Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 135
Filter
1.
HLA ; 103(2): e15387, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38358031

ABSTRACT

The immunogenome is the part of the genome that underlies immune mechanisms and evolves under various selective pressures. Two complex regions of the immunogenome, major histocompatibility complex (MHC) and natural killer cell receptor (NKR) genes, play an important role in the response to selective pressures of pathogens. Their importance is expressed by their genetic polymorphism at the molecular level, and their diversity associated with different types of diseases at the population level. Findings of associations between specific combinations of MHC/NKR haplotypes with different diseases in model species suggest that these gene complexes did not evolve independently. No such associations have been described in horses so far. The aim of the study was to detect associations between MHC and NKR gene/microsatellite haplotypes in three horse breed groups (Camargue, African, and Romanian) by statistical methods; chi-square test, Fisher's exact test, Pearson's goodness-of-fit test and logistic regression. Associations were detected for both MHC/NKR genes and microsatellites; the most significant associations were found between the most variable KLRA3 gene and the EQCA-1 or EQCA-2 genes. This finding supports the assumption that the KLRA3 is an important receptor for MHC I and that interactions of these molecules play important roles in the horse immunity and reproduction. Despite some limitations of the study such as low numbers of horses or lack of knowledge of the selected genes functions, the results were consistent across different statistical methods and remained significant even after overconservative Bonferroni corrections. We therefore consider them biologically plausible.


Subject(s)
Major Histocompatibility Complex , Polymorphism, Genetic , Animals , Horses/genetics , Humans , Receptors, Natural Killer Cell/genetics , Alleles , Major Histocompatibility Complex/genetics , Breeding
2.
J Immunol Res ; 2022: 7972039, 2022.
Article in English | MEDLINE | ID: mdl-35652109

ABSTRACT

Cancer immunotherapies are preferred over conventional treatments which are highly cytotoxic to normal cells. Focus has been on T cells but natural killer (NK) cells have equal potential. Concepts in cancer control and influence of sex require further investigation to improve successful mobilization of immune cells in cancer patients. Acute lymphoblastic leukemia (ALL) is a hematological malignancy mainly of B cell (B-ALL) and T cell (T-ALL) subtypes. Influence of ALL on NK cell is still unclear. Targeted next-generation sequencing was conducted on 62 activating/inhibitory receptors, ligands, effector, and exhaustion molecules on T-ALL (6 males) and normal controls (NC) (4 males and 4 females). Quantitative PCR (q-PCR) further investigated copy number variation (CNV), methylation index (MI), and mRNA expression of significant genes in T-ALL (14 males), NC (12 males and 12 females), and B-ALL samples (N = 12 males and 12 females). Bioinformatics revealed unique variants particularly rs2253849 (T>C) in KLRC1 and rs1141715 (A>G) in KLRC2 only among T-ALL (allele frequency 0.8-1.0). Gene amplification was highest in female B-ALL compared to male B-ALL (KLRC2, KLRC4, and NCR3, p < 0.05) and lowest in male T-ALL cumulating in deletion of KLRD1 and CD69. MI was higher in male ALL of both subtypes compared to normal (KIR2DL1-2 and 4 and KIR2DS2 and 4, p < 0.05) as well as to female B-ALL (KIR3DL2 and KIR2DS2, p < 0.05). mRNA expressions were low. Thus, ALL subtypes potentially regulated NK cell suppression by different mechanisms which should be considered in future immunotherapies for ALL.


Subject(s)
Precursor T-Cell Lymphoblastic Leukemia-Lymphoma , DNA Copy Number Variations , Female , Humans , Killer Cells, Natural , Male , NK Cell Lectin-Like Receptor Subfamily C/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , RNA, Messenger/metabolism , Receptors, Natural Killer Cell/genetics , Receptors, Natural Killer Cell/metabolism
3.
Front Immunol ; 13: 879124, 2022.
Article in English | MEDLINE | ID: mdl-35720328

ABSTRACT

Infection with the human immunodeficiency virus (HIV), when left untreated, typically leads to disease progression towards acquired immunodeficiency syndrome. Some people living with HIV (PLWH) control their virus to levels below the limit of detection of standard viral load assays, without treatment. As such, they represent examples of a functional HIV cure. These individuals, called Elite Controllers (ECs), are rare, making up <1% of PLWH. Genome wide association studies mapped genes in the major histocompatibility complex (MHC) class I region as important in HIV control. ECs have potent virus specific CD8+ T cell responses often restricted by protective MHC class I antigens. Natural Killer (NK) cells are innate immune cells whose activation state depends on the integration of activating and inhibitory signals arising from cell surface receptors interacting with their ligands on neighboring cells. Inhibitory NK cell receptors also use a subset of MHC class I antigens as ligands. This interaction educates NK cells, priming them to respond to HIV infected cell with reduced MHC class I antigen expression levels. NK cells can also be activated through the crosslinking of the activating NK cell receptor, CD16, which binds the fragment crystallizable portion of immunoglobulin G. This mode of activation confers NK cells with specificity to HIV infected cells when the antigen binding portion of CD16 bound immunoglobulin G recognizes HIV Envelope on infected cells. Here, we review the role of NK cells in antibody independent and antibody dependent HIV control.


Subject(s)
HIV Infections , HIV-1 , Genome-Wide Association Study , HLA Antigens , Histocompatibility Antigens Class I/genetics , Humans , Immunoglobulin G/genetics , Killer Cells, Natural , Ligands , Receptors, Natural Killer Cell/genetics
4.
HLA ; 99(2): 93-104, 2022 02.
Article in English | MEDLINE | ID: mdl-34921518

ABSTRACT

Chronic myeloid leukemia (CML) is a myeloproliferative neoplasm treated with tyrosine kinase inhibitors (TKIs). Although survival rates have improved, response to these treatments is highly heterogeneous. Variations in response rates may be due to different causes such as, treatment adherence, mutations in the BCR-ABL1 gene, clonal evolution and amplification of the BCR-ABL1 gene, but innate immune response is also considered to play a very important role and, specifically, NK cell activity through their receptors and ligands, could be determinant. The aim of this retrospective study was to explore the role of different activating and inhibiting KIR genes as well as the activating NKG2D receptor, present in NK cells, and also their respective ligands, HLA-A, -B, -C, -G, -F, MICA and MICB, in the progression of 190 patients with CML and treated at two hospitals from Barcelona between 2000 and 2019. Early molecular response (EMR), major molecular response (MMR) or MR3.0 and deep molecular response (DMR) or MR4.0 were correlated. As control samples, healthy donors from the Barcelona Blood Bank were analyzed. The presence of KIR2DL2/KIR2DS2 was associated with the achievement of EMR, MR3.0, and MR4.0. Carriers of the higher expression NKG2D variant and MICA*009:01 were also likely to achieve molecular response (MR). The most remarkable difference between CML patients and controls was a higher frequency of the lower expression NKG2D variant in CML patients. In summary, our results showed that activating NK receptor phenotypes might help to achieve MR and DMR in CML patients treated with TKIs although confirmatory studies are necessary.


Subject(s)
Leukemia, Myelogenous, Chronic, BCR-ABL Positive , NK Cell Lectin-Like Receptor Subfamily K , Alleles , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Ligands , NK Cell Lectin-Like Receptor Subfamily K/genetics , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Receptors, Natural Killer Cell/genetics , Retrospective Studies
5.
Front Immunol ; 12: 796072, 2021.
Article in English | MEDLINE | ID: mdl-34956230

ABSTRACT

Natural killer cells are important effector cells in the immune response against myeloid malignancies. Previous studies show that the expression of activating NK cell receptors is pivotal for efficient recognition of blasts from patients with acute myeloid leukemia (AML) and that high expression levels impact favorably on patient survival. This study investigated the potential impact of activating receptor gene variants on NK cell receptor expression and survival in a cohort of AML patients receiving relapse-preventive immunotherapy with histamine dihydrochloride and low-dose IL-2 (HDC/IL-2). Patients harboring the G allele of rs1049174 in the KLRK1 gene encoding NKG2D showed high expression of NKG2D by CD56bright NK cells and a favorable clinical outcome in terms of overall survival. For DNAM-1, high therapy-induced receptor expression entailed improved survival, while patients with high DNAM-1 expression before immunotherapy associated with unfavorable clinical outcome. The previously reported SNPs in NCR3 encoding NKp30, which purportedly influence mRNA splicing into isoforms with discrete functions, did not affect outcome in this study. Our results imply that variations in genes encoding activating NK cell receptors determine receptor expression and clinical outcome in AML immunotherapy.


Subject(s)
Killer Cells, Natural/immunology , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/immunology , Receptors, Natural Killer Cell/genetics , Adolescent , Adult , Aged , Female , Genotype , Humans , Immunotherapy , Male , Middle Aged , Polymorphism, Single Nucleotide , Treatment Outcome , Young Adult
6.
Front Immunol ; 12: 713158, 2021.
Article in English | MEDLINE | ID: mdl-34394116

ABSTRACT

Immune checkpoint inhibitors (ICI) revolutionized the field of immuno-oncology and opened new avenues towards the development of novel assets to achieve durable immune control of cancer. Yet, the presence of tumor immune evasion mechanisms represents a challenge for the development of efficient treatment options. Therefore, combination therapies are taking the center of the stage in immuno-oncology. Such combination therapies should boost anti-tumor immune responses and/or target tumor immune escape mechanisms, especially those created by major players in the tumor microenvironment (TME) such as tumor-associated macrophages (TAM). Natural killer (NK) cells were recently positioned at the forefront of many immunotherapy strategies, and several new approaches are being designed to fully exploit NK cell antitumor potential. One of the most relevant NK cell-activating receptors is NKG2D, a receptor that recognizes 8 different NKG2D ligands (NKG2DL), including MICA and MICB. MICA and MICB are poorly expressed on normal cells but become upregulated on the surface of damaged, transformed or infected cells as a result of post-transcriptional or post-translational mechanisms and intracellular pathways. Their engagement of NKG2D triggers NK cell effector functions. Also, MICA/B are polymorphic and such polymorphism affects functional responses through regulation of their cell-surface expression, intracellular trafficking, shedding of soluble immunosuppressive isoforms, or the affinity of NKG2D interaction. Although immunotherapeutic approaches that target the NKG2D-NKG2DL axis are under investigation, several tumor immune escape mechanisms account for reduced cell surface expression of NKG2DL and contribute to tumor immune escape. Also, NKG2DL polymorphism determines functional NKG2D-dependent responses, thus representing an additional challenge for leveraging NKG2DL in immuno-oncology. In this review, we discuss strategies to boost MICA/B expression and/or inhibit their shedding and propose that combination strategies that target MICA/B with antibodies and strategies aimed at promoting their upregulation on tumor cells or at reprograming TAM into pro-inflammatory macrophages and remodeling of the TME, emerge as frontrunners in immuno-oncology because they may unleash the antitumor effector functions of NK cells and cytotoxic CD8 T cells (CTL). Pursuing several of these pipelines might lead to innovative modalities of immunotherapy for the treatment of a wide range of cancer patients.


Subject(s)
GPI-Linked Proteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Neoplasms/etiology , Neoplasms/metabolism , Animals , Combined Modality Therapy , Disease Management , Disease Susceptibility , GPI-Linked Proteins/genetics , Gene Expression Regulation, Neoplastic , Humans , Intercellular Signaling Peptides and Proteins/genetics , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Ligands , Molecular Targeted Therapy , Neoplasms/pathology , Neoplasms/therapy , Receptors, Natural Killer Cell/genetics , Receptors, Natural Killer Cell/metabolism , Signal Transduction , Tumor Escape , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
7.
Front Immunol ; 12: 680480, 2021.
Article in English | MEDLINE | ID: mdl-34295330

ABSTRACT

Natural killer (NK) cells are innate lymphocytes that can kill diseased- or virally-infected cells, mediate antibody dependent cytotoxicity and produce type I immune-associated cytokines upon activation. NK cells also contribute to the allo-immune response upon kidney transplantation either by promoting allograft rejection through lysis of cells of the transplanted organ or by promoting alloreactive T cells. In addition, they protect against viral infections upon transplantation which may be especially relevant in patients receiving high dose immune suppression. NK cell activation is tightly regulated through the integrated balance of signaling via inhibitory- and activating receptors. HLA class I molecules are critical regulators of NK cell activation through the interaction with inhibitory- as well as activating NK cell receptors, hence, HLA molecules act as critical immune checkpoints for NK cells. In the current review, we evaluate how NK cell alloreactivity and anti-viral immunity are regulated by NK cell receptors belonging to the KIR family and interacting with classical HLA class I molecules, or by NKG2A/C and LILRB1/KIR2DL4 engaging non-classical HLA-E or -G. In addition, we provide an overview of the methods to determine genetic variation in these receptors and their HLA ligands.


Subject(s)
Disease Susceptibility/immunology , Histocompatibility Antigens Class I/immunology , Kidney Transplantation/adverse effects , Killer Cells, Natural/immunology , Virus Diseases/etiology , Animals , Biomarkers , Histocompatibility Testing , Humans , Immune Checkpoint Proteins/immunology , Immune Checkpoint Proteins/metabolism , Isoantibodies/immunology , Killer Cells, Natural/metabolism , Ligands , Lymphocyte Activation/immunology , Prognosis , Protein Binding , Receptors, Natural Killer Cell/genetics , Receptors, Natural Killer Cell/metabolism , Transplantation Immunology , Treatment Outcome , Virus Diseases/metabolism
8.
Int J Mol Sci ; 22(13)2021 Jun 22.
Article in English | MEDLINE | ID: mdl-34206399

ABSTRACT

Key features of chronic lymphocytic leukemia (CLL) are defects in the immune system and the ability of leukemic cells to evade immune defenses and induce immunosuppression, resulting in increased susceptibility to infections and disease progression. Several immune effectors are impaired in CLL, including T and natural killer (NK) cells. The role of T cells in defense against CLL and in CLL progression and immunotherapy has been extensively studied. Less is known about the role of NK cells in this leukemia, and data on NK cell alterations in CLL are contrasting. Besides studies showing that NK cells have intrinsic defects in CLL, there is a large body of evidence indicating that NK cell dysfunctions in CLL mainly depend on the escape mechanisms employed by leukemic cells. In keeping, it has been shown that NK cell functions, including antibody-dependent cellular cytotoxicity (ADCC), can be retained and/or restored after adequate stimulation. Therefore, due to their preserved ADCC function and the reversibility of CLL-related dysfunctions, NK cells are an attractive source for novel immunotherapeutic strategies in this disease, including chimeric antigen receptor (CAR) therapy. Recently, satisfying clinical responses have been obtained in CLL patients using cord blood-derived CAR-NK cells, opening new possibilities for further exploring NK cells in the immunotherapy of CLL. However, notwithstanding the promising results of this clinical trial, more evidence is needed to fully understand whether and in which CLL cases NK cell-based immunotherapy may represent a valid, alternative/additional therapeutic option for this leukemia. In this review, we provide an overview of the current knowledge about phenotypic and functional alterations of NK cells in CLL and the mechanisms by which CLL cells circumvent NK cell-mediated immunosurveillance. Additionally, we discuss the potential relevance of using NK cells in CLL immunotherapy.


Subject(s)
Disease Susceptibility , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/etiology , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Biomarkers , Cell Communication , Disease Management , Humans , Immune System/immunology , Immune System/metabolism , Immunotherapy/adverse effects , Immunotherapy/methods , Leukemia, Lymphocytic, Chronic, B-Cell/diagnosis , Leukemia, Lymphocytic, Chronic, B-Cell/therapy , Ligands , Protein Binding , Receptors, Natural Killer Cell/genetics , Receptors, Natural Killer Cell/metabolism , Treatment Outcome , Tumor Escape/genetics , Tumor Escape/immunology
9.
Signal Transduct Target Ther ; 6(1): 236, 2021 06 23.
Article in English | MEDLINE | ID: mdl-34158475

ABSTRACT

Despite the successful use of the humanized monoclonal antibody trastuzumab (Herceptin) in the clinical treatment of human epidermal growth factor receptor 2 (HER2)-overexpressing breast cancer, the frequently occurring drug resistance remains to be overcome. The regulatory mechanisms of trastuzumab-elicited immune response in the tumor microenvironment remain largely uncharacterized. Here, we found that the nonclassical histocompatibility antigen HLA-G desensitizes breast cancer cells to trastuzumab by binding to the natural killer (NK) cell receptor KIR2DL4. Unless engaged by HLA-G, KIR2DL4 promotes antibody-dependent cell-mediated cytotoxicity and forms a regulatory circuit with the interferon-γ (IFN-γ) production pathway, in which IFN-γ upregulates KIR2DL4 via JAK2/STAT1 signaling, and then KIR2DL4 synergizes with the Fcγ receptor to increase IFN-γ secretion by NK cells. Trastuzumab treatment of neoplastic and NK cells leads to aberrant cytokine production characterized by excessive tumor growth factor-ß (TGF-ß) and IFN-γ, which subsequently reinforce HLA-G/KIR2DL4 signaling. In addition, TGF-ß and IFN-γ impair the cytotoxicity of NK cells by upregulating PD-L1 on tumor cells and PD-1 on NK cells. Blockade of HLA-G/KIR2DL4 signaling improved the vulnerability of HER2-positive breast cancer to trastuzumab treatment in vivo. These findings provide novel insights into the mechanisms underlying trastuzumab resistance and demonstrate the applicability of combined HLA-G and PD-L1/PD-1 targeting in the treatment of trastuzumab-resistant breast cancer.


Subject(s)
Breast Neoplasms/drug therapy , HLA-G Antigens/genetics , Receptor, ErbB-2/genetics , Receptors, KIR2DL4/genetics , Trastuzumab/pharmacology , Adult , Aged , Antibodies, Monoclonal, Humanized/genetics , Antibodies, Monoclonal, Humanized/immunology , Antibody-Dependent Cell Cytotoxicity/genetics , Antibody-Dependent Cell Cytotoxicity/immunology , Breast Neoplasms/genetics , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Drug Resistance, Neoplasm/immunology , Female , Humans , Interferon-gamma/genetics , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Middle Aged , Receptors, Natural Killer Cell/genetics , Receptors, Natural Killer Cell/immunology , Trastuzumab/adverse effects , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
10.
J Clin Invest ; 131(13)2021 07 01.
Article in English | MEDLINE | ID: mdl-34043588

ABSTRACT

Peripheral T cell lymphomas (PTCLs) represent a significant unmet medical need with dismal clinical outcomes. The T cell receptor (TCR) is emerging as a key driver of T lymphocyte transformation. However, the role of chronic TCR activation in lymphomagenesis and in lymphoma cell survival is still poorly understood. Using a mouse model, we report that chronic TCR stimulation drove T cell lymphomagenesis, whereas TCR signaling did not contribute to PTCL survival. The combination of kinome, transcriptome, and epigenome analyses of mouse PTCLs revealed a NK cell-like reprogramming of PTCL cells with expression of NK receptors (NKRs) and downstream signaling molecules such as Tyrobp and SYK. Activating NKRs were functional in PTCLs and dependent on SYK activity. In vivo blockade of NKR signaling prolonged mouse survival, demonstrating the addiction of PTCLs to NKRs and downstream SYK/mTOR activity for their survival. We studied a large collection of human primary samples and identified several PTCLs recapitulating the phenotype described in this model by their expression of SYK and the NKR, suggesting a similar mechanism of lymphomagenesis and establishing a rationale for clinical studies targeting such molecules.


Subject(s)
Lymphoma, T-Cell, Peripheral/immunology , Receptors, Antigen, T-Cell/immunology , Receptors, Natural Killer Cell/immunology , Animals , Carcinogenesis/genetics , Carcinogenesis/immunology , Cellular Reprogramming/genetics , Cellular Reprogramming/immunology , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Genes, p53 , Humans , Killer Cells, Natural/immunology , Lymphoma, T-Cell, Peripheral/genetics , Lymphoma, T-Cell, Peripheral/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasms, Experimental/genetics , Neoplasms, Experimental/immunology , Neoplasms, Experimental/metabolism , Receptors, Antigen, T-Cell/genetics , Receptors, Natural Killer Cell/genetics , Signal Transduction/genetics , Signal Transduction/immunology , Syk Kinase/metabolism , T-Lymphocytes/immunology
11.
Int J Immunogenet ; 48(2): 157-171, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33352617

ABSTRACT

Natural killer (NK) cells are of major significance in patients after allogeneic haematopoietic stem cell transplantation (HSCT). They are the first subset of lymphocytes to appear in peripheral blood after transplantation and play an important role in the immune responses against cancer and viral infections. The function of NK cells is controlled by various surface receptors, of which type I integral proteins with immunoglobulin-like domains (killer-cell immunoglobulin-like receptors, KIRs) have been the most extensively studied. The present review focuses on less studied NK cell receptors, such as type II integral proteins with lectin-like domains (CD94/NKG2, NKG2D), natural cytotoxicity receptors (NCRs), immunoglobulin-like transcripts (ILTs) and their ligands. Their potential role in patients with haematological disorders subjected to HSC transplant procedure in the context of post-transplant complications such as viral reactivation and acute graft-versus-host disease (GvHD) will be presented and discussed.


Subject(s)
Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/immunology , Killer Cells, Natural/immunology , Receptors, Natural Killer Cell/immunology , Allografts , Cytotoxicity, Immunologic , Graft vs Host Disease/immunology , Hematologic Diseases/immunology , Hematologic Diseases/therapy , Humans , Lymphocyte Activation , Receptors, Immunologic/immunology , Receptors, Natural Killer Cell/classification , Receptors, Natural Killer Cell/genetics , Virus Activation
12.
Genes (Basel) ; 13(1)2021 12 29.
Article in English | MEDLINE | ID: mdl-35052430

ABSTRACT

Conventional genome-wide association studies (GWASs) of complex traits, such as Multiple Sclerosis (MS), are reliant on per-SNP p-values and are therefore heavily burdened by multiple testing correction. Thus, in order to detect more subtle alterations, ever increasing sample sizes are required, while ignoring potentially valuable information that is readily available in existing datasets. To overcome this, we used penalised regression incorporating elastic net with a stability selection method by iterative subsampling to detect the potential interaction of loci with MS risk. Through re-analysis of the ANZgene dataset (1617 cases and 1988 controls) and an IMSGC dataset as a replication cohort (1313 cases and 1458 controls), we identified new association signals for MS predisposition, including SNPs above and below conventional significance thresholds while targeting two natural killer receptor loci and the well-established HLA loci. For example, rs2844482 (98.1% iterations), otherwise ignored by conventional statistics (p = 0.673) in the same dataset, was independently strongly associated with MS in another GWAS that required more than 40 times the number of cases (~45 K). Further comparison of our hits to those present in a large-scale meta-analysis, confirmed that the majority of SNPs identified by the elastic net model reached conventional statistical GWAS thresholds (p < 5 × 10-8) in this much larger dataset. Moreover, we found that gene variants involved in oxidative stress, in addition to innate immunity, were associated with MS. Overall, this study highlights the benefit of using more advanced statistical methods to (re-)analyse subtle genetic variation among loci that have a biological basis for their contribution to disease risk.


Subject(s)
HLA Antigens/genetics , Multiple Sclerosis/genetics , Polymorphism, Single Nucleotide , Quantitative Trait Loci , Receptors, Natural Killer Cell/genetics , Case-Control Studies , Cohort Studies , Female , Genome-Wide Association Study , Humans , Male , Multiple Sclerosis/pathology , Regression Analysis
13.
Front Immunol ; 12: 816499, 2021.
Article in English | MEDLINE | ID: mdl-35087536

ABSTRACT

Natural killer (NK) cells represent a subset of CD3- CD7+ CD56+/dim lymphocytes with cytotoxic and suppressor activity against virus-infected cells and cancer cells. The overall potential of NK cells has brought them to the spotlight of targeted immunotherapy in solid and hematological malignancies, including multiple myeloma (MM). Nonetheless, NK cells are subjected to a variety of cancer defense mechanisms, leading to impaired maturation, chemotaxis, target recognition, and killing. This review aims to summarize the available and most current knowledge about cancer-related impairment of NK cell function occurring in MM.


Subject(s)
Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Multiple Myeloma/etiology , Multiple Myeloma/metabolism , Tumor Microenvironment/immunology , Animals , Biomarkers , Cytotoxicity, Immunologic , Disease Management , Disease Susceptibility , Humans , Immunity , Immunomodulation/drug effects , Molecular Targeted Therapy , Multiple Myeloma/diagnosis , Multiple Myeloma/therapy , Prognosis , Receptors, Natural Killer Cell/genetics , Receptors, Natural Killer Cell/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Treatment Outcome , Tumor Microenvironment/drug effects
14.
Clin Exp Immunol ; 202(1): 11-27, 2020 10.
Article in English | MEDLINE | ID: mdl-32544282

ABSTRACT

Natural killer (NK) cells are innate immune effectors which play a crucial role in recognizing and eliminating virally infected and cancerous cells. They effectively distinguish between healthy and distressed self through the integration of signals delivered by germline-encoded activating and inhibitory cell surface receptors. The frequent up-regulation of stress markers on genetically unstable cancer cells has prompted the development of novel immunotherapies that exploit such innate receptors. One prominent example entails the development of chimeric antigen receptors (CAR) that detect cell surface ligands bound by NK receptors, coupling this engagement to the delivery of tailored immune activating signals. Here, we review strategies to engineer CARs in which specificity is conferred by natural killer group 2D (NKG2D) or other NK receptor types. Multiple preclinical studies have demonstrated the remarkable ability of chimeric NK receptor-targeted T cells and NK cells to effectively and specifically eliminate cancer cells and to reject established tumour burdens. Importantly, such systems act not only acutely but, in some cases, they also incite immunological memory. Moreover, CARs targeted with the NKG2D ligand binding domain have also been shown to disrupt the tumour microenvironment, through the targeting of suppressive T regulatory cells, myeloid-derived suppressor cells and tumour vasculature. Collectively, these findings have led to the initiation of early-phase clinical trials evaluating both autologous and allogeneic NKG2D-targeted CAR T cells in the haematological and solid tumour settings.


Subject(s)
Immunotherapy, Adoptive , Killer Cells, Natural , Myeloid-Derived Suppressor Cells , Neoplasms , Receptors, Chimeric Antigen , Receptors, Natural Killer Cell , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/pathology , Killer Cells, Natural/transplantation , Myeloid-Derived Suppressor Cells/immunology , Myeloid-Derived Suppressor Cells/pathology , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/pathology , Neoplasms/therapy , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/therapeutic use , Receptors, Natural Killer Cell/genetics , Receptors, Natural Killer Cell/immunology
15.
Br J Haematol ; 191(3): 433-441, 2020 11.
Article in English | MEDLINE | ID: mdl-32352166

ABSTRACT

There is currently no biomarker that reliably predicts treatment-free remission (TFR) in chronic myeloid leukaemia (CML). We characterised effector and suppressor immune responses at the time of tyrosine kinase inhibitor (TKI) cessation in patients from the CML8 and CML10 clinical studies. Natural killer (NK) cells with increased expression of activating NK receptors were higher in patients who achieved TFR. There was no difference in the proportion of CD4+ or CD8+ T cells. Furthermore, we found that FoxP3+ regulatory T cells (T reg) and monocytic myeloid-derived suppressor cells (Mo-MDSCs) were concomitantly decreased in TFR patients, suggesting that the effector and suppressor arms of the immune system work in concert to mediate TFR. A discovery cohort (CML10) was used to generate a predictive model, using logistic regression. Patients classified into the high-risk group were more likely to relapse when compared with the low-risk group (HR 7·4, 95% CI 2·9-19·1). The model was successfully validated on the independent CML8 cohort (HR 8·3, 95% CI 2·2-31·3). Effective prediction of TFR success may be obtained with an effector-suppressor score, calculated using absolute NK cell, T reg, and Mo-MDSC counts, at TKI cessation, reflecting the contribution of both immune suppressors and effectors in the immunobiology underlying successful TFR.


Subject(s)
Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/immunology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Myeloid-Derived Suppressor Cells/immunology , Myeloid-Derived Suppressor Cells/metabolism , Adult , Aged , Aged, 80 and over , Antineoplastic Agents, Immunological/administration & dosage , Antineoplastic Agents, Immunological/adverse effects , Antineoplastic Agents, Immunological/therapeutic use , Biomarkers , Female , Humans , Immunomodulation/drug effects , Killer Cells, Natural/drug effects , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/mortality , Leukocyte Count , Male , Middle Aged , Myeloid-Derived Suppressor Cells/drug effects , Prognosis , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/therapeutic use , Receptors, Natural Killer Cell/genetics , Receptors, Natural Killer Cell/metabolism , Recurrence , Remission Induction , Treatment Outcome
16.
Front Immunol ; 11: 197, 2020.
Article in English | MEDLINE | ID: mdl-32153566

ABSTRACT

Increasing evidence points to a key role for NK cells in controlling adaptive immune responses. In studies examining the role of CD1d on CD4+ T cell responses, we found that a line of CD1d-deficient mice on the C57BL/6J background had a homozygous 129 locus on chromosome 6 containing the entire NK cell gene cluster. Mice possessing this locus (C57BL/6.NKC129) displayed a >10-fold reduction in antigen-specific CD4+ T cell responses after intracranial infection with lymphocytic choriomeningitis virus (LCMV). Neither parental strain displayed defects in viral-specific CD4+ T cell responses. Interestingly, following infection, increased numbers of NK cells accumulated in the lymph nodes of C57BL/6.NKC129 mice and displayed enhanced in vivo functionality. Moreover, depletion of NK cells with anti-asialo-GM-1 antibody in C57BL/6.NKC129 mice resulted in a >20-fold increase in viral-specific CD4+ T cell responses. Mechanistically, we found that dendritic cell antigen presentation and early type I IFN production were significantly decreased in C57BL/6.NKC129 mice, but were restored in perforin-deficient C57BL/6.NKC129 mice or following NK depletion. Together, these data reveal that the variable genomic regions containing the activating/inhibitory NK cell receptors are key determinants of antigen-specific CD4+ T cell responses, controlling type I IFN production and the antigen-presenting capacity of dendritic cells.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Genetic Loci , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Receptors, Natural Killer Cell/genetics , Animals , Antigen Presentation/genetics , Antigens, CD1d/genetics , Dendritic Cells/immunology , Interferon Type I/biosynthesis , Killer Cells, Natural/immunology , Lymph Nodes/immunology , Lymphocyte Activation/genetics , Lymphocytic Choriomeningitis/virology , Mice , Mice, Inbred C57BL , Mice, Knockout , Pore Forming Cytotoxic Proteins/deficiency , Pore Forming Cytotoxic Proteins/genetics
17.
Immunobiology ; 225(3): 151909, 2020 05.
Article in English | MEDLINE | ID: mdl-32051096

ABSTRACT

NK cells are important in the onset of acute myocardial infarction (AMI) by their ability to secrete IFN-γ and other inflammatory cytokines. They also participate in regulating pathological cardiac remodeling after myocardial infarction. Mechanisms of regulation, however, are incompletely understood. Herein, the aim of this study is to explore the possible association between the expression pattern of different NK cell receptors (phenotype), as well as the cytotoxic function of NK cells from AMI patients with their myocardial function after three months follow-up. We analyzed the phenotype and function of both CD56dimCD16+ and CD56brightCD16- NK cells from twenty-one patients within the first 72 h after ST-elevation AMI and three-month follow-up, as well as fifteen healthy controls. Clinical characteristics and ventricular function determined by echocardiography were also evaluated. NK cells from AMI patients showed an activated phenotype, characterized by high TNF-α production and low percentages of the activating receptor NKG2D. Interestingly, AMI patients display higher levels of circulating IL-10+ NK cells. Three-month follow-up showed that NK cells exhibit a diminished cytotoxic function. These data show that NK cells may have a role mediating myocardial remodeling by regulating the inflammatory response, mainly by the production of IL-10. We also propose that NKG2D may have a role in the onset of the inflammatory response immediately after AMI. The precise regulation of NK cells function may represent an important step in recovery of myocardial function.


Subject(s)
Disease Susceptibility , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Myocardial Infarction/etiology , Myocardial Infarction/metabolism , Receptors, Natural Killer Cell/metabolism , Biomarkers , CD56 Antigen/metabolism , Cytokines/blood , Cytokines/metabolism , Cytotoxicity Tests, Immunologic , Female , Follow-Up Studies , Humans , Inflammation Mediators/metabolism , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Male , Myocardial Infarction/blood , Receptors, IgG/metabolism , Receptors, Natural Killer Cell/genetics
18.
Virology ; 539: 26-37, 2020 01 02.
Article in English | MEDLINE | ID: mdl-31670188

ABSTRACT

In addition to direct anti-viral activity, NK cells regulate viral pathogenesis by virtue of their cytolytic attack on activated CD4 and CD8 T cells. To gain insight into which differentiated T cell subsets are preferred NK targets, transgenic T cells were differentiated in vitro into Th0, Th1, Th2, Th17, Treg, Tc1, and Tc2 effector cells and then tested for lysis by enriched populations of lymphocytic choriomeningitis virus (LCMV)-induced activated NK cells. There was a distinct hierarchy of cytotoxicity in vitro and in vivo, with Treg, Th17, and Th2 cells being more sensitive and Th0 and Th1 cells more resistant. Some distinctions between in vitro vs in vivo generated T cells were explainable by type 1 interferon induction of class 1 histocompatibility antigens on the effector T cell subsets. NK receptor (NKR)-deficient mice and anti-NKR antibody studies identified no one essential NKR for killing, though there could be redundancies.


Subject(s)
Cytotoxicity, Immunologic , Killer Cells, Natural/immunology , Killer Cells, Natural/virology , T-Lymphocyte Subsets/immunology , Animals , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Cytotoxicity, Immunologic/drug effects , Interferons/genetics , Interferons/pharmacology , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Lymphocytic choriomeningitis virus/pathogenicity , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Receptors, Natural Killer Cell/antagonists & inhibitors , Receptors, Natural Killer Cell/genetics , T-Lymphocyte Subsets/drug effects
19.
Front Immunol ; 10: 177, 2019.
Article in English | MEDLINE | ID: mdl-30837985

ABSTRACT

Natural killer (NK) cells have diverse roles in hominid immunity and reproduction. Modulating these functions are the interactions between major histocompatibility complex (MHC) class I molecules that are ligands for two NK cell surface receptor types. Diverse killer cell immunoglobulin-like receptors (KIR) bind specific motifs encoded within the polymorphic MHC class I cell surface glycoproteins, while, in more conserved interactions, CD94:NKG2A receptors recognize MHC-E with bound peptides derived from MHC class I leader sequences. The hominid lineage presents a choreographed co-evolution of KIR with their MHC class I ligands. MHC-A, -B, and -C are present in all great apes with species-specific haplotypic variation in gene content. The Bw4 epitope recognized by lineage II KIR is restricted to MHC-B but also present on some gorilla and human MHC-A. Common to great apes, but rare in humans, are MHC-B possessing a C1 epitope recognized by lineage III KIR. MHC-C arose from duplication of MHC-B and is fixed in all great apes except orangutan, where it exists on approximately 50% of haplotypes and all allotypes are C1-bearing. Recent study showed that gorillas possess yet another intermediate MHC organization compared to humans. Like orangutans, but unlike the Pan-Homo species, duplication of MHC-B occurred. However, MHC-C is fixed, and the MHC-C C2 epitope (absent in orangutans) emerges. The evolution of MHC-C drove expansion of its cognate lineage III KIR. Recently, position -21 of the MHC-B leader sequence has been shown to be critical in determining NK cell educational outcome. In humans, methionine (-21M) results in CD94:NKG2A-focused education whereas threonine (-21T) produces KIR-focused education. This is another dynamic position among hominids. Orangutans have exclusively -21M, consistent with their intermediate stage in lineage III KIR-focused evolution. Gorillas have both -21M and -21T, like humans, but they are unequally encoded by their duplicated B genes. Chimpanzees have near-fixed -21T, indicative of KIR-focused NK education. Harmonious with this observation, chimpanzee KIR exhibit strong binding and, compared to humans, smaller differences between binding levels of activating and inhibitory KIR. Consistent between these MHC-NK cell receptor systems over the course of hominid evolution is the evolution of polymorphism favoring the more novel and dynamic KIR system.


Subject(s)
Biological Evolution , Histocompatibility Antigens/immunology , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Receptors, Natural Killer Cell/metabolism , Alleles , Animals , Evolution, Molecular , Gene Duplication , Haplotypes , Histocompatibility Antigens/chemistry , Histocompatibility Antigens/genetics , Hominidae/classification , Hominidae/physiology , Humans , Ligands , Phylogeny , Polymorphism, Genetic , Protein Binding , Protein Sorting Signals , Receptors, Natural Killer Cell/chemistry , Receptors, Natural Killer Cell/genetics , Reproduction/genetics
20.
Cell Mol Immunol ; 16(5): 430-441, 2019 05.
Article in English | MEDLINE | ID: mdl-30778167

ABSTRACT

NK cells play important roles in innate defenses against viruses and in the control of tumor growth and metastasis. The regulation/induction of NK cell function is mediated by an array of activating or inhibitory surface receptors. In humans, major activating receptors involved in target cell killing are the natural cytotoxicity receptors (NCRs) and NKG2D. Activating receptors recognize ligands that are overexpressed or expressed de novo upon cell stress, viral infection, or tumor transformation. The HLA-class I-specific inhibitory receptors, including KIRs recognizing HLA-class I allotypic determinants and CD94/NKG2A recognizing the class-Ib HLA-E, constitute a fail-safe mechanism to avoid unwanted NK-mediated damage to healthy cells. Other receptors such as PD-1, primarily expressed by activated T lymphocytes, are important inhibitory checkpoints of immune responses that ensure T-cell tolerance. PD-1 also may be expressed by NK cells in cancer patients. Since PD-1 ligand (PD-L1) may be expressed by different tumors, PD-1/PD-L1 interactions inactivate both T and NK cells. Thus, the reliable evaluation of PD-L1 expression in tumors has become a major issue to select patients who may benefit from therapy with mAbs disrupting PD-1/PD-L1 interactions. Recently, NKG2A was revealed to be an important checkpoint controlling both NK and T-cell activation. Since most tumors express HLA-E, mAbs targeting NKG2A has been used alone or in combination with other therapeutic mAbs targeting PD-1 or tumor antigens (e.g., EGFR), with encouraging results. The translational value of NK cells and their receptors is evidenced by the extraordinary therapeutic success of haploidentical HSCT to cure otherwise fatal high-risk leukemias.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Hematopoietic Stem Cell Transplantation , Immunotherapy/methods , Killer Cells, Natural/physiology , Leukemia/therapy , Receptors, KIR/metabolism , Receptors, Natural Killer Cell/metabolism , B7-H1 Antigen/metabolism , Humans , Leukemia/immunology , Lymphocyte Activation , Programmed Cell Death 1 Receptor/metabolism , Receptors, Natural Killer Cell/genetics , T-Lymphocytes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...