Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
PLoS One ; 9(6): e100362, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24941045

RESUMEN

BACKGROUND: Gastrointestinal (GI) inflammation in mice and men are frequently accompanied by distinct changes of the GI microbiota composition at sites of inflammation. Helicobacter (H.) pylori infection results in gastric immunopathology accompanied by colonization of stomachs with bacterial species, which are usually restricted to the lower intestine. Potential microbiota shifts distal to the inflammatory process following long-term H. pylori infection, however, have not been studied so far. METHODOLOGY/PRINCIPAL FINDINGS: For the first time, we investigated microbiota changes along the entire GI tract of Mongolian gerbils after 14 months of infection with H. pylori B8 wildtype (WT) or its isogenic ΔcagY mutant (MUT) strain which is defective in the type IV secretion system and thus unable to modulate specific host pathways. Comprehensive cultural analyses revealed that severe gastric diseases such as atrophic pangastritis and precancerous transformations were accompanied by elevated luminal loads of E. coli and enterococci in the caecum and together with Bacteroides/Prevotella spp. in the colon of H. pylori WT, but not MUT infected gerbils as compared to naïve animals. Strikingly, molecular analyses revealed that Akkermansia, an uncultivable species involved in mucus degradation, was exclusively abundant in large intestines of H. pylori WT, but not MUT infected nor naïve gerbils. CONCLUSION/SIGNIFICANCE: Taken together, long-term infection of Mongolian gerbils with a H. pylori WT strain displaying an intact type IV secretion system leads to distinct shifts of the microbiota composition in the distal uninflamed, but not proximal inflamed GI tract. Hence, H. pylori induced immunopathogenesis of the stomach, including hypochlorhydria and hypergastrinemia, might trigger large intestinal microbiota changes whereas the exact underlying mechanisms need to be further unraveled.


Asunto(s)
Aclorhidria/microbiología , Infecciones por Helicobacter/microbiología , Helicobacter pylori/patogenicidad , Intestino Grueso/microbiología , Microbiota/inmunología , Estómago/microbiología , Aclorhidria/complicaciones , Aclorhidria/inmunología , Aclorhidria/patología , Animales , Sistemas de Secreción Bacterianos/inmunología , Bacteroides/inmunología , Bacteroides/patogenicidad , Chlamydiaceae/inmunología , Chlamydiaceae/patogenicidad , Escherichia coli/inmunología , Escherichia coli/patogenicidad , Femenino , Gerbillinae , Infecciones por Helicobacter/complicaciones , Infecciones por Helicobacter/inmunología , Infecciones por Helicobacter/patología , Helicobacter pylori/inmunología , Inmunidad Innata , Intestino Grueso/inmunología , Intestino Grueso/patología , Prevotella/inmunología , Prevotella/patogenicidad , Estómago/inmunología , Estómago/patología
2.
PLoS One ; 7(7): e40761, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22808254

RESUMEN

BACKGROUND: Campylobacter jejuni is a leading cause of foodborne bacterial enterocolitis worldwide. Investigation of immunopathology is hampered by a lack of suitable vertebrate models. We have recently shown that gnotobiotic mice as well as conventional IL-10(-/-) animals are susceptible to C. jejuni infection and develop intestinal immune responses. However, clinical symptoms of C. jejuni infection were rather subtle and did not reflect acute bloody diarrhea seen in human campylobacteriosis. METHODOLOGY/PRINCIPAL FINDINGS: In order to overcome these limitations we generated gnotobiotic IL-10(-/-) mice by quintuple antibiotic treatment starting right after weaning. The early treatment was essential to prevent these animals from chronic colitis. Following oral infection C. jejuni colonized the gastrointestinal tract at high levels and induced acute enterocolitis within 7 days as indicated by bloody diarrhea and pronounced histopathological changes of the colonic mucosa. Immunopathology was further characterized by increased numbers of apoptotic cells, regulatory T-cells, T- and B-lymphocytes as well as elevated TNF-α, IFN-γ, and MCP-1 concentrations in the inflamed colon. The induction of enterocolitis was specific for C. jejuni given that control animals infected with a commensal E. coli strain did not display any signs of disease. Most strikingly, intestinal immunopathology was ameliorated in mice lacking Toll-like-receptors-2 or -4 indicating that C. jejuni lipoproteins and lipooligosaccharide are essential for induction and progression of immunopathology. CONCLUSION/SIGNIFICANCE: Gnotobiotic IL-10(-/-) mice develop acute enterocolitis following C. jejuni infection mimicking severe episodes of human campylobacteriosis and are thus well suited to further dissect mechanisms underlying Campylobacter infections in vivo.


Asunto(s)
Campylobacter jejuni/fisiología , Enterocolitis/inmunología , Enterocolitis/microbiología , Vida Libre de Gérmenes , Interleucina-10/deficiencia , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 4/metabolismo , Enfermedad Aguda , Animales , Traslocación Bacteriana , Infecciones por Campylobacter/inmunología , Infecciones por Campylobacter/microbiología , Campylobacter jejuni/crecimiento & desarrollo , Colon/inmunología , Colon/microbiología , Colon/patología , Recuento de Colonia Microbiana , Citocinas/metabolismo , Enterocolitis/patología , Humanos , Inmunidad/inmunología , Inflamación/microbiología , Inflamación/patología , Mediadores de Inflamación/metabolismo , Interleucina-10/metabolismo , Ratones , Ratones Endogámicos C57BL , Transducción de Señal/inmunología , Receptor Toll-Like 2/deficiencia , Receptor Toll-Like 4/deficiencia
3.
PLoS One ; 7(5): e35988, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22563475

RESUMEN

BACKGROUND: The zoonotic pathogen Campylobacter jejuni is a leading cause of bacterial foodborne enterocolitis in humans worldwide. The understanding of immunopathology underlying human campylobacteriosis is hampered by the fact that mice display strong colonization resistance against the pathogen due to their host specific gut microbiota composition. METHODOLOGY/PRINCIPAL FINDINGS: Since the microbiota composition changes significantly during intestinal inflammation we dissected factors contributing to colonization resistance against C. jejuni in murine ileitis, colitis and in infant mice. In contrast to healthy animals C. jejuni could stably colonize mice suffering from intestinal inflammation. Strikingly, in mice with Toxoplasma gondii-induced acute ileitis, C. jejuni disseminated to mesenteric lymphnodes, spleen, liver, kidney, and blood. In infant mice C. jejuni infection induced enterocolitis. Mice suffering from intestinal inflammation and C. jejuni susceptible infant mice displayed characteristical microbiota shifts dominated by increased numbers of commensal Escherichia coli. To further dissect the pivotal role of those distinct microbiota shifts in abrogating colonization resistance, we investigated C. jejuni infection in healthy adult mice in which the microbiota was artificially modified by feeding live commensal E. coli. Strikingly, in animals harboring supra-physiological intestinal E. coli loads, colonization resistance was significantly diminished and C. jejuni infection induced enterocolitis mimicking key features of human campylobacteriosis. CONCLUSION/SIGNIFICANCE: Murine colonization resistance against C. jejuni is abrogated by changes in the microbiota composition towards elevated E. coli loads during intestinal inflammation as well as in infant mice. Intestinal inflammation and microbiota shifts thus represent potential risk factors for C. jejuni infection. Corresponding interplays between C. jejuni and microbiota might occur in human campylobacteriosis. Murine models introduced here mimick key features of human campylobacteriosis and allow for further analysis of immunological and molecular mechanisms of C. jejuni-host interactions.


Asunto(s)
Infecciones por Campylobacter/microbiología , Campylobacter jejuni/fisiología , Escherichia coli/fisiología , Intestinos/microbiología , Animales , Traslocación Bacteriana/inmunología , Traslocación Bacteriana/fisiología , Infecciones por Campylobacter/genética , Infecciones por Campylobacter/inmunología , Campylobacter jejuni/crecimiento & desarrollo , Campylobacter jejuni/inmunología , Colitis/genética , Colitis/inmunología , Colitis/microbiología , Colon/inmunología , Colon/metabolismo , Colon/microbiología , Escherichia coli/crecimiento & desarrollo , Escherichia coli/inmunología , Heces/microbiología , Interacciones Huésped-Patógeno/inmunología , Humanos , Ileítis/etiología , Ileítis/inmunología , Ileítis/microbiología , Mediadores de Inflamación/inmunología , Mediadores de Inflamación/metabolismo , Interferón gamma/inmunología , Interferón gamma/metabolismo , Interleucina-10/deficiencia , Interleucina-10/genética , Interleucina-10/inmunología , Intestinos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Ratones Noqueados , Factores de Tiempo , Toxoplasmosis Animal/complicaciones , Factor de Necrosis Tumoral alfa/inmunología , Factor de Necrosis Tumoral alfa/metabolismo
4.
PLoS One ; 6(6): e20953, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21698299

RESUMEN

BACKGROUND: Although Campylobacter jejuni-infections have a high prevalence worldwide and represent a significant socioeconomic burden, it is still not well understood how C. jejuni causes intestinal inflammation. Detailed investigation of C. jejuni-mediated intestinal immunopathology is hampered by the lack of appropriate vertebrate models. In particular, mice display colonization resistance against this pathogen. METHODOLOGY/PRINCIPAL FINDINGS: To overcome these limitations we developed a novel C. jejuni-infection model using gnotobiotic mice in which the intestinal flora was eradicated by antibiotic treatment. These animals could then be permanently associated with a complete human (hfa) or murine (mfa) microbiota. After peroral infection C. jejuni colonized the gastrointestinal tract of gnotobiotic and hfa mice for six weeks, whereas mfa mice cleared the pathogen within two days. Strikingly, stable C. jejuni colonization was accompanied by a pro-inflammatory immune response indicated by increased numbers of T- and B-lymphocytes, regulatory T-cells, neutrophils and apoptotic cells, as well as increased concentrations of TNF-α, IL-6, and MCP-1 in the colon mucosa of hfa mice. Analysis of MyD88(-/-), TRIF(-/-), TLR4(-/-), and TLR9(-/-) mice revealed that TLR4- and TLR9-signaling was essential for immunopathology following C. jejuni-infection. Interestingly, C. jejuni-mutant strains deficient in formic acid metabolism and perception induced less intestinal immunopathology compared to the parental strain infection. In summary, the murine gut flora is essential for colonization resistance against C. jejuni and can be overcome by reconstitution of gnotobiotic mice with human flora. Detection of C. jejuni-LPS and -CpG-DNA by host TLR4 and TLR9, respectively, plays a key role in immunopathology. Finally, the host immune response is tightly coupled to bacterial formic acid metabolism and invasion fitness. CONCLUSION/SIGNIFICANCE: We conclude that gnotobiotic and "humanized" mice represent excellent novel C. jejuni-infection and -inflammation models and provide deep insights into the immunological and molecular interplays between C. jejuni, microbiota and innate immunity in human campylobacteriosis.


Asunto(s)
Infecciones por Campylobacter/inmunología , Campylobacter jejuni/aislamiento & purificación , Modelos Animales de Enfermedad , Inmunidad Innata , Animales , Infecciones por Campylobacter/metabolismo , Infecciones por Campylobacter/microbiología , Citocinas/metabolismo , Ratones , Ratones Endogámicos C57BL
5.
PLoS One ; 5(12): e15099, 2010 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-21151942

RESUMEN

BACKGROUND: The health beneficial effects of Resveratrol, Curcumin and Simvastatin have been demonstrated in various experimental models of inflammation. We investigated the potential anti-inflammatory and immunomodulatory mechanisms of the above mentioned compounds in a murine model of hyper-acute Th1-type ileitis following peroral infection with Toxoplasma gondii. METHODOLOGY/PRINCIPAL FINDINGS: Here we show that after peroral administration of Resveratrol, Curcumin or Simvastatin, mice were protected from ileitis development and survived the acute phase of inflammation whereas all Placebo treated controls died. In particular, Resveratrol treatment resulted in longer-term survival. Resveratrol, Curcumin or Simvastatin treated animals displayed significantly increased numbers of regulatory T cells and augmented intestinal epithelial cell proliferation/regeneration in the ileum mucosa compared to placebo control animals. In contrast, mucosal T lymphocyte and neutrophilic granulocyte numbers in treated mice were reduced. In addition, levels of the anti-inflammatory cytokine IL-10 in ileum, mesenteric lymph nodes and spleen were increased whereas pro-inflammatory cytokine expression (IL-23p19, IFN-γ, TNF-α, IL-6, MCP-1) was found to be significantly lower in the ileum of treated animals as compared to Placebo controls. Furthermore, treated animals displayed not only fewer pro-inflammatory enterobacteria and enterococci but also higher anti-inflammatory lactobacilli and bifidobacteria loads. Most importantly, treatment with all three compounds preserved intestinal barrier functions as indicated by reduced bacterial translocation rates into spleen, liver, kidney and blood. CONCLUSION/SIGNIFICANCE: Oral treatment with Resveratrol, Curcumin or Simvastatin ameliorates acute small intestinal inflammation by down-regulating Th1-type immune responses and prevents bacterial translocation by maintaining gut barrier function. These findings provide novel and potential prophylaxis and treatment options of patients with inflammatory bowel diseases.


Asunto(s)
Antiinflamatorios/farmacología , Curcumina/farmacología , Simvastatina/farmacología , Estilbenos/farmacología , Administración Oral , Animales , Antiinflamatorios no Esteroideos/farmacología , Modelos Animales de Enfermedad , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Ileítis/metabolismo , Ileítis/microbiología , Inflamación , Intestino Delgado/efectos de los fármacos , Intestino Delgado/metabolismo , Ratones , Ratones Endogámicos C57BL , Resveratrol , Células TH1/efectos de los fármacos , Toxoplasma/metabolismo
6.
Gut ; 59(8): 1079-87, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20639251

RESUMEN

BACKGROUND: The bacterial microflora aggravates graft-versus-host-disease (GvHD) after allogeneic stem cell transplantation, but the underlying mechanisms of manifestations of intestinal GvHD (iGvHD) in the gut remain poorly understood. AIM: To analyse the gut flora composition and the impact of bacterial sensing via Toll-like receptors (TLRs) in iGvHD. METHODS: By mimicking clinical low-intensity conditioning regimens used in humans, a novel irradiation independent, treosulfan and cyclophosphamide-based murine allogeneic transplantation model was established. A global survey of the intestinal microflora by cultural and molecular methods was performed, the intestinal immunopathology in TLR-deficient recipient mice with iGvHD investigated and finally, the impact of anti-TLR9 treatment on iGvHD development assessed. RESULTS: The inflammatory responses in iGvHD were accompanied by gut flora shifts towards enterobacteria, enterococci and Bacteroides/Prevotella spp. Analysis of iGvHD in MyD88(-/-), TRIF(-/-), TLR2/4(-/-), and TLR9(-/-) recipient mice showed that bacterial sensing via TLRs was essential for iGvHD development. Acute iGvHD was characterised by increasing numbers of apoptotic cells, proliferating cells, T cells and neutrophils within the colon. These responses were significantly reduced in MyD88(-/-), TLR2/4(-/-), TRIF(-/-) and TLR9(-/-) mice, as compared with wild-type controls. However, TRIF(-/-) and TLR2/4(-/-) mice were not protected from mortality, whereas TLR9(-/-) mice displayed increased survival rates. The important role of TLR9-mediated immunopathology was independently confirmed by significantly reduced macroscopic disease symptoms and colonic apoptosis as well as by reduced T-cell and neutrophil numbers within the colon after treatment with a synthetic inhibitory oligonucleotide. CONCLUSIONS: These results emphasise the critical role of gut microbiota, innate immunity and TLR9 in iGvHD and highlight anti-TLR9 strategies as novel therapeutic options.


Asunto(s)
Colitis/microbiología , Enfermedad Injerto contra Huésped/inmunología , Enfermedad Injerto contra Huésped/microbiología , Factor 88 de Diferenciación Mieloide/inmunología , Receptor Toll-Like 9/inmunología , Enfermedad Aguda , Animales , Apoptosis/inmunología , Trasplante de Médula Ósea , Proliferación Celular , Colitis/inmunología , Colitis/patología , Colitis/prevención & control , Modelos Animales de Enfermedad , Femenino , Enfermedad Injerto contra Huésped/patología , Enfermedad Injerto contra Huésped/prevención & control , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/deficiencia , Oligonucleótidos Antisentido/uso terapéutico , Bazo/trasplante , Receptor Toll-Like 9/deficiencia , Acondicionamiento Pretrasplante
7.
J Exp Med ; 206(13): 3047-59, 2009 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-19995958

RESUMEN

Peroral infection with Toxoplasma gondii leads to the development of small intestinal inflammation dependent on Th1 cytokines. The role of Th17 cells in ileitis is unknown. We report interleukin (IL)-23-mediated gelatinase A (matrixmetalloproteinase [MMP]-2) up-regulation in the ileum of infected mice. MMP-2 deficiency as well as therapeutic or prophylactic selective gelatinase blockage protected mice from the development of T. gondii-induced immunopathology. Moreover, IL-23-dependent up-regulation of IL-22 was essential for the development of ileitis, whereas IL-17 was down-regulated and dispensable. CD4(+) T cells were the main source of IL-22 in the small intestinal lamina propria. Thus, IL-23 regulates small intestinal inflammation via IL-22 but independent of IL-17. Gelatinases may be useful targets for treatment of intestinal inflammation.


Asunto(s)
Interleucina-17/fisiología , Interleucina-23/fisiología , Interleucinas/fisiología , Intestino Delgado/patología , Metaloproteinasa 2 de la Matriz/fisiología , Toxoplasma/patogenicidad , Animales , Femenino , Intestino Delgado/inmunología , Metaloproteinasa 9 de la Matriz/fisiología , Ratones , Ratones Endogámicos C57BL , Piperazinas/farmacología , Pirimidinas/farmacología , Toxoplasmosis Animal/inmunología , Interleucina-22
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...