Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.465
Filter
Add more filters

Publication year range
1.
Nat Genet ; 18(4): 325-30, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9537413

ABSTRACT

Stable transduction of mammalian cells typically involves random integration of viral vectors by non-homologous recombination. Here we report that vectors based on adeno-associated virus (AAV) can efficiently modify homologous human chromosomal target sequences. Both integrated neomycin phosphotransferase genes and the hypoxanthine phosphoribosyltransferase gene were targeted by AAV vectors. Site-specific genetic modifications could be introduced into approximately 1% of cells, with the highest targeting rates occurring in normal human fibroblasts. These results suggest that AAV vectors could be used to introduce specific genetic changes into the genomic DNA of a wide variety of mammalian cells, including therapeutic gene targeting applications.


Subject(s)
Adenoviridae/genetics , Gene Targeting , Genetic Vectors/genetics , Dose-Response Relationship, Drug , Gene Transfer Techniques , Genes/genetics , Genes, Viral/genetics , Genetic Vectors/pharmacology , Humans , Hypoxanthine Phosphoribosyltransferase/genetics , Male , Mutation/genetics , Neomycin , Phosphotransferases (Alcohol Group Acceptor)/genetics , Recombinant Proteins/genetics , Recombination, Genetic/drug effects , Transfection/genetics , Transfection/methods , Tumor Cells, Cultured
2.
Nat Med ; 4(6): 685-90, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9623977

ABSTRACT

The transfer of apoptosis genes to tumors is one of the most promising strategies for cancer gene therapy. We have shown that massive apoptosis occurs when wild-type p53 expression is induced in glioma cells carrying a p53 gene mutation. However, adenovirus-mediated p53 gene transfer is ineffective in causing apoptosis in glioma cells that retain a wild-type p53 genotype. We evaluated the effect of E2F-1 overexpression on the growth of gliomas in vitro and in vivo. In the in vitro study, the adenovirus-mediated transfer of exogenous E2F-1 protein precipitated generalized apoptosis in gliomas. The treatment with Ad5CMV-E2F-1 of nude mice carrying subcutaneous gliomas arrested tumor growth. Our results indicate that E2F-1 has anti-glioma activity in vitro and in vivo.


Subject(s)
Apoptosis/physiology , Carrier Proteins , Glioma/genetics , Transcription Factors/genetics , Adenoviruses, Human/genetics , Animals , Apoptosis/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/physiology , Cell Death/genetics , Cell Death/physiology , Cell Survival/genetics , Cell Survival/physiology , Cyclin-Dependent Kinase Inhibitor p16/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Disease Models, Animal , E2F Transcription Factors , E2F1 Transcription Factor , Gene Expression/genetics , Genes, Tumor Suppressor , Genetic Therapy , Genetic Vectors/genetics , Glioma/physiopathology , Glioma/therapy , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Oncogene Protein p21(ras)/metabolism , Recombinant Fusion Proteins/genetics , Retinoblastoma Protein/metabolism , Retinoblastoma-Binding Protein 1 , Transcription Factor DP1 , Transcription Factors/physiology , Transfection/genetics , Tumor Cells, Cultured
3.
J Exp Med ; 172(1): 273-83, 1990 Jul 01.
Article in English | MEDLINE | ID: mdl-2358778

ABSTRACT

The relative importance of 11 polymorphic positions in the HLA-DR7 beta 1 chain in T cell recognition of foreign antigens was investigated using transfectants expressing mutant DR7 beta 1 chains as APC for five rabies virus-specific T cell clones. The results indicate that multiple amino acids, located in both the beta-strands and alpha-helix of DR7 beta 1 in the model of a class II molecule, are involved in DR7-restricted T cell recognition of these antigens. Many of the substitutions appeared to reduce the affinity of an antigenic peptide for the mutant DR7 molecules but did not prevent binding. The heterogeneity of responses of the three G-specific T cell clones to presentation of the G11.3 peptide by several of the mutant DR7 molecules indicates that the T cell receptor (TCR) of each these clones requires a different view of the G11.3/DR7 complex and raises the possibility that the G11.3 peptide may bind to the DR7 molecule in more than one conformation.


Subject(s)
Antigen-Presenting Cells/immunology , Antigens, Viral/immunology , HLA-DR7 Antigen/immunology , Polymorphism, Genetic/immunology , T-Lymphocytes/immunology , Amino Acid Sequence , HLA-DR7 Antigen/genetics , Humans , Lymphocyte Activation/immunology , Molecular Sequence Data , Mutation , Peptides/chemical synthesis , Rabies virus/immunology , Transfection/genetics
4.
J Exp Med ; 177(3): 797-809, 1993 Mar 01.
Article in English | MEDLINE | ID: mdl-8436910

ABSTRACT

Mice with transgenes containing an antibody H chain V region (VHDJH) gene were used in an analysis of the cis-acting elements required for hypermutation of immunoglobulin (Ig) V genes. These transgenes can somatically recombine with endogenous IgH DNA, leading to the formation of functional heavy (H) chains partially encoded by the transgenic VHDJH. The transgenomes in the five different lines of mice analyzed contain as little as 150 bp, and as much as 2.8 kb of natural DNA flanking the 5' side of the VH and either 1.5 or 2.3 kb (including the intronic enhancer and 5' matrix attachment region [MAR]) flanking the 3' side of VH. Hybridomas were constructed from immunized transgenic mice, and transgenes present in these hybridomas that had or had not recombined to form functional H chain loci were sequenced. The data obtained show that: (a) the recombined transgenes contain hypermutated VH genes; and (b) among such transgenes, even those containing only 150 bp of natural VH 5' flanking sequence and several kilobases of 5' plasmid vector sequence display a frequency, distribution, and type of mutation characteristic of conventional IgH loci. The data also indicate that transgenic VHDJH genes that have not recombined with endogenous IgH DNA are not substrates for hypermutation, even if they are flanked by 2.8 kb of natural 5' DNA, and 2.3 kb of natural 3' DNA, including the JH2-JH4 region, a MAR, and the intronic enhancer. Collectively, the data suggest that sequences 5' of the VH promoter are dispensable, a VH promoter and the intronic IgH enhancer region are not sufficient, and a region(s) within or 3' of the IgH constant region locus is requisite, for hypermutation of Ig VH transgenes.


Subject(s)
DNA/genetics , Enhancer Elements, Genetic/genetics , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Variable Region/genetics , Mutation/genetics , Promoter Regions, Genetic/genetics , Transfection/genetics , Animals , Base Sequence , Chromosome Mapping , Female , Gene Expression Regulation, Neoplastic/genetics , Genetic Vectors , Hybridomas/pathology , Male , Mice , Mice, Transgenic , Molecular Sequence Data
5.
J Exp Med ; 189(1): 51-62, 1999 Jan 04.
Article in English | MEDLINE | ID: mdl-9874563

ABSTRACT

The HIV-1 virion-associated accessory protein Vpr affects both viral replication and cellular transcription, proliferation, and differentiation. We report that Vpr enhances the activity of glucocorticoids in lymphoid and muscle-derived cell lines by interacting directly with the glucocorticoid receptor and general transcription factors, acting as a coactivator. Vpr contains the signature motif LXXLL also present in cellular nuclear receptor coactivators, such as steroid receptor coactivator 1 and p300/CREB-binding protein, which mediates their interaction with the glucocorticoid and other nuclear hormone receptors. A mutant Vpr molecule with disruption of this coactivator signature motif lost its ability to influence transcription of glucocorticoid-responsive genes and became a dominant-negative inhibitor of Vpr, possibly by retaining its general transcription factor-binding activities. The glucocorticoid coactivator activity of Vpr may contribute to increased tissue glucocorticoid sensitivity in the absence of hypercortisolism and to the pathogenesis of AIDS.


Subject(s)
Gene Products, vpr/metabolism , HIV-1/metabolism , Receptors, Glucocorticoid/metabolism , Transcriptional Activation/genetics , Cell Line , Dexamethasone/pharmacology , Genes, Reporter/genetics , Glucocorticoids/metabolism , Humans , Transcription Factor TFIID , Transcription Factors, TFII/genetics , Transcription Factors, TFII/immunology , Transfection/genetics , Viral Proteins/metabolism , vpr Gene Products, Human Immunodeficiency Virus
6.
J Exp Med ; 189(1): 195-205, 1999 Jan 04.
Article in English | MEDLINE | ID: mdl-9874576

ABSTRACT

The T cell antigen receptor (TCR) mediates recognition of peptide antigens bound in the groove of major histocompatibility complex (MHC) molecules. This dual recognition is mediated by the complementarity-determining residue (CDR) loops of the alpha and beta chains of a single TCR which contact exposed residues of the peptide antigen and amino acids along the MHC alpha helices. The recent description of T cells that recognize hydrophobic microbial lipid antigens has challenged immunologists to explain, in molecular terms, the nature of this interaction. Structural studies on the murine CD1d1 molecule revealed an electrostatically neutral putative antigen-binding groove beneath the CD1 alpha helices. Here, we demonstrate that alpha/beta TCRs, when transferred into TCR-deficient recipient cells, confer specificity for both the foreign lipid antigen and CD1 isoform. Sequence analysis of a panel of CD1-restricted, lipid-specific TCRs reveals the incorporation of template-independent N nucleotides that encode diverse sequences and frequent charged basic residues at the V(D)J junctions. These sequences permit a model for recognition in which the TCR CDR3 loops containing charged residues project between the CD1 alpha helices, contacting the lipid antigen hydrophilic head moieties as well as adjacent CD1 residues in a manner that explains antigen specificity and CD1 restriction.


Subject(s)
Antigens/immunology , Lipids/immunology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , Animals , Antigens, CD1/chemistry , Antigens, CD1/immunology , Clone Cells/immunology , Cloning, Molecular , Humans , Mice , Models, Molecular , Polymerase Chain Reaction , Protein Structure, Secondary , Transfection/genetics
7.
J Exp Med ; 189(2): 309-18, 1999 Jan 18.
Article in English | MEDLINE | ID: mdl-9892613

ABSTRACT

PIR-A and PIR-B, paired immunoglobulin-like receptors encoded, respectively, by multiple Pira genes and a single Pirb gene in mice, are relatives of the human natural killer (NK) and Fc receptors. Monoclonal and polyclonal antibodies produced against a recombinant PIR protein identified cell surface glycoproteins of approximately 85 and approximately 120 kD on B cells, granulocytes, and macrophages. A disulfide-linked homodimer associated with the cell surface PIR molecules was identified as the Fc receptor common gamma (FcRgammac) chain. Whereas PIR-B fibroblast transfectants expressed cell surface molecules of approximately 120 kD, PIR-A transfectants expressed the approximately 85-kD molecules exclusively intracellularly; PIR-A and FcRgammac cotransfectants expressed the PIR-A/ FcRgammac complex on their cell surface. Correspondingly, PIR-B was normally expressed on the cell surface of splenocytes from FcRgammac-/- mice whereas PIR-A was not. Cell surface levels of PIR molecules on myeloid and B lineage cells increased with cellular differentiation and activation. Dendritic cells, monocytes/macrophages, and mast cells expressed the PIR molecules in varying levels, but T cells and NK cells did not. These experiments define the coordinate cellular expression of PIR-B, an inhibitory receptor, and PIR-A, an activating receptor; demonstrate the requirement of FcRgammac chain association for cell surface PIR-A expression; and suggest that the level of FcRgammac chain expression could differentially affect the PIR-A/PIR-B equilibrium in different cell lineages.


Subject(s)
Receptors, Immunologic/immunology , Animals , Antibodies/immunology , Base Sequence , Cells, Cultured , Dendritic Cells/immunology , Flow Cytometry , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Receptors, Cell Surface/immunology , Receptors, Fc/immunology , Recombinant Proteins/immunology , Spleen/immunology , Transfection/genetics
8.
J Exp Med ; 188(10): 1841-8, 1998 Nov 16.
Article in English | MEDLINE | ID: mdl-9815261

ABSTRACT

Natural killer (NK) cells preferentially lyse targets that express reduced levels of major histocompatibility complex (MHC) class I proteins. To date, the only known mouse NK receptors for MHC class I belong to the Ly49 family of C-type lectin homodimers. Here, we report the cloning of mouse NKG2A, and demonstrate it forms an additional and distinct class I receptor, a CD94/NKG2A heterodimer. Using soluble tetramers of the nonclassical class I molecule Qa-1(b), we provide direct evidence that CD94/NKG2A recognizes Qa-1(b). We further demonstrate that NK recognition of Qa-1(b) results in the inhibition of target cell lysis. Inhibition appears to depend on the presence of Qdm, a Qa-1(b)-binding peptide derived from the signal sequences of some classical class I molecules. Mouse NKG2A maps adjacent to CD94 in the heart of the NK complex on mouse chromosome six, one of a small cluster of NKG2-like genes. Our findings suggest that mouse NK cells, like their human counterparts, use multiple mechanisms to survey class I expression on target cells.


Subject(s)
Antigens, CD/genetics , Histocompatibility Antigens Class I/immunology , Killer Cells, Natural/immunology , Lectins, C-Type , Membrane Glycoproteins/genetics , Receptors, Cell Surface/immunology , Animals , COS Cells , Chromosome Mapping , Cloning, Molecular , Flow Cytometry , Mice , NK Cell Lectin-Like Receptor Subfamily D , Protein Conformation , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Transfection/genetics
9.
J Exp Med ; 185(11): 1939-50, 1997 Jun 02.
Article in English | MEDLINE | ID: mdl-9166423

ABSTRACT

The receptor for granulocyte/macrophage colony-stimulating factor (GM-CSF) is composed of two chains, alpha and betac. Both chains belong to the superfamily of cytokine receptors characterized by a common structural feature, i.e., the presence of at least two fibronectin-like folds in the extracellular domain, which was first identified in the growth hormone receptor. The GM-CSF receptor (GMR)-alpha chain confers low affinity binding only (5-10 nM), whereas the other chain, betac, does not bind GM-CSF by itself but confers high affinity binding when associated with GMR-alpha (25-100 pM). The present study was designed to define the assembly of the GMR complex at the molecular level through site-directed mutagenesis guided by homology modeling with the growth hormone receptor complex. In our three-dimensional model, R280 of GMR-alpha, located in the F'-G' loop and close to the WSSWS motif, is in the vicinity of the ligand Asp112, suggesting the possibility of electrostatic interaction between these two residues. Through site directed mutagenesis, we provide several lines of evidence indicating the importance of electrostatic interaction in ligand-receptor recognition. First, mutagenesis of GMR-alphaR280 strikingly ablated ligand binding in the absence of beta common (betac); ligand binding was restored in the presence of betac with, nonetheless, a significant shift from high (26 pM) toward low affinity (from 2 to 13 nM). The rank order of the dissociation constant for the different GMR-alphaR280 mutations where Lys > Gln > Met > Asp, suggesting the importance of the charge at this position. Second, a mutant GM-CSF with charge reversal mutation at position Asp112 exhibited a 1,000-fold decrease in affinity in receptor binding, whereas charge ablation or conservative mutations were the least affected (10-20-fold). Third, removal of the charge at position R280 of GMR-alpha introduced a 10-fold decrease in the association rate constant and only a 2-fold change in the dissociation rate constant, suggesting that R280 is implicated in ligand recognition, possibly through interaction with Asp112 of GM-CSF. For all R280 mutants, the half-efficient concentrations of GM-CSF required for membrane (receptor binding) to nuclear events (c-fos promoter activation) and cell proliferation (thymidine incorporation) were in the same range, indicating that the threshold for biologic activity is governed mainly by the affinity of ligand-receptor interaction. Furthermore, mutation of other residues in the immediate vicinity of R280 was less drastic. Sequence alignment and modeling of interleukin (IL)-3R and IL-5R identified an arginine residue at the tip of a beta turn in a highly divergent context at the F'-G' loop, close to a conserved structural element, the WSXWS motif, suggesting the possibility of a ligand association mechanism similar to the one described herein for GMR.


Subject(s)
Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/chemistry , 3T3 Cells , Amino Acid Sequence , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/metabolism , CHO Cells , Cricetinae , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Kinetics , Ligands , Mice , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Binding , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Sequence Homology, Amino Acid , Software , Transfection/genetics
10.
J Exp Med ; 185(11): 2025-32, 1997 Jun 02.
Article in English | MEDLINE | ID: mdl-9166431

ABSTRACT

Antigen receptor gene rearrangement is directed by DNA motifs consisting of a conserved heptamer and nonamer separated by a nonconserved spacer of either 12 or 23 base pairs (12 or 23 recombination signal sequences [RSS]). V(D)J recombination requires that the rearranging DNA segments be flanked by RSSs of different spacer lengths, a phenomenon known as the 12/23 rule. Recent studies have shown that this restriction operates at the level of DNA cleavage, which is mediated by the products of the recombination activating genes RAG1 and RAG2. Here, we show that RAG1 and RAG2 are not sufficient for 12/23 dependent cleavage, whereas RAG1 and RAG2 complemented with whole cell extract faithfully recapitulates the 12/23 rule. In addition, HMG box containing proteins HMG1 and HMG2 enhance RAG1- and RAG2-mediated cleavage of substrates containing 23 RSS but not of substrates containing only 12 RSS. These results suggest the existence of a nucleoprotein complex at the cleavage site, consisting of architectural, catalytic, and regulatory components.


Subject(s)
DNA-Binding Proteins , DNA/metabolism , Homeodomain Proteins , Nucleoproteins/metabolism , Proteins/metabolism , Receptors, Antigen, T-Cell/genetics , Recombination, Genetic/genetics , T-Lymphocytes/metabolism , DNA/chemistry , DNA Nucleotidyltransferases/metabolism , Electrophoresis, Polyacrylamide Gel , High Mobility Group Proteins/metabolism , Humans , Nuclear Proteins , Proteins/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Transfection/genetics , VDJ Recombinases
11.
J Exp Med ; 189(1): 111-21, 1999 Jan 04.
Article in English | MEDLINE | ID: mdl-9874568

ABSTRACT

Antigen receptor-triggered T-cell activation is mediated by the sequential action of the Src and Syk/Zap-70 families of protein tyrosine kinases (PTKs). Previously, we reported that another PTK termed p50(csk) was a potent negative regulator of T-cell receptor (TCR) signaling because of its ability to inactivate Src-related kinases. This inhibitory effect required the catalytic activity of Csk, as well as its Src homology (SH)3 and SH2 domains. Subsequent studies uncovered that, via its SH3 domain, p50(csk) was associated with PEP, a proline-enriched protein tyrosine phosphatase (PTP) of unknown function expressed in hemopoietic cells. Herein, we have attempted to identify the role of the Csk-PEP complex in T lymphocytes. The results of our experiments showed that, like Csk, PEP was a strong repressor of TCR signaling. This property was dependent on the phosphatase activity of PEP, as well as on the sequence mediating its binding to p50(csk). Through reconstitution experiments in Cos-1 cells, evidence was obtained that Csk and PEP act synergistically to inhibit protein tyrosine phosphorylation by Src-related kinases, and that this effect requires their association. Finally, experiments with a substrate-trapping mutant of PEP suggested that PEP functions by dephosphorylating and inactivating the PTKs responsible for T-cell activation. In addition to giving novel insights into the mechanisms involved in the negative regulation of T-cell activation, these findings indicate that the association of an inhibitory PTK with a PTP constitutes a more efficient means of inhibiting signal transduction by Src family kinases in vivo.


Subject(s)
Protein Tyrosine Phosphatases/metabolism , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/metabolism , src-Family Kinases/metabolism , Animals , Binding Sites , COS Cells , CSK Tyrosine-Protein Kinase , Hybridomas/metabolism , Mice , Peptide Mapping , Phosphoproteins/analysis , Phosphorylation , Protein Binding , Protein-Tyrosine Kinases , Signal Transduction , Transfection/genetics , src Homology Domains
12.
J Exp Med ; 189(1): 169-78, 1999 Jan 04.
Article in English | MEDLINE | ID: mdl-9874573

ABSTRACT

The mechanisms underlying initiation and maintenance of CD4 T cell responses after DNA vaccination were studied using a construct coding for nonsecreted fifth component of complement (C5) protein, thus restricting the availability of antigen. The only cell types to express C5 were keratinocytes at the site of DNA application and a small number of dendritic cells present in the draining lymph nodes. Antigen expression persisted for up to 12 wk in keratinocytes, but dendritic cells did not express C5 beyond 2 wk after vaccination. Cross-priming of dendritic cells by C5 expressed in keratinocytes did not occur unless keratinocyte death was induced by irradiation in vitro. CD4 T cells were activated in the draining lymph nodes only and subsequently migrated to the spleen, where memory T cells persisted for longer than 40 wk despite the absence of a source of persistent antigen. While DNA vaccination resulted in transfection of a small proportion of dendritic cells only, it led to general activation of all dendritic cells, thus providing optimal conditions for effective T cell activation and maintenance of memory.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Immunity/immunology , Transfection/genetics , Vaccines, DNA/immunology , Animals , Complement C5/genetics , Complement C5/immunology , DNA, Complementary/genetics , DNA, Complementary/immunology , Flow Cytometry , Interleukin-2/metabolism , Lymph Nodes/immunology , Mice , Mice, Transgenic , Polymerase Chain Reaction , Spleen/immunology , Vaccines, DNA/genetics
13.
J Exp Med ; 189(2): 241-52, 1999 Jan 18.
Article in English | MEDLINE | ID: mdl-9892607

ABSTRACT

L-selectin mediates leukocyte rolling on vascular endothelium during inflammation. Although vascular endothelium can be activated with inflammatory cytokines to express functional L-selectin ligands, these ligands have not been well characterized. In this study, fucosyltransferase VII cDNA (Fuc-TVII) transfection of the EA.hy926 human vascular endothelial cell line (926-FtVII) induced functional L-selectin ligand expression and expression of sialyl Lewisx (sLex), as defined by HECA-452 (cutaneous lymphocyte antigen; CLA) and CSLEX-1 mAbs. Cytokine activation of human umbilical vein endothelial cells (HUVEC) also induced functional L-selectin ligand expression, with increased CLA expression and Fuc-TVII transcription. The majority of L-selectin-dependent lymphocyte attachment to activated HUVEC and 926-FtVII cells was blocked specifically by treating the endothelial cells with the HECA-452 mAb, but not the CSLEX-1 mAb. CLA-bearing ligands on vascular endothelium also required sulfation and appropriate molecular scaffolds for functional activity, but were distinct from the L-selectin ligands previously identified by the MECA-79 mAb. These findings demonstrate that the HECA-452- defined antigen, CLA, is an essential carbohydrate component of vascular L-selectin ligands.


Subject(s)
Endothelium, Vascular/immunology , L-Selectin/immunology , Ligands , Membrane Glycoproteins/immunology , Antibodies, Monoclonal/immunology , Antigens, Differentiation, T-Lymphocyte , Antigens, Neoplasm , Cell Adhesion/immunology , Cell Line , Fluorescent Antibody Technique , Fucosyltransferases/genetics , Humans , Immunoglobulin M/genetics , Immunoglobulin M/immunology , Inflammation/immunology , Leukocytes/immunology , Lymphocytes/metabolism , Oligosaccharides/immunology , Recombinant Fusion Proteins/genetics , Sialyl Lewis X Antigen , Transfection/genetics , Tumor Necrosis Factor-alpha/pharmacology
14.
J Exp Med ; 189(2): 395-402, 1999 Jan 18.
Article in English | MEDLINE | ID: mdl-9892621

ABSTRACT

We have previously reported (Badolato, R., J.M. Wang, W.J. Murphy, A. R. Lloyd, D.F. Michiel, L.L. Bausserman, D.J. Kelvin, and J.J. Oppenheim. 1994. J. Exp. Med. 180:203; Xu, L., R. Badolato, W.J. Murphy, D.L. Longo, M. Anver, S. Hale, J.J. Oppenheim, and J.M. Wang. 1995. J. Immunol. 155:1184.) that the acute phase protein serum amyloid A (SAA) is a potent chemoattractant for human leukocytes in vitro and mouse phagocytes in vivo. To identify the signaling mechanisms, we evaluated patterns of cross-desensitization between SAA and other leukocyte chemoattractants. We found that the chemotactic bacterial peptide, N-formyl- methionyl-leucyl-phenylalanine (fMLP), was able to specifically attenuate Ca2+ mobilization in human phagocytes induced by SAA, but only at very high concentrations, suggesting that SAA uses a low affinity fMLP receptor. Here we demonstrate that SAA selectively induced Ca2+ mobilization and migration of HEK cells expressing FPRL1, a human seven-transmembrane domain phagocyte receptor with low affinity for fMLP, and high affinity for lipoxin A4. Furthermore, radiolabeled SAA specifically bound to human phagocytes and FPRL1-transfected 293 cells. In contrast, SAA was not a ligand or agonist for FPR, the high affinity fMLP receptor. Thus, SAA is the first chemotactic ligand identified for FPRL1. Our results suggest that FPRL1 mediates phagocyte migration in response to SAA.


Subject(s)
Apolipoproteins/pharmacology , Chemotaxis/physiology , GTP-Binding Proteins/metabolism , Phagocytes/metabolism , Receptors, Immunologic/metabolism , Receptors, Lipoxin , Receptors, Peptide/metabolism , Serum Amyloid A Protein/pharmacology , Amino Acid Sequence , Calcium/metabolism , Cell Line , Chemotactic Factors/pharmacology , Humans , Molecular Sequence Data , Monocytes/metabolism , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Receptors, Formyl Peptide , Transfection/genetics
15.
J Exp Med ; 188(10): 1769-74, 1998 Nov 16.
Article in English | MEDLINE | ID: mdl-9815254

ABSTRACT

Each member of the rab guanosine triphosphatase protein family assists in the regulation of a specific step within the biosynthetic or endocytic pathways. We have found that the early endosome-associated rab4 protein controls a step critical for receptor-mediated antigen processing in a murine A20 B cell line. Expression of the dominant negative rab4N121I mutant dramatically inhibited the processing and presentation of ovalbumin, lambda cI repressor, or rabbit immunoglobulin G internalized as antigens by B cell antigen receptors or transfected Fc receptors. This defect did not reflect a block in antigen endocytosis or degradation, and transfected cells remained completely capable of presenting exogenously added ovalbumin and lambda repressor peptides. Most remarkably, rab4N121I-expressing cells were undiminished in their ability to present each of these antigens when whole proteins were internalized at high concentration by fluid-phase endocytosis. Thus, expression of the rab4N121I selectively inactivated a portion of the endocytic pathway required for the processing of receptor-bound, but not nonspecifically internalized, antigens. These results suggest that elements of the early endosome-recycling pathway play an important and selective role in physiologically relevant forms of antigen processing in B cells.


Subject(s)
Antigen Presentation/immunology , B-Lymphocytes/immunology , DNA-Binding Proteins , GTP-Binding Proteins/immunology , Animals , Cell Line , Endocytosis/physiology , Endosomes/metabolism , Gene Expression Regulation/immunology , Immunoglobulin G/immunology , Mice , Microscopy, Fluorescence , Mutation/genetics , Ovalbumin/immunology , Receptors, Fc/immunology , Repressor Proteins/immunology , Transfection/genetics , Viral Proteins , Viral Regulatory and Accessory Proteins , rab4 GTP-Binding Proteins
16.
J Exp Med ; 187(3): 367-78, 1998 Feb 02.
Article in English | MEDLINE | ID: mdl-9449717

ABSTRACT

T cells specific for nucleosomal autoepitopes are selectively expanded in lupus mice and these Th cells drive autoimmune B cells to produce pathogenic antinuclear antibodies. We transfected the TCR-alpha and -beta chain genes of a representative, pathogenic autoantibody-inducing Th clone specific for the nucleosomal core histone peptide H471-94 into TCR-negative recipient cells. Although the autoimmune TCRs were originally derived from SNF1 (I-Ad/q) mice, the transfectants could recognize the nucleosomal autoepitope presented by APC-bearing I-A molecules of all haplotypes tested, as well as human DR molecules. Competition assays indicated that the autoepitopes bound to the MHC class II groove. Most remarkably, MHC-unrestricted recognition of the nucleosomal peptide epitope was conferred by the lupus TCR-alpha chain even when it paired with a TCR-beta chain of irrelevant specificity. Several other disease-relevant Th clones and splenic T cells of lupus mice had similar properties. The TCR-alpha chains of these murine lupus Th clones shared related motifs and charged residues in their CDRs, and similar motifs were apparent even in TCR-alpha chains of human lupus Th clones. The lupus TCR-alpha chains probably contact the nucleosomal peptide complexed with MHC with relatively high affinity/avidity to sustain TCR signaling, because CD4 coreceptor was not required for promiscuous recognition. Indeed, pathogenic autoantibody-inducing, CD4-negative, TCR-alphabeta+ Th cells are expanded in systemic lupus erythematosus. These results have implications regarding thymic selection and peripheral expansion of nucleosome-specific T cells in lupus. They also suggest that universally tolerogenic epitopes could be designed for therapy of lupus patients with diverse HLA alleles. We propose to designate nucleosomes and other antigens bearing universal epitopes "Pantigens" (for promiscuous antigens).


Subject(s)
Autoantigens/immunology , Autoimmunity/immunology , Lupus Vulgaris/immunology , Nucleosomes/immunology , Receptors, Antigen, T-Cell/metabolism , Amino Acid Sequence , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/metabolism , Autoantibodies/immunology , CD4 Antigens/immunology , Cloning, Molecular , Epitopes/immunology , Histocompatibility Antigens Class II/immunology , Histones/chemistry , Humans , Interleukin-2/metabolism , Mice , Mice, Inbred Strains , Molecular Sequence Data , Peptide Fragments/immunology , Receptors, Antigen, T-Cell/chemistry , Transfection/genetics
17.
J Exp Med ; 187(3): 433-8, 1998 Feb 02.
Article in English | MEDLINE | ID: mdl-9449723

ABSTRACT

The majority of T cell responses are restricted to peptide antigens bound by polymorphic major histocompatibility complex (MHC) molecules. However, peptide antigens can be presented to T cells by murine non-MHC-encoded CD1d (mCD1) molecules, and human T cell lines specific for nonpeptide antigens presented on CD1 isoforms have been identified. It is shown here that antigen-specific, mCD1-restricted lymphocytes can be generated in vivo by immunizing mice with a combination of plasmids encoding chicken ovalbumin, murine CD1d, and costimulatory molecules. Splenocytes from immunized mice have CD1d-restricted, MHC- unrestricted, ovalbumin-specific cytolytic activity that can be inhibited by anti-CD1 antibodies as well as a competing CD1-binding peptide. These results suggest a physiologic role for murine CD1d to present exogenous protein antigens.


Subject(s)
Antigens, CD1/immunology , T-Lymphocytes, Cytotoxic/metabolism , Amino Acid Sequence , Animals , Antibodies/immunology , Antibodies/pharmacology , Antigen Presentation/physiology , CD8 Antigens/immunology , Immunization , Major Histocompatibility Complex/immunology , Mice , Molecular Sequence Data , Ovalbumin/genetics , Ovalbumin/immunology , Peptides/immunology , Peptides/pharmacology , Plasmids/genetics , Plasmids/immunology , Protein Binding , Spleen/immunology , T-Lymphocytes, Cytotoxic/immunology , Transfection/genetics
18.
J Exp Med ; 187(7): 973-84, 1998 Apr 06.
Article in English | MEDLINE | ID: mdl-9529314

ABSTRACT

The c-rel protooncogene encodes a member of the Rel/nuclear factor (NF)-kappaB family of transcriptional factors. To assess the role of the transcriptional activation domain of c-Rel in vivo, we generated mice expressing a truncated c-Rel (Deltac-Rel) that lacks the COOH-terminal region, but retains a functional Rel homology domain. Mice with an homozygous mutation in the c-rel region encoding the COOH terminus of c-Rel (c-relDeltaCT/DeltaCT) display marked defects in proliferative and immune functions. c-relDeltaCT/DeltaCT animals present histopathological alterations of hemopoietic tissues, such as an enlarged spleen due to lymphoid hyperplasia, extramedullary hematopoiesis, and bone marrow hypoplasia. In older c-relDeltaCT/DeltaCT mice, lymphoid hyperplasia was also detected in lymph nodes, liver, lung, and stomach. These animals present a more severe phenotype than mice lacking the entire c-Rel protein. Thus, in c-relDeltaCT/DeltaCT mice, the lack of c-Rel activity is less efficiently compensated by other NF-kappaB proteins.


Subject(s)
Hematopoiesis/genetics , Proto-Oncogene Proteins/genetics , Pseudolymphoma/genetics , Transcriptional Activation/genetics , Animals , B-Lymphocytes/immunology , Bone Marrow/pathology , Cell Line , DNA-Binding Proteins/analysis , Flow Cytometry , Listeria monocytogenes/pathogenicity , Macrophages, Peritoneal/metabolism , Mice , Mice, Knockout , NF-kappa B/metabolism , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins c-rel , Sequence Deletion/genetics , Spleen/pathology , T-Lymphocytes/immunology , Thymus Gland/pathology , Transfection/genetics
19.
J Exp Med ; 187(7): 1029-35, 1998 Apr 06.
Article in English | MEDLINE | ID: mdl-9529319

ABSTRACT

Protein C is an important regulatory mechanism of blood coagulation. Protein C functions as an anticoagulant when converted to the active serine protease form on the endothelial cell surface. Thrombomodulin (TM), an endothelial cell surface receptor specific for thrombin, has been identified as an essential component for protein C activation. Although protein C can be activated directly by the thrombin-TM complex, the conversion is known as a relatively low-affinity reaction. Therefore, protein C activation has been believed to occur only in microcirculation. On the other hand, we have identified and cloned a novel endothelial cell surface receptor (EPCR) that is capable of high-affinity binding of protein C and activated protein C. In this study, we demonstrate the constitutive, endothelial cell-specific expression of EPCR in vivo. Abundant expression was particularly detected in the aorta and large arteries. In vitro cultured, arterial endothelial cells were also found to express abundant EPCR and were capable of promoting significant levels of protein C activation. EPCR was found to greatly accelerate protein C activation by examining functional activity in transfected cell lines expressing EPCR and/or TM. EPCR decreased the dissociation constant and increased the maximum velocity for protein C activation mediated by the thrombin-TM complex. By these mechanisms, EPCR appears to enable significant levels of protein C activation in large vessels. These results suggest that the protein C anticoagulation pathway is important for the regulation of blood coagulation not only in microvessels but also in large vessels.


Subject(s)
Blood Coagulation Factors , Endothelium, Vascular/metabolism , Protein C/metabolism , Receptors, Cell Surface/metabolism , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Anticoagulants/pharmacology , Blood Coagulation/physiology , Cell Line , Endothelium, Vascular/cytology , Enzyme Activation/physiology , Flow Cytometry , Humans , Immunohistochemistry , Kinetics , Models, Biological , Serine Endopeptidases/metabolism , Thrombin/physiology , Thrombomodulin/physiology , Transfection/genetics
20.
J Exp Med ; 189(1): 37-50, 1999 Jan 04.
Article in English | MEDLINE | ID: mdl-9874562

ABSTRACT

Adhesion receptors that are known to initiate contact (tethering) between blood-borne leukocytes and their endothelial counterreceptors are frequently concentrated on the microvilli of leukocytes. Other adhesion molecules are displayed either randomly or preferentially on the planar cell body. To determine whether ultrastructural distribution plays a role during tethering in vivo, we used pre-B cell transfectants expressing L- or E-selectin ectodomains linked to transmembrane/intracellular domains that mediated different surface distribution patterns. We analyzed the frequency and velocity of transfectant rolling in high endothelial venules of peripheral lymph nodes using an intravital microscopy model. Ectodomains on microvilli conferred a higher efficiency at initiating rolling than random distribution which, in turn, was more efficient than preferential expression on the cell body. The role of microvillous presentation was less accentuated in venules below 20 micrometers in diameter than in larger venules. In the narrow venules, tethering of cells with cell body expression may have been aided by forced margination through collision with erythrocytes. L-selectin transfected cells rolled 10-fold faster than E-selectin transfectants. Interestingly, rolling velocity histograms of cell lines expressing equivalent copy numbers of the same ectodomain were always similar, irrespective of the topographic distribution. Our data indicate that the distribution of adhesion receptors has a dramatic impact on contact initiation between leukocytes and endothelial cells, but does not play a role once rolling has been established.


Subject(s)
B-Lymphocytes/metabolism , Cell Adhesion/physiology , L-Selectin/metabolism , Microvilli/metabolism , Animals , B-Lymphocytes/ultrastructure , Cell Adhesion Molecules/metabolism , Cell Movement/physiology , E-Selectin/genetics , E-Selectin/metabolism , Flow Cytometry , Hemodynamics , Humans , L-Selectin/genetics , Lymph Nodes/blood supply , Lymph Nodes/metabolism , Mice , Receptors, Cell Surface/metabolism , Transfection/genetics
SELECTION OF CITATIONS
SEARCH DETAIL