Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 120(40): e2306761120, 2023 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-37756335

RESUMO

Natural killer (NK) cells and type 1 innate lymphoid cells (ILC1) require signal transducer and activator of transcription 4 (STAT4) to elicit rapid effector responses and protect against pathogens. By combining genetic and transcriptomic approaches, we uncovered divergent roles for STAT4 in regulating effector differentiation of these functionally related cell types. Stat4 deletion in Ncr1-expressing cells led to impaired NK cell terminal differentiation as well as to an unexpected increased generation of cytotoxic ILC1 during intestinal inflammation. Mechanistically, Stat4-deficient ILC1 exhibited upregulation of gene modules regulated by STAT5 in vivo and an aberrant effector differentiation upon in vitro stimulation with IL-2, used as a prototypical STAT5 activator. Moreover, STAT4 expression in NCR+ innate lymphocytes restrained gut inflammation in the dextran sulfate sodium-induced colitis model limiting pathogenic production of IL-13 from adaptive CD4+ T cells in the large intestine. Collectively, our data shed light on shared and distinctive mechanisms of STAT4-regulated transcriptional control in NK cells and ILC1 required for intestinal inflammatory responses.


Assuntos
Antineoplásicos , Fator de Transcrição STAT5 , Humanos , Imunidade Inata , Diferenciação Celular , Células Matadoras Naturais , Inflamação , Fator de Transcrição STAT4/genética
2.
Proc Natl Acad Sci U S A ; 118(10)2021 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-33649222

RESUMO

Natural killer (NK) cells are innate effectors armed with cytotoxic and cytokine-secreting capacities whose spontaneous antitumor activity is key to numerous immunotherapeutic strategies. However, current mouse models fail to mirror the extensive immune system variation that exists in the human population which may impact on NK cell-based therapies. We performed a comprehensive profiling of NK cells in the Collaborative Cross (CC), a collection of novel recombinant inbred mouse strains whose genetic diversity matches that of humans, thereby providing a unique and highly diverse small animal model for the study of immune variation. We demonstrate that NK cells from CC strains displayed a breadth of phenotypic and functional variation reminiscent of that reported for humans with regards to cell numbers, key marker expression, and functional capacities. We took advantage of the vast genetic diversity of the CC and identified nine genomic loci through quantitative trait locus mapping driving these phenotypic variations. SNP haplotype patterns and variant effect analyses identified candidate genes associated with lung NK cell numbers, frequencies of CD94+ NK cells, and expression levels of NKp46. Thus, we demonstrate that the CC represents an outstanding resource to study NK cell diversity and its regulation by host genetics.


Assuntos
Antígenos Ly , Regulação da Expressão Gênica/imunologia , Células Matadoras Naturais/imunologia , Subfamília D de Receptores Semelhantes a Lectina de Células NK , Receptor 1 Desencadeador da Citotoxicidade Natural , Polimorfismo de Nucleotídeo Único , Locos de Características Quantitativas/imunologia , Animais , Antígenos Ly/genética , Antígenos Ly/imunologia , Cruzamentos Genéticos , Camundongos , Camundongos Endogâmicos , Subfamília D de Receptores Semelhantes a Lectina de Células NK/genética , Subfamília D de Receptores Semelhantes a Lectina de Células NK/imunologia , Receptor 1 Desencadeador da Citotoxicidade Natural/genética , Receptor 1 Desencadeador da Citotoxicidade Natural/imunologia
3.
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
4.
Methods Mol Biol ; 1884: 161-176, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30465202

RESUMO

Over the last decades, it has been established that the immune system is crucial for the impediment of cancer development by recognizing and destroying transformed cells. This process has been termed cancer immunosurveillance. Small animal models have significantly facilitated our understanding of it. Dissecting the contribution of any specific immune cell type participating in this process requires the ability to specifically target it while leaving the other immune components as well as the cancer model system unperturbed in vivo. Here, we provide a simple and rapid protocol for the generation of transgenic mice expressing Cre recombinase in a cell type-specific manner-in our example we chose cells expressing Ncr1, which encodes for the surface protein NKp46-and the use of those mice to ablate NKp46+ cells in order to study their role in a model of cancer immunosurveillance against experimental pulmonary metastases. This protocol can easily be adapted to target other cell types and other cancer models.


Assuntos
Antígenos Ly/genética , Vigilância Imunológica , Integrases/genética , Células Matadoras Naturais/imunologia , Neoplasias Pulmonares/imunologia , Receptor 1 Desencadeador da Citotoxicidade Natural/genética , Animais , Antígenos Ly/imunologia , Antígenos Ly/metabolismo , Linhagem Celular Tumoral , Feminino , Genes Reporter/genética , Microscopia Intravital/instrumentação , Microscopia Intravital/métodos , Células Matadoras Naturais/metabolismo , Luciferases de Vaga-Lume/química , Luciferases de Vaga-Lume/genética , Medições Luminescentes/instrumentação , Medições Luminescentes/métodos , Neoplasias Pulmonares/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Receptor 1 Desencadeador da Citotoxicidade Natural/imunologia , Receptor 1 Desencadeador da Citotoxicidade Natural/metabolismo , Neoplasias Experimentais/genética , Neoplasias Experimentais/imunologia
5.
Curr Opin Immunol ; 44: 61-68, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-28359987

RESUMO

Innate lymphoid cells (ILCs) are lineage- and antigen receptor-negative lymphocytes including natural killer (NK) cells and at least three distinguishable cell subsets (ILC1, ILC2, ILC3) that rapidly produce cytokines (IFN-γ, IL-5, IL-13, IL-17A, IL-22) upon activation. As such, ILCs can act as first-line defenders in the context of infection, inflammation and cancer. Because of the strong conservation between the expression of key transcription factors that can drive signature cytokine outputs in ILCs and differentiated helper T cells, it has been proposed that ILCs represent innate counterparts of the latter. Several distinct ILC precursors (ILCP) with pan-ILC (giving rise to all ILCs) or subset-restricted potentials have been described in both mouse and man. How and where these different ILCP give rise to more mature tissue-resident ILCs remains unclear. Recently, environmental signals have been shown to epigenetically influence canonical ILC differentiation pathways, generating substantial functional plasticity. These new results suggest that while ILC differentiation may be 'fixed' in principle, it remains 'flexible' in practice. A more comprehensive knowledge in the molecular mechanisms that regulate ILC development and effector functions may allow for therapeutic manipulation of ILCs for diverse disease conditions.


Assuntos
Imunoterapia , Inflamação/imunologia , Linfócitos/fisiologia , Células Progenitoras Linfoides/fisiologia , Neoplasias/imunologia , Animais , Diferenciação Celular , Plasticidade Celular , Citocinas/metabolismo , Epigênese Genética , Humanos , Imunidade Inata , Ativação Linfocitária , Camundongos , Linfócitos T Auxiliares-Indutores/fisiologia
6.
Kidney Int ; 91(5): 1146-1158, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28111009

RESUMO

Crescentic glomerulonephritis is a life-threatening renal disease that has been extensively studied by the experimental anti-glomerular basement membrane glomerulonephritis (anti-GBM-GN) model. Although T cells have a significant role in this model, athymic/nude mice and rats still develop severe renal disease. Here we further explored the contribution of intrinsic renal cells in the development of T-cell-independent GN lesions. Anti-GBM-GN was induced in three strains of immune-deficient mice (Rag2-/-, Rag2-/-Il2rg-/-, and Rag2-/-Il2rb-/-) that are devoid of either T/B cells or T/B/NK cells. The Rag2-/-Il2rg-/- or Rag2-/-Il2rb-/- mice harbor an additional deletion of either the common gamma chain (γC) or the interleukin-2 receptor ß subunit (IL-2Rß), respectively, impairing IL-15 signaling in particular. As expected, all these strains developed severe anti-GBM-GN. Additionally, bone marrow replenishment experiments allowed us to deduce a protective role for the glomerular-expressed γC during anti-GBM-GN. Given that IL-15 has been found highly expressed in nephritic kidneys despite the absence of lymphocytes, we then studied this cytokine in vitro on primary cultured podocytes from immune-deficient mice (Rag2-/-Il2rg-/- and Rag2-/-Il2rb-/-) compared to controls. IL-15 induced downstream activation of JAK1/3 and SYK in primary cultured podocytes. IL-15-dependent JAK/SYK induction was impaired in the absence of γC or IL-2Rß. We found γC largely induced on podocytes during human glomerulonephritis. Thus, renal lesions are indeed modulated by intrinsic glomerular cells through the γC/IL-2Rß receptor response, to date classically described only in immune cells.


Assuntos
Proteínas de Ligação a DNA/imunologia , Glomerulonefrite/imunologia , Subunidade gama Comum de Receptores de Interleucina/imunologia , Subunidade beta de Receptor de Interleucina-2/imunologia , Glomérulos Renais/imunologia , Podócitos/imunologia , Animais , Autoanticorpos/toxicidade , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Modelos Animais de Doenças , Imunofluorescência , Glomerulonefrite/induzido quimicamente , Glomerulonefrite/metabolismo , Humanos , Subunidade gama Comum de Receptores de Interleucina/genética , Subunidade gama Comum de Receptores de Interleucina/metabolismo , Interleucina-15/imunologia , Interleucina-15/metabolismo , Subunidade beta de Receptor de Interleucina-2/genética , Janus Quinase 1/metabolismo , Janus Quinase 3/metabolismo , Glomérulos Renais/citologia , Glomérulos Renais/metabolismo , Células Matadoras Naturais , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Nus , Podócitos/metabolismo , Cultura Primária de Células , Transdução de Sinais , Quinase Syk/metabolismo
7.
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
8.
J Immunol ; 196(11): 4731-8, 2016 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-27183613

RESUMO

Group 3 innate lymphoid cells (ILC3) actively participate in mucosal defense and homeostasis through prompt secretion of IL-17A, IL-22, and IFN-γ. Reports identify two ILC3 lineages: a CCR6(+)T-bet(-) subset that appears early in embryonic development and promotes lymphoid organogenesis and a CCR6(-)T-bet(+) subset that emerges after microbial colonization and harbors NKp46(+) ILC3. We demonstrate that NKp46 expression in the ILC3 subset is highly unstable. Cell fate mapping using Ncr1(CreGFP) × Rosa26(RFP) mice revealed the existence of an intestinal RFP(+) ILC3 subset (Ncr1(FM)) lacking NKp46 expression at the transcript and protein levels. Ncr1(FM) ILC3 produced more IL-22 and were distinguishable from NKp46(+) ILC3 by differential CD117, CD49a, DNAX accessory molecule-1, and, surprisingly, CCR6 expression. Ncr1(FM) ILC3 emerged after birth and persisted in adult mice following broad-spectrum antibiotic treatment. These results identify an unexpected phenotypic instability within NKp46(+) ILC3 that suggests a major role for environmental signals in tuning ILC3 functional plasticity.


Assuntos
Antígenos Ly/imunologia , Imunidade Inata/imunologia , Intestinos/imunologia , Linfócitos/imunologia , Receptor 1 Desencadeador da Citotoxicidade Natural/imunologia , Animais , Células Cultivadas , Intestinos/citologia , Linfócitos/citologia , Camundongos , Camundongos Transgênicos , Fenótipo
9.
Nat Rev Immunol ; 15(7): 415-28, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26065585

RESUMO

Innate lymphoid cells (ILCs) are a recently described family of lymphoid effector cells that have important roles in immune defence, inflammation and tissue remodelling. It has been proposed that ILCs represent 'innate' homologues of differentiated effector T cells, and they have been categorized into three groups ­ namely, ILC1s, ILC2s and ILC3s ­ on the basis of their expression of cytokines and transcription factors that are typically associated with T helper 1 (T(H)1)-, T(H)2- and T(H)17-type immune responses, respectively. Indeed, remarkable similarity is seen between the specific transcription factors required for the development and diversification of different ILC groups and those that drive effector T cell differentiation. The recent identification of dedicated ILC precursors has provided a view of the mechanisms that control this first essential stage of ILC development. Here, we discuss the transcriptional mechanisms that regulate ILC development and diversification into distinct effector subsets with key roles in immunity and tissue homeostasis. We further caution against the current distinction between 'helper' versus 'killer' subsets in the evolving area of ILC nomenclature.


Assuntos
Diferenciação Celular/imunologia , Linhagem da Célula , Imunidade Inata/imunologia , Linfócitos/citologia , Linfócitos/imunologia , Transcrição Gênica/imunologia , Animais , Humanos
10.
Cell Rep ; 10(12): 2043-54, 2015 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-25801035

RESUMO

Innate lymphoid cells (ILCs) are a family of effectors that originate from a common innate lymphoid cell progenitor. However, the transcriptional program that sets the identity of the ILC lineage remains elusive. Here, we show that NFIL3 is a critical regulator of the common helper-like innate lymphoid cell progenitor (CHILP). Cell-intrinsic Nfil3 ablation led to variably impaired development of fetal and adult ILC subsets. Conditional gene targeting demonstrated that NFIL3 exerted its function prior to ILC subset commitment. Accordingly, NFIL3 ablation resulted in loss of ID2(+) CHILP and PLZF(+) ILC progenitors. Nfil3 expression in lymphoid progenitors was under the control of the mesenchyme-derived hematopoietin IL-7, and NFIL3 exerted its function via direct Id2 regulation in the CHILP. Moreover, ectopic Id2 expression in Nfil3-null precursors rescued defective ILC lineage development in vivo. Our data establish NFIL3 as a key regulator of common helper-like ILC progenitors as they emerge during early lymphopoiesis.


Assuntos
Fatores de Transcrição de Zíper de Leucina Básica/metabolismo , Diferenciação Celular/fisiologia , Imunidade Inata , Células Matadoras Naturais/citologia , Linfócitos/citologia , Células Progenitoras Linfoides/citologia , Linfopoese/imunologia , Animais , Diferenciação Celular/genética , Linhagem da Célula/fisiologia , Células Matadoras Naturais/imunologia , Linfócitos/imunologia , Camundongos Endogâmicos C57BL
11.
Blood ; 125(9): 1427-34, 2015 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-25525117

RESUMO

The impairment of cytotoxic activity of lymphocytes disturbs immune surveillance and leads to the development of hemophagocytic lymphohistiocytic syndrome (HLH). Although cytotoxic T lymphocyte (CTL) control of HLH development is well documented, the role for natural killer (NK)-cell effector functions in the pathogenesis of this immune disorder remains unclear. In this study, we specifically targeted a defect in cytotoxicity to either CTL or NK cells in mice so as to dissect the contribution of these lymphocyte subsets to HLH-like disease severity after lymphocytic choriomeningitis virus (LCMV) infection. We found that NK-cell cytotoxicity was sufficient to protect mice from the fatal outcome that characterizes HLH-like disease and was also sufficient to reduce HLH-like manifestations. Mechanistically, NK-cell cytotoxicity reduced tissue infiltration by inflammatory macrophages and downmodulated LCMV-specific T-cell responses by limiting hyperactivation of CTL. Interestingly, the critical protective effect of NK cells on HLH was independent of interferon-γ secretion and changes in viral load. Therefore our findings identify a crucial role of NK-cell cytotoxicity in limiting HLH-like immunopathology, highlighting the important role of NK cytotoxic activity in immune homeostasis.


Assuntos
Citotoxicidade Imunológica/imunologia , Células Matadoras Naturais/imunologia , Coriomeningite Linfocítica/imunologia , Vírus da Coriomeningite Linfocítica/imunologia , Linfo-Histiocitose Hemofagocítica/imunologia , Linfócitos T Citotóxicos/imunologia , Animais , Proliferação de Células , Células Cultivadas , Interferon gama/metabolismo , Células Matadoras Naturais/patologia , Células Matadoras Naturais/virologia , Coriomeningite Linfocítica/patologia , Coriomeningite Linfocítica/virologia , Linfo-Histiocitose Hemofagocítica/patologia , Linfo-Histiocitose Hemofagocítica/virologia , Camundongos , Camundongos Endogâmicos C57BL , Baço/imunologia , Baço/patologia , Baço/virologia , Carga Viral
12.
Eur J Immunol ; 44(11): 3380-91, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25142413

RESUMO

To study gene functions specifically in NKp46+ cells we developed novel Cre mice allowing for conditional gene targeting in cells expressing Ncr1 (encoding NKp46). We generated transgenic Ncr1(greenCre) mice carrying an EGFPcre fusion under the control of a proximal Ncr1 promoter that faithfully directed EGFPcre expression to NKp46+ cells from lymphoid and nonlymphoid tissues. This approach allowed for direct detection of Cre-expressing NKp46+ cells via their GFP signature by flow cytometry and histology. Cre was functional as evidenced by the NKp46+ cell-specific expression of RFP in Ncr1(greenCre) Rosa-dtRFP reporter mice. We generated Ncr1(greenCre) Il2rg(fl/fl) mice that lack NKp46+ cells in an otherwise intact hematopoietic environment. Il2rg encodes the common gamma chain (γc ), which is an essential receptor subunit for cytokines (IL-2, -4, -7, -9, -15, and -21) that stimulate lymphocyte development and function. In Ncr1(greenCre) Il2rg(fl/fl) mice, NK cells are severely reduced and the few remaining NKp46+ cells escaping γc deletion failed to express GFP. Using this new NK-cell-deficient model, we demonstrate that the homeostasis of NKp46+ cells from all tissues (including the recently described intraepithelial ILC1 subset) requires Il2rg. Finally, Ncr1(greenCre) Il2rg(fl/fl) mice are unable to reject B16 lung metastases demonstrating the essential role of NKp46+ cells in antimelanoma immune responses.


Assuntos
Antígenos Ly/genética , Células Matadoras Naturais/imunologia , Neoplasias Pulmonares/secundário , Melanoma Experimental/patologia , Receptor 1 Desencadeador da Citotoxicidade Natural/genética , Animais , Antígenos Ly/biossíntese , Linfócitos B/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Proteínas de Fluorescência Verde/genética , Subunidade gama Comum de Receptores de Interleucina/genética , Neoplasias Pulmonares/imunologia , Melanoma Experimental/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptor 1 Desencadeador da Citotoxicidade Natural/biossíntese
13.
J Exp Med ; 211(2): 199-208, 2014 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-24419270

RESUMO

Group 3 innate lymphoid cells (ILC3) include IL-22-producing NKp46(+) cells and IL-17A/IL-22-producing CD4(+) lymphoid tissue inducerlike cells that express RORγt and are implicated in protective immunity at mucosal surfaces. Whereas the transcription factor Gata3 is essential for T cell and ILC2 development from hematopoietic stem cells (HSCs) and for IL-5 and IL-13 production by T cells and ILC2, the role for Gata3 in the generation or function of other ILC subsets is not known. We found that abundant GATA-3 protein is expressed in mucosa-associated ILC3 subsets with levels intermediate between mature B cells and ILC2. Chimeric mice generated with Gata3-deficient fetal liver hematopoietic precursors lack all intestinal RORγt(+) ILC3 subsets, and these mice show defective production of IL-22 early after infection with the intestinal pathogen Citrobacter rodentium, leading to impaired survival. Further analyses demonstrated that ILC3 development requires cell-intrinsic Gata3 expression in fetal liver hematopoietic precursors. Our results demonstrate that Gata3 plays a generalized role in ILC lineage determination and is critical for the development of gut RORγt(+) ILC3 subsets that maintain mucosal barrier homeostasis. These results further extend the paradigm of Gata3-dependent regulation of diversified innate ILC and adaptive T cell subsets.


Assuntos
Fator de Transcrição GATA3/imunologia , Imunidade Inata , Subpopulações de Linfócitos/imunologia , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/imunologia , Imunidade Adaptativa , Animais , Citrobacter rodentium , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/imunologia , Infecções por Enterobacteriaceae/imunologia , Feminino , Desenvolvimento Fetal/imunologia , Fator de Transcrição GATA3/deficiência , Fator de Transcrição GATA3/genética , Imunidade nas Mucosas , Interleucinas/metabolismo , Fígado/citologia , Fígado/embriologia , Fígado/imunologia , Subpopulações de Linfócitos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/metabolismo , Gravidez , Interleucina 22
14.
Mol Ther ; 21(10): 1950-7, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23975040

RESUMO

The development of innovative therapeutic strategies for muscular dystrophies, particularly cell-based approaches, is still a developing field. Although positive results have been obtained in animal models, they have rarely been confirmed in patients and resulted in very limited clinical improvements, suggesting some specificity in humans. These findings emphasized the need for an appropriate animal model (i.e., immunodeficient and dystrophic) to investigate in vivo the behavior of transplanted human myogenic stem cells. We report a new model, the Rag2(-)Il2rb(-)Dmd(-) mouse, which lacks T, B, and NK cells, and also carries a mutant Dmd allele that prevents the production of any dystrophin isoform. The dystrophic features of this new model are comparable with those of the classically used mdx mouse, but with the total absence of any revertant dystrophin positive fiber. We show that Rag2(-)Il2rb(-)Dmd(-) mice allow long-term xenografts of human myogenic cells. Altogether, our findings indicate that the Rag2(-)Il2rb(-)Dmd(-) mouse represents an ideal model to gain further insights into the behavior of human myogenic stem cells in a dystrophic context, and can be used to assess innovative therapeutic strategies for muscular dystrophies.


Assuntos
Proteínas de Ligação a DNA/genética , Modelos Animais de Doenças , Distrofina/genética , Subunidade beta de Receptor de Interleucina-2/genética , Camundongos Endogâmicos mdx/genética , Distrofias Musculares/patologia , Distrofia Muscular Animal/patologia , Animais , Terapia Baseada em Transplante de Células e Tecidos/métodos , Técnicas de Inativação de Genes , Humanos , Recém-Nascido , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Distrofias Musculares/terapia , Distrofia Muscular Animal/terapia , Mioblastos/transplante , Transplante Heterólogo , Ensaios Antitumorais Modelo de Xenoenxerto
15.
Proc Natl Acad Sci U S A ; 110(25): 10240-5, 2013 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-23733962

RESUMO

Group 2 innate lymphoid cells (ILC2s; also called nuocytes, innate helper cells, or natural helper cells) provide protective immunity during helminth infection and play an important role in influenza-induced and allergic airway hyperreactivity. Whereas the transcription factor GATA binding protein 3 (Gata3) is important for the production of IL-5 and -13 by ILC2s in response to IL-33 or -25 stimulation, it is not known whether Gata3 is required for ILC2 development from hematopoietic stem cells. Here, we show that chimeric mice generated with Gata3-deficient fetal liver hematopoietic stem cells fail to develop systemically dispersed ILC2s. In these chimeric mice, in vivo administration of IL-33 or -25 fails to expand ILC2 numbers or to induce characteristic ILC2-dependent IL-5 or -13 production. Moreover, cell-intrinsic Gata3 expression is required for ILC2 development in vitro and in vivo. Using mutant and transgenic mice in which Gata3 gene copy number is altered, we show that ILC2 generation from common lymphoid progenitors, as well as ILC2 homeostasis and cytokine production, is regulated by Gata3 expression levels in a dose-dependent fashion. Collectively, these results identify Gata3 as a critical early regulator of ILC2 development, thereby extending the paradigm of Gata3-dependent control of type 2 immunity to include both innate and adaptive lymphocytes.


Assuntos
Fator de Transcrição GATA3/genética , Interleucina-13/genética , Interleucina-5/genética , Linfócitos/imunologia , Animais , Asma/genética , Asma/imunologia , Fator de Transcrição GATA3/imunologia , Dosagem de Genes/genética , Dosagem de Genes/imunologia , Homeostase/imunologia , Imunidade Inata/genética , Imunidade Inata/imunologia , Inflamação/induzido quimicamente , Inflamação/imunologia , Interleucina-13/imunologia , Interleucina-33 , Interleucina-5/imunologia , Interleucinas/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
16.
Curr Opin Immunol ; 25(2): 130-8, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23490162

RESUMO

In recent years we have witnessed a blooming interest in innate lymphoid cell (ILC) biology thanks to the discovery of novel lineages of ILC that are phenotypically and functionally distinct from NK cells. While the importance of these novel ILC subsets as essential functional components of the early immune responses are now clearly established, many questions remain as to how early ILC developmental fates are determined and how specific effector functions associated with individual ILC subsets are achieved. As the founding member of the ILC family, properties of NK cells have defining attributes that characterize this group of innate effectors. Analysing their developmental rules may provide clues to principles that guide ILC development in general.


Assuntos
Imunidade Inata/imunologia , Células Matadoras Naturais/citologia , Células Matadoras Naturais/imunologia , Linfócitos/citologia , Linfócitos/imunologia , Animais , Humanos
17.
Eur J Immunol ; 41(3): 780-6, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21341264

RESUMO

The natural cytotoxicity receptor NKp46 is an activating receptor expressed by several distinct innate lymphoid cell (ILC) subsets, including NK cells, some γδ T cells and intestinal RORγt(+) IL-22(+) cells (NCR22 cells, IL-22-producing NKp46(+) cell). NCR22 cells may play a role in mucosal barrier function through IL-22-mediated production of anti-bacterial peptides from intestinal epithelial cells. Previous studies identified a predominant proportion of NCR22 cells in gut cryptopatches (CP), lymphoid structures that are strategically positioned to collect and integrate signals from luminal microbes; however, whether CP or other lymphoid structures condition NCR22 cell differentiation is not known. Programmed and inducible lymphoid tissue development requires cell-surface-expressed lymphotoxin (LT)α(1) ß(2) heterotrimers (provided by lymphoid tissue inducer (LTi) cells) to signal lymphotoxin-ß receptor (LTR)(+) stromal cells. Here, we analyzed NCR22 cells in LTßR-deficient Ncr1(GFP/+) mice that lack organized secondary lymphoid tissues. We found that NCR22 cells develop in the absence of LTßR, become functionally competent and localize to the lamina propria under steady-state conditions. Following infection of LTßR(-/-) mice with the Gram-negative pathogen Citrobacter rodentium, IL-22 production from NCR22 cells was not affected. These results indicate that organized lymphoid tissue structures are not critical for the generation of an intact and fully functional intestinal NCR22 cell compartment.


Assuntos
Antígenos Ly/metabolismo , Interleucinas/biossíntese , Mucosa Intestinal/citologia , Mucosa Intestinal/imunologia , Células Matadoras Naturais/imunologia , Receptor beta de Linfotoxina/imunologia , Receptor 1 Desencadeador da Citotoxicidade Natural/metabolismo , Animais , Citrobacter rodentium , Infecções por Enterobacteriaceae/imunologia , Infecções por Enterobacteriaceae/patologia , Imunidade Inata , Subunidade alfa de Receptor de Interleucina-7/metabolismo , Células Matadoras Naturais/classificação , Células Matadoras Naturais/metabolismo , Receptor beta de Linfotoxina/deficiência , Receptor beta de Linfotoxina/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/metabolismo , Transdução de Sinais , Interleucina 22
18.
Eur J Immunol ; 40(12): 3347-57, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21110318

RESUMO

Chronic inflammation is associated with promotion of malignancy and tumor progression. Many tumors enhance the accumulation of myeloid-derived suppressor cells (MDSC), which contribute to tumor progression and growth by suppressing anti-tumor immune responses. Tumor-derived IL-1ß secreted into the tumor microenvironment has been shown to induce the accumulation of MDSC possessing an enhanced capacity to suppress T cells. In this study, we found that the enhanced suppressive potential of IL-1ß-induced MDSC was due to the activity of a novel subset of MDSC lacking Ly6C expression. This subset was present at low frequency in tumor-bearing mice in the absence of IL-1ß-induced inflammation; however, under inflammatory conditions, Ly6C(neg) MDSC were predominant. Ly6C(neg) MDSC impaired NK cell development and functions in vitro and in vivo. These results identify a novel IL-1ß-induced subset of MDSC with unique functional properties. Ly6C(neg) MDSC mediating NK cell suppression may thus represent useful targets for therapeutic interventions.


Assuntos
Carcinoma/metabolismo , Interleucina-1beta/metabolismo , Células Matadoras Naturais/metabolismo , Células Mieloides/metabolismo , Neoplasias Experimentais/metabolismo , Animais , Antígenos de Diferenciação/biossíntese , Antígenos de Diferenciação/metabolismo , Antígenos Ly/biossíntese , Carcinoma/genética , Carcinoma/imunologia , Carcinoma/patologia , Diferenciação Celular , Linhagem Celular Tumoral , Citotoxicidade Imunológica , Tolerância Imunológica , Vigilância Imunológica , Mediadores da Inflamação/imunologia , Interleucina-1beta/genética , Interleucina-1beta/imunologia , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Células Mieloides/imunologia , Células Mieloides/patologia , Neoplasias Experimentais/genética , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/patologia , Evasão Tumoral
19.
J Immunol ; 185(9): 4993-7, 2010 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-20889548

RESUMO

Although NK cells in the mouse are thought to develop in the bone marrow, a small population of NK cells in the thymus has been shown to derive from a GATA3-dependent pathway. Characteristically, thymic NK cells express CD127 and few Ly49 molecules and lack CD11b. Because these NK cells develop in the thymus, the question of their relationship to the T cell lineage has been raised. Using several different mouse models, we find that unlike T cells, thymic NK cells are not the progeny of Rorc-expressing progenitors and do not express Rag2 or rearrange the TCRγ locus. We further demonstrate that thymic NK cells develop independently of the Notch signaling pathway, supporting the idea that thymic NK cells represent bona fide NK cells that can develop independently of all T cell precursors.


Assuntos
Diferenciação Celular/imunologia , Linhagem da Célula/imunologia , Células-Tronco Hematopoéticas/citologia , Células Matadoras Naturais/citologia , Timo/citologia , Animais , Separação Celular , Citometria de Fluxo , Células-Tronco Hematopoéticas/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Reação em Cadeia da Polimerase , Células Precursoras de Linfócitos T/citologia
20.
Immunol Rev ; 238(1): 126-37, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20969589

RESUMO

T lymphocytes depend on the thymic microenvironment for initiation of the T-cell developmental program. As the progenitors in the thymus have lost the capacity to self-renew, this process depends on the constant influx of hematopoietic progenitors that originate in the bone marrow. Nevertheless, thymic emigrants are heterogeneous and retain developmental plasticity for both the myeloid and lymphoid lineages. It is the role of the thymic microenvironment to steer these uncommitted progenitors toward a T-cell fate. Still, the thymus also generates a unique population of thymic NK cells, thus raising the question of how the T versus NK lymphoid cell fate is determined intrathymically. Many factors have been implicated in the developmental pathways in the thymus, and the processes are characterized by both subtle and not so subtle modifications in gene expression. In this review, we consider the crucial factors governing lineage determination of T cells versus NK cells from bi-potent thymic NK/T precursors. Recent reports have shed new light on the complex interactions of cytokines and transcription factors at different cell fate decision branch points in thymopoiesis. We discuss the implications of these findings and propose a model that may be applicable at this critical thymic NK/T juncture.


Assuntos
Células Matadoras Naturais/imunologia , Linfócitos T/imunologia , Timo/imunologia , Animais , Diferenciação Celular , Linhagem da Célula , Citocinas/imunologia , Humanos , Células Progenitoras Linfoides/imunologia , Modelos Imunológicos , Células Progenitoras Mieloides/imunologia , Fatores de Transcrição/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...