Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 14(7): e0215883, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31291255

RESUMO

Innate CD8αα+ cells, also referred to as iCD8α cells, are TCR-negative intraepithelial lymphocytes (IEL) possessing cytokine and chemokine profiles and functions related to innate immune cells. iCD8α cells constitute an important source of osteopontin in the intestinal epithelium. Osteopontin is a pleiotropic cytokine with diverse roles in bone and tissue remodeling, but also has relevant functions in the homeostasis of immune cells. In this report, we present evidence for the role of iCD8α cells in the homeostasis of TCR-negative NKp46+NK1.1+ IEL (ILC1-like). We also show that the effect of iCD8α cells on ILC1-like IEL is enhanced in vitro by osteopontin. We show that in the absence of iCD8α cells, the number of NKp46+NK1.1+ IEL is significantly reduced. These ILC1-like cells are involved in intestinal pathogenesis in the anti-CD40 mouse model of intestinal inflammation. Reduced iCD8α cell numbers results in a milder form of intestinal inflammation in this disease model, whereas treatment with osteopontin increases disease severity. Collectively, our results suggest that iCD8α cells promote survival of NKp46+NK1.1+ IEL, which significantly impacts the development of intestinal inflammation.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Mucosa Intestinal/imunologia , Linfócitos Intraepiteliais/imunologia , Animais , Antígenos CD8/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/patologia , Sobrevivência Celular/imunologia , Gastroenterite/etiologia , Gastroenterite/imunologia , Gastroenterite/patologia , Homeostase/imunologia , Imunidade Inata , Mucosa Intestinal/patologia , Linfócitos Intraepiteliais/metabolismo , Linfócitos Intraepiteliais/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Osteopontina/metabolismo , Receptores de Antígenos de Linfócitos T/metabolismo
2.
Cancer Res ; 79(7): 1600-1611, 2019 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-30696658

RESUMO

Helicobacter pylori (H. pylori) is the strongest known risk for gastric cancer. The H. pylori cag type IV secretion system is an oncogenic locus that translocates peptidoglycan into host cells, where it is recognized by NOD1, an innate immune receptor. Beyond this, the role of NOD1 in H. pylori-induced cancer remains undefined. To address this knowledge gap, we infected two genetic models of Nod1 deficiency with the H. pylori cag + strain PMSS1: C57BL/6 mice, which rarely develop cancer, and INS-GAS FVB/N mice, which commonly develop cancer. Infected C57BL/6 Nod1-/- and INS-GAS Nod1-/- mice acutely developed more severe gastritis, and INS-GAS Nod1-/- mice developed gastric dysplasia more frequently compared with Nod1+/+ mice. Because Nod1 genotype status did not alter microbial phenotypes of in vivo-adapted H. pylori, we investigated host immunologic responses. H. pylori infection of Nod1-/- mice led to significantly increased gastric mucosal levels of Th1, Th17, and Th2 cytokines compared with Nod1 wild-type (WT) mice. To define the role of specific innate immune cells, we quantified cytokine secretion from H. pylori-infected primary gastric organoids generated from WT or Nod1-/- mice that were cocultured with or without WT or Nod1-/- macrophages. Infection increased cytokine production from gastric epithelial cells and macrophages and elevations were significantly increased with Nod1 deficiency. Furthermore, H. pylori infection altered the polarization status of Nod1-/- macrophages compared with Nod1+/+ macrophages. Collectively, these studies demonstrate that loss of Nod1 augments inflammatory and injury responses to H. pylori. Nod1 may exert its restrictive role by altering macrophage polarization, leading to immune evasion and microbial persistence. SIGNIFICANCE: These findings suggest that manipulation of NOD1 may represent a novel strategy to prevent or treat pathologic outcomes induced by H. pylori infection.


Assuntos
Helicobacter pylori/patogenicidade , Proteína Adaptadora de Sinalização NOD1/fisiologia , Neoplasias Gástricas/microbiologia , Animais , Carcinogênese , Citocinas/biossíntese , Mucosa Gástrica/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína Adaptadora de Sinalização NOD1/genética , Neoplasias Gástricas/imunologia
3.
J Pathol ; 239(4): 399-410, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27125972

RESUMO

Spasmolytic polypeptide-expressing metaplasia (SPEM) and intestinal metaplasia are considered neoplastic precursors of gastric adenocarcinoma in humans. Loss of parietal cells causes the development of SPEM in the gastric corpus and then chronic inflammation drives SPEM toward a more proliferative lineage. Mongolian gerbils infected with Helicobacter pylori develop chronic gastritis and metaplasia, mimicking aspects of human gastritis with H. pylori infection. We therefore examined metaplastic lineages in the gastric corpus mucosa of gerbils infected by H. pylori strain 7.13, which produces rapid onset of severe inflammation. Six weeks following H. pylori infection, Griffonia simplicifolia lectin II (GSII)-positive SPEM developed in the base of oxyntic glands in association with parietal cell loss and inflammation. In association with severe inflammation, SPEM glands evolved into aberrant phenotypes, including branched lesions, dilated lesions, and penetrating invasive glands. Mucin 4 (MUC4) was up-regulated in SPEM and progressive SPEM. Clusterin was expressed in the tips of branched and dilated lesions and throughout regions of invasive glands. Intriguingly, clusterin-positive regions in these lesions expressed Ki67 and matrix metalloproteinase 7 (MMP-7). These same regions were also positive for expression of phospho-IkBα, suggestive of activated NFkB signalling. These findings suggest that clusterin-positive regions in progressive phenotypes of SPEM have invasive characteristics. Thus, H. pylori infection in gerbils induces SPEM, which then can progress to further aberrant and invasive metaplastic phenotypes. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Assuntos
Mucosa Gástrica/patologia , Infecções por Helicobacter/patologia , Helicobacter pylori , Animais , Clusterina/metabolismo , Mucosa Gástrica/metabolismo , Mucosa Gástrica/microbiologia , Gerbillinae , Infecções por Helicobacter/complicações , Infecções por Helicobacter/microbiologia , Inflamação/metabolismo , Inflamação/microbiologia , Inflamação/patologia , Masculino , Metaplasia/etiologia , Metaplasia/microbiologia , Metaplasia/patologia , Mucina-4/metabolismo
4.
Gut ; 62(10): 1446-55, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22833394

RESUMO

OBJECTIVE: The myeloid translocation genes (MTGs) are transcriptional corepressors with both Mtg8(-/-) and Mtgr1(-/-) mice showing developmental and/or differentiation defects in the intestine. We sought to determine the role of MTG16 in intestinal integrity. METHODS: Baseline and stress induced colonic phenotypes were examined in Mtg16(-/-) mice. To unmask phenotypes, we treated Mtg16(-/-) mice with dextran sodium sulphate (DSS) or infected them with Citrobacter rodentium and the colons were examined for ulceration and for changes in proliferation, apoptosis and inflammation. RESULTS: Mtg16(-/-) mice have altered immune subsets, suggesting priming towards Th1 responses. Mtg16(-/-) mice developed increased weight loss, diarrhoea, mortality and histological colitis and there were increased innate (Gr1(+), F4/80(+), CD11c(+) and MHCII(+); CD11c(+)) and Th1 adaptive (CD4) immune cells in Mtg16(-/-) colons after DSS treatment. Additionally, there was increased apoptosis and a compensatory increased proliferation in Mtg16(-/-) colons. Compared with wild-type mice, Mtg16(-/-) mice exhibited increased colonic CD4;IFN-γ cells in vehicle-treated and DSS-treated mice. Adoptive transfer of wild-type marrow into Mtg16(-/-) recipients did not rescue the Mtg16(-/-) injury phenotype. Isolated colonic epithelial cells from DSS-treated Mtg16(-/-) mice exhibited increased KC (Cxcl1) mRNA expression when compared with wild-type mice. Mtg16(-/-) mice infected with C rodentium had more severe colitis and greater bacterial colonisation. Last, MTG16 mRNA levels were reduced in human ulcerative colitis versus normal colon tissues. CONCLUSIONS: These observations indicate that MTG16 is critical for colonocyte survival and regeneration in response to intestinal injury and provide evidence that this transcriptional corepressor regulates inflammatory recruitment in response to injury.


Assuntos
Colite/patologia , Proteínas Nucleares/fisiologia , Fatores de Transcrição/fisiologia , Imunidade Adaptativa , Transferência Adotiva , Animais , Transplante Ósseo , Proliferação de Células , Colite/induzido quimicamente , Colite/imunologia , Colite/fisiopatologia , Colite Ulcerativa/metabolismo , Colo/imunologia , Sulfato de Dextrana , Enterócitos/patologia , Feminino , Humanos , Imunidade Inata , Imunofenotipagem , Absorção Intestinal/fisiologia , Mucosa Intestinal/patologia , Mucosa Intestinal/fisiopatologia , Masculino , Camundongos , Camundongos Knockout , Proteínas Nucleares/deficiência , Proteínas Nucleares/genética , Permeabilidade , Proteínas Repressoras , Células Th1/imunologia , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética
5.
Int J Cancer ; 128(3): 668-75, 2011 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-20473875

RESUMO

Identification of biomarkers is needed for development of screening programs to prevent gastric cancer. Because racial differences exist in cancer rates, we aimed to evaluate the association between polymorphisms in inflammation-related genes and gastric preneoplastic lesions in African Americans and Caucasians from Louisiana, USA. Gastric biopsies from 569 adults (361 African Americans and 208 Caucasians) undergoing diagnostic endoscopy were used for histological diagnosis and genomic DNA extraction. Polymorphisms within eight genes (IL1B, IL8, IL6, TNF, PTGS2, ARG1, IL10 and TGFB1) were investigated by TaqMan. The cagA status of Helicobacter pylori infection was assessed by PCR. Haplotype logistic regression models were used to identify variables associated with intestinal metaplasia or dysplasia. African Americans carrying the haplotype IL1B-511T/-31C/+3954T, which includes the three risk-associated alleles at the IL1B locus, were more likely to being diagnosed with intestinal metaplasia or dysplasia than those carrying the most common haplotype T-C-C (adjusted OR: 2.51, 95% CI: 1.1-5.5). None of the polymorphisms were associated with intestinal metaplasia and dysplasia in Caucasians. Age and cagA-positive status were independent factors associated with these lesions. Haplotypes at the IL1B locus may participate in mediating the susceptibility to gastric carcinogenesis and might be useful as markers of advanced premalignant lesions in African Americans. Interestingly, carriage of IL1B+3954T allele seems to be the key factor, even though the role played by other polymorphisms cannot be excluded.


Assuntos
População Negra/genética , Haplótipos , Inflamação/genética , Polimorfismo de Nucleotídeo Único , Lesões Pré-Cancerosas/genética , Neoplasias Gástricas/genética , População Branca/genética , Adulto , Índice de Massa Corporal , DNA de Neoplasias/genética , DNA de Neoplasias/isolamento & purificação , Feminino , Mucosa Gástrica/patologia , Infecções por Helicobacter/complicações , Infecções por Helicobacter/epidemiologia , Helicobacter pylori , Humanos , Interleucina-18/genética , Louisiana , Masculino , Pessoa de Meia-Idade , Polimorfismo Genético , Neoplasias Gástricas/complicações , Neoplasias Gástricas/patologia
6.
Gastroenterology ; 139(4): 1333-43, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20547161

RESUMO

BACKGROUND & AIMS: Infection with the gastric mucosal pathogen Helicobacter pylori is the strongest identified risk factor for distal gastric cancer. These bacteria colonize a significant part of the world's population. We investigated the molecular mechanisms of p53 regulation in H pylori-infected cells. METHODS: Mongolian gerbils were challenged with H pylori and their gastric tissues were analyzed by immunohistochemistry and immunoblotting with p53 antibodies. Gastric epithelial cells were co-cultured with H pylori and the regulation of p53 was assessed by real-time polymerase chain reaction, immunoblotting, immunofluorescence, and cell survival assays. Short hairpin RNA and dominant-negative mutants were used to inhibit activities of Human Double Minute 2 (HDM2) and AKT1 proteins. RESULTS: We found that in addition to previously reported up-regulation of p53, H pylori can also negatively regulate p53 by increasing ubiquitination and proteasomal degradation via activation of the serine/threonine kinase AKT1, which phosphorylates and activates the ubiquitin ligase HDM2. These effects were mediated by the bacterial virulence factor CagA; ectopic expression of CagA in gastric epithelial cells increased phosphorylation of HDM2 along with the ubiquitination and proteasomal degradation of p53. The decrease in p53 levels increased survival of gastric epithelial cells that had sustained DNA damage. CONCLUSIONS: H pylori is able to inhibit the tumor suppressor p53. H pylori activates AKT1, resulting in phosphorylation and activation of HDM2 and subsequent degradation of p53 in gastric epithelial cells. H pylori-induced dysregulation of p53 is a potential mechanism by which the microorganism increases the risk of gastric cancer in infected individuals.


Assuntos
Mucosa Gástrica/microbiologia , Helicobacter pylori/patogenicidade , Proteína Supressora de Tumor p53/análise , Animais , Antígenos de Bactérias/fisiologia , Proteínas de Bactérias/fisiologia , Linhagem Celular Tumoral , Gerbillinae , Humanos , Proteínas Proto-Oncogênicas c-akt/fisiologia , Proteínas Proto-Oncogênicas c-mdm2/fisiologia , Neoplasias Gástricas/etiologia , Virulência
7.
Cancer ; 112(8): 1688-98, 2008 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-18311783

RESUMO

BACKGROUND: Upper gastrointestinal adenocarcinomas are a common cause of cancer-related deaths. In this study, the authors investigated the prevalence and biological significance of Aurora Kinase A (AURKA) overexpression in upper gastrointestinal adenocarcinomas. METHODS: Quantitative real-time polymerase chain reaction (qRT-PCR) and immunohistochemical staining on tumor tissue microarrays (TMA) were used to study the expression of AURKA in upper gastrointestinal adenocarcinomas. To investigate the biological and signaling impact of AURKA, the authors used multiple in vitro assays that included 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide (MTT), TUNEL (terminal deoxynucleotidyl transferase-mediated nick-end labeling), cytochrome C release, flow cytometry, luciferase reporter, and Western blot analysis. RESULTS: Frequent overexpression of AURKA transcript in upper gastrointestinal adenocarcinomas was detected compared with normal samples (47%; P= .001). The immunohistochemical analysis of 130 tumors demonstrated moderate-to-strong immunostaining of AURKA in >50% of upper gastrointestinal adenocarcinomas. By using camptothecin as a drug-induced apoptosis in vitro model, the authors demonstrated that the expression of AURKA provided protection against apoptosis to gastrointestinal cancer cells (AGS and RKO) (P= .006) and RIE-1 primary intestinal epithelial cells (P= .001). The AURKA overexpression mediated an increase in phosphorylation of AKT(Ser473) with an increase in HDM2 level. The shRNA-knockdown of AKT in AURKA-overexpressing cells reversed this effect and showed a significant increase in the p53 protein level, indicating a possible nexus of AURKA/AKT/p53. Indeed, overexpression of AURKA led to a remarkable reduction in the transcription activity of p53, with subsequent reductions in transcript and protein levels of its downstream proapoptotic transcription targets (p21, BAX, NOXA, and PUMA). CONCLUSIONS: Study results indicated that AURKA provides potent antiapoptotic properties to gastrointestinal cells by regulating levels of p53 through the AKT/HDM2 axis.


Assuntos
Adenocarcinoma/enzimologia , Apoptose/fisiologia , Neoplasias Esofágicas/enzimologia , Proteínas Serina-Treonina Quinases/análise , Neoplasias Gástricas/enzimologia , Apoptose/genética , Aurora Quinase A , Aurora Quinases , Biomarcadores Tumorais/análise , Western Blotting , Camptotecina/farmacologia , Corantes , Citocromos c/análise , Inibidores Enzimáticos/farmacologia , Citometria de Fluxo , Regulação Enzimológica da Expressão Gênica , Humanos , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Luciferases , Substâncias Luminescentes , Reação em Cadeia da Polimerase , Análise Serial de Proteínas , Sais de Tetrazólio , Tiazóis , Proteína Supressora de Tumor p53/análise
8.
Gastroenterology ; 134(5): 1412-23, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18343378

RESUMO

BACKGROUND & AIMS: Although the p53 tumor suppressor has been extensively studied, many critical questions remain unanswered about the biological functions of p53 homologs, p73 and p63. Accumulating evidence suggests that both p73 and p63 play important roles in regulation of apoptosis, cell differentiation, and therapeutic drug sensitivity. METHODS: Gastric epithelial cells were cocultured with Helicobacter pylori, and the roles of p63 and p73 proteins were assessed by luciferase reporter, real-time polymerase chain reaction, immunoblotting, and cell survival assays. Short hairpin RNA and dominant-negative mutants were used to inhibit activity of p73 and p63 isoforms. Human and murine gastric tissues were analyzed by immunohistochemistry with p73 and p63 antibodies and modified Steiner's silver method. RESULTS: Interaction of H pylori with gastric epithelial cells leads to robust up-regulation of p73 protein in vitro and in vivo in human gastritis specimens and H pylori-infected mice. The p73 increase resulted in up-regulation of pro-apoptotic genes, NOXA, PUMA, and FAS receptor in gastric epithelial cells. Down-regulation of p73 activity suppressed cell death and Fas receptor induced by H pylori. Bacterial virulence factors within the cag pathogenicity island, c-Abl tyrosine kinase, and interaction with p63 isoforms control the activity of p73. CONCLUSION: Our findings implicate p73 in H pylori-induced apoptosis and more generally suggest that the p53 family may play a role in the epithelial cell response to H pylori infection.


Assuntos
Células Epiteliais/microbiologia , Mucosa Gástrica/microbiologia , Infecções por Helicobacter/microbiologia , Helicobacter pylori/fisiologia , Proteína Supressora de Tumor p53/fisiologia , Animais , Anticorpos Antibacterianos/análise , Apoptose , Proteínas Reguladoras de Apoptose/biossíntese , Proteínas Reguladoras de Apoptose/genética , Técnicas de Cocultura , Proteínas de Ligação a DNA/fisiologia , Modelos Animais de Doenças , Citometria de Fluxo , Gastrite/microbiologia , Vetores Genéticos , Humanos , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas Nucleares/fisiologia , Proteínas Proto-Oncogênicas/biossíntese , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-bcl-2/biossíntese , Proteínas Proto-Oncogênicas c-bcl-2/genética , RNA/genética , Proteína Tumoral p73 , Proteínas Supressoras de Tumor/fisiologia , Receptor fas/biossíntese , Receptor fas/genética
9.
Int J Cancer ; 118(3): 649-57, 2006 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-16114018

RESUMO

Several polymorphisms of the IL1B and IL10 gene promoters have been reported to be associated with gastric cancer risk in Caucasians. However, studies in other populations have shown differing results. We aimed to test for associations between polymorphisms in IL1B (-31 and +3954), IL10-592 and IL1RN variable number of tandem repeats (VNTR) and risk of gastric cancer in a Mexican population. DNA was extracted from sera of 183 gastric adenocarcinoma patients and 377 controls. The IL1B-31, IL1B+3954 and IL10-592 biallelic polymorphisms were discriminated using 5' Nuclease (TaqMan) assays and Pyrosequencing. The IL1RN penta-allelic VNTR polymorphism was genotyped using PCR followed by GeneScan analysis. A significant interaction was found between IL1B-31 and CagA status for the risk of intestinal-type gastric cancer (p = 0.023). Among CagA positive subjects, those with IL1B-31CC genotype had an increased risk of intestinal-type gastric cancer (OR 3.19, 95%CI = 1.05-9.68), compared to carriers of IL1B-31TT genotype. In contrast, among CagA negative subjects, no significant association of IL1B-31CC genotype with gastric cancer was observed. The IL10-592CC genotype was associated with more than doubling of the risk of the intestinal-type gastric cancer (OR, 2.20, 95%CI = 1.04-4.65). A nonsignificantly increased risk for intestinal-type gastric cancer was found in IL1RN*2 carriers (OR 1.49, 95%CI = 0.89-2.50). None of these polymorphisms was significantly related to the risk of diffuse-type gastric cancer. No significant association was found between risk of gastric cancer and the IL1B+3954 polymorphism. Individuals carrying 2 or more of the risk-associated alleles (IL1B-31C, IL1RN *2 and IL10-592C) were at increased risk for intestinal-type gastric cancer, compared to those with 0 or 1 risk-associated allele. The risk from multiple risk-associated alleles was especially high in subjects infected with CagA positive H. pylori. Our results support the identification of the IL1B-31 promoter polymorphism as a useful marker for risk of intestinal type gastric cancer in persons with CagA positive H. pylori infections.


Assuntos
Adenocarcinoma/genética , Antígenos de Bactérias/imunologia , Proteínas de Bactérias/imunologia , Infecções por Helicobacter/genética , Interleucina-10/genética , Interleucina-1/genética , Polimorfismo Genético , Neoplasias Gástricas/genética , Adenocarcinoma/epidemiologia , Adenocarcinoma/microbiologia , Estudos de Casos e Controles , Feminino , Genótipo , Infecções por Helicobacter/microbiologia , Helicobacter pylori/patogenicidade , Humanos , Masculino , México/epidemiologia , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase , Receptores de Interleucina-1/genética , Fatores de Risco , Sensibilidade e Especificidade , Neoplasias Gástricas/epidemiologia , Neoplasias Gástricas/microbiologia , Sequências de Repetição em Tandem
10.
Cancer Res ; 64(16): 5839-49, 2004 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-15313928

RESUMO

T cells infiltrating tumors have a decreased expression of signal transduction proteins, a diminished ability to proliferate, and a decreased production of cytokines. The mechanisms causing these changes have remained unclear. We demonstrated recently that peritoneal macrophages stimulated with interleukin 4 + interleukin 13 produce arginase I, which decreases the expression of the T-cell receptor CD3zeta chain and impairs T-cell responses. Using a 3LL murine lung carcinoma model we tested whether arginase I was produced in the tumor microenvironment and could decrease CD3zeta expression and impair T-cell function. The results show that a subpopulation of mature tumor-associated myeloid cells express high levels of arginase I, whereas tumor cells and infiltrating lymphocytes do not. Arginase I expression in the tumor was seen on day 7 after tumor injection. Tumor-associated myeloid cells also expressed high levels of cationic amino acid transporter 2B, which allowed them to rapidly incorporate L-Arginine (L-Arg) and deplete extracellular L-Arg in vitro. L-Arg depletion by tumor-associated myeloid cells blocked the re-expression of CD3zeta in stimulated T cells and inhibited antigen-specific proliferation of OT-1 and OT-2 cells. The injection of the arginase inhibitor N-hydroxy-nor-L-Arg blocked growth of s.c. 3LL lung carcinoma in mice. High levels of arginase I were also found in tumor samples of patients with non-small cell carcinoma. Therefore, arginase I production by mature myeloid cells in the tumor microenvironment may be a central mechanism for tumor evasion and may represent a target for new therapies.


Assuntos
Arginase/biossíntese , Arginase/imunologia , Carcinoma Pulmonar de Lewis/imunologia , Neoplasias Pulmonares/imunologia , Células Mieloides/imunologia , Receptores de Antígenos de Linfócitos T/antagonistas & inibidores , Sequência de Aminoácidos , Animais , Arginase/antagonistas & inibidores , Complexo CD3/biossíntese , Complexo CD3/imunologia , Carcinoma Pulmonar de Lewis/enzimologia , Carcinoma Pulmonar de Lewis/patologia , Carcinoma Pulmonar de Células não Pequenas/enzimologia , Carcinoma Pulmonar de Células não Pequenas/imunologia , Carcinoma Pulmonar de Células não Pequenas/patologia , Divisão Celular/fisiologia , Epitopos de Linfócito T/imunologia , Feminino , Humanos , Neoplasias Pulmonares/enzimologia , Neoplasias Pulmonares/patologia , Ativação Linfocitária/imunologia , Camundongos , Dados de Sequência Molecular , Células Mieloides/enzimologia , Receptores de Antígenos de Linfócitos T/biossíntese
11.
Oncol Rep ; 10(4): 1059-62, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12792770

RESUMO

To estimate the incidence of Epstein-Barr virus-associated gastric carcinoma (EBV-GC) in Colombia and to clarify its clinicopathological features, we examined 178 consecutive gastric carcinoma cases, diagnosed during the period from 1996 to 1998, at Hospital Universitario del Valle in Cali, Colombia. The mean age of the cases was 60 years in males and 58 years in females. Using in situ hybridization assay of EBV-encoded small RNA-1 in paraffin-embedded tissue samples, we identified 23 cases of EBV-GC (13%). After excluding remnant carcinoma, which was found to be EBV-negative in this series, there were 19 (18%) male and 4 (6%) female EBV-GC cases, and the male predominance was statistically significant (P=0.004). The proportion of EBV-GCs decreased with age (P for trend = 0.022). Using sex- and age-specific proportions of EBV-GCs estimated by logistic models and gastric cancer incidence in Cali, which was obtained from tumor registry during the period 1987-1991, we estimated sex- and age-specific incidence of EBV-GCs. The incidence of EBV-GCs (per 100,000 person-years) was 4.1 and 1.4 among men and women, respectively, after age adjustment using the standard world population. Pathological features of EBV-GCs were also examined. EBV-GCs accounted for 33% (8/24) of carcinomas located in the stomach cardia, 14% (6/43) of carcinomas in the middle-part of the stomach, and 7% (6/81) of carcinomas in the antrum. The difference by tumor location was statistically significant (P=0.009). Histology-specific analysis using Lauren classification revealed that the proportion of EBV-GCs was not different in intestinal- and diffuse-type carcinomas (13% in both types). When the classification scheme of the Japanese Research Society for Gastric Cancer was used, EBV-GCs were identified more frequently in moderately differentiated tubular adenocarcinoma, and solid poorly differentiated adenocarcinoma when compared to other histological types. No lymphoepitelioma-like histology was found in the present series. The frequency of EBV-GC was slightly higher in advanced tumors, which involved serosa. Further analysis of clinico-pathological features of EBV-GC using a larger number of cases would give invaluable insights into its etiology.


Assuntos
Adenocarcinoma/virologia , Infecções por Vírus Epstein-Barr/virologia , Herpesvirus Humano 4/isolamento & purificação , RNA Viral/genética , Neoplasias Gástricas/virologia , Adenocarcinoma/epidemiologia , Adenocarcinoma/patologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Colômbia/epidemiologia , Infecções por Vírus Epstein-Barr/epidemiologia , Infecções por Vírus Epstein-Barr/patologia , Feminino , Herpesvirus Humano 4/patogenicidade , Humanos , Técnicas Imunoenzimáticas , Hibridização In Situ , Masculino , Pessoa de Meia-Idade , Prognóstico , RNA Viral/isolamento & purificação , Neoplasias Gástricas/epidemiologia , Neoplasias Gástricas/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...