Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Nat Immunol ; 9(7): 753-60, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18536718

ABSTRACT

To provide insight into the structural and functional properties of human complement component 5 (C5), we determined its crystal structure at a resolution of 3.1 A. The core of C5 adopted a structure resembling that of C3, with the domain arrangement at the position corresponding to the C3 thioester being very well conserved. However, in contrast to C3, the convertase cleavage site in C5 was ordered and the C345C domain flexibly attached to the core of C5. Binding of the tick C5 inhibitor OmCI to C5 resulted in stabilization of the global conformation of C5 but did not block the convertase cleavage site. The structure of C5 may render possible a structure-based approach for the design of new selective complement inhibitors.


Subject(s)
Complement C5/chemistry , Complement C5/metabolism , Insect Proteins/metabolism , Protein Structure, Quaternary , Animals , Arthropod Proteins , Binding Sites , Carrier Proteins , Complement C3 , Crystallography, X-Ray , Humans , Insect Proteins/chemistry , Surface Plasmon Resonance
2.
EMBO J ; 30(3): 606-16, 2011 Feb 02.
Article in English | MEDLINE | ID: mdl-21217642

ABSTRACT

Complement acts as a danger-sensing system in the innate immune system, and its activation initiates a strong inflammatory response and cleavage of the proteins C3 and C5 by proteolytic enzymes, the convertases. These contain a non-catalytic substrate contacting subunit (C3b or C4b) in complex with a protease subunit (Bb or C2a). We determined the crystal structures of the C3b homologue cobra venom factor (CVF) in complex with C5, and in complex with C5 and the inhibitor SSL7 at 4.3 Å resolution. The structures reveal a parallel two-point attachment between C5 and CVF, where the presence of SSL7 only slightly affects the C5-CVF interface, explaining the IgA dependence for SSL7-mediated inhibition of C5 cleavage. CVF functions as a relatively rigid binding scaffold inducing a conformational change in C5, which positions its cleavage site in proximity to the serine protease Bb. A general model for substrate recognition by the convertases is presented based on the C5-CVF and C3b-Bb-SCIN structures. Prior knowledge concerning interactions between the endogenous convertases and their substrates is rationalized by this model.


Subject(s)
Complement C3-C5 Convertases/metabolism , Complement C5/metabolism , Elapid Venoms/metabolism , Exotoxins/metabolism , Models, Molecular , Multiprotein Complexes/metabolism , Protein Conformation , Complement C3-C5 Convertases/chemistry , Complement C5/chemistry , Crystallography , Elapid Venoms/chemistry , Exotoxins/chemistry , Humans , Multiprotein Complexes/chemistry
3.
Proc Natl Acad Sci U S A ; 109(38): 15425-30, 2012 Sep 18.
Article in English | MEDLINE | ID: mdl-22949645

ABSTRACT

An essential aspect of innate immunity is recognition of molecular patterns on the surface of pathogens or altered self through the lectin and classical pathways, two of the three well-established activation pathways of the complement system. This recognition causes activation of the MASP-2 or the C1s serine proteases followed by cleavage of the protein C4. Here we present the crystal structures of the 203-kDa human C4 and the 245-kDa C4·MASP-2 substrate·enzyme complex. When C4 binds to MASP-2, substantial conformational changes in C4 are induced, and its scissile bond region becomes ordered and inserted into the protease catalytic site in a manner canonical to serine proteases. In MASP-2, an exosite located within the CCP domains recognizes the C4 C345C domain 60 Å from the scissile bond. Mutations in C4 and MASP-2 residues at the C345C-CCP interface inhibit the intermolecular interaction and C4 cleavage. The possible assembly of the huge in vivo enzyme-substrate complex consisting of glycan-bound mannan-binding lectin, MASP-2, and C4 is discussed. Our own and prior functional data suggest that C1s in the classical pathway of complement activated by, e.g., antigen-antibody complexes, also recognizes the C4 C345C domain through a CCP exosite. Our results provide a unified structural framework for understanding the early and essential step of C4 cleavage in the elimination of pathogens and altered self through two major pathways of complement activation.


Subject(s)
Complement C4/chemistry , Binding Sites , Crystallography/methods , HEK293 Cells , Humans , Immunity, Innate , Mannans/chemistry , Mannose-Binding Protein-Associated Serine Proteases/chemistry , Molecular Conformation , Mutation , Protein Binding , Protein Conformation , Protein Structure, Tertiary , Proteins/chemistry , Proteolysis , Recombinant Proteins/chemistry , Static Electricity , Substrate Specificity
4.
Proc Natl Acad Sci U S A ; 107(8): 3681-6, 2010 Feb 23.
Article in English | MEDLINE | ID: mdl-20133685

ABSTRACT

Staphylococcus aureus secretes the SSL7 protein as part of its immune evasion strategy. The protein binds both complement C5 and IgA, yet it is unclear whether SSL7 cross-links these two proteins and, if so, what purpose this serves the pathogen. We have isolated a stable IgA-SSL7-C5 complex, and our crystal structure of the C5-SSL7 complex confirms that binding to C5 occurs exclusively through the C-terminal beta-grasp domain of SSL7 leaving the OB domain free to interact with IgA. SSL7 interacts with C5 >70 A from the C5a cleavage site without inducing significant conformational changes in C5, and efficient inhibition of convertase cleavage of C5 is shown to be IgA dependent. Inhibition of C5a production and bacteriolysis are all shown to require C5 and IgA binding while inhibition of hemolysis is achieved by the C5 binding SSL7 beta-grasp domain alone. These results provide a conceptual and structural basis for the development of a highly specific complement inhibitor preventing only the formation of the lytic membrane attack complex without affecting the important signaling functions of C5a.


Subject(s)
Complement C5/antagonists & inhibitors , Complement C5/chemistry , Exotoxins/immunology , Staphylococcus aureus/immunology , Animals , Complement C5/immunology , Crystallography, X-Ray , Humans , Mutation , Protein Structure, Tertiary , Rabbits , Staphylococcus aureus/pathogenicity
5.
Angew Chem Int Ed Engl ; 51(14): 3340-4, 2012 Apr 02.
Article in English | MEDLINE | ID: mdl-22290936

ABSTRACT

I'm your Venus: the crystal structure of the human methylamine-induced form of α(2)-macroglobulin (α(2)M) shows its large central cavity can accommodate two medium-sized proteinases. Twelve major entrances provide access for small substrates to the cavity and the still-active trapped "prey". The structure unveils the molecular basis of the unique "venus flytrap" mechanism of α(2)M.


Subject(s)
alpha-Macroglobulins/chemistry , Crystallography, X-Ray , Humans , Methylamines/chemistry , Protease Inhibitors/chemistry , Protein Multimerization , Protein Structure, Quaternary , Protein Structure, Tertiary , alpha-Macroglobulins/metabolism
6.
J Mol Biol ; 361(1): 115-27, 2006 Aug 04.
Article in English | MEDLINE | ID: mdl-16831446

ABSTRACT

The third component of complement (C3) is a 190 kDa glycoprotein essential for eliciting the complement response. The protein consists of two polypeptide chains (alpha and beta) held together with a single disulfide bridge. The beta-chain is composed of six MG domains, one of which is shared with the alpha-chain. The disulfide bridge connecting the chains is positioned in the shared MG domain. The alpha-chain consists of the anaphylatoxin domain, three MG domains, a CUB domain, an alpha(6)/alpha(6)-barrel domain and the C-terminal C345c domain. An internal thioester in the alpha-chain of C3 (present in C4 but not in C5) is cleaved during complement activation. This mediates covalent attachment of the activated C3b to immune complexes and invading microorganisms, thereby opsonizing the target. We present the structure of bovine C3 determined at 3 Angstroms resolution. The structure shows that the ester is buried deeply between the thioester domain and the properdin binding domain, in agreement with the human structure. This domain interface is broken upon activation, allowing nucleophile access. The structure of bovine C3 clearly demonstrates that the main chain around the thioester undergoes a helical transition upon activation. This rearrangement is proposed to be the basis for the high level of reactivity of the thioester group. A strictly conserved glutamate residue is suggested to function catalytically in thioester proteins. Structure-based design of inhibitors of C3 activation may target a conserved pocket between the alpha-chain and the beta-chain of C3, which appears essential for conformational changes in C3.


Subject(s)
Complement C3/chemistry , Complement C3/physiology , Animals , Cattle , Complement Activation , Complement C3/metabolism , Crystallography, X-Ray , Esters , Molecular Sequence Data , Protein Structure, Tertiary , Structure-Activity Relationship
7.
FEBS Lett ; 560(1-3): 147-52, 2004 Feb 27.
Article in English | MEDLINE | ID: mdl-14988014

ABSTRACT

By proteolytic cleavage of insulin-like growth factor binding proteins, the metalloproteinase pregnancy-associated plasma protein-A (PAPP-A) is able to control the biological activity of insulin-like growth factors. PAPP-A circulates in pregnancy as a proteolytically inactive complex, disulfide bound to the proform of eosinophil major basic protein (proMBP). We here demonstrate that co-transfection of mammalian cells with PAPP-A and proMBP cDNA results in the formation of a covalent PAPP-A/proMBP complex in which PAPP-A is inhibited. Formation of the complex also occurs when PAPP-A and proMBP synthesized separately are incubated. Complex formation was monitored by Western blotting, and by using an immunoassay specific for the complex. Using mutagenesis, we further demonstrate that the complex forms in a specific manner and depends on the presence of two proMBP cysteine residues. Mutated proMBP, in which Cys-51 and -169 are replaced by serine, is unable to form the covalent complex with PAPP-A. Of particular interest, such mutated proMBP further lacks the ability to inhibit PAPP-A. For the first time, this conclusively demonstrates that proMBP is a proteinase inhibitor. We further conclude that proMBP inhibits PAPP-A in an unusual manner, not paralleled by other proteinase inhibitors of our knowledge, which requires proMBP to be covalently bound to PAPP-A by disulfide bonds. ProMBP binding to PAPP-A most likely either abrogates substrate access to the active site of PAPP-A or induces a conformational change in the structure of PAPP-A, as we, by further mutagenesis, were able to exclude that the inhibitory mechanism of proMBP is based on a cysteine switch-like mechanism.


Subject(s)
Blood Proteins/metabolism , Pregnancy-Associated Plasma Protein-A/metabolism , Protease Inhibitors/metabolism , Ribonucleases/metabolism , Amino Acid Substitution , Blood Proteins/genetics , Cell Line , DNA, Complementary/genetics , Disulfides/metabolism , Eosinophil Granule Proteins , Eosinophils/chemistry , Eosinophils/metabolism , Female , Humans , Kidney/cytology , Kidney/embryology , Kinetics , Plasmids , Pregnancy , Pregnancy-Associated Plasma Protein-A/chemistry , Protein Binding , Recombinant Proteins/metabolism , Ribonucleases/genetics , Serine/metabolism , Transfection
8.
Methods Mol Biol ; 1100: 93-102, 2014.
Article in English | MEDLINE | ID: mdl-24218252

ABSTRACT

Complement C5 is cleaved by proteolysis in the terminal phase of complement activation generating the pro-inflammatory C5a and membrane attack complex nucleator C5b. Whereas purification of its paralogues C3 and C4 from plasma is relatively straightforward, C5 purification is more complicated due to the lower amounts present and overlaps with the much more abundant C3 during several chromatographic steps. Here we describe our procedure for purifying homogenous, monodisperse, and crystallizable C5.


Subject(s)
Complement C5/isolation & purification , Chromatography, Liquid/methods , Complement C5/chemistry , Humans
9.
Protein Expr Purif ; 48(2): 261-73, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16529943

ABSTRACT

Although pregnancy-associated plasma protein-A (PAPP-A), a modulator of insulin-like growth factor (IGF) activity through its cleavage of IGF-binding protein (IGFBP)-4 and -5, has been known for more than two decades, knowledge about its domain architecture is still incomplete. Using position-specific iterative BLAST, we have identified distant relatives of the PAPP-A N-terminal sequence stretch of 250 residues. We present evidence that a protein domain with weak similarity to known laminin G-like (LG) modules is contained within this region, and we propose that PAPP-A and PAPP-A2 are new and unique members in the group of LG proteins as the pappalysins represent the first examples where LG modules are associated with proteinases. Fourteen beta-strands characteristic for the LG structure were tentatively located within the PAPP-A LG (PA-LG) module using secondary structure prediction and sequence alignment. Upon mammalian expression of PAPP-A truncation mutants, we defined domain boundaries showing that PA-LG is an autonomously folding unit, which spans the first 243 residues. We were unable to express PAPP-A variants which lack the PA-LG module, suggesting a possible role in stabilization of the proteolytic domain. To obtain larger amounts of protein for functional and structural analysis, the defined PA-LG domain was expressed in bacteria and folded in vitro. In addition, the availability of recombinant PA-LG module may potentially improve diagnostic assays based on the measurement of PAPP-A antigen, and also facilitate the study of PAPP-A in animal model systems.


Subject(s)
Laminin/chemistry , Laminin/classification , Pregnancy-Associated Plasma Protein-A/chemistry , Pregnancy-Associated Plasma Protein-A/genetics , Amino Acid Sequence , Binding Sites , Escherichia coli/genetics , Female , Gene Expression , Humans , Insulin-Like Growth Factor Binding Protein 4/metabolism , Insulin-Like Growth Factor Binding Protein 5/metabolism , Molecular Sequence Data , Peptide Library , Pregnancy , Pregnancy-Associated Plasma Protein-A/classification , Pregnancy-Associated Plasma Protein-A/metabolism , Protein Structure, Tertiary , Sequence Homology, Amino Acid
10.
J Biol Chem ; 280(11): 9823-32, 2005 Mar 18.
Article in English | MEDLINE | ID: mdl-15647258

ABSTRACT

The metzincin metalloproteinase pregnancy-associated plasma protein A (PAPP-A, pappalysin-1) promotes cell growth by the cleavage of insulin-like growth factor-binding proteins-4 and -5, causing the release of bound insulin-like growth factors. The proteolytic activity of PAPP-A is inhibited by the proform of eosinophil major basic protein (pro-MBP), which forms a covalent 2:2 proteinase-inhibitor complex based on disulfide bonds. To understand the process of complex formation, we determined the status of cysteine residues in both of the uncomplexed molecules. A comparison of the disulfide structure of the reactants with the known disulfide structure of the PAPP-A.pro-MBP complex reveals that six cysteine residues of the pro-MBP subunit (Cys-51, Cys-89, Cys-104, Cys-107, Cys-128, and Cys-169) and two cysteine residues of the PAPP-A subunit (Cys-381 and Cys-652) change their status from the uncomplexed to the complexed states. Upon complex formation, three disulfide bonds of pro-MBP, which connect the acidic propiece with the basic, mature portion, are disrupted. In the PAPP-A.pro-MBP complex, two of these form the basis of both two interchain disulfide bonds between the PAPP-A and the pro-MBP subunits and two disulfide bonds responsible for pro-MBP dimerization, respectively. Based on the status of the reactants, we investigated the role of individual cysteine residues upon complex formation by mutagenesis of specific cysteine residues of both subunits. Our findings allow us to depict a hypothetical model of how the PAPPA.pro-MBP complex is formed. In addition, we have demonstrated that complex formation is greatly enhanced by the addition of micromolar concentrations of reductants. It is therefore possible that the activity in vivo of PAPP-A is controlled by the redox potential, and it is further tempting to speculate that such mechanism operates under pathological conditions of altered redox potential.


Subject(s)
Enzyme Inhibitors/pharmacology , Eosinophil Major Basic Protein/physiology , Peptide Hydrolases/pharmacology , Pregnancy-Associated Plasma Protein-A/chemistry , Protein Precursors/chemistry , Blotting, Western , Cell Line , Cell Proliferation , Chromatography , Chromatography, High Pressure Liquid , Cysteine/chemistry , Cytokines/chemistry , Dimerization , Disulfides/chemistry , Dose-Response Relationship, Drug , Electrophoresis, Polyacrylamide Gel , Enzyme-Linked Immunosorbent Assay , Glutathione/metabolism , Humans , Models, Biological , Models, Chemical , Models, Genetic , Mutagenesis, Site-Directed , Oxidation-Reduction , Oxygen/metabolism , Peptides/chemistry , Pregnancy-Associated Plasma Protein-A/metabolism , Protein Binding , Protein Structure, Tertiary , Time Factors , Transfection
11.
J Biol Chem ; 278(4): 2106-17, 2003 Jan 24.
Article in English | MEDLINE | ID: mdl-12421832

ABSTRACT

Pregnancy-associated plasma protein-A (PAPP-A) is a metzincin superfamily metalloproteinase responsible for cleavage of insulin-like growth factor-binding protein-4, thus causing release of bound insulin-like growth factor. PAPP-A is secreted as a dimer of 400 kDa but circulates in pregnancy as a disulfide-bound 500-kDa 2:2 complex with the proform of eosinophil major basic protein (pro-MBP), recently shown to function as a proteinase inhibitor of PAPP-A. Except for PAPP-A2, PAPP-A does not share global similarity with other proteins. Three lin-notch (LNR or LIN-12) modules and five complement control protein modules (also known as SCR modules) have been identified in PAPP-A by sequence similarity with other proteins, but no data are available that allow unambiguous prediction of disulfide bonds of these modules. To establish the connectivities of cysteine residues of the PAPP-A.pro-MBP complex, biochemical analyses of peptides derived from purified protein were performed. The PAPP-A subunit contains a total of 82 cysteine residues, of which 81 have been accounted for. The pro-MBP subunit contains 12 cysteine residues, of which 10 have been accounted for. Within the 2:2 complex, PAPP-A is dimerized by a single disulfide bond; pro-MBP is dimerized by two disulfides, and each PAPP-A subunit is connected to a pro-MBP subunit by two disulfide bonds. All other disulfides are intrachain bridges. We also show that of 13 potential sites for N-linked carbohydrate substitution of the PAPP-A subunit, 11 are occupied. The large number of disulfide bonds of the PAPP-A.pro-MBP complex imposes many restraints on polypeptide folding, and knowledge of the disulfide pattern of PAPP-A will facilitate structural studies based on recombinant expression of individual, putative PAPP-A domains. Furthermore, it will allow rational experimental design of functional studies aimed at understanding the formation of the PAPP-A.pro-MBP complex, as well as the inhibitory mechanism of pro-MBP.


Subject(s)
Blood Proteins/chemistry , Pregnancy-Associated Plasma Protein-A/chemistry , Ribonucleases , Amino Acid Sequence , Amino Acids/chemistry , Animals , Blood Proteins/metabolism , Blotting, Western , Carbohydrates/chemistry , Cations , Chromatography, Gel , Chromatography, High Pressure Liquid , Chromatography, Ion Exchange , Cyanogen Bromide/pharmacology , Cysteine/chemistry , DNA, Complementary/metabolism , Disulfides/chemistry , Electrophoresis, Polyacrylamide Gel , Eosinophil Granule Proteins , Humans , Mass Spectrometry , Molecular Sequence Data , Mutagenesis, Site-Directed , Peptides/chemistry , Pregnancy-Associated Plasma Protein-A/metabolism , Protein Binding , Protein Folding , Protein Structure, Tertiary , Recombinant Fusion Proteins/chemistry , Recombinant Proteins/chemistry , Sequence Homology, Amino Acid , Transfection
12.
J Biol Chem ; 277(46): 43698-706, 2002 Nov 15.
Article in English | MEDLINE | ID: mdl-12218066

ABSTRACT

The primary structure determination of the dimeric invertebrate alpha(2)-macroglobulin (alpha(2)M) from Limulus polyphemus has been completed by determining its sites of glycosylation and disulfide bridge pattern. Of seven potential glycosylation sites for N-linked glycosylation, six (Asn(275), Asn(307), Asn(866), Asn(896), Asn(1089), and Asn(1145)) carry common glucosamine-based carbohydrates groups, whereas one (Asn(80)) carries a carbohydrate chain containing both glucosamine and galactosamine. Nine disulfide bridges, which are homologues with bridges in human alpha(2)M, have been identified (Cys(228)-Cys(269), Cys(456)-Cys(580), Cys(612)-Cys(799), Cys(657)-Cys(707), Cys(849)-Cys(876), Cys(874)-Cys(910), Cys(946)-Cys(1328), Cys(1104)-Cys(1155), and Cys(1362)-Cys(1475)). In addition to these bridges, Limulus alpha(2)M contains three unique bridges that connect Cys(361) and Cys(382), Cys(1370) and Cys(1374), respectively, and Cys(719) in one subunit with the same residue in the other subunit of the dimer. The latter bridge forms the only interchain disulfide bridge in Limulus alpha(2)M. The location of this bridge within the bait region is discussed and compared with other alpha-macroglobulins. Several peptides identified in the course of determining the disulfide bridge pattern provided evidence for the existence of two forms of Limulus alpha(2)M. The two forms have a high degree of sequence identity, but they differ extensively in large parts of their bait regions suggesting that they have different inhibitory spectra. The two forms (Limulus alpha(2)M-1 and -2) are most likely present in an approximately 2:1 ratio in the hemolymph of each animal, and they can be partially separated on a Mono Q column at pH 7.4 by applying a shallow gradient of NaCl.


Subject(s)
Carbohydrates/chemistry , Disulfides , Horseshoe Crabs/metabolism , alpha-Macroglobulins/chemistry , alpha-Macroglobulins/metabolism , Amino Acid Sequence , Animals , Asparagine/chemistry , Binding Sites , Chromatography, Gel , Chromatography, High Pressure Liquid , Chromatography, Ion Exchange , Cloning, Molecular , Cysteine/chemistry , DNA, Complementary/metabolism , Disulfides/metabolism , Electrophoresis, Polyacrylamide Gel , Glycosylation , Hemolymph , Humans , Hydrogen-Ion Concentration , Methylamines/pharmacology , Molecular Sequence Data , Polymerase Chain Reaction , Protein Binding , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Sodium Chloride/pharmacology , Trypsin/pharmacology
13.
Eur J Biochem ; 271(8): 1525-35, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15066178

ABSTRACT

The metalloproteinase pregnancy-associated plasma protein-A (PAPP-A) cleaves a subset of insulin-like growth factor binding proteins (IGFBP), which inhibit the activities of insulin-like growth factor (IGF). Through this proteolytic activity, PAPP-A is believed to regulate IGF bioavailability in several biological systems, including the human reproductive system and the cardiovascular system. PAPP-A adheres to mammalian cells by interactions with glycosaminoglycan (GAG), thus targeting the proteolytic activity of PAPP-A to the cell surface. Based on site-directed mutagenesis, we here delineate the PAPP-A GAG-binding site in the C-terminal modules CCP3 and CCP4. Using heparin affinity chromatography, commonly employed in such studies, we define three clusters of arginines and lysines of CCP3, which are important for the interaction of PAPP-A with heparin. In a model of PAPP-A CCP3-CCP4, basic residues of these sequence clusters form a contiguous patch located on one side of the structure. Binding to the unknown, natural cell surface receptor of PAPP-A, assessed by flow cytometry, also depends on residues of these three basic clusters. However, single or double residue substitutions generally have a modest effect on PAPP-A heparin binding assessed by chromatography, but cell surface adhesion was critically reduced by several of these substitutions, emphasizing the relevance of analysis by flow cytometry. The contributions of positively charged residues located in CCP4 were all minor when analyzed by heparin affinity chromatography. However, the mutation of CCP4 residues Arg1459 and Lys1460 to Ala almost abrogated cell surface adhesion. Furthermore, when acidic residues of the homologous proteinase PAPP-A2 (Asp1547, Glu1555 and Glu1567) were introduced into the corresponding positions in the sequence of PAPP-A, located in each of the three basic clusters of CCP3, binding to heparin was strongly impaired and cell surface binding was abrogated. This explains, at least in part, why PAPP-A2 lacks the ability of cell surface adhesion, and further emphasizes the role of the basic clusters defined in PAPP-A.


Subject(s)
Amino Acids, Basic/metabolism , Cell Adhesion/physiology , Pregnancy-Associated Plasma Protein-A/chemistry , Pregnancy-Associated Plasma Protein-A/metabolism , Amino Acid Sequence , Amino Acids, Acidic/genetics , Amino Acids, Acidic/metabolism , Amino Acids, Basic/chemistry , Amino Acids, Basic/genetics , Binding Sites , Cell Line , Cell Membrane/metabolism , Chromatography, Affinity/methods , Flow Cytometry , Glycosaminoglycans/metabolism , Heparin/metabolism , Humans , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Pregnancy-Associated Plasma Protein-A/genetics , Protein Binding , Protein Subunits , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid
14.
J Biol Chem ; 279(37): 38525-31, 2004 Sep 10.
Article in English | MEDLINE | ID: mdl-15262980

ABSTRACT

The Lin12-Notch repeat (LNR) module of about 35 residues is a hallmark of the Notch receptor family. Three copies, arranged in tandem, are invariably present in the extracellular portion of the Notch receptors. Although their function is unknown, genetic and biochemical data indicate that the LNR modules participate in the regulation of ligand-induced proteolytic cleavage of the Notch receptor, a prerequisite to intramembrane cleavage and Notch signaling. Outside the Notch receptor family, the LNR module is present only in the metalloproteinase pregnancy-associated plasma protein-A (PAPP-A) and its homologue PAPP-A2, which also contain three copies. Curiously, LNR modules 1 and 2 are present within the proteolytic domain of PAPP-A/A2, but LNR3 is separated from LNR2 by more than 1000 amino acids. The growth factor antagonists insulin-like growth factor-binding protein (IGFBP)-4 and -5 are both substrates of PAPP-A. We provide here evidence that the PAPP-A LNR modules function together to determine the proteolytic specificity of PAPP-A. Analysis of C-terminally truncated PAPP-A mutants followed by the analysis of LNR deletion mutants demonstrated that each of the three PAPP-A LNR modules is strictly required for proteolytic activity against IGFBP-4 but not for proteolytic activity against IGFBP-5. Individual substitution of conserved LNR residues predicted to participate in calcium coordination caused elimination (D341A, D356A, D389A, D1484A, D1499A, and D1502A) or a significant reduction (D359A and E392A) of IGFBP-4 proteolysis, whereas IGFBP-5 proteolysis was unaffected. The activity of the latter mutants against IGFBP-4 could be partially rescued by calcium, and the addition of the calcium-binding protein calbindin D9k to wild-type PAPP-A eliminated activity against IGFBP-4 but not against IGFBP-5, demonstrating that the PAPP-A LNR modules bind calcium ions. We propose a model in which LNR3 is spatially localized in proximity to LNR1 and -2, forming a single functional unit.


Subject(s)
Calcium/metabolism , Pregnancy-Associated Plasma Protein-A/chemistry , Amino Acid Sequence , Blotting, Western , Cell Line , Dose-Response Relationship, Drug , Endopeptidases/chemistry , Enzyme-Linked Immunosorbent Assay , Humans , Insulin-Like Growth Factor Binding Protein 4/metabolism , Insulin-Like Growth Factor Binding Protein 5/metabolism , Ions , Ligands , Membrane Proteins/metabolism , Molecular Sequence Data , Mutation , Plasmids/metabolism , Pregnancy-Associated Plasma Protein-A/metabolism , Protein Binding , Protein Structure, Tertiary , Receptors, Notch , Sequence Homology, Amino Acid , Transfection
15.
J Biol Chem ; 277(49): 47225-34, 2002 Dec 06.
Article in English | MEDLINE | ID: mdl-12370176

ABSTRACT

The activities of insulin-like growth factor (IGF)-I and -II are regulated by IGF-binding proteins (IGFBPs). Cleavage of IGFBP-4 by the metalloproteinase pregnancy-associated plasma protein-A (PAPP-A) causes release of bound IGF and has been established in several biological systems including the human reproductive system. Using flow cytometry, we first demonstrate that PAPP-A reversibly binds to the cell surface of several cell types analyzed. Heparin and heparan sulfate, but not dermatan or chondroitin sulfate, effectively compete for PAPP-A surface binding, and because incubation of cells with heparinase abrogated PAPP-A adhesion, binding is probably mediated by a cell surface heparan sulfate proteoglycan. Furthermore, the proteolytic activity of PAPP-A is preserved while bound to cells, suggesting that adhesion functions to target its activity to the vicinity of the IGF receptor, decreasing the probability that released IGF is captured by another IGFBP molecule before receptor binding. This mechanism potentially functions in both autocrine and paracrine regulation, as PAPP-A need not be synthesized in a cell to which it adheres. A truncated PAPP-A variant without the five short consensus repeats in the C-terminal third of the 1547-residue PAPP-A subunit, lacked surface binding. We also show that PAPP-A2, a recently discovered IGFBP-5 proteinase with homology to PAPP-A, does not bind cells. This finding allowed further mapping of the PAPP-A adhesion site to short consensus repeat modules 3 and 4 by the expression and analysis of nine PAPP-A/PAPP-A2 chimeras. Interestingly, the proteolytically inactive, disulfide-bound complex of PAPP-A and the proform of eosinophil major basic protein (proMBP), PAPP-A.proMBP, shows only weak surface binding, probably because the adhesion site of PAPP-A is occupied by heparan sulfate, known to be covalently bound to proMBP. This hypothesis was further substantiated by demonstrating that heparinase treatment of PAPP-A.proMBP restores surface binding. We finally propose a model in which IGF bioactivity is regulated by reversible cell surface binding of PAPP-A, which in turn is regulated by proMBP.


Subject(s)
Blood Proteins/chemistry , Pregnancy-Associated Plasma Protein-A/chemistry , Ribonucleases , Binding Sites , Blood Proteins/metabolism , Blotting, Western , Cell Adhesion , Cell Line , Cell Membrane/metabolism , Cysteine/chemistry , DNA, Complementary/metabolism , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Eosinophil Granule Proteins , Flow Cytometry , Glycosaminoglycans/pharmacology , Heparin/metabolism , Heparitin Sulfate/metabolism , Humans , Insulin-Like Growth Factor I/metabolism , Models, Biological , Plasmids/metabolism , Pregnancy-Associated Plasma Protein-A/metabolism , Protein Binding , Protein Structure, Tertiary , Transfection
16.
Eur J Biochem ; 269(8): 2247-56, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11985604

ABSTRACT

Murine pregnancy-associated plasma protein-A (PAPP-A) cDNA encoding a 1545 amino-acid protein has been cloned. We have also identified and cloned cDNA that encodes a novel variant of PAPP-A, PAPP-Ai, carrying a 29-residue highly basic insert. The point of insertion corresponds to a junction between two exons in the human PAPP-A gene. The human intron flanked by these exons does not encode a homologous corresponding insert, which is unique to the mouse. The overall sequence identity between murine and human PAPP-A is 91%, and murine PAPP-A contains sequence motifs previously described in the sequence of human PAPP-A. Through expression in mammalian cells, we show that murine PAPP-A and PAPP-Ai are active metalloproteinases, both capable of cleaving insulin-like growth factor binding protein (IGFBP)-4 and -5. Cleavage of IGFBP-4 is dramatically enhanced by the addition of IGF, whereas cleavage of IGFBP-5 is slightly inhibited by IGF, as previously established with human PAPP-A. Surprisingly, however, quantitative analyses demonstrate that the murine PAPP-Ai cleaves IGFBP-4 very slowly compared to PAPP-A, even though its ability to cleave IGFBP-5 is unaffected by the presence of the insert. By RT-PCR analysis, we find that both variants are expressed in several tissues. The level of mRNA in the murine placenta does not exceed the levels of other tissues analyzed. Furthermore, the IGFBP-4-proteolytic activity of murine pregnancy serum is not elevated. This is in striking contrast to the increase seen in human pregnancy serum, and the expression of PAPP-A in the human placenta, which exceeds other tissues at least 250-fold. Interestingly, the position of the insert of PAPP-Ai, within the proteolytic domain, lies in close proximity to the cysteine residue, which in human PAPP-A forms a disulfide bond with the proform of eosinophil major basic protein (proMBP). ProMBP functions as a proteinase inhibitor in the PAPP-A-proMBP complex, but whether any mechanistic parallel on regulation of proteolytic activity can be drawn between the insert of PAPP-Ai and the linkage to proMBP is not known. Importantly, these data support the development of the mouse as a model organism for the study of PAPP-A, which must take into account the differences between the mouse and the human.


Subject(s)
Pregnancy-Associated Plasma Protein-A/biosynthesis , Recombinant Proteins/biosynthesis , Amino Acid Sequence , Animals , Female , Humans , Insulin-Like Growth Factor Binding Protein 4/metabolism , Insulin-Like Growth Factor Binding Protein 5/metabolism , Mice , Molecular Sequence Data , Organ Specificity/physiology , Pregnancy-Associated Plasma Protein-A/genetics , Recombinant Proteins/genetics , Sequence Alignment , Sequence Homology , Substrate Specificity/physiology
17.
Biochem J ; 367(Pt 1): 31-40, 2002 Oct 01.
Article in English | MEDLINE | ID: mdl-12241545

ABSTRACT

Human pregnancy-associated plasma protein-A (PAPP-A) cleaves insulin-like growth factor (IGF) binding protein-4 (IGFBP-4), causing a dramatic reduction in its affinity for IGF-I and -II. Through this mechanism, PAPP-A is a regulator of IGF bioactivity in several systems, including the human ovary and the cardiovascular system. PAPP-A belongs to the metzincin superfamily of zinc metalloproteinases, and is the founding member of a fifth metzincin family, the pappalysins. Herein, we first determined that PAPP-A cleaves IGFBP-4 at a single site (Met-135/Lys-136), and we analysed the influence of ionic strength, pH and zinc ion concentration on the cleavage reaction. Secondly, we sought to delineate the role of substrate residues in PAPP-A-mediated cleavage by the construction and analysis of 30 IGFBP-4 mutants in which various residues were replaced by alanine, by the analysis of eight mutants of IGFBP-5 (found recently to be a second PAPP-A substrate), and by cleavage analysis of synthetic peptides derived from IGFBP-4. Our data reveal a complex mode of substrate recognition and/or binding, pointing at important roles for several basic residues located up to 16 residues N-terminal to the scissile bond. An unexpected parallel can be drawn with an intracellular enzyme, the mitochondrial processing peptidase, that may help us to understand properties of the pappalysins. Further, proteinase-resistant variants of IGFBP-4 and -5, presented here, will be useful tools for the study of proteolysis in cell-based systems, and our finding that a synthetic peptide can be cleaved by PAPP-A provides the basis for development of quantitative assays for the investigation of PAPP-A enzyme kinetics.


Subject(s)
Mutagenesis, Site-Directed , Peptides/chemistry , Pregnancy-Associated Plasma Protein-A/chemistry , Amino Acid Sequence , Binding Sites , Cell Line , Dose-Response Relationship, Drug , Humans , Hydrogen-Ion Concentration , Insulin-Like Growth Factor Binding Protein 4/metabolism , Insulin-Like Growth Factor Binding Protein 5/metabolism , Ions , Kinetics , Molecular Sequence Data , Mutation , Plasmids/metabolism , Pregnancy-Associated Plasma Protein-A/metabolism , Proteins/chemistry , Sequence Homology, Amino Acid , Substrate Specificity , Transfection , Zinc/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL