Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Nat Immunol ; 15(1): 80-7, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24241693

RESUMO

The subclassification of immunology into innate and adaptive immunity is challenged by innate-like T lymphocytes that use innate receptors to respond rapidly to stress despite expressing T cell antigen receptors (TCRs), a hallmark of adaptive immunity. In studies that explain how such cells can straddle innate and adaptive immunity, we found that signaling via antigen receptors, whose conventional role is to facilitate clonal T cell activation, was critical for the development of innate-like T cells but then was rapidly attenuated, which accommodated the cells' innate responsiveness. These findings permitted the identification of a previously unknown innate-like T cell subset and indicate that T cell hyporesponsiveness, a state traditionally linked to tolerance, may be fundamental to T cells entering the innate compartment and thereby providing lymphoid stress surveillance.


Assuntos
Imunidade Adaptativa/imunologia , Imunidade Inata/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Subpopulações de Linfócitos T/imunologia , Animais , Animais Recém-Nascidos , Células Cultivadas , Citometria de Fluxo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Interleucina-17/imunologia , Interleucina-17/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares/genética , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares/imunologia , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares/metabolismo , Receptores de Antígenos de Linfócitos T/metabolismo , Receptores de Antígenos de Linfócitos T gama-delta/imunologia , Receptores de Antígenos de Linfócitos T gama-delta/metabolismo , Transdução de Sinais/imunologia , Subpopulações de Linfócitos T/metabolismo , Proteína-Tirosina Quinase ZAP-70/imunologia , Proteína-Tirosina Quinase ZAP-70/metabolismo
2.
Immunity ; 36(3): 427-37, 2012 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-22425250

RESUMO

The thymic medulla provides a specialized microenvironment for the negative selection of T cells, with the presence of autoimmune regulator (Aire)-expressing medullary thymic epithelial cells (mTECs) during the embryonic-neonatal period being both necessary and sufficient to establish long-lasting tolerance. Here we showed that emergence of the first cohorts of Aire(+) mTECs at this key developmental stage, prior to αß T cell repertoire selection, was jointly directed by Rankl(+) lymphoid tissue inducer cells and invariant Vγ5(+) dendritic epidermal T cell (DETC) progenitors that are the first thymocytes to express the products of gene rearrangement. In turn, generation of Aire(+) mTECs then fostered Skint-1-dependent, but Aire-independent, DETC progenitor maturation and the emergence of an invariant DETC repertoire. Hence, our data attributed a functional importance to the temporal development of Vγ5(+) γδ T cells during thymus medulla formation for αß T cell tolerance induction and demonstrated a Rank-mediated reciprocal link between DETC and Aire(+) mTEC maturation.


Assuntos
Células Precursoras de Linfócitos T/citologia , Células Precursoras de Linfócitos T/imunologia , Receptor Ativador de Fator Nuclear kappa-B/imunologia , Receptores de Antígenos de Linfócitos T gama-delta/metabolismo , Fatores de Transcrição/imunologia , Animais , Diferenciação Celular/imunologia , Microambiente Celular , Células Epiteliais/imunologia , Feminino , Feto/citologia , Feto/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Gravidez , Transdução de Sinais/imunologia , Timo/citologia , Timo/imunologia , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética , Proteína AIRE
3.
Immunity ; 35(1): 59-68, 2011 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-21737317

RESUMO

Murine T cell development begins with the generation of a unique Vγ5(+)Vδ1(+) epidermal γδ T cell compartment and a unique, more broadly distributed Vγ6(+)Vδ1(+) subset that is an important source of interleukin-17 (IL-17). This study showed that these respective functional programs were determined by Skint-1, a thymic epithelial cell determinant. By engaging Skint-1(+) cells, Vγ5(+)Vδ1(+) thymocytes induced an Egr3-mediated pathway, provoking differentiation and the potential to produce IFN-γ while suppressing the γδ T cell lineage factor, Sox13, and a RORγt transcription factor-associated IL-17-producing capacity. Hence, the functions of the earliest T cells are substantially preprogrammed in the thymus. Additionally, the phenotype of Skint-1-selected fetal thymocytes permitted identification in the adult thymus of an analogous gene regulatory network regulated by the γδ T cell receptor. Hence, these observations describe a molecular pathway by which distinct stress-responsive lymphocyte repertoires may emerge throughout ontogeny and offer parallels with emerging perspectives on the functional selection of other lymphoid cells.


Assuntos
Células Epiteliais/metabolismo , Imunoglobulinas/metabolismo , Receptores de Antígenos de Linfócitos T gama-delta/metabolismo , Linfócitos T/metabolismo , Timo/patologia , Animais , Autoantígenos/metabolismo , Diferenciação Celular , Células Cultivadas , Proteína 3 de Resposta de Crescimento Precoce/metabolismo , Células Epiteliais/imunologia , Células Epiteliais/patologia , Regulação da Expressão Gênica/imunologia , Imunoglobulinas/genética , Imunoglobulinas/imunologia , Interferon gama/metabolismo , Interleucina-17/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Mutação/genética , Receptores de Antígenos de Linfócitos T gama-delta/imunologia , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Linfócitos T/imunologia , Linfócitos T/patologia
4.
J Immunol ; 200(11): 3762-3768, 2018 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-29661825

RESUMO

NK cells are a subset of group 1 innate lymphocytes that recognize and eliminate virus-infected and transformed cells. During the course of their development, NK cells acquire a repertoire of activating and inhibitory receptors, which ultimately define their reactivity against target cells. The array of receptors and their specificity during early developmental stages will control and imprint functional properties of NK cells, a process known as "NK cell education." Innate lymphoid cells (ILCs) are a diverse group of lymphocytes, which, like NK cells, do not rely on somatically rearranged Ag receptors for recognition. Among ILC subsets, ILC1s are most like NK cells functionally. Prototypic ILC1s reside in the liver, and a large part of their function is attributed to the expression of TRAIL, a TNF superfamily member with a well-documented antitumor activity. In this article, we show that TRAIL expression on mouse ILC1s is controlled by an activating receptor NKp46, which has been previously shown to control NK cell education. In the absence of NKp46, ILC1s fail to express normal levels of TRAIL on the surface, which results in diminished cytotoxicity toward TRAIL receptor-positive targets. To our knowledge, these findings provide the first evidence of a role of NKp46 in ILC1s that calibrates their antitumor response.


Assuntos
Antígenos Ly/imunologia , Citotoxicidade Imunológica/imunologia , Imunidade Inata/imunologia , Linfócitos/imunologia , Receptor 1 Desencadeador da Citotoxicidade Natural/imunologia , Neoplasias/imunologia , Ligante Indutor de Apoptose Relacionado a TNF/imunologia , Animais , Linhagem Celular , Humanos , Células Matadoras Naturais/imunologia , Fígado/imunologia , Camundongos , Camundongos Endogâmicos C57BL
5.
PLoS Genet ; 11(11): e1005645, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26544571

RESUMO

Proper development of the immune system is an intricate process dependent on many factors, including an intact DNA damage response. The DNA double-strand break signaling kinase ATM and its cofactor NBS1 are required during T cell development and for the maintenance of genomic stability. The role of a second ATM cofactor, ATMIN (also known as ASCIZ) in T cells is much less clear, and whether ATMIN and NBS1 function in synergy in T cells is unknown. Here, we investigate the roles of ATMIN and NBS1, either alone or in combination, using murine models. We show loss of NBS1 led to a developmental block at the double-positive stage of T cell development, as well as reduced TCRα recombination, that was unexpectedly neither exacerbated nor alleviated by concomitant loss of ATMIN. In contrast, loss of both ATMIN and NBS1 enhanced DNA damage that drove spontaneous peripheral T cell hyperactivation, proliferation as well as excessive production of proinflammatory cytokines and chemokines, leading to a highly inflammatory environment. Intriguingly, the disease causing T cells were largely proficient for both ATMIN and NBS1. In vivo this resulted in severe intestinal inflammation, colitis and premature death. Our findings reveal a novel model for an intestinal bowel disease phenotype that occurs upon combined loss of the DNA repair cofactors ATMIN and NBS1.


Assuntos
Proteínas de Ciclo Celular/fisiologia , Reparo do DNA , Ativação Linfocitária/fisiologia , Proteínas Nucleares/fisiologia , Linfócitos T/imunologia , Fatores de Transcrição/fisiologia , Animais , Colite/imunologia , Dano ao DNA , Proteínas de Ligação a DNA , Imunofenotipagem , Camundongos , Espécies Reativas de Oxigênio/metabolismo , Receptores de Antígenos de Linfócitos T alfa-beta/genética , Recombinação Genética , Baço/citologia , Baço/metabolismo
6.
J Immunol ; 193(4): 1737-46, 2014 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-25015818

RESUMO

Constitutive expression of Krüppel-like factor 3 (KLF3, BKLF) increases marginal zone (MZ) B cell numbers, a phenotype shared with mice lacking KLF2. Ablation of KLF3, known to interact with serum response factor (SRF), or SRF itself, results in fewer MZ B cells. It is unknown how these functional equivalences result. In this study, it is shown that KLF3 acts as transcriptional repressor for the leukocyte-specific integrin ß7 (Itgb7, Ly69) by binding to the ß7 promoter, as revealed by chromatin immunoprecipitation. KLF2 overexpression antagonizes this repression and also binds the ß7 promoter, indicating that these factors may compete for target sequence(s). Whereas ß7 is identified as direct KLF target, its repression by KLF3 is not connected to the MZ B cell increase because ß7-deficient mice have a normal complement of these and the KLF3-driven increase still occurs when ß7 is deleted. Despite this, KLF3 overexpression abolishes lymphocyte homing to Peyer's patches, much like ß7 deficiency does. Furthermore, KLF3 expression alone overcomes the MZ B cell deficiency when SRF is absent. SRF is also dispensable for the KLF3-mediated repression of ß7. Thus, despite the shared phenotype of KLF3 and SRF-deficient mice, cooperation of these factors appears neither relevant for the formation of MZ B cells nor for the regulation of ß7. Finally, a potent negative regulatory feedback loop limiting KLF3 expression is shown in this study, mediated by KLF3 directly repressing its own gene promoter. In summary, KLFs use regulatory circuits to steer lymphocyte maturation and homing and directly control leukocyte integrin expression.


Assuntos
Linfócitos B/imunologia , Cadeias beta de Integrinas/genética , Fatores de Transcrição Kruppel-Like/genética , Linfopoese/imunologia , Animais , Antígenos CD11/biossíntese , Diferenciação Celular/genética , Células Cultivadas , Proteínas de Ligação a DNA , Regulação da Expressão Gênica/imunologia , Cadeias alfa de Integrinas/biossíntese , Fatores de Transcrição Kruppel-Like/biossíntese , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Nódulos Linfáticos Agregados/imunologia , Regiões Promotoras Genéticas , Ligação Proteica , Fator de Resposta Sérica/genética
7.
Trends Immunol ; 32(12): 567-73, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22056207

RESUMO

γδ cells have been conserved across ∼450 million years of evolution, from which they share the distinction, alongside αß T cells and B cells, of forming antigen receptors by somatic gene recombination. However, much about these cells remains unclear. Indeed, although γδ cells display 'innate-like' characteristics exemplified by rapid tissue-localised responses to stress-associated stimuli, their huge capacity for T cell receptor (TCR)γδ diversity also suggests 'adaptive-like' potential. Clarity requires a better understanding of TCRγδ itself, not only through identification of TCR ligands, but also by correlating thymic TCRγδ signalling with commitment to γδ effector fates. Here, we propose that thymic TCRγδ-ligand engagement versus ligand-independent signalling differentially imprints innate-like versus adaptive-like characteristics on developing γδ cells, which fundamentally dictate their peripheral effector properties.


Assuntos
Receptores de Antígenos de Linfócitos T gama-delta/imunologia , Transdução de Sinais , Linfócitos T/imunologia , Imunidade Adaptativa , Animais , Linhagem da Célula , Humanos , Imunidade Inata , Receptores de Antígenos de Linfócitos T gama-delta/metabolismo , Linfócitos T/citologia , Linfócitos T/metabolismo
8.
Proc Natl Acad Sci U S A ; 108(8): 3330-5, 2011 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-21300860

RESUMO

αß T-cell repertoire selection is mediated by peptide-MHC complexes presented by thymic epithelial or myeloid cells, and by lipid-CD1 complexes expressed by thymocytes. γδ T-cell repertoire selection, by contrast, is largely unresolved. Mice mutant for Skint-1, a unique Ig superfamily gene, do not develop canonical Vγ5Vδ1(+) dendritic epidermal T cells. This study shows that transgenic Skint-1, across a broad range of expression levels, precisely and selectively determines the Vγ5Vδ1(+) dendritic epidermal T-cell compartment. Skint-1 is expressed by medullary thymic epithelial cells, and unlike lipid-CD1 complexes, must be expressed by stromal cells to function efficiently. Its unusual transmembrane-cytoplasmic regions severely limit cell surface expression, yet increasing this or, conversely, retaining Skint1 intracellularly markedly compromises function. Each Skint1 domain appears nonredundant, including a unique decamer specifying IgV-domain processing. This investigation of Skint-1 biology points to complex events underpinning the positive selection of an intraepithelial γδ repertoire.


Assuntos
Epiderme/imunologia , Imunoglobulinas/imunologia , Linfócitos T/imunologia , Animais , Células Dendríticas , Células Epidérmicas , Células Epiteliais , Expressão Gênica/imunologia , Imunoglobulinas/genética , Camundongos , Camundongos Transgênicos , Receptores de Antígenos de Linfócitos T alfa-beta/imunologia , Subpopulações de Linfócitos T
9.
Blood ; 117(14): 3780-92, 2011 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-21297003

RESUMO

Splenic marginal zone (MZ) B cells are a lineage distinct from follicular and peritoneal B1 B cells. They are located next to the marginal sinus where blood is released. Here they pick up antigens and shuttle the load onto follicular dendritic cells inside the follicle. On activation, MZ B cells rapidly differentiate into plasmablasts secreting antibodies, thereby mediating humoral immune responses against blood-borne type 2 T-independent antigens. As Krüppel-like factors are implicated in cell differentiation/function in various tissues, we studied the function of basic Krüppel-like factor (BKLF/KLF3) in B cells. Whereas B-cell development in the bone marrow of KLF3-transgenic mice was unaffected, MZ B-cell numbers in spleen were increased considerably. As revealed in chimeric mice, this occurred cell autonomously, increasing both MZ and peritoneal B1 B-cell subsets. Comparing KLF3-transgenic and nontransgenic follicular B cells by RNA-microarray revealed that KLF3 regulates a subset of genes that was similarly up-regulated/down-regulated on normal MZ B-cell differentiation. Indeed, KLF3 expression overcame the lack of MZ B cells caused by different genetic alterations, such as CD19-deficiency or blockade of B-cell activating factor-receptor signaling, indicating that KLF3 may complement alternative nuclear factor-κB signaling. Thus, KLF3 is a driving force toward MZ B-cell maturation.


Assuntos
Diferenciação Celular/genética , Fatores de Transcrição Kruppel-Like/fisiologia , Células Progenitoras Linfoides/metabolismo , Células Progenitoras Linfoides/fisiologia , Linfopoese/genética , Mucosa/imunologia , Animais , Antígenos CD19/genética , Antígenos CD19/metabolismo , Células Cultivadas , Análise por Conglomerados , Feminino , Perfilação da Expressão Gênica , Técnicas de Transferência de Genes , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Tecido Linfoide/imunologia , Tecido Linfoide/metabolismo , Tecido Linfoide/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Análise em Microsséries , Mucosa/metabolismo , Mucosa/fisiologia
10.
Front Immunol ; 14: 1085895, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37153600

RESUMO

Members of the Regulator of G-protein signaling (Rgs) family regulate the extent and timing of G protein signaling by increasing the GTPase activity of Gα protein subunits. The Rgs family member Rgs1 is one of the most up-regulated genes in tissue-resident memory (TRM) T cells when compared to their circulating T cell counterparts. Functionally, Rgs1 preferentially deactivates Gαq, and Gαi protein subunits and can therefore also attenuate chemokine receptor-mediated immune cell trafficking. The impact of Rgs1 expression on tissue-resident T cell generation, their maintenance, and the immunosurveillance of barrier tissues, however, is only incompletely understood. Here we report that Rgs1 expression is readily induced in naïve OT-I T cells in vivo following intestinal infection with Listeria monocytogenes-OVA. In bone marrow chimeras, Rgs1 -/- and Rgs1 +/+ T cells were generally present in comparable frequencies in distinct T cell subsets of the intestinal mucosa, mesenteric lymph nodes, and spleen. After intestinal infection with Listeria monocytogenes-OVA, however, OT-I Rgs1 +/+ T cells outnumbered the co-transferred OT-I Rgs1- /- T cells in the small intestinal mucosa already early after infection. The underrepresentation of the OT-I Rgs1 -/- T cells persisted to become even more pronounced during the memory phase (d30 post-infection). Remarkably, upon intestinal reinfection, mice with intestinal OT-I Rgs1 +/+ TRM cells were able to prevent the systemic dissemination of the pathogen more efficiently than those with OT-I Rgs1 -/- TRM cells. While the underlying mechanisms are not fully elucidated yet, these data thus identify Rgs1 as a critical regulator for the generation and maintenance of tissue-resident CD8+ T cells as a prerequisite for efficient local immunosurveillance in barrier tissues in case of reinfections with potential pathogens.


Assuntos
Linfócitos T CD8-Positivos , Proteínas de Ligação ao GTP , Listeria monocytogenes , Animais , Camundongos , Proteínas de Ligação ao GTP/metabolismo , Subunidades Proteicas/metabolismo , Subpopulações de Linfócitos T
11.
EMBO J ; 27(9): 1333-44, 2008 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-18369315

RESUMO

Upon B-cell antigen receptor (BCR) activation, the protein tyrosine kinase Syk phosphorylates the adaptor protein SH2 domain-containing leukocyte protein of 65 kDa (SLP-65), thus coupling the BCR to diverse signalling pathways. Here, we report that SLP-65 is not only a downstream target and substrate of Syk but also a direct binding-partner and activator of this kinase. This positive feedback is mediated by the binding of the SH2 domain of SLP-65 to an autophosphorylated tyrosine of Syk. The mutant B cells that cannot form the Syk/SLP-65 complex are defective in BCR-induced extracellular signal-regulated kinase, nuclear factor kappa B and nuclear factor of activated T cells, but not Akt activation, and are blocked in B-cell development. Furthermore, we show that formation of the Syk/SLP-65 complex is required for sustained Ca(2+) responses in activated B cells. We suggest that after activation and internalization of the BCR, Syk remains active as part of a membrane-bound Syk/SLP-65 complex controlling sustained signalling and calcium influx.


Assuntos
Sinalização do Cálcio/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas Tirosina Quinases/metabolismo , Receptores de Antígenos de Linfócitos B/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Cálcio/metabolismo , Sinalização do Cálcio/genética , Diferenciação Celular , Linhagem Celular , MAP Quinases Reguladas por Sinal Extracelular/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Citometria de Fluxo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/genética , Luciferases/genética , Luciferases/metabolismo , Camundongos , Camundongos Knockout , Modelos Biológicos , Mutação , NF-kappa B/genética , NF-kappa B/metabolismo , Fosforilação , Ligação Proteica , Proteínas Tirosina Quinases/genética , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores de Antígenos de Linfócitos B/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Quinase Syk , Transfecção , Tirosina/genética , Tirosina/metabolismo , Domínios de Homologia de src/genética
12.
Dev Cell ; 57(15): 1847-1865.e9, 2022 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-35803280

RESUMO

Immune surveillance is critical to prevent tumorigenesis. Gliomas evade immune attack, but the underlying mechanisms remain poorly understood. We show that glioma cells can sustain growth independent of immune system constraint by reducing Notch signaling. Loss of Notch activity in a mouse model of glioma impairs MHC-I and cytokine expression and curtails the recruitment of anti-tumor immune cell populations in favor of immunosuppressive tumor-associated microglia/macrophages (TAMs). Depletion of T cells simulates Notch inhibition and facilitates tumor initiation. Furthermore, Notch-depleted glioma cells acquire resistance to interferon-γ and TAMs re-educating therapy. Decreased interferon response and cytokine expression by human and mouse glioma cells correlate with low Notch activity. These effects are paralleled by upregulation of oncogenes and downregulation of quiescence genes. Hence, suppression of Notch signaling enables gliomas to evade immune surveillance and increases aggressiveness. Our findings provide insights into how brain tumor cells shape their microenvironment to evade immune niche control.


Assuntos
Neoplasias Encefálicas , Glioma , Animais , Neoplasias Encefálicas/metabolismo , Transformação Celular Neoplásica , Citocinas , Glioma/genética , Glioma/metabolismo , Glioma/patologia , Humanos , Evasão da Resposta Imune , Interferon gama/metabolismo , Camundongos , Receptores Notch , Microambiente Tumoral/fisiologia
13.
Nat Commun ; 11(1): 1794, 2020 04 14.
Artigo em Inglês | MEDLINE | ID: mdl-32286285

RESUMO

Although group 3 innate lymphoid cells (ILC3s) are efficient inducers of T cell responses in the spleen, they fail to induce CD4+ T cell proliferation in the gut. The signals regulating ILC3-T cell responses remain unknown. Here, we show that transcripts associated with MHC II antigen presentation are down-modulated in intestinal natural cytotoxicity receptor (NCR)- ILC3s. Further data implicate microbiota-induced IL-23 as a crucial signal for reversible silencing of MHC II in ILC3s, thereby reducing the capacity of ILC3s to present antigen to T cells in the intestinal mucosa. Moreover, IL-23-mediated MHC II suppression is dependent on mTORC1 and STAT3 phosphorylation in NCR- ILC3s. By contrast, splenic interferon-γ induces MHC II expression and CD4+ T cell stimulation by NCR- ILC3s. Our results thus identify biological circuits for tissue-specific regulation of ILC3-dependent T cell responses. These pathways may have implications for inducing or silencing T cell responses in human diseases.


Assuntos
Apresentação de Antígeno/imunologia , Imunidade Inata , Linfócitos/imunologia , Microbiota , Baço/citologia , Animais , Células Apresentadoras de Antígenos/citologia , Células Apresentadoras de Antígenos/imunologia , Antígenos CD/metabolismo , Polaridade Celular , Regulação para Baixo , Antígenos de Histocompatibilidade Classe II/metabolismo , Interferon gama/metabolismo , Interleucina-23/metabolismo , Ativação Linfocitária/imunologia , Linfócitos/citologia , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Camundongos , Microbiota/genética , Microbiota/imunologia , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fenótipo , Fosforilação , Análise de Componente Principal , Regiões Promotoras Genéticas/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fator de Transcrição STAT3/metabolismo , Linfócitos T/imunologia , Transativadores/genética , Transativadores/metabolismo , Transcrição Gênica
14.
Front Immunol ; 6: 416, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26322047

RESUMO

Innate lymphoid cells (ILCs) have emerged as a new family of immune cells with crucial functions in innate and adaptive immunity. ILC subsets mirror the cytokine and transcriptional profile of CD4(+) T helper (TH) cell subsets. Hence, group 1 (ILC1), group 2 (ILC2), and group 3 (ILC3) ILCs can be distinguished by the production of TH1, TH2, and TH17-type cytokines, respectively. Cytokine release by ILCs not only shapes early innate immunity but can also orchestrate TH immune responses to microbial or allergen exposure. Recent studies have identified an unexpected effector function of ILCs as antigen presenting cells. Both ILC2s and ILC3s are able to process and present foreign antigens (Ags) via major histocompatibility complex class II, and to induce cognate CD4(+) T cell responses. In addition, Ag-stimulated T cells promote ILC activation and effector functions indicating a reciprocal interaction between the adaptive and innate immune system. A fundamental puzzle in ILC function is how ILC/T cell interactions promote host protection and prevent autoimmune diseases. Furthermore, the way in which microenvironmental and inflammatory signals determine the outcome of ILC/T cell immune responses in various tissues is not yet understood. This review focuses on recent advances in understanding the mechanisms that coordinate the collaboration between ILCs and T cells under homeostatic and inflammatory conditions. We also discuss the potential roles of T cells and other immune cells to regulate ILC functions and to maintain homeostasis in mucosal tissues.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA