Secretoras de Gastrina/metabolismo , Gastrinas/genética , Proteínas Fluorescentes Verdes/genética , Helicobacter felis/genética , Envejecimiento/metabolismo , Animales , Azoximetano , Carcinógenos , Neoplasias del Colon/inducido químicamente , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Sulfato de Dextran , Regulación hacia Abajo , Ayuno , Feto/metabolismo , Ácido Gástrico/metabolismo , Gastrinas/deficiencia , Regulación del Desarrollo de la Expresión Génica , Genes Reporteros , Proteínas Fluorescentes Verdes/metabolismo , Infecciones por Helicobacter/genética , Infecciones por Helicobacter/metabolismo , Ratones , Ratones Transgénicos , Antro Pilórico/metabolismo , Antro Pilórico/patología , Somatostatina/administración & dosificación , Distribución Tisular , Transcripción Genética , Transgenes , Regulación hacia Arriba
4.
Gastroenterology ; 138(3): 1022-34.e1-10, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19962981

RESUMEN

BACKGROUND & AIMS: The nuclear factor kappaB (NF-kappaB)/IkappaB-kinase-beta (IKKbeta) pathway has been shown to represent a key link between inflammation and cancer, inducing pro-inflammatory cytokines in myeloid cells and anti-apoptotic pathways in epithelial cells. However, the role of NF-kappaB pathway in gastric carcinogenesis and injury has not been well-defined. We derived mice with a conditional knockout of IKKbeta in gastric epithelial cells (GECs) and myeloid cells, and examined responses to ionizing radiation (IR) and Helicobacter felis infection. METHODS: Ikkbeta(Deltastom) mice were generated by crossing Foxa3-Cre mice to Ikkbeta(F/F) mice. Cellular stress was induced with IR and H felis in Ikkbeta(Deltastom), Ikkbeta(F/F), and cis-NF-kappaB-enhanced green fluorescent protein (GFP) reporter mice. Gastric histopathology, apoptosis, proliferation, necrosis, reactive oxygen species, and expression of cytokines, chemokines, and anti-apoptotic genes were assessed. The role of myeloid IKKbeta in these models was studied by crosses with LysM-Cre mice. RESULTS: NF-kappaB activity was upregulated in myeloid cells with acute H felis infection, but in GECs by IR or long-term H felis infection during progression to dysplasia. Deletion of IKKbeta in GECs led to increased apoptosis, reactive oxygen species, and cellular necrosis, and resulted in up-regulation of interleukin-1alpha and down-regulation of anti-apoptotic genes. Loss of IKKbeta in GECs resulted in worse inflammation and more rapid progression to gastric preneoplasia, while loss of IKKbeta in myeloid cells inhibited development of gastric atrophy. CONCLUSIONS: The loss of IKKbeta/NF-kappaB signaling in GECs results in increased apoptosis and necrosis in response to cellular stress, and accelerated development of dysplasia by Helicobacter infection.


Asunto(s)
Apoptosis , Proliferación Celular , Mucosa Gástrica/enzimología , Infecciones por Helicobacter/enzimología , Helicobacter felis/patogenicidad , Quinasa I-kappa B/deficiencia , Lesiones Precancerosas/enzimología , Neoplasias Gástricas/enzimología , Animales , Apoptosis/efectos de la radiación , Proteínas Reguladoras de la Apoptosis/metabolismo , Atrofia , Proliferación Celular/efectos de la radiación , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Mucosa Gástrica/microbiología , Mucosa Gástrica/patología , Mucosa Gástrica/efectos de la radiación , Genes Reporteros , Infecciones por Helicobacter/genética , Infecciones por Helicobacter/microbiología , Infecciones por Helicobacter/patología , Factor Nuclear 3-gamma del Hepatocito/genética , Quinasa I-kappa B/genética , Mediadores de Inflamación/metabolismo , Integrasas/genética , Interleucina-1alfa/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Mieloides/enzimología , Células Mieloides/microbiología , Células Mieloides/patología , FN-kappa B/genética , FN-kappa B/metabolismo , Necrosis , Estrés Oxidativo , Lesiones Precancerosas/genética , Lesiones Precancerosas/microbiología , Lesiones Precancerosas/patología , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Neoplasias Gástricas/genética , Neoplasias Gástricas/microbiología , Neoplasias Gástricas/patología , Factores de Tiempo
5.
Stem Cells ; 27(9): 2301-11, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19591219

RESUMEN

Bone marrow mesenchymal stem cells (MSCs) have been shown to have immune modulatory effects. Despite efforts to identify these cells in vivo, to date, MSCs have been defined mainly by their in vitro cell characteristics. Here, we show that Lin(-)CD44(hi)Sca1(-)cKit+CD34(-) cells make up approximately 0.5%-1% of murine whole bone marrow cells and yield nearly an equal amount of fibroblastic colony-forming units (CFU-F) as whole bone marrow. After transplantation into lethally irradiated recipients, Lin(-)CD44(hi)Sca1(-)cKit+CD34(-) cells engrafted in the bone marrow long-term and demonstrated characteristics of MSCs, including capacity to differentiate into osteoblasts and adipocytes. To examine whether Lin(-)CD44(hi)Sca1(-)cKit+CD34(-) cells have immune modulatory effects, in vitro coculture with activated CD4+ T-cells resulted in decreased Th17 cell differentiation by Lin(-)CD44(hi)Sca1(-)cKit+CD34(-) cells. Furthermore, serial infusions with Lin(-)CD44(hi)Sca1(-)cKit+CD34(-) cells reduced the progression to low-grade gastric dysplasia in mice infected with chronic Helicobacter felis (p = .038). This correlated with reduced gastric interleukin (IL)-17F, IL-22, and ROR-gammat gene expression in responding mice (p < .05). These data suggest that bone marrow derived Lin(-)CD44(hi)Sca1(-)cKit+CD34(-) cells have characteristics of MSCs and reduce progression of early gastric tumorigenesis induced by chronic H. felis infection. The prevention of dysplastic changes may occur through inhibition of Th17-dependent pathways.


Asunto(s)
Células de la Médula Ósea/fisiología , Helicobacter felis/fisiología , Células Madre Mesenquimatosas/fisiología , Neoplasias Gástricas/prevención & control , Neoplasias Gástricas/terapia , Animales , Células de la Médula Ósea/citología , Trasplante de Médula Ósea , Linfocitos T CD4-Positivos/inmunología , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Células Cultivadas , Femenino , Citometría de Flujo , Infecciones por Helicobacter/patología , Masculino , Células Madre Mesenquimatosas/citología , Ratones , Ratones Endogámicos C57BL , Células Madre , Neoplasias Gástricas/microbiología
6.
Gastroenterology ; 135(4): 1277-87, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18789941

RESUMEN

BACKGROUND & AIMS: Chronic pancreatitis is a significant cause of morbidity and a known risk factor for pancreatic adenocarcinoma. Interleukin-1beta is a proinflammatory cytokine involved in pancreatic inflammation. We sought to determine whether targeted overexpression of interleukin-1beta in the pancreas could elicit localized inflammatory responses and chronic pancreatitis. METHODS: We created a transgenic mouse model (elastase sshIL-1beta) in which the rat elastase promoter drives the expression of human interleukin-1beta. Mice were followed up for up to 2 years. Pancreata of elastase sshIL-1beta mice were analyzed for chronic pancreatitis-associated histologic and molecular changes. To study the potential effect of p53 mutation in chronic pancreatitis, elastase sshIL-1beta mice were crossed with p53(R172H) mice. RESULTS: Three transgenic lines were generated, and in each line the pancreas was atrophic and occasionally showed dilation of pancreatic and biliary ducts secondary to proximal fibrotic stenosis. Pancreatic histology showed typical features of chronic pancreatitis. There was evidence for increased acinar proliferation and apoptosis, along with prominent expression of tumor necrosis factor-alpha; chemokine (C-X-C motif) ligand 1; stromal cell-derived factor 1; transforming growth factor-beta1; matrix metallopeptidase 2, 7, and 9; inhibitor of metalloproteinase 1; and cyclooxygenase 2. The severity of the lesions correlated well with the level of human interleukin-1beta expression. Older mice displayed acinar-ductal metaplasia but did not develop mouse pancreatic intraepithelial neoplasia or tumors. Elastase sshIL-1beta*p53(R172H/+) mice had increased frequency of tubular complexes, some of which were acinar-ductal metaplasia. CONCLUSIONS: Overexpression of interleukin-1beta in the murine pancreas induces chronic pancreatitis. Elastase sshIL-1beta mice consistently develop severe chronic pancreatitis and constitute a promising model for studying chronic pancreatitis and its relationship with pancreatic adenocarcinoma.


Asunto(s)
Modelos Animales de Enfermedad , Interleucina-1beta/genética , Ratones Transgénicos , Páncreas/fisiopatología , Pancreatitis Crónica/fisiopatología , Animales , Femenino , Fibrosis , Expresión Génica/fisiología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Páncreas/patología , Elastasa Pancreática/genética , Pancreatitis Crónica/genética , Pancreatitis Crónica/patología , Fenotipo , Regiones Promotoras Genéticas/genética , Ratas , Índice de Severidad de la Enfermedad
7.
Mol Cell Biol ; 24(17): 7758-68, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15314181

RESUMEN

Emerging evidence suggests that the gamma subunit composition of an individual G protein contributes to the specificity of the hundreds of known receptor signaling pathways. Among the twelve gamma subtypes, gamma3 is abundantly and widely expressed in the brain. To identify specific functions and associations for gamma3, a gene-targeting approach was used to produce mice lacking the Gng3 gene (Gng3-/-). Confirming the efficacy and specificity of gene targeting, Gng3-/- mice show no detectable expression of the Gng3 gene, but expression of the divergently transcribed Bscl2 gene is not affected. Suggesting unique roles for gamma3 in the brain, Gng3-/- mice display increased susceptibility to seizures, reduced body weights, and decreased adiposity compared to their wild-type littermates. Predicting possible associations for gamma3, these phenotypic changes are associated with significant reductions in beta2 and alphai3 subunit levels in certain regions of the brain. The finding that the Gng3-/- mice and the previously reported Gng7-/- mice display distinct phenotypes and different alphabetagamma subunit associations supports the notion that even closely related gamma subtypes, such as gamma3 and gamma7, perform unique functions in the context of the organism.


Asunto(s)
Subunidades gamma de la Proteína de Unión al GTP/genética , Subunidades gamma de la Proteína de Unión al GTP/metabolismo , Convulsiones/genética , Adenilil Ciclasas/metabolismo , Tejido Adiposo , Animales , Peso Corporal , Encéfalo/metabolismo , Femenino , Subunidades beta de la Proteína de Unión al GTP/metabolismo , Marcación de Gen , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Proteínas de Unión al GTP Heterotriméricas/genética , Proteínas de Unión al GTP Heterotriméricas/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Convulsiones/metabolismo , Transducción de Señal/fisiología
8.
Cancer Cell ; 19(2): 257-72, 2011 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-21316604

RESUMEN

Carcinoma-associated fibroblasts (CAFs) that express α-smooth muscle actin (αSMA) contribute to cancer progression, but their precise origin and role are unclear. Using mouse models of inflammation-induced gastric cancer, we show that at least 20% of CAFs originate from bone marrow (BM) and derive from mesenchymal stem cells (MSCs). αSMA+ myofibroblasts (MFs) are niche cells normally present in BM and increase markedly during cancer progression. MSC-derived CAFs that are recruited to the dysplastic stomach express IL-6, Wnt5α and BMP4, show DNA hypomethylation, and promote tumor growth. Moreover, CAFs are generated from MSCs and are recruited to the tumor in a TGF-ß- and SDF-1α-dependent manner. Therefore, carcinogenesis involves expansion and relocation of BM-niche cells to the tumor to create a niche to sustain cancer progression.


Asunto(s)
Células de la Médula Ósea/citología , División Celular , Células Madre Mesenquimatosas/citología , Neoplasias Experimentales/patología , Neoplasias Gástricas/patología , Actinas/metabolismo , Animales , Transformación Celular Neoplásica , Modelos Animales de Enfermedad , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Ratones , Neoplasias Experimentales/metabolismo , Receptores CXCR4/metabolismo , Neoplasias Gástricas/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Regulación hacia Arriba
9.
Cancer Res ; 70(21): 8435-45, 2010 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-20959488

RESUMEN

Chronic infectious diseases, such as Helicobacter pylori infection, can promote cancer in a large part through induction of chronic inflammation. Oncogenic K-ras mutation in epithelial cells activates inflammatory pathways, which could compensate for a lack of infectious stimulus. Gastric histopathology and putative progenitor markers [doublecortin and calcium/calmodulin-dependent protein kinase-like 1 (Dcamkl1) and keratin 19 (K19)] in K19-K-ras-V12 (K19-kras) transgenic mice were assessed at 3, 6, 12, and 18 months of age, in comparison with Helicobacter felis-infected wild-type littermates. Inflammation was evaluated by reverse transcription-PCR of proinflammatory cytokines, and K19-kras mice were transplanted with green fluorescent protein (GFP)-labeled bone marrow. Both H. felis infection and K-ras mutation induced upregulation of proinflammatory cytokines, expansion of Dcamkl1(+) cells, and progression to oxyntic atrophy, metaplasia, hyperplasia, and high-grade dysplasia. K19-kras transgenic mice uniquely displayed mucous metaplasia as early as 3 months and progressed to high-grade dysplasia and invasive intramucosal carcinoma by 20 months. In bone marrow-transplanted K19-kras mice that progressed to dysplasia, a large proportion of stromal cells were GFP(+) and bone marrow-derived, but only rare GFP(+) epithelial cells were observed. GFP(+) bone marrow-derived cells included leukocytes and CD45(-) stromal cells that expressed vimentin or α smooth muscle actin and were often found surrounding clusters of Dcamkl1(+) cells at the base of gastric glands. In conclusion, the expression of mutant K-ras in K19(+) gastric epithelial cells can induce chronic inflammation and promote the development of dysplasia.


Asunto(s)
Carcinoma in Situ/etiología , Inflamación/etiología , Mutación/genética , Lesiones Precancerosas/etiología , Proteínas Proto-Oncogénicas/fisiología , Células Madre/patología , Neoplasias Gástricas/etiología , Proteínas ras/fisiología , Actinas/metabolismo , Animales , Western Blotting , Trasplante de Médula Ósea , Carcinoma in Situ/patología , Proliferación Celular , Quimiocinas/metabolismo , Citocinas/metabolismo , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Mucosa Gástrica/metabolismo , Infecciones por Helicobacter/metabolismo , Infecciones por Helicobacter/patología , Infecciones por Helicobacter/virología , Helicobacter felis , Humanos , Hiperplasia/etiología , Hiperplasia/patología , Hiperplasia/virología , Técnicas para Inmunoenzimas , Hibridación in Situ , Inflamación/patología , Queratina-19/genética , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/patología , Metaplasia/etiología , Metaplasia/patología , Metaplasia/virología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Músculo Liso/citología , Músculo Liso/metabolismo , Invasividad Neoplásica , Lesiones Precancerosas/patología , Proteínas Proto-Oncogénicas p21(ras) , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Madre/metabolismo , Células Madre/virología , Estómago/patología , Estómago/virología , Neoplasias Gástricas/patología , Neoplasias Gástricas/virología , Células del Estroma/metabolismo , Células del Estroma/patología , Células del Estroma/virología
10.
Cancer Cell ; 14(5): 408-19, 2008 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-18977329

RESUMEN

Polymorphisms of interleukin-1beta (IL-1beta) are associated with an increased risk of solid malignancies. Here, we show that stomach-specific expression of human IL-1beta in transgenic mice leads to spontaneous gastric inflammation and cancer that correlate with early recruitment of myeloid-derived suppressor cells (MDSCs) to the stomach. IL-1beta activates MDSCs in vitro and in vivo through an IL-1RI/NF-kappaB pathway. IL-1beta transgenic mice deficient in T and B lymphocytes develop gastric dysplasia accompanied by a marked increase in MDSCs in the stomach. Antagonism of IL-1 receptor signaling inhibits the development of gastric preneoplasia and suppresses MDSC mobilization. These results demonstrate that pathologic elevation of a single proinflammatory cytokine may be sufficient to induce neoplasia and provide a direct link between IL-1beta, MDSCs, and carcinogenesis.


Asunto(s)
Gastritis/etiología , Inflamación/etiología , Interleucina-1beta/metabolismo , Ratones Transgénicos/inmunología , Células Mieloides/inmunología , Neoplasias Gástricas/etiología , Animales , Médula Ósea/inmunología , Médula Ósea/metabolismo , Médula Ósea/patología , Células Cultivadas , Proteínas de Unión al ADN/fisiología , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Gastritis/metabolismo , Gastritis/patología , ATPasa Intercambiadora de Hidrógeno-Potásio/fisiología , Infecciones por Helicobacter/inmunología , Infecciones por Helicobacter/metabolismo , Infecciones por Helicobacter/virología , Helicobacter felis/patogenicidad , Inflamación/metabolismo , Proteína Antagonista del Receptor de Interleucina 1/genética , Interleucina-1beta/inmunología , Linfocitos/inmunología , Linfocitos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos/metabolismo , Ratones Transgénicos/virología , FN-kappa B/antagonistas & inhibidores , FN-kappa B/genética , FN-kappa B/metabolismo , Lesiones Precancerosas/inmunología , Lesiones Precancerosas/metabolismo , Lesiones Precancerosas/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Interleucina-1/antagonistas & inhibidores , Receptores de Interleucina-1/genética , Receptores de Interleucina-1/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología , Transfección
11.
Lab Invest ; 86(10): 1037-51, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16894354

RESUMEN

Hypergastrinemia in INS-GAS mice leads to accelerated carcinogenesis of the stomach, but the mechanisms have not been well defined. We investigated the possible role of gastrin-induced gastric cell apoptosis in the development of gastric cancer. We examined apoptosis and the expression of Bcl-2 family proteins in INS-GAS mice of different ages, as well as in gastrin-deficient (GAS-KO) mice after gastrin-17 (G-17) infusion. In addition, we studied the effects of the gastrin/cholecystokinin-2 (CCK-2) receptor antagonist YF476 and/or histamine H2 (H-2) receptor antagonist loxtidine on apoptosis and atrophy in INS-GAS mice with or without Helicobacter felis (H. felis) infection. INS-GAS mice had age-associated increases in Bax protein expression and decreases in Bcl-2 protein expression, along with increased glandular and epithelial cell apoptosis. At 8-week gastrin infusions in GAS-KO mice resulted in a similar pattern of altered Bax and Bcl-2 expression, followed by gastric cell apoptosis. H. felis infection of INS-GAS mice led to increased apoptosis and the development of atrophy, whereas treatment with either YF476 and/or loxtidine strongly inhibited both apoptosis and atrophy. In vitro studies with Fas-expressing RGM1 cells showed that gastrin stimulation alone directly induced apoptosis via gastrin/CCK-2 receptor and synergized with FasL stimulation. These results indicate that gastrin can induce apoptosis in gastric epithelial cells and contribute to the development of gastric carcinogenesis.


Asunto(s)
Apoptosis/fisiología , Atrofia/fisiopatología , Gastrinas/fisiología , Receptor de Colecistoquinina B/fisiología , Receptores Histamínicos H2/fisiología , Neoplasias Gástricas/fisiopatología , Animales , Atrofia/inmunología , Modelos Animales de Enfermedad , Infecciones por Helicobacter , Helicobacter felis , Masculino , Ratones , Ratones Noqueados , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Neoplasias Gástricas/microbiología , Neoplasias Gástricas/patología
12.
J Biol Chem ; 278(8): 6575-9, 2003 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-12488442

RESUMEN

The G protein beta gamma-dimer is required for receptor interaction and effector regulation. However, previous approaches have not identified the physiologic roles of individual subtypes in these processes. We used a gene knockout approach to demonstrate a unique role for the G protein gamma(7)-subunit in mice. Notably, deletion of Gng7 caused behavioral changes that were associated with reductions in the alpha(olf)-subunit content and adenylyl cyclase activity of the striatum. These data demonstrate that an individual gamma-subunit contributes to the specificity of a given signaling pathway and controls the formation or stability of a particular G protein heterotrimer.


Asunto(s)
Cuerpo Estriado/metabolismo , AMP Cíclico/metabolismo , Proteínas de Unión al GTP Heterotriméricas/genética , Proteínas de Unión al GTP Heterotriméricas/metabolismo , Animales , Secuencia de Bases , Cartilla de ADN , Dimerización , Femenino , Subunidades alfa de la Proteína de Unión al GTP , Proteínas de Unión al GTP Heterotriméricas/deficiencia , Masculino , Ratones , Ratones Noqueados , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA