Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Nat Immunol ; 23(6): 904-915, 2022 06.
Article in English | MEDLINE | ID: mdl-35618834

ABSTRACT

Malignancy can be suppressed by the immune system. However, the classes of immunosurveillance responses and their mode of tumor sensing remain incompletely understood. Here, we show that although clear cell renal cell carcinoma (ccRCC) was infiltrated by exhaustion-phenotype CD8+ T cells that negatively correlated with patient prognosis, chromophobe RCC (chRCC) had abundant infiltration of granzyme A-expressing intraepithelial type 1 innate lymphoid cells (ILC1s) that positively associated with patient survival. Interleukin-15 (IL-15) promoted ILC1 granzyme A expression and cytotoxicity, and IL-15 expression in chRCC tumor tissue positively tracked with the ILC1 response. An ILC1 gene signature also predicted survival of a subset of breast cancer patients in association with IL-15 expression. Notably, ILC1s directly interacted with cancer cells, and IL-15 produced by cancer cells supported the expansion and anti-tumor function of ILC1s in a murine breast cancer model. Thus, ILC1 sensing of cancer cell IL-15 defines an immunosurveillance mechanism of epithelial malignancies.


Subject(s)
Breast Neoplasms , Interleukin-15/metabolism , Animals , Breast Neoplasms/genetics , CD8-Positive T-Lymphocytes , Female , Granzymes , Humans , Immunity, Innate , Lymphocytes , Mice
3.
Nat Immunol ; 17(12): 1388-1396, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27798617

ABSTRACT

Fibroblastic reticular cells (FRCs) of secondary lymphoid organs form distinct niches for interaction with hematopoietic cells. We found here that production of the cytokine IL-15 by FRCs was essential for the maintenance of group 1 innate lymphoid cells (ILCs) in Peyer's patches and mesenteric lymph nodes. Moreover, FRC-specific ablation of the innate immunological sensing adaptor MyD88 unleashed IL-15 production by FRCs during infection with an enteropathogenic virus, which led to hyperactivation of group 1 ILCs and substantially altered the differentiation of helper T cells. Accelerated clearance of virus by group 1 ILCs precipitated severe intestinal inflammatory disease with commensal dysbiosis, loss of intestinal barrier function and diminished resistance to colonization. In sum, FRCs act as an 'on-demand' immunological 'rheostat' by restraining activation of group 1 ILCs and thereby preventing immunopathological damage in the intestine.


Subject(s)
Citrobacter rodentium/immunology , Coronavirus Infections/immunology , Enterobacteriaceae Infections/immunology , Fibroblasts/immunology , Interleukin-15/metabolism , Lymphocytes/immunology , Murine hepatitis virus/immunology , Animals , Cells, Cultured , Immunity, Innate , Lymph Nodes/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Peyer's Patches/pathology , Th1 Cells/immunology , Toll-Like Receptor 7/genetics , Toll-Like Receptor 7/metabolism
4.
Immunity ; 48(2): 286-298.e6, 2018 02 20.
Article in English | MEDLINE | ID: mdl-29396162

ABSTRACT

Glucocorticoids are steroid hormones with strong anti-inflammatory and immunosuppressive effects that are produced in a diurnal fashion. Although glucocorticoids have the potential to induce interleukin-7 receptor (IL-7R) expression in T cells, whether they control T cell homeostasis and responses at physiological concentrations remains unclear. We found that glucocorticoid receptor signaling induces IL-7R expression in mouse T cells by binding to an enhancer of the IL-7Rα locus, with a peak at midnight and a trough at midday. This diurnal induction of IL-7R supported the survival of T cells and their redistribution between lymph nodes, spleen, and blood by controlling expression of the chemokine receptor CXCR4. In mice, T cell accumulation in the spleen at night enhanced immune responses against soluble antigens and systemic bacterial infection. Our results reveal the immunoenhancing role of glucocorticoids in adaptive immunity and provide insight into how immune function is regulated by the diurnal rhythm.


Subject(s)
Circadian Rhythm/physiology , Glucocorticoids/pharmacology , Receptors, CXCR4/physiology , Receptors, Interleukin-7/physiology , T-Lymphocytes/immunology , Animals , Cells, Cultured , Chemokine CXCL12/biosynthesis , Female , Immunologic Memory , Lymphocyte Activation , Male , Mice , Mice, Inbred C57BL , Receptors, Glucocorticoid/physiology
5.
Proc Natl Acad Sci U S A ; 120(36): e2215941120, 2023 09 05.
Article in English | MEDLINE | ID: mdl-37639581

ABSTRACT

Group 2 innate lymphoid cells (ILC2s) are critical for the immune response against parasite infection and tissue homeostasis and involved in the pathogenesis of allergy and inflammatory diseases. Although multiple molecules positively regulating ILC2 development and activation have been extensively investigated, the factors limiting their population size and response remain poorly studied. Here, we found that CD45, a membrane-bound tyrosine phosphatase essential for T cell development, negatively regulated ILC2s in a cell-intrinsic manner. ILC2s in CD45-deficient mice exhibited enhanced proliferation and maturation in the bone marrow and hyperactivated phenotypes in the lung with high glycolytic capacity. Furthermore, CD45 signaling suppressed the type 2 inflammatory response by lung ILC2s and alleviated airway inflammation and pulmonary fibrosis. Finally, the interaction with galectin-9 influenced CD45 signaling in ILC2s. These results demonstrate that CD45 is a cell-intrinsic negative regulator of ILC2s and prevents lung inflammation and fibrosis via ILC2s.


Subject(s)
Pulmonary Fibrosis , Animals , Mice , Pulmonary Fibrosis/prevention & control , Immunity, Innate , Lymphocytes , Inflammation , Signal Transduction
6.
Int Immunol ; 35(11): 513-530, 2023 Nov 07.
Article in English | MEDLINE | ID: mdl-37493250

ABSTRACT

Interleukin-7 (IL-7) is a cytokine critical for the development and maintenance of group 2 innate lymphoid cells (ILC2s). ILC2s are resident in peripheral tissues such as the intestine and lung. However, whether IL-7 produced in the lung plays a role in the maintenance and function of lung ILC2s during airway inflammation remains unknown. IL-7 was expressed in bronchoalveolar epithelial cells and lymphatic endothelial cells (LECs). To investigate the role of local IL-7 in lung ILC2s, we generated two types of IL-7 conditional knockout (IL-7cKO) mice: Sftpc-Cre (SPC-Cre) IL-7cKO mice specific for bronchial epithelial cells and type 2 alveolar epithelial cells and Lyve1-Cre IL-7cKO mice specific for LECs. In steady state, ILC2s were located near airway epithelia, although lung ILC2s were unchanged in the two lines of IL-7cKO mice. In papain-induced airway inflammation dependent on innate immunity, lung ILC2s localized near bronchia via CCR4 expression, and eosinophil infiltration and type 2 cytokine production were reduced in SPC-Cre IL-7cKO mice. In contrast, in house dust mite (HDM)-induced airway inflammation dependent on adaptive immunity, lung ILC2s localized near lymphatic vessels via their CCR2 expression 2 weeks after the last challenge. Furthermore, lung ILC2s were decreased in Lyve1-Cre IL-7cKO mice in the HDM-induced inflammation because of decreased cell survival and proliferation. Finally, administration of anti-IL-7 antibody attenuated papain-induced inflammation by suppressing the activation of ILC2s. Thus, this study demonstrates that IL-7 produced by bronchoalveolar epithelial cells and LECs differentially controls the activation and maintenance of lung ILC2s, where they are localized in airway inflammation.


Subject(s)
Immunity, Innate , Interleukin-7 , Mice , Animals , Endothelial Cells/metabolism , Papain , Lymphocytes , Lung , Adaptive Immunity , Inflammation , Cytokines/metabolism , Interleukin-33
7.
J Immunol ; 209(6): 1083-1094, 2022 09 15.
Article in English | MEDLINE | ID: mdl-35977797

ABSTRACT

Asthma is more common in females than males after adolescence. However, the mechanism of the sex bias in the prevalence of asthma remains unknown. To test whether sex steroid hormones have some roles in T cells during development of asthma, we analyzed airway inflammation in T cell-specific androgen receptor (AR)- and estrogen receptor (ER)-deficient mice. T cell-specific AR-deficient male mice developed severer house dust mite-induced allergic airway inflammation than did control male mice, whereas T cell-specific ERα- and ERß-deficient female mice exhibited a similar degree of inflammation as for control female mice. Furthermore, administration of dihydrotestosterone reduced cytokine production of Th2 cells from control, but not AR-deficient, naive T cells. Transfer of OT-II transgenic AR-deficient Th2 cells into wild-type mice induced severer allergic airway inflammation by OVA than transfer of control Th2 cells. Gene expression profiling suggested that the expression of genes related with cell cycle and Th2 differentiation was elevated in AR-deficient Th2 cells, whereas expression of dual specificity phosphatase (DUSP)-2, a negative regulator of p38, was downregulated. In addition, a chromatin immunoprecipitation assay suggested that AR bound to an AR motif in the 5' untranslated region of the Dusp2 gene in Th2 cells. Furthermore, the Dusp2 promoter with a wild-type AR motif, but not a mutated motif, was transactivated by dihydrotestosterone in a reporter assay. Finally, forced expression of DUSP-2 by retrovirus vector reduced IL-4 expression in Th2 cells. Thus, these results suggest that androgen signaling suppresses cytokine production of Th2 cells by inducing DUSP-2, explaining, in part, the sex bias of asthma after adolescence.


Subject(s)
Asthma , Hypersensitivity , 5' Untranslated Regions , Androgens/metabolism , Animals , Asthma/genetics , Asthma/metabolism , Dihydrotestosterone , Disease Models, Animal , Dual-Specificity Phosphatases/metabolism , Estrogen Receptor alpha/genetics , Estrogen Receptor beta/genetics , Female , Hypersensitivity/metabolism , Inflammation/metabolism , Interleukin-4/genetics , Interleukin-4/metabolism , Male , Mice , Mice, Knockout , Receptors, Androgen/genetics , Receptors, Estrogen/genetics , Th17 Cells/metabolism , Th2 Cells/metabolism
8.
Adv Exp Med Biol ; 1444: 111-127, 2024.
Article in English | MEDLINE | ID: mdl-38467976

ABSTRACT

Recently, considerable attention has been directed toward innate-like T cells (ITCs) and innate lymphoid cells (ILCs) owing to their indispensable contributions to immune responses, tissue homeostasis, and inflammation. Innate-like T cells include NKT cells, MAIT cells, and γδ T cells, whereas ILCs include NK cells, type 1 ILCs (ILC1s), type 2 ILCs (ILC2s), and type 3 ILCs (ILC3s). Many of these ITCs and ILCs are distributed to specific tissues and remain tissue-resident, while others, such as NK cells and some γδ T cells, circulate through the bloodstream. Nevertheless, recent research has shed light on novel subsets of innate immune cells that exhibit characteristics intermediate between tissue-resident and circulating states under normal and pathological conditions. The local microenvironment frequently influences the development, distribution, and function of these innate immune cells. This review aims to consolidate the current knowledge on the functional heterogeneity of ITCs and ILCs, shaped by local environmental cues, with particular emphasis on IL-15, which governs the activities of the innate immune cells involved in type 1 immune responses.


Subject(s)
Immunity, Innate , Lymphocytes , Humans , Killer Cells, Natural , Inflammation
9.
Curr Top Microbiol Immunol ; 434: 83-101, 2021.
Article in English | MEDLINE | ID: mdl-34850283

ABSTRACT

Lymphoid organs consist of immune cells and stromal cells. The stromal cells produce various cytokines that support the development, maintenance, and response of the immune cells. IL-7 and IL-15 are the major cytokines produced by stromal cells and are essential for the development and maintenance of lymphocytes and innate lymphoid cells (ILCs). In addition, IL-7 is indispensable for the organogenesis of lymphoid organs. However, because the amount of these two cytokines is relatively low, it has been difficult to directly detect their expression. Recently, several groups succeeded in establishing IL-7 and IL-15 reporter mouse lines. As expected, IL-7 and IL-15 were detected in mesenchymal stromal cells in the bone marrow and lymph nodes and in epithelial cells in the thymus. Furthermore, IL-7 and IL-15 were differentially expressed in lymphatic endothelial cells and blood endothelial cells, respectively. In addition to their expression, many groups have analyzed the local functions of IL-7 and IL-15 by using cell-type-specific knockout mice. From these experiments, CXCL12-expressing mesenchymal stromal cells were identified as the major niche for early B cell precursors. Single-cell RNA sequencing (scRNA-seq) analysis has revealed different subpopulations of stromal cells in the lymphoid organs, including those that express both IL-7 and IL-15. Future research is still needed to elucidate which stromal cells serve as the niche for the early precursors of ILCs and NK cells in the bone marrow.


Subject(s)
Interleukin-15 , Interleukin-7 , Animals , Endothelial Cells , Immunity, Innate , Interleukin-15/genetics , Interleukin-7/genetics , Killer Cells, Natural , Mice
10.
Curr Top Microbiol Immunol ; 434: 103-121, 2021.
Article in English | MEDLINE | ID: mdl-34850284

ABSTRACT

Lymph nodes (LNs) are secondary lymphoid organs that function as the first line of defense against invasive foreign substances. Within the LNs, different types of immune cells are strategically localized to induce immune responses efficiently. Such a sophisticated tissue structure is a complex of functionally specialized niches, constructed by a variety of fibroblastic stromal cells. Elucidating the characteristics and functions of the niches and stromal cells will facilitate comprehension of the immune response induced in the LNs. Three recent studies offered novel insights into specialized stromal cells. In our discussion of these surprisingly diverse stromal cells, we will integrate information from these studies to improve knowledge about the structure and niches of LN.


Subject(s)
Lymph Nodes , Stromal Cells , Immunity
11.
J Immunol ; 204(10): 2671-2684, 2020 05 15.
Article in English | MEDLINE | ID: mdl-32238459

ABSTRACT

TCR signaling is required for homeostasis of naive αß T cells. However, whether such a signal is necessary for γδ T cell homeostasis in the periphery remains unknown. In this study, we present evidence that a portion of Vγ2+ γδ T cells, one of the major γδ T cell subsets in the secondary lymphoid organs, requires TCR signaling for homeostasis. To attenuate γδTCR signals, we generated mice lacking Eγ4 (Eγ4-/-), an enhancer located at the 3'-most end of the TCRγ locus. Overall, we found that in thymus, Eγ4 loss altered V-J rearrangement, chromatin accessibility, and transcription of the TCRγ locus in a distance-dependent manner. Vγ2+ γδ T cells in Eγ4-/- mice developed normally both fetal and adult mouse thymi but were relatively reduced in number in spleen and lymph nodes. Although Vγ2 TCR transcription decreased in all subpopulations of Eγ4-/- mice, the number of Vγ2+ γδ T cells decreased and TCR signaling was attenuated only in the innate-like CD27+CD45RBhigh subpopulation in peripheral lymphoid organs. Consistently, CD27+CD45RBhigh Vγ2+ γδ T cells from Eγ4-/- mice transferred into Rag2-deficient mice were not efficiently recovered, suggesting that continuous TCR signaling is required for their homeostasis. Finally, CD27+CD45RBhigh Vγ2+ γδ T cells from Eγ4-/- mice showed impaired TCR-induced activation and antitumor responses. These results suggest that normal homeostasis of innate-like CD27+CD45RBhigh Vγ2+ γδ T cells in peripheral lymphoid organs requires TCR signaling.


Subject(s)
Germinal Center/immunology , Lymph Nodes/immunology , Lymphoid Tissue/immunology , Receptors, Antigen, T-Cell, gamma-delta/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocytes/immunology , Animals , Cells, Cultured , Enhancer Elements, Genetic/genetics , Homeostasis , Immunity, Innate , Leukocyte Common Antigens/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Peripheral Tolerance , Receptors, Antigen, T-Cell, gamma-delta/genetics , Signal Transduction , Tumor Necrosis Factor Receptor Superfamily, Member 7/metabolism
12.
J Immunol ; 204(4): 844-857, 2020 02 15.
Article in English | MEDLINE | ID: mdl-31924648

ABSTRACT

T cell development and homeostasis requires IL-7R α-chain (IL-7Rα) signaling. Tyrosine Y449 of the IL-7Rα is essential to activate STAT5 and PI3K, whereas PI3K recruitment requires IL-7Rα methionine M452. How IL-7Rα activates and regulates both signaling pathways differentially remains unclear. To characterize differential signaling, we established two lines of IL-7Rα mutant mice: IL-7R-Y449F mice and IL-7R-M452L mice. IL-7R-Y449F mice showed decreased PI3K and STAT5 signals, whereas IL-7R-M452L mice showed decreased PI3K but significantly increased STAT5 signaling, owing to a competition between PI3K and STAT5 signaling through Y449 of IL-7Rα. The number of T, B, and mature innate lymphoid cells were markedly reduced in IL-7R-Y449F mice, whereas IL-7R-M452L mice showed impaired early T cell development and memory precursor effector T cell maintenance with the downregulation of transcription factor T cell factor-1. Peripheral T cell numbers increased in IL-7R-M452L mice with enhanced survival and homeostatic proliferation. Furthermore, although wild type and IL-7R-Y449F mice showed comparable Th1/Th2 differentiation, IL-7R-M452L mice exhibited impaired Th17 differentiation. We conclude that PI3K competes with STAT5 under IL-7Rα and maintains an appropriate signal balance for modulating T cell development and homeostasis. To our knowledge, this study provides a new insight into complex regulation of IL-7Rα signaling, which supports immune development and responses.


Subject(s)
Homeostasis/immunology , Phosphatidylinositol 3-Kinases/immunology , Receptors, Interleukin-7/immunology , STAT5 Transcription Factor/immunology , T-Lymphocytes/immunology , Animals , Cell Differentiation/immunology , Immunity, Innate , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Phosphatidylinositol 3-Kinases/metabolism , Receptors, Interleukin-7/metabolism , STAT5 Transcription Factor/metabolism , Signal Transduction/immunology
13.
Int Immunol ; 32(5): 307-319, 2020 05 08.
Article in English | MEDLINE | ID: mdl-31875880

ABSTRACT

Interleukin-15 (IL-15) is a cytokine critical for maintenance of intestinal intra-epithelial lymphocytes (IELs), especially CD8αα + IELs (CD8αα IELs). In the intestine, IL-15 is produced by intestinal epithelial cells (IECs), blood vascular endothelial cells (BECs) and hematopoietic cells. However, the precise role of intestinal IL-15 on IELs is still unknown. To address the question, we generated two kinds of IL-15 conditional knockout (IL-15cKO) mice: villin-Cre (Vil-Cre) and Tie2-Cre IL-15cKO mice. IEC-derived IL-15 was specifically deleted in Vil-Cre IL-15cKO mice, whereas IL-15 produced by BECs and hematopoietic cells was deleted in Tie2-Cre IL-15cKO mice. The cell number and frequency of CD8αα IELs and NK IELs were significantly reduced in Vil-Cre IL-15cKO mice. By contrast, CD8αα IELs were unchanged in Tie2-Cre IL-15cKO mice, indicating that IL-15 produced by BECs and hematopoietic cells is dispensable for CD8αα IELs. Expression of an anti-apoptotic factor, Bcl-2, was decreased, whereas Fas expression was increased in CD8αα IELs of Vil-Cre IL-15cKO mice. Forced expression of Bcl-2 by a Bcl-2 transgene partially restored CD8αα IELs in Vil-Cre IL-15cKO mice, suggesting that some IL-15 signal other than Bcl-2 is required for maintenance of CD8αα IELs. Furthermore, granzyme B production was reduced, whereas PD-1 expression was increased in CD8αα IELs of Vil-Cre IL-15cKO mice. These results collectively suggested that IEC-derived IL-15 is essential for homeostasis of IELs by promoting their survival and functional maturation.


Subject(s)
Endothelial Cells/immunology , Interleukin-15/immunology , Intestines/cytology , Intestines/immunology , Intraepithelial Lymphocytes/cytology , Intraepithelial Lymphocytes/immunology , Animals , Female , Interleukin-15/deficiency , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic
14.
J Immunol ; 195(7): 3129-38, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26336149

ABSTRACT

The IL-7R plays critical roles in lymphocyte development and homeostasis. Although IL-7R expression is strictly regulated during lymphocyte differentiation and the immune response, little is known regarding its in vivo regulation. To address this issue, we established a mouse line with targeted deletion of the conserved non-coding sequence 1 (CNS1) element found 3.6 kb upstream of the IL-7Rα promoter. We report that IL-7Rα is expressed normally on T and B cells in thymus and bone marrow of CNS1(-/-) mice except for in regulatory T cells. In contrast, these mice show reduced IL-7Rα expression in conventional CD4 and CD8 T cells as well as regulatory T, NKT, and γδ T cells in the periphery. CD4 T cells of CNS1(-/-) mice showed IL-7Rα upregulation in the absence of growth factors and IL-7Rα downregulation by IL-7 or TCR stimulation, although the expression levels were lower than those in control mice. Naive CD4 and CD8 T cells of CNS1(-/-) mice show attenuated survival by culture with IL-7 and reduced homeostatic proliferation after transfer into lymphopenic hosts. CNS1(-/-) mice exhibit impaired maintenance of Ag-stimulated T cells. Furthermore, IL-7Rα upregulation by glucocorticoids and TNF-α was abrogated in CNS1(-/-) mice. This work demonstrates that the CNS1 element controls IL-7Rα expression and maintenance of peripheral T cells, suggesting differential regulation of IL-7Rα expression between central and peripheral lymphoid organs.


Subject(s)
CD4-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/cytology , Enhancer Elements, Genetic , Interleukin-7 Receptor alpha Subunit/genetics , Regulatory Sequences, Nucleic Acid/genetics , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Cell Proliferation/genetics , Cells, Cultured , Dexamethasone/pharmacology , Gene Expression/drug effects , Gene Expression/genetics , Interleukin-7 Receptor alpha Subunit/biosynthesis , Lymphocyte Activation/immunology , Mice , Mice, Knockout , Promoter Regions, Genetic/genetics , Sequence Deletion/genetics , Signal Transduction/immunology , Transcription, Genetic/genetics , Tumor Necrosis Factor-alpha/pharmacology
15.
Proc Natl Acad Sci U S A ; 111(5): 1915-20, 2014 Feb 04.
Article in English | MEDLINE | ID: mdl-24449915

ABSTRACT

IL-15 is a cytokine critical for development, maintenance, and response of T cells, natural killer (NK) cells, NK T cells, and dendritic cells. However, the identity and distribution of IL-15-expressing cells in lymphoid organs are not well understood. To address these questions, we established and analyzed IL-15-CFP knock-in mice. We found that IL-15 was highly expressed in thymic medulla, and medullary thymic epithelial cells with high MHC class II expression were the major source of IL-15. In bone marrow, IL-15 was detected primarily in VCAM-1(+)PDGFRß(+)CD31(-)Sca-1(-) stromal cells, which corresponded to previously described CXCL12-abundant reticular cells. In lymph nodes, IL-15-expressing cells were mainly distributed in the T-cell zone and medulla. IL-15 was expressed in some fibroblastic reticular cells and gp38(-)CD31(-) double-negative stromal cells in the T-cell zone. Blood endothelial cells, including all high endothelial venules, also expressed high IL-15 levels in lymph nodes, whereas lymphatic endothelial cells (LECs) lacked IL-15 expression. In spleen, IL-15 was expressed in VCAM-1(+) stromal cells, where its expression increased as mice aged. Finally, IL-15 expression in blood and LECs of peripheral lymphoid organs significantly increased in LPS-induced inflammation. Overall, we have identified and characterized several IL-15-expressing cells in primary and secondary lymphoid organs, providing a unique perspective of IL-15 niche in immune microenvironment. This study also suggests that some stromal cells express IL-7 and IL-15 differentially and suggests a way to functionally classify different stromal cell subsets.


Subject(s)
Interleukin-15/metabolism , Lymphoid Tissue/metabolism , Aging/immunology , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Gene Knock-In Techniques , Inflammation/pathology , Lipopolysaccharides/pharmacology , Lymph Nodes/cytology , Lymph Nodes/drug effects , Lymph Nodes/metabolism , Lymphoid Tissue/cytology , Lymphoid Tissue/drug effects , Mesoderm/cytology , Mesoderm/drug effects , Mice , Mucous Membrane/cytology , Mucous Membrane/drug effects , Mucous Membrane/metabolism , Spleen/cytology , Spleen/growth & development , Spleen/metabolism , Stromal Cells/cytology , Stromal Cells/drug effects , Stromal Cells/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Thymus Gland/cytology , Thymus Gland/drug effects , Thymus Gland/metabolism
16.
Front Immunol ; 15: 1349184, 2024.
Article in English | MEDLINE | ID: mdl-38440725

ABSTRACT

Invariant natural killer T (iNKT) cells are a distinct subpopulation of innate-like T lymphocytes. They are characterized by semi-invariant T cell receptors (TCRs) that recognize both self and foreign lipid antigens presented by CD1d, a non-polymorphic MHC class I-like molecule. iNKT cells play a critical role in stimulating innate and adaptive immune responses, providing an effective defense against infections and cancers, while also contributing to chronic inflammation. The functions of iNKT cells are specific to their location, ranging from lymphoid to non-lymphoid tissues, such as the thymus, lung, liver, intestine, and adipose tissue. This review aims to provide insights into the heterogeneity of development and function in iNKT cells. First, we will review the expression of master transcription factors that define subsets of iNKT cells and their production of effector molecules such as cytokines and granzymes. In this article, we describe the gene expression profiles contributing to the kinetics, distribution, and cytotoxicity of iNKT cells across different tissue types. We also review the impact of cytokine production in distinct immune microenvironments on iNKT cell heterogeneity, highlighting a recently identified circulating iNKT cell subset. Additionally, we explore the potential of exploiting iNKT cell heterogeneity to create potent immunotherapies for human cancers in the future.


Subject(s)
Natural Killer T-Cells , Neoplasms , Humans , Cues , Adipose Tissue , Cell Membrane , Tumor Microenvironment
17.
Elife ; 122023 Jun 22.
Article in English | MEDLINE | ID: mdl-37352115

ABSTRACT

Group 1 innate lymphoid cells (G1-ILCs), including circulating natural killer (NK) cells and tissue-resident type 1 ILCs (ILC1s), are innate immune sentinels critical for responses against infection and cancer. In contrast to relatively uniform NK cells through the body, diverse ILC1 subsets have been characterized across and within tissues in mice, but their developmental and functional heterogeneity remain unsolved. Here, using multimodal in vivo approaches including fate-mapping and targeting of the interleukin 15 (IL-15)-producing microenvironment, we demonstrate that liver parenchymal niches support the development of a cytotoxic ILC1 subset lacking IL-7 receptor (7 R- ILC1s). During ontogeny, fetal liver (FL) G1-ILCs arise perivascularly and then differentiate into 7 R- ILC1s within sinusoids. Hepatocyte-derived IL-15 supports parenchymal development of FL G1-ILCs to maintain adult pool of 7 R- ILC1s. IL-7R+ (7R+) ILC1s in the liver, candidate precursors for 7 R- ILC1s, are not essential for 7 R- ILC1 development in physiological conditions. Functionally, 7 R- ILC1s exhibit killing activity at steady state through granzyme B expression, which is underpinned by constitutive mTOR activity, unlike NK cells with exogenous stimulation-dependent cytotoxicity. Our study reveals the unique ontogeny and functions of liver-specific ILC1s, providing a detailed interpretation of ILC1 heterogeneity.


Subject(s)
Interleukin-15 , Lymphocytes , Mice , Animals , Lymphocytes/metabolism , Interleukin-15/metabolism , Immunity, Innate , Receptors, Interleukin-7/metabolism , Killer Cells, Natural , Liver
18.
Cell Rep ; 42(9): 113127, 2023 09 26.
Article in English | MEDLINE | ID: mdl-37729919

ABSTRACT

Natural killer (NK) cells are innate immune cells critical for protective immune responses against infection and cancer. Although NK cells differentiate in the bone marrow (BM) in an interleukin-15 (IL-15)-dependent manner, the cellular source of IL-15 remains elusive. Using NK cell reporter mice, we show that NK cells are localized in the BM in scattered and clustered manners. NK cell clusters overlap with monocyte and dendritic cell accumulations, whereas scattered NK cells require CXCR4 signaling. Using cell-specific IL-15-deficient mice, we show that hematopoietic cells, but not stromal cells, support NK cell development in the BM through IL-15. In particular, IL-15 produced by monocytes and dendritic cells appears to contribute to NK cell development. These results demonstrate that hematopoietic cells are the IL-15 niche for NK cell development in the BM and that BM NK cells are present in scattered and clustered compartments by different mechanisms, suggesting their distinct functions in the immune response.


Subject(s)
Bone Marrow , Interleukin-15 , Mice , Animals , Bone Marrow Cells , Cell Differentiation , Killer Cells, Natural
19.
Nat Commun ; 13(1): 863, 2022 02 22.
Article in English | MEDLINE | ID: mdl-35194029

ABSTRACT

The bacterial microbiota works as a community that consists of many individual organisms, i.e., cells. To fully understand the function of bacterial microbiota, individual cells must be identified; however, it is difficult with current techniques. Here, we develop a method, Barcoding Bacteria for Identification and Quantification (BarBIQ), which classifies single bacterial cells into taxa-named herein cell-based operational taxonomy units (cOTUs)-based on cellularly barcoded 16S rRNA sequences with single-base accuracy, and quantifies the cell number for each cOTU in the microbiota in a high-throughput manner. We apply BarBIQ to murine cecal microbiotas and quantify in total 3.4 × 105 bacterial cells containing 810 cOTUs. Interestingly, we find location-dependent global differences in the cecal microbiota depending on the dietary vitamin A deficiency, and more differentially abundant cOTUs at the proximal location than the distal location. Importantly, these location differences are not clearly shown by conventional 16S rRNA gene-amplicon sequencing methods, which quantify the 16S rRNA genes, not the cells. Thus, BarBIQ enables microbiota characterization with the identification and quantification of individual constituent bacteria, which is a cornerstone for microbiota studies.


Subject(s)
High-Throughput Nucleotide Sequencing , Microbiota , Animals , Bacteria/genetics , DNA, Bacterial/genetics , High-Throughput Nucleotide Sequencing/methods , Mice , Microbiota/genetics , RNA, Ribosomal, 16S/genetics , Sequence Analysis, DNA
20.
Sci Immunol ; 7(76): eabj8760, 2022 10 28.
Article in English | MEDLINE | ID: mdl-36269840

ABSTRACT

Invariant natural killer T (iNKT) cells are a group of innate-like T lymphocytes that recognize lipid antigens. They are supposed to be tissue resident and important for systemic and local immune regulation. To investigate the heterogeneity of iNKT cells, we recharacterized iNKT cells in the thymus and peripheral tissues. iNKT cells in the thymus were divided into three subpopulations by the expression of the natural killer cell receptor CD244 and the chemokine receptor CXCR6 and designated as C0 (CD244-CXCR6-), C1 (CD244-CXCR6+), or C2 (CD244+CXCR6+) iNKT cells. The development and maturation of C2 iNKT cells from C0 iNKT cells strictly depended on IL-15 produced by thymic epithelial cells. C2 iNKT cells expressed high levels of IFN-γ and granzymes and exhibited more NK cell-like features, whereas C1 iNKT cells showed more T cell-like characteristics. C2 iNKT cells were influenced by the microbiome and aging and suppressed the expression of the autoimmune regulator AIRE in the thymus. In peripheral tissues, C2 iNKT cells were circulating that were distinct from conventional tissue-resident C1 iNKT cells. Functionally, C2 iNKT cells protected mice from the tumor metastasis of melanoma cells by enhancing antitumor immunity and promoted antiviral immune responses against influenza virus infection. Furthermore, we identified human CD244+CXCR6+ iNKT cells with high cytotoxic properties as a counterpart of mouse C2 iNKT cells. Thus, this study reveals a circulating subset of iNKT cells with NK cell-like properties distinct from conventional tissue-resident iNKT cells.


Subject(s)
Natural Killer T-Cells , Mice , Humans , Animals , Natural Killer T-Cells/metabolism , Natural Killer T-Cells/pathology , Interleukin-15 , Antiviral Agents , Granzymes , Receptors, Natural Killer Cell , Receptors, Chemokine/metabolism , Lipids
SELECTION OF CITATIONS
SEARCH DETAIL