Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Cells ; 12(20)2023 10 11.
Article in English | MEDLINE | ID: mdl-37887277

ABSTRACT

Innate CD8 T cells are proinflammatory effector T cells that achieve functional maturation in the thymus prior to their export into and maturation in peripheral tissues. Innate CD8 T cells produce the Th1 cytokine IFNγ but depend on the Th2 cytokine IL-4 for their generation. Thus, innate CD8 T cells can permute the intrathymic cytokine milieu by consuming a Th2 cytokine but driving a Th1 cytokine response. The cellular source of IL-4 is the NKT2 subset of invariant NKT (iNKT) cells. Consequently, NKT2 deficiency results in the lack of innate CD8 T cells. Whether NKT2 is the only iNKT subset and whether IL-4 is the only cytokine required for innate CD8 T cell generation, however, remains unclear. Here, we employed a mouse model of NKT1 deficiency, which is achieved by overexpression of the cytokine receptor IL-2Rß, and assessed the role of other iNKT subsets and cytokines in innate CD8 T cell differentiation. Because IL-2Rß-transgenic mice failed to generate both NKT1 and innate CD8 T cells, we postulated an in vivo requirement for IFNγ-producing NKT1 cells for innate CD8 T cell development. In-depth analyses of IL-2Rß-transgenic mice and IFNγ-deficient mice, however, demonstrated that neither NKT1 nor IFNγ was required to induce Eomes or to drive innate CD8 T cell generation. Instead, in vivo administration of recombinant IL-4 sufficed to restore the development of innate CD8 T cells in NKT1-deficient mice, affirming that intrathymic IL-4, and not IFNγ, is the limiting factor and key regulator of innate CD8 T cell generation in the thymus.


Subject(s)
Interleukin-4 , Thymus Gland , Mice , Animals , CD8-Positive T-Lymphocytes , Cytokines , Mice, Transgenic , Interferon-gamma , T-Box Domain Proteins/genetics
2.
J Autoimmun ; 138: 103059, 2023 07.
Article in English | MEDLINE | ID: mdl-37216869

ABSTRACT

Innate CD8 T cells correspond to a population of terminally differentiated effector T cells that phenotypically appear as antigen-experienced memory cells and functionally resemble proinflammatory CD8 T cells, expressing copious amounts of IFNγ. Innate CD8 T cells, however, are distinct from conventional effector-memory CD8 T cells as they acquire functional maturity during their generation in the thymus. Understanding the molecular mechanisms that drive their thymic development and differentiation is an intensely studied subject in T cell immunity, and here we identified the cytokine receptor γc as a critical mediator of innate CD8 T cell generation that promotes their selection even in the absence of classical MHC-I molecules. Consequently, overexpression of γc resulted in a dramatic increase of innate CD8 T cells in KbDb-deficient mice. We mapped its underlying mechanism to the expansion of IL-4-producing invariant NKT cells, so that it is the increased availability of intrathymic IL-4 which augments the selection of innate CD8 T cells. Collectively, these results unravel the selection of innate CD8 T cells being mediated by non-classical MHC-I molecules and being modulated by the abundance of the γc cytokine, IL-4.


Subject(s)
Natural Killer T-Cells , Receptors, Cytokine , Mice , Animals , Histocompatibility Antigens Class I/genetics , Interleukin-4 , Mice, Knockout , CD8-Positive T-Lymphocytes , Thymus Gland , Cell Differentiation , Mice, Inbred C57BL
3.
Cell Rep ; 38(2): 110219, 2022 01 11.
Article in English | MEDLINE | ID: mdl-35021100

ABSTRACT

Understanding the homeostatic mechanism of invariant natural killer T (iNKT) cells is a critical issue in iNKT cell biology. Because interleukin (IL)-15 is required for the thymic generation of iNKT cells, IL-15 has also been considered necessary for the homeostasis of peripheral iNKT cells. Here, we delineated the in vivo cytokine requirement for iNKT cells, and we came to the surprising conclusion that IL-7, not IL-15, is the homeostatic cytokine for iNKT cells. Employing a series of experimental mouse models where the availability of IL-7 or IL-15 was manipulated in peripheral tissues, either by genetic tools or by adult thymectomy and cytokine pump installation, we demonstrate that the abundance of IL-7, and not IL-15, limits the size of the peripheral iNKT cell pool. These results redefine the cytokine requirement for iNKT cells and indicate competition for IL-7 between iNKT and conventional αß T cells.


Subject(s)
Cell Differentiation/immunology , Interleukin-7/metabolism , Natural Killer T-Cells/metabolism , Animals , Cytokines/metabolism , Female , Homeostasis , Interleukin-7/immunology , Male , Mice , Mice, Inbred C57BL , Natural Killer T-Cells/immunology
4.
Cell Mol Life Sci ; 79(1): 17, 2021 Dec 31.
Article in English | MEDLINE | ID: mdl-34971407

ABSTRACT

Innate-like T (iT) cells comprise a population of immunoregulatory T cells whose effector function is imposed during their development in the thymus to provide protective immunity prior to antigen encounter. The molecular mechanism that drives the generation of iT cells remains unclear. Here, we report that the cytokine receptor γc plays a previously unappreciated role for thymic iT cells by controlling their cellular abundance, lineage commitment, and subset differentiation. As such, γc overexpression on thymocytes dramatically altered iT cell generation in the thymus, as it skewed the subset composition of invariant NKT (iNKT) cells and promoted the generation of IFNγ-producing innate CD8 T cells. Mechanistically, we found that the γc-STAT6 axis drives the differentiation of IL-4-producing iNKT cells, which in turn induced the generation of innate CD8 T cells. Collectively, these results reveal a cytokine-driven circuity of thymic iT cell differentiation that is controlled by the abundance of γc proteins.


Subject(s)
Immunity, Innate , Interleukin Receptor Common gamma Subunit/metabolism , T-Lymphocytes/metabolism , Thymus Gland/cytology , Animals , CD8-Positive T-Lymphocytes , Cell Differentiation , Cytokines/metabolism , Mice, Transgenic , Natural Killer T-Cells/metabolism , Promyelocytic Leukemia Zinc Finger Protein , STAT6 Transcription Factor/metabolism , Signal Transduction , Thymocytes/metabolism
5.
Front Immunol ; 12: 642856, 2021.
Article in English | MEDLINE | ID: mdl-34054809

ABSTRACT

Invariant NKT (iNKT) cells are thymus-generated innate-like T cells, comprised of three distinct subsets with divergent effector functions. The molecular mechanism that drives the lineage trifurcation of immature iNKT cells into the NKT1, NKT2, and NKT17 subsets remains a controversial issue that remains to be resolved. Because cytokine receptor signaling is necessary for iNKT cell generation, cytokines are proposed to contribute to iNKT subset differentiation also. However, the precise roles and requirements of cytokines in these processes are not fully understood. Here, we show that IL-2Rß, a nonredundant component of the IL-15 receptor complex, plays a critical role in both the development and differentiation of thymic iNKT cells. While the induction of IL-2Rß expression on postselection thymocytes is necessary to drive the generation of iNKT cells, surprisingly, premature IL-2Rß expression on immature iNKT cells was detrimental to their development. Moreover, while IL-2Rß is necessary for NKT1 generation, paradoxically, we found that the increased abundance of IL-2Rß suppressed NKT1 generation without affecting NKT2 and NKT17 cell differentiation. Thus, the timing and abundance of IL-2Rß expression control iNKT lineage fate and development, thereby establishing cytokine receptor expression as a critical regulator of thymic iNKT cell differentiation.


Subject(s)
Interleukin-2 Receptor beta Subunit/physiology , Natural Killer T-Cells/physiology , Thymus Gland/immunology , Animals , Cell Differentiation , Interleukin-15/pharmacology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Natural Killer T-Cells/classification , Natural Killer T-Cells/cytology , STAT5 Transcription Factor/physiology
7.
Exp Mol Med ; 41(3): 161-70, 2009 Mar 31.
Article in English | MEDLINE | ID: mdl-19293635

ABSTRACT

Increasing importance is being given to the stimulation of Th1 response in cancer immunotherapy because its presence can shift the direction of adaptive immune responses toward protective immunity. Based on chemokine receptor expression, CXCR3(+) CCR4(-) CD4(+) T cells as Th1-type cells were investigated its capacity in monocyte-derived dendritic cell (DC) maturation and polarization, and induction of antigen specific cytotoxic T lymphocytes (CTL) in vitro. The levels of IL-4, IL-5 and IL-10 were decreased to the basal level compared with high production of IFN-gamma, TNF-alpha, and IL-2 in CXCR3+CCR4-CD4+ T cells stimulated with anti-CD3 and anti-CD28 antibodies. Co-incubation of activated CD4(+) or CXCR3(+) CCR4-CD4(+) T cells with DC (CD4(+/) DC or CXCR3(+) CD4(+/) DC, respectively) particularly up-regulated IL-12 and CD80 expression compared with DC matured with TNF-a and LPS (mDC). Although there was no significant difference between the effects of the CXCR3(+) CCR4(-) CD4(+) and CD4(+) T cells on DC phenotype expression, CXCR3(+) CD4(+/) DC in CTL culture were able to expand number of CD8(+) T cells and increased frequencies of IFN-gamma secreting cells and overall cytolytic activity against tumor antigen WT-1. These results demonstrated that the selective addition of CXCR3(+) CCR4(-) CD4(+) T cells to CTL cultures could enhance the induction of CTLs by DC in vitro, and implicated on a novel strategy for adoptive T cell therapy.


Subject(s)
CD4 Antigens/immunology , Dendritic Cells/immunology , Receptors, CCR4/immunology , Receptors, CXCR3/immunology , T-Lymphocytes, Cytotoxic/cytology , Th1 Cells/immunology , Cell Line , Cells, Cultured , Cytokines/immunology , Cytotoxicity, Immunologic , Dendritic Cells/cytology , Humans , Interferon-gamma/biosynthesis , T-Lymphocytes, Cytotoxic/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...