Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
mBio ; 15(4): e0007824, 2024 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-38470269

RESUMO

Inflammatory bowel disease (IBD) is a group of inflammatory conditions of the gastrointestinal tract. The etiology of IBD remains elusive, but the disease is suggested to arise from the interaction of environmental and genetic factors that trigger inadequate immune responses and inflammation in the intestine. The gut microbiome majorly contributes to disease as an environmental variable, and although some causative bacteria are identified, little is known about which specific members of the microbiome aid in the intestinal epithelial barrier function to protect from disease. While chemically inducing colitis in mice from two distinct animal facilities, we serendipitously found that mice in one facility showed remarkable resistance to disease development, which was associated with increased markers of epithelial barrier integrity. Importantly, we show that Akkermansia muciniphila and Parabacteroides distasonis were significantly increased in the microbiota of resistant mice. To causally connect these microbes to protection against disease, we colonized susceptible mice with the two bacterial species. Our results demonstrate that A. muciniphila and P. distasonis synergistically drive a protective effect in both acute and chronic models of colitis by boosting the frequency of type 3 innate lymphoid cells in the colon and by improving gut epithelial integrity. Altogether, our work reveals a combined effort of commensal microbes in offering protection against severe intestinal inflammation by shaping gut immunity and by enhancing intestinal epithelial barrier stability. Our study highlights the beneficial role of gut bacteria in dictating intestinal homeostasis, which is an important step toward employing microbiome-driven therapeutic approaches for IBD clinical management. IMPORTANCE: The contribution of the gut microbiome to the balance between homeostasis and inflammation is widely known. Nevertheless, the etiology of inflammatory bowel disease, which is known to be influenced by genetics, immune response, and environmental cues, remains unclear. Unlocking novel players involved in the dictation of a protective gut, namely, in the microbiota component, is therefore crucial to develop novel strategies to tackle IBD. Herein, we revealed a synergistic interaction between two commensal bacterial strains, Akkermansia muciniphila and Parabacteroides distasonis, which induce protection against both acute and chronic models of colitis induction, by enhancing epithelial barrier integrity and promoting group 3 innate lymphoid cells in the colonic mucosa. This study provides a novel insight on how commensal bacteria can beneficially act to promote intestinal homeostasis, which may open new avenues toward the use of microbiome-derived strategies to tackle IBD.


Assuntos
Bacteroidetes , Colite , Doenças Inflamatórias Intestinais , Animais , Camundongos , Imunidade Inata , Linfócitos , Colite/microbiologia , Doenças Inflamatórias Intestinais/microbiologia , Inflamação , Verrucomicrobia/genética , Akkermansia
2.
Nat Rev Immunol ; 23(1): 9-23, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-35534624

RESUMO

The prevalence of autoimmune diseases (ADs) worldwide has rapidly increased over the past few decades. Thus, in addition to the classical risk factors for ADs, such as genetic polymorphisms, infections and smoking, environmental triggers have been considered. Recent sequencing-based approaches have revealed that patients with extra-intestinal ADs, such as multiple sclerosis, rheumatoid arthritis, type 1 diabetes and systemic lupus erythematosus, have distinct gut microbiota compositions compared to healthy controls. Faecal microbiota transplantation or inoculation with specific microbes in animal models of ADs support the hypothesis that alterations of gut microbiota influence autoimmune responses and disease outcome. Here, we describe the compositional and functional changes in the gut microbiota in patients with extra-intestinal AD and discuss how the gut microbiota affects immunity. Moreover, we examine how the gut microbiota might be modulated in patients with ADs as a potential preventive or therapeutic approach.


Assuntos
Doenças Autoimunes , Diabetes Mellitus Tipo 1 , Microbioma Gastrointestinal , Enteropatias , Lúpus Eritematoso Sistêmico , Animais , Humanos , Lúpus Eritematoso Sistêmico/etiologia , Lúpus Eritematoso Sistêmico/terapia , Fatores de Risco , Disbiose
3.
mSystems ; 6(6): e0071721, 2021 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-34726493

RESUMO

Food safety has considerably improved worldwide, yet infections with foodborne human enteric pathogens, such as Listeria spp. and Salmonella spp., still cause numerous hospitalizations and fatalities. Since dietary alterations, including fiber deficiency, might impact the colonization resistance mediated by the gut microbiome, studying the diet-microbiome-pathogen axis holds promise in further understanding the pathogenesis mechanisms. Using a gnotobiotic mouse model containing a 14-member synthetic human gut microbiota (14SM), we have previously shown that dietary fiber deprivation promotes proliferation of mucin-degrading bacteria, leading to a microbiome-mediated erosion of the colonic mucus barrier, which results in an increased susceptibility toward the rodent enteric pathogen Citrobacter rodentium. Here, we sought to understand how a low-fiber diet affects susceptibility to Listeria monocytogenes and Salmonella enterica serovar Typhimurium by using our 14SM gnotobiotic mouse model in BALB/c and C57BL/6 mouse backgrounds, respectively. Intriguingly, and in contrast to our results with C. rodentium, we observed that depriving mice of dietary fiber protected them from infections with both pathogens, compared to mice fed a standard chow. The microbiome delayed the overall pathogenicity compared to the onset of disease observed in germfree control mice. Nevertheless, we observed the same effect of diet on germfree mice, suggesting that the susceptibility is directly driven by the diet itself even in the absence of the gut microbiome. Our study points out an important observation, namely, that dietary fiber plays a crucial role in either the host's susceptibility, the virulence of these pathogens, or both. It would be judicious to design and interpret future studies on this basis. IMPORTANCE The human enteric pathogens Listeria monocytogenes and Salmonella Typhimurium are employed as classical models in rodent hosts to understand the pathogenesis mechanisms of foodborne pathogens. Research in the past decade has stressed the importance of the gut microbial composition in modulating susceptibility to these pathogens. The results of our study-using gnotobiotic mice and germfree control animals-additionally suggest that the dietary fiber components can dominate the impact of enteropathogenic virulence over the pathogenicity-modulating properties of the gut microbiome. The significance of our research is that there is a need to carefully choose a certain chow when performing the enteropathogen-associated mouse experiments and to cautiously match the rodent diets when trying to replicate experiments across different laboratories. Finally, our data underscore the importance of using germfree control animals to study these pathogens, as our findings would have been prone to misinterpretation in the absence of these controls.

4.
Gut Microbes ; 13(1): 1966263, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34530674

RESUMO

The change of dietary habits in Western societies, including reduced consumption of fiber, is linked to alterations in gut microbial ecology. Nevertheless, mechanistic connections between diet-induced microbiota changes that affect colonization resistance and enteric pathogen susceptibility are still emerging. We sought to investigate how a diet devoid of soluble plant fibers impacts the structure and function of a conventional gut microbiota in specific-pathogen-free (SPF) mice and how such changes alter susceptibility to a rodent enteric pathogen. We show that absence of dietary fiber intake leads to shifts in the abundances of specific taxa, microbiome-mediated erosion of the colonic mucus barrier, a reduction of intestinal barrier-promoting short-chain fatty acids, and increases in markers of mucosal barrier integrity disruption. Importantly, our results highlight that these low-fiber diet-induced changes in the gut microbial ecology collectively contribute to a lethal colitis by the mucosal pathogen Citrobacter rodentium, which is used as a mouse model for enteropathogenic and enterohemorrhagic Escherichia coli (EPEC and EHEC, respectively). Our study indicates that modern, low-fiber Western-style diets might make individuals more prone to infection by enteric pathogens via the disruption of mucosal barrier integrity by diet-driven changes in the gut microbiota, illustrating possible implications for EPEC and EHEC infections.


Assuntos
Citrobacter rodentium/crescimento & desenvolvimento , Colite/microbiologia , Dieta Ocidental/efeitos adversos , Fibras na Dieta/análise , Mucosa Intestinal/microbiologia , Junções Íntimas/fisiologia , Animais , Bactérias/classificação , Bactérias/crescimento & desenvolvimento , Bactérias/isolamento & purificação , Disbiose/microbiologia , Infecções por Enterobacteriaceae/microbiologia , Infecções por Enterobacteriaceae/patologia , Ácidos Graxos Voláteis/metabolismo , Comportamento Alimentar/fisiologia , Feminino , Mucosa Intestinal/patologia , Camundongos , Camundongos Endogâmicos C57BL , Organismos Livres de Patógenos Específicos
5.
Nat Rev Gastroenterol Hepatol ; 18(12): 885-902, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34580480

RESUMO

Autoimmune diseases, including inflammatory bowel disease, multiple sclerosis and rheumatoid arthritis, have distinct clinical presentations but share underlying patterns of gut microbiome perturbation and intestinal barrier dysfunction. Their potentially common microbial drivers advocate for treatment strategies aimed at restoring appropriate microbiome function, but individual variation in host factors makes a uniform approach unlikely. In this Perspective, we consolidate knowledge on diet-microbiome interactions in local inflammation, gut microbiota imbalance and host immune dysregulation. By understanding and incorporating the effects of individual dietary components on microbial metabolic output and host physiology, we examine the potential for diet-based therapies for autoimmune disease prevention and treatment. We also discuss tools targeting the gut microbiome, such as faecal microbiota transplantation, probiotics and orthogonal niche engineering, which could be optimized using custom dietary interventions. These approaches highlight paths towards leveraging diet for precise engineering of the gut microbiome at a time of increasing autoimmune disease.


Assuntos
Doenças Autoimunes/microbiologia , Doenças Autoimunes/terapia , Dieta/métodos , Microbioma Gastrointestinal/imunologia , Microbioma Gastrointestinal/fisiologia , Doenças Autoimunes/imunologia , Doenças Autoimunes/fisiopatologia , Terapia Combinada , Transplante de Microbiota Fecal , Humanos , Prebióticos , Prevenção Primária/métodos , Probióticos/uso terapêutico
6.
Int J Mol Sci ; 22(13)2021 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-34202227

RESUMO

The consumption of prebiotic fibers to modulate the human gut microbiome is a promising strategy to positively impact health. Nevertheless, given the compositional complexity of the microbiome and its inter-individual variances, generalized recommendations on the source or amount of fiber supplements remain vague. This problem is further compounded by availability of tractable in vitro and in vivo models to validate certain fibers. We employed a gnotobiotic mouse model containing a 14-member synthetic human gut microbiome (SM) in vivo, characterized a priori for their ability to metabolize a collection of fibers in vitro. This SM contains 14 different strains belonging to five distinct phyla. Since soluble purified fibers have been a common subject of studies, we specifically investigated the effects of dietary concentrated raw fibers (CRFs)-containing fibers from pea, oat, psyllium, wheat and apple-on the compositional and functional alterations in the SM. We demonstrate that, compared to a fiber-free diet, CRF supplementation increased the abundance of fiber-degraders, namely Eubacterium rectale, Roseburia intestinalis and Bacteroides ovatus and decreased the abundance of the mucin-degrader Akkermansia muciniphila. These results were corroborated by a general increase of bacterial fiber-degrading α-glucosidase enzyme activity. Overall, our results highlight the ability of CRFs to enhance the microbial fiber-degrading capacity.


Assuntos
Fibras na Dieta/metabolismo , Microbioma Gastrointestinal , Prebióticos , Animais , Bactérias , Dieta , Suplementos Nutricionais , Ácidos Graxos Voláteis/metabolismo , Fezes/microbiologia , Humanos , Camundongos , Polissacarídeos/metabolismo
7.
STAR Protoc ; 2(2): 100607, 2021 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-34179836

RESUMO

Reproducible in vivo models are necessary to address functional aspects of the gut microbiome in various diseases. Here, we present a gnotobiotic mouse model that allows for the investigation of specific microbial functions within the microbiome. We describe how to culture 14 different well-characterized human gut species and how to verify their proper colonization in germ-free mice. This protocol can be modified to add or remove certain species of interest to investigate microbial mechanistic details in various disease models. For complete details on the use and execution of this protocol, please refer to Desai et al. (2016).


Assuntos
Microbioma Gastrointestinal , Interações Hospedeiro-Patógeno , Animais , Bactérias/classificação , Bactérias/crescimento & desenvolvimento , Bactérias/isolamento & purificação , Vida Livre de Germes , Humanos , Camundongos , Filogenia
8.
STAR Protoc ; 2(1): 100326, 2021 03 19.
Artigo em Inglês | MEDLINE | ID: mdl-33665621

RESUMO

The gut microbiome expresses a multitude of enzymes degrading polysaccharides in dietary plant fibers and in host-secreted mucus. The quantitative detection of these glycan-degrading enzymes in fecal samples is important to elucidate the functional activity of the microbiome in health and disease. We describe a protocol for detection of glycan-degrading enzyme activity in mouse and human fecal samples, namely sulfatase and four carbohydrate-active enzymes. Assessing their activity can inform treatment strategies for diseases linked to the gut microbiome. For complete details on the use and execution of this protocol, please refer to Desai et al. (2016).


Assuntos
Bactérias/enzimologia , Proteínas de Bactérias/metabolismo , Fezes , Glicosídeo Hidrolases/metabolismo , Microbiota , Animais , Fezes/enzimologia , Fezes/microbiologia , Humanos , Camundongos
9.
Front Immunol ; 11: 612336, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33542719

RESUMO

Intestinal commensal bacteria can have a large impact on the state of health and disease of the host. Regulation of Th17 cell development by gut commensals is known to contribute to their dichotomous role in promoting gut homeostasis and host defense, or development of autoimmune diseases. Yet, the underlying mechanisms remain to be fully elucidated. One candidate factor contributing to Th17 differentiation, and the expression of which could be influenced by commensals is the atypical nuclear IκB protein IκBζ. IκBζ acts as a transcriptional regulator of the expression of Th17-related secondary response genes in many cell types including dendritic cells (DCs). Insights into the regulation of IκBζ in DCs could shed light on how these immune sentinel cells at the interface between commensals, innate and adaptive immune system drive an immune-tolerogenic or inflammatory Th17 cell response. In this study, the influence of two gut commensals of low (Bacteroides vulgatus) or high (Escherichia coli) immunogenicity on IκBζ expression in DCs and its downstream effects was analyzed. We observed that the amount of IκBζ expression and secretion of Th17-inducing cytokines correlated with the immunogenicity of these commensals. However, under immune-balanced conditions, E. coli also strongly induced an IκBζ-dependent secretion of anti-inflammatory IL-10, facilitating a counter-regulative Treg response as assessed in in vitro CD4+ T cell polarization assays. Yet, in an in vivo mouse model of T cell-induced colitis, prone to inflammatory and autoimmune conditions, administration of E. coli promoted an expansion of rather pro-inflammatory T helper cell subsets whereas administration of B. vulgatus resulted in the induction of protective T helper cell subsets. These findings might contribute to the development of new therapeutic strategies for the treatment of autoimmune diseases using commensals or commensal-derived components.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/imunologia , Células Dendríticas/imunologia , Microbioma Gastrointestinal/imunologia , Células Th17/imunologia , Animais , Doenças Autoimunes/imunologia , Bacteroides/imunologia , Linfócitos T CD4-Positivos/imunologia , Diferenciação Celular/imunologia , Células Cultivadas , Colite/imunologia , Citocinas/imunologia , Escherichia coli/imunologia , Feminino , Inflamação/imunologia , Interleucina-10/imunologia , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos C57BL
10.
Mol Ther ; 27(11): 1974-1991, 2019 11 06.
Artigo em Inglês | MEDLINE | ID: mdl-31416777

RESUMO

Generated by gram-negative bacteria, lipopolysaccharides (LPSs) are one of the most abundant and potent immunomodulatory substances present in the intestinal lumen. Interaction of agonistic LPS with the host myeloid-differentiation-2/Toll-like receptor 4 (MD-2/TLR4) receptor complex results in nuclear factor κB (NF-κB) activation, followed by the robust induction of pro-inflammatory immune responses. Here we have isolated LPS from a common gut commensal, Bacteroides vulgatus mpk (BVMPK), which provides only weak agonistic activity. This weak agonistic activity leads to the amelioration of inflammatory immune responses in a mouse model for experimental colitis, and it was in sharp contrast to strong agonists and antagonists. In this context, the administration of BVMPK LPS into mice with severe intestinal inflammation re-established intestinal immune homeostasis within only 2 weeks, resulting in the clearance of all symptoms of inflammation. These inflammation-reducing properties of weak agonistic LPS are grounded in the induction of a special type of endotoxin tolerance via the MD-2/TLR4 receptor complex axis in intestinal lamina propria CD11c+ cells. Thus, weak agonistic LPS represents a promising agent to treat diseases involving pathological overactivation of the intestinal immune system, e.g., in inflammatory bowel diseases.


Assuntos
Homeostase/imunologia , Imunidade nas Mucosas , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , Lipopolissacarídeos/imunologia , Animais , Biomarcadores , Antígeno CD11c/metabolismo , Colite/etiologia , Colite/metabolismo , Colite/patologia , Modelos Animais de Doenças , Microbioma Gastrointestinal/imunologia , Homeostase/efeitos dos fármacos , Humanos , Doenças Inflamatórias Intestinais/diagnóstico por imagem , Doenças Inflamatórias Intestinais/etiologia , Doenças Inflamatórias Intestinais/metabolismo , Doenças Inflamatórias Intestinais/patologia , Mucosa Intestinal/efeitos dos fármacos , Lipídeo A/imunologia , Lipopolissacarídeos/farmacologia , Camundongos , Camundongos Knockout , Tomografia por Emissão de Pósitrons
11.
PLoS Biol ; 17(6): e3000334, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31206517

RESUMO

Escherichia coli represents a classical intestinal gram-negative commensal. Despite this commensalism, different E. coli strains can mediate disparate immunogenic properties in a given host. Symbiotic E. coli strains such as E. coli Nissle 1917 (EcN) are attributed beneficial properties, e.g., promotion of intestinal homeostasis. Therefore, we aimed to identify molecular features derived from symbiotic bacteria that might help to develop innovative therapeutic alternatives for the treatment of intestinal immune disorders. This study was performed using the dextran sodium sulphate (DSS)-induced colitis mouse model, which is routinely used to evaluate potential therapeutics for the treatment of Inflammatory Bowel Diseases (IBDs). We focused on the analysis of flagellin structures of different E. coli strains. EcN flagellin was found to harbor a substantially longer hypervariable region (HVR) compared to other commensal E. coli strains, and this longer HVR mediated symbiotic properties through stronger activation of Toll-like receptor (TLR)5, thereby resulting in interleukin (IL)-22-mediated protection of mice against DSS-induced colitis. Furthermore, using bone-marrow-chimeric mice (BMCM), CD11c+ cells of the colonic lamina propria (LP) were identified as the main mediators of these flagellin-induced symbiotic effects. We propose flagellin from symbiotic E. coli strains as a potential therapeutic to restore intestinal immune homeostasis, e.g., for the treatment of IBD patients.


Assuntos
Escherichia coli/metabolismo , Flagelina/genética , Simbiose/genética , Animais , Colite/induzido quimicamente , Colite/imunologia , Modelos Animais de Doenças , Escherichia coli/genética , Infecções por Escherichia coli/microbiologia , Proteínas de Escherichia coli/genética , Feminino , Flagelina/metabolismo , Mucosa Intestinal , Intestinos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais/imunologia , Simbiose/fisiologia , Receptor 5 Toll-Like/metabolismo
12.
Gut Microbes ; 9(1): 1-12, 2018 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-28686482

RESUMO

The Gram negative intestinal symbiont Bacteroides vulgatus mpk is able to prevent from induction of colonic inflammation in Rag1-/- mice and promotes immune balance in Il2-/- mice. These inflammation-silencing effects are associated with B. vulgatus mpk-mediated induction of semi-mature dendritic cells, especially in the colonic lamina propria (cLP). However the beneficial interaction of bacteria with host immune cells is limited due to the existence of a large mucus layer covering the intestinal epithelium. How can intestinal bacteria overcome this physical barrier and contact the host immune system? One mechanism is the production of outer membrane vesicles (OMVs) via ubiquitous blebbing of the outer membrane. These proteoliposomes have the ability to traverse the mucus layer. Hence, OMVs play an important role in immunomodulation and the maintenance of a balanced gut microbiota. Here we demonstrate that the stimulation of bone marrow derived dendritic cells (BMDCs) with isolated OMVs originated from B. vulgatus mpk leads to the induction of a tolerant semi-mature phenotype. Thereby, microbe- associated molecular patterns (MAMPs) delivered by OMVs are crucial for the interaction and the resulting maturation of immune cells. Additional to the binding to host TLR4, a yet unknown ligand to TLR2 is indispensable for the conversion of immature BMDCs into a semi-mature state. Thus, crossing the epithelial mucus layer and directly contact host cells, OMV mediate cross-tolerance via the transport of various Toll-like receptor antigens. These features make OMVs to a key attribute of B. vulgatus mpk for a vigorous acellular prevention and treatment of systemic diseases.


Assuntos
Bacteroides/imunologia , Membrana Celular/ultraestrutura , Células Dendríticas/imunologia , Células Dendríticas/microbiologia , Exossomos/metabolismo , Tolerância Imunológica , Animais , Bacteroides/metabolismo , Bacteroides/ultraestrutura , Membrana Celular/metabolismo , Células Cultivadas , Escherichia coli/imunologia , Exossomos/imunologia , Células HEK293 , Humanos , Fatores Imunológicos/metabolismo , Interleucina-6/análise , Camundongos , Mutação , Transdução de Sinais/imunologia , Receptor 2 Toll-Like/genética , Receptor 2 Toll-Like/metabolismo , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/metabolismo , Fator de Necrose Tumoral alfa/análise
14.
J Autoimmun ; 75: 82-95, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27484364

RESUMO

Cathepsin S (CTSS) is a lysosomal protease whose activity regulation is important for MHC-II signaling and subsequent activation of CD4+ T cell mediated immune responses. Dysregulation of its enzymatic activity or enhanced secretion into extracellular environments is associated with the induction or progression of several autoimmune diseases. Here we demonstrate that commensal intestinal bacteria influence secretion rates and intracellular activity of host CTSS and that symbiotic bacteria, i.e. Bacteroides vulgatus mpk, may actively regulate this process and help to maintain physiological levels of CTSS activities in order to prevent from induction of pathological inflammation. The symbiont-controlled regulation of CTSS activity is mediated by anticipating reactive oxygen species induction in dendritic cells which, in turn, maintains cystatin C (CysC) monomer binding to CTSS. CysC monomers are potent endogenous CTSS inhibitors. This Bacteroides vulgatus caused and CysC dependent CTSS activity regulation is involved in the generation of tolerant intestinal dendritic cells contributing to prevention of T-cell mediated induction of colonic inflammation. Taken together, we demonstrate that symbionts of the intestinal microbiota regulate host CTSS activity and secretion and might therefore be an attractive approach to deal with CTSS associated autoimmune diseases.


Assuntos
Bactérias/imunologia , Catepsinas/imunologia , Microbioma Gastrointestinal/imunologia , Simbiose/imunologia , Animais , Bacteroides/imunologia , Bacteroides/fisiologia , Infecções por Bacteroides/imunologia , Infecções por Bacteroides/microbiologia , Benzopiranos/farmacologia , Western Blotting , Células da Medula Óssea/imunologia , Células da Medula Óssea/metabolismo , Células da Medula Óssea/microbiologia , Carbamatos/farmacologia , Catepsinas/antagonistas & inibidores , Catepsinas/genética , Células Cultivadas , Colite/imunologia , Colite/metabolismo , Citocinas/imunologia , Citocinas/metabolismo , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Células Dendríticas/microbiologia , Microbioma Gastrointestinal/fisiologia , Expressão Gênica/imunologia , Interações Hospedeiro-Patógeno/imunologia , Tolerância Imunológica/imunologia , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Espécies Reativas de Oxigênio/imunologia , Espécies Reativas de Oxigênio/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
15.
Genome Biol Evol ; 8(4): 1197-207, 2016 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-27071651

RESUMO

Like many other Bacteroides species, Bacteroides vulgatus strain mpk, a mouse fecal isolate which was shown to promote intestinal homeostasis, utilizes a variety of mobile elements for genome evolution. Based on sequences collected by Pacific Biosciences SMRT sequencing technology, we discuss the challenges of assembling and studying a bacterial genome of high plasticity. Additionally, we conducted comparative genomics comparing this commensal strain with the B. vulgatus type strain ATCC 8482 as well as multiple other Bacteroides and Parabacteroides strains to reveal the most important differences and identify the unique features of B. vulgatus mpk. The genome of B. vulgatus mpk harbors a large and diverse set of mobile element proteins compared with other sequenced Bacteroides strains. We found evidence of a number of different horizontal gene transfer events and a genome landscape that has been extensively altered by different mobilization events. A CRISPR/Cas system could be identified that provides a possible mechanism for preventing the integration of invading external DNA. We propose that the high genome plasticity and the introduced genome instabilities of B. vulgatus mpk arising from the various mobilization events might play an important role not only in its adaptation to the challenging intestinal environment in general, but also in its ability to interact with the gut microbiota.


Assuntos
Bacteroides/genética , Evolução Molecular , Transferência Genética Horizontal , Genoma Bacteriano , Camundongos/microbiologia , Animais , Sequência de Bases , Sistemas CRISPR-Cas , DNA Bacteriano/genética , Intestinos/microbiologia , Filogenia
16.
Int J Med Microbiol ; 306(5): 290-301, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27009633

RESUMO

Lipopolysaccharides (LPS) of Gram negative bacteria are one of the most potent stimulators of the host innate immune system and LPS recognition is essential for the host organism to clear infections of invading bacterial pathogens. Here we review on the latest research on how LPS is sensed by host cells and how distinct LPS structures differentially modulate the strength of the host immune response. Much is known about host immunological reactions towards pathogens via recognition of their LPS, as well as strategies of pathogens to modulate their LPS structure in order to evade the immune system. However, less is known about differential sensing of lipopolysaccharides of commensal bacteria in the intestine and how this contributes to manifestation or destruction of the intestinal homeostasis. LPS sensing is necessary to fight pathogens. However, sensing of LPS of gut commensal bacteria can simultaneously be disadvantageous for the genetically predisposed host, since this might lead to damage of the intestinal homeostasis and therefore to chronic intestinal inflammation. However, less immunogenic LPS could also serve as therapeutics to antagonize an overreacting innate immune system. Therefore, commensal gut bacteria-derived LPS could prevent from uncontrolled intestinal immune response in the intestine which makes LPS an attractive therapeutical approach to treat e.g. IBD.


Assuntos
Interações Hospedeiro-Patógeno , Lipídeo A/imunologia , Lipídeo A/metabolismo , Microbiota/imunologia , Simbiose , Animais , Humanos
17.
J Immunol Res ; 2016: 1958650, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26981546

RESUMO

How does the host manage to tolerate its own intestinal microbiota? A simple question leading to complicated answers. In order to maintain balanced immune responses in the intestine, the host immune system must tolerate commensal bacteria in the gut while it has to simultaneously keep the ability to fight pathogens and to clear infections. If this tender equilibrium is disturbed, severe chronic inflammatory reactions can result. Tolerogenic intestinal dendritic cells fulfil a crucial role in balancing immune responses and therefore creating homeostatic conditions and preventing from uncontrolled inflammation. Although several dendritic cell subsets have already been characterized to play a pivotal role in this process, less is known about definite molecular mechanisms of how intestinal dendritic cells are converted into tolerogenic ones. Here we review how gut commensal bacteria interact with intestinal dendritic cells and why this bacteria-host cell interaction is crucial for induction of dendritic cell tolerance in the intestine. Hereby, different commensal bacteria can have distinct effects on the phenotype of intestinal dendritic cells and these effects are mainly mediated by impacting toll-like receptor signalling in dendritic cells.


Assuntos
Células Dendríticas/imunologia , Microbioma Gastrointestinal/fisiologia , Tolerância Imunológica , Mucosa Intestinal/imunologia , Intestinos/microbiologia , Animais , Microbioma Gastrointestinal/imunologia , Homeostase , Inflamação , Doenças Inflamatórias Intestinais/imunologia , Doenças Inflamatórias Intestinais/terapia , Mucosa Intestinal/citologia , Intestinos/imunologia , Camundongos , Fenótipo , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Simbiose , Receptores Toll-Like/genética , Receptores Toll-Like/imunologia
18.
J Immunol Methods ; 432: 87-94, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26899824

RESUMO

Cathepsin S (CTSS) is a eukaryotic protease mostly expressed in professional antigen presenting cells (APCs). Since CTSS activity regulation plays a role in the pathogenesis of various autoimmune diseases like multiple sclerosis, atherosclerosis, Sjögren's syndrome and psoriasis as well as in cancer progression, there is an ongoing interest in the reliable detection of cathepsin S activity. Various applications have been invented for specific detection of this enzyme. However, most of them have only been shown to be suitable for human samples, do not deliver quantitative results or the experimental procedure requires technical equipment that is not commonly available in a standard laboratory. We have tested a fluorogen substrate, Mca-GRWPPMGLPWE-Lys(Dnp)-DArg-NH2, that has been described to specifically detect CTSS activities in human APCs for its potential use for mouse samples. We have modified the protocol and thereby offer a cheap, easy, reproducible and quick activity assay to detect CTSS activities in mouse APCs. Since most of basic research on CTSS is performed in mice, this method closes a gap and offers a possibility for reliable and quantitative CTSS activity detection that can be performed in almost every laboratory.


Assuntos
Células Apresentadoras de Antígenos/enzimologia , Catepsinas/metabolismo , Infecções por Escherichia coli/enzimologia , Corantes Fluorescentes/metabolismo , Peptídeos/metabolismo , Espectrometria de Fluorescência , Animais , Células Apresentadoras de Antígenos/efeitos dos fármacos , Células Apresentadoras de Antígenos/imunologia , Células Apresentadoras de Antígenos/microbiologia , Catepsinas/antagonistas & inibidores , Catepsinas/deficiência , Catepsinas/genética , Células Cultivadas , Inibidores de Cisteína Proteinase/farmacologia , Infecções por Escherichia coli/imunologia , Infecções por Escherichia coli/microbiologia , Hidrólise , Leucina/análogos & derivados , Leucina/farmacologia , Camundongos Knockout , Reprodutibilidade dos Testes , Especificidade por Substrato , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...