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










Database
Language
Publication year range
1.
Cancer Res ; 78(13): 3589-3603, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29691253

ABSTRACT

Natural killer (NK) cells express receptors specific for MHC class I (MHC-I) molecules involved in "missing-self" recognition of cancer and virus-infected cells. Here we elucidate the role of MHC-I-independent NKR-P1B:Clr-b interactions in the detection of oncogenic transformation by NK cells. Ras oncogene overexpression was found to promote a real-time loss of Clr-b on mouse fibroblasts and leukemia cells, mediated in part via the Raf/MEK/ERK and PI3K pathways. Ras-driven Clr-b downregulation occurred at the level of the Clrb (Clec2d) promoter, nascent Clr-b transcripts, and cell surface Clr-b protein, in turn promoting missing-self recognition via the NKR-P1B inhibitory receptor. Both Ras- and c-Myc-mediated Clr-b loss selectively augmented cytotoxicity of oncogene-transformed leukemia cells by NKR-P1B+ NK cells in vitro and enhanced rejection by WT mice in vivo Interestingly, genetic ablation of either one (Clr-b+/-) or two Clr-b alleles (Clr-b-/-) enhanced survival of Eµ-cMyc transgenic mice in a primary lymphoma model despite preferential rejection of Clr-b-/- hematopoietic cells previously observed following adoptive transfer into naïve wild-type mice in vivo Collectively, these findings suggest that the inhibitory NKR-P1B:Clr-b axis plays a beneficial role in innate detection of oncogenic transformation via NK-cell-mediated cancer immune surveillance, in addition to a pathologic role in the immune escape of primary lymphoma cells in Eµ-cMyc mice in vivo These results provide a model for the human NKR-P1A:LLT1 system in cancer immunosurveillance in patients with lymphoma and suggest it may represent a target for immune checkpoint therapy.Significance: A mouse model shows that an MHC-independent NK-cell recognition axis enables the detection of leukemia cells, with implications for a novel immune checkpoint therapy target in human lymphoma. Cancer Res; 78(13); 3589-603. ©2018 AACR.


Subject(s)
Immunologic Surveillance , Killer Cells, Natural/immunology , Lectins, C-Type/metabolism , Lymphoma/immunology , NK Cell Lectin-Like Receptor Subfamily B/metabolism , Receptors, Cell Surface/metabolism , Receptors, Immunologic/metabolism , Signal Transduction/immunology , Animals , Cell Line, Tumor , Cell Transformation, Neoplastic/immunology , Disease Models, Animal , Down-Regulation , HEK293 Cells , Humans , Lectins, C-Type/immunology , Lymphoma/genetics , Lymphoma/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , NIH 3T3 Cells , NK Cell Lectin-Like Receptor Subfamily B/immunology , Receptors, Cell Surface/immunology , Receptors, Immunologic/immunology
2.
Sci Rep ; 7(1): 3501, 2017 06 14.
Article in English | MEDLINE | ID: mdl-28615725

ABSTRACT

For many years, human peripheral blood natural killer (NK) cells have been divided into functionally distinct CD3- CD56bright CD16- and CD3- CD56dim CD16+ subsets. Recently, several groups of innate lymphoid cells (ILC), distinct from NK cells in development and function, have been defined in mouse. A signature of genes present in mouse ILC except NK cells, defined by Immunological Genome Project studies, is significantly over-represented in human CD56bright cells, by gene set enrichment analysis. Conversely, the signature genes of mouse NK cells are enriched in human CD56dim cells. Correlations are based upon large differences in expression of a few key genes. CD56bright cells show preferential expression of ILC-associated IL7R (CD127), TNFSF10 (TRAIL), KIT (CD117), IL2RA (CD25), CD27, CXCR3, DPP4 (CD26), GPR183, and MHC class II transcripts and proteins. This could indicate an ontological relationship between human CD56bright cells and mouse CD127+ ILC, or conserved networks of transcriptional regulation. In line with the latter hypothesis, among transcription factors known to impact ILC or NK cell development, GATA3, TCF7 (TCF-1), AHR, SOX4, RUNX2, and ZEB1 transcript levels are higher in CD56bright cells, while IKZF3 (AIOLOS), TBX21 (T-bet), NFIL3 (E4BP4), ZEB2, PRDM1 (BLIMP1), and RORA mRNA levels are higher in CD56dim cells.


Subject(s)
CD3 Complex/genetics , CD56 Antigen/genetics , Interleukin-7 Receptor alpha Subunit/genetics , Killer Cells, Natural/metabolism , Transcriptome , Animals , CD3 Complex/blood , CD3 Complex/immunology , CD56 Antigen/blood , CD56 Antigen/immunology , Gene Expression Profiling , Humans , Interleukin-7 Receptor alpha Subunit/blood , Interleukin-7 Receptor alpha Subunit/immunology , Killer Cells, Natural/immunology , Mice , Species Specificity
3.
Cell ; 169(1): 58-71.e14, 2017 03 23.
Article in English | MEDLINE | ID: mdl-28340350

ABSTRACT

Natural killer (NK) cells play a key role in innate immunity by detecting alterations in self and non-self ligands via paired NK cell receptors (NKRs). Despite identification of numerous NKR-ligand interactions, physiological ligands for the prototypical NK1.1 orphan receptor remain elusive. Here, we identify a viral ligand for the inhibitory and activating NKR-P1 (NK1.1) receptors. This murine cytomegalovirus (MCMV)-encoded protein, m12, restrains NK cell effector function by directly engaging the inhibitory NKR-P1B receptor. However, m12 also interacts with the activating NKR-P1A/C receptors to counterbalance m12 decoy function. Structural analyses reveal that m12 sequesters a large NKR-P1 surface area via a "polar claw" mechanism. Polymorphisms in, and ablation of, the viral m12 protein and host NKR-P1B/C alleles impact NK cell responses in vivo. Thus, we identify the long-sought foreign ligand for this key immunoregulatory NKR family and reveal how it controls the evolutionary balance of immune recognition during host-pathogen interplay.


Subject(s)
Killer Cells, Natural/immunology , Muromegalovirus/immunology , Receptors, Natural Killer Cell/immunology , Viral Proteins/metabolism , Animals , Antigens, Ly/metabolism , Cell Line , HEK293 Cells , Host-Pathogen Interactions , Humans , Immune Evasion , Immunity, Innate , Mice , NIH 3T3 Cells , NK Cell Lectin-Like Receptor Subfamily B/metabolism , Rats
4.
J Innate Immun ; 9(4): 343-358, 2017.
Article in English | MEDLINE | ID: mdl-28288457

ABSTRACT

Natural killer (NK) cells are innate lymphocytes that aid in self-nonself discrimination by recognizing cells undergoing pathological alterations. The NKR-P1B inhibitory receptor recognizes Clr-b, a self-encoded marker of cell health downregulated during viral infection. Here, we show that Clr-b loss during mouse cytomegalovirus (MCMV) infection is predicated by a loss of Clr-b (Clec2d) promoter activity and nascent transcripts, driven in part by MCMV ie3 (M122) activity. In contrast, uninfected bystander cells near MCMV-infected fibroblasts reciprocally upregulate Clr-b expression due to paracrine type-I interferon (IFN) signaling. Exposure of fibroblasts to type-I IFN augments Clec2d promoter activity and nascent Clr-b transcripts, dependent upon a cluster of IRF3/7/9 motifs located ∼200 bp upstream of the transcriptional start site. Cells deficient in type-I IFN signaling components revealed IRF9 and STAT1 as key transcription factors involved in Clr-b upregulation. In chromatin immunoprecipitation experiments, the Clec2d IRF cluster recruited STAT2 upon IFN-α exposure, confirming the involvement of ISGF3 (IRF9/STAT1/STAT2) in positively regulating the Clec2d promoter. These findings demonstrate that Clr-b is an IFN-stimulated gene on healthy bystander cells, in addition to a missing-self marker on MCMV-infected cells, and thereby enhances the dynamic range of innate self-nonself discrimination by NK cells.


Subject(s)
Fibroblasts/physiology , Herpesviridae Infections/immunology , Killer Cells, Natural/immunology , Lectins, C-Type/metabolism , Muromegalovirus/immunology , NK Cell Lectin-Like Receptor Subfamily B/metabolism , Receptors, Immunologic/metabolism , STAT1 Transcription Factor/genetics , Animals , Cytotoxicity, Immunologic , Immunity, Innate , Interferon Type I/metabolism , Interferon-Stimulated Gene Factor 3, gamma Subunit/genetics , Lectins, C-Type/genetics , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , NIH 3T3 Cells , Paracrine Communication , Promoter Regions, Genetic/genetics , Receptors, Immunologic/genetics , STAT2 Transcription Factor/genetics
5.
J Innate Immun ; 7(6): 584-600, 2015.
Article in English | MEDLINE | ID: mdl-26044139

ABSTRACT

Viruses are known to induce pathological cellular states that render infected cells susceptible or resistant to immune recognition. Here, we characterize an MHC-I-independent natural killer (NK) cell recognition mechanism that involves modulation of inhibitory NKR-P1B:Clr-b receptor-ligand interactions in response to mouse cytomegalovirus (MCMV) infection. We demonstrate that mouse Clr-b expression on healthy cells is rapidly lost at the cell surface and transcript levels in a time- and dose-dependent manner upon MCMV infection. In addition, cross-species infections using rat cytomegalovirus (RCMV) infection of mouse fibroblasts and MCMV infection of rat fibroblasts suggest that this response is conserved during host-pathogen interactions. Active viral infection appears to be necessary for Clr-b loss, as cellular stimulation using UV-inactivated whole virus or agonists of many innate pattern recognition receptors failed to elicit efficient Clr-b downregulation. Notably, Clr-b loss could be partially blocked by titrated cycloheximide treatment, suggesting that early viral or nascent host proteins are required for Clr-b downregulation. Interestingly, reporter cell assays suggest that MCMV may encode a novel Clr-b-independent immunoevasin that functionally engages the NKR-P1B receptor. Together, these data suggest that Clr-b modulation is a conserved innate host cell response to virus infection that is subverted by multiple CMV immune evasion strategies.


Subject(s)
Calcitonin Receptor-Like Protein/immunology , Gene Expression Regulation/immunology , Herpesviridae Infections/immunology , Immune Evasion , Muromegalovirus/immunology , NK Cell Lectin-Like Receptor Subfamily B/immunology , Animals , Calcitonin Receptor-Like Protein/genetics , Herpesviridae Infections/genetics , Herpesviridae Infections/pathology , Mice , NIH 3T3 Cells , NK Cell Lectin-Like Receptor Subfamily B/genetics , Rats
6.
J Immunol ; 194(6): 2909-18, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25681346

ABSTRACT

MHC-I-specific receptors play a vital role in NK cell-mediated "missing-self" recognition, which contributes to NK cell activation. In contrast, MHC-independent NK recognition mechanisms are less well characterized. In this study, we investigated the role of NKR-P1B:Clr-b (Klrb1:Clec2d) interactions in determining the outcome of murine hematopoietic cell transplantation in vivo. Using a competitive transplant assay, we show that Clr-b(-/-) bone marrow (BM) cells were selectively rejected by wild-type B6 recipients, to a similar extent as H-2D(b-/-) MHC-I-deficient BM cells. Selective rejection of Clr-b(-/-) BM cells was mitigated by NK depletion of recipient mice. Competitive rejection of Clr-b(-/-) BM cells also occurred in allogeneic transplant recipients, where it was reversed by selective depletion of NKR-P1B(hi) NK cells, leaving the remaining NKR-P1B(lo) NK subset and MHC-I-dependent missing-self recognition intact. Moreover, competitive rejection of Clr-b(-/-) hematopoietic cells was abrogated in Nkrp1b-deficient recipients, which lack the receptor for Clr-b. Of interest, similar to MHC-I-deficient NK cells, Clr-b(-/-) NK cells were hyporesponsive to both NK1.1 (NKR-P1C)-stimulated and IL-12/18 cytokine-primed IFN-γ production. These findings support a unique and nonredundant role for NKR-P1B:Clr-b interactions in missing-self recognition of normal hematopoietic cells and suggest that optimal BM transplant success relies on MHC-independent tolerance mechanisms. These findings provide a model for human NKR-P1A:LLT1 (KLRB1:CLEC2D) interactions in human hematopoietic cell transplants.


Subject(s)
Bone Marrow Transplantation/methods , Killer Cells, Natural/immunology , Lectins, C-Type/immunology , Membrane Proteins/immunology , NK Cell Lectin-Like Receptor Subfamily B/immunology , Animals , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Cell Line, Tumor , Flow Cytometry , Gene Expression/immunology , Graft Rejection/genetics , Graft Rejection/immunology , Hematopoietic Stem Cell Transplantation/methods , Histocompatibility Antigen H-2D/genetics , Histocompatibility Antigen H-2D/immunology , Histocompatibility Antigen H-2D/metabolism , Humans , Interferon-gamma/immunology , Interferon-gamma/metabolism , Killer Cells, Natural/metabolism , Lectins, C-Type/deficiency , Lectins, C-Type/genetics , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice, Inbred C57BL , Mice, Knockout , Models, Animal , NK Cell Lectin-Like Receptor Subfamily B/deficiency , NK Cell Lectin-Like Receptor Subfamily B/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transplantation, Homologous
7.
Front Immunol ; 5: 214, 2014.
Article in English | MEDLINE | ID: mdl-24917862

ABSTRACT

The NKR-P1 receptors were identified as prototypical natural killer (NK) cell surface antigens and later shown to be conserved from rodents to humans on NK cells and subsets of T cells. C-type lectin-like in nature, they were originally shown to be capable of activating NK cell function and to recognize ligands on tumor cells. However, certain family members have subsequently been shown to be capable of inhibiting NK cell activity, and to recognize proteins encoded by a family of genetically linked C-type lectin-related ligands. Some of these ligands are expressed by normal, healthy cells, and modulated during transformation, infection, and cellular stress, while other ligands are upregulated during the immune response and during pathological circumstances. Here, we discuss historical and recent developments in NKR-P1 biology that demonstrate this NK receptor-ligand system to be far more complex and diverse than originally anticipated.

SELECTION OF CITATIONS
SEARCH DETAIL
...