Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 116
Filtrar
1.
Cell Rep ; 35(2): 108995, 2021 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-33852847

RESUMO

The complement fragment C5a is closely associated with adaptive immune induction in the mucosa. However, the mechanisms that control CD8+ T cell responses by C5a have not been extensively explored. This study reveals that C5/C5a in the Peyer's patch (PP) subepithelial dome increases upon oral Listeria infection. We hypothesize that C5aR+ PP cells play an important role in the induction of antigen-specific T cell immunity. Using single-cell RNA sequencing, we identify C5aR- and lysozyme-expressing dendritic cells (C5aR+ LysoDCs) in PP and examine their role in CD8+ T cell immune induction. Stimulation of C5aR+ LysoDCs by C5a increases reactive oxygen species levels, leading to efficient antigen cross-presentation, which elicits an antigen-specific CD8+ T cell response. In C5-deficient mice, oral co-administration of C5a and Listeria enhances Listeria-specific cytotoxic T cell levels. Collectively, these findings suggest a role of the complement system in intestinal T cell immunity.


Assuntos
Complemento C5a/imunologia , Apresentação Cruzada , Mucosa Intestinal/imunologia , Listeria monocytogenes/imunologia , Nódulos Linfáticos Agregados/imunologia , Receptor da Anafilatoxina C5a/genética , Linfócitos T Citotóxicos/imunologia , Imunidade Adaptativa , Animais , Apresentação de Antígeno , Complemento C5a/genética , Complemento C5a/farmacologia , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Células Dendríticas/microbiologia , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Imunidade nas Mucosas , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/microbiologia , Listeria monocytogenes/patogenicidade , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Monócitos/efeitos dos fármacos , Monócitos/imunologia , Monócitos/microbiologia , Muramidase/genética , Muramidase/imunologia , Nódulos Linfáticos Agregados/efeitos dos fármacos , Nódulos Linfáticos Agregados/microbiologia , Espécies Reativas de Oxigênio/imunologia , Espécies Reativas de Oxigênio/metabolismo , Receptor da Anafilatoxina C5a/imunologia , Análise de Célula Única , Linfócitos T Citotóxicos/efeitos dos fármacos , Linfócitos T Citotóxicos/microbiologia
2.
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
3.
Sci Rep ; 8(1): 14881, 2018 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-30291258

RESUMO

The lipid sensor oleoylethanolamide (OEA), an endogenous high-affinity agonist of peroxisome proliferator-activated receptor-α (PPAR-α) secreted in the proximal intestine, is endowed with several distinctive homeostatic properties, such as control of appetite, anti-inflammatory activity, stimulation of lipolysis and fatty acid oxidation. When administered exogenously, OEA has beneficial effects in several cognitive paradigms; therefore, in all respects, OEA can be considered a hormone of the gut-brain axis. Here we report an unexplored modulatory effect of OEA on the intestinal microbiota and on immune response. Our study shows for the first time that sub-chronic OEA administration to mice fed a normal chow pellet diet, changes the faecal microbiota profile, shifting the Firmicutes:Bacteroidetes ratio in favour of Bacteroidetes (in particular Bacteroides genus) and decreasing Firmicutes (Lactobacillus), and reduces intestinal cytokines expression by immune cells isolated from Peyer's patches. Our results suggest that sub-chronic OEA treatment modulates gut microbiota composition towards a "lean-like phenotype", and polarises gut-specific immune responses mimicking the effect of a diet low in fat and high in polysaccharides content.


Assuntos
Endocanabinoides/farmacologia , Microbioma Gastrointestinal/efeitos dos fármacos , Fatores Imunológicos/farmacologia , Ácidos Oleicos/farmacologia , PPAR alfa/agonistas , Nódulos Linfáticos Agregados/efeitos dos fármacos , Animais , Citocinas/análise , Citocinas/imunologia , Endocanabinoides/administração & dosagem , Fatores Imunológicos/administração & dosagem , Masculino , Camundongos , Ácidos Oleicos/administração & dosagem , Nódulos Linfáticos Agregados/imunologia , Nódulos Linfáticos Agregados/microbiologia
4.
PLoS One ; 13(6): e0198359, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29894476

RESUMO

Oxaliplatin is a platinum-based chemotherapeutic used for cancer treatment. Its use associates with peripheral neuropathies and chronic gastrointestinal side-effects. Oxaliplatin induces immunogenic cell death by provoking the presentation of damage associated molecular patterns. The damage associated molecular patterns high-mobility group box 1 (HMGB1) protein exerts pro-inflammatory cytokine-like activity and binds to toll-like receptors (namely TLR4). Gastrointestinal microbiota may influence chemotherapeutic efficacy and contribute to local and systemic inflammation. We studied effects of oxaliplatin treatment on 1) TLR4 and high-mobility group box 1 expression within the colon; 2) gastrointestinal microbiota composition; 3) inflammation within the colon; 4) changes in Peyer's patches and mesenteric lymph nodes immune populations in mice. TLR4+ cells displayed pseudopodia-like extensions characteristic of antigen sampling co-localised with high-mobility group box 1 -overexpressing cells in the colonic lamina propria from oxaliplatin-treated animals. Oxaliplatin treatment caused significant reduction in Parabacteroides and Prevotella1, but increase in Prevotella2 and Odoribacter bacteria at the genus level. Downregulation of pro-inflammatory cytokines and chemokines in colon samples, a reduction in macrophages and dendritic cells in mesenteric lymph nodes were found after oxaliplatin treatment. In conclusion, oxaliplatin treatment caused morphological changes in TLR4+ cells, increase in gram-negative microbiota and enhanced HMGB1 expression associated with immunosuppression in the colon.


Assuntos
Bactérias/classificação , Colo/metabolismo , Microbioma Gastrointestinal/efeitos dos fármacos , Proteína HMGB1/metabolismo , Oxaliplatina/efeitos adversos , Nódulos Linfáticos Agregados/metabolismo , Receptor 4 Toll-Like/metabolismo , Animais , Bactérias/efeitos dos fármacos , Bactérias/genética , Colo/efeitos dos fármacos , Colo/microbiologia , DNA Bacteriano/genética , DNA Ribossômico/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Sequenciamento de Nucleotídeos em Larga Escala , Antígenos Comuns de Leucócito/metabolismo , Masculino , Camundongos , Nódulos Linfáticos Agregados/efeitos dos fármacos , Nódulos Linfáticos Agregados/microbiologia , Filogenia , RNA Ribossômico 16S/genética , Análise de Sequência de DNA
5.
Food Funct ; 9(5): 2989-2997, 2018 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-29774357

RESUMO

Specific Lactobacillus reuteri is autochthonous Lactobacillus species in humans with potential application in food production as a probiotic. The difference in colonizing Peyer's patches (PP) might decide their health-promoting properties. We aimed to investigate the difference between PP- and lumen-specific L. reuteri on antimicrobial peptide expression in this study. L. reuteri strains were isolated from PP and the lumen of C57BL/6J mice, which were used to treat mice. PP-specific L. reuteri cells stimulate RegIIIγ mRNA expression of the crypt epithelial sample. PP-specific L. reuteri induces accumulation of extracellular DNA (eDNA) in the bottom of crypts. eDNA was extracted from the small-intestinal mucus, the yield of which was significantly increased after the PP-specific L. reuteri treatment. And it increased cytokine production in RAW264.7 murine macrophages. PP-specific L. reuteri significantly increased the relative abundance of Bacteroidetes-, Lactobacillus-, and Proteobacteria-derived eDNA. However, the levels of Strentrophomonas-derived eDNA decreased. These results provide a rationale for the screening of human derived L. reuteri with an immune-modulatory property.


Assuntos
Peptídeos Catiônicos Antimicrobianos/metabolismo , DNA Bacteriano/genética , Intestino Delgado/metabolismo , Limosilactobacillus reuteri/genética , Nódulos Linfáticos Agregados/metabolismo , Animais , Citocinas/metabolismo , DNA Bacteriano/metabolismo , Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Humanos , Intestino Delgado/microbiologia , Limosilactobacillus reuteri/classificação , Limosilactobacillus reuteri/isolamento & purificação , Limosilactobacillus reuteri/metabolismo , Macrófagos/metabolismo , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Nódulos Linfáticos Agregados/microbiologia , Filogenia , Células RAW 264.7 , Especificidade da Espécie
6.
J Vet Med Sci ; 79(11): 1826-1835, 2017 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-28993550

RESUMO

The mechanism by which indigenous bacteria on the follicle-associated epithelium (FAE) of lymphatic follicles (LFs) accelerate the differentiation of microvillous columnar epithelial cells (MV) into M-cells was immunohistochemically investigated in rat Peyer's patches. The results showed that the number of Toll-like receptor (TLR) -4+ M-cells was greater in the FAE with expansion of bacterial colonies (LFs with bacterial colonies on the FAE: b-LF) than the FAE without expansion of bacterial colonies (nb-LF). TLR-4 was also expressed in the striated borders of MV upstream next to M-cells in the FAE of the b-LF. TLR-4+ vesicles were frequently detected in the cytoplasms of MV with TLR-4+ striated borders upstream next to TLR-4+ M-cells in the FAE of b-LF. These findings suggest that TLR-4+ MV take up TLR-4 ligands and differentiate into M-cells in the b-LF. Neither the distribution of RANK nor that of RANKL was coincident with that of M-cells in the b-LF. Moreover, RANK, but not RANKL, was expressed in intestinal villi, whereas cleaved caspase-3 was immunonegative in the MV and M-cells of the FAE, unlike in villous epithelial cells. Therefore, RANK/RANKL signaling in the LF might contribute to the down-regulation of epithelial apoptosis to facilitate the differentiation of MV into M-cells in rat Peyer's patches.


Assuntos
Bactérias/crescimento & desenvolvimento , Diferenciação Celular/fisiologia , Nódulos Linfáticos Agregados/crescimento & desenvolvimento , Nódulos Linfáticos Agregados/microbiologia , Animais , Células Epiteliais , Imuno-Histoquímica , Mucosa Intestinal/citologia , Masculino , Ligante RANK , Ratos Wistar , Organismos Livres de Patógenos Específicos , Receptor 4 Toll-Like
7.
J Control Release ; 264: 55-65, 2017 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-28842314

RESUMO

We describe a novel live oral vaccine type. Conceptually, this vaccine is based on a non-lytic, recombinant filamentous bacteriophage that displays an antigen of interest. To provide proof of concept we used the amino-terminal part of a conserved influenza A virus epitope, i.e. matrix protein 2 ectodomain (M2e) residues 2 to 16, as the antigen of interest. Rather than using the phages as purified virus-like particles as a vaccine, these phages were delivered to intestinal Peyer's patches as a live bacterium-phage combination that comprises Escherichia coli cells that conditionally express invasin derived from Yersinia pseudotuberculosis. Invasin-expressing E. coli cells were internalized by mammalian Hep-2 cells in vitro and adhered to mouse intestinal microfold (M) cells ex vivo. Invasin-expressing E. coli cells were permissive for recombinant filamentous bacteriophage f88 that displays M2e and became persistently infected. Oral administration of the live engineered E. coli-invasin-phage combination to mice induced M2e-specific serum IgG antibodies. Mice that had been immunized with invasin-expressing E. coli cells that carried M2e2-16 displaying fd phages seroconverted to M2e and showed partial protection against challenge with influenza A virus. Oral delivery of a live vaccine comprising a bacterial host that is targeted to Peyer's patches and is persistently infected with an antigen-displaying phage, can thus be exploited as an oral vaccine.


Assuntos
Antígenos/imunologia , Bacteriófagos/imunologia , Escherichia coli/virologia , Vírus da Influenza A/imunologia , Vacinas contra Influenza , Proteínas da Matriz Viral/imunologia , Adesinas Bacterianas/imunologia , Administração Oral , Animais , Linhagem Celular Tumoral , Escherichia coli/imunologia , Feminino , Humanos , Imunoglobulina G/sangue , Camundongos Endogâmicos BALB C , Nódulos Linfáticos Agregados/microbiologia , Domínios Proteicos/imunologia
8.
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
9.
Sci Rep ; 7: 44655, 2017 03 20.
Artigo em Inglês | MEDLINE | ID: mdl-28317910

RESUMO

Enterohemorrhagic Escherichia coli (EHEC) are major food-borne pathogens whose survival and virulence in the human digestive tract remain unclear owing to paucity of relevant models. EHEC interact with the follicle-associated epithelium of Peyer's patches of the distal ileum and translocate across the intestinal epithelium via M-cells, but the underlying molecular mechanisms are still unknown. Here, we investigated the involvement of Long polar fimbriae (Lpf) in EHEC pathogenesis. Of the 236 strains tested, a significant association was observed between the presence of lpf operons and pathogenicity. In sophisticated in vitro models of the human gastro-intestinal tract, lpf expression was induced during transit through the simulated stomach and small intestine, but not in the colonic compartment. To investigate the involvement of Lpf in EHEC pathogenesis, lpf isogenic mutants and their relative trans-complemented strains were generated. Translocation across M-cells, interactions with murine ileal biopsies containing Peyer's patches and the number of hemorrhagic lesions were significantly reduced with the lpf mutants compared to the wild-type strain. Complementation of lpf mutants fully restored the wild-type phenotypes. Our results indicate that (i) EHEC might colonize the terminal ileum at the early stages of infection, (ii) Lpf are an important player in the interactions with Peyer's patches and M-cells, and could contribute to intestinal colonization.


Assuntos
Escherichia coli Êntero-Hemorrágica/patogenicidade , Infecções por Escherichia coli/microbiologia , Fímbrias Bacterianas/metabolismo , Nódulos Linfáticos Agregados/microbiologia , Nódulos Linfáticos Agregados/patologia , Animais , Aderência Bacteriana/genética , Translocação Bacteriana , Células CACO-2 , Escherichia coli Êntero-Hemorrágica/classificação , Escherichia coli Êntero-Hemorrágica/genética , Células Epiteliais/microbiologia , Células Epiteliais/patologia , Escherichia coli O157 , Proteínas de Escherichia coli/metabolismo , Regulação Bacteriana da Expressão Gênica , Genes Bacterianos , Humanos , Intestino Delgado/microbiologia , Intestino Delgado/patologia , Masculino , Camundongos , Modelos Biológicos , Óperon/genética , Sorotipagem , Estômago/microbiologia , Estômago/patologia , Virulência
10.
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
11.
J Microbiol Biotechnol ; 26(6): 1035-45, 2016 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-26975767

RESUMO

To evaluate the effects of lactic acid bacteria (LAB) on Peyer's patch cells, mice were treated with a high dose of kanamycin to disturb the gut microbial environment. The overarching goal was to explore the potential of LAB for use as a dietary probiotic that buffers the negative consequences of antibiotic treatment. In vitro, LAB stimulated the production of immunoglobulin A (IgA) from isolated Peyer's patch cells. Inflammation-related genes (TNF-α, IL-1ß, and IL-8) were up-regulated in Caco-2 cells stimulated with lipopolysaccharide (LPS), while tight-junction-related genes (ZO-1 and occludin) were down-regulated; the effects of LPS on inflammatory gene and tight-junction gene expression were reversed by treatment with LAB. Mice treated with a high dose of kanamycin showed increased serum IgE levels and decreases in serum IgA and fecal IgA levels; the number of Peyer's patch cells decreased with kanamycin treatment. However, subsequent LAB treatment was effective in reducing the serum IgE level and recovering the serum IgA and fecal IgA levels, as well as the number of Peyer's patch cells. In addition, ZO-1 and occludin mRNA levels were up-regulated in the ileum tissues of mice receiving LAB treatment. Lactic acid bacteria can enhance the intestinal immune system by improving the integrity of the intestinal barrier and increasing the production of IgA in Peyer's patches. Lactic acid bacteria should be considered a potential probiotic candidate for improving intestinal immunity, particularly in mitigating the negative consequences of antibiotic use.


Assuntos
Microbioma Gastrointestinal , Lactobacillus/fisiologia , Nódulos Linfáticos Agregados/imunologia , Nódulos Linfáticos Agregados/fisiologia , Probióticos/uso terapêutico , Junções Íntimas/genética , Animais , Antibacterianos/administração & dosagem , Antibacterianos/efeitos adversos , Células CACO-2 , Humanos , Imunoglobulina A/biossíntese , Imunoglobulina A/sangue , Imunoglobulina A/imunologia , Imunoglobulina E/sangue , Interleucina-1beta/genética , Interleucina-8/genética , Canamicina/administração & dosagem , Canamicina/efeitos adversos , Lipopolissacarídeos/farmacologia , Camundongos , Ocludina/genética , Nódulos Linfáticos Agregados/citologia , Nódulos Linfáticos Agregados/microbiologia , Fator de Necrose Tumoral alfa/genética , Proteína da Zônula de Oclusão-1/genética
12.
PLoS One ; 11(1): e0148216, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26820624

RESUMO

BACKGROUND & AIMS: Intestinal microfold (M) cells are specialized epithelial cells that act as gatekeepers of luminal antigens in the intestinal tract. They play a critical role in the intestinal mucosal immune response through transport of viruses, bacteria and other particles and antigens across the epithelium to immune cells within Peyer's patch regions and other mucosal sites. Recent studies in mice have demonstrated that M cells are generated from Lgr5+ intestinal stem cells (ISCs), and that infection with Salmonella enterica serovar Typhimurium increases M cell formation. However, it is not known whether and how these findings apply to primary human small intestinal epithelium propagated in an in vitro setting. METHODS: Human intestinal crypts were grown as monolayers with growth factors and treated with recombinant RANKL, and assessed for mRNA transcripts, immunofluorescence and uptake of microparticles and S. Typhimurium. RESULTS: Functional M cells were generated by short-term culture of freshly isolated human intestinal crypts in a dose- and time-dependent fashion. RANKL stimulation of the monolayer cultures caused dramatic induction of the M cell-specific markers, SPIB, and Glycoprotein-2 (GP2) in a process primed by canonical WNT signaling. Confocal microscopy demonstrated a pseudopod phenotype of GP2-positive M cells that preferentially take up microparticles. Furthermore, infection of the M cell-enriched cultures with the M cell-tropic enteric pathogen, S. Typhimurium, led to preferential association of the bacteria with M cells, particularly at lower inoculum sizes. Larger inocula caused rapid induction of M cells. CONCLUSIONS: Human intestinal crypts containing ISCs can be cultured and differentiate into an epithelial layer with functional M cells with characteristic morphological and functional properties. This study is the first to demonstrate that M cells can be induced to form from primary human intestinal epithelium, and that S. Typhimurium preferentially infect these cells in an in vitro setting. We anticipate that this model can be used to generate large numbers of M cells for further functional studies of these key cells of intestinal immune induction and their impact on controlling enteric pathogens and the intestinal microbiome.


Assuntos
Mucosa Intestinal/citologia , Intestino Delgado/citologia , Nódulos Linfáticos Agregados/citologia , Células-Tronco/citologia , Técnicas de Cultura de Células/métodos , Diferenciação Celular , Células Cultivadas , Humanos , Imunidade nas Mucosas , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Intestino Delgado/imunologia , Intestino Delgado/microbiologia , Nódulos Linfáticos Agregados/imunologia , Nódulos Linfáticos Agregados/microbiologia , Ligante RANK/imunologia , Salmonella typhimurium/imunologia , Células-Tronco/imunologia
13.
Nutrition ; 32(2): 265-72, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26620713

RESUMO

OBJECTIVE: Diet-induced inflammation in the small intestine may represent an early event that precedes and predisposes to obesity and insulin resistance. This is related to decrease of lactobacilli in Peyer's patches (PP) revealed in our previous study. The present study aimed to clarify specific changes of PP Lactobacillus on the strain level and related biological activity. METHODS: C57 BL/6 J male mice were fed with either low-fat diet (control [CT]; 10% calories from fat) or high-fat diet (HFD; 50% calories from fat) for 25 wk, and the HFD-fed mice were classified into obesity prone (OP) or obesity resistant (OR) on the basis of their body weight gain. Lactobacillus was isolated from PP using a selective medium. Oxidative resistance and cytokine-inducing effect were analyzed in vitro. RESULTS: We obtained 52, 18, and 22 isolates from CT, OP, and OR mice, respectively. They belonged to 13 different types according to enterobacterial repetitive intergenic consensus sequence-PCR analysis. Lactobacillus reuteri was the most abundant strain, but its abundance in OP mice was much lower than that in CT and OR mice. This strain includes eight subgroups according to genotyping. L. reuteri L3 and L. reuteri L8 were the specific strains found in CT and OP mice, respectively. Oxidative-resistant L. reuteri was much higher in HFD-fed mice. When co-cultured with PP cells, L8 induced higher production of proinflammatory cytokines such as interleukin (IL)-6, IL-12, and tumor necrosis factor-α, whereas L3 induced higher production of an anti-inflammatory cytokine (IL-10). CONCLUSION: HFD may induce oxidative stress that drives strain selection of Lactobacillus strains, resulting in decreased anti-inflammatory response in PP.


Assuntos
Dieta Hiperlipídica/efeitos adversos , Limosilactobacillus reuteri , Obesidade/fisiopatologia , Estresse Oxidativo , Nódulos Linfáticos Agregados/microbiologia , Animais , Anti-Inflamatórios/metabolismo , Dieta com Restrição de Gorduras , Genótipo , Resistência à Insulina , Interleucina-10/sangue , Interleucina-12/sangue , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Microbiota , Obesidade/microbiologia , Fator de Necrose Tumoral alfa/sangue
14.
Clin Nutr ; 35(5): 1066-72, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-26302851

RESUMO

BACKGROUND: The hyperglycemia leads to increased oxidative stress, causing lipid peroxidation and imbalance in the immune system. AIMS: To investigate the effect of Kefir - a probiotic containing different strains - on metabolic parameters, cytokines, nitric oxide (NO) production, phagocytic activity of peritoneal macrophages and intestinal modulation in diabetes. METHODS: Wistar rats received injection of streptozotocin (45 mg/kg, intravenously) and diabetes was defined as glycemia ≥200 mg/dL. The animals were distributed in four groups: control (CTL); control Kefir (CTLK); diabetic (DM); diabetic Kefir (DMK). Kefir was given at 1.8 mL/day by gavage, started on the 5th day of diabetes, during 8 weeks. The animals were allocated in metabolic cages, pre and post treatment with Kefir, for measurement of the metabolic parameters, such as water intake, food intake, diuresis, glycemia, body mass, insulin and lipid profile, these last two were only measured at the end of Kefir protocol. After treatment, the animals were euthanized and the peritoneal cavity was prepared, resident macrophages were collected and cultured for analysis of the phagocytic activity, cytokines (IL-10, TNF-α, IL-17, IL-1ß) and NO. The intestinal modulation was performed by the quantification of Peyer's patches (PP) in the small intestine. The data were presented as mean ± SEM, with significance of p < 0.05. RESULTS: DM when compared to CTL showed increase in water intake (133 ± 7 vs. 28 ± 1 mL, p < 0.0001), food intake (40 ± 2 vs. 16 ± 1 g, p < 0.0001), diuresis (102 ± 5 vs. 13 ± 1 mL, p < 0.0001) and glycemia (567 ± 12 vs. 84 ± 3 mg/dL, p < 0.0001), while in DMK group all these metabolic parameters were decreased (96 ± 14; 36 ± 1; 86 ± 7 and 407 ± 19, respectively, p < 0.0001), presenting increase of body mass (42 ± 5 vs. 16 ± 4Δ, p < 0.0001) and insulin levels (0.3 ± 0.8 vs. 0.1 ± 0.04 ng/mL, p < 0.0001) compared to DM. The lipid profile of the diabetic groups showed tendency to increase compared to the respective controls. In relation to function of peritoneal macrophages, DMK group vs. DM showed improvement in phagocytic capacity (70 ± 5 vs. 51 ± 7%, p = 0,0023) and increased concentration of all the cytokines analyzed (pg/mL), as IL-10 (926 ± 69 vs. 556 ± 92, p = 0.0004), TNF-α (178 ± 20 vs. 109 ± 20, p = 0.005), IL-17 (33 ± 1 vs. 9 ± 1, p = 0.0001) and IL-1ß (102 ± 14 vs. 70 ± 5, p = 0.0129), after 24 h of LPS stimulation; including NO bioavailability after 24 h (102 ± 9 vs. 66 ± 5 µM/mL, p = 0.0029) or 48 h (143 ± 8 vs. 119 ± 4 µM/mL, p = 0.0102) of LPS stimulation. Moreover, the number of PP in the whole small intestine of DMK group was also increased as compared to DM (22 ± 1 vs. 18 ± 1, p = 0.0292). CONCLUSION: These results show that Kefir has a potential to modulate the immune response and activate peritoneal macrophages in diabetic animals, which suggests that it could enhance the immunocompetence of patients affected by diabetes mellitus. The hypoglycemic effect of this probiotic could be used as a tool to control glycemia, reducing or delaying the onset of complications associated with this disease.


Assuntos
Diabetes Mellitus Experimental/terapia , Microbioma Gastrointestinal , Imunomodulação , Macrófagos Peritoneais/microbiologia , Óxido Nítrico/metabolismo , Probióticos/administração & dosagem , Animais , Glicemia/metabolismo , Sobrevivência Celular , Citocinas/metabolismo , Diabetes Mellitus Experimental/microbiologia , Intestino Delgado/metabolismo , Intestino Delgado/microbiologia , Kefir , Masculino , Nódulos Linfáticos Agregados/metabolismo , Nódulos Linfáticos Agregados/microbiologia , Ratos , Ratos Wistar
15.
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
16.
Inflamm Bowel Dis ; 22(1): 68-81, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26512715

RESUMO

BACKGROUND: Adherent-invasive Escherichia coli (AIEC) associated with Crohn's disease target M cells lining Peyer's patches (PPs) through the expression of long polar fimbriae (LPF) and survive macrophage killing. Invasion of PPs constitutes a way to colonize the mucosa for bacteria able to escape or resist killing of underlying immune cells. We aimed to identify new virulence factors involved in PPs colonization by AIEC. METHODS: The presence of gipA (Growth in PPs) gene was determined by polymerase chain reaction. In vivo experiments were performed using CEABAC10 transgenic mice. Intramacrophagic behavior of AIEC was assessed in murine bone marrow-derived macrophages and human monocyte-derived macrophages. Cytokines production was quantified by ELISA. RESULTS: A higher prevalence of gipA-positive E. coli was observed in patients with Crohn's disease (27.3%) compared with controls (17.2%). Unlike non-AIEC strains, all gipA-positive AIEC strains also harbored lpfA. GipA deletion impaired AIEC translocation across M cells and their replication inside macrophages. GipA expression was induced by gastrointestinal (bile salts) and phagolysosomal (reactive oxygen species and acid pH) conditions. GipA deletion decreased lpfA mRNA level in AIEC bacteria. Survival of AIEC-ΔgipA bacteria was reduced in medium containing H2O2 or acidic pH. GipA deletion impaired AIEC colonization of PPs and dissemination to mesenteric lymph nodes in mice. CONCLUSIONS: GipA is required for optimal colonization of mouse PPs and survival within macrophages by AIEC, suggesting that this factor plays a role in AIEC promotion of Crohn's disease. Detection of gipA and lpfA could be a predictor for the presence of AIEC.


Assuntos
Aderência Bacteriana , Doença de Crohn/metabolismo , Infecções por Escherichia coli/metabolismo , Escherichia coli/patogenicidade , Nódulos Linfáticos Agregados/microbiologia , Fatores de Virulência/metabolismo , Sequência de Aminoácidos , Animais , Estudos de Casos e Controles , Doença de Crohn/complicações , Doença de Crohn/microbiologia , Escherichia coli/genética , Escherichia coli/isolamento & purificação , Infecções por Escherichia coli/etiologia , Infecções por Escherichia coli/patologia , Fímbrias Bacterianas/metabolismo , Fímbrias Bacterianas/microbiologia , Fímbrias Bacterianas/patologia , Seguimentos , Humanos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Mucosa Intestinal/patologia , Macrófagos/metabolismo , Macrófagos/microbiologia , Macrófagos/patologia , Camundongos , Dados de Sequência Molecular , Nódulos Linfáticos Agregados/metabolismo , Homologia de Sequência de Aminoácidos
17.
Res Microbiol ; 166(8): 626-32, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26272025

RESUMO

Lactobacillus plantarum C4, previously isolated from kefir and characterized as a potential probiotic strain, was tested for its protective and immunomodulatory capacity in a murine model of yersiniosis. The inoculation of BALB/c mice with a low pathogenicity serotype O9 strain of Yersinia enterocolitica results in a prolonged intestinal infection with colonization of Peyer's patches. Pretreatment with C4 was without effect on fecal excretion of yersiniae, but shortened the colonization of Peyer's patches. This protective effect was associated with pro-inflammatory status in the intestinal mucosa (TNF-α production in infected mice was increased by C4) and an increase in total IgA secretion. At a systemic level, C4 did not promote a pro-inflammatory response, although production of the immunoregulatory cytokine IFN-γ was enhanced. These findings suggest that L. plantarum C4 can increase resistance to intestinal infections through its immunomodulatory activity.


Assuntos
Produtos Fermentados do Leite/microbiologia , Lactobacillus plantarum/isolamento & purificação , Lactobacillus plantarum/fisiologia , Probióticos , Yersiniose/prevenção & controle , Yersinia enterocolitica , Animais , Citocinas/imunologia , Modelos Animais de Doenças , Fezes/microbiologia , Feminino , Imunoglobulina A Secretora/imunologia , Imunomodulação , Interferon gama/sangue , Interferon gama/imunologia , Intestinos/imunologia , Intestinos/microbiologia , Lactobacillus plantarum/crescimento & desenvolvimento , Camundongos Endogâmicos BALB C , Nódulos Linfáticos Agregados/imunologia , Nódulos Linfáticos Agregados/microbiologia , Fator de Necrose Tumoral alfa/imunologia , Yersiniose/imunologia , Yersinia enterocolitica/crescimento & desenvolvimento , Yersinia enterocolitica/imunologia
18.
BMC Vet Res ; 11: 74, 2015 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-25889716

RESUMO

BACKGROUND: Paratuberculosis caused by Mycobacterium avium subsp. paratuberculosis (MAP) is difficult to control due to a long phase of clinically non-apparent (latent) infection for which sensitive diagnostics are lacking. A defined animal model for this phase of the infection can help to investigate host-MAP interactions in apparently healthy animals and identify surrogate markers for disease progress and might also serve as challenge model for vaccines. To establish such a model in goats, different age at inoculation and doses of oral inoculum of MAP were compared. Clinical signs, faecal shedding as well as MAP-specific antibody, IFN-γ and IL-10 responses were used for in vivo monitoring. At necropsy, about one year after inoculation (pi), pathomorphological findings and bacterial organ burden (BOB) were scored. RESULTS: MAP infection manifested in 26/27 inoculated animals irrespective of age at inoculation and dose. Clinical signs developed in three goats. Faecal shedding, IFN-γ and antibody responses emerged 6, 10-14 and 14 wpi, respectively, and continued with large inter-individual variation. One year pi, lesions were detected in 26 and MAP was cultured from tissues of 23 goats. Positive animals subdivided in those with high and low overall BOB. Intestinal findings resembled paucibacillary lesions in 23 and multibacillary in 4 goats. Caseous and calcified granulomas predominated in intestinal LNN. BOB and lesion score corresponded well in intestinal mucosa and oGALT but not in intestinal LNN. CONCLUSIONS: A defined experimental infection model for the clinically non-apparent phase of paratuberculosis was established in goats as suitable basis for future studies.


Assuntos
Doenças das Cabras/microbiologia , Mycobacterium avium subsp. paratuberculosis , Paratuberculose/patologia , Animais , Formação de Anticorpos , Infecções Assintomáticas , Derrame de Bactérias , Progressão da Doença , Doenças das Cabras/patologia , Cabras/microbiologia , Interferon gama/sangue , Interleucina-10/sangue , Linfonodos/microbiologia , Linfonodos/patologia , Masculino , Paratuberculose/microbiologia , Nódulos Linfáticos Agregados/microbiologia , Nódulos Linfáticos Agregados/patologia
19.
Mucosal Immunol ; 8(3): 650-60, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25336168

RESUMO

The microfold (M) cell residing in the follicle-associated epithelium is a specialized epithelial cell that initiates mucosal immune responses by sampling luminal antigens. The differentiation process of M cells remains unclear due to limitations of analytical methods. Here we found that M cells were classified into two functionally different subtypes based on the expression of Glycoprotein 2 (GP2) by newly developed image cytometric analysis. GP2-high M cells actively took up luminal microbeads, whereas GP2-negative or low cells scarcely ingested them, even though both subsets equally expressed the other M-cell signature genes, suggesting that GP2-high M cells represent functionally mature M cells. Further, the GP2-high mature M cells were abundant in Peyer's patch but sparse in the cecal patch: this was most likely due to a decrease in the nuclear translocation of RelB, a downstream transcription factor for the receptor activator of nuclear factor-κB signaling. Given that murine cecum contains a protrusion of beneficial commensals, the restriction of M-cell activity might contribute to preventing the onset of any excessive immune response to the commensals through decelerating the M-cell-dependent uptake of microorganisms.


Assuntos
Imunidade nas Mucosas , Animais , Ceco/citologia , Ceco/imunologia , Ceco/microbiologia , Diferenciação Celular , Linhagem da Célula/imunologia , Quimiocinas CC/genética , Quimiocinas CC/imunologia , Citocinas/genética , Citocinas/imunologia , Células Epiteliais/citologia , Células Epiteliais/imunologia , Células Epiteliais/microbiologia , Proteínas Ligadas por GPI/genética , Proteínas Ligadas por GPI/imunologia , Regulação da Expressão Gênica , Mucosa Intestinal/citologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Proteínas Inflamatórias de Macrófagos/genética , Proteínas Inflamatórias de Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Microbiota/imunologia , Microscopia Confocal , NF-kappa B/genética , NF-kappa B/imunologia , Nódulos Linfáticos Agregados/citologia , Nódulos Linfáticos Agregados/imunologia , Nódulos Linfáticos Agregados/microbiologia , Fagocitose/genética , Fagocitose/imunologia , Ligante RANK/genética , Ligante RANK/imunologia , Receptor Ativador de Fator Nuclear kappa-B/genética , Receptor Ativador de Fator Nuclear kappa-B/imunologia , Transdução de Sinais , Fator de Transcrição RelB/genética , Fator de Transcrição RelB/imunologia , Fatores de Necrose Tumoral/genética , Fatores de Necrose Tumoral/imunologia
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA