Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 84
Filter
1.
Cell Immunol ; 371: 104467, 2022 01.
Article in English | MEDLINE | ID: mdl-34896761

ABSTRACT

OBJECTIVE: To elucidate the role of the functional unknown gene C6orf120 in the pathogenesis of AIH and its mechanism of action, using C6orf120 knockout rats. METHODS: An autoimmune hepatitis model was established with 35 mg/kg intravenous injection of concanavalin A (Con A) in C6orf120-knockout (C6orf120-/-) and wild-type (WT) rats. Rats were sacrificed after administering Con A for 0, 12, and 24 h. The peripheral blood, liver, spleen, and mesenteric lymph nodes were collected for follow-up studies. RESULTS: C6orf120 knockout significantly decreased the serum levels of alanine aminotransferase (ALT) and aspartate aminotransferase (AST) and improved the histological damage in Con A-induced autoimmune liver injury.Loss of C6orf120 function significantly increased the frequency of CD3+ CD161+ NKT cells in the peripheral blood, liver, and spleen; downregulated the expression of CD314 (NKG2D) in the liver, spleen, and mesenteric lymph nodes; reduced the expression of inflammatory cytokines and chemokines; and suppressed the mRNA and protein expression of Fas and FasL in the liver. Additionally, C6orf120 knockout significantly downregulated the expression of p-JAK1, p-JAK2, p-STAT1, and p-STAT3 in liver tissue. CONCLUSION: The protective effect of C6orf120 knockout against Con A-induced hepatitis may be due to the inhibition of NKT cell activation, restriction of cytokine and chemokine activities, inhibition of JAK-STAT and Fas/FasL signaling pathway activation, and reduction in liver inflammation and hepatocyte apoptosis.


Subject(s)
Concanavalin A/toxicity , Glycoproteins/genetics , Hepatitis, Autoimmune/immunology , Hepatitis, Autoimmune/pathology , Natural Killer T-Cells/immunology , Alanine Transaminase/blood , Animals , Aspartate Aminotransferases/blood , Chemical and Drug Induced Liver Injury/prevention & control , Cytokines/analysis , Disease Models, Animal , Fas Ligand Protein/biosynthesis , Fas-Associated Death Domain Protein/biosynthesis , Gene Knockout Techniques , Janus Kinases/biosynthesis , Liver/pathology , Lymph Nodes/pathology , Male , Mice , NK Cell Lectin-Like Receptor Subfamily K/biosynthesis , Rats, Sprague-Dawley , Rats, Transgenic , STAT Transcription Factors/biosynthesis , Spleen/pathology
2.
Clin Exp Immunol ; 206(3): 366-377, 2021 12.
Article in English | MEDLINE | ID: mdl-34562314

ABSTRACT

Hepatocellular carcinoma (HCC) is the world's leading cause of tumor-related mortalities. Natural killer (NK) cells play a critical role at the first immunological defense line against HCC initiation and progression. NK cell dysfunction is therefore an important mechanism for immune evasion of HCC cells. In the present study using a murine HCC model, we revealed the down-regulation of PR/SET Domain 10 (PRDM10) in hepatic NK cells that were phenotypically and functionally exhausted. PRDM10 silencing diminished the expression of natural killer group 2 member D (NKG2D) and tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL), augmented T cell immunoglobulin and ITIM domain (TIGIT) expression, and decreased the expression of interferon (IFN)-γ, perforin and granzyme B in normal hepatic NK cells in vitro. Consistently, PRDM10-deficient NK cells exhibited impaired cytotoxicity on target cells. In contrast, PRDM10 over-expression promoted NKG2D and Fas ligand (FasL) expression, reduced CD96 expression and enhanced transcripts of IFN-γ, perforin and granzyme B in NK cells in vivo. Moreover, PRDM10 silencing and PRDM10 over-expression down-regulated and up-regulated Eomesodermin (Eomes) expression, respectively. In summary, this study reveals PRDM10 down-regulation as a novel mechanism underlying NK cell dysfunction and identifies PRDM10 as a supporting factor of NK cell function.


Subject(s)
Carcinoma, Hepatocellular/pathology , Killer Cells, Natural/immunology , Liver Neoplasms/pathology , Transcription Factors/biosynthesis , Tumor Escape/genetics , Animals , Carcinoma, Hepatocellular/immunology , Cells, Cultured , Disease Models, Animal , Down-Regulation/genetics , Granzymes/biosynthesis , Interferon-gamma/biosynthesis , Liver Neoplasms/immunology , Mice , Mice, Inbred C57BL , NK Cell Lectin-Like Receptor Subfamily K/biosynthesis , Perforin/biosynthesis , T-Box Domain Proteins/biosynthesis , TNF-Related Apoptosis-Inducing Ligand/metabolism , Transcription Factors/genetics , Tumor Escape/immunology
3.
Cancer Immunol Res ; 9(2): 170-183, 2021 02.
Article in English | MEDLINE | ID: mdl-33303573

ABSTRACT

In this study, we explored whether Nutlin-3a, a well-known, nontoxic small-molecule compound antagonizing the inhibitory interaction of MDM2 with the tumor suppressor p53, may restore ligands for natural killer (NK) cell-activating receptors (NK-AR) on neuroblastoma cells to enhance the NK cell-mediated killing. Neuroblastoma cell lines were treated with Nutlin-3a, and the expression of ligands for NKG2D and DNAM-1 NK-ARs and the neuroblastoma susceptibility to NK cells were evaluated. Adoptive transfer of human NK cells in a xenograft neuroblastoma-bearing NSG murine model was assessed. Two data sets of neuroblastoma patients were explored to correlate p53 expression with ligand expression. Luciferase assays and chromatin immunoprecipitation analysis of p53 functional binding on PVR promoter were performed. Primary neuroblastoma cells were also treated with Nutlin-3a, and neuroblastoma spheroids obtained from one high-risk patient were assayed for NK-cell cytotoxicity. We provide evidence showing that the Nutlin-3a-dependent rescue of p53 function in neuroblastoma cells resulted in (i) increased surface expression of ligands for NK-ARs, thus rendering neuroblastoma cell lines significantly more susceptible to NK cell-mediated killing; (ii) shrinkage of human neuroblastoma tumor masses that correlated with overall survival upon adoptive transfer of NK cells in neuroblastoma-bearing mice; (iii) and increased expression of ligands in primary neuroblastoma cells and boosting of NK cell-mediated disaggregation of neuroblastoma spheroids. We also found that p53 was a direct transcription factor regulating the expression of PVR ligand recognized by DNAM-1. Our findings demonstrated an immunomodulatory role of Nutlin-3a, which might be prospectively used for a novel NK cell-based immunotherapy for neuroblastoma.


Subject(s)
Antigens, Differentiation, T-Lymphocyte/immunology , Imidazoles/pharmacology , Killer Cells, Natural/immunology , NK Cell Lectin-Like Receptor Subfamily K/immunology , Neuroblastoma/drug therapy , Piperazines/pharmacology , Tumor Suppressor Protein p53/metabolism , Animals , Antigens, Differentiation, T-Lymphocyte/biosynthesis , Cell Line, Tumor , Cytotoxicity, Immunologic , Female , Humans , Ligands , Mice , Mice, Inbred NOD , NK Cell Lectin-Like Receptor Subfamily K/biosynthesis , Neuroblastoma/immunology , Neuroblastoma/pathology , Receptors, Natural Killer Cell/metabolism , Xenograft Model Antitumor Assays
4.
Pathol Oncol Res ; 26(1): 223-231, 2020 Jan.
Article in English | MEDLINE | ID: mdl-29948616

ABSTRACT

Regional lymph nodes (LN)s represent important immunological barriers in spreading of malignant tumors. However, they are the most frequent early metastatic site in melanoma. Immunomodulatory agents including cytokines have been included in therapy of melanoma and have shown severe side effects and toxicity. In this sense, there is a growing need for bringing these agents to further in vitro testing that may enlighten aspects of their regional application. Therefore, the aim of this study was to investigate the effect of interleukin (IL)-2 and IL-15, the two cytokines with similar immune-enhancing effects, on the expression of activating NKG2D, inhibitory CD158a and CD158b receptors on CD8+ T, NKT-like and NK cell lymphocyte subsets from regional LNs of melanoma patients. In this study, we showed significant effects of IL-2 and IL-15 cytokine treatments on the expression of activating NKG2D and on inhibitory CD158a and CD158b receptors on lymphocytes, CD8+ T, NKT-like and NK cell lymphocyte subsets originating from regional LNs of melanoma patients. Furthermore, IL-2 and IL-15 by inducing the expression of NKG2D activating receptor on innate and on adaptive lymphocyte subsets and by augmenting NK cell antitumor cytotoxicity that correlated with the cytokine-induced NKG2D expression, increased antitumor potential of immune cells in regional LNs of melanoma patients irrespective of LN involvement. These findings indicate the importance of immune cell population from regional LNs of melanoma patients in the development of immune intervention strategies that may if applied locally increase antitumor potential to the level that controls tumor progressions.


Subject(s)
Interleukin-15/pharmacology , Interleukin-2/pharmacology , Lymph Nodes/drug effects , Lymphocyte Subsets/drug effects , Melanoma/immunology , Skin Neoplasms/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Female , Humans , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Lymph Nodes/immunology , Lymphocyte Subsets/immunology , Male , Melanoma/pathology , NK Cell Lectin-Like Receptor Subfamily K/biosynthesis , Natural Killer T-Cells/drug effects , Natural Killer T-Cells/immunology , Receptors, KIR2DL1/biosynthesis , Receptors, KIR2DL3/biosynthesis , Skin Neoplasms/pathology , Melanoma, Cutaneous Malignant
5.
Front Immunol ; 10: 569, 2019.
Article in English | MEDLINE | ID: mdl-30972064

ABSTRACT

The functional plasticity and anti-tumor potential of human γδ T cells have been widely studied. However, the epigenetic regulation of γδ T-cell/tumor cell interactions has been poorly investigated. In the present study, we show that treatment with the histone deacetylase inhibitor Valproic acid (VPA) significantly enhanced the expression and/or release of the NKG2D ligands MICA, MICB and ULBP-2, but not ULBP-1 in the pancreatic carcinoma cell line Panc89 and the prostate carcinoma cell line PC-3. Under in vitro tumor co-culture conditions, the expression of full length and the truncated form of the NKG2D receptor in γδ T cells was significantly downregulated. Furthermore, using a newly established flow cytometry-based method to analyze histone acetylation (H3K9ac) in γδ T cells, we showed constitutive H3K9aclow and inducible H3K9achigh expression in Vδ2 T cells. The detailed analysis of H3K9aclow Vδ2 T cells revealed a significant reversion of TEMRA to TEM phenotype during in vitro co-culture with pancreatic ductal adenocarcinoma cells. Our study uncovers novel mechanisms of how epigenetic modifiers modulate γδ T-cell differentiation during interaction with tumor cells. This information is important when considering combination therapy of VPA with the γδ T-cell-based immunotherapy for the treatment of certain types of cancer.


Subject(s)
Histone Deacetylase Inhibitors/pharmacology , Intraepithelial Lymphocytes/immunology , NK Cell Lectin-Like Receptor Subfamily K/biosynthesis , Receptors, Antigen, T-Cell, gamma-delta/immunology , Valproic Acid/pharmacology , Acetylation , Cell Line, Tumor , GPI-Linked Proteins/metabolism , Histocompatibility Antigens Class I/metabolism , Histones/metabolism , Humans , Immunologic Memory/immunology , Intercellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Lymphocyte Activation/immunology , Male , PC-3 Cells , Pancreatic Neoplasms/immunology , Prostatic Neoplasms/immunology , Pancreatic Neoplasms
6.
Virology ; 510: 9-21, 2017 10.
Article in English | MEDLINE | ID: mdl-28689087

ABSTRACT

In pilot HIV-1 eradication studies, patients' immune responses were ineffective at killing viral reservoirs reactivated through latency reversing agents (LRAs) like suberoylanilide hydroxamic acid (SAHA). We hypothesized that T cells harboring reactivated HIV-1 express MIC and ULBP ligands for the activating NKG2D receptor of natural killer (NK) cells. Here, we demonstrated that MICA/B and ULBP2 are induced by SAHA on primary T cells harboring reactivated virus. Using latently HIV-1-infected J-Lat 6.3/8.4/9.2 and J1.1 cell lines, we showed that SAHA reverts latency and, simultaneously, up-regulates MICA/B and ULBP2 acting at the transcriptional level and through ATR activation, thus sensitizing T cells with reactivated virus to NKG2D-mediated killing by NK cells. Moreover, IL-2 and IL-15 potently boosted NKG2D expression and cytotoxicity of NK cells against SAHA-reactivated p24+ target cells. Therefore, immunotherapy with cytokines enhancing NKG2D-mediated NK-cell cytotoxicity combined with administration of LRAs up-modulating NKG2D ligands, represents a promising approach towards HIV-1 eradication.


Subject(s)
HIV-1/physiology , Histone Deacetylase Inhibitors/metabolism , Hydroxamic Acids/metabolism , Killer Cells, Natural/immunology , NK Cell Lectin-Like Receptor Subfamily K/biosynthesis , Virus Latency/drug effects , Cells, Cultured , GPI-Linked Proteins/analysis , Gene Expression Profiling , HIV Infections/therapy , Histocompatibility Antigens Class I/analysis , Humans , Immunotherapy/methods , Intercellular Signaling Peptides and Proteins/analysis , T-Lymphocytes/drug effects , T-Lymphocytes/virology , Up-Regulation , Vorinostat
7.
Anticancer Res ; 37(8): 4093-4101, 2017 08.
Article in English | MEDLINE | ID: mdl-28739693

ABSTRACT

BACKGROUND/AIM: We investigated the relationship between the expression of natural killer group 2, member D ligands (NKG2DLs) and the antitumor effects of protein-bound polysaccharide-K (PSK). MATERIALS AND METHODS: PSK was administered to evaluate its effectiveness against tumor growth. The expression of Rae-1 and H60 were analyzed in multiple cell lines. RESULTS: PSK showed the highest antitumor effects in mice implanted with cells expressing neither Rae-1 nor H60. PSK had little antitumor effect in mice implanted with cells expressing both Rae-1 and H60. A correlation between the expression of NKG2DLs and the antitumor effect of PSK was observed. After PSK administration, INF-γ production in CD8+ T cells increased in mice with cells expressing neither Rae-1 nor H60, but did not change in mice implanted with cells expressing both Rae-1 and H60. CONCLUSION: We demonstrated that the expression of NKG2DLs affects tumor immunity and the efficacy of immuno therapy in tumor-bearing mouse model.


Subject(s)
Fungal Proteins/administration & dosage , Minor Histocompatibility Antigens/genetics , NK Cell Lectin-Like Receptor Subfamily K/genetics , Neoplasms/drug therapy , Nuclear Matrix-Associated Proteins/genetics , Nucleocytoplasmic Transport Proteins/genetics , Polysaccharides/administration & dosage , Animals , CD8-Positive T-Lymphocytes/drug effects , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Humans , Ligands , Mice , Minor Histocompatibility Antigens/biosynthesis , NK Cell Lectin-Like Receptor Subfamily K/biosynthesis , Neoplasms/genetics , Neoplasms/pathology , Nuclear Matrix-Associated Proteins/biosynthesis , Nucleocytoplasmic Transport Proteins/biosynthesis , Xenograft Model Antitumor Assays
8.
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
9.
Int J Oncol ; 50(5): 1810-1820, 2017 May.
Article in English | MEDLINE | ID: mdl-28339029

ABSTRACT

Hydroxychloroquine (HCQ) is the only autophagy inhibitor in clinical use and it has shown great potential in treating chronic myeloid leukemia (CML). By inhibiting autophagy, HCQ enhances the anti-CML efficiency of chemotherapy. In the present study, we demonstrated that HCQ sensitized CML cells to Vγ9Vδ2 T cell-mediated lysis. HCQ inhibited autophagy in CML cells, but the sensitizing effects of HCQ were autophagy-independent. Since the sensitization was not mimicked by ATG7 knockdown and even occurred in the absence of ATG7. We revealed that in a time-dependent manner HCQ induced the expression of NKG2D ligand ULBP4 on the surface of CML cells. This marks the leukemia cell for recognition by Vγ9Vδ2 T cells. Blocking the interaction of NKG2D with its ligands deleted the sensitizing effects of HCQ. In addition, we showed that HCQ did not affect the synthesis or degradation of ULBP4, but induced the translocation of ULBP4 from the cytoplasm to the cell membrane. Our results uncovered a previously unknown mechanism for HCQ in CML treatment that underlines the ability of HCQ to modulate the immune visibility of CML cells, and pave the way to the development of new combination treatments with HCQ and Vγ9Vδ2 T cells.


Subject(s)
Carrier Proteins/genetics , Histocompatibility Antigens Class I/genetics , Hydroxychloroquine/administration & dosage , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Membrane Proteins/genetics , NK Cell Lectin-Like Receptor Subfamily K/genetics , Autophagy/genetics , Autophagy-Related Protein 7/genetics , Carrier Proteins/biosynthesis , Cell Membrane/genetics , Cytoplasm/genetics , Drug Resistance, Neoplasm , Gene Expression Regulation, Leukemic , Histocompatibility Antigens Class I/biosynthesis , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Membrane Proteins/biosynthesis , NK Cell Lectin-Like Receptor Subfamily K/biosynthesis , Signal Transduction/drug effects , T-Lymphocytes/drug effects , T-Lymphocytes/pathology
10.
Cytokine ; 95: 43-50, 2017 07.
Article in English | MEDLINE | ID: mdl-28235675

ABSTRACT

Interleukin (IL)-15, a key manipulator of T-cell function also modulates B-1a cell activity by augmenting activation markers, turning them towards type 1 polarization and immunoglobulin (Ig) expression which is significant in the context of gut immunity. Here we show, for the first time, IL-15 mediated up-regulation of the activation receptor NKG2D and its adaptor DAP10 in B-1a cells indicating their essential coupling with IL-15 receptor signaling pathway. Our results demonstrate IL-15 treatment increases phosphorylation of STAT5 and p38 leading to translocation of NF-κB onto the nucleus, an attribute that delineates activation of B-1a cells and its role in inflammation. In parallel, increase of anti-apoptotic Bcl-xL suggests its role in long term survival of B-1a cells in culture by IL-15. The cytokine induced overexpression of the plasma cell differentiation transcription factor BLIMP-1 while reducing PAX-5a that could be responsible for the spontaneous Ig secretion by B-1a cells. Up-regulation of IgM transcripts in presence of IL-15 validates mucosal response of the cells through natural Abs to counter pathogens.


Subject(s)
B-Lymphocyte Subsets/immunology , Immunoglobulin M/biosynthesis , Interleukin-15/physiology , NK Cell Lectin-Like Receptor Subfamily K/biosynthesis , Animals , B-Lymphocyte Subsets/metabolism , Cells, Cultured , Female , Gene Expression Regulation , Immunoglobulin M/genetics , Male , Mice, Inbred C57BL , NF-kappa B/metabolism , NK Cell Lectin-Like Receptor Subfamily K/genetics , Positive Regulatory Domain I-Binding Factor 1/metabolism , RNA, Messenger/metabolism , Receptors, Immunologic/biosynthesis , Receptors, Immunologic/genetics , STAT5 Transcription Factor/metabolism , Signal Transduction , Up-Regulation , p38 Mitogen-Activated Protein Kinases/metabolism
11.
PLoS One ; 12(1): e0167640, 2017.
Article in English | MEDLINE | ID: mdl-28076376

ABSTRACT

The role of natural killer (NK) cell function in HIV disease especially in the setting of long-term antiretroviral therapy (ART) and viral suppression is not fully understood. In the current study, we have investigated NK cell activation in healthy controls and aviremic ART-treated HIV+ subjects with different degrees of immune restoration. We performed a cross sectional study in 12 healthy controls and 24 aviremic ART-treated HIV-infected subjects including 13 HIV+ subjects with CD4+ T cells above 500 cells/µL defined as "immunologic responders" and 11 HIV+ subjects with CD4+ T cells below 350 cells/µL defined as "immunologic non-responders". We analyzed NK cell number, subset, and activation by expression of CD107a and NKG2D and co-expression of CD38 and HLA-DR. NK cell-mediated cytotoxicity against uninfected CD4+ T cells was tested in vitro. We found that NK cell absolute number, percentage of NK cells, and percentage of NK cell subsets were similar in the three study groups. The increased NK cell activation was found predominantly in CD56dimCD16+ subset of immunologic non-responders but not immunologic responders compared to healthy controls. The activation of NK cells was inversely correlated with the peripheral CD4+ T cell count in HIV+ subjects, even after controlling for chronic T cell activation, sex, and age, potential contributors for CD4+ T cell counts in HIV disease. Interestingly, NK cells from immunologic non-responders mediated cytotoxicity against uninfected CD4+ T cells ex vivo. NK cells may play a role in blunted CD4+ T cell recovery in ART-treated HIV disease.


Subject(s)
Anti-Retroviral Agents/administration & dosage , CD4-Positive T-Lymphocytes/immunology , HIV Infections/drug therapy , HIV Infections/immunology , HIV-1/immunology , Killer Cells, Natural/immunology , Lymphocyte Activation/drug effects , ADP-ribosyl Cyclase 1/biosynthesis , ADP-ribosyl Cyclase 1/immunology , Adult , Age Factors , CD4 Lymphocyte Count , CD4-Positive T-Lymphocytes/metabolism , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , HIV Infections/blood , Humans , Killer Cells, Natural/metabolism , Lysosomal-Associated Membrane Protein 1/biosynthesis , Lysosomal-Associated Membrane Protein 1/immunology , Male , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/immunology , Middle Aged , NK Cell Lectin-Like Receptor Subfamily K/biosynthesis , NK Cell Lectin-Like Receptor Subfamily K/immunology , Sex Factors
12.
J Asthma ; 54(5): 456-466, 2017 Jun.
Article in English | MEDLINE | ID: mdl-27736259

ABSTRACT

OBJECTIVE: The three main types of killer cells - CD8+ T cells, NK cells and NKT cells - have been linked to asthma and chronic obstructive pulmonary disease (COPD). However, their role in a small subset of asthma patients displaying fixed airway obstruction (FAO), similar to that seen in COPD, has not been explored. The objective of the present study was to investigate killer cell numbers, phenotype and function in peripheral blood from asthma patients with FAO, asthma patients without FAO, and healthy individuals. METHODS: Peripheral CD8+ T cells (CD8+CD3+CD56-), NK cells (CD56+CD3-) and NKT-like cells (CD56+CD3+) of 14 asthma patients with FAO (post-bronchodilator FEV/FVC <0.7, despite clinician-optimised treatment), 7 asthma patients without FAO (post-bronchodilator FEV/FVC ≥ 0.7), and 9 healthy individuals were studied. RESULTS: No significant differences were seen between the number, receptor expression, MAPK signalling molecule expression, cytotoxic mediator expression, and functional cytotoxicity of peripheral killer cells from asthma patients with FAO, asthma patients without FAO and healthy individuals. CONCLUSIONS: Peripheral killer cell numbers or functions do not differentiate between asthma patients with or without fixed airway obstruction.


Subject(s)
Airway Obstruction/immunology , Asthma/immunology , CD8-Positive T-Lymphocytes/metabolism , Killer Cells, Natural/metabolism , Natural Killer T-Cells/metabolism , Aged , Female , Humans , Leukocytes, Mononuclear/metabolism , MAP Kinase Signaling System/immunology , Male , Middle Aged , NK Cell Lectin-Like Receptor Subfamily K/biosynthesis , Protein Array Analysis , Pulmonary Disease, Chronic Obstructive/immunology , Receptors, KIR3DL1/biosynthesis
13.
J Leukoc Biol ; 100(6): 1285-1296, 2016 12.
Article in English | MEDLINE | ID: mdl-27381007

ABSTRACT

Activation of NK cells depends on a balance between activating and inhibitory signals. Class Ia PI3K are heterodimeric proteins with a catalytic and a regulatory subunit and have a central role in cell signaling by associating with tyrosine kinase receptors to trigger signaling cascades. The regulatory p85 subunit participates in signaling through NKG2D, one of the main activating receptors on NK cells, via its interaction with the adaptor protein DAP10. Although the effects of inhibiting catalytic subunits or deleting the regulatory p85α subunit have been studied, little attention has focused on the role of the p85ß subunit in NK cells. Using p85ß knockout mice, we found that p85ß deficiency does not alter NK cell differentiation and maturation in spleen or bone marrow. NK cells from p85ß-/- mice nonetheless produced more IFN-γ and degranulated more effectively when stimulated with anti-NKG2D antibody. These cells also degranulated and killed NKG2D ligand-expressing target cells more efficiently. We show that p85ß deficiency impaired NKG2D internalization, which could contribute to the activated phenotype. Decreasing p85ß subunit protein levels might thus constitute a therapeutic target to promote NK cell activity toward NKG2D ligand-expressing cells.


Subject(s)
Killer Cells, Natural/cytology , Lymphocyte Activation , NK Cell Lectin-Like Receptor Subfamily K/immunology , Phosphatidylinositol 3-Kinases/deficiency , Animals , Bone Marrow/immunology , Cell Degranulation , Cells, Cultured , Down-Regulation , Interferon-gamma/biosynthesis , Lymphopoiesis , MAP Kinase Signaling System , Mice , Mice, Inbred C57BL , Mice, Knockout , NK Cell Lectin-Like Receptor Subfamily K/biosynthesis , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/physiology , Protein Subunits , Receptors, Immunologic/immunology , Specific Pathogen-Free Organisms , Spleen/cytology , Spleen/immunology
14.
Int J Oncol ; 49(2): 763-72, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27279019

ABSTRACT

To elucidate the immunological mechanisms critical for tumor progression, we bred novel mouse strains, different in the NKC and H-2D domains. We used inbreeding to generate hybrids of Balb/c and C57BL/6 of stable H-2Db+d-NK1.1neg and H-2Db-d+NK1.1high phenotypes. We analyzed the growth of three established MHC class I-deficient tumor cell lines: TC-1/A9 tumor (HPV-associated) and B16F10 melanoma, both syngeneic to C57BL/6, and the MCB8 (3-methycholanthrene-induced tumor) syngeneic to Balb/c. Furthermore, we induced colorectal carcinoma by azoxymethane-DSS treatment to test the susceptibility to chemically-induced primary cancer. We found that the novel strains spontaneously regressed the tumor transplants syngeneic to both Balb/c (MCB8) and C57BL/6 (B16F10 and TC-1/A9) mice. The H2-Db+d-NK1.1neg, but not the H2-Db-d+NK1.1high strain was also highly resistant to chemically-induced colorectal cancer in comparison to the parental mice. The immune changes during TC-1/A9 cancer development involved an increase of the NK cell distribution in the peripheral blood and spleen along with higher expression of NKG2D activation antigen; this was in correlation with the time-dependent rise of cytotoxic activity in comparison to C57BL/6 mice. The TC-1/A9 cancer regression was accompanied by higher proportion of B cells in the spleen and B220+/CD86+ activated antigen-presenting B cells distributed in the lymphoid organs, as well as in the periphery. The changes in the T-cell population were represented mainly by the prevalence of T helper cells reflected by grown CD4/CD8 ratio, most prominent in the b+d-NK1.1neg strain. The results of the present study imply usefulness of the two novel mouse strains as an experimental model for further studies of tumor resistance mechanisms.


Subject(s)
Histocompatibility Antigens Class I/immunology , Killer Cells, Natural/immunology , Neoplasms, Experimental/genetics , Animals , Female , Gene Expression Regulation, Neoplastic/genetics , Histocompatibility Antigens Class I/genetics , Humans , Killer Cells, Natural/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , NK Cell Lectin-Like Receptor Subfamily K/biosynthesis , NK Cell Lectin-Like Receptor Subfamily K/immunology , Neoplasms, Experimental/immunology , Neoplasms, Experimental/pathology
15.
J Leukoc Biol ; 100(5): 1181-1189, 2016 11.
Article in English | MEDLINE | ID: mdl-27256566

ABSTRACT

γδ T cells play a critical role in early anti-tumor immunity and perform cytotoxicity via NKG2D for recognition and multiple cytotoxic factors for tumor killing. Recent studies have demonstrated pivotal roles of mTOR-mediated metabolism in the maturation, differentiation, and effector function of diverse immune cells, including DCs, NK cells, CD4+ T cell subsets, and CD8+ T cells, but the role of mTOR signaling in γδ T cells is barely known. Here, we showed that suppressing mTOR signaling in in vitro-expanded Vγ4 γδ T cells via the mechanistic inhibitor rapamycin enhanced their cytotoxicity against multiple tumor cell lines, and these cells performed better tumor-suppressing effects upon adoptive therapy. Further investigation revealed that elevated cytotoxicity was a result of up-regulation of NKG2D and TNF-α. Moreover, rapamycin treatment significantly decreased the expression of CISH and increased pSTAT5. The inhibition of STAT5 pathways via siRNA interference or a specific inhibitor eliminated the up-regulation of NKG2D and TNF-α in rapamycin-treated Vγ4 γδ T cells. These results uncovered an important role of mTOR signaling in the cytotoxic effector function of γδ T cells and provided a potential strategy to improve γδ T cell-based cancer immunotherapy.


Subject(s)
Melanoma, Experimental/immunology , NK Cell Lectin-Like Receptor Subfamily K/biosynthesis , Receptors, Antigen, T-Cell, gamma-delta , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Cytotoxic/immunology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Tumor Necrosis Factor-alpha/biosynthesis , Animals , Cell Line, Tumor , Coculture Techniques , Cytotoxicity, Immunologic , Gene Expression Regulation/drug effects , Immunotherapy, Adoptive , Melanoma, Experimental/pathology , Melanoma, Experimental/therapy , Mice , Mice, Knockout , NK Cell Lectin-Like Receptor Subfamily K/genetics , RNA Interference , RNA, Small Interfering/genetics , Receptors, Antigen, T-Cell, gamma-delta/deficiency , Receptors, Antigen, T-Cell, gamma-delta/genetics , STAT5 Transcription Factor/antagonists & inhibitors , STAT5 Transcription Factor/genetics , STAT5 Transcription Factor/physiology , Sirolimus/pharmacology , Specific Pathogen-Free Organisms , T-Lymphocyte Subsets/drug effects , T-Lymphocytes, Cytotoxic/drug effects , TOR Serine-Threonine Kinases/physiology , Tumor Necrosis Factor-alpha/genetics , Up-Regulation/drug effects
16.
Cancer Biother Radiopharm ; 31(4): 119-24, 2016 May.
Article in English | MEDLINE | ID: mdl-27183434

ABSTRACT

INTRODUCTION: Natural killer (NK) cells are a subset of lymphocytes in humans that release cytokines such as tumor necrosis factor alpha and interferon gamma-γ during infection. NKG2D is one of the most important stimulating NK receptors binding MIC-A, MIC-B, and ULBPs, which leads to activation of NK cells against tumor cells. In this study, the authors evaluated the effect of G2 adjuvant on gene expression and delivery of NKG2D receptor on NK cells in peripheral blood. MATERIALS AND METHODS: Peripheral blood mononuclear cells were isolated from venous blood obtained from healthy volunteers after adding G2 adjuvant within 12, 24, and 48 hours of incubation. Then, total RNA was extracted from the cells, cDNA synthesis was performed, and gene expression was evaluated by real-time PCR. In addition, NK cells were stained with the appropriate monoclonal antibodies, and the receptors expressed on cell surface were quantified. RESULTS: G2 adjuvant leads to upregulation of gene expression and increases the expression of NKG2D receptor on the surface of NK cells after incubation. CONCLUSION: The findings of this study demonstrated that G2 adjuvant can increase NK cell cytotoxicity. It may play an important role in killing tumor cells, preventing tumor growth and metastasis.


Subject(s)
Adjuvants, Immunologic/pharmacology , Killer Cells, Natural/drug effects , NK Cell Lectin-Like Receptor Subfamily K/biosynthesis , Flow Cytometry , Gene Expression/drug effects , Gene Expression/immunology , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , NK Cell Lectin-Like Receptor Subfamily K/blood , NK Cell Lectin-Like Receptor Subfamily K/genetics , Real-Time Polymerase Chain Reaction
17.
Life Sci ; 151: 174-181, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26968781

ABSTRACT

AIMS: Natural killer (NK) cells play critical roles in antitumor immunity. Our previous study showed that over-expression of miR-30c-1* enhanced NKL cell cytotoxicity through up-regulation of tumor necrosis factor-α via directly targeting transcription factor homeobox containing 1. MiR-30c, the complimentary microRNA of miR-30c-1*, has been found to exert regulatory effect on T cell function. However, the effect of miR-30c on NK cells is unknown. Therefore, this study aimed to investigate whether miR-30c could play a role to enhance NK cell activation and cytotoxicity. MAIN METHODS: Chemosynthesis exogenous miR-30c mimics and miR-30c inhibitor were transfected into NKL cells and isolated human peripheral blood NK cells, respectively. The expression levels of NK group 2, member D (NKG2D), CD107a and FasL on cell surface and cytotoxic ability of miRNAs transfected NKL cells against SMMC-7721 cells were evaluated. KEY FINDINGS: MiR-30c could increase the expression of NKG2D and CD107a on NKL cells, and enhance cytotoxic ability of NKL cells to kill SMMC-7721 cells. Moreover, miR-30c could up-regulate the expression of FasL on both NKL cells and human peripheral blood NK cells. However, the peripheral blood NK cells from only four in ten healthy donors appeared high expression levels of NKG2D and CD107a after miR-30c transfection. SIGNIFICANCE: MiR-30c could promote the cytotoxicity of NKL cells in vitro by up-regulating the expression levels of NKG2D, CD107a and FasL. However, the effect of miR-30c on ex vivo NK cells from different human individuals is diverse, indicating that miR-30c may play complicate and fine adjustment in immune system.


Subject(s)
Cytotoxicity, Immunologic/drug effects , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , MicroRNAs/pharmacology , NK Cell Lectin-Like Receptor Subfamily K/biosynthesis , Up-Regulation/drug effects , Cells, Cultured , Fas Ligand Protein/biosynthesis , Humans , Killer Cells, Natural/metabolism , Lysosomal-Associated Membrane Protein 1/biosynthesis
18.
Oncotarget ; 7(3): 2220-8, 2016 Jan 19.
Article in English | MEDLINE | ID: mdl-26745675

ABSTRACT

Immune suppression following major thermal injury directly impacts the recovery potential. Limited data from past reports indicate that natural killer cells might be suppressed due to a putative soluble factor that has remained elusive up to date. Here we comparatively study cohorts of patients with Major and Non-Major Burns as well as healthy donors. MICB and ULBP1 are stress ligands of NKG2D that can be induced by heat stress. Remarkably, serum concentration levels of MICB and ULBP1 are increased by 3-fold and 20-fold, respectively, already within 24h post major thermal injury, and are maintained high for 28 days. In contrast, milder thermal injuries do not similarly enhance the serum levels of MICB and ULBP1. This kinetics coincides with a significant downregulation of NKG2D expression among peripheral blood NK cells. Downregulation of NKG2D by high concentration of soluble MICB occurs in cancer patients and during normal pregnancy due to over production by cancer cells or extravillous trophoblasts, respectively, as an active immune-evasion mechanism. In burn patients this seems an incidental outcome of extensive thermal injury, leading to reduced NKG2D expression. Enhanced susceptibility of these patients to opportunistic viral infections, particularly herpes viruses, could be explained by the reduced NKG2D expression. Further studies are warranted for translation into innovative diagnostic or therapeutic technologies.


Subject(s)
Burns/pathology , Histocompatibility Antigens Class I/blood , Intracellular Signaling Peptides and Proteins/blood , Killer Cells, Natural/immunology , NK Cell Lectin-Like Receptor Subfamily K/immunology , Adult , Burns/blood , Burns/immunology , Female , GPI-Linked Proteins/blood , Humans , Male , Middle Aged , NK Cell Lectin-Like Receptor Subfamily K/biosynthesis , Neoplasms/metabolism , Opportunistic Infections/immunology , Young Adult
19.
Endocrine ; 51(3): 478-89, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26100786

ABSTRACT

Regulatory T (Treg) cells play an important role in the pathogenesis of autoimmune thyroid disorders (AITD). New subsets of CD4(+)CD69(+) and CD4(+)NKG2D(+) T lymphocytes that behave as regulatory cells have been recently reported. The role of these immunoregulatory lymphocytes has not been previously explored in AITD. We analyzed by multi-parametric flow cytometry different Treg cell subsets in peripheral blood from 32 patients with AITD and 19 controls, and in thyroid tissue from seven patients. The suppressive activity was measured by an assay of inhibition of lymphocyte activation. We found a significant increased percentage of CD4(+)CD69(+)IL-10(+), CD4(+)CD69(+)NKG2D(+), and CD4(+)CD69(+)IL-10(+)NKG2D(+) cells, in peripheral blood from GD patients compared to controls. The increase in CD4(+)CD69(+)IL-10(+) and CD4(+)CD69(+)IL-10(+)NKG2D(+) T cells was especially remarkable in patients with active Graves' ophthalmopathy (GO), and a significant positive correlation between GO activity and CD4(+)CD69(+)IL-10(+) or CD4(+)CD69(+)IL-10(+)NKG2D(+) cells was also found. In addition, these cells were increased in patients with a more severe and/or prolonged disease. Thyroid from AITD patients showed an increased proportion of CD69(+) regulatory T cells subpopulations compared to autologous peripheral blood. The presence of CD69(+), NKG2D(+), and IL-10(+) cells was confirmed by immunofluorescence microscopy. In vitro functional assays showed that CD69(+) Treg cells exerted an important suppressive effect on the activation of T effector cells in controls, but not in AITD patients. Our findings suggest that the levels of CD69(+) regulatory lymphocytes are increased in AITD patients, but they are apparently unable to down-modulate the autoimmune response and tissue damage.


Subject(s)
Antigens, CD/biosynthesis , Antigens, Differentiation, T-Lymphocyte/biosynthesis , Interleukin-10/biosynthesis , Lectins, C-Type/biosynthesis , NK Cell Lectin-Like Receptor Subfamily K/biosynthesis , T-Lymphocytes, Regulatory/metabolism , Thyroiditis, Autoimmune/metabolism , Adult , Aged , Cells, Cultured , Female , Goiter/metabolism , Graves Ophthalmopathy/metabolism , Hashimoto Disease/metabolism , Humans , Lymphocyte Activation , Male , Middle Aged , T-Lymphocytes, Regulatory/ultrastructure , Thyroid Gland/metabolism
20.
Tumour Biol ; 37(4): 5455-66, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26563374

ABSTRACT

Cancers constitutively produce and secrete into the blood and other biofluids 30-150 nm-sized endosomal vehicles called exosomes. Cancer-derived exosomes exhibit powerful influence on a variety of biological mechanisms to the benefit of the tumors that produce them. We studied the immunosuppressive ability of epithelial ovarian cancer (EOC) exosomes on two cytotoxic pathways of importance for anticancer immunity-the NKG2D receptor-ligand pathway and the DNAM-1-PVR/nectin-2 pathway. Using exosomes, isolated from EOC tumor explant and EOC cell-line culture supernatants, and ascitic fluid from EOC patients, we studied the expression of NKG2D and DNAM-1 ligands on EOC exosomes and their ability to downregulate the cognate receptors. Our results show that EOC exosomes differentially and constitutively express NKG2D ligands from both MICA/B and ULBP families on their surface, while DNAM-1 ligands are more seldom expressed and not associated with the exosomal membrane surface. Consequently, the NKG2D ligand-bearing EOC exosomes significantly downregulated the NKG2D receptor expression on peripheral blood mononuclear cells (PBMC) while the DNAM-1 receptor was unaffected. The downregulation of NKG2D receptor expression was coupled to inhibition of NKG2D receptor-ligand-mediated degranulation and cytotoxicity measured in vitro with OVCAR-3 and K562 cells as targets. The EOC exosomes acted as a decoy impairing the NKG2D mediated cytotoxicity while the DNAM-1 receptor-ligand system remained unchanged. Taken together, our results support and explain the mechanism behind the recently reported finding that in EOC, NK-cell recognition and killing of tumor cells was mainly dependent on DNAM-1 signaling while the contribution of the NKG2D receptor-ligand pathway was complementary and uncertain.


Subject(s)
Antigens, Differentiation, T-Lymphocyte/genetics , Exosomes/genetics , NK Cell Lectin-Like Receptor Subfamily K/genetics , Neoplasms, Glandular and Epithelial/genetics , Ovarian Neoplasms/genetics , Antigens, Differentiation, T-Lymphocyte/biosynthesis , Carcinoma, Ovarian Epithelial , Exosomes/metabolism , Gene Expression Regulation, Neoplastic , Humans , K562 Cells , Killer Cells, Natural/metabolism , Killer Cells, Natural/pathology , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/pathology , Ligands , NK Cell Lectin-Like Receptor Subfamily K/biosynthesis , Neoplasms, Glandular and Epithelial/pathology , Ovarian Neoplasms/pathology , Signal Transduction/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...