Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
1.
Exp Dermatol ; 33(5): e15087, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38685821

RESUMO

Hidradenitis Suppurativa (HS) is a chronic autoinflammatory skin disease with activated keratinocytes, tunnel formation and a complex immune infiltrate in tissue. The HS microbiome is polymicrobial with an abundance of commensal gram-positive facultative (GPs) Staphylococcus species and gram-negative anaerobic (GNA) bacteria like Prevotella, Fusobacterium and Porphyromonas with increasing predominance of GNAs with disease severity. We sought to define the keratinocyte response to bacteria commonly isolated from HS lesions to probe pathogenic relationships between HS and the microbiome. Type strains of Prevotella nigrescens, Prevotella melaninogenica, Prevotella intermedia, Prevotella asaccharolytica, Fusobacterium nucleatum, as well as Staphylococcus aureus and the normal skin commensal Staphylococcus epidermidis were heat-killed and co-incubated with normal human keratinocytes. RNA was collected and analysed using RNAseq and RT-qPCR. The supernatant was collected from cell culture for protein quantification. Transcriptomic profiles between HS clinical samples and stimulated keratinocytes were compared. Co-staining of patient HS frozen sections was used to localize bacteria in lesions. A mouse intradermal injection model was used to investigate early immune recruitment. TLR4 and JAK inhibitors were used to investigate mechanistic avenues of bacterial response inhibition. GNAs, especially F. nucleatum, stimulated vastly higher CXCL8, IL17C, CCL20, IL6, TNF and IL36γ transcription in normal skin keratinocytes than the GPs S. epidermidis and S. aureus. Using RNAseq, we found that F. nucleatum (and Prevotella) strongly induced the IL-17 pathway in keratinocytes and overlapped with transcriptome profiles of HS patient clinical samples. Bacteria were juxtaposed to activated keratinocytes in vivo, and F. nucleatum strongly recruited murine neutrophil and macrophage migration. Both the TLR4 and pan-JAK inhibitors reduced cytokine production. Detailed transcriptomic profiling of healthy skin keratinocytes exposed to GNAs prevalent in HS revealed a potent, extensive inflammatory response vastly stronger than GPs. GNAs stimulated HS-relevant genes, including many genes in the IL-17 response pathway, and were significantly associated with HS tissue transcriptomes. The close association of activated keratinocytes with bacteria in HS lesions and innate infiltration in murine skin cemented GNA pathogenic potential. These novel mechanistic insights could drive future targeted therapies.


Assuntos
Hidradenite Supurativa , Queratinócitos , Queratinócitos/imunologia , Queratinócitos/microbiologia , Queratinócitos/metabolismo , Humanos , Animais , Camundongos , Hidradenite Supurativa/microbiologia , Hidradenite Supurativa/imunologia , Staphylococcus aureus/imunologia , Staphylococcus epidermidis/imunologia , Fusobacterium nucleatum/imunologia , Transcriptoma , Citocinas/metabolismo , Bactérias Anaeróbias , Interleucina-17/metabolismo , Microbiota , Prevotella/imunologia
2.
Anaerobe ; 68: 102237, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-32721554

RESUMO

The human intestinal tract is colonized by a large number of diverse microorganisms that play various important physiologic functions. In inflammatory gut diseases including celiac disease (CeD), a dysbiotic state of microbiome has been observed. Interestingly, this perturbed microbiome is normalized towards eubiosis in patients showing recovery after treatment. The treatment has been observed to increase the abundance of beneficial microbes in comparison to non-treated patients. In this study, we investigated the effect of Prevotella histicola or Prevotella melaninogenica, isolated from the duodenum of a treated CeD patient, on the induction and maintenance of oral tolerance to gliadin, a CeD associated subgroup of gluten proteins, in NOD.DQ8.ABo transgenic mice. Conventionally raised mice on a gluten free diet were orally gavaged with bacteria before and after injection with pepsin trypsin digested gliadin (PTD-gliadin). P. histicola suppressed the cellular response to gliadin, whereas P. melaninogenica failed to suppress an immune response against gliadin. Interestingly, tolerance to gliadin in NOD.DQ8.ABo mice may be associated with gut microbiota as mice gavaged with P melaninogenica harbored a different microbial diversity as compared to P. histicola treated mice. This study provides experimental evidence that gut microbes like P. histicola from treated patients can suppress the immune response against gliadin epitopes.


Assuntos
Doença Celíaca/imunologia , Doença Celíaca/microbiologia , Microbioma Gastrointestinal , Gliadina/imunologia , Linfócitos T/imunologia , Animais , Feminino , Humanos , Tolerância Imunológica , Masculino , Camundongos , Camundongos Endogâmicos NOD , Prevotella/imunologia , Prevotella/fisiologia , Prevotella melaninogenica/imunologia , Prevotella melaninogenica/fisiologia
3.
Mucosal Immunol ; 14(1): 113-124, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32433514

RESUMO

Diverse microbial signatures within the intestinal microbiota have been associated with intestinal and systemic inflammatory diseases, but whether these candidate microbes actively modulate host phenotypes or passively expand within the altered microbial ecosystem is frequently not known. Here we demonstrate that colonization of mice with a member of the genus Prevotella, which has been previously associated to colitis in mice, exacerbates intestinal inflammation. Our analysis revealed that Prevotella intestinalis alters composition and function of the ecosystem resulting in a reduction of short-chain fatty acids, specifically acetate, and consequently a decrease in intestinal IL-18 levels during steady state. Supplementation of IL-18 to Prevotella-colonized mice was sufficient to reduce intestinal inflammation. Hence, we conclude that intestinal Prevotella colonization results in metabolic changes in the microbiota, which reduce IL-18 production and consequently exacerbate intestinal inflammation, and potential systemic autoimmunity.


Assuntos
Infecções por Bacteroidaceae/imunologia , Infecções por Bacteroidaceae/microbiologia , Microbioma Gastrointestinal/imunologia , Interações Hospedeiro-Patógeno/imunologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Prevotella/imunologia , Imunidade Adaptativa , Animais , Citocinas/metabolismo , Modelos Animais de Doenças , Suscetibilidade a Doenças , Mediadores da Inflamação/metabolismo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Metagenoma , Metagenômica/métodos , Camundongos , Camundongos Knockout , Mucosite/etiologia , Mucosite/metabolismo , Mucosite/patologia
4.
J Immunol Res ; 2020: 9607328, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33204736

RESUMO

Th17-mediated mucosal inflammation is related to increased Prevotella bacterial abundance. The actual involvement of Prevotella in the development and accumulation of intestinal Th17 cells at a steady state, however, remains undefined. Herein, we investigated the role of Prevotella in inducing intestinal Th17 cells in mice. Mice were treated with a combination of broad-spectrum antibiotics (including ampicillin, neomycin sulfate, vancomycin hydrochloride, and metronidazole) in their drinking water for 4 weeks and then gavaged with Prevotella for 4 weeks. After inoculation, 16S rDNA sequencing was used to verify the colonization of Prevotella in the colon of mice. The IL-17A as well as IL-17A-expressing T cells was localized and quantified by an immunofluorescence assay (IFA) of colon sections. Th17 cells in the mesenteric lymph nodes of mice were counted by flow cytometry. Systemic immune response to Prevotella colonization was evaluated based on the serum levels of IL-6, TNF-α, IL-1ß, IL-17A, IL-10, IL-4, IFN-γ, and IL-2. Th17-polarizing cytokines (IL-6, TNF-α, IL-1ß, and IL-2) induced by Prevotella were evaluated by stimulation of bone marrow-derived dendritic cells (BMDCs). Results revealed that after inoculation, Prevotella successfully colonized the intestine of mice and induced the production and accumulation of colonic Th17 cells in the colon. Moreover, Prevotella elevated some of the Th17-related cytokines in the serum of mice. And Th17-polarizing cytokines (IL-6 and IL-1ß) produced by BMDCs were mediated mainly through the interaction between Prevotella and Toll-like receptor 2 (TLR2). In conclusion, our data suggest that Prevotella induces the production of Th17 cells in the colon of mice, thus highlighting the potential role of Prevotella in training the intestinal immune system.


Assuntos
Colo/imunologia , Colo/metabolismo , Microbioma Gastrointestinal/imunologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , Prevotella/imunologia , Células Th17/imunologia , Células Th17/metabolismo , Animais , Antibacterianos/farmacologia , Colo/microbiologia , Citocinas/biossíntese , Citocinas/sangue , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Microbioma Gastrointestinal/efeitos dos fármacos , Regulação da Expressão Gênica , Imunofenotipagem , Interleucina-17/genética , Interleucina-17/metabolismo , Ativação Linfocitária/efeitos dos fármacos , Ativação Linfocitária/imunologia , Metagenoma , Metagenômica/métodos , Camundongos , Camundongos Transgênicos , Prevotella/efeitos dos fármacos
5.
PLoS One ; 15(10): e0235803, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33031374

RESUMO

Cystic Fibrosis (CF), caused by mutations affecting the CFTR gene, is characterised by viscid secretions in multiple organ systems. CF airways contain thick mucus, creating a gradient of hypoxia, which promotes the establishment of polymicrobial infection. Such inflammation predisposes to further infection, a self-perpetuating cycle in mediated by NF-κB. Anaerobic Gram-negative Prevotella spp. are found in sputum from healthy volunteers and CF patients and in CF lungs correlate with reduced levels of inflammation. Prevotella histicola (P. histicola) can suppress murine lung inflammation, however, no studies have examined the role of P. histicola in modulating infection and inflammation in the CF airways. We investigated innate immune signalling and NF-kB activation in CF epithelial cells CFBE41o- in response to clinical stains of P. histicola and Pseudomonas aeruginosa (P. aeruginosa). Toll-Like Receptor (TLR) expressing HEK-293 cells and siRNA assays for TLRs and IKKα were used to confirm signalling pathways. We show that P. histicola infection activated the alternative NF-kB signalling pathway in CF bronchial epithelial cells inducing HIF-1α protein. TLR5 signalling was responsible for the induction of the alternative NF-kB pathway through phosphorylation of IKKα. The induction of transcription factor HIF-1α was inversely associated with the induction of the alternative NF-kB pathway and knockdown of IKKα partially restored canonical NF-kB activation in response to P. histicola. This study demonstrates that different bacterial species in the respiratory microbiome can contribute differently to inflammation, either by activating inflammatory cascades (P. aeruginosa) or by muting the inflammatory response by modulating similar or related pathways (P. histicola). Further work is required to assess the complex interactions of the lung microbiome in response to mixed bacterial infections and their effects in people with CF.


Assuntos
Brônquios/imunologia , Fibrose Cística/imunologia , NF-kappa B/metabolismo , Prevotella/imunologia , Infecções por Pseudomonas/complicações , Pseudomonas aeruginosa/imunologia , Receptores Toll-Like/metabolismo , Brônquios/metabolismo , Brônquios/microbiologia , Brônquios/patologia , Fibrose Cística/metabolismo , Fibrose Cística/microbiologia , Fibrose Cística/patologia , Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Células Epiteliais/patologia , Humanos , Interleucina-8/metabolismo , NF-kappa B/genética , Prevotella/isolamento & purificação , Infecções por Pseudomonas/microbiologia , Pseudomonas aeruginosa/isolamento & purificação , Transdução de Sinais , Receptores Toll-Like/imunologia
6.
Cancer Immunol Immunother ; 69(12): 2665-2671, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-32761425

RESUMO

INTRODUCTION: A hypersensitivity response akin to immune reconstitution inflammatory syndrome (IRIS) has been proposed as a mechanism responsible for anti-PD-1 therapy-induced tuberculosis. IRIS is associated with enhanced activation of IL-17A-expressing CD4 + T cells (Th17). Gut microbiota is thought to be linked to pulmonary inflammation through the gut-lung axis. MATERIALS AND METHODS: We used ImmuCellAI to investigate the T cell population in lung cancer and tuberculosis samples. Then, we applied flow cytometry to monitor the expression levels of the Th17 cell activation marker CD38 in the peripheral blood of a patient experiencing adverse events, including tuberculosis, in response to pembrolizumab. The gut microbiome was examined by 16S rRNA sequencing to examine the alterations caused by pembrolizumab. RESULTS: The percentage of Th17 cells was increased in both lung cancer and tuberculosis. FACS analysis showed that pembrolizumab induced substantial CD38 expression in Th17 cells. The patient's fecal samples showed that the diversity of the gut microbiota was significantly increased in response to the pembrolizumab cycle. One enriched genus was Prevotella, which has previously been linked to lung inflammation and Th17 immune activation. DISCUSSION: The observed Th17 activation in our patient was consistent with a role of Th17-mediated IRIS in pembrolizumab-triggered tuberculosis. Pembrolizumab might trigger airway inflammation with a Th17 phenotype through microbiota interactions in the gut-lung axis.


Assuntos
Anticorpos Monoclonais Humanizados/efeitos adversos , Microbioma Gastrointestinal/imunologia , Síndrome Inflamatória da Reconstituição Imune/imunologia , Neoplasias Pulmonares/tratamento farmacológico , Células Th17/imunologia , Tuberculose/imunologia , Antituberculosos/uso terapêutico , DNA Bacteriano/isolamento & purificação , Conjuntos de Dados como Assunto , Microbioma Gastrointestinal/genética , Humanos , Síndrome Inflamatória da Reconstituição Imune/sangue , Síndrome Inflamatória da Reconstituição Imune/induzido quimicamente , Síndrome Inflamatória da Reconstituição Imune/microbiologia , Pulmão/diagnóstico por imagem , Pulmão/imunologia , Pulmão/microbiologia , Pulmão/patologia , Neoplasias Pulmonares/sangue , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/microbiologia , Masculino , Pessoa de Meia-Idade , Mycobacterium tuberculosis/imunologia , Mycobacterium tuberculosis/isolamento & purificação , Prevotella/genética , Prevotella/imunologia , Prevotella/isolamento & purificação , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Receptor de Morte Celular Programada 1/imunologia , RNA Ribossômico 16S/genética , Células Th17/efeitos dos fármacos , Tomografia Computadorizada por Raios X , Tuberculose/induzido quimicamente , Tuberculose/tratamento farmacológico , Tuberculose/microbiologia
7.
J Infect Dis ; 222(12): 2082-2092, 2020 11 13.
Artigo em Inglês | MEDLINE | ID: mdl-32515473

RESUMO

BACKGROUND: Prevotella species are commonly isolated from the reproductive tract of women with obstetric/gynecologic health complications. However, contributions of this genus to changes in local microenvironment are not well characterized. Our objective was to evaluate species-specific effects of Prevotella on the human endometrial epithelium. METHODS: Thirteen Prevotella strains, originally isolated from the human oral cavity, amniotic fluid, endometrium, or vagina (including women with bacterial vaginosis), were obtained from BEI and ATCC resources. Bacteria were evaluated in silico and in vitro using human endometrial epithelial cells (EEC) grown as monolayers or a 3-dimensional (3D) model. RESULTS: Genomic characterization illustrated metabolic and phylogenetic diversity of Prevotella genus. Among tested species, P. disiens exhibited cytotoxicity. Scanning electron microscopy analysis of the 3D EEC model revealed species-specific colonization patterns and alterations of ultracellular structures. Infection with sialidase-producing P. timonensis resulted in elongated microvilli, and increased MUC3 and MUC4 expression. Infections with Prevotella species, including P. bivia, did not result in significant proinflammatory activation of EEC. CONCLUSIONS: Collectively, findings indicate that Prevotella species are metabolically diverse and overall not cytotoxic or overtly inflammatory in EEC; however, these bacteria can form biofilms, alter barrier properties of the endometrial epithelium, and ultimately impact colonization of secondary colonizers.


Assuntos
Células Epiteliais/microbiologia , Imunidade Inata , Prevotella/genética , Prevotella/patogenicidade , Linhagem Celular Tumoral , Endométrio/citologia , Células Epiteliais/imunologia , Feminino , Humanos , Microscopia Eletrônica de Varredura , Mucinas/genética , Prevotella/imunologia , Especificidade da Espécie
8.
J Immunol ; 205(3): 842-852, 2020 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-32571839

RESUMO

Secretory Ig A (sIgA) plays an important role in the maintenance of intestinal homeostasis via cross-talk with gut microbiota. The defects in sIgA production could elicit dysbiosis of commensal microbiota and subsequently facilitate the development of inflammatory bowel disease. Our previous study revealed activating transcription factor 3 (ATF3) as an important regulator of follicular helper T (TFH) cells in gut. ATF3 deficiency in CD4+ T cells impaired the development of gut TFH cells, and therefore diminished sIgA production, which increased the susceptibility to murine colitis. However, the potential role of microbiota in ATF3-mediated gut homeostasis remains incompletely understood. In this study, we report that both Atf3-/- and CD4creAtf3fl/fl mice displayed profound dysbiosis of gut microbiota when compared with their littermate controls. The proinflammatory Prevotella taxa, especially Prevotella copri, were more abundant in ATF3-deficient mice when compared with littermate controls. This phenotype was obviously abrogated by adoptive transfer of either TFH cells or IgA+ B cells. Importantly, depletion of gut microbiota dramatically alleviated the severity of colitis in Atf3-/- mice, whereas transfer of microbiota from Atf3-/- mice to wild-type recipients increased their susceptibility to colitis. Collectively, these observations indicate the importance of IgA-microbiota interaction in ATF3-mediated gut homeostasis.


Assuntos
Fator 3 Ativador da Transcrição/imunologia , Linfócitos B/imunologia , Microbioma Gastrointestinal/imunologia , Homeostase/imunologia , Imunoglobulina A/imunologia , Células T Auxiliares Foliculares/imunologia , Fator 3 Ativador da Transcrição/genética , Animais , Disbiose/genética , Disbiose/imunologia , Disbiose/microbiologia , Homeostase/genética , Imunoglobulina A/genética , Camundongos , Camundongos Knockout , Prevotella/imunologia
9.
Clin Immunol ; 216: 108463, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32437923

RESUMO

OBJECTIVES: Patients with Crohn's disease often produce antibodies against flagellated intestinal bacteria. There are mixed data as to whether such antibodies are present in patients with spondyloarthritis. Our objectives were to evaluate for the presence of antibodies against intestinal organisms in children with enthesitis related arthritis (ERA). METHODS: Children with ERA and healthy controls were recruited at three sites. Sera were plated on a nitrocellulose array and incubated with labelled antibodies to human IgA and IgG. RESULTS: At UAB, patients and controls had similar antibody levels against the majority of the bacteria selected, with the exception of increased IgA antibodies among ERA patients against Prevotella oralis (1231 [IQR 750, 2566] versus 706 [IQR 428, 1106], p = .007.) These findings were partially validated at a second but not at a third site. CONCLUSIONS: ERA patients may produce increased IgA antibodies against P. oralis. The possible significance of this finding bears further exploration.


Assuntos
Antígenos de Bactérias/sangue , Antígenos de Bactérias/imunologia , Artrite Juvenil/sangue , Artrite Juvenil/imunologia , Prevotella/imunologia , Artrite Juvenil/microbiologia , Criança , Doença de Crohn/imunologia , Doença de Crohn/microbiologia , Feminino , Humanos , Imunoglobulina A/sangue , Imunoglobulina G/sangue , Masculino
10.
Nat Commun ; 11(1): 1999, 2020 04 24.
Artigo em Inglês | MEDLINE | ID: mdl-32332850

RESUMO

Emerging evidence suggests associations between the vaginal microbiota (VMB) composition, human papillomavirus (HPV) infection, and cervical intraepithelial neoplasia (CIN); however, causal inference remains uncertain. Here, we use bacterial DNA sequencing from serially collected vaginal samples from a cohort of 87 adolescent and young women aged 16-26 years with histologically confirmed, untreated CIN2 lesions to determine whether VMB composition affects rates of regression over 24 months. We show that women with a Lactobacillus-dominant microbiome at baseline are more likely to have regressive disease at 12 months. Lactobacillus spp. depletion and presence of specific anaerobic taxa including Megasphaera, Prevotella timonensis and Gardnerella vaginalis are associated with CIN2 persistence and slower regression. These findings suggest that VMB composition may be a future useful biomarker in predicting disease outcome and tailoring surveillance, whilst it may offer rational targets for the development of new prevention and treatment strategies.


Assuntos
Microbiota/imunologia , Infecções por Papillomavirus/microbiologia , Displasia do Colo do Útero/microbiologia , Neoplasias do Colo do Útero/microbiologia , Vagina/microbiologia , Adolescente , Adulto , Colo do Útero/patologia , Colo do Útero/virologia , Estudos de Coortes , DNA Bacteriano/isolamento & purificação , Feminino , Seguimentos , Gardnerella vaginalis/genética , Gardnerella vaginalis/imunologia , Gardnerella vaginalis/isolamento & purificação , Humanos , Lactobacillus/genética , Lactobacillus/imunologia , Lactobacillus/isolamento & purificação , Microbiota/genética , Estadiamento de Neoplasias , Papillomaviridae/imunologia , Papillomaviridae/isolamento & purificação , Infecções por Papillomavirus/imunologia , Infecções por Papillomavirus/patologia , Prevotella/genética , Prevotella/imunologia , Prevotella/isolamento & purificação , RNA Ribossômico 16S/genética , Fatores de Risco , Neoplasias do Colo do Útero/imunologia , Neoplasias do Colo do Útero/patologia , Adulto Jovem , Displasia do Colo do Útero/imunologia , Displasia do Colo do Útero/patologia
11.
J Reprod Immunol ; 138: 103085, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32004804

RESUMO

Dysbiosis of the vaginal microbiome as a result of overgrowth of anaerobic bacteria leads to bacterial vaginosis (BV) which is associated with increased inflammation in the genital mucosa. Moreover, BV increases susceptibility to sexual transmitted infections (STIs) and is associated with adverse pregnancy outcomes. It remains unclear how specific vaginal aerobic and anaerobic bacteria affect health and disease. We selected different vaginal bacteria ranging from true commensals to species associated with dysbiosis and investigated their effects on activation of dendritic cells (DCs). Commensal Lactobacilli crispatus did not induce DC maturation nor led to production of pro-inflammatory cytokines. In contrast, BV-associated bacteria Megasphaera elsdenii and Prevotella timonensis induced DC maturation and increased levels of pro-inflammatory cytokines. Notably, DCs stimulated with Prevotella timonensis suppressed Th2 responses and induced Th1 skewing, typically associated with preterm birth. In contrast, Lactobacillus crispatus and Megasphaera elsdenii did not affect Th cell polarization. These results strongly indicate that the interaction of vaginal bacteria with mucosal DCs determines mucosal inflammation and we have identified the anaerobic bacterium Prevotella timonensis as a strong inducer of inflammatory responses. Specifically targeting these inflammation-inducing bacteria might be a therapeutic strategy to prevent BV and associated risks in STI susceptibility and preterm birth.


Assuntos
Células Dendríticas/imunologia , Disbiose/complicações , Megasphaera elsdenii/imunologia , Prevotella/imunologia , Vaginose Bacteriana/imunologia , Células Cultivadas , Citocinas/metabolismo , Células Dendríticas/metabolismo , Disbiose/imunologia , Disbiose/microbiologia , Feminino , Humanos , Mediadores da Inflamação/metabolismo , Leucócitos Mononucleares , Prevotella/isolamento & purificação , Cultura Primária de Células , Vagina/citologia , Vagina/imunologia , Vagina/microbiologia , Vaginose Bacteriana/microbiologia
12.
Nat Commun ; 10(1): 5001, 2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31676759

RESUMO

Asthma is believed to arise through early life aberrant immune development in response to environmental exposures that may influence the airway microbiota. Here, we examine the airway microbiota during the first three months of life by 16S rRNA gene amplicon sequencing in the population-based Copenhagen Prospective Studies on Asthma in Childhood 2010 (COPSAC2010) cohort consisting of 700 children monitored for the development of asthma since birth. Microbial diversity and the relative abundances of Veillonella and Prevotella in the airways at age one month are associated with asthma by age 6 years, both individually and with additional taxa in a multivariable model. Higher relative abundance of these bacteria is furthermore associated with an airway immune profile dominated by reduced TNF-α and IL-1ß and increased CCL2 and CCL17, which itself is an independent predictor for asthma. These findings suggest a mechanism of microbiota-immune interactions in early infancy that predisposes to childhood asthma.


Assuntos
Asma/imunologia , Bactérias/imunologia , Citocinas/imunologia , Microbiota/imunologia , Sistema Respiratório/imunologia , Asma/diagnóstico , Asma/microbiologia , Bactérias/classificação , Bactérias/crescimento & desenvolvimento , Criança , Citocinas/metabolismo , Feminino , Humanos , Lactente , Recém-Nascido , Masculino , Microbiota/genética , Microbiota/fisiologia , Prevotella/genética , Prevotella/imunologia , Estudos Prospectivos , RNA Ribossômico 16S/genética , Sistema Respiratório/microbiologia , Veillonella/genética , Veillonella/imunologia
13.
Ann Rheum Dis ; 78(5): 590-593, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30760471

RESUMO

OBJECTIVES: Rheumatoid arthritis (RA) has been associated with a relative expansion of faecal Prevotellaceae. To determine the microbiome composition and prevalence of Prevotella spp. in a group of individuals at increased risk for RA, but prior to the development of the disease. METHODS: In an ongoing cohort study of first-degree relatives (FDRs) of patients with RA, we identified 'FDR controls', asymptomatic and without autoantibodies, and individuals in pre-clinical RA stages, who had either developed anticitrullinated peptide antibodies or rheumatoid factor positivity and/or symptoms and signs associated with possible RA. Stool sampling and culture-independent microbiota analyses were performed followed by descriptive statistics and statistical analyses of community structures. RESULTS: A total of 133 participants were included, of which 50 were categorised as 'FDR controls' and 83 in 'pre-clinical RA stages'. The microbiota of individuals in 'pre-clinical RA stages' was significantly altered compared with FDR controls. We found a significant enrichment of the bacterial family Prevotellaceae, particularly Prevotella spp., in the 'pre-clinical RA' group (p=0.04). CONCLUSIONS: Prevotella spp. enrichment in individuals in pre-clinical stages of RA, before the onset of RA, suggests a role of intestinal dysbiosis in the development of RA.


Assuntos
Artrite Reumatoide/microbiologia , Fezes/microbiologia , Microbioma Gastrointestinal/imunologia , Prevotella/imunologia , Idoso , Artrite Reumatoide/genética , Artrite Reumatoide/imunologia , Autoanticorpos/sangue , Autoanticorpos/imunologia , Estudos de Casos e Controles , Disbiose/sangue , Disbiose/imunologia , Disbiose/microbiologia , Feminino , Predisposição Genética para Doença , Humanos , Masculino , Pessoa de Meia-Idade , Linhagem , Fator Reumatoide/sangue , Fatores de Risco
14.
Nat Commun ; 9(1): 4832, 2018 12 03.
Artigo em Inglês | MEDLINE | ID: mdl-30510245

RESUMO

The gut microbiota has been causally linked to cancer, yet how intestinal microbes influence progression of extramucosal tumors is poorly understood. Here we provide evidence implying that Prevotella heparinolytica promotes the differentiation of Th17 cells colonizing the gut and migrating to the bone marrow (BM) of transgenic Vk*MYC mice, where they favor progression of multiple myeloma (MM). Lack of IL-17 in Vk*MYC mice, or disturbance of their microbiome delayed MM appearance. Similarly, in smoldering MM patients, higher levels of BM IL-17 predicted faster disease progression. IL-17 induced STAT3 phosphorylation in murine plasma cells, and activated eosinophils. Treatment of Vk*MYC mice with antibodies blocking IL-17, IL-17RA, and IL-5 reduced BM accumulation of Th17 cells and eosinophils and delayed disease progression. Thus, in Vk*MYC mice, commensal bacteria appear to unleash a paracrine signaling network between adaptive and innate immunity that accelerates progression to MM, and can be targeted by already available therapies.


Assuntos
Eosinófilos/imunologia , Microbioma Gastrointestinal/imunologia , Interleucina-17/imunologia , Mieloma Múltiplo/imunologia , Células Th17/imunologia , Animais , Medula Óssea/imunologia , Medula Óssea/metabolismo , Diferenciação Celular/imunologia , Movimento Celular/imunologia , Progressão da Doença , Eosinófilos/metabolismo , Humanos , Interleucina-17/genética , Interleucina-17/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Mieloma Múltiplo/metabolismo , Mieloma Múltiplo/patologia , Prevotella/imunologia , Células Th17/metabolismo
15.
Gut ; 67(9): 1716-1725, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29934437

RESUMO

The microbiome has received increasing attention over the last 15 years. Although gut microbes have been explored for several decades, investigations of the role of microorganisms that reside in the human gut has attracted much attention beyond classical infectious diseases. For example, numerous studies have reported changes in the gut microbiota during not only obesity, diabetes, and liver diseases but also cancer and even neurodegenerative diseases. The human gut microbiota is viewed as a potential source of novel therapeutics. Between 2013 and 2017, the number of publications focusing on the gut microbiota was, remarkably, 12 900, which represents four-fifths of the total number of publications over the last 40 years that investigated this topic. This review discusses recent evidence of the impact of the gut microbiota on metabolic disorders and focus on selected key mechanisms. This review also aims to provide a critical analysis of the current knowledge in this field, identify putative key issues or problems and discuss misinterpretations. The abundance of metagenomic data generated on comparing diseased and healthy subjects can lead to the erroneous claim that a bacterium is causally linked with the protection or the onset of a disease. In fact, environmental factors such as dietary habits, drug treatments, intestinal motility and stool frequency and consistency are all factors that influence the composition of the microbiota and should be considered. The cases of the bacteria Prevotella copri and Akkermansia muciniphila will be discussed as key examples.


Assuntos
Microbioma Gastrointestinal/imunologia , Doenças Metabólicas/imunologia , Doenças Metabólicas/microbiologia , Diabetes Mellitus/imunologia , Diabetes Mellitus/microbiologia , Disbiose/prevenção & controle , Medicina Baseada em Evidências , Humanos , Hepatopatias/imunologia , Hepatopatias/microbiologia , Doenças Metabólicas/metabolismo , Doenças Metabólicas/prevenção & controle , Neoplasias/imunologia , Neoplasias/microbiologia , Doenças Neurodegenerativas/imunologia , Doenças Neurodegenerativas/microbiologia , Obesidade/imunologia , Obesidade/microbiologia , Prevotella/imunologia , Verrucomicrobia/imunologia
16.
Cell Rep ; 20(6): 1269-1277, 2017 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-28793252

RESUMO

The human gut is colonized by a large number of microorganisms (∼1013 bacteria) that support various physiologic functions. A perturbation in the healthy gut microbiome might lead to the development of inflammatory diseases, such as multiple sclerosis (MS). Therefore, gut commensals might provide promising therapeutic options for treating MS and other diseases. We report the identification of human gut-derived commensal bacteria, Prevotella histicola, which can suppress experimental autoimmune encephalomyelitis (EAE) in a human leukocyte antigen (HLA) class II transgenic mouse model. P. histicola suppresses disease through the modulation of systemic immune responses. P. histicola challenge led to a decrease in pro-inflammatory Th1 and Th17 cells and an increase in the frequencies of CD4+FoxP3+ regulatory T cells, tolerogenic dendritic cells, and suppressive macrophages. Our study provides evidence that the administration of gut commensals may regulate a systemic immune response and may, therefore, have a possible role in treatment strategies for MS.


Assuntos
Encefalomielite Autoimune Experimental/terapia , Microbioma Gastrointestinal , Prevotella/imunologia , Probióticos/uso terapêutico , Animais , Linfócitos T CD4-Positivos/imunologia , Células Cultivadas , Células Dendríticas/imunologia , Encefalomielite Autoimune Experimental/microbiologia , Fatores de Transcrição Forkhead/metabolismo , Genes MHC da Classe II , Humanos , Macrófagos/imunologia , Camundongos , Prevotella/patogenicidade , Células Th1/imunologia , Células Th17/imunologia
17.
Immunology ; 151(4): 363-374, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28542929

RESUMO

The microbiota plays a central role in human health and disease by shaping immune development, immune responses and metabolism, and by protecting from invading pathogens. Technical advances that allow comprehensive characterization of microbial communities by genetic sequencing have sparked the hunt for disease-modulating bacteria. Emerging studies in humans have linked the increased abundance of Prevotella species at mucosal sites to localized and systemic disease, including periodontitis, bacterial vaginosis, rheumatoid arthritis, metabolic disorders and low-grade systemic inflammation. Intriguingly, Prevotella abundance is reduced within the lung microbiota of patients with asthma and chronic obstructive pulmonary disease. Increased Prevotella abundance is associated with augmented T helper type 17 (Th17) -mediated mucosal inflammation, which is in line with the marked capacity of Prevotella in driving Th17 immune responses in vitro. Studies indicate that Prevotella predominantly activate Toll-like receptor 2, leading to production of Th17-polarizing cytokines by antigen-presenting cells, including interleukin-23 (IL-23) and IL-1. Furthermore, Prevotella stimulate epithelial cells to produce IL-8, IL-6 and CCL20, which can promote mucosal Th17 immune responses and neutrophil recruitment. Prevotella-mediated mucosal inflammation leads to systemic dissemination of inflammatory mediators, bacteria and bacterial products, which in turn may affect systemic disease outcomes. Studies in mice support a causal role of Prevotella as colonization experiments promote clinical and inflammatory features of human disease. When compared with strict commensal bacteria, Prevotella exhibit increased inflammatory properties, as demonstrated by augmented release of inflammatory mediators from immune cells and various stromal cells. These findings indicate that some Prevotella strains may be clinically important pathobionts that can participate in human disease by promoting chronic inflammation.


Assuntos
Infecções por Bacteroidaceae/imunologia , Imunidade , Inflamação/imunologia , Mucosa Intestinal/imunologia , Microbiota/imunologia , Prevotella/imunologia , Células Th17/imunologia , Animais , Infecções por Bacteroidaceae/diagnóstico , Doença Crônica , Citocinas/metabolismo , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Imunomodulação , Mucosa Intestinal/microbiologia , Prevotella/genética , Receptor 2 Toll-Like/metabolismo
18.
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
19.
Arthritis Rheumatol ; 69(5): 964-975, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-27863183

RESUMO

OBJECTIVE: Prevotella copri, an intestinal microbe, may overexpand in stool samples from patients with new-onset rheumatoid arthritis (RA), but it is not yet clear whether the organism has immune relevance in RA pathogenesis. METHODS: HLA-DR-presented peptides (T cell epitopes) from P copri were sought directly in the patients' synovial tissue or peripheral blood mononuclear cell (PBMC) samples using tandem mass spectrometry. The antigenicity of peptides or their source proteins was examined in samples from the RA patients or comparison groups. T cell reactivity was determined by enzyme-linked immunospot assay; antibody responses were measured by enzyme-linked immunosorbent assay, and cytokine/chemokine determinations were made by bead-based assays. Serum and synovial fluid samples were examined for 16S ribosomal DNA for P copri using nested polymerase chain reaction analysis. RESULTS: In PBMCs, we identified an HLA-DR-presented peptide from a 27-kd protein of P copri (Pc-p27), which stimulated Th1 responses in 42% of patients with new-onset RA. In both new-onset RA patients and chronic RA patients, 1 subgroup had IgA antibody responses to either Pc-p27 or the whole organism, which correlated with Th17 cytokine responses and frequent anti-citrullinated protein antibodies (ACPAs). The other subgroup had IgG P copri antibodies, which were associated with Prevotella DNA in synovial fluid, P copri-specific Th1 responses, and less frequent ACPAs. In contrast, P copri antibody responses were rarely found in patients with other rheumatic diseases or in healthy controls. CONCLUSION: Subgroups of RA patients have differential IgG or IgA immune reactivity with P copri, which appears to be specific for this disease. These observations provide evidence that P copri is immune-relevant in RA pathogenesis.


Assuntos
Artrite Reumatoide/imunologia , Proteínas de Bactérias/imunologia , Epitopos de Linfócito T/imunologia , Microbioma Gastrointestinal/imunologia , Leucócitos Mononucleares/imunologia , Prevotella/imunologia , Membrana Sinovial/imunologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Artrite Reumatoide/microbiologia , DNA Ribossômico/genética , DNA Ribossômico/metabolismo , Ensaio de Imunoadsorção Enzimática , ELISPOT , Feminino , Microbioma Gastrointestinal/genética , Humanos , Imunoglobulina A/imunologia , Imunoglobulina G/imunologia , Masculino , Pessoa de Meia-Idade , Compostos Organotiofosforados/imunologia , Peptídeos/imunologia , Reação em Cadeia da Polimerase , Prevotella/genética , Líquido Sinovial/metabolismo , Espectrometria de Massas em Tandem , Células Th1/imunologia , Adulto Jovem
20.
Mucosal Immunol ; 10(2): 299-306, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27966551

RESUMO

The lungs are not sterile or free from bacteria; rather, they harbor a distinct microbiome whose composition is driven by different ecological rules than for the gastrointestinal tract. During disease, there is often a shift in community composition towards Gammaproteobacteria, the bacterial class that contains many common lung-associated gram-negative "pathogens." Numerous byproducts of host inflammation are growth factors for these bacteria. The extracellular nutrient supply for bacteria in the lungs, which is severely limited during health, markedly increases due to the presence of mucus and vascular permeability. While Gammaproteobacteria benefit from airway inflammation, they also encode molecular components that promote inflammation, potentially creating a cyclical inflammatory mechanism. In contrast, Prevotella species that are routinely acquired via microaspiration from the oral cavity may participate in immunologic homeostasis of the airways.vAreas of future research include determining for specific lung diseases (1) whether an altered lung microbiome initiates disease pathogenesis, promotes chronic inflammation, or is merely a marker of injury and inflammation, (2) whether the lung microbiome can be manipulated therapeutically to change disease progression, (3) what molecules (metabolites) generated during an inflammatory response promote cross-kingdom signaling, and (4) how the lung "ecosystem" collapses during pneumonia, to be dominated by a single pathogen.


Assuntos
Disbiose/imunologia , Gammaproteobacteria/imunologia , Pneumopatias/microbiologia , Microbiota , Pneumonia/microbiologia , Prevotella/imunologia , Sistema Respiratório/microbiologia , Animais , Permeabilidade Capilar , Homeostase , Interações Hospedeiro-Patógeno , Humanos , Pneumopatias/imunologia , Pneumonia/imunologia , Sistema Respiratório/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...