Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
J Struct Biol ; 203(3): 247-254, 2018 09.
Article in English | MEDLINE | ID: mdl-29763735

ABSTRACT

Cleavage of collagen by collagenases such as matrix metalloproteinase 1 (MMP-1) is a key step in development, tissue remodeling, and tumor proliferation. The abundant heterotrimeric type I collagen composed of two α1(I) chains and one α2(I) chain is efficiently cleaved by MMP-1 at a unique site in the triple helix, a process which may be initiated by local unfolding within the peptide chains. Atypical homotrimers of the α1(I) chain, found in embryonic and cancer tissues, are very resistant to MMP cleavage. To investigate MMP-1 cleavage, recombinant homotrimers were constructed with sequences from the MMP cleavage regions of human collagen chains inserted into a host bacterial collagen protein system. All triple-helical constructs were cleaved by MMP-1, with α2(I) homotrimers cleaved efficiently at a rate similar to that seen for α1(II) and α1(III) homotrimers, while α1(I) homotrimers were cleaved at a much slower rate. The introduction of destabilizing Gly to Ser mutations within the human collagenase susceptible region of the α2(I) chain did not interfere with MMP-1 cleavage. Molecular dynamics simulations indicated a greater degree of transient hydrogen bond breaking in α2(I) homotrimers compared with α1(I) homotrimers at the MMP-1 cleavage site, and showed an extensive disruption of hydrogen bonding in the presence of a Gly to Ser mutation, consistent with chymotrypsin digestion results. This study indicates that α2(I) homotrimers are susceptible to MMP-1, proves that the presence of an α1(I) chain is not a requirement for α2(I) cleavage, and supports the importance of local unfolding of α2(I) in collagenase cleavage.


Subject(s)
Collagen Type I/chemistry , Collagenases/chemistry , Matrix Metalloproteinase 1/chemistry , Neoplasms/genetics , Amino Acid Sequence/genetics , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Binding Sites , Cell Proliferation/genetics , Collagen/chemistry , Collagen/genetics , Collagen Type I/genetics , Collagenases/genetics , Humans , Hydrogen Bonding , Matrix Metalloproteinase 1/genetics , Molecular Dynamics Simulation , Neoplasms/pathology , Protein Binding , Protein Conformation , Protein Conformation, alpha-Helical/genetics , Streptococcus pyogenes/chemistry
2.
Proc Natl Acad Sci U S A ; 113(39): 10884-9, 2016 09 27.
Article in English | MEDLINE | ID: mdl-27630193

ABSTRACT

It is well established that the expression profiles of multiple and possibly redundant matrix-remodeling proteases (e.g., collagenases) differ strongly in health, disease, and development. Although enzymatic redundancy might be inferred from their close similarity in structure, their in vivo activity can lead to extremely diverse tissue-remodeling outcomes. We observed that proteolysis of collagen-rich natural extracellular matrix (ECM), performed uniquely by individual homologous proteases, leads to distinct events that eventually affect overall ECM morphology, viscoelastic properties, and molecular composition. We revealed striking differences in the motility and signaling patterns, morphology, and gene-expression profiles of cells interacting with natural collagen-rich ECM degraded by different collagenases. Thus, in contrast to previous notions, matrix-remodeling systems are not redundant and give rise to precise ECM-cell crosstalk. Because ECM proteolysis is an abundant biochemical process that is critical for tissue homoeostasis, these results improve our fundamental understanding its complexity and its impact on cell behavior.


Subject(s)
Extracellular Matrix/metabolism , Matrix Metalloproteinase 13/metabolism , Matrix Metalloproteinase 1/metabolism , Proteolysis , Sequence Homology, Amino Acid , Animals , Cell-Matrix Junctions/metabolism , Collagen/metabolism , Collagen/ultrastructure , Elasticity , Extracellular Matrix/ultrastructure , Fibroblasts/metabolism , Humans , Imaging, Three-Dimensional , Principal Component Analysis , Rats , Rheology , Viscosity
3.
J Biol Chem ; 291(42): 22160-22172, 2016 Oct 14.
Article in English | MEDLINE | ID: mdl-27582494

ABSTRACT

Tissue inhibitor of metalloproteinases-3 (TIMP-3) is a central inhibitor of matrix-degrading and sheddase families of metalloproteinases. Extracellular levels of the inhibitor are regulated by the balance between its retention on the extracellular matrix and its endocytic clearance by the scavenger receptor low density lipoprotein receptor-related protein 1 (LRP1). Here, we used molecular modeling to predict TIMP-3 residues potentially involved in binding to LRP1 based on the proposed LRP1 binding motif of 2 lysine residues separated by about 21 Å and mutated the candidate lysine residues to alanine individually and in pairs. Of the 22 mutants generated, 13 displayed a reduced rate of uptake by HTB94 chondrosarcoma cells. The two mutants (TIMP-3 K26A/K45A and K42A/K110A) with lowest rates of uptake were further evaluated and found to display reduced binding to LRP1 and unaltered inhibitory activity against prototypic metalloproteinases. TIMP-3 K26A/K45A retained higher affinity for sulfated glycosaminoglycans than K42A/K110A and exhibited increased affinity for ADAMTS-5 in the presence of heparin. Both mutants inhibited metalloproteinase-mediated degradation of cartilage at lower concentrations and for longer than wild-type TIMP-3, indicating that their increased half-lives improved their ability to protect cartilage. These mutants may be useful in treating connective tissue diseases associated with increased metalloproteinase activity.


Subject(s)
Bone Neoplasms/metabolism , Chondrosarcoma/metabolism , Endocytosis , Extracellular Matrix/metabolism , Neoplasm Proteins/metabolism , Tissue Inhibitor of Metalloproteinase-3/metabolism , ADAMTS5 Protein/genetics , ADAMTS5 Protein/metabolism , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Cartilage/metabolism , Cartilage/pathology , Cell Line, Tumor , Chondrosarcoma/genetics , Chondrosarcoma/pathology , Extracellular Matrix/genetics , Extracellular Matrix/pathology , Heparin/metabolism , Humans , Low Density Lipoprotein Receptor-Related Protein-1/genetics , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Neoplasm Proteins/genetics , Tissue Inhibitor of Metalloproteinase-3/genetics
4.
Matrix Biol ; 56: 57-73, 2016 12.
Article in English | MEDLINE | ID: mdl-27084377

ABSTRACT

Matrix metalloproteinase 13 (MMP-13) degrades collagenous extracellular matrix and its aberrant activity associates with diseases such as arthritis, cancer, atherosclerosis and fibrosis. The wide range of MMP-13 proteolytic capacity suggests that it is a powerful, potentially destructive proteinase and thus it has been believed that MMP-13 is not produced in most adult human tissues in the steady state. Present study has revealed that human chondrocytes isolated from healthy adults constitutively express and secrete MMP-13, but that it is rapidly endocytosed and degraded by chondrocytes. Both pro- and activated MMP-13 bind to clusters II and III of low-density lipoprotein (LDL) receptor-related protein 1 (LRP1). Domain deletion studies indicated that the hemopexin domain is responsible for this interaction. Binding competition between MMP-13 and ADAMTS-4, -5 or TIMP-3, which also bind to cluster II, further shown that the MMP-13 binding site within cluster II is different from those of ADAMTS-4, -5 or TIMP-3. MMP-13 is therefore co-endocytosed with ADAMTS-5 and TIMP-3 by human chondrocytes. These findings indicate that MMP-13 may play a role on physiological turnover of cartilage extracellular matrix and that LRP1 is a key modulator of extracellular levels of MMP-13 and its internalization is independent of the levels of ADAMTS-4, -5 and TIMP-3.


Subject(s)
ADAMTS5 Protein/metabolism , Chondrocytes/enzymology , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Matrix Metalloproteinase 13/metabolism , Tissue Inhibitor of Metalloproteinase-3/metabolism , ADAMTS5 Protein/chemistry , Binding, Competitive , Endocytosis , HEK293 Cells , Humans , Low Density Lipoprotein Receptor-Related Protein-1/chemistry , Matrix Metalloproteinase 13/chemistry , Protein Binding , Protein Interaction Domains and Motifs , Protein Transport , Tissue Inhibitor of Metalloproteinase-3/chemistry
5.
J Biol Chem ; 289(35): 24091-101, 2014 Aug 29.
Article in English | MEDLINE | ID: mdl-25008319

ABSTRACT

Remodeling of collagen by matrix metalloproteinases (MMPs) is crucial to tissue homeostasis and repair. MMP-13 is a collagenase with a substrate preference for collagen II over collagens I and III. It recognizes a specific, well-known site in the tropocollagen molecule where its binding locally perturbs the triple helix, allowing the catalytic domain of the active enzyme to cleave the collagen α chains sequentially, at Gly(775)-Leu(776) in collagen II. However, the specific residues upon which collagen recognition depends within and surrounding this locus have not been systematically mapped. Using our triple-helical peptide Collagen Toolkit libraries in solid-phase binding assays, we found that MMP-13 shows little affinity for Collagen Toolkit III, but binds selectively to two triple-helical peptides of Toolkit II. We have identified the residues required for the adhesion of both proMMP-13 and MMP-13 to one of these, Toolkit peptide II-44, which contains the canonical collagenase cleavage site. MMP-13 was unable to bind to a linear peptide of the same sequence as II-44. We also discovered a second binding site near the N terminus of collagen II (starting at helix residue 127) in Toolkit peptide II-8. The pattern of binding of the free hemopexin domain of MMP-13 was similar to that of the full-length enzyme, but the free catalytic subunit bound none of our peptides. The susceptibility of Toolkit peptides to proteolysis in solution was independent of the very specific recognition of immobilized peptides by MMP-13; the enzyme proved able to cleave a range of dissolved collagen peptides.


Subject(s)
Collagen Type II/metabolism , Matrix Metalloproteinase 13/metabolism , Peptides/metabolism , Amino Acid Sequence , Base Sequence , Collagen Type II/chemistry , DNA Primers , Matrix Metalloproteinase 13/chemistry , Molecular Sequence Data , Proteolysis , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Substrate Specificity
6.
FASEB J ; 27(11): 4395-405, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23913860

ABSTRACT

Matrix metalloproteinase (MMP)-13 is one of the mammalian collagenases that play key roles in tissue remodelling and repair and in progression of diseases such as cancer, arthritis, atherosclerosis, and aneurysm. For collagenase to cleave triple helical collagens, the triple helical structure has to be locally unwound before hydrolysis, but this process is not well understood. We report crystal structures of catalytically inactive full-length human MMP-13(E223A) in complex with peptides of 14-26 aa derived from the cleaved prodomain during activation. Peptides are bound to the active site of the enzyme by forming an extended ß-strand with Glu(40) or Tyr(46) inserted into the S1' specificity pocket. The structure of the N-terminal part of the peptides is variable and interacts with different parts of the catalytic domain. Those areas are designated substrate-dependent exosites, in that they accommodate different peptide structures, whereas the precise positioning of the substrate backbone is maintained in the active site. These modes of peptide-MMP-13 interactions have led us to propose how triple helical collagen strands fit into the active site cleft of the collagenase.


Subject(s)
Catalytic Domain , Collagen/chemistry , Matrix Metalloproteinase 13/chemistry , Molecular Docking Simulation , Peptides/chemistry , Amino Acid Motifs , Amino Acid Sequence , Collagen/metabolism , Crystallography, X-Ray , Glutamic Acid/chemistry , Humans , Matrix Metalloproteinase 13/metabolism , Molecular Sequence Data , Mutation, Missense , Peptides/metabolism , Protein Binding , Tyrosine/chemistry
7.
Proc Natl Acad Sci U S A ; 109(31): 12461-6, 2012 Jul 31.
Article in English | MEDLINE | ID: mdl-22761315

ABSTRACT

Collagenases of the matrix metalloproteinase (MMP) family play major roles in morphogenesis, tissue repair, and human diseases, but how they recognize and cleave the collagen triple helix is not fully understood. Here, we report temperature-dependent binding of a catalytically inactive MMP-1 mutant (E200A) to collagen through the cooperative action of its catalytic and hemopexin domains. Contact between the two molecules was mapped by screening the Collagen Toolkit peptide library and by hydrogen/deuterium exchange. The crystal structure of MMP-1(E200A) bound to a triple-helical collagen peptide revealed extensive interactions of the 115-Å-long triple helix with both MMP-1 domains. An exosite in the hemopexin domain, which binds the leucine 10 residues C-terminal to the scissile bond, is critical for collagenolysis and represents a unique target for inhibitor development. The scissile bond is not correctly positioned for hydrolysis in the crystallized complex. A productive binding mode is readily modeled, without altering the MMP-1 structure or the exosite interactions, by axial rotation of the collagen homotrimer. Interdomain flexing of the enzyme and a localized excursion of the collagen chain closest to the active site, facilitated by thermal loosening of the substrate, may lead to the first transition state of collagenolysis.


Subject(s)
Collagen/chemistry , Matrix Metalloproteinase 1/chemistry , Models, Molecular , Proteolysis , Amino Acid Substitution , Collagen/metabolism , Humans , Matrix Metalloproteinase 1/genetics , Matrix Metalloproteinase 1/metabolism , Mutation, Missense , Peptide Library , Protein Structure, Quaternary , Protein Structure, Tertiary
8.
J Biol Chem ; 287(27): 22988-97, 2012 Jun 29.
Article in English | MEDLINE | ID: mdl-22573319

ABSTRACT

Degradation of fibrillar collagens is important in many physiological and pathological events. These collagens are resistant to most proteases due to the tightly packed triple-helical structure, but are readily cleaved at a specific site by collagenases, selected members of the matrix metalloproteinases (MMPs). To investigate the structural requirements for collagenolysis, varying numbers of GXY triplets from human type III collagen around the collagenase cleavage site were inserted between two triple helix domains of the Scl2 bacterial collagen protein. The original bacterial CL domain was not cleaved by MMP-1 (collagenase 1) or MMP-13 (collagenase 3). The minimum type III sequence necessary for cleavage by the two collagenases was 5 GXY triplets, including 4 residues before and 11 residues after the cleavage site (P4-P11'). Cleavage of these chimeric substrates was not achieved by the catalytic domain of MMP-1 or MMP-13, nor by full-length MMP-3. Kinetic analysis of the chimeras indicated that the rate of cleavage by MMP-1 of the chimera containing six triplets (P7-P11') of collagen III was similar to that of native collagen III. The collagenase-susceptible chimeras were cleaved very slowly by trypsin, a property also seen for native collagen III, supporting a local structural relaxation of the triple helix near the collagenase cleavage site. The recombinant bacterial-human collagen system characterized here is a good model to investigate the specificity and mechanism of action of collagenases.


Subject(s)
Collagen Type III/genetics , Collagen Type III/metabolism , Matrix Metalloproteinase 13/genetics , Matrix Metalloproteinase 13/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Amino Acid Sequence , Catalytic Domain/physiology , Collagen Type III/chemistry , Collagenases/genetics , Collagenases/metabolism , Escherichia coli/enzymology , Escherichia coli/genetics , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , HEK293 Cells , Humans , Matrix Metalloproteinase 13/chemistry , Molecular Sequence Data , Protein Structure, Tertiary , Substrate Specificity , Trypsin/metabolism
9.
Eur J Med Chem ; 47(1): 143-52, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22088955

ABSTRACT

Osteoarthritis (OA) is the leading cause of joint pain and disability in middle-aged and elderly patients, and is characterized by progressive loss of articular cartilage. Among the various matrix metalloproteinases (MMPs), MMP-13 is specifically expressed in the cartilage of human OA patients and is not present in normal adult cartilage. Thus, MMP-13-selective inhibitors are promising candidates in osteoarthritis therapy. Recently, we designed an N-isopropoxy-arylsulfonamide-based hydroxamate inhibitor, which showed low nanomolar activity and high selectivity for MMP-13. In parallel to further studies aiming to assess the in vivo activity of our compound, we screened the Life Chemicals database through computational docking to seek for novel scaffolds as zinc-chelating non-hydroxamate inhibitors. Experimental evaluation of 20 selected candidate compounds verified five novel leads with IC(50) in the low µM range. These newly discovered inhibitors are structurally unrelated to the ones known so far and provide useful scaffolds to develop compounds with more desirable properties. Finally, a first round of structure-based optimization on lead 1 was accomplished and led to an increase in potency of more than 5 fold.


Subject(s)
Drug Design , Matrix Metalloproteinase Inhibitors , Protease Inhibitors/chemistry , Protease Inhibitors/pharmacology , Catalytic Domain , Crystallography, X-Ray , Drug Evaluation, Preclinical , Humans , Inhibitory Concentration 50 , Matrix Metalloproteinase 13/chemistry , User-Computer Interface
10.
J Biol Chem ; 286(9): 7587-600, 2011 Mar 04.
Article in English | MEDLINE | ID: mdl-21193411

ABSTRACT

Homodimerization is an essential step for membrane type 1 matrix metalloproteinase (MT1-MMP) to activate proMMP-2 and to degrade collagen on the cell surface. To uncover the molecular basis of the hemopexin (Hpx) domain-driven dimerization of MT1-MMP, a crystal structure of the Hpx domain was solved at 1.7 Å resolution. Two interactions were identified as potential biological dimer interfaces in the crystal structure, and mutagenesis studies revealed that the biological dimer possesses a symmetrical interaction where blades II and III of molecule A interact with blades III and II of molecule B. The mutations of amino acids involved in the interaction weakened the dimer interaction of Hpx domains in solution, and incorporation of these mutations into the full-length enzyme significantly inhibited dimer-dependent functions on the cell surface, including proMMP-2 activation, collagen degradation, and invasion into the three-dimensional collagen matrix, whereas dimer-independent functions, including gelatin film degradation and two-dimensional cell migration, were not affected. These results shed light on the structural basis of MT1-MMP dimerization that is crucial to promote cellular invasion.


Subject(s)
Extracellular Matrix/enzymology , Hemopexin/chemistry , Hemopexin/metabolism , Matrix Metalloproteinase 14/chemistry , Matrix Metalloproteinase 14/metabolism , Animals , COS Cells , Chlorocebus aethiops , Crystallography , Dimerization , Enzyme Activation/physiology , HeLa Cells , Hemopexin/genetics , Humans , Matrix Metalloproteinase 14/genetics , Membrane Proteins/chemistry , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mutagenesis , Protein Structure, Tertiary , Solubility , Structure-Activity Relationship
11.
Biochem J ; 431(1): 113-22, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20645923

ABSTRACT

We have reported previously that reactive-site mutants of N-TIMP-3 [N-terminal inhibitory domain of TIMP-3 (tissue inhibitor of metalloproteinases 3)] modified at the N-terminus, selectively inhibited ADAM17 (a disintegrin and metalloproteinase 17) over the MMPs (matrix metalloproteinases). The primary aggrecanases ADAMTS (ADAM with thrombospondin motifs) -4 and -5 are ADAM17-related metalloproteinases which are similarly inhibited by TIMP-3, but are poorly inhibited by other TIMPs. Using a newly developed recombinant protein substrate based on the IGD (interglobular domain) of aggrecan, gst-IGD-flag, these reactive-site mutants were found to similarly inhibit ADAMTS-4 and ADAMTS-5. Further mutations of N-TIMP-3 indicated that up to two extra alanine residues can be attached to the N-terminus before the Ki (app) for ADAMTS-4 and ADAMTS-5 increased to over 100 nM. No other residues tested at the [-1] position produced inhibitors as potent as the alanine mutant. The mutants N-TIMP-3(T2G), [-1A]N-TIMP-3 and [-2A]N-TIMP-3 were effective inhibitors of aggrecan degradation, but not of collagen degradation in both IL-1α (interleukin-1α)-stimulated porcine articular cartilage explants and IL-1α with oncostatin M-stimulated human cartilage explants. Molecular modelling studies indicated that the [-1A]N-TIMP-3 mutant has additional stabilizing interactions with the catalytic domains of ADAM17, ADAMTS-4 and ADAMTS-5 that are absent from complexes with MMPs. These observations suggest that further mutation of the residues of N-TIMP-3 which make unique contacts with these metalloproteinases may allow discrimination between them.


Subject(s)
ADAM Proteins/antagonists & inhibitors , Procollagen N-Endopeptidase/antagonists & inhibitors , Procollagen N-Endopeptidase/chemistry , Tissue Inhibitor of Metalloproteinase-3/chemistry , Tissue Inhibitor of Metalloproteinase-3/genetics , ADAM Proteins/chemistry , ADAM Proteins/genetics , ADAMTS4 Protein , Aggrecans/metabolism , Animals , Cartilage, Articular/metabolism , Catalytic Domain , Cells, Cultured , Humans , Mutation , Procollagen N-Endopeptidase/genetics , Swine , Tissue Inhibitor of Metalloproteinase-3/metabolism
12.
J Biol Chem ; 285(29): 22276-81, 2010 Jul 16.
Article in English | MEDLINE | ID: mdl-20463013

ABSTRACT

Type I collagen cleavage is crucial for tissue remodeling, but its homotrimeric isoform is resistant to all collagenases. The homotrimers occur in fetal tissues, fibrosis, and cancer, where their collagenase resistance may play an important physiological role. To understand the mechanism of this resistance, we studied interactions of alpha1(I)(3) homotrimers and normal alpha1(I)(2)alpha2(I) heterotrimers with fibroblast collagenase (MMP-1). Similar MMP-1 binding to the two isoforms and similar cleavage efficiency of unwound alpha1(I) and alpha2(I) chains suggested increased stability and less efficient unwinding of the homotrimer triple helix at the collagenase cleavage site. The unwinding, necessary for placing individual chains inside the catalytic cleft of the enzyme, was the rate-limiting cleavage step for both collagen isoforms. Comparative analysis of the homo- and heterotrimer cleavage kinetics revealed that MMP-1 binding promotes stochastic helix unwinding, resolving the controversy between different models of collagenase action.


Subject(s)
Collagen Type I/chemistry , Collagen Type I/metabolism , Matrix Metalloproteinase 1/metabolism , Protein Multimerization , Animals , Fibrillar Collagens/metabolism , Humans , Kinetics , Mice , Microscopy, Confocal , Models, Biological , Protein Binding , Protein Structure, Tertiary , Recombinant Proteins/metabolism , Temperature
13.
Cancer Res ; 70(11): 4366-74, 2010 Jun 01.
Article in English | MEDLINE | ID: mdl-20460529

ABSTRACT

Collagen fibers affect metastasis in two opposing ways, by supporting invasive cells but also by generating a barrier to invasion. We hypothesized that these functions might be performed by different isoforms of type I collagen. Carcinomas are reported to contain alpha1(I)(3) homotrimers, a type I collagen isoform normally not present in healthy tissues, but the role of the homotrimers in cancer pathophysiology is unclear. In this study, we found that these homotrimers were resistant to all collagenolytic matrix metalloproteinases (MMP). MMPs are massively produced and used by cancer cells and cancer-associated fibroblasts for degrading stromal collagen at the leading edge of tumor invasion. The MMP-resistant homotrimers were produced by all invasive cancer cell lines tested, both in culture and in tumor xenografts, but they were not produced by cancer-associated fibroblasts, thereby comprising a specialized fraction of tumor collagen. We observed the homotrimer fibers to be resistant to pericellular degradation, even upon stimulation of the cells with proinflammatory cytokines. Furthermore, we confirmed an enhanced proliferation and migration of invasive cancer cells on the surface of homotrimeric versus normal (heterotrimeric) type I collagen fibers. In summary, our findings suggest that invasive cancer cells may use homotrimers for building MMP-resistant invasion paths, supporting local proliferation and directed migration of the cells whereas surrounding normal stromal collagens are cleaved. Because the homotrimers are universally secreted by cancer cells and deposited as insoluble, MMP-resistant fibers, they offer an appealing target for cancer diagnostics and therapy.


Subject(s)
Collagen Type I/metabolism , Fibrosarcoma/metabolism , Fibrosarcoma/pathology , Matrix Metalloproteinases/metabolism , Animals , Cell Growth Processes/physiology , Cell Movement/physiology , Fibroblasts/metabolism , Fibrosarcoma/enzymology , Humans , Isoenzymes , Mice , Mice, Nude , Neoplasm Invasiveness
14.
J Med Chem ; 52(15): 4757-73, 2009 Aug 13.
Article in English | MEDLINE | ID: mdl-19606871

ABSTRACT

Matrix metalloproteinase-13 (MMP-13) is a key enzyme implicated in the degradation of the extracellular matrix in osteoarthritis (OA). For this reason, MMP-13 synthetic inhibitors are being sought as potential therapeutic agents to prevent cartilage degradation and to halt the progression of OA. Herein, we report the synthesis and in vitro evaluation of a new series of selective MMP-13 inhibitors possessing an arylsulfonamidic scaffold. Among these potential inhibitors, a very promising compound was discovered exhibiting nanomolar activity for MMP-13 and was highly selective for this enzyme compared to MMP-1, -14, and TACE. This compound acted as a slow-binding inhibitor of MMP-13 and was demonstrated to be effective in an in vitro collagen assay and in a model of cartilage degradation. Furthermore, a docking study was conducted for this compound in order to investigate its binding interactions with MMP-13 and the reasons for its selectivity toward MMP-13 versus other MMPs.


Subject(s)
Drug Design , Hydroxamic Acids/chemical synthesis , Matrix Metalloproteinase Inhibitors , Osteoarthritis/drug therapy , Protease Inhibitors/chemical synthesis , ADAM Proteins/chemistry , ADAM17 Protein , Cartilage/metabolism , Hydroxamic Acids/pharmacology , Hydroxamic Acids/therapeutic use , Matrix Metalloproteinase 1/chemistry , Matrix Metalloproteinase 13/chemistry , Matrix Metalloproteinase 14/chemistry , Models, Molecular , Protease Inhibitors/pharmacology , Protease Inhibitors/therapeutic use , Structure-Activity Relationship
15.
Biochemistry ; 47(2): 537-47, 2008 Jan 15.
Article in English | MEDLINE | ID: mdl-18081311

ABSTRACT

Human ADAM12 (a disintegrin and metalloproteinase) is a multidomain zinc metalloproteinase expressed at high levels during development and in human tumors. ADAM12 exists as two splice variants: a classical type 1 membrane-anchored form (ADAM12-L) and a secreted splice variant (ADAM12-S) consisting of pro, catalytic, disintegrin, cysteine-rich, and EGF domains. Here we present a novel activity of recombinant ADAM12-S and its domain deletion mutants on S-carboxymethylated transferrin (Cm-Tf). Cleavage of Cm-Tf occurred at multiple sites, and N-terminal sequencing showed that the enzyme exhibits restricted specificity but a consensus sequence could not be defined as its subsite requirements are promiscuous. Kinetic analysis revealed that the noncatalytic C-terminal domains are important regulators of Cm-Tf activity and that ADAM12-PC consisting of the pro domain and catalytic domain is the most active on this substrate. It was also observed that NaCl inhibits ADAM12. Among the tissue inhibitors of metalloproteinases (TIMP) examined, the N-terminal domain of TIMP-3 (N-TIMP-3) inhibits ADAM12-S and ADAM12-PC with low nanomolar Ki(app) values while TIMP-2 inhibits them with a slightly lower affinity (9-44 nM). However, TIMP-1 is a much weaker inhibitor. N-TIMP-3 variants that lack MMP inhibitory activity but retained the ability to inhibit ADAM17/TACE failed to inhibit ADAM12. These results indicate unique enzymatic properties of ADAM12 among the members of the ADAM family of metalloproteinases.


Subject(s)
ADAM Proteins/chemistry , ADAM Proteins/metabolism , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Mutant Proteins/metabolism , Sequence Deletion , ADAM Proteins/antagonists & inhibitors , ADAM Proteins/isolation & purification , ADAM12 Protein , Amino Acid Sequence , Animals , Calcium/pharmacology , Catalysis , Electrophoresis, Polyacrylamide Gel , Guinea Pigs , Humans , Hydrogen-Ion Concentration , Kinetics , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/isolation & purification , Metals/pharmacology , Molecular Sequence Data , Mutant Proteins/antagonists & inhibitors , Mutant Proteins/chemistry , Mutant Proteins/isolation & purification , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/chemistry , Protein Isoforms/isolation & purification , Protein Isoforms/metabolism , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Sequence Analysis, Protein , Sodium Chloride/pharmacology , Substrate Specificity/drug effects , Tissue Inhibitor of Metalloproteinases/metabolism , Transferrin/metabolism
16.
J Biol Chem ; 281(50): 38302-13, 2006 Dec 15.
Article in English | MEDLINE | ID: mdl-17065155

ABSTRACT

The hydrolysis of collagen (collagenolysis) is one of the committed steps in extracellular matrix turnover. Within the matrix metalloproteinase (MMP) family distinct preferences for collagen types are seen. The substrate determinants that may guide these specificities are unknown. In this study, we have utilized 12 triple-helical substrates in combination with 10 MMPs to better define the contributions of substrate sequence and thermal stability toward triple helicase activity and collagen specificity. In general, MMP-13 was found to be distinct from MMP-8 and MT1-MMP(Delta279-523), in that enhanced substrate thermal stability has only a modest effect on activity, regardless of sequence. This result correlates to the unique collagen specificity of MMP-13 compared with MMP-8 and MT1-MMP, in that MMP-13 hydrolyzes type II collagen efficiently, whereas MMP-8 and MT1-MMP are similar in their preference for type I collagen. In turn, MMP-1 was the least efficient of the collagenolytic MMPs at processing increasingly thermal stable triple helices and thus favors type III collagen, which has a relatively flexible cleavage site. Gelatinases (MMP-2 and MMP-9(Delta444-707)) appear incapable of processing more stable helices and are thus mechanistically distinct from collagenolytic MMPs. The collagen specificity of MMPs appears to be based on a combination of substrate sequence and thermal stability. Analysis of the hydrolysis of triple-helical peptides by an MMP mutant indicated that Tyr(210) functions in triple helix binding and hydrolysis, but not in processing triple helices of increasing thermal stabilities. Further exploration of MMP active sites and exosites, in combination with substrate conformation, may prove valuable for additional dissection of collagenolysis and yield information useful in the design of more selective MMP inhibitors.


Subject(s)
Collagen/metabolism , Matrix Metalloproteinases/metabolism , Peptide Hydrolases/metabolism , Amino Acid Sequence , Chromatography, High Pressure Liquid , Circular Dichroism , Enzyme Stability , Hydrolysis , Kinetics , Molecular Sequence Data , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Substrate Specificity
17.
J Mol Biol ; 362(1): 78-88, 2006 Sep 08.
Article in English | MEDLINE | ID: mdl-16890240

ABSTRACT

The extracellular matrix is a dynamic environment that constantly undergoes remodelling and degradation during vital physiological processes such as angiogenesis, wound healing, and development. Unbalanced extracellular matrix breakdown is associated with many diseases such as arthritis, cancer and fibrosis. Interstitial collagen is degraded by matrix metalloproteinases with collagenolytic activity by MMP-1, MMP-8 and MMP-13, collectively known as the collagenases. Matrix metalloproteinase 1 (MMP-1) plays a pivotal role in degradation of interstitial collagen types I, II, and III. Here, we report the crystal structure of the active form of human MMP-1 at 2.67 A resolution. This is the first MMP-1 structure that is free of inhibitor and a water molecule essential for peptide hydrolysis is observed coordinated with the active site zinc. Comparing this structure with the human proMMP-1 shows significant structural differences, mainly in the relative orientation of the hemopexin domain, between the pro form and active form of the human enzyme.


Subject(s)
Matrix Metalloproteinase 1/chemistry , Protein Structure, Tertiary , Animals , Catalytic Domain , Collagenases/chemistry , Collagenases/genetics , Crystallography, X-Ray , Enzyme Precursors/chemistry , Enzyme Precursors/genetics , Hemopexin/chemistry , Hemopexin/genetics , Humans , Hydrogen Bonding , Matrix Metalloproteinase 1/genetics , Matrix Metalloproteinase 1/metabolism , Models, Molecular , Molecular Sequence Data , Swine
18.
Cardiovasc Res ; 69(3): 562-73, 2006 Feb 15.
Article in English | MEDLINE | ID: mdl-16405877

ABSTRACT

Matrix metalloproteinases (MMPs), also called matrixins, function in the extracellular environment of cells and degrade both matrix and non-matrix proteins. They play central roles in morphogenesis, wound healing, tissue repair and remodelling in response to injury, e.g. after myocardial infarction, and in progression of diseases such as atheroma, arthritis, cancer and chronic tissue ulcers. They are multi-domain proteins and their activities are regulated by tissue inhibitors of metalloproteinases (TIMPs). This review introduces the members of the MMP family and discusses their domain structure and function, proenyme activation, the mechanism of inhibition by TIMPs and their significance in physiology and pathology.


Subject(s)
Cardiovascular System/enzymology , Extracellular Matrix/enzymology , Matrix Metalloproteinases/chemistry , Tissue Inhibitor of Metalloproteinases/chemistry , Animals , Cardiovascular Diseases/enzymology , Humans , Matrix Metalloproteinase Inhibitors , Structure-Activity Relationship , Tissue Inhibitor of Metalloproteinases/metabolism
19.
J Biol Chem ; 280(10): 9578-85, 2005 Mar 11.
Article in English | MEDLINE | ID: mdl-15611040

ABSTRACT

Vertebrate collagenases, members of the matrix metalloproteinase (MMP) family, initiate interstitial fibrillar collagen breakdown. It is essential in many biological processes, and unbalanced collagenolysis is associated with diseases such as arthritis, cancer, atherosclerosis, aneurysm, and fibrosis. These metalloproteinases are secreted from the cell as inactive precursors, procollagenases (proMMPs). To gain insights into the structural basis of their activation mechanisms and collagen binding, we have crystallized recombinant human proMMP-1 and determined its structure to 2.2 A resolution. The catalytic metalloproteinase domain and the C-terminal hemopexin (Hpx) domain show the classical MMP-fold, but the structure has revealed new features in surface loops and domain interaction. The prodomain is formed by a three-helix bundle and gives insight into the stepwise activation mechanism of proMMP-1. The prodomain interacts with the Hpx domain, which affects the position of the Hpx domain relative to the catalytic domain. This interaction results in a "closed" configuration of proMMP-1 in contrast to the "open" configuration observed previously for the structure of active MMP-1. This is the first evidence of mobility of the Hpx domain in relation to the catalytic domain, providing an important clue toward the understanding of the collagenase-collagen interaction and subsequent collagenolysis.


Subject(s)
Collagen/metabolism , Collagenases/chemistry , Collagenases/metabolism , Enzyme Precursors/chemistry , Enzyme Precursors/metabolism , Amino Acid Sequence , Binding Sites , Crystallography, X-Ray , Humans , Hydrogen Bonding , Matrix Metalloproteinase 1 , Models, Molecular , Molecular Sequence Data , Protein Structure, Secondary
20.
EMBO J ; 23(15): 3020-30, 2004 Aug 04.
Article in English | MEDLINE | ID: mdl-15257288

ABSTRACT

Breakdown of triple-helical interstitial collagens is essential in embryonic development, organ morphogenesis and tissue remodelling and repair. Aberrant collagenolysis may result in diseases such as arthritis, cancer, atherosclerosis, aneurysm and fibrosis. In vertebrates, it is initiated by collagenases belonging to the matrix metalloproteinase (MMP) family. The three-dimensional structure of a prototypic collagenase, MMP-1, indicates that the substrate-binding site of the enzyme is too narrow to accommodate triple-helical collagen. Here we report that collagenases bind and locally unwind the triple-helical structure before hydrolyzing the peptide bonds. Mutation of the catalytically essential residue Glu200 of MMP-1 to Ala resulted in a catalytically inactive enzyme, but in its presence noncollagenolytic proteinases digested collagen into typical 3/4 and 1/4 fragments, indicating that the MMP-1(E200A) mutant unwinds the triple-helical collagen. The study also shows that MMP-1 preferentially interacts with the alpha2(I) chain of type I collagen and cleaves the three alpha chains in succession. Our results throw light on the basic mechanisms that control a wide range of biological and pathological processes associated with tissue remodelling.


Subject(s)
Collagen/chemistry , Collagen/metabolism , Matrix Metalloproteinase 1/chemistry , Matrix Metalloproteinase 1/metabolism , Amino Acid Sequence , Binding Sites , Catalytic Domain , Glutamic Acid/genetics , Glutamic Acid/metabolism , Hydrolysis , Matrix Metalloproteinase 1/genetics , Matrix Metalloproteinase 3/chemistry , Matrix Metalloproteinase 3/genetics , Matrix Metalloproteinase 3/metabolism , Models, Molecular , Molecular Sequence Data , Mutation/genetics , Protein Structure, Tertiary , Temperature
SELECTION OF CITATIONS
SEARCH DETAIL
...