Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 119(31): e2120028119, 2022 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-35878027

RESUMO

Type 1 diabetes (T1D) is an autoimmune disease characterized by the destruction of pancreatic ß-cells. One of the earliest aspects of this process is the development of autoantibodies and T cells directed at an epitope in the B-chain of insulin (insB:9-23). Analysis of microbial protein sequences with homology to the insB:9-23 sequence revealed 17 peptides showing >50% identity to insB:9-23. Of these 17 peptides, the hprt4-18 peptide, found in the normal human gut commensal Parabacteroides distasonis, activated both human T cell clones from T1D patients and T cell hybridomas from nonobese diabetic (NOD) mice specific to insB:9-23. Immunization of NOD mice with P. distasonis insB:9-23 peptide mimic or insB:9-23 peptide verified immune cross-reactivity. Colonization of female NOD mice with P. distasonis accelerated the development of T1D, increasing macrophages, dendritic cells, and destructive CD8+ T cells, while decreasing FoxP3+ regulatory T cells. Western blot analysis identified P. distasonis-reacting antibodies in sera of NOD mice colonized with P. distasonis and human T1D patients. Furthermore, adoptive transfer of splenocytes from P. distasonis-treated mice to NOD/SCID mice enhanced disease phenotype in the recipients. Finally, analysis of human children gut microbiome data from a longitudinal DIABIMMUNE study revealed that seroconversion rates (i.e., the proportion of individuals developing two or more autoantibodies) were consistently higher in children whose microbiome harbored sequences capable of producing the hprt4-18 peptide compared to individuals who did not harbor it. Taken together, these data demonstrate the potential role of a gut microbiota-derived insB:9-23-mimic peptide as a molecular trigger of T1D pathogenesis.


Assuntos
Diabetes Mellitus Tipo 1 , Microbioma Gastrointestinal , Mimetismo Molecular , Peptídeos , Animais , Autoanticorpos/imunologia , Bacteroidetes , Linfócitos T CD8-Positivos , Criança , Diabetes Mellitus Tipo 1/patologia , Feminino , Humanos , Insulina/genética , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Peptídeos/química
2.
EBioMedicine ; 69: 103443, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-34186487

RESUMO

BACKGROUND: Birth by caesarean section (CS) is associated with aberrant gut microbiome development and greater disease susceptibility later in life. We investigated whether oral administration of maternal vaginal microbiota to infants born by CS could restore their gut microbiome development in a pilot single-blinded, randomised placebo-controlled trial (Australian New Zealand Clinical Trials Registry, ACTRN12618000339257). METHODS: Pregnant women scheduled for a CS underwent comprehensive antenatal pathogen screening. At birth, healthy neonates were randomised to receive a 3 ml solution of either maternal vaginal microbes (CS-seeded, n = 12) or sterile water (CS-placebo, n = 13). Vaginally-born neonates were used as the reference control (VB, n = 22). Clinical assessments occurred within the first 2 h of birth, and at 1 month and 3 months of age. Infant stool samples and maternal vaginal extracts from CS women underwent shotgun metagenomic sequencing. The primary outcome was gut microbiome composition at 1 month of age. Secondary outcomes included maternal strain engraftment, functional potential of the gut microbiome, anthropometry, body composition, and adverse events. FINDINGS: Despite the presence of viable microbial cells within transplant solutions, there were no observed differences in gut microbiome composition or functional potential between CS-seeded and CS-placebo infants at 1 month or 3 months of age. Both CS groups displayed the characteristic signature of low Bacteroides abundance, which contributed to a number of biosynthesis pathways being underrepresented when compared with VB microbiomes. Maternal vaginal strain engraftment was rare. Vaginal seeding had no observed effects on anthropometry or body composition. There were no serious adverse events associated with treatment. INTERPRETATION: Our pilot findings question the value of vaginal seeding given that oral administration of maternal vaginal microbiota did not alter early gut microbiome development in CS-born infants. The limited colonisation of maternal vaginal strains suggest that other maternal sources, such as the perianal area, may play a larger role in seeding the neonatal gut microbiome. FUNDING: Health Research Council of New Zealand, A Better Start - National Science Challenge.


Assuntos
Cesárea/efeitos adversos , Transplante de Microbiota Fecal/métodos , Microbioma Gastrointestinal , Doenças do Recém-Nascido/microbiologia , Vagina/microbiologia , Administração Oral , Adulto , Bacteroides/patogenicidade , Transplante de Microbiota Fecal/efeitos adversos , Feminino , Humanos , Recém-Nascido , Doenças do Recém-Nascido/etiologia , Doenças do Recém-Nascido/prevenção & controle , Masculino
3.
Gastroenterology ; 159(6): 2193-2202.e5, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32860788

RESUMO

BACKGROUND & AIMS: Crohn's disease (CD) is a chronic gastrointestinal disease resulting from the dysfunctional interplay between genetic susceptibility, the immune system, and commensal intestinal microbiota. Emerging evidence suggests that treatment by suppression of the immune response and replacement of the microbiota through fecal microbiota transplantation (FMT) is a promising approach for the treatment of CD. METHODS: We obtained stool metagenomes from CD patients in remission and assessed gut microbiome composition before and after FMT at the species and strain levels. Longitudinal follow-up evaluation allowed us to identify the gain, loss, and strain replacement of specific species and link these events to the maintenance of remission in CD. RESULTS: We found that FMT had a significant long-term effect on patient microbial compositions, although this was primarily driven by the engraftment of donor species, which remained at low abundance. Thirty-eight percent of FMT-driven changes were strain replacements, emphasizing the importance of detailed profiling methods, such as metagenomics. Several instances of long-term coexistence between donor and patient strains were also observed. Engraftment of some Actinobacteria, and engraftment or loss of Proteobacteria, were related to better disease outcomes in CD patients who received FMT, and transmission of Bacteroidetes was deleterious. CONCLUSIONS: Our results suggest clades that may be beneficial to transmit/eliminate through FMT, and provide criteria that may help identify personalized FMT donors to more effectively maintain remission in CD patients. The framework established here creates a foundation for future studies centered around the application of FMT and defined microbial communities as a therapeutic approach for treating CD.


Assuntos
Doença de Crohn/terapia , Transplante de Microbiota Fecal/métodos , Microbioma Gastrointestinal/genética , Adulto , Doença de Crohn/imunologia , Doença de Crohn/microbiologia , Conjuntos de Dados como Assunto , Fezes/microbiologia , Feminino , Microbioma Gastrointestinal/imunologia , Haplótipos , Humanos , Masculino , Metagenômica , Pessoa de Meia-Idade , Tipagem Molecular , Filogenia , Indução de Remissão/métodos , Resultado do Tratamento , Adulto Jovem
4.
Nat Microbiol ; 4(2): 293-305, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30531976

RESUMO

The inflammatory bowel diseases (IBDs), which include Crohn's disease (CD) and ulcerative colitis (UC), are multifactorial chronic conditions of the gastrointestinal tract. While IBD has been associated with dramatic changes in the gut microbiota, changes in the gut metabolome-the molecular interface between host and microbiota-are less well understood. To address this gap, we performed untargeted metabolomic and shotgun metagenomic profiling of cross-sectional stool samples from discovery (n = 155) and validation (n = 65) cohorts of CD, UC and non-IBD control patients. Metabolomic and metagenomic profiles were broadly correlated with faecal calprotectin levels (a measure of gut inflammation). Across >8,000 measured metabolite features, we identified chemicals and chemical classes that were differentially abundant in IBD, including enrichments for sphingolipids and bile acids, and depletions for triacylglycerols and tetrapyrroles. While > 50% of differentially abundant metabolite features were uncharacterized, many could be assigned putative roles through metabolomic 'guilt by association' (covariation with known metabolites). Differentially abundant species and functions from the metagenomic profiles reflected adaptation to oxidative stress in the IBD gut, and were individually consistent with previous findings. Integrating these data, however, we identified 122 robust associations between differentially abundant species and well-characterized differentially abundant metabolites, indicating possible mechanistic relationships that are perturbed in IBD. Finally, we found that metabolome- and metagenome-based classifiers of IBD status were highly accurate and, like the vast majority of individual trends, generalized well to the independent validation cohort. Our findings thus provide an improved understanding of perturbations of the microbiome-metabolome interface in IBD, including identification of many potential diagnostic and therapeutic targets.


Assuntos
Microbioma Gastrointestinal , Doenças Inflamatórias Intestinais/metabolismo , Doenças Inflamatórias Intestinais/microbiologia , Biodiversidade , Biomarcadores/metabolismo , Colite Ulcerativa/imunologia , Colite Ulcerativa/metabolismo , Colite Ulcerativa/microbiologia , Doença de Crohn/imunologia , Doença de Crohn/metabolismo , Doença de Crohn/microbiologia , Fezes/química , Fezes/microbiologia , Microbioma Gastrointestinal/genética , Microbioma Gastrointestinal/imunologia , Humanos , Inflamação/metabolismo , Inflamação/microbiologia , Doenças Inflamatórias Intestinais/imunologia , Complexo Antígeno L1 Leucocitário/análise , Metaboloma , Metagenoma
5.
Inflamm Bowel Dis ; 22(9): 2182-90, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27542133

RESUMO

BACKGROUND: The microbiota in the lumen of patients with Crohn's disease (CD) is characterized by reduced diversity, particularly Firmicutes and Bacteroidetes. It is unknown whether the introduction of the intestinal microbiota from healthy individuals could correct this dysbiosis and reverse mucosal inflammation. We investigated the response to fecal microbial transplantation (FMT) from healthy individuals to subjects with active CD. METHODS: We performed a prospective open-label study (uncontrolled) of FMT from healthy donors to subjects with active CD. A single FMT was performed by colonoscopy. Recipients' microbial diversity, mucosal T-cell phenotypes, and clinical and inflammatory parameters were measured over 12 weeks, and safety over 26 weeks. RESULTS: Nineteen subjects were treated with FMT and completed the study follow-up. Fifty-eight percent (11/19) demonstrated a clinical response (Harvey-Bradshaw Index decrease >3) following FMT. Fifteen subjects had sufficient pre/postfecal samples for analysis. A significant increase in microbial diversity occurred after FMT (P = 0.02). This was greater in clinical responders than nonresponders. Patients who experienced a clinical response demonstrated a significant shift in fecal microbial composition toward their donor's profile as assessed by the Bray-Curtis index at 4 weeks (P = 0.003). An increase in regulatory T cells (CD4CD25CD127lo) was also noted in recipients' lamina propria following FMT. No serious adverse events were noted over the 26-week study period. CONCLUSIONS: In this open-label study, FMT led to an expansion in microbial bacterial diversity in patients with active CD. FMT was overall safe, although the clinical response was variable. Determining donor microbial factors that influence clinical response is needed before randomized clinical trials of FMT in CD.


Assuntos
Doença de Crohn/microbiologia , Doença de Crohn/terapia , Disbiose/terapia , Transplante de Microbiota Fecal , Microbioma Gastrointestinal , Adulto , Seleção do Doador , Fezes/microbiologia , Feminino , Humanos , Mucosa Intestinal/imunologia , Intestinos/microbiologia , Masculino , Redes e Vias Metabólicas , Pessoa de Meia-Idade , Estudos Prospectivos , Indução de Remissão , Linfócitos T/classificação , Estados Unidos , Sequenciamento Completo do Genoma , Adulto Jovem
6.
Genome Med ; 8(1): 75, 2016 07 13.
Artigo em Inglês | MEDLINE | ID: mdl-27412252

RESUMO

BACKGROUND: Gut microbiome dysbiosis has been demonstrated in subjects with newly diagnosed and chronic inflammatory bowel disease (IBD). In this study we sought to explore longitudinal changes in dysbiosis and ascertain associations between dysbiosis and markers of disease activity and treatment outcome. METHODS: We performed a prospective cohort study of 19 treatment-naïve pediatric IBD subjects and 10 healthy controls, measuring fecal calprotectin and assessing the gut microbiome via repeated stool samples. Associations between clinical characteristics and the microbiome were tested using generalized estimating equations. Random forest classification was used to predict ultimate treatment response (presence of mucosal healing at follow-up colonoscopy) or non-response using patients' pretreatment samples. RESULTS: Patients with Crohn's disease had increased markers of inflammation and dysbiosis compared to controls. Patients with ulcerative colitis had even higher inflammation and dysbiosis compared to those with Crohn's disease. For all cases, the gut microbial dysbiosis index associated significantly with clinical and biological measures of disease severity, but did not associate with treatment response. We found differences in specific gut microbiome genera between cases/controls and responders/non-responders including Akkermansia, Coprococcus, Fusobacterium, Veillonella, Faecalibacterium, and Adlercreutzia. Using pretreatment microbiome data in a weighted random forest classifier, we were able to obtain 76.5 % accuracy for prediction of responder status. CONCLUSIONS: Patient dysbiosis improved over time but persisted even among those who responded to treatment and achieved mucosal healing. Although dysbiosis index was not significantly different between responders and non-responders, we found specific genus-level differences. We found that pretreatment microbiome signatures are a promising avenue for prediction of remission and response to treatment.


Assuntos
Bactérias/classificação , Colite Ulcerativa/tratamento farmacológico , Doença de Crohn/tratamento farmacológico , Disbiose/tratamento farmacológico , Fatores Imunológicos/uso terapêutico , Mucosa Intestinal/efeitos dos fármacos , Adolescente , Bactérias/genética , Bactérias/isolamento & purificação , Biomarcadores/metabolismo , Estudos de Casos e Controles , Criança , Pré-Escolar , Colite Ulcerativa/diagnóstico , Colite Ulcerativa/imunologia , Colite Ulcerativa/microbiologia , Doença de Crohn/diagnóstico , Doença de Crohn/imunologia , Doença de Crohn/microbiologia , Disbiose/diagnóstico , Disbiose/imunologia , Disbiose/microbiologia , Fezes/química , Fezes/microbiologia , Feminino , Microbioma Gastrointestinal/efeitos dos fármacos , Microbioma Gastrointestinal/genética , Trato Gastrointestinal/efeitos dos fármacos , Trato Gastrointestinal/imunologia , Trato Gastrointestinal/microbiologia , Humanos , Inflamação , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Complexo Antígeno L1 Leucocitário/metabolismo , Estudos Longitudinais , Masculino , Índice de Gravidade de Doença , Resultado do Tratamento
7.
Science ; 352(6285): 565-9, 2016 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-27126040

RESUMO

Deep sequencing of the gut microbiomes of 1135 participants from a Dutch population-based cohort shows relations between the microbiome and 126 exogenous and intrinsic host factors, including 31 intrinsic factors, 12 diseases, 19 drug groups, 4 smoking categories, and 60 dietary factors. These factors collectively explain 18.7% of the variation seen in the interindividual distance of microbial composition. We could associate 110 factors to 125 species and observed that fecal chromogranin A (CgA), a protein secreted by enteroendocrine cells, was exclusively associated with 61 microbial species whose abundance collectively accounted for 53% of microbial composition. Low CgA concentrations were seen in individuals with a more diverse microbiome. These results are an important step toward a better understanding of environment-diet-microbe-host interactions.


Assuntos
Bactérias/classificação , Microbioma Gastrointestinal/genética , Trato Gastrointestinal/microbiologia , Bactérias/genética , Bactérias/isolamento & purificação , Cromogranina A/análise , Cromogranina A/metabolismo , Dieta , Células Enteroendócrinas/metabolismo , Fezes/química , Fezes/microbiologia , Marcadores Genéticos , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Metagenômica , Países Baixos , Filogenia , RNA Ribossômico 16S/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA