Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 80
Filter
1.
Immunohorizons ; 7(11): 760-770, 2023 Nov 01.
Article in English | MEDLINE | ID: mdl-37971282

ABSTRACT

NK cells are a key focus in immuno-oncology, based on their ability to eliminate malignant cells without prior sensitization. Dogs are valuable models for translational immunotherapy studies, especially for NK cells, where critical species differences exist between mice and humans. Given that the mechanism for recognition of "self" by canine NK cells is currently unknown, we sought to evaluate expression of Ly49 in canine NK cells using in silico and high-throughput techniques. We interrogated the identified polymorphism/mutation in canine Ly49 and assessed the potential impact on structure using computational modeling of three-dimensional protein structure and protein-protein docking of canine Ly49 with MHC class I (MHC-I). Bulk and single-cell RNA-sequencing analysis was performed to detect gene expression of Ly49/KLRA1 in resting and activated NK cells. Tertiary protein structure demonstrated significant structural similarity to the known murine system. Molecular docking of canine Ly49 with MHC-I was favorable, converging at a single low-energy conformation. RNA sequencing revealed expression of Ly49/KLRA1 in both resting and activated NK cells and demonstrated almost exclusive expression of the gene in the NK cluster at the single-cell level. Despite prior reports of a mutated, nonfunctional canine Ly49, our data support that the protein product is predicted to bind to MHC-I in a comparable conformation to the murine system and is expressed in canine NK cells with upregulation following activation. Taken together, these data suggest that Ly49 is capable of recognizing MHC-I and therefore regulating NK cell function in dogs.


Subject(s)
Histocompatibility Antigens Class I , Neoplasms , Animals , Mice , Dogs , Humans , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/metabolism , NK Cell Lectin-Like Receptor Subfamily A/genetics , NK Cell Lectin-Like Receptor Subfamily A/metabolism , Molecular Docking Simulation , Killer Cells, Natural , Neoplasms/genetics
2.
J Immunol ; 209(4): 751-759, 2022 08 15.
Article in English | MEDLINE | ID: mdl-35896339

ABSTRACT

MHC class I (MHC I) expression in the host influences NK cells in a process termed education. The result of this education is reflected in the responsiveness of NK cells at the level of individual cells as well as in the repertoire of inhibitory MHC I-specific receptors at the NK cell system level. The presence of MHC I molecules in the host environment gives rise to a skewed receptor repertoire in spleen NK cells where subsets expressing few (one or two) inhibitory receptors are expanded whereas subsets with many (three or more) receptors are contracted. It is not known whether this MHC I-dependent skewing is imposed during development or after maturation of NK cells. In this study, we tested the hypothesis that the NK cell receptor repertoire is shaped already early during NK cell development in the bone marrow. We used mice with a repertoire imposed by a single MHC I allele, as well as a C57BL/6 mutant strain with exaggerated repertoire skewing, to investigate Ly49 receptor repertoires at different stages of NK cell differentiation. Our results show that NK cell inhibitory receptor repertoire skewing can indeed be observed in the bone marrow, even during the earliest developmental steps where Ly49 receptors are expressed. This may partly be accounted for by selective proliferation of certain NK cell subsets, but other mechanisms must also be involved. We propose a model for how repertoire skewing is established during a developmental phase in the bone marrow, based on sequential receptor expression as well as selective proliferation.


Subject(s)
Bone Marrow , NK Cell Lectin-Like Receptor Subfamily A , Animals , Antigens, Ly/metabolism , Bone Marrow/metabolism , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/metabolism , Killer Cells, Natural , Lectins, C-Type/metabolism , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , NK Cell Lectin-Like Receptor Subfamily A/genetics , NK Cell Lectin-Like Receptor Subfamily A/metabolism , Receptors, NK Cell Lectin-Like/metabolism , Receptors, Natural Killer Cell/metabolism
3.
Mol Immunol ; 135: 217-225, 2021 07.
Article in English | MEDLINE | ID: mdl-33932686

ABSTRACT

Ly49Q is an ITIM-bearing MHC class I receptor that is highly expressed in plasmacytoid dendritic cells (pDCs). Ly49Q is required for the TLR9-mediated IFN-I production in pDCs, although the mechanism is not fully understood. We here demonstrate that Ly49Q protects pDCs from pyroptotic cell death induced by CpG oligodeoxynucleotides (CpG). In the Ly49Q-deficient (Klra17-/-) mouse spleen, the number of ssDNA-positive pDCs increased significantly after CpG treatment, strongly suggesting that Klra17-/- pDCs were susceptible to CpG-induced cell death. In Klra17-/- bone-marrow-derived dendritic cells (BMDCs), CpG-induced cell death was accompanied by increased cathepsin B leakage from the vesicular compartments into the cytoplasm. Concurrently, IL-1ß secretion increased in the CpG-treated Klra17-/- BMDCs, strongly suggesting that the CpG-induced cell death in these cells is pyroptotic in nature. Consistent with these observations, inhibiting cathepsin B or caspase 1 in CpG-stimulated Klra17-/- BMDCs reversed the increase in cell death. Pyroptotic cell death and IL-1ß secretion were also observed in BMDCs derived from transgenic mice expressing an ITIM-less Ly49Q (Ly49Q-YF Tg). CpG also increased the IL-1ß production and cell death in B2m-/- BMDCs. These results suggest that Ly49Q and MHC class I play important roles for protecting pyroptosis-like cell death of DCs by influencing lysosome state.


Subject(s)
Dendritic Cells/immunology , Lysosomes/metabolism , NK Cell Lectin-Like Receptor Subfamily A/metabolism , Oligodeoxyribonucleotides/pharmacology , Pyroptosis/immunology , Animals , Caspase 1/metabolism , Cathepsin B/metabolism , Cell Membrane/physiology , Cells, Cultured , CpG Islands/genetics , Interleukin-1beta/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , NK Cell Lectin-Like Receptor Subfamily A/genetics , Oligodeoxyribonucleotides/genetics
4.
J Immunol ; 206(6): 1215-1227, 2021 03 15.
Article in English | MEDLINE | ID: mdl-33495236

ABSTRACT

Previous studies of NK cell inhibitory Ly-49 genes showed their expression is stochastic. However, relatively few studies have examined the mechanisms governing acquisition of inhibitory receptors in conjunction with activating Ly-49 receptors and NK cell development. We hypothesized that the surface expression of activating Ly-49 receptors is nonrandom and is influenced by inhibitory Ly-49 receptors. We analyzed NK cell "clusters" defined by combinatorial expression of activating (Ly-49H and Ly-49D) and inhibitory (Ly-49I and Ly-49G2) receptors in C57BL/6 mice. Using the product rule to evaluate the interdependencies of the Ly-49 receptors, we found evidence for a tightly regulated expression at the immature NK cell stage, with the highest interdependencies between clusters that express at least one activating receptor. Further analysis demonstrated that certain NK clusters predominated at the immature (CD27+CD11b-), transitional (CD27+CD11b+), and mature (CD27-CD11b-) NK cell stages. Using parallel in vitro culture and in vivo transplantation of sorted NK clusters, we discovered nonrandom expression of Ly-49 receptors, suggesting that prescribed pathways of NK cluster differentiation exist. Our data infer that surface expression of Ly-49I is an important step in NK cell maturation. Ki-67 expression and cell counts confirmed that immature NK cells proliferate more than mature NK cells. We found that MHC class I is particularly important for regulation of Ly-49D and Ly-49G2, even though no known MHC class I ligand for these receptors is present in B6 mice. Our data indicate that surface expression of both activating and inhibitory Ly-49 receptors on NK cell clusters occurs in a nonrandom process correlated to their maturation stage.


Subject(s)
Cell Differentiation/genetics , Killer Cells, Natural/immunology , NK Cell Lectin-Like Receptor Subfamily A/genetics , Adoptive Transfer , Animals , Cell Differentiation/immunology , Cell Proliferation/genetics , Female , Gene Expression Regulation/immunology , Killer Cells, Natural/metabolism , Killer Cells, Natural/transplantation , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NK Cell Lectin-Like Receptor Subfamily A/metabolism
5.
Sci Rep ; 10(1): 13153, 2020 08 04.
Article in English | MEDLINE | ID: mdl-32753654

ABSTRACT

Domestic animal populations are often characterised by high rates of inbreeding and low effective population sizes due to selective breeding practices. These practices can result in otherwise rare recessive deleterious alleles drifting to high frequencies, resulting in reduced fertility rates. This study aimed to identify potential recessive lethal haplotypes in the Thoroughbred horse breed, a closed population that has been selectively bred for racing performance. In this study, we identified a haplotype in the LY49B gene that shows strong evidence of being homozygous lethal, despite having high frequencies of heterozygotes in Thoroughbreds and other domestic horse breeds. Variant analysis of whole-genome sequence data identified two SNPs in the 3'UTR of the LY49B gene that may result in loss of function. Analysis of transcriptomic data from equine embryonic tissue revealed that LY49B is expressed in the trophoblast during placentation stage of development. These findings suggest that LY49B may have an essential, but as yet unknown function in the implantation stage of equine development. Further investigation of this region may allow for the development of a genetic test to improve fertility rates in horse populations. Identification of other lethal variants could assist in improving natural levels of fertility in horse populations.


Subject(s)
3' Untranslated Regions , Breeding , Haplotypes , Horses/genetics , NK Cell Lectin-Like Receptor Subfamily A/genetics , Polymorphism, Single Nucleotide , Animals , Female , Fertility/genetics , Genome-Wide Association Study , Male
6.
J Immunol ; 204(9): 2455-2463, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32213565

ABSTRACT

Cattle possess the most diverse repertoire of NK cell receptor genes among all mammals studied to date. Killer cell receptor genes encoded within the NK complex and killer cell Ig-like receptor genes encoded within the leukocyte receptor complex have both been expanded and diversified. Our previous studies identified two divergent and polymorphic KLRA alleles within the NK complex in the Holstein-Friesian breed of dairy cattle. By examining a much larger cohort and other ruminant species, we demonstrate the emergence and fixation of two KLRA allele lineages (KLRA*01 and -*02) at a single locus during ruminant speciation. Subsequent recombination events between these allele lineages have increased the frequency of KLRA*02 extracellular domains. KLRA*01 and KLRA*02 transcription levels contrasted in response to cytokine stimulation, whereas homozygous animals consistently transcribed higher levels of KLRA, regardless of the allele lineage. KLRA*02 mRNA levels were also generally higher than KLRA*01 Collectively, these data point toward alternative functional roles governed by KLRA genotype and allele lineage. On a background of high genetic diversity of NK cell receptor genes, this KLRA allele fixation points to fundamental and potentially differential function roles.


Subject(s)
NK Cell Lectin-Like Receptor Subfamily A/genetics , Ruminants/genetics , Transcription, Genetic/genetics , Alleles , Animals , Cattle , Gene Frequency/genetics , Gene Frequency/immunology , Genotype , Killer Cells, Natural/immunology , NK Cell Lectin-Like Receptor Subfamily A/immunology , RNA, Messenger/genetics , RNA, Messenger/immunology , Ruminants/immunology , Transcription, Genetic/immunology
7.
Front Immunol ; 11: 602783, 2020.
Article in English | MEDLINE | ID: mdl-33488602

ABSTRACT

The role of Ly49+CD8 T-cells in the immune system is not clear. Previously, several papers suggested Ly49+CD8 T-cells as immunosuppressors, while multiple studies also suggested their role as potent participants of the immune response. The mechanism of Ly49 expression on CD8 T-cells is also not clear. We investigated phenotype, functions, and regulation of Ly49 expression on murine CD8 T-cells in both normal state and during LCMV infection. CD8 T-cells express different Ly49 receptors compared with NK-cells. In intact mice, Ly49+CD8 T-cells have a phenotype similar to resting central memory CD8 T-cells and do not show impaired proliferation and cytokine production. Conventional CD8 T-cells upregulate Ly49 receptors during TCR-induced stimulation, and IL-2, as well as IL-15, affect it. At the same time, Ly49+CD8 T-cells change the Ly49 expression profile dramatically upon re-stimulation downregulating inhibitory and upregulating activating Ly49 receptors. We observed the expression of Ly49 receptors on the virus-specific CD8 T-cells during LCMV infection, especially marked in the early stages, and participation of Ly49+CD8 T-cells in the anti-viral response. Thus, CD8 T-cells acquire Ly49 receptors during the T-cell activation and show dynamic regulation of Ly49 receptors during stimulation.


Subject(s)
CD8-Positive T-Lymphocytes/virology , Lymphocyte Activation , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/pathogenicity , NK Cell Lectin-Like Receptor Subfamily A/metabolism , Adoptive Transfer , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/transplantation , Cell Proliferation , Chlorocebus aethiops , Cytokines/metabolism , Disease Models, Animal , Host-Pathogen Interactions , Immunologic Memory , Lymphocytic Choriomeningitis/genetics , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/metabolism , Lymphocytic choriomeningitis virus/immunology , Mice, Inbred C57BL , Mice, Knockout , NK Cell Lectin-Like Receptor Subfamily A/genetics , Phenotype , Signal Transduction , Vero Cells
8.
Nat Commun ; 10(1): 5010, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31676749

ABSTRACT

Mice lacking MHC class-I (MHC-I) display severe defects in natural killer (NK) cell functional maturation, a process designated as "education". Whether self-MHC-I specific Ly49 family receptors and NKG2A, which are closely linked within the NK gene complex (NKC) locus, are essential for NK cell education is still unclear. Here we show, using CRISPR/Cas9-mediated gene deletion, that mice lacking all members of the Ly49 family exhibit a moderate defect in NK cell activity, while mice lacking only two inhibitory Ly49 members, Ly49C and Ly49I, have comparable phenotypes. Furthermore, the deficiency of NKG2A, which recognizes non-classical MHC-Ib molecules, mildly impairs NK cell function. Notably, the combined deletion of NKG2A and the Ly49 family severely compromises the ability of NK cells to mediate "missing-self" and "induced-self" recognition. Therefore, our data provide genetic evidence supporting that NKG2A and the inhibitory members of Ly49 family receptors synergize to regulate NK cell education.


Subject(s)
Killer Cells, Natural/immunology , Multigene Family , NK Cell Lectin-Like Receptor Subfamily A/immunology , NK Cell Lectin-Like Receptor Subfamily C/immunology , Animals , CRISPR-Cas Systems , Gene Editing/methods , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , Killer Cells, Natural/metabolism , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , NK Cell Lectin-Like Receptor Subfamily A/genetics , NK Cell Lectin-Like Receptor Subfamily A/metabolism , NK Cell Lectin-Like Receptor Subfamily C/genetics , NK Cell Lectin-Like Receptor Subfamily C/metabolism
9.
J Immunol ; 203(7): 1981-1988, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31444264

ABSTRACT

NK cells can recognize target cells such as virus-infected and tumor cells through integration of activation and inhibitory receptors. Recognition by NK cells can lead to direct lysis of the target cell and production of the signature cytokine IFN-γ. However, it is unclear whether stimulation through activation receptors alone is sufficient for IFN-γ production. In this study, we show that NK activation receptor engagement requires additional signals for optimal IFN-γ production, which could be provided by IFN-ß or IL-12. Stimulation of murine NK cells with soluble Abs directed against NK1.1, Ly49H, Ly49D, or NKp46 required additional stimulation with cytokines, indicating that a range of activation receptors with distinct adaptor molecules require additional stimulation for IFN-γ production. The requirement for multiple signals extends to stimulation with primary m157-transgenic target cells, which triggers the activation receptor Ly49H, suggesting that NK cells do require multiple signals for IFN-γ production in the context of target cell recognition. Using quantitative PCR and RNA flow cytometry, we found that cytokines, not activating ligands, act on NK cells to express Ifng transcripts. Ly49H engagement is required for IFN-γ translational initiation. Results using inhibitors suggest that the proteasome-ubiquitin-IKK-TPL2-MNK1 axis was required during activation receptor engagement. Thus, this study indicates that activation receptor-dependent IFN-γ production is regulated on the transcriptional and translational levels.


Subject(s)
Interferon-gamma/immunology , Killer Cells, Natural/immunology , Proteasome Endopeptidase Complex/immunology , Protein Biosynthesis/immunology , Signal Transduction/immunology , Transcription, Genetic/immunology , Animals , Antigens, Ly/genetics , Antigens, Ly/immunology , Interferon-gamma/genetics , Killer Cells, Natural/cytology , Mice , Mice, Knockout , NK Cell Lectin-Like Receptor Subfamily A/genetics , NK Cell Lectin-Like Receptor Subfamily A/immunology , NK Cell Lectin-Like Receptor Subfamily B/genetics , NK Cell Lectin-Like Receptor Subfamily B/immunology , Natural Cytotoxicity Triggering Receptor 1/genetics , Natural Cytotoxicity Triggering Receptor 1/immunology , Proteasome Endopeptidase Complex/genetics , Signal Transduction/genetics
10.
Immunity ; 50(6): 1381-1390.e5, 2019 06 18.
Article in English | MEDLINE | ID: mdl-31103381

ABSTRACT

The process of affinity maturation, whereby T and B cells bearing antigen receptors with optimal affinity to the relevant antigen undergo preferential expansion, is a key feature of adaptive immunity. Natural killer (NK) cells are innate lymphocytes capable of "adaptive" responses after cytomegalovirus (CMV) infection. However, whether NK cells are similarly selected on the basis of their avidity for cognate ligand is unknown. Here, we showed that NK cells with the highest avidity for the mouse CMV glycoprotein m157 were preferentially selected to expand and comprise the memory NK cell pool, whereas low-avidity NK cells possessed greater capacity for interferon-γ (IFN-γ) production. Moreover, we provide evidence for avidity selection occurring in human NK cells during human CMV infection. These results delineate how heterogeneity in NK cell avidity diversifies NK cell effector function during antiviral immunity, and how avidity selection might serve to produce the most potent memory NK cells.


Subject(s)
Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/virology , Cytomegalovirus/immunology , Host-Pathogen Interactions/immunology , Killer Cells, Natural/immunology , Animals , Cytomegalovirus Infections/metabolism , Cytotoxicity, Immunologic , Gene Expression Regulation , Herpesviridae Infections/immunology , Herpesviridae Infections/metabolism , Herpesviridae Infections/virology , Host-Pathogen Interactions/genetics , Humans , Immunologic Memory , Killer Cells, Natural/metabolism , Lymphocyte Activation/immunology , Mice , Mice, Knockout , Muromegalovirus/immunology , NK Cell Lectin-Like Receptor Subfamily A/genetics , NK Cell Lectin-Like Receptor Subfamily A/metabolism , T-Cell Antigen Receptor Specificity
11.
Proc Natl Acad Sci U S A ; 115(45): 11579-11584, 2018 11 06.
Article in English | MEDLINE | ID: mdl-30249666

ABSTRACT

Adaptive natural killer (NK) cell memory represents a new frontier in immunology. Work over the last decade has discovered and confirmed the existence of NK cells with antigen-specific memories, which had previously been considered a unique property of T and B cells. These findings have shown that antigen-specific NK cells gain their specificity without the use of RAG proteins, representing a novel mechanism for generating antigen specificity, but the details of this mechanism have remained a mystery. We have discovered that members of the Ly49 family of surface receptors are critically involved in both the sensitization and the challenge phases of an NK cell memory response, as is antigen presentation from their binding partner, the class I MHC. Moreover, we demonstrate that the Ly49-interacting component of a presented antigen dictates the specificity of the NK cell memory response, implicating Ly49 receptors themselves in antigen-specific recognition. Finally, we demonstrate that adaptive NK cell memories can protect against an otherwise lethal melanoma without T cell or B cell support. These findings offer insight into the mechanism behind NK cell antigen specificity and demonstrate the clinical potential of this adaptive immune cell.


Subject(s)
Dermatitis, Contact/prevention & control , Immunologic Memory , Killer Cells, Natural/immunology , Melanoma, Experimental/therapy , NK Cell Lectin-Like Receptor Subfamily A/genetics , Peptides/immunology , Adaptive Immunity/drug effects , Amino Acid Sequence , Animals , Antigen Presentation , Cancer Vaccines/administration & dosage , Dermatitis, Contact/genetics , Dermatitis, Contact/immunology , Dermatitis, Contact/pathology , Dinitrofluorobenzene/administration & dosage , Female , Homeodomain Proteins/genetics , Homeodomain Proteins/immunology , Killer Cells, Natural/cytology , Killer Cells, Natural/drug effects , Male , Melanoma, Experimental/genetics , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , NK Cell Lectin-Like Receptor Subfamily A/immunology , Oxazoles/administration & dosage , Peptides/administration & dosage , Peptides/chemical synthesis , Vaccination
12.
J Immunol ; 199(5): 1567-1571, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28760883

ABSTRACT

NK cells play a critical role in host defense against viruses. In this study, we investigated the role of NKG2D in the expansion of NK cells after mouse CMV (MCMV) infection. Wild-type and NKG2D-deficient (Klrk1-/- ) Ly49H+ NK cells proliferated robustly when infected with MCMV strains engineered to allow expression of NKG2D ligands, which enhanced the response of wild-type NK cells. Naive NK cells exclusively express NKG2D-L, which pairs only with DAP10, whereas NKG2D-S expressed by activated NK cells pairs with DAP10 and DAP12, similar to Ly49H. However, NKG2D alone was unable to drive robust expansion of Ly49H- NK cells when mice were infected with these MCMV strains, likely because NKG2D-S was only transiently expressed postinfection. These findings demonstrate that NKG2D augments Ly49H-dependent proliferation of NK cells; however, NKG2D signaling alone is inadequate for expansion of NK cells, likely due to only transient expression of the NKG2D-DAP12 complex.


Subject(s)
Herpesviridae Infections/immunology , Killer Cells, Natural/immunology , Muromegalovirus/immunology , NK Cell Lectin-Like Receptor Subfamily A/metabolism , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Proliferation , Cells, Cultured , Immunity, Innate , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , NK Cell Lectin-Like Receptor Subfamily A/genetics , NK Cell Lectin-Like Receptor Subfamily K/genetics , Protein Binding , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Signal Transduction
13.
Immunol Lett ; 189: 40-47, 2017 09.
Article in English | MEDLINE | ID: mdl-28414184

ABSTRACT

Cytomegaloviruses (CMVs) have dedicated a large portion of their genome towards immune evasion targeting many aspects of the host immune system, particularly NK cells. However, the host managed to cope with the infection by developing multiple mechanisms to recognize viral threat and counterattack it, thus illustrating never-ending evolutionary interplay between CMV and its host. In this review, we will focus on several mechanisms of NK cell evasion by mouse CMV (MCMV), the role of host inhibitory and activating Ly49 receptors involved in the virus control and acquisition of adaptive features by NK cells as a consequence of MCMV infection.


Subject(s)
Herpesviridae Infections/immunology , Muromegalovirus/immunology , NK Cell Lectin-Like Receptor Subfamily A/metabolism , Animals , Autoantigens/immunology , Cytotoxicity, Immunologic , Evolution, Molecular , Histocompatibility Antigens Class I/metabolism , Host-Pathogen Interactions , Humans , Immune Evasion , Mice , NK Cell Lectin-Like Receptor Subfamily A/genetics
14.
Biochem J ; 474(1): 179-194, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27831490

ABSTRACT

Natural killer (NK) cells are lymphocytes of the innate immune system that eliminate virally infected or malignantly transformed cells. NK cell function is regulated by diverse surface receptors that are both activating and inhibitory. Among them, the homodimeric Ly49 receptors control NK cell cytotoxicity by sensing major histocompatibility complex class I molecules (MHC-I) on target cells. Although crystal structures have been reported for Ly49/MHC-I complexes, the underlying binding mechanism has not been elucidated. Accordingly, we carried out thermodynamic and kinetic experiments on the interaction of four NK Ly49 receptors (Ly49G, Ly49H, Ly49I and Ly49P) with two MHC-I ligands (H-2Dd and H-2Dk). These Ly49s embrace the structural and functional diversity of the highly polymorphic Ly49 family. Combining surface plasmon resonance, fluorescence anisotropy and far-UV circular dichroism (CD), we determined that the best model to describe both inhibitory and activating Ly49/MHC-I interactions is one in which the two MHC-I binding sites of the Ly49 homodimer present similar binding constants for the two sites (∼106 M-1) with a slightly positive co-operativity in some cases, and without far-UV CD observable conformational changes. Furthermore, Ly49/MHC-I interactions are diffusion-controlled and enthalpy-driven. These features stand in marked contrast with the activation-controlled and entropy-driven interaction of Ly49s with the viral immunoevasin m157, which is characterized by strong positive co-operativity and conformational selection. These differences are explained by the distinct structures of Ly49/MHC-I and Ly49/m157 complexes. Moreover, they reflect the opposing roles of NK cells to rapidly scan for virally infected cells and of viruses to escape detection using immunoevasins such as m157.


Subject(s)
Histocompatibility Antigen H-2D/chemistry , Multiprotein Complexes/chemistry , NK Cell Lectin-Like Receptor Subfamily A/chemistry , Animals , Histocompatibility Antigen H-2D/genetics , Histocompatibility Antigen H-2D/immunology , Kinetics , Mice , Mice, Inbred BALB C , Multiprotein Complexes/genetics , Multiprotein Complexes/immunology , NK Cell Lectin-Like Receptor Subfamily A/genetics , NK Cell Lectin-Like Receptor Subfamily A/immunology , Surface Plasmon Resonance , Thermodynamics
15.
Cancer Immunol Immunother ; 65(11): 1365-1375, 2016 11.
Article in English | MEDLINE | ID: mdl-27585789

ABSTRACT

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.


Subject(s)
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
16.
PLoS Biol ; 14(8): e1002526, 2016 08.
Article in English | MEDLINE | ID: mdl-27500644

ABSTRACT

It has recently been appreciated that NK cells exhibit many features reminiscent of adaptive immune cells. Considerable heterogeneity exists with respect to the ligand specificity of individual NK cells and as such, a subset of NK cells can respond, expand, and differentiate into memory-like cells in a ligand-specific manner. MHC I-binding inhibitory receptors, including those belonging to the Ly49 and KIR families, are expressed in a variegated manner, which creates ligand-specific diversity within the NK cell pool. However, how NK cells determine which inhibitory receptors to express on their cell surface during a narrow window of development is largely unknown. In this manuscript, we demonstrate that signals from activating receptors are critical for induction of Ly49 and KIR receptors during NK cell development; activating receptor-derived signals increased the probability of the Ly49 bidirectional Pro1 promoter to transcribe in the forward versus the reverse direction, leading to stable expression of Ly49 receptors in mature NK cells. Our data support a model where the balance of activating and inhibitory receptor signaling in NK cells selects for the induction of appropriate inhibitory receptors during development, which NK cells use to create a diverse pool of ligand-specific NK cells.


Subject(s)
Killer Cells, Natural/immunology , NK Cell Lectin-Like Receptor Subfamily A/immunology , Receptors, KIR/immunology , Signal Transduction/immunology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/immunology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cells, Cultured , Flow Cytometry , Genetic Variation/immunology , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , Humans , Killer Cells, Natural/metabolism , Ligands , Mice, Inbred C57BL , Mice, Knockout , NK Cell Lectin-Like Receptor Subfamily A/genetics , NK Cell Lectin-Like Receptor Subfamily A/metabolism , Phosphoproteins/genetics , Phosphoproteins/immunology , Phosphoproteins/metabolism , RNA Interference , Receptors, KIR/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics
17.
Sci Rep ; 6: 30564, 2016 07 29.
Article in English | MEDLINE | ID: mdl-27469529

ABSTRACT

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.


Subject(s)
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
18.
J Biol Chem ; 291(36): 18740-52, 2016 09 02.
Article in English | MEDLINE | ID: mdl-27385590

ABSTRACT

Murine natural killer (NK) cells are regulated by the interaction of Ly49 receptors with major histocompatibility complex class I molecules (MHC-I). Although the ligands for inhibitory Ly49 were considered to be restricted to classical MHC (MHC-Ia), we have shown that the non-classical MHC molecule (MHC-Ib) H2-M3 was a ligand for the inhibitory Ly49A. Here we establish that another MHC-Ib, H2-Q10, is a bona fide ligand for the inhibitory Ly49C receptor. H2-Q10 bound to Ly49C with a marginally lower affinity (∼5 µm) than that observed between Ly49C and MHC-Ia (H-2K(b)/H-2D(d), both ∼1 µm), and this recognition could be prevented by cis interactions with H-2K in situ To understand the molecular details underpinning Ly49·MHC-Ib recognition, we determined the crystal structures of H2-Q10 and Ly49C bound H2-Q10. Unliganded H2-Q10 adopted a classical MHC-I fold and possessed a peptide-binding groove that exhibited features similar to those found in MHC-Ia, explaining the diverse peptide binding repertoire of H2-Q10. Ly49C bound to H2-Q10 underneath the peptide binding platform to a region that encompassed residues from the α1, α2, and α3 domains, as well as the associated ß2-microglobulin subunit. This docking mode was conserved with that previously observed for Ly49C·H-2K(b) Indeed, structure-guided mutation of Ly49C indicated that Ly49C·H2-Q10 and Ly49C·H-2K(b) possess similar energetic footprints focused around residues located within the Ly49C ß4-stand and L5 loop, which contact the underside of the peptide-binding platform floor. Our data provide a structural basis for Ly49·MHC-Ib recognition and demonstrate that MHC-Ib represent an extended family of ligands for Ly49 molecules.


Subject(s)
Histocompatibility Antigen H-2D/chemistry , Killer Cells, Natural/chemistry , NK Cell Lectin-Like Receptor Subfamily A/chemistry , Animals , Crystallography, X-Ray , H-2 Antigens/chemistry , H-2 Antigens/genetics , H-2 Antigens/immunology , Histocompatibility Antigen H-2D/genetics , Histocompatibility Antigen H-2D/immunology , Killer Cells, Natural/immunology , Mice , Mice, Knockout , NK Cell Lectin-Like Receptor Subfamily A/genetics , NK Cell Lectin-Like Receptor Subfamily A/immunology , Protein Domains , Protein Structure, Quaternary
19.
Genes Immun ; 17(6): 349-57, 2016 09.
Article in English | MEDLINE | ID: mdl-27467282

ABSTRACT

The variegated expression of murine Ly49 loci has been associated with the probabilistic behavior of an upstream promoter active in immature cells, the Pro1 element. However, recent data suggest that Pro1 may be active in mature natural killer (NK) cells and function as an enhancer element. To assess directly if Pro1 transcripts are present in mature Ly49-expressing NK cells, RNA-sequencing of the total transcript pool was performed on freshly isolated splenic NK cells sorted for expression of either Ly49G or Ly49I. No Pro1 transcripts were detected from the Ly49a, Ly49c or Ly49i genes in mature Ly49(+) NK cells that contained high levels of Pro2 transcripts. Low levels of Ly49g Pro1 transcripts were found in both Ly49G(+) and Ly49G(-) populations, consistent with the presence of a small population of mature NK cells undergoing Ly49g gene activation, as previously demonstrated by culture of splenic NK cells in interleukin-2. Ly49 gene reporter constructs containing Pro1 failed to show any enhancer activity of Pro1 on Pro2 in a mature Ly49-expressing cell line. Taken together, the results are consistent with Pro1 transcription having a role in gene activation in developing NK, and argue against a role for Pro1 in Ly49 gene transcription by mature NK cells.


Subject(s)
Gene Expression Regulation , Killer Cells, Natural/metabolism , NK Cell Lectin-Like Receptor Subfamily A/genetics , Promoter Regions, Genetic , Animals , Cell Line , Enhancer Elements, Genetic , Female , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Sequence Analysis, RNA , Spleen/cytology , Spleen/metabolism , Transcription, Genetic
20.
J Immunol ; 197(1): 128-40, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27233959

ABSTRACT

NK cells possess inhibitory receptors that are responsible for self-MHC class I recognition; beyond their inhibitory function, accumulating evidence indicates that such receptors confer NK cell functional competence through an unclear process termed "licensing." Ly49C is the main self-specific inhibitory Ly49 receptor in H-2(b) C57BL/6 (B6) mice. We used B6 Ly49C-transgenic and B6 ß2 microglobulin (ß2m)-knockout Ly49C-transgenic mice to investigate the impact of licensing through this inhibitory receptor in precursor and mature NK cells. We found that self-specific inhibitory receptors affected NK cell precursor survival and proliferation at particular developmental stages in an MHC class I-dependent manner. The presence of Ly49C impacted the NK cell repertoire in a ß2m-dependent manner, with reduced Ly49A(+), Ly49G2(+), and Ly49D(+) subsets, an increased DNAM-1(+) subset, and higher NKG2D expression. Licensed NK cells displayed a skewed distribution of the maturation stages, which was characterized by differential CD27 and CD11b expression, toward the mature phenotypes. We found that Ly49C-mediated licensing induced a split effect on NK cell functions, with increased cytokine-production capabilities following engagement of various activating receptors while cytotoxicity remained unchanged. Analysis of licensed NK cell functions in vivo, in a system of mouse CMV infection, indicated that licensing did not play a major role in the NK cell antiviral response during acute infection, but it strongly impaired the generation and/or persistence of memory NK cells. This study unravels multifaceted effects of licensing on NK cell populations and their functions.


Subject(s)
Herpesviridae Infections/immunology , Killer Cells, Natural/immunology , Lymphocyte Subsets/immunology , Muromegalovirus/immunology , NK Cell Lectin-Like Receptor Subfamily A/metabolism , Animals , Antigens, Differentiation, T-Lymphocyte/metabolism , Cell Differentiation , Cells, Cultured , Cytotoxicity, Immunologic , Histocompatibility Antigens Class I/genetics , Immunity, Innate , Immunologic Memory , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , NK Cell Lectin-Like Receptor Subfamily A/genetics , NK Cell Lectin-Like Receptor Subfamily K/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...