Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Curr Protoc ; 4(7): e1092, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-39007482

RESUMEN

The intestinal inflammation induced by injection of naïve CD4+ T cells into lymphocyte-deficient hosts (more commonly known as the T cell transfer model of colitis) shares many features of idiopathic inflammatory bowel disease (IBD) in humans, such as epithelial cell hyperplasia, crypt abscess formation, and dense lamina propria lymphocyte infiltration. As such, it provides a useful tool for studying mucosal immune regulation as it relates to the pathogenesis and treatment of IBD in humans. In the IBD model described here, colitis is induced in Rag (recombination-activating gene)-deficient mice by reconstitution of these mice with naïve CD4+CD45RBhi T cells through adoptive T cell transfer. Although different recipient hosts of cell transfer can be used, Rag-deficient mice are the best characterized and support studies that are both flexible and reproduceable. As described in the Basic Protocol, in most studies the transferred cells consist of naïve CD4+ T cells (CD45RBhi T cells) derived by fluorescence-activated cell sorting from total CD4+ T cells previously purified using immunomagnetic negative selection beads. In a Support Protocol, methods to characterize colonic disease progression are described, including the monitoring of weight loss and diarrhea and the histological assessment of colon pathology. © 2024 The Author(s). Current Protocols published by Wiley Periodicals LLC. Basic Protocol: Induction of IBD in Rag-deficient mice by the transfer of naïve CD4+CD45RBhi T cells Support Protocol: Monitoring development of colitis.


Asunto(s)
Linfocitos T CD4-Positivos , Modelos Animales de Enfermedad , Enfermedades Inflamatorias del Intestino , Animales , Ratones , Linfocitos T CD4-Positivos/inmunología , Enfermedades Inflamatorias del Intestino/inmunología , Enfermedades Inflamatorias del Intestino/patología , Colitis/inmunología , Colitis/inducido químicamente , Colitis/patología , Traslado Adoptivo
2.
Nat Microbiol ; 7(4): 590-599, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35365791

RESUMEN

Experimental mouse models are central to basic biomedical research; however, variability exists across genetically identical mice and mouse facilities making comparisons difficult. Whether specific indigenous gut bacteria drive immunophenotypic variability in mouse models of human disease remains poorly understood. We performed a large-scale experiment using 579 genetically identical laboratory mice from a single animal facility, designed to identify the causes of disease variability in the widely used dextran sulphate sodium mouse model of inflammatory bowel disease. Commonly used treatment endpoint measures-weight loss and intestinal pathology-showed limited correlation and varied across mouse lineages. Analysis of the gut microbiome, coupled with machine learning and targeted anaerobic culturing, identified and isolated two previously undescribed species, Duncaniella muricolitica and Alistipes okayasuensis, and demonstrated that they exert dominant effects in the dextran sulphate sodium model leading to variable treatment endpoint measures. We show that the identified gut microbial species are common, but not ubiquitous, in mouse facilities around the world, and suggest that researchers monitor for these species to provide experimental design opportunities for improved mouse models of human intestinal diseases.


Asunto(s)
Colitis , Microbioma Gastrointestinal , Enfermedades Inflamatorias del Intestino , Animales , Bacteroidetes , Colitis/inducido químicamente , Colitis/microbiología , Modelos Animales de Enfermedad , Enfermedades Inflamatorias del Intestino/microbiología , Ratones
3.
Cell Rep ; 38(13): 110565, 2022 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-35354043

RESUMEN

Interleukin (IL)-10 is considered a prototypical anti-inflammatory cytokine, significantly contributing to the maintenance and reestablishment of immune homeostasis. Accordingly, it has been shown in the intestine that IL-10 produced by Tregs can act on effector T cells, thereby limiting inflammation. Herein, we investigate whether this role also applies to IL-10 produced by T cells during central nervous system (CNS) inflammation. During neuroinflammation, both CNS-resident and -infiltrating cells produce IL-10; yet, as IL-10 has a pleotropic function, the exact contribution of the different cellular sources is not fully understood. We find that T-cell-derived IL-10, but not other relevant IL-10 sources, can promote inflammation in experimental autoimmune encephalomyelitis. Furthermore, in the CNS, T-cell-derived IL-10 acts on effector T cells, promoting their survival and thereby enhancing inflammation and CNS autoimmunity. Our data indicate a pro-inflammatory role of T-cell-derived IL-10 in the CNS.


Asunto(s)
Interleucina-10 , Linfocitos T , Animales , Linfocitos T CD4-Positivos , Supervivencia Celular , Sistema Nervioso Central , Inflamación , Interleucina-10/fisiología , Ratones
4.
Immunology ; 159(1): 4-14, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31777071

RESUMEN

The past two decades have seen an explosion in research that aims to understand how the dynamic interplay with the gut microbiota impacts host health and disease, establishing a role for the gut microbiota in a plethora of pathologies. Understanding how health-promoting microbiota are established and how beneficial host-microbiota interactions are maintained is of immense biomedical importance. Despite the enormous progress that has been made, our knowledge of the specific microbiota members that mediate these effects and the mechanisms underlying these interactions is rudimentary. The dearth of information regarding the nature of advantageous host-microbiota interactions, and the factors that cause these relationships to go awry, has hampered our ability to realize the therapeutic potential of the microbiota. Here we discuss key issues that limit current knowledge and describe a path forwards to improving our understanding of the contributions of the microbiota to host health.


Asunto(s)
Microbioma Gastrointestinal/inmunología , Interacciones Microbiota-Huesped/inmunología , Inmunidad , Animales , Vida Libre de Gérmenes/inmunología , Homeostasis/inmunología , Humanos , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Ratones , Modelos Animales
5.
Immunity ; 48(6): 1070-1072, 2018 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-29924970

RESUMEN

Shigella pathogenesis has confounded researchers for years because of its narrow host selectivity and extraordinary infectious capability. In this issue of Immunity, Xu et al. (2018) identify a cunning mechanism whereby Shigella hijacks human α-defensin 5 to enhance its adhesion and subsequent invasion.


Asunto(s)
Adhesión Bacteriana , Disentería Bacilar , Humanos , Shigella , alfa-Defensinas
6.
Autophagy ; 14(8): 1460-1461, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29799774

RESUMEN

Genome-wide association studies (GWAS) linking polymorphisms in ATG16L1 with susceptibility to inflammatory bowel disease (IBD) have prompted mucosal immunologists to investigate the functional roles of macroautophagy/autophagy in different cell types in the gut. Here we present a recent study that addressed 2 key questions: in which cell type is autophagy deficiency most detrimental during chronic colitis and what is the functional role of autophagy in those cells? We report that autophagy in intestinal epithelial cells (IECs) acts to limit intestinal inflammation by protecting them from TNF-induced apoptosis and we discuss the potential implications for IBD treatment.


Asunto(s)
Autofagia , Colitis , Animales , Apoptosis , Células Epiteliales , Estudio de Asociación del Genoma Completo , Ratones
7.
J Infect Dis ; 218(8): 1314-1323, 2018 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-29800313

RESUMEN

The balance between pro- and antiinflammatory mechanisms is essential to limit immune-mediated pathology, and CD4+ forkhead box P3 (Foxp3+) regulatory T cells (Treg) play an important role in this process. The expression of inhibitory receptors regulates cytokine production by Plasmodium vivax-specific T cells. Our goal was to assess the induction of programmed death-1 (PD-1) and cytotoxic T-lymphocyte antigen (CTLA-4) on Treg during malaria and to evaluate their function. We found that P. vivax infection triggered an increase in circulating Treg and their expression of CTLA-4 and PD-1. Functional analysis demonstrated that Treg from malaria patients had impaired suppressive ability and PD-1+Treg displayed lower levels of Foxp3 and Helios, but had higher frequencies of T-box transcription factor+ and interferon-gamma+ cells than PD-1-Treg. Thus malaria infection alters the function of circulating Treg by triggering increased expression of PD-1 on Treg that is associated with decreased regulatory function and increased proinflammatory characteristics.


Asunto(s)
Malaria Vivax/inmunología , Malaria Vivax/parasitología , Linfocitos T Reguladores/fisiología , Adulto , Proliferación Celular , Citocinas/genética , Citocinas/metabolismo , Femenino , Regulación de la Expresión Génica/inmunología , Humanos , Inmunofenotipificación , Masculino , Persona de Mediana Edad , Plasmodium vivax , Reticulocitos/parasitología , Reticulocitos/fisiología , Adulto Joven
8.
Front Immunol ; 9: 499, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29636751

RESUMEN

Chronic inflammation contributes to tumor initiation in colitis-associated colorectal cancer (CRC). Indeed, inflammatory bowel disease (IBD) patients show an increased risk of developing CRC. Cancer immune evasion is a major issue in CRC and preclinical and clinical evidence has defined a critical role for myeloid-derived suppressor cells (MDSCs) that contribute to tumor growth and progression by suppressing T-cells and modulating innate immune responses. MDSCs comprise a heterogeneous population of immature myeloid cells that can be distinct in two subtypes: CD11b+Ly6G+Ly6Clow with granulocytic phenotype (G-MDSCs) and CD11b+Ly6G-Ly6Chigh with monocytic phenotype (M-MDSCs). Hydrogen sulfide (H2S) is an endogenous gaseous signaling molecule that regulates various physiological and pathophysiological functions. In particular, several studies support its anti-inflammatory activity in experimental colitis and ulcer. However, the role of the H2S pathway in innate immune-mediated IBD has not yet been elucidated. To better define a possible link between MDSCs and H2S pathway in colitis-associated CRC development, we used an innate immune-mediated IBD model induced by infection with the bacterium Helicobacter hepaticus (Hh), closely resembling human IBD. Here, we demonstrated an involvement of MDSCs in colitis development. A significant time-dependent increase of both G-MDSCs and M-MDSCs was observed in the colon and in the spleen of Hh-infected mice. Following, we observed that chronic oral administration of the H2S donor DATS reduced colon inflammation by limiting the recruitment of G-MDSCs in the colon of Hh-infected mice. Thus, we identify the metabolic pathway l-cysteine/H2S as a possible new player in the immunosuppressive mechanism responsible for the MDSCs-promoted colitis-associated cancer development.


Asunto(s)
Colitis/inmunología , Infecciones por Helicobacter/inmunología , Helicobacter hepaticus/inmunología , Sulfuro de Hidrógeno/farmacología , Inmunidad Celular/efectos de los fármacos , Células Supresoras de Origen Mieloide/inmunología , Animales , Colitis/genética , Colitis/microbiología , Colitis/patología , Colon/inmunología , Colon/microbiología , Colon/patología , Modelos Animales de Enfermedad , Infecciones por Helicobacter/patología , Inflamación/genética , Inflamación/inmunología , Inflamación/microbiología , Inflamación/patología , Ratones , Ratones Noqueados , Células Supresoras de Origen Mieloide/patología
9.
Front Immunol ; 9: 540, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29599783

RESUMEN

A controlled and self-limiting inflammatory reaction generally results in removal of the injurious agent and repair of the damaged tissue. However, in chronic inflammation, immune responses become dysregulated and prolonged, leading to tissue destruction. The role of metabolic reprogramming in orchestrating appropriate immune responses has gained increasing attention in recent years. Proliferation and differentiation of the T cell subsets that are needed to address homeostatic imbalance is accompanied by a series of metabolic adaptations, as T cells traveling from nutrient-rich secondary lymphoid tissues to sites of inflammation experience a dramatic shift in microenvironment conditions. How T cells integrate information about the local environment, such as nutrient availability or oxygen levels, and transfer these signals to functional pathways remains to be fully understood. In this review, we discuss how distinct subsets of CD4+ T cells metabolically adapt to the conditions of inflammation and whether these insights may pave the way to new treatments for human inflammatory diseases.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , Inflamación/metabolismo , Animales , Linfocitos T CD4-Positivos/inmunología , Humanos , Inflamación/tratamiento farmacológico , Inflamación/inmunología
10.
Front Immunol ; 8: 1168, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28979266

RESUMEN

Pattern recognition receptors (PRR), such as NOD-like receptors (NLRs), sense conserved microbial signatures, and host danger signals leading to the coordination of appropriate immune responses. Upon activation, a subset of NLR initiate the assembly of a multimeric protein complex known as the inflammasome, which processes pro-inflammatory cytokines and mediates a specialized form of cell death known as pyroptosis. The identification of inflammasome-associated genes as inflammatory bowel disease susceptibility genes implicates a role for the inflammasome in intestinal inflammation. Despite the fact that the functional importance of inflammasomes within immune cells has been well established, the contribution of inflammasome expression in non-hematopoietic cells remains comparatively understudied. Given that intestinal epithelial cells (IEC) act as a barrier between the host and the intestinal microbiota, inflammasome expression by these cells is likely important for intestinal immune homeostasis. Accumulating evidence suggests that the inflammasome plays a key role in shaping epithelial responses at the host-lumen interface with many inflammasome components highly expressed by IEC. Recent studies have exposed functional roles of IEC inflammasomes in mucosal immune defense, inflammation, and tumorigenesis. In this review, we present the main features of the predominant inflammasomes and their effector mechanisms contributing to intestinal homeostasis and inflammation. We also discuss existing controversies in the field and open questions related to their implications in disease. A comprehensive understanding of the molecular basis of intestinal inflammasome signaling could hold therapeutic potential for clinical translation.

12.
Nat Med ; 23(5): 579-589, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28368383

RESUMEN

Inflammatory bowel diseases (IBD), including Crohn's disease (CD) and ulcerative colitis (UC), are complex chronic inflammatory conditions of the gastrointestinal tract that are driven by perturbed cytokine pathways. Anti-tumor necrosis factor-α (TNF) antibodies are mainstay therapies for IBD. However, up to 40% of patients are nonresponsive to anti-TNF agents, which makes the identification of alternative therapeutic targets a priority. Here we show that, relative to healthy controls, inflamed intestinal tissues from patients with IBD express high amounts of the cytokine oncostatin M (OSM) and its receptor (OSMR), which correlate closely with histopathological disease severity. The OSMR is expressed in nonhematopoietic, nonepithelial intestinal stromal cells, which respond to OSM by producing various proinflammatory molecules, including interleukin (IL)-6, the leukocyte adhesion factor ICAM1, and chemokines that attract neutrophils, monocytes, and T cells. In an animal model of anti-TNF-resistant intestinal inflammation, genetic deletion or pharmacological blockade of OSM significantly attenuates colitis. Furthermore, according to an analysis of more than 200 patients with IBD, including two cohorts from phase 3 clinical trials of infliximab and golimumab, high pretreatment expression of OSM is strongly associated with failure of anti-TNF therapy. OSM is thus a potential biomarker and therapeutic target for IBD, and has particular relevance for anti-TNF-resistant patients.


Asunto(s)
Enfermedades Inflamatorias del Intestino/genética , Subunidad beta del Receptor de Oncostatina M/genética , Oncostatina M/genética , Adulto , Anciano , Animales , Anticuerpos Monoclonales/uso terapéutico , Estudios de Casos y Controles , Quimiocinas , Colitis/genética , Colitis/inmunología , Modelos Animales de Enfermedad , Femenino , Citometría de Flujo , Fármacos Gastrointestinales/uso terapéutico , Perfilación de la Expresión Génica , Humanos , Immunoblotting , Inmunohistoquímica , Inflamación , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Enfermedades Inflamatorias del Intestino/inmunología , Enfermedades Inflamatorias del Intestino/metabolismo , Infliximab/uso terapéutico , Molécula 1 de Adhesión Intercelular/inmunología , Interleucina-6/inmunología , Masculino , Ratones , Ratones Noqueados , Persona de Mediana Edad , Oncostatina M/inmunología , Oncostatina M/metabolismo , Subunidad beta del Receptor de Oncostatina M/inmunología , Subunidad beta del Receptor de Oncostatina M/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Adulto Joven
13.
Microbiol Spectr ; 5(1)2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-28102120

RESUMEN

The intestine is the tissue of the body with the highest constitutive exposure to foreign antigen and is also a common entry portal for many local and systemic pathogens. Therefore, the local immune system has the unenviable task of balancing efficient responses to dangerous pathogens with tolerance toward beneficial microbiota and food antigens. As in most tissues, the decision between tolerance and immunity is critically governed by the activity of local myeloid cells. However, the unique challenges posed by the intestinal environment have necessitated the development of several specialized mononuclear phagocyte populations with distinct phenotypic and functional characteristics that have vital roles in maintaining barrier function and immune homeostasis in the intestine. Intestinal mononuclear phagocyte populations, comprising dendritic cells and macrophages, are crucial for raising appropriate active immune responses against ingested pathogens. Recent technical advances, including microsurgical approaches allowing collection of cells migrating in intestinal lymph, intravital microscopy, and novel gene-targeting approaches, have led to clearer distinctions between mononuclear phagocyte populations in intestinal tissue. In this review, we present an overview of the various subpopulations of intestinal mononuclear phagocytes and discuss their phenotypic and functional characteristics. We also outline their roles in host protection from infection and their regulatory functions in maintaining immune tolerance toward beneficial intestinal antigens.


Asunto(s)
Enfermedades Intestinales/inmunología , Enfermedades Intestinales/patología , Intestinos/inmunología , Intestinos/fisiología , Leucocitos Mononucleares/inmunología , Fagocitos/inmunología , Animales , Humanos , Tolerancia Inmunológica , Inmunidad Innata
14.
Front Immunol ; 7: 240, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27446072

RESUMEN

The gastrointestinal tract presents a unique challenge to the mucosal immune system, which has to constantly monitor the vast surface for the presence of pathogens, while at the same time maintaining tolerance to beneficial or innocuous antigens. In the intestinal mucosa, specialized innate and adaptive immune components participate in directing appropriate immune responses toward these diverse challenges. Recent studies provide compelling evidence that the process of autophagy influences several aspects of mucosal immune responses. Initially described as a "self-eating" survival pathway that enables nutrient recycling during starvation, autophagy has now been connected to multiple cellular responses, including several aspects of immunity. Initial links between autophagy and host immunity came from the observations that autophagy can target intracellular bacteria for degradation. However, subsequent studies indicated that autophagy plays a much broader role in immune responses, as it can impact antigen processing, thymic selection, lymphocyte homeostasis, and the regulation of immunoglobulin and cytokine secretion. In this review, we provide a comprehensive overview of mucosal immune cells and discuss how autophagy influences many aspects of their physiology and function. We focus on cell type-specific roles of autophagy in the gut, with a particular emphasis on the effects of autophagy on the intestinal T cell compartment. We also provide a perspective on how manipulation of autophagy may potentially be used to treat mucosal inflammatory disorders.

15.
Trends Immunol ; 37(9): 569-570, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27452288

RESUMEN

Although best known for inducing inflammasome formation and pyroptosis in myeloid cells, increasing evidence highlights additional roles for Nod-like receptors (NLR) in nonhematopoietic cells. Two recent studies demonstrate that NLRX1 functions as an intrinsic tumor suppressor in intestinal epithelial cells (IEC), by regulating their responses to proliferative signals following intestinal injury.


Asunto(s)
Neoplasias del Colon/inmunología , Gastritis/inmunología , Mucosa Intestinal/metabolismo , Meningitis/inmunología , Proteínas Mitocondriales/metabolismo , Neumonía/inmunología , Proteínas Supresoras de Tumor/metabolismo , Animales , Proliferación Celular , Homeostasis , Humanos , Inmunidad Innata , Mucosa Intestinal/patología , Ratones , Ratones Noqueados , Proteínas Mitocondriales/genética , Transducción de Señal , Proteínas Supresoras de Tumor/genética
17.
Elife ; 5: e12444, 2016 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-26910010

RESUMEN

A polymorphism in the autophagy gene Atg16l1 is associated with susceptibility to inflammatory bowel disease (IBD); however, it remains unclear how autophagy contributes to intestinal immune homeostasis. Here, we demonstrate that autophagy is essential for maintenance of balanced CD4(+) T cell responses in the intestine. Selective deletion of Atg16l1 in T cells in mice resulted in spontaneous intestinal inflammation that was characterized by aberrant type 2 responses to dietary and microbiota antigens, and by a loss of Foxp3(+) Treg cells. Specific ablation of Atg16l1 in Foxp3(+) Treg cells in mice demonstrated that autophagy directly promotes their survival and metabolic adaptation in the intestine. Moreover, we also identify an unexpected role for autophagy in directly limiting mucosal TH2 cell expansion. These findings provide new insights into the reciprocal control of distinct intestinal TH cell responses by autophagy, with important implications for understanding and treatment of chronic inflammatory disorders.


Asunto(s)
Proteínas Portadoras/metabolismo , Enfermedades Inflamatorias del Intestino/patología , Linfocitos T Reguladores/inmunología , Células Th2/inmunología , Animales , Proteínas Relacionadas con la Autofagia , Proteínas Portadoras/genética , Eliminación de Gen , Ratones , Ratones Noqueados
18.
Trends Immunol ; 36(8): 442-50, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26166583

RESUMEN

While the functional importance of inflammasomes in blood-derived cell types is well established, it remains poorly understood how inflammasomes in nonhematopoietic cells contribute to mucosal immunity. Recent studies have revealed functional roles of inflammasomes - particularly NAIP/NLRC4, NLRP6, and noncanonical caspase-4 (caspase-11) - within epithelial cells of the gut in mucosal immune defense, inflammation, and tumorigenesis. Here, we review and discuss these findings in the broader context of tissue compartment-specific mucosal immunity. We propose several models whereby activities of the intestinal epithelial inflammasomes converge on mechanisms to remove compromised epithelial cells, maintain host-microbiota mutualism, and communicate with immune cells of the underlying lamina propria.


Asunto(s)
Inflamasomas/inmunología , Mucosa Intestinal/inmunología , Animales , Humanos , Inmunidad Mucosa
19.
Methods Mol Biol ; 1193: 199-211, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25151008

RESUMEN

Inflammatory bowel diseases (IBD) represent idiopathic chronic inflammatory disorders of the intestinal tract that are associated with aberrant immune responses against intestinal bacteria. Here, we describe two T cell-dependent models of experimental murine IBD. In the "T cell transfer" model, lymphopenic (scid or Rag (-/-) ) mice develop colitis upon adoptive transfer of naïve CD4(+) T cells. This model has also been extensively employed to identify mechanisms through which CD4(+)CD25(+) regulatory T cells suppress intestinal inflammation in vivo. We also describe a model of T cell-dependent IBD in immunocompetent mice, induced by infection with the intestinal bacterium Helicobacter hepaticus and concomitant treatment with a blocking αIL-10R mAb, which leads to the development of chronic inflammation of the caecum and colon (typhlocolitis). Both models reproduce many facets of human IBD pathology, including epithelial hyperplasia, goblet cell depletion, and leukocyte infiltration. These models provide reliable and tractable systems for the analyses of the induction and regulation of chronic inflammation in the gut.


Asunto(s)
Modelos Animales de Enfermedad , Enfermedades Inflamatorias del Intestino/inmunología , Ratones , Linfocitos T/inmunología , Linfocitos T/trasplante , Animales , Progresión de la Enfermedad , Citometría de Flujo , Helicobacter hepaticus/inmunología , Proteínas de Homeodominio/genética , Enfermedades Inflamatorias del Intestino/patología , Ratones Endogámicos C57BL , Ratones SCID , Ratones Transgénicos , Receptores de Interleucina-10/inmunología , Linfocitos T/citología
20.
Trends Immunol ; 35(11): 507-17, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25172617

RESUMEN

The immune system must constantly monitor the gastrointestinal tract for the presence of pathogens while tolerating trillions of commensal microbiota. It is clear that intestinal microbiota actively modulate the immune system to maintain a mutually beneficial relation, but the mechanisms that maintain homeostasis are not fully understood. Recent advances have begun to shed light on the cellular and molecular factors involved, revealing that a range of microbiota derivatives can influence host immune functions by targeting various cell types, including intestinal epithelial cells, mononuclear phagocytes, innate lymphoid cells, and B and T lymphocytes. Here, we review these findings, highlighting open questions and important challenges to overcome in translating this knowledge into new therapies for intestinal and systemic immune disorders.


Asunto(s)
Inmunomodulación , Intestinos/inmunología , Intestinos/microbiología , Microbiota , Animales , Diferenciación Celular , Homeostasis , Humanos , Inmunidad Innata , Inmunidad Mucosa , Inmunoglobulina A Secretora/biosíntesis , Inmunoglobulina A Secretora/inmunología , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Linfocitos/citología , Linfocitos/inmunología , Linfocitos/metabolismo , Fagocitos/inmunología , Fagocitos/metabolismo , Fagocitos/microbiología , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA