Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Nature ; 575(7781): 224-228, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31666699

RESUMO

The human gastrointestinal tract consists of a dense and diverse microbial community, the composition of which is intimately linked to health. Extrinsic factors such as diet and host immunity are insufficient to explain the constituents of this community, and direct interactions between co-resident microorganisms have been implicated as important drivers of microbiome composition. The genomes of bacteria derived from the gut microbiome contain several pathways that mediate contact-dependent interbacterial antagonism1-3. Many members of the Gram-negative order Bacteroidales encode the type VI secretion system (T6SS), which facilitates the delivery of toxic effector proteins into adjacent cells4,5. Here we report the occurrence of acquired interbacterial defence (AID) gene clusters in Bacteroidales species that reside within the human gut microbiome. These clusters encode arrays of immunity genes that protect against T6SS-mediated intra- and inter-species bacterial antagonism. Moreover, the clusters reside on mobile elements, and we show that their transfer is sufficient to confer resistance to toxins in vitro and in gnotobiotic mice. Finally, we identify and validate the protective capability of a recombinase-associated AID subtype (rAID-1) that is present broadly in Bacteroidales genomes. These rAID-1 gene clusters have a structure suggestive of active gene acquisition and include predicted immunity factors of toxins derived from diverse organisms. Our data suggest that neutralization of contact-dependent interbacterial antagonism by AID systems helps to shape human gut microbiome ecology.


Assuntos
Bacteroidetes , Microbioma Gastrointestinal , Trato Gastrointestinal/microbiologia , Interações Microbianas , Sistemas de Secreção Tipo VI/antagonistas & inibidores , Animais , Bacteroidetes/genética , Bacteroidetes/imunologia , Feminino , Microbioma Gastrointestinal/imunologia , Trato Gastrointestinal/imunologia , Genes Bacterianos/genética , Humanos , Camundongos , Interações Microbianas/genética , Interações Microbianas/imunologia , Família Multigênica/genética , Sistemas de Secreção Tipo VI/genética , Sistemas de Secreção Tipo VI/imunologia
2.
BMC Microbiol ; 24(1): 237, 2024 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-38961326

RESUMO

OBJECTIVE: Bladder cancer(BCa) was a disease that seriously affects patients' quality of life and prognosis. To address this issue, many researches suggested that the gut microbiota modulated tumor response to treatment; however, this had not been well-characterized in bladder cancer. In this study, our objective was to determine whether the diversity and composition of the gut microbiota or the density of specific bacterial genera influence the prognosis of patients with bladder cancer. METHODS: We collected fecal samples from a total of 50 bladder cancer patients and 22 matched non-cancer individuals for 16S rDNA sequencing to investigate the distribution of Parabacteroides in these two groups. Further we conducted follow-up with cancer patients to access the impact of different genera of microorganisms on patients survival. We conducted a Fecal Microbiota Transplantation (FMT) and mono-colonization experiment with Parabacteroides distasonis to explore its potential enhancement of the efficacy of anti-PD-1 immunotherapy in MB49 tumor-bearing mice. Immunohistochemistry, transcriptomics and molecular experiment analyses were employed to uncover the underlying mechanisms. RESULTS: The 16S rDNA showed that abundance of the genus Parabacteroides was elevated in the non-cancer control group compared to bladder cancer group. The results of tumor growth curves showed that a combination therapy of P. distasonis and ICIs treatment significantly delayed tumor growth and increased the intratumoral densities of both CD4+T and CD8+T cells. The results of transcriptome analysis demonstrated that the pathways associated with antitumoral immune response were remarkably upregulated in the P. distasonis gavage group. CONCLUSION: P. distasonis delivery combined with α-PD-1 mAb could be a new strategy to enhance the effect of anti-PD-1 immunotherapy. This effect might be achieved by activating immune and antitumor related pathways.


Assuntos
Bacteroidetes , Transplante de Microbiota Fecal , Microbioma Gastrointestinal , Imunoterapia , Neoplasias da Bexiga Urinária , Neoplasias da Bexiga Urinária/terapia , Neoplasias da Bexiga Urinária/imunologia , Neoplasias da Bexiga Urinária/microbiologia , Animais , Humanos , Camundongos , Imunoterapia/métodos , Bacteroidetes/genética , Bacteroidetes/imunologia , Feminino , Masculino , RNA Ribossômico 16S/genética , Fezes/microbiologia , Pessoa de Meia-Idade , Idoso , Camundongos Endogâmicos C57BL
3.
Gastroenterology ; 162(1): 166-178, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34606847

RESUMO

BACKGROUND & AIMS: Fecal microbiota transplantation (FMT) is an emerging treatment modality for ulcerative colitis (UC). Several randomized controlled trials have shown efficacy for FMT in the treatment of UC, but a better understanding of the transferable microbiota and their immune impact is needed to develop more efficient microbiome-based therapies for UC. METHODS: Metagenomic analysis and strain tracking was performed on 60 donor and recipient samples receiving FMT for active UC. Sorting and sequencing of immunoglobulin (Ig) A-coated microbiota (called IgA-seq) was used to define immune-reactive microbiota. Colonization of germ-free or genetically engineered mice with patient-derived strains was performed to determine the mechanism of microbial impact on intestinal immunity. RESULTS: Metagenomic analysis defined a core set of donor-derived transferable bacterial strains in UC subjects achieving clinical response, which predicted response in an independent trial of FMT for UC. IgA-seq of FMT recipient samples and gnotobiotic mice colonized with donor microbiota identified Odoribacter splanchnicus as a transferable strain shaping mucosal immunity, which correlated with clinical response and the induction of mucosal regulatory T cells. Colonization of mice with O splanchnicus led to an increase in Foxp3+/RORγt+ regulatory T cells, induction of interleukin (IL) 10, and production of short chain fatty acids, all of which were required for O splanchnicus to limit colitis in mouse models. CONCLUSIONS: This work provides the first evidence of transferable, donor-derived strains that correlate with clinical response to FMT in UC and reveals O splanchnicus as a key component promoting both metabolic and immune cell protection from colitis. These mechanistic features will help enable strategies to enhance the efficacy of microbial therapy for UC. Clinicaltrials.gov ID NCT02516384.


Assuntos
Bacteroidetes/imunologia , Colite/terapia , Colo/microbiologia , Transplante de Microbiota Fecal , Microbioma Gastrointestinal , Imunoglobulina A/imunologia , Mucosa Intestinal/microbiologia , Animais , Bacteroidetes/genética , Bacteroidetes/metabolismo , Ensaios Clínicos como Assunto , Colite/imunologia , Colite/metabolismo , Colite/microbiologia , Colite Ulcerativa/diagnóstico , Colite Ulcerativa/imunologia , Colite Ulcerativa/metabolismo , Colite Ulcerativa/microbiologia , Colo/imunologia , Colo/metabolismo , Modelos Animais de Doenças , Fatores de Transcrição Forkhead/metabolismo , Microbioma Gastrointestinal/genética , Microbioma Gastrointestinal/imunologia , Vida Livre de Germes , Humanos , Imunidade nas Mucosas , Imunoglobulina A/genética , Imunoglobulina A/metabolismo , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , Linfócitos Intraepiteliais/imunologia , Linfócitos Intraepiteliais/metabolismo , Linfócitos Intraepiteliais/microbiologia , Metagenoma , Metagenômica , Camundongos Endogâmicos C57BL , Camundongos Knockout , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/metabolismo , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Linfócitos T Reguladores/microbiologia , Resultado do Tratamento
4.
Int J Med Sci ; 18(2): 511-519, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33390820

RESUMO

The association between body composition and gut microbiota in type 2 diabetes mellitus (DM) remains unknown. To elucidate the correlation of body composition and gut microbiota, we conducted a clinical study to enroll 179 patients with type 2 DM. Body composition of lean tissue index (LTI) and fat tissue index was measured by Body Composition Monitor. Eight pairs of 16S rRNA gene primers specific to Firmicutes, Bacteroidetes, the Clostridium leptum group, Bacteroides, Bifidobacterium, Akkermansia muciniphila, Escherichia coli, and Faecalibacterium prausnitzii were used to measure their abundance by quantitative polymerase chain reaction. The results showed that type 2 DM with higher abundance of phylum Firmicutes and a higher ratio of phyla Firmicutes to Bacteroidetes (phyla F/B ratio) had higher LTI. This significant correlation between phyla F/B ratio and LTI was especially evident in type 2 DM with high body mass index, and independent of glycemic control or dipeptidyl peptidase-4 inhibitor usage. In conclusion, our study demonstrated the positive association of LTI with the abundance of phylum Firmicutes and the phyla F/B ratio in type 2 DM.


Assuntos
Composição Corporal/imunologia , Diabetes Mellitus Tipo 2/imunologia , Disbiose/complicações , Microbioma Gastrointestinal/imunologia , Idoso , Bacteroidetes/genética , Bacteroidetes/imunologia , Bacteroidetes/isolamento & purificação , DNA Bacteriano/isolamento & purificação , Diabetes Mellitus Tipo 2/microbiologia , Disbiose/diagnóstico , Disbiose/imunologia , Disbiose/microbiologia , Feminino , Firmicutes/genética , Firmicutes/imunologia , Firmicutes/isolamento & purificação , Microbioma Gastrointestinal/genética , Humanos , Masculino , Pessoa de Meia-Idade , Prevalência , RNA Ribossômico 16S/genética , Fatores de Risco
5.
Infect Immun ; 87(10)2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31308086

RESUMO

As important players in the host defense system, commensal microbes and the microbiota influence multiple aspects of host physiology. Bordetella pertussis infection is highly contagious among humans. However, the roles of the microbiota in B. pertussis pathogenesis are poorly understood. Here, we show that antibiotic-mediated depletion of the microbiota results in increased susceptibility to B. pertussis infection during the early stage. The increased susceptibility was associated with a marked impairment of the systemic IgG, IgG2a, and IgG1 antibody responses to B. pertussis infection after antibiotic treatment. Furthermore, the microbiota impacted the short-lived plasma cell responses as well as the recall responses of memory B cells to B. pertussis infection. Finally, we found that the dysbiosis caused by antibiotic treatment affects CD4+ T cell generation and PD-1 expression on CD4+ T cells and thereby perturbs plasma cell differentiation. Our results have revealed the importance of commensal microbes in modulating host immune responses to B. pertussis infection and support the possibility of controlling the severity of B. pertussis infection in humans by manipulating the microbiota.


Assuntos
Bordetella pertussis/imunologia , Disbiose/imunologia , Microbioma Gastrointestinal/imunologia , Imunidade Humoral , Simbiose/imunologia , Coqueluche/imunologia , Ampicilina/farmacologia , Animais , Antibacterianos/farmacologia , Anticorpos Antibacterianos/biossíntese , Anticorpos Antibacterianos/classificação , Bacteroidetes/classificação , Bacteroidetes/efeitos dos fármacos , Bacteroidetes/crescimento & desenvolvimento , Bacteroidetes/imunologia , Bordetella pertussis/crescimento & desenvolvimento , Bordetella pertussis/patogenicidade , Disbiose/microbiologia , Disbiose/fisiopatologia , Feminino , Firmicutes/classificação , Firmicutes/efeitos dos fármacos , Firmicutes/crescimento & desenvolvimento , Firmicutes/imunologia , Microbioma Gastrointestinal/efeitos dos fármacos , Imunidade Inata , Imunoglobulina G/biossíntese , Imunoglobulina G/classificação , Metronidazol/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Neomicina/farmacologia , Proteobactérias/classificação , Proteobactérias/efeitos dos fármacos , Proteobactérias/crescimento & desenvolvimento , Proteobactérias/imunologia , Simbiose/efeitos dos fármacos , Vancomicina/farmacologia , Coqueluche/microbiologia , Coqueluche/fisiopatologia
6.
Clin Exp Allergy ; 49(2): 227-238, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30312497

RESUMO

BACKGROUND: House dust mite (HDM) is the major source of indoor allergens that cause airway disease. Recent evidence suggests that Gram-negative/positive bacteria produce nano-sized extracellular vesicles (EVs) containing diverse components, including various immunostimulatory molecules. However, the association between bacteria-derived EVs and development of airway disease is unclear. OBJECTIVE: To identify and isolate HDM-derived EVs and to evaluate their effect on the development of airway inflammation. METHODS: Extracellular vesicles were isolated from crude HDM extracts by ultra-centrifugation, and their physical and immunological characteristics and roles in airway inflammation were tested in vitro and in murine models of airway inflammation. In addition, 16s metagenome analysis of nucleic acid from EVs was performed to identify their origin. RESULTS: Round, bilayered vesicles measuring 80-100 nanometres and containing abundant amounts of LPS were isolated. These vesicles induced innate immune responses both in vitro and in vivo. Intranasal exposure of naïve mice to HDM EVs induced production of cytokines associated with development of Th2-mediated and mixed (Th1-/Th2-/Th17-mediated) airway inflammation to allergen. Metagenome analysis identified Bacteroidetes and Proteobacteria as the probable sources of HDM EVs. CONCLUSION: House dust mite EVs originating from Gram-negative bacteria may play an important role on the development of airway inflammation.


Assuntos
Asma , Bacteroidetes , Vesículas Extracelulares , Proteobactérias , Pyroglyphidae , Linfócitos T Auxiliares-Indutores , Animais , Asma/metabolismo , Asma/microbiologia , Asma/patologia , Bacteroidetes/genética , Bacteroidetes/imunologia , Modelos Animais de Doenças , Vesículas Extracelulares/química , Vesículas Extracelulares/imunologia , Vesículas Extracelulares/microbiologia , Metagenoma , Camundongos , Camundongos Knockout , Proteobactérias/genética , Proteobactérias/imunologia , Pyroglyphidae/química , Pyroglyphidae/imunologia , Pyroglyphidae/microbiologia , Linfócitos T Auxiliares-Indutores/imunologia , Linfócitos T Auxiliares-Indutores/patologia
7.
Proc Natl Acad Sci U S A ; 113(13): 3627-32, 2016 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-26951680

RESUMO

Type VI secretion systems (T6SSs) are multiprotein complexes best studied in Gram-negative pathogens where they have been shown to inhibit or kill prokaryotic or eukaryotic cells and are often important for virulence. We recently showed that T6SS loci are also widespread in symbiotic human gut bacteria of the order Bacteroidales, and that these T6SS loci segregate into three distinct genetic architectures (GA). GA1 and GA2 loci are present on conserved integrative conjugative elements (ICE) and are transferred and shared among diverse human gut Bacteroidales species. GA3 loci are not contained on conserved ICE and are confined to Bacteroides fragilis Unlike GA1 and GA2 T6SS loci, most GA3 loci do not encode identifiable effector and immunity proteins. Here, we studied GA3 T6SSs and show that they antagonize most human gut Bacteroidales strains analyzed, except for B. fragilis strains with the same T6SS locus. A combination of mutation analyses,trans-protection analyses, and in vitro competition assays, allowed us to identify novel effector and immunity proteins of GA3 loci. These proteins are not orthologous to known proteins, do not contain identified motifs, and most have numerous predicted transmembrane domains. Because the genes encoding effector and immunity proteins are contained in two variable regions of GA3 loci, GA3 T6SSs of the species B. fragilis are likely the source of numerous novel effector and immunity proteins. Importantly, we show that the GA3 T6SS of strain 638R is functional in the mammalian gut and provides a competitive advantage to this organism.


Assuntos
Bacteroides fragilis/fisiologia , Bacteroidetes/fisiologia , Microbioma Gastrointestinal/fisiologia , Sistemas de Secreção Tipo VI/fisiologia , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/imunologia , Proteínas de Bactérias/fisiologia , Bacteroides fragilis/genética , Bacteroides fragilis/imunologia , Bacteroidetes/genética , Bacteroidetes/imunologia , Microbioma Gastrointestinal/imunologia , Genes Bacterianos , Humanos , Camundongos , Mutação , Sistemas de Secreção Tipo VI/genética , Sistemas de Secreção Tipo VI/imunologia
8.
Eur J Immunol ; 47(7): 1188-1199, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28543188

RESUMO

Dysbiosis in gut microbiome has been shown to be associated with inflammatory and autoimmune diseases. Previous studies from our laboratory demonstrated the pivotal role played by CD44 in the regulation of EAE, a murine model of multiple sclerosis. In the current study, we determined whether these effects resulted from an alteration in gut microbiota and the short-chain fatty acid (SCFA) production in CD44 knockout (CD44KO) mice. Fecal transfer from naïve CD44KO but not C57BL/6 wild type (CD44WT) mice, into EAE-induced CD44WT mice, led to significant amelioration of EAE. High-throughput bacterial 16S rRNA gene sequencing, followed by clustering sequences into operational taxonomic units (OTUs) and biochemical analysis, revealed that EAE-induced CD44KO mice showed significant diversity, richness, and evenness when compared to EAE-induced CD44WT mice at the phylum level, with dominant Bacteroidetes (68.5%) and low Firmicutes (26.8%). Further, data showed a significant change in the abundance of SCFAs, propionic acid, and i-butyric acid in EAE-CD44KO compared to EAE-CD44WT mice. In conclusion, our results demonstrate that the attenuation of EAE seen following CD44 gene deletion in mice may result from alterations in the gut microbiota and SCFAs. Furthermore, our studies also demonstrate that the phenotype of gene knock-out animals may be shaped by gut microbiota.


Assuntos
Encefalomielite Autoimune Experimental/imunologia , Microbioma Gastrointestinal/imunologia , Receptores de Hialuronatos/genética , Receptores de Hialuronatos/imunologia , Animais , Bacteroidetes/genética , Bacteroidetes/imunologia , Bacteroidetes/isolamento & purificação , Modelos Animais de Doenças , Disbiose , Encefalomielite Autoimune Experimental/fisiopatologia , Ácidos Graxos Voláteis/imunologia , Transplante de Microbiota Fecal , Fezes/microbiologia , Firmicutes/genética , Firmicutes/imunologia , Firmicutes/isolamento & purificação , Microbioma Gastrointestinal/genética , Deleção de Genes , Metagenômica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Propionatos/metabolismo , RNA Ribossômico 16S
9.
J Autoimmun ; 93: 57-65, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29960834

RESUMO

The incidence of type 1 diabetes (T1D) is determined by both genetic and environmental factors. In recent years, the gut microbiota have been identified to be an important environmental factor that could modify diabetes susceptibility. We have previously shown that Myeloid differentiation primary response gene 88 (MyD88), a major adaptor protein downstream of most innate immune Toll-like receptor (TLR) signaling, is important for mediating diabetes susceptibility in the non-obese diabetic (NOD) mouse model of human T1D. Here we report the role of TIR-domain-containing adapter-inducing interferon-ß (TRIF) in T1D development, as TRIF is an important adaptor protein downstream of TLR3 and TLR4 signaling. We found that TRIF-deficient (TRIF-/-) NOD mice were protected from development of diabetes, but only when housed with TRIF-deficient (TRIF-/-) NOD mice. When housed with TRIF-sufficient wild type (WT, i.e., TRIF+/+) NOD mice, the mice developed diabetes. We further investigated the gut microbiota as a potential cause for the altered diabetes development. Interestingly, TRIF-/-NOD mice had a different microbiota composition compared to WT NOD mice, only if they were housed with TRIF-/-NOD mice. However, the composition of gut microbiota in the TRIF-/-NOD mice was indistinguishable from WT NOD mice, if they were housed with WT NOD mice. The difference in the gut microbiota in TRIF-/-NOD mice, due to cohousing, accorded with the diabetes development in TRIF-/-NOD mice. Comparing the gut microbiota in TRIF-/- and WT NOD mice, we identified changes in percentage of Sutterella, Rikenella and Turicibacter species. Moreover, bacteria from WT NOD mice induced significantly stronger inflammatory immune responses in vitro compared to those from TRIF-/-NOD mice. Further immunological analysis revealed impaired function of dendritic cells and reduced T cell activation and proliferation in TRIF-/-NOD mice. Our data show that TRIF-deficiency protects NOD mice from diabetes development through alteration of the gut microbiota and reduced immune cell activation; however, that protection is over-ridden upon exposure to WT NOD bacteria. Therefore exposure to different microbiota can modify disease susceptibility determined by genetic factors related to innate immunity.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/genética , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/microbiologia , Microbioma Gastrointestinal/imunologia , Fator 88 de Diferenciação Mieloide/genética , Linfócitos T/imunologia , Proteínas Adaptadoras de Transporte Vesicular/deficiência , Proteínas Adaptadoras de Transporte Vesicular/imunologia , Transferência Adotiva , Animais , Bacteroidetes/imunologia , Burkholderiales/imunologia , Proliferação de Células , Células Dendríticas/imunologia , Células Dendríticas/patologia , Diabetes Mellitus Experimental/imunologia , Diabetes Mellitus Experimental/patologia , Suscetibilidade a Doenças , Feminino , Firmicutes/imunologia , Regulação da Expressão Gênica , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos NOD , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/imunologia , Transdução de Sinais , Linfócitos T/patologia , Linfócitos T/transplante , Receptor 3 Toll-Like/genética , Receptor 3 Toll-Like/imunologia , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/imunologia
10.
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
11.
Appl Microbiol Biotechnol ; 102(21): 9317-9329, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30151605

RESUMO

Immune function is influenced by the diversity and stability of the intestinal microbiota. A likely trade-off of immune function for growth has been demonstrated in heavier breeds of poultry that have been genetically selected for growth and feed efficiency traits. We investigated the expression of selected innate immune genes and genes encoding products involved in intestinal barrier function to determine whether function changes could be consistently linked to the phenotypic expression of feed conversion ratio (FCR), a common measure of performance within poultry broiler flocks. In addition, we compared individual cecal microbial composition with innate immune gene expression. Samples were utilised from two replicate trials termed P1E1 and P1E2. High (n = 12) and low (n = 12) performing birds were selected based on their individual FCR data from each replicate and combined for microbiota phylogenetic composition and immune gene expression analysis. Toll-like receptor 1 (TLR1La) and zonula occludens 1 (ZO1) were differentially expressed between high- and low-performing broilers. Several taxa were correlated with FCR; of these, unclassified YS2 and ZO1 were also positively correlated with each other. Interactions between taxa and differentially expressed innate immune genes between P1E1 and P1E2 were much greater compared to relationships between high- and low-performing birds. At the level of phylum, reciprocal correlations between tight junction proteins and Toll-like receptors with Bacteroidetes and Firmicutes were evident, as were correlations at the genus level.


Assuntos
Ceco/imunologia , Ceco/microbiologia , Microbioma Gastrointestinal/imunologia , Imunidade Inata/genética , Intestinos/imunologia , Aves Domésticas/imunologia , Ração Animal/microbiologia , Animais , Bacteroidetes/imunologia , Dieta , Firmicutes/imunologia , Microbioma Gastrointestinal/genética , Expressão Gênica/genética , Expressão Gênica/imunologia , Imunidade Inata/imunologia , Intestinos/microbiologia , Filogenia , Aves Domésticas/genética , Aves Domésticas/microbiologia , Probióticos , Proteínas de Junções Íntimas/metabolismo , Receptores Toll-Like/metabolismo
12.
Gut ; 66(4): 633-643, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-26992426

RESUMO

OBJECTIVE: A signature that unifies the colorectal cancer (CRC) microbiota across multiple studies has not been identified. In addition to methodological variance, heterogeneity may be caused by both microbial and host response differences, which was addressed in this study. DESIGN: We prospectively studied the colonic microbiota and the expression of specific host response genes using faecal and mucosal samples ('ON' and 'OFF' the tumour, proximal and distal) from 59 patients undergoing surgery for CRC, 21 individuals with polyps and 56 healthy controls. Microbiota composition was determined by 16S rRNA amplicon sequencing; expression of host genes involved in CRC progression and immune response was quantified by real-time quantitative PCR. RESULTS: The microbiota of patients with CRC differed from that of controls, but alterations were not restricted to the cancerous tissue. Differences between distal and proximal cancers were detected and faecal microbiota only partially reflected mucosal microbiota in CRC. Patients with CRC can be stratified based on higher level structures of mucosal-associated bacterial co-abundance groups (CAGs) that resemble the previously formulated concept of enterotypes. Of these, Bacteroidetes Cluster 1 and Firmicutes Cluster 1 were in decreased abundance in CRC mucosa, whereas Bacteroidetes Cluster 2, Firmicutes Cluster 2, Pathogen Cluster and Prevotella Cluster showed increased abundance in CRC mucosa. CRC-associated CAGs were differentially correlated with the expression of host immunoinflammatory response genes. CONCLUSIONS: CRC-associated microbiota profiles differ from those in healthy subjects and are linked with distinct mucosal gene-expression profiles. Compositional alterations in the microbiota are not restricted to cancerous tissue and differ between distal and proximal cancers.


Assuntos
Colo/microbiologia , Neoplasias do Colo/microbiologia , Pólipos do Colo/microbiologia , Microbioma Gastrointestinal , RNA Ribossômico 16S/análise , Neoplasias Retais/microbiologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Antígenos de Bactérias/análise , Bacteroidetes/imunologia , Bacteroidetes/isolamento & purificação , Estudos de Casos e Controles , Quimiocina CCL20/genética , Quimiocina CXCL1/genética , Neoplasias do Colo/genética , Pólipos do Colo/genética , Fezes/microbiologia , Feminino , Firmicutes/imunologia , Firmicutes/isolamento & purificação , Microbioma Gastrointestinal/genética , Expressão Gênica , Humanos , Interleucina-17/genética , Interleucina-23/genética , Mucosa Intestinal/microbiologia , Masculino , Pessoa de Meia-Idade , Inibidor 1 de Ativador de Plasminogênio/genética , Prevotella/imunologia , Prevotella/isolamento & purificação , Estudos Prospectivos , Neoplasias Retais/genética
13.
Curr Opin Rheumatol ; 29(4): 374-377, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28376066

RESUMO

PURPOSE OF REVIEW: Systemic lupus erythematosus (SLE) is an autoimmune disease of unknown cause. In recent years, with the emergence of microbiome research, changes in the gut microbiota composition have been correlated with a variety of autoimmune disorders, and several mechanisms linking these together have been suggested, including the hygiene theory, immune system activation and hormonal effects. It has therefore been suggested that gut microbiota may play a role in SLE. In this review, we summarize recent findings on the SLE-related microbiota compositions in both humans and rodents. Evidence linking microbiome with SLE opens a new avenue in researching the cause of SLE as well as improved future treatments. RECENT FINDINGS: Although two studies found a lower Firmicutes/Bacteroidetes ratio in SLE patients vs. controls, there were inconsistencies regarding significant differences in the abundance of specific genera or species. Studies of mouse disease models have shown some correlations between microbial compositions and disease states, also indicating differences between males and females. SUMMARY: Current data support an association between microbiota composition and SLE. Further research is needed to fully unravel this connection, potentially shedding light on mechanisms in SLE development and on the female bias of the disease, improving diagnosis and treatment.


Assuntos
Microbioma Gastrointestinal/imunologia , Lúpus Eritematoso Sistêmico/imunologia , Animais , Bacteroidetes/imunologia , Modelos Animais de Doenças , Feminino , Firmicutes/imunologia , Humanos , Lúpus Eritematoso Sistêmico/microbiologia , Masculino , Camundongos
14.
Appl Environ Microbiol ; 82(14): 4264-78, 2016 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-27208127

RESUMO

UNLABELLED: Intestinal homeostasis mechanisms must protect the host intestinal tissue from endogenous lipopolysaccharides (LPSs) produced by the intestinal microbiota. In this report, we demonstrate that murine intestinal fecal lipids effectively block Toll-like receptor 4 (TLR4) responses to naturally occurring Bacteroidetes sp. LPS. Cardiolipin (CL) represents a significant proportion of the total intestinal and fecal lipids and, furthermore, potently antagonizes TLR4 activation by reducing LPS binding at the lipopolysaccharide binding protein (LBP), CD14, and MD-2 steps of the TLR4 signaling pathway. It is further demonstrated that intestinal lipids and CL are less effective at neutralizing more potent Enterobacteriaceae-type LPS, which is enriched in feces obtained from mice with dextran sodium sulfate (DSS)-treated inflammatory bowel disease. The selective inhibition of naturally occurring LPS structures by intestinal lipids may represent a novel homeostasis mechanism that blocks LPS activation in response to symbiotic but not dysbiotic microbial communities. IMPORTANCE: The guts of animals harbor a variety of Gram-negative bacteria associated with both states of intestinal health and states of disease. Environmental factors, such as dietary habits, can drive the microbial composition of the host animal's intestinal bacterial community toward a more pathogenic state. Both beneficial and harmful Gram-negative bacteria are capable of eliciting potentially damaging inflammatory responses from the host intestinal tissues via a lipopolysaccharide (LPS)-dependent pathway. Physical mucosal barriers and antibodies produced by the intestinal immune system protect against the undesired inflammatory effects of LPS, although it is unknown why some bacteria are more effective at overcoming the protective barriers than others. This report describes the discovery of a lipid-type protective barrier in the intestine that reduces the deleterious effects of LPSs from beneficial bacteria but is less effective in dampening the inflammatory effects of LPSs from harmful bacteria, providing a novel mechanistic insight into inflammatory intestinal disorders.


Assuntos
Cardiolipinas/metabolismo , Fatores Imunológicos/metabolismo , Intestinos/imunologia , Intestinos/microbiologia , Lipopolissacarídeos/imunologia , Receptor 4 Toll-Like/antagonistas & inibidores , Animais , Bacteroidetes/imunologia , Enterobacteriaceae/imunologia , Camundongos
15.
Immunol Rev ; 245(1): 27-44, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22168412

RESUMO

The healthy gut tolerates very large numbers of diverse bacterial species belonging mainly to the Bacteroidetes and Firmicutes phyla. These bacteria normally coexist peacefully with the gut and help maintain immune homeostasis and tolerance. The mechanisms promoting tolerance affect various cell populations, including the epithelial cells lining the gut, resident dendritic cells (DCs), and gut-homing T cells. Gut bacteria also influence multiple signaling pathways from Toll-like receptors to nuclear factor κB and regulate the functionality of DCs and T cells. Several bacterial species have been identified that promote T-cell differentiation, in particular T-helper 17 and T-regulatory cells. Insight into the molecular mechanisms by which bacteria mediate these effects will be very important in identifying new ways of treating intestinal and extra-intestinal immune-mediated diseases. These diseases are increasing dramatically in the human population and require new treatments. It may be possible in the future to identify specific bacterial species or strains that can correct for T-cell imbalances in the gut and promote immune homeostasis, both locally and systemically. In addition, new information describing microbial genomes affords the opportunity to mine for functional genes that may lead to new generation drugs relevant to a range of inflammatory disease conditions.


Assuntos
Bacteroidetes/imunologia , Terapia Biológica , Inflamação/terapia , Enteropatias/imunologia , Enteropatias/microbiologia , Intestinos/imunologia , Intestinos/microbiologia , Probióticos/uso terapêutico , Animais , Antígenos de Bactérias/imunologia , Autoimunidade , Células Dendríticas/imunologia , Células Dendríticas/microbiologia , Humanos , Tolerância Imunológica , Imunomodulação , Enteropatias/terapia , Simbiose , Linfócitos T/imunologia , Linfócitos T/microbiologia
16.
Infect Immun ; 83(12): 4582-93, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26371120

RESUMO

The American Heart Association supports an association between periodontal diseases and atherosclerosis but not a causal association. This study explores the use of the integrin ß6(-/-) mouse model to study the causality. We investigated the ability of a polymicrobial consortium of Porphyromonas gingivalis, Treponema denticola, Tannerella forsythia, and Fusobacterium nucleatum to colonize the periodontium and induce local and systemic inflammatory responses. Polymicrobially infected Itgß6(-/-) mice demonstrate greater susceptibility to gingival colonization/infection, with severe gingival inflammation, apical migration of the junctional epithelium, periodontal pocket formation, alveolar bone resorption, osteoclast activation, bacterial invasion of the gingiva, a greater propensity for the bacteria to disseminate hematogenously, and a strong splenic T cell cytokine response. Levels of atherosclerosis risk factors, including serum nitric oxide, oxidized low-density lipoprotein, serum amyloid A, and lipid peroxidation, were significantly altered by polybacterial infection, demonstrating an enhanced potential for atherosclerotic plaque progression. Aortic gene expression revealed significant alterations in specific Toll-like receptor (TLR) and nucleotide-binding domain- and leucine-rich-repeat-containing receptor (NLR) pathway genes in response to periodontal bacterial infection. Histomorphometry of the aorta demonstrated larger atherosclerotic plaques in Itgß6(-/-) mice than in wild-type (WT) mice but no significant difference in atherosclerotic plaque size between mice with polybacterial infection and mice with sham infection. Fluorescence in situ hybridization demonstrated active invasion of the aortic adventitial layer by P. gingivalis. Our observations suggest that polybacterial infection elicits distinct aortic TLR and inflammasome signaling and significantly increases local aortic oxidative stress. These results are the first to demonstrate the mechanism of the host aortic inflammatory response induced by polymicrobial infection with well-characterized periodontal pathogens.


Assuntos
Túnica Adventícia/patologia , Antígenos de Neoplasias/imunologia , Aorta/patologia , Aterosclerose/complicações , Integrinas/imunologia , Periodontite/complicações , Placa Aterosclerótica/complicações , Túnica Adventícia/imunologia , Túnica Adventícia/microbiologia , Animais , Antígenos de Neoplasias/genética , Aorta/imunologia , Aorta/microbiologia , Aterosclerose/imunologia , Aterosclerose/microbiologia , Aterosclerose/patologia , Bacteroidetes/crescimento & desenvolvimento , Bacteroidetes/imunologia , Bacteroidetes/patogenicidade , Reabsorção Óssea , Modelos Animais de Doenças , Fusobacterium nucleatum/crescimento & desenvolvimento , Fusobacterium nucleatum/imunologia , Fusobacterium nucleatum/patogenicidade , Expressão Gênica , Gengiva/imunologia , Gengiva/microbiologia , Gengiva/patologia , Hibridização in Situ Fluorescente , Inflamassomos , Integrinas/deficiência , Integrinas/genética , Lipoproteínas LDL/genética , Lipoproteínas LDL/imunologia , Camundongos , Camundongos Knockout , Consórcios Microbianos , Periodontite/imunologia , Periodontite/microbiologia , Periodontite/patologia , Periodonto/imunologia , Periodonto/microbiologia , Periodonto/patologia , Placa Aterosclerótica/imunologia , Placa Aterosclerótica/microbiologia , Placa Aterosclerótica/patologia , Porphyromonas gingivalis/crescimento & desenvolvimento , Porphyromonas gingivalis/imunologia , Porphyromonas gingivalis/patogenicidade , Treponema denticola/crescimento & desenvolvimento , Treponema denticola/imunologia , Treponema denticola/patogenicidade
17.
Circ J ; 79(9): 1882-90, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26212124

RESUMO

Atherosclerosis is considered a chronic inflammatory disease and an intervention targeting the inflammatory process could be a new therapeutic strategy for preventing atherosclerotic cardiovascular diseases (CVD). We hypothesized that the intestine, which is considered the biggest immune organ in the human body, could be a therapeutic target for preventing CVD. We demonstrated that oral administration of anti-CD3 antibody or an active form of vitamin D3 reduced atherosclerosis in mice via induction of regulatory T cells and tolerogenic dendritic cells in the gut-associated lymphoid tissues. Similar to regulatory immune responses achieved by oral tolerance, our method had systemic effects that ultimately contributed towards atherosclerosis reduction. Recently, we have been interested in the gut microbiota, which have been reported as highly associated with intestinal immunity and systemic metabolic disorders, including obesity and diabetes. Notably, the guts of obese individuals are predominantly colonized by Firmicutes over Bacteroidetes. The association between atherosclerosis and microbiota has been attracting increased attention, and gut microbiota have been shown to participate in the metabolism of a proatherogenic compound called trimethylamine-N-oxide (TMAO) and aggravate CVD. Our investigation of the relationship between susceptibility to CVD and the gut microbiota revealed a characteristic flora type. Here, we discuss the evidence for the relationship between the gut microbiota and cardiometabolic diseases, and consider the gut microbiota as new potential therapeutic targets for treating CVD. (Circ J 2015; 79: 1882-1890).


Assuntos
Aterosclerose , Bacteroidetes/imunologia , Firmicutes/imunologia , Microbioma Gastrointestinal/imunologia , Imunidade nas Mucosas , Intestinos , Animais , Aterosclerose/imunologia , Aterosclerose/microbiologia , Humanos , Intestinos/imunologia , Intestinos/microbiologia , Metilaminas/imunologia , Camundongos
18.
Amino Acids ; 46(10): 2403-13, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25023447

RESUMO

This study was conducted to determine effects of dietary supplementation with 1 % L-glutamine for 14 days on the abundance of intestinal bacteria and the activation of intestinal innate immunity in mice. The measured variables included (1) the abundance of Bacteroidetes, Firmicutes, Lactobacillus, Streptococcus and Bifidobacterium in the lumen of the small intestine; (2) the expression of toll-like receptors (TLRs), pro-inflammatory cytokines, and antibacterial substances secreted by Paneth cells and goblet cells in the jejunum, ileum and colon; and (3) the activation of TLR4-nuclear factor kappa B (NF-κB), mitogen-activated protein kinases (MAPK), and phosphoinositide-3-kinases (PI3K)/PI3K-protein kinase B (Akt) signaling pathways in the jejunum and ileum. In the jejunum, glutamine supplementation decreased the abundance of Firmicutes, while increased mRNA levels for antibacterial substances in association with the activation of NF-κB and PI3K-Akt pathways. In the ileum, glutamine supplementation induced a shift in the Firmicutes:Bacteroidetes ratio in favor of Bacteroidetes, and enhanced mRNA levels for Tlr4, pro-inflammatory cytokines, and antibacterial substances participating in NF-κB and JNK signaling pathways. These results indicate that the effects of glutamine on the intestine vary with its segments and compartments. Collectively, dietary glutamine supplementation of mice beneficially alters intestinal bacterial community and activates the innate immunity in the small intestine through NF-κB, MAPK and PI3K-Akt signaling pathways.


Assuntos
Suplementos Nutricionais , Glutamina/administração & dosagem , Imunidade Inata , Fatores Imunológicos/administração & dosagem , Mucosa Intestinal/microbiologia , Intestino Delgado/microbiologia , Animais , Bacteroidetes/classificação , Bacteroidetes/crescimento & desenvolvimento , Bacteroidetes/imunologia , Bacteroidetes/isolamento & purificação , Colo/imunologia , Colo/metabolismo , Citocinas/genética , Citocinas/metabolismo , Feminino , Células Caliciformes/imunologia , Células Caliciformes/metabolismo , Bactérias Gram-Positivas/classificação , Bactérias Gram-Positivas/crescimento & desenvolvimento , Bactérias Gram-Positivas/imunologia , Bactérias Gram-Positivas/isolamento & purificação , Íleo/citologia , Íleo/imunologia , Íleo/metabolismo , Íleo/microbiologia , Mucosa Intestinal/citologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , Intestino Delgado/citologia , Intestino Delgado/imunologia , Intestino Delgado/metabolismo , Jejuno/citologia , Jejuno/imunologia , Jejuno/metabolismo , Jejuno/microbiologia , Camundongos Endogâmicos ICR , Tipagem Molecular , Celulas de Paneth/imunologia , Celulas de Paneth/metabolismo , Distribuição Aleatória , Transdução de Sinais , Organismos Livres de Patógenos Específicos , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/metabolismo
19.
Infect Immun ; 81(4): 1198-206, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23357386

RESUMO

Tannerella forsythia is an anaerobic, Gram-negative bacterium involved in the so-called "red complex," which is associated with severe and chronic periodontitis. The surface layer (S-layer) of T. forsythia is composed of cell surface glycoproteins, such as TfsA and TfsB, and is known to play a role in adhesion/invasion and suppression of proinflammatory cytokine expression. Here we investigated the association of this S-layer with serum resistance and coaggregation with other oral bacteria. The growth of the S-layer-deficient mutant in a bacterial medium containing more than 20% non-heat-inactivated calf serum (CS) or more than 40% non-heat-inactivated human serum was significantly suppressed relative to that of the wild type (WT). Next, we used confocal microscopy to perform quantitative analysis on the effect of serum. The survival ratio of the mutant exposed to 100% non-heat-inactivated CS (76% survival) was significantly lower than that of the WT (97% survival). Furthermore, significant C3b deposition was observed in the mutant but not in the WT. In a coaggregation assay, the mutant showed reduced coaggregation with Streptococcus sanguinis, Streptococcus salivarius, and Porphyromonas gingivalis but strong coaggregation with Fusobacterium nucleatum. These results indicated that the S-layer of T. forsythia plays multiple roles in virulence and may be associated with periodontitis.


Assuntos
Aderência Bacteriana , Bacteroidetes/imunologia , Bacteroidetes/fisiologia , Glicoproteínas de Membrana/metabolismo , Viabilidade Microbiana/efeitos dos fármacos , Soro/imunologia , Soro/microbiologia , Bacteroidetes/genética , Complemento C3b/imunologia , Complemento C3b/metabolismo , Humanos , Glicoproteínas de Membrana/genética
20.
Eur J Clin Microbiol Infect Dis ; 32(10): 1333-40, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23660697

RESUMO

Chronic periodontitis (CP) is considered to be a multifactorial disease influenced by microbial and genetic factors. The aim of the present study was to investigate whether the genetic susceptibility to CP in individuals with the IL8 ATC/TTC haplotype is associated with subgingival levels of periodontopathogens. Sixty-five individuals, grouped according to the presence (n = 28) or absence (n = 37) of the IL8 haplotype, were evaluated. After clinical periodontal evaluation, each group was subdivided according to the presence (CP) or absence (H) of periodontitis. Four subgingival samples were obtained from CP and two samples per subject from H patients. The levels and proportions of Porphyromonas gingivalis, Tannerella forsythia, and Treponema denticola were analyzed using quantitative real-time polymerase chain reaction (q-PCR). No differences were found in the proportion of periodontopathogenic bacteria between groups with the presence or absence of the IL8 haplotype. However, in the CP groups, the levels of periodontopathogens were significantly higher in the individuals without the IL8 haplotype than in the individuals with the IL8 haplotype. These results suggest that periodontal destruction may occur in patients who are considered to be genetically susceptible to CP with a lower microbial challenge because of the presence of the IL8 ATC/TTC haplotype than in patients without this haplotype.


Assuntos
Carga Bacteriana , Bacteroidetes/isolamento & purificação , Periodontite Crônica/imunologia , Predisposição Genética para Doença , Interleucina-8/genética , Porphyromonas gingivalis/isolamento & purificação , Treponema denticola/isolamento & purificação , Adulto , Bacteroidetes/imunologia , Periodontite Crônica/microbiologia , Feminino , Haplótipos , Humanos , Interleucina-8/imunologia , Masculino , Pessoa de Meia-Idade , Porphyromonas gingivalis/imunologia , Reação em Cadeia da Polimerase em Tempo Real , Treponema denticola/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA