Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 187
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Exp Med ; 181(6): 2259-64, 1995 Jun 01.
Article in English | MEDLINE | ID: mdl-7539045

ABSTRACT

L-selectin, a cell surface adhesion molecule that is expressed by most leukocytes, mediates leukocyte rolling along vascular endothelium at sites of inflammation. The contribution of L-selectin to leukocyte migration in models of chronic inflammation was assessed by using mice that lack cell surface L-selectin expression. Significant inhibition of neutrophil (56-62%), lymphocyte (70-75%), and monocyte (72-78%) migration into an inflamed peritoneum was observed 24 and 48 h after administration of thioglycollate, an inflammatory stimulus. L-selectin-deficient mice were also significantly impaired in delayed-type hypersensitivity reactions. Footpad swelling in response to sheep red blood cell challenge was reduced 75% in L-selectin-deficient mice compared with wild-type mice. Ear swelling in a model of contact hypersensitivity induced by oxazolone challenge was also reduced by 69% compared to wild-type mice. Consistent with L-selectin-mediating leukocyte migration into diverse vascular beds during inflammation, L-selectin-deficient mice were significantly resistant to death resulting from lipopolysaccharide (LPS)-induced toxic shock. LPS administration resulted in a 90% mortality rate in control mice after 24 h, while there was a 90% survival rate in L-selectin-deficient mice. These results demonstrate that L-selectin plays a prominent role in leukocyte homing to nonlymphoid tissues during inflammation and that blocking this process can be beneficial during pathological inflammatory responses.


Subject(s)
Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/physiology , Dermatitis, Contact/immunology , Inflammation/immunology , Leukocytes/physiology , Animals , Cell Adhesion , Crosses, Genetic , Dermatitis, Contact/physiopathology , Endothelium, Vascular/physiology , Endothelium, Vascular/physiopathology , Inflammation/physiopathology , L-Selectin , Leukocytes/immunology , Lymphocytes/immunology , Lymphocytes/physiology , Macrophages/immunology , Macrophages/physiology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Neutrophils/immunology , Neutrophils/physiology , Oxazolone
2.
J Exp Med ; 182(2): 519-30, 1995 Aug 01.
Article in English | MEDLINE | ID: mdl-7543141

ABSTRACT

L-selectin mediates leukocyte rolling on vascular endothelium at sites of inflammation and lymphocyte migration to peripheral lymph nodes. L-selectin is rapidly shed from the cell surface after leukocyte activation by a proteolytic mechanism that cleaves the receptor in a membrane proximal extracellular region. This process may allow rapid leukocyte detachment from the endothelial surface before entry into tissues. In this study, the structural requirements for regulation of human L-selectin endoproteolytic release were examined through analysis of chimeric selectin molecules and mutant L-selectin receptors. The use of chimeric selectins and a cytoplasmic tail truncation mutant demonstrated that the extracellular membrane-proximal 15-amino acid region of L-selectin is required for endoproteolytic release. The introduction of alanine-scanning mutations within this membrane-proximal region did not prevent endoproteolytic release, indicating that a specific amino acid motif was not an absolute requirement for cleavage. Furthermore, alterations within the putative primary cleavage site (K283-S284) resulted in either constitutive endoproteolytic release of the receptor or inhibition of cell activation-induced shedding to variable extents. The length of the membrane-proximal region was also critical since truncations of this region completely abolished endoproteolytic release. Thus, release of L-selectin is likely to be regulated by the generation of an appropriate tertiary conformation within the membrane-proximal region of the receptor which allows recognition by a membrane-bound endoprotease with relaxed sequence specificity that cleaves the receptor at a specific distance from the plasma membrane. These observations suggest a generalized protein-processing pathway involved in the endoproteolytic release of specific transmembrane proteins which harbor widely differing primary sequences at or neighboring their cleavage sites.


Subject(s)
Cell Adhesion Molecules/metabolism , Endopeptidases/metabolism , Receptors, Lymphocyte Homing/metabolism , Amino Acid Sequence , Cell Adhesion Molecules/chemistry , Cell Membrane/metabolism , L-Selectin , Membrane Glycoproteins/metabolism , Molecular Sequence Data , Mutagenesis, Site-Directed , Receptors, Lymphocyte Homing/chemistry , Recombinant Fusion Proteins , Sequence Alignment , Sequence Homology, Amino Acid , Solubility , Structure-Activity Relationship , Tetradecanoylphorbol Acetate/pharmacology
3.
J Exp Med ; 181(4): 1581-6, 1995 Apr 01.
Article in English | MEDLINE | ID: mdl-7535343

ABSTRACT

CD22 is a B cell-restricted member of the immunoglobulin (Ig) superfamily that functions as an adhesion receptor for leukocytes and erythrocytes. CD22 is unique among members of the Ig superfamily in that it has been suggested to bind a series of sialic acid-dependent ligands, potentially through different functional domains expressed by different splice variants of CD22. In this study, the epitopes identified by a large panel of function-blocking and non-function-blocking CD22 monoclonal antibodies were localized to specific Ig-like domains, revealing that all function-blocking monoclonal antibodies bound to the first and/or second Ig-like domains. Consistent with a single ligand-binding region, the two amino-terminal domains were the functional unit that mediated CD22 adhesion with lymphocytes, neutrophils, monocytes, and erythrocytes. The predominant cell surface species of CD22 was a full length 140,000 relative molecular mass seven Ig-like domain glycoprotein and a minor 130,000 relative molecular mass form lacking the fourth domain. While the two amino-terminal Ig-like domains of CD22 are structurally similar to those found in other members of the Ig superfamily involved in cell adhesion and containing an amino acid sequence motif associated with integrin recognition, site-directed mutagenesis of critical residues surrounding this motif did not disrupt CD22-mediated adhesion. These results demonstrate that the unique ligand-binding properties of CD22 are distinct from those of other members of the Ig superfamily involved in integrin-mediated cell adhesion.


Subject(s)
Antigens, CD/chemistry , Antigens, Differentiation, B-Lymphocyte/chemistry , Cell Adhesion Molecules , Lectins , Protein Structure, Tertiary , Sialic Acids/physiology , Amino Acid Sequence , Animals , Antibodies, Monoclonal/immunology , Antigens, CD/immunology , Antigens, CD/metabolism , Antigens, Differentiation, B-Lymphocyte/immunology , Antigens, Differentiation, B-Lymphocyte/metabolism , Binding Sites , Cell Adhesion , Cell Line, Transformed , Chlorocebus aethiops , DNA Mutational Analysis , DNA, Complementary/genetics , Epitopes/chemistry , Epitopes/immunology , Erythrocytes/metabolism , Humans , Immunoglobulins , Integrins/chemistry , Leukocytes, Mononuclear/metabolism , Ligands , Mice , Molecular Sequence Data , Molecular Weight , Multigene Family , N-Acetylneuraminic Acid , Protein Folding , Recombinant Fusion Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Sialic Acid Binding Ig-like Lectin 2 , Tumor Cells, Cultured
4.
J Exp Med ; 175(6): 1789-92, 1992 Jun 01.
Article in English | MEDLINE | ID: mdl-1375271

ABSTRACT

The receptors that mediate monocyte adhesion to cytokine-stimulated endothelial monolayers were assessed using a nonstatic (rotating) cell-attachment assay. In this system, leukocyte adhesion molecule-1 (LAM-1) (L-selectin) mediated a major portion (87 +/- 15% at 37 degrees C) of monocyte attachment to activated endothelium. mAb blocking of endothelial leukocyte adhesion molecule-1 (41% inhibition), CD18 (36%), and vascular cell adhesion molecule-1 (25%) function had lesser effects on attachment. These results suggest that LAM-1 may serve an important role in monocyte attachment to endothelium at sites of inflammation.


Subject(s)
Cell Adhesion Molecules/physiology , Cell Adhesion , Endothelium, Vascular/physiology , Monocytes/physiology , Antibodies, Monoclonal , Antigens, CD/physiology , CD18 Antigens , Cell Adhesion/drug effects , Cell Adhesion Molecules/biosynthesis , Cell Adhesion Molecules/immunology , Cell Line , Endothelium, Vascular/drug effects , Humans , L-Selectin , Monocytes/drug effects , Receptors, Leukocyte-Adhesion/physiology , Tumor Necrosis Factor-alpha/drug effects , Umbilical Veins , Vascular Cell Adhesion Molecule-1
5.
J Exp Med ; 177(3): 833-8, 1993 Mar 01.
Article in English | MEDLINE | ID: mdl-7679710

ABSTRACT

L-selectin (leukocyte adhesion molecule 1/MEL-14), a member of the selectin family of cell adhesion molecules, mediates leukocyte rolling and leukocyte adhesion to endothelium at sites of inflammation. In addition, L-selectin mediates the binding of lymphocytes to high endothelial venules (HEV) of peripheral lymph nodes. The strong amino acid sequence conservation of the cytoplasmic domain of L-selectin between humans and mice suggests an important role for this region. Deletion of the COOH-terminal 11 amino acids from the approximately 17 amino acid cytoplasmic domain of L-selectin eliminated binding of lymphocytes to HEV in the in vitro frozen section assay, and also abolished leukocyte rolling in vivo in exteriorized rat mesenteric venules, but did not alter the lectin activity of L-selectin. Pretreatment of cells with cytochalasin B, which disrupts actin microfilaments, also abolished adhesion without affecting carbohydrate recognition. Therefore, the cytoplasmic domain of L-selectin regulates leukocyte adhesion to endothelium independent of ligand recognition, by controlling cytoskeletal interactions and/or receptor avidity.


Subject(s)
Cell Adhesion Molecules/pharmacology , Endothelium, Vascular/cytology , Leukocytes/cytology , Actin Cytoskeleton/drug effects , Amino Acid Sequence , Animals , Base Sequence , Cell Adhesion/drug effects , Cell Adhesion/physiology , Cell Adhesion Molecules/analysis , Cell Line , Cell Movement/drug effects , Cell Movement/physiology , Cytochalasin B/pharmacology , Cytoplasm/chemistry , DNA/genetics , Endothelium, Vascular/physiology , Endothelium, Vascular/ultrastructure , Flow Cytometry , Inflammation/pathology , L-Selectin , Leukocytes/physiology , Leukocytes/ultrastructure , Lymphatic System , Mice , Molecular Sequence Data , Precipitin Tests
6.
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
7.
J Exp Med ; 176(5): 1405-14, 1992 Nov 01.
Article in English | MEDLINE | ID: mdl-1383386

ABSTRACT

Epstein-Barr virus (EBV) adsorption to human B lymphocytes is mediated by the viral envelope glycoprotein, gp350/220, which binds to the cell surface protein, CD21, also known as the CR2 complement receptor. Human epithelial cells also express an EBV receptor. A candidate surface molecule of 195 kD has previously been identified on an epithelial cell line and explanted epithelial tissue by reactivity with the CD21 specific monoclonal antibody (mAb), HB-5a. In experiments to further characterize the epithelial cell EBV receptor, we have found that two human epithelial cell lines, RHEK-1 and HeLa, specifically bind intact EB virions. A 145-kD protein, similar in size to B lymphocyte CD21, was specifically precipitated from surface iodinated RHEK-1 cells using the HB-5a mAb, or using purified soluble gp350/220 coupled to agarose beads. The previously identified 195-kD protein did not bind to gp350/220 or react with two other anti-CD21 mAbs. CD21 homologous RNA, similar in size to the B lymphocyte CD21 mRNA, was detected in both RHEK-1 and HeLa cells. The nucleotide sequence of the epithelial cell cDNA was identical to B lymphocyte CD21. The longest clone differs from previously reported CD21 cDNAs in having additional 5' untranslated sequence. Polymerase chain reaction amplification of RHEK-1- or B lymphoblastoid-derived cDNA verified that most CD21 transcripts are initiated at least 30-50 nucleotides upstream of the previously reported mRNA cap site. These experiments demonstrate that human epithelial cells can express CD21, and that CD21 is likely to mediate EBV adsorption to epithelial cells.


Subject(s)
Epithelium/chemistry , Herpesvirus 4, Human/metabolism , Receptors, Complement 3d/analysis , Receptors, Virus/analysis , Antigens, CD/analysis , Antigens, CD19 , Antigens, Differentiation , Antigens, Differentiation, B-Lymphocyte/analysis , Base Sequence , HeLa Cells/chemistry , Humans , Keratinocytes/chemistry , Molecular Sequence Data , Precipitin Tests , RNA, Messenger/analysis , Receptors, Complement 3d/genetics , Receptors, Complement 3d/immunology
8.
J Exp Med ; 186(11): 1923-31, 1997 Dec 01.
Article in English | MEDLINE | ID: mdl-9382890

ABSTRACT

The CD19 cell surface molecule regulates signal transduction events critical for B lymphocyte development and humoral immunity. Increasing the density of CD19 expression renders B lymphocytes hyper-responsive to transmembrane signals, and transgenic mice that overexpress CD19 have increased levels of autoantibodies. The role of CD19 in tolerance regulation and autoantibody generation was therefore examined by crossing mice that overexpress a human CD19 transgene with transgenic mice expressing a model autoantigen (soluble hen egg lysozyme, sHEL) and high-affinity HEL-specific IgMa and IgDa (IgHEL) antigen receptors. In this model of peripheral tolerance, B cells in sHEL/IgHEL double-transgenic mice are functionally anergic and do not produce autoantibodies. However, it was found that overexpression of CD19 in sHEL/IgHEL double-transgenic mice resulted in a breakdown of peripheral tolerance and the production of anti-HEL antibodies at levels similar to those observed in IgHEL mice lacking the sHEL autoantigen. Therefore, altered signaling thresholds due to CD19 overexpression resulted in the breakdown of peripheral tolerance. Thus, CD19 overexpression shifts the balance between tolerance and immunity to autoimmunity by augmenting antigen receptor signaling.


Subject(s)
Antigens, CD19/physiology , Autoantibodies/biosynthesis , Autoimmunity/immunology , B-Lymphocyte Subsets/immunology , Immune Tolerance/immunology , Immunoglobulin D/immunology , Immunoglobulin M/immunology , Receptors, Antigen, B-Cell/immunology , Signal Transduction/physiology , Animals , Antibody Specificity , Antigens, CD19/genetics , Autoantibodies/immunology , Autoantigens/genetics , Autoantigens/immunology , Autoimmune Diseases/etiology , Chickens/genetics , Chickens/immunology , Clonal Anergy/immunology , Complement C3d/immunology , Crosses, Genetic , Humans , Inflammation/complications , Inflammation/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Muramidase/genetics , Muramidase/immunology , Receptors, Complement 3d/immunology
9.
J Exp Med ; 173(1): 55-64, 1991 Jan 01.
Article in English | MEDLINE | ID: mdl-1702139

ABSTRACT

The complement system augments the humoral immune response, possibly by a mechanism that involves the B lymphocyte membrane receptor, CR2, which binds the C3dg fragment of C3 and triggers several B cell responses in vitro. The present study demonstrates that CR2 associates with a complex of membrane proteins that may mediate signal transduction by ligated CR2. Monoclonal antibodies to CR2 immunoprecipitated from digitonin lysates of Raji B lymphoblastoid cells a membrane complex containing CR2, approximately equimolar amounts of CD19, which is a member of the immunoglobulin superfamily, and three unidentified components: p130, p50, and p20. The complex, which was immunoprecipitated also with anti-CD19, could be dissociated by Nonidet P-40, accounting for its absence in previous studies of CR2. Expression of recombinant CR2 and CD19 in K562 erythroleukemia cells led to formation of a complex that contained not only these two proteins but also p130, p50, and p20, and another component, p14. These unidentified components of the CR2/CD19 complex coimmunoprecipitated with CD19 and not with CR2 from singly transfected cells, indicating primary association with the former. CD19 replicated the capacity of CR2 to interact synergistically with mIgM for increasing free intracellular Ca2+, suggesting that the complex mediates this function of CR2. Therefore, CR2 associates directly with CD19 to become a ligand-binding subunit of a pre-existing signal transduction complex of the B cell that may be representative of a family of membrane protein complexes. This interaction between the complement and immune systems differs from that between immunoglobulin and Clq by involving membrane rather than plasma proteins, and by having complement involved in the afferent phase of the immune response.


Subject(s)
Antigens, Differentiation, B-Lymphocyte/physiology , B-Lymphocytes/immunology , Receptors, Complement/physiology , Antigens, CD/metabolism , Antigens, CD/physiology , Antigens, CD19 , Antigens, Differentiation, B-Lymphocyte/metabolism , Digitonin , Humans , Immunoglobulin M/metabolism , Leukemia, Erythroblastic, Acute , Macromolecular Substances , Membrane Proteins/metabolism , Receptors, Complement/metabolism , Receptors, Complement 3d , Signal Transduction/immunology , Transfection
10.
J Exp Med ; 181(2): 669-75, 1995 Feb 01.
Article in English | MEDLINE | ID: mdl-7530761

ABSTRACT

Leukocyte recruitment into inflammatory sites is initiated by a reversible transient adhesive contact with the endothelium called leukocyte rolling, which is thought to be mediated by the selectin family of adhesion molecules. Selectin-mediated rolling precedes inflammatory cell emigration, which is significantly impaired in both P- and L-selectin gene-deficient mice. We report here that approximately 13% of all leukocytes passing venules of the cremaster muscle of wild-type mice roll along the endothelium at < 20 min after surgical dissection. Rolling leukocyte flux fraction reaches a maximum of 28% at 40-60 min and returns to 13% at 80-120 min. In P-selectin-deficient mice, rolling is absent initially and reaches 5% at 80-120 min. Rolling flux fraction in L-selectin-deficient mice is similar to wild type initially and declines to 5% at 80-120 min. In both wild-type and L-selectin-deficient mice, initial leukocyte rolling (0-60 min) is completely blocked by the P-selectin monoclonal antibody (mAb) RB40.34, but unaffected by L-selectin mAb MEL-14. Conversely, rolling at later time points (60-120 min) is inhibited by mAb MEL-14 but not by mAb RB40.34. After treatment with tumor necrosis factor (TNF)-alpha for 2 h, approximately 24% of all passing leukocytes roll in cremaster venules of wild-type and P-selectin gene-deficient mice. Rolling in TNF-alpha-treated mice is unaffected by P-selectin mAb or E-selectin mAb 10E9.6. By contrast, rolling in TNF-alpha-treated P-selectin-deficient mice is completely blocked by L-selectin mAb. These data show that P-selectin is important during the initial induction of leukocyte rolling after tissue trauma. At later time points and in TNF-alpha-treated preparations, rolling is largely L-selectin dependent. Under the conditions tested, we are unable to find evidence for involvement of E-selectin in leukocyte rolling in mice.


Subject(s)
Cell Adhesion Molecules/physiology , Leukocytes/physiology , Platelet Membrane Glycoproteins/physiology , Antibodies, Monoclonal/pharmacology , Cell Adhesion , Cell Line , Chemotaxis, Leukocyte , Humans , L-Selectin , P-Selectin , Tumor Necrosis Factor-alpha/pharmacology , Venules/cytology , Venules/drug effects
11.
J Exp Med ; 188(7): 1385-90, 1998 Oct 05.
Article in English | MEDLINE | ID: mdl-9763619

ABSTRACT

L-selectin binding activity for its ligand expressed by vascular endothelium is rapidly and transiently increased after leukocyte activation. To identify mechanisms for upregulation and assess how this influences leukocyte/endothelial cell interactions, cell-surface dimers of L-selectin were induced using the coumermycin-GyrB dimerization strategy for cross-linking L-selectin cytoplasmic domains in L-selectin cDNA-transfected lymphoblastoid cells. Coumermycin- induced L-selectin dimerization resulted in an approximately fourfold increase in binding of phosphomanan monoester core complex (PPME), a natural mimic of an L-selectin ligand, comparable to that observed after leukocyte activation. Moreover, L-selectin dimerization significantly increased (by approximately 700%) the number of lymphocytes rolling on vascular endothelium under a broad range of physiological shear stresses, and significantly slowed their rolling velocities. Therefore, L-selectin dimerization may explain the rapid increase in ligand binding activity that occurs after leukocyte activation and may directly influence leukocyte migration to peripheral lymphoid tissues or to sites of inflammation. Inducible oligomerization may also be a common mechanism for rapidly upregulating the adhesive or ligand-binding function of other cell-surface receptors.


Subject(s)
L-Selectin/metabolism , Leukocytes/metabolism , Cells, Cultured , DNA Gyrase , DNA Topoisomerases, Type II/genetics , DNA Topoisomerases, Type II/metabolism , Dimerization , Endothelium, Vascular , Humans , L-Selectin/genetics , Mannans/metabolism , Mannosephosphates/metabolism
12.
J Exp Med ; 188(8): 1413-9, 1998 Oct 19.
Article in English | MEDLINE | ID: mdl-9782118

ABSTRACT

Leukocyte migration into sites of inflammation involves multiple molecular interactions between leukocytes and vascular endothelial cells, mediating sequential leukocyte capture, rolling, and firm adhesion. In this study, we tested the role of molecular interactions between fractalkine (FKN), a transmembrane mucin-chemokine hybrid molecule expressed on activated endothelium, and its receptor (CX3CR1) in leukocyte capture, firm adhesion, and activation under physiologic flow conditions. Immobilized FKN fusion proteins captured resting peripheral blood mononuclear cells at physiologic wall shear stresses and induced firm adhesion of resting monocytes, resting and interleukin (IL)-2-activated CD8(+) T lymphocytes and IL-2-activated NK cells. FKN also induced cell shape change in firmly adherent monocytes and IL-2-activated lymphocytes. CX3CR1-transfected K562 cells, but not control K562 cells, firmly adhered to FKN-expressing ECV-304 cells (ECV-FKN) and tumor necrosis factor alpha-activated human umbilical vein endothelial cells. This firm adhesion was not inhibited by pertussis toxin, EDTA/EGTA, or antiintegrin antibodies, indicating that the firm adhesion was integrin independent. In summary, FKN mediated the rapid capture, integrin-independent firm adhesion, and activation of circulating leukocytes under flow. Thus, FKN and CX3CR1 mediate a novel pathway for leukocyte trafficking.


Subject(s)
Chemokines, CX3C , Chemokines, CXC/physiology , Leukocytes/physiology , Membrane Proteins/physiology , Receptors, Chemokine/physiology , Cell Adhesion , Cell Movement , Chemokine CX3CL1 , Endothelium, Vascular/cytology , Humans , Integrins/physiology , Interleukin-2/pharmacology , Tumor Necrosis Factor-alpha/pharmacology
13.
J Exp Med ; 170(1): 123-33, 1989 Jul 01.
Article in English | MEDLINE | ID: mdl-2473156

ABSTRACT

A cDNA encoding a new human lymphocyte cell surface molecule has been isolated and shown to identify a fourth member of a recently discovered family of adhesion proteins. This lymphocyte-associated molecule (LAM-1) is uniquely composed of multiple distinct domains, one domain homologous with animal lectins, one homologous with epidermal growth factor, and two short consensus repeat units similar to those found in C3/C4 binding proteins. This cDNA clone hybridized with RNAs found in B cell lines and T lymphocytes, but not with RNA from other cell types. The amino acid sequence of LAM-1 is 77% homologous with the sequence of the mouse lymphocyte homing receptor, suggesting that LAM-1 may function in human lymphocyte adhesion. The LAM-1 gene is located on chromosome 1q23-25, as is another member of this adhesion family, suggesting that this new family of proteins may be encoded by a cluster of "adhesion protein" loci.


Subject(s)
Antigens, Surface/genetics , Chromosomes, Human, Pair 1 , DNA/genetics , Lymphocytes/immunology , Membrane Glycoproteins/genetics , Receptors, Immunologic/genetics , Amino Acid Sequence , Animals , Base Sequence , Blotting, Northern , Cell Adhesion , Cell Adhesion Molecules , Chromosome Mapping , Cloning, Molecular , DNA/isolation & purification , Humans , L-Selectin , Mice , Molecular Sequence Data , Nucleic Acid Hybridization , Receptors, Lymphocyte Homing , Restriction Mapping , Sequence Homology, Nucleic Acid
14.
Am J Transplant ; 10(12): 2596-603, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21070606

ABSTRACT

In murine models, the adoptive transfer of CD4(+) /CD25(+) regulatory T cells (T(regs) ) inhibited graft-versus-host disease (GvHD). Previous work has indicated a critical role for the adhesion molecule L-selectin (CD62L) in the function of T(regs) in preventing GvHD. Here we examined the capacity of naive wild-type (WT), CD62L(-/-) and ex vivo expanded CD62L(Lo) T(regs) to inhibit acute GvHD. Surprisingly, we found that CD62L(-/-) T(regs) were potent suppressors of GvHD, whereas CD62L(Lo) T(regs) were unable to inhibit disease despite being functionally competent to suppress allo T cell responses in vitro. Concomitant with improved outcomes, WT and CD62L(-/-) T(regs) significantly reduced liver pathology and systemic pro-inflammatory cytokine production, although CD62L(-/-) T(regs) were less effective in reducing lung pathology. While accumulation of CD62L(-/-) T(regs) in GvHD target organs was equivalent to WT T(regs) , CD62L(-/-) T(regs) did not migrate as well as WT T(regs) to peripheral lymph nodes (PLNs) over the first 2 weeks posttransplantation. This work demonstrated that CD62L was dispensable for T(reg) -mediated protection from GvHD.


Subject(s)
Bone Marrow Transplantation/immunology , Graft vs Host Disease/prevention & control , L-Selectin/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Cell Migration Assays , Male , Mice , Mice, Inbred C57BL , Receptors, Chemokine/biosynthesis
15.
J Cell Biol ; 119(1): 229-38, 1992 Oct.
Article in English | MEDLINE | ID: mdl-1382078

ABSTRACT

L-selectin expressed by granulocytes, lymphocytes, and monocytes is responsible for initial leukocyte attachment to inflamed endothelium and high endothelial venules of peripheral lymph nodes. After leukocyte activation in vitro, L-selectin is rapidly shed from the cell surface. In this study, shed L-selectin (sL-selectin) from both lymphocytes and neutrophils was demonstrated to be present in high levels in human plasma by Western blot analysis and using a quantitative ELISA. In serum from normal human blood donors, a mean sL-selectin level of 1.6 +/- 0.8 micrograms/ml (n = 63) was found by ELISA. In addition, semipurified sL-selectin from plasma inhibited L-selectin-specific attachment of lymphocytes to cytokine-activated endothelium in a dose-dependent manner. L-selectin-dependent leukocyte attachment was completely inhibited at sL-selectin concentrations of 8-15 micrograms/ml, while physiological concentrations of sL-selectin caused a small but consistent inhibition of lymphocyte attachment. sL-selectin in plasma also inhibited anti-L-selectin mAb (2-5 micrograms/ml) binding to the surface of leukocytes. Interestingly, one epitope present within the EGF-like domain of L-selectin was lost in sL-selectin, suggesting a conformational change in the structure of the receptor after shedding. The presence of serum sL-selectin with functional activity indicates a potential role for sL-selectin in the regulation of leukocyte attachment to endothelium.


Subject(s)
Cell Adhesion Molecules/metabolism , Cell Adhesion/physiology , Endothelium/metabolism , Lymphocytes/metabolism , Membrane Glycoproteins/metabolism , Blotting, Western , Cell Adhesion Molecules/blood , Cells, Cultured , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Humans , L-Selectin , Membrane Glycoproteins/blood
16.
J Cell Biol ; 114(2): 351-8, 1991 Jul.
Article in English | MEDLINE | ID: mdl-1712791

ABSTRACT

The human lymphocyte homing receptor LAM-1, like its murine counterpart MEL-14, functions as a mammalian lectin, and mediates the binding of leukocytes to specialized high endothelial cells in lymphoid organs (HEV). LAM-1 is a member of a new family of cell adhesion molecules, termed selectins or LEC-CAMs, which also includes ELAM-1 and PAD-GEM (GMP-140/CD62). To localize the regions of LAM-1 that are involved in cell adhesion, we developed chimeric selectins, in which various domains of PAD-GEM were substituted into LAM-1, and used these chimeric proteins to define the domain requirements for carbohydrate binding, and to localize the regions recognized by several mAb which inhibit the adhesion of lymphocytes to lymph node HEV. The binding of PPME or fucoidin, soluble complex carbohydrates that specifically define the lectin activity of LAM-1 and MEL-14, required only the lectin domain of LAM-1. The LAM1-1, LAM1-3, and LAM1-6 mAb each strongly inhibit the binding of lymphocytes to HEV in the in vitro frozen section assay, and defined three independent epitopes on LAM-1. Blocking of PPME or fucoidin binding by LAM1-3 indicated that this site is identical, or in close proximity, to the carbohydrate binding site, and analysis of the binding of LAM1-3 to chimeric selectins showed that the epitope detected by LAM1-3 is located within the lectin domain. Although the LAM1-6 epitope is also located in the lectin domain, LAM1-6 did not affect the binding of PPME or fucoidin. The LAM1-1 epitope was located in, or required, the EGF domain, and, importantly, binding of LAM1-1 significantly enhanced the binding of both PPME and fucoidin. These results suggest that adhesion mediated by LAM-1 may involve cooperativity between functionally and spatially distinct sites, and support previous data suggesting a role for the EGF domain of LAM-1 in lymphocyte adhesion to HEV.


Subject(s)
Cell Adhesion Molecules/genetics , Chromosome Mapping , Endothelium, Vascular/ultrastructure , Leukocytes/ultrastructure , Receptors, Leukocyte-Adhesion/genetics , Amino Acid Sequence , Animals , Antibodies, Monoclonal/immunology , Anticoagulants/metabolism , B-Lymphocytes/metabolism , B-Lymphocytes/ultrastructure , Carbohydrate Metabolism , Cell Adhesion/physiology , Cell Adhesion Molecules/metabolism , Cell Adhesion Molecules/physiology , Cell Line , Chimera/genetics , Chimera/physiology , DNA/genetics , Endothelium, Vascular/metabolism , Endothelium, Vascular/physiology , Epitopes , L-Selectin , Lectins/metabolism , Leukocytes/metabolism , Leukocytes/physiology , Mannans/metabolism , Mice , Molecular Sequence Data , P-Selectin , Plant Lectins , Platelet Membrane Glycoproteins/genetics , Platelet Membrane Glycoproteins/metabolism , Platelet Membrane Glycoproteins/physiology , Polysaccharides/metabolism , Receptors, Leukocyte-Adhesion/metabolism , Receptors, Leukocyte-Adhesion/physiology
17.
J Cell Biol ; 125(1): 215-22, 1994 Apr.
Article in English | MEDLINE | ID: mdl-7511143

ABSTRACT

Vascular cell adhesion molecule 1 (VCAM-1), a member of the Ig superfamily originally identified on activated endothelium, binds to the integrin very late antigen-4 (VLA-4), also known as alpha 4 beta 1 or CD49d/CD29, to support cell-cell adhesion. Studies based on cell adhesion to two alternatively spliced forms of VCAM-1 or to chimeric molecules generated from them and intercellular adhesion molecule-1 (ICAM-1) have demonstrated two VLA-4 binding sites on the predominate form of VCAM-1. Here, we studied VLA-4-dependent adhesion of the lymphoid tumor cell line Ramos to cells expressing wild type and mutant forms of VCAM-1. Results based on domain deletion mutants demonstrated the existence and independence of two VLA-4-binding sites located in the first and fourth domains of VCAM-1. Results based on amino acid substitution mutants demonstrated that residues within a linear sequence of six amino acids found in both domain 1 and 4 were required for VLA-4 binding to either domain. Five of these amino acids represent a conserved motif also found in ICAM domains. We propose that integrin binding to these Ig-like domains depends on residues within this conserved motif. Specificity of integrin binding to Ig-like domains may be regulated by a set of nonconserved residues distinct from the conserved motif.


Subject(s)
Cell Adhesion Molecules/metabolism , Receptors, Very Late Antigen/metabolism , Amino Acid Sequence , Animals , Base Sequence , Cell Adhesion Molecules/chemistry , DNA Primers/chemistry , Humans , Intercellular Adhesion Molecule-1 , Mice , Molecular Sequence Data , Protein Binding , Recombinant Fusion Proteins/metabolism , Sequence Deletion , Structure-Activity Relationship , Vascular Cell Adhesion Molecule-1
18.
J Cell Biol ; 125(6): 1417-27, 1994 Jun.
Article in English | MEDLINE | ID: mdl-7515891

ABSTRACT

Leukocyte interactions with vascular endothelium at sites of inflammation can be dynamically regulated by activation-dependent adhesion molecules. Current models, primarily based on studies with polymorphonuclear leukocytes, suggest the involvement of multiple members of the selectin, integrin, and immunoglobulin gene families, sequentially, in the process of initial attachment (rolling), stable adhesion (arrest), spreading and ultimate diapedesis. In the current study, IL-4-activated human umbilical vein endothelium, which selectively expresses VCAM-1 and an L-selectin ligand but not E-selectin, and appropriate function blocking monoclonal antibodies, were used to study monocyte-endothelial interactions in an in vitro model that mimics microcirculatory flow conditions. In this system, L-selectin mediates monocyte rolling and also facilitates alpha 4 beta 1-integrin-dependent arrest, whereas beta 2-integrins are required for spreading of firmly attached monocytes on the endothelial cell surface but not their arrest. These findings provide the first in vitro evidence for human monocyte rolling on cytokine-activated endothelium, and suggest a sequential requirement for both beta 1- and beta 2-integrin-dependent adhesive mechanisms in monocyte-endothelial interactions.


Subject(s)
Cell Adhesion/physiology , Endothelium, Vascular/physiology , Monocytes/physiology , Signal Transduction , Antigens, CD/analysis , Blood Circulation/physiology , CD18 Antigens , Cell Adhesion Molecules/metabolism , Endothelium, Vascular/drug effects , Humans , Integrin alpha4beta1 , Integrin beta1 , Integrins/metabolism , Interleukin-4/pharmacology , L-Selectin , Physical Stimulation , Vascular Cell Adhesion Molecule-1
19.
J Cell Biol ; 121(5): 1121-32, 1993 Jun.
Article in English | MEDLINE | ID: mdl-7684739

ABSTRACT

CD20 is a plasma membrane phosphoprotein expressed exclusively by B lymphocytes. mAb binding to CD20 alters cell cycle progression and differentiation, indicating that CD20 plays an essential role in B lymphocyte function. Whole-cell patch clamp and fluorescence microscopy measurements of plasma membrane ionic conductance and cytosolic-free Ca2+ activity, respectively, were used to directly examine CD20 function. Transfection of human T and mouse pre-B lymphoblastoid cell lines with CD20 cDNA and subsequent stable expression of CD20 specifically increased transmembrane Ca2+ conductance. Transfection of CD20 cDNA and subsequent expression of CD20 in nonlymphoid cells (human K562 erythroleukemia cells and mouse NIH-3T3 fibroblasts) also induced the expression of an identical transmembrane Ca2+ conductance. The binding of a CD20-specific mAb to CD20+ lymphoblastoid cells also enhanced the transmembrane Ca2+ conductance. The mAb-enhanced Ca2+ currents had the same conductance characteristics as the CD20-associated Ca2+ currents in CD20 cDNA-transfected cells. C20 is structurally similar to several ion channels; each CD20 monomer possesses four membrane spanning domains, and both the amino and carboxy termini reside within the cytoplasm. Biochemical cross-linking of cell-surface molecules with subsequent immunoprecipitation analysis of CD20 suggests that CD20 may be present as a multimeric oligomer within the membrane, as occurs with several known membrane channels. Taken together, these findings indicate that CD20 directly regulates transmembrane Ca2+ conductance in B lymphocytes, and suggest that multimeric complexes of CD20 may form Ca2+ conductive ion channels in the plasma membrane of B lymphoid cells.


Subject(s)
Antigens, CD/physiology , Antigens, Differentiation, B-Lymphocyte/physiology , B-Lymphocytes/physiology , Calcium/metabolism , Antibodies, Monoclonal , Antigens, CD/chemistry , Antigens, CD20 , Antigens, Differentiation, B-Lymphocyte/chemistry , Cell Cycle , Cytosol/metabolism , Electric Conductivity , Humans , In Vitro Techniques , Ion Channel Gating , Lymphocyte Activation , Macromolecular Substances , Phosphorylation , Tetradecanoylphorbol Acetate/pharmacology , Transfection , Tumor Cells, Cultured
20.
J Cell Biol ; 124(4): 609-18, 1994 Feb.
Article in English | MEDLINE | ID: mdl-7508943

ABSTRACT

The selectin family of adhesion molecules mediates the initial interactions of leukocytes with endothelium. The extracellular region of each selectin contains an amino-terminal C-type lectin domain, followed by an EGF-like domain and multiple short consensus repeat units (SCR). Previous studies have indirectly suggested a role for each of the extracellular domains of the selectins in cell adhesion. In this study, a panel of chimeric selectins created by exchange of domains between L- and P-selectin was used to directly examine the role of the extracellular domains in cell adhesion. Exchange of only the lectin domains between L- and P-selectin conferred the adhesive and ligand recognition functions of the lectin domain of the parent molecule. However, chimeric selectins which contained both the lectin domain of L-selectin and the EGF-like domain of P-selectin exhibited dual ligand-binding specificity. These chimeric proteins supported adhesion both to myeloid cells and to high endothelial venules (HEV) of lymph nodes and mesenteric venules in vivo. Exchange of the SCR domains had no detectable effect on receptor function or specificity. Thus, the EGF-like domain of P-selectin may play a direct role in ligand recognition and leukocyte adhesion mediated by P-selectin, with the lectin plus EGF-like domains collectively forming a functional ligand recognition unit.


Subject(s)
Cell Adhesion , Epidermal Growth Factor/metabolism , Platelet Membrane Glycoproteins/metabolism , Animals , Cell Adhesion Molecules/chemistry , Cell Adhesion Molecules/metabolism , Cell Line , Cloning, Molecular , Humans , L-Selectin , Ligands , P-Selectin , Platelet Membrane Glycoproteins/chemistry , Protein Conformation , Rats , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL