Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 364
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Nat Immunol ; 22(4): 510-519, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33707780

RESUMO

Fibroblastic reticular cells (FRCs) determine the organization of lymphoid organs and control immune cell interactions. While the cellular and molecular mechanisms underlying FRC differentiation in lymph nodes and the splenic white pulp have been elaborated to some extent, in Peyer's patches (PPs) they remain elusive. Using a combination of single-cell transcriptomics and cell fate mapping in advanced mouse models, we found that PP formation in the mouse embryo is initiated by an expansion of perivascular FRC precursors, followed by FRC differentiation from subepithelial progenitors. Single-cell transcriptomics and cell fate mapping confirmed the convergence of perivascular and subepithelial FRC lineages. Furthermore, lineage-specific loss- and gain-of-function approaches revealed that the two FRC lineages synergistically direct PP organization, maintain intestinal microbiome homeostasis and control anticoronavirus immune responses in the gut. Collectively, this study reveals a distinct mosaic patterning program that generates key stromal cell infrastructures for the control of intestinal immunity.


Assuntos
Linhagem da Célula , Fibroblastos/imunologia , Imunidade nas Mucosas , Mucosa Intestinal/imunologia , Intestino Delgado/imunologia , Nódulos Linfáticos Agregados/imunologia , Animais , Comunicação Celular , Células Cultivadas , Infecções por Coronavirus/imunologia , Infecções por Coronavirus/metabolismo , Infecções por Coronavirus/virologia , Modelos Animais de Doenças , Fibroblastos/metabolismo , Microbioma Gastrointestinal , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Interações Hospedeiro-Patógeno , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Mucosa Intestinal/virologia , Intestino Delgado/metabolismo , Intestino Delgado/microbiologia , Intestino Delgado/virologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Vírus da Hepatite Murina/imunologia , Vírus da Hepatite Murina/patogenicidade , Nódulos Linfáticos Agregados/metabolismo , Nódulos Linfáticos Agregados/microbiologia , Nódulos Linfáticos Agregados/virologia , Fenótipo , Análise de Célula Única , Transcriptoma
2.
Immunity ; 50(2): 446-461.e9, 2019 02 19.
Artigo em Inglês | MEDLINE | ID: mdl-30709742

RESUMO

Production of interleukin-17 (IL-17) and IL-22 by T helper 17 (Th17) cells and group 3 innate lymphoid cells (ILC3s) in response to the gut microbiota ensures maintenance of intestinal barrier function. Here, we examined the mechanisms whereby the immune system detects microbiota in the steady state. A Syk-kinase-coupled signaling pathway in dendritic cells (DCs) was critical for commensal-dependent production of IL-17 and IL-22 by CD4+ T cells. The Syk-coupled C-type lectin receptor Mincle detected mucosal-resident commensals in the Peyer's patches (PPs), triggered IL-6 and IL-23p19 expression, and thereby regulated function of intestinal Th17- and IL-17-secreting ILCs. Mice deficient in Mincle or with selective depletion of Syk in CD11c+ cells had impaired production of intestinal RegIIIγ and IgA and increased systemic translocation of gut microbiota. Consequently, Mincle deficiency led to liver inflammation and deregulated lipid metabolism. Thus, sensing of commensals by Mincle and Syk signaling in CD11c+ cells reinforces intestinal immune barrier and promotes host-microbiota mutualism, preventing systemic inflammation.


Assuntos
Células Dendríticas/imunologia , Microbioma Gastrointestinal/imunologia , Interleucina-17/imunologia , Interleucinas/imunologia , Lectinas Tipo C/imunologia , Proteínas de Membrana/imunologia , Quinase Syk/imunologia , Animais , Células Dendríticas/metabolismo , Microbioma Gastrointestinal/fisiologia , Humanos , Interleucina-17/metabolismo , Interleucinas/metabolismo , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Nódulos Linfáticos Agregados/imunologia , Nódulos Linfáticos Agregados/metabolismo , Nódulos Linfáticos Agregados/microbiologia , Transdução de Sinais/imunologia , Quinase Syk/genética , Quinase Syk/metabolismo , Células Th17/imunologia , Células Th17/metabolismo , Interleucina 22
3.
Immunity ; 43(3): 527-40, 2015 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-26362264

RESUMO

The interrelationship between IgAs and microbiota diversity is still unclear. Here we show that BALB/c mice had higher abundance and diversity of IgAs than C57BL/6 mice and that this correlated with increased microbiota diversity. We show that polyreactive IgAs mediated the entrance of non-invasive bacteria to Peyer's patches, independently of CX3CR1(+) phagocytes. This allowed the induction of bacteria-specific IgA and the establishment of a positive feedback loop of IgA production. Cohousing of mice or fecal transplantation had little or no influence on IgA production and had only partial impact on microbiota composition. Germ-free BALB/c, but not C57BL/6, mice already had polyreactive IgAs that influenced microbiota diversity and selection after colonization. Together, these data suggest that genetic predisposition to produce polyreactive IgAs has a strong impact on the generation of antigen-specific IgAs and the selection and maintenance of microbiota diversity.


Assuntos
Antígenos de Bactérias/imunologia , Variação Genética/imunologia , Imunoglobulina A/imunologia , Microbiota/imunologia , Animais , Bactérias/classificação , Bactérias/genética , Bactérias/imunologia , DNA Bacteriano/química , DNA Bacteriano/genética , Fezes/microbiologia , Citometria de Fluxo , Interações Hospedeiro-Patógeno/imunologia , Imunização , Imunoglobulina A/sangue , Imunoglobulina A/metabolismo , Metagenômica/métodos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Microbiota/genética , Nódulos Linfáticos Agregados/imunologia , Nódulos Linfáticos Agregados/metabolismo , Nódulos Linfáticos Agregados/microbiologia , Filogenia , RNA Ribossômico 16S/genética , Salmonella typhimurium/genética , Salmonella typhimurium/imunologia , Salmonella typhimurium/fisiologia , Especificidade da Espécie
4.
Eur J Immunol ; 50(6): 783-794, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32065660

RESUMO

In humans and mice, mucosal immune responses are dominated by IgA antibodies and the cytokine TGF-ß, suppressing unwanted immune reactions but also targeting Ig class switching to IgA. It had been suggested that eosinophils promote the generation and maintenance of mucosal IgA-expressing plasma cells. Here, we demonstrate that not eosinophils, but specific bacteria determine mucosal IgA production. Co-housing of eosinophil-deficient mice with mice having high intestinal IgA levels, as well as the intentional microbiota transfer induces TGF-ß expression in intestinal T follicular helper cells, thereby promoting IgA class switching in Peyer's patches, enhancing IgA+ plasma cell numbers in the small intestinal lamina propria and levels of mucosal IgA. We show that bacteria highly enriched for the genus Anaeroplasma are sufficient to induce these changes and enhance IgA levels when adoptively transferred. Thus, specific members of the intestinal microbiota and not the microbiota as such regulate gut homeostasis, by promoting the expression of immune-regulatory TGF-ß and of mucosal IgA.


Assuntos
Microbioma Gastrointestinal/imunologia , Imunidade nas Mucosas , Imunoglobulina A/imunologia , Mucosa Intestinal , Nódulos Linfáticos Agregados , Linfócitos T Auxiliares-Indutores/imunologia , Animais , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Nódulos Linfáticos Agregados/imunologia , Nódulos Linfáticos Agregados/microbiologia , Tenericutes/imunologia
5.
BMC Microbiol ; 21(1): 68, 2021 02 27.
Artigo em Inglês | MEDLINE | ID: mdl-33639835

RESUMO

BACKGROUND: The genus Lactobacillus is an important component of the gastrointestinal tract of human and animals and commonly considered as probiotic. L. taiwanensis has long been proposed to be a probiotic whereas understanding on this species is still in its infancy. Genomic information of L. taiwanensis is fairly limited. Extensive characterization of its beneficial traits is needed. RESULTS: A new strain CLG01 of L. taiwanensis was isolated from mouse Peyer's patches. We established its probiotic profile through in vitro experiments. Complete genome of this strain was also sequenced and analyzed. L. taiwanensis CLG01 showed robust tolerance to acid and a degree of tolerance to bile salt with a promising antibacterial activity against a broad spectrum of pathogenic bacteria. In vitro treatment of mouse RAW 264.7 macrophage cells with heat-killed bacteria and bacterial supernatant of L. taiwanensis CLG01 resulted in enhancement of immune responses and upregulated expression of TNF-α and IL-6. The strain CLG01 also increased the IL-10 production of macrophages when co-treated with lipopolysaccharide (LPS). Complete genome of L. taiwanensis CLG01 contained a 1.89 Mb chromosome and two plasmids. Further genomic analysis revealed the presence of genes related to its resistance to different stresses and the beneficial effects mentioned above. Moreover, biosynthetic gene clusters (BGCs) encoding antimicrobial peptides, like bacteriocin, linear azol(in)e-containing peptide (LAP) and lanthipeptide, were also identified in the genome of L. taiwanensis CLG01. CONCLUSIONS: L. taiwanensis CLG01, isolated from mouse Peyer's patches, is the first L. taiwanensis strain with both phenotypes and genotypes systematically studied. These preliminary data confirmed the role of L. taiwanensis CLG01 as a potential probiotic candidate with antibacterial and immunomodulatory activity, which provide insight for further investigation to this species.


Assuntos
Antibacterianos , Genoma Bacteriano/genética , Fatores Imunológicos , Lactobacillus/genética , Lactobacillus/metabolismo , Nódulos Linfáticos Agregados/microbiologia , Probióticos , Animais , Antibacterianos/isolamento & purificação , Células Cultivadas , Regulação da Expressão Gênica/imunologia , Fatores Imunológicos/isolamento & purificação , Interleucina-6/genética , Camundongos , Fator de Necrose Tumoral alfa/genética
6.
Proc Natl Acad Sci U S A ; 114(5): E791-E800, 2017 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-28096329

RESUMO

Pathogenic bacteria need to rapidly adjust their virulence and fitness program to prevent eradication by the host. So far, underlying adaptation processes that drive pathogenesis have mostly been studied in vitro, neglecting the true complexity of host-induced stimuli acting on the invading pathogen. In this study, we developed an unbiased experimental approach that allows simultaneous monitoring of genome-wide infection-linked transcriptional alterations of the host and colonizing extracellular pathogens. Using this tool for Yersinia pseudotuberculosis-infected lymphatic tissues, we revealed numerous alterations of host transcripts associated with inflammatory and acute-phase responses, coagulative activities, and transition metal ion sequestration, highlighting that the immune response is dominated by infiltrating neutrophils and elicits a mixed TH17/TH1 response. In consequence, the pathogen's response is mainly directed to prevent phagocytic attacks. Yersinia up-regulates the gene and expression dose of the antiphagocytic type III secretion system (T3SS) and induces functions counteracting neutrophil-induced ion deprivation, radical stress, and nutritional restraints. Several conserved bacterial riboregulators were identified that impacted this response. The strongest influence on virulence was found for the loss of the carbon storage regulator (Csr) system, which is shown to be essential for the up-regulation of the T3SS on host cell contact. In summary, our established approach provides a powerful tool for the discovery of infection-specific stimuli, induced host and pathogen responses, and underlying regulatory processes.


Assuntos
Interações Hospedeiro-Patógeno/genética , Transcriptoma , Infecções por Yersinia pseudotuberculosis/genética , Yersinia pseudotuberculosis/genética , Animais , Feminino , Camundongos Endogâmicos BALB C , Nódulos Linfáticos Agregados/metabolismo , Nódulos Linfáticos Agregados/microbiologia , RNA Mensageiro/genética , Análise de Sequência de RNA , Fatores de Virulência/genética , Yersinia pseudotuberculosis/metabolismo , Yersinia pseudotuberculosis/fisiologia , Infecções por Yersinia pseudotuberculosis/imunologia
7.
J Immunol ; 199(4): 1382-1392, 2017 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-28716827

RESUMO

Yersinia enterocolitica is an enteropathogenic bacterium that causes gastrointestinal disorders, as well as extraintestinal manifestations. To subvert the host's immune response, Y. enterocolitica uses a type III secretion system consisting of an injectisome and effector proteins, called Yersinia outer proteins (Yops), that modulate activation, signaling, and survival of immune cells. In this article, we show that galectin-1 (Gal-1), an immunoregulatory lectin widely expressed in mucosal tissues, contributes to Y. enterocolitica pathogenicity by undermining protective antibacterial responses. We found higher expression of Gal-1 in the spleen and Peyer's patches of mice infected orogastrically with Y. enterocolitica serotype O:8 compared with noninfected hosts. This effect was prevented when mice were infected with Y. enterocolitica lacking YopP or YopH, two critical effectors involved in bacterial immune evasion. Consistent with a regulatory role for this lectin during Y. enterocolitica pathogenesis, mice lacking Gal-1 showed increased weight and survival, lower bacterial load, and attenuated intestinal pathology compared with wild-type mice. These protective effects involved modulation of NF-κB activation, TNF production, and NO synthesis in mucosal tissue and macrophages, as well as systemic dysregulation of IL-17 and IFN-γ responses. In vivo neutralization of these proinflammatory cytokines impaired bacterial clearance and eliminated host protection conferred by Gal-1 deficiency. Finally, supplementation of recombinant Gal-1 in mice lacking Gal-1 or treatment of wild-type mice with a neutralizing anti-Gal-1 mAb confirmed the immune inhibitory role of this endogenous lectin during Y. enterocolitica infection. Thus, targeting Gal-1-glycan interactions may contribute to reinforce antibacterial responses by reprogramming innate and adaptive immune mechanisms.


Assuntos
Galectina 1/metabolismo , Interações Hospedeiro-Patógeno , Yersiniose/imunologia , Yersinia enterocolitica/imunologia , Animais , Carga Bacteriana , Proteínas da Membrana Bacteriana Externa/genética , Proteínas de Bactérias/genética , Galectina 1/antagonistas & inibidores , Galectina 1/genética , Galectina 1/imunologia , Interferon gama/sangue , Interferon gama/imunologia , Interleucina-17/sangue , Interleucina-17/imunologia , Intestinos/imunologia , Intestinos/microbiologia , Intestinos/patologia , Camundongos , NF-kappa B/metabolismo , Óxido Nítrico/biossíntese , Nódulos Linfáticos Agregados/imunologia , Nódulos Linfáticos Agregados/microbiologia , Nódulos Linfáticos Agregados/patologia , Proteínas Tirosina Fosfatases/deficiência , Proteínas Tirosina Fosfatases/genética , Baço/imunologia , Baço/microbiologia , Fator de Necrose Tumoral alfa/biossíntese
8.
Immunity ; 31(4): 677-89, 2009 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-19833089

RESUMO

Microbiota-induced cytokine responses participate in gut homeostasis, but the cytokine balance at steady-state and the role of individual bacterial species in setting the balance remain elusive. Herein, systematic analysis of gnotobiotic mice indicated that colonization by a whole mouse microbiota orchestrated a broad spectrum of proinflammatory T helper 1 (Th1), Th17, and regulatory T cell responses whereas most tested complex microbiota and individual bacteria failed to efficiently stimulate intestinal T cell responses. This function appeared the prerogative of a restricted number of bacteria, the prototype of which is the segmented filamentous bacterium, a nonculturable Clostridia-related species, which could largely recapitulate the coordinated maturation of T cell responses induced by the whole mouse microbiota. This bacterium, already known as a potent inducer of mucosal IgA, likely plays a unique role in the postnatal maturation of gut immune functions. Changes in the infant flora may thus influence the development of host immune responses.


Assuntos
Clostridium/imunologia , Citocinas/metabolismo , Intestinos/imunologia , Nódulos Linfáticos Agregados/imunologia , Linfócitos T Reguladores/imunologia , Células Th1/imunologia , Animais , Bacteroidetes/imunologia , Citocinas/imunologia , Escherichia coli/imunologia , Feminino , Expressão Gênica , Vida Livre de Germes , Interleucina-17/imunologia , Intestinos/microbiologia , Intestinos/ultraestrutura , Camundongos , Camundongos Endogâmicos C3H , Microscopia Eletrônica de Varredura , Nódulos Linfáticos Agregados/metabolismo , Nódulos Linfáticos Agregados/microbiologia , Linfócitos T Reguladores/microbiologia , Células Th1/microbiologia
9.
Biol Pharm Bull ; 41(2): 190-197, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29386479

RESUMO

Application of food-grade Lactococcus lactis (L. lactis) as a safe delivery tool for DNA vaccines and therapeutic proteins has been well investigated. Although some studies showed that eukaryotic expression plasmids were transferred from L. lactis to enterocytes, the precise mechanism of the DNA transfer remains unknown. In this study, we generated an invasive L. lactis strain that expresses "murinized" Internalin A, an invasin of intracellular bacteria Listeria monocytogenes with two amino acid alterations for invasion into murine cells, and confirmed that this L. lactis strain delivered DNA in an invasin-dependent manner into a monolayer of epithelial cells polarized to mimic the gastrointestinal tract environment. Although invasive L. lactis inoculated orally can deliver DNA into enterocytes in the gastrointestinal tract of mice, the efficiency of DNA transfer was similar to that of non-invasive L. lactis strain, suggesting that the in vivo DNA transfer from L. lactis occurs invasin-independently. A ligated-intestinal loop assay, a method for a short-term culturing of the whole intestine filled with materials to evaluate the interaction of the materials with intestinal cells, demonstrated that both non-invasive and invasive L. lactis strains were present in the Peyer's patches of the small intestine. On the other hand, few L. lactis was detected in the non-Peyer's patch epithelial region. Thus, our observations lead us to speculate that DNA transfer from L. lactis occurs predominantly in the Peyer's patches in an invasin-independent manner.


Assuntos
Proteínas de Bactérias/metabolismo , DNA Recombinante/metabolismo , Sistemas de Liberação de Medicamentos , Lactococcus lactis/fisiologia , Microrganismos Geneticamente Modificados/fisiologia , Nódulos Linfáticos Agregados/metabolismo , Vacinas de DNA/metabolismo , Administração Oral , Animais , Proteínas de Bactérias/administração & dosagem , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Translocação Bacteriana , Transporte Biológico , Células CACO-2 , Linhagem Celular , Polaridade Celular , DNA Recombinante/administração & dosagem , Feminino , Microbiologia de Alimentos , Humanos , Mucosa Intestinal/citologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Intestino Delgado/citologia , Intestino Delgado/metabolismo , Intestino Delgado/microbiologia , Lactococcus lactis/citologia , Lactococcus lactis/genética , Listeria monocytogenes/citologia , Listeria monocytogenes/genética , Listeria monocytogenes/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Microrganismos Geneticamente Modificados/citologia , Microrganismos Geneticamente Modificados/genética , Nódulos Linfáticos Agregados/citologia , Nódulos Linfáticos Agregados/microbiologia , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/metabolismo , Vacinas de DNA/administração & dosagem
10.
Cell Mol Life Sci ; 74(8): 1503-1509, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-27838744

RESUMO

To sustain the bio-energetic demands of growth, proliferation, and effector functions, the metabolism of immune cells changes dramatically in response to immunologic stimuli. In this review, I focus on B cell metabolism, especially regarding the production of intestinal IgA antibody. Accumulating evidence has implicated not only host-derived factors (e.g., cytokines) but also gut environmental factors, including the possible involvement of commensal bacteria and diet, in the control of B cell metabolism during intestinal IgA antibody production. These findings yield new insights into the regulation of immunosurveillance and homeostasis in the gut.


Assuntos
Linfócitos B/imunologia , Imunoglobulina A/imunologia , Intestinos/imunologia , Intestinos/microbiologia , Animais , Linfócitos B/citologia , Linfócitos B/metabolismo , Diferenciação Celular , Metabolismo Energético , Humanos , Imunidade Celular , Imunoglobulina A/metabolismo , Mucosa Intestinal/metabolismo , Nódulos Linfáticos Agregados/imunologia , Nódulos Linfáticos Agregados/metabolismo , Nódulos Linfáticos Agregados/microbiologia , Tiamina/imunologia , Tiamina/metabolismo
11.
J Infect Dis ; 216(6): 752-760, 2017 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-28329174

RESUMO

Background: To successfully limit pathogen dissemination, an immunological link between the entry tissue of the pathogen and the underlying secondary lymphoid organs (SLOs) needs to be established to prime adaptive immune responses. Here, the prerequisite of CCR7 to mount host immune responses within SLOs during gastrointestinal Yersinia pseudotuberculosis infection to limit pathogen spread was investigated. Methods: Survival, bacterial dissemination, and intestinal and systemic pathology of wild-type and CCR7-/- mice were assessed and correlated to the presence of immune cell subsets and cytokine responses throughout the course of infection. Results: The CCR7-/- mice show a significantly higher morbidity and are more prone to pathogen dissemination and intestinal and systemic inflammation during the oral route of infection. Significant impact of CCR7 deficiency over the course of infection on several immunological parameters were observed (ie, elevated neutrophil-dominated innate immune response in Peyer's patches, limited dendritic cell migration to mesenteric lymph nodes [mLNs] causing reduced T cell-mediated adaptive immune responses (in particular Th17-like responses) in mLNs). Conclusions: Our work indicates that CCR7 is required to mount a robust immune response against enteropathogenic Y. pseudotuberculosis by promoting Th17-like responses in mLNs.


Assuntos
Predisposição Genética para Doença , Receptores CCR7/imunologia , Células Th17/imunologia , Infecções por Yersinia pseudotuberculosis/imunologia , Animais , Movimento Celular , Células Dendríticas/imunologia , Interações Hospedeiro-Patógeno/genética , Intestinos/imunologia , Intestinos/microbiologia , Linfonodos/imunologia , Linfonodos/microbiologia , Camundongos , Células Mieloides/imunologia , Nódulos Linfáticos Agregados/imunologia , Nódulos Linfáticos Agregados/microbiologia , Receptores CCR7/genética , Yersinia pseudotuberculosis , Infecções por Yersinia pseudotuberculosis/genética
12.
Cell Microbiol ; 18(2): 195-210, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26242223

RESUMO

Candida albicans is the most frequent yeast responsible for systemic infections in humans. These infections mainly originate from the gastrointestinal tract where C. albicans can invade the gut epithelial barrier to gain access to the bloodstream. Along the gut, pathogens can use Microfold (M) cells as a portal of entry to cross the epithelial barrier. M cells are specialized cells mainly located in the follicule-associated epithelium of Peyer patches. In this study, we used scanning electron and fluorescence microscopy, adhesion and invasion assays and fungal mutants to investigate the interactions of C. albicans with M cells obtained in an established in vitro model whereby enterocyte-like Caco-2 cells co-cultured with the Raji B cell line undergo a phenotypic switch to morphologically and functionally resembling M cells. Our data demonstrate that C. albicans co-localizes with and invades preferentially M cells, providing evidence that the fungus can use M cells as a portal of entry into the intestinal barrier. In addition to active penetration, F-actin dependent endocytosis contributes to internalization of the fungus into M cells through a mechanism involving hypha-associated invasins including Ssa1 and Als3.


Assuntos
Candida albicans/fisiologia , Candidemia/microbiologia , Trato Gastrointestinal/microbiologia , Interações Hospedeiro-Patógeno , Nódulos Linfáticos Agregados/microbiologia , Linfócitos B/fisiologia , Adesão Celular , Linhagem Celular , Técnicas de Cocultura , Endocitose , Células Epiteliais/microbiologia , Células Epiteliais/fisiologia , Humanos , Microscopia Eletrônica de Varredura , Microscopia de Fluorescência
13.
Am J Physiol Gastrointest Liver Physiol ; 310(4): G240-8, 2016 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-26680736

RESUMO

Organotypic tissue slices provide seminatural, three-dimensional microenvironments for use in ex vivo study of specific organs and have advanced investigative capabilities compared with isolated cell cultures. Several characteristics of the gastrointestinal tract have made in vitro models for studying the intestine challenging, such as maintaining the intricate structure of microvilli, the intrinsic enteric nervous system, Peyer's patches, the microbiome, and the active contraction of gut muscles. In the present study, an organotypic intestinal slice model was developed that allows for functional investigation across regions of the intestine. Intestinal tissue slices were maintained ex vivo for several days in a physiologically relevant environment that preserved normal enterocyte structure, intact and proliferating crypt cells, submucosal organization, and muscle wall composure. Cell death was measured by a membrane-impermeable DNA binding indicator, ethidium homodimer, and less than 5% of cells were labeled in all regions of the villi and crypt epithelia at 24 h ex vivo. This tissue slice model demonstrated intact myenteric and submucosal neuronal plexuses and functional interstitial cells of Cajal to the extent that nonstimulated, segmental contractions occurred for up to 48 h ex vivo. To detect changes in physiological responses, slices were also assessed for segmental contractions in the presence and absence of antibiotic treatment, which resulted in slices with lesser or greater amounts of commensal bacteria, respectively. Segmental contractions were significantly greater in slices without antibiotics and increased native microbiota. This model renders mechanisms of neuroimmune-microbiome interactions in a complex gut environment available to direct observation and controlled perturbation.


Assuntos
Intestinos/imunologia , Intestinos/inervação , Animais , Antibacterianos/farmacologia , Bloqueadores dos Canais de Cálcio/farmacologia , Proliferação de Células/efeitos dos fármacos , Enterócitos/efeitos dos fármacos , Enterócitos/fisiologia , Enterócitos/ultraestrutura , Feminino , Mucosa Intestinal/imunologia , Mucosa Intestinal/inervação , Mucosa Intestinal/microbiologia , Mucosa Intestinal/fisiologia , Intestinos/microbiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microbiota , Modelos Biológicos , Contração Muscular/efeitos dos fármacos , Contração Muscular/fisiologia , Músculo Liso/fisiologia , Nicardipino/farmacologia , Técnicas de Cultura de Órgãos , Nódulos Linfáticos Agregados/imunologia , Nódulos Linfáticos Agregados/inervação , Nódulos Linfáticos Agregados/microbiologia
14.
PLoS Pathog ; 10(7): e1004270, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25079958

RESUMO

Vaccination represents an important instrument to control typhoid fever in humans and protects mice from lethal infection with mouse pathogenic serovars of Salmonella species. Mixed infections with tagged Salmonella can be used in combination with probabilistic models to describe the dynamics of the infection process. Here we used mixed oral infections with tagged Salmonella strains to identify bottlenecks in the infection process in naïve and vaccinated mice. We established a next generation sequencing based method to characterize the composition of tagged Salmonella strains which offers a fast and reliable method to characterise the composition of genome-tagged Salmonella strains. We show that initial colonization of Salmonella was distinguished by a non-Darwinian selection of few bacteria setting up the infection independently in gut associated lymphoid tissue and systemic compartments. Colonization of Peyer's patches fuels the sustained spread of bacteria into mesenteric lymph nodes via dendritic cells. In contrast, infection of liver and spleen originated from an independent pool of bacteria. Vaccination only moderately reduced invasion of Peyer's patches but potently uncoupled bacterial populations present in different systemic compartments. Our data indicate that vaccination differentially skews the capacity of Salmonella to colonize systemic and gut immune compartments and provide a framework for the further dissection of infection dynamics.


Assuntos
Trato Gastrointestinal/microbiologia , Mucosa Intestinal/microbiologia , Nódulos Linfáticos Agregados/microbiologia , Salmonelose Animal/microbiologia , Salmonella typhimurium/patogenicidade , Baço/microbiologia , Administração Oral , Animais , DNA Bacteriano/genética , Trato Gastrointestinal/imunologia , Sequenciamento de Nucleotídeos em Larga Escala , Camundongos , Camundongos Endogâmicos C57BL , Nódulos Linfáticos Agregados/imunologia , Salmonelose Animal/imunologia , Salmonelose Animal/prevenção & controle , Salmonella typhimurium/genética , Vacinação
15.
PLoS Pathog ; 10(6): e1004194, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24945271

RESUMO

Type III secretion systems (T3SS) are essential for virulence in dozens of pathogens, but are not required for growth outside the host. Therefore, the T3SS of many bacterial species are under tight regulatory control. To increase our understanding of the molecular mechanisms behind T3SS regulation, we performed a transposon screen to identify genes important for T3SS function in the food-borne pathogen Yersinia pseudotuberculosis. We identified two unique transposon insertions in YPTB2860, a gene that displays 79% identity with the E. coli iron-sulfur cluster regulator, IscR. A Y. pseudotuberculosis iscR in-frame deletion mutant (ΔiscR) was deficient in secretion of Ysc T3SS effector proteins and in targeting macrophages through the T3SS. To determine the mechanism behind IscR control of the Ysc T3SS, we carried out transcriptome and bioinformatic analysis to identify Y. pseudotuberculosis genes regulated by IscR. We discovered a putative IscR binding motif upstream of the Y. pseudotuberculosis yscW-lcrF operon. As LcrF controls transcription of a number of critical T3SS genes in Yersinia, we hypothesized that Yersinia IscR may control the Ysc T3SS through LcrF. Indeed, purified IscR bound to the identified yscW-lcrF promoter motif and mRNA levels of lcrF and 24 other T3SS genes were reduced in Y. pseudotuberculosis in the absence of IscR. Importantly, mice orally infected with the Y. pseudotuberculosis ΔiscR mutant displayed decreased bacterial burden in Peyer's patches, mesenteric lymph nodes, spleens, and livers, indicating an essential role for IscR in Y. pseudotuberculosis virulence. This study presents the first characterization of Yersinia IscR and provides evidence that IscR is critical for virulence and type III secretion through direct regulation of the T3SS master regulator, LcrF.


Assuntos
Sistemas de Secreção Bacterianos/genética , Proteínas de Escherichia coli/genética , Fatores de Transcrição/genética , Fatores de Virulência/genética , Yersinia pseudotuberculosis/genética , Yersinia pseudotuberculosis/patogenicidade , Sequência de Aminoácidos , Animais , Sítios de Ligação/genética , Elementos de DNA Transponíveis/genética , Escherichia coli/genética , Perfilação da Expressão Gênica , Regulação Bacteriana da Expressão Gênica , Fígado/imunologia , Fígado/microbiologia , Linfonodos/imunologia , Linfonodos/microbiologia , Camundongos , Dados de Sequência Molecular , Nódulos Linfáticos Agregados/imunologia , Nódulos Linfáticos Agregados/microbiologia , Regiões Promotoras Genéticas/genética , Ligação Proteica , Alinhamento de Sequência , Baço/imunologia , Baço/microbiologia , Transcrição Gênica , Transcriptoma/genética , Infecções por Yersinia pseudotuberculosis/imunologia , Infecções por Yersinia pseudotuberculosis/patologia
16.
Int J Med Microbiol ; 306(6): 357-66, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27107739

RESUMO

Enteropathogenic Yersinia enterocolitica (Ye) enters the host via contaminated food. After colonisation of the small intestine Ye invades the Peyer's patches (PPs) via M cells and disseminates to the mesenteric lymph nodes (MLNs), spleen and liver. Whether Ye uses other invasion routes and which pathogenicity factors are required remains elusive. Oral infection of lymphotoxin-ß-receptor deficient mice lacking PPs and MLNs with Ye revealed similar bacterial load in the spleen 1h post infection as wild-type mice, demonstrating a PP-independent dissemination route for Ye. Immunohistological analysis of the small intestine revealed Ye in close contact with mononuclear phagocytes (MPs), specifically CX3CR1(+) monocyte-derived cells (MCs) as well as CD103(+) dendritic cells (DCs). This finding was confirmed by flow cytometry and imaging flow cytometry analysis of lamina propria (LP) leukocytes showing CD103(+) DCs and MCs with intracellular Ye. Uptake of Ye by LP CD103(+) DCs and MCs was dependent on the pathogenicity factor invasin, whereas the adhesin YadA was dispensable as demonstrated by Ye deletion mutants. Furthermore, Ye were found exclusively associated with CD103(+) DCs in the MLNs from wild-type mice, but not from CCR7(-/-) mice, demonstrating a CCR7 dependent transport of Ye by CD103(+) DCs from LP to the MLNs. In contrast, dissemination of Ye to the spleen was dependent on MCs as significantly less Ye could be recovered from the spleen of CX3CR1(GFP/GFP) mice compared to wild-type mice. Altogether, MCs and CD103(+) DCs contribute to immediate invasion and dissemination of Ye. This together with data from other bacteria suggests MPs as general pathogenic entry site in the intestine.


Assuntos
Interações Hospedeiro-Patógeno , Intestino Delgado/patologia , Fagócitos/microbiologia , Yersiniose/patologia , Yersinia enterocolitica/imunologia , Yersinia enterocolitica/fisiologia , Animais , Carga Bacteriana , Feminino , Citometria de Fluxo , Imuno-Histoquímica , Intestino Delgado/imunologia , Intestino Delgado/microbiologia , Fígado/microbiologia , Linfonodos/imunologia , Linfonodos/microbiologia , Camundongos Endogâmicos C57BL , Nódulos Linfáticos Agregados/imunologia , Nódulos Linfáticos Agregados/microbiologia , Baço/microbiologia , Fatores de Tempo , Yersiniose/imunologia , Yersiniose/microbiologia
17.
FASEB J ; 29(2): 684-95, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25392266

RESUMO

Ingestion of a commensal bacteria, Lactobacillus rhamnosus JB-1, has potent immunoregulatory effects, and changes nerve-dependent colon migrating motor complexes (MMCs), enteric nerve function, and behavior. How these alterations occur is unknown. JB-1 microvesicles (MVs) are enriched for heat shock protein components such as chaperonin 60 heat-shock protein isolated from Escherichia coli (GroEL) and reproduce regulatory and neuronal effects in vitro and in vivo. Ingested labeled MVs were detected in murine Peyer's patch (PP) dendritic cells (DCs) within 18 h. After 3 d, PP and mesenteric lymph node DCs assumed a regulatory phenotype and increased functional regulatory CD4(+)25(+)Foxp3+ T cells. JB-1, MVs, and GroEL similarly induced phenotypic change in cocultured DCs via multiple pathways including C-type lectin receptors specific intercellular adhesion molecule-3 grabbing non-integrin-related 1 and Dectin-1, as well as TLR-2 and -9. JB-1 and MVs also decreased the amplitude of neuronally dependent MMCs in an ex vivo model of peristalsis. Gut epithelial, but not direct neuronal application of, MVs, replicated functional effects of JB-1 on in situ patch-clamped enteric neurons. GroEL and anti-TLR-2 were without effect in this system, suggesting the importance of epithelium neuron signaling and discrimination between pathways for bacteria-neuron and -immune communication. Together these results offer a mechanistic explanation of how Gram-positive commensals and probiotics may influence the host's immune and nervous systems.


Assuntos
Sistema Nervoso Entérico/fisiologia , Trato Gastrointestinal/inervação , Sistema Imunitário/fisiologia , Lacticaseibacillus rhamnosus/imunologia , Animais , Células da Medula Óssea/citologia , Linfócitos T CD4-Positivos/citologia , Chaperonina 60/metabolismo , Técnicas de Cocultura , Células Dendríticas/citologia , Células Dendríticas/microbiologia , Fatores de Transcrição Forkhead/metabolismo , Subunidade alfa de Receptor de Interleucina-2/metabolismo , Lectinas Tipo C/metabolismo , Linfonodos/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Neurônios/metabolismo , Peristaltismo , Nódulos Linfáticos Agregados/microbiologia , Fenótipo , Probióticos , Proteômica , Transdução de Sinais
18.
Hepatobiliary Pancreat Dis Int ; 15(4): 399-405, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27498580

RESUMO

BACKGROUND: Acute liver injury is a common clinical disorder associated with intestinal barrier injury and disturbance of intestinal microbiota. Probiotic supplementation has been reported to reduce liver injury; however, it is unclear whether enteropathogen infection exacerbates liver injury. The purpose of this study was to address this unanswered question using a rat model. METHODS: Oral supplementation with Salmonella enterica serovar enteritidis (S. enteritidis) was given to rats for 7 days. Different degrees of acute liver injury were then induced by intraperitoneal injection of D-galactosamine. The presence and extent of liver injury was assayed by measuring the concentrations of serum alanine aminotransferase, aspartate aminotransferase, and total bilirubin. Histology was used to observe liver tissue damage. Additionally, we measured the changes in plasma endotoxin, serum cytokines and bacterial translocation to clarify the mechanisms underlying intestinal microbiota associated liver injury. RESULTS: The levels of liver damage and endotoxin were significantly increased in the Salmonella infected rats with severe liver injury compared with the no infection rats with severe liver injury (P<0.01); The peyer's patch CD3+ T cell counts were increased significantly when the Salmonella infection with severe injury group was compared with the normal group (P<0.05). S. enteritidis pretreatment enhanced intestinal barrier impairment and bacterial translocation. CONCLUSIONS: Oral S. enteritidis administration exacerbates acute liver injury, especially when injury was severe. Major factors of the exacerbation include inflammatory and oxidative stress injuries induced by the translocated bacteria and associated endotoxins, as well as over-activation of the immune system in the intestine and liver.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas/microbiologia , Fígado/microbiologia , Infecções por Salmonella/microbiologia , Salmonella enteritidis/patogenicidade , Doença Aguda , Alanina Transaminase/sangue , Animais , Aspartato Aminotransferases/sangue , Translocação Bacteriana , Bilirrubina/sangue , Biomarcadores/sangue , Complexo CD3/imunologia , Doença Hepática Induzida por Substâncias e Drogas/sangue , Doença Hepática Induzida por Substâncias e Drogas/imunologia , Doença Hepática Induzida por Substâncias e Drogas/patologia , Citocinas/imunologia , Modelos Animais de Doenças , Endotoxinas/metabolismo , Galactosamina , Interações Hospedeiro-Patógeno , Fígado/metabolismo , Fígado/patologia , Masculino , Nódulos Linfáticos Agregados/imunologia , Nódulos Linfáticos Agregados/microbiologia , Ratos Sprague-Dawley , Infecções por Salmonella/sangue , Infecções por Salmonella/imunologia , Infecções por Salmonella/patologia , Salmonella enteritidis/imunologia , Salmonella enteritidis/metabolismo , Linfócitos T/imunologia , Linfócitos T/microbiologia , Fatores de Tempo
19.
Int J Food Sci Nutr ; 67(6): 641-9, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27198983

RESUMO

Probiotics exert numerous effects on human well-being. Here, heat-killed Lactobacillus plantarum BF-LP284 (H-Lp) was isolated as a potent immuno-modulator among 15 strains of lactobacilli in terms of TNF-α induction ability in peritoneal macrophages. In vitro TNF-α and IFN-γ induction in Peyer's patch (PP) cells was higher when incubated with H-Lp than with live L. plantarum BF-LP284 (L-Lp). Suppression of syngeneic Meth-A tumors in a murine model by oral administration of H-Lp was also greater than that of L-Lp and of controls. H-Lp stimulated IFN-γ production in spleen cells, which displayed inhibited tumor growth in Winn assays when treated with H-Lp. Moreover, H-Lp increased the ratio of CD3(+ )cells among peripheral blood mononuclear cells in Meth-A tumor-bearing mice, suggesting an H-Lp-mediated anti-tumor mechanism whereby immune cells that are activated by H-Lp in PP and acquire anti-tumor activity in the spleen migrate to tumor sites through lymphocyte homing to inhibit tumor growth.


Assuntos
Antineoplásicos/uso terapêutico , Lactobacillus plantarum , Neoplasias Experimentais/terapia , Probióticos , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Feminino , Temperatura Alta , Interferon gama/metabolismo , Leucócitos Mononucleares/metabolismo , Leucócitos Mononucleares/microbiologia , Macrófagos Peritoneais/microbiologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos ICR , Nódulos Linfáticos Agregados/citologia , Nódulos Linfáticos Agregados/metabolismo , Nódulos Linfáticos Agregados/microbiologia , Baço/citologia , Baço/metabolismo , Baço/microbiologia , Fator de Necrose Tumoral alfa/metabolismo
20.
Appl Microbiol Biotechnol ; 99(21): 9097-110, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26084888

RESUMO

Enterohemorrhagic Escherichia coli (EHEC) are major food-borne pathogens responsible for serious infections ranging from mild diarrhea to hemorrhagic colitis and life-threatening complications. Shiga toxins (Stxs) are the main virulence factor of EHEC. The antagonistic effect of a prophylactic treatment with the probiotic strain Saccharomyces cerevisiae against EHEC O157:H7 was investigated using complementary in vitro human colonic model and in vivo murine ileal loop assays. In vitro, the probiotic treatment had no effect on O157:H7 survival but favorably influenced gut microbiota activity through modulation of short-chain fatty acid production, increasing acetate production and decreasing that of butyrate. Both pathogen and probiotic strains had individual-dependent effects on human gut microbiota. For the first time, stx expression was followed in human colonic environment: at 9 and 12 h post EHEC infection, probiotic treatment significantly decreased stx mRNA levels. Besides, in murine ileal loops, the probiotic yeast specifically exerted a trophic effect on intestinal mucosa and inhibited O157:H7 interactions with Peyer's patches and subsequent hemorrhagic lesions. Taken together, the results suggest that S. cerevisiae may be useful in the fight against EHEC infection and that host associated factors such as microbiota could influence clinical evolution of EHEC infection and the effectiveness of probiotics.


Assuntos
Antibiose , Escherichia coli Êntero-Hemorrágica/crescimento & desenvolvimento , Infecções por Escherichia coli/prevenção & controle , Microbioma Gastrointestinal , Profilaxia Pré-Exposição/métodos , Probióticos/administração & dosagem , Saccharomyces cerevisiae/crescimento & desenvolvimento , Animais , Colo/microbiologia , Modelos Animais de Doenças , Ácidos Graxos Voláteis/metabolismo , Expressão Gênica , Perfilação da Expressão Gênica , Humanos , Íleo/microbiologia , Camundongos , Modelos Biológicos , Nódulos Linfáticos Agregados/microbiologia , Saccharomyces cerevisiae/fisiologia , Toxina Shiga/biossíntese , Fatores de Tempo , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA