Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
J Mol Biol ; 436(16): 168667, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38901640

ABSTRACT

The excessive deposition of fibrillar collagens is a hallmark of fibrosis. Collagen fibril formation requires proteolytic maturations by Procollagen N- and C-proteinases (PNPs and PCPs) to remove the N- and C-propeptides which maintain procollagens in the soluble form. Procollagen C-Proteinase Enhancer-1 (PCPE-1, a glycoprotein composed of two CUB domains and one NTR domain) is a regulatory protein that activates the C-terminal processing of procollagens by the main PCPs. It is often up-regulated in fibrotic diseases and represents a promising target for the development of novel anti-fibrotic strategies. Here, our objective was to develop the first antagonists of PCPE-1, based on the nanobody scaffold. Using both an in vivo selection through the immunization of a llama and an in vitro selection with a synthetic library, we generated 18 nanobodies directed against the CUB domains of PCPE1, which carry its enhancing activity. Among them, I5 from the immune library and H4 from the synthetic library have a high affinity for PCPE-1 and inhibit its interaction with procollagens. The crystal structure of the complex formed by PCPE-1, H4 and I5 showed that they have distinct epitopes and enabled the design of a biparatopic fusion, the diabody diab-D1. Diab-D1 has a sub-nanomolar affinity for PCPE-1 and is a potent antagonist of its activity, preventing the stimulation of procollagen cleavage in vitro. Moreover, Diab-D1 is also effective in reducing the proteolytic maturation of procollagen I in cultures of human dermal fibroblasts and hence holds great promise as a tool to modulate collagen deposition in fibrotic conditions.

2.
Nat Commun ; 14(1): 8020, 2023 Dec 04.
Article in English | MEDLINE | ID: mdl-38049428

ABSTRACT

BMP-1/tolloid-like proteinases (BTPs) are major players in tissue morphogenesis, growth and repair. They act by promoting the deposition of structural extracellular matrix proteins and by controlling the activity of matricellular proteins and TGF-ß superfamily growth factors. They have also been implicated in several pathological conditions such as fibrosis, cancer, metabolic disorders and bone diseases. Despite this broad range of pathophysiological functions, the putative existence of a specific endogenous inhibitor capable of controlling their activities could never be confirmed. Here, we show that procollagen C-proteinase enhancer-2 (PCPE-2), a protein previously reported to bind fibrillar collagens and to promote their BTP-dependent maturation, is primarily a potent and specific inhibitor of BTPs which can counteract their proteolytic activities through direct binding. PCPE-2 therefore differs from the cognate PCPE-1 protein and extends the possibilities to fine-tune BTP activities, both in physiological conditions and in therapeutic settings.


Subject(s)
Glycoproteins , Peptide Hydrolases , Humans , Glycoproteins/metabolism , Extracellular Matrix Proteins/metabolism , Morphogenesis , Intercellular Signaling Peptides and Proteins
3.
PLoS Pathog ; 19(5): e1011368, 2023 05.
Article in English | MEDLINE | ID: mdl-37155700

ABSTRACT

The bacterial human pathogen Helicobacter pylori produces a type IV secretion system (cagT4SS) to inject the oncoprotein CagA into gastric cells. The cagT4SS external pilus mediates attachment of the apparatus to the target cell and the delivery of CagA. While the composition of the pilus is unclear, CagI is present at the surface of the bacterium and required for pilus formation. Here, we have investigated the properties of CagI by an integrative structural biology approach. Using Alpha Fold 2 and Small Angle X-ray scattering, it was found that CagI forms elongated dimers mediated by rod-shape N-terminal domains (CagIN) prolonged by globular C-terminal domains (CagIC). Three Designed Ankyrin Repeat Proteins (DARPins) K2, K5 and K8 selected against CagI interacted with CagIC with subnanomolar affinities. The crystal structures of the CagI:K2 and CagI:K5 complexes were solved and identified the interfaces between the molecules, thereby providing a structural explanation for the difference in affinity between the two binders. Purified CagI and CagIC were found to interact with adenocarcinoma gastric (AGS) cells, induced cell spreading and the interaction was inhibited by K2. The same DARPin inhibited CagA translocation by up to 65% in AGS cells while inhibition levels were 40% and 30% with K8 and K5, respectively. Our study suggests that CagIC plays a key role in cagT4SS-mediated CagA translocation and that DARPins targeting CagI represent potent inhibitors of the cagT4SS, a crucial risk factor for gastric cancer development.


Subject(s)
Helicobacter Infections , Helicobacter pylori , Humans , Bacterial Proteins/metabolism , Antigens, Bacterial/metabolism , Type IV Secretion Systems/genetics , Type IV Secretion Systems/metabolism , Designed Ankyrin Repeat Proteins , Helicobacter pylori/metabolism , Helicobacter Infections/microbiology
4.
Sci Rep ; 12(1): 14850, 2022 09 01.
Article in English | MEDLINE | ID: mdl-36050373

ABSTRACT

Sizzled (Szl) is both a secreted frizzled related protein (sFRP) and a naturally occurring inhibitor of the zinc metalloproteinase bone morphogenetic protein-1 (BMP-1), a key regulator of extracellular matrix assembly and growth factor activation. Here we present a new crystal structure for Szl which differs from that previously reported by a large scale (90°) hinge rotation between its cysteine-rich and netrin-like domains. We also present results of a molecular docking analysis showing interactions likely to be involved in the inhibition of BMP-1 activity by Szl. When compared with known structures of BMP-1 in complex with small molecule inhibitors, this reveals features that may be helpful in the design of new inhibitors to prevent the excessive accumulation of extracellular matrix that is the hallmark of fibrotic diseases.


Subject(s)
Intercellular Signaling Peptides and Proteins , Xenopus Proteins , Bone Morphogenetic Protein 1/metabolism , Bone Morphogenetic Protein 2/metabolism , Bone Morphogenetic Protein 7/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Molecular Docking Simulation , Protein Binding , Xenopus Proteins/metabolism
5.
Matrix Biol Plus ; 11: 100062, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34435180

ABSTRACT

The correct balance between collagen synthesis and degradation is essential for almost every aspect of life, from development to healthy aging, reproduction and wound healing. When this balance is compromised by external or internal stress signals, it very often leads to disease as is the case in fibrotic conditions. Fibrosis occurs in the context of defective tissue repair and is characterized by the excessive, aberrant and debilitating deposition of fibril-forming collagens. Therefore, the numerous proteins involved in the biosynthesis of fibrillar collagens represent a potential and still underexploited source of therapeutic targets to prevent fibrosis. One such target is procollagen C-proteinase enhancer-1 (PCPE-1) which has the unique ability to accelerate procollagen maturation by BMP-1/tolloid-like proteinases (BTPs) and contributes to trigger collagen fibrillogenesis, without interfering with other BTP functions or the activities of other extracellular metalloproteinases. This role is achieved through a fine-tuned mechanism of action that is close to being elucidated and offers promising perspectives for drug design. Finally, the in vivo data accumulated in recent years also confirm that PCPE-1 overexpression is a general feature and early marker of fibrosis. In this review, we describe the results which presently support the driving role of PCPE-1 in fibrosis and discuss the questions that remain to be solved to validate its use as a biomarker or therapeutic target.

6.
FEBS Open Bio ; 8(12): 2011-2021, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30524951

ABSTRACT

BMP-1/tolloid-like proteinases belong to the astacin family of human metalloproteinases, together with meprins and ovastacin. They represent promising targets to treat or prevent a wide range of diseases such as fibrotic disorders or cancer. However, the study of their pathophysiological roles is still impaired by the lack of well-characterized inhibitors and the questions that remain regarding their selectivity and in vivo efficiency. As a first step towards the identification of suitable tools to be used in functional studies, we have undertaken a systematic comparison of seven molecules known to affect the proteolytic activity of human astacins including three hydroxamates (FG-2575, UK383,367, S33A), the protein sizzled, a new phosphinic inhibitor (RXP-1001) and broad-spectrum protease inhibitors (GM6001, actinonin). Their efficacy in vitro, their cellular toxicity and efficacy in cell cultures were thoroughly characterized. We found that these molecules display very different potency and selectivity profiles, with hydroxamate FG-2575 and the protein sizzled being very powerful and selective inhibitors of BMP-1, whereas phosphinic peptide RXP-1001 behaves as a broad-spectrum inhibitor of astacins. Their use should therefore be carefully considered in agreement with the aim of the study to avoid result misinterpretation.

7.
Structure ; 26(10): 1384-1392.e3, 2018 10 02.
Article in English | MEDLINE | ID: mdl-30078642

ABSTRACT

Procollagen C-proteinase enhancer-1 (PCPE-1) is a secreted protein that specifically accelerates proteolytic release of the C-propeptides from fibrillar procollagens, a crucial step in fibril assembly. As such, it is a potential therapeutic target to improve tissue repair and prevent fibrosis, a major cause of mortality worldwide. Here we present the crystal structure of the active CUB1CUB2 fragment of PCPE-1 bound to the C-propeptide trimer of procollagen III (CPIII). This shows that the two CUB domains bind to two different chains of CPIII and that the N-terminal region of one CPIII chain, close to the proteolytic cleavage site, lies in the cleft between CUB1 and CUB2. This suggests that enhancing activity involves unraveling of this chain from the rest of the trimer, thus facilitating the action of the proteinase involved. Support for this hypothesis comes from site-directed mutagenesis, enzyme assays, binding studies, and molecular modeling.


Subject(s)
Collagen Type III/metabolism , Extracellular Matrix Proteins/chemistry , Extracellular Matrix Proteins/metabolism , Glycoproteins/chemistry , Glycoproteins/metabolism , Mutagenesis, Site-Directed/methods , Binding Sites , Crystallography, X-Ray , Extracellular Matrix Proteins/genetics , Female , Glycoproteins/genetics , HEK293 Cells , Humans , Models, Molecular , Protein Binding , Proteolysis
8.
Sci Rep ; 7(1): 16958, 2017 12 05.
Article in English | MEDLINE | ID: mdl-29209066

ABSTRACT

The defence collagens C1q and mannose-binding lectin (MBL) are immune recognition proteins that associate with the serine proteinases C1r/C1s and MBL-associated serine proteases (MASPs) to trigger activation of complement, a major innate immune system. Bone morphogenetic protein-1 (BMP-1)/tolloid-like proteinases (BTPs) are metalloproteinases with major roles in extracellular matrix assembly and growth factor signalling. Despite their different functions, C1r/C1s/MASPs and BTPs share structural similarities, including a specific CUB-EGF-CUB domain arrangement found only in these enzymes that mediates interactions with collagen-like proteins, suggesting a possible functional relationship. Here we investigated the potential interactions between the defence collagens C1q and MBL and the BTPs BMP-1 and mammalian tolloid-like-1 (mTLL-1). C1q and MBL bound to immobilized BMP-1 and mTLL-1 with nanomolar affinities. These interactions involved the collagen-like regions of the defence collagens and were inhibited by pre-incubation of C1q or MBL with their cognate complement proteinases. Soluble BMP-1 and mTLL-1 did not inhibit complement activation and the defence collagens were neither substrates nor inhibitors of BMP-1. Finally, C1q co-localized with BMP-1 in skin biopsies following melanoma excision and from patients with recessive dystrophic epidermolysis bullosa. The observed interactions provide support for a functional link between complement and BTPs during inflammation and tissue repair.


Subject(s)
Bone Morphogenetic Protein 1/metabolism , Complement C1q/metabolism , Mannose-Binding Lectin/metabolism , Tolloid-Like Metalloproteinases/metabolism , Binding Sites , Bone Morphogenetic Protein 1/genetics , Complement Activation , Epidermolysis Bullosa Dystrophica/metabolism , Epidermolysis Bullosa Dystrophica/pathology , Humans , Melanoma/metabolism , Melanoma/pathology , Skin Neoplasms/metabolism , Skin Neoplasms/pathology
9.
Nat Commun ; 8: 14671, 2017 03 10.
Article in English | MEDLINE | ID: mdl-28281531

ABSTRACT

Fibrillar collagen molecules are synthesized as precursors, procollagens, with large propeptide extensions. While a homotrimeric form (three α1 chains) has been reported in embryonic tissues as well as in diseases (cancer, fibrosis, genetic disorders), collagen type I usually occurs as a heterotrimer (two α1 chains and one α2 chain). Inside the cell, the role of the C-terminal propeptides is to gather together the correct combination of three α chains during molecular assembly, but how this occurs for different forms of the same collagen type is so far unknown. Here, by structural and mutagenic analysis, we identify key amino acid residues in the α1 and α2 C-propeptides that determine homo- and heterotrimerization. A naturally occurring mutation in one of these alters the homo/heterotrimer balance. These results show how the C-propeptide of the α2 chain has specifically evolved to permit the appearance of heterotrimeric collagen I, the major extracellular building block among the metazoa.


Subject(s)
Collagen Type I/chemistry , Amino Acid Sequence , Binding Sites , Cloning, Molecular , Collagen Type I/genetics , Collagen Type I/metabolism , Collagen Type I, alpha 1 Chain , Crystallography, X-Ray , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , HEK293 Cells , Humans , Models, Molecular , Mutation , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Protein Multimerization , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid
10.
FASEB J ; 30(5): 1741-56, 2016 05.
Article in English | MEDLINE | ID: mdl-26740262

ABSTRACT

A disintegrin and metalloproteinase with thrombospondin type I motif (ADAMTS)2, 3, and 14 are collectively named procollagen N-proteinases (pNPs) because of their specific ability to cleave the aminopropeptide of fibrillar procollagens. Several reports also indicate that they could be involved in other biological processes, such as blood coagulation, development, and male fertility, but the potential substrates associated with these activities remain unknown. Using the recently described N-terminal amine isotopic labeling of substrate approach, we analyzed the secretomes of human fibroblasts and identified 8, 17, and 22 candidate substrates for ADAMTS2, 3, and 14, respectively. Among these newly identified substrates, many are components of the extracellular matrix and/or proteins related to cell signaling such as latent TGF-ß binding protein 1, TGF-ß RIII, and dickkopf-related protein 3. Candidate substrates for the 3 ADAMTS have been biochemically validated in different contexts, and the implication of ADAMTS2 in the control of TGF-ß activity has been further demonstrated in human fibroblasts. Finally, the cleavage site specificity was assessed showing a clear and unique preference for nonpolar or slightly hydrophobic amino acids. This work shows that the activities of the pNPs extend far beyond the classically reported processing of the aminopropeptide of fibrillar collagens and that they should now be considered as multilevel regulators of matrix deposition and remodeling.-Bekhouche, M., Leduc, C., Dupont, L., Janssen, L., Delolme, F., Vadon-Le Goff, S., Smargiasso, N., Baiwir, D., Mazzucchelli, G., Zanella-Cleon, I., Dubail, J., De Pauw, E., Nusgens, B., Hulmes, D. J. S., Moali, C., Colige, A. Determination of the substrate repertoire of ADAMTS2, 3, and 14 significantly broadens their functions and identifies extracellular matrix organization and TGF-ß signaling as primary targets.


Subject(s)
ADAMTS Proteins/metabolism , Extracellular Matrix/metabolism , Procollagen N-Endopeptidase/metabolism , Transforming Growth Factor beta/metabolism , ADAMTS Proteins/genetics , Adaptor Proteins, Signal Transducing , Chemokines , Gene Expression Regulation/physiology , HEK293 Cells , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Latent TGF-beta Binding Proteins/genetics , Latent TGF-beta Binding Proteins/metabolism , Procollagen N-Endopeptidase/genetics , Proteoglycans/genetics , Proteoglycans/metabolism , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction/physiology , Transforming Growth Factor beta/genetics
11.
Matrix Biol ; 44-46: 14-23, 2015.
Article in English | MEDLINE | ID: mdl-25701650

ABSTRACT

Bone morphogenetic protein-1 (BMP-1)/tolloid-like proteinases, here called BTPs, include the proteases originally identified for their roles in the C-terminal maturation of fibrillar procollagens ("procollagen C-proteinase"). Though numerous other substrates have since been discovered, the BTPs remain the main proteases involved in extracellular matrix assembly with little or no implication in matrix degradation. During the same period however, the BTPs have also become established as important proteases in the activation of growth factors, including TGF-ß1, BMP-2/-4, GDF-8/-11 and IGFs, as well as the release of anti-angiogenic fragments from parent proteins. The BTPs are therefore key players in many pathophysiological processes such as morphogenesis, tissue repair and tumor progression. This mini-review summarizes our current knowledge of the functions of BTPs, their substrates and unusual mechanisms of regulation, and discusses their potential as new targets for future therapies.


Subject(s)
Bone Morphogenetic Protein 1/metabolism , Extracellular Matrix/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Angiogenesis Inhibitors/metabolism , Animals , Extracellular Matrix Proteins/metabolism , Humans , Morphogenesis , Regeneration
12.
Cell Mol Life Sci ; 72(5): 1009-27, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25260970

ABSTRACT

The metalloproteinase BMP-1 (bone morphogenetic protein-1) plays a major role in the control of extracellular matrix (ECM) assembly and growth factor activation. Most of the growth factors activated by BMP-1 are members of the TGF-ß superfamily known to regulate multiple biological processes including embryonic development, wound healing, inflammation and tumor progression. In this study, we used an iTRAQ (isobaric tags for relative and absolute quantification)-based quantitative proteomic approach to reveal the release of proteolytic fragments from the cell surface or the ECM by BMP-1. Thirty-eight extracellular proteins were found in significantly higher or lower amounts in the conditioned medium of HT1080 cells overexpressing BMP-1 and thus, could be considered as candidate substrates. Strikingly, three of these new candidates (betaglycan, CD109 and neuropilin-1) were TGF-ß co-receptors, also acting as antagonists when released from the cell surface, and were chosen for further substrate validation. Betaglycan and CD109 proved to be directly cleaved by BMP-1 and the corresponding cleavage sites were extensively characterized using a new mass spectrometry approach. Furthermore, we could show that the ability of betaglycan and CD109 to interact with TGF-ß was altered after cleavage by BMP-1, leading to increased and prolonged SMAD2 phosphorylation in BMP-1-overexpressing cells. Betaglycan processing was also observed in primary corneal keratocytes, indicating a general and novel mechanism by which BMP-1 directly affects signaling by controlling TGF-ß co-receptor activity. The proteomic data have been submitted to ProteomeXchange with the identifier PXD000786 and doi: 10.6019/PXD000786 .


Subject(s)
Bone Morphogenetic Protein 1/metabolism , Proteomics , Receptors, Transforming Growth Factor beta/metabolism , Antigens, CD/metabolism , Bone Morphogenetic Protein 1/genetics , Cell Line, Tumor , Chromatography, High Pressure Liquid , Extracellular Matrix/metabolism , GPI-Linked Proteins/metabolism , Humans , Neoplasm Proteins/metabolism , Neuropilin-1/metabolism , Peptides/analysis , Phosphorylation , Protein Binding , Proteoglycans/metabolism , Proteolysis , Signal Transduction , Smad2 Protein/metabolism , Spectrometry, Mass, Electrospray Ionization , Transforming Growth Factor beta/metabolism
13.
Proc Natl Acad Sci U S A ; 110(35): 14219-24, 2013 Aug 27.
Article in English | MEDLINE | ID: mdl-23940311

ABSTRACT

Type I fibrillar collagen is the most abundant protein in the human body, crucial for the formation and strength of bones, skin, and tendon. Proteolytic enzymes are essential for initiation of the assembly of collagen fibrils by cleaving off the propeptides. We report that Mep1a(-/-) and Mep1b(-/-) mice revealed lower amounts of mature collagen I compared with WT mice and exhibited significantly reduced collagen deposition in skin, along with markedly decreased tissue tensile strength. While exploring the mechanism of this phenotype, we found that cleavage of full-length human procollagen I heterotrimers by either meprin α or meprin ß led to the generation of mature collagen molecules that spontaneously assembled into collagen fibrils. Thus, meprin α and meprin ß are unique in their ability to process and release both C- and N-propeptides from type I procollagen in vitro and in vivo and contribute to the integrity of connective tissue in skin, with consequent implications for inherited connective tissue disorders.


Subject(s)
Collagen Type I/metabolism , Metalloendopeptidases/metabolism , Procollagen N-Endopeptidase/metabolism , Tensile Strength , Animals , CHO Cells , Cricetinae , Cricetulus , HEK293 Cells , Humans , Metalloendopeptidases/genetics , Mice , Mice, Knockout , Proteolysis , Skin/metabolism
14.
Proc Natl Acad Sci U S A ; 110(16): 6394-9, 2013 Apr 16.
Article in English | MEDLINE | ID: mdl-23550162

ABSTRACT

Tight regulation of collagen fibril deposition in the extracellular matrix is essential for normal tissue homeostasis and repair, defects in which are associated with several degenerative or fibrotic disorders. A key regulatory step in collagen fibril assembly is the C-terminal proteolytic processing of soluble procollagen precursors. This step, carried out mainly by bone morphogenetic protein-1/tolloid-like proteinases, is itself subject to regulation by procollagen C-proteinase enhancer proteins (PCPEs) which can dramatically increase bone morphogenetic protein-1/tolloid-like proteinase activity, in a substrate-specific manner. Although it is known that this enhancing activity requires binding of PCPE to the procollagen C-propeptide trimer, identification of the precise binding site has so far remained elusive. Here, use of small-angle X-ray scattering provides structural data on this protein complex indicating that PCPE binds to the stalk region of the procollagen C-propeptide trimer, where the three polypeptide chains associate together, at the junction with the base region. This is supported by site-directed mutagenesis, which identifies two highly conserved, surface-exposed lysine residues in this region of the trimer that are essential for binding, thus revealing structural parallels with the interactions of Complement C1r/C1s, Uegf, BMP-1 (CUB) domain-containing proteins in diverse biological systems such as complement activation, receptor signaling, and transport. Together with detailed kinetics and interaction analysis, these results provide insights into the mechanism of action of PCPEs and suggest clear strategies for the development of novel antifibrotic therapies.


Subject(s)
Bone Morphogenetic Protein 1/metabolism , Collagen Type III/metabolism , Extracellular Matrix/metabolism , Recombinant Proteins/metabolism , Animals , Binding Sites/genetics , Bone Morphogenetic Protein 1/genetics , Chromatography, Gel , Circular Dichroism , Collagen Type III/genetics , Electrophoresis, Polyacrylamide Gel , Extracellular Matrix Proteins , Glycoproteins , HEK293 Cells , Humans , Kinetics , Mutagenesis, Site-Directed , Polymerase Chain Reaction , Scattering, Small Angle , Surface Plasmon Resonance
15.
J Biol Chem ; 287(40): 33581-93, 2012 Sep 28.
Article in English | MEDLINE | ID: mdl-22825851

ABSTRACT

BMP-1/tolloid-like proteinases (BTPs) are major enzymes involved in extracellular matrix assembly and activation of bioactive molecules, both growth factors and anti-angiogenic molecules. Although the control of BTP activity by several enhancing molecules is well established, the possibility that regulation also occurs through endogenous inhibitors is still debated. Secreted frizzled-related proteins (sFRPs) have been studied as possible candidates, with highly contradictory results, after the demonstration that sizzled, a sFRP found in Xenopus and zebrafish, was a potent inhibitor of Xenopus and zebrafish tolloid-like proteases. In this study, we demonstrate that mammalian sFRP-1, -2, and -4 do not modify human BMP-1 activity on several of its known substrates including procollagen I, procollagen III, pN-collagen V, and prolysyl oxidase. In contrast, Xenopus sizzled appears as a tight binding inhibitor of human BMP-1, with a K(i) of 1.5 ± 0.5 nM, and is shown to strongly inhibit other human tolloid isoforms mTLD and mTLL-1. Because sizzled is the most potent inhibitor of human tolloid-like proteinases known to date, we have studied its mechanism of action in detail and shown that the frizzled domain of sizzled is both necessary and sufficient for inhibitory activity and that it acts directly on the catalytic domain of BMP-1. Residues in sizzled required for inhibition include Asp-92, which is shared by sFRP-1 and -2, and also Phe-94, Ser-43, and Glu-44, which are specific to sizzled, thereby providing a rational basis for the absence of inhibitory activity of human sFRPs.


Subject(s)
Bone Morphogenetic Protein 1/metabolism , Glycoproteins/metabolism , Xenopus Proteins/metabolism , Amino Acid Sequence , Animals , Extracellular Matrix/metabolism , Humans , Intracellular Signaling Peptides and Proteins , Matrix Metalloproteinases/metabolism , Mice , Models, Molecular , Molecular Sequence Data , Peptide Hydrolases/metabolism , Protease Inhibitors/pharmacology , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Signal Transduction , Surface Plasmon Resonance , Tissue Inhibitor of Metalloproteinases/metabolism , Wnt Proteins/metabolism , Xenopus laevis/metabolism
16.
J Biol Chem ; 286(45): 38932-8, 2011 Nov 11.
Article in English | MEDLINE | ID: mdl-21940633

ABSTRACT

Bone morphogenetic protein-1 (BMP-1) and the tolloid-like metalloproteinases control several aspects of embryonic development and tissue repair. Unlike other proteinases whose activities are regulated mainly by endogenous inhibitors, regulation of BMP-1/tolloid-like proteinases relies mostly on proteins that stimulate activity. Among these, procollagen C-proteinase enhancers (PCPEs) markedly increase BMP-1/tolloid-like proteinase activity on fibrillar procollagens, in a substrate-specific manner. Here, we performed a detailed quantitative study of the binding of PCPE-1 and of its minimal active fragment (CUB1-CUB2) to three regions of the procollagen III molecule: the triple helix, the C-telopeptide, and the C-propeptide. Contrary to results described elsewhere, we found the PCPE-1-binding sites to be located exclusively in the C-propeptide region. In addition, binding and enhancing activities were found to be independent of the glycosylation state of the C-propeptide. These data exclude previously proposed mechanisms for the action of PCPEs and also suggest new mechanisms to explain how these proteins can stimulate BMP-1/tolloid-like proteinases by up to 20-fold.


Subject(s)
Bone Morphogenetic Protein 1/metabolism , Collagen Type III/metabolism , Extracellular Matrix Proteins/metabolism , Glycoproteins/metabolism , Protein Processing, Post-Translational/physiology , Binding Sites , Bone Morphogenetic Protein 1/genetics , Cell Line, Transformed , Collagen Type III/genetics , Extracellular Matrix Proteins/genetics , Glycoproteins/genetics , Humans , Protein Structure, Secondary
17.
J Invest Dermatol ; 130(12): 2727-35, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20631730

ABSTRACT

Meprins α and ß, a subgroup of zinc metalloproteinases belonging to the astacin family, are known to cleave components of the extracellular matrix, either during physiological remodeling or in pathological situations. In this study we present a new role for meprins in matrix assembly, namely the proteolytic processing of procollagens. Both meprins α and ß release the N- and C-propeptides from procollagen III, with such processing events being critical steps in collagen fibril formation. In addition, both meprins cleave procollagen III at exactly the same site as the procollagen C-proteinases, including bone morphogenetic protein-1 (BMP-1) and other members of the tolloid proteinase family. Indeed, cleavage of procollagen III by meprins is more efficient than by BMP-1. In addition, unlike BMP-1, whose activity is stimulated by procollagen C-proteinase enhancer proteins (PCPEs), the activity of meprins on procollagen III is diminished by PCPE-1. Finally, following our earlier observations of meprin expression by human epidermal keratinocytes, meprin α is also shown to be expressed by human dermal fibroblasts. In the dermis of fibrotic skin (keloids), expression of meprin α increases and meprin ß begins to be detected. Our study suggests that meprins could be important players in several remodeling processes involving collagen fiber deposition.


Subject(s)
Collagen Type III/metabolism , Extracellular Matrix Proteins/metabolism , Keloid/pathology , Keratinocytes/physiology , Metalloendopeptidases/metabolism , Bone Morphogenetic Protein 1/metabolism , Cells, Cultured , Collagen Type III/genetics , Dermis/cytology , Dermis/physiology , Fibroblasts/cytology , Fibroblasts/physiology , Fibrosis , HEK293 Cells , Humans , Keloid/physiopathology , Keratinocytes/cytology , Substrate Specificity
18.
J Biol Chem ; 285(21): 15950-9, 2010 May 21.
Article in English | MEDLINE | ID: mdl-20207734

ABSTRACT

The netrin-like (NTR) domain is a feature of several extracellular proteins, most notably the N-terminal domain of tissue inhibitors of metalloproteinases (TIMPs), where it functions as a strong inhibitor of matrix metalloproteinases and some other members of the metzincin superfamily. The presence of a C-terminal NTR domain in procollagen C-proteinase enhancers (PCPEs), proteins that stimulate the activity of astacin-like tolloid proteinases, raises the possibility that this might also have inhibitory activity. Here we show that both long and short forms of the PCPE-1 NTR domain, the latter beginning at the N-terminal cysteine known to be critical for TIMP activity, show no inhibition, at micromolar concentrations, of several members of the metzincin superfamily, including matrix metalloproteinase-2, bone morphogenetic protein-1 (a tolloid proteinase), and different ADAMTS (a disintegrin and a metalloproteinase with thrombospondin motifs) proteinases from the adamalysin family. In contrast, we report that the NTR domain within PCPE-1 leads to superstimulation of bone morphogenetic protein-1 activity in the presence of heparin and heparan sulfate. These observations point to a new mechanism whereby binding to cell surface-associated or extracellular heparin-like sulfated glycosaminoglycans might provide a means to accelerate procollagen processing in specific cellular and extracellular microenvironments.


Subject(s)
ADAM Proteins/metabolism , Extracellular Matrix Proteins/metabolism , Glycoproteins/metabolism , Tolloid-Like Metalloproteinases/metabolism , ADAM Proteins/chemistry , ADAM Proteins/genetics , Cell Line , Extracellular Matrix Proteins/chemistry , Extracellular Matrix Proteins/genetics , Glycoproteins/chemistry , Glycoproteins/genetics , Humans , Procollagen/chemistry , Procollagen/genetics , Procollagen/metabolism , Protein Structure, Tertiary , Tissue Inhibitor of Metalloproteinases/chemistry , Tissue Inhibitor of Metalloproteinases/genetics , Tissue Inhibitor of Metalloproteinases/metabolism , Tolloid-Like Metalloproteinases/chemistry , Tolloid-Like Metalloproteinases/genetics
19.
J Biol Chem ; 284(48): 33437-46, 2009 Nov 27.
Article in English | MEDLINE | ID: mdl-19801683

ABSTRACT

Procollagen C-proteinase enhancers (PCPE-1 and -2) specifically activate bone morphogenetic protein-1 (BMP-1) and other members of the tolloid proteinase family during C-terminal processing of fibrillar collagen precursors. PCPEs consist of two CUB domains (CUB1 and CUB2) and one NTR domain separated by one short and one long linker. It was previously shown that PCPEs can strongly interact with procollagen molecules, but the exact mechanism by which they enhance BMP-1 activity remains largely unknown. Here, we used a series of deletion mutants of PCPE-1 and two chimeric constructs with repetitions of the same CUB domain to study the role of each domain and linker. Out of all the forms tested, only those containing both CUB1 and CUB2 were capable of enhancing BMP-1 activity and binding to a mini-procollagen substrate with nanomolar affinity. Both these properties were lost by individual CUB domains, which had dissociation constants at least three orders of magnitude higher. In addition, none of the constructs tested could inhibit PCPE activity, although CUB2CUB2NTR was found to modulate BMP-1 activity through direct complex formation with the enzyme, resulting in a decreased rate of substrate processing. Finally, increasing the length of the short linker between CUB1 and CUB2 was without detrimental effect on both activity and substrate binding. These data support the conclusion that CUB1 and CUB2 bind to the procollagen substrate in a cooperative manner, involving the short linker that provides a flexible tether linking the two binding regions.


Subject(s)
Bone Morphogenetic Protein 1/metabolism , Extracellular Matrix Proteins/metabolism , Glycoproteins/metabolism , Procollagen/metabolism , Amino Acid Sequence , Binding Sites , Binding, Competitive , Bone Morphogenetic Protein 1/genetics , Cell Line , Circular Dichroism , Electrophoresis, Polyacrylamide Gel , Extracellular Matrix Proteins/genetics , Glycoproteins/genetics , Humans , Kinetics , Mutation , Protein Binding , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...