Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Int Immunopharmacol ; 97: 107677, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33933844

ABSTRACT

Natural killer (NK) cells are involved in innate and acquired immunity, stimulating and enhancing immune responses via secretion of IFN-γ and TNF-α. NKG2D is among the most important NK's stimulant receptors, the ligands of which are elevated on cancerous and virus-infected cells. We analyzed effect of 5-ALA on gene expression and receptor presentation of NKG2D, which is present on peripheral blood NK cells. Mononuclear cells were isolated from the venous blood samples of healthy individuals. RNA extraction and cDNA synthesis were performed after exposure of samples to 5-ALA, and gene expression was evaluated using Real-Time PCR, and the receptor presence rate on the cell surface was evaluated by flow-cytometry analysis. The results showed the gene expression of NKG2D and the presence of its receptor on NK cells were increased.5-ALA can be used to activate NK cells in their killing activity, preventing the growth and metastasis of cancerous cells.


Subject(s)
Aminolevulinic Acid/pharmacology , Killer Cells, Natural/drug effects , NK Cell Lectin-Like Receptor Subfamily K/agonists , Adult , Cell Differentiation/drug effects , Cell Differentiation/immunology , Cells, Cultured , Drug Screening Assays, Antitumor , Gene Expression Regulation/immunology , Healthy Volunteers , Humans , Interferon-gamma/metabolism , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Lymphocyte Activation/drug effects , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Neoplasms/drug therapy , Neoplasms/immunology , Primary Cell Culture
2.
J Hematol Oncol ; 13(1): 74, 2020 06 09.
Article in English | MEDLINE | ID: mdl-32517713

ABSTRACT

BACKGROUND: Melanoma patients who have detectable serum soluble NKG2D ligands either at the baseline or post-treatment of PD1/PDL1 blockade exhibit poor overall survival. Among families of soluble human NKG2D ligands, the soluble human MHC I chain-related molecule (sMIC) was found to be elevated in melanoma patients and mostly associated with poor response to PD1/PDL1 blockade therapy. METHODS: In this study, we aim to investigate whether co-targeting tumor-released sMIC enhances the therapeutic outcome of PD1/PDL1 blockade therapy for melanoma. We implanted sMIC-expressing B16F10 melanoma tumors into syngeneic host and evaluated therapeutic efficacy of anti-sMIC antibody and anti-PDL1 antibody combination therapy in comparison with monotherapy. We analyzed associated effector mechanism. We also assessed sMIC/MIC prevalence in metastatic human melanoma tumors. RESULTS: We found that the combination therapy of the anti-PDL1 antibody with an antibody targeting sMIC significantly improved animal survival as compared to monotherapies and that the effect of combination therapy depends significantly on NK cells. We show that combination therapy significantly increased IL-2Rα (CD25) on NK cells which sensitizes NK cells to low dose IL-2 for survival. We demonstrate that sMIC negatively reprograms gene expression related to NK cell homeostatic survival and proliferation and that antibody clearing sMIC reverses the effect of sMIC and reprograms NK cell for survival. We further show that sMIC/MIC is abundantly present in metastatic human melanoma tumors. CONCLUSIONS: Our findings provide a pre-clinical proof-of-concept and a new mechanistic understanding to underscore the significance of antibody targeting sMIC to improve therapeutic efficacy of anti-PD1/PDL1 antibody for MIC/sMIC+ metastatic melanoma patients.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , B7-H1 Antigen/antagonists & inhibitors , Histocompatibility Antigens Class I/immunology , Immune Checkpoint Inhibitors/therapeutic use , Killer Cells, Natural/cytology , Melanoma, Experimental/drug therapy , NK Cell Lectin-Like Receptor Subfamily K/agonists , Neoplasm Proteins/antagonists & inhibitors , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Animals , Antineoplastic Agents, Immunological/pharmacology , CD8-Positive T-Lymphocytes/immunology , Cell Survival , Drug Synergism , Histocompatibility Antigens Class I/physiology , Homeostasis , Humans , Immune Checkpoint Inhibitors/pharmacology , Interleukin-2 Receptor alpha Subunit/biosynthesis , Interleukin-2 Receptor alpha Subunit/genetics , Ligands , Lung Neoplasms/prevention & control , Lung Neoplasms/secondary , Male , Melanoma, Experimental/immunology , Melanoma, Experimental/secondary , Mice , Mice, Transgenic , Neoplasm Proteins/physiology , Solubility , Specific Pathogen-Free Organisms , Transplantation, Isogeneic , Tumor Microenvironment , Up-Regulation
3.
J Immunother ; 43(6): 175-188, 2020.
Article in English | MEDLINE | ID: mdl-32349046

ABSTRACT

B-cell maturation antigen (BCMA) is a highly plasma cell-selective protein expressed on malignant plasma cells of patients with multiple myeloma (MM), and it is a defined therapeutic target. Major histocompatibility complex class I-related chain A (MICA) is frequently expressed in lymphoproliferative malignancies including MM. MICA activates natural killer (NK) cells and costimulates T cells by interaction with its immunoreceptor NK cell receptor G2D (NKG2D). Nonetheless, during full-blown MM, tumor cells promote efficient MICA shedding, which evokes NKG2D internalization and immune suppression. To enhance the directional killing efficacy of immune cells against myeloma cells, we constructed a novel bispecific antibody 2A9-MICA and explored its potential antimyeloma activity against MM. 2A9-MICA consists of human MICA extracellular region and a single-chain antibody fragment (scFv) that targets BCMA generated by phage display technology. In vitro, 2A9-MICA activated NK cell-mediated cytotoxicity and induced NK cells to kill BCMA-positive human myeloma cells. Moreover, in BCMA-positive, MM-bearing nude mice, 2A9-MICA specifically targeted tumor tissue, where it effectively recruited immune cells and inhibited tumor tissue growth showed superior antitumor activity. Taken together, bispecific antibody 2A9-MICA provides a new approach for MM-targeting immunotherapy and has attractive potential for clinical applications.


Subject(s)
Antibodies, Bispecific/pharmacology , Antineoplastic Agents, Immunological/pharmacology , B-Cell Maturation Antigen/antagonists & inhibitors , Multiple Myeloma/metabolism , NK Cell Lectin-Like Receptor Subfamily K/agonists , Antibodies, Bispecific/isolation & purification , Antibodies, Bispecific/therapeutic use , Antibody Affinity/immunology , Antibody Specificity/immunology , Antineoplastic Agents, Immunological/isolation & purification , Antineoplastic Agents, Immunological/therapeutic use , Cell Degranulation/drug effects , Cell Degranulation/immunology , Cell Line, Tumor , Cytokines/metabolism , Cytotoxicity, Immunologic/immunology , Drug Design , Drug Screening Assays, Antitumor , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Molecular Targeted Therapy , Multiple Myeloma/drug therapy , Multiple Myeloma/etiology , Recombinant Proteins/chemistry , Recombinant Proteins/pharmacology
4.
J Cancer Res Clin Oncol ; 145(5): 1179-1190, 2019 May.
Article in English | MEDLINE | ID: mdl-30778749

ABSTRACT

PURPOSE: Bi-specific antibody (BsAb) is an emerging novel format of antibody. We aimed to develop the natural killer (NK) cell receptor NK group 2, member D (NKG2D)-mediated, immune surveillance system. In this system, the NKG2D ligand MHC class I-related chain A (MICA) was fused with BsAb, which targeted a cluster of differentiation 24 (CD24), a tumor-initiating cell marker that is over-expressed on hepatocellular carcinoma (HCC). METHODS: The Homo MICA extracellular domains (hMICA) were fused to the end of the heavy chain of cG7 with the flexible pentapeptide (Gly-Gly-Gly-Gly-Ser; G4S), which formed the cG7-MICA that was further identified using sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) and western blotting (WB). The targeting specificity was characterized using the Surface Plasmon Resonance (SPR) technology and a flow cytometry assay. Furthermore, the design of BsAb cG7-MICA that targeted CD24 and NKG2D was proven to enhance antibody-dependent, cell-mediated cytotoxicity (ADCC) in vitro by the CytoTox 96 Nonradioactive Cytotoxicity assay. Degranulation and a cytokine production assay of NK cells demonstrated that NK cells were activated effectively by cG7-MICA. Further, in HCC-bearing nude mice, the anti-tumor effects of cG7-MICA combined with sorafenib were verified again. RESULTS: We purified cG7-MICA successfully, and it has a high affinity. In vivo, cG7-MICA recruited NK cells to the tumor site and improved the anti-tumor efficacy of sorafenib. cG7-MICA also activated NK cells to release interferon γ (IFN-γ) and tumor necrosis factor α (TNF-α), and it increased the CD107a expression on the surface of the NK cells in vitro. CONCLUSION: NK cells play a major role in the natural, innate immune system, and they have the function of identifying and killing target cells. cG7-MICA remodels the function of MICA molecules to activate NK cells, which provides a possible strategy for HCC-targeting immunotherapy.


Subject(s)
Antibodies, Bispecific/pharmacology , Antineoplastic Agents, Immunological/pharmacology , CD24 Antigen/antagonists & inhibitors , NK Cell Lectin-Like Receptor Subfamily K/agonists , Neoplasms/immunology , Neoplasms/metabolism , Animals , Antibodies, Bispecific/genetics , Antibody Affinity/immunology , Antibody Specificity/genetics , Antibody-Dependent Cell Cytotoxicity/immunology , Cell Degranulation/immunology , Cell Line, Tumor , Cytokines/metabolism , Disease Models, Animal , Drug Synergism , Genetic Vectors/genetics , Histocompatibility Antigens Class I/metabolism , Humans , Immunoglobulin Fc Fragments/immunology , Immunoglobulin Fc Fragments/metabolism , Immunotherapy , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Leukocytes, Mononuclear , Mice , Neoplasms/drug therapy , Neoplasms/pathology , Receptors, Fc/metabolism , Signal Transduction , Sorafenib/pharmacology , Xenograft Model Antitumor Assays
5.
Front Immunol ; 9: 476, 2018.
Article in English | MEDLINE | ID: mdl-29662484

ABSTRACT

The activating receptor NKG2D is peculiar in its capability to bind to numerous and highly diversified MHC class I-like self-molecules. These ligands are poorly expressed on normal cells but can be induced on damaged, transformed or infected cells, with the final NKG2D ligand expression resulting from multiple levels of regulation. Although redundant molecular mechanisms can converge in the regulation of all NKG2D ligands, different stimuli can induce specific cellular responses, leading to the expression of one or few ligands. A large body of evidence demonstrates that NK cell activation can be triggered by different NKG2D ligands, often expressed on the same cell, suggesting a functional redundancy of these molecules. However, since a number of evasion mechanisms can reduce membrane expression of these molecules both on virus-infected and tumor cells, the co-expression of different ligands and/or the presence of allelic forms of the same ligand guarantee NKG2D activation in various stressful conditions and cell contexts. Noteworthy, NKG2D ligands can differ in their ability to down-modulate NKG2D membrane expression in human NK cells supporting the idea that NKG2D transduces different signals upon binding various ligands. Moreover, whether proteolytically shed and exosome-associated soluble NKG2D ligands share with their membrane-bound counterparts the same ability to induce NKG2D-mediated signaling is still a matter of debate. Here, we will review recent studies on the NKG2D/NKG2D ligand biology to summarize and discuss the redundancy and/or diversity in ligand expression, regulation, and receptor specificity.


Subject(s)
Histocompatibility Antigens Class I/metabolism , Killer Cells, Natural/immunology , NK Cell Lectin-Like Receptor Subfamily K/immunology , Neoplasms/immunology , Virus Diseases/immunology , Animals , Cytotoxicity, Immunologic , Humans , Ligands , NK Cell Lectin-Like Receptor Subfamily K/agonists , NK Cell Lectin-Like Receptor Subfamily K/genetics , Signal Transduction
6.
J Biol Chem ; 292(50): 20472-20480, 2017 12 15.
Article in English | MEDLINE | ID: mdl-28986447

ABSTRACT

Self/non-self-discrimination by the innate immune system relies on germline-encoded, non-rearranging receptors expressed by innate immune cells recognizing conserved pathogen-associated molecular patterns. The natural killer group 2D (NKG2D) receptor is a potent immune-activating receptor that binds human genome-encoded ligands, whose expression is negligible in normal tissues, but increased in stress and disease conditions for reasons that are incompletely understood. Here it is not clear how the immune system reconciles receptor binding of self-proteins with self/non-self-discrimination to avoid autoreactivity. We now report that increased expression of NKG2D ligands after virus infection depends on interferon response factors activated by the detection of viral double-stranded RNA by pattern-recognition receptors (RIG-I/MDA-5) and that NKG2D ligand up-regulation can be blocked by the expression of viral dsRNA-binding proteins. Thus, innate immunity-mediated recognition of viral nucleic acids triggers the infected cell to release interferon for NK cell recruitment and to express NKG2D ligands to become more visible to the immune system. Finally, the observation that NKG2D-ligand induction is a consequence of signaling by pattern-recognition receptors that have been selected over evolutionary time to be highly pathogen-specific explains how the risks of autoreactivity in this system are minimized.


Subject(s)
Gene Expression Regulation , Immunity, Innate , Killer Cells, Natural/metabolism , Lentivirus/physiology , NK Cell Lectin-Like Receptor Subfamily K/agonists , RNA, Viral/metabolism , Amino Acid Substitution , Animals , Cell Line , Cells, Cultured , Cricetinae , DEAD Box Protein 58/chemistry , DEAD Box Protein 58/genetics , DEAD Box Protein 58/metabolism , Gene Expression Regulation, Viral , Genes, Reporter , Humans , Interferon-Induced Helicase, IFIH1/chemistry , Interferon-Induced Helicase, IFIH1/genetics , Interferon-Induced Helicase, IFIH1/metabolism , Killer Cells, Natural/immunology , Killer Cells, Natural/virology , Lentivirus/immunology , Ligands , Mutation , NK Cell Lectin-Like Receptor Subfamily K/genetics , NK Cell Lectin-Like Receptor Subfamily K/metabolism , RNA/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Receptors, Immunologic , Recombinant Proteins/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism
7.
J Immunother ; 40(5): 164-174, 2017 06.
Article in English | MEDLINE | ID: mdl-28452850

ABSTRACT

Multitargeted tyrosine kinase inhibitors (MTKIs) have been shown to combine with natural killer (NK) cell adoptive transfer for the treatment in various cancers. MTKIs sensitize cancer cells to NK cell therapy through upregulation of nature killer group 2 member D ligands (NKG2DLs) on tumor cells. However, the molecular mechanism of MTKIs-mediated upregulation of NKG2DLs is still unknown. In this study, we confirmed sunitinib induced downregulation of its targets, such as vascular endothelial growth factor, platelet-derived growth factor, and c-kit in multiple-drug-resistant nasopharyngeal carcinoma cell line CNE2/DDP and hepatoma cell line HepG2. Then, we further showed sunitinib induced cell proliferation inhibition, apoptosis, and DNA damage in CNE2/DDP and HepG2 cells. Coculture experiments showed that sunitinib-treated CNE2/DDP and HepG2 cells were able to increase the activation and cytotoxicity of NK cells. Quantitative polymerase chain reaction results showed that sunitinib upregulated NKG2DLs, apoptotic genes, DNA damage repair genes, and nuclear factor (NF)-κß family genes. Silencing of NF-κß1, NF-κß2, or RelB (NF-κß pathway) inhibited sunitinib-induced upregulation of NKG2DLs. Taken together, we concluded that sunitinib upregulated NKG2DLs through NF-κß signaling noncanonical pathway which might mediate higher cytotoxic sensitivity of CNE2/DDP and HepG2 cells to NK cells.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/metabolism , Carcinoma/metabolism , Immunologic Surveillance/drug effects , Indoles/pharmacology , Killer Cells, Natural/drug effects , Liver Neoplasms/metabolism , NK Cell Lectin-Like Receptor Subfamily K/biosynthesis , Nasopharyngeal Neoplasms/metabolism , Protein Kinase Inhibitors/pharmacology , Pyrroles/pharmacology , Apoptosis/drug effects , Carcinoma/therapy , Carcinoma, Hepatocellular/therapy , Coculture Techniques , Combined Modality Therapy , Cytotoxicity, Immunologic/drug effects , Gene Expression , Hep G2 Cells , Humans , Immunotherapy, Adoptive , Killer Cells, Natural/immunology , Killer Cells, Natural/transplantation , Ligands , Liver Neoplasms/therapy , NF-kappa B/genetics , NF-kappa B/metabolism , NK Cell Lectin-Like Receptor Subfamily K/agonists , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/therapy , RNA, Small Interfering/genetics , Signal Transduction , Sunitinib
8.
J Immunol ; 198(9): 3662-3670, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28356383

ABSTRACT

NK cells are part of the innate immune system, and are able to identify and kill hazardous cells. The discrimination between normal and hazardous cells is possible due to an array of inhibitory and activating receptors. NKG2D is one of the prominent activating receptors expressed by all human NK cells. This receptor binds stress-induced ligands, including human MICA, MICB, and UL16-binding proteins 1-6. The interaction between NKG2D and its ligands facilitates the elimination of cells under cellular stress, such as tumor transformation. However, the mechanisms regulating the expression of these ligands are still not well understood. Under normal conditions, the NKG2D ligands were shown to be posttranscriptionally regulated by cellular microRNAs and RNA-binding proteins (RBPs). Thus far, only the 3' untranslated regions (UTRs) of MICA, MICB, and UL16-binding protein 2 were shown to be regulated by RBPs and microRNAs, usually resulting in their downregulation. In this study we investigated whether MICB expression is controlled by RBPs through its 5'UTR. We used an RNA pull-down assay followed by mass spectrometry and identified vigilin, a ubiquitously expressed multifunctional RNA-binding protein. We demonstrated that vigilin binds and negatively regulates MICB expression through its 5'UTR. Additionally, vigilin downregulation in target cells led to a significant increase in NK cell activation against said target cells. Taken together, we have discovered a novel mode of MICB regulation.


Subject(s)
Histocompatibility Antigens Class I/metabolism , Immunologic Surveillance , Killer Cells, Natural/immunology , RNA-Binding Proteins/metabolism , Stress, Physiological/immunology , 5' Untranslated Regions/genetics , Cell Line, Tumor , Gene Expression Regulation/genetics , Histocompatibility Antigens Class I/genetics , Humans , Ligands , Lymphocyte Activation , NK Cell Lectin-Like Receptor Subfamily K/agonists , Protein Binding , RNA, Small Interfering/genetics , RNA-Binding Proteins/genetics
9.
Tissue Antigens ; 84(3): 293-303, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24962621

ABSTRACT

The major histocompatibility complex (MHC) class I-related chain A (MICA) and B (MICB) are ligands for the natural killer group 2, member D (NKG2D) activating receptor expressed on natural killer (NK) cells, natural killer T (NKT) cells, CD8+ T cells and γδ T cells. Natural killer group 2, member D (NKG2D) ligand expression is stress-related and upregulated by infected or oncogenic cells leading to cytolysis. MICA and MICB genes display considerable polymorphism among individuals and studies have investigated allelic association with disease and relevance of MICA in transplantation, with variable success. It is now known that promoters of MICA and MICB are polymorphic with some polymorphisms associating with reduced expression. We sequenced International Histocompatibility Workshop (IHW) cell line DNA to determine promoter types and alleles encoded by exons 2-6. We found 8 of 12 known MICA promoter polymorphisms and although promoter P7 dominated, other promoters associated with the same allele. For example, MICA*002:01 had promoters P3, P4 or P7 and the common MICA*008:01/04 type had P1, P6 or P7. Similarly, we sequenced 8 of 12 known MICB promoter haplotypes. Some coding region defined MICB alleles had a single promoter, for example, MICB*002:01 and promoter P9, whereas the promiscuous MICB*005 allele had promoters P1, P2, P5, P6, P10 or P12. The results indicate potential for variation in expression of MICA and MICB ligands between individuals with the same allelic types. If differential expression by polymorphic MICA and MICB promoters is confirmed by functional studies, involvement of these genes in disease susceptibility or adverse transplantation outcomes may require knowledge of both promoter and allelic types to make meaningful conclusions.


Subject(s)
Histocompatibility Antigens Class I/genetics , Killer Cells, Natural/physiology , NK Cell Lectin-Like Receptor Subfamily K/agonists , Promoter Regions, Genetic/genetics , Cytotoxicity, Immunologic , DNA Mutational Analysis , Gene Frequency , Genetic Predisposition to Disease , Haplotypes , Histocompatibility Antigens Class I/metabolism , Humans , Ligands , Polymorphism, Genetic , United Kingdom
10.
Blood ; 123(23): 3585-95, 2014 Jun 05.
Article in English | MEDLINE | ID: mdl-24677544

ABSTRACT

Cytarabine (cytosine arabinoside) is one of the most effective drugs for the treatment of patients diagnosed with acute myeloid leukemia (AML). Despite its efficiency against AML cells, the emergence of drug resistance due to prolonged chemotherapy in most patients is still a major obstacle. Several studies have shown that drug resistance mechanisms alter the sensitivity of leukemia cells to immune system effector cells. To investigate this phenomenon, parental acute myeloid cell lines, HL-60 and KG-1, were continuously exposed to increasing doses of cytarabine in order to establish equivalent resistant cell lines, HL-60(R) and KG-1(R). Our data indicate that cytarabine-resistant cells are more susceptible to natural killer (NK)-mediated cell lysis as compared with parental cytarabine-sensitive cells. The increased susceptibility correlates with the induction of UL-16 binding proteins (ULBP) 1/2/3 and NK group 2, member D (NKG2D) ligands on target cells by a mechanism involving c-Myc induction. More importantly, chromatin immunoprecipitation assay revealed that ULBP1/3 are direct targets of c-Myc. Using drug-resistant primary AML blasts as target cells, inhibition of c-Myc resulted in decreased expression of NKG2D ligands and the subsequent impairment of NK cell lysis. This study provides for the first time, the c-Myc dependent regulation of NKG2D ligands in AML.


Subject(s)
Cytotoxicity, Immunologic/genetics , Intercellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/genetics , Killer Cells, Natural/immunology , Leukemia, Myeloid, Acute/genetics , Proto-Oncogene Proteins c-myc/physiology , Antimetabolites, Antineoplastic/pharmacology , Cell Death/genetics , Cells, Cultured , Cytarabine/pharmacology , Drug Resistance, Neoplasm/genetics , Drug Resistance, Neoplasm/immunology , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Gene Expression Regulation, Leukemic , HL-60 Cells , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Killer Cells, Natural/metabolism , Leukemia, Myeloid, Acute/metabolism , Ligands , NK Cell Lectin-Like Receptor Subfamily K/agonists , NK Cell Lectin-Like Receptor Subfamily K/metabolism
11.
Biol Chem ; 394(10): 1325-31, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23787466

ABSTRACT

Evasion of apoptosis is a hallmark of cancer cells. Inhibitor of apoptosis proteins (IAPs) act as endogenous inhibitors of programmed cell death and are overexpressed in several tumors including Hodgkin lymphoma (HL). Preclinical studies indicate antitumor activity of IAP antagonists and clinical studies in hematological malignancies are underway. Here, we investigate the impact of the small molecule IAP antagonist LCL161 on HL cell lines. Although the antagonist caused rapid degradation of cIAP1 leading to TNFα secretion, LCL161 did not promote apoptosis significantly. However, LCL161 induced expression of MICA and MICB, ligands for the activating immune receptor NKG2D, and enhanced the susceptibility of HL cells to NKG2D-dependent lysis by NK cells. MICA/B upregulation was dependent on activation of the DNA damage response upon LCL161 treatment. Taken together, we demonstrate a novel link between IAP inhibition, DNA damage and immune recognition.


Subject(s)
DNA Damage/immunology , Hodgkin Disease/physiopathology , Immunity, Innate/immunology , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , NK Cell Lectin-Like Receptor Subfamily K/agonists , Thiazoles/pharmacology , Up-Regulation , Blotting, Western , Cell Line, Tumor , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Gene Expression Regulation, Neoplastic , Hodgkin Disease/immunology , Humans , NK Cell Lectin-Like Receptor Subfamily K/genetics , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Tumor Necrosis Factor-alpha/metabolism
12.
J Exp Med ; 208(13): 2633-40, 2011 Dec 19.
Article in English | MEDLINE | ID: mdl-22143889

ABSTRACT

Although carcinoembryonic antigen (CEA)-related cell adhesion molecule 1 (CEACAM1) has been viewed as a tumor suppressor, increasing clinical evidence shows that high levels of CEACAM1 expression on tumors correlates with poor prognosis and high risk of metastasis. Here, we examined the consequences of CEACAM1 expression on tumor cells. We show that tumor cell-associated CEACAM1 causes intracellular retention of various NKG2D ligands in mouse and human tumor cells. CEACAM1-silenced tumor cells expressed more cell surface NKG2D ligands and exhibited greater sensitivity to natural killer cell-mediated cytolysis in vitro and rejection in vivo. Our studies reveal a novel mechanism through which CEACAM1-bearing tumor cells may escape immune-surveillance.


Subject(s)
Antigens, CD/immunology , Carcinoembryonic Antigen/immunology , Cell Adhesion Molecules/immunology , Gene Expression Regulation, Neoplastic/immunology , NK Cell Lectin-Like Receptor Subfamily K/agonists , Neoplasms/immunology , Tumor Escape/immunology , Animals , Antigens, CD/genetics , Carcinoembryonic Antigen/genetics , Cell Adhesion Molecules/genetics , Cell Line, Tumor , Humans , Immunity, Cellular , Killer Cells, Natural/immunology , Killer Cells, Natural/pathology , Ligands , Mice , Mice, Knockout , NK Cell Lectin-Like Receptor Subfamily K/genetics , NK Cell Lectin-Like Receptor Subfamily K/immunology , Neoplasm Metastasis , Neoplasms/pathology
13.
Gastroenterology ; 141(1): 217-26, 226.e1-2, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21600899

ABSTRACT

BACKGROUND & AIMS: The natural killer group 2 member D (NKG2D) is a stimulatory receptor expressed on a subset of mucosal and peripheral CD4+ T cells in patients with Crohn's disease (CD) and other inflammatory diseases. Ligand activation of NKG2D in patients induces CD4+ T cells to release T-helper (Th) 1 cytokines and become cytotoxic. We investigated the Th17 cytokines produced by T cells that express NKG2D in blood and intestinal mucosa samples from patients with CD. METHODS: We isolated CD4+ T cells from peripheral blood and lamina propria samples of patients with CD or ulcerative colitis (UC) and healthy individuals (controls). We analyzed the phenotype and functions of the CD4+NKG2D+ T cells and the cytokines they produce in response to NKG2D stimulation. RESULTS: In patients with CD, CD4+ T cells that express NKG2D produced high levels of interleukin (IL)-17 and IL-22 and expressed high levels of CCR6, the IL-23 receptor, CD161, and RORC (a transcription factor that regulates expression of Th17 cytokines). CD4+ T cells that produced IL-17 expressed high levels of NKG2D and CD161. Costimulation of NKG2D and the T-cell receptor (TCR) significantly increased production of IL-17 and tumor necrosis factor α by CD4+ T cells, compared with activation of only the TCR. CD4+NKG2D+ T cells also responded to Th17 polarization. CONCLUSIONS: NKG2D is a functional marker of CD4+ T cells that produce IL-17 in patients with CD, via costimulation of the TCR and NKG2D. Reagents developed to block NKG2D might reduce gastrointestinal inflammation in patients with CD.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Colitis, Ulcerative/immunology , Crohn Disease/immunology , Cytokines/metabolism , Inflammation Mediators/metabolism , Intestinal Mucosa/immunology , NK Cell Lectin-Like Receptor Subfamily K/agonists , Th17 Cells/immunology , Adolescent , Adult , Animals , Case-Control Studies , Cell Line, Tumor , Colitis, Ulcerative/blood , Colitis, Ulcerative/genetics , Crohn Disease/blood , Crohn Disease/genetics , Cytokines/genetics , Female , Flow Cytometry , France , Humans , Immunophenotyping/methods , Ligands , Male , Mice , Middle Aged , NK Cell Lectin-Like Receptor Subfamily B/immunology , Phenotype , Receptors, Antigen, T-Cell/agonists , Young Adult
14.
Blood ; 117(24): 6571-81, 2011 Jun 16.
Article in English | MEDLINE | ID: mdl-21518928

ABSTRACT

Although innate immune signals shape the activation of naive T cells, it is unclear how innate signals influence effector T-cell function. This study determined the effects of stimulating the NKG2D receptor in conjunction with the TCR on human effector CD8(+) T cells. Stimulation of CD8(+) T cells through CD3 and NKG2D simultaneously or through a chimeric NKG2D receptor, which consists of NKG2D fused to the intracellular region of CD3ζ, activated ß-catenin and increased expression of ß-catenin-induced genes, whereas T cells stimulated through the TCR or a combination of the TCR and CD28 did not. Activation by TCR and NKG2D prevented expression and production of anti-inflammatory cytokines IL-10, IL-9, IL-13, and VEGF-α in a ß-catenin- and PPARγ- dependent manner. NKG2D stimulation also modulated the cytokine secretion of T cells activated simultaneously through CD3 and CD28. These data indicate that activating CD8(+) T cells through the NKG2D receptor along with the TCR modulates signal transduction and the production of anti-inflammatory cytokines. Thus, human effector T cells alter their function depending on which innate receptors are engaged in conjunction with the TCR complex.


Subject(s)
Cytokines/metabolism , NK Cell Lectin-Like Receptor Subfamily K/physiology , T-Lymphocytes, Helper-Inducer/metabolism , Animals , Anti-Inflammatory Agents/metabolism , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/physiology , Cells, Cultured , Cytokines/genetics , Gene Expression Regulation/physiology , Humans , Lymphocyte Activation/genetics , Lymphocyte Activation/physiology , Mice , NK Cell Lectin-Like Receptor Subfamily K/agonists , NK Cell Lectin-Like Receptor Subfamily K/genetics , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Oncogene Protein v-akt/metabolism , Oncogene Protein v-akt/physiology , PPAR gamma/metabolism , PPAR gamma/physiology , U937 Cells , beta Catenin/metabolism , beta Catenin/physiology
15.
Int Immunol ; 20(8): 981-91, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18544572

ABSTRACT

UL16-binding proteins [ULBPs, also termed as retinoic acid early transcripts (RAET1) molecules] are frequently expressed by malignant transformed cells and stimulate anti-tumor immune responses mediated by NKG2D-positive NK cells, CD8(+) alphabeta T cells and gammadelta T cells in vitro and in vivo. In this study, we identified four novel functional splice variants of ULBPs including ULBP4-I, ULBP4-II, ULBP4-III and RAET1G3 in HepG2 liver carcinoma cells, WISH human amnion cells, Hep-2 larynx carcinoma cells and K562 leukemia cells, respectively, by reverse transcription-PCR and T vector cloning strategy. Analysis of alignments of amino acid sequences of the splice variants illustrated that there were important modifications between splice variants and their individual parental ULBP. All ULBP4 splice variants (ULBP4-I, ULBP4-II and ULBP4-III) were type 1 membrane-spanning molecules and had the ability to bind with human NKG2D receptor in vitro. Ectopic expressions of ULBP4 and ULBP4 splice variants resulted in the enhanced cytotoxic sensitivity of target cells against NK cells, which could be blocked by anti-NKG2D mAb. Moreover, co-culture-free soluble forms of ULBP4 splice variants (their alpha1 + alpha2 ectodomains) and RAET1G3 (soluble splice variant of RAET1G2) with NK cells down-regulated the cell surface expression of NKG2D. Finally, immobilized in a plate-bound form of RAET1G3 stimulated NK cells to secrete IFN-gamma. Taken together, all the identified functional splice variants will help to advance our knowledge regarding the overall functions of ULBP gene family.


Subject(s)
Carrier Proteins/genetics , Histocompatibility Antigens Class I/genetics , Membrane Proteins/genetics , NK Cell Lectin-Like Receptor Subfamily K/agonists , Recombinant Proteins/genetics , Amino Acid Sequence/genetics , Carrier Proteins/immunology , Carrier Proteins/metabolism , Cell Line, Tumor , Cytotoxicity, Immunologic/genetics , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , Humans , Interferon-gamma/genetics , Interferon-gamma/metabolism , K562 Cells , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Ligands , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Membrane Proteins/immunology , Membrane Proteins/metabolism , Molecular Sequence Data , NK Cell Lectin-Like Receptor Subfamily K/biosynthesis , NK Cell Lectin-Like Receptor Subfamily K/genetics , NK Cell Lectin-Like Receptor Subfamily K/immunology , Protein Binding , Protein Isoforms/genetics , Protein Isoforms/immunology , Protein Isoforms/metabolism , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Sequence Alignment , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL