Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Infect Immun ; 80(3): 921-8, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22184416

RESUMO

Helicobacter cinaedi, a common human intestinal bacterium, has been implicated in various enteric and systemic diseases in normal and immunocompromised patients. Protection against oxidative stress is a crucial component of bacterium-host interactions. Alkyl hydroperoxide reductase C (AhpC) is an enzyme responsible for detoxification of peroxides and is important in protection from peroxide-induced stress. H. cinaedi possesses a single ahpC, which was investigated with respect to its role in bacterial survival during oxidative stress. The H. cinaedi ahpC mutant had diminished resistance to organic hydroperoxide toxicity but increased hydrogen peroxide resistance compared with the wild-type (WT) strain. The mutant also exhibited an oxygen-sensitive phenotype and was more susceptible to killing by macrophages than the WT strain. In vivo experiments in BALB/c and BALB/c interleukin-10 (IL-10)(-/-) mice revealed that the cecal colonizing ability of the ahpC mutant was significantly reduced. The mutant also had diminished ability to induce bacterium-specific immune responses in vivo, as shown by immunoglobulin (IgG2a and IgG1) serum levels. Collectively, these data suggest that H. cinaedi ahpC not only contributes to protecting the organism against oxidative stress but also alters its pathogenic properties in vivo.


Assuntos
Proteínas de Bactérias/metabolismo , Infecções por Helicobacter/microbiologia , Helicobacter/patogenicidade , Interleucina-10/imunologia , Viabilidade Microbiana , Estresse Oxidativo , Peroxidases/metabolismo , Estresse Fisiológico , Animais , Carga Bacteriana , Proteínas de Bactérias/genética , Ceco/microbiologia , Feminino , Deleção de Genes , Helicobacter/efeitos dos fármacos , Helicobacter/enzimologia , Interações Hospedeiro-Patógeno , Peróxido de Hidrogênio/toxicidade , Interleucina-10/deficiência , Macrófagos/imunologia , Macrófagos/microbiologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Oxigênio/toxicidade , Peroxidases/genética , Peroxirredoxinas/genética , Peroxirredoxinas/metabolismo , Fatores de Virulência/genética , Fatores de Virulência/metabolismo
2.
Immunology ; 133(2): 165-78, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21426337

RESUMO

To model inflammatory bowel disease, we assessed infection with Helicobacter hepaticus 3B1 (ATCC 51449) and a potential probiotic Lactobacillus reuteri (ATCC PTA-6475) in gnotobiotic B6.129P2-IL-10(tm1Cgn) (IL-10(-/-) ) mice. No typhlocolitis developed in germ-free controls (n=21) or in L. reuteri (n=8) or H. hepaticus (n=18) mono-associated mice for 20 weeks post-infection. As positive controls, three specific pathogen-free IL-10(-/-) mice dosed with H. hepaticus developed severe typhlocolitis within 11 weeks. Because L. reuteri PTA-6475 has anti-inflammatory properties in vitro, it was unexpected to observe significant typhlocolitis (P<0·0001) in mice that had been infected with L. reuteri followed in 1 week by H. hepaticus (n=16). The H. hepaticus colonization was not affected through 20 weeks post-infection but L. reuteri colonization was lower in co-infected compared with L. reuteri mono-associated mice at 8-11 weeks post-infection (P<0·05). Typhlocolitis was associated with an increased T helper type 1 serum IgG2c response to H. hepaticus in co-infected mice compared with H. hepaticus mono-associated mice (P<0·005) and similarly, mRNA expression in caecal-colonic tissue was elevated at least twofold for chemokine ligands and pro-inflammatory interleukin-1α (IL-1α), IL-1ß, IL-12 receptor, tumour necrosis factor-α and inducible nitric oxide synthase. Anti-inflammatory transforming growth factor-ß, lactotransferrin, peptidoglycan recognition proteins, Toll-like receptors 4, 6, 8 and particularly 9 gene expression, were also elevated only in co-infected mice (P<0·05). These data support that the development of typhlocolitis in H. hepaticus-infected IL-10(-/-) mice required co-colonization with other microbiota and in this study, required only L. reuteri. Although the effects other microbiota may have on H. hepaticus virulence properties remain speculative, further investigations using this gnotobiotic model are now possible.


Assuntos
Vida Livre de Germes , Infecções por Helicobacter/microbiologia , Helicobacter hepaticus/patogenicidade , Doenças Inflamatórias Intestinais/etiologia , Doenças Inflamatórias Intestinais/microbiologia , Interleucina-10/genética , Limosilactobacillus reuteri/fisiologia , Imunidade Adaptativa , Animais , Linfócitos B/imunologia , Ensaio de Imunoadsorção Enzimática , Imunidade Inata , Imunoglobulina A/sangue , Imunoglobulina G/sangue , Imuno-Histoquímica , Doenças Inflamatórias Intestinais/imunologia , Camundongos , Camundongos Knockout , Reação em Cadeia da Polimerase
3.
Comp Med ; 70(3): 216-232, 2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32349859

RESUMO

Helicobacter bilis (Hb) causes hepatitis in some strains of inbred mice. The current study confirmed that Hb directly causes portal hepatitis in outbred gnotobiotic Swiss Webster (SW) mice, as we previously reported for conventional SW mice. Hbmonoassociated SW mice also developed mild enterocolitis, expanded gut-associated lymphoid tissue (GALT), and tertiary lymphoid tissue in the lower bowel. At 1 and 10 mo after infection, Hb-induced GALT hyperplasia exhibited well-organized, ectopic germinal centers with increased mononuclear cell apoptosis, MHC class II antigen presentation, and pronounced endothelial venule formation, consistent with features of tertiary lymphoid tissue. In the lower bowel, Hb induced mainly B220+ cells as well as CD4+ IL17+, CD4+ IFNγ+, and CD4+ FoxP3+ regulatory T cells and significantly increased IL10 mRNA expression. This gnotobiotic model confirmed that Hb causes portal hepatitis in outbred SW mice but stimulated GALT with an antiinflammatory bias. Because Hb had both anti- and proinflammatory effects on GALT, it should be considered a 'pathosymbiont provocateur' and merits further evaluation in mouse models of human disease.


Assuntos
Enterocolite/microbiologia , Infecções por Helicobacter/imunologia , Helicobacter/imunologia , Hepatite/microbiologia , Animais , Ceco/microbiologia , Colo/microbiologia , Enterocolite/imunologia , Feminino , Vida Livre de Germes , Infecções por Helicobacter/microbiologia , Hepatite/imunologia , Masculino , Camundongos , Camundongos Endogâmicos
4.
Infect Immun ; 76(5): 1866-76, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18285497

RESUMO

Helicobacter hepaticus causes hepatitis in susceptible strains of mice. Previous studies indicated that A/JCr mice are susceptible and C57BL/6NCr mice are resistant to H. hepaticus-induced hepatitis. We used F1 hybrid mice derived from A/J and C57BL/6 matings to investigate their phenotype and determine their hepatic gene expression profile in response to H. hepaticus infection. F1 hybrid mice, as well as parental A/J and C57BL/6 mice, were divided equally into control and H. hepaticus-infected groups and euthanized at 18 months postinoculation. Hepatic lesions were evaluated histologically and the differential hepatic gene expression in F1 mice was determined by microarray-based global gene expression profiling analysis. H. hepaticus-infected parental strains including A/J and C57BL/6 mice, as well as F1 mice, developed significant hepatitis. Overall, hepatocellular carcinomas or dysplastic liver lesions were observed in 69% of H. hepaticus-infected F1 male mice and H. hepaticus was isolated from hepatic tissues of all F1 mice with liver tumors. Liver tumors, characterized by hepatic steatosis, developed in livers with high hepatitis scores. To identify gene expression specific to H. hepaticus-induced hepatitis and progression to hepatocellular carcinoma in F1 mice, a method using comparative group transcriptome analysis was utilized. The canonical pathway most significantly enriched was immunological disease. Fatty acid synthase and steaoryl-coenzyme A desaturase, the two rate-limiting enzymes in lipogenesis, were upregulated in neoplastic relative to dysplastic livers. This study suggests a synergistic interaction between hepatic steatosis and infectious hepatitis leading to hepatocellular carcinoma. The use of AB6F1 and B6AF1 mice, as well as genetically engineered mice, on a C57BL/6 background will allow studies investigating the role of chronic microbial hepatitis and steatohepatitis in the pathogenesis of liver cancer.


Assuntos
Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/imunologia , Predisposição Genética para Doença , Infecções por Helicobacter/genética , Hepatite Crônica/genética , Padrões de Herança , Animais , Carcinoma Hepatocelular/microbiologia , Ácido Graxo Sintases/biossíntese , Fígado Gorduroso/patologia , Feminino , Perfilação da Expressão Gênica , Infecções por Helicobacter/complicações , Infecções por Helicobacter/imunologia , Infecções por Helicobacter/microbiologia , Helicobacter hepaticus/fisiologia , Hepatite Crônica/complicações , Hepatite Crônica/imunologia , Hepatite Crônica/microbiologia , Fígado/microbiologia , Fígado/patologia , Masculino , Camundongos , Camundongos Endogâmicos A , Camundongos Endogâmicos C57BL , Análise de Sequência com Séries de Oligonucleotídeos , Índice de Gravidade de Doença , Estearoil-CoA Dessaturase/biossíntese , Regulação para Cima
5.
Oncotarget ; 7(19): 28624-36, 2016 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-27086921

RESUMO

Much of the global cancer burden is associated with longstanding inflammation accompanied by release of DNA-damaging reactive oxygen and nitrogen species. Here, we report that the Mbd4 DNA glycosylase is protective in the azoxymethane/dextran sodium sulfate (AOM/DSS) mouse model of inflammation-driven colon cancer. Mbd4 excises T and U from T:G and U:G mismatches caused by deamination of 5-methylcytosine and cytosine. Since the rate of deamination is higher in inflamed tissues, we investigated the role of Mbd4 in inflammation-driven tumorigenesis. In the AOM/DSS assay, Mbd4-/- mice displayed more severe clinical symptoms, decreased survival, and a greater tumor burden than wild-type (WT) controls. The increased tumor burden in Mbd4-/- mice did not arise from impairment of AOM-induced apoptosis in the intestinal crypt. Histopathological analysis indicated that the colonic epithelium of Mbd4-/- mice is more vulnerable than WT to DSS-induced tissue damage. We investigated the role of the Mbd4-/- immune system in AOM/DSS-mediated carcinogenesis by repeating the assay on WT and Mbd4-/- mice transplanted with WT bone marrow. Mbd4-/- mice with WT bone marrow behaved similarly to Mbd4-/- mice. Together, our results indicate that the colonic epithelium of Mbd4-/- mice is more vulnerable to DSS-induced injury, which exacerbates inflammation-driven tissue injury and cancer.


Assuntos
Colo/metabolismo , Neoplasias do Colo/metabolismo , DNA Glicosilases/metabolismo , Endodesoxirribonucleases/metabolismo , Inflamação/metabolismo , Animais , Apoptose/genética , Azoximetano , Colo/patologia , Neoplasias do Colo/genética , DNA Glicosilases/genética , Sulfato de Dextrana , Endodesoxirribonucleases/genética , Humanos , Inflamação/induzido quimicamente , Inflamação/genética , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Estimativa de Kaplan-Meier , Camundongos Knockout , Carga Tumoral/genética
6.
Inflamm Bowel Dis ; 19(10): 2041-50, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23820270

RESUMO

BACKGROUND: Wiskott-Aldrich syndrome protein-deficient patients and mice are immunodeficient and can develop inflammatory bowel disease. The intestinal microbiome is critical to the development of colitis in most animal models, in which Helicobacter spp. have been implicated in disease pathogenesis. We sought to determine the role of Helicobacter spp. in colitis development in Wiskott-Aldrich syndrome protein-deficient (WKO) mice. METHODS: Feces from WKO mice raised under specific pathogen-free conditions were evaluated for the presence of Helicobacter spp., after which a subset of mice were rederived in Helicobacter spp.-free conditions. Helicobacter spp.-free WKO animals were subsequently infected with Helicobacter bilis. RESULTS: Helicobacter spp. were detected in feces from WKO mice. After rederivation in Helicobacter spp.-free conditions, WKO mice did not develop spontaneous colitis but were susceptible to radiation-induced colitis. Moreover, a T-cell transfer model of colitis dependent on Wiskott-Aldrich syndrome protein-deficient innate immune cells also required Helicobacter spp. colonization. Helicobacter bilis infection of rederived WKO mice led to typhlitis and colitis. Most notably, several H. bilis-infected animals developed dysplasia with 10% demonstrating colon carcinoma, which was not observed in uninfected controls. CONCLUSIONS: Spontaneous and T-cell transfer, but not radiation-induced, colitis in WKO mice is dependent on the presence of Helicobacter spp. Furthermore, H. bilis infection is sufficient to induce typhlocolitis and colon cancer in Helicobacter spp.-free WKO mice. This animal model of a human immunodeficiency with chronic colitis and increased risk of colon cancer parallels what is seen in human colitis and implicates specific microbial constituents in promoting immune dysregulation in the intestinal mucosa.


Assuntos
Colite/etiologia , Neoplasias do Colo/etiologia , Modelos Animais de Doenças , Infecções por Helicobacter/complicações , Inflamação/etiologia , Linfócitos T/imunologia , Proteína da Síndrome de Wiskott-Aldrich/fisiologia , Animais , Colite/metabolismo , Colite/patologia , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , DNA Viral/genética , Feminino , Helicobacter/classificação , Helicobacter/genética , Helicobacter/patogenicidade , Infecções por Helicobacter/microbiologia , Infecções por Helicobacter/patologia , Humanos , Técnicas Imunoenzimáticas , Inflamação/metabolismo , Inflamação/patologia , Masculino , Camundongos , Camundongos Knockout , Reação em Cadeia da Polimerase , Especificidade da Espécie , Irradiação Corporal Total
7.
Vaccine ; 25(30): 5583-90, 2007 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-17360081

RESUMO

Streptococcus equi, a clonal descendent of an ancestral S. zooepidemicus, causes equine strangles, a highly contagious purulent lymphadenitis of the head and neck. The aim of this study was to evaluate as vaccine components novel surface exposed or secreted S. equi proteins identified in an expression gene library with sera from resistant horses. Six proteins expressed by S. equi CF32 but not by S. zooepidemicus 631 were used to vaccinate one group of eight ponies. A second pony group was immunized with five adhesin and other proteins encoded by genes of Linkage Gr 1. All ponies made strong serum antibody responses to each protein as measured by ELISA but none were resistant to subsequent comingling challenge with S. equi CF32. These results in combination with evidence that recovered horses rapidly clear intranasally inoculated S. equi and do not make detectable serum antibody responses to its surface proteins suggest that acquired immune-mediated tonsillar clearance and not serum antibody must be stimulated by an effective strangles vaccine.


Assuntos
Proteínas de Bactérias/imunologia , Doenças dos Cavalos/prevenção & controle , Proteínas de Membrana/imunologia , Infecções Estreptocócicas/veterinária , Vacinas Estreptocócicas/imunologia , Streptococcus equi/imunologia , Animais , Anticorpos Antibacterianos/sangue , Proteínas de Bactérias/genética , DNA Bacteriano/química , DNA Bacteriano/genética , Ensaio de Imunoadsorção Enzimática , Doenças dos Cavalos/imunologia , Doenças dos Cavalos/microbiologia , Cavalos , Proteínas de Membrana/genética , Dados de Sequência Molecular , Análise de Sequência de DNA , Infecções Estreptocócicas/imunologia , Infecções Estreptocócicas/prevenção & controle , Streptococcus equi/genética , Vacinas de Subunidades Antigênicas/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA