Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 103
Filter
Add more filters










Publication year range
1.
Leuk Lymphoma ; 32(5-6): 467-74, 1999 Feb.
Article in English | MEDLINE | ID: mdl-10048419

ABSTRACT

Soluble forms of Fc gammaR type III (sFc gammaRIII or sCD16) are present in many biological fluids. Their main ligand is IgG in the form of complexes. In plasma, sCD16 essentially derive from cleavage of membrane CD16 (or Fc gammaRIII) present on neutrophils and, to a lesser extent, on NK cells. Determination of sCD16 serum level during monoclonal gammopathies has demonstrated markedly reduced levels in multiple myeloma and in monoclonal gammopathy of undetermined significance (MGUS) rapidly evolving to multiple myeloma, compared to stable MGUS or controls, indicating a prognostic value for this biological parameter. The biology and functions of sCD16 are described, together with the biological significance of modifications of the sCD16 serum level in monoclonal gammopathies.


Subject(s)
Paraproteinemias/metabolism , Receptors, IgG/blood , Receptors, IgG/physiology , Biomarkers/blood , Humans , Paraproteinemias/blood , Paraproteinemias/immunology , Solubility
2.
Biochem J ; 336 ( Pt 2): 299-303, 1998 Dec 01.
Article in English | MEDLINE | ID: mdl-9820804

ABSTRACT

Leflunomide is currently in phase-III clinical trials for the treatment of rheumatoid arthritis. In this study, we have focused our efforts on the study of the mechanism of action of the active metabolite of leflunomide, A77 1726, in cells and tissue of human origin. The human high-affinity binding protein for radiolabelled A77 1726 was purified from solubilized U937 membranes by following the binding activity through the purification process and was characterized as the mitochondrial enzyme dihydro-orotate dehydrogenase (DHO-DH). The human and murine enzyme displayed identical pI and molecular mass values on SDS/PAGE (43 kDa), which contrasts notably with previous reports suggesting a molecular mass of 50 kDa for the human enzyme. DHO-DH activity was inhibited by A77 1726 and its analogue HR325 with similar potency in U937 and human spleen membrane preparations. HR325 was found to be anti-proliferative for phytohaemagglutinin-stimulated human peripheral blood mononuclear cells, at the same concentrations that caused accumulation of DHO and depletion of uridine. Supplementation of the cultures with exogenous uridine led to partial abrogation of the anti-proliferative effect. This is in line with our recent demonstration that the anti-proliferative effect in vitro of A77 1726 on lipopolysaccharide-stimulated mouse spleen cells was mediated by DHO-DH inhibition [Williamson, Yea, Robson, Curnock, Gadher, Hambleton, Woodward, Bruneau, Hambleton, Moss et al., (1995) J. Biol. Chem. 270, 22467-22472]. Thus, DHO-DH inhibition by A77 1726 and its analogues is responsible for the anti-proliferative effects in vitro of the compounds on human cells and is likely to be responsible for some of its effects in vivo.


Subject(s)
Aniline Compounds/pharmacology , Hydroxybutyrates/pharmacology , Oxidoreductases Acting on CH-CH Group Donors , Oxidoreductases/antagonists & inhibitors , Oxidoreductases/isolation & purification , Aniline Compounds/metabolism , Binding Sites , Binding, Competitive , Cell Division/drug effects , Cell Membrane/drug effects , Cell Membrane/enzymology , Crotonates , Dihydroorotate Dehydrogenase , Humans , Hydroxybutyrates/metabolism , Lymphoma/enzymology , Nitriles , Oxidoreductases/metabolism , Spleen/enzymology , Toluidines , Tumor Cells, Cultured , Uridine/metabolism , Uridine/pharmacology
3.
Eur J Immunol ; 28(7): 2101-7, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9692878

ABSTRACT

CD16 (FcgammaR type III) is a low-affinity IgG Fc receptor (R) that exists in two isoforms, a transmembrane FcgammaRIIIa expressed by NK cells and monocytes, and a phosphatidylinositol-linked FcgammaRIIIb expressed by neutrophils. A soluble form of CD16 (sCD16) circulates in plasma. The cleavage site and the nature of the enzyme(s) involved in production of sCD16 were investigated. Soluble CD16 was purified to apparent homogeneity from human serum by eight steps, including anion exchange and immunoaffinity chromatography. Serum sCD16 was sequenced at both ends, as well as a recombinant form of sCD16 used as control. N-terminal sequencing demonstrated that serum sCD16 originates from neutrophil FcgammaRIIIb and C-terminal sequencing suggested that the cleavage site is between Val 196 and Ser 197, close to the membrane anchor. Addition of a hydroxamate-based inhibitor of Zn2+ metalloproteinases (RU36156) led to a dramatic decrease of sCD16 production by phorbol 12-myristate 13-acetate-activated neutrophils, whereas inhibitors of serine proteinases had no significant effect, showing the metalloproteinase dependence of this cleavage process.


Subject(s)
Receptors, IgG/biosynthesis , Amino Acid Sequence , Humans , Molecular Sequence Data , Neutrophils/metabolism , Receptors, IgG/chemistry , Serine Proteinase Inhibitors/pharmacology , Tetradecanoylphorbol Acetate/pharmacology
4.
Immunotechnology ; 4(1): 71-87, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9661816

ABSTRACT

BACKGROUND: Few antibodies are available to study the function of the Fc gamma RII murine immunoglobulin receptor. Human phage display libraries represent a potential source of single-chain Fv (sFv) to facilitate the study of the Fc gamma RII murine immunoglobulin receptor. OBJECTIVES: To isolate human sFv specific for mouse Fc gamma RII. STUDY DESIGN: Two human phage display libraries were selected for reactivity to mouse Fc gamma RII. Those human anti-mouse Fc gamma RII sFv that were derived from the libraries were characterized with respect to kinetics, cellular binding, epitope specificity and amino acid sequence. RESULTS: Nine anti-mouse Fc gamma RII sFv molecules were isolated from two human phage display libraries (Marks et al., J Mol Biol 1991;222:581-597; Sheets et al., Proc Natl Acad Sci USA, in press). Surface plasmon resonance (SPR) analysis revealed that the human anti-mouse Fc gamma RII sFv had off-rates ranging from 10(-2) to 10(-3) s-1, with KD values calculated to range between 10(-7) and 10(-9) M. The binding of the FITC-labeled human anti-mouse Fc gamma RII sFv to mouse peritoneal neutrophils was not detected by flow cytometry, due to the rapid off-rates of these monomeric proteins. However, when the human anti-mouse Fc gamma RII sFv were coated on yellow-green latex particles, all of the human sFv were found to specifically bind to mouse peritoneal neutrophils. Deglycosylation of mouse Fc gamma RII did not diminish the binding of these sFv, suggesting that the sFv molecules recognize a polypeptide epitope on murine Fc gamma RII. In contrast, denaturation of mouse Fc gamma RII dramatically reduced the binding of the human sFv, suggesting that the epitopes are conformational. Sequence analysis of the human anti-mouse Fc gamma RII sFv revealed a high degree of structural similarity among the nine sFv. The DP73 VH gene segment was utilized by four of the nine sFv, while seven of the nine sFv contained the DPL16 V lambda gene segment. The sequence similarities between these sFv suggested that several of the human sFv may recognize a common epitope on mouse Fc gamma RII. Epitope mapping studies demonstrated that eight of the nine human anti-mouse Fc gamma RII sFv recognized overlapping epitopes. All of these human anti-mouse Fc gamma RII sFv competed with the 2.4G2 rat monoclonal anti-mouse Fc gamma RII/III antibody for binding with mouse Fc gamma RII, suggesting that the targeted epitopes reside in or near the Fc binding pocket of mouse Fc gamma RII. CONCLUSIONS: The availability of novel sFv recognizing mouse Fc gamma RII will facilitate the study of receptor triggering events. Such sFv may prove useful to engage murine Fc gamma RII for targeted cytotoxicity or immunization strategies.


Subject(s)
Bacteriophages/genetics , Gene Library , Immunoglobulin Fragments/genetics , Immunoglobulin Variable Region/genetics , Receptors, IgG/immunology , Amino Acid Sequence , Animals , Antibodies/genetics , Antibodies, Monoclonal/immunology , Antigen-Antibody Reactions/immunology , Bacteriophages/ultrastructure , Base Sequence , Carbohydrates/analysis , Cell Membrane/chemistry , Epitope Mapping , Epitopes/chemistry , Epitopes/immunology , Humans , Immunoglobulin Fragments/analysis , Immunoglobulin Fragments/immunology , Kinetics , Mice , Molecular Sequence Data
5.
Glycoconj J ; 15(9): 905-14, 1998 Sep.
Article in English | MEDLINE | ID: mdl-10052594

ABSTRACT

N-glycans of a recombinant mouse soluble Fcgamma receptor II (sFcgammaRII) expressed in baby hamster kidney cells were released from glycopeptides by digestion with glycoamidase A (from sweet almond), and the reducing ends of the oligosaccharides were reductively aminated with 2-aminopyridine. The derivatized N-glycans were separated and structurally identified by a three-dimensional high-performance liquid chromatography (HPLC) mapping technique on three kinds of HPLC columns [Takahashi, et al. (1995) Anal. Biochem. 226:139-46]. Eighteen different major N-glycan structures were identified, of which six were neutral (45%), five mono-sialyl (49%), one di-sialyl (4.6%), five tri-sialyl (1.1%), and one tetra-sialyl (0.3%). All N-glycan structures determined were complex type with fucosylation at the N-acetylglucosamine residue of the reducing end, and N-acetylneuraminic acid, when present, was alpha-(2,3)-linked. The existence of a unique structure containing both N-acetylgalactosamine and alpha-(2,3)-N-acetylneuraminic acid residues at the reducing ends, as below, was confirmed by MALDI-TOF mass spectrometry. Carbohydrate sequence [see text]


Subject(s)
Polysaccharides/chemistry , Receptors, IgG/chemistry , Animals , Carbohydrate Conformation , Carbohydrate Sequence , Cells, Cultured , Chromatography, High Pressure Liquid , Glycoproteins/chemistry , Mice , Molecular Sequence Data , Oligosaccharides/chemistry , Receptors, IgG/genetics , Recombinant Proteins/chemistry , Solubility , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
6.
J Immunol ; 159(9): 4155-64, 1997 Nov 01.
Article in English | MEDLINE | ID: mdl-9379008

ABSTRACT

The cell cycle events accompanying TGF-beta1-induced growth arrest of normal mouse resting B lymphocytes stimulated by LPS were investigated. We showed that TGF-beta1 prevents the retinoblastoma protein (pRb) phosphorylation and induces growth arrest in mid- to late G1. To explore the molecular basis of the effect of TGF-beta1, we analyzed the in vitro kinase activities of cyclin/cyclin-dependent kinase (cdk) complexes involved in the progression through G1 phase and in the G1/S transition, by using the glutathione S-transferase-pRb fusion protein as a substrate. Cdk2-associated kinase activity was strongly induced in mitogen-treated B cells. It was dramatically inhibited by TGF-beta1 as were the cyclin E- and cyclin A-dependent kinase activities. TGF-beta1 treatment had no significant effect on the expression of two G1/S phase proteins, cyclin E and cdk2. In contrast, the appearance of cyclin A, occuring in late G1 phase, was almost totally inhibited by TGF-beta1. We also showed that expression of the cdk inhibitor protein p27Kip1 decreased as cells progressed through the G1 phase. An accumulation of p27 was found in TGF-beta1-treated cells, showing that TGF-beta1 prevented LPS-induced decline of p27. Finally we found that the lack of kinase activity associated with cyclin E/cdk2 complexes was correlated with increased amounts of cdk2- and cyclin E-bound p27. Overall, these results suggest that both cyclin A and cdk2 may be active participants in the TGF-beta1-induced cell cycle arrest in normal mouse B cells and indicate the involvement of p27 in this mechanism.


Subject(s)
B-Lymphocytes/metabolism , Cell Cycle Proteins/metabolism , Cell Cycle/immunology , Lipopolysaccharides/pharmacology , Lymphocyte Activation/drug effects , Transforming Growth Factor beta/pharmacology , Animals , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Cell Cycle Proteins/immunology , Flow Cytometry , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Signal Transduction/immunology
7.
Eur J Immunol ; 27(8): 1928-32, 1997 Aug.
Article in English | MEDLINE | ID: mdl-9295028

ABSTRACT

Binding of the Fc region of IgG antibodies to low affinity Fc gamma receptors (Fc gammaR) triggers important effector functions in the immune system. The type IIIb Fc gammaR (Fc gammaRIIIb or CD16) is a heavily glycosylated protein anchored to the membrane of neutrophils by a glycosylphosphatidylinositol link. This receptor contributes to cell activation by IgG immune complexes. To better understand the nature of the ligand-receptor association, we have studied the affinity and kinetics of the interaction between human IgG subclasses and two soluble forms of Fc gammaRIIIb (sFc gammaRIIIb or sCD16) corresponding to the 188 N-terminal amino acids of the extracellular region of the receptor, a glycosylated one made in eucaryotic cells (euc.sCD16) and a non-glycosylated one (proc.sCD16) made in Escherichia coli. Experiments using a BIAcore instrument, to measure protein binding in real time, showed that monomeric human IgG1 and IgG3, but not IgG2, IgG4, IgA and divalent antigen-binding fragments (F(ab')2) of IgG1, bound to immobilized euc.sCD16 with an affinity constant (K(A)) of 1.3 +/- 0.6 x 10(6) M(-1) and 2.6 +/- 0.4 x 10(5) M(-1), respectively. The affinity constant of proc.sCD16 for human IgG1 was in the same range (1.1 +/- 0.2 x 10(6) M(-1)), whereas that for human IgG3 was twofold higher (4.2 +/- 0.4 x 10(5) M(-1)). The specificity of the non-glycosylated receptor for human IgG subclasses bound to Sepharose was IgG1 > IgG3 >> IgG4 >>> IgG2. Thus, the extracellular polypeptide of Fc gammaRIIIb dictates the interaction of the receptor with IgG subclasses although glycosylation plays an inhibitory role in the interaction with human IgG3.


Subject(s)
Immunoglobulin G/metabolism , Receptors, IgG/metabolism , Animals , Cricetinae , Escherichia coli/genetics , Eukaryotic Cells , Glycosylation , Humans , Immunoglobulin G/chemistry , Immunoglobulin G/classification , In Vitro Techniques , Kinetics , Prokaryotic Cells , Protein Binding , Receptors, IgG/chemistry , Receptors, IgG/classification , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Solubility
8.
Clin Cancer Res ; 3(8): 1443-51, 1997 Aug.
Article in English | MEDLINE | ID: mdl-9815830

ABSTRACT

gp130 acts as a common transducing signal chain for all receptors belonging to the interleukin (IL)-6 receptor family. The IL-6-related cytokines [IL-6, IL-11, oncostatin M (OSM), leukemia inhibitory factor, ciliary neurotrophic factor, and cardiotrophin] often modulate tumor phenotype and control the proliferation of many tumor cell lines. We demonstrate that melanoma cell lines release, in vitro and in vivo (when transplanted in nude mice), soluble gp130 (sgp130), a potential antagonist of cytokines from the IL-6 family. Biochemical analysis revealed that sgp130 derived from melanoma patients' sera or from culture supernatants of melanoma cell lines is a Mr 104,000 protein that resolved after deglycosylation as a Mr 58,000 protein. PCR and Northern blot analysis only identified one gp130 membrane mRNA, suggesting that the soluble form of gp130 is generated by proteolytic cleavage. OSM reproducibly increases sgp130 released by melanoma cell lines, whereas leukemia inhibitory factor stimulates the production of sgp130 in only one of three cell lines tested. This tumor-derived sgp130 is functional because it binds in solution to the IL-6-soluble IL-6 receptor (gp80) complex. Recombinant sgp130 inhibits the growth inhibitory activity of the IL-6-soluble IL-6 receptor complex and OSM on some melanoma cell lines. Therefore, this soluble gp130 represents a natural antagonist of cytokines from the IL-6 family.


Subject(s)
Antigens, CD/physiology , Cytokines/antagonists & inhibitors , Interleukin-6/antagonists & inhibitors , Melanoma/physiopathology , Membrane Glycoproteins/physiology , Receptors, Interleukin-6/physiology , Animals , Antigens, CD/biosynthesis , Ciliary Neurotrophic Factor , Cytokine Receptor gp130 , Female , Growth Inhibitors/antagonists & inhibitors , Humans , Interleukin-11/antagonists & inhibitors , Leukemia Inhibitory Factor , Lymphokines/antagonists & inhibitors , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/metabolism , Mice , Mice, Nude , Nerve Tissue Proteins/antagonists & inhibitors , Oncostatin M , Peptides/antagonists & inhibitors , Receptors, Interleukin-6/antagonists & inhibitors , Signal Transduction , Transplantation, Heterologous , Tumor Cells, Cultured
9.
Adv Exp Med Biol ; 417: 345-52, 1997.
Article in English | MEDLINE | ID: mdl-9286384

ABSTRACT

Fc gamma RIII (CD16), a low affinity FcR which binds IgG-containing immune-complexes, exists under membrane-associated forms and under a soluble form (sFc gamma RIII). The latter, present in biological fluids (serum, saliva), is generated by proteolytic cleavage of the two membrane-associated Fc gamma RIII isoforms, Fc gamma RIII-A (expressed by macrophages and NK cells) and Fc gamma RIII-B (expressed exclusively by neutrophils). Herein we demonstrate that dendritic cells (DCs), generated by culturing monocytes with GM-CSF and IL-4, bind biotinylated recombinant sFc gamma RIII. This binding is specific and involves the complement receptor CR3 (CD11b/CD18) and CR4 (CD11c/CD18). Indeed, preincubation of DCs with anti-CD11b and anti-CD11c mAbs decreased by 52% and 62% respectively the binding with sFc gamma RIII. Moreover, electron microscopy showed that binding of gold-labeled sFc gamma RIII to DCs maintained at 4 degrees C occurred within clathrin-coated pits. Once internalized, at 37 degrees C, sFc gamma RIII entered the endocytic pathway and reached the MHC class II compartments. Furthermore, DCs incubated for 48 h with multivalent sFc gamma RIII expressed increased levels of CD40, CD80, CD86, CD54, CD58, HLA class I and class II molecules and decreased levels of CD23 and CD32. These effects result in an increased capacity of DCs to trigger proliferative responses by CD4+ CD45RA+ allogeneic T cells. RT-PCR amplification demonstrated that incubation of DCs for 20 h in the presence of multivalent sFc gamma RIII induced the appearance of GM-CSF and IL-12 p40 mRNA. Among the cytokines constitutively expressed, IL-1 beta and IL-8 were strongly up-regulated whereas IL-6 and IL-12 p35 mRNA were increased to a lesser extent and the expression of MIP-1 alpha mRNA remained constant. Finally, ELISA tests demonstrated that DCs incubated with multivalent sFc gamma RIII secreted the cytokines IL-1 beta, IL-6, IL-8, GM-CSF and IL-12 p75. Thus, while becoming internalized sFc gamma RIII could affect the capacity of DCs to present antigens and, via the induction of accessory molecules and the release of the IL-12 p75 protein, could initiate Th1 type immune response.


Subject(s)
Cytokines/biosynthesis , Dendritic Cells/cytology , Dendritic Cells/immunology , Interleukin-12/biosynthesis , Membrane Proteins , Receptors, IgG/metabolism , Antigen Presentation , Binding Sites , Cell Adhesion Molecules/metabolism , Cell Differentiation , Endocytosis , HLA Antigens/metabolism , Histocompatibility Antigens Class II/metabolism , Humans , In Vitro Techniques , Isoantigens , Lymphocyte Culture Test, Mixed , Receptors, Complement/metabolism , Receptors, IgE/metabolism , Solubility , Th1 Cells/immunology
10.
Int Rev Immunol ; 16(1-2): 87-111, 1997.
Article in English | MEDLINE | ID: mdl-9651787

ABSTRACT

Soluble Fc gamma receptors are produced by cleavage of the membrane receptors or by alternative splicing. They are found in biologic fluids. After a brief description of the structure and mode of production of soluble Fc gamma R, we address the question of ligands and function of the soluble Fc gamma R by using recombinant molecules and transgenic animals. We show that soluble Fc gamma R are not only IgG-binding factors which interfere with, and block, Fc-dependent immune reactions but also molecules that interact, in vitro, with non-Ig-ligands such as CR3 and CR4 and are trigger or regulate immune functions via these receptors.


Subject(s)
Receptors, IgG/immunology , Animals , Humans , Ligands , Mice , Mice, Transgenic , Solubility
11.
Int Rev Immunol ; 16(1-2): 187-203, 1997.
Article in English | MEDLINE | ID: mdl-9651791

ABSTRACT

Immature dendritic cells are antigen presenting cells highly specialized for capturing and processing foreign protein antigens. These cells express Fc gamma RII and Fc epsilon RI which, by their ability to internalize and use the endocytic pathway, increase their capacity to process antigens. Immature dendritic cells, such as epidermal Langerhans cells, also release soluble forms of Fc gamma RII. These latter molecules are likely to compete with the membrane-associated Fc gamma R to diminish or abrogate the capacity of dendritic cells to present immune complexes, as suggested by our in vitro experiments using both human and mouse epidermal Langerhans cells. However, when dendritic cells mature in vitro and become efficient stimulators of resting T cells, they rapidly down-regulate and sometimes completely abolish the expression of their membrane-associated Fc gamma R and Fc epsilon RI. Consequently, they lose or at least strongly diminish their capacity to capture immune complexes. At this stage, the release of soluble Fc gamma R by dendritic cells is also markedly diminished. One can hypothesize that the membrane-associated Fc gamma RII and the soluble Fc gamma RII are molecules expressed when dendritic cells are potent capturing and processing cells, the soluble Fc gamma RII molecule acting by competition as a negative regulatory element on the Fc gamma RII-mediated internalization of IgG-containing immune complexes. Thus, the expression of membrane-associated Fc gamma R and Fc epsilon RI, as well as the release of soluble Fc gamma R, would seem to characterize the immature stage of dendritic cells.


Subject(s)
Langerhans Cells/chemistry , Receptors, Fc/physiology , Humans
12.
Hybridoma ; 16(6): 519-28, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9455704

ABSTRACT

Four mouse anti-human Fc gamma RII (CD32) (6C4, 2B2, 3D3, 93.4) (IgG1, kappa) and one anti-human Fc gamma RIII (CD16) (7.5.4) IgG1, kappa) MAbs were raised. An in vitro switch variant, 7.5.4Sw50 (IgG2b, kappa), was also derived from the 7.5.4 MAb. 6C4, 2B2, and 3D3 MAbs bind both Fc gamma RIIa and Fc gamma RIIb isoforms. Two of them (6C4 and 2B2 MAbs) allow a complete blockade of the binding of immune complexes to Fc gamma RII. All three MAbs immunoprecipitate the receptor and bind both its glycosylated and nonglycosylated forms. The fourth anti Fc gamma RII MAb, 93.4, directed against the intracellular region of Fc gamma RIIa1/2, allows its detection by Western blotting only when it is not phosphorylated. The 7.5.4 MAb binds both Fc gamma RIIIa and Fc gamma RIIIb, can be used in Western blotting and does not inhibit aggregated IgG binding. ELISA using IV.3 (anti-Fc gamma RIIa1/2)/6C4 and 3G8 (anti-Fc gamma RIIIa/b)/7.5.4Sw50 MAb pairs make it possible to detect soluble Fc gamma RIIa1/2 and Fc gamma RIII, with a sensitivity of 200 pg/mL and 1 ng/mL, respectively. Surface plasmon resonance analyses indicated that the KD of two of the three anti-Fc gamma RII and of the anti-Fc gamma RIII are in the same order of magnitude (6C4: 0.78 nM, 2B2: 0.28 nM, 7.5.4: 0.47 nM). The anti-Fc gamma RII 3D3 MAb exhibits an off-rate constant higher than the 6C4 and 2B2 MAbs and a KD of 2.19 nM.


Subject(s)
Antibodies, Monoclonal/immunology , Immunoglobulin Fc Fragments/immunology , Receptors, IgG/immunology , Animals , Antibodies, Monoclonal/metabolism , Binding Sites , Blotting, Western , CHO Cells , Cricetinae , Enzyme-Linked Immunosorbent Assay , Humans , Mice , Mice, Inbred BALB C , Precipitin Tests
13.
Br J Haematol ; 95(4): 660-5, 1996 Dec.
Article in English | MEDLINE | ID: mdl-8982042

ABSTRACT

There are no well-defined host markers to determine which patients with a diagnosis of monoclonal gammopathy of undetermined significance (MGUS) will progress to multiple myeloma (MM). In this preliminary study we measured plasmatic soluble Fe gamma receptor type III (sFe gamma RIII or sCD16) in 54 individuals with MGUS. 35 patients with multiple myeloma (MM) and 29 healthy controls. We confirmed, through receiver operating characteristic (ROC) curve analysis, that a low level of sCD16 discriminates MM patients from controls. Indeed, for a sCD16 value of 1.3 micrograms/ml, the sensitivity, as well as the specificity, of this discrimination were both equal to 83%, i.e. 83% of MM patients had a plasmatic sCD16 value < 1.3 micrograms/ml compared with only 17% of controls. Moreover, ROC curve analysis showed that a low sCD16 level also identifies among MGUS patients a subgroup of patients who rapidly progress towards multiple myeloma: in this comparison, for a sCD16 level of 1.3 micrograms/ml. sensitivity and specificity were 70% and 79% respectively. Therefore a low sCD16 level in MGUS indicated a high likelihood of rapid evolution of MM. In contrast to sCD16, soluble IL-6R did not appear to be discriminant in this study.


Subject(s)
Multiple Myeloma/diagnosis , Paraproteinemias/diagnosis , Receptors, IgG/blood , Adult , Aged , Aged, 80 and over , Biomarkers , Disease Progression , Enzyme-Linked Immunosorbent Assay , Female , Humans , Interleukin-6/blood , Male , Middle Aged , Prognosis , Sensitivity and Specificity
15.
J Immunol ; 157(10): 4707-16, 1996 Nov 15.
Article in English | MEDLINE | ID: mdl-8906852

ABSTRACT

A quantitative trait locus for increased IgG serum levels in the NOD mouse strain was mapped to distal chromosome 1, close to the fcgr2 locus encoding the low-affinity type II receptor for the Fc portion of IgG (Fc gamma RII). Expression of membrane-inserted (b2) and soluble (b3) isoforms of Fc gamma RII was strongly decreased in macrophages of NOD compared with C57BL/6 (B6) mice. In contrast, B cell-specific (Fc gamma RIIb1) isoform was only slightly decreased and Fc gamma RIII was not altered. This Fc gamma RII regulatory defect was cis-encoded by fcgr2 or by a closely linked locus, occurred at the mRNA level, and was associated with multiple mutations in the fcgr2 gene promoter. In relation with this defect, binding of IgG1- and IgG2b- but not IgG2a-opsonized RBC by macrophages of NOD and congenic B6.NOD-fcgr2 mice was severely impaired, but was normal in macrophages of NOD.B6-fcgr2 congenic mice, indicating that Fc gamma RII plays a nondispensable role in binding of IgG1 and IgG2b isotypes. Likewise, serum levels of IgG1 and IgG2b but not IgG2a were up-regulated in NOD compared with NOD.B6-fcgr2 congenic mice. These findings indicate that macrophage Fc gamma RII may regulate serum IgG1 and IgG2b through their catabolism, and validate the NOD strain as a model to investigate the functions of Fc gamma RII isoforms.


Subject(s)
Gene Expression Regulation/immunology , Genes, Immunoglobulin , Genetic Linkage/immunology , Immunoglobulin G/blood , Immunoglobulin G/genetics , Macrophages/immunology , Receptors, IgG/biosynthesis , Receptors, IgG/genetics , Up-Regulation/immunology , Animals , Base Sequence , Chromosome Mapping , Macrophages/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Inbred NOD , Molecular Sequence Data
16.
Int J Cancer ; 68(2): 219-27, 1996 Oct 09.
Article in English | MEDLINE | ID: mdl-8900432

ABSTRACT

We have previously shown that Fc gamma receptor type II B1 (Fc(gamma)RIIB1), when expressed on non-lymphoid tumor cells, significantly enhanced their tumorigenic phenotype. This study elucidates the role of the intracellular domain of Fc(gamma)RIIB1 in the enhancement of the malignant phenotype of polyoma-transformed 3T3 cells. We investigated the tumorigenic potential conferred by different variants of the receptor: Fc(gamma)RIIB1, a full-length receptor (B1) whose intracellular region is encoded by exons 8, 9 and 10; Fc(gamma)RIIB2, a spliced variant (B2) whose cytoplasmic domain comprises exons 9 and 10 and lacks exon 8; and Fc(gamma)RIIB1-CT53, a deleted mutant whose cytoplasmic domain contains the fragment encoded by exon 8 alone. We have investigated various properties of cells transfected with each of the above variants: tumorigenicity in syngeneic mice, formation of colonies in soft agar, growth rate, production of soluble receptor and capping of the ligand-bound receptor. Results show that while the presence of exon 8 did not enhance growth rate in vitro or production of soluble Fc(gamma)R, it did enhance the tumorigenic phenotype of transfected cells (both in vivo and in vitro growth in soft agar). B1-expressing cells exhibited a significantly higher tumorigenic phenotype than B2 cells. The presence of exon 8 alone (CT53 mutant) conferred the transfected cells a higher tumorigenic phenotype than Fc(gamma)R-negative control cells but lower than intact B1 or B2 cells, indicating that the presence of B1-specific exon 8 is not sufficient but that the presence of an intact B1 intracellular domain is essential, for conferring the high tumorigenicity phenotype upon cells. We conclude that the capping, following ligand binding contributed by exon 8, and the function contributed by the specific localization of exons 9 and 10 in B1 cells may determine their malignant phenotype.


Subject(s)
Antigens, CD/physiology , Neoplasms, Experimental/etiology , Receptors, IgG/physiology , 3T3 Cells , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Cell Line, Transformed , Disease Progression , Female , Genetic Vectors , Mice , Mice, Inbred BALB C , Phenotype , Receptors, IgG/genetics , Receptors, IgG/metabolism , Transfection , Tumor Stem Cell Assay
17.
J Immunol ; 157(3): 1184-92, 1996 Aug 01.
Article in English | MEDLINE | ID: mdl-8757624

ABSTRACT

The type III-B Fcgamma receptor (FcgammaRIII-B) is a glycosyl-phosphatidylinositol-linked receptor found on human neutrophils. A soluble form of FcgammaRIII-B (sCD16) corresponding to the extracellular region of the receptor circulates in plasma. In the present work, we have identified membrane receptors for sCD16. Soluble CD16 bound to CR3 (CDllb/CD18)- and CR4 (CDllc/CD18)- positive leukocytes and cell lines, the labeling was inhibited by anti-CD11b, CD11c or CD18 mAbs, and the up-regulation of CR3 and CR4 led to an increased fixation of sCD16. Transfected eukaryotic cells expressing recombinant CD11b/CD18 or CD11c/CD18 heterodimers but not those expressing CD11a/CD18 bound sCD16. Moreover, the lectin-like binding site of CR3 is probably involved in the interaction with sCD16, as suggested by inhibition studies using mAbs against CR3 or sugars such as N-acetyl D-glucosamine, alpha- or beta-methyl D-glucoside, alpha- or beta-methyl D-mannoside, or zymosan. Thus, the complement receptors CR3 and CR4 are membrane receptors for sCD16. Through this interaction, sCD16 induces a CR3-dependent production of IL-6 and IL-8 by monocytes. These results suggest that sCD16 plays a regulatory role in inflammatory processes and provide a molecular basis for the interaction between FcgammaRIII-B and CR3 described on the cell membrane.


Subject(s)
Monocytes/immunology , Neutrophil Activation , Neutrophils/immunology , Receptors, Complement/metabolism , Receptors, IgG/metabolism , Antibodies, Monoclonal/immunology , CD11 Antigens/metabolism , CD18 Antigens/metabolism , Cell Line , Flow Cytometry , Humans , Interleukin-6/metabolism , Interleukin-8/metabolism , Macrophage-1 Antigen/metabolism , Monocytes/metabolism , Neutrophils/metabolism , Structure-Activity Relationship , Up-Regulation
18.
Int J Cancer ; 65(2): 221-9, 1996 Jan 17.
Article in English | MEDLINE | ID: mdl-8567121

ABSTRACT

The murine receptor for the Fc portion of IgG is a molecule expressed by cells of the immune system. This study suggests the hypothesis that Fc gamma receptor type II B I (Fc gamma RIIB I) functions as a progression-enhancing factor when expressed ectopically on non-lymphoid tumor cells. It has been shown previously that BALB/c 3T3 cells transformed in vitro with polyoma virus (PyV) do not express Fc gamma RII but acquire the expression of this receptor following an in vivo passage in syngeneic mice. The specific Fc gamma RII transcript present in tumor cells was identified in this report as Fc gamma RIIB I (BI). In order to determine whether or not the ectopically expressed Fc gamma RII plays a role in the progression of these transformed cells, PyV-transformed 3T3 cells were transfected with BI-cDNA. The BI transfected cells were tested for their ability to form local tumors in syngeneic mice, as compared to transfected cells which express the co-transfecting neomycine resistance (neores) DNA alone or together with the lacZ gene. Fc gamma RIIB I expressors exhibited a significantly higher tumorigenic phenotype than FcR-negative controls, though both types of cells exhibited the same growth curve in vitro. The ability of Fc gamma RIIB I to act as a potentially tumorgenicity-enhancing factor was also demonstrated as Fc gamma RII was expressed by tumor cells, originating from inoculated Fc gamma RIIB I-transfected cells, or from inoculation of a mixture of receptor-positive and -negative cells. B I-expressing cells dominated the tumor-cell population over non-expressors. This dominance strengthened the hypothesis that FcR plays a role in tumor progression in vivo.


Subject(s)
Cell Transformation, Neoplastic/immunology , Cell Transformation, Viral/immunology , DNA, Complementary/genetics , Polyomavirus , Receptors, IgG/biosynthesis , 3T3 Cells , Animals , Cell Transformation, Neoplastic/genetics , DNA, Complementary/isolation & purification , Gene Transfer Techniques , Mice , Mice, Nude , Neoplasm Transplantation , Receptors, IgG/genetics
19.
Immunobiology ; 196(4): 387-98, 1996.
Article in English | MEDLINE | ID: mdl-9061379

ABSTRACT

CD16, the low affinity receptor for monomeric IgG (Fc gamma RIIIA), is a well characterized activation molecule on NK cells. In this study we investigated the role of CD16 in NK cell-mediated regulation of immunoglobulin production. Cocultures of the CD16+ human NK clone CNK6 and highly purified SAC/IL-2-activated B lymphocytes with various CD16 antibodies showed significantly diminished NK-enhanced immunoglobulin production in a dose-dependent manner, indicating that CD16 is relevant in NK-B cell interaction. Similarly, recombinant soluble CD16 incubated with B cells before cultures, suppressed the NK cell-stimulated B cell antibody response. Enhanced immunoglobulin production was also inhibited by Fc-specific F(ab')2 anti-body fragments. Coculture of NK cells with B lymphocytes resulted in induction of mRNA for IFN-gamma and TNF-alpha. The accumulation of mRNA for these cytokines was prevented by addition of CD16 and Fc-specific antibodies. It is proposed that interaction of CD16 on NK cells with B cell bound immunoglobulin leads to induction of cytokines in NK cells which stimulate immunoglobulin production by B cells.


Subject(s)
B-Lymphocytes/immunology , Cell Communication/immunology , Killer Cells, Natural/immunology , Receptors, IgG/physiology , Antibodies, Monoclonal/pharmacology , B-Lymphocytes/metabolism , Coculture Techniques , Cytokines/biosynthesis , Cytokines/genetics , Humans , Immunoglobulins/biosynthesis , Killer Cells, Natural/metabolism , Lymphocyte Activation , Protein Binding/immunology , Receptors, IgG/immunology , Receptors, IgG/metabolism , Solubility
20.
J Exp Med ; 182(6): 1717-26, 1995 Dec 01.
Article in English | MEDLINE | ID: mdl-7500016

ABSTRACT

Immunoglobulin G-binding factors (IgG-BF), which are produced by cells of the immune system, inhibit antibody production. In this paper, we show that transforming growth factor-beta (TGF-beta) suppresses secondary in vitro anti-sheep red blood cell responses of mouse splenocytes and lipopolysaccharide- or anti-IgM-stimulated mouse B cell responses in a way similar to, and with the same kinetics as, rodent IgG-BF. Moreover, the immunosuppressive activity of IgG-BF was totally neutralized by polyclonal and monoclonal anti-TGF-beta antibodies and it eluted with TGF-beta by gel exclusion chromatography, suggesting that a TGF-beta-like immunosuppressive factor is present in IgG-BF. We also show that TGF-beta behaves as an IgG-BF since it binds to insolubilized IgG, but not to insolubilized F(ab')2 or bovine serum albumin. Altogether, the data support the concept of a biological role for TGF-beta in the IgG-mediated negative feedback of antibody responses.


Subject(s)
Antibody Formation/drug effects , B-Lymphocytes/immunology , Immunosuppressive Agents , Lymphokines/physiology , Prostatic Secretory Proteins , Receptors, IgG/physiology , Transforming Growth Factor beta/physiology , Animals , Cells, Cultured , Heterozygote , Lymphocyte Activation/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Receptors, IgG/chemistry , Solubility , Spleen/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...