Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 124
Filter
1.
J Exp Med ; 170(2): 481-97, 1989 Aug 01.
Article in English | MEDLINE | ID: mdl-2526846

ABSTRACT

A low affinity receptor for IgG immune complexes, Fc gamma RIII(CD16), is expressed on human NK cells as an integral membrane glycoprotein anchored through a transmembrane peptide; on polymorphonuclear neutrophils (PMN) the receptor is anchored through a phosphatidylinositol (PI) linkage. The protein on NK cells has a molecular mass 6-10 kD larger than that on PMN, and, unlike the latter, is resistant to PI-specific phospholipase C (PI-PLC). Fc gamma RIII(CD16) transcripts isolated from PMN and NK cells of single donors revealed multiple single nucleotide differences, one of which converts an in frame UGA termination codon to a CGA codon. The resulting open reading frame encodes a longer cytoplasmic domain for Fc gamma RIII(CD16) in NK cells, contributing to its transmembrane anchor. Two nearly identical, linked genes that encode these transcripts have been cloned for Fc gamma RIII(CD16), one of which (III-1) is allelic for NA-1 and NA-2. The allelic sites have been mapped to two single nucleotides in the extracellular domain. These genes are transcribed in a cell type-specific fashion to generate the alternatively anchored forms of this receptor.


Subject(s)
Antigens, Differentiation/genetics , Killer Cells, Natural/physiology , Neutrophils/physiology , Receptors, Fc/genetics , Amino Acid Sequence , Base Sequence , Cloning, Molecular , DNA/genetics , Genes , Glycoside Hydrolases/pharmacology , Humans , Membrane Glycoproteins/genetics , Molecular Sequence Data , Oligonucleotide Probes , Peptide Chain Termination, Translational , Phosphatidylinositols/physiology , RNA, Messenger/genetics , Receptors, IgG , Type C Phospholipases/pharmacology
2.
J Exp Med ; 182(3): 801-10, 1995 Sep 01.
Article in English | MEDLINE | ID: mdl-7650486

ABSTRACT

The putative factors that couple the signal transduction from surface receptors to the activation of cytokine synthesis in natural killer (NK) cells have not been elucidated. We report here that the nuclear factor of activated T cells (NFATp), a cyclosporin A (CsA)-sensitive factor that regulates the transcription of several cytokines, mediates CD16-induced activation of cytokine genes in human NK cells. CD16 (Fc gamma RIIIA)-induced expression of cytokine mRNA in NK cells occurs via a CsA-sensitive and Ca(2+)-dependent mechanism. Stimulation of NK cells with CD16 ligands induces NFAT-like DNA binding activity in the nuclear extracts from these cells, as detected in electrophoretic mobility shift assays. This occurs with fast kinetics after stimulation, via a CsA-sensitive and Ca(2+)-dependent mechanism that does not require de novo protein synthesis. NK cell NFAT is present in the cytosol of nonstimulated cells, migrates to the nucleus upon stimulation, and can associate with AP-1. Two distinct molecules, NFATp and NFATc, have been reported to mediate NFAT activity. The results of supershift assays using NFATp- and NFATc- specific antibodies indicate that NK cell activation early after CD16 ligand binding involves primarily, if not exclusively, NFATp, and Western blot analysis shows that this has the same electrophoretic mobility (approximately 120 kD) as that of T lymphocytes. NK cells do not express NFATc constitutively, but NFATc mRNA accumulation is induced in these cells within 2 h of stimulation with CD16 ligands. However, supershift assays using the available mAb recognizing the T cell NFATc revealed no detectable NFATc protein in nuclear and cytoplasmic extracts from CD16- or phorbol ester-stimulated cells at any time tested, up to 4 h. These results provide the first direct evidence that both CsA-sensitive transcription factors, NFATp and NFATc, are expressed in human NK cells, and that their activation and/or expression can be regulated in primary cells by a single stimulus, that, in the case of CD16 in NK cells, results in early activation of NFATp and subsequently induced expression of NFATc mRNA.


Subject(s)
DNA-Binding Proteins/biosynthesis , Gene Expression Regulation/drug effects , Killer Cells, Natural/immunology , Nuclear Proteins , Receptors, IgG/physiology , Transcription Factors/biosynthesis , Animals , Antibodies, Monoclonal/pharmacology , Antigen-Antibody Complex/immunology , B-Lymphocytes , Base Sequence , Cell Line, Transformed , Chlorocebus aethiops , Cyclosporine/pharmacology , Cytokines/biosynthesis , Cytokines/genetics , DNA-Binding Proteins/genetics , Humans , Interleukin-12/pharmacology , Interleukin-2/pharmacology , Killer Cells, Natural/drug effects , Killer Cells, Natural/metabolism , Leukemia-Lymphoma, Adult T-Cell/pathology , Ligands , Lymphocyte Activation/drug effects , Mice , Molecular Sequence Data , NFATC Transcription Factors , Transcription Factors/genetics , Tumor Cells, Cultured
3.
J Exp Med ; 184(3): 1027-35, 1996 Sep 01.
Article in English | MEDLINE | ID: mdl-9064320

ABSTRACT

Cross-linking the receptors for the Fc domain of IgG (Fc gamma R) on leukocytes induces activation of protein tyrosine kinases. The intermediary molecules that transduce to the nucleus the signals leading to induction of the diverse biological responses mediated by these receptors are not clearly identified. We have investigated whether mitogen-activated protein kinases (MAPK) are involved in transmembrane signaling via the three Fc gamma R present on monocytic, polymorphonuclear, and natural killer (NK) cells. Our results indicate that occupancy of Fc gamma RI and Fc gamma RII on the monocytic cell line THP-I and on polymorphonuclear leukocytes (PMN) induces, transiently and with fast kinetics, MAPK phosphorylation, as indicated by decreased electrophoretic mobility in sodium dodecyl sulfate-polyacrylamide gel electrophoresis, and increased amounts of the proteins in antiphosphotyrosine antibody immunoprecipitates. This, associated with increased enzymatic activity, also occurs upon stimulation of the transmembrane isoform of CD16 (Fc gamma RIIIA) in NK cells and in a T cell line expressing transfected Fc gamma RIIIA alpha ligand-binding chain in association with zeta, but not upon stimulation of the glycosil-phosphatidylinositol-anchored Fc gamma RIIIB on PMN. Using the specific MAP kinase kinase inhibitor-PD 098059, we show that activation of MAPK is necessary for the Fc gamma R-dependent induction of c-fos and tumor necrosis factor alpha mRNA expression in monocytes and NK cells. These results underscore the role of MAPK as signal-transducing molecules controlling the expression of different genes relevant to leukocyte biology upon Fc gamma R stimulation.


Subject(s)
Gene Expression Regulation, Enzymologic , Leukocytes/enzymology , Mitogen-Activated Protein Kinases , Receptors, IgG/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/biosynthesis , Animals , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Electrophoresis, Polyacrylamide Gel , Enzyme Activation , Humans , Killer Cells, Natural/metabolism , Kinetics , Mice , Mitogen-Activated Protein Kinase 1 , Mitogen-Activated Protein Kinase 3 , Phosphorylation , Th1 Cells/metabolism , Tumor Cells, Cultured
4.
J Exp Med ; 161(5): 1152-68, 1985 May 01.
Article in English | MEDLINE | ID: mdl-3838767

ABSTRACT

Incubation of human peripheral blood lymphocytes with bone marrow cells resulted in significant inhibition of colony formation by committed myeloid and erythroid cells. Using positively selected homogeneous natural killer (NK) cell preparations and lymphocyte subpopulations depleted of or enriched for NK cells, we definitively characterize as NK cells the cells in normal peripheral blood that are responsible for inhibition of bone marrow colony growth. The inhibitory effect of NK cells on hematopoiesis can be mediated by a soluble factor that is produced only by NK cells upon culture with HLA-DR+ hematopoietic cells and with NK-sensitive cell lines. Both NK cells and the NK-produced, colony-inhibiting activity (NK-CIA) are suppressive for allogeneic and autologous bone marrow CFU-GEMM (colony-forming units, granulocyte, erythroid, monocyte, megakaryocyte), CFU-E (CFU, erythroid), and early CFU-GM (CFU, granulocyte, monocyte), but not for either BFU-E (burst-forming units, erythroid) or late CFU-GM. [3H]Thymidine incorporation was inhibited by NK-CIA-containing supernatants in HLA-DR+ but not HLA-DR- bone marrow cell populations stimulated to proliferative by colony-stimulating factor (CSF). These data suggest that the NK cell-mediated inhibitory effect on proliferation and differentiation of hematopoietic precursor cells is mediated in part or completely by the secreted NK-CIA. The concentration of NK-CIA reached in the supernatant of the mixture of NK cell-containing lymphocyte populations with bone marrow cells is sufficient to account for the inhibitory effect mediated by NK cells. Our data support the hypothesis that human NK cells play a major role in the control of hematopoiesis, down-regulating it under conditions in which the NK cells are functionally activated.


Subject(s)
Bone Marrow Cells , Growth Inhibitors/physiology , Hematopoiesis , Killer Cells, Natural/physiology , Lipoproteins/physiology , Proteins/physiology , Animals , Bone Marrow/pathology , Cell Line , Cell-Free System , Colony-Forming Units Assay , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/pathology , Humans , Killer Cells, Natural/metabolism , Killer Factors, Yeast , Kinetics , Mice
5.
J Exp Med ; 164(6): 2061-76, 1986 Dec 01.
Article in English | MEDLINE | ID: mdl-3097239

ABSTRACT

We show here that human peripheral blood NK cells contain a pore-forming protein (PFP) with an Mr of 70,000-72,000 that assembles structural lesions (with an average internal diameter of 150-170 A) and forms functional channels. The PFP was isolated by affinity chromatography from human NK cells, using a specific anti-C9 antiserum as the immunoadsorbent. The NK cells were isolated from PBL by positive or negative selection by indirect rosetting using a panel of monoclonal antibodies directed against different NK and T cell surface antigens. PFP was identified in NK cells freshly isolated and isolated from cultured PBL, both stimulated with interleukin 2, but not in NK cell-depleted lymphocytes. In planar bilayers, the channels formed by the NK cell-derived PFP are highly voltage resistant, with most channels persisting in the open state once they have inserted into the bilayer. The unit conductances of these channels range 0.3-1 nS in 0.1 M NaCl. The channels show poor selectivity for monovalent and divalent ions. The PFP is also released from human NK cells stimulated with the calcium ionophore A23187, suggesting that this protein, like the one produced by murine CTL lines, may be similarly secreted during cell-mediated killing. Its identification in primary human NK cell cultures indicates that this protein may play an active role in NK cell-mediated killing.


Subject(s)
Killer Cells, Natural/analysis , Membrane Glycoproteins , Membrane Proteins/analysis , Calcimycin/pharmacology , Cell Line , Chromatography, Affinity , Cytotoxicity, Immunologic , Electrophoresis, Polyacrylamide Gel , Humans , Immunosorbent Techniques , Kidney Tubules/ultrastructure , Killer Cells, Natural/immunology , Molecular Weight , Perforin , Phenotype , Pore Forming Cytotoxic Proteins , Rosette Formation
6.
J Exp Med ; 176(6): 1745-50, 1992 Dec 01.
Article in English | MEDLINE | ID: mdl-1281217

ABSTRACT

Binding of ligand to the alpha subunit of Fc gamma RIIIA(CD16), expressed at the natural killer (NK) cell membrane in association with homo or heterodimers of proteins of the zeta family, results in phosphorylation of several proteins on tyrosine residues. We have analyzed the role of protein tyrosine phosphorylation in the regulation of molecular events induced upon stimulation of Fc gamma RIIIA in NK cells and in T cells expressing the Fc gamma RIII alpha chain in association with endogenous zeta 2 homodimers and devoid of other (CD3, CD2) transducing molecules. Our data indicate that treatment of these cells with protein tyrosine kinase inhibitors prevents not only Fc gamma RIIIA-induced protein tyrosine phosphorylation but also phosphatidylinositol 4,5 diphosphate hydrolysis and increased intracellular Ca2+ concentration, indicating a primary role of tyrosine kinase(s) in the induction of these early activation events. Occupancy of Fc gamma RIIIA by ligand results in phospholipase C (PLC)-gamma 1 tyrosine phosphorylation in NK cells and in Fc gamma RIIIA-transfected CD3-/CD2- T cells, and induces functional activation of p56lck in Fc gamma RIIIA alpha/zeta 2-transfected T cells, suggesting the possibility that the receptor-induced PLC-gamma 1 activation occurs upon phosphorylation of its tyrosine residues mediated by this kinase and is, at least in part, responsible for the signal transduction mediated via CD16 upon ligand binding.


Subject(s)
Isoenzymes/metabolism , Protein-Tyrosine Kinases/metabolism , Receptors, IgG/metabolism , Type C Phospholipases/metabolism , Antibodies, Monoclonal , Cell Membrane/immunology , Humans , Killer Cells, Natural/immunology , Lymphocyte Specific Protein Tyrosine Kinase p56(lck) , Macromolecular Substances , Phosphorylation , Phosphotyrosine , Protein-Tyrosine Kinases/antagonists & inhibitors , Receptors, IgG/genetics , Transfection , Tumor Cells, Cultured , Tyrosine/analogs & derivatives , Tyrosine/analysis
7.
J Exp Med ; 158(4): 1092-113, 1983 Oct 01.
Article in English | MEDLINE | ID: mdl-6225822

ABSTRACT

We report here that FcR for human monomeric IgG1 can be induced on cells of myeloid origin cultured in the presence of IFN gamma for 8 h. Supernatant fluids from cultures of lymphocytes infected with a variety of viruses or cocultured with cell lines have the same FcR enhancing effect as IFN gamma. We identify the factor in the supernatant fluid responsible for the induction as immune interferon. Among the different types of IFN, only the gamma type (both purified and recombinant) specifically induces the appearance of FcR for monomeric IgG1 on normal and leukemic myeloid cells but not on cells of lymphoid origin. This effect is also evident on mature PMN. We show that the specificity and the affinity of the receptor induced on HL-60 promyelocytic cells, peripheral blood monocytes, and PMN are identical to those of the receptor spontaneously present on the same cells, except for PMN, which do not spontaneously express this type of receptor. The results of inhibition experiments performed with mouse IgG of and IgG3. These results suggest that the receptor present on human monocytes different isotypes indicate that the receptor can be inhibited by murine IgG2a or immature myeloid cells, selectively inducible by IFN gamma, has a specificity similar to the FcR1 described on mouse macrophages.


Subject(s)
Interferon-gamma/physiology , Monocytes/metabolism , Neutrophils/metabolism , Receptors, Fc/analysis , Bone Marrow Cells , Cell Line , Humans , Immunoglobulin G/classification , Influenza, Human/immunology , Leukemia, Myeloid, Acute/immunology , Melanoma/immunology , Molecular Weight , Receptors, IgG , Rosette Formation
8.
J Exp Med ; 167(2): 452-72, 1988 Feb 01.
Article in English | MEDLINE | ID: mdl-2831292

ABSTRACT

We report evidence that FcR(CD16) on human NK cells are signal-transducing molecules that, upon ligand binding, induce transcription of genes encoding surface activation molecules [IL-2-R(CD25)] and cytokines (IFN-gamma and TNF) relevant to NK cell biology and functions. Homogeneous NK and T cell populations purified from short-term bulk cultures of PBMC with irradiated B lymphoblastoid cell lines were cultured in the presence of FcR ligands (particulate immune complexes or immobilized anti-CD16 antibodies) alone or with rIL-2. Upon 18 h of stimulation, NK cells express Tac, TfR, and 4F2 antigens and produce IFN-gamma and TNF; both effects are synergistically enhanced in the presence of rIL-2, which is itself ineffective. Treatment of NK cells with FcR(CD16) ligands induces accumulation of mRNA for IFN-gamma and TNF and, to a lesser extent, IL-2-R with fast kinetics also in the absence of de novo protein synthesis. rIL-2 and FcR(CD16) ligands synergize to induce mRNA accumulation. mRNA accumulation and transcription of TNF and IFN-gamma genes induced by FcR(CD16) ligands are greater than those induced by rIL-2, and the reverse is true for IL-2-R. The two stimuli do not synergize at the transcriptional level. These observations indicate that the mechanisms through which FcR(CD16) ligands and rIL-2 induce NK cell activation are, in part, distinct. Both operate at the transcriptional level, although other mechanisms are probably induced by the FcR ligand stimulus per se or in combination with other lymphokines and synergize at a post-transcriptional or translational level to enhance NK cell activation.


Subject(s)
Killer Cells, Natural/metabolism , Lymphokines/genetics , Receptors, Fc/physiology , Receptors, Immunologic/genetics , Transcription, Genetic , Animals , Antigens, Surface/biosynthesis , Biological Products/biosynthesis , Biological Products/metabolism , Cell Line , Cell Membrane/metabolism , Cells, Cultured , Cross-Linking Reagents , Cytokines , Drug Synergism , Humans , Interleukin-2/pharmacology , Killer Cells, Natural/immunology , Kinetics , Mice , Protein Biosynthesis , RNA, Messenger/metabolism , Receptors, Immunologic/metabolism , Receptors, Interleukin-2 , Recombinant Proteins/pharmacology , Tumor Necrosis Factor Receptor Superfamily, Member 7
9.
J Exp Med ; 179(2): 551-8, 1994 Feb 01.
Article in English | MEDLINE | ID: mdl-8294866

ABSTRACT

Induced activation of protein tyrosine kinase(s) is a central event in signal transduction mediated via the low affinity receptor for IgG (Fc gamma RIIIA, CD16) in natural killer (NK) cells. Tyrosine phosphorylation may affect the function of several protein directly, or indirectly by inducing their association with other tyrosine phosphorylated proteins. Here, we report that Fc gamma RIII stimulation induces activation of phosphatidylinositol (PI)-3 kinase in NK cells. Phosphotyrosine immunoprecipitates from Fc gamma RIII-stimulated NK cells contain PI-kinase activity and PI-3 kinase can be directly precipitated from them. Conversely, a series of tyrosine-phosphorylated proteins is coprecipitated with PI-3 kinase from the stimulated, but not from control cells. Analogous results obtained using Jurkat T cells expressing transfected Fc gamma RIIIA alpha ligand binding chain in association with gamma 2 or zeta 2 homodimers indicate that both complexes transduce this effect, although the Fc gamma RIIIA-zeta 2 complexes do so with greater efficiency. Accumulation of phosphoinositide D3 phosphorylated products in stimulated cells confirms PI-3 kinase activation, indicating the participation of this enzyme in Fc gamma RIIIA-mediated signal transduction.


Subject(s)
Immunoglobulin G/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Receptors, IgG/metabolism , 1-Phosphatidylinositol 4-Kinase , Antibodies, Monoclonal , Cell Line , Enzyme Activation , Humans , Ligands , Phosphatidylinositols/metabolism , Phosphoproteins/metabolism , Precipitin Tests , Signal Transduction , Tyrosine/metabolism
10.
J Exp Med ; 164(4): 1206-25, 1986 Oct 01.
Article in English | MEDLINE | ID: mdl-3093627

ABSTRACT

We show that the cytotoxins tumor necrosis factor (TNF) or lymphotoxin (LT), at concentrations of approximately 10(-11) M induce monocytic differentiation of human myeloid cell lines. After 5 d of culture in the presence of rTNF and LT, a significant proportion of the myeloid cell lines express monocyte differentiation antigens and ANAE activity, and become able to reduce nitroblue tetrazolium (NBT) and mediate low levels of ADCC against tumor target cells. These markers of differentiation, however, are maximally induced when rIFN-gamma, at concentrations as low as 4 U/ml, is present simultaneously with the cytotoxins, and the two classes of cytokines act synergistically to induce terminal differentiation. The appearance of monocytic antigens is accompanied by acquisition of morphology and other functional properties of mature monocytic cells, such as chemiluminescence and phagocytosis, and by expression of FcR for monomeric IgG. A decrease in cell proliferation accompanies induced differentiation, and is not due to the cytotoxic properties of TNF or LT, as indicated in simultaneous analysis of surface phenotype and cell cycle. The lack of cytotoxicity of TNF on the HL-60 cell line is also demonstrated by the enhancing effect of TNF on HL-60 cell growth and nucleoside uptake in the first 2 d of culture. These data show that the cytotoxins TNF and LT mediate complex effects on cells of the myelomonocytic lineage and, in synergy with IFN-gamma, can fully induce immature myeloid cells to differentiate into cells with phenotypic, functional, and proliferative characteristics of terminally differentiated myelomonocytic cells.


Subject(s)
Glycoproteins/pharmacology , Interferon-gamma/pharmacology , Leukemia, Myeloid, Acute/pathology , Lymphotoxin-alpha/pharmacology , Antibody-Dependent Cell Cytotoxicity , Cell Cycle/drug effects , Cell Differentiation/drug effects , Cell Line , Culture Media , Drug Synergism , Humans , Leukemia, Myeloid, Acute/immunology , Luminescent Measurements , Lymphotoxin-alpha/isolation & purification , Molecular Weight , Phagocytosis , Phytohemagglutinins/pharmacology , Receptors, Fc/analysis , Receptors, Transferrin/analysis , Tumor Necrosis Factor-alpha
11.
J Exp Med ; 164(1): 180-95, 1986 Jul 01.
Article in English | MEDLINE | ID: mdl-3014033

ABSTRACT

The role of HLA-DR+ cells in NK activity against CMV-infected FS4 foreskin fibroblasts and K562 erythroleukemia cells was examined. When nonadherent PBMC were depleted of either HLA-DR+ or Leu-11b+ cells by treatment with mAbs plus C, NK activity against CMV-FS4 target cells was markedly reduced. In contrast, depletion of HLA-DR+ cells had no effect on NK activity against K562 target cells. When HLA-DR-depleted cells were added to Leu-11b-depleted cells, NK activity against CMV-FS4 was restored. Negative selection experiments indicated that the HLA-DR+ cells contributing to NK activity against CMV-FS4 are not B or T cells, while negative and positive selection experiments excluded a role for monocytes. Experiments in which HLA-DR- and Leu-11b- cells were mixed in varying proportions indicated that NK(CMV-FS4) is mediated by Leu-11b+ cells, while HLA-DR+ cells provide an accessory function. Irradiation (50 GY) abolished the NK effector function of Leu-11b+ cells, but not the accessory function of HLA-DR+ cells. The NK activity against CMV-FS4 of HLA-DR- cells was restored by the addition of rIFN-alpha or of cell-free supernatants generated by coculturing PBMC or Leu-11b- cells with CMV-FS4. The ability of these supernatants to restore NK activity of HLA-DR- cells was completely abrogated by the addition of neutralizing amounts of antibody to IFN-alpha. In related experiments, neutralization of IFN-alpha in NK assays had little or no effect on NK activity against CMV-FS4, suggesting that the accessory function of HLA-DR+ cells might be mediated by alternative mechanisms in addition to the secretion of extracellular IFN-alpha.


Subject(s)
Antigen-Presenting Cells/immunology , Cytomegalovirus/immunology , Cytotoxicity, Immunologic , Fibroblasts/immunology , Histocompatibility Antigens Class II , Killer Cells, Natural/immunology , Antigen-Presenting Cells/classification , Antigen-Presenting Cells/radiation effects , Antigens, Differentiation, T-Lymphocyte , Antigens, Surface , Cell-Free System , Cytotoxicity Tests, Immunologic , Cytotoxicity, Immunologic/radiation effects , HLA-DR Antigens , Humans , Immune Sera/pharmacology , Interferon Type I/immunology , Interferon Type I/physiology , Phenotype
12.
J Exp Med ; 162(5): 1512-30, 1985 Nov 01.
Article in English | MEDLINE | ID: mdl-3932579

ABSTRACT

We characterize the natural killer (NK) cell colony-inhibiting activity (CIA) produced in supernatants from cultures of human peripheral blood lymphocytes (PBL) with NK-sensitive target cell lines, and study its relationship with NK cell-derived cytotoxic factor (NKCF). Using monoclonal antibodies (mAb) specific for NK cells or other lymphocyte populations, we unambiguously identify NK cells as the only PBL subset able to produce both NKCF and NK-CIA. We present functional and biochemical data suggesting that NKCF and NK-CIA represent the same molecule: (a) a highly significant positive correlation exists between the quantity of NKCF and NK-CIA in supernatants independently produced by different PBL subsets; (b) both NK-CIA and NKCF are induced by culture of PBL with NK-sensitive, but not with NK-insensitive cell lines, and with HLA-DR+ bone marrow cells; (c) both NKCF and NK-CIA are absorbed on the same cell lines or bone marrow cell types; (d) the two activities coelute in the same gel filtration fractions; (e) D-mannose-6-phosphate blocks both NKCF and NK-CIA activity, and prevents their absorption by K562 cells; and (f) both NKCF and NK-CIA activity are lost after 2 d at 37 degrees C. The NK-CIA-containing preparations are devoid of antiviral activity, and antiinterferon (anti-IFN) antibodies do not block the inhibitor activity of NK-CIA. The effect of NK-CIA on day 14 (early) colony-forming units of granulocytes and macrophages (CFU-GM) is synergistic with that of IFN-gamma, and this synergy is also evident on day 7 (late) CFU-GM growth. A combination of NK-CIA and IFN-gamma suppresses late CFU-GM, at concentrations of the two lymphokines that are completely ineffective when used independently. No synergy between NK-CIA and IFN-alpha or -beta was observed, due to a direct inhibitory effect of these two IFN types on late CFU-GM. Antibodies specific for tumor necrosis factor (TNF), but not those specific for lymphotoxins, inhibit both NK-CIA and NKCF activity in the NK cell-derived supernatant. Recombinant TNF, in the range of concentrations corresponding to that of the cytotoxic activity on L-929 cells present in supernatants, mediated both NKCF and NK-CIA activity.(ABSTRACT TRUNCATED AT 400 WORDS)


Subject(s)
Cytotoxicity, Immunologic , Glycoproteins/immunology , Growth Inhibitors/immunology , Interferon-gamma/immunology , Killer Cells, Natural/immunology , Animals , Bone Marrow/immunology , Bone Marrow Cells , Burkitt Lymphoma/immunology , Cell Line , Cells, Cultured , Colony-Forming Units Assay , Drug Synergism , Humans , Leukemia, Myeloid/immunology , Lymphocytes/immunology , Lymphokines/immunology , Mast-Cell Sarcoma/immunology , Mice , Tumor Necrosis Factor-alpha
13.
J Exp Med ; 188(12): 2375-80, 1998 Dec 21.
Article in English | MEDLINE | ID: mdl-9858524

ABSTRACT

Mature natural killer (NK) cells use Ca2+-dependent granule exocytosis and release of cytotoxic proteins, Fas ligand (FasL), and membrane-bound or secreted cytokines (tumor necrosis factor [TNF]-alpha) to induce target cell death. Fas belongs to the TNF receptor family of molecules, containing a conserved intracytoplasmic "death domain" that indirectly activates the caspase enzymatic cascade and ultimately apoptotic mechanisms in numerous cell types. Two additional members of this family, DR4 and DR5, transduce apoptotic signals upon binding soluble TNF-related apoptosis-inducing ligand (TRAIL) that, like FasL, belongs to the growing TNF family of molecules. Here, we report that TRAIL produced or expressed by different populations of primary human NK cells is functional, and represents a marker of differentiation or activation of these, and possibly other, cytotoxic leukocytes. During differentiation NK cells, sequentially and differentially, use distinct members of the TNF family or granule exocytosis to mediate target cell death. Phenotypically immature CD161(+)/CD56(-) NK cells mediate TRAIL-dependent but not FasL- or granule release-dependent cytotoxicity, whereas mature CD56(+) NK cells mediate the latter two.


Subject(s)
Cytotoxicity, Immunologic , Killer Cells, Natural/immunology , Membrane Glycoproteins/physiology , Tumor Necrosis Factor-alpha/physiology , Antibodies, Monoclonal/pharmacology , Antigens, CD/analysis , Apoptosis/drug effects , Apoptosis Regulatory Proteins , Calcium/metabolism , Cell Degranulation , Cell Differentiation , Cells, Cultured , Cytotoxicity, Immunologic/drug effects , Fas Ligand Protein , Humans , Interleukins/pharmacology , Killer Cells, Natural/cytology , Killer Cells, Natural/drug effects , Membrane Glycoproteins/genetics , RNA, Messenger/analysis , Receptors, Tumor Necrosis Factor/metabolism , Recombinant Fusion Proteins/pharmacology , TNF-Related Apoptosis-Inducing Ligand , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , fas Receptor/physiology
14.
J Exp Med ; 177(5): 1475-80, 1993 May 01.
Article in English | MEDLINE | ID: mdl-8478617

ABSTRACT

The transmembrane receptor for immunoglobulin G immune complexes on natural killer (NK) cells and macrophages, Fc gamma RIIIA (CD16), mediates cellular activation through a tyrosine kinase-dependent pathway. We show that Fc gamma RIII crosslinking results in activation of the src-related kinase p56lck in NK cells and demonstrate a physical association of p56lck with Fc gamma RIIIA in immunoprecipitates from NK cells obtained using anti-Fc gamma RIII antibodies or immune complexes. Our studies show that the zeta chain, the signal transducing subunit of Fc gamma RIIIA and of T cell receptor, associates with p56lck and, in NK cells, is a substrate for this kinase. Such direct association of p56lck with the zeta subunit as confirmed by demonstrating the interaction in heterologous cells transfected with cDNA expressing p56lck and zeta. Our findings demonstrate both functional and physical association of p56lck with Fc gamma RIIIA, through direct interaction of the kinase with the zeta and/or the gamma signal transducer subunits of the receptor. These data suggest a possible mechanism by which activation via Fc gamma RIIIA occurs.


Subject(s)
Killer Cells, Natural/metabolism , Protein-Tyrosine Kinases/metabolism , Receptors, IgG/metabolism , Amino Acid Sequence , Cells, Cultured , Humans , Killer Cells, Natural/enzymology , Lymphocyte Specific Protein Tyrosine Kinase p56(lck) , Molecular Sequence Data , Phosphorylation , Signal Transduction
15.
J Exp Med ; 184(5): 1845-56, 1996 Nov 01.
Article in English | MEDLINE | ID: mdl-8920872

ABSTRACT

Human natural killer (NK) cell differentiation from immature lineage negative (Lin-) umbilical cord blood cells was examined in vitro. Cells expressing differentiation antigens of mature NK cells (CD56, CD16, CD2, CD8, NKR-P1A) were generated from Lin- cells cultured with interleukin (IL)-2 and a murine bone marrow stromal cell line expressing the human membrane-bound form of stem cell factor. Two subsets of NK cells were identified in these cultures: one expressed both NKR-P1A and CD56 and, in variable proportions, all other NK cell differentiation antigens; the second subset expressed only NKR-P1A and, unlike the former, was not cytotoxic. Neither subset expressed interferon (IFN)-gamma mRNA even after stimulation with phorbol di-ester and Ca2+ ionophore, but both expressed tumor necrosis factor alpha mRNA and the cytotoxic granule-associated proteins TIA-1, perforin, and serine esterase-1. After 10-d culture with IL-2, IL-12, and irradiated B lymphoblastoid cells, approximately 45% of the NKR-P1A+/ CD56- cells became CD56+, and the same cultures contained cells capable of cytotoxicity and of IFN-gamma production. These results indicate that NKR-P1A expression in the absence of other NK cell markers defines an intermediate, functionally immature stage of NK cell differentiation, and that effector functions develop in these cells, concomitantly with CD56 expression, in the presence of IL-12. These cells likely represent the counterpart of a CD3-/NKR-P1A+/ CD56-/CD16- cell subset that, as shown here, is present both in adult and neonatal circulating lymphocytes.


Subject(s)
Antigens, Differentiation/analysis , Hematopoietic Stem Cells/drug effects , Interleukin-12/pharmacology , Killer Cells, Natural/drug effects , Lectins, C-Type , Lymphocyte Subsets/drug effects , Adult , Animals , Antigens, Surface/analysis , CD3 Complex/analysis , CD56 Antigen/analysis , Cell Differentiation , Cell Lineage , Culture Techniques/methods , Cytotoxicity, Immunologic , Fetal Blood , Humans , Interferon-gamma/biosynthesis , Leukocytes, Mononuclear , Mice , NK Cell Lectin-Like Receptor Subfamily B , Receptors, IgG/analysis
16.
J Exp Med ; 165(6): 1581-94, 1987 Jun 01.
Article in English | MEDLINE | ID: mdl-3108447

ABSTRACT

We present evidence that human peripheral blood lymphocytes, free of contaminating monocytes, rapidly produce high levels of tumor necrosis factor (TNF) when stimulated with phorbol diester and calcium ionophore, and lower but significant levels of TNF when stimulated with mitogens. These two types of inducers act preferentially on T cells, both CD4+ and CD8+. NK cells produce TNF only when stimulated with phorbol diester and calcium ionophore, and they do so at a much lower level than T cells. The procedures used in the purification of lymphocytes and the differential ability to respond to various inducers allow us to exclude that monocytes or basophils contaminating the lymphocyte preparation participate in the production of TNF. In particular, LPS, a potent inducer of TNF production from monocytes, is unable to induce significant levels of TNF in the lymphocyte preparations. The TNF produced by lymphocytes has antigenic, physicochemical, and biochemical characteristics identical to those of the TNF produced by myeloid cell lines or monocytes upon stimulation with LPS. LT is also produced by lymphocyte preparations. Production of TNF and LT proteins in response to the different inducers is paralleled by accumulation of cytoplasmic TNF and LT mRNA. Both at mRNA and at protein levels, stimulation of T lymphocytes with phorbol diester and calcium ionophore preferentially induces TNF, whereas mitogen stimulation preferentially induces LT. Our data suggest that the TNF and LT genes, two closely linked genes encoding two partially homologous proteins with almost identical biological functions, are independently regulated in lymphocytes.


Subject(s)
Glycoproteins/biosynthesis , Lymphocytes/metabolism , Lymphotoxin-alpha/biosynthesis , Calcimycin/pharmacology , Glycoproteins/genetics , Humans , Interferon-gamma/pharmacology , Killer Cells, Natural/metabolism , Lipopolysaccharides/pharmacology , Lymphocytes/classification , Lymphotoxin-alpha/genetics , Monocytes/metabolism , Phorbol 12,13-Dibutyrate , Phorbol Esters/pharmacology , Phytohemagglutinins/pharmacology , RNA, Messenger/analysis , Transcription, Genetic , Tumor Necrosis Factor-alpha
17.
J Exp Med ; 169(2): 549-67, 1989 Feb 01.
Article in English | MEDLINE | ID: mdl-2536067

ABSTRACT

In this study, we present evidence that interaction of Fc gamma R(CD16) with ligands (immune complexes or anti-CD16 antibodies) induces a rapid rise in [Ca2+]i and fast production of both inositol 1,4,5 triphosphate (IP3) and IP4 in homogeneous NK cell preparations. Part of the initial [Ca2+]i rise observed upon stimulation of NK cells with either anti-CD16 antibodies alone or after their crosslinking at the cell membrane depends on Ca2+ mobilization from intracellular stores, but sustained [Ca2+]i levels are maintained, after the initial spike, through influx of extracellular Ca2+. The [Ca2+]i rise is mediated, at least in part, by increases in IP3 after receptor-induced hydrolysis of membrane polyphosphoinositides (PPI). The role of extracellular Ca2+ in Fc gamma R(CD16)-dependent induction of lymphokine gene expression has been tested by evaluating production, mRNA accumulation and transcription of IFN-gamma and TNF in NK cells stimulated with Fc gamma R(CD16) ligands and/or rIL-2 in the presence of EGTA. Under these conditions, accumulation and transcription of both IFN-gamma and TNF mRNA induced by CD16 ligands, but not that induced by rIL-2, is completely abolished and neither cytokine can be detected at significant levels in the supernatant fluids of cells so treated. These data confirm that NK cell activation by specific ligands occurs through mechanisms distinct from those induced by IL-2, and indicate that extracellular Ca2+ represents a stringent requirement for cytokine production induced in NK cells through specific (Fc gamma R) stimulation. Our data also indicate that the [Ca2+]i rise induced upon Fc gamma R(CD16) crosslinking, though necessary, is not sufficient per se to induce activation of lymphokine genes, compatible with the hypothesis that Fc gamma R(CD16) crosslinking generates additional transducing signals that synergize with IL-2 to maximally activate NK cells.


Subject(s)
Antigens, Differentiation/physiology , Calcium/physiology , Killer Cells, Natural/physiology , Lymphocyte Activation , Lymphokines/genetics , Phosphatidylinositols/physiology , Receptors, Fc/physiology , Ethers/pharmacology , Gene Expression Regulation , Humans , In Vitro Techniques , Interferon-gamma/genetics , Ionomycin , Ligands , RNA, Messenger/genetics , Receptors, IgG , Receptors, Interleukin-2/physiology , Transcription, Genetic , Tumor Necrosis Factor-alpha/genetics
18.
J Exp Med ; 160(4): 1147-69, 1984 Oct 01.
Article in English | MEDLINE | ID: mdl-6434688

ABSTRACT

The present study shows that recombinant interleukin 2 (IL-2) purified to homogeneity induces a rapid and potent enhancement of spontaneous cytotoxicity of human peripheral blood lymphocytes. The cells mediating cytotoxicity after 18-h treatment with IL-2 have surface markers of natural killer (NK) cells and are generated from the peripheral blood subset containing spontaneous cytotoxic cells. A parallel production of gamma interferon (IFN-gamma) is induced by recombinant IL-2 (rIL-2), and NK cells appear to be the major producer cells, whereas T cells are unable to produce IFN-gamma under these experimental conditions. However, the kinetics of the enhancement of cytotoxicity are faster than those of IFN-gamma production, and monoclonal anti-IFN-gamma antibodies do not suppress this effect, making it unlikely that the IFN-gamma produced is responsible for the enhancement. The enhancement of NK cell activity induced by rIL-2 precedes any proliferative response of the lymphocytes, which is instead observed in longer-term cultures of both NK and T cells.


Subject(s)
Cytotoxicity, Immunologic , Interleukin-2/physiology , Killer Cells, Natural/immunology , Lymphocyte Activation , Adjuvants, Immunologic/physiology , Antibodies, Monoclonal/immunology , Antigens, Surface/analysis , Growth Substances/physiology , Humans , Interferon-gamma/biosynthesis , Interleukin-2/isolation & purification , Interphase , Killer Cells, Natural/cytology , Killer Cells, Natural/metabolism , Kinetics , Phenotype
19.
J Exp Med ; 158(6): 2058-80, 1983 Dec 01.
Article in English | MEDLINE | ID: mdl-6417261

ABSTRACT

Conditioned medium from phytohemagglutinin-stimulated human leukocytes contains a factor that can induce promyelocytic cell lines and certain acute myelogenous leukemia cells to differentiate along the monocytic pathway. In this report, we show that immature myeloid cells from normal bone marrow or the peripheral blood of patients with chronic myelogenous leukemia can be induced to differentiate to monocyte-like cells by immune gamma interferon (IFN gamma). We have identified IFN gamma as the predominant differentiation factor contained in the conditioned medium. Purified or recombinant IFN gamma, but not various preparations of IFN alpha or beta, can induce monocytic differentiation in myeloid cells. In cultures containing conditioned medium, the cells fail to continue myeloid maturation, and are induced to express monocyte markers and functions, such as monocyte-specific surface antigens, HLA-DR antigens, Fc receptors for monomeric immunoglobulins, nonspecific esterase, and the ability to mediate antibody-dependent, cell-mediated cytotoxicity. Even myeloid cells as mature as metamyelocytes or band cells can be induced by IFN gamma to undergo monocyte differentiation, but monocyte-specific or HLA-DR antigens are not induced in mature neutrophils. These findings reveal a previously unknown, specific function of human IFN gamma and offer new insights to the regulation of monocyte recruitment and differentiation during a virus infection or immune response.


Subject(s)
Interferon-gamma/pharmacology , Leukemia, Myeloid, Acute/pathology , Leukocytes/immunology , Bone Marrow Cells , Cell Differentiation , Cell Line , Culture Media , Histocompatibility Antigens Class II/immunology , Humans , Leukemia, Myeloid, Acute/immunology , Lymphocyte Activation , Monocytes/immunology
20.
J Exp Med ; 179(6): 1855-65, 1994 Jun 01.
Article in English | MEDLINE | ID: mdl-8195713

ABSTRACT

The p210bcr/abl tyrosine kinase appears to be responsible for initiating and maintaining the leukemic phenotype in chronic myelogenous leukemia (CML) patients. p21ras-p120GAP interactions play a central role in transducing mitogenic signals. Therefore, we investigated whether p21ras and p120GAP are regulated by p210bcr/abl, and whether this activation is functionally significant for CML cell proliferation. We report that transient expression of p210bcr/abl in fibroblast-like cells induces simultaneous activation of p21ras and inhibition of GTPase-promoting activity of p120GAP, and confirm these data showing that downregulation of p210bcr/abl expression in CML cells with bcr/abl antisense oligodeoxynucleotides induces both inhibition of p21ras activation and stimulation of GTPase-promoting activity of p120GAP. Tyrosine phosphorylation of two p120GAP-associated proteins, p190 and p62, which may affect p120GAP activity, also depends on p210bcr/abl tyrosine kinase expression. Direct dependence of these effects on the kinase activity is proven in experiments in which expression of c-MYB protein in fibroblast-like cells or downregulation of c-MYB expression resulting in analogous inhibition of CML cell proliferation does not result in the same changes. Use of specific antisense oligodeoxynucleotides to downregulate p21ras expression revealed a requirement for functional p21ras in the proliferation of Philadelphia chromosome-positive CML primary cells. Thus, the p210bcr/abl-dependent regulation of p120GAP activity is responsible, in part, for the maintenance of p21ras in the active GTP-bound form, a crucial requirement for CML cell proliferation.


Subject(s)
Cell Division/physiology , Fusion Proteins, bcr-abl/metabolism , GTP Phosphohydrolases/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Oligonucleotides, Antisense/pharmacology , Philadelphia Chromosome , Proteins/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Base Sequence , Cell Division/drug effects , Cell Line , Chromosomes, Human, Pair 15 , Chromosomes, Human, Pair 17 , Chromosomes, Human, Pair 22 , Chromosomes, Human, Pair 9 , GTPase-Activating Proteins , Homeostasis , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Molecular Sequence Data , Oncogenes/drug effects , Proto-Oncogenes/drug effects , Signal Transduction , Translocation, Genetic , Tumor Cells, Cultured , ras GTPase-Activating Proteins
SELECTION OF CITATIONS
SEARCH DETAIL