Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27.234
Filtrar
Más filtros

Intervalo de año de publicación
1.
Annu Rev Immunol ; 30: 149-73, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22224779

RESUMEN

The lumen of the gastrointestinal (GI) tract is home to an enormous quantity of different bacterial species, our microbiota, that thrive in an often symbiotic relationship with the host. Given that the healthy host must regulate contact between the microbiota and its immune system to avoid overwhelming systemic immune activation, humans have evolved several mechanisms to attenuate systemic microbial translocation (MT) and its consequences. However, several diseases are associated with the failure of one or more of these mechanisms, with consequent immune activation and deleterious effects on health. Here, we discuss the mechanisms underlying MT, diseases associated with MT, and therapeutic interventions that aim to decrease it.


Asunto(s)
Tracto Gastrointestinal/inmunología , Tracto Gastrointestinal/microbiología , Metagenoma/fisiología , Animales , Antiinfecciosos/farmacología , Antiinfecciosos/uso terapéutico , Enfermedades del Sistema Digestivo/inmunología , Enfermedades del Sistema Digestivo/microbiología , Enfermedades del Sistema Digestivo/terapia , Tracto Gastrointestinal/efectos de los fármacos , Interacciones Huésped-Patógeno/inmunología , Humanos , Factores Inmunológicos/farmacología , Factores Inmunológicos/uso terapéutico , Metagenoma/efectos de los fármacos , Probióticos/uso terapéutico
2.
Cell ; 174(6): 1388-1405.e21, 2018 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-30193112

RESUMEN

Empiric probiotics are commonly consumed by healthy individuals as means of life quality improvement and disease prevention. However, evidence of probiotic gut mucosal colonization efficacy remains sparse and controversial. We metagenomically characterized the murine and human mucosal-associated gastrointestinal microbiome and found it to only partially correlate with stool microbiome. A sequential invasive multi-omics measurement at baseline and during consumption of an 11-strain probiotic combination or placebo demonstrated that probiotics remain viable upon gastrointestinal passage. In colonized, but not germ-free mice, probiotics encountered a marked mucosal colonization resistance. In contrast, humans featured person-, region- and strain-specific mucosal colonization patterns, hallmarked by predictive baseline host and microbiome features, but indistinguishable by probiotics presence in stool. Consequently, probiotics induced a transient, individualized impact on mucosal community structure and gut transcriptome. Collectively, empiric probiotics supplementation may be limited in universally and persistently impacting the gut mucosa, meriting development of new personalized probiotic approaches.


Asunto(s)
Microbioma Gastrointestinal , Probióticos/administración & dosificación , Adolescente , Adulto , Anciano , Animales , Bacterias/genética , Bacterias/aislamiento & purificación , Heces/microbiología , Femenino , Mucosa Gástrica/microbiología , Humanos , Mucosa Intestinal/microbiología , Masculino , Metagenómica , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Efecto Placebo , Análisis de Componente Principal , ARN Ribosómico 16S/genética , ARN Ribosómico 16S/metabolismo , Transcriptoma , Adulto Joven
3.
Cell ; 174(6): 1406-1423.e16, 2018 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-30193113

RESUMEN

Probiotics are widely prescribed for prevention of antibiotics-associated dysbiosis and related adverse effects. However, probiotic impact on post-antibiotic reconstitution of the gut mucosal host-microbiome niche remains elusive. We invasively examined the effects of multi-strain probiotics or autologous fecal microbiome transplantation (aFMT) on post-antibiotic reconstitution of the murine and human mucosal microbiome niche. Contrary to homeostasis, antibiotic perturbation enhanced probiotics colonization in the human mucosa but only mildly improved colonization in mice. Compared to spontaneous post-antibiotic recovery, probiotics induced a markedly delayed and persistently incomplete indigenous stool/mucosal microbiome reconstitution and host transcriptome recovery toward homeostatic configuration, while aFMT induced a rapid and near-complete recovery within days of administration. In vitro, Lactobacillus-secreted soluble factors contributed to probiotics-induced microbiome inhibition. Collectively, potential post-antibiotic probiotic benefits may be offset by a compromised gut mucosal recovery, highlighting a need of developing aFMT or personalized probiotic approaches achieving mucosal protection without compromising microbiome recolonization in the antibiotics-perturbed host.


Asunto(s)
Antibacterianos/farmacología , Microbioma Gastrointestinal/efectos de los fármacos , Probióticos/administración & dosificación , Adolescente , Adulto , Anciano , Animales , Trasplante de Microbiota Fecal , Heces/microbiología , Femenino , Humanos , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/microbiología , Lactobacillus/efectos de los fármacos , Lactobacillus/genética , Lactobacillus/aislamiento & purificación , Lactococcus/genética , Lactococcus/aislamiento & purificación , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , ARN Ribosómico 16S/análisis , ARN Ribosómico 16S/genética , ARN Ribosómico 16S/metabolismo , Adulto Joven
4.
Nat Immunol ; 20(10): 1279-1290, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31501577

RESUMEN

The revolution in microbiota research over the past decade has provided invaluable knowledge about the function of the microbial species that inhabit the human body. It has become widely accepted that these microorganisms, collectively called 'the microbiota', engage in networks of interactions with each other and with the host that aim to benefit both the microbial members and the mammalian members of this unique ecosystem. The lungs, previously thought to be sterile, are now known to harbor a unique microbiota and, additionally, to be influenced by microbial signals from distal body sites, such as the intestine. Here we review the role of the lung and gut microbiotas in respiratory health and disease and highlight the main pathways of communication that underlie the gut-lung axis.


Asunto(s)
Microbioma Gastrointestinal , Enfermedades Pulmonares/microbiología , Pulmón/microbiología , Microbiota , Probióticos/uso terapéutico , Acinetobacter , Animales , Bifidobacterium , Suplementos Dietéticos , Femenino , Interacciones Huésped-Patógeno , Humanos , Lactobacillus , Pulmón/inmunología , Enfermedades Pulmonares/dietoterapia , Enfermedades Pulmonares/inmunología , Exposición Materna , Embarazo
5.
Nat Immunol ; 24(5): 732, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37095381
7.
Nature ; 633(8031): 878-886, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39294375

RESUMEN

Persistent colonization and outgrowth of potentially pathogenic organisms in the intestine can result from long-term antibiotic use or inflammatory conditions, and may perpetuate dysregulated immunity and tissue damage1,2. Gram-negative Enterobacteriaceae gut pathobionts are particularly recalcitrant to conventional antibiotic treatment3,4, although an emerging body of evidence suggests that manipulation of the commensal microbiota may be a practical alternative therapeutic strategy5-7. Here we isolated and down-selected commensal bacterial consortia from stool samples from healthy humans that could strongly and specifically suppress intestinal Enterobacteriaceae. One of the elaborated consortia, comprising 18 commensal strains, effectively controlled ecological niches by regulating gluconate availability, thereby re-establishing colonization resistance and alleviating Klebsiella- and Escherichia-driven intestinal inflammation in mice. Harnessing these activities in the form of live bacterial therapies may represent a promising solution to combat the growing threat of proinflammatory, antimicrobial-resistant Enterobacteriaceae infection.


Asunto(s)
Infecciones por Enterobacteriaceae , Enterobacteriaceae , Microbioma Gastrointestinal , Simbiosis , Animales , Humanos , Ratones , Enterobacteriaceae/crecimiento & desarrollo , Enterobacteriaceae/patogenicidad , Infecciones por Enterobacteriaceae/microbiología , Infecciones por Enterobacteriaceae/prevención & control , Infecciones por Enterobacteriaceae/terapia , Escherichia/crecimiento & desarrollo , Escherichia/patogenicidad , Heces/microbiología , Microbioma Gastrointestinal/fisiología , Gluconatos/metabolismo , Inflamación/microbiología , Inflamación/prevención & control , Inflamación/terapia , Intestinos/microbiología , Klebsiella/crecimiento & desarrollo , Klebsiella/patogenicidad , Ratones Endogámicos C57BL , Probióticos/uso terapéutico , Simbiosis/fisiología , Farmacorresistencia Bacteriana
8.
Nature ; 620(7973): 381-385, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37532933

RESUMEN

The human gut microbiota has gained interest as an environmental factor that may contribute to health or disease1. The development of next-generation probiotics is a promising strategy to modulate the gut microbiota and improve human health; however, several key candidate next-generation probiotics are strictly anaerobic2 and may require synergy with other bacteria for optimal growth. Faecalibacterium prausnitzii is a highly prevalent and abundant human gut bacterium associated with human health, but it has not yet been developed into probiotic formulations2. Here we describe the co-isolation of F. prausnitzii and Desulfovibrio piger, a sulfate-reducing bacterium, and their cross-feeding for growth and butyrate production. To produce a next-generation probiotic formulation, we adapted F. prausnitzii to tolerate oxygen exposure, and, in proof-of-concept studies, we demonstrate that the symbiotic product is tolerated by mice and humans (ClinicalTrials.gov identifier: NCT03728868 ) and is detected in the human gut in a subset of study participants. Our study describes a technology for the production of next-generation probiotics based on the adaptation of strictly anaerobic bacteria to tolerate oxygen exposures without a reduction in potential beneficial properties. Our technology may be used for the development of other strictly anaerobic strains as next-generation probiotics.


Asunto(s)
Biotecnología , Microbioma Gastrointestinal , Probióticos , Animales , Humanos , Ratones , Butiratos/metabolismo , Oxígeno/metabolismo , Oxígeno/farmacología , Probióticos/metabolismo , Aerobiosis , Faecalibacterium prausnitzii/efectos de los fármacos , Faecalibacterium prausnitzii/metabolismo , Simbiosis , Biotecnología/métodos
9.
Nature ; 620(7975): 881-889, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37558878

RESUMEN

Dendritic cells (DCs) have a role in the development and activation of self-reactive pathogenic T cells1,2. Genetic variants that are associated with the function of DCs have been linked to autoimmune disorders3,4, and DCs are therefore attractive therapeutic targets for such diseases. However, developing DC-targeted therapies for autoimmunity requires identification of the mechanisms that regulate DC function. Here, using single-cell and bulk transcriptional and metabolic analyses in combination with cell-specific gene perturbation studies, we identify a regulatory loop of negative feedback that operates in DCs to limit immunopathology. Specifically, we find that lactate, produced by activated DCs and other immune cells, boosts the expression of NDUFA4L2 through a mechanism mediated by hypoxia-inducible factor 1α (HIF-1α). NDUFA4L2 limits the production of mitochondrial reactive oxygen species that activate XBP1-driven transcriptional modules in DCs that are involved in the control of pathogenic autoimmune T cells. We also engineer a probiotic that produces lactate and suppresses T cell autoimmunity through the activation of HIF-1α-NDUFA4L2 signalling in DCs. In summary, we identify an immunometabolic pathway that regulates DC function, and develop a synthetic probiotic for its therapeutic activation.


Asunto(s)
Enfermedades Autoinmunes , Sistema Nervioso Central , Células Dendríticas , Subunidad alfa del Factor 1 Inducible por Hipoxia , Ácido Láctico , Humanos , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/metabolismo , Enfermedades Autoinmunes/prevención & control , Autoinmunidad , Sistema Nervioso Central/citología , Sistema Nervioso Central/inmunología , Sistema Nervioso Central/patología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/química , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Ácido Láctico/metabolismo , Probióticos/uso terapéutico , Especies Reactivas de Oxígeno/metabolismo , Linfocitos T/inmunología , Retroalimentación Fisiológica , Lactasa/genética , Lactasa/metabolismo , Análisis de la Célula Individual
10.
Nature ; 620(7973): 386-392, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37495692

RESUMEN

Transient molecules in the gastrointestinal tract such as nitric oxide and hydrogen sulfide are key signals and mediators of inflammation. Owing to their highly reactive nature and extremely short lifetime in the body, these molecules are difficult to detect. Here we develop a miniaturized device that integrates genetically engineered probiotic biosensors with a custom-designed photodetector and readout chip to track these molecules in the gastrointestinal tract. Leveraging the molecular specificity of living sensors1, we genetically encoded bacteria to respond to inflammation-associated molecules by producing luminescence. Low-power electronic readout circuits2 integrated into the device convert the light emitted by the encapsulated bacteria to a wireless signal. We demonstrate in vivo biosensor monitoring in the gastrointestinal tract of small and large animal models and the integration of all components into a sub-1.4 cm3 form factor that is compatible with ingestion and capable of supporting wireless communication. With this device, diseases such as inflammatory bowel disease could be diagnosed earlier than is currently possible, and disease progression could be more accurately tracked. The wireless detection of short-lived, disease-associated molecules with our device could also support timely communication between patients and caregivers, as well as remote personalized care.


Asunto(s)
Biomarcadores , Técnicas Biosensibles , Sulfuro de Hidrógeno , Inflamación , Óxido Nítrico , Animales , Biomarcadores/análisis , Biomarcadores/metabolismo , Técnicas Biosensibles/instrumentación , Técnicas Biosensibles/métodos , Enfermedades Inflamatorias del Intestino/diagnóstico , Enfermedades Inflamatorias del Intestino/metabolismo , Modelos Animales , Tracto Gastrointestinal/metabolismo , Tracto Gastrointestinal/microbiología , Cápsulas/administración & dosificación , Probióticos/metabolismo , Bacterias/metabolismo , Luminiscencia , Progresión de la Enfermedad , Inflamación/diagnóstico , Inflamación/metabolismo , Óxido Nítrico/análisis , Óxido Nítrico/metabolismo , Sulfuro de Hidrógeno/análisis , Sulfuro de Hidrógeno/metabolismo , Tecnología Inalámbrica/instrumentación , Administración Oral , Tecnología de Sensores Remotos/instrumentación , Tecnología de Sensores Remotos/métodos , Factores de Tiempo , Humanos , Tamaño Corporal
11.
Cell ; 155(7): 1451-63, 2013 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-24315484

RESUMEN

Neurodevelopmental disorders, including autism spectrum disorder (ASD), are defined by core behavioral impairments; however, subsets of individuals display a spectrum of gastrointestinal (GI) abnormalities. We demonstrate GI barrier defects and microbiota alterations in the maternal immune activation (MIA) mouse model that is known to display features of ASD. Oral treatment of MIA offspring with the human commensal Bacteroides fragilis corrects gut permeability, alters microbial composition, and ameliorates defects in communicative, stereotypic, anxiety-like and sensorimotor behaviors. MIA offspring display an altered serum metabolomic profile, and B. fragilis modulates levels of several metabolites. Treating naive mice with a metabolite that is increased by MIA and restored by B. fragilis causes certain behavioral abnormalities, suggesting that gut bacterial effects on the host metabolome impact behavior. Taken together, these findings support a gut-microbiome-brain connection in a mouse model of ASD and identify a potential probiotic therapy for GI and particular behavioral symptoms in human neurodevelopmental disorders.


Asunto(s)
Trastornos Generalizados del Desarrollo Infantil/microbiología , Tracto Gastrointestinal/microbiología , Animales , Ansiedad/metabolismo , Ansiedad/microbiología , Bacteroides fragilis , Conducta Animal , Encéfalo/fisiología , Niño , Trastornos Generalizados del Desarrollo Infantil/metabolismo , Modelos Animales de Enfermedad , Femenino , Tracto Gastrointestinal/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Microbiota , Probióticos/administración & dosificación
12.
Immunol Rev ; 326(1): 203-218, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39285525

RESUMEN

The increasing prevalence of immune-mediated non-communicable chronic diseases, such as food allergies, has prompted a deeper investigation into the role of the gut microbiome in modulating immune responses. Here, we explore the complex interactions between commensal microbes and the host immune system, highlighting the critical role of gut bacteria in maintaining immune homeostasis. We examine how modern lifestyle practices and environmental factors have disrupted co-evolved host-microbe interactions and discuss how changes in microbiome composition impact epithelial barrier function, responses to food allergens, and susceptibility to allergic diseases. Finally, we examine the potential of bioengineered microbiome-based therapies, and live biotherapeutic products, for reestablishing immune homeostasis to prevent or treat food allergies.


Asunto(s)
Hipersensibilidad a los Alimentos , Microbioma Gastrointestinal , Simbiosis , Humanos , Animales , Microbioma Gastrointestinal/inmunología , Hipersensibilidad a los Alimentos/inmunología , Simbiosis/inmunología , Homeostasis , Alérgenos/inmunología , Alimentos , Inmunomodulación , Interacciones Microbiota-Huesped/inmunología , Probióticos/uso terapéutico
13.
Annu Rev Microbiol ; 76: 435-460, 2022 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-35655344

RESUMEN

Extensive research has elucidated the influence of the gut microbiota on human health and disease susceptibility and resistance. We review recent clinical and laboratory-based experimental studies associating the gut microbiota with certain human diseases. We also highlight ongoing translational advances that manipulate the gut microbiota to treat human diseases and discuss opportunities and challenges in translating microbiome research from and to the bedside.


Asunto(s)
Enfermedad , Microbioma Gastrointestinal , Terapéutica , Trasplante de Microbiota Fecal , Humanos , Probióticos/uso terapéutico , Terapéutica/tendencias
14.
Immunity ; 49(6): 1116-1131.e7, 2018 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-30446387

RESUMEN

Nutritional supplementation with probiotics can prevent pathologic bone loss. Here we examined the impact of supplementation with Lactobacillus rhamnosus GG (LGG) on bone homeostasis in eugonadic young mice. Micro-computed tomography revealed that LGG increased trabecular bone volume in mice, which was due to increased bone formation. Butyrate produced in the gut following LGG ingestion, or butyrate fed directly to germ-free mice, induced the expansion of intestinal and bone marrow (BM) regulatory T (Treg) cells. Interaction of BM CD8+ T cells with Treg cells resulted in increased secretion of Wnt10b, a bone anabolic Wnt ligand. Mechanistically, Treg cells promoted the assembly of a NFAT1-SMAD3 transcription complex in CD8+ cells, which drove expression of Wnt10b. Reducing Treg cell numbers, or reconstitution of TCRß-/- mice with CD8+ T cells from Wnt10b-/- mice, prevented butyrate-induced bone formation and bone mass acquisition. Thus, butyrate concentrations regulate bone anabolism via Treg cell-mediated regulation of CD8+ T cell Wnt10b production.


Asunto(s)
Butiratos/farmacología , Osteogénesis/efectos de los fármacos , Linfocitos T Reguladores/metabolismo , Proteínas Wnt/metabolismo , Animales , Butiratos/metabolismo , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/metabolismo , Comunicación Celular , Proliferación Celular/efectos de los fármacos , Femenino , Lacticaseibacillus rhamnosus/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Osteoblastos/citología , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Probióticos/administración & dosificación , Probióticos/metabolismo , Linfocitos T Reguladores/citología , Proteínas Wnt/genética
15.
Nature ; 598(7882): 662-666, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34616044

RESUMEN

The availability of L-arginine in tumours is a key determinant of an efficient anti-tumour T cell response1-4. Consequently, increases of typically low L-arginine concentrations within the tumour may greatly potentiate the anti-tumour responses of immune checkpoint inhibitors, such as programmed death-ligand 1 (PD-L1)-blocking antibodies5. However, currently no means are available to locally increase intratumoural L-arginine levels. Here we used a synthetic biology approach to develop an engineered probiotic Escherichia coli Nissle 1917 strain that colonizes tumours and continuously converts ammonia, a metabolic waste product that accumulates in tumours6, to L-arginine. Colonization of tumours with these bacteria increased intratumoural L-arginine concentrations, increased the number of tumour-infiltrating T cells and had marked synergistic effects with PD-L1 blocking antibodies in the clearance of tumours. The anti-tumour effect of these bacteria was mediated by L-arginine and was dependent on T cells. These results show that engineered microbial therapies enable metabolic modulation of the tumour microenvironment leading to enhanced efficacy of immunotherapies.


Asunto(s)
Inmunoterapia/métodos , Ingeniería Metabólica , Microorganismos Modificados Genéticamente , Neoplasias Experimentales/terapia , Traslado Adoptivo , Animales , Arginina/metabolismo , Antígeno B7-H1/antagonistas & inhibidores , Línea Celular Tumoral , Escherichia coli , Femenino , Linfocitos Infiltrantes de Tumor/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/microbiología , Probióticos , Proteoma , Biología Sintética , Linfocitos T/inmunología , Microambiente Tumoral/inmunología
16.
Proc Natl Acad Sci U S A ; 121(43): e2402352121, 2024 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-39401350

RESUMEN

Empirical studies from laboratory systems and humans show that the gut microbiota is linked to host health. Similar evidence for effects on traits linked to fitness in nature is rare, not least because experimentally manipulating the gut microbiota is challenging. We isolated, characterized, and cultured a bacterial strain, Lactobacillus kimchicus APC4233, directly from a wild bird (the great tit Parus major) and provided it as a self-administered dietary supplement. We assessed the impact of the treatment on the host microbiota community, on weight, and tested whether the treatment affected a previous result linking microbiota alpha diversity to weight in nestlings. The treatment dramatically increased L. kimchicus abundance in the gut microbiota and increased alpha diversity. This effect was strongest in the youngest birds, validating earlier findings pointing to a brief developmental window when the gut microbiota are most sensitive. In time-lagged models, nestling weight was higher in the treatment birds suggesting L. kimchicus may have probiotic potential. There was also a positive time-lagged relationship between diversity and weight in control birds but not in the treatment birds, suggesting L. kimchicus helped birds compensate for low alpha diversity. We discuss why ecological context is likely key when predicting impacts of the microbiome. The manipulation of the gut microbiota with a host native strain in this wild population provides direct evidence for the role of the microbiota in the ecology and evolution of natural populations.


Asunto(s)
Microbioma Gastrointestinal , Aumento de Peso , Animales , Microbioma Gastrointestinal/genética , Lactobacillus/genética , Animales Salvajes/microbiología , Probióticos , Passeriformes/microbiología , Biodiversidad
17.
Brief Bioinform ; 25(4)2024 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-39038938

RESUMEN

With the increasing prevalence of age-related chronic diseases burdening healthcare systems, there is a pressing need for innovative management strategies. Our study focuses on the gut microbiota, essential for metabolic, nutritional, and immune functions, which undergoes significant changes with aging. These changes can impair intestinal function, leading to altered microbial diversity and composition that potentially influence health outcomes and disease progression. Using advanced metagenomic sequencing, we explore the potential of personalized probiotic supplements in 297 older adults by analyzing their gut microbiota. We identified distinctive Lactobacillus and Bifidobacterium signatures in the gut microbiota of older adults, revealing probiotic patterns associated with various population characteristics, microbial compositions, cognitive functions, and neuroimaging results. These insights suggest that tailored probiotic supplements, designed to match individual probiotic profile, could offer an innovative method for addressing age-related diseases and functional declines. Our findings enhance the existing evidence base for probiotic use among older adults, highlighting the opportunity to create more targeted and effective probiotic strategies. However, additional research is required to validate our results and further assess the impact of precision probiotics on aging populations. Future studies should employ longitudinal designs and larger cohorts to conclusively demonstrate the benefits of tailored probiotic treatments.


Asunto(s)
Envejecimiento , Suplementos Dietéticos , Microbioma Gastrointestinal , Probióticos , Probióticos/uso terapéutico , Probióticos/administración & dosificación , Humanos , Anciano , Femenino , Masculino , Anciano de 80 o más Años , Persona de Mediana Edad , Lactobacillus/genética , Metagenómica/métodos , Bifidobacterium
18.
Brief Bioinform ; 25(2)2024 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-38487846

RESUMEN

Beneficial bacteria remain largely unexplored. Lacking systematic methods, understanding probiotic community traits becomes challenging, leading to various conclusions about their probiotic effects among different publications. We developed language model-based metaProbiotics to rapidly detect probiotic bins from metagenomes, demonstrating superior performance in simulated benchmark datasets. Testing on gut metagenomes from probiotic-treated individuals, it revealed the probioticity of intervention strains-derived bins and other probiotic-associated bins beyond the training data, such as a plasmid-like bin. Analyses of these bins revealed various probiotic mechanisms and bai operon as probiotic Ruminococcaceae's potential marker. In different health-disease cohorts, these bins were more common in healthy individuals, signifying their probiotic role, but relevant health predictions based on the abundance profiles of these bins faced cross-disease challenges. To better understand the heterogeneous nature of probiotics, we used metaProbiotics to construct a comprehensive probiotic genome set from global gut metagenomic data. Module analysis of this set shows that diseased individuals often lack certain probiotic gene modules, with significant variation of the missing modules across different diseases. Additionally, different gene modules on the same probiotic have heterogeneous effects on various diseases. We thus believe that gene function integrity of the probiotic community is more crucial in maintaining gut homeostasis than merely increasing specific gene abundance, and adding probiotics indiscriminately might not boost health. We expect that the innovative language model-based metaProbiotics tool will promote novel probiotic discovery using large-scale metagenomic data and facilitate systematic research on bacterial probiotic effects. The metaProbiotics program can be freely downloaded at https://github.com/zhenchengfang/metaProbiotics.


Asunto(s)
Metagenoma , Probióticos , Humanos , Algoritmos , Metagenómica/métodos , Bacterias/genética , Lenguaje
19.
PLoS Pathog ; 20(9): e1012541, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39250508

RESUMEN

Inflammatory bowel disease (IBD) is an immune system disorder primarily characterized by colitis, the exact etiology of which remains unclear. Traditional treatment approaches currently yield limited efficacy and are associated with significant side effects. Extensive research has indicated the potent therapeutic effects of probiotics, particularly Lactobacillus strains, in managing colitis. However, the mechanisms through which Lactobacillus strains ameliorate colitis require further exploration. In our study, we selected Lactobacillus gasseri ATCC33323 from the intestinal microbiota to elucidate the specific mechanisms involved in modulation of colitis. Experimental findings in a DSS-induced colitis mouse model revealed that L. gasseri ATCC33323 significantly improved physiological damage in colitic mice, reduced the severity of colonic inflammation, decreased the production of inflammatory factors, and preserved the integrity of the intestinal epithelial structure and function. It also maintained the expression and localization of adhesive proteins while improving intestinal barrier permeability and restoring dysbiosis in the gut microbiota. E-cadherin, a critical adhesive protein, plays a pivotal role in this protective mechanism. Knocking down E-cadherin expression within the mouse intestinal tract significantly attenuated the ability of L. gasseri ATCC33323 to regulate colitis, thus confirming its protective role through E-cadherin. Finally, transcriptional analysis and in vitro experiments revealed that L. gasseri ATCC33323 regulates CDH1 transcription by affecting NR1I3, thereby promoting E-cadherin expression. These findings contribute to a better understanding of the specific mechanisms by which Lactobacillus strains alleviate colitis, offering new insights for the potential use of L. gasseri as an alternative therapy for IBD, particularly in dietary supplementation.


Asunto(s)
Cadherinas , Colitis , Sulfato de Dextran , Mucosa Intestinal , Lactobacillus gasseri , Probióticos , Animales , Colitis/inducido químicamente , Colitis/microbiología , Colitis/metabolismo , Colitis/terapia , Cadherinas/metabolismo , Ratones , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Sulfato de Dextran/toxicidad , Probióticos/farmacología , Lactobacillus gasseri/metabolismo , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad , Microbioma Gastrointestinal , Humanos
20.
Immunity ; 46(6): 977-979, 2017 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-28636967

RESUMEN

Whereas strong evidence supports the notion that the microbiota promotes immune system maturation in multiple tissues, the identity of the specific microbes that elicit protective immunity to different infections is less clear. In a recent issue of Cell Host & Microbe, Thiemann et al. (2017) report the identification of specific gut bacteria that protect from Salmonella infection by priming host IFN-γ responses.


Asunto(s)
Microbioma Gastrointestinal , Interferón gamma/metabolismo , Infecciones por Salmonella/inmunología , Salmonella/inmunología , Simbiosis , Animales , Biodiversidad , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Exposición a Riesgos Ambientales , Humanos , Inmunidad Innata , Interferón gamma/genética , Ratones , Probióticos , Especificidad de la Especie
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA