Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Helicobacter ; 24(4): e12598, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31111570

RESUMO

BACKGROUND: Cation transport regulator 1 (CHAC1), a newly discovered enzyme that degrades glutathione, is induced in Helicobacter pylori (H. pylori)-infected gastric epithelial cells in culture. The CHAC1-induced decrease in glutathione leads to an accumulation of reactive oxygen species and somatic mutations in TP53. We evaluated the possible correlation between H. pylori infection and CHAC1 expression in human gastric mucosa. MATERIALS AND METHODS: Both fresh-frozen and formalin-fixed paraffin-embedded tissue samples of gastric mucosa with or without H. pylori infection were obtained from 41 esophageal cancer patients that underwent esophago-gastrectomy. Fresh samples were used for real-time polymerase chain reaction for H. pylori DNA and CHAC1 mRNA, and formalin-fixed samples were used for immunohistochemistry with anti-CHAC1 and anti-H. pylori monoclonal antibodies. Double-enzyme or fluorescence immunohistochemistry and immuno-electron microscopy were used for further analysis. RESULTS: Significant CHAC1 overexpression was detected in H. pylori-infected parietal cells that expressed the human proton pump/H,K-ATPase α subunit, whereas a constitutively low level of CHAC1 mRNA expression was observed in the other samples regardless of the H. pylori infection status, reflecting the weak CHAC1 expression detected by immunohistochemistry in the fundic-gland areas. Immuno-electron microscopy revealed intact H. pylori cells in the secretory canaliculi of infected parietal cells. Some parietal cells exhibited positive nuclear signals for Ki67 in the neck zone of the gastric fundic-gland mucosa with H. pylori infection. CONCLUSION: Cation transport regulator 1 overexpression in H. pylori-infected parietal cells may cause the H. pylori-induced somatic mutations that contribute to the development of gastric cancer.


Assuntos
Mucosa Gástrica/metabolismo , Infecções por Helicobacter/genética , Helicobacter pylori/fisiologia , gama-Glutamilciclotransferase/genética , Mucosa Gástrica/microbiologia , Mucosa Gástrica/patologia , Infecções por Helicobacter/metabolismo , Infecções por Helicobacter/microbiologia , Infecções por Helicobacter/patologia , Humanos , Células Parietais Gástricas/metabolismo , Células Parietais Gástricas/microbiologia , Células Parietais Gástricas/patologia , gama-Glutamilciclotransferase/metabolismo
2.
J Pediatr Gastroenterol Nutr ; 68(6): e99-e104, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30664567

RESUMO

OBJECTIVES: Autoimmune gastritis (AIG) is a chronic inflammatory condition of the gastric mucosa, mainly described in adults presenting with pernicious anemia. It results from antibody-mediated destruction of parietal cells, but the precise initiating event is unknown. The pathogenicity of Helicobacter pylori (H pylori) has been suggested but not established. This study aimed to better characterize AIG in pediatric patients and to address the possible role of H pylori infection. METHODS: Descriptive single-center study, retrospectively describing 20 patients with a diagnosis of AIG based on positivity for anti-parietal cell autoantibodies, in addition to analytical and/or histological findings of oxyntic mucosa atrophy. RESULTS: In the majority (18/20), AIG diagnosis was suggested during investigation of refractory iron-deficient anemia. One patient had dyspepsia and none of the others had gastrointestinal symptoms. Fifty-five percent (11/20) were H pylori positive, but there were no significant differences regarding mean hemoglobin values at presentation (10.6 ±â€Š2.5 vs 9.5 ±â€Š1.0 g/dL, P > 0.05), analytical indicators of gastric atrophy (gastrin, 564.4 ±â€Š184 vs 721.2 ±â€Š220.6 pg/mL, P > 0.05), or in the presence or the grade of oxyntic mucosa atrophy. CONCLUSIONS: Our findings highlight that AIG may have an age-dependent presentation; thus, we can consider a pediatric phenotype that in contrast to adults, is manifested by refractory iron-deficient anemia and associated with parietal cell autoantibody positivity, but not intrinsic factor autoantibodies. A correlation between H pylori and AIG was not evident in the current study and it is still unclear whether H pylori is a trigger for AIG.


Assuntos
Doenças Autoimunes/microbiologia , Gastrite/microbiologia , Infecções por Helicobacter/imunologia , Helicobacter pylori/imunologia , Adolescente , Autoanticorpos/sangue , Doenças Autoimunes/imunologia , Criança , Pré-Escolar , Feminino , Gastrite/imunologia , Infecções por Helicobacter/epidemiologia , Humanos , Masculino , Células Parietais Gástricas/imunologia , Células Parietais Gástricas/microbiologia , Portugal/epidemiologia , Prevalência , Estudos Retrospectivos
4.
J Oral Pathol Med ; 43(8): 576-8, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24761808

RESUMO

OBJECTIVES: Oral lichen planus is a T-cell-mediated mucosal disease of unknown etiology. Numerous predisposing factors have been put forward in the etiology of this disease. This includes stress, drugs, genetic susceptibility, certain viruses, and bacterial infections. Recently, there have been studies published on possible role of Helicobacter pylori infection in pathogenesis of mucocutaneous diseases including oral lichen planus (OLP). The aim of this study was to detect immunohistochemically the presence of Helicobacter pylori in oral lichen planus. MATERIALS AND METHODS: Paraffin-embedded tissue blocks of 50 cases of OLP and 10 cases of normal buccal mucosal biopsies and 6 endoscopic biopsies of patients with peptic ulcer (control group) were sectioned and stained by hematoxylin and eosin. Serial sections of same were stained immunohistochemically using Anti-Helicobacter pylori antibody and observed under microscope for presence or absence of Helicobacter pylori. RESULTS: Except for the control group, none of the cases of OLP and normal buccal mucosal biopsies showed positivity for Helicobacter pylori. CONCLUSION: As we did not detect the presence of Helicobacter pylori in any of the OLP cases, we question the role of these organisms in the pathogenesis of OLP planus if any.


Assuntos
Helicobacter pylori/isolamento & purificação , Líquen Plano Bucal/microbiologia , Adolescente , Adulto , Idoso , Anticorpos Antibacterianos , Criança , Feminino , Mucosa Gástrica/microbiologia , Infecções por Helicobacter/microbiologia , Helicobacter pylori/imunologia , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Mucosa Bucal/microbiologia , Células Parietais Gástricas/microbiologia , Úlcera Péptica/microbiologia , Adulto Jovem
5.
Am J Physiol Gastrointest Liver Physiol ; 306(7): G606-13, 2014 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-24503769

RESUMO

Acute Helicobacter pylori infection of gastric epithelial cells induces CagA oncoprotein- and peptidoglycan (SLT)-dependent mobilization of NF-κB p50 homodimers that bind to H-K-ATPase α-subunit (HKα) promoter and repress HKα gene transcription. This process may facilitate gastric H. pylori colonization by induction of transient hypochlorhydria. We hypothesized that H. pylori also regulates HKα expression posttranscriptionally by miRNA interaction with HKα mRNA. In silico analysis of the HKα 3' untranslated region (UTR) identified miR-1289 as a highly conserved putative HKα-regulatory miRNA. H. pylori infection of AGS cells transfected with HKα 3' UTR-Luc reporter construct repressed luciferase activity by 70%, whereas ΔcagA or Δslt H. pylori infections partially abrogated repression. Transfection of AGS cells expressing HKα 3' UTR-Luc construct with an oligoribonucleotide mimetic of miR-1289 induced maximal repression (54%) of UTR activity within 30 min; UTR activity was unchanged by nontargeting siRNA transfection. Gastric biopsies from patients infected with cagA(+) H. pylori showed a significant increase in miR-1289 expression compared with uninfected patients or those infected with cagA(-) H. pylori. Finally, miR-1289 expression was necessary and sufficient to attenuate biopsy HKα protein expression in the absence of infection. Taken together, these data indicate that miR-1289 is upregulated by H. pylori in a CagA- and SLT-dependent manner and targets HKα 3' UTR, affecting HKα mRNA translation. The sensitivity of HKα mRNA 3' UTR to binding of miR-1289 identifies a novel regulatory mechanism of gastric acid secretion and offers new insights into mechanisms underlying transient H. pylori-induced hypochlorhydria.


Assuntos
Mucosa Gástrica/enzimologia , ATPase Trocadora de Hidrogênio-Potássio/metabolismo , Infecções por Helicobacter/enzimologia , Helicobacter pylori/metabolismo , MicroRNAs/metabolismo , Subunidade p50 de NF-kappa B/metabolismo , Células Parietais Gástricas/enzimologia , Processamento Pós-Transcricional do RNA , Regiões 3' não Traduzidas , Acloridria/enzimologia , Acloridria/microbiologia , Antígenos de Bactérias/genética , Antígenos de Bactérias/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Sítios de Ligação , Linhagem Celular , Mucosa Gástrica/microbiologia , Regulação Enzimológica da Expressão Gênica , Genes Reporter , ATPase Trocadora de Hidrogênio-Potássio/genética , Infecções por Helicobacter/complicações , Infecções por Helicobacter/genética , Infecções por Helicobacter/microbiologia , Helicobacter pylori/genética , Helicobacter pylori/patogenicidade , Interações Hospedeiro-Patógeno , Humanos , Subunidade p50 de NF-kappa B/genética , Células Parietais Gástricas/microbiologia , Peptidoglicano/metabolismo , Interferência de RNA , RNA Mensageiro/metabolismo , Fatores de Tempo , Transfecção , Virulência
6.
Am J Physiol Gastrointest Liver Physiol ; 304(8): G715-22, 2013 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-23392237

RESUMO

Acute Helicobacter pylori infection produces hypochlorhydria. The decrease in acid facilitates survival of the bacterium and its colonization of the stomach. The present study was designed to identify the pathways in oxyntic mucosa by which acute H. pylori infection inhibits acid secretion. In rat fundic sheets in an Ussing chamber, perfusion of the luminal surface with H. pylori in spent broth (10(3)-10(8) cfu/ml) or spent broth alone (1:10(5) to 1:10(0) final dilution) caused a concentration-dependent increase in somatostatin (SST; maximal: 200 ± 20 and 194 ± 9% above basal; P < 0.001) and decrease in histamine secretion (maximal: 45 ± 5 and 48 ± 2% below basal; P < 0.001); the latter was abolished by SST antibody, implying that changes in histamine secretion reflected changes in SST secretion. Both responses were abolished by the axonal blocker tetrodotoxin (TTX), the sensory neurotoxin capsaicin, or the CGRP antagonist CGRP8-37, implying that the reciprocal changes in SST and histamine secretion were due to release of CGRP from sensory neurons. In isolated rabbit oxyntic glands, H. pylori inhibited basal and histamine-stimulated acid secretion in a concentration-dependent manner; the responses were not affected by TTX or SST antibody, implying that H. pylori can directly inhibit parietal cell function. In conclusion, acute administration of H. pylori is capable of inhibiting acid secretion directly as well as indirectly by activating intramural CGRP sensory neurons coupled to stimulation of SST and inhibition of histamine secretion. Activation of neural pathways provides one explanation as to how initial patchy colonization of the superficial gastric mucosa by H. pylori can acutely inhibit acid secretion.


Assuntos
Acloridria/microbiologia , Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Infecções por Helicobacter/metabolismo , Helicobacter pylori/metabolismo , Células Receptoras Sensoriais/metabolismo , Somatostatina/metabolismo , Acloridria/metabolismo , Animais , Peptídeo Relacionado com Gene de Calcitonina/antagonistas & inibidores , Peptídeo Relacionado com Gene de Calcitonina/farmacologia , Modelos Animais de Doenças , Ácido Gástrico/metabolismo , Fundo Gástrico/inervação , Fundo Gástrico/metabolismo , Fundo Gástrico/microbiologia , Mucosa Gástrica/inervação , Mucosa Gástrica/metabolismo , Mucosa Gástrica/microbiologia , Células HeLa , Infecções por Helicobacter/microbiologia , Histamina/metabolismo , Humanos , Células Parietais Gástricas/metabolismo , Células Parietais Gástricas/microbiologia , Fragmentos de Peptídeos/farmacologia , Coelhos , Ratos , Ratos Sprague-Dawley , Células Receptoras Sensoriais/efeitos dos fármacos , Células Receptoras Sensoriais/microbiologia , Bloqueadores dos Canais de Sódio/farmacologia , Somatostatina/farmacologia , Tetrodotoxina/farmacologia
7.
Mol Med Rep ; 7(1): 299-305, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23128938

RESUMO

The aim of the present study was to explore the effects of IL-1ß on the proliferation and apoptosis of gastric epithelial cells and acid secretion from isolated rabbit parietal cells. The mechanisms by which these effects are mediated were also investigated. Parietal cells were isolated from rabbit gastric mucosa by elutriation. The AGS human gastric cancer cell line, the GES-1 human gastric epithelial cell line and parietal cells were treated with interleukin (IL)-1ß in the presence or absence of Helicobacter pylori (H. pylori) for the times indicated. MTT assay and flow cytometry (FCM) were used to determine the levels of proliferation and apoptosis. The expression levels of cyclooxygenase-2 (COX-2) mRNA and protein were examined by RT-PCR and FCM. Acid secretion by parietal cells was examined using 14C-aminopyrine (14C­AP) accumulation. H+/K+ATPase α subunit mRNA expression was assessed by RT-PCR. The results demonstrated that IL-1ß (10 ng/ml) stimul-ated cellular proliferation and inhibited H. pylori-induced apoptosis in GES-1 and AGS cell lines. IL-1ß (10 ng/ml) upregulated the mRNA and protein expression of COX­2 in GES-1 and AGS cells. Acid secretion stimulated by histamine was identified as significantly inhi-bited and mRNA expression of H+/K+ATPase α subunit was downregulated by treatment with IL-1ß (10 ng/ml) for 30 min and 16 h compared with the control in isolated rabbit parietal cells. The present study demonstrates that IL-1ß plays a significant role in H. pylori-induced gastric carcinogenesis through 2 main mechanisms: i) IL-1ß may interfere in gastric epithelial cell growth by upregulating COX-2 expression; ii) IL-1ß may inhibit the acid secretion from parietal cells by downregulating H+/K+ATPase expression.


Assuntos
Apoptose/efeitos dos fármacos , Ácido Gástrico/metabolismo , Interleucina-1beta/farmacologia , Células Parietais Gástricas/efeitos dos fármacos , Células Parietais Gástricas/metabolismo , Animais , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , ATPase Trocadora de Hidrogênio-Potássio/genética , ATPase Trocadora de Hidrogênio-Potássio/metabolismo , Helicobacter pylori/metabolismo , Humanos , Masculino , Células Parietais Gástricas/microbiologia , Coelhos
8.
PLoS One ; 7(11): e50194, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23185574

RESUMO

There is increasing evidence from clinical and population studies for a role of H. pylori infection in the aetiology of iron deficiency. Rodent models of Helicobacter infection are helpful for investigating any causal links and mechanisms of iron deficiency in the host. The aim of this study was to investigate the effects of gastric Helicobacter infection on iron deficiency and host iron metabolism/transport gene expression in hypergastrinemic INS-GAS mice. INS-GAS mice were infected with Helicobacter felis for 3, 6 and 9 months. At post mortem, blood was taken for assessment of iron status and gastric mucosa for pathology, immunohistology and analysis of gene expression. Chronic Helicobacter infection of INS- GAS mice resulted in decreased serum iron, transferrin saturation and hypoferritinemia and increased Total iron binding capacity (TIBC). Decreased serum iron concentrations were associated with a concomitant reduction in the number of parietal cells, strengthening the association between hypochlorhydria and gastric Helicobacter-induced iron deficiency. Infection with H. felis for nine months was associated with decreased gastric expression of iron metabolism regulators hepcidin, Bmp4 and Bmp6 but increased expression of Ferroportin 1, the iron efflux protein, iron absorption genes such as Divalent metal transporter 1, Transferrin receptor 1 and also Lcn2 a siderophore-binding protein. The INS-GAS mouse is therefore a useful model for studying Helicobacter-induced iron deficiency. Furthermore, the marked changes in expression of gastric iron transporters following Helicobacter infection may be relevant to the more rapid development of carcinogenesis in the Helicobacter infected INS-GAS model.


Assuntos
Anemia Ferropriva/metabolismo , Infecções por Helicobacter/metabolismo , Helicobacter felis/metabolismo , Ferro/metabolismo , Células Parietais Gástricas/metabolismo , Proteínas de Fase Aguda/genética , Proteínas de Fase Aguda/metabolismo , Anemia Ferropriva/complicações , Anemia Ferropriva/microbiologia , Anemia Ferropriva/patologia , Animais , Peptídeos Catiônicos Antimicrobianos/genética , Peptídeos Catiônicos Antimicrobianos/metabolismo , Proteína Morfogenética Óssea 4/genética , Proteína Morfogenética Óssea 4/metabolismo , Proteína Morfogenética Óssea 6/genética , Proteína Morfogenética Óssea 6/metabolismo , Proteínas de Transporte de Cátions/genética , Proteínas de Transporte de Cátions/metabolismo , Gastrinas/genética , Regulação da Expressão Gênica , Infecções por Helicobacter/complicações , Infecções por Helicobacter/microbiologia , Infecções por Helicobacter/patologia , Helicobacter felis/patogenicidade , Hepcidinas , Insulina/genética , Lipocalina-2 , Lipocalinas/genética , Lipocalinas/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Proteínas Oncogênicas/genética , Proteínas Oncogênicas/metabolismo , Células Parietais Gástricas/microbiologia , Células Parietais Gástricas/patologia , Receptores da Transferrina/genética , Receptores da Transferrina/metabolismo , Transdução de Sinais
9.
Am J Physiol Gastrointest Liver Physiol ; 303(8): G915-26, 2012 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-22899822

RESUMO

CD24 is expressed in the putative stem cells within several tissues and is overexpressed in gastric and colonic adenocarcinomas. Perturbed CD24 expression may therefore alter the response of gastrointestinal epithelia to damage-inducing stimuli that induce cancer. We have investigated the effects of CD24 deletion on gastric responses to Helicobacter felis infection and γ-irradiation using CD24-null mice. Gastric CD24 expression was determined by immunohistochemistry in C57BL/6 mice. Female CD24-null and C57BL/6 mice were infected with H. felis for 6 wk, and inflammation, proliferation, apoptosis, and parietal cell numbers were assessed in gastric tissue sections. Apoptosis and proliferation were analyzed on a cell-positional basis in stomach, small intestine, and colon of CD24-null and C57BL/6 mice following γ-irradiation. Apoptosis was also assessed in HT29 cells following CD24 siRNA transfection. Of CD24-positive cells in the gastric corpus, 98% were H(+)-K(+)-ATPase-expressing parietal cells. CD24-null mice showed more prominent gastric H. felis colonization than C57BL/6 mice but displayed a marked reduction in corpus inflammation, reduced Ki67 labeling, and less gastric atrophy 6 wk following infection. Corpus apoptosis was elevated in CD24-null mice, but this did not increase further with H. felis infection as observed in C57BL/6 mice. More apoptotic cells were found following γ-irradiation in the stomach, small intestine, and colon of CD24-null mice and following CD24 knockdown in vitro. In conclusion, CD24 is expressed in gastric parietal cells, where it modulates gastric responses to H. felis and γ-radiation. CD24 also regulates susceptibility to apoptosis in the distal murine gastrointestinal tract.


Assuntos
Apoptose/fisiologia , Antígeno CD24/metabolismo , Mucosa Gástrica/metabolismo , Infecções por Helicobacter/metabolismo , Células Parietais Gástricas/metabolismo , Animais , Antígeno CD24/genética , Feminino , Raios gama , Mucosa Gástrica/microbiologia , Mucosa Gástrica/efeitos da radiação , Infecções por Helicobacter/genética , Infecções por Helicobacter/patologia , Helicobacter felis , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Parietais Gástricas/microbiologia , Células Parietais Gástricas/efeitos da radiação , Estômago/microbiologia , Estômago/efeitos da radiação
10.
Turk J Gastroenterol ; 22(2): 134-8, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21796548

RESUMO

BACKGROUND/AIMS: Helicobacter pylori-associated corpus atrophy and autoimmune gastric atrophy share similar histopathologic and clinical aspects. In our study, the relation between Helicobacter pylori and autoimmune gastritis was investigated. METHODS: Eighty-two consecutive histologically and serologically Helicobacter pylori-positive and 96 Helicobacter pylori-negative patients were enrolled in the study. All patients underwent diagnostic upper esophagogastroduodenal endoscopy. Three biopsy specimens from the antrum and corpus greater curvature were obtained for histologic evaluation. Serum samples were collected for detection of anti-parietal cell antibody, anti-Helicobacter pylori IgG and vitamin B12. Statistical analyses were determined with Student t-test and chi-square test. Statistical significance was determined with a p-value <0.05. RESULTS: Of 82 Helicobacter pylori-positive patients, 45 were female and 36 were male, with a mean age 45.1 ± 15.1. There was no significant difference in age, gender and corpus atrophy between the Helicobacter pylori-positive and -negative groups. Eleven Helicobacter pylori-positive patients (13.4%) and 14 (14.6%) Helicobacter pylori-negative patients were positive for anti-parietal cell antibody; the difference between the two groups was not statistically significant (p>0.05). Differences in esophagogastroduodenal endoscopy findings, antrum and corpus inflammation, antrum and corpus atrophy, and vitamin B12 levels were found to be insignificant between parietal cell antibody-positive and -negative groups (p>0.05). CONCLUSIONS: We did not find any relation between Helicobacter pylori infection and anti-parietal cell antibody, a marker of autoimmune gastritis. Long-term follow-up of Helicobacter pylori-infected patients and also determination of the relation between eradication of Helicobacter pylori and autoimmune atrophic gastritis are needed.


Assuntos
Doenças Autoimunes/epidemiologia , Gastrite/epidemiologia , Infecções por Helicobacter/epidemiologia , Helicobacter pylori/imunologia , Adulto , Idoso , Anticorpos Antibacterianos/sangue , Autoanticorpos/sangue , Doenças Autoimunes/imunologia , Doenças Autoimunes/microbiologia , Estudos de Casos e Controles , Di-Hidrotaquisterol , Feminino , Mucosa Gástrica/imunologia , Mucosa Gástrica/microbiologia , Mucosa Gástrica/patologia , Gastrite/imunologia , Gastrite/patologia , Infecções por Helicobacter/imunologia , Infecções por Helicobacter/patologia , Humanos , Imunoglobulina G/sangue , Masculino , Pessoa de Meia-Idade , Células Parietais Gástricas/imunologia , Células Parietais Gástricas/microbiologia , Prevalência , Fatores de Risco , Estudos Soroepidemiológicos , Vitamina B 12/sangue
11.
Vet Res ; 42: 42, 2011 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-21366900

RESUMO

In dogs Helicobacter spp. are found in all gastric regions usually localized in the surface mucus, gastric glands and parietal cells. The aim of this study was to detail the distribution of Helicobacter spp. in the fundic mucosa of asymptomatic Beagle dogs and their intracellular localization within parietal cells, in order to evaluate species-specific pathogenetic effects on gastric cells. The presence of Helicobacter spp. was investigated by immunohistochemistry, TEM, and PCR in the fundic mucosa of six Beagle dogs. Helicobacter spp. were found in all dogs examined, and H. bizzozeronii and H. felis were identified by PCR and confirmed by TEM. In the lumen of the fundic glands, co-localization was common. H. bizzozeronii was present in larger numbers than H. felis in both intraluminal and intraparietal localization. The amounts of H. bizzozeronii were similar in superficial and basal portions of the glands. H. felis was predominantly localized in the superficial portions of gastric glands but almost absent from the base. Within parietal cells, most Helicobacter organisms were intracanalicular, but intact and degenerate Helicobacter organisms were also visualized free in the cytoplasm or in secondary lysosomes. No specific degenerative lesions were found in infected parietal cells. Helicobacter organisms were also observed within macrophages in the lamina propria. In conclusion, there is a differential distribution of H. bizzozeronii and H. felis in the fundic mucosa of Beagle dogs, and their intracellular localization in parietal cells and macrophages suggests novel pathogenic scenarios for the development of immune response and maintenance of chronic gastritis in dogs.


Assuntos
Doenças do Cão/microbiologia , Mucosa Gástrica/microbiologia , Infecções por Helicobacter/veterinária , Helicobacter/fisiologia , Animais , Doenças do Cão/imunologia , Doenças do Cão/patologia , Cães , Feminino , Mucosa Gástrica/patologia , Mucosa Gástrica/ultraestrutura , Gastrite/imunologia , Gastrite/microbiologia , Gastrite/patologia , Gastrite/veterinária , Helicobacter/genética , Helicobacter/isolamento & purificação , Infecções por Helicobacter/imunologia , Infecções por Helicobacter/microbiologia , Infecções por Helicobacter/patologia , Imuno-Histoquímica/veterinária , Masculino , Microscopia Eletrônica de Transmissão/veterinária , Células Parietais Gástricas/microbiologia , Células Parietais Gástricas/patologia , Células Parietais Gástricas/ultraestrutura , Reação em Cadeia da Polimerase/veterinária , Especificidade da Espécie
12.
PLoS One ; 5(11): e14083, 2010 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-21124878

RESUMO

BACKGROUND: "Helicobacter (H.) heilmannii" type 1 is the most prevalent gastric non-H. pylori Helicobacter species in humans suffering from gastric disease. It has been shown to be identical to H. suis, a bacterium which is mainly associated with pigs. To obtain better insights into the long-term pathogenesis of infections with this micro-organism, experimental infections were carried out in different rodent models. METHODOLOGY/PRINCIPAL FINDINGS: Mongolian gerbils and mice of two strains (BALB/c and C57BL/6) were infected with H. suis and sacrificed at 3 weeks, 9 weeks and 8 months after infection. Gastric tissue samples were collected for PCR analysis, histological and ultrastructural examination. In gerbils, bacteria mainly colonized the antrum and a narrow zone in the fundus near the forestomach/stomach transition zone. In both mice strains, bacteria colonized the entire glandular stomach. Colonization with H. suis was associated with necrosis of parietal cells in all three animal strains. From 9 weeks after infection onwards, an increased proliferation rate of mucosal epithelial cells was detected in the stomach regions colonized with H. suis. Most gerbils showed a marked lymphocytic infiltration in the antrum and in the forestomach/stomach transition zone, becoming more pronounced in the course of time. At 8 months post infection, severe destruction of the normal antral architecture at the inflamed sites and development of mucosa-associated lymphoid tissue (MALT) lymphoma-like lesions were observed in some gerbils. In mice, the inflammatory response was less pronounced than in gerbils, consisting mainly of mononuclear cell infiltration and being most severe in the fundus. CONCLUSIONS/SIGNIFICANCE: H. suis causes death of parietal cells, epithelial cell hyperproliferation and severe inflammation in mice and Mongolian gerbil models of human gastric disease. Moreover, MALT lymphoma-like lesions were induced in H. suis-infected Mongolian gerbils. Therefore, the possible involvement of this micro-organism in human gastric disease should not be neglected.


Assuntos
Mucosa Gástrica/microbiologia , Helicobacter heilmannii/fisiologia , Gastropatias/microbiologia , Estômago/microbiologia , Animais , Proliferação de Células , Modelos Animais de Doenças , Mucosa Gástrica/patologia , Mucosa Gástrica/ultraestrutura , Gerbillinae , Centro Germinativo/microbiologia , Centro Germinativo/patologia , ATPase Trocadora de Hidrogênio-Potássio/metabolismo , Interações Hospedeiro-Patógeno , Humanos , Imuno-Histoquímica , Linfoma de Zona Marginal Tipo Células B/etiologia , Linfoma de Zona Marginal Tipo Células B/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Microscopia Eletrônica de Transmissão , Células Parietais Gástricas/metabolismo , Células Parietais Gástricas/microbiologia , Células Parietais Gástricas/patologia , Especificidade da Espécie , Estômago/patologia , Gastropatias/complicações , Gastropatias/patologia
13.
Am J Physiol Gastrointest Liver Physiol ; 297(2): G312-22, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19520737

RESUMO

Helicobacter pylori is a major cause of the transdifferentiation into intestinal metaplasia that may develop gastric cancer. However, the molecular pathogenesis of this transdifferentiation is poorly understood. A SRY-related HMG box protein Sox2 is an essential transcription factor of organ development in brain, lung, and stomach. Our aim of this study was to investigate the mechanism responsible for regulation of Sox2 in host Th1-dominant response to H. pylori. Sox2 protein was immunohistochemically expressed in both human oxyntic and pyloric glands with H. pylori infection, but not in intestinal metaplasia. Western immunoblotting of gastric epithelial cell lines showed that IL-4, a Th2-related cytokine, dose dependently enhanced Sox2 expression among H. pylori infection-mediated cytokines. Small changes of Sox2 expression were observed after each treatment with IFN-gamma, IL-1beta, or TNF-alpha. IL-4-mediated Sox2 induction was suppressed by the inhibition of STAT6 activation with STAT6 RNA interference, and electrophoretic mobility shift assay indicated that activation of the Sox2 promoter by IL-4 occurred through the action of STAT6. Furthermore, H. pylori and IFN-gamma inhibited the phosphorylation of STAT6, resulting in the suppression of IL-4-mediated Sox2 expression. Immunohistochemical analyses showed significantly the suppressed STAT6 activity in H. pylori-infected human gastric mucosa. Additionally, downregulation of Sox2 by knockdown experiments led to intestinal phenotype with expressions of Cdx2 and MUC2. These results suggest that H. pylori and IFN-gamma interfere with the differentiation into oxyntic and pyloric glands by the downregulation of Sox2 on IL-4/STAT6 signaling, which may contribute to the transdifferentiation into intestinal metaplasia.


Assuntos
Transdiferenciação Celular , Mucosa Gástrica/metabolismo , Infecções por Helicobacter/metabolismo , Helicobacter pylori , Interleucina-4/metabolismo , Lesões Pré-Cancerosas/metabolismo , Fatores de Transcrição SOXB1/metabolismo , Neoplasias Gástricas/metabolismo , Sequência de Bases , Sítios de Ligação , Fator de Transcrição CDX2 , Regulação para Baixo , Mucosa Gástrica/microbiologia , Mucosa Gástrica/patologia , Infecções por Helicobacter/microbiologia , Infecções por Helicobacter/patologia , Proteínas de Homeodomínio/metabolismo , Humanos , Imuno-Histoquímica , Interferon gama/metabolismo , Interleucina-1beta/metabolismo , Antígeno Ki-67/metabolismo , Metaplasia , Dados de Sequência Molecular , Mucina-2/metabolismo , Células Parietais Gástricas/metabolismo , Células Parietais Gástricas/microbiologia , Células Parietais Gástricas/patologia , Fosforilação , Lesões Pré-Cancerosas/microbiologia , Lesões Pré-Cancerosas/patologia , Regiões Promotoras Genéticas , Interferência de RNA , Fatores de Transcrição SOXB1/genética , Fator de Transcrição STAT6/metabolismo , Transdução de Sinais , Neoplasias Gástricas/microbiologia , Neoplasias Gástricas/patologia , Fatores de Tempo , Fator de Necrose Tumoral alfa/metabolismo
14.
J Biol Chem ; 283(39): 26714-25, 2008 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-18625712

RESUMO

Helicobacter pylori persistently colonize the human stomach and have been linked to atrophic gastritis and gastric carcinoma. Although it is well known that H. pylori infection can result in hypochlorhydria, the molecular mechanisms underlying this phenomenon remain poorly understood. Here we show that VacA permeabilizes the apical membrane of gastric parietal cells and induces hypochlorhydria. The functional consequences of VacA infection on parietal cell physiology were studied using freshly isolated rabbit gastric glands and cultured parietal cells. Secretory activity of parietal cells was judged by an aminopyrine uptake assay and confocal microscopic examination. VacA permeabilization induces an influx of extracellular calcium, followed by activation of calpain and subsequent proteolysis of ezrin at Met(469)-Thr(470), which results in the liberation of ezrin from the apical membrane of the parietal cells. VacA treatment inhibits acid secretion by preventing the recruitment of H,K-ATPase-containing tubulovesicles to the apical membrane of gastric parietal cells. Electron microscopic examination revealed that VacA treatment disrupts the radial arrangement of actin filaments in apical microvilli due to the loss of ezrin integrity in parietal cells. Significantly, expression of calpain-resistant ezrin restored the functional activity of parietal cells in the presence of VacA. Proteolysis of ezrin in VacA-infected parietal cells is a novel mechanism underlying H. pylori-induced inhibition of acid secretion. Our results indicate that VacA disrupts the apical membrane-cytoskeletal interactions in gastric parietal cells and thereby causes hypochlorhydria.


Assuntos
Acloridria/metabolismo , Citoesqueleto de Actina/metabolismo , Proteínas de Bactérias/metabolismo , Membrana Celular/metabolismo , Infecções por Helicobacter/metabolismo , Helicobacter pylori/metabolismo , Células Parietais Gástricas/metabolismo , Acloridria/microbiologia , Citoesqueleto de Actina/ultraestrutura , Animais , Cálcio/metabolismo , Calpaína/metabolismo , Membrana Celular/microbiologia , Membrana Celular/ultraestrutura , Permeabilidade da Membrana Celular , Células Cultivadas , Proteínas do Citoesqueleto/metabolismo , Gastrite Atrófica/metabolismo , Gastrite Atrófica/microbiologia , Gastrite Atrófica/patologia , Infecções por Helicobacter/microbiologia , Infecções por Helicobacter/patologia , Humanos , Células Parietais Gástricas/microbiologia , Células Parietais Gástricas/ultraestrutura , Coelhos , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/microbiologia , Neoplasias Gástricas/ultraestrutura
15.
Gastroenterology ; 133(4): 1210-8, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17919495

RESUMO

BACKGROUND & AIMS: The MUC1 mucin is expressed on the cell surface of epithelial cells lining the gastric mucosa. Epidemiologic studies suggest that functional allelic variations in the MUC1 gene may play a role in human susceptibility to Helicobacter pylori-associated pathologies, including gastric adenocarcinoma. We have evaluated the impact of Muc1 expression on the colonization and pathogenesis of gastric Helicobacter infections. METHODS: Wild-type and Muc1-deficient mice were infected with H pylori and colonization and gastritis levels determined. Primary gastric cells were used to examine the impact of Muc1 expression on bacterial adherence. RESULTS: Mice lacking Muc1 were colonized by 5-fold more H pylori within 1 day of infection, and this difference was maintained for at least 2 months postinfection. Mice heterozygous for the null Muc1 allele developed intermediate bacterial colonization. Although wild-type mice developed only a mild gastritis when infected for 2 months with H pylori, Muc1(-/-) mice developed an atrophic gastritis marked by loss of parietal cells. We demonstrate H pylori adhesion to purified MUC1 and significantly increased adhesion to cultured murine Muc1 null gastric epithelial cells, suggesting that Muc1 acts as a decoy limiting binding to the cell surface. CONCLUSIONS: Muc1 provides a protective barrier, which limits both acute and chronic colonization by H pylori, as well as playing a major role in limiting the inflammation induced by Helicobacter infection. We propose that Muc1 restricts access of H pylori to the epithelial surface, hence reducing exposure of the host to proinflammatory bacterial products.


Assuntos
Mucosa Gástrica/microbiologia , Gastrite/microbiologia , Infecções por Helicobacter/microbiologia , Helicobacter pylori/patogenicidade , Mucina-1/metabolismo , Animais , Formação de Anticorpos , Aderência Bacteriana , Linhagem Celular Tumoral , Células Cultivadas , Contagem de Colônia Microbiana , Modelos Animais de Doenças , Mucosa Gástrica/metabolismo , Mucosa Gástrica/patologia , Gastrite/metabolismo , Gastrite/patologia , Gastrite/prevenção & controle , Infecções por Helicobacter/complicações , Infecções por Helicobacter/imunologia , Infecções por Helicobacter/metabolismo , Infecções por Helicobacter/patologia , Helicobacter pylori/isolamento & purificação , Humanos , Imunidade Celular , Camundongos , Camundongos Knockout , Mucina-1/genética , Células Parietais Gástricas/metabolismo , Células Parietais Gástricas/microbiologia , Ligação Proteica , Índice de Gravidade de Doença , Fatores de Tempo
16.
FEMS Immunol Med Microbiol ; 50(2): 268-72, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17488330

RESUMO

Helicobacter heilmannii has been reported to cause gastric low-grade mucosa-associated lymphoid tissue-type (MALT) lymphoma, but its precise pathophysiological mechanism remains to be clarified. We recently established a model of gastric B-cell MALT lymphoma in C57BL/6 mice by means of peroral infection of H. heilmannii primarily obtained from cynomolgus monkeys. Using this model, macroscopic, immunohistochemical, and electron microscopic observations of MALT lymphomas were carried out in order to examine the development of apoptosis and angiogenesis. Enhancement of the microvascular network and an increase in vascular endothelial growth factor-A were detected in the central region of the MALT lymphoma tissue in the infected mouse stomach, while vascular endothelial growth factor-C was detected at the margins of the MALT lymphomas. In addition, many H. heilmannii-invaded parietal cells showed caspase-3 immunoreactivity in the fundic mucosal tissue surrounding the MALT lymphoma. In conclusion, in H. heilmannii-induced MALT lymphoma, enhanced immunoreactivity of vascular endothelial growth factor-A and factor-C was observed in areas encircled by increased parietal cell apoptosis, which indicates the pathophysiological relevance of both angiogenesis and apoptosis in MALT lymphoma formation.


Assuntos
Apoptose , Mucosa Gástrica/patologia , Infecções por Helicobacter/complicações , Helicobacter heilmannii/crescimento & desenvolvimento , Linfoma de Zona Marginal Tipo Células B/patologia , Neovascularização Patológica , Animais , Caspase 3/análise , Modelos Animais de Doenças , Infecções por Helicobacter/patologia , Helicobacter heilmannii/isolamento & purificação , Imuno-Histoquímica , Linfoma de Zona Marginal Tipo Células B/microbiologia , Macaca fascicularis , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Eletrônica de Transmissão , Células Parietais Gástricas/microbiologia , Fator A de Crescimento do Endotélio Vascular/análise , Fator C de Crescimento do Endotélio Vascular/análise
17.
J Comp Pathol ; 135(4): 226-36, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17069831

RESUMO

In contrast to Helicobacter(H.) pylori, little is known about the pathogenic mechanisms of gastric non-H. pylori Helicobacter species. Mongolian gerbils were inoculated intragastrically with H. felis or H. bizzozeronii and killed at different timepoints post-inoculation (p.i.), stomach tissue being taken for light and transmission electron microscopy (TEM) and polymerase chain reaction (PCR) analysis. Parietal cells (PCs), apoptosis, cell proliferation and nuclear factor-kappaB (NF-kappaB) activation were "visualized" immunohistochemically. Inflammation consisted of neutrophilic granulocytes, mainly in the antrum, and lymphocytic infiltrates around the limiting ridge and throughout the stomach mucosa and submucosa. From day 11 p.i. onwards, H. felis-inoculated animals showed moderate to severe loss of PCs extending from the limiting ridge into the fundus. Apoptotic cells, spiral bacteria, cell proliferation, and NF-kappaB activation were detected at the transition zone between affected and normal PCs. TEM revealed interaction of H. felis flagella with PCs and chief cells. Moreover, H. felis was seen in proximity to, and inside, necrotic cells. At 10 weeks p.i., some H. felis-infected gerbils showed complete loss of fundic glands, and mucous metaplasia of the epithelium. H. bizzozeronii, which made no flagellar contact with epithelial cells, was associated with only mild PC loss. The mechanism by which H. felis induces PC necrosis and apoptosis remains unclear. The observed flagellar contact and NF-kappaB activation may play an important role in H. felis-associated inflammation.


Assuntos
Mucosa Gástrica/microbiologia , Gerbillinae , Infecções por Helicobacter/patologia , Helicobacter felis , Helicobacter , Células Parietais Gástricas/microbiologia , Animais , Modelos Animais de Doenças , Feminino , Mucosa Gástrica/anatomia & histologia , Mucosa Gástrica/patologia , Infecções por Helicobacter/induzido quimicamente , Técnicas Histológicas , Imuno-Histoquímica , Microscopia Eletrônica de Transmissão , Células Parietais Gástricas/patologia
18.
Microbes Infect ; 8(2): 503-10, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16311055

RESUMO

Non-pylori helicobacter infections are associated with gastritis, gastric ulcers and MALT lymphomas in man. Approximately 50% of these are caused by helicobacters commonly found in dogs and cats, including Helicobacter felis, Helicobacter bizzozeronii and H. salomonis. In contrast to Helicobacter pylori, the virulence mechanisms of these species are unknown. In this study the virulence of H. felis, H. bizzozeronii and H. salomonis was investigated in Mongolian gerbils. Female SPF gerbils were inoculated intragastrically with H. felis, H. bizzozeronii or H. salomonis and sacrificed 3 weeks later. Fundus and antrum samples were taken for bacterial detection by PCR. A longitudinal strip covering all stomach regions was taken for histology. Gastric colonization, inflammation, apoptosis, loss of parietal cells and cell proliferation were assessed. Controls and H. salomonis inoculated gerbils were negative in PCR. H. felis and H. bizzozeronii inoculated animals were positive. H. felis inoculated animals showed loss of parietal cells extending from the limiting ridge into the fundus. A high cell proliferation rate was noticed in the mucosal area devoid of parietal cells. A dense band of apoptotic cells and large numbers of Helicobacter bacteria were seen at the transition zone between affected and normal parietal cells. In H. bizzozeronii infected gerbils, this was less pronounced. Focal apoptotic loss of gastric epithelial cells was spatially associated with the presence of bacteria especially in H. felis and to a lesser extent in H. bizzozeronii infected gerbils. This loss of cells may lead to intestinal metaplasia.


Assuntos
Infecções por Helicobacter/patologia , Helicobacter felis/patogenicidade , Helicobacter/patogenicidade , Células Parietais Gástricas/patologia , Animais , DNA Bacteriano/análise , Feminino , Gerbillinae , Helicobacter/classificação , Helicobacter/genética , Infecções por Helicobacter/microbiologia , Infecções por Helicobacter/fisiopatologia , Helicobacter felis/genética , Helicobacter felis/isolamento & purificação , Células Parietais Gástricas/microbiologia , Reação em Cadeia da Polimerase , Organismos Livres de Patógenos Específicos
19.
Scand J Clin Lab Invest ; 65(4): 291-9, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16076684

RESUMO

OBJECTIVE: Low serum pepsinogen I (PG I) values are common in subjects with advanced corpus atrophy with or without parietal cell antibodies (PCA). Elevated values are usual during Helicobacter pylori infection. MATERIAL AND METHODS: PG I levels were determined in two randomly selected cross-sectional adult population samples using the Gastroset PGI test kits. The sera (408 in 1973 and 504 in 1994), tested earlier for H. pylori infection and now for PCA, represented subjects living in Vammala, Finland. RESULTS: In the PCA-negative population, the mean (+/-SD) PG I level was significantly higher in men than in women among both H. pylori-negative (88.13+/-34.16 microg/l versus 72.43+/-29.31 microg/l; p<0.0001) and H. pylori-positive (110.50+/-50.59 microg/l, versus 97.74+/-44.82 microg/l, p<0.0001) subjects; the difference between all H. pylori-positive and -negative subjects was also significant (p<0.001). In the 10-year age groups, age had no impact on the mean PG I levels in H. pylori-negative subjects (p=0.860). In the PCA-positive population, the 10 H. pylori-positive subjects had higher mean PG I levels (112.96+/-53.62 microg/l) than the 13 H. pylori-negative subjects (32.57+/-27.59 microg/l; p=0.002); the latter mean was also significantly lower than that of the PCA- and H. pylori-negative subjects (80.08+/-32.69 microg/l; p<0.0001). CONCLUSIONS: Men had higher normal PG I values than women, but there was no significant variation by age. H. pylori infection was associated with elevated PG I levels and a small decrease with increasing age. Non-infected PCA-positive subjects showed the lowest mean PG I level.


Assuntos
Autoanticorpos/sangue , Infecções por Helicobacter/sangue , Helicobacter pylori , Células Parietais Gástricas/imunologia , Pepsinogênio A/sangue , Kit de Reagentes para Diagnóstico/normas , Adolescente , Adulto , Fatores Etários , Idoso , Biomarcadores , Química Clínica/normas , Estudos Transversais , Feminino , Gastrite Atrófica/imunologia , Gastrite Atrófica/microbiologia , Infecções por Helicobacter/imunologia , Humanos , Masculino , Pessoa de Meia-Idade , Células Parietais Gástricas/microbiologia , Valores de Referência , Fatores Sexuais
20.
Vestn Ross Akad Med Nauk ; (4): 23-8, 2004.
Artigo em Russo | MEDLINE | ID: mdl-15154325

RESUMO

Microbiological examinations of the human-colon parietal mucin were made in different sections of the human gastrointestinal tract (GIT). Biopsy samples of the human colon mucus tunic were used as the research material. Convincing data were found by research on the importance of the anatomic-and-morphological factor in shaping-up of microbiocenosis of the colon-wall parietal mucin. It was established that, depending on a GIT section, not only the quantitative and qualitative composition of normal microflora but also the nature of inter-microbe interactions are subject to changes. In particular, biopsy materials of the small-intestine parietal mucin are much different from that of the colon. The colon section from the ascending colon to the sigmoid colon has, with some exceptions, an identical microbial composition. At the same time, the rectum is significantly different from other colon sections. The results are suggestive of the below cluster-type pattern of parietal biological material: microorganisms are clustered as small domains with certain specific and quantitative compositions. It was established that, although feces and parietal mucin have a similar species composition of microorganisms, still, there is a number of essential differences between them in as far as the frequency ratio and the microorganism concentration are concerned, which signifies a certain degree of isolation of the above biopsy materials. Finally, a certain isolation degree of the feces biopsy materials and of the parietal ones was established.


Assuntos
Trato Gastrointestinal/metabolismo , Trato Gastrointestinal/patologia , Mucinas/metabolismo , Células Parietais Gástricas/metabolismo , Células Parietais Gástricas/patologia , Bactérias/classificação , Bactérias/isolamento & purificação , Trato Gastrointestinal/microbiologia , Humanos , Células Parietais Gástricas/microbiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA