Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Annu Rev Neurosci ; 40: 21-49, 2017 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-28301775

RESUMEN

The microbiota is increasingly recognized for its ability to influence the development and function of the nervous system and several complex host behaviors. In this review, we discuss emerging roles for the gut microbiota in modulating host social and communicative behavior, stressor-induced behavior, and performance in learning and memory tasks. We summarize effects of the microbiota on host neurophysiology, including brain microstructure, gene expression, and neurochemical metabolism across regions of the amygdala, hippocampus, frontal cortex, and hypothalamus. We further assess evidence linking dysbiosis of the gut microbiota to neurobehavioral diseases, such as autism spectrum disorder and major depression, drawing upon findings from animal models and human trials. Finally, based on increasing associations between the microbiota, neurophysiology, and behavior, we consider whether investigating mechanisms underlying the microbiota-gut-brain axis could lead to novel approaches for treating particular neurological conditions.


Asunto(s)
Encéfalo/fisiología , Microbioma Gastrointestinal/fisiología , Tracto Gastrointestinal/fisiología , Trastornos Mentales/fisiopatología , Animales , Humanos , Trastornos Mentales/microbiología
2.
Immunity ; 44(3): 634-646, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26982365

RESUMEN

Physical separation between the mammalian immune system and commensal bacteria is necessary to limit chronic inflammation. However, selective species of commensal bacteria can reside within intestinal lymphoid tissues of healthy mammals. Here, we demonstrate that lymphoid-tissue-resident commensal bacteria (LRC) colonized murine dendritic cells and modulated their cytokine production. In germ-free and antibiotic-treated mice, LRCs colonized intestinal lymphoid tissues and induced multiple members of the IL-10 cytokine family, including dendritic-cell-derived IL-10 and group 3 innate lymphoid cell (ILC3)-derived IL-22. Notably, IL-10 limited the development of pro-inflammatory Th17 cell responses, and IL-22 production enhanced LRC colonization in the steady state. Furthermore, LRC colonization protected mice from lethal intestinal damage in an IL-10-IL-10R-dependent manner. Collectively, our data reveal a unique host-commensal-bacteria dialog whereby selective subsets of commensal bacteria interact with dendritic cells to facilitate tissue-specific responses that are mutually beneficial for both the host and the microbe.


Asunto(s)
Infecciones por Bordetella/inmunología , Bordetella/inmunología , Células Dendríticas/inmunología , Interleucina-10/metabolismo , Intestinos/inmunología , Tejido Linfoide/inmunología , Células Th17/inmunología , Animales , Células Cultivadas , Citocinas/metabolismo , Células Dendríticas/microbiología , Interleucina-10/genética , Interleucinas/genética , Interleucinas/metabolismo , Intestinos/microbiología , Tejido Linfoide/microbiología , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Microbiota , Receptores de Interleucina-10/genética , Receptores de Interleucina-10/metabolismo , Simbiosis/genética , Células Th17/microbiología , Interleucina-22
3.
Neurobiol Dis ; 136: 104714, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31846737

RESUMEN

Intestinal inflammatory disorders are associated with neurophysiological and behavioral symptoms. Conversely, many disorders of the central nervous system (CNS) are accompanied by intestinal complications. These observations suggest that intestinal and nervous system physiologies are functionally linked. Indeed, a growing body of literature has revealed multiple pathways mediating bidirectional communication between the intestine and the CNS, collectively referred to as the gut-brain axis. In particular, microbes naturally colonizing the mammalian gastrointestinal (GI) tract, termed the gut microbiota, not only correlate with but also play a causative role in regulating CNS function, development and host behavior. Despite these findings, our understanding of the cellular and molecular mechanisms that mediate gut-brain communication remains in its infancy. However, members of the gut microbiota have been established as potent modulators of intestinal, systemic and CNS-resident immune cell function, suggesting that gut-brain interactions may involve the host immune system. Multiple CNS disorders with gut microbiota associations, including neuroinflammatory, neuropsychiatric and neurodegenerative disorders, also have significant inflammatory manifestations. In this review, I discuss recent advances exploring the role of microbiota-immune interactions as a critical regulator of the gut-brain axis in the context of CNS and related disorders.


Asunto(s)
Encéfalo/inmunología , Enfermedades del Sistema Nervioso Central/inmunología , Microbioma Gastrointestinal/fisiología , Tracto Gastrointestinal/inmunología , Animales , Encéfalo/metabolismo , Enfermedades del Sistema Nervioso Central/metabolismo , Tracto Gastrointestinal/metabolismo , Humanos , Mediadores de Inflamación/inmunología , Mediadores de Inflamación/metabolismo , Microbiota/fisiología
4.
Nature ; 498(7452): 113-7, 2013 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-23698371

RESUMEN

Innate lymphoid cells (ILCs) are a recently characterized family of immune cells that have critical roles in cytokine-mediated regulation of intestinal epithelial cell barrier integrity. Alterations in ILC responses are associated with multiple chronic human diseases, including inflammatory bowel disease, implicating a role for ILCs in disease pathogenesis. Owing to an inability to target ILCs selectively, experimental studies assessing ILC function have predominantly used mice lacking adaptive immune cells. However, in lymphocyte-sufficient hosts ILCs are vastly outnumbered by CD4(+) T cells, which express similar profiles of effector cytokines. Therefore, the function of ILCs in the presence of adaptive immunity and their potential to influence adaptive immune cell responses remain unknown. To test this, we used genetic or antibody-mediated depletion strategies to target murine ILCs in the presence of an adaptive immune system. We show that loss of retinoic-acid-receptor-related orphan receptor-γt-positive (RORγt(+)) ILCs was associated with dysregulated adaptive immune cell responses against commensal bacteria and low-grade systemic inflammation. Remarkably, ILC-mediated regulation of adaptive immune cells occurred independently of interleukin (IL)-17A, IL-22 or IL-23. Genome-wide transcriptional profiling and functional analyses revealed that RORγt(+) ILCs express major histocompatibility complex class II (MHCII) and can process and present antigen. However, rather than inducing T-cell proliferation, ILCs acted to limit commensal bacteria-specific CD4(+) T-cell responses. Consistent with this, selective deletion of MHCII in murine RORγt(+) ILCs resulted in dysregulated commensal bacteria-dependent CD4(+) T-cell responses that promoted spontaneous intestinal inflammation. These data identify that ILCs maintain intestinal homeostasis through MHCII-dependent interactions with CD4(+) T cells that limit pathological adaptive immune cell responses to commensal bacteria.


Asunto(s)
Bacterias/inmunología , Linfocitos T CD4-Positivos/inmunología , Inmunidad Innata/inmunología , Intestinos/inmunología , Intestinos/microbiología , Simbiosis , Animales , Presentación de Antígeno/inmunología , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/patología , Proliferación Celular , Antígenos de Histocompatibilidad Clase II/inmunología , Antígenos de Histocompatibilidad Clase II/metabolismo , Humanos , Inflamación/patología , Interleucina-17/metabolismo , Interleucina-23/metabolismo , Interleucinas/metabolismo , Intestinos/patología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Interleucina-22
5.
Immunol Rev ; 260(1): 35-49, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24942680

RESUMEN

The mammalian gastrointestinal (GI) tract is colonized by trillions of beneficial commensal bacteria that are essential for promoting normal intestinal physiology. While the majority of commensal bacteria are found in the intestinal lumen, many species have also adapted to colonize different anatomical locations in the intestine, including the surface of intestinal epithelial cells (IECs) and the interior of gut-associated lymphoid tissues. These distinct tissue localization patterns permit unique interactions with the mammalian immune system and collectively influence intestinal immune cell homeostasis. Conversely, dysregulated localization of commensal bacteria can lead to inappropriate activation of the immune system and is associated with numerous chronic infectious, inflammatory, and metabolic diseases. Therefore, regulatory mechanisms that control proper anatomical containment of commensal bacteria are essential to maintain tissue homeostasis and limit pathology. In this review, we propose that commensal bacteria associated with the mammalian GI tract can be anatomically defined as (i) luminal, (ii) epithelial-associated, or (iii) lymphoid tissue-resident, and we discuss the role and regulation of these microbial populations in health and disease.


Asunto(s)
Susceptibilidad a Enfermedades , Tracto Gastrointestinal/inmunología , Tracto Gastrointestinal/microbiología , Homeostasis , Tejido Linfoide/inmunología , Microbiota , Membrana Mucosa/inmunología , Membrana Mucosa/microbiología , Animales , Tracto Gastrointestinal/virología , Humanos , Inmunidad Mucosa , Tejido Linfoide/citología , Membrana Mucosa/virología
6.
J Biol Chem ; 290(15): 9886-95, 2015 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-25694432

RESUMEN

Protein tyrosine phosphatase α (PTPα) promotes integrin-stimulated cell migration in part through the role of Src-phosphorylated PTPα-Tyr(P)-789 in recruiting and localizing p130Cas to focal adhesions. The growth factor IGF-1 also stimulates PTPα-Tyr-789 phosphorylation to positively regulate cell movement. This is in contrast to integrin-induced PTPα phosphorylation, that induced by IGF-1 can occur in cells lacking Src family kinases (SFKs), indicating that an unknown kinase distinct from SFKs can target PTPα. We show that this IGF-1-stimulated tyrosine kinase is Abl. We found that PTPα binds to the scaffold protein RACK1 and that RACK1 coordinates the IGF-1 receptor, PTPα, and Abl in a complex to enable IGF-1-stimulated and Abl-dependent PTPα-Tyr-789 phosphorylation. In cells expressing SFKs, IGF-1-stimulated phosphorylation of PTPα is mediated by RACK1 but is Abl-independent. Furthermore, expressing the SFKs Src and Fyn in SFK-deficient cells switches IGF-1-induced PTPα phosphorylation to occur in an Abl-independent manner, suggesting that SFK activity dominantly regulates IGF-1/IGF-1 receptor signaling to PTPα. RACK1 is a molecular scaffold that integrates growth factor and integrin signaling, and our identification of PTPα as a RACK1 binding protein suggests that RACK1 may coordinate PTPα-Tyr-789 phosphorylation in these signaling networks to promote cell migration.


Asunto(s)
Proteínas de Unión al GTP/metabolismo , Factor I del Crecimiento Similar a la Insulina/farmacología , Proteínas de Neoplasias/metabolismo , Proteínas Proto-Oncogénicas c-abl/metabolismo , Proteínas Tirosina Fosfatasas Clase 4 Similares a Receptores/metabolismo , Receptores de Superficie Celular/metabolismo , Animales , Línea Celular , Línea Celular Tumoral , Células Cultivadas , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Proteínas de Unión al GTP/genética , Humanos , Immunoblotting , Células MCF-7 , Ratones , Proteínas de Neoplasias/genética , Fosforilación/efectos de los fármacos , Unión Proteica , Proteínas Proto-Oncogénicas c-abl/genética , Pirimidinas/farmacología , Interferencia de ARN , Receptor IGF Tipo 1/genética , Receptor IGF Tipo 1/metabolismo , Proteínas Tirosina Fosfatasas Clase 4 Similares a Receptores/genética , Receptores de Cinasa C Activada , Receptores de Superficie Celular/genética , Tirosina/metabolismo , Familia-src Quinasas/genética , Familia-src Quinasas/metabolismo
7.
J Immunol ; 193(7): 3717-25, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25156365

RESUMEN

Type 2 inflammation underlies allergic diseases such as atopic dermatitis, which is characterized by the accumulation of basophils and group 2 innate lymphoid cells (ILC2s) in inflamed skin lesions. Although murine studies have demonstrated that cutaneous basophil and ILC2 responses are dependent on thymic stromal lymphopoietin, whether these cell populations interact to regulate the development of cutaneous type 2 inflammation is poorly defined. In this study, we identify that basophils and ILC2s significantly accumulate in inflamed human and murine skin and form clusters not observed in control skin. We demonstrate that murine basophil responses precede ILC2 responses and that basophils are the dominant IL-4-enhanced GFP-expressing cell type in inflamed skin. Furthermore, basophils and IL-4 were necessary for the optimal accumulation of ILC2s and induction of atopic dermatitis-like disease. We show that ILC2s express IL-4Rα and proliferate in an IL-4-dependent manner. Additionally, basophil-derived IL-4 was required for cutaneous ILC2 responses in vivo and directly regulated ILC2 proliferation ex vivo. Collectively, these data reveal a previously unrecognized role for basophil-derived IL-4 in promoting ILC2 responses during cutaneous inflammation.


Asunto(s)
Basófilos/inmunología , Dermatitis Atópica/inmunología , Inmunidad Innata , Linfocitos/inmunología , Piel/inmunología , Animales , Basófilos/patología , Proliferación Celular , Citocinas/genética , Citocinas/inmunología , Dermatitis Atópica/genética , Dermatitis Atópica/patología , Femenino , Humanos , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Interleucina-4/genética , Interleucina-4/inmunología , Linfocitos/patología , Masculino , Ratones , Ratones Noqueados , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/inmunología , Piel/patología , Linfopoyetina del Estroma Tímico
8.
PLoS Pathog ; 9(6): e1003400, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23762026

RESUMEN

Inflammasome activation is important for antimicrobial defense because it induces cell death and regulates the secretion of IL-1 family cytokines, which play a critical role in inflammatory responses. The inflammasome activates caspase-1 to process and secrete IL-1ß. However, the mechanisms governing IL-1α release are less clear. Recently, a non-canonical inflammasome was described that activates caspase-11 and mediates pyroptosis and release of IL-1α and IL-1ß. Caspase-11 activation in response to Gram-negative bacteria requires Toll-like receptor 4 (TLR4) and TIR-domain-containing adaptor-inducing interferon-ß (TRIF)-dependent interferon production. Whether additional bacterial signals trigger caspase-11 activation is unknown. Many bacterial pathogens use specialized secretion systems to translocate effector proteins into the cytosol of host cells. These secretion systems can also deliver flagellin into the cytosol, which triggers caspase-1 activation and pyroptosis. However, even in the absence of flagellin, these secretion systems induce inflammasome activation and the release of IL-1α and IL-1ß, but the inflammasome pathways that mediate this response are unclear. We observe rapid IL-1α and IL-1ß release and cell death in response to the type IV or type III secretion systems of Legionella pneumophila and Yersinia pseudotuberculosis. Unlike IL-1ß, IL-1α secretion does not require caspase-1. Instead, caspase-11 activation is required for both IL-1α secretion and cell death in response to the activity of these secretion systems. Interestingly, whereas caspase-11 promotes IL-1ß release in response to the type IV secretion system through the NLRP3/ASC inflammasome, caspase-11-dependent release of IL-1α is independent of both the NAIP5/NLRC4 and NLRP3/ASC inflammasomes as well as TRIF and type I interferon signaling. Furthermore, we find both overlapping and non-redundant roles for IL-1α and IL-1ß in mediating neutrophil recruitment and bacterial clearance in response to pulmonary infection by L. pneumophila. Our findings demonstrate that virulent, but not avirulent, bacteria trigger a rapid caspase-11-dependent innate immune response important for host defense.


Asunto(s)
Sistemas de Secreción Bacterianos/inmunología , Caspasas/inmunología , Citosol/inmunología , Legionella pneumophila/inmunología , Enfermedad de los Legionarios/inmunología , Macrófagos/inmunología , Animales , Proteínas Reguladoras de la Apoptosis/inmunología , Proteínas de Unión al Calcio/inmunología , Proteínas Portadoras/inmunología , Caspasas/genética , Caspasas Iniciadoras , Línea Celular , Citosol/microbiología , Activación Enzimática/inmunología , Inmunidad Innata/inmunología , Inflamasomas/genética , Inflamasomas/inmunología , Interleucina-1alfa/inmunología , Interleucina-1beta/inmunología , Legionella pneumophila/patogenicidad , Enfermedad de los Legionarios/microbiología , Enfermedad de los Legionarios/patología , Macrófagos/microbiología , Macrófagos/patología , Ratones , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR
9.
Infect Immun ; 82(10): 4325-36, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25092908

RESUMEN

Legionella pneumophila, an intracellular pathogen responsible for the severe pneumonia Legionnaires' disease, uses its dot/icm-encoded type IV secretion system (T4SS) to translocate effector proteins that promote its survival and replication into the host cell cytosol. However, by introducing bacterial products into the host cytosol, L. pneumophila also activates cytosolic immunosurveillance pathways, thereby triggering robust proinflammatory responses that mediate the control of infection. Thus, the pulmonary cell types that L. pneumophila infects not only may act as an intracellular niche that facilitates its pathogenesis but also may contribute to the immune response against L. pneumophila. The identity of these host cells remains poorly understood. Here, we developed a strain of L. pneumophila producing a fusion protein consisting of ß-lactamase fused to the T4SS-translocated effector RalF, which allowed us to track cells injected by the T4SS. Our data reveal that alveolar macrophages and neutrophils both are the primary recipients of T4SS-translocated effectors and harbor viable L. pneumophila during pulmonary infection of mice. Moreover, both alveolar macrophages and neutrophils from infected mice produced tumor necrosis factor and interleukin-1α in response to T4SS-sufficient, but not T4SS-deficient, L. pneumophila. Collectively, our data suggest that alveolar macrophages and neutrophils are both an intracellular reservoir for L. pneumophila and a source of proinflammatory cytokines that contribute to the host immune response against L. pneumophila during pulmonary infection.


Asunto(s)
Sistemas de Secreción Bacterianos , Legionella pneumophila/inmunología , Legionella pneumophila/fisiología , Macrófagos Alveolares/inmunología , Macrófagos Alveolares/microbiología , Neutrófilos/inmunología , Neutrófilos/microbiología , Animales , Citosol/metabolismo , Citosol/microbiología , Modelos Animales de Enfermedad , Femenino , Interacciones Huésped-Patógeno , Interleucina-1alfa/metabolismo , Enfermedad de los Legionarios/inmunología , Enfermedad de los Legionarios/microbiología , Ratones , Ratones Endogámicos C57BL , Factor de Necrosis Tumoral alfa/metabolismo
10.
Science ; 368(6487): 186-189, 2020 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-32273468

RESUMEN

Bleeding and altered iron distribution occur in multiple gastrointestinal diseases, but the importance and regulation of these changes remain unclear. We found that hepcidin, the master regulator of systemic iron homeostasis, is required for tissue repair in the mouse intestine after experimental damage. This effect was independent of hepatocyte-derived hepcidin or systemic iron levels. Rather, we identified conventional dendritic cells (cDCs) as a source of hepcidin that is induced by microbial stimulation in mice, prominent in the inflamed intestine of humans, and essential for tissue repair. cDC-derived hepcidin acted on ferroportin-expressing phagocytes to promote local iron sequestration, which regulated the microbiota and consequently facilitated intestinal repair. Collectively, these results identify a pathway whereby cDC-derived hepcidin promotes mucosal healing in the intestine through means of nutritional immunity.


Asunto(s)
Células Dendríticas/metabolismo , Microbioma Gastrointestinal , Hepcidinas/metabolismo , Enfermedades Intestinales/microbiología , Mucosa Intestinal/microbiología , Mucosa Intestinal/fisiología , Hierro/metabolismo , Animales , Proteínas de Transporte de Catión/metabolismo , Trasplante de Microbiota Fecal , Eliminación de Gen , Hepcidinas/genética , Homeostasis , Ratones , Ratones Mutantes , Fagocitos/metabolismo
11.
Nat Microbiol ; 4(12): 2064-2073, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31477894

RESUMEN

The gut microbiota regulates levels of serotonin (5-hydroxytryptamine (5-HT)) in the intestinal epithelium and lumen1-5. However, whether 5-HT plays a functional role in bacteria from the gut microbiota remains unknown. We demonstrate that elevating levels of intestinal lumenal 5-HT by oral supplementation or genetic deficiency in the host 5-HT transporter (SERT) increases the relative abundance of spore-forming members of the gut microbiota, which were previously reported to promote host 5-HT biosynthesis. Within this microbial community, we identify Turicibacter sanguinis as a gut bacterium that expresses a neurotransmitter sodium symporter-related protein with sequence and structural homology to mammalian SERT. T. sanguinis imports 5-HT through a mechanism that is inhibited by the selective 5-HT reuptake inhibitor fluoxetine. 5-HT reduces the expression of sporulation factors and membrane transporters in T. sanguinis, which is reversed by fluoxetine exposure. Treating T. sanguinis with 5-HT or fluoxetine modulates its competitive colonization in the gastrointestinal tract of antibiotic-treated mice. In addition, fluoxetine reduces the membership of T. sanguinis in the gut microbiota of conventionally colonized mice. Host association with T. sanguinis alters intestinal expression of multiple gene pathways, including those important for lipid and steroid metabolism, with corresponding reductions in host systemic triglyceride levels and inguinal adipocyte size. Together, these findings support the notion that select bacteria indigenous to the gut microbiota signal bidirectionally with the host serotonergic system to promote their fitness in the intestine.


Asunto(s)
Fluoxetina/administración & dosificación , Microbioma Gastrointestinal/efectos de los fármacos , Intestinos/microbiología , Inhibidores Selectivos de la Recaptación de Serotonina/administración & dosificación , Agonistas de Receptores de Serotonina/administración & dosificación , Serotonina/administración & dosificación , Administración Oral , Animales , Bacterias/efectos de los fármacos , Heces/química , Heces/microbiología , Femenino , Firmicutes/efectos de los fármacos , Variación Genética , Interacciones Microbiota-Huesped/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Organismos Libres de Patógenos Específicos
12.
Nat Neurosci ; 20(2): 145-155, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28092661

RESUMEN

The diverse collection of microorganisms that inhabit the gastrointestinal tract, collectively called the gut microbiota, profoundly influences many aspects of host physiology, including nutrient metabolism, resistance to infection and immune system development. Studies investigating the gut-brain axis demonstrate a critical role for the gut microbiota in orchestrating brain development and behavior, and the immune system is emerging as an important regulator of these interactions. Intestinal microbes modulate the maturation and function of tissue-resident immune cells in the CNS. Microbes also influence the activation of peripheral immune cells, which regulate responses to neuroinflammation, brain injury, autoimmunity and neurogenesis. Accordingly, both the gut microbiota and immune system are implicated in the etiopathogenesis or manifestation of neurodevelopmental, psychiatric and neurodegenerative diseases, such as autism spectrum disorder, depression and Alzheimer's disease. In this review, we discuss the role of CNS-resident and peripheral immune pathways in microbiota-gut-brain communication during health and neurological disease.


Asunto(s)
Trastorno del Espectro Autista/inmunología , Encéfalo/inmunología , Tracto Gastrointestinal/inmunología , Microbiota/inmunología , Enfermedades del Sistema Nervioso/inmunología , Animales , Autoinmunidad/fisiología , Humanos , Enfermedades del Sistema Nervioso/fisiopatología
13.
Science ; 348(6238): 1031-5, 2015 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-25908663

RESUMEN

Inflammatory CD4(+) T cell responses to self or commensal bacteria underlie the pathogenesis of autoimmunity and inflammatory bowel disease (IBD), respectively. Although selection of self-specific T cells in the thymus limits responses to mammalian tissue antigens, the mechanisms that control selection of commensal bacteria-specific T cells remain poorly understood. Here, we demonstrate that group 3 innate lymphoid cell (ILC3)-intrinsic expression of major histocompatibility complex class II (MHCII) is regulated similarly to thymic epithelial cells and that MHCII(+) ILC3s directly induce cell death of activated commensal bacteria-specific T cells. Further, MHCII on colonic ILC3s was reduced in pediatric IBD patients. Collectively, these results define a selection pathway for commensal bacteria-specific CD4(+) T cells in the intestine and suggest that this process is dysregulated in human IBD.


Asunto(s)
Bacterias/inmunología , Linfocitos T CD4-Positivos/inmunología , Colon/microbiología , Antígenos de Histocompatibilidad Clase II/inmunología , Inmunidad Innata , Enfermedades Inflamatorias del Intestino/microbiología , Animales , Apoptosis/inmunología , Autoinmunidad , Femenino , Flagelina/genética , Flagelina/inmunología , Humanos , Enfermedades Inflamatorias del Intestino/inmunología , Activación de Linfocitos , Masculino , Ratones , Ratones Endogámicos C57BL , Simbiosis , Timo/inmunología
14.
J Clin Invest ; 124(5): 1945-55, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24667637

RESUMEN

Inflammatory bowel disease (IBD) pathogenesis is associated with dysregulated CD4⁺ Th cell responses, with intestinal homeostasis depending on the balance between IL-17-producing Th17 and Foxp3⁺ Tregs. Differentiation of naive T cells into Th17 and Treg subsets is associated with specific gene expression profiles; however, the contribution of epigenetic mechanisms to controlling Th17 and Treg differentiation remains unclear. Using a murine T cell transfer model of colitis, we found that T cell-intrinsic expression of the histone lysine methyltransferase G9A was required for development of pathogenic T cells and intestinal inflammation. G9A-mediated dimethylation of histone H3 lysine 9 (H3K9me2) restricted Th17 and Treg differentiation in vitro and in vivo. H3K9me2 was found at high levels in naive Th cells and was lost following Th cell activation. Loss of G9A in naive T cells was associated with increased chromatin accessibility and heightened sensitivity to TGF-ß1. Pharmacological inhibition of G9A methyltransferase activity in WT T cells promoted Th17 and Treg differentiation. Our data indicate that G9A-dependent H3K9me2 is a homeostatic epigenetic checkpoint that regulates Th17 and Treg responses by limiting chromatin accessibility and TGF-ß1 responsiveness, suggesting G9A as a therapeutic target for treating intestinal inflammation.


Asunto(s)
Diferenciación Celular/inmunología , Colitis/inmunología , N-Metiltransferasa de Histona-Lisina/inmunología , Linfocitos T Reguladores/inmunología , Células Th17/inmunología , Animales , Diferenciación Celular/genética , Cromatina/genética , Cromatina/inmunología , Colitis/tratamiento farmacológico , Colitis/genética , Colitis/patología , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Antígenos de Histocompatibilidad/genética , Antígenos de Histocompatibilidad/inmunología , N-Metiltransferasa de Histona-Lisina/antagonistas & inhibidores , N-Metiltransferasa de Histona-Lisina/genética , Histonas/genética , Histonas/inmunología , Metilación/efectos de los fármacos , Ratones , Ratones Noqueados , Linfocitos T Reguladores/patología , Células Th17/patología , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/inmunología
15.
Science ; 336(6086): 1321-5, 2012 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-22674331

RESUMEN

The mammalian intestinal tract is colonized by trillions of beneficial commensal bacteria that are anatomically restricted to specific niches. However, the mechanisms that regulate anatomical containment remain unclear. Here, we show that interleukin-22 (IL-22)-producing innate lymphoid cells (ILCs) are present in intestinal tissues of healthy mammals. Depletion of ILCs resulted in peripheral dissemination of commensal bacteria and systemic inflammation, which was prevented by administration of IL-22. Disseminating bacteria were identified as Alcaligenes species originating from host lymphoid tissues. Alcaligenes was sufficient to promote systemic inflammation after ILC depletion in mice, and Alcaligenes-specific systemic immune responses were associated with Crohn's disease and progressive hepatitis C virus infection in patients. Collectively, these data indicate that ILCs regulate selective containment of lymphoid-resident bacteria to prevent systemic inflammation associated with chronic diseases.


Asunto(s)
Alcaligenes/fisiología , Interleucinas/inmunología , Intestinos/inmunología , Linfocitos/inmunología , Tejido Linfoide/inmunología , Tejido Linfoide/microbiología , Adulto , Alcaligenes/inmunología , Alcaligenes/aislamiento & purificación , Animales , Traslocación Bacteriana , Enfermedad de Crohn/inmunología , Enfermedad de Crohn/microbiología , Hepatitis C Crónica/inmunología , Hepatitis C Crónica/microbiología , Humanos , Inmunidad Innata , Inflamación , Interleucinas/administración & dosificación , Interleucinas/biosíntesis , Intestinos/microbiología , Complejo de Antígeno L1 de Leucocito/metabolismo , Hígado/microbiología , Ganglios Linfáticos/inmunología , Macaca mulatta , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Bazo/microbiología , Adulto Joven , Interleucina-22
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA