Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Int J Oncol ; 50(1): 66-74, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27878235

ABSTRACT

Asbestos exposure causes malignant tumors such as lung cancer and malignant mesothelioma. Based on our hypothesis in which continuous exposure to asbestos of immune cells cause reduction of antitumor immunity, the decrease of natural killer cell killing activity with reduction of NKp46 activating receptor expression, inhibition of cytotoxic T cell clonal expansion, reduced CXCR3 chemokine receptor expression and production of interferon-γ production in CD4+ T cells were reported using cell line models, freshly isolated peripheral blood immune cells from health donors as well as asbestos exposed patients such as pleural plaque and mesothelioma. In addition to these findings, regulatory T cells (Treg) showed enhanced function through cell-cell contact and increased secretion of typical soluble factors, interleukin (IL)-10 and transforming growth factor (TGF)-ß, in a cell line model using the MT-2 human polyclonal T cells and its sublines exposed continuously to asbestos fibers. Since these sublines showed a remarkable reduction of FoxO1 transcription factor, which regulates various cell cycle regulators in asbestos-exposed sublines, the cell cycle progression in these sublines was examined and compared with that of the original MT-2 cells. Results showed that cyclin D1 expression was markedly enhanced, and various cyclin-dependent kinase-inhibitors were reduced with increased S phases in the sublines. Furthermore, the increase of cyclin D1 expression was regulated by FoxO1. The overall findings indicate that antitumor immunity in asbestos-exposed individuals may be reduced in Treg through changes in the function and volume of Treg.


Subject(s)
Cyclin D1/immunology , Forkhead Box Protein O1/biosynthesis , Lung Neoplasms/immunology , Mesothelioma/immunology , T-Lymphocytes, Regulatory/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , Cell Cycle/drug effects , Cell Cycle/immunology , Cyclin D1/biosynthesis , Cyclin D1/blood , Forkhead Box Protein O1/immunology , Gene Expression Regulation, Neoplastic , Humans , Interleukin-10/biosynthesis , Interleukin-10/blood , Interleukin-10/immunology , Killer Cells, Natural/immunology , Lung Neoplasms/blood , Lung Neoplasms/chemically induced , Lung Neoplasms/pathology , Mesothelioma/blood , Mesothelioma/chemically induced , Mesothelioma/pathology , Mesothelioma, Malignant , Natural Cytotoxicity Triggering Receptor 1/biosynthesis , Natural Cytotoxicity Triggering Receptor 1/blood , Natural Cytotoxicity Triggering Receptor 1/immunology , Receptors, CXCR3/biosynthesis , Receptors, CXCR3/immunology , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Regulatory/drug effects , Transforming Growth Factor beta/biosynthesis , Transforming Growth Factor beta/blood , Transforming Growth Factor beta/immunology
2.
Vet Immunol Immunopathol ; 169: 79-84, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26827843

ABSTRACT

The amount, distribution and phenotype of ovine NCR1+ cells were investigated during developing GALT from day 70 of gestation. Antibodies against CD3 and CD79 were used to identify the compartments of GALT, and the localization of NCR1+ cells were correlated within these structures. Markers CD34 and c-kit, in addition to Ki67, were used to investigate possible origin and the stage of development of the NCR1+ cells. NCR1+ cells were present as single cells in the subepithelial tissue as early as 70 days of gestation, and were predominantly present in the T cell rich IFAs and domes as these intestinal wall compartments developed. While NCR1+ cells proliferated more intensively at mid-gestation (70-104 days), the number of NCR1+ cells also expressing c-kit, increased at the end of gestation. In conclusion, NCR1+ cells appeared early in T cell areas of the gut and displayed a phenotype consistent with intermediate stages of cNK cells and/or a subpopulation of ILC22.


Subject(s)
Intestines/embryology , Lymphoid Tissue/embryology , Natural Cytotoxicity Triggering Receptor 1/biosynthesis , Proto-Oncogene Proteins c-kit/biosynthesis , Animals , Intestinal Mucosa/cytology , Intestinal Mucosa/embryology , Intestines/cytology , Lymphocyte Subsets/cytology , Lymphocyte Subsets/metabolism , Lymphocytes/cytology , Lymphocytes/metabolism , Lymphoid Tissue/cytology , Phenotype , Sheep
3.
Oncotarget ; 6(40): 42569-74, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26544512

ABSTRACT

In a phase IV trial, eighty-four patients (age 18-79) with acute myeloid leukemia (AML) in first complete remission (CR) received cycles of immunotherapy with histamine dihydrochloride (HDC) and low-dose human recombinant interleukin-2 (IL-2) to prevent relapse in the post-consolidation phase. Aspects of natural killer (NK) cell biology were analyzed before and during immunotherapy with focus on outcome in older patients. In younger (<60 years old, n = 37) and older patients (>60 years old, n = 47), treatment with HDC/IL-2 resulted in an expansion of CD56(bright) and CD16+ NK cells in blood along with an increased NK cell expression of the natural cytotoxicity receptors (NCR) NKp30 and NKp46. In older patients, a high expression of NKp30 or NKp46 on CD16+ NK cells before and during therapy predicted leukemia-free and overall survival. These results suggest that NK cell functions determine relapse risk and survival in older AML patients and point to biomarkers of efficacy in protocols for remission maintenance.


Subject(s)
Immunotherapy/methods , Killer Cells, Natural/immunology , Leukemia, Myeloid, Acute/immunology , Lymphocyte Subsets/immunology , Adolescent , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , CD56 Antigen/biosynthesis , CD56 Antigen/immunology , Female , Flow Cytometry , GPI-Linked Proteins/biosynthesis , GPI-Linked Proteins/immunology , Histamine/administration & dosage , Humans , Interleukin-2/administration & dosage , Interleukin-2/analogs & derivatives , Kaplan-Meier Estimate , Leukemia, Myeloid, Acute/mortality , Leukemia, Myeloid, Acute/therapy , Maintenance Chemotherapy/methods , Male , Middle Aged , Natural Cytotoxicity Triggering Receptor 1/biosynthesis , Natural Cytotoxicity Triggering Receptor 1/immunology , Natural Cytotoxicity Triggering Receptor 3/biosynthesis , Natural Cytotoxicity Triggering Receptor 3/immunology , Neoplasm Recurrence, Local/immunology , Receptors, IgG/biosynthesis , Receptors, IgG/immunology , Recombinant Proteins/administration & dosage , Remission Induction , Young Adult
4.
Clin Lab ; 61(9): 1221-9, 2015.
Article in English | MEDLINE | ID: mdl-26554241

ABSTRACT

BACKGROUND: Severe aplastic anemia (SAA) is a kind of bone marrow failure caused by complex pathogenesis, mainly characterized by severe pancytopenia which causes anemia, hemorrhage, and infection. Natural killer (NK) cells, derived from hematopoietic stem cells (HSCs) or common lymphoid progenitors (CLP), play an important role in the innate immunity and adaptive immune responses. Of the receptors on NK cells, the NKp46/NCR1 is considered to be an important activating receptor for NK cells. However, the quantity and function of NKp46/NCR1 remains unknown. METHODS: The quantity of NKp46/NCR1 on NK cells in patients with SAA before and after immunosuppressive therapy (IST) was investigated by flow cytometry, quantitative real-time PCR, and western blot. After knockdown of the NKp46/NCR1 gene, NK cells were cultured with K562 cells to detect the function of NK cells. RESULTS: The results showed that the expression of NKp46/NCR1 in NK cells was significantly higher in untreated SAA patients than those in remission SAA and controls by FCM, qRT-PCR, and WB. After co-culturing with NK cells knockdown with siRNA-NKp46/NCR1, the apoptosis rate of K562 cells was significantly lower compared with the siRNA-scr group and control groups (7.08 ± 5.23% vs. 11.31 ± 7.20% and 10.30 ± 6.08%, p < 0.05). CONCLUSIONS: We concluded that the decrease of total NK cells and the higher expressions of NKp46/NCR1 on them may be the reason for the hyperfunction of the immune system in SAA patients.


Subject(s)
Anemia, Aplastic/immunology , Killer Cells, Natural/immunology , Natural Cytotoxicity Triggering Receptor 1/biosynthesis , Adolescent , Adult , Anemia, Aplastic/drug therapy , Anemia, Aplastic/metabolism , Apoptosis , Coculture Techniques , Female , Gene Expression Regulation/immunology , Gene Knockdown Techniques , Humans , Immunosuppressive Agents/therapeutic use , K562 Cells , Killer Cells, Natural/metabolism , Male , Middle Aged , Natural Cytotoxicity Triggering Receptor 1/antagonists & inhibitors , Natural Cytotoxicity Triggering Receptor 1/genetics , RNA Interference , RNA, Small Interfering/pharmacology , Young Adult
5.
J Cutan Pathol ; 42(3): 199-205, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25407699

ABSTRACT

Primary cutaneous aggressive epidermotropic T-cell lymphoma (PCAETCL) is a very rare lymphoma characterized by rapidly growing necrotic cutaneous lesions with an epidermotropic CD8+ T-cell neoplastic infiltrate observed histopathologically. It is associated with a very poor outcome, despite aggressive multi-agent chemotherapy. We report a 49-year-old human immunodeficiency virus (HIV)-infected patient who developed PCAETCL with associated marked vascular injury leading to diffuse purpuric and necrotic lesions complicated by recalcitrant hemophagocytic activation syndrome. The lymphoma strongly and diffusely expressed CD158k/KIR3DL2 at the protein and transcript level and NKp46 transcripts, in addition to CD8 and cytotoxic proteins. We observed a diffuse CD158k/KIR3DL2 protein expression in another case of PAETCL, not associated with immunodeficiency, which was used as a positive control. PCAETCL can develop in HIV-infected patients and may present in vasculitis-like fashion. The possible role of immunosuppression and/or HIV in oncogenesis can be postulated, as patients infected with HIV may develop anti-HIV cytotoxic CD8+ lymphoproliferations. The frequency of CD158k/KIR3DL2 and NKp46 expression in PCAECL remains to be studied in a series of cases, and may represent interesting targets for future treatments.


Subject(s)
CD8-Positive T-Lymphocytes/pathology , Carrier State/pathology , HIV Infections/pathology , HIV/isolation & purification , Lymphoma, T-Cell, Cutaneous/pathology , Natural Cytotoxicity Triggering Receptor 1/biosynthesis , Receptors, KIR3DL2/biosynthesis , Biopsy , Carrier State/virology , HIV Infections/genetics , Humans , Immunohistochemistry , Lymphoma, T-Cell, Cutaneous/virology , Male , Middle Aged , Prognosis , Skin Neoplasms/pathology , Skin Neoplasms/virology
6.
Immunity ; 41(5): 776-88, 2014 Nov 20.
Article in English | MEDLINE | ID: mdl-25456160

ABSTRACT

Interleukin-22 (IL-22) plays a critical role in mucosal defense, although the molecular mechanisms that ensure IL-22 tissue distribution remain poorly understood. We show that the CXCL16-CXCR6 chemokine-chemokine receptor axis regulated group 3 innate lymphoid cell (ILC3) diversity and function. CXCL16 was constitutively expressed by CX3CR1(+) intestinal dendritic cells (DCs) and coexpressed with IL-23 after Citrobacter rodentium infection. Intestinal ILC3s expressed CXCR6 and its ablation generated a selective loss of the NKp46(+) ILC3 subset, a depletion of intestinal IL-22, and the inability to control C. rodentium infection. CD4(+) ILC3s were unaffected by CXCR6 deficiency and remained clustered within lymphoid follicles. In contrast, the lamina propria of Cxcr6(-/-) mice was devoid of ILC3s. The loss of ILC3-dependent IL-22 epithelial stimulation reduced antimicrobial peptide expression that explained the sensitivity of Cxcr6(-/-) mice to C. rodentium. Our results delineate a critical CXCL16-CXCR6 crosstalk that coordinates the intestinal topography of IL-22 secretion required for mucosal defense.


Subject(s)
Chemokine CXCL6/immunology , Enterobacteriaceae Infections/immunology , Interleukins/immunology , Mucous Membrane/immunology , Receptors, CXCR/immunology , Animals , Antigens, Ly/biosynthesis , CD4-Positive T-Lymphocytes/immunology , CX3C Chemokine Receptor 1 , Chemokine CXCL16 , Chemokine CXCL6/biosynthesis , Citrobacter rodentium/immunology , Dendritic Cells/immunology , Interleukin-23/biosynthesis , Interleukins/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Transgenic , Natural Cytotoxicity Triggering Receptor 1/biosynthesis , Receptors, CXCR/biosynthesis , Receptors, CXCR/genetics , Receptors, CXCR6 , Receptors, Chemokine/biosynthesis , Receptors, Chemokine/immunology , Interleukin-22
7.
Eur J Immunol ; 44(11): 3380-91, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25142413

ABSTRACT

To study gene functions specifically in NKp46+ cells we developed novel Cre mice allowing for conditional gene targeting in cells expressing Ncr1 (encoding NKp46). We generated transgenic Ncr1(greenCre) mice carrying an EGFPcre fusion under the control of a proximal Ncr1 promoter that faithfully directed EGFPcre expression to NKp46+ cells from lymphoid and nonlymphoid tissues. This approach allowed for direct detection of Cre-expressing NKp46+ cells via their GFP signature by flow cytometry and histology. Cre was functional as evidenced by the NKp46+ cell-specific expression of RFP in Ncr1(greenCre) Rosa-dtRFP reporter mice. We generated Ncr1(greenCre) Il2rg(fl/fl) mice that lack NKp46+ cells in an otherwise intact hematopoietic environment. Il2rg encodes the common gamma chain (γc ), which is an essential receptor subunit for cytokines (IL-2, -4, -7, -9, -15, and -21) that stimulate lymphocyte development and function. In Ncr1(greenCre) Il2rg(fl/fl) mice, NK cells are severely reduced and the few remaining NKp46+ cells escaping γc deletion failed to express GFP. Using this new NK-cell-deficient model, we demonstrate that the homeostasis of NKp46+ cells from all tissues (including the recently described intraepithelial ILC1 subset) requires Il2rg. Finally, Ncr1(greenCre) Il2rg(fl/fl) mice are unable to reject B16 lung metastases demonstrating the essential role of NKp46+ cells in antimelanoma immune responses.


Subject(s)
Antigens, Ly/genetics , Killer Cells, Natural/immunology , Lung Neoplasms/secondary , Melanoma, Experimental/pathology , Natural Cytotoxicity Triggering Receptor 1/genetics , Animals , Antigens, Ly/biosynthesis , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Green Fluorescent Proteins/genetics , Interleukin Receptor Common gamma Subunit/genetics , Lung Neoplasms/immunology , Melanoma, Experimental/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Natural Cytotoxicity Triggering Receptor 1/biosynthesis
8.
Med Microbiol Immunol ; 203(5): 303-14, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24824867

ABSTRACT

We have characterized the NK/NKT-like cells in patients with self-limiting hepatitis E infection. The distribution of peripheral NK/NKT-like cells, expressions of activation receptors, cytotoxic potential and effector function of NK/NKT-like cells from fresh peripheral blood mononuclear cells of 86 acute patients, 101 recovered and 54 control individuals were assessed. Activated NKT-like (CD16(+) CD56(+) CD3(+)) cells were high in the patient groups. On CD56(+) CD3(-) cells, NKp44 and NKp46 expressions were high in the acute patients, whereas NKp30, NKp44, NKp46 and NKG2D were high in the recovered individuals. On CD56(+) CD3(+) cells, NKp44, NKp46 and NKG2D expressions were high in the recovered but NKp30 was low in both the patient groups. Collectively, the current study elucidates the role of NK/NKT-like cells demonstrating phenotypic alterations of activated NKT-like cells and activation receptors, lack of CD107a expression and functional impairment of peripheral NK/NKT-like cells in self-limiting hepatitis E infection.


Subject(s)
Gene Expression , Hepatitis E/pathology , Killer Cells, Natural/physiology , Natural Cytotoxicity Triggering Receptor 1/biosynthesis , Natural Cytotoxicity Triggering Receptor 2/biosynthesis , Natural Killer T-Cells/physiology , Adolescent , Adult , Aged , Antigens, CD/analysis , Female , Gene Expression Profiling , Hepatitis E/immunology , Humans , Killer Cells, Natural/chemistry , Lymphocyte Activation , Male , Middle Aged , NK Cell Lectin-Like Receptor Subfamily K/analysis , Natural Cytotoxicity Triggering Receptor 3/analysis , Natural Killer T-Cells/chemistry , Young Adult
9.
Melanoma Res ; 24(4): 295-304, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24769842

ABSTRACT

Although natural killer (NK) cells play an important antitumor role, melanoma cells may affect their effector functions. In this study, we analyzed the expression of various receptors and effector molecules in NK cells and their subsets in metastatic melanoma (MM) patients compared with healthy controls (HCs). In HC and MM patients, we analyzed NK cell activity using a chromium release assay and the expression of CD107a degranulation marker, activating NKG2D, NKp46, DNAM-1, and inhibitory CD158a and CD158b receptors, IL-12R beta 1, IL-12R beta 2, intracellular interferon (IFN)-γ, perforin, and STAT-1 in CD3-CD56+ NK cells, and cytotoxic CD3-CD56 and immunoregulatory CD3-CD56 subsets by flow cytometry. MM patients compared with HC not only had significantly decreased NK cell activity, lower expression of CD107a, and impaired IFN-γ production but also had decreased expression of activating NKG2D, NKp46, and DNAM-1 receptors, which was followed by lower expression of perforin, STAT-1, and both IL-12R subunits in NK cells. In MM patients only, there was a positive correlation between NKG2D expression and degranulation capacity, as well as IFN-γ production in NK cells. Analysis of the expression of various parameters of NK cell effector functions between MM patients with different localization of distant metastases showed that patients in the unfavorable M1c subclass had decreased expression of NKG2D and NKp46 on NK cells compared with patients in the M1a+b group. Downregulated NKG2D, NKp46, and DNAM-1 receptors associated with impaired NK cell effector function are important biomarkers of advanced disease with a poor prognosis in melanoma patients.


Subject(s)
Antigens, Differentiation, T-Lymphocyte/biosynthesis , Killer Cells, Natural/metabolism , Melanoma/metabolism , NK Cell Lectin-Like Receptor Subfamily K/biosynthesis , Natural Cytotoxicity Triggering Receptor 1/biosynthesis , Perforin/biosynthesis , STAT1 Transcription Factor/biosynthesis , Adult , Aged , Female , Humans , Killer Cells, Natural/immunology , Male , Melanoma/immunology , Middle Aged , Prognosis , Transcription Factors
10.
Am J Reprod Immunol ; 71(4): 359-67, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24495049

ABSTRACT

PROBLEM: To investigate the relationship between the expression of natural cytotoxicity receptors (NCRs) on peritoneal fluid (PF) natural killer (NK) (pfNK) cells and cytokine production by pfNK cells in women with endometriosis. METHOD OF STUDY: Peritoneal fluid was collected from women with endometriosis undergoing laparoscopic surgery (n = 21) and controls without endometriosis (n = 28). The expression of NK cell surface antigens such as CD16 and NCRs (NKp46, NKp44 and NKp30) on pfNK cells, and cytokines production by pfNK cells [tumor necrosis factor (TNF)-α, IFN-γ, IL-4, IL-10, GM-CSF and transforming growth factor (TGF)-ß1] were measured using multicolor flow cytometry. RESULTS: The percentages of CD56(+)/NKp46(+) cells and CD56(dim) /NKp46(+) cells in severe endometriosis group were significantly lower than that in controls. TNF-α and IFN-γ production by pfNK cells in severe endometriosis group was significantly higher than those in controls. CONCLUSION: The differential expression of NKp46, TNF-α, and IFN-γ on pfNK cells in women with severe endometriosis may allow the proliferation and angiogenesis of endometriotic cells.


Subject(s)
Endometriosis/immunology , Interferon-gamma/biosynthesis , Killer Cells, Natural/immunology , Natural Cytotoxicity Triggering Receptor 1/biosynthesis , Tumor Necrosis Factor-alpha/biosynthesis , Adult , Antigens, Surface/biosynthesis , Ascitic Fluid/immunology , Cell Proliferation , Cytokines/biosynthesis , Endometriosis/pathology , Female , Humans , Laparoscopy , Neovascularization, Pathologic , Receptors, Natural Cytotoxicity Triggering/biosynthesis
11.
Am J Clin Pathol ; 140(6): 853-66, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24225754

ABSTRACT

OBJECTIVES: To evaluate the expression of CD335 (NKp46), an activation receptor that is selectively expressed on natural killer (NK) cells. METHODS: We assessed CD335's potential utility as a diagnostic marker in 657 cases by flow cytometry and 410 cases by immunohistochemistry. RESULTS: We observed that CD335 was highly specific for NK cells in nonneoplastic tissues. Moreover, 61 (90%) of 68 of NK cell neoplasms demonstrated CD335 expression, whereas B-cell, myelomonocytic, and plasma cell neoplasms lacked expression. Notably, 16 (20%) of 82 mature T-cell neoplasms, particularly T-cell large granular lymphocytic leukemia, mycosis fungoides, and ALK+ anaplastic large cell lymphoma, aberrantly expressed CD335. CONCLUSIONS: Collectively, these data support the diagnostic utility of CD335 in evaluating hematopoietic malignancies and suggest that CD335 could be a useful target for selective immunotherapy in patients with mature NK and T-cell neoplasms.


Subject(s)
Biomarkers, Tumor/analysis , Hematologic Neoplasms/diagnosis , Killer Cells, Natural/immunology , Natural Cytotoxicity Triggering Receptor 1/biosynthesis , T-Lymphocytes/immunology , Adolescent , Adult , Aged , Aged, 80 and over , Child , Child, Preschool , Female , Flow Cytometry , Hematologic Neoplasms/immunology , Hematologic Neoplasms/metabolism , Humans , Immunohistochemistry , Infant , Killer Cells, Natural/metabolism , Killer Cells, Natural/pathology , Male , Middle Aged , Natural Cytotoxicity Triggering Receptor 1/analysis , T-Lymphocytes/metabolism , T-Lymphocytes/pathology
12.
J Immunol ; 191(3): 1486-95, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23817426

ABSTRACT

In this study, we explore the hypothesis that enhanced production of lactate by tumor cells, because of high glycolytic activity, results in inhibition of host immune response to tumor cells. Lactate dehydrogenase-A (LDH-A), responsible for conversion of pyruvate to lactate, is highly expressed in tumor cells. Lentiviral vector-mediated LDH-A short hairpin RNA knockdown Pan02 pancreatic cancer cells injected in C57BL/6 mice developed smaller tumors than mice injected with Pan02 cells. A decrease occurred in the frequency of myeloid-derived suppressor cells (MDSCs) in the spleens of mice carrying LDH-A-depleted tumors. NK cells from LDH-A-depleted tumors had improved cytolytic function. Exogenous lactate increased the frequency of MDSCs generated from mouse bone marrow cells with GM-CSF and IL-6 in vitro. Lactate pretreatment of NK cells in vitro inhibited cytolytic function of both human and mouse NK cells. This reduction of NK cytotoxic activity was accompanied by lower expression of perforin and granzyme in NK cells. The expression of NKp46 was decreased in lactate-treated NK cells. These studies strongly suggest that tumor-derived lactate inhibits NK cell function via direct inhibition of cytolytic function as well as indirectly by increasing the numbers of MDSCs that inhibit NK cytotoxicity. Depletion of glucose levels using a ketogenic diet to lower lactate production by glycolytic tumors resulted in smaller tumors, decreased MDSC frequency, and improved antitumor immune response. These studies provide evidence for an immunosuppressive role of tumor-derived lactate in inhibiting innate immune response against developing tumors via regulation of MDSC and NK cell activity.


Subject(s)
Killer Cells, Natural/immunology , L-Lactate Dehydrogenase/metabolism , Lactic Acid/metabolism , Neoplasms/immunology , Tumor Escape , Animals , Antigens, Ly/biosynthesis , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Cell Line, Tumor , Cytotoxicity, Immunologic , Female , Glucose/biosynthesis , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Granzymes/biosynthesis , Humans , Interleukin-6/metabolism , Isoenzymes/genetics , Isoenzymes/metabolism , Killer Cells, Natural/drug effects , L-Lactate Dehydrogenase/genetics , Lactate Dehydrogenase 5 , Lactic Acid/pharmacology , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Myeloid Cells/immunology , Natural Cytotoxicity Triggering Receptor 1/biosynthesis , Neoplasms/genetics , Neoplasms/metabolism , Perforin/biosynthesis , RNA Interference , RNA, Small Interfering , Spleen/immunology
13.
J Biol Chem ; 287(10): 7324-34, 2012 Mar 02.
Article in English | MEDLINE | ID: mdl-22253448

ABSTRACT

Natural cytotoxicity receptor 1 (NCR1), also known as NKp46, is a natural killer (NK) lymphocyte-activating receptor. It is involved in major aspects of NK immune function and shows a high degree of lineage specificity in blood and bone marrow. The nature of its NK-restricted expression is not well understood. In this study, we confirm that human NCR1 NK-specific expression is achieved at the mRNA level. We found two key cis-regulatory elements in the immediate vicinity upstream of the gene. One element acts as an essential promoter, whereas the other acts as a tissue-dependent enhancer/repressor. This latter regulatory element contains a runt related-transcription factor (RUNX) recognition motif that preferentially binds RUNX3. Interfering with RUNX proteins using a dominant negative form results in decreased Ncr1 expression. RUNX3 overexpression had the opposite effect. These findings shed light on the role of RUNX3 in the control of an important NK-activating receptor.


Subject(s)
Core Binding Factor Alpha 3 Subunit/metabolism , Killer Cells, Natural/metabolism , Natural Cytotoxicity Triggering Receptor 1/biosynthesis , RNA, Messenger/biosynthesis , Response Elements/physiology , Animals , Core Binding Factor Alpha 3 Subunit/genetics , HEK293 Cells , Humans , K562 Cells , Killer Cells, Natural/cytology , Mice , NIH 3T3 Cells , Natural Cytotoxicity Triggering Receptor 1/genetics , Organ Specificity/physiology , RNA, Messenger/genetics
14.
Cancer Immunol Immunother ; 61(9): 1395-405, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22270713

ABSTRACT

BACKGROUND: IRX-2 is a primary biologic which has been used for the therapy of head and neck squamous cell cancer (HNSCC) with promising clinical results. Since NK-cell function is compromised in HNSCC patients, we tested the effects of IRX-2 on the restoration of human NK-cell functions in vitro. METHODS: Peripheral blood mononuclear cells (PBMC) were isolated from 23 HNSCC patients and 10 normal controls (NC). The NK-cell phenotype and functions were compared before and after culture ± IRX-2 or ± 50 IU/ml rhIL-2. Flow cytometry was used to study the NK-cell phenotype, cytotoxic activity and cytokine expression. RESULTS: Impaired NK-cell cytotoxicity in HNSCC patients was related to lower expression of NKG2D, NKp30 and NKp46 receptors (P < 0.05) and not to a decreased frequency of NK cells. Incubation of patients' NK cells with IRX-2 up-regulated the percentage of receptor-positive NK cells (P < 0.05). It also up-regulated cytotoxicity of patients' NK cells (P < 0.01) more effectively than rhIL-2 (P < 0.01). IRX-2, but not rhIL-2, protected NK cells from suppression mediated by TGF-ß, and it restored (P < 0.05) expression of activating NK-cell receptors and NK-cell cytotoxicity suppressed by TGF-ß. Expression of pSMAD was decreased in NK cells treated with IRX-2 but not in those treated with rhIL-2. CONCLUSIONS: IRX-2 was more effective than IL-2 in enhancing NK-cell cytotoxicity and protecting NK-cell function of HNSCC patients in vitro, emphasizing the potential advantage of IRX-2 as a component of future therapies for HNSCC.


Subject(s)
Carcinoma, Squamous Cell/immunology , Cytokines/pharmacology , Head and Neck Neoplasms/immunology , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Carcinoma, Squamous Cell/blood , Carcinoma, Squamous Cell/therapy , Cytokines/immunology , Cytotoxicity, Immunologic/drug effects , Cytotoxicity, Immunologic/immunology , Flow Cytometry , Head and Neck Neoplasms/blood , Head and Neck Neoplasms/therapy , Humans , Immunotherapy , Interleukin-2/immunology , Interleukin-2/pharmacology , K562 Cells , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , NK Cell Lectin-Like Receptor Subfamily K/biosynthesis , NK Cell Lectin-Like Receptor Subfamily K/immunology , Natural Cytotoxicity Triggering Receptor 1/biosynthesis , Natural Cytotoxicity Triggering Receptor 1/immunology , Natural Cytotoxicity Triggering Receptor 3/biosynthesis , Natural Cytotoxicity Triggering Receptor 3/immunology , Phosphorylation/drug effects , Smad Proteins/biosynthesis , Smad Proteins/immunology , Transforming Growth Factor beta1/immunology , Transforming Growth Factor beta1/pharmacology , Up-Regulation/drug effects , Up-Regulation/immunology
15.
J Leukoc Biol ; 91(2): 321-31, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22124136

ABSTRACT

HDACi are being used as a novel, therapeutic approach for leukemias and other hematological malignancies. However, their effect on immune cells remains ill-defined, as HDACi may impair immune surveillance. In this work, we demonstrate that TSA, VPA, and NaB inhibited IFN-γ production by CD56(dim) and CD56(bright) NK cells and NK cell-mediated cytotoxicity against K562 target cells. HDACi promoted minor NK cell apoptosis but inhibited nuclear mobilization of NF-κB p50, which was accompanied by a robust down-regulation of NKG2D and NKp46 on resting NK cells and of NKG2D, NKp44, NKp46, and CD25 on cytokine-activated NK cells. Decreased CD25 expression promoted a weakened IFN-γ secretion upon restimulation of NK cells with IL-2, whereas reduced expression of NKG2D and NKp46 was accompanied by an impaired NKG2D- and NKp46-dependent cytotoxicity. Moreover, NK cells from normal mice treated in vivo with TSA displayed a diminished expression of NK1.1, NKG2D, and NKp46 and secreted reduced amounts of IFN-γ upon ex vivo stimulation with cytokines. Thus, our preclinical results indicate that HDACi exert deleterious effects on NK cell function, which may weaken immune surveillance and facilitate relapse of the malignant disease in HDACi-treated patients.


Subject(s)
Antineoplastic Agents/pharmacology , Butyrates/pharmacology , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Immunologic Surveillance/drug effects , Killer Cells, Natural/drug effects , Lymphocyte Activation/drug effects , Animals , Antineoplastic Agents/adverse effects , Apoptosis/drug effects , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Cells, Cultured/drug effects , Cells, Cultured/immunology , Cells, Cultured/metabolism , Cytoplasmic Granules/drug effects , Cytoplasmic Granules/metabolism , Cytotoxicity, Immunologic , Histone Deacetylase Inhibitors/adverse effects , Humans , Interferon-gamma/metabolism , Interleukins/pharmacology , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Mice , Mice, Inbred C57BL , NK Cell Lectin-Like Receptor Subfamily K/biosynthesis , NK Cell Lectin-Like Receptor Subfamily K/genetics , Natural Cytotoxicity Triggering Receptor 1/biosynthesis , Natural Cytotoxicity Triggering Receptor 1/genetics , Vorinostat
16.
Eur J Immunol ; 41(6): 1563-72, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21469096

ABSTRACT

Here, we identify cells within human adult secondary lymphoid tissues that are comparable in phenotype and location to the lymphoid tissue inducer (LTi) cells that persist in the adult mouse. Identified as CD117(+) CD3(-) CD56(-) cells, like murine LTi cells, they lack expression of many common lineage markers and express CD127, OX40L and TRANCE. These cells were detected at the interface between the B- and T- zones, as well as at the subcapsular sinus in LNs, the location where LTi cells reside in murine spleen and LNs. Furthermore, like murine LTi cells, these cells expressed high levels of IL-22 and upregulated IL-22 expression upon IL-23 stimulation. Importantly, these cells were not an NK cell subset since they showed no expression of IFN-γ and perforin. Interestingly, a subset of the CD117(+) CD3(-) CD56(-) OX40L(+) population expressed NKp46, again similar to recent findings in mice. Finally, these cells supported memory CD4(+) T-cell survival in an OX40L-dependent manner. Combined, these data indicate that the CD117(+) CD3(-) CD56(-) OX40L(+) cells in human secondary lymphoid tissues are comparable in phenotype, location and function to the LTi cells that persist within adult murine secondary lymphoid tissues.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Interleukins/metabolism , OX40 Ligand/metabolism , Palatine Tonsil/cytology , Th17 Cells/metabolism , Animals , Antigens, CD/biosynthesis , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , Cell Differentiation , Cell Separation , Cell Survival , Cells, Cultured , Flow Cytometry , Humans , Interleukin-23/immunology , Interleukin-23/metabolism , Interleukins/genetics , Interleukins/immunology , Lymph Nodes/cytology , Mice , Natural Cytotoxicity Triggering Receptor 1/biosynthesis , OX40 Ligand/immunology , Th17 Cells/cytology , Th17 Cells/immunology , Interleukin-22
17.
Scand J Immunol ; 74(2): 107-13, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21375557

ABSTRACT

The neuropeptide substance P (SP) can regulate a number of immunological functions in vitro and in vivo and may regulate natural killer (NK) cell activity. Here, we investigated whether SP has a role in regulating NK92-MI cell function in vitro, and how it influences NK cell activity. We found that SP dose dependently increased the cytotoxicity of NK92-MI cells and had a maximal effect at a concentration of 10(-12) and 10(-10) m. Furthermore, the expression of cytotoxic-associated molecules (perforin, granzyme) and activating receptor NKp46 [a member of natural cytotoxicity receptors (NCRs)] was observed to be upregulated by SP at optimal concentration, at which SP enhanced the cytotoxicity of NK92-MI cells. Neurokinin-1 receptor (NK-1R), a functional receptor of SP, was found on NK92-MI cells, and the observed effects of SP on NK92-MI cells could be more partially blocked by an NK-1R antagonist. Our data suggest that SP induces NK92-MI cell cytotoxicity by directly increasing the expression of cytotoxic granules and upregulates NK92-MI cell receptor-mediated functions indirectly. Thus, SP may regulate NK cell function mainly through NK-1R.


Subject(s)
Cytotoxicity, Immunologic , Killer Cells, Natural/immunology , Substance P/immunology , Cell Line , Granzymes/biosynthesis , Granzymes/immunology , Humans , Killer Cells, Natural/drug effects , Natural Cytotoxicity Triggering Receptor 1/biosynthesis , Natural Cytotoxicity Triggering Receptor 1/immunology , Neurokinin-1 Receptor Antagonists , Perforin/biosynthesis , Perforin/immunology , Receptors, Neurokinin-1/immunology , Substance P/pharmacology , Up-Regulation/drug effects , Up-Regulation/immunology
18.
Transfusion ; 51(2): 293-305, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20723170

ABSTRACT

BACKGROUND: The human cytokine granulocyte-colony stimulatory factor (G-CSF) has found widespread application in the medical treatment of neutropenia and to mobilize hematopoietic stem cells used for transplantation. So far, the effect of G-CSF on natural killer (NK) cells has not been fully investigated. STUDY DESIGN AND METHODS: The effect of G-CSF on the phenotype, cytokine secretion profile, and cytotoxicity of NK cells was assessed. NK cells incubated in vitro in presence of G-CSF for 48 hours as well as NK cells isolated from peripheral blood of G-CSF-mobilized stem cell donors (in vivo) were used. RESULTS: In vitro, G-CSF caused a strongly altered phenotype in NK cells with 49% down regulation of NKp44 frequency. Furthermore, the expression of the activating receptors NKp46 and NKG2D decreased 40 and 64%, respectively. The expression of KIR2DL1 and KIR2DL2 decreased by 46% each. In cytotoxicity assays, the lytic capacity of G-CSF-exposed NK cells is reduced by up to 68% in vitro and up to 83% in vivo. Accordingly, granzyme B levels of in vivo G-CSF-exposed NK cells were reduced by up to 87% in comparison to nonstimulated NK cells. Cytokine production of in vitro and in vivo incubated NK cells was strongly decreased for interferon-γ, tumor necrosis factor-α, and granulocyte macrophage colony-stimulating factor as well as interleukin (IL)-6 and IL-8. Furthermore, we observed a reduction in proliferation and a positive feedback loop that increased the expression of the G-CSF receptor. CONCLUSION: G-CSF was demonstrated to be a strong inhibitor of NK cells activity and may prevent their graft-versus-leukemia effect after transplantation.


Subject(s)
Cytotoxicity, Immunologic/drug effects , Granulocyte Colony-Stimulating Factor/pharmacology , Killer Cells, Natural/drug effects , Cell Division/drug effects , Cells, Cultured/drug effects , Cells, Cultured/immunology , Cytokines/biosynthesis , Cytokines/genetics , Depression, Chemical , Gene Expression Regulation/drug effects , Graft vs Leukemia Effect/drug effects , Granulocyte Colony-Stimulating Factor/adverse effects , Granzymes/biosynthesis , Granzymes/genetics , Humans , Killer Cells, Natural/metabolism , Lenograstim , NK Cell Lectin-Like Receptor Subfamily K/biosynthesis , NK Cell Lectin-Like Receptor Subfamily K/genetics , Natural Cytotoxicity Triggering Receptor 1/biosynthesis , Natural Cytotoxicity Triggering Receptor 1/genetics , Natural Cytotoxicity Triggering Receptor 2/biosynthesis , Natural Cytotoxicity Triggering Receptor 2/genetics , Receptors, Granulocyte Colony-Stimulating Factor/biosynthesis , Receptors, Granulocyte Colony-Stimulating Factor/genetics , Recombinant Proteins/adverse effects , Recombinant Proteins/pharmacology
19.
Anim Biotechnol ; 21(3): 156-63, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20665287

ABSTRACT

NCR1 (NKp46) is expressed on the surfaces of natural killer cells and recognizes hemagglutinin on the influenza virus. We cloned the NCR1 gene in pigs and found that porcine NCR1 was minimally expressed in the thymus, suggesting that NCR1 could be a useful marker of natural killer cells in pigs. We observed three nonsynonymous single nucleotide polymorphisms and one deletion of three nucleotides in the coding sequence of porcine NCR1; these may affect the function of NCR1. The polymorphisms detected here may be useful markers for breeding for influenza resistance in pigs.


Subject(s)
Natural Cytotoxicity Triggering Receptor 1/genetics , Swine/genetics , Animals , Base Sequence , Chromosomes, Artificial, Bacterial , Cloning, Molecular , Female , Genetic Variation , Male , Molecular Sequence Data , Natural Cytotoxicity Triggering Receptor 1/biosynthesis , Natural Cytotoxicity Triggering Receptor 1/immunology , Polymorphism, Single Nucleotide , RNA/chemistry , RNA/genetics , Reverse Transcriptase Polymerase Chain Reaction/veterinary , Sequence Alignment , Sequence Analysis, DNA , Swine/immunology , Thymus Gland/metabolism , Thymus Gland/physiology
20.
J Virol ; 84(15): 7822-31, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20484512

ABSTRACT

Natural killer (NK) cells are the effectors of innate immunity and are recruited into the lung 48 h after influenza virus infection. Functional NK cell activation can be triggered by the interaction between viral hemagglutinin (HA) and natural cytotoxicity receptors NKp46 and NKp44 on the cell surface. Recently, novel subtypes of influenza viruses, such as H5N1 and 2009 pandemic H1N1, transmitted directly to the human population, with unusual mortality and morbidity rates. Here, the human NK cell responses to these viruses were studied. Differential activation of heterogeneous NK cells (upregulation of CD69 and CD107a and gamma interferon [IFN-gamma] production as well as downregulation of NKp46) was observed following interactions with H5N1, 1918 H1N1, and 2009 H1N1 pseudotyped particles (pps), respectively, and the responses of the CD56(dim) subset predominated. Much stronger NK activation was triggered by H5N1 and 1918 H1N1 pps than by 2009 H1N1 pps. The interaction of pps with NK cells and subsequent internalization were mediated by NKp46 partially. The NK cell activation by pps showed a dosage-dependent manner, while an increasing viral HA titer attenuated NK activation phenotypes, cytotoxicity, and IFN-gamma production. The various host innate immune responses to different influenza virus subtypes or HA titers may be associated with disease severity.


Subject(s)
Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H5N1 Subtype/immunology , Influenza, Human/immunology , Influenza, Human/virology , Killer Cells, Natural/immunology , Adult , Antigens, CD/biosynthesis , Antigens, Differentiation, T-Lymphocyte/biosynthesis , CD56 Antigen/analysis , Cytotoxicity, Immunologic , Down-Regulation , Gene Expression , Humans , Interferon-gamma/metabolism , Lectins, C-Type/biosynthesis , Lymphocyte Subsets/chemistry , Lymphocyte Subsets/immunology , Lysosomal-Associated Membrane Protein 1/biosynthesis , Natural Cytotoxicity Triggering Receptor 1/biosynthesis , Up-Regulation , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...