Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Res Sq ; 2023 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-37961431

RESUMEN

Persistent colonization and outgrowth of pathogenic organisms in the intestine may occur due to long-term antibiotic usage or inflammatory conditions, which perpetuate dysregulated immunity and tissue damage1,2. Gram-negative Enterobacteriaceae gut pathobionts are particularly recalcitrant to conventional antibiotic treatment3,4, though an emerging body of evidence suggests that manipulation of the commensal microbiota may be a practical alternative therapeutic strategy5-7. In this study, we rationally isolated and down-selected commensal bacterial consortia from healthy human stool samples capable of strongly and specifically suppressing intestinal Enterobacteriaceae. One of the elaborated consortia, consisting of 18 commensal strains, effectively controlled ecological niches by regulating gluconate availability, thereby reestablishing colonization resistance and alleviating antibiotic-resistant Klebsiella-driven intestinal inflammation in mice. Harnessing these microbial activities in the form of live bacterial therapeutics may represent a promising solution to combat the growing threat of proinflammatory, antimicrobial-resistant bacterial infection.

2.
Science ; 376(6596): 934-936, 2022 05 27.
Artículo en Inglés | MEDLINE | ID: mdl-35617380

RESUMEN

Oral microbiota form complex biofilms that can affect local and systemic health.


Asunto(s)
Biopelículas , Enfermedad , Microbioma Gastrointestinal , Boca , Microbioma Gastrointestinal/fisiología , Salud , Humanos , Boca/microbiología
3.
Annu Rev Immunol ; 40: 559-587, 2022 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-35113732

RESUMEN

The immune system employs recognition tools to communicate with its microbial evolutionary partner. Among all the methods of microbial perception, T cells enable the widest spectrum of microbial recognition resolution, ranging from the crudest detection of whole groups of microbes to the finest detection of specific antigens. The application of this recognition capability to the crucial task of combatting infections has been the focus of classical immunology. We now appreciate that the coevolution of the immune system and the microbiota has led to development of a lush immunological decision tree downstream of microbial recognition, of which an inflammatory response is but one branch. In this review we discuss known T cell-microbe interactions in the gut and place them in the context of an algorithmic framework of recognition, context-dependent interpretation, and response circuits across multiple levels of microbial recognition resolution. The malleability of T cells in response to the microbiota presents an opportunity to edit immune response cellularity, identity, and functionality by utilizing microbiota-controlled pathways to promote human health.


Asunto(s)
Microbiota , Linfocitos T , Animales , Humanos
4.
Eur J Immunol ; 51(9): 2120-2136, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34242413

RESUMEN

Fundamental asymmetries between the host and its microbiome in enzymatic activities and nutrient storage capabilities have promoted mutualistic adaptations on both sides. As a result, the enteric immune system has evolved so as not to cause a zero-sum sterilization of non-self, but rather achieve a non-zero-sum self-reinforcing cooperation with its evolutionary partner the microbiome. In this review, we attempt to integrate the accumulated knowledge of immune-microbiome interactions into an evolutionary framework and trace the pattern of positive immune-microbiome feedback loops across epithelial, enteric nervous system, innate, and adaptive immune circuits. Indeed, the immune system requires commensal signals for its development and function, and reciprocally protects the microbiome from nutrient shortage and pathogen outgrowth. In turn, a healthy microbiome is the result of immune system curatorship as well as microbial ecology. The paradigms of host-microbiome asymmetry and the cooperative nature of their interactions identified in the gut are applicable across all tissues influenced by microbial activities. Incorporation of immune system influences into models of microbiome ecology will be a step forward toward defining what constitutes a healthy human microbiome and guide discoveries of novel host-microbiome mutualistic adaptations that may be harnessed for the promotion of human health.


Asunto(s)
Sistema Nervioso Entérico/fisiología , Microbioma Gastrointestinal/inmunología , Tracto Gastrointestinal/inmunología , Tracto Gastrointestinal/microbiología , Inmunidad Adaptativa/inmunología , Tracto Gastrointestinal/inervación , Tracto Gastrointestinal/fisiología , Humanos , Inmunidad Innata/inmunología , Simbiosis/inmunología
5.
Cell ; 182(6): 1441-1459.e21, 2020 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-32888430

RESUMEN

Throughout a 24-h period, the small intestine (SI) is exposed to diurnally varying food- and microbiome-derived antigenic burdens but maintains a strict immune homeostasis, which when perturbed in genetically susceptible individuals, may lead to Crohn disease. Herein, we demonstrate that dietary content and rhythmicity regulate the diurnally shifting SI epithelial cell (SIEC) transcriptional landscape through modulation of the SI microbiome. We exemplify this concept with SIEC major histocompatibility complex (MHC) class II, which is diurnally modulated by distinct mucosal-adherent SI commensals, while supporting downstream diurnal activity of intra-epithelial IL-10+ lymphocytes regulating the SI barrier function. Disruption of this diurnally regulated diet-microbiome-MHC class II-IL-10-epithelial barrier axis by circadian clock disarrangement, alterations in feeding time or content, or epithelial-specific MHC class II depletion leads to an extensive microbial product influx, driving Crohn-like enteritis. Collectively, we highlight nutritional features that modulate SI microbiome, immunity, and barrier function and identify dietary, epithelial, and immune checkpoints along this axis to be potentially exploitable in future Crohn disease interventions.


Asunto(s)
Enfermedad de Crohn/microbiología , Células Epiteliales/metabolismo , Microbioma Gastrointestinal , Antígenos de Histocompatibilidad Clase II/metabolismo , Intestino Delgado/inmunología , Intestino Delgado/microbiología , Transcriptoma/genética , Animales , Antibacterianos/farmacología , Relojes Circadianos/fisiología , Enfermedad de Crohn/inmunología , Enfermedad de Crohn/metabolismo , Dieta , Células Epiteliales/citología , Células Epiteliales/inmunología , Citometría de Flujo , Microbioma Gastrointestinal/efectos de los fármacos , Microbioma Gastrointestinal/genética , Perfilación de la Expresión Génica , Antígenos de Histocompatibilidad Clase II/genética , Homeostasis , Hibridación Fluorescente in Situ , Interleucina-10/metabolismo , Interleucina-10/farmacología , Intestino Delgado/fisiología , Linfocitos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Periodicidad , Linfocitos T/inmunología , Transcriptoma/fisiología
6.
Nat Microbiol ; 5(4): 610-619, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32015497

RESUMEN

Although much research has been done on the diversity of the gut microbiome, little is known about how it influences intestinal homeostasis under normal and pathogenic conditions. Epigenetic mechanisms have recently been suggested to operate at the interface between the microbiota and the intestinal epithelium. We performed whole-genome bisulfite sequencing on conventionally raised and germ-free mice, and discovered that exposure to commensal microbiota induced localized DNA methylation changes at regulatory elements, which are TET2/3-dependent. This culminated in the activation of a set of 'early sentinel' response genes to maintain intestinal homeostasis. Furthermore, we demonstrated that exposure to the microbiota in dextran sodium sulfate-induced acute inflammation results in profound DNA methylation and chromatin accessibility changes at regulatory elements, leading to alterations in gene expression programs enriched in colitis- and colon-cancer-associated functions. Finally, by employing genetic interventions, we show that microbiota-induced epigenetic programming is necessary for proper intestinal homeostasis in vivo.


Asunto(s)
Colitis/genética , ADN/genética , Epigénesis Genética , Microbioma Gastrointestinal/fisiología , Genoma , Simbiosis/genética , Animales , Colitis/inducido químicamente , Colitis/microbiología , Colitis/patología , Colon/metabolismo , Colon/microbiología , ADN/metabolismo , Metilación de ADN , Sulfato de Dextran/administración & dosificación , Vida Libre de Gérmenes , Homeostasis/genética , Inflamación , Masculino , Ratones , Ratones Endogámicos C57BL , Secuenciación Completa del Genoma
7.
EMBO Rep ; 20(4)2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30877136

RESUMEN

Host-microbiome interactions constitute key determinants of host physiology, while their dysregulation is implicated in a wide range of human diseases. The microbiome undergoes diurnal variation in composition and function, and this in turn drives oscillations in host gene expression and functions. In this review, we discuss the newest developments in understanding circadian host-microbiome interplays, and how they may be relevant in health and disease contexts. We summarize the molecular mechanisms by which the microbiome influences host function in a diurnal manner, and inversely describe how the host orchestrates circadian rhythmicity of the microbiome. Furthermore, we highlight the future perspectives and challenges in studying this new and exciting facet of host-microbiome interactions. Finally, we illustrate how the elucidation of the microbiome chronobiology may pave the way for novel therapeutic approaches.


Asunto(s)
Ritmo Circadiano , Susceptibilidad a Enfermedades , Homeostasis , Microbiota , Animales , Relojes Circadianos , Retroalimentación Fisiológica , Microbioma Gastrointestinal , Humanos
8.
Semin Immunol ; 32: 74-81, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28431920

RESUMEN

The commensal microbiome constitutes an important modulator of host physiology and risk of disease, including cancer development and progression. Lately, the microbiome has been suggested to modulate the efficacy of anti-cancer treatment. Examples include chemotherapy and total body irradiation-induced barrier function disruption, leading to microbial efflux that drives activation of anti-tumorigenic T cells; Microbiome-driven release of reactive oxygen species contributing to the efficacy of platinum salts; and microbiome-induced immune priming promoting the anti-tumor effects of alkylating chemotherapy and immune checkpoint inhibitors. Furthermore, selected commensals are able to colonize solid tumors. This 'tumor microbiome' may further impact local tumor responses to treatment and potentially be harnessed for tumor-specific targeting and therapeutic delivery. In this review, we present recent advances in understanding of the intricate role of microbiome in modulating efficacy of a number of anti-cancer treatments, and discuss how anti-cancer treatment approaches utilizing the tumor microbiome may enhance oncological treatment efficacy.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos Alquilantes/uso terapéutico , Inmunomodulación , Inmunoterapia/métodos , Microbiota/inmunología , Neoplasias/microbiología , Neoplasias/terapia , Animales , Carcinogénesis , Receptores Coestimuladores e Inhibidores de Linfocitos T/inmunología , Humanos , Neoplasias/inmunología , Simbiosis , Resultado del Tratamiento
9.
Cell ; 167(6): 1495-1510.e12, 2016 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-27912059

RESUMEN

The intestinal microbiota undergoes diurnal compositional and functional oscillations that affect metabolic homeostasis, but the mechanisms by which the rhythmic microbiota influences host circadian activity remain elusive. Using integrated multi-omics and imaging approaches, we demonstrate that the gut microbiota features oscillating biogeographical localization and metabolome patterns that determine the rhythmic exposure of the intestinal epithelium to different bacterial species and their metabolites over the course of a day. This diurnal microbial behavior drives, in turn, the global programming of the host circadian transcriptional, epigenetic, and metabolite oscillations. Surprisingly, disruption of homeostatic microbiome rhythmicity not only abrogates normal chromatin and transcriptional oscillations of the host, but also incites genome-wide de novo oscillations in both intestine and liver, thereby impacting diurnal fluctuations of host physiology and disease susceptibility. As such, the rhythmic biogeography and metabolome of the intestinal microbiota regulates the temporal organization and functional outcome of host transcriptional and epigenetic programs.


Asunto(s)
Ritmo Circadiano , Colon/microbiología , Microbioma Gastrointestinal , Transcriptoma , Animales , Cromatina/metabolismo , Colon/metabolismo , Vida Libre de Gérmenes , Hígado/metabolismo , Ratones , Microscopía Electrónica de Rastreo
10.
World J Clin Oncol ; 7(2): 200-13, 2016 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-27081642

RESUMEN

"The forgotten organ", the human microbiome, comprises a community of microorganisms that colonizes various sites of the human body. Through coevolution of bacteria, archaea and fungi with the human host over thousands of years, a complex host-microbiome relationship emerged in which many functions, including metabolism and immune responses, became codependent. This coupling becomes evident when disruption in the microbiome composition, termed dysbiosis, is mirrored by the development of pathologies in the host. Among the most serious consequences of dysbiosis, is the development of cancer. As many as 20% of total cancers worldwide are caused by a microbial agent. To date, a vast majority of microbiome-cancer studies focus solely on the microbiome of the large intestine and the development of gastrointestinal cancers. Here, we will review the available evidence implicating microbiome involvement in the development and progression of non-gastrointestinal cancers, while distinguishing between viral and bacterial drivers of cancer, as well as "local" and "systemic", "cancer-stimulating" and "cancer-suppressing" effects of the microbiome. Developing a system-wide approach to cancer-microbiome studies will be crucial in understanding how microbiome influences carcinogenesis, and may enable to employ microbiome-targeting approaches as part of cancer treatment.

11.
Nat Methods ; 11(6): 653-5, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24793455

RESUMEN

Deep profiling of antibody and T cell-receptor repertoires by means of high-throughput sequencing has become an attractive approach for adaptive immunity studies, but its power is substantially compromised by the accumulation of PCR and sequencing errors. Here we report MIGEC (molecular identifier groups-based error correction), a strategy for high-throughput sequencing data analysis. MIGEC allows for nearly absolute error correction while fully preserving the natural diversity of complex immune repertoires.


Asunto(s)
Dermatoglifia del ADN/métodos , Secuenciación de Nucleótidos de Alto Rendimiento/normas , Receptores de Antígenos de Linfocitos T/genética , Proyectos de Investigación , Dermatoglifia del ADN/normas , Reacción en Cadena de la Polimerasa/normas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...