Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 18 de 18
1.
Int J Mol Sci ; 23(19)2022 Sep 26.
Article En | MEDLINE | ID: mdl-36232644

The ability of natural killer (NK) cells to kill tumor cells without prior sensitization makes them a rising player in immunotherapy. Increased understanding of the development and functioning of NK cells will improve their clinical utilization. As opposed to murine NK cell development, human NK cell development is still less understood. Here, we studied the role of thioredoxin-interacting protein (TXNIP) in human NK cell differentiation by stable TXNIP knockdown or overexpression in cord blood hematopoietic stem cells, followed by in vitro NK cell differentiation. TXNIP overexpression only had marginal effects, indicating that endogenous TXNIP levels are sufficient in this process. TXNIP knockdown, however, reduced proliferation of early differentiation stages and greatly decreased NK cell numbers. Transcriptome analysis and experimental confirmation showed that reduced protein synthesis upon TXNIP knockdown likely caused this low proliferation. Contrary to its profound effects on the early differentiation stages, TXNIP knockdown led to limited alterations in NK cell phenotype, and it had no effect on NK cell cytotoxicity or cytokine production. Thus, TXNIP promotes human NK cell differentiation by affecting protein synthesis and proliferation of early NK cell differentiation stages, but it is redundant for functional NK cell maturation.


Killer Cells, Natural , Thioredoxins , Animals , Carrier Proteins/genetics , Cell Differentiation/genetics , Cytokines/metabolism , Gene Expression , Humans , Killer Cells, Natural/metabolism , Mice , Thioredoxins/genetics , Thioredoxins/metabolism
2.
Elife ; 112022 07 06.
Article En | MEDLINE | ID: mdl-35793229

Natural killer (NK) cells are innate lymphocytes that eliminate virus-infected and cancer cells by cytotoxicity and cytokine secretion. In addition to circulating NK cells, distinct tissue-resident NK subsets have been identified in various organs. Although transcription factors regulating NK cell development and function have been extensively studied in mice, the role of RUNX2 in these processes has not been investigated, neither in mice nor in human. Here, by manipulating RUNX2 expression with either knockdown or overexpression in human haematopoietic stem cell-based NK cell differentiation cultures, combined with transcriptomic and ChIP-sequencing analyses, we established that RUNX2 drives the generation of NK cells, possibly through induction of IL-2Rß expression in NK progenitor cells. Importantly, RUNX2 promotes tissue residency in human NK cells. Our findings have the potential to improve existing NK cell-based cancer therapies and can impact research fields beyond NK cell biology, since tissue-resident subsets have also been described in other lymphocyte subpopulations.


Transcription Factors , Humans , Core Binding Factor Alpha 1 Subunit/metabolism , Gene Expression Regulation , Killer Cells, Natural/metabolism , Transcription Factors/metabolism
3.
Front Immunol ; 12: 732511, 2021.
Article En | MEDLINE | ID: mdl-34630413

T-bet and Eomes are transcription factors that are known to be important in maturation and function of murine natural killer (NK) cells. Reduced T-BET and EOMES expression results in dysfunctional NK cells and failure to control tumor growth. In contrast to mice, the current knowledge on the role of T-BET and EOMES in human NK cells is rudimentary. Here, we ectopically expressed either T-BET or EOMES in human hematopoietic progenitor cells. Combined transcriptome, chromatin accessibility and protein expression analyses revealed that T-BET or EOMES epigenetically represses hematopoietic stem cell quiescence and non-NK lineage differentiation genes, while activating an NK cell-specific transcriptome and thereby drastically accelerating NK cell differentiation. In this model, the effects of T-BET and EOMES are largely overlapping, yet EOMES shows a superior role in early NK cell maturation and induces faster NK receptor and enhanced CD16 expression. T-BET particularly controls transcription of terminal maturation markers and epigenetically controls strong induction of KIR expression. Finally, NK cells generated upon T-BET or EOMES overexpression display improved functionality, including increased IFN-γ production and killing, and especially EOMES overexpression NK cells have enhanced antibody-dependent cellular cytotoxicity. Our findings reveal novel insights on the regulatory role of T-BET and EOMES in human NK cell maturation and function, which is essential to further understand human NK cell biology and to optimize adoptive NK cell therapies.


Cell Differentiation , Hematopoietic Stem Cells/metabolism , Killer Cells, Natural/metabolism , T-Box Domain Proteins/metabolism , Animals , Antibody-Dependent Cell Cytotoxicity , Cell Lineage , Chromatin Assembly and Disassembly , Coculture Techniques , Epigenesis, Genetic , Fetal Blood/cytology , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Hematopoietic Stem Cells/immunology , Humans , Interferon-gamma/metabolism , K562 Cells , Killer Cells, Natural/immunology , Mice , Phenotype , Receptors, IgG/genetics , Receptors, IgG/metabolism , Receptors, KIR/genetics , Receptors, KIR/metabolism , T-Box Domain Proteins/genetics , Transcriptome
4.
Blood ; 136(3): 288-298, 2020 07 16.
Article En | MEDLINE | ID: mdl-32350509

Natural killer (NK) cells are important in the immune defense against tumor cells and pathogens, and they regulate other immune cells by cytokine secretion. Although murine NK cell biology has been extensively studied, knowledge about transcriptional circuitries controlling human NK cell development and maturation is limited. By generating ETS1-deficient human embryonic stem cells and by expressing the dominant-negative ETS1 p27 isoform in cord blood hematopoietic progenitor cells, we show that the transcription factor ETS1 is critically required for human NK cell differentiation. Genome-wide transcriptome analysis determined by RNA-sequencing combined with chromatin immunoprecipitation-sequencing analysis reveals that human ETS1 directly induces expression of key transcription factors that control NK cell differentiation (ie, E4BP4, TXNIP, TBET, GATA3, HOBIT, BLIMP1). In addition, ETS1 regulates expression of genes involved in apoptosis and NK cell activation. Our study provides important molecular insights into the role of ETS1 as an important regulator of human NK cell development and terminal differentiation.


Cell Differentiation/immunology , Gene Expression Regulation/immunology , Human Embryonic Stem Cells/immunology , Killer Cells, Natural/immunology , Lymphocyte Activation , Proto-Oncogene Protein c-ets-1/immunology , Apoptosis/genetics , Apoptosis/immunology , Cell Differentiation/genetics , Cell Line , Gene Expression Profiling , Genome-Wide Association Study , Human Embryonic Stem Cells/cytology , Humans , Killer Cells, Natural/cytology , Protein Isoforms/genetics , Protein Isoforms/immunology , Proto-Oncogene Protein c-ets-1/genetics
5.
Eur Respir J ; 52(6)2018 12.
Article En | MEDLINE | ID: mdl-30385529

Although type 1, 2 and 3 innate lymphoid cells (ILC1s, ILC2s and ILC3s, respectively) are emerging as important cell populations regulating tissue homeostasis, remodelling and inflammation, a vast majority of our knowledge stems from in vitro and murine experiments, and requires thorough confirmation in human diseases.Relative levels of ILCs were evaluated by means of flow cytometry in freshly resected human upper airways mucosa of patients with chronic rhinosinusitis without nasal polyps (CRSsNP) and with nasal polyps (CRSwNP), taking into account the patient's clinical parameters and disease comorbidities.We report that the CD117 and interleukin-receptor type I (IL-1RI) expression status of human ILC2s depends on the local tissue environment. Only CD117+ IL-1RI+ ILC2s, exclusively present in CRSwNP, possess an interrelationship with type 2 T-helper cell cytokine and eosinophil levels in human upper airway mucosa. In CRSsNP, mainly CD117-IL-1RI- ILC2s are increased, yielding lower eosinophilia in this disease despite the high levels of ILC2s.These data unveil that the CD117- and CD117+ fractions within the native human ILC2 population are not a random phenomenon, in contrast to what could be concluded from in vitro data, and that the IL-1RI expression is not ubiquitous in ILC2s in vivo in humans, which cannot be assessed via in vitro and murine experiments.


Lymphocytes/cytology , Proto-Oncogene Proteins c-kit/metabolism , Receptors, Interleukin-1 Type I/metabolism , Rhinitis/immunology , Sinusitis/immunology , Adolescent , Adult , Aged , Chronic Disease , Eosinophils/cytology , Eosinophils/metabolism , Female , Flow Cytometry , Humans , Lymphocytes/metabolism , Male , Middle Aged , Nasal Polyps/complications , Rhinitis/complications , Sinusitis/complications , Young Adult
6.
Cell Rep ; 19(7): 1431-1443, 2017 05 16.
Article En | MEDLINE | ID: mdl-28514662

The Ly49E receptor is preferentially expressed on murine innate-like lymphocytes, such as epidermal Vγ3 T cells, intestinal intraepithelial CD8αα+ T lymphocytes, and CD49a+ liver natural killer (NK) cells. As the latter have recently been shown to be distinct from conventional NK cells and have innate lymphoid cell type 1 (ILC1) properties, we investigated Ly49E expression on intestinal ILC populations. Here, we show that Ly49E expression is very low on known ILC populations, but it can be used to define a previously unrecognized intraepithelial innate lymphoid population. This Ly49E-positive population is negative for NKp46 and CD8αα, expresses CD49a and CD103, and requires T-bet expression and IL-15 signaling for differentiation and/or survival. Transcriptome analysis reveals a group 1 ILC gene profile, different from NK cells, iCD8α cells, and intraepithelial ILC1. Importantly, NKp46-CD8αα-Ly49E+ cells produce interferon (IFN)-γ, suggesting that this previously unrecognized population may contribute to Th1-mediated immunity.


Antigens, Ly/metabolism , Epithelial Cells/metabolism , Immunity, Innate , Intestines/cytology , Lymphocytes/cytology , Lymphocytes/metabolism , Natural Cytotoxicity Triggering Receptor 1/metabolism , Animals , Antigens, CD/metabolism , Cell Shape , Cytotoxicity, Immunologic , Epithelial Cells/cytology , Interferon-gamma/biosynthesis , Killer Cells, Natural/metabolism , Mice , NK Cell Lectin-Like Receptor Subfamily A/metabolism , Phenotype , Signal Transduction , Transcription Factors/metabolism , Transcriptome/genetics
7.
Front Immunol ; 7: 472, 2016.
Article En | MEDLINE | ID: mdl-27891126

The protozoan parasite Trypanosoma cruzi circulates in the blood upon infection and invades various cells. Parasites intensively multiply during the acute phase of infection and persist lifelong at low levels in tissues and blood during the chronic phase. Natural killer (NK) and NKT cells play an important role in the immune control of T. cruzi infection, mainly by releasing the cytokine IFN-γ that activates the microbicidal action of macrophages and other cells and shapes a protective type 1 immune response. The mechanisms by which immune cells are regulated to produce IFN-γ during T. cruzi infection are still incompletely understood. Here, we show that urokinase plasminogen activator (uPA) is induced early upon T. cruzi infection and remains elevated until day 20 post-infection. We previously demonstrated that the inhibitory receptor Ly49E, which is expressed, among others, on NK and NKT cells, is triggered by uPA. Therefore, we compared wild type (WT) to Ly49E knockout (KO) mice for their control of experimental T. cruzi infection. Our results show that young, i.e., 4- and 6-week-old, Ly49E KO mice control the infection better than WT mice, indicated by a lower parasite load and less cachexia. The beneficial effect of Ly49E depletion is more obvious in 4-week-old male than in female mice and weakens in 8-week-old mice. In young mice, the lower T. cruzi parasitemia in Ly49E KO mice is paralleled by higher IFN-γ production compared to their WT controls. Our data indicate that Ly49E receptor expression inhibits the immune control of T. cruzi infection. This is the first demonstration that the inhibitory Ly49E receptor can interfere with the immune response to a pathogen in vivo.

8.
Cancer Immunol Immunother ; 65(11): 1365-1375, 2016 11.
Article En | MEDLINE | ID: mdl-27585789

Ly49E is a member of the Ly49 family of NK receptors and is distinct from other members of this family on the basis of its structural properties, expression pattern and ligand recognition. Importantly, Ly49E receptor expression is high on small intestinal and colonic intraepithelial lymphocytes (IELs). Intestinal IELs are regulators of the mucosal immune system and contribute to front-line defense at the mucosal barrier, including anti-tumor immune response. Whereas most Ly49 receptors have MHC class-I ligands, we showed that Ly49E is instead triggered by urokinase plasminogen activator (uPA). uPA has been extensively implicated in tumor development, where increased uPA expression correlates with poor prognosis. As such, we investigated the role of Ly49E receptor expression on intestinal IELs in the anti-tumor immune response. For this purpose, we compared Ly49E wild-type mice to Ly49E knockout mice in two established tumor models: ApcMin/+-mediated and azoxymethane-induced intestinal cancer. Our results indicate that Ly49E expression on IELs does not influence the development or progression of intestinal cancer.


Carcinoma in Situ/immunology , Epithelium/immunology , Intestinal Neoplasms/immunology , Lymphocytes/immunology , NK Cell Lectin-Like Receptor Subfamily A/metabolism , Adenomatous Polyposis Coli Protein/genetics , Animals , Azoxymethane , Carcinogenesis , Carcinoma in Situ/chemically induced , Carcinoma in Situ/genetics , Disease Models, Animal , Epithelium/pathology , Gene Expression Regulation, Neoplastic , Immunity, Cellular , Intestinal Neoplasms/chemically induced , Intestinal Neoplasms/genetics , Mice , Mice, Inbred Strains , Mice, Knockout , NK Cell Lectin-Like Receptor Subfamily A/genetics , Tumor Burden , Urokinase-Type Plasminogen Activator/genetics , Urokinase-Type Plasminogen Activator/metabolism
9.
Sci Rep ; 6: 30564, 2016 07 29.
Article En | MEDLINE | ID: mdl-27469529

Natural killer (NK) lymphocytes are part of the innate immune system and are important in immune protection against tumourigenesis. NK cells display a broad repertoire of activating and inhibitory cell surface receptors that regulate NK cell activity. The Ly49 family of NK receptors is composed of several members that recognize major histocompatibility complex class I (MHC-I) or MHC-I-related molecules. Ly49E is a unique inhibitory member, being triggered by the non-MHC-I-related protein urokinase plasminogen activator (uPA) in contrast to the known MHC-I-triggering of the other inhibitory Ly49 receptors. Ly49E also has an uncommon expression pattern on NK cells, including high expression on liver DX5(-) NK cells. Furthermore, Ly49E is the only Ly49 member expressed by epidermal γδ T cells. As γδ T cells and/or NK cells have been shown to be involved in the regulation of cutaneous, pulmonary and liver malignancies, and as uPA is involved in tumourigenesis, we investigated the role of the inhibitory Ly49E receptor in the anti-tumour immune response. We demonstrate that, although Ly49E is highly expressed on epidermal γδ T cells and liver NK cells, this receptor does not play a major role in the control of skin tumour formation or in lung and liver tumour development.


Immunity, Cellular , Killer Cells, Natural/immunology , Liver Neoplasms/immunology , Lung Neoplasms/immunology , NK Cell Lectin-Like Receptor Subfamily A/immunology , Neoplasm Proteins/immunology , Skin Neoplasms/immunology , T-Lymphocytes/immunology , Animals , Killer Cells, Natural/pathology , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice , Mice, Knockout , NK Cell Lectin-Like Receptor Subfamily A/genetics , Neoplasm Proteins/genetics , Receptors, Antigen, T-Cell, gamma-delta/genetics , Receptors, Antigen, T-Cell, gamma-delta/immunology , Skin Neoplasms/genetics , Skin Neoplasms/pathology , T-Lymphocytes/pathology
10.
PLoS One ; 9(10): e110015, 2014.
Article En | MEDLINE | ID: mdl-25310588

The Ly49E NK receptor is a unique inhibitory receptor, presenting with a high degree of conservation among mouse strains and expression on both NK cells and intraepithelial-localised T cells. Amongst intraepithelial-localised T cells, the Ly49E receptor is abundantly expressed on CD8αα-expressing innate-like intestinal intraepithelial lymphocytes (iIELs), which contribute to front-line defense at the mucosal barrier. Inflammatory bowel diseases (IBDs), encompassing Crohn's disease and ulcerative colitis, have previously been suggested to have an autoreactive origin and to evolve from a dysbalance between regulatory and effector functions in the intestinal immune system. Here, we made use of Ly49E-deficient mice to characterize the role of Ly49E receptor expression on CD8αα-expressing iIELs in the development and progression of IBD. For this purpose we used the dextran sodium sulphate (DSS)- and trinitrobenzenesulfonic-acid (TNBS)-induced colitis models, and the TNFΔARE ileitis model. We show that Ly49E is expressed on a high proportion of CD8αα-positive iIELs, with higher expression in the colon as compared to the small intestine. However, Ly49E expression on small intestinal and colonic iIELs does not influence the development or progression of inflammatory bowel diseases.


CD8 Antigens/metabolism , Disease Progression , Epithelial Cells/pathology , Inflammatory Bowel Diseases/pathology , Intestines/pathology , Lymphocytes/pathology , NK Cell Lectin-Like Receptor Subfamily A/metabolism , Animals , Cell Count , Colitis/metabolism , Colitis/pathology , Colon/metabolism , Colon/pathology , Dextran Sulfate , Ileitis/metabolism , Ileitis/pathology , Inflammatory Bowel Diseases/chemically induced , Inflammatory Bowel Diseases/metabolism , Mice, Inbred C57BL , Mice, Knockout , Receptors, Natural Killer Cell/metabolism , Trinitrobenzenesulfonic Acid , Tumor Necrosis Factor-alpha/metabolism
11.
PLoS One ; 9(1): e87463, 2014.
Article En | MEDLINE | ID: mdl-24498110

Natural killer (NK) cells have different roles in the host response against Plasmodium-induced malaria depending on the stage of infection. Liver NK cells have a protective role during the initial hepatic stage of infection by production of the TH1-type cytokines IFN-γ and TNF-α. In the subsequent erythrocytic stage of infection, NK cells also induce protection through Th1-type cytokines but, in addition, may also promote development of cerebral malaria via CXCR3-induction on CD8(+) T cells resulting in migration of these cells to the brain. We have recently shown that the regulatory Ly49E NK receptor is expressed on liver NK cells in particular. The main objective of this study was therefore to examine the role of Ly49E expression in the immune response upon Plasmodium berghei ANKA infection, for which we compared wild type (WT) to Ly49E knockout (KO) mice. We show that the parasitemia was higher at the early stage, i.e. at days 6-7 of Plasmodium berghei ANKA infection in Ly49E KO mice, which correlated with lower induction of CD69, IFN-γ and TNF-α in DX5(-) liver NK cells at day 5 post-infection. At later stages, these differences faded. There was also no difference in the kinetics and the percentage of cerebral malaria development and in lymphocyte CXCR3 expression in WT versus Ly49E KO mice. Collectively, we show that the immune response against Plasmodium berghei ANKA infection is not drastically affected in Ly49E KO mice. Although NK cells play a crucial role in Plasmodium infection and Ly49E is highly expressed on liver NK cells, the Ly49E NK receptor only has a temporarily role in the immune control of this parasite.


Killer Cells, Natural/immunology , Liver/immunology , Liver/parasitology , Malaria/immunology , Malaria/parasitology , Plasmodium berghei/immunology , Animals , Antigens, CD/immunology , Antigens, Differentiation, T-Lymphocyte/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/parasitology , Female , Interferon-gamma/immunology , Lectins, C-Type/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Parasitemia/immunology , Parasitemia/parasitology , Receptors, CXCR3/immunology , Tumor Necrosis Factor-alpha/immunology
12.
J Leukoc Biol ; 93(5): 699-711, 2013 May.
Article En | MEDLINE | ID: mdl-23475576

The NKR Ly49E has several unique characteristics. Unlike most NKRs, Ly49E is highly expressed on fetal NK cells, whereas expression is decreased on bone marrow-derived NK cells in adult mice. To investigate a possible role for Ly49E in NK cell differentiation and function, we have generated an Ly49E KO mouse. Our results show that bone marrow and splenic NK cells are present in normal numbers in Ly49E KO mice, expressing an unaltered panel of NKRs and differentiation markers. Furthermore, cytokine production and cytotoxicity by these cells are unaffected. Surprisingly, WT DX5(-) liver NK cells express high Ly49E levels in fetal and adult mice. Ly49E(+)DX5(-) liver NK cells transferred into Rag-2(-/-)/gc(-/-) mice maintain high Ly49E expression in the liver and differentiate into DX5(+) NK cells in spleen and bone marrow. Ly49E expression is not crucial for liver NK cell differentiation during ontogeny, as the DX5(-)/DX5(+) ratio, the NKR repertoire, and the granzyme B and TRAIL levels are comparable in Ly49E KO versus WT mice, except for lower TRAIL expression on DX5(-) liver NK cells in 20-day-old mice. The TRAIL-, perforin-, and FasL-mediated cytolysis by liver NK cells is unaffected in Ly49E KO mice. Collectively, we show that in addition to high Ly49E expression on fetal NK cells versus low Ly49E expression on conventional NK cells in adult life, Ly49E remains highly expressed on DX5(-) liver NK cells. However, Ly49E expression does not have a crucial role in differentiation and/or function of these NK cells.


Killer Cells, Natural/immunology , Liver/immunology , NK Cell Lectin-Like Receptor Subfamily A/physiology , Adoptive Transfer , Animals , Cell Differentiation , Granzymes/analysis , Integrin alpha2/physiology , Interferon-gamma/biosynthesis , Killer Cells, Natural/cytology , Mice , Mice, Inbred C57BL , Mice, Knockout , TNF-Related Apoptosis-Inducing Ligand/analysis
13.
J Immunol ; 190(5): 1982-90, 2013 Mar 01.
Article En | MEDLINE | ID: mdl-23338239

The Ly49 NK receptor family in mice is composed of several members that recognize MHC class I (MHC-I) or MHC-I-related molecules. We and others have shown before that Ly49E is a unique member, with a different expression pattern on NK cells and being triggered by the non-MHC-I-related protein urokinase plasminogen activator. Among the entire Ly49 receptor family, Ly49E is the only Ly49 member expressed by epidermal-localized γδ T cells and their fetal thymic TCRγδ precursors, and it is the most abundantly expressed member on intestinal intraepithelial γδ T cell lymphocytes. In this study, we provide mechanistic insights into the regulation of Ly49e expression in γδ T cells. First, we demonstrate that TCR-mediated activation of intraepithelial γδ T cells significantly increases Ly49E expression. This results from de novo Ly49E expression and is highly selective, because no other Ly49 family members are induced. TCR-mediated Ly49E induction is a conserved feature of skin- and gut-residing intraepithelial-localized γδ T cell subsets, whereas it is not observed in spleen γδ T cells. By investigating Ly49e promoter activities and lymphotoxin (LT) αß dependency in resting versus TCR-activated intraepithelial γδ T cells, we reveal two separate regulatory pathways for Ly49E expression, as follows: a LTαß-dependent pathway leading to basal Ly49E expression in resting cells that is induced by Pro2-mediated Ly49e transcription, and a LTαß-independent pathway leading to elevated, Pro3-driven Ly49E expression in TCR-stimulated cells.


Epithelium/drug effects , Killer Cells, Natural/drug effects , NK Cell Lectin-Like Receptor Subfamily A/genetics , Receptors, Antigen, T-Cell, gamma-delta/immunology , T-Lymphocytes/drug effects , Transcription, Genetic/drug effects , Animals , Epidermal Cells , Epidermis/drug effects , Epidermis/immunology , Epithelium/immunology , Gene Expression Regulation , Intestines/cytology , Intestines/drug effects , Intestines/immunology , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Lymphotoxin alpha1, beta2 Heterotrimer/pharmacology , Mice , Mice, Transgenic , NK Cell Lectin-Like Receptor Subfamily A/immunology , Promoter Regions, Genetic , Receptors, Antigen, T-Cell, gamma-delta/genetics , Signal Transduction/drug effects , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Urokinase-Type Plasminogen Activator/pharmacology
14.
Blood ; 118(2): 339-47, 2011 Jul 14.
Article En | MEDLINE | ID: mdl-21613250

The engagement of inhibitory receptors specific for major histocompatibility complex class I (MHC-I) molecules educates natural killer (NK) cells, meaning the improvement of the response of activation receptors to subsequent stimulation. It is not known whether inhibitory MHC-I receptors educate only NK cells or whether they improve the responsiveness of all cell types, which express them. To address this issue, we analyzed the expression of inhibitory MHC-I receptors on intestinal intraepithelial lymphocytes (iIELs) and show that T-cell receptor (TCR)-αß CD8αα iIELs express multiple inhibitory receptors specific for MHC-I molecules, including CD94/NKG2A, Ly49A, and Ly49G2. However, the presence of MHC-I ligand for these receptors did not improve the response of iIELs to activation via the TCR. The absence of iIEL education by MHC-I receptors was not related to a lack of inhibitory function of these receptors in iIELs and a failure of these receptors to couple to the TCR. Thus, unlike NK cells, iIELs do not undergo an MHC-I-guided education process. These data suggest that education is an NK cell-specific function of inhibitory MHC-I receptors.


Histocompatibility Antigens Class I/immunology , Intestinal Mucosa/immunology , Killer Cells, Natural/immunology , Lymphocyte Activation/genetics , Receptors, Natural Killer Cell/physiology , T-Lymphocytes/immunology , Animals , CD8 Antigens/metabolism , Cells, Cultured , Histocompatibility Antigens Class I/genetics , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Killer Cells, Natural/metabolism , Killer Cells, Natural/physiology , Lymphocyte Activation/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, Natural Killer Cell/genetics , Receptors, Natural Killer Cell/immunology , Receptors, Natural Killer Cell/metabolism , Substrate Specificity , T-Lymphocytes/metabolism , T-Lymphocytes/physiology
15.
J Leukoc Biol ; 90(1): 61-8, 2011 Jul.
Article En | MEDLINE | ID: mdl-21486908

This study tested the hypothesis that Vγ3 TCR-bearing T cells are influenced by LCs. Vγ3 T cells and LCs are located in the epidermis of mice. Vγ3 T cells represent the main T cell population in the skin epithelium and play a crucial role in maintaining the skin integrity, whereas LCs are professional APCs. Although Vγ3 T cells and LCs form an interdigitating network in the epidermis, not much is known about their reciprocal influence and/or interdependence. We used two different LC-deficient mouse models, in which LCs are constitutively or inducibly depleted, to investigate the role of LCs in maturation, homeostasis, and function of Vγ3 T cells. We show that Vγ3 T cell numbers are unaltered by LC deficiency, and Vγ3 T cells isolated from LC-deficient mice are phenotypically and upon in vitro stimulation, functionally indistinguishable from Vγ3 T cells isolated from WT mice based on their cytotoxic potential and cytokine production. Additionally, in vivo skin-wounding experiments show no major difference in response of Vγ3 T cells to wounding in the absence or presence of LCs. These observations indicate that Vγ3 T cells develop and function independently of LCs.


Homeostasis/immunology , Langerhans Cells/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology , Skin/immunology , T-Lymphocytes/immunology , Animals , Cell Differentiation/immunology , Cell Separation , Epidermal Cells , Epidermis/immunology , Flow Cytometry , Immunohistochemistry , Langerhans Cells/cytology , Mice , Mice, Transgenic , Skin/cytology , T-Lymphocytes/cytology
16.
Immunol Cell Biol ; 89(7): 803-11, 2011 Oct.
Article En | MEDLINE | ID: mdl-21283110

Natural killer (NK) cells are part of the first line defense against tumors, parasites and virus-infected cells. Therefore, factors that control NK-cell numbers and their function are important. CD27 is constitutively expressed on NK cells and its expression correlates with sequential phases in NK-cell development, discriminating phenotypically and functionally different subsets within the NK-cell population. Although CD27 has been described to have an important regulatory role in effector and memory T and B lymphocytes, its role in NK-cell biology remains to be addressed. In this study, we used CD27(-/-) mice to investigate the role of CD27 in NK-cell development and function, both during the resting state and upon stimulation. The results show that NK-cell numbers are not impaired in CD27(-/-) mice. Moreover, CD27(-/-) NK cells reach full phenotypic maturity, evidenced by normal expression of CD49b, CD43 and CD11b. Expression of activating receptors is unaltered, whereas expression of several inhibitory receptors is increased. Cytotoxicity and interferon-γ production by NK cells from CD27(-/-) mice in the resting state are normal. However, upon in vivo anti-CD40- or poly-I:C-mediated activation, or in vitro interleukin-15 priming plus anti-NKp46 stimulation, the absence of CD27 results in decreased cytolytic activity and cytokine production by spleen and liver NK cells. In conclusion, this study demonstrates that CD27 is dispensable for the development of functional NK cells. However, upon stimulation of NK cells, CD27 displays an important role in their activation and functionality.


Cytotoxicity, Immunologic , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Lymphocyte Activation , Tumor Necrosis Factor Receptor Superfamily, Member 7/immunology , Animals , Antibodies, Monoclonal , Antigens, Ly/immunology , CD11b Antigen/biosynthesis , CD40 Antigens/immunology , Cell Differentiation , Cytokines/biosynthesis , Integrin alpha2/biosynthesis , Interferon-gamma/biosynthesis , Interleukin-15/pharmacology , Killer Cells, Natural/metabolism , Leukosialin/biosynthesis , Lymphocyte Count , Mice , Mice, Inbred C57BL , Mice, Knockout , Natural Cytotoxicity Triggering Receptor 1/immunology , Poly I-C/pharmacology , Receptors, Immunologic , Tumor Necrosis Factor Receptor Superfamily, Member 7/deficiency , Tumor Necrosis Factor Receptor Superfamily, Member 7/genetics
17.
Eur J Immunol ; 40(4): 1107-17, 2010 Apr.
Article En | MEDLINE | ID: mdl-20140903

NK cells are important mediators of the early defense. In mice, immature and mature NK (mNK) cells constitutively express the TNF receptor family member CD27; however, mNK cells eventually lose CD27 expression and become resting NK cells. Interaction of CD27 with its ligand, CD70, enhances proliferation and effector functions of NK cells. We used mice that constitutively express CD70 on B cells (CD70-Tg) to study the in vivo effects of continuous triggering of CD27 on NK cells. Continuous CD70-CD27 interaction resulted in strongly down-modulated CD27 expression on NK cells and gradually reduced absolute NK cell numbers. This reduction was most prominent in the mNK cell subpopulation and was at least partially due to increased apoptosis. Residual NK cells showed lower expression of activating Ly49 receptors and normal (liver) or decreased (spleen) IFN-gamma production. Nevertheless, NK cells from CD70-Tg mice displayed higher YAC-1 killing capacities. CD70-Tg NK cells exhibited up-regulated expression of NKG2D, which is in accordance with the increased YAC-1 lysis, as this is mainly NKG2D-dependent. Taken together, this study is the first to demonstrate that continuous CD70 triggering of CD27 on NK cells in vivo results in a severe reduction of NK cells. On a single cell basis, however, residual NK cells display enhanced cytotoxicity.


Killer Cells, Natural/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 7/immunology , Animals , Antigens, Ly/immunology , Apoptosis , B-Lymphocytes/immunology , CD27 Ligand/genetics , CD27 Ligand/immunology , Crosses, Genetic , Cytotoxicity, Immunologic , Female , Gene Expression Regulation/immunology , Immunophenotyping , Liver/cytology , Liver/immunology , Lymphocyte Count , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , NK Cell Lectin-Like Receptor Subfamily K/biosynthesis , NK Cell Lectin-Like Receptor Subfamily K/genetics , NK Cell Lectin-Like Receptor Subfamily K/immunology , Receptors, Immunologic/immunology , Spleen/cytology , Spleen/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 7/biosynthesis , Tumor Necrosis Factor Receptor Superfamily, Member 7/genetics
18.
Blood ; 112(13): 5046-51, 2008 Dec 15.
Article En | MEDLINE | ID: mdl-18784372

The Ly49 natural killer (NK)-cell receptor family comprises both activating and inhibitory members, which recognize major histocompatibility complex (MHC) class I or MHC class I-related molecules and are involved in target recognition. As previously shown, the Ly49E receptor fails to bind to a variety of soluble or cell-bound MHC class I molecules, indicating that its ligand is not an MHC class I molecule. Using BWZ.36 reporter cells, we demonstrate triggering of Ly49E by the completely distinct, non-MHC-related protein urokinase plasminogen activator (uPA). uPA is known to be secreted by a variety of cells, including epithelial and hematopoietic cells, and levels are up-regulated during tissue remodeling, infections, and tumorigenesis. Here we show that addition of uPA to Ly49E-positive adult and fetal NK cells inhibits interferon-gamma secretion and reduces their cytotoxic potential, respectively. These uPA-mediated effects are Ly49E-dependent, as they are reversed by addition of anti-Ly49E monoclonal antibody and by down-regulation of Ly49E expression using RNA interference. Our results suggest that uPA, besides its established role in fibrinolysis, tissue remodeling, and tumor metastasis, could be involved in NK cell-mediated immune surveillance and tumor escape.


Killer Cells, Natural/immunology , NK Cell Lectin-Like Receptor Subfamily A/physiology , Urokinase-Type Plasminogen Activator/physiology , Animals , Cytotoxicity, Immunologic , Immunologic Surveillance , Interferon-gamma/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Tumor Escape
...