Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Malawi Med J ; 34(1): 17-24, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-37265823

RESUMO

Introduction: Little is known about specific bacterial characteristics of Helicobacter pylori (H. pylori) infection influencing gastric carcinogenesis in Zambia. The aim of this study was to evaluate the associations between pre-selected H. pylori antibodies with gastric cancer, premalignant lesions and active gastritis. Methods: This was cross-sectional study with multiple comparisons of patients with gastric cancer (GC), gastric premalignant (GP) lesions and active or chronic gastritis. A fluorescent bead-based antibody multiplex serology assay was used to quantify antibodies to thirteen immunogenic H. pylori antigens. Logistic regression models were used to examine the associations. Results: Included were 295 patients with: 59 GC, 27 GP lesions, 48 active and 161 chronic gastritis. Overall, 257/295 (87%) were H. pylori positive. H. pylori seropositivity was not associated with sex, age, body mass index, socio-economic status, HIV infection, alcohol consumption or cigarette smoking (p-values all above 0.05). When compared to the patients with chronic gastritis, the presence of catalase and cinnamyl alcohol dehydrogenase (Cad) antibodies was positively associated with GP lesions (OR 3.53; 95% CI 1.52-8.17 and OR 2.47; 95% CI 1.08-5.67 respectively). However, seropositivity to Cad antibodies was significantly lower in GC patients (OR 0.28; 95% CI 0.09-0.83). Compared to chronic, active gastritis was significantly associated with (p<0.05) H. pylori sero-positivity (OR 9.46; 95% CI 1.25-71.52) and specific antibodies including cytotoxin-associated gene A, vacuolating cytotoxin A, Helicobacter cysteine-rich protein C, hypothetical protein HP0305 and outer membrane protein HP1564. Conclusions: Among Zambian patients seen at a single center, antibodies to H. pylori (CagA, VacA, Omp, HcpC, HP0305 and HpaA) were associated with active gastritis.


Assuntos
Gastrite , Infecções por HIV , Helicobacter pylori , Neoplasias Gástricas , Humanos , Helicobacter pylori/genética , Zâmbia/epidemiologia , Estudos Transversais , Universidades , Antígenos de Bactérias/genética , Gastrite/epidemiologia , Gastrite/microbiologia , Hospitais de Ensino
2.
Chembiochem ; 22(18): 2783-2790, 2021 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-34169626

RESUMO

Chronic infection with Helicobacter pylori increases risk of gastric diseases including gastric cancer. Despite development of a robust immune response, H. pylori persists in the gastric niche. Progression of gastric inflammation to serious disease outcomes is associated with infection with H. pylori strains which encode the cag Type IV Secretion System (cag T4SS). The cag T4SS is responsible for translocating the oncogenic protein CagA into host cells and inducing pro-inflammatory and carcinogenic signaling cascades. Our previous work demonstrated that nutrient iron modulates the activity of the T4SS and biogenesis of T4SS pili. In response to H. pylori infection, the host produces a variety of antimicrobial molecules, including the iron-binding glycoprotein, lactoferrin. Our work shows that apo-lactoferrin exerts antimicrobial activity against H. pylori under iron-limited conditions, while holo-lactoferrin enhances bacterial growth. Culturing H. pylori in the presence of holo-lactoferrin prior to co-culture with gastric epithelial cells, results in repression of the cag T4SS activity. Concomitantly, a decrease in biogenesis of cag T4SS pili at the host-pathogen interface was observed under these culture conditions by high-resolution electron microscopy analyses. Taken together, these results indicate that acquisition of alternate sources of nutrient iron plays a role in regulating the pro-inflammatory activity of a bacterial secretion system and present novel therapeutic targets for the treatment of H. pylori-related disease.


Assuntos
Helicobacter pylori/efeitos dos fármacos , Lactoferrina/farmacologia , Sistemas de Secreção Tipo IV/metabolismo , Animais , Modelos Animais de Doenças , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Mucosa Gástrica/citologia , Mucosa Gástrica/metabolismo , Gerbillinae , Infecções por Helicobacter/tratamento farmacológico , Infecções por Helicobacter/microbiologia , Helicobacter pylori/metabolismo , Imunidade Inata , Interleucina-8/metabolismo , Ferro/metabolismo , Lactoferrina/química , Lactoferrina/metabolismo , Lactoferrina/uso terapêutico , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/farmacologia , Isoformas de Proteínas/uso terapêutico , Sistemas de Secreção Tipo IV/antagonistas & inibidores
3.
Virchows Arch ; 479(4): 679-686, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-33990867

RESUMO

The use of Operative Link on Gastritis Assessment (OLGA) and Operative Link on Gastritis Assessment based on Intestinal Metaplasia (OLGIM) staging system is recommended for identifying subjects at risk for developing gastric cancer; usually high-risk lesions are considered only as stages III and IV. Accumulating evidence suggests that incomplete intestinal metaplasia (IM) is important in the development of gastric cancer. Our aim has been to identify the prevalence of incomplete IM in patients with low-risk OLGA/OLGIM stages among a high-risk general population. Healthy adult volunteers aged 40-64 years were invited to undergo upper endoscopy within a regional GISTAR pilot study in Kazakhstan (n = 166). Gastric lesions were staged according to OLGA/OLGIM staging system. High iron diamine-alcian blue (HID-AB) was used for subtyping IM. IM prevalence overall was 45.8%. Incomplete IM was present in 52.6% (type II in 30.3% and type III in 22.3%), whereas complete IM was found in 47.4% individuals. The prevalence of OLGIM I and II stage were 39.8 and 4.8%, respectively, whereas OLGIM III was observed in 1.2%. The prevalence of incomplete IM in patients stratified to OLGIM I was 54.5% (type II in 31.8% and type III in 22.7%). High prevalence of incomplete IM was detected not only in subjects with extensive IM, but in those stratified as at the OLGIM I stage. Without IM subtyping, patients with high risk of gastric cancer development would be missed for surveillance.


Assuntos
Detecção Precoce de Câncer/métodos , Intestinos/patologia , Neoplasias Gástricas/diagnóstico , Adulto , Biópsia , Feminino , Gastrite/patologia , Voluntários Saudáveis , Infecções por Helicobacter/patologia , Helicobacter pylori , Humanos , Masculino , Metaplasia/patologia , Pessoa de Meia-Idade , Projetos Piloto , Lesões Pré-Cancerosas/patologia , Medição de Risco , Fatores de Risco , Índice de Gravidade de Doença , Neoplasias Gástricas/metabolismo
4.
Dig Dis Sci ; 65(7): 1899-1903, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32356261

RESUMO

Patients with gastric precancerous lesions (atrophic gastritis and intestinal metaplasia) have increased risk of developing gastric cancer, and adequate management and surveillance of these patients should allow to reduce gastric cancer-related mortality. The guidelines on the management of these patients have been recently published by the European Societies (MAPS II guidelines) and by the American Gastroenterological Association (AGA). The aim of this commentary is to compare these two guidelines by highlighting the common points and differences between them. Both guidelines recommend a systematic detection and eradication of Helicobacter pylori in all patients with gastric atrophy. However, there is a major difference in the recommendations for surveillance: while the MAPS II guidelines recommend systematic endoscopic surveillance in all patients with severe gastric atrophy (with or without intestinal metaplasia), the AGA guidelines focus only on intestinal metaplasia and plead against systematic surveillance, leaving the possibility of surveillance in individual patients based on shared decision between clinicians and patients. The difference between two guidelines comes essentially from the different arguments used by two authorities (randomized control studies by AGA and observational cohort studies by the European Societies), and may be, at least in part, related to the difference between the European and American health care systems and potential economic burden.


Assuntos
Adenocarcinoma/patologia , Gastrite Atrófica/terapia , Gastroscopia , Infecções por Helicobacter/diagnóstico , Lesões Pré-Cancerosas/terapia , Neoplasias Gástricas/patologia , Estômago/patologia , Adenocarcinoma/diagnóstico , Gerenciamento Clínico , Europa (Continente) , Gastrite Atrófica/patologia , Gastroenterologia , Infecções por Helicobacter/tratamento farmacológico , Helicobacter pylori , Humanos , Metaplasia , Guias de Prática Clínica como Assunto , Lesões Pré-Cancerosas/patologia , Sociedades Médicas , Neoplasias Gástricas/diagnóstico , Estados Unidos
5.
J Clin Invest ; 130(5): 2422-2434, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-32250340

RESUMO

Approximately half of the world's population is infected with the stomach pathogen Helicobacter pylori. Infection with H. pylori is the main risk factor for distal gastric cancer. Bacterial virulence factors, such as the oncoprotein CagA, augment cancer risk. Yet despite high infection rates, only a fraction of H. pylori-infected individuals develop gastric cancer. This raises the question of defining the specific host and bacterial factors responsible for gastric tumorigenesis. To investigate the tumorigenic determinants, we analyzed gastric tissues from human subjects and animals infected with H. pylori bacteria harboring different CagA status. For laboratory studies, well-defined H. pylori strain B128 and its cancerogenic derivative strain 7.13, as well as various bacterial isogenic mutants were employed. We found that H. pylori compromises key tumor suppressor mechanisms: the host stress and apoptotic responses. Our studies showed that CagA induces phosphorylation of XIAP E3 ubiquitin ligase, which enhances ubiquitination and proteasomal degradation of the host proapoptotic factor Siva1. This process is mediated by the PI3K/Akt pathway. Inhibition of Siva1 by H. pylori increases survival of human cells with damaged DNA. It occurs in a strain-specific manner and is associated with the ability to induce gastric tumor.


Assuntos
Antígenos de Bactérias/metabolismo , Proteínas de Bactérias/metabolismo , Células Epiteliais/metabolismo , Mucosa Gástrica/metabolismo , Helicobacter pylori/metabolismo , Neoplasias Gástricas/metabolismo , Antígenos de Bactérias/genética , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/metabolismo , Proteínas de Bactérias/genética , Células Epiteliais/microbiologia , Células Epiteliais/patologia , Mucosa Gástrica/microbiologia , Mucosa Gástrica/patologia , Células HCT116 , Helicobacter pylori/genética , Humanos , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteólise , Neoplasias Gástricas/genética , Neoplasias Gástricas/patologia , Ubiquitinação , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/genética , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/metabolismo
7.
Gut ; 64(7): 1028-39, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25107557

RESUMO

OBJECTIVE: In this study, we investigated the role of Trefoil factor 1 (TFF1) in regulating cell proliferation and tumour development through ß-catenin signalling using in vivo and in vitro models of gastric tumorigenesis. DESIGN: Tff1-knockout (Tff1-KO) mice, immunohistochemistry, luciferase reporter, qRT-PCR, immunoblot, and phosphatase assays were used to examine the role of TFF1 on ß-catenin signalling pathway. RESULTS: Nuclear localisation of ß-catenin with transcriptional upregulation of its target genes, c-Myc and Ccnd1, was detected in hyperplastic tissue at an early age of 4-6 weeks and maintained during all stages of gastric tumorigenesis in the Tff1-KO mice. The reconstitution of TFF1 or TFF1 conditioned media significantly inhibited the ß-catenin/T-cell factor (TCF) transcription activity in MKN28 gastric cancer cells. In agreement with these results, we detected a reduction in the levels of nuclear ß-catenin with downregulation of c-MYC and CCND1 mRNA. Analysis of signalling molecules upstream of ß-catenin revealed a decrease in phosphorylated glycogen synthase kinase 3ß (p-GSK3ß) (Ser9) and p-AKT (Ser473) protein levels following the reconstitution of TFF1 expression; this was consistent with the increase of p-ß-catenin (Ser33/37/Thr41) and decrease of p-ß-catenin (Ser552). This TFF1-induced reduction in phosphorylation of GSK3ß, and AKT was dependent on protein phosphatase 2A (PP2A) activity. The treatment with okadaic acid or knockdown of PP2A abrogated these effects. Consistent with the mouse data, we observed loss of TFF1 and an increase in nuclear localisation of ß-catenin in stages of human gastric tumorigenesis. CONCLUSIONS: Our data indicate that loss of TFF1 promotes ß-catenin activation and gastric tumorigenesis through regulation of PP2A, a major regulator of AKT-GSK3ß signalling.


Assuntos
Inibidores do Crescimento/fisiologia , Peptídeos/fisiologia , Proteína Fosfatase 2/fisiologia , Proteínas Proto-Oncogênicas c-akt/fisiologia , Neoplasias Gástricas/patologia , Neoplasias Gástricas/fisiopatologia , beta Catenina/fisiologia , Animais , Linhagem Celular Tumoral , Proliferação de Células , Regulação para Baixo/fisiologia , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Imuno-Histoquímica , Camundongos , Camundongos Knockout , Ativação Transcricional/fisiologia , Fator Trefoil-1
8.
Gut ; 64(7): 1040-8, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25080447

RESUMO

OBJECTIVE: Infection with Helicobacter pylori is the strongest known risk factor for adenocarcinoma of the stomach. Tumorigenic transformation of gastric epithelium induced by H. pylori is a highly complex process driven by an active interplay between bacterial virulence and host factors, many aspects of which remain obscure. In this work, we investigated the degradation of p53 tumour suppressor induced by H. pylori. DESIGN: Expression of p53 protein in gastric biopsies was assessed by immunohistochemistry. Gastric cells were co-cultured with H. pylori strains isolated from high-gastric risk and low-gastric risk areas and assessed for expression of p53, p14ARF and cytotoxin-associated gene A (CagA) by immunoblotting. siRNA was used to inhibit activities of ARF-BP1 and Human Double Minute 2 (HDM2) proteins. RESULTS: Our analysis demonstrated that H. pylori strains expressing high levels of CagA virulence factor and associated with a higher gastric cancer risk more strongly suppress p53 compared with low-risk strains in vivo and in vitro. We found that degradation of p53 induced by bacterial CagA protein is mediated by host HDM2 and ARF-BP1 E3 ubiquitin ligases, while the p14ARF protein counteracts H. pylori-induced signalling. CONCLUSIONS: Our results provide novel evidence that tumorigenicity associated with H. pylori infection is linked to inhibition of p53 protein by CagA. We propose a model in which CagA-induced degradation of p53 protein is determined by a relative level of p14ARF. In cells in which p14ARF levels were decreased due to hypermethylation or deletion of the p14ARF gene, H. pylori efficiently degraded p53, whereas p53 is protected in cells expressing high levels of p14ARF.


Assuntos
Antígenos de Bactérias/fisiologia , Proteínas de Bactérias/fisiologia , Neoplasias Gástricas/microbiologia , Proteína Supressora de Tumor p14ARF/fisiologia , Proteína Supressora de Tumor p53/metabolismo , Antígenos de Bactérias/classificação , Proteínas de Bactérias/classificação , Linhagem Celular Tumoral , Epitélio/metabolismo , Mucosa Gástrica/microbiologia , Humanos , Imuno-Histoquímica , Neoplasias Gástricas/fisiopatologia
9.
Colomb. med ; 44(3): 192-201, July-Sept. 2013. ilus, tab
Artigo em Inglês | LILACS | ID: lil-700502

RESUMO

Abstract Gastric cancer ranks fourth in incidence and second in mortality among all cancers worldwide. Despite the decrease in incidence in some regions of the world, gastric cancer continues to present a major clinical challenge due to most cases being diagnosed in advanced stages with poor prognosis and limited treatment options. The development of gastric cancer is a complex and multifactorial process involving a number of etiological factors and multiple genetic and epigenetic alterations. Among the predisposing factors are: Helicobacter pylori infection, high salt intake, smoking, and in a small percentage of patients, a familial genetic component. More than 95% of stomach cancer cases are adenocarcinomas, which are classified into two major histologic types: intestinal and diffuse. Intestinal type adenocarcinoma is preceded by a sequence of gastric lesions known as Correa´s cascade and is the histologic type associated with the global decrease in gastric cancer rates. Diffuse type adenocarcinomas have a more aggressive behavior and worse prognosis than those of the intestinal type. According to the anatomical location, adenocarcinomas are classified as proximal (originating in the cardia) and distal (originating in the body and antrum). This classification seems to recognize two different clinical entities. Surgical resection of the tumor at an early stage is the only effective treatment method. Therefore, the identification and surveillance of patients at risk may play a significant role in survival rates. Anti-Helicobacter pylori therapy has been shown to be an effective measure in the prevention of gastric cancer.


Resumen El cáncer gástrico ocupa el cuarto lugar en incidencia y el segundo en mortalidad entre todos los cánceres en todo el mundo. A pesar de la disminución de la incidencia en algunas regiones del mundo, el cáncer gástrico continúa siendo un reto clínico debido a que la mayoría de los casos se diagnostican en etapas avanzadas con mal pronóstico y las opciones de tratamiento limitadas. El desarrollo de cáncer gástrico es un proceso complejo y multifactorial que implica un número de factores etiológicos y múltiples alteraciones genéticas y epigenéticas. Entre los factores predisponentes están: infección por Helicobacter pylori, alto consumo de sal, tabaquismo, y en un pequeño porcentaje de los pacientes, un componente genético familiar. Más del 95% de los casos de cáncer de estómago son adenocarcinomas, que se clasifican en dos principales tipos histológicos: intestinal y difuso. Adenocarcinoma de tipo intestinal es precedida por una secuencia de lesiones gástricas conocidas como cascada de Correa y es el tipo histológico asociado con la disminución global de las tasas de cáncer gástrico. Los Adenocarcinomas de tipo difuso tienen un comportamiento más agresivo y peor pronóstico que aquellos del tipo intestinal. De acuerdo con la localización anatómica, los adenocarcinomas se clasifican como proximal (originario en el cardias) y distal (que se origina en el cuerpo y antro). Esta clasificación parece reconocer dos entidades clínicas diferentes. La resección quirúrgica del tumor en una etapa temprana es el método de tratamiento eficaz. Por lo tanto, la identificación y vigilancia de los pacientes de alto riesgo pueden desempeñar un papel importante en las tasas de supervivencia. La terapia anti-Helicobacter pylori ha demostrado ser una medida eficaz en la prevención del cáncer gástrico.

10.
Rev. esp. patol ; 43(1): 8-15, ene.-mar. 2010. tab, ilus
Artigo em Espanhol | IBECS | ID: ibc-79241

RESUMO

AntecedentesEl esófago de Barrett es una reconocida lesión precursora de adenocarcinoma esofágico. Aunque generalmente asociada al reflujo gastroesofágico, los mecanismos patogénicos de la enfermedad no son bien conocidos. El objetivo del presente estudio es explorar la historia natural e identificar marcadores de progreso del proceso precanceroso.Material y métodosSe utilizaron cortes histológicos de 67 especímenes de esófago correspondientes a 14 pacientes con esófago de Barrett, a los que se siguió entre 1–9 años. Se clasificaron las lesiones en: esófago de Barrett sin displasia, indefinido para displasia o con displasia. Se evaluó la expresión de diferentes mucinas en las células caliciformes y en las columnares usando técnicas de histoquímica e inmunohistoquímica.ResultadosEn todos los casos se comprobó la presencia de metaplasia intestinal incompleta. Las células columnares dentro del epitelio metaplásico contenían mucinas neutras. A mayor severidad de la lesión se encontró significativamente menor expresión de sialomucinas en las células columnares (p de tendencia igual a 0,03). En sujetos con lesiones indefinidas para displasia se observó un mayor contenido de sulfomucinas en las células caliciformes (p=0,034) y de MUC2 en las células columnares (p=0,029) que en sujetos con esófago de Barrett sin displasia. Se observó expresión de la mucina intestinal MUC2 y de la mucina gástrica MUC5AC en todas las muestras. MUC6, una mucina de las glándulas profundas gástricas, se presentó ocasionalmente.ConclusiónLa evaluación de los perfiles de mucinas en el esófago de Barrett sugiere una transición gradual del fenotipo del epitelio metaplásico a medida que la lesión avanza en el tiempo(AU)


Background: Barrett’s esophagus is a known precursor lesion of esophageal adenocarcinoma. Although itis generally associated with gastroesophageal reflux, the pathogenic mechanisms of the disease are notwell understood. The aim of this study was to explore the natural history and to identify markers ofprogression of the precancerous process.Material andmethods: Histological sections of 67 esophageal specimens were used in this study. Theywere obtained from 14 subjects with Barrett’s esophagus who were followed from 1 to 9 years. The lesionswere histologically classified as: Barrett’s esophagus without dysplasia,indefinitefordysplasia,ordysplasia. Expression of various mucins in goblet and columnar cells was assessed by histochemistry andimmunohistochemistry.Results: Incomplete intestinal metaplasia was observed in all the specimens. Columnar cells with in themetaplastic epithelium expressed neutral mucins. Sialo mucins were significantly less expressed incolumnar cells as the lesions increased in severity(p trend=0.03). Subjects with indefinite dysplasia lesionshad significantly higher expression of sulphomucins in goblet cells(p=0.034)and of MUC2 in columnarcells (p=0.029)than subjects with Barrett’s esophagus without dysplasia. Expression of the intestinalmucin MUC2 and gastricmucin MUC5AC was observed in all specimens. MUC6, a mucin of the deep gastricglands, was occasionally expressed.Conclusion: The evaluation of the mucin profiles in Barrett’s esophagus suggests a gradual transition of themetaplastic epithelium phenotype as the lesion advances in time(AU)


Assuntos
Humanos , Masculino , Feminino , Pessoa de Meia-Idade , Fenótipo , Mucinas/isolamento & purificação , Esôfago de Barrett/patologia , Adenocarcinoma/patologia , Metaplasia/patologia , Imuno-Histoquímica , Refluxo Gastroesofágico/complicações , Refluxo Gastroesofágico/patologia
11.
Rev Esp Patol ; 48(1): 8-15, 2010 Jan 01.
Artigo em Espanhol | MEDLINE | ID: mdl-21804831

RESUMO

ANTECEDENTES: El esófago de Barrett es una reconocida lesión precursora de adenocarcinoma esofágico. Aunque generalmente asociada al reflujo gastroesofágico, los mecanismos patogénicos de la enfermedad no son bien conocidos. El objetivo del presente estudio es explorar la historia natural e identificar marcadores de progreso del proceso precanceroso. MATERIAL Y M#ENTITYSTARTX000E9;TODOS: Se utilizaron cortes histológicos de 67 especímenes de esófago correspondientes a 14 pacientes con esófago de Barrett, a los que se siguió entre 1 - 9 años. Se clasificaron las lesiones en: esófago de Barrett sin displasia, indefinido para displasia o con displasia. Se evaluó la expresión de diferentes mucinas en las células caliciformes y en las columnares usando técnicas de histoquímica e inmunohistoquímica. RESULTADOS: En todos los casos se comprobó la presencia de metaplasia intestinal incompleta. Las células columnares dentro del epitelio metaplásico contenían mucinas neutras. A mayor severidad de la lesión se encontró significativamente menor expresión de sialomucinas en las células columnares (p de tendencia igual a 0,03). En sujetos con lesiones indefinidas para displasia se observó un mayor contenido de sulfomucinas en las células caliciformes (p=0,034) y de MUC2 en las células columnares (p=0,029) que en sujetos con esófago de Barrett sin displasia. Se observó expresión de la mucina intestinal MUC2 y de la mucina gástrica MUC5AC en todas las muestras. MUC6, una mucina de las glándulas profundas gástricas, se presentó ocasionalmente. CONCLUSI#ENTITYSTARTX000F3;N: La evaluación de los perfiles de mucinas en el esófago de Barrett sugiere una transición gradual del fenotipo del epitelio metaplásico a medida que la lesión avanza en el tiempo.

12.
Cancer Res ; 68(2): 379-87, 2008 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-18199531

RESUMO

Helicobacter pylori is the strongest known risk factor for gastric adenocarcinoma, and strains that possess the cag secretion system, which translocates the bacterial effector CagA into host cells, augment cancer risk. H. pylori strains that express the vacuolating cytotoxin or the outer membrane protein OipA are similarly associated with severe pathologic outcomes. We previously reported that an in vivo adapted H. pylori strain, 7.13, induces gastric adenocarcinoma in rodent models of gastritis. In the current study, we used carcinogenic strain 7.13 as a prototype to define the role of virulence constituents in H. pylori-mediated carcinogenesis. Mongolian gerbils were infected with wild-type strain 7.13 or cagA(-), vacA(-), or oipA(-) mutants for 12 to 52 weeks. All infected gerbils developed gastritis; however, inflammation was significantly attenuated in animals infected with the cagA(-) but not the vacA(-) or oipA(-) strains. Gastric dysplasia and cancer developed in >50% of gerbils infected with either the wild-type or vacA(-) strain but in none of the animals infected with the cagA(-) strain. Inactivation of oipA decreased beta-catenin nuclear localization in vitro and reduced the incidence of cancer in gerbils. OipA expression was detected significantly more frequently among H. pylori strains isolated from human subjects with gastric cancer precursor lesions versus persons with gastritis alone. These results indicate that loss of CagA prevents the development of cancer in this model. Inactivation of oipA attenuates beta-catenin nuclear translocation and also decreases the incidence of carcinoma. In addition to defining factors that mediate H. pylori-induced cancer, these results provide insight into mechanisms that may regulate the development of other malignancies arising within the context of inflammatory states.


Assuntos
Adenocarcinoma/etiologia , Helicobacter pylori/genética , Neoplasias Gástricas/etiologia , Fatores de Virulência/fisiologia , Adenocarcinoma/microbiologia , Adesinas Bacterianas/genética , Adesinas Bacterianas/fisiologia , Adulto , Animais , Antígenos de Bactérias/genética , Antígenos de Bactérias/fisiologia , Aderência Bacteriana/genética , Proteínas da Membrana Bacteriana Externa/genética , Proteínas da Membrana Bacteriana Externa/fisiologia , Proteínas de Bactérias/genética , Proteínas de Bactérias/fisiologia , Transformação Celular Neoplásica/genética , Células Cultivadas , Gastrite/genética , Gastrite/microbiologia , Regulação Bacteriana da Expressão Gênica , Gerbillinae , Humanos , Masculino , Pessoa de Meia-Idade , Proteínas Mutantes/genética , Neoplasias Gástricas/microbiologia , Fatores de Virulência/genética , beta Catenina/metabolismo
13.
Colomb Med (Cali) ; 39(1): 58-65, 2008 Jan.
Artigo em Espanhol | MEDLINE | ID: mdl-25267864

RESUMO

INTRODUCTION: Multiple sampling from different sites of the stomach as well as the number of fragments of gastric mucosa available for histopathologic evaluation are important sources of variation when classifying and grading chronic gastritis. OBJECTIVE: To estimate the sensitivity of the number of fragments of gastric mucosa necessary to establish the diagnosis of atrophic gastritis with intestinal metaplasia, gastric dysplasia and H. pylori infection. In addition, this study will attempt to assess the intra-observer variability in the classification of these premalignant gastric lesions. METHODS: This is a 6 year-cohort study, wherein 1958 gastric endoscopic procedures performed by two gastroenterologists were reviewed. Five gastric biopsy samples were obtained from the antrum, body and lesser curvature during each procedure. One pathologist was in charge of reviewing the five histopathology samples for each subject and providing a definitive diagnosis which was used as the gold standard. Each gastric mucosa sample reviewed led to an individual diagnosis for that sample which was compared with the gold standard. Intra-observer variability was assessed in 127 individuals who correspond to a random sample of 20% of the total endoscopic procedures performed during the 72 month-follow-up. RESULTS: The sensitivity of the diagnosis of intestinal metaplasia (IM) and gastric dysplasia increased proportionally with the number of gastric mucosa samples reviewed. The lesser curvature of the stomach had the highest sensitivity for the diagnosis of IM and dysplasia, among all the stomach regions studied. Just one sample of gastric mucosa attained a sensitivity of 95.9% for the detection of H. pylori infection. The intra-observer agreement for the diagnosis of multifocal atrophic gastritis was 86.1% and the kappa value was 0.79 (95% CI 0.76-0.85). Alcohol-fixed biopsy specimens were inadequate to diagnose H. pylori infection and to assess dysplasia. CONCLUSION: The number of mucosa gastric fragments reviewed, the fixation method used, and the biopsy site are all important factors in order to ensure a correct classification of chronic gastritis.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...