Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Front Immunol ; 11: 960, 2020.
Article in English | MEDLINE | ID: mdl-32582150

ABSTRACT

NKG2D is a potent activating immunoreceptor expressed on nearly all cytotoxic lymphocytes promoting their cytotoxicity against self-cells expressing NKG2D ligands (NKG2DLs). NKG2DLs are MHC class I-like glycoproteins that usually are not expressed on "healthy" cells. Rather, their surface expression is induced by various forms of cellular stress, viral infection, or malignant transformation. Hence, cell surface NKG2DLs mark "dangerous" cells for elimination by cytotoxic lymphocytes and therefore can be considered as "kill-me" signals. In addition, NKG2DLs are up-regulated on activated leukocytes, which facilitates containment of immune responses. While the NKG2D receptor is conserved among mammals, NKG2DL genes have rapidly diversified during mammalian speciation, likely due to strong selective pressures exerted by species-specific pathogens. Consequently, NKG2DL genes are not conserved in man and mice, although their NKG2D-binding domains maintained structural homology. Human NKG2DLs comprise two members of the MIC (MICA/MICB) and six members of the ULBP family of glycoproteins (ULBP1-6) with MICA representing the best-studied human NKG2DLs by far. Many of these studies implicate a role of MICA in various malignant, infectious, or autoimmune diseases. However, conclusions from these studies often were limited in default of supporting in vivo experiments. Here, we report a MICA transgenic (MICAgen) mouse model that replicates central features of human MICA expression and function and, therefore, constitutes a novel tool to critically assess and extend conclusions from previous in vitro studies on MICA. Similarly to humans, MICA transcripts are broadly present in organs of MICAgen mice, while MICA glycoproteins are barely detectable. Upon activation, hematopoietic cells up-regulate and proteolytically shed surface MICA. Shed soluble MICA (sMICA) is also present in plasma of MICAgen mice but affects neither surface NKG2D expression of circulating NK cells nor their functional recognition of MICA-expressing tumor cells. Accordingly, MICAgen mice also show a delayed growth of MICA-expressing B16F10 tumors, not accompanied by an emergence of MICA-specific antibodies. Such immunotolerance for the xenoantigen MICA ideally suits MICAgen mice for anti-MICA-based immunotherapies. Altogether, MICAgen mice represent a valuable model to study regulation, function, disease relevance, and therapeutic targeting of MICA in vivo.


Subject(s)
Histocompatibility Antigens Class I/metabolism , Lymphocyte Activation , Lymphocytes/metabolism , Melanoma, Experimental/metabolism , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Animals , Cell Line, Tumor , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Immune Tolerance , Isoantibodies/immunology , Isoantibodies/metabolism , Ligands , Lymphocytes/immunology , Melanoma, Experimental/genetics , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice, Inbred C57BL , Mice, Transgenic , NK Cell Lectin-Like Receptor Subfamily K/deficiency , NK Cell Lectin-Like Receptor Subfamily K/genetics , Signal Transduction , Tumor Burden
2.
Am J Transplant ; 19(6): 1652-1662, 2019 06.
Article in English | MEDLINE | ID: mdl-30549427

ABSTRACT

Stem cell-based approaches have the potential to address the organ shortage in transplantation. Whereas both embryonic stem cells and induced pluripotent stem cells have been utilized as cellular sources for differentiation and lineage specification, their relative ability to be recognized by immune effector cells is unclear. We determined the expression of immune recognition molecules on hepatocyte-like cells (HLC) generated from murine embryonic stem cells and induced pluripotent stem cells, compared to adult hepatocytes, and we evaluated the impact on recognition by natural killer (NK) cells. We report that HLC lack MHC class I expression, and that embryonic stem cell-derived HLC have higher expression of the NK cell activating ligands Rae1, H60, and Mult1 than induced pluripotent stem cell-derived HLC and adult hepatocytes. Moreover, the lack of MHC class I renders embryonic stem cell-derived HLC, and induced pluripotent stem cell-derived HLC, susceptible to killing by syngeneic and allogeneic NK cells. Both embryonic stem cell-derived HLC, and induced pluripotent stem cell-derived HLC, are killed by NK cells at higher levels than adult hepatocytes. Finally, we demonstrate that the NK cell activation receptor, NKG2D, plays a key role in NK cell cytotoxicity of embryonic stem cell-derived HLC, but not induced pluripotent stem cell-derived HLC.


Subject(s)
Embryonic Stem Cells/immunology , Hepatocytes/immunology , Hepatocytes/transplantation , Induced Pluripotent Stem Cells/immunology , NK Cell Lectin-Like Receptor Subfamily K/immunology , Allografts , Animals , Cell Differentiation , Cell Transplantation/methods , Cytotoxicity, Immunologic , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Female , Gene Expression Profiling , Hepatocytes/cytology , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Isografts , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Ligands , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Mice, Transgenic , Models, Animal , NK Cell Lectin-Like Receptor Subfamily K/deficiency , NK Cell Lectin-Like Receptor Subfamily K/genetics
3.
Eur J Immunol ; 47(7): 1123-1135, 2017 07.
Article in English | MEDLINE | ID: mdl-28378389

ABSTRACT

Natural killer group 2 member D (NKG2D) is an activating receptor that is expressed on most cytotoxic cells of the immune system, including NK cells, γδ, and CD8+ T cells. It is still a matter of debate whether and how NKG2D mediates priming of CD8+ T cells in vivo, due to a lack of studies where NKG2D is eliminated exclusively in these cells. Here, we studied the impact of NKG2D on effector CD8+ T-cell formation. NKG2D deficiency that is restricted to murine CD8+ T cells did not impair antigen-specific T-cell expansion following mouse CMV and lymphocytic choriomeningitis virus infection, but reduced their capacity to produce cytokines. Upon infection, conventional dendritic cells induce NKG2D ligands, which drive cytokine production on CD8+ T cells via the Dap10 signaling pathway. T-cell development, homing, and proliferation were not affected by NKG2D deficiency and cytotoxicity was only impaired when strong T-cell receptor (TCR) stimuli were used. Transfer of antigen-specific CD8+ T cells demonstrated that NKG2D deficiency attenuated their capacity to reduce viral loads. The inability of NKG2D-deficient cells to produce cytokines could be overcome with injection of IL-15 superagonist during priming. In summary, our data show that NKG2D has a nonredundant role in priming of CD8+ T cells to produce antiviral cytokines.


Subject(s)
Arenaviridae Infections/immunology , CD8-Positive T-Lymphocytes/immunology , Cytokines/immunology , Herpesviridae Infections/immunology , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Animals , Cytokines/biosynthesis , Cytotoxicity, Immunologic , Dendritic Cells/immunology , Killer Cells, Natural/immunology , Lymphocyte Activation , Lymphocytic choriomeningitis virus/immunology , Mice , Muromegalovirus , NK Cell Lectin-Like Receptor Subfamily K/deficiency , NK Cell Lectin-Like Receptor Subfamily K/genetics , Receptors, Immunologic/immunology , Receptors, Immunologic/metabolism , Signal Transduction
4.
J Immunol ; 198(4): 1531-1542, 2017 02 15.
Article in English | MEDLINE | ID: mdl-28087665

ABSTRACT

NKG2D is a potent activating receptor that is expressed on cytotoxic immune cells such as CD8 T and NK cells, where it promotes cytotoxicity after binding stress ligands on infected or transformed cells. On NK cell precursors NKG2D modulates proliferation and maturation. Previously, we observed that NKG2D deficiency affects peripheral B cell numbers. In this study, we show that NKG2D regulates B1a cell development and function. We find that mice deficient for NKG2D have a strong reduction of B1a cell numbers. As a result, NKG2D-deficient mice produce significantly less Ag-specific IgM Abs upon immunization with T cell-independent Ags, and they are more susceptible to Gram-negative sepsis. Klrk1-/- B1a cells are also functionally impaired and they fail to provide protection against Francisella novicida upon adoptive transfer. Using mixed bone marrow chimeric mice, we show that the impact of NKG2D deficiency on B1a cell development is cell intrinsic. No changes in homeostatic turnover and homing of B cells were detectable, limiting the effects of NKG2D to modulation of the hematopoietic development of B1a cells. Using conditional ablation, we demonstrate that the effect of NKG2D on B1a cell development occurs at a developmental stage that precedes the common lymphoid progenitor. Our findings reveal an unexpected new role for NKG2D in the regulation of B1a cell development. The protective effects of this activating receptor therefore reach beyond that of cytotoxic cells, stimulating the immune system to fight bacterial infections by promoting development of innate-like B cells.


Subject(s)
B-Lymphocytes/physiology , Cell Differentiation , Gram-Negative Bacterial Infections/immunology , NK Cell Lectin-Like Receptor Subfamily K/immunology , Adoptive Transfer , Animals , B-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Francisella/immunology , Gram-Negative Bacterial Infections/prevention & control , Histocompatibility Antigens Class I/immunology , Immunoglobulin M/immunology , Killer Cells, Natural/immunology , Mice , NK Cell Lectin-Like Receptor Subfamily K/deficiency , Spleen/cytology , Spleen/immunology
5.
J Immunol ; 196(11): 4805-13, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27183590

ABSTRACT

It is now clear that recognition of nascent tumors by the immune system is critical for survival of the host against cancer. During cancer immunoediting, the ability of the tumor to escape immune recognition is important for tumor development. The immune system recognizes tumors via the presence of classical Ags and also by conserved innate mechanisms. One of these mechanisms is the NKG2D receptor that recognizes ligands whose expression is induced by cell transformation. In this study, we show that in NKG2D receptor-deficient mice, increasing numbers of B cells begin to express NKG2D ligands as they age. Their absence in wild-type mice suggests that these cells are normally cleared by NKG2D-expressing cells. NKG2D-deficient mice and mice constitutively expressing NKG2D ligands had increased incidence of B cell tumors, confirming that the inability to clear NKG2D ligand-expressing cells was important in tumor suppression and that NKG2D ligand expression is a marker of nascent tumors. Supporting a role for NKG2D ligand expression in controlling the progression of early-stage B cell lymphomas in humans, we found higher expression of a microRNA that inhibits human NKG2D ligand expression in tumor cells from high-grade compared with low-grade follicular lymphoma patients.


Subject(s)
Lymphoma, B-Cell/metabolism , Lymphoma, B-Cell/pathology , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Animals , Disease Progression , Humans , Ligands , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , MicroRNAs/genetics , NK Cell Lectin-Like Receptor Subfamily K/antagonists & inhibitors , NK Cell Lectin-Like Receptor Subfamily K/deficiency , NK Cell Lectin-Like Receptor Subfamily K/genetics
6.
Mol Cell Proteomics ; 15(3): 1007-16, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26637539

ABSTRACT

T-helper cells are differentiated from CD4+ T cells and are traditionally characterized by inflammatory or immunosuppressive responses in contrast to cytotoxic CD8+ T cells. Mass-spectrometry studies on T-helper cells are rare. In this study, we aimed to identify the proteomes of human Th1 and Th1/Th17 clones derived from intestinal biopsies of Crohn's disease patients and to identify differentially expressed proteins between the two phenotypes. Crohn's disease is an inflammatory bowel disease, with predominantly Th1- and Th17-mediated response where cells of the "mixed" phenotype Th1/Th17 have also been commonly found. High-resolution mass spectrometry was used for protein identification and quantitation. In total, we identified 7401 proteins from Th1 and Th1/Th17 clones, where 334 proteins were differentially expressed. Major differences were observed in cytotoxic proteins that were overrepresented in the Th1 clones. The findings were validated by flow cytometry analyses using staining with anti-granzyme B and anti-perforin and by a degranulation assay, confirming higher cytotoxic features of Th1 compared with Th1/Th17 clones. By testing a larger panel of T-helper cell clones from seven different Crohn's disease patients, we concluded that only a subgroup of the Th1 cell clones had cytotoxic features, and these expressed the surface markers T-cell-specific surface glycoprotein CD28 and were negative for expression of natural killer group 2 member D.


Subject(s)
CD28 Antigens/metabolism , Crohn Disease/metabolism , NK Cell Lectin-Like Receptor Subfamily K/deficiency , Proteomics/methods , Th1 Cells/metabolism , Th17 Cells/metabolism , CD4-Positive T-Lymphocytes/metabolism , Crohn Disease/pathology , Gene Expression Regulation , Gene Regulatory Networks , Humans , Intestinal Mucosa/metabolism , Mass Spectrometry , Proteome/immunology , Proteome/isolation & purification , T-Lymphocytes, Cytotoxic/metabolism
7.
Clin Immunol ; 149(3): 432-9, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24211717

ABSTRACT

The NKG2D activating receptor has been implicated in numerous autoimmune diseases. We tested the role of NKG2D in models of autoimmunity and inflammation using NKG2D knockout mice and antibody blockade experiments. The severity of experimental autoimmune encephalitis (EAE) was decreased in NKG2D-deficient mice when the disease was induced with a limiting antigen dose, but unchanged with an optimal antigen dose. Surprisingly, however, NKG2D deficiency had no detectable effect in several other models, including two models of type 1 diabetes, and a model of intestinal inflammation induced by poly(I:C). NKG2D antibody blockade in normal mice also failed to inhibit disease in the NOD diabetes model or the intestinal inflammation model. Published evidence using NKG2D knockout mice demonstrated a role for NKG2D in mouse models of atherosclerosis and liver inflammation, as well as in chronic obstructive pulmonary disease. Therefore, our results suggest that NKG2D plays selective roles in inflammatory diseases.


Subject(s)
Diabetes Mellitus, Type 1/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Inflammatory Bowel Diseases/immunology , Killer Cells, Natural/immunology , NK Cell Lectin-Like Receptor Subfamily K/immunology , Amino Acid Sequence , Animals , Antibodies, Neutralizing/pharmacology , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/pathology , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Gene Expression , Inflammatory Bowel Diseases/chemically induced , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/pathology , Killer Cells, Natural/pathology , Male , Mice , Mice, Inbred NOD , Mice, Knockout , Molecular Sequence Data , Myelin-Oligodendrocyte Glycoprotein , NK Cell Lectin-Like Receptor Subfamily K/deficiency , NK Cell Lectin-Like Receptor Subfamily K/genetics , Peptide Fragments , Poly I-C , Severity of Illness Index
8.
J Immunol ; 191(2): 961-70, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23772039

ABSTRACT

Infections with DNA tumor viruses, including members of the polyomavirus family, often result in tumor formation in immune-deficient hosts. The complex control involved in antiviral and antitumor immune responses during these infections can be studied in murine polyomavirus (PyV)-infected mice as a model. We found that NK cells efficiently kill cells derived from PyV-induced salivary gland tumors in vitro in an NKG2D (effector cell)-RAE-1 (target cell)-dependent manner; but in T cell-deficient mice, NK cells only delay but do not prevent the development of PyV-induced tumors. In this article, we show that the PyV-induced tumors have infiltrating functional NK cells. The freshly removed tumors, however, lack surface RAE-1 expression, and the tumor tissues produce soluble factors that downregulate RAE-1. These factors include the proinflammatory cytokines IL-1α, IL-1ß, IL-33, and TNF. Each of these cytokines downregulates RAE-1 expression and susceptibility to NK cell-mediated cytotoxicity. CD11b(+)F4/80(+) macrophages infiltrating the PyV-induced tumors produce high amounts of IL-1ß and TNF. Thus, our data suggest a new mechanism whereby inflammatory cytokines generated in the tumor environment lead to evasion of NK cell-mediated control of virus-induced tumors.


Subject(s)
Killer Cells, Natural/immunology , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Polyomavirus/immunology , Salivary Gland Neoplasms/immunology , Tumor Escape , Animals , CD11b Antigen/metabolism , Cell Line, Tumor , Cytotoxicity, Immunologic , Down-Regulation , Interleukin-1alpha/metabolism , Interleukin-1beta/metabolism , Interleukin-33 , Interleukins/metabolism , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, SCID , NK Cell Lectin-Like Receptor Subfamily K/deficiency , NK Cell Lectin-Like Receptor Subfamily K/genetics , Nuclear Matrix-Associated Proteins/metabolism , Nucleocytoplasmic Transport Proteins/metabolism , Polyomavirus Infections/immunology , Salivary Gland Neoplasms/virology , Tumor Microenvironment/immunology , Tumor Necrosis Factors/metabolism , Tumor Virus Infections/immunology
9.
Stem Cells ; 31(9): 1829-39, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23733329

ABSTRACT

Cell replacement therapy holds promise for a number of untreatable neurological or psychiatric diseases but the immunogenicity of cellular grafts remains controversial. Emerging stem cell and reprogramming technologies can be used to generate autologous grafts that minimize immunological concerns but autologous grafts may carry an underlying genetic vulnerability that reduces graft efficacy or survival. Healthy allogeneic grafts are an attractive and commercially scalable alternative if immunological variables can be controlled. Stem cells and immature neural progenitor cells (NPC) do not express major histocompatibility complex (MHC) antigens and can evade adaptive immune surveillance. Nevertheless, in an experimental murine model, allogeneic NPCs do not survive and differentiate as well as syngeneic grafts, even when traditional immunosuppressive treatments are used. In this study, we show that natural killer (NK) cells recognize the lack of self-MHC antigens on NPCs and pose a barrier to NPC transplantation. NK cells readily target both syngeneic and allogeneic NPC, and killing is modulated primarily by NK-inhibiting "self" class I MHC and NK-activating NKG2D-ligand expression. The absence of NKG2D signaling in NK cells significantly improves NPC-derived neuron survival and differentiation. These data illustrate the importance of innate immune mechanisms in graft outcome and the potential value of identifying and targeting NK cell-activating ligands that may be expressed by stem cell derived grafts.


Subject(s)
Immunity, Innate/immunology , Killer Cells, Natural/immunology , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Neural Stem Cells/immunology , Neural Stem Cells/transplantation , Stem Cell Transplantation , Animals , Antigens, Ly/metabolism , Cell Movement , Cytotoxicity, Immunologic , Histocompatibility Antigens Class I/metabolism , Ligands , Mice , Mice, Inbred C57BL , NK Cell Lectin-Like Receptor Subfamily K/deficiency , Natural Cytotoxicity Triggering Receptor 1/metabolism , Neural Stem Cells/cytology , Neurons/cytology , Neurons/metabolism , Transplantation, Homologous
10.
Blood ; 121(25): 5025-33, 2013 Jun 20.
Article in English | MEDLINE | ID: mdl-23649470

ABSTRACT

The immunoreceptors NKG2D and NKp46 are known for their capacity to activate natural killer (NK) cell cytotoxicity and secretory responses in the contexts of tumors and infections, yet their roles in NK cell education remain unclear. Here, we provide the first characterization of mice deficient for both NKG2D and NKp46 receptors to address the relevance of their concomitant absence during NK cell development and function. Our findings reveal that NK cells develop normally in double-mutant (DKO) mice. Mice lacking NKG2D but not NKp46 showed subtle differences in the percentages of NK cells expressing inhibitory Ly49 receptors and the adhesion molecule DNAM-1. A slightly increased percentage of terminally differentiated NK cells and functional response to in vitro stimuli was observed in some experiments. These alterations were modest and did not affect NK cell function in vivo in response to mouse cytomegalovirus infection. NKp46 deficiency alone, or in combination with NKG2D deficiency, had no effect on frequency or function of NK cells.


Subject(s)
Antigens, Ly/immunology , Killer Cells, Natural/immunology , Lymphocyte Subsets/immunology , NK Cell Lectin-Like Receptor Subfamily K/immunology , Natural Cytotoxicity Triggering Receptor 1/immunology , Animals , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , NK Cell Lectin-Like Receptor Subfamily K/deficiency , Natural Cytotoxicity Triggering Receptor 1/deficiency
11.
J Exp Med ; 209(13): 2409-22, 2012 Dec 17.
Article in English | MEDLINE | ID: mdl-23166357

ABSTRACT

The NKG2D stimulatory receptor expressed by natural killer cells and T cell subsets recognizes cell surface ligands that are induced on transformed and infected cells and facilitate immune rejection of tumor cells. We demonstrate that expression of retinoic acid early inducible gene 1 (RAE-1) family NKG2D ligands in cancer cell lines and proliferating normal cells is coupled directly to cell cycle regulation. Raet1 genes are directly transcriptionally activated by E2F family transcription factors, which play a central role in regulating cell cycle entry. Induction of RAE-1 occurred in primary cell cultures, embryonic brain cells in vivo, and cells in healing skin wounds and, accordingly, wound healing was delayed in mice lacking NKG2D. Transcriptional activation by E2Fs is likely coordinated with posttranscriptional regulation by other stress responses. These findings suggest that cellular proliferation, as occurs in cancer cells but also other pathological conditions, is a key signal tied to immune reactions mediated by NKG2D-bearing lymphocytes.


Subject(s)
Cell Cycle Checkpoints/physiology , E2F Transcription Factors/metabolism , Membrane Proteins/metabolism , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Animals , Cell Cycle Checkpoints/immunology , Cell Line, Tumor , Cell Proliferation , Cells, Cultured , Gene Expression , Ligands , Lymphocytes/immunology , Lymphocytes/metabolism , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , NK Cell Lectin-Like Receptor Subfamily K/deficiency , NK Cell Lectin-Like Receptor Subfamily K/genetics , Signal Transduction , Stress, Physiological , Transcription, Genetic , Wound Healing/genetics , Wound Healing/immunology , Wound Healing/physiology
12.
Int J Oncol ; 40(4): 1285-90, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22200673

ABSTRACT

Natural-killer group 2 (NKG2), a natural killer (NK) cell receptor, plays a critical role in regulating NK cytotoxicity. In this study, we investigated the expression levels of natural killer group 2 member A (NKG2A) and natural killer group 2 member D (NKG2D) in NK cells as well as the regulatory function of NKG2D in patients with colorectal cancer (CRC). Sixty-two CRC patients and 32 healthy controls were enrolled in this study. The expression levels of NKG2A and NKG2D mRNA in peripheral blood mononuclear cells (PBMCs) were investigated using real-time PCR. Flow cytometry was performed to assay the levels of NKG2A and NKG2D proteins in NK cells. The levels of NKG2D mRNA in PBMCs in the patients were significantly lower than those in the controls [mean ± SD, 1.11 ± 0.60 (CRC patients) vs. 1.65 ± 0.71 (healthy controls); p < 0.01], whereas the 2 groups showed no apparent difference in the levels of NKG2A mRNA (p>0.05). In addition, the patients showed significantly lower NKG2D levels in NK cells than the controls did (71.23% ± 8.31% [CRC patients] vs. 79.39% ± 5.58% [healthy controls]; p < 0.01). However, we observed no distinct difference in the NKG2A expression levels in NK cells between the 2 groups (p> 0.05). Notably, blockage of NKG2D signaling with anti-NKG2D antibodies ex vivo resulted in decreased cytotoxicity and CD107a degranulation. Our data revealed that the decrease in NKG2D expression levels may have been associated with suppression of NK cell activity in CRC patients.


Subject(s)
Colorectal Neoplasms/immunology , Killer Cells, Natural/immunology , NK Cell Lectin-Like Receptor Subfamily K/deficiency , Case-Control Studies , Cell Line, Tumor , Colorectal Neoplasms/metabolism , Female , HT29 Cells , Humans , Killer Cells, Natural/metabolism , Male , Middle Aged , NK Cell Lectin-Like Receptor Subfamily K/biosynthesis , NK Cell Lectin-Like Receptor Subfamily K/immunology
13.
J Immunol ; 186(6): 3304-8, 2011 Mar 15.
Article in English | MEDLINE | ID: mdl-21307295

ABSTRACT

It is uncertain whether NK cells modulate T cell memory differentiation. By using a genetic model that allows the selective depletion of NK cells, we show in this study that NK cells shape CD8(+) T cell fate by killing recently activated CD8(+) T cells in an NKG2D- and perforin-dependent manner. In the absence of NK cells, the differentiation of CD8(+) T cells is strongly biased toward a central memory T cell phenotype. Although, on a per-cell basis, memory CD8(+) T cells generated in the presence or the absence of NK cells have similar functional features and recall capabilities, NK cell deletion resulted in a significantly higher number of memory Ag-specific CD8(+) T cells, leading to more effective control of tumors carrying model Ags. The enhanced memory responses induced by the transient deletion of NK cells may provide a rational basis for the design of new vaccination strategies.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunologic Memory , Killer Cells, Natural/immunology , Lymphocyte Activation/immunology , Lymphocyte Depletion/methods , Up-Regulation/immunology , Animals , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/metabolism , Cells, Cultured , Coculture Techniques , Female , Humans , Immunologic Memory/genetics , Killer Cells, Natural/metabolism , Killer Cells, Natural/pathology , Lymphocyte Activation/genetics , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , NK Cell Lectin-Like Receptor Subfamily K/deficiency , NK Cell Lectin-Like Receptor Subfamily K/metabolism , NK Cell Lectin-Like Receptor Subfamily K/physiology , Perforin , Pore Forming Cytotoxic Proteins/biosynthesis , Pore Forming Cytotoxic Proteins/genetics , Pore Forming Cytotoxic Proteins/physiology , Up-Regulation/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...