Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Int J Mol Sci ; 20(21)2019 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-31690060

RESUMO

Innate lymphoid cells (ILC) are important players of early immune defenses in situations like lymphoid organogenesis or in case of immune response to inflammation, infection and cancer. Th1 and Th2 antagonism is crucial for the regulation of immune responses, however mechanisms are still unclear for ILC functions. ILC2 and NK cells were reported to be both involved in allergic airway diseases and were shown to be able to interplay in the regulation of the immune response. CXCR6 is a common chemokine receptor expressed by all ILC, and its deficiency affects ILC2 and ILC1/NK cell numbers and functions in lungs in both steady-state and inflammatory conditions. We determined that the absence of a specific ILC2 KLRG1+ST2- subset in CXCR6-deficient mice is probably dependent on CXCR6 for its recruitment to the lung under inflammation. We show that despite their decreased numbers, lung CXCR6-deficient ILC2 are even more activated cells producing large amount of type 2 cytokines that could drive eosinophilia. This is strongly associated to the decrease of the lung Th1 response in CXCR6-deficient mice.


Assuntos
Subpopulações de Linfócitos/imunologia , Pneumonia/imunologia , Receptores CXCR6/metabolismo , Animais , Células Cultivadas , Interferon gama/genética , Interferon gama/metabolismo , Camundongos , Papaína/toxicidade , Pneumonia/etiologia , Receptores CXCR6/genética
2.
Immunity ; 50(4): 1054-1068.e3, 2019 04 16.
Artigo em Inglês | MEDLINE | ID: mdl-30926235

RESUMO

Innate lymphoid cell (ILC) development proposes that ILC precursors (ILCPs) segregate along natural killer (NK) cell versus helper cell (ILC1, ILC2, ILC3) pathways, the latter depending on expression of Id2, Zbtb16, and Gata3. We have developed an Id2-reporter strain expressing red fluorescent protein (RFP) in the context of normal Id2 expression to re-examine ILCP phenotype and function. We show that bone-marrow ILCPs were heterogeneous and harbored extensive NK-cell potential in vivo and in vitro. By multiplexing Id2RFP with Zbtb16CreGFP and Bcl11btdTomato strains, we made a single-cell dissection of the ILCP compartment. In contrast with the current model, we have demonstrated that Id2+Zbtb16+ ILCPs included multi-potent ILCPs that retained NK-cell potential. Late-stage ILC2P and ILC3P compartments could be defined by differential Zbtb16 and Bcl11b expression. We suggest a revised model for ILC differentiation that redefines the cell-fate potential of helper-ILC-restricted Zbtb16+ ILCPs.


Assuntos
Regulação da Expressão Gênica/imunologia , Células-Tronco Hematopoéticas/citologia , Imunidade Inata , Proteína 2 Inibidora de Diferenciação/genética , Linfopoese/genética , Transferência Adotiva , Animais , Linhagem da Célula , Fator de Transcrição GATA3/biossíntese , Fator de Transcrição GATA3/genética , Fator de Transcrição GATA3/fisiologia , Genes Reporter , Células-Tronco Hematopoéticas/metabolismo , Proteína 2 Inibidora de Diferenciação/biossíntese , Células Matadoras Naturais/citologia , Proteínas Luminescentes/análise , Proteínas Luminescentes/genética , Camundongos , Camundongos Endogâmicos C57BL , Modelos Imunológicos , Proteína com Dedos de Zinco da Leucemia Promielocítica/biossíntese , Proteína com Dedos de Zinco da Leucemia Promielocítica/genética , Proteína com Dedos de Zinco da Leucemia Promielocítica/fisiologia , Análise de Célula Única , Linfócitos T Auxiliares-Indutores/citologia , Transcrição Gênica , Proteína Vermelha Fluorescente
3.
Nat Immunol ; 18(10): 1139-1149, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28825702

RESUMO

The molecular events that initiate lymphoid-lineage specification remain unidentified because the stages of differentiation during which lineage commitment occurs are difficult to characterize. We isolated fetal liver progenitor cells undergoing restriction of their differentiation potential toward the T cell-innate lymphoid cell lineage or the B cell lineage. Transcripts that defined the molecular signatures of these two subsets were sequentially upregulated in lympho-myeloid precursor cells and in common lymphoid progenitor cells, respectively, and this preceded lineage restriction; this indicates that T cell-versus-B cell commitment is not a binary fate 'decision'. The T cell-bias and B cell-bias transcriptional programs were frequently co-expressed in common lymphoid progenitor cells and were segregated in subsets biased toward T cell differentiation or B cell differentiation, after interleukin 7 (IL-7) signaling that controlled the number of progenitor cells engaging in T cell differentiation versus B cell differentiation.


Assuntos
Linfócitos B/citologia , Linhagem da Célula , Fígado/citologia , Linfopoese , Linfócitos T/citologia , Animais , Linfócitos B/metabolismo , Biomarcadores , Diferenciação Celular/genética , Linhagem da Célula/genética , Análise por Conglomerados , Feto , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Imunofenotipagem , Interleucina-7/metabolismo , Fígado/embriologia , Células Progenitoras Linfoides/citologia , Células Progenitoras Linfoides/metabolismo , Linfopoese/genética , Camundongos , Camundongos Transgênicos , Transdução de Sinais , Linfócitos T/metabolismo , Transcriptoma
4.
J Vis Exp ; (119)2017 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-28191880

RESUMO

Gene expression heterogeneity is an interesting feature to investigate in lymphoid populations. Gene expression in these cells varies during cell activation, stress, or stimulation. Single-cell multiplex gene expression enables the simultaneous assessment of tens of genes1,2,3. At the single-cell level, multiplex gene expression determines population heterogeneity4,5. It allows for the distinction of population heterogeneity by determining both the probable mix of diverse precursor stages among mature cells and also the diversity of cell responses to stimuli. Innate lymphoid cells (ILC) have been recently described as a population of innate effectors of the immune response6,7. In this protocol, cell heterogeneity of the ILC hepatic compartment is investigated during homeostasis. Currently, the most widely used technique to assess gene expression is RT-qPCR. This method measures gene expression only one gene at a time. Additionally, this method cannot estimate heterogeneity of gene expression, since multiple cells are needed for one test. This leads to the measurement of the average gene expression of the population. When assessing large numbers of genes, RT-qPCR becomes a time-, reagent-, and sample-consuming method. Hence, the trade-offs limit the number of genes or cell populations that can be evaluated, increasing the risk of missing the global picture. This manuscript describes how single-cell multiplex RT-qPCR can be used to overcome these limitations. This technique has benefited from recent microfluidics technological advances1,2. Reactions occurring in multiplex RT-qPCR chips do not exceed the nanoliter-level. Hence, single-cell gene expression, as well as simultaneous multiple gene expression, can be performed in a reagent-, sample-, and cost-effective manner. It is possible to test cell gene signature heterogeneity at the clonal level between cell subsets within a population at different developmental stages or under different conditions4,5. Working on rare populations with large numbers of conditions at the single-cell level is no longer a restriction.


Assuntos
Perfilação da Expressão Gênica/métodos , Linfócitos/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Análise de Célula Única , Diferenciação Celular , Humanos
5.
Sci Signal ; 9(426): ra45, 2016 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-27141929

RESUMO

The Notch signaling pathway is conserved throughout evolution, and it controls various processes, including cell fate determination, differentiation, and proliferation. Innate lymphoid cells (ILCs) are lymphoid cells lacking antigen receptors that fulfill effector and regulatory functions in innate immunity and tissue remodeling. Type 3 ILCs (ILC3s) reinforce the epithelial barrier and maintain homeostasis with intestinal microbiota. We demonstrated that the population of natural cytotoxicity receptor-positive (NCR(+)) ILC3s in mice is composed of two subsets that have distinct developmental requirements. A major subset depended on the activation of Notch2 in NCR(-) ILC3 precursors in the lamina propria of the small intestine to stimulate expression of the genes encoding the transcription factors T-bet, RORγt, and aryl hydrocarbon receptor (AhR). Notch signaling contributed to the transition of NCR(-) cells into NCR(+) cells, the more proinflammatory subset, in a cell-autonomous manner. In the absence of Notch signaling, this subset of NCR(-) ILC3s did not acquire the gene expression profile of NCR(+) ILC3s. A second subset of NCR(+) ILC3s did not depend on Notch for their development or for increased transcription factor abundance; however, their production of cytokines and cell surface abundance of NCRs were decreased in the absence of Notch signaling. Together, our data suggest that Notch is a regulator of the plasticity of ILC3s by controlling NCR(+) cell fate.


Assuntos
Linfócitos/citologia , Linfócitos/metabolismo , Receptores Notch/metabolismo , Animais , Linhagem da Célula , Proliferação de Células , Células Cultivadas , Citocinas/metabolismo , Humanos , Imunidade Inata , Interleucinas/metabolismo , Intestinos , Camundongos , Camundongos Knockout , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo
6.
Cell Rep ; 14(6): 1500-1516, 2016 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-26832410

RESUMO

T and innate lymphoid cells (ILCs) share some aspects of their developmental programs. However, although Notch signaling is strictly required for T cell development, it is dispensable for fetal ILC development. Constitutive activation of Notch signaling, at the common lymphoid progenitor stage, drives T cell development and abrogates ILC development by preventing Id2 expression. By combining single-cell transcriptomics and clonal culture strategies, we characterize two heterogeneous α4ß7-expressing lymphoid progenitor compartments. αLP1 (Flt3(+)) still retains T cell potential and comprises the global ILC progenitor, while αLP2 (Flt3(-)) consists of ILC precursors that are primed toward the different ILC lineages. Only a subset of αLP2 precursors is sensitive to Notch signaling required for their proliferation. Our study identifies, in a refined manner, the diversity of transitional stages of ILC development, their transcriptional signatures, and their differential dependence on Notch signaling.


Assuntos
Subpopulações de Linfócitos B/imunologia , Regulação da Expressão Gênica no Desenvolvimento , Imunidade Inata , Transdução de Sinais , Subpopulações de Linfócitos T/imunologia , Animais , Subpopulações de Linfócitos B/citologia , Diferenciação Celular , Linhagem da Célula/imunologia , Proliferação de Células , Feto , Perfilação da Expressão Gênica , Proteína 2 Inibidora de Diferenciação/genética , Proteína 2 Inibidora de Diferenciação/imunologia , Integrinas/genética , Integrinas/imunologia , Camundongos , Camundongos Transgênicos , Receptores Notch/genética , Receptores Notch/imunologia , Análise de Célula Única , Células-Tronco/citologia , Células-Tronco/imunologia , Subpopulações de Linfócitos T/citologia , Transcrição Gênica , Transcriptoma , Tirosina Quinase 3 Semelhante a fms/genética , Tirosina Quinase 3 Semelhante a fms/imunologia
7.
Nat Immunol ; 17(3): 269-76, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26779601

RESUMO

The precise lineage relationship between innate lymphoid cells (ILCs) and lymphoid tissue-inducer (LTi) cells is poorly understood. Using single-cell multiplex transcriptional analysis of 100 lymphoid genes and single-cell cultures of fetal liver precursor cells, we identified the common proximal precursor to these lineages and found that its bifurcation was marked by differential induction of the transcription factors PLZF and TCF1. Acquisition of individual effector programs specific to the ILC subsets ILC1, ILC2 and ILC3 was initiated later, at the common ILC precursor stage, by transient expression of mixed ILC1, ILC2 and ILC3 transcriptional patterns, whereas, in contrast, the development of LTi cells did not go through multilineage priming. Our findings provide insight into the divergent mechanisms of the differentiation of the ILC lineage and LTi cell lineage and establish a high-resolution 'blueprint' of their development.


Assuntos
Linhagem da Célula/imunologia , Subpopulações de Linfócitos/imunologia , Linfócitos/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Animais , Diferenciação Celular/genética , Citometria de Fluxo , Perfilação da Expressão Gênica , Fator 1-alfa Nuclear de Hepatócito/imunologia , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/imunologia , Tecido Linfoide/citologia , Camundongos , Reação em Cadeia da Polimerase Multiplex , Proteína com Dedos de Zinco da Leucemia Promielocítica , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise de Célula Única
8.
Mediators Inflamm ; 2015: 368427, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26494947

RESUMO

Innate lymphoid cells are present at mucosal sites and represent the first immune barrier against infections, but what contributes to their circulation and homing is still unclear. Using Rag2(-/-) Cxcr6(Gfp/+) reporter mice, we assessed the expression and role of CXCR6 in the circulation of ILC precursors and their progeny. We identify CXCR6 expressing ILC precursors in the bone marrow and characterize their significant increase in CXCR6-deficient mice at steady state, indicating their partial retention in the bone marrow after CXCR6 ablation. Circulation was also impaired during embryonic life as fetal liver from CXCR6-deficient embryos displayed decreased numbers of ILC3 precursors. When injected, fetal CXCR6-deficient ILC3 precursors also fail to home and reconstitute ILC compartments in vivo. We show that adult intestinal ILC subsets have heterogeneous expression pattern of CXCR6, integrin α 4 ß 7, CD62L, CD69, and CD44, with ILC1 and ILC3 being more likely tissue resident lymphocytes. Intestinal ILC subsets were unchanged in percentages and numbers in both mice. We demonstrate that the ILC frequency is maintained due to a significant increase of ILC peripheral proliferation, as well as an increased proliferation of the in situ ILC precursors to compensate their retention in the bone marrow.


Assuntos
Regulação da Expressão Gênica , Linfócitos/metabolismo , Receptores CXCR/genética , Receptores CXCR/metabolismo , Animais , Medula Óssea/metabolismo , Medula Óssea/patologia , Células da Medula Óssea/citologia , Proliferação de Células , Meios de Cultura , Citometria de Fluxo , Imunidade Inata/imunologia , Intestinos/imunologia , Subpopulações de Linfócitos/imunologia , Camundongos , Camundongos Transgênicos , Mucosa , Receptores CXCR6
9.
J Immunol ; 188(3): 1245-54, 2012 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-22210913

RESUMO

RANK and its ligand RANKL play important roles in the development and regulation of the immune system. We show that mice transgenic for Rank in hair follicles display massive postnatal growth of skin-draining lymph nodes. The proportions of hematopoietic and nonhematopoietic stromal cells and their organization are maintained, with the exception of an increase in B cell follicles. The hematopoietic cells are not activated and respond to immunization by foreign Ag and adjuvant. We demonstrate that soluble RANKL is overproduced from the transgenic hair follicles and that its neutralization normalizes lymph node size, inclusive area, and numbers of B cell follicles. Reticular fibroblastic and vascular stromal cells, important for secondary lymphoid organ formation and organization, express RANK and undergo hyperproliferation, which is abrogated by RANKL neutralization. In addition, they express higher levels of CXCL13 and CCL19 chemokines, as well as MAdCAM-1 and VCAM-1 cell-adhesion molecules. These findings highlight the importance of tissue-derived cues for secondary lymphoid organ homeostasis and identify RANKL as a key molecule for controlling the plasticity of the immune system.


Assuntos
Proliferação de Células , Linfonodos/crescimento & desenvolvimento , Ligante RANK/fisiologia , Células Estromais/citologia , Animais , Quimiocina CCL19 , Quimiocina CXCL13 , Folículo Piloso , Homeostase , Sistema Imunitário/fisiologia , Camundongos , Camundongos Transgênicos
10.
Nat Immunol ; 12(10): 949-58, 2011 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-21909092

RESUMO

The transcription factor RORγt is required for the development of several innate lymphoid populations, such as lymphoid tissue-inducer cells (LTi cells) and cells that secrete interleukin 17 (IL-17) or IL-22. The progenitor cells as well as the developmental stages that lead to the emergence of RORγt(+) innate lymphoid cells (ILCs) remain undefined. Here we identify the chemokine receptor CXCR6 as an additional marker of the development of ILCs and show that common lymphoid progenitors lost B cell and T cell potential as they successively acquired expression of the integrin α(4)ß(7) and CXCR6. Whereas fetal RORγt(+) cells matured in the fetal liver environment, adult bone marrow-derived RORγt(+) ILCs matured outside the bone marrow, in a Notch2-dependent manner. Therefore, fetal and adult environments influence the differentiation of RORγt(+) cells differently.


Assuntos
Feto/imunologia , Linfócitos/imunologia , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/fisiologia , Receptor Notch2/fisiologia , Transdução de Sinais , Animais , Diferenciação Celular , Células Cultivadas , Proteínas de Ligação a DNA/fisiologia , Imunidade Inata , Integrinas/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Receptores CXCR/fisiologia , Receptores CXCR6
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA