Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Immunol ; 211(3): 389-402, 2023 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-37272847

RESUMO

The impact of endemic parasitic infection on vaccine efficacy is an important consideration for vaccine development and deployment. We have examined whether intestinal infection with the natural murine helminth Heligmosomoides polygyrus bakeri alters Ag-specific Ab and cellular immune responses to oral and parenteral vaccination in mice. Oral vaccination of mice with a clinically relevant, live, attenuated, recombinant Salmonella vaccine expressing chicken egg OVA (Salmonella-OVA) induced the accumulation of activated, OVA-specific T effector cells rather than OVA-specific regulatory T cells in the GALT. Intestinal helminth infection significantly reduced Th1-skewed Ab responses to oral vaccination with Salmonella-OVA. Activated, adoptively transferred, OVA-specific CD4+ T cells accumulated in draining mesenteric lymph nodes of vaccinated mice, regardless of their helminth infection status. However, helminth infection increased the frequencies of adoptively transferred OVA-specific CD4+ T cells producing IL-4 and IL-10 in the mesenteric lymph node. Ab responses to the oral Salmonella-OVA vaccine were reduced in helminth-free mice adoptively transferred with OVA-specific CD4+ T cells harvested from mice with intestinal helminth infection. Intestinal helminth infection also significantly reduced Th2-skewed Ab responses to parenteral vaccination with OVA adsorbed to alum. These findings suggest that vaccine-specific CD4+ T cells induced in the context of helminth infection retain durable immunomodulatory properties and may promote blunted Ab responses to vaccination. They also underscore the potential need to treat parasitic infection before mass vaccination campaigns in helminth-endemic areas.


Assuntos
Helmintíase , Enteropatias Parasitárias , Camundongos , Animais , Eficácia de Vacinas , Linfócitos T CD4-Positivos , Vacinas Sintéticas , Ovalbumina , Camundongos Endogâmicos BALB C
2.
Immunity ; 55(11): 2027-2043.e9, 2022 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-36243007

RESUMO

T helper 17 (Th17) cells regulate mucosal barrier defenses but also promote multiple autoinflammatory diseases. Although many molecular determinants of Th17 cell differentiation have been elucidated, the transcriptional programs that sustain Th17 cells in vivo remain obscure. The transcription factor RORγt is critical for Th17 cell differentiation; however, it is not clear whether the closely related RORα, which is co-expressed in Th17 cells, has a distinct role. Here, we demonstrated that although dispensable for Th17 cell differentiation, RORα was necessary for optimal Th17 responses in peripheral tissues. The absence of RORα in T cells led to reductions in both RORγt expression and effector function among Th17 cells. Cooperative binding of RORα and RORγt to a previously unidentified Rorc cis-regulatory element was essential for Th17 lineage maintenance in vivo. These data point to a non-redundant role of RORα in Th17 lineage maintenance via reinforcement of the RORγt transcriptional program.


Assuntos
Encefalomielite Autoimune Experimental , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares , Diferenciação Celular , Encefalomielite Autoimune Experimental/metabolismo , Regulação da Expressão Gênica , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/genética , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/metabolismo , Células Th17/metabolismo , Fatores de Transcrição/metabolismo
3.
Immunity ; 54(8): 1745-1757.e7, 2021 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-34348118

RESUMO

Environmental enteric dysfunction (EED) is a gastrointestinal inflammatory disease caused by malnutrition and chronic infection. EED is associated with stunting in children and reduced efficacy of oral vaccines. To study the mechanisms of oral vaccine failure during EED, we developed a microbiota- and diet-dependent mouse EED model. Analysis of E. coli-labile toxin vaccine-specific CD4+ T cells in these mice revealed impaired CD4+ T cell responses in the small intestine and but not the lymph nodes. EED mice exhibited increased frequencies of small intestine-resident RORγT+FOXP3+ regulatory T (Treg) cells. Targeted deletion of RORγT from Treg cells restored small intestinal vaccine-specific CD4 T cell responses and vaccine-mediated protection upon challenge. However, ablation of RORγT+FOXP3+ Treg cells made mice more susceptible to EED-induced stunting. Our findings provide insight into the poor efficacy of oral vaccines in EED and highlight how RORγT+FOXP3+ Treg cells can regulate intestinal immunity while leaving systemic responses intact.


Assuntos
Toxinas Bacterianas/imunologia , Vacinas contra Escherichia coli/imunologia , Gastroenteropatias/imunologia , Intestino Delgado/imunologia , Linfócitos T Reguladores/imunologia , Administração Oral , Animais , Linhagem Celular , Modelos Animais de Doenças , Drosophila , Escherichia coli/imunologia , Feminino , Fatores de Transcrição Forkhead/metabolismo , Gastroenteropatias/microbiologia , Gastroenteropatias/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/metabolismo , Vacinação
5.
Immunohorizons ; 4(2): 57-71, 2020 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-32034085

RESUMO

The Tec kinase IL-2-inducible T cell kinase (ITK) regulates the expression of TCR-induced genes. Itk-/- T cell responses are impaired but not absent. ITK inhibition prevented colitis disease progression and impaired T cell migration to the colon in mice. To examine the function of ITK in T cell migration to the intestine, we examined the number of gut T cells in Itk-/- mice and then evaluated their expression of gut-homing receptors. Combined with in vitro murine T cell stimulation and in vivo migration assay using congenic B6 mice, we demonstrated an essential role for ITK in T cell migration to the intestine in mice. Reconstitution of Itk-/- mouse CD8+ T cells with IFN regulatory factor 4 restored gut-homing properties, providing mechanistic insight into the function of ITK-mediated signaling in CD8+ T cell migration to the intestinal mucosa in mice.


Assuntos
Linfócitos T CD8-Positivos/fisiologia , Quimiotaxia de Leucócito , Intestinos/imunologia , Proteínas Tirosina Quinases/metabolismo , Transdução de Sinais , Transferência Adotiva , Animais , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/transplante , Células Cultivadas , Fatores Reguladores de Interferon/metabolismo , Intestinos/virologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Tirosina Quinases/deficiência , Receptores de Retorno de Linfócitos/metabolismo , Rhadinovirus/fisiologia , Replicação Viral
6.
Cell ; 180(1): 79-91.e16, 2020 01 09.
Artigo em Inglês | MEDLINE | ID: mdl-31866067

RESUMO

Lymphoid cells that produce interleukin (IL)-17 cytokines protect barrier tissues from pathogenic microbes but are also prominent effectors of inflammation and autoimmune disease. T helper 17 (Th17) cells, defined by RORγt-dependent production of IL-17A and IL-17F, exert homeostatic functions in the gut upon microbiota-directed differentiation from naive CD4+ T cells. In the non-pathogenic setting, their cytokine production is regulated by serum amyloid A proteins (SAA1 and SAA2) secreted by adjacent intestinal epithelial cells. However, Th17 cell behaviors vary markedly according to their environment. Here, we show that SAAs additionally direct a pathogenic pro-inflammatory Th17 cell differentiation program, acting directly on T cells in collaboration with STAT3-activating cytokines. Using loss- and gain-of-function mouse models, we show that SAA1, SAA2, and SAA3 have distinct systemic and local functions in promoting Th17-mediated inflammatory diseases. These studies suggest that T cell signaling pathways modulated by the SAAs may be attractive targets for anti-inflammatory therapies.


Assuntos
Síndrome do Intestino Irritável/metabolismo , Proteína Amiloide A Sérica/metabolismo , Células Th17/metabolismo , Adulto , Animais , Doenças Autoimunes/metabolismo , Diferenciação Celular/imunologia , Citocinas/metabolismo , Encefalomielite Autoimune Experimental/metabolismo , Feminino , Humanos , Inflamação/metabolismo , Interleucina-17/metabolismo , Síndrome do Intestino Irritável/sangue , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/metabolismo , Células Th1 , Células Th17/imunologia
7.
Immunity ; 51(1): 185-197.e6, 2019 07 16.
Artigo em Inglês | MEDLINE | ID: mdl-31278058

RESUMO

Innate lymphoid cells (ILCs) promote tissue homeostasis and immune defense but also contribute to inflammatory diseases. ILCs exhibit phenotypic and functional plasticity in response to environmental stimuli, yet the transcriptional regulatory networks (TRNs) that control ILC function are largely unknown. Here, we integrate gene expression and chromatin accessibility data to infer regulatory interactions between transcription factors (TFs) and genes within intestinal type 1, 2, and 3 ILC subsets. We predicted the "core" TFs driving ILC identities, organized TFs into cooperative modules controlling distinct gene programs, and validated roles for c-MAF and BCL6 as regulators affecting type 1 and type 3 ILC lineages. The ILC network revealed alternative-lineage-gene repression, a mechanism that may contribute to reported plasticity between ILC subsets. By connecting TFs to genes, the TRNs suggest means to selectively regulate ILC effector functions, while our network approach is broadly applicable to identifying regulators in other in vivo cell populations.


Assuntos
Intestinos/fisiologia , Subpopulações de Linfócitos/fisiologia , Linfócitos/fisiologia , Animais , Diferenciação Celular , Linhagem da Célula , Plasticidade Celular , Montagem e Desmontagem da Cromatina , Repressão Epigenética , Redes Reguladoras de Genes , Imunidade Inata , Imunomodulação , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas Proto-Oncogênicas c-bcl-6/genética , Proteínas Proto-Oncogênicas c-maf/genética , Transcriptoma
8.
Nat Commun ; 10(1): 784, 2019 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-30770814

RESUMO

Innate lymphoid cells (ILC) are lymphocytes that lack an antigen-specific receptor and are preferentially localized in non-lymphoid tissues, such as mucosal barriers. In these locations ILC respond to tissue perturbations by producing factors that promote tissue repair and improve barrier integrity. We show that mice lacking the Tec kinase ITK have impaired intestinal tissue integrity, and a reduced ability to restore homeostasis after tissue damage. This defect is associated with a substantial loss of Type 2 ILC (ILC2) in the intestinal lamina propria. Adoptive transfer of bone marrow ILC2 precursors confirms a cell-intrinsic role for ITK. Intestinal ILC2 numbers in Itk-/- mice are restored by the administration of IL-2 complexes, also leading to improved intestinal tissue damage repair. Reduced Bcl-2 expression in intestinal Itk-/- ILC2 is also restored to WT levels after IL-2 complex treatment, indicating a tissue-specific role for ITK in ILC2 survival in the intestine.


Assuntos
Intestinos/citologia , Linfócitos/metabolismo , Proteínas Tirosina Quinases/metabolismo , Animais , Células Cultivadas , Colite/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Subunidade gama Comum de Receptores de Interleucina/genética , Subunidade gama Comum de Receptores de Interleucina/metabolismo , Interleucina-2/metabolismo , Interleucina-33/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Tirosina Quinases/genética
9.
Genome Res ; 29(3): 449-463, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30696696

RESUMO

Transcriptional regulatory networks (TRNs) provide insight into cellular behavior by describing interactions between transcription factors (TFs) and their gene targets. The assay for transposase-accessible chromatin (ATAC)-seq, coupled with TF motif analysis, provides indirect evidence of chromatin binding for hundreds of TFs genome-wide. Here, we propose methods for TRN inference in a mammalian setting, using ATAC-seq data to improve gene expression modeling. We test our methods in the context of T Helper Cell Type 17 (Th17) differentiation, generating new ATAC-seq data to complement existing Th17 genomic resources. In this resource-rich mammalian setting, our extensive benchmarking provides quantitative, genome-scale evaluation of TRN inference, combining ATAC-seq and RNA-seq data. We refine and extend our previous Th17 TRN, using our new TRN inference methods to integrate all Th17 data (gene expression, ATAC-seq, TF knockouts, and ChIP-seq). We highlight newly discovered roles for individual TFs and groups of TFs ("TF-TF modules") in Th17 gene regulation. Given the popularity of ATAC-seq, which provides high-resolution with low sample input requirements, we anticipate that our methods will improve TRN inference in new mammalian systems, especially in vivo, for cells directly from humans and animal models.


Assuntos
Cromatina/genética , Redes Reguladoras de Genes , Células Th17/metabolismo , Fatores de Transcrição/metabolismo , Diferenciação Celular , Cromatina/química , Montagem e Desmontagem da Cromatina , Humanos , Ligação Proteica , Software , Células Th17/citologia
10.
Nature ; 562(7725): 150, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29973715

RESUMO

Change History: This Article has been retracted; see accompanying Retraction. Corrected online 20 January: In this Article, author Frank Rigo was incorrectly listed with a middle initial; this has been corrected in the online versions of the paper.

11.
Sci Rep ; 7: 40775, 2017 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-28098217

RESUMO

Development of mice with hepatocyte knockout of lanosterol 14α-demethylase (HCyp51-/-) from cholesterol synthesis is characterized by the progressive onset of liver injury with ductular reaction and fibrosis. These changes begin during puberty and are generally more aggravated in the knockout females. However, a subgroup of (pre)pubertal knockout mice (runts) exhibits a pronounced male prevalent liver dysfunction characterized by downregulated amino acid metabolism and elevated Casp12. RORC transcriptional activity is diminished in livers of all runt mice, in correlation with the depletion of potential RORC ligands subsequent to CYP51 disruption. Further evidence for this comes from the global analysis that identified a crucial overlap between hepatic Cyp51-/- and Rorc-/- expression profiles. Additionally, the reduction in RORA and RORC transcriptional activity was greater in adult HCyp51-/- females than males, which correlates well with their downregulated amino and fatty acid metabolism. Overall, we identify a global and sex-dependent transcriptional de-regulation due to the block in cholesterol synthesis during development of the Cyp51 knockout mice and provide in vivo evidence that sterol intermediates downstream of lanosterol may regulate the hepatic RORC activity.


Assuntos
Colesterol/biossíntese , Família 51 do Citocromo P450/genética , Hepatócitos/metabolismo , Hepatopatias/etiologia , Hepatopatias/metabolismo , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/metabolismo , Transdução de Sinais , Animais , Modelos Animais de Doenças , Progressão da Doença , Feminino , Fibrose , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Hepatopatias/patologia , Masculino , Camundongos , Camundongos Knockout , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/genética , Caracteres Sexuais , Esteróis/metabolismo , Resposta a Proteínas não Dobradas
14.
Nature ; 528(7583): 517-22, 2015 Dec 24.
Artigo em Inglês | MEDLINE | ID: mdl-26675721

RESUMO

T helper 17 (TH17) lymphocytes protect mucosal barriers from infections, but also contribute to multiple chronic inflammatory diseases. Their differentiation is controlled by RORγt, a ligand-regulated nuclear receptor. Here we identify the RNA helicase DEAD-box protein 5 (DDX5) as a RORγt partner that coordinates transcription of selective TH17 genes, and is required for TH17-mediated inflammatory pathologies. Surprisingly, the ability of DDX5 to interact with RORγt and coactivate its targets depends on intrinsic RNA helicase activity and binding of a conserved nuclear long noncoding RNA (lncRNA), Rmrp, which is mutated in patients with cartilage-hair hypoplasia. A targeted Rmrp gene mutation in mice, corresponding to a gene mutation in cartilage-hair hypoplasia patients, altered lncRNA chromatin occupancy, and reduced the DDX5-RORγt interaction and RORγt target gene transcription. Elucidation of the link between Rmrp and the DDX5-RORγt complex reveals a role for RNA helicases and lncRNAs in tissue-specific transcriptional regulation, and provides new opportunities for therapeutic intervention in TH17-dependent diseases.


Assuntos
RNA Helicases DEAD-box/metabolismo , RNA Longo não Codificante/metabolismo , Células Th17/imunologia , Células Th17/metabolismo , Animais , Cromatina/genética , Cromatina/metabolismo , RNA Helicases DEAD-box/genética , Feminino , Regulação da Expressão Gênica/genética , Cabelo/anormalidades , Doença de Hirschsprung/genética , Humanos , Síndromes de Imunodeficiência/genética , Inflamação/imunologia , Inflamação/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mutação/genética , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/metabolismo , Especificidade de Órgãos , Osteocondrodisplasias/congênito , Osteocondrodisplasias/genética , Doenças da Imunodeficiência Primária , Ligação Proteica , RNA Longo não Codificante/genética , Transcrição Gênica/genética
15.
Cell ; 163(2): 381-93, 2015 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-26411290

RESUMO

RORγt(+) Th17 cells are important for mucosal defenses but also contribute to autoimmune disease. They accumulate in the intestine in response to microbiota and produce IL-17 cytokines. Segmented filamentous bacteria (SFB) are Th17-inducing commensals that potentiate autoimmunity in mice. RORγt(+) T cells were induced in mesenteric lymph nodes early after SFB colonization and distributed across different segments of the gastrointestinal tract. However, robust IL-17A production was restricted to the ileum, where SFB makes direct contact with the epithelium and induces serum amyloid A proteins 1 and 2 (SAA1/2), which promote local IL-17A expression in RORγt(+) T cells. We identified an SFB-dependent role of type 3 innate lymphoid cells (ILC3), which secreted IL-22 that induced epithelial SAA production in a Stat3-dependent manner. This highlights the critical role of tissue microenvironment in activating effector functions of committed Th17 cells, which may have important implications for how these cells contribute to inflammatory disease.


Assuntos
Microbioma Gastrointestinal , Interleucinas/metabolismo , Intestinos/imunologia , Receptores de Interleucina/metabolismo , Proteína Amiloide A Sérica/metabolismo , Células Th17/imunologia , Animais , Imunidade Inata , Interleucinas/imunologia , Intestinos/anatomia & histologia , Intestinos/microbiologia , Linfócitos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Receptores de Interleucina/imunologia , Transdução de Sinais , Interleucina 22
17.
J Exp Med ; 210(10): 1961-76, 2013 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-23999499

RESUMO

Dendritic cells (DCs) comprise distinct populations with specialized immune-regulatory functions. However, the environmental factors that determine the differentiation of these subsets remain poorly defined. Here, we report that retinoic acid (RA), a vitamin A derivative, controls the homeostasis of pre-DC (precursor of DC)-derived splenic CD11b(+)CD8α(-)Esam(high) DCs and the developmentally related CD11b(+)CD103(+) subset within the gut. Whereas mice deprived of RA signaling significantly lost both of these populations, neither pre-DC-derived CD11b(-)CD8α(+) and CD11b(-)CD103(+) nor monocyte-derived CD11b(+)CD8α(-)Esam(low) or CD11b(+)CD103(-) DC populations were deficient. In fate-tracking experiments, transfer of pre-DCs into RA-supplemented hosts resulted in near complete conversion of these cells into the CD11b(+)CD8α(-) subset, whereas transfer into vitamin A-deficient (VAD) hosts caused diversion to the CD11b(-)CD8α(+) lineage. As vitamin A is an essential nutrient, we evaluated retinoid levels in mice and humans after radiation-induced mucosal injury and found this conditioning led to an acute VAD state. Consequently, radiation led to a selective loss of both RA-dependent DC subsets and impaired class II-restricted auto and antitumor immunity that could be rescued by supplemental RA. These findings establish a critical role for RA in regulating the homeostasis of pre-DC-derived DC subsets and have implications for the management of patients with immune deficiencies resulting from malnutrition and irradiation.


Assuntos
Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Homeostase/imunologia , Tretinoína/metabolismo , Animais , Diferenciação Celular/imunologia , Proliferação de Células , Sobrevivência Celular , Células Dendríticas/citologia , Células Dendríticas/efeitos da radiação , Feminino , Antígenos de Histocompatibilidade Classe II/imunologia , Humanos , Imunofenotipagem , Mucosa Intestinal/metabolismo , Intestinos/imunologia , Intestinos/efeitos da radiação , Camundongos , Neoplasias/imunologia , Neoplasias/metabolismo , Especificidade de Órgãos/imunologia , Fenótipo , Receptores do Ácido Retinoico/metabolismo , Transdução de Sinais , Baço/imunologia , Baço/metabolismo , Baço/efeitos da radiação , Vitamina A/metabolismo , Irradiação Corporal Total/efeitos adversos
18.
J Biol Chem ; 288(20): 14080-14086, 2013 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-23546877

RESUMO

Members of the Amt family of channels mediate the transport of ammonium. The form of ammonium, NH3 or NH4(+), carried by these proteins remains controversial, and the mechanism by which they select against K(+) ions is unclear. We describe here a set of Escherichia coli AmtB proteins carrying mutations at the conserved twin-histidine site within the conduction pore that have altered substrate specificity and now transport K(+). Subsequent work established that AmtB-mediated K(+) uptake occurred against a concentration gradient and was membrane potential-dependent. These findings indicate that the twin-histidine element serves as a filter to prevent K(+) conduction and strongly support the notion that Amt proteins transport cations (NH4(+) or, in mutant proteins, K(+)) rather than NH3 gas molecules through their conduction pores.


Assuntos
Proteínas de Transporte de Cátions/metabolismo , Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Potássio/metabolismo , Transporte Biológico , Citoplasma/metabolismo , Relação Dose-Resposta a Droga , Concentração de Íons de Hidrogênio , Íons , Potenciais da Membrana , Mutação , Conformação Proteica , Compostos de Amônio Quaternário/metabolismo , Especificidade por Substrato
19.
Science ; 337(6098): 1115-9, 2012 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-22837383

RESUMO

Intestinal commensal bacteria induce protective and regulatory responses that maintain host-microbial mutualism. However, the contribution of tissue-resident commensals to immunity and inflammation at other barrier sites has not been addressed. We found that in mice, the skin microbiota have an autonomous role in controlling the local inflammatory milieu and tuning resident T lymphocyte function. Protective immunity to a cutaneous pathogen was found to be critically dependent on the skin microbiota but not the gut microbiota. Furthermore, skin commensals tuned the function of local T cells in a manner dependent on signaling downstream of the interleukin-1 receptor. These findings underscore the importance of the microbiota as a distinctive feature of tissue compartmentalization, and provide insight into mechanisms of immune system regulation by resident commensal niches in health and disease.


Assuntos
Metagenoma/imunologia , Dermatopatias Bacterianas/imunologia , Pele/imunologia , Pele/microbiologia , Linfócitos T/imunologia , Animais , Interações Hospedeiro-Patógeno , Humanos , Imunidade , Intestinos/imunologia , Intestinos/microbiologia , Intestinos/patologia , Camundongos , Dermatopatias Bacterianas/patologia
20.
J Clin Invest ; 121(11): 4503-15, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21965331

RESUMO

Tregs not only keep immune responses to autoantigens in check, but also restrain those directed toward pathogens and the commensal microbiota. Control of peripheral immune homeostasis by Tregs relies on their capacity to accumulate at inflamed sites and appropriately adapt to their local environment. To date, the factors involved in the control of these aspects of Treg physiology remain poorly understood. Here, we show that the canonical Th2 transcription factor GATA3 is selectively expressed in Tregs residing in barrier sites including the gastrointestinal tract and the skin. GATA3 expression in both murine and human Tregs was induced upon TCR and IL-2 stimulation. Although GATA3 was not required to sustain Treg homeostasis and function at steady state, GATA3 played a cardinal role in Treg physiology during inflammation. Indeed, the intrinsic expression of GATA3 by Tregs was required for their ability to accumulate at inflamed sites and to maintain high levels of Foxp3 expression in various polarized or inflammatory settings. Furthermore, our data indicate that GATA3 limits Treg polarization toward an effector T cell phenotype and acquisition of effector cytokines in inflamed tissues. Overall, our work reveals what we believe to be a new facet in the complex role of GATA3 in T cells and highlights what may be a fundamental role in controlling Treg physiology during inflammation.


Assuntos
Fatores de Transcrição Forkhead/imunologia , Fator de Transcrição GATA3/imunologia , Inflamação/imunologia , Inflamação/metabolismo , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Animais , Citocinas/deficiência , Citocinas/genética , Citocinas/imunologia , Feminino , Fator de Transcrição GATA3/deficiência , Fator de Transcrição GATA3/genética , Trato Gastrointestinal/imunologia , Trato Gastrointestinal/patologia , Humanos , Inflamação/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Receptores de Antígenos de Linfócitos T/metabolismo , Pele/imunologia , Pele/patologia , Linfócitos T Reguladores/patologia , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA