Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Gut Microbes ; 16(1): 2338947, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38717824

RESUMEN

The gut microbiota has coevolved with the host for hundreds of millions of years, playing a beneficial role in host health. Human parasitic helminths are widespread and pose a pervasive global public health issue. Although Type 2 immunity provides partial resistance to helminth infections, the composition of the gut microbiota can change correspondingly. Therefore, it raises the question of what role the gut microbiota plays during helminth infection. Akkermansia muciniphila has emerged as a notable representative of beneficial microorganisms in the gut microbiota. Recent studies indicate that A. muciniphila is not merely associated with helminth infection but is also causally linked to infection. Here, we provide an overview of the crosstalk between A. muciniphila and enteric helminth infection. Our goal is to enhance our understanding of the interplay among A. muciniphila, helminths, and their hosts while also exploring the potential underlying mechanisms.


Asunto(s)
Akkermansia , Microbioma Gastrointestinal , Animales , Humanos , Helmintiasis/inmunología , Helmintos/inmunología , Helmintos/genética , Verrucomicrobia/genética , Verrucomicrobia/inmunología
2.
Proc Natl Acad Sci U S A ; 117(40): 24998-25007, 2020 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-32958643

RESUMEN

Infections elicit immune adaptations to enable pathogen resistance and/or tolerance and are associated with compositional shifts of the intestinal microbiome. However, a comprehensive understanding of how infections with pathogens that exhibit distinct capability to spread and/or persist differentially change the microbiome, the underlying mechanisms, and the relative contribution of individual commensal species to immune cell adaptations is still lacking. Here, we discovered that mouse infection with a fast-spreading and persistent (but not a slow-spreading acute) isolate of lymphocytic choriomeningitis virus induced large-scale microbiome shifts characterized by increased Verrucomicrobia and reduced Firmicute/Bacteroidetes ratio. Remarkably, the most profound microbiome changes occurred transiently after infection with the fast-spreading persistent isolate, were uncoupled from sustained viral loads, and were instead largely caused by CD8 T cell responses and/or CD8 T cell-induced anorexia. Among the taxa enriched by infection with the fast-spreading virus, Akkermansia muciniphila, broadly regarded as a beneficial commensal, bloomed upon starvation and in a CD8 T cell-dependent manner. Strikingly, oral administration of A. muciniphila suppressed selected effector features of CD8 T cells in the context of both infections. Our findings define unique microbiome differences after chronic versus acute viral infections and identify CD8 T cell responses and downstream anorexia as driver mechanisms of microbial dysbiosis after infection with a fast-spreading virus. Our data also highlight potential context-dependent effects of probiotics and suggest a model in which changes in host behavior and downstream microbiome dysbiosis may constitute a previously unrecognized negative feedback loop that contributes to CD8 T cell adaptations after infections with fast-spreading and/or persistent pathogens.


Asunto(s)
Anorexia/inmunología , Antígenos CD8/inmunología , Memoria Inmunológica/inmunología , Coriomeningitis Linfocítica/inmunología , Virosis/inmunología , Akkermansia , Animales , Anorexia/microbiología , Anorexia/virología , Antígenos CD8/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/microbiología , Disbiosis/inmunología , Disbiosis/microbiología , Disbiosis/virología , Firmicutes/inmunología , Firmicutes/metabolismo , Microbioma Gastrointestinal/inmunología , Humanos , Coriomeningitis Linfocítica/microbiología , Coriomeningitis Linfocítica/patología , Virus de la Coriomeningitis Linfocítica/patogenicidad , Ratones , Linfocitos T/inmunología , Linfocitos T/microbiología , Verrucomicrobia/inmunología , Verrucomicrobia/patogenicidad , Virosis/microbiología , Virosis/patología
3.
Cell Host Microbe ; 26(6): 779-794.e8, 2019 12 11.
Artículo en Inglés | MEDLINE | ID: mdl-31784260

RESUMEN

Fecal transfer from healthy donors is being explored as a microbiome modality. MicroRNAs (miRNAs) have been found to affect the microbiome. Multiple sclerosis (MS) patients have been shown to have an altered gut microbiome. Here, we unexpectedly found that transfer of feces harvested at peak disease from the experimental autoimmune encephalomyelitis (EAE) model of MS ameliorates disease in recipients in a miRNA-dependent manner. Specifically, we show that miR-30d is enriched in the feces of peak EAE and untreated MS patients. Synthetic miR-30d given orally ameliorates EAE through expansion of regulatory T cells (Tregs). Mechanistically, miR-30d regulates the expression of a lactase in Akkermansia muciniphila, which increases Akkermansia abundance in the gut. The expanded Akkermansia in turn increases Tregs to suppress EAE symptoms. Our findings report the mechanistic underpinnings of a miRNA-microbiome axis and suggest that the feces of diseased subjects might be enriched with miRNAs with therapeutic properties.


Asunto(s)
Encefalomielitis Autoinmune Experimental , Trasplante de Microbiota Fecal , MicroARNs/uso terapéutico , Esclerosis Múltiple/tratamiento farmacológico , Verrucomicrobia , Administración Oral , Akkermansia , Animales , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/inmunología , Heces , Microbioma Gastrointestinal/inmunología , Interacciones Microbiota-Huesped , Humanos , Lactasa/metabolismo , Ratones , Ratones Endogámicos C57BL , MicroARNs/metabolismo , Linfocitos T Reguladores/metabolismo , Verrucomicrobia/crecimiento & desarrollo , Verrucomicrobia/inmunología , Verrucomicrobia/metabolismo
4.
Nat Commun ; 10(1): 5711, 2019 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-31836714

RESUMEN

In order to improve targeted therapeutic approaches for asthma patients, insights into the molecular mechanisms that differentially contribute to disease phenotypes, such as obese asthmatics or severe asthmatics, are required. Here we report immunological and microbiome alterations in obese asthmatics (n = 50, mean age = 45), non-obese asthmatics (n = 53, mean age = 40), obese non-asthmatics (n = 51, mean age = 44) and their healthy counterparts (n = 48, mean age = 39). Obesity is associated with elevated proinflammatory signatures, which are enhanced in the presence of asthma. Similarly, obesity or asthma induced changes in the composition of the microbiota, while an additive effect is observed in obese asthma patients. Asthma disease severity is negatively correlated with fecal Akkermansia muciniphila levels. Administration of A. muciniphila to murine models significantly reduces airway hyper-reactivity and airway inflammation. Changes in immunological processes and microbiota composition are accentuated in obese asthma patients due to the additive effects of both disease states, while A. muciniphila may play a non-redundant role in patients with a severe asthma phenotype.


Asunto(s)
Asma/inmunología , Microbioma Gastrointestinal/inmunología , Interacciones Microbiota-Huesped/inmunología , Obesidad/inmunología , Verrucomicrobia/inmunología , Adulto , Akkermansia , Animales , Asma/complicaciones , Asma/diagnóstico , Asma/microbiología , Modelos Animales de Enfermedad , Heces/microbiología , Femenino , Volumen Espiratorio Forzado , Voluntarios Sanos , Humanos , Masculino , Ratones , Persona de Mediana Edad , Obesidad/complicaciones , Obesidad/microbiología , Sistema Respiratorio/inmunología , Índice de Severidad de la Enfermedad , Verrucomicrobia/aislamiento & purificación
5.
Science ; 364(6446): 1179-1184, 2019 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-31221858

RESUMEN

Intestinal adaptive immune responses influence host health, yet only a few intestinal bacteria species that induce cognate adaptive immune responses during homeostasis have been identified. Here, we show that Akkermansia muciniphila, an intestinal bacterium associated with systemic effects on host metabolism and PD-1 checkpoint immunotherapy, induces immunoglobulin G1 (IgG1) antibodies and antigen-specific T cell responses in mice. Unlike previously characterized mucosal responses, T cell responses to A. muciniphila are limited to T follicular helper cells in a gnotobiotic setting, without appreciable induction of other T helper fates or migration to the lamina propria. However, A. muciniphila-specific responses are context dependent and adopt other fates in conventional mice. These findings suggest that, during homeostasis, contextual signals influence T cell responses to the microbiota and modulate host immune function.


Asunto(s)
Inmunidad Adaptativa , Microbioma Gastrointestinal/inmunología , Homeostasis , Intestinos/inmunología , Verrucomicrobia/inmunología , Animales , Movimiento Celular/inmunología , Femenino , Vida Libre de Gérmenes , Inmunidad Mucosa , Inmunoglobulina G/inmunología , Mucosa Intestinal/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Linfocitos T Colaboradores-Inductores/inmunología
6.
Microb Biotechnol ; 12(6): 1109-1125, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31006995

RESUMEN

Akkermansia muciniphila (A. muciniphila), an intestinal symbiont colonizing in the mucosal layer, is considered to be a promising candidate as probiotics. A. muciniphila is known to have an important value in improving the host metabolic functions and immune responses. Moreover, A. muciniphila may have a value in modifying cancer treatment. However, most of the current researches focus on the correlation between A. muciniphila and diseases, and little is known about the causal relationship between them. Few intervention studies on A. muciniphila are limited to animal experiments, and limited studies have explored its safety and efficacy in humans. Therefore, a critical analysis of the current knowledge in A. muciniphila will play an important foundation for it to be defined as a new beneficial microbe. This article will review the bacteriological characteristics and safety of A. muciniphila, as well as its causal relationship with metabolic disorders, immune diseases and cancer therapy.


Asunto(s)
Probióticos/administración & dosificación , Verrucomicrobia/inmunología , Verrucomicrobia/metabolismo , Akkermansia , Animales , Humanos , Factores Inmunológicos/administración & dosificación , Mucosa Intestinal/microbiología
7.
Sci Rep ; 8(1): 12072, 2018 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-30104612

RESUMEN

In spite of the extensive contribution of intestinal pathology to the pathophysiology of schistosomiasis, little is known of the impact of schistosome infection on the composition of the gut microbiota of its mammalian host. Here, we characterised the fluctuations in the composition of the gut microbial flora of the small and large intestine, as well as the changes in abundance of individual microbial species, of mice experimentally infected with Schistosoma mansoni with the goal of identifying microbial taxa with potential roles in the pathophysiology of infection and disease. Bioinformatic analyses of bacterial 16S rRNA gene data revealed an overall reduction in gut microbial alpha diversity, alongside a significant increase in microbial beta diversity characterised by expanded populations of Akkermansia muciniphila (phylum Verrucomicrobia) and lactobacilli, in the gut microbiota of S. mansoni-infected mice when compared to uninfected control animals. These data support a role of the mammalian gut microbiota in the pathogenesis of hepato-intestinal schistosomiasis and serves as a foundation for the design of mechanistic studies to unravel the complex relationships amongst parasitic helminths, gut microbiota, pathophysiology of infection and host immunity.


Asunto(s)
Microbioma Gastrointestinal/inmunología , Intestinos/microbiología , Schistosoma mansoni/patogenicidad , Esquistosomiasis mansoni/inmunología , Animales , Bacteroides/genética , Bacteroides/inmunología , Bacteroides/aislamiento & purificación , ADN Bacteriano/aislamiento & purificación , Modelos Animales de Enfermedad , Femenino , Microbioma Gastrointestinal/genética , Humanos , Intestinos/patología , Lactobacillus/genética , Lactobacillus/inmunología , Lactobacillus/aislamiento & purificación , Ratones , ARN Ribosómico 16S/genética , Schistosoma mansoni/inmunología , Esquistosomiasis mansoni/microbiología , Esquistosomiasis mansoni/parasitología , Esquistosomiasis mansoni/patología , Verrucomicrobia/genética , Verrucomicrobia/inmunología , Verrucomicrobia/aislamiento & purificación
8.
Gut ; 67(9): 1716-1725, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29934437

RESUMEN

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.


Asunto(s)
Microbioma Gastrointestinal/inmunología , Enfermedades Metabólicas/inmunología , Enfermedades Metabólicas/microbiología , Diabetes Mellitus/inmunología , Diabetes Mellitus/microbiología , Disbiosis/prevención & control , Medicina Basada en la Evidencia , Humanos , Hepatopatías/inmunología , Hepatopatías/microbiología , Enfermedades Metabólicas/metabolismo , Enfermedades Metabólicas/prevención & control , Neoplasias/inmunología , Neoplasias/microbiología , Enfermedades Neurodegenerativas/inmunología , Enfermedades Neurodegenerativas/microbiología , Obesidad/inmunología , Obesidad/microbiología , Prevotella/inmunología , Verrucomicrobia/inmunología
9.
Science ; 359(6371): 91-97, 2018 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-29097494

RESUMEN

Immune checkpoint inhibitors (ICIs) targeting the PD-1/PD-L1 axis induce sustained clinical responses in a sizable minority of cancer patients. We found that primary resistance to ICIs can be attributed to abnormal gut microbiome composition. Antibiotics inhibited the clinical benefit of ICIs in patients with advanced cancer. Fecal microbiota transplantation (FMT) from cancer patients who responded to ICIs into germ-free or antibiotic-treated mice ameliorated the antitumor effects of PD-1 blockade, whereas FMT from nonresponding patients failed to do so. Metagenomics of patient stool samples at diagnosis revealed correlations between clinical responses to ICIs and the relative abundance of Akkermansia muciniphila Oral supplementation with A. muciniphila after FMT with nonresponder feces restored the efficacy of PD-1 blockade in an interleukin-12-dependent manner by increasing the recruitment of CCR9+CXCR3+CD4+ T lymphocytes into mouse tumor beds.


Asunto(s)
Trasplante de Microbiota Fecal , Microbioma Gastrointestinal/inmunología , Inmunoterapia/métodos , Neoplasias/terapia , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Animales , Antibacterianos/uso terapéutico , Anticuerpos Monoclonales/uso terapéutico , Antígenos CD4/inmunología , Heces/microbiología , Microbioma Gastrointestinal/genética , Humanos , Interleucina-12/inmunología , Metagenoma/genética , Ratones , Receptores CCR/inmunología , Receptores CXCR3/inmunología , Linfocitos T/inmunología , Verrucomicrobia/genética , Verrucomicrobia/inmunología
11.
PLoS One ; 12(3): e0173004, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28249045

RESUMEN

Gut barrier function is key in maintaining a balanced response between the host and its microbiome. The microbiota can modulate changes in gut barrier as well as metabolic and inflammatory responses. This highly complex system involves numerous microbiota-derived factors. The gut symbiont Akkermansia muciniphila is positively correlated with a lean phenotype, reduced body weight gain, amelioration of metabolic responses and restoration of gut barrier function by modulation of mucus layer thickness. However, the molecular mechanisms behind its metabolic and immunological regulatory properties are unexplored. Herein, we identify a highly abundant outer membrane pili-like protein of A. muciniphila MucT that is directly involved in immune regulation and enhancement of trans-epithelial resistance. The purified Amuc_1100 protein and enrichments containing all its associated proteins induced production of specific cytokines through activation of Toll-like receptor (TLR) 2 and TLR4. This mainly leads to high levels of IL-10 similar to those induced by the other beneficial immune suppressive microorganisms such as Faecalibacterium prausnitzii A2-165 and Lactobacillus plantarum WCFS1. Together these results indicate that outer membrane protein composition and particularly the newly identified highly abundant pili-like protein Amuc_1100 of A. muciniphila are involved in host immunological homeostasis at the gut mucosa, and improvement of gut barrier function.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/inmunología , Mucosa Intestinal/inmunología , Verrucomicrobia/inmunología , Proteínas de la Membrana Bacteriana Externa/genética , Línea Celular , Células Cultivadas , Citocinas/genética , Citocinas/metabolismo , Microbioma Gastrointestinal , Humanos , Mucosa Intestinal/microbiología , Receptor Toll-Like 2/genética , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo , Verrucomicrobia/patogenicidad
12.
Sci Rep ; 6: 39026, 2016 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-27976725

RESUMEN

Age-associated immunological dysfunction (immunosenescence) is closely linked to perturbation of the gut microbiota. Here, we investigated whether syringaresinol (SYR), a polyphenolic lignan, modulates immune aging and the gut microbiota associated with this effect in middle-aged mice. Compared with age-matched control mice, SYR treatment delayed immunosenescence by enhancing the numbers of total CD3+ T cells and naïve T cells. SYR treatment induced the expression of Bim as well as activation of FOXO3 in Foxp3+ regulatory T cells (Tregs). Furthermore, SYR treatment significantly enhanced the Firmicutes/Bacteroidetes ratio compared with that in age-matched controls by increasing beneficial bacteria, Lactobacillus and Bifidobacterium, while reducing the opportunistic pathogenic genus, Akkermansia. In addition, SYR treatment reduced the serum level of lipopolysaccharide-binding protein, an inflammatory marker, and enhanced humoral immunity against influenza vaccination to the level of young control mice. Taken together, these findings suggest that SYR may rejuvenate the immune system through modulation of gut integrity and microbiota diversity as well as composition in middle-aged mice, which may delay the immunosenescence associated with aging.


Asunto(s)
Envejecimiento/inmunología , Furanos/farmacología , Microbioma Gastrointestinal/efectos de los fármacos , Inmunosenescencia/efectos de los fármacos , Lignanos/farmacología , Animales , Área Bajo la Curva , Bifidobacterium/efectos de los fármacos , Bifidobacterium/inmunología , Bifidobacterium/fisiología , Complejo CD3/inmunología , Complejo CD3/metabolismo , Femenino , Proteína Forkhead Box O3/inmunología , Proteína Forkhead Box O3/metabolismo , Furanos/farmacocinética , Microbioma Gastrointestinal/inmunología , Microbioma Gastrointestinal/fisiología , Inmunosenescencia/inmunología , Lactobacillus/efectos de los fármacos , Lactobacillus/inmunología , Lactobacillus/fisiología , Lignanos/farmacocinética , Masculino , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Ratas Sprague-Dawley , Subgrupos de Linfocitos T/efectos de los fármacos , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Verrucomicrobia/efectos de los fármacos , Verrucomicrobia/inmunología , Verrucomicrobia/fisiología
13.
Microbiome ; 4: 17, 2016 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-27114075

RESUMEN

BACKGROUND: The development of anti-islet cell autoimmunity precedes clinical type 1 diabetes and occurs very early in life. During this early period, dietary factors strongly impact on the composition of the gut microbiome. At the same time, the gut microbiome plays a central role in the development of the infant immune system. A functional model of the association between diet, microbial communities, and the development of anti-islet cell autoimmunity can provide important new insights regarding the role of the gut microbiome in the pathogenesis of type 1 diabetes. RESULTS: A novel approach was developed to enable the analysis of the microbiome on an aggregation level between a single microbial taxon and classical ecological measures analyzing the whole microbial population. Microbial co-occurrence networks were estimated at age 6 months to identify candidates for functional microbial communities prior to islet autoantibody development. Stratification of children based on these communities revealed functional associations between diet, gut microbiome, and islet autoantibody development. Two communities were strongly associated with breast-feeding and solid food introduction, respectively. The third community revealed a subgroup of children that was dominated by Bacteroides abundances compared to two subgroups with low Bacteroides and increased Akkermansia abundances. The Bacteroides-dominated subgroup was characterized by early introduction of non-milk diet, increased risk for early autoantibody development, and by lower abundances of genes for the production of butyrate via co-fermentation of acetate. By combining our results with information from the literature, we provide a refined functional hypothesis for a protective role of butyrate in the pathogenesis of type 1 diabetes. CONCLUSIONS: Based on functional traits of microbial communities estimated from co-occurrence networks, we provide evidence that alterations in the composition of mucin degrading bacteria associate with early development of anti-islet cell autoimmunity. We hypothesize that lower levels of Bacteroides in favor of increased levels of Akkermansia lead to a competitive advantage of acetogens compared to sulfate reducing bacteria, resulting in increased butyrate production via co-fermentation of acetate. This hypothesis suggests that butyrate has a protective effect on the development of anti-islet cell autoantibodies.


Asunto(s)
Bacteroides/metabolismo , Ácido Butírico/metabolismo , Diabetes Mellitus Tipo 1/microbiología , Microbioma Gastrointestinal/inmunología , Tracto Gastrointestinal/microbiología , Verrucomicrobia/metabolismo , Ácido Acético/inmunología , Ácido Acético/metabolismo , Adulto , Autoanticuerpos/biosíntesis , Autoinmunidad , Bacteroides/inmunología , Lactancia Materna , Ácido Butírico/inmunología , Niño , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/patología , Dieta , Femenino , Fermentación , Tracto Gastrointestinal/inmunología , Humanos , Inmunidad Innata , Lactante , Islotes Pancreáticos/inmunología , Masculino , Verrucomicrobia/inmunología
14.
Appl Environ Microbiol ; 81(11): 3655-62, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25795669

RESUMEN

Akkermansia muciniphila is a Gram-negative mucin-degrading bacterium that resides in the gastrointestinal tracts of humans and animals. A. muciniphila has been linked with intestinal health and improved metabolic status in obese and type 2 diabetic subjects. Specifically, A. muciniphila has been shown to reduce high-fat-diet-induced endotoxemia, which develops as a result of an impaired gut barrier. Despite the accumulating evidence of the health-promoting effects of A. muciniphila, the mechanisms of interaction of the bacterium with the host have received little attention. In this study, we used several in vitro models to investigate the adhesion of A. muciniphila to the intestinal epithelium and its interaction with the host mucosa. We found that A. muciniphila adheres strongly to the Caco-2 and HT-29 human colonic cell lines but not to human colonic mucus. In addition, A. muciniphila showed binding to the extracellular matrix protein laminin but not to collagen I or IV, fibronectin, or fetuin. Importantly, A. muciniphila improved enterocyte monolayer integrity, as shown by a significant increase in the transepithelial electrical resistance (TER) of cocultures of Caco-2 cells with the bacterium. Further, A. muciniphila induced interleukin 8 (IL-8) production by enterocytes at cell concentrations 100-fold higher than those for Escherichia coli, suggesting a very low level of proinflammatory activity in the epithelium. In conclusion, our results demonstrate that A. muciniphila adheres to the intestinal epithelium and strengthens enterocyte monolayer integrity in vitro, suggesting an ability to fortify an impaired gut barrier. These results support earlier associative in vivo studies and provide insights into the interaction of A. muciniphila with the host.


Asunto(s)
Adhesión Bacteriana , Enterocitos/microbiología , Células Epiteliales/fisiología , Verrucomicrobia/fisiología , Línea Celular , Enterocitos/inmunología , Enterocitos/metabolismo , Humanos , Interleucina-8/metabolismo , Verrucomicrobia/inmunología
15.
Benef Microbes ; 3(1): 43-50, 2012 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-22348908

RESUMEN

Enterococcus faecium NCIMB 10415 reduces diarrhoea incidence and duration in animals and human study subjects. We tested whether the strain is also capable of reducing chronic gut inflammation and aimed to identify mechanisms that are involved in possible probiotic effects. To identify health-promoting mechanisms of the strain, we used interleukin-10-deficient mice that spontaneously develop gut inflammation and fed these mice a diet containing NCIMB 10415 for 3, 8 and 24 weeks, respectively. Control mice were fed a diet which was identically composed but did not contain the strain. After 3 weeks of intervention the experimental animals were less inflamed in the caecum than the control animals. This effect was not observed in the colon and there were no differences between experimental and control mice at any other time point. The application of the strain was associated with higher expression levels of interferon gamma and interferon gamma-induced protein 10 after 3 and 24 but not after 8 weeks of feeding. No differences between the animals were observed in intestinal barrier function or intestinal microbiota composition. However, we observed a low abundance of the mucin-degrading bacterium Akkermansia muciniphila in the mice that were fed NCIMB 10415 for 8 weeks. These low cell numbers were associated with a significantly lower caecal inflammation score and improved paracellular permeability as compared to the NCIMB-treated mice that were killed after 3 and 24 weeks of intervention. In conclusion, NCIMB 10415 is not capable of reducing gut inflammation in the IL-10-/- mouse model. The exact role of A. muciniphila and of a possible interaction between this bacterium, NCIMB 10415 and the host in gut inflammation requires further investigation.


Asunto(s)
Enterococcus faecium/inmunología , Tracto Gastrointestinal/microbiología , Inflamación/microbiología , Interleucina-10/inmunología , Animales , Carga Bacteriana , Permeabilidad de la Membrana Celular , Quimiocina CXCL10/inmunología , Enfermedad Crónica/terapia , Enterococcus faecium/crecimiento & desarrollo , Tracto Gastrointestinal/inmunología , Inflamación/inmunología , Inflamación/terapia , Interferón gamma/inmunología , Ratones , Ratones Noqueados , Probióticos/administración & dosificación , Verrucomicrobia/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...