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












Base de datos
Intervalo de año de publicación
1.
BMC Microbiol ; 24(1): 237, 2024 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-38961326

RESUMEN

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.


Asunto(s)
Bacteroidetes , Trasplante de Microbiota Fecal , Microbioma Gastrointestinal , Inmunoterapia , Neoplasias de la Vejiga Urinaria , Neoplasias de la Vejiga Urinaria/terapia , Neoplasias de la Vejiga Urinaria/inmunología , Neoplasias de la Vejiga Urinaria/microbiología , Animales , Humanos , Ratones , Inmunoterapia/métodos , Bacteroidetes/genética , Bacteroidetes/inmunología , Femenino , Masculino , ARN Ribosómico 16S/genética , Heces/microbiología , Persona de Mediana Edad , Anciano , Ratones Endogámicos C57BL
2.
Science ; 377(6606): 660-666, 2022 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-35926021

RESUMEN

The microbiome contributes to the development and maturation of the immune system. In response to commensal bacteria, intestinal CD4+ T lymphocytes differentiate into functional subtypes with regulatory or effector functions. The development of small intestine intraepithelial lymphocytes that coexpress CD4 and CD8αα homodimers (CD4IELs) depends on the microbiota. However, the identity of the microbial antigens recognized by CD4+ T cells that can differentiate into CD4IELs remains unknown. We identified ß-hexosaminidase, a conserved enzyme across commensals of the Bacteroidetes phylum, as a driver of CD4IEL differentiation. In a mouse model of colitis, ß-hexosaminidase-specific lymphocytes protected against intestinal inflammation. Thus, T cells of a single specificity can recognize a variety of abundant commensals and elicit a regulatory immune response at the intestinal mucosa.


Asunto(s)
Bacteroidetes , Linfocitos T CD4-Positivos , Colitis , Mucosa Intestinal , beta-N-Acetilhexosaminidasas , Animales , Bacteroidetes/enzimología , Bacteroidetes/inmunología , Linfocitos T CD4-Positivos/inmunología , Antígenos CD8/inmunología , Colitis/inmunología , Colitis/microbiología , Modelos Animales de Enfermedad , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Ratones , Ratones Endogámicos C57BL , beta-N-Acetilhexosaminidasas/inmunología
4.
Gastroenterology ; 162(1): 166-178, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34606847

RESUMEN

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.


Asunto(s)
Bacteroidetes/inmunología , Colitis/terapia , Colon/microbiología , Trasplante de Microbiota Fecal , Microbioma Gastrointestinal , Inmunoglobulina A/inmunología , Mucosa Intestinal/microbiología , Animales , Bacteroidetes/genética , Bacteroidetes/metabolismo , Ensayos Clínicos como Asunto , Colitis/inmunología , Colitis/metabolismo , Colitis/microbiología , Colitis Ulcerosa/diagnóstico , Colitis Ulcerosa/inmunología , Colitis Ulcerosa/metabolismo , Colitis Ulcerosa/microbiología , Colon/inmunología , Colon/metabolismo , Modelos Animales de Enfermedad , Factores de Transcripción Forkhead/metabolismo , Microbioma Gastrointestinal/genética , Microbioma Gastrointestinal/inmunología , Vida Libre de Gérmenes , Humanos , Inmunidad Mucosa , Inmunoglobulina A/genética , Inmunoglobulina A/metabolismo , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Linfocitos Intraepiteliales/inmunología , Linfocitos Intraepiteliales/metabolismo , Linfocitos Intraepiteliales/microbiología , Metagenoma , Metagenómica , Ratones Endogámicos C57BL , Ratones Noqueados , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Linfocitos T Reguladores/microbiología , Resultado del Tratamiento
5.
Sci Rep ; 11(1): 8966, 2021 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-33903700

RESUMEN

Since alterations in the intestinal microbiota may induce systemic inflammation and polarization of macrophages to the M1 state, the microbiome role in atherosclerosis, an M1-driven disease, requires evaluation. We aimed to determine if antibiotic (Abx) induced alterations to the intestinal microbiota interferes with atherosclerotic plaque inflammation resolution after lipid-lowering in mice. Hyperlipidemic Apoe-/- mice were fed a western diet to develop aortic atherosclerosis with aortas then transplanted into normolipidemic wild-type (WT) mice to model clinically aggressive lipid management and promote atherosclerosis inflammation resolution. Gut microbial composition pre and post-transplant was altered via an enteral antibiotic or not. Post aortic transplant, after Abx treatment, while plaque size did not differ, compared to Apoe-/- mice, Abx- WT recipient mice had a 32% reduction in CD68-expressing cells (p = 0.02) vs. a non-significant 12% reduction in Abx+ WT mice. A trend toward an M1 plaque CD68-expresing cell phenotype was noted in Abx+ mice. By 16S rRNA sequence analysis, the Abx+ mice had reduced alpha diversity and increased Firmicutes/Bacteroidetes relative abundance ratio with a correlation between gut Firmicutes abundance and plaque CD68-expressing cell content (p < 0.05). These results indicate that in a murine atherosclerotic plaque inflammation resolution model, antibiotic-induced microbiome perturbation may blunt the effectiveness of lipid-lowering to reduce the content of plaque inflammatory CD68-expressing cells.


Asunto(s)
Aterosclerosis , Bacteroidetes , Firmicutes , Microbioma Gastrointestinal/inmunología , Placa Aterosclerótica , Animales , Aterosclerosis/genética , Aterosclerosis/inmunología , Aterosclerosis/microbiología , Bacteroidetes/genética , Bacteroidetes/inmunología , Modelos Animales de Enfermedad , Firmicutes/genética , Firmicutes/inmunología , Inflamación/genética , Inflamación/inmunología , Inflamación/microbiología , Ratones , Ratones Noqueados para ApoE , Placa Aterosclerótica/genética , Placa Aterosclerótica/inmunología , Placa Aterosclerótica/microbiología
6.
Carbohydr Polym ; 255: 117388, 2021 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-33436217

RESUMEN

Pectins are a part of daily diet as well as food additives that are indigestible polysaccharides by human enzymes, however, they can be easily degraded by gut bacteria with the production of short chain fatty acids (SCFAs). Knowledge of pectin gut homeostasis and further how pectin affect gut bacterial communities is insufficient and limited. This review focuses on providing the whole story of how pectin functions as prebiotics in the gut. Understanding the interplay between functional and immunological responses inside animal or human gut as influenced by pectin in diets is provided. The interaction between pectin and gut microbiota is presented from both sides, in terms of how pectin affects gut microbiome and or the fermentation products produced in response by gut bacteria. This knowledge can be used to define preferred dietary pectins, targeting beneficial bacteria, and favoring balanced microbiota communities in the gut to maximize pectins' health benefits.


Asunto(s)
Microbioma Gastrointestinal/inmunología , Homeostasis/inmunología , Inmunomodulación/fisiología , Pectinas/farmacología , Polisacáridos/administración & dosificación , Prebióticos/administración & dosificación , Animales , Bacteroidetes/genética , Bacteroidetes/inmunología , Biotransformación , Ensayos Clínicos como Asunto , Dieta/métodos , Ácidos Grasos Volátiles/biosíntesis , Fermentación , Firmicutes/genética , Firmicutes/inmunología , Humanos , Pectinas/inmunología , Pectinas/metabolismo , Polisacáridos/análisis , Prebióticos/análisis
7.
Int J Med Sci ; 18(2): 511-519, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33390820

RESUMEN

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.


Asunto(s)
Composición Corporal/inmunología , Diabetes Mellitus Tipo 2/inmunología , Disbiosis/complicaciones , Microbioma Gastrointestinal/inmunología , Anciano , Bacteroidetes/genética , Bacteroidetes/inmunología , Bacteroidetes/aislamiento & purificación , ADN Bacteriano/aislamiento & purificación , Diabetes Mellitus Tipo 2/microbiología , Disbiosis/diagnóstico , Disbiosis/inmunología , Disbiosis/microbiología , Femenino , Firmicutes/genética , Firmicutes/inmunología , Firmicutes/aislamiento & purificación , Microbioma Gastrointestinal/genética , Humanos , Masculino , Persona de Mediana Edad , Prevalencia , ARN Ribosómico 16S/genética , Factores de Riesgo
8.
Cells ; 9(9)2020 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-32947881

RESUMEN

Alterations in the gut microbiota composition and diversity seem to play a role in the development of chronic diseases, including inflammatory bowel disease (IBD), leading to gut barrier disruption and induction of proinflammatory immune responses. This opens the door for the use of novel health-promoting bacteria. We selected five Parabacteroides distasonis strains isolated from human adult and neonates gut microbiota. We evaluated in vitro their immunomodulation capacities and their ability to reinforce the gut barrier and characterized in vivo their protective effects in an acute murine model of colitis. The in vitro beneficial activities were highly strain dependent: two strains exhibited a potent anti-inflammatory potential and restored the gut barrier while a third strain reinstated the epithelial barrier. While their survival to in vitro gastric conditions was variable, the levels of P. distasonis DNA were higher in the stools of bacteria-treated animals. The strains that were positively scored in vitro displayed a strong ability to rescue mice from colitis. We further showed that two strains primed dendritic cells to induce regulatory T lymphocytes from naïve CD4+ T cells. This study provides better insights on the functionality of commensal bacteria and crucial clues to design live biotherapeutics able to target inflammatory chronic diseases such as IBD.


Asunto(s)
Bacteroidetes/genética , Bacteroidetes/inmunología , Colitis/inducido químicamente , Colitis/microbiología , Microbioma Gastrointestinal/inmunología , Ácido Trinitrobencenosulfónico/efectos adversos , Adulto , Animales , Bacteroidetes/aislamiento & purificación , Células CACO-2 , Colitis/inmunología , ADN Bacteriano/genética , ADN Bacteriano/metabolismo , Modelos Animales de Enfermedad , Heces/microbiología , Femenino , Humanos , Recién Nacido , Enfermedades Inflamatorias del Intestino/inmunología , Enfermedades Inflamatorias del Intestino/microbiología , Mucosa Intestinal/inmunología , Ratones , Ratones Endogámicos BALB C , Reacción en Cadena en Tiempo Real de la Polimerasa , Linfocitos T Reguladores/inmunología
9.
Nutrients ; 12(4)2020 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-32230951

RESUMEN

Altered intestinal microbiota is associated with systemic and intestinal diseases, such as inflammatory bowel disease (IBD). Dysbiotic microbiota with enhanced proinflammatory capacity is characterized by depletion of anaerobic commensals, increased proportion of facultatively anaerobic bacteria, as well as reduced diversity and stability. In this study, we developed a high-throughput in vitro screening assay to isolate intestinal commensal bacteria with anti-inflammatory capacity from a healthy fecal microbiota transplantation donor. Freshly isolated gut bacteria were screened for their capacity to attenuate Escherichia coli lipopolysaccharide (LPS)-induced interleukin 8 (IL-8) release from HT-29 cells. The screen yielded a number of Bacteroides and Parabacteroides isolates, which were identified as P. distasonis, B. caccae, B. intestinalis, B. uniformis, B. fragilis, B. vulgatus and B. ovatus using whole genome sequencing. We observed that a cell-cell contact with the epithelium was not necessary to alleviate in vitro inflammation as spent culture media from the isolates were also effective and the anti-inflammatory action did not correlate with the enterocyte adherence capacity of the isolates. The anti-inflammatory isolates also exerted enterocyte monolayer reinforcing action and lacked essential genes to synthetize hexa-acylated, proinflammatory lipid A, part of LPS. Yet, the anti-inflammatory effector molecules remain to be identified. The Bacteroides strains isolated and characterized in this study have potential to be used as so-called next-generation probiotics.


Asunto(s)
Antiinflamatorios/metabolismo , Bacteroides , Microbioma Gastrointestinal/inmunología , Adulto , Bacteroides/clasificación , Bacteroides/inmunología , Bacteroides/aislamiento & purificación , Bacteroides/metabolismo , Bacteroidetes/clasificación , Bacteroidetes/inmunología , Bacteroidetes/aislamiento & purificación , Bacteroidetes/metabolismo , Células CACO-2 , Heces/microbiología , Femenino , Ensayos Analíticos de Alto Rendimiento , Homeostasis/inmunología , Humanos , Interleucina-8/análisis , Interleucina-8/metabolismo , Lipopolisacáridos/metabolismo , Probióticos
10.
Curr Opin Allergy Clin Immunol ; 20(3): 323-328, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32250972

RESUMEN

PURPOSE OF REVIEW: To perform a nonsystematic review of the literature on the microbiota in the different types of non-IgE-mediated food allergy. RECENT FINDINGS: The commonest non-IgE-mediated disorders managed by allergists include: eosinophilic esophagitis, food protein-induced enteropathy, food protein-induced enterocolitis syndrome, and food protein-induced allergic proctocolitis. The review of the literature describes how at phylum level we observe an increase of Proteobacteria in eosinophilic esophagitis esophageal microbiota and in food protein-induced enterocolitis syndrome, and food protein-induced allergic proctocolitis gut microbiota, while we observe an increase of Bacteroidetes in healthy controls. Several studies endorse the concept that a bloom of Proteobacteria in the gut reflects dysbiosis or an unstable gut microbial community structure. In several studies, the type of diet, the use of probiotics and in a single experience the use of fecal microbiota transplantation has produced significant variations of the microbiota. SUMMARY: Genetic factors alone cannot account for the rapid rise in food allergy prevalence and the microbiome might be contributing to allergy risk. Our review showed that common features of the pathological microbiota among different types of non-IgE-mediated food allergy can be identified. These evidences suggest a possible role of the microbiota in the pathogenesis and non-IgE-mediated food allergies and the need to understand the effects of its modulation on the disorders themselves.


Asunto(s)
Disbiosis/inmunología , Hipersensibilidad a los Alimentos/inmunología , Microbioma Gastrointestinal/inmunología , Bacteroidetes/inmunología , Proteínas en la Dieta/inmunología , Disbiosis/diagnóstico , Disbiosis/microbiología , Enteritis/epidemiología , Enteritis/inmunología , Enteritis/microbiología , Eosinofilia/epidemiología , Eosinofilia/inmunología , Eosinofilia/microbiología , Esofagitis Eosinofílica/epidemiología , Esofagitis Eosinofílica/inmunología , Esofagitis Eosinofílica/microbiología , Heces/microbiología , Hipersensibilidad a los Alimentos/epidemiología , Hipersensibilidad a los Alimentos/microbiología , Gastritis/epidemiología , Gastritis/inmunología , Gastritis/microbiología , Humanos , Prevalencia , Proctocolitis/epidemiología , Proctocolitis/inmunología , Proctocolitis/microbiología , Proteobacteria/inmunología , Proteobacteria/aislamiento & purificación
11.
Sci Rep ; 10(1): 2232, 2020 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-32042047

RESUMEN

Microbial dysbiosis has long been postulated to be associated with the pathogenesis of inflammatory bowel disease (IBD). Although evidence supporting the anti-colitic effects of melatonin have been accumulating, it is not clear how melatonin affects the microbiota. Herein, we investigated the effects of melatonin on the microbiome in colitis and identified involvement of Toll-like receptor (TLR) 4 signalling in the effects. Melatonin improved dextran sulfate sodium (DSS)-induced colitis and reverted microbial dysbiosis in wild-type (WT) mice but not in TLR4 knockout (KO) mice. Induction of goblet cells was observed with melatonin administration, which was accompanied by suppression of Il1b and Il17a and induction of melatonin receptor and Reg3ß, an antimicrobial peptide (AMP) against Gram-negative bacteria. In vitro, melatonin treatment of HT-29 intestinal epithelial cells promotes mucin and wound healing and inhibits growth of Escherichia coli. Herein, we showed that melatonin significantly increases goblet cells, Reg3ß, and the ratio of Firmicutes to Bacteriodetes by suppressing Gram-negative bacteria through TLR4 signalling. Our study suggests that sensing of bacteria through TLR4 and regulation of bacteria through altered goblet cells and AMPs is involved in the anti-colitic effects of melatonin. Melatonin may have use in therapeutics for IBD.


Asunto(s)
Colitis Ulcerosa/tratamiento farmacológico , Disbiosis/tratamiento farmacológico , Microbioma Gastrointestinal/efectos de los fármacos , Células Caliciformes/efectos de los fármacos , Melatonina/administración & dosificación , Receptor Toll-Like 4/metabolismo , Animales , Bacteroidetes/efectos de los fármacos , Bacteroidetes/inmunología , Bacteroidetes/aislamiento & purificación , Diferenciación Celular/efectos de los fármacos , Colitis Ulcerosa/inducido químicamente , Colitis Ulcerosa/inmunología , Colitis Ulcerosa/microbiología , Sulfato de Dextran/toxicidad , Modelos Animales de Enfermedad , Disbiosis/inmunología , Disbiosis/microbiología , Disbiosis/patología , Firmicutes/efectos de los fármacos , Firmicutes/inmunología , Firmicutes/aislamiento & purificación , Microbioma Gastrointestinal/inmunología , Células Caliciformes/inmunología , Células Caliciformes/microbiología , Células Caliciformes/fisiología , Células HT29 , Humanos , Masculino , Ratones , Ratones Noqueados , Proteínas Asociadas a Pancreatitis/inmunología , Proteínas Asociadas a Pancreatitis/metabolismo , Receptores de Melatonina/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Receptor Toll-Like 4/genética
12.
Nat Microbiol ; 5(2): 304-313, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31907407

RESUMEN

The microbiota primes immune defences but the identity of specific commensal microorganisms that protect against infection is unclear. Conversely, how pathogens compete with the microbiota to establish their host niche is also poorly understood. In the present study, we investigate the antagonism between the microbiota and Klebsiella pneumoniae during colonization and transmission. We discover that maturation of the microbiota drives the development of distinct immune defence programmes in the upper airways and intestine to limit K. pneumoniae colonization within these niches. Immune protection in the intestine depends on the development of Bacteroidetes, interleukin (IL)-36 signalling and macrophages. This effect of Bacteroidetes requires the polysaccharide utilization locus of their conserved commensal colonization factor. Conversely, in the upper airways, Proteobacteria prime immunity through IL-17A, but K. pneumoniae overcomes these defences through encapsulation to effectively colonize this site. Ultimately, we find that host-to-host spread of K. pneumoniae occurs principally from its intestinal reservoir, and that commensal-colonization-factor-producing Bacteroidetes are sufficient to prevent transmission between hosts through IL-36. Thus, our study provides mechanistic insight into when, where and how commensal Bacteroidetes protect against K. pneumoniae colonization and contagion, providing insight into how these protective microorganisms could be harnessed to confer population-level protection against K. pneumoniae infection.


Asunto(s)
Bacteroidetes/inmunología , Interleucina-1/inmunología , Infecciones por Klebsiella/prevención & control , Klebsiella pneumoniae , Microbiota/inmunología , Animales , Animales Recién Nacidos , Microbioma Gastrointestinal/inmunología , Interacciones Microbiota-Huesped/inmunología , Interleucina-17/inmunología , Infecciones por Klebsiella/inmunología , Infecciones por Klebsiella/transmisión , Klebsiella pneumoniae/patogenicidad , Ratones , Modelos Biológicos , Sistema Respiratorio/inmunología , Sistema Respiratorio/microbiología , Transducción de Señal/inmunología
13.
J Diabetes Complications ; 34(2): 107449, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31677982

RESUMEN

AIM: The aim of this study was to determine and compare the levels of both Bacteroidetes and Firmicutes in the gut microbiota and TLR2/TLR4 gene expression in the blood of patients with type 1 diabetes mellitus (T1DM) and healthy individuals. These results may serve as a preliminary assessment to guide future research. METHOD: Between January and October 2014, stool and blood samples were collected from 53 adult T1DM patients and 53 age- and gender-matched healthy individuals. Bacteroidetes and Firmicutes levels were assessed from stool sample DNA and TLR2 and TLR4 expression levels were analyzed via qPCR using RNA from EDTA blood samples from both patients and healthy controls. RESULTS: The amounts of Bacteroidetes and Firmicutes were statistically significantly higher and lower, respectively, in the T1DM group than in the healthy control group (p < 0.001 and p < 0.001, respectively). In addition, the Firmicutes/Bacteroidetes ratios in patients with T1DM were significantly lower than in healthy controls. The TLR4 and TLR2 gene expression levels in T1DM patients were significantly upregulated and downregulated, respectively, compared to those in the control group. CONCLUSION: Our data are the first to show a relationship between T1DM and gut microbiota in our country. In addition, our results provide information about the connections between T1DM, gut microbiota, and TLR2 and TLR4 expression. We believe that Bacteroidetes and Firmicutes in the gut microbiota may play a role in the autoimmune process of T1DM and that these findings should be further investigated in the future.


Asunto(s)
Bacteroidetes/inmunología , Diabetes Mellitus Tipo 1 , Firmicutes/inmunología , Microbioma Gastrointestinal , Receptor Toll-Like 2/genética , Receptor Toll-Like 4/genética , Adulto , Bacteroidetes/aislamiento & purificación , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/microbiología , Femenino , Firmicutes/aislamiento & purificación , Microbioma Gastrointestinal/genética , Microbioma Gastrointestinal/inmunología , Tracto Gastrointestinal/inmunología , Tracto Gastrointestinal/microbiología , Expresión Génica , Humanos , Masculino , Persona de Mediana Edad , Receptor Toll-Like 2/inmunología , Receptor Toll-Like 4/inmunología , Turquía , Adulto Joven
14.
Nature ; 575(7781): 224-228, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31666699

RESUMEN

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.


Asunto(s)
Bacteroidetes , Microbioma Gastrointestinal , Tracto Gastrointestinal/microbiología , Interacciones Microbianas , Sistemas de Secreción Tipo VI/antagonistas & inhibidores , Animales , Bacteroidetes/genética , Bacteroidetes/inmunología , Femenino , Microbioma Gastrointestinal/inmunología , Tracto Gastrointestinal/inmunología , Genes Bacterianos/genética , Humanos , Ratones , Interacciones Microbianas/genética , Interacciones Microbianas/inmunología , Familia de Multigenes/genética , Sistemas de Secreción Tipo VI/genética , Sistemas de Secreción Tipo VI/inmunología
15.
Front Immunol ; 10: 1772, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31417552

RESUMEN

The active form of vitamin D (1,25(OH)2D) suppresses experimental models of inflammatory bowel disease in part by regulating the microbiota. In this study, the role of vitamin D in the regulation of microbe induced RORγt/FoxP3+ T regulatory (reg) cells in the colon was determined. Vitamin D sufficient (D+) mice had significantly higher frequencies of FoxP3+ and RORγt/FoxP3+ T reg cells in the colon compared to vitamin D deficient (D-) mice. The higher frequency of RORγt/FoxP3+ T reg cells in D+ colon correlated with higher numbers of bacteria from the Clostridium XIVa and Bacteroides in D+ compared to D- cecum. D- mice with fewer RORγt/FoxP3+ T reg cells were significantly more susceptible to colitis than D+ mice. Transfer of the cecal bacteria from D+ or D- mice to germfree recipients phenocopied the higher numbers of RORγt/FoxP3+ cells and reduced susceptibility to colitis in D+ vs. D- recipient mice. 1,25(OH)2D treatment of the D- mice beginning at 3 weeks of age did not completely recover RORγt/FoxP3+ T reg cells or the Bacteriodes, Bacteriodes thetaiotaomicron, and Clostridium XIVa numbers to D+ values. Early vitamin D status shapes the microbiota to optimize the population of colonic RORγt/FoxP3+ T reg cells important for resistance to colitis.


Asunto(s)
Calcitriol/farmacología , Colitis , Colon , Microbioma Gastrointestinal , Linfocitos T Reguladores/inmunología , Animales , Bacteroidetes/inmunología , Clostridium/inmunología , Colitis/inmunología , Colitis/microbiología , Colitis/patología , Colon/inmunología , Colon/microbiología , Colon/patología , Factores de Transcripción Forkhead/inmunología , Microbioma Gastrointestinal/efectos de los fármacos , Microbioma Gastrointestinal/inmunología , Ratones , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/inmunología , Linfocitos T Reguladores/patología
16.
Infect Immun ; 87(10)2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31308086

RESUMEN

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.


Asunto(s)
Bordetella pertussis/inmunología , Disbiosis/inmunología , Microbioma Gastrointestinal/inmunología , Inmunidad Humoral , Simbiosis/inmunología , Tos Ferina/inmunología , Ampicilina/farmacología , Animales , Antibacterianos/farmacología , Anticuerpos Antibacterianos/biosíntesis , Anticuerpos Antibacterianos/clasificación , Bacteroidetes/clasificación , Bacteroidetes/efectos de los fármacos , Bacteroidetes/crecimiento & desarrollo , Bacteroidetes/inmunología , Bordetella pertussis/crecimiento & desarrollo , Bordetella pertussis/patogenicidad , Disbiosis/microbiología , Disbiosis/fisiopatología , Femenino , Firmicutes/clasificación , Firmicutes/efectos de los fármacos , Firmicutes/crecimiento & desarrollo , Firmicutes/inmunología , Microbioma Gastrointestinal/efectos de los fármacos , Inmunidad Innata , Inmunoglobulina G/biosíntesis , Inmunoglobulina G/clasificación , Metronidazol/farmacología , Ratones , Ratones Endogámicos BALB C , Neomicina/farmacología , Proteobacteria/clasificación , Proteobacteria/efectos de los fármacos , Proteobacteria/crecimiento & desarrollo , Proteobacteria/inmunología , Simbiosis/efectos de los fármacos , Vancomicina/farmacología , Tos Ferina/microbiología , Tos Ferina/fisiopatología
17.
Clin Exp Allergy ; 49(2): 227-238, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30312497

RESUMEN

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.


Asunto(s)
Asma , Bacteroidetes , Vesículas Extracelulares , Proteobacteria , Pyroglyphidae , Linfocitos T Colaboradores-Inductores , Animales , Asma/metabolismo , Asma/microbiología , Asma/patología , Bacteroidetes/genética , Bacteroidetes/inmunología , Modelos Animales de Enfermedad , Vesículas Extracelulares/química , Vesículas Extracelulares/inmunología , Vesículas Extracelulares/microbiología , Metagenoma , Ratones , Ratones Noqueados , Proteobacteria/genética , Proteobacteria/inmunología , Pyroglyphidae/química , Pyroglyphidae/inmunología , Pyroglyphidae/microbiología , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Colaboradores-Inductores/patología
18.
Mucosal Immunol ; 11(6): 1591-1605, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30115998

RESUMEN

Human mucosal-associated invariant T (MAIT) cell receptors (TCRs) recognize bacterial riboflavin pathway metabolites through the MHC class 1-related molecule MR1. However, it is unclear whether MAIT cells discriminate between many species of the human microbiota. To address this, we developed an in vitro functional assay through human T cells engineered for MAIT-TCRs (eMAIT-TCRs) stimulated by MR1-expressing antigen-presenting cells (APCs). We then screened 47 microbiota-associated bacterial species from different phyla for their eMAIT-TCR stimulatory capacities. Only bacterial species that encoded the riboflavin pathway were stimulatory for MAIT-TCRs. Most species that were high stimulators belonged to Bacteroidetes and Proteobacteria phyla, whereas low/non-stimulator species were primarily Actinobacteria or Firmicutes. Activation of MAIT cells by high- vs low-stimulating bacteria also correlated with the level of riboflavin they secreted or after bacterial infection of macrophages. Remarkably, we found that human T-cell subsets can also present riboflavin metabolites to MAIT cells in a MR1-restricted fashion. This T-T cell-mediated signaling also induced IFNγ, TNF and granzyme B from MAIT cells, albeit at lower level than professional APC. These findings suggest that MAIT cells can discriminate and categorize complex human microbiota through computation of TCR signals depending on antigen load and presenting cells, and fine-tune their functional responses.


Asunto(s)
Bacteroidetes/inmunología , Macrófagos/inmunología , Microbiota/inmunología , Células T Invariantes Asociadas a Mucosa/inmunología , Proteobacteria/inmunología , Riboflavina/metabolismo , Presentación de Antígeno , Antígenos Bacterianos/inmunología , Células Cultivadas , Ingeniería Genética , Antígenos de Histocompatibilidad Clase I/metabolismo , Humanos , Interferón gamma/metabolismo , Activación de Linfocitos , Macrófagos/microbiología , Antígenos de Histocompatibilidad Menor/metabolismo , Células T Invariantes Asociadas a Mucosa/microbiología , Receptores de Antígenos de Linfocitos T/genética , Transducción de Señal , Factor de Necrosis Tumoral alfa/metabolismo
19.
Appl Microbiol Biotechnol ; 102(21): 9317-9329, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30151605

RESUMEN

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.


Asunto(s)
Ciego/inmunología , Ciego/microbiología , Microbioma Gastrointestinal/inmunología , Inmunidad Innata/genética , Intestinos/inmunología , Aves de Corral/inmunología , Alimentación Animal/microbiología , Animales , Bacteroidetes/inmunología , Dieta , Firmicutes/inmunología , Microbioma Gastrointestinal/genética , Expresión Génica/genética , Expresión Génica/inmunología , Inmunidad Innata/inmunología , Intestinos/microbiología , Filogenia , Aves de Corral/genética , Aves de Corral/microbiología , Probióticos , Proteínas de Uniones Estrechas/metabolismo , Receptores Toll-Like/metabolismo
20.
J Autoimmun ; 93: 57-65, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29960834

RESUMEN

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.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/genética , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/microbiología , Microbioma Gastrointestinal/inmunología , Factor 88 de Diferenciación Mieloide/genética , Linfocitos T/inmunología , Proteínas Adaptadoras del Transporte Vesicular/deficiencia , Proteínas Adaptadoras del Transporte Vesicular/inmunología , Traslado Adoptivo , Animales , Bacteroidetes/inmunología , Burkholderiales/inmunología , Proliferación Celular , Células Dendríticas/inmunología , Células Dendríticas/patología , Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Experimental/patología , Susceptibilidad a Enfermedades , Femenino , Firmicutes/inmunología , Regulación de la Expresión Génica , Activación de Linfocitos , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/inmunología , Transducción de Señal , Linfocitos T/patología , Linfocitos T/trasplante , Receptor Toll-Like 3/genética , Receptor Toll-Like 3/inmunología , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...