Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Am J Physiol Gastrointest Liver Physiol ; 310(1): G13-25, 2016 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-26514775

RESUMO

We tested whether the T helper (Th) type 2 (Th2) cell agonist and allergenic ligand IL-33 was associated with eosinophilic esophagitis (EoE) development in a pediatric cohort and whether IL-33 protein could induce disease symptoms in mice. Biopsies from EoE patients or controls were used to measure IL-33 mRNA and protein expression. Increased expression of IL-33 mRNA was found in the esophageal mucosa in EoE. IL-33 protein was detected in cells negative for CD45, mast cells, and epithelial cell markers near blood vessels. Circulating levels of IL-33 were not increased. The time course for IL-33 gene expression was quantified in an established Aspergillus fumigatus allergen mouse model of EoE. Because IL-33 induction was transient in this model and chronicity of IL-33 expression has been demonstrated in humans, naive mice were treated with recombinant IL-33 for 1 wk and esophageal pathology was evaluated. IL-33 application produced changes consistent with phenotypically early EoE, including transmural eosinophilia, mucosal hyperproliferation, and upregulation of eosinophilic genes and chemokines. Th2 cytokines, including IL-13, along with innate lymphoid cell group 2, Th1/17, and M2 macrophage marker genes, were increased after IL-33 application. IL-33-induced eosinophilia was ablated in IL-13 null mice. In addition, IL-33 induced a profound inhibition of the regulatory T cell gene signature. We conclude that IL-33 gene expression is associated with pediatric EoE development and that application of recombinant protein in mice phenocopies the early clinical phase of the human disease in an IL-13-dependent manner. IL-33 inhibition of esophageal regulatory T cell function may induce loss of antigenic tolerance, thereby providing a mechanistic rationale for EoE development.


Assuntos
Esofagite Eosinofílica/induzido quimicamente , Esofagite Eosinofílica/metabolismo , Esôfago/metabolismo , Mediadores da Inflamação/metabolismo , Interleucina-33/metabolismo , Imunidade Adaptativa , Adolescente , Animais , Aspergillus fumigatus/patogenicidade , Biópsia , Estudos de Casos e Controles , Proliferação de Células , Quimiocina CCL26 , Quimiocinas CC/metabolismo , Criança , Pré-Escolar , Modelos Animais de Doenças , Células Endoteliais/imunologia , Células Endoteliais/metabolismo , Esofagite Eosinofílica/genética , Esofagite Eosinofílica/imunologia , Esofagite Eosinofílica/microbiologia , Esofagite Eosinofílica/patologia , Esôfago/imunologia , Esôfago/microbiologia , Esôfago/patologia , Humanos , Tolerância Imunológica , Imunidade Inata , Interleucina-13/deficiência , Interleucina-13/genética , Interleucina-33/genética , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos Endogâmicos BALB C , Camundongos Knockout , Fenótipo , RNA Mensageiro/metabolismo , Linfócitos T Auxiliares-Indutores/imunologia , Linfócitos T Auxiliares-Indutores/metabolismo , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Fatores de Tempo , Regulação para Cima
2.
Oncogene ; 34(22): 2856-66, 2015 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-25088199

RESUMO

Runt domain transcription factor 3 (RUNX3) is widely regarded as a tumour-suppressor gene inactivated by DNA hypermethylation of its canonical CpG (cytidine-phosphate-guanidine) island (CGI) promoter in gastric cancer (GC). Absence of RUNX3 expression from normal gastric epithelial cells (GECs), the progenitors to GC, coupled with frequent RUNX3 overexpression in GC progression, challenge this longstanding paradigm. However, epigenetic models to better describe RUNX3 deregulation in GC have not emerged. Here, we identify lineage-specific DNA methylation at an alternate, non-CGI promoter (P1) as a new mechanism of RUNX3 epigenetic control. In normal GECs, P1 was hypermethylated and repressed, whereas in immune lineages P1 was hypomethylated and widely expressed. In human GC development, we detected aberrant P1 hypomethylation signatures associated with the early inflammatory, preneoplastic and tumour stages. Aberrant P1 hypomethylation was fully recapitulated in mouse models of gastric inflammation and tumorigenesis. Cell sorting showed that P1 hypomethylation reflects altered cell-type composition of the gastric epithelium/tumour microenvironment caused by immune cell recruitment, not methylation loss. Finally, via long-term culture of gastric tumour epithelium, we revealed that de novo methylation of the RUNX3 canonical CGI promoter is a bystander effect of oncogenic immortalization and not likely causal in GC pathogenesis as previously argued. We propose a new model of RUNX3 epigenetic control in cancer, based on immune-specific, non-CGI promoter hypomethylation. This novel epigenetic signature may have utility in early detection of GC and possibly other epithelial cancers with premalignant immune involvement.


Assuntos
Linhagem da Célula/genética , Subunidade alfa 3 de Fator de Ligação ao Core/genética , Metilação de DNA , Lesões Pré-Cancerosas/genética , Lesões Pré-Cancerosas/imunologia , Neoplasias Gástricas/genética , Neoplasias Gástricas/imunologia , Animais , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/imunologia , Células Cultivadas , Ilhas de CpG , Mucosa Gástrica/imunologia , Mucosa Gástrica/metabolismo , Mucosa Gástrica/patologia , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Especificidade de Órgãos/genética , Especificidade de Órgãos/imunologia , Regiões Promotoras Genéticas , Neoplasias Gástricas/patologia
3.
Biochim Biophys Acta ; 1793(11): 1623-33, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19665497

RESUMO

Cytokine signalling pathways that depend on gp130 are dysregulated in several epithelial cancers including gastric cancer. It has been established that blockade of SHP2 activation of MAPK signalling results in hyperactivation of STAT3 resulting in increased cell proliferation, angiogenesis, inflammation and inhibition of both immunocyte and epithelial cell apoptosis. Additionally, key genes regulated downstream of gp130 via MAPK activation such as the stomach-specific tumor suppressor gene tff1 are suppressed, contributing to the oncogenic outcome. The main cytokine driver of gp130 signalling in the stomach is IL-11, with IL-6 having little activity in the antral stomach in which most pathology initiates. IL-11 is up-regulated in both mouse and human gastric cancer and in pre-neoplastic mucosa. A characteristic gene signature specifically associated with IL-11 drive has been observed, although the prognostic value of the signature has not yet been assessed. Infection of human or mouse stomach with Helicobacter pylori, especially that expressing the CagA cytotoxin, produces constitutive MAPK activation, but also activated STAT3 and increases IL-11 expression. The possibility of designing and utilising small molecule inhibitors of either IL-11 or STAT3 activation may be worthwhile in developing new cancer therapeutics.


Assuntos
Receptor gp130 de Citocina/metabolismo , Interleucina-11/metabolismo , Interleucina-6/metabolismo , Sistema de Sinalização das MAP Quinases , Neoplasias Gástricas/metabolismo , Animais , Antígenos de Bactérias/metabolismo , Apoptose , Proteínas de Bactérias/metabolismo , Proliferação de Células , Ativação Enzimática , Mucosa Gástrica/metabolismo , Regulação Neoplásica da Expressão Gênica , Infecções por Helicobacter/tratamento farmacológico , Infecções por Helicobacter/metabolismo , Helicobacter pylori , Humanos , Interleucina-11/antagonistas & inibidores , Camundongos , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/metabolismo , Neovascularização Patológica/microbiologia , Peptídeos/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 11/metabolismo , Fator de Transcrição STAT3/antagonistas & inibidores , Fator de Transcrição STAT3/metabolismo , Neoplasias Gástricas/tratamento farmacológico , Neoplasias Gástricas/microbiologia , Fator Trefoil-1 , Proteínas Supressoras de Tumor/metabolismo
4.
J Pathol ; 213(2): 140-51, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17724739

RESUMO

H. pylori infection accounts for most cases of gastric cancer, but the initiating events remain unclear. The principal H. pylori pathogenicity-associated CagA protein disrupts intracellular SHP-2 signalling pathways including those used by the IL-6 family cytokines, IL-6 and IL-11. Imbalanced IL-6 family cytokine signalling in the gp130(757FF) mouse model of gastric cancer arising from hyperactivation of oncogenic STAT3 after altered SHP-2 : ERK1/2 signalling produces dysplastic antral tumours preceded by gastritis and metaplasia. In a cohort of patient gastric biopsies with known H. pylori and CagA status, we investigated whether (i) STAT3 and ERK1/2 activation is altered in H. pylori-dependent gastritis; (ii) these profiles are more pronounced in CagA+ H. pylori infection; and (iii) the expression of pro-inflammatory cytokines that activate STAT3 and ERK 1/2 pathways is associated with progression to gastric cancer. IL-6, IL-11, and activated STAT3 and ERK1/2 were quantified in antral biopsies from gastritic stomach, metaplastic tissue, and resected gastric cancer tissues. We observed significantly increased STAT3 and ERK1/2 activation (p = 0.001) in H. pylori-dependent gastritis, which was further enhanced in the presence of CagA+ H. pylori strains. Of known gastric ligands that drive STAT3 activation, IL-6 expression was increased after H. pylori infection and both IL-6 and IL-11 were strongly up-regulated in the gastric cancer biopsies. This suggests a mechanism by which IL-11 drives STAT3 activation and proliferation during gastric cancer progression. We addressed this using an in vitro approach, demonstrating that recombinant human IL-11 activates STAT3 and concomitantly increases proliferation of MKN28 gastric epithelial cells. In summary, we show increased STAT3 and ERK1/2 activation in H. pylori-dependent gastritis that is likely driven in an IL-6-dependent fashion. IL-11 expression is associated with adenocarcinoma development, but not gastritic lesions, and we identify a novel mechanism for IL-11 as a potent inducer of proliferation in the human gastric cancer setting.


Assuntos
Interleucina-6/metabolismo , Neoplasias Gástricas/imunologia , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/microbiologia , Adenocarcinoma/patologia , Antígenos de Bactérias/metabolismo , Proteínas de Bactérias/metabolismo , Biópsia , Proliferação de Células , Progressão da Doença , Ativação Enzimática , Mucosa Gástrica/metabolismo , Mucosa Gástrica/microbiologia , Gastrite/metabolismo , Gastrite/microbiologia , Regulação Neoplásica da Expressão Gênica , Infecções por Helicobacter/complicações , Helicobacter pylori , Humanos , Interleucina-11/metabolismo , Interleucina-8/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Proteínas de Neoplasias/metabolismo , Inibidores da Bomba de Prótons , Antro Pilórico/microbiologia , Antro Pilórico/patologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Neoplasias Gástricas/genética , Neoplasias Gástricas/microbiologia , Neoplasias Gástricas/patologia , Proteína 3 Supressora da Sinalização de Citocinas , Proteínas Supressoras da Sinalização de Citocina/metabolismo
5.
Artigo em Inglês | MEDLINE | ID: mdl-16935852

RESUMO

Gastric trefoil peptides mediate mucosal repair by stimulating cell migration, inhibiting apoptosis and inflammation, and likely augmenting the barrier function of mucus. One of these, tff1, is a gastric-specific tumor suppressor gene, which when repressed is associated with gastric cancer progression. IL-6 family cytokines play an important role in maintaining gastric homeostasis by regulating tff1 and other mediators of mucosal proliferation, inflammation, angiogenesis, and apoptosis. In this review the signaling cascades downstream of the common IL-6 cytokine family coreceptor gp130 that contribute to control of this homeostasis are described, as are the pathological outcomes of imbalancing these pathways.


Assuntos
Mucosa Gástrica/citologia , Mucosa Gástrica/fisiologia , Interleucina-6/fisiologia , Animais , Estrogênios/fisiologia , Homeostase , Humanos , Transdução de Sinais , Fator Trefoil-1 , Proteínas Supressoras de Tumor/fisiologia
6.
J Pathol ; 207(1): 43-52, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15983982

RESUMO

H+/K+-ATPase beta-subunit-deficient mice (129/Sv background) display numerous pathologies in the stomach. Expression of the mutation in BALB/cCrSlc mice results in the development of an aberrant 'mucus-rich' cell population. 'Mucus-rich' cells have been described in stomachs of mice with autoimmune gastritis, a disease mediated by CD4+ T cells. Other pathological features of autoimmune gastritis are similar to those in H+/K+ beta-deficient mice and include a mononuclear cell infiltrate in the gastric mucosa, non-functional or absent parietal cells, depletion of zymogenic cells, hypergastrinaemia, and gastric unit hypertrophy caused by immature cell hyperplasia. The present study investigates further the aberrant gastric 'mucus-rich' cell lineage and analyses the mRNA expression of mucus cell products TFF1 and TFF2. 'Mucus-rich' cells stained for both acidic and neutral mucins, and with a TFF2-specific antibody. Stomachs from both models expressed decreased TFF1 mRNA and reciprocally increased TFF2 mRNA. The involvement of gastrin in regulating trefoil mRNA expression was also investigated using gastrin-deficient mice. In contrast to previous findings, gastrin did not positively regulate TFF1 mRNA expression, but there was possible augmentation of TFF2. Additionally, a clear role for inflammation was established involving both polymorphonuclear and mononuclear cells in these models, and a link was found between mucosal hypertrophy and increased interleukin-11 (IL-11) expression.


Assuntos
Mucosa Gástrica/patologia , Gastrite/metabolismo , Mucinas/metabolismo , Proteínas Musculares/metabolismo , Peptídeos/metabolismo , Animais , Doenças Autoimunes/metabolismo , Citocinas/biossíntese , Citocinas/genética , Citocinas/fisiologia , Modelos Animais de Doenças , Mucosa Gástrica/metabolismo , Gastrinas/sangue , Regulação da Expressão Gênica , ATPase Trocadora de Hidrogênio-Potássio/deficiência , ATPase Trocadora de Hidrogênio-Potássio/genética , Hiperplasia/metabolismo , Hipertrofia/metabolismo , Interleucina-11/biossíntese , Interleucina-11/genética , Interleucina-11/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Mutantes , Mucinas/genética , Mucinas/fisiologia , Proteínas Musculares/genética , Proteínas Musculares/fisiologia , Peptídeos/genética , Peptídeos/fisiologia , RNA Mensageiro/genética , Especificidade da Espécie , Fator Trefoil-1 , Fator Trefoil-2
7.
Am J Physiol Gastrointest Liver Physiol ; 281(6): G1502-11, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11705756

RESUMO

The gastric H(+)/K(+)-ATPase is essential for normal development of parietal cells. Here we have directly assessed the role of the H(+)/K(+)-ATPase beta-subunit (H/K-beta) on epithelial cell development by detailed quantitation of the epithelial cell types of the gastric mucosa of H/K-beta-deficient mice. H/K-beta-deficient mice had a 3.1-fold increase in the number of immature cells per gastric unit; however, the numbers of surface mucous and parietal cells were similar to those in the gastric units of wild-type mice. The effect of elevated gastrin levels in the H/K-beta-deficient mice was determined by producing mice that are also deficient in gastrin. We demonstrated that the increased production of immature cells and resulting hypertrophy is caused by the overproduction of gastrin. However, the depletion of zymogenic cells, which is another feature of H/K-beta-deficient mice, is independent of hypergastrinemia. Significantly, parietal cells of H/K-beta- and gastrin-deficient mice had abnormal secretory membranes and were devoid of resting tubulovesicular membranes. Together these data suggest a homeostatic mechanism limiting the number of immature cells that can develop into end-stage epithelial cells and indicate a direct role for H/K-beta in the development of mature parietal cells.


Assuntos
Mucosa Gástrica/patologia , Gastrinas/deficiência , ATPase Trocadora de Hidrogênio-Potássio/deficiência , Animais , Contagem de Células , Morte Celular , Divisão Celular , Ciclinas/análise , Células Epiteliais/patologia , Gastrinas/genética , Gastrinas/fisiologia , ATPase Trocadora de Hidrogênio-Potássio/genética , ATPase Trocadora de Hidrogênio-Potássio/fisiologia , Concentração de Íons de Hidrogênio , Hipertrofia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Microscopia Eletrônica , Células Parietais Gástricas/patologia , Fenótipo , Antígeno Nuclear de Célula em Proliferação/análise
8.
Immunology ; 104(4): 392-401, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11899424

RESUMO

Autoimmune gastritis, in which the H+/K(+)-ATPase of parietal cells is the major antigen, is one of the most common autoimmune diseases. Here we examined if specific properties of the H+/K(+)-ATPase or parietal cells are involved in rendering them autoimmune targets. The model antigens beta-galactosidase and ovalbumin (OVA) were expressed in parietal cells of transgenic mice. On experimental induction of autoimmune gastritis by neonatal thymectomy, autoantibodies to beta-galactosidase developed in mice expressing beta-galactosidase in parietal cells, a response that was independent of either the response to the gastric H+/K(+)-ATPase or gastric inflammation. In contrast, mice that expressed OVA in parietal cells did not exhibit an antibody response to OVA after thymectomy. However, increasing the frequency of anti-OVA T lymphocytes in OVA-expressing mice resulted in autoantibodies to OVA and gastritis. These studies indicate that parietal cells can present a variety of antigens to the immune system. Factors such as the identity and expression level of the autoantigen and the frequency of autoreactive T cells play a role in determining the prevalence and outcome of the particular immune response. In addition, as not all mice of a particular genotype displayed autoimmunity, random events are involved in determining the target of autoimmune recognition.


Assuntos
Autoantígenos/imunologia , Doenças Autoimunes/imunologia , Gastrite/imunologia , Estômago/imunologia , Animais , Doenças Autoimunes/patologia , Autoimunidade , Feminino , Gastrite/patologia , Expressão Gênica/imunologia , ATPase Trocadora de Hidrogênio-Potássio/imunologia , Tolerância Imunológica , Imunoglobulina G/biossíntese , Masculino , Camundongos , Camundongos Transgênicos , Ovalbumina/metabolismo , Células Parietais Gástricas/imunologia , Timo/imunologia , Transgenes , beta-Galactosidase/metabolismo
9.
Gastroenterology ; 117(3): 605-18, 1999 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10464136

RESUMO

BACKGROUND & AIMS: Parietal cells of the gastric mucosa contain a complex and extensive secretory membrane system that harbors gastric H(+),K(+)-adenosine triphosphatase (ATPase), the enzyme primarily responsible for acidification of the gastric lumen. We have produced mice deficient in the H(+),K(+)-ATPase beta subunit to determine the role of the protein in the biosynthesis of this membrane system and the biology of gastric mucosa. METHODS: Mice deficient in the H(+), K(+)-ATPase beta subunit were produced by gene targeting. RESULTS: The stomachs of H(+),K(+)-ATPase beta subunit-deficient mice were achlorhydric. Histological and immunocytochemical analyses with antibodies to the H(+),K(+)-ATPase alpha subunit revealed that parietal cell development during ontogeny was retarded in H(+), K(+)-ATPase beta subunit-deficient mice. In 15-day-old mice, cells with secretory canaliculi were observed in wild-type but not in H(+), K(+)-ATPase beta subunit-deficient mice. Parietal cells of H(+), K(+)-ATPase beta subunit-deficient mice 17 days and older contained an abnormal canaliculus that was dilated and contained fewer and shorter microvilli than normal. In older parietal cells, the abnormal canaliculus was massive (25 micrometer in diameter) and contained few microvilli. We did not observe typical tubulovesicular membranes in any parietal cell from H(+),K(+)-ATPase beta subunit-deficient mice. Histopathologic alterations were only observed in the stomach. CONCLUSIONS: The H(+),K(+)-ATPase beta subunit is required for acid-secretory activity of parietal cells in vivo, normal development and cellular homeostasis of the gastric mucosa, and attainment of the normal structure of the secretory membranes.


Assuntos
Mucosa Gástrica/citologia , ATPase Trocadora de Hidrogênio-Potássio/fisiologia , Células Parietais Gástricas/fisiologia , Animais , Mucosa Gástrica/enzimologia , Mucosa Gástrica/metabolismo , Gastrinas/sangue , ATPase Trocadora de Hidrogênio-Potássio/genética , Concentração de Íons de Hidrogênio , Immunoblotting , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia Eletrônica , Células Parietais Gástricas/metabolismo , Células Parietais Gástricas/ultraestrutura , RNA/isolamento & purificação , Reação em Cadeia da Polimerase Via Transcriptase Reversa
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA