Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 422
Filter
1.
J Immunol ; 212(11): 1819-1828, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38619282

ABSTRACT

NK cells are cytotoxic innate lymphocytes that play a critical role in antitumor immunity. NK cells recognize target cells by using a repertoire of activating NK receptors and exert the effector functions. Although the magnitude of activation signals through activating NK receptors controls NK cell function, it has not been fully understood how these activating signals are modulated in NK cells. In this study, we found that a scaffold protein, THEMIS2, inhibits activating NK receptor signaling. Overexpression of THEMIS2 attenuated the effector function of human NK cells, whereas knockdown of THEMIS2 enhanced it. Mechanistically, THEMIS2 binds to GRB2 and phosphorylated SHP-1 and SHP-2 at the proximity of activating NK receptors DNAM-1 and NKG2D. Knockdown of THEMIS2 in primary human NK cells promoted the effector functions. Furthermore, Themis2-deficient mice showed low metastatic burden in an NK cell-dependent manner. These findings demonstrate that THEMIS2 has an inhibitory role in the antitumor activity of NK cells, suggesting that THEMIS2 might be a potential therapeutic target for NK cell-mediated cancer immunotherapy.


Subject(s)
Killer Cells, Natural , Signal Transduction , Killer Cells, Natural/immunology , Humans , Animals , Mice , Signal Transduction/immunology , Mice, Knockout , NK Cell Lectin-Like Receptor Subfamily K/metabolism , NK Cell Lectin-Like Receptor Subfamily K/immunology , Receptors, Natural Killer Cell/metabolism , Mice, Inbred C57BL , Neoplasms/immunology , Cytotoxicity, Immunologic , Lymphocyte Activation/immunology , Cell Line, Tumor , Receptors, Immunologic
2.
PLoS Comput Biol ; 18(5): e1010114, 2022 05.
Article in English | MEDLINE | ID: mdl-35584138

ABSTRACT

Natural Killer (NK) cells provide key resistance against viral infections and tumors. A diverse set of activating and inhibitory NK cell receptors (NKRs) interact with cognate ligands presented by target host cells, where integration of dueling signals initiated by the ligand-NKR interactions determines NK cell activation or tolerance. Imaging experiments over decades have shown micron and sub-micron scale spatial clustering of activating and inhibitory NKRs. The mechanistic roles of these clusters in affecting downstream signaling and activation are often unclear. To this end, we developed a predictive in silico framework by combining spatially resolved mechanistic agent based modeling, published TIRF imaging data, and parameter estimation to determine mechanisms by which formation and spatial movements of activating NKG2D microclusters affect early time NKG2D signaling kinetics in a human cell line NKL. We show co-clustering of NKG2D and the guanosine nucleotide exchange factor Vav1 in NKG2D microclusters plays a dominant role over ligand (ULBP3) rebinding in increasing production of phospho-Vav1(pVav1), an activation marker of early NKG2D signaling. The in silico model successfully predicts several scenarios of inhibition of NKG2D signaling and time course of NKG2D spatial clustering over a short (~3 min) interval. Modeling shows the presence of a spatial positive feedback relating formation and centripetal movements of NKG2D microclusters, and pVav1 production offers flexibility towards suppression of activating signals by inhibitory KIR ligands organized in inhomogeneous spatial patterns (e.g., a ring). Our in silico framework marks a major improvement in developing spatiotemporal signaling models with quantitatively estimated model parameters using imaging data.


Subject(s)
Killer Cells, Natural , NK Cell Lectin-Like Receptor Subfamily K , Proto-Oncogene Proteins c-vav , Cluster Analysis , Computer Simulation , Humans , Killer Cells, Natural/immunology , Kinetics , Ligands , Lymphocyte Activation , NK Cell Lectin-Like Receptor Subfamily K/immunology , Proto-Oncogene Proteins c-vav/immunology , Signal Transduction/immunology
3.
Sci Rep ; 12(1): 3234, 2022 02 25.
Article in English | MEDLINE | ID: mdl-35217717

ABSTRACT

Leukemic cells proliferate faster than non-transformed counterparts. This requires them to change their metabolism to adapt to their high growth. This change can stress cells and facilitate recognition by immune cells such as cytotoxic lymphocytes, which express the activating receptor Natural Killer G2-D (NKG2D). The tumor suppressor gene p53 regulates cell metabolism, but its role in the expression of metabolism-induced ligands, and subsequent recognition by cytotoxic lymphocytes, is unknown. We show here that dichloroacetate (DCA), which induces oxidative phosphorylation (OXPHOS) in tumor cells, induces the expression of such ligands, e.g. MICA/B, ULBP1 and ICAM-I, by a wtp53-dependent mechanism. Mutant or null p53 have the opposite effect. Conversely, DCA sensitizes only wtp53-expressing cells to cytotoxic lymphocytes, i.e. cytotoxic T lymphocytes and NK cells. In xenograft in vivo models, DCA slows down the growth of tumors with low proliferation. Treatment with DCA, monoclonal antibodies and NK cells also decreased tumors with high proliferation. Treatment of patients with DCA, or a biosimilar drug, could be a clinical option to increase the effectiveness of CAR T cell or allogeneic NK cell therapies.


Subject(s)
Antineoplastic Agents , Leukemia , Tumor Suppressor Protein p53 , Antineoplastic Agents/metabolism , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Leukemia/immunology , Leukemia/metabolism , Ligands , NK Cell Lectin-Like Receptor Subfamily K/immunology , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Tumor Suppressor Protein p53/immunology , Tumor Suppressor Protein p53/metabolism
4.
J Extracell Vesicles ; 11(1): e12176, 2022 01.
Article in English | MEDLINE | ID: mdl-34973063

ABSTRACT

Natural killer (NK) cells are innate cytotoxic lymphocytes that play a key role in cancer immunosurveillance thanks to their ability to recognize and kill cancer cells. NKG2D is an activating receptor that binds to MIC and ULBP molecules typically induced on damaged, transformed or infected cells. The release of NKG2D ligands (NKG2DLs) in the extracellular milieu through protease-mediated cleavage or by extracellular vesicle (EV) secretion allows cancer cells to evade NKG2D-mediated immunosurveillance. In this work, we investigated the immunomodulatory properties of the NKG2D ligand MICA*008 associated to distinct populations of EVs (i.e., small extracellular vesicles [sEVs] and medium size extracellular vesicles [mEVs]). By using as model a human MICA*008-transfected multiple myeloma (MM) cell line, we found that this ligand is present on both vesicle populations. Interestingly, our findings reveal that NKG2D is specifically involved in the uptake of vesicles expressing its cognate ligand. We provide evidence that MICA*008-expressing sEVs and mEVs are able on one hand to activate NK cells but, following prolonged stimulation induce a sustained NKG2D downmodulation leading to impaired NKG2D-mediated functions. Moreover, our findings show that MICA*008 can be transferred by vesicles to NK cells causing fratricide. Focusing on MM as a clinically and biologically relevant model of tumour-NK cell interactions, we found enrichment of EVs expressing MICA in the bone marrow of a cohort of patients. All together our results suggest that the accumulation of NKG2D ligands associated to vesicles in the tumour microenvironment could favour the suppression of NK cell activity either by NKG2D down-modulation or by fratricide of NK cell dressed with EV-derived NKG2D ligands.


Subject(s)
Extracellular Vesicles/immunology , Histocompatibility Antigens Class I/immunology , Immunologic Surveillance , Killer Cells, Natural/immunology , Multiple Myeloma/immunology , Aged , Aged, 80 and over , Bone Marrow/immunology , Cell Death/immunology , Cell Line , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Humans , Immunomodulation , Interferon-gamma/metabolism , Ligands , Male , Middle Aged , NK Cell Lectin-Like Receptor Subfamily K/immunology , Tumor Escape
5.
Int Immunopharmacol ; 104: 108497, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34999394

ABSTRACT

BACKGROUND: Venetoclax, a selective B-cell lymphoma-2 (BCL2) inhibitor, has a potential therapeutic effect when combined with demethylating agents in the first-line setting of unfit elderly patients with acute myeloid leukaemia (AML); however, efficacy is still limited in refractory/recurrent AML. Therefore, exploration of a suitable novel treatment scheme is urgently needed.However, combining venetoclax with NK cell-based immunotherapy has not been studied. METHODS: The cytotoxicity of NK cell combined with venetoclax was assessed in vitro using flow cytometry. Venetoclax-induced natural killer group 2 member D (NKG2D) ligand (NKG2DL) expression was detected by flow cytometry and western blotting. Mechanisms underlying venetoclax-induced NKG2DL expression were found by GSE127200 analysis and investigated using real-time PCR (Q-PCR) and western blotting. RESULTS: Flow cytometric analysis showed that combining venetoclax with NK cells produced synergistic anti-leukaemia effects similar to those of venetoclax + azacitidine. Venetoclax could render AML cell lines and primary AML cells sensitive to NK cell killing by promoting NK cell degranulation, NK-AML cell recognition and NK cell secretion of interferon (IFN)-γ and granzyme B. The synergistic effect resulted from venetoclax-induced NKG2DL upregulation in AML cells and could be undermined by blocking NKG2D on NK cells. This finding suggests that venetoclax enhances NK cell killing activity by activating the NKG2D/NKG2DL ligand-receptor pathway. Furthermore, the nuclear factor-kappa-B (NFKB) signalling pathway was involved in venetoclax-induced NKG2DL upregulation. CONCLUSIONS: Collectively, our data confirm that venetoclax combined with NK cells induces synergistic AML cell cytolysis and preliminarily revealed that venetoclax could selectively induce NKG2DLs on AML cells via NFKB signalling pathway.


Subject(s)
Antineoplastic Agents/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Killer Cells, Natural/drug effects , Leukemia, Myeloid, Acute/immunology , NK Cell Lectin-Like Receptor Subfamily K/immunology , Sulfonamides/pharmacology , Adult , Aged , Cell Line, Tumor , Child , Female , Humans , Killer Cells, Natural/immunology , Leukemia, Myeloid, Acute/genetics , Male , Middle Aged , NK Cell Lectin-Like Receptor Subfamily K/genetics , Young Adult
6.
Front Cell Infect Microbiol ; 12: 1084841, 2022.
Article in English | MEDLINE | ID: mdl-36590594

ABSTRACT

Introduction: Hantaan virus (HTNV) can cause endothelium injury in hemorrhagic fever with renal syndrome (HFRS) patients. Bystander activation of CD8+ T cells by virus infection has been shown that was involved in host injury, but it is unclear during HTNV infection. This project aimed to study the effect of bystander-activated CD8+ T cell responses in HTNV infection. Methods: The in vitro infection model was established to imitate the injury of endothelium in HFRS patients. Flow cytometry was performed to detect the expression of markers of tetramer+ CD8+ T cells and human umbilical vein endothelial cells (HUVECs). The levels of interleukin-15 (IL-15) in serum and supermanant were detected using ELISA kit. The expression of MICA of HUVECs was respectively determined by flow cytometry and western blot. The cytotoxicity of CD8+ T cells was assessed through the cytotoxicity assay and antibody blocking assay. Results: EBV or CMV-specific CD8+ T cells were bystander activated after HTNV infection in HFRS patients. HTNV-infected HUVECs in vitro could produce high levels of IL-15, which was positively correlated with disease severity and the expression of NKG2D on bystander-activated CD8+ T cells. Moreover, the elevated IL-15 could induce activation of CD122 (IL-15Rß)+NKG2D+ EBV/CMV-specific CD8+ T cells. The expression of IL-15Rα and ligand for NKG2D were upregulated on HTNV-infected HUVECs. Bystander-activated CD8+ T cells could exert cytotoxicity effects against HTNV-infected HUVECs, which could be enhanced by IL-15 stimulation and blocked by NKG2D antibody. Discussion: IL-15 induced bystander activation of CD8+ T cells through NKG2D, which may mediate endothelium injury during HTNV infection in HFRS patients.


Subject(s)
Bystander Effect , CD8-Positive T-Lymphocytes , Endothelium , Hemorrhagic Fever with Renal Syndrome , Interleukin-15 , NK Cell Lectin-Like Receptor Subfamily K , Humans , CD8-Positive T-Lymphocytes/immunology , Cytomegalovirus Infections , Endothelium/immunology , Endothelium/injuries , Endothelium/physiopathology , Hantaan virus/immunology , Hemorrhagic Fever with Renal Syndrome/genetics , Hemorrhagic Fever with Renal Syndrome/immunology , Hemorrhagic Fever with Renal Syndrome/virology , Human Umbilical Vein Endothelial Cells , Interleukin-15/genetics , Interleukin-15/immunology , NK Cell Lectin-Like Receptor Subfamily K/genetics , NK Cell Lectin-Like Receptor Subfamily K/immunology , Bystander Effect/immunology
7.
Cell Immunol ; 371: 104454, 2022 01.
Article in English | MEDLINE | ID: mdl-34773897

ABSTRACT

Immune dysregulation is commonly observed in patients with coronavirus disease 2019 (COVID-19). Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) induces severe lung inflammation and innate immune cell dysregulation. However, the precise interaction between SARS-CoV-2 and the innate immune system is currently unknown. To understand the interaction between SARS-CoV-2 and natural killer (NK) cells, several SARS-CoV-2 S protein peptides capable of binding to the NKG2D receptor were screened by in silico analysis. Among them, two peptides, cov1 and cov2, bound to NK cells and NKG2D receptors. These cov peptides increased NK cytotoxicity toward lung cancer cells, stimulated interferon gamma (IFN-γ) production by NK cells, and likely mediated these responses through the phosphorylation of Vav1, a key downstream-signaling molecule of NKG2D and NK activation genes. The direct interaction between SARS-CoV-2 and NK cells is a novel finding, and modulation of this interaction has potential clinical application as a therapeutic target for COVID-19.


Subject(s)
COVID-19/immunology , Killer Cells, Natural/immunology , NK Cell Lectin-Like Receptor Subfamily K/immunology , Peptides/immunology , SARS-CoV-2/immunology , Spike Glycoprotein, Coronavirus/immunology , Amino Acid Sequence , COVID-19/metabolism , COVID-19/virology , Cell Line, Tumor , Cytotoxicity, Immunologic/immunology , Humans , Interferon-gamma/immunology , Interferon-gamma/metabolism , Killer Cells, Natural/metabolism , Lung/immunology , Lung/pathology , Lung/virology , Lymphocyte Activation/immunology , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Peptides/metabolism , Protein Binding , SARS-CoV-2/metabolism , SARS-CoV-2/physiology , Signal Transduction/immunology , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/metabolism
8.
ACS Appl Mater Interfaces ; 13(36): 42370-42381, 2021 Sep 15.
Article in English | MEDLINE | ID: mdl-34486371

ABSTRACT

Natural killer (NK) cells serve as key innate effectors and their activity has been considered a prognostic biomarker in diverse human diseases. Currently, NK cell functional assays have several problems primarily related to adequate preparation, labeling, or treatment of target cells, which are cumbersome and often hamper consistent sensitivity for NK cells. Here, bispecific antibodies (BsAb's) targeting NKG2D and 2B4 receptors, whose combination mounts selective cytotoxicity and IFN-γ production of NK cells, are developed as acellular, consistent, and easy-to-use strategies for assessing NK cell functions. These NK cell activator BsAb's (NKABs) are constructed in symmetric dual bivalent formats with different interdomain spacings [immunoglobulin G (IgG)-single-chain variable fragment (scFv) and dual-variable domain (DVD)-Ig] and kappa constant (Cκ)-scFv format linking two scFv's with a Cκ domain. These NKABs are specific and superior to a combination of monospecific antibodies for NK cell activation. NKAB elicits both direct cytotoxicity and IFN-γ production via integration of NKG2D and 2B4 signals. Moreover, stimulation with NKAB IgG-scFv and Cκ-scFv reveals defective NK cell functions in X-linked lymphoproliferative disease involving 2B4 dysfunction in NK cells and multiple myeloma in peripheral blood mononuclear cells and whole blood, respectively. Hence, this work provides a proof of concept that NKAB facilitates the reliable and comprehensive measurement of NK cell function in clinical settings for diagnostic and prognostic purposes.


Subject(s)
Antibodies, Bispecific/immunology , Killer Cells, Natural/metabolism , Lymphocyte Activation/immunology , NK Cell Lectin-Like Receptor Subfamily K/immunology , Signaling Lymphocytic Activation Molecule Family/immunology , HEK293 Cells , Humans , Interferon-gamma/metabolism , Killer Cells, Natural/immunology , Lymphoproliferative Disorders/diagnosis , Lymphoproliferative Disorders/immunology , Multiple Myeloma/diagnosis , Multiple Myeloma/immunology , Proof of Concept Study , Single-Chain Antibodies/immunology
9.
Cells ; 10(6)2021 06 07.
Article in English | MEDLINE | ID: mdl-34200375

ABSTRACT

Natural Killer (NK) cells are natural cytotoxic, effector cells of the innate immune system. They can recognize transformed or infected cells. NK cells are armed with a set of activating and inhibitory receptors which are able to bind to their ligands on target cells. The right balance between expression and activation of those receptors is fundamental for the proper functionality of NK cells. One of the best known activating receptors is NKG2D, a member of the CD94/NKG2 family. Due to a specific NKG2D binding with its eight different ligands, which are overexpressed in transformed, infected and stressed cells, NK cells are able to recognize and attack their targets. The NKG2D receptor has an enormous significance in various, autoimmune diseases, viral and bacterial infections as well as for transplantation outcomes and complications. This review focuses on the NKG2D receptor, the mechanism of its action, clinical relevance of its gene polymorphisms and a potential application in various clinical settings.


Subject(s)
Bacterial Infections , Immunity, Cellular , Killer Cells, Natural/immunology , NK Cell Lectin-Like Receptor Subfamily K , Organ Transplantation , Polymorphism, Genetic , Virus Diseases , Bacterial Infections/genetics , Bacterial Infections/immunology , Humans , Ligands , NK Cell Lectin-Like Receptor Subfamily K/genetics , NK Cell Lectin-Like Receptor Subfamily K/immunology , Virus Diseases/genetics , Virus Diseases/immunology
10.
Molecules ; 26(13)2021 Jun 28.
Article in English | MEDLINE | ID: mdl-34203519

ABSTRACT

Histone acetylation is an epigenetic mechanism that regulates the expression of various genes, such as natural killer group 2, member D (NKG2D) ligands. These NKG2D ligands are the key molecules that activate immune cells expressing the NKG2D receptor. It has been observed that cancer cells overexpress histone deacetylases (HDACs) and show reduced acetylation of nuclear histones. Furthermore, HDAC inhibitors are known to upregulate the expression of NKG2D ligands. Humans have 18 known HDAC enzymes that are divided into four classes. At present, it is not clear which types of HDAC are involved in the expression of NKG2D ligands. We hypothesized that specific types of HDAC genes might be responsible for altering the expression of NKG2D ligands. In this study, we monitored the expression of NKG2D ligands and major histocompatibility complex (MHC) class I molecules in lung cancer cells which were treated with six selective HDAC inhibitors and specific small interfering RNAs (siRNAs). We observed that treatment with FK228, which is a selective HDAC1/2 inhibitor, also known as Romidepsin, induced NKG2D ligand expression at the transcriptional and proteomic levels in two different lung cancer cell lines. It also caused an increase in the susceptibility of NCI-H23 cells to NK cells. Silencing HDAC1 or HDAC2 using specific siRNAs increased NKG2D ligand expression. In conclusion, it appears that HDAC1 and HDAC2 might be the key molecules regulating the expression of NKG2D ligands. These results imply that specifically inhibiting HDAC1 and HDAC2 could induce the expression of NKG2D ligands and improve the NK cell-mediated anti-cancer immunity.


Subject(s)
Gene Expression Regulation, Neoplastic/immunology , Histone Deacetylase 1/immunology , Histone Deacetylase 2/immunology , Immunity, Cellular/immunology , Lung Neoplasms/immunology , NK Cell Lectin-Like Receptor Subfamily K/immunology , Neoplasm Proteins/immunology , A549 Cells , Histone Deacetylase 1/genetics , Histone Deacetylase 2/genetics , Humans , Killer Cells, Natural , Lung Neoplasms/genetics , Lung Neoplasms/pathology , NK Cell Lectin-Like Receptor Subfamily K/genetics , Neoplasm Proteins/genetics
11.
Mol Immunol ; 137: 94-104, 2021 09.
Article in English | MEDLINE | ID: mdl-34242922

ABSTRACT

The signaling adapter MyD88 is critical for immune cell activation in response to viral or bacterial pathogens via several TLRs, IL-1ßR and IL-18R. However, the essential role of MyD88 during activations mediated by germline-encoded NK cell receptors (NKRs), such as Ly49H or NKG2D, has yet to be investigated. To define the NK cell-intrinsic function of MyD88, we generated a novel NK cell conditional knockout mouse for MyD88 (Myd88fl/flNcr1Cre/+). Phenotypic characterization of these mice demonstrated that MyD88 is dispensable for NK cell development and maturation. However, the MyD88-deficient NK cells exhibited significantly reduced cytotoxic potentials in vivo. In addition, the lack of MyD88 significantly reduced the NKG2D-mediated inflammatory cytokine production in vitro. Consistent with this, mice lacking MyD88 were unable to respond and clear MCMV infection. Transcriptomic analyses of splenic NK cells following MCMV infection revealed that inflammatory gene signatures were upregulated in Ly49H+. In contrast, Ly49H- NK cells have significant enrichment in G2M checkpoint genes, revealing distinct transcriptomic profiles of these subsets. Our results identify a central role for MyD88 in Ly49H-dependent gene signatures, including alterations in genes regulating proliferation in Ly49H+ NK cells. In summary, our study reveals a previously unknown function of MyD88 in Ly49H-dependent signaling and in vivo functions of NK cells.


Subject(s)
Herpesviridae Infections/immunology , Killer Cells, Natural/immunology , Muromegalovirus/immunology , Myeloid Differentiation Factor 88/immunology , Animals , Cell Proliferation/physiology , Cytokines/immunology , Female , Inflammation/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , NK Cell Lectin-Like Receptor Subfamily K/immunology , Receptors, Natural Killer Cell/immunology , Signal Transduction/immunology , Transcriptome/immunology
12.
Front Immunol ; 12: 650788, 2021.
Article in English | MEDLINE | ID: mdl-34220808

ABSTRACT

Numerous studies reported a small subpopulation of TCRαß+CD4-CD8- (double-negative) T cells that exert regulatory functions in the peripheral lymphocyte population. However, the origin of these double-negative T (DNT) cells is controversial. Some researchers reported that DNT cells originated from the thymus, and others argued that these cells are derived from peripheral immune induction. We report a possible mechanism for the induction of nonregulatory CD4+ T cells to become regulatory double-negative T (iDNT) cells in vitro. We found that immature bone marrow dendritic cells (CD86+MHC-II- DCs), rather than mature DCs (CD86+MHC-II+), induced high levels of iDNT cells. The addition of an anti-MHC-II antibody to the CD86+MHC-II+ DC group significantly increased induction. These iDNT cells promoted B cell apoptosis and inhibited B cell proliferation and plasma cell formation. A subgroup of iDNT cells expressed NKG2D. Compared to NKG2D- iDNT cells, NKG2D+ iDNT cells released more granzyme B to enhance B cell regulation. This enhancement may function via NKG2D ligands expressed on B cells following lipopolysaccharide stimulation. These results demonstrate that MHC-II impedes induction, and iDNT cells may be MHC independent. NKG2D expression on iDNT cells enhanced the regulatory function of these cells. Our findings elucidate one possible mechanism of the induction of peripheral immune tolerance and provide a potential treatment for chronic allograft rejection in the future.


Subject(s)
B-Lymphocytes/immunology , CD4 Antigens/immunology , CD8 Antigens/immunology , NK Cell Lectin-Like Receptor Subfamily K/immunology , T-Lymphocytes/immunology , Animals , B7-2 Antigen/genetics , B7-2 Antigen/immunology , B7-2 Antigen/metabolism , CD4 Antigens/genetics , CD4 Antigens/metabolism , CD8 Antigens/genetics , CD8 Antigens/metabolism , Cells, Cultured , Cytokines/genetics , Cytokines/immunology , Cytokines/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Gene Expression/immunology , Granzymes/genetics , Granzymes/immunology , Granzymes/metabolism , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/immunology , Histocompatibility Antigens Class II/metabolism , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Models, Immunological , NK Cell Lectin-Like Receptor Subfamily K/genetics , NK Cell Lectin-Like Receptor Subfamily K/metabolism , T-Lymphocytes/metabolism
13.
Clin Immunol ; 230: 108802, 2021 09.
Article in English | MEDLINE | ID: mdl-34298181

ABSTRACT

Ataxia-telangiectasia (A-T) is a multisystem disorder caused by biallelic pathogenic variants in the gene encoding A-T mutated (ATM) kinase, a master regulator of the DNA damage response (DDR) pathway. Most A-T patients show cellular and/or humoral immunodeficiency that has been associated with cancer risk and reduced survival, but NK cells have not been thoroughly studied. Here we investigated NK cells of A-T patients with a special focus on the NKG2D receptor that triggers cytotoxicity upon engagement by its ligands (NKG2DLs) commonly induced via the DDR pathway on infected, transformed, and variously stressed cells. Using flow cytometry, we examined the phenotype and function of NK cells in 6 A-T patients as compared with healthy individuals. NKG2D expression was evaluated also by western blotting and RT-qPCR; plasma soluble NKG2DLs (sMICA, sMICB, sULBP1, ULBP2) were measured by ELISA. Results showed that A-T NK cells were skewed towards the CD56neg anergic phenotype and displayed decreased expression of NKG2D and perforin. NKG2D was reduced at the protein but not at the mRNA level and resulted in impaired NKG2D-mediated cytotoxicity in 4/6 A-T patients. Moreover, in A-T plasma we found 24-fold and 2-fold increase of sMICA and sULBP1, respectively, both inversely correlated with NKG2D expression. Overall, NK cells are disturbed in A-T patients showing reduced NKG2D expression, possibly caused by persistent engagement of its ligands, that may contribute to susceptibility to cancer and infections and represent novel targets for therapeutic interventions.


Subject(s)
Ataxia Telangiectasia/immunology , Killer Cells, Natural/immunology , NK Cell Lectin-Like Receptor Subfamily K/immunology , Adolescent , Ataxia Telangiectasia/genetics , Ataxia Telangiectasia/metabolism , Case-Control Studies , Cell Line , Child , Cytotoxicity, Immunologic , Down-Regulation , Female , Flow Cytometry , GPI-Linked Proteins/blood , Histocompatibility Antigens Class I/blood , Humans , Intercellular Signaling Peptides and Proteins/blood , Intracellular Signaling Peptides and Proteins/blood , K562 Cells , Killer Cells, Natural/metabolism , Ligands , Male , NK Cell Lectin-Like Receptor Subfamily K/genetics , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Young Adult
14.
J Immunol ; 207(1): 333-343, 2021 07 01.
Article in English | MEDLINE | ID: mdl-34155069

ABSTRACT

Ex vivo expansion followed by reinfusion of tumor-infiltrating leukocytes (TILs) has been used successfully for the treatment of multiple malignancies. Most protocols rely on the use of the cytokine IL-2 to expand TILs prior to reinfusion. In addition, TIL administration relies on systemic administration of IL-2 after reinfusion to support transferred cell survival. The use of IL-2, however, can be problematic because of its preferential expansion of regulatory T and myeloid cells as well as its systemic side effects. In this study, we describe the use of a novel IL-2 mutant retargeted to NKG2D rather than the high-affinity IL-2R for TIL-mediated immunotherapy in a murine model of malignant melanoma. We demonstrate that the NKG2D-retargeted IL-2 (called OMCPmutIL-2) preferentially expands TIL-resident CTLs, such as CD8+ T cells, NK cells, and γδT cells, whereas wild-type IL-2 provides a growth advantage for CD4+Foxp3+ T cells as well as myeloid cells. OMCPmutIL-2-expanded CTLs express higher levels of tumor-homing receptors, such as LFA-1, CD49a, and CXCR3, which correlate with TIL localization to the tumor bed after i.v. injection. Consistent with this, OMCPmutIL-2-expanded TILs provided superior tumor control compared with those expanded in wild-type IL-2. Our data demonstrate that adoptive transfer immunotherapy can be improved by rational retargeting of cytokine signaling to NKG2D-expressing CTLs rather than indiscriminate expansion of all TILs.


Subject(s)
Adoptive Transfer , Interleukin-2/immunology , Leukocytes/immunology , Melanoma/immunology , Melanoma/therapy , NK Cell Lectin-Like Receptor Subfamily K/genetics , Animals , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , NK Cell Lectin-Like Receptor Subfamily K/immunology , Signal Transduction/immunology
15.
Nat Commun ; 12(1): 2715, 2021 05 11.
Article in English | MEDLINE | ID: mdl-33976157

ABSTRACT

Efficient immune responses rely on heterogeneity, which in CD8+ T cells, amongst other mechanisms, is achieved by asymmetric cell division (ACD). Here we find that ageing, known to negatively impact immune responses, impairs ACD in murine CD8+ T cells, and that this phenotype can be rescued by transient mTOR inhibition. Increased ACD rates in mitotic cells from aged mice restore the expansion and memory potential of their cellular progenies. Further characterization of the composition of CD8+ T cells reveals that virtual memory cells (TVM cells), which accumulate during ageing, have a unique proliferation and metabolic profile, and retain their ability to divide asymmetrically, which correlates with increased memory potential. The opposite is observed for naive CD8+ T cells from aged mice. Our data provide evidence on how ACD modulation contributes to long-term survival and function of T cells during ageing, offering new insights into how the immune system adapts to ageing.


Subject(s)
Aging/genetics , Asymmetric Cell Division/genetics , CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/genetics , TOR Serine-Threonine Kinases/genetics , Aging/immunology , Animals , Asymmetric Cell Division/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/drug effects , Cell Proliferation/drug effects , Gene Expression Regulation , Immunity, Innate , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Interleukin-2 Receptor beta Subunit/genetics , Interleukin-2 Receptor beta Subunit/immunology , Lectins, C-Type/genetics , Lectins, C-Type/immunology , Lymphocyte Activation , Mice , NK Cell Lectin-Like Receptor Subfamily K/genetics , NK Cell Lectin-Like Receptor Subfamily K/immunology , Receptors, CXCR3/genetics , Receptors, CXCR3/immunology , Receptors, Immunologic/genetics , Receptors, Immunologic/immunology , Receptors, Interleukin-7/genetics , Receptors, Interleukin-7/immunology , Signal Transduction , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/immunology
16.
Front Immunol ; 12: 532484, 2021.
Article in English | MEDLINE | ID: mdl-33897679

ABSTRACT

Human cytomegalovirus (HCMV) infection often leads to systemic disease in immunodeficient patients and congenitally infected children. Despite its clinical significance, the exact mechanisms contributing to HCMV pathogenesis and clinical outcomes have yet to be determined. One of such mechanisms involves HCMV-mediated NK cell immune response, which favors viral immune evasion by hindering NK cell-mediated cytolysis. This process appears to be dependent on the extent of HCMV genetic variation as high levels of variability in viral genes involved in immune escape have an impact on viral pathogenesis. However, the link between viral genome variations and their functional effects has so far remained elusive. Thus, here we sought to determine whether inter-host genetic variability of HCMV influences its ability to modulate NK cell responses to infection. For this purpose, five HCMV clinical isolates from a previously characterized cohort of pediatric patients with confirmed HCMV congenital infection were evaluated by next-generation sequencing (NGS) for genetic polymorphisms, phylogenetic relationships, and multiple-strain infection. We report variable levels of genetic characteristics among the selected clinical strains, with moderate variations in genome regions associated with modulation of NK cell functions. Remarkably, we show that different HCMV clinical strains differentially modulate the expression of several ligands for the NK cell-activating receptors NKG2D, DNAM-1/CD226, and NKp30. Specifically, the DNAM-1/CD226 ligand PVR/CD155 appears to be predominantly upregulated by fast-replicating ("aggressive") HCMV isolates. On the other hand, the NGK2D ligands ULBP2/5/6 are downregulated regardless of the strain used, while other NK cell ligands (i.e., MICA, MICB, ULBP3, Nectin-2/CD112, and B7-H6) are not significantly modulated. Furthermore, we show that IFN-γ; production by NK cells co-cultured with HCMV-infected fibroblasts is directly proportional to the aggressiveness of the HCMV clinical isolates employed. Interestingly, loss of NK cell-modulating genes directed against NK cell ligands appears to be a common feature among the "aggressive" HCMV strains, which also share several gene variants across their genomes. Overall, even though further studies based on a higher number of patients would offer a more definitive scenario, our findings provide novel mechanistic insights into the impact of HCMV genetic variability on NK cell-mediated immune responses.


Subject(s)
Cytomegalovirus Infections/immunology , Cytomegalovirus/immunology , Intercellular Signaling Peptides and Proteins/immunology , Interferon-gamma/immunology , Killer Cells, Natural/immunology , NK Cell Lectin-Like Receptor Subfamily K/immunology , Cells, Cultured , Cytomegalovirus/genetics , Cytomegalovirus/physiology , Cytomegalovirus Infections/metabolism , Cytomegalovirus Infections/virology , Cytotoxicity, Immunologic/genetics , Cytotoxicity, Immunologic/immunology , GPI-Linked Proteins/genetics , GPI-Linked Proteins/immunology , GPI-Linked Proteins/metabolism , Gene Expression , Genetic Variation , High-Throughput Nucleotide Sequencing/methods , Humans , Immune Evasion/genetics , Immune Evasion/immunology , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Interferon-gamma/metabolism , Killer Cells, Natural/metabolism , Killer Cells, Natural/virology , Ligands , Male , NK Cell Lectin-Like Receptor Subfamily K/genetics , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Reverse Transcriptase Polymerase Chain Reaction
17.
J Vis Exp ; (168)2021 02 21.
Article in English | MEDLINE | ID: mdl-33682851

ABSTRACT

Within the same patient, absence of NKG2D ligands (NKG2DL) surface expression was shown to distinguish leukemic subpopulations with stem cell properties (so called leukemic stem cells, LSCs) from more differentiated counterpart leukemic cells that lack disease initiation potential although they carry similar leukemia specific genetic mutations. NKG2DL are biochemically highly diverse MHC class I-like self-molecules. Healthy cells in homeostatic conditions generally do not express NKG2DL on the cell surface. Instead, expression of these ligands is induced upon exposure to cellular stress (e.g., oncogenic transformation or infectious stimuli) to trigger elimination of damaged cells via lysis through NKG2D-receptor-expressing immune cells such as natural killer (NK) cells. Interestingly, NKG2DL surface expression is selectively suppressed in LSC subpopulations, allowing these cells to evade NKG2D-mediated immune surveillance. Here, we present a side-by-side analysis of two different flow cytometry methods that allow the investigation of NKG2DL surface expression on cancer cells i.e., a method involving pan-ligand recognition and a method involving staining with multiple antibodies against single ligands. These methods can be used to separate viable NKG2DL negative cellular subpopulations with putative cancer stem cell properties from NKG2DL positive non-LSC.


Subject(s)
Flow Cytometry/methods , Leukemia, Myeloid, Acute/pathology , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Antibodies, Neoplasm/metabolism , Biotinylation , Cell Count , Humans , Ligands , NK Cell Lectin-Like Receptor Subfamily K/genetics , NK Cell Lectin-Like Receptor Subfamily K/immunology , Recombinant Fusion Proteins/metabolism , Staining and Labeling , Tumor Cells, Cultured
18.
BMC Cancer ; 21(1): 126, 2021 Feb 06.
Article in English | MEDLINE | ID: mdl-33549054

ABSTRACT

BACKGROUND: Breast cancer is one of the most frequently diagnosed cancers among women worldwide. Alterations in the tumor microenvironment (TME) have been increasingly recognized as key in the development and progression of breast cancer in recent years. To deeply comprehend the gene expression profiling of the TME and identify immunological targets, as well as determine the relationship between gene expression and different prognoses is highly critical. METHODS: The stromal/immune scores of breast cancer patients from The Cancer Genome Atlas (TCGA) were employed to comprehensively evaluate the TME. Then, TME characteristics were assessed, overlapping genes of the top 3 Gene Ontology (GO) terms and upregulated differentially expressed genes (DEGs) were analyzed. Finally, through combined analyses of overall survival, time-dependent receiver operating characteristic (ROC), and protein-protein interaction (PPI) network, novel immune related genes with good prognosis were screened and validated in both TCGA and GEO database. RESULTS: Although the TME did not correlate with the stages of breast cancer, it was closely associated with the subtypes of breast cancer and gene mutations (CDH1, TP53 and PTEN), and had immunological characteristics. Based on GO functional enrichment analysis, the upregulated genes from the high vs low immune score groups were mainly involved in T cell activation, the external side of the plasma membrane, and receptor ligand activity. The top GO terms of the upregulated DEGs from the high vs low immune score groups exhibited better prognosis in breast cancer; 15 of them were related to good prognosis in breast cancer, especially CD226 and KLRC4-KLRK1. CONCLUSIONS: High CD226 and KLRC4-KLRK1 expression levels were identified and validated to correlate with better overall survival in specific stages or subtypes of breast cancer. CD226, KLRC4-KLRK1 and other new targets seem to be promising avenues for promoting antitumor targeted immunotherapy in breast cancer.


Subject(s)
Antigens, Differentiation, T-Lymphocyte/genetics , Breast Neoplasms/genetics , Gene Expression Profiling , NK Cell Lectin-Like Receptor Subfamily C/genetics , NK Cell Lectin-Like Receptor Subfamily K/genetics , Tumor Microenvironment/genetics , Antigens, CD/genetics , Antigens, Differentiation, T-Lymphocyte/immunology , Breast Neoplasms/immunology , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Cadherins/genetics , Cell Membrane/genetics , Cell Membrane/immunology , Databases, Genetic , Female , Genes, p53 , Humans , Lymphocyte Activation , Mutation , NK Cell Lectin-Like Receptor Subfamily C/immunology , NK Cell Lectin-Like Receptor Subfamily K/immunology , PTEN Phosphohydrolase/genetics , Prognosis , Protein Interaction Maps/genetics , Protein Interaction Maps/immunology , ROC Curve , Stromal Cells/pathology , T-Lymphocytes/immunology , Tumor Microenvironment/immunology , Up-Regulation
19.
J Cancer Res Clin Oncol ; 147(3): 713-723, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33392659

ABSTRACT

BACKGROUND: Nasopharyngeal carcinoma (NPC) is a rare epithelial carcinoma arising from the nasopharyngeal region. The pathogenesis of NPC is linked to Epstein-Barr virus (EBV) infection, although genetics and lifestyle factors appears to be also implicated. NKG2D is an immunoreceptor expressed by NK and T-cell subsets that recognizes MICA protein and other ligands on tumor cells. NKG2D interaction with MICA plays a role in the immunosurveillance to viruses and cancer. METHODS: We investigated potential associations between functional polymorphisms in NKG2D and MICA genes with NPC susceptibility. We conducted a case-control study including 255 Vietnamese patients with EBV + non-differentiated NPC and 220 healthy controls. RESULTS: We observed a significant association between the LNK/LNK genotype of rs1049174 (a variant associated with lower NKG2D receptor expression and reduced NK cell cytotoxicity) and increased susceptibility to NPC (adjusted OR = 1.66, 95% CI 1.07-2.59; p = 0.024). Similarly, the AA genotype of MICA rs2596542 was significantly associated with NPC (adjusted OR = 2.12; 95% CI 1.22-3.81; p = 0.009). In addition, tumor specimens of NPC patients with the AA genotype displayed a higher expression level of MICA proteins and showed higher EBV titers compared with tumor tissues from patients with the GG or GA genotypes. Higher EBV copy numbers were also observed in tumors with the A allele of MICA rs1051792 (also known as MICA-129 Met/Val) compared with those with the G allele; however, MICA rs1051792 variants were not associated with NPC susceptibility. These results suggest that genetic variants in components of the NKG2D axis may influence the individual susceptibility to EBV-induced NPC.


Subject(s)
Epstein-Barr Virus Infections/genetics , NK Cell Lectin-Like Receptor Subfamily K/genetics , Nasopharyngeal Carcinoma/virology , Nasopharyngeal Neoplasms/virology , Adult , Case-Control Studies , DNA, Viral/analysis , Epstein-Barr Virus Infections/immunology , Epstein-Barr Virus Infections/pathology , Female , Genetic Predisposition to Disease , Genetic Variation , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/isolation & purification , Histocompatibility Antigens Class I/biosynthesis , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Humans , Immunohistochemistry , Killer Cells, Natural/immunology , Male , Middle Aged , NK Cell Lectin-Like Receptor Subfamily K/immunology , Nasopharyngeal Carcinoma/genetics , Nasopharyngeal Carcinoma/immunology , Nasopharyngeal Carcinoma/pathology , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/immunology , Nasopharyngeal Neoplasms/pathology , Polymorphism, Single Nucleotide
20.
Protein Expr Purif ; 178: 105783, 2021 02.
Article in English | MEDLINE | ID: mdl-33122138

ABSTRACT

Natural killer (NK) cells are potent cytotoxic effector cells of the innate immune system and play an important role in tumor immunosurveillance and control. NKG2D is an activating receptor of NK cells. The NKG2D receptor-ligand system has contributed to immune cells recognizing tumor cells and the tumor microenvironment. In order to stretch the application of NK cells on adoptive immunotherapy for B-cell malignancies, we designed and produced a novel bispecific ULBP1×CD19-scFv fusion protein, in which the extracellular domain of NKG2D ligand ULBP1 was fused to a single chain variable fragment (scFv) of anti-CD19. The vector expressing ULBP1×CD19-scFv protein was constructed and expressed in Pichia pastoris. Effects of medium composition, concentration of methanol as the inducer, induction time and broth content in shake flask on the expression of the recombinant protein were investigated. The results showed that the optimized conditions for ULBP1×CD19-scFv expression were 1% methanol induction for 96 h with 15% broth content. The secreted recombinant protein was purified using ammonium sulfate fractionation and Ni-NTA affinity chromatography and the purity is about 93%. The cytotoxicity of NK92-MI cells against CD19+ Raji cells was enhanced in the presence of purified ULBP1×CD19-scFv protein. These results indicated that ULBP1 could be used as an activating element of bispecific killer engagers (BiKEs) and Pichia pastoris yeast might be an alternative expression host for BiKEs production.


Subject(s)
Antibodies, Bispecific , Antigens, CD19/immunology , Killer Cells, Natural/immunology , Lymphocyte Activation , NK Cell Lectin-Like Receptor Subfamily K , Single-Chain Antibodies , Antibodies, Bispecific/chemistry , Antibodies, Bispecific/immunology , Antibodies, Bispecific/isolation & purification , Hep G2 Cells , Humans , NK Cell Lectin-Like Receptor Subfamily K/antagonists & inhibitors , NK Cell Lectin-Like Receptor Subfamily K/immunology , Recombinant Fusion Proteins , Single-Chain Antibodies/chemistry , Single-Chain Antibodies/immunology , Single-Chain Antibodies/isolation & purification
SELECTION OF CITATIONS
SEARCH DETAIL
...