Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 282(2): 1296-304, 2007 Jan 12.
Article in English | MEDLINE | ID: mdl-17085439

ABSTRACT

Collagen-related peptide is a selective agonist for the platelet collagen receptor Glycoprotein VI. The triple helical peptide contains ten GPO triplets/strand (single letter amino acid nomenclature, where O is hydroxyproline) and so over-represents GPO compared with native collagen sequence. To investigate the ability of Glycoprotein VI to recognize GPO triplets in a setting more representative of the collagens, we synthesized a set of triple helical peptides containing fewer GPO triplets, varying their number and spacing within an inert (GPP)n backbone. The adhesion of recombinant human Glycoprotein VI ectodo-main, like that of human platelets, to these peptides increased with their GPO content, and platelet adhesion was abolished by the specific anti-Glycoprotein VI-blocking antibody, 10B12. Platelet aggregation and protein tyrosine phosphorylation were induced only by cross-linked peptides and only those that contained two or more GPO triplets. Such peptides were less potent than cross-linked collagen-related peptide. Our data suggest that both the sequences GPOGPO and GPO.........GPO represent functional Glycoprotein VI recognition motifs within collagen. Furthermore, we propose that the (GPO)4 motif can support simultaneous binding of two glycoprotein VI molecules, in either a parallel or anti-parallel stacking arrangement, which could play an important role in activation of signaling.


Subject(s)
Blood Platelets/metabolism , Collagen/chemistry , Collagen/metabolism , Platelet Adhesiveness/physiology , Platelet Membrane Glycoproteins/metabolism , Binding Sites/physiology , Cross-Linking Reagents/chemistry , Cross-Linking Reagents/metabolism , Glycine/metabolism , Humans , Hydroxyproline/metabolism , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Phosphorylation , Platelet Aggregation/physiology , Platelet Membrane Glycoproteins/chemistry , Proline/metabolism , Protein Binding/physiology , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction/physiology , Structure-Activity Relationship , Tyrosine/metabolism
2.
J Immunol Methods ; 316(1-2): 75-83, 2006 Oct 20.
Article in English | MEDLINE | ID: mdl-17027020

ABSTRACT

We report the development of an expression system for the production of soluble, calmodulin (CaM)-tagged proteins in Drosophila Schneider S2 cells and the subsequent use of these proteins for the selection of phage displayed antibodies. The CaM-tag permitted the purification of recombinant protein to >90% purity in a single step at yields of >20 mg/l. Using platelet glycoprotein VI (GP6) as a model, we demonstrated that the recombinant CaM-tagged protein was post-translationally N-glycosylated and had identical ligand specificity to native protein. A novel selection strategy, exploiting the CaM tag, was then used to isolate four single chain Fv fragments (scFvs) specific for GP6 from a non-immune phage display library. In contrast to other selection methods, which can result in antibodies that do not recognise native protein, all of the scFvs we selected bound cell surface expressed GP6. In conclusion, the production of CaM-tagged proteins in Drosophila Schneider S2 cells and the selection strategy reported here offer advantages over previously published methods, including simple culture conditions, rapid protein purification, specific elution of phage antibodies and preferential selection of phage antibodies that recognise native, cell surface expressed protein.


Subject(s)
Antigens/biosynthesis , Calmodulin/genetics , Drosophila melanogaster/genetics , Immunoglobulin Fragments/biosynthesis , Peptide Library , Platelet Membrane Glycoproteins/genetics , Recombinant Proteins/biosynthesis , Amino Acid Sequence , Animals , Antigens/genetics , Blotting, Western , Calmodulin/metabolism , Cell Line , Cloning, Molecular , Drosophila melanogaster/metabolism , Enzyme-Linked Immunosorbent Assay , Humans , Immunoglobulin Fragments/genetics , Immunoglobulin Fragments/immunology , Molecular Sequence Data , Platelet Membrane Glycoproteins/immunology , Recombinant Proteins/chemical synthesis , Recombinant Proteins/genetics
3.
J Biol Chem ; 279(46): 47763-72, 2004 Nov 12.
Article in English | MEDLINE | ID: mdl-15345717

ABSTRACT

Only three recognition motifs, GFOGER, GLOGER, and GASGER, all present in type I collagen, have been identified to date for collagen-binding integrins, such as alpha(2)beta(1). Sequence alignment was used to investigate the occurrence of related motifs in other human fibrillar collagens, and located a conserved array of novel GER motifs within their triple helical domains. We compared the integrin binding properties of synthetic triple helical peptides containing examples of such sequences (GLSGER, GMOGER, GAOGER, and GQRGER) or the previously identified motifs. Recombinant inserted (I) domains of integrin subunits alpha(1), alpha(2) and alpha(11) all bound poorly to all motifs other than GFOGER and GLOGER. Similarly, alpha(2)beta(1) -containing resting platelets adhered well only to GFOGER and GLOGER, while ADP-activated platelets, HT1080 cells and two active alpha(2)I domain mutants (E318W, locked open) bound all motifs well, indicating that affinity modulation determines the sequence selectivity of integrins. GxO/SGER peptides inhibited platelet adhesion to collagen monomers with order of potency F >/= L >/= M > A. These results establish GFOGER as a high affinity sequence, which can interact with the alpha(2)I domain in the absence of activation and suggest that integrin reactivity of collagens may be predicted from their GER content.


Subject(s)
Amino Acid Motifs , Collagen Type I/metabolism , Integrin alpha2beta1/metabolism , Peptides/metabolism , Animals , Blood Platelets/metabolism , Cell Line , Collagen Type I/chemistry , Collagen Type I/genetics , Humans , Integrin alpha Chains/genetics , Integrin alpha Chains/metabolism , Integrin alpha2beta1/genetics , Peptides/chemistry , Peptides/genetics , Platelet Adhesiveness/physiology , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Alignment
5.
J Mol Biol ; 335(4): 1019-28, 2004 Jan 23.
Article in English | MEDLINE | ID: mdl-14698296

ABSTRACT

We have determined the 1.8A crystal structure of a triple helical integrin-binding collagen peptide (IBP) with sequence (Gly-Pro-Hyp)(2)-Gly-Phe-Hyp-Gly-Glu-Arg-(Gly-Pro-Hyp)(3). The central GFOGER hexapeptide is recognised specifically by the integrins alpha2beta1, alpha1beta1, alpha10beta1 and alpha11beta1. These integrin/collagen interactions are implicated in a number of key physiological processes including cell adhesion, cell growth and differentiation, and pathological states such as thrombosis and tumour metastasis. Comparison of the IBP structure with the previously determined structure of an identical collagen peptide in complex with the integrin alpha2-I domain (IBP(c)) allows the first detailed examination of collagen in a bound and an unbound state. The IBP structure shows a direct and a water-mediated electrostatic interaction between Glu and Arg side-chains from adjacent strands, but no intra-strand interactions. The interactions between IBP Glu and Arg side-chains are disrupted upon integrin binding. A comparison of IBP and IBP(c) main-chain conformation reveals the flexible nature of the triple helix backbone in the imino-poor GFOGER region. This flexibility could be important to the integrin-collagen interaction and provides a possible explanation for the unique orientation of the three GFOGER strands observed in the integrin-IBP(c) complex crystal structure.


Subject(s)
Collagen/chemistry , Collagen/metabolism , Integrin alpha2beta1/metabolism , Peptides/chemistry , Peptides/metabolism , Amino Acid Motifs , Amino Acid Sequence , Cell Adhesion , Crystallography, X-Ray , Hydrogen Bonding , Models, Molecular , Pliability , Protein Structure, Quaternary , Static Electricity
6.
Blood ; 103(3): 903-11, 2004 Feb 01.
Article in English | MEDLINE | ID: mdl-14504096

ABSTRACT

Glycoprotein (GP) VI is the major receptor responsible for platelet activation by collagen, but the collagen-binding surface of GPVI is unknown. To address this issue we expressed, from insect cells, the immunoglobulin (Ig)-like ectodomains (residues 1-185) of human and murine GPVI, called hD1D2 and mD1D2, respectively. Both proteins bound specifically to collagen-related peptide (CRP), a GPVI-specific ligand, but hD1D2 bound CRP more strongly than did mD1D2. Molecular modeling and sequence comparison identified key differences between hD1D2 and mD1D2. Ten mutant hD1D2s were expressed, of which 4 had human residues replaced by their murine counterpart, and 6 had replacements by alanine. CRP binding studies with these mutants demonstrated that the exchange of lysine at position 59 for the corresponding murine glutamate substantially reduced binding to CRP. The position of lysine59 on the apical surface of GPVI suggests a mode of CRP binding analogous to that used by the related killer cell Ig-like receptors to bind HLA. This surface was confirmed as critical for collagen binding by epitope mapping of an inhibitory phage antibody against GPVI. This anti-GPVI, clone 10B12, gave dose-dependent inhibition of the hD1D2-collagen interaction. Clone 10B12 inhibited activation of platelets by CRP and collagen in aggregometry and thrombus formation by the latter in whole blood perfusion. Antibody 10B12 showed significantly reduced binding to the hD1D2-E59, and, on that basis, the GPVI:10B12 interface was modeled.


Subject(s)
Carrier Proteins/metabolism , Peptides , Platelet Membrane Glycoproteins/chemistry , Platelet Membrane Glycoproteins/genetics , Amino Acid Sequence , Animals , Antibodies, Blocking , Base Sequence , Binding Sites/genetics , Collagen/metabolism , DNA, Complementary/genetics , HLA Antigens/metabolism , Humans , In Vitro Techniques , Ligands , Lysine/chemistry , Mice , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Platelet Membrane Glycoproteins/immunology , Platelet Membrane Glycoproteins/metabolism , Protein Structure, Tertiary , Receptors, Immunologic/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Species Specificity
7.
Biochem Soc Symp ; (70): 81-94, 2003.
Article in English | MEDLINE | ID: mdl-14587284

ABSTRACT

The collagen-platelet interaction is central to haemostasis and may be a critical determinant of arterial thrombosis, where subendothelium is exposed after rupture of atherosclerotic plaque. Recent research has capitalized on the cloning of an important signalling receptor for collagen, glycoprotein VI, which is expressed only on platelets, and on the use of collagen-mimetic peptides as specific tools for both glycoprotein VI and integrin alpha 2 beta 1. We have identified sequences, GPO and GFOGER (where O denotes hydroxyproline), within collagen that are recognized by the collagen receptors glycoprotein VI and integrin alpha 2 beta 1 respectively, allowing their signalling properties and specific functional roles to be examined. Triple-helical peptides containing these sequences were used to show the signalling potential of integrin alpha 2 beta 1, and to confirm its important contribution to platelet adhesion. Glycoprotein VI appears to operate functionally on the platelet surface as a dimer, which recognizes GPO motifs that are separated by four triplets of collagen sequence. These advances will allow the relationship between the structure of collagen and its haemostatic activity to be established.


Subject(s)
Blood Platelets/metabolism , Collagen/metabolism , Signal Transduction , Collagen/chemistry , Humans , Models, Molecular , Protein Conformation
8.
J Biol Chem ; 278(41): 40213-23, 2003 Oct 10.
Article in English | MEDLINE | ID: mdl-12832416

ABSTRACT

The transcription factor activator protein (AP)-1 plays crucial roles in proliferation, cell death, and the immune response. c-JUN is an important component of AP-1, but only very few c-JUN response genes have been identified to date. Activity of c-JUN is controlled by NH2-terminal phosphorylation (JNP) of its transactivation domain by a family of JUN-NH2-terminal protein kinases (JNK). JNK form a stable complex with c-JUN in vitro and in vivo. We have targeted this interaction by means of a cell-permeable peptide containing the JNK-binding (delta) domain of human c-JUN. This peptide strongly and specifically induced apoptosis in HeLa tumor cells, which was paralleled by inhibition of serum-induced c-JUN phosphorylation and up-regulation of the cell cycle inhibitor p21cip/waf. Application of the c-JUN peptide to interleukin (IL)-1-stimulated human primary fibroblasts resulted in up-regulation of four genes, namely COX-2, MnSOD, I kappa B alpha, and MAIL and down-regulation of 10 genes, namely CCL8, mPGES, SAA1, hIAP-1, hIAP-2, pent(r)axin-3, CXCL10, IL-1 beta, ICAM-1, and CCL2. Only a small group of genes, namely pent(r)axin-3, CXCL10, ICAM-1, and IL-1 beta, was inhibited by both the c-JUN peptide and the JNK inhibitor SP600125. Thereby, and by additional experiments using small interfering RNA to suppress endogenous c-JUN we identify for the first time three distinct groups of inflammatory genes whose IL-1-induced expression depends on c-JUN, on JNK, or on both. These results shed further light on the complexity of c-JUN-JNK-mediated gene regulation and also highlight the potential use of dissecting signaling downstream from JNK to specifically target proliferative diseases or the inflammatory response.


Subject(s)
Apoptosis/drug effects , Inflammation/genetics , Mitogen-Activated Protein Kinases/metabolism , Peptide Fragments/pharmacology , Proto-Oncogene Proteins c-jun/metabolism , Amino Acid Sequence , Base Sequence , Cells, Cultured , DNA, Complementary/genetics , Gene Expression Regulation/drug effects , HeLa Cells , Humans , In Vitro Techniques , Interleukin-1/pharmacology , JNK Mitogen-Activated Protein Kinases , Macromolecular Substances , Mitogen-Activated Protein Kinases/chemistry , Mitogen-Activated Protein Kinases/genetics , Molecular Sequence Data , Peptide Fragments/chemistry , Peptide Fragments/genetics , Phosphorylation , Protein Structure, Tertiary , Proto-Oncogene Proteins c-jun/chemistry , Proto-Oncogene Proteins c-jun/genetics , Recombinant Proteins/pharmacology , Signal Transduction , Transcription Factor AP-1/chemistry , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism
9.
Blood ; 102(6): 2085-92, 2003 Sep 15.
Article in English | MEDLINE | ID: mdl-12791669

ABSTRACT

Integrin alpha2beta1 is the principal adhesive receptor for collagen but platelets also adhere through glycoprotein VI (GPVI). Integrin alphaIIbbeta3 may augment platelet adhesion. We have shown that disulfide exchange is necessary for platelet adhesion to fibrinogen, fibronectin, and collagen. However 2 questions remained: (1) Can activated alphaIIbbeta3 explain the observed role of disulfide exchange in adhesion to collagen, or is this role common to other integrins? (2) Is disulfide dependence specific to the integrin receptors or shared with GPVI? To discriminate adhesive functions of alpha2beta1 from those of alphaIIbbeta3 we used Glanzmann platelets and alphaIIbbeta3-specific antibodies applied to normal platelets. To resolve adhesive events mediated by alpha2beta1 from those of GPVI we used synthetic peptides specific to each receptor. We addressed direct integrin ligation using purified alpha2beta1 and recombinant I domain. We observed the following: adhesion to the alpha2beta1-specific peptide was disulfide-exchange dependent and protein disulfide isomerase (PDI) mediated; membrane-impermeant thiol blockers inhibited alpha2beta1, but not GPVI mediated, adhesion; direct blockade of PDI revealed that it is involved in adhesion through alpha2beta1 but not GPVI; and purified alpha2beta1, but not recombinant I domain, depended on free thiols for ligation. These data suggest that the enzymatically catalyzed adhesion-associated reorganization of disulfide bonds is common to members of the integrin family and specific to this family.


Subject(s)
Collagen/metabolism , Disulfides/metabolism , Integrin alpha2beta1/metabolism , Platelet Adhesiveness/physiology , 4-Chloromercuribenzenesulfonate/pharmacology , Anti-Bacterial Agents/pharmacology , Antibodies/pharmacology , Bacitracin/pharmacology , Carrier Proteins/metabolism , Catalysis , Cross-Linking Reagents/metabolism , Dithionitrobenzoic Acid/pharmacology , Dose-Response Relationship, Drug , Humans , Integrin alpha2beta1/chemistry , Ligands , Peptides/metabolism , Piperazines/pharmacology , Piperidines/pharmacology , Platelet Glycoprotein GPIIb-IIIa Complex/immunology , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Platelet Membrane Glycoproteins/metabolism , Protein Binding/drug effects , Protein Binding/physiology , Protein Disulfide-Isomerases/antagonists & inhibitors , Protein Disulfide-Isomerases/immunology , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sulfhydryl Compounds/metabolism , Sulfhydryl Reagents/pharmacology
10.
Eur J Biochem ; 270(11): 2394-403, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12755694

ABSTRACT

Three mammalian ADAMTS enzymes, ADAMTS-1, -4 and -5, are known to cleave aggrecan at certain glutamyl bonds and are considered to be largely responsible for cartilage aggrecan catabolism observed during the development of arthritis. We have previously reported that certain catechins, polyphenolic compounds found in highest concentration in green tea (Camellia sinensis), are capable of inhibiting cartilage aggrecan breakdown in an in vitro model of cartilage degradation. We have now cloned and expressed recombinant human ADAMTS-1, -4 and -5 and report here that the catechin gallate esters found in green tea potently inhibit the aggrecan-degrading activity of these enzymes, with submicromolar IC50 values. Moreover, the concentration needed for total inhibition of these members of the ADAMTS group is approximately two orders of magnitude lower than that which is needed to partially inhibit collagenase or ADAM-10 activity. Catechin gallate esters therefore provide selective inhibition of certain members of the ADAMTS group of enzymes and could constitute an important nutritional aid in the prevention of arthritis as well as being part of an effective therapy in the treatment of joint disease and other pathologies involving the action of these enzymes.


Subject(s)
Catechin/analogs & derivatives , Catechin/metabolism , Disintegrins/antagonists & inhibitors , Esters/metabolism , Metalloendopeptidases/antagonists & inhibitors , ADAM Proteins , ADAM10 Protein , ADAMTS1 Protein , ADAMTS4 Protein , ADAMTS5 Protein , Amino Acid Sequence , Amyloid Precursor Protein Secretases , Animals , Blotting, Western , Cattle , Cell Line , Culture Media, Conditioned/pharmacology , Disintegrins/metabolism , Dose-Response Relationship, Drug , Electrophoresis, Polyacrylamide Gel , Endopeptidases/metabolism , Enzyme Inhibitors/pharmacology , Genetic Vectors , Humans , Inhibitory Concentration 50 , Insecta , Membrane Proteins/metabolism , Metalloendopeptidases/metabolism , Molecular Sequence Data , Procollagen N-Endopeptidase , Protein Structure, Tertiary , Recombinant Proteins/metabolism , Tea
11.
J Biol Chem ; 278(15): 12609-12, 2003 Apr 11.
Article in English | MEDLINE | ID: mdl-12538645

ABSTRACT

Neutrophil proteinase 3 (Pr3) and elastase (NE) may cause lung tissue destruction in emphysema and cystic fibrosis. These serine proteinases have similar P(1) specificities. We have compared their catalytic activity using acyl-tetrapeptide-p-nitroanilides, which occupy the S(5)-S'(1) subsites of their substrate binding site, and intramolecularly quenched fluorogenic heptapeptides, which bind at S(5)-S'(4). Most p-nitroanilide substrates are turned over slowly by Pr3 as compared with NE. These differences disappear with the fluorogenic heptapeptides, some of which are hydrolyzed even faster by Pr3 than by NE. Elongation of substrates strongly increases the catalytic efficiency of Pr3, whereas it has little effect on NE catalysis. These different sensitivities to S'-P' interactions show that Pr3 and NE are not interchangeable enzymes despite their similar P(1) specificity.


Subject(s)
Leukocyte Elastase/metabolism , Serine Endopeptidases/metabolism , Amino Acid Sequence , Catalysis , Granulomatosis with Polyangiitis , Humans , Hydrolysis , Kinetics , Myeloblastin , Oligopeptides/chemistry , Oligopeptides/metabolism , Substrate Specificity
12.
J Biol Chem ; 278(9): 7270-7, 2003 Feb 28.
Article in English | MEDLINE | ID: mdl-12496264

ABSTRACT

The integrins alpha(1)beta(1), alpha(2)beta(1), alpha(10)beta(1), and alpha(11)beta(1) are referred to as a collagen receptor subgroup of the integrin family. Recently, both alpha(1)beta(1) and alpha(2)beta(1) integrins have been shown to recognize triple-helical GFOGER (where single letter amino acid nomenclature is used, O = hydroxyproline) or GFOGER-like motifs found in collagens, despite their distinct binding specificity for various collagen subtypes. In the present study we have investigated the mechanism whereby the latest member in the integrin family, alpha(11)beta(1), recognizes collagens using C2C12 cells transfected with alpha(11) cDNA and the bacterially expressed recombinant alpha(11) I domain. The ligand binding properties of alpha(11)beta(1) were compared with those of alpha(2)beta(1). Mg(2+)-dependent alpha(11)beta(1) binding to type I collagen required micromolar Ca(2+) but was inhibited by 1 mm Ca(2+), whereas alpha(2)beta(1)-mediated binding was refractory to millimolar concentrations of Ca(2+). The bacterially expressed recombinant alpha(11) I domain preference for fibrillar collagens over collagens IV and VI was the same as the alpha(2) I domain. Despite the difference in Ca(2+) sensitivity, alpha(11)beta(1)-expressing cells and the alpha(11) I domain bound to helical GFOGER sequences in a manner similar to alpha(2)beta(1)-expressing cells and the alpha(2) I domain. Modeling of the alpha I domain-collagen peptide complexes could partially explain the observed preference of different I domains for certain GFOGER sequence variations. In summary, our data indicate that the GFOGER sequence in fibrillar collagens is a common recognition motif used by alpha(1)beta(1), alpha(2)beta(1), and also alpha(11)beta(1) integrins. Although alpha(10) and alpha(11) chains show the highest sequence identity, alpha(2) and alpha(11) are more similar with regard to collagen specificity. Future studies will reveal whether alpha(2)beta(1) and alpha(11)beta(1) integrins also show overlapping biological functions.


Subject(s)
Collagen/chemistry , Collagen/metabolism , Integrins/chemistry , Integrins/metabolism , Receptors, Collagen/chemistry , Receptors, Collagen/metabolism , Amino Acid Motifs , Animals , Calcium/metabolism , Cell Adhesion , Cells, Cultured , DNA, Complementary/metabolism , Dose-Response Relationship, Drug , Humans , Kinetics , Magnesium/metabolism , Mice , Models, Molecular , Peptides/chemistry , Phenylalanine/chemistry , Precipitin Tests , Protein Binding , Protein Structure, Tertiary , Recombinant Proteins/metabolism
13.
J Biol Chem ; 277(50): 48210-9, 2002 Dec 13.
Article in English | MEDLINE | ID: mdl-12372841

ABSTRACT

ADAMs (a disintegrin and metalloprotease domains) are metalloprotease and disintegrin domain-containing transmembrane glycoproteins with proteolytic, cell adhesion, cell fusion, and cell signaling properties. ADAM8 was originally cloned from monocytic cells, and its distinct expression pattern indicates possible roles in both immunology and neuropathology. Here we describe our analysis of its biochemical properties. In transfected COS-7 cells, ADAM8 is localized to the plasma membrane and processed into two forms derived either by prodomain removal or as remnant protein comprising the extracellular region with the disintegrin domain at the N terminus. Proteolytic removal of the ADAM8 propeptide was completely blocked in mutant ADAM8 with a Glu(330) to Gln exchange (EQ-A8) in the Zn(2+) binding motif (HE(330)LGHNLGMSHD), arguing for autocatalytic prodomain removal. In co-transfection experiments, the ectodomain but not the entire MP domain of ADAM8 was able to remove the prodomain from EQ-ADAM8. With cells expressing ADAM8, cell adhesion to a substrate-bound recombinant ADAM8 disintegrin/Cys-rich domain was observed in the absence of serum, blocked by an antibody directed against the ADAM8 disintegrin domain. Soluble ADAM8 protease, consisting of either the metalloprotease domain or the complete ectodomain, cleaved myelin basic protein and a fluorogenic peptide substrate, and was inhibited by batimastat (BB-94, IC(50) approximately 50 nm) but not by recombinant tissue inhibitor of matrix metalloproteinases 1, 2, 3, and 4. Our findings demonstrate that ADAM8 processing by autocatalysis leads to a potential sheddase and to a form of ADAM8 with a function in cell adhesion.


Subject(s)
Antigens, CD/metabolism , Membrane Proteins , Metalloendopeptidases/metabolism , ADAM Proteins , Amino Acid Sequence , Animals , Antigens, CD/physiology , Base Sequence , COS Cells , Catalysis , Cell Adhesion/physiology , DNA Primers , Hydrolysis , Metalloendopeptidases/physiology , Mice , Tumor Cells, Cultured
14.
FEBS Lett ; 524(1-3): 154-8, 2002 Jul 31.
Article in English | MEDLINE | ID: mdl-12135759

ABSTRACT

The ADAM family of proteases are type I transmembrane proteins with both metalloproteinase and disintegrin containing extracellular domains. ADAMs are implicated in the proteolytic processing of membrane-bound precursors and involved in modulating cell-cell and cell-matrix interactions. ADAM8 (MS2, CD156) has been identified in myeloid and B cells. In this report we demonstrate that soluble ADAM8 is an active metalloprotease in vitro and is able to hydrolyse myelin basic protein and a variety of peptide substrates based on the cleavage sites of membrane-bound cytokines, growth factors and receptors which are known to be processed by metalloproteinases. Interestingly, although ADAM8 was inhibited by a number of peptide analogue hydroxamate inhibitors, it was not inhibited by the tissue inhibitors of metalloproteinases (TIMPs). We also demonstrate that the activity of recombinant soluble ADAM9 (meltrin-gamma, MDC9) lacks inhibition by the TIMPs, but can be inhibited by hydroxamate inhibitors. The lack of TIMP inhibition of ADAM8 and 9 contrasts with other membrane-associated metalloproteinases characterised to date in this respect (ADAM10, 12, 17, and the membrane-type metalloproteinases) which have been implicated in protein processing at the cell surface.


Subject(s)
Antigens, CD , Antigens, Surface/metabolism , Disintegrins/metabolism , Membrane Proteins , Metalloendopeptidases/metabolism , Tissue Inhibitor of Metalloproteinases/metabolism , ADAM Proteins , Amino Acid Sequence , Antigens, Surface/genetics , Antigens, Surface/isolation & purification , Catalysis , Chromatography, Affinity , Electrophoresis, Polyacrylamide Gel , Metalloendopeptidases/antagonists & inhibitors , Metalloendopeptidases/genetics , Metalloendopeptidases/isolation & purification , Molecular Sequence Data , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism
15.
J Biol Chem ; 277(30): 26872-8, 2002 Jul 26.
Article in English | MEDLINE | ID: mdl-11973338

ABSTRACT

The binding of certain growth factors and cytokines to components of the extracellular matrix can regulate their local availability and modulate their biological activities. We show that mesenchymal cell-derived keratinocyte growth factor (KGF), a key stimulator of epithelial cell proliferation during wound healing, preferentially binds to collagens I, III, and VI. Binding is inhibited in a dose-dependent manner by denatured single collagen chains and collagen cyanogen bromide peptides. This interaction is saturable with dissociation constants of approximately 10(-8) to 10(-9) m and estimated molar ratios of up to three molecules of KGF bound to one molecule of triple helical collagen. Furthermore, collagen-bound KGF stimulated the proliferation of transformed keratinocyte or HaCaT cells. Ligand blotting of collagen-derived peptides points to a limited set of collagenous consensus sequences that bind KGF. By using synthetic collagen peptides, we defined the consensus sequence (Gly-Pro-Hyp)(n) as the collagen binding motif. We conclude that the preferential binding of KGF to the abundant collagens leads to a spatial pattern of bioavailable KGF that is dictated by the local organization of the collagenous extracellular matrix. The defined collagenous consensus peptide or its analogue may be useful in wound healing by increasing KGF bioactivity and thus modulating local epithelial remodeling and regeneration.


Subject(s)
Collagen/metabolism , Fibroblast Growth Factors/metabolism , Amino Acid Motifs , Collagen/chemistry , Collodion/chemistry , Cyanogen Bromide/chemistry , Dose-Response Relationship, Drug , Fibroblast Growth Factor 7 , Glycine/chemistry , Humans , Hydroxyproline/chemistry , Kinetics , Ligands , Peptides/metabolism , Proline/chemistry , Protein Binding , Recombinant Proteins/metabolism , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...