51
|
Kumar S, Rao N, Ge R. Emerging Roles of ADAMTSs in Angiogenesis and Cancer. Cancers (Basel) 2012; 4:1252-99. [PMID: 24213506 PMCID: PMC3712723 DOI: 10.3390/cancers4041252] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2012] [Revised: 11/21/2012] [Accepted: 11/23/2012] [Indexed: 12/18/2022] Open
Abstract
A Disintegrin-like And Metalloproteinase with ThromboSpondin motifs—ADAMTSs—are a multi-domain, secreted, extracellular zinc metalloproteinase family with 19 members in humans. These extracellular metalloproteinases are known to cleave a wide range of substrates in the extracellular matrix. They have been implicated in various physiological processes, such as extracellular matrix turnover, melanoblast development, interdigital web regression, blood coagulation, ovulation, etc. ADAMTSs are also critical in pathological processes such as arthritis, atherosclerosis, cancer, angiogenesis, wound healing, etc. In the past few years, there has been an explosion of reports concerning the role of ADAMTS family members in angiogenesis and cancer. To date, 10 out of the 19 members have been demonstrated to be involved in regulating angiogenesis and/or cancer. The mechanism involved in their regulation of angiogenesis or cancer differs among different members. Both angiogenesis-dependent and -independent regulation of cancer have been reported. This review summarizes our current understanding on the roles of ADAMTS in angiogenesis and cancer and highlights their implications in cancer therapeutic development.
Collapse
Affiliation(s)
- Saran Kumar
- Department of Biological Sciences, National University of Singapore, Singapore 117543, Singapore.
| | | | | |
Collapse
|
52
|
Bourhis JM, Mariano N, Zhao Y, Walter TS, El Omari K, Delolme F, Moali C, Hulmes DJS, Aghajari N. Production and crystallization of the C-propeptide trimer from human procollagen III. Acta Crystallogr Sect F Struct Biol Cryst Commun 2012; 68:1209-13. [PMID: 23027749 PMCID: PMC3497981 DOI: 10.1107/s1744309112035294] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2012] [Accepted: 08/09/2012] [Indexed: 12/27/2022]
Abstract
The C-propeptide domains of the fibrillar procollagens, which are present throughout the Metazoa in the form of ∼90 kDa trimers, play crucial roles in both intracellular molecular assembly and extracellular formation of collagen fibrils. The first crystallization of a C-propeptide domain, that from human procollagen III, is described. Following transient expression in mammalian 293T cells of both the native protein and a selenomethionine derivative, two crystal forms of the homotrimer were obtained: an orthorhombic form (P2(1)2(1)2(1)) that diffracted to 1.7 Å resolution and a trigonal form (P321) that diffracted to 3.5 Å resolution. Characterization by MALDI-TOF mass spectrometry allowed the efficiency of selenomethionine incorporation to be determined.
Collapse
Affiliation(s)
- J.-M. Bourhis
- FRE 3310, Institut de Biologie et Chimie des Protéines, CNRS/Université Lyon 1, 69367 Lyon CEDEX 7, France
- Biologie Structurale des Interactions entre Virus et Cellule-Hôte, Université Joseph Fourier/European Molecular Biology Laboratory/CNRS UMI3265, 38042 Grenoble CEDEX 9, France
| | - N. Mariano
- FRE 3310, Institut de Biologie et Chimie des Protéines, CNRS/Université Lyon 1, 69367 Lyon CEDEX 7, France
| | - Y. Zhao
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, England
| | - T. S. Walter
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, England
| | - K. El Omari
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, England
| | - F. Delolme
- FR3302, Institut de Biologie et Chimie des Protéines, CNRS/Université Lyon 1, 69367 Lyon CEDEX 7, France
| | - C. Moali
- FRE 3310, Institut de Biologie et Chimie des Protéines, CNRS/Université Lyon 1, 69367 Lyon CEDEX 7, France
| | - D. J. S. Hulmes
- FRE 3310, Institut de Biologie et Chimie des Protéines, CNRS/Université Lyon 1, 69367 Lyon CEDEX 7, France
| | - N. Aghajari
- UMR5086, Institut de Biologie et Chimie des Protéines, CNRS/Université Lyon 1, 69367 Lyon CEDEX 7, France
| |
Collapse
|
53
|
Paulissen G, El Hour M, Rocks N, Guéders MM, Bureau F, Foidart JM, Lopez-Otin C, Noel A, Cataldo DD. Control of allergen-induced inflammation and hyperresponsiveness by the metalloproteinase ADAMTS-12. THE JOURNAL OF IMMUNOLOGY 2012; 189:4135-43. [PMID: 22962682 DOI: 10.4049/jimmunol.1103739] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
A disintegrin and metalloproteinase with thrombospondin motifs (ADAMTS) constitute a family of endopeptidases related to matrix metalloproteinases. These proteinases have been largely implicated in tissue remodeling associated with pathological processes. Among them, ADAMTS12 was identified as an asthma-associated gene in a human genome screening program. However, its functional implication in asthma is not yet documented. The present study aims at investigating potential ADAMTS-12 functions in experimental models of allergic airways disease. Two different in vivo protocols of allergen-induced airways disease were applied to the recently generated Adamts12-deficient mice and corresponding wild-type mice. In this study, we provide evidence for a protective effect of ADAMTS-12 against bronchial inflammation and hyperresponsiveness. In the absence of Adamts12, challenge with different allergens (OVA and house dust mite) led to exacerbated eosinophilic inflammation in the bronchoalveolar lavage fluid and in lung tissue, along with airway dysfunction assessed by increased airway responsiveness following methacholine exposure. Furthermore, mast cell counts and ST2 receptor and IL-33 levels were higher in the lungs of allergen-challenged Adamts12-deficient mice. The present study provides, to our knowledge, the first experimental evidence for a contribution of ADAMTS-12 as a key mediator in airways disease, interfering with immunological processes leading to inflammation and airway hyperresponsiveness.
Collapse
Affiliation(s)
- Geneviève Paulissen
- Laboratory of Tumor and Developmental Biology, Interdisciplinary Group of Applied Genoproteomics-Cancer (GIGA-Cancer), University of Liège and University Hospital of Liège, 4000 Liège, Belgium
| | | | | | | | | | | | | | | | | |
Collapse
|
54
|
Noël A, Gutiérrez-Fernández A, Sounni NE, Behrendt N, Maquoi E, Lund IK, Cal S, Hoyer-Hansen G, López-Otín C. New and paradoxical roles of matrix metalloproteinases in the tumor microenvironment. Front Pharmacol 2012; 3:140. [PMID: 22822400 PMCID: PMC3398411 DOI: 10.3389/fphar.2012.00140] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2012] [Accepted: 06/27/2012] [Indexed: 12/12/2022] Open
Abstract
Processes such as cell proliferation, angiogenesis, apoptosis, or invasion are strongly influenced by the surrounding microenvironment of the tumor. Therefore, the ability to change these surroundings represents an important property through which tumor cells are able to acquire specific functions necessary for tumor growth and dissemination. Matrix metalloproteinases (MMPs) constitute key players in this process, allowing tumor cells to modify the extracellular matrix (ECM) and release cytokines, growth factors, and other cell-surface molecules, ultimately facilitating protease-dependent tumor progression. Remodeling of the ECM by collagenolytic enzymes such as MMP1, MMP8, MMP13, or the membrane-bound MT1-MMP as well as by other membrane-anchored proteases is required for invasion and recruitment of novel blood vessels. However, the multiple roles of the MMPs do not all fit into a simple pattern. Despite the pro-tumorigenic function of certain metalloproteinases, recent studies have shown that other members of these families, such as MMP8 or MMP11, have a protective role against tumor growth and metastasis in animal models. These studies have been further expanded by large-scale genomic analysis, revealing that the genes encoding metalloproteinases, such as MMP8, MMP27, ADAM7, and ADAM29, are recurrently mutated in specific tumors, while several ADAMTSs are epigenetically silenced in different cancers. The importance of these proteases in modifying the tumor microenvironment highlights the need for a deeper understanding of how stroma cells and the ECM can modulate tumor progression.
Collapse
Affiliation(s)
- Agnès Noël
- Laboratory of Tumor and Development Biology, GIGA-Cancer, University of Liège Liège, Belgium
| | | | | | | | | | | | | | | | | |
Collapse
|
55
|
In Vivo Evidence for a Bridging Role of a Collagen V Subtype at the Epidermis–Dermis Interface. J Invest Dermatol 2012; 132:1841-9. [DOI: 10.1038/jid.2012.56] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
56
|
Gao W, Zhu J, Westfield LA, Tuley EA, Anderson PJ, Sadler JE. Rearranging exosites in noncatalytic domains can redirect the substrate specificity of ADAMTS proteases. J Biol Chem 2012; 287:26944-52. [PMID: 22707719 DOI: 10.1074/jbc.m112.380535] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
ADAMTS proteases typically employ some combination of ancillary C-terminal disintegrin-like, thrombospondin-1, cysteine-rich, and spacer domains to bind substrates and facilitate proteolysis by an N-terminal metalloprotease domain. We constructed chimeric proteases and substrates to examine the role of C-terminal domains of ADAMTS13 and ADAMTS5 in the recognition of their physiological cleavage sites in von Willebrand factor (VWF) and aggrecan, respectively. ADAMTS5 cleaves Glu(373)-Ala(374) and Glu(1480)-Gly(1481) bonds in bovine aggrecan but does not cleave VWF. Conversely, ADAMTS13 cleaves the Tyr(1605)-Met(1606) bond of VWF, which is exposed by fluid shear stress but cannot cleave aggrecan. Replacing the thrombospondin-1/cysteine-rich/spacer domains of ADAMTS5 with those of ADAMTS13 conferred the ability to cleave the Glu(1615)-Ile(1616) bond of VWF domain A2 in peptide substrates or VWF multimers that had been sheared; native (unsheared) VWF multimers were resistant. Thus, by recombining exosites, we engineered ADAMTS5 to cleave a new bond in VWF, preserving physiological regulation by fluid shear stress. The results demonstrate that noncatalytic thrombospondin-1/cysteine-rich/spacer domains are principal modifiers of substrate recognition and cleavage by both ADAMTS5 and ADAMTS13. Noncatalytic domains may perform similar functions in other ADAMTS family members.
Collapse
Affiliation(s)
- Weiqiang Gao
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | | | | | | | | | | |
Collapse
|
57
|
Lewis R, Ravindran S, Wirthlin L, Traeger G, Fernandes RJ, McAlinden A. Disruption of the developmentally-regulated Col2a1 pre-mRNA alternative splicing switch in a transgenic knock-in mouse model. Matrix Biol 2012; 31:214-26. [PMID: 22248926 PMCID: PMC3295890 DOI: 10.1016/j.matbio.2011.12.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2011] [Revised: 12/16/2011] [Accepted: 12/19/2011] [Indexed: 12/17/2022]
Abstract
The present study describes the generation of a knock-in mouse model to address the role of type II procollagen (Col2a1) alternative splicing in skeletal development and maintenance. Alternative splicing of Col2a1 precursor mRNA is a developmentally-regulated event that only occurs in chondrogenic tissue. Normally, chondroprogenitor cells synthesize predominantly exon 2-containing mRNA isoforms (type IIA and IID) while Col2a1 mRNA devoid of exon 2 (type IIB) is the major isoform produced by differentiated chondrocytes. Another isoform, IIC, has also been identified that contains a truncated exon 2 and is not translated into protein. The biological significance of this IIA/IID to IIB splicing switch is not known. Utilizing a splice site targeting knock-in approach, a 4 nucleotide mutation was created to convert the 5' splice site of Col2a1 exon 2 from a weak, non-consensus sequence to a strong, consensus splice site. This resulted in apparent expression of only the IIA mRNA isoform, as confirmed in vitro by splicing of a type II procollagen mini-gene containing the 5' splice site mutation. To test the splice site targeting approach in vivo, homozygote mice engineered to retain IIA exon 2 (Col2a1(+ex2)) were generated. Chondrocytes from hindlimb epiphyseal cartilage of homozygote mice were shown to express only IIA mRNA and protein at all pre- and post-natal developmental stages analyzed (E12.5, E16.5, P0, P3, P7, P14, P28 and P70). As expected, type IIB procollagen was the major isoform produced in wild type cartilage at all post-natal time points. Col2a1(+ex2) homozygote mice are viable, appear healthy and display no overt phenotype to date. However, research is currently underway to investigate the biological consequence of persistent expression of the exon 2-encoded conserved cysteine-rich domain in post-natal skeletal tissues.
Collapse
Affiliation(s)
- Renate Lewis
- Department of Neurology, Washington University School of Medicine, St Louis, MO
| | - Soumya Ravindran
- Department of Orthopaedic Surgery, Washington University School of Medicine, St Louis, MO
| | - Louisa Wirthlin
- Department of Orthopaedic Surgery, Washington University School of Medicine, St Louis, MO
| | - Geoffrey Traeger
- Department of Orthopaedic and Sports Medicine, University of Washington, Seattle, WA
| | - Russell J. Fernandes
- Department of Orthopaedic and Sports Medicine, University of Washington, Seattle, WA
| | - Audrey McAlinden
- Department of Orthopaedic Surgery, Washington University School of Medicine, St Louis, MO
- Department of Cell Biology and Physiology, Washington University School of Medicine, St Louis, MO
| |
Collapse
|
58
|
|
59
|
Bahney CS, Hsu CW, Yoo JU, West JL, Johnstone B. A bioresponsive hydrogel tuned to chondrogenesis of human mesenchymal stem cells. FASEB J 2011; 25:1486-96. [PMID: 21282205 DOI: 10.1096/fj.10-165514] [Citation(s) in RCA: 97] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Cartilage tissue engineering aims to replace damaged or diseased tissue with a functional regenerate that restores joint function. Scaffolds are used to deliver cells and facilitate tissue development, but they can also interfere with the structural assembly of the cartilage matrix. Biodegradable scaffolds have been proposed as a means to improve matrix deposition and the biomechanical properties of neocartilage. The challenge is designing scaffolds with appropriate degradation rates, ideally such that scaffold degradation is proportional to matrix deposition. In this study, we developed a bioresponsive hydrogel with cell-mediated degradation aligned to the chondrogenic differentiation of human mesenchymal stem cells (hMSCs). We identified matrix metalloproteinase 7 (MMP7) as an enzyme with a temporal expression pattern that corresponded with cartilage development. By embedding MMP7 peptide substrates within a poly(ethylene glycol) diacrylate backbone, we built MMP7-sensitive hydrogels with distinct degradation rates. When MMP7-sensitive scaffolds were compared with nondegradable scaffolds in vitro, photoencapsulated hMSCs produced neocartilage constructs with more extensive collagenous matrices, as demonstrated through immunohistochemistry and biochemical quantification of matrix molecules. Furthermore, these changes translated into an increased dynamic compressive modulus. This work presents a practical strategy for designing biomaterials uniquely tuned to individual biological processes.
Collapse
Affiliation(s)
- Chelsea S Bahney
- Department of Orthopaedics and Rehabilitation, Oregon Health and Science University, Portland, Oregon 97239, USA
| | | | | | | | | |
Collapse
|
60
|
Abstract
Classic Ehlers-Danlos syndrome is a heritable connective tissue disorder characterized by skin hyperextensibility, fragile and soft skin, delayed wound healing with formation of atrophic scars, easy bruising, and generalized joint hypermobility. It comprises Ehlers-Danlos syndrome type I and Ehlers-Danlos syndrome type II, but it is now apparent that these form a continuum of clinical findings and differ only in phenotypic severity. It is currently estimated that approximately 50% of patients with a clinical diagnosis of classic Ehlers-Danlos syndrome harbor mutations in the COL5A1 and the COL5A2 gene, encoding the α1 and the α2-chain of type V collagen, respectively. However, because no prospective molecular studies of COL5A1 and COL5A2 have been performed in a clinically well-defined patient group, this number may underestimate the real proportion of patients with classic Ehlers-Danlos syndrome harboring a mutation in one of these genes. In the majority of patients with molecularly characterized classic Ehlers-Danlos syndrome, the disease is caused by a mutation leading to a nonfunctional COL5A1 allele and resulting in haploinsufficiency of type V collagen. A smaller proportion of patients harbor a structural mutation in COL5A1 or COL5A2, causing the production of a functionally defective type V collagen protein. Most mutations identified so far result in a reduced amount of type V collagen in the connective tissues available for collagen fibrillogenesis. Inter- and intrafamilial phenotypic variability is observed, but no genotype-phenotype correlations have been observed. No treatment for the underlying defect is presently available for Ehlers-Danlos syndrome. However, a series of preventive guidelines are applicable.
Collapse
|
61
|
Identification of binding partners interacting with the α1-N-propeptide of type V collagen. Biochem J 2011; 433:371-81. [PMID: 20979576 DOI: 10.1042/bj20101061] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The predominant form of type V collagen is the [α1(V)]₂α2(V) heterotrimer. Mutations in COL5A1 or COL5A2, encoding respectively the α1(V)- and α2(V)-collagen chain, cause classic EDS (Ehlers-Danlos syndrome), a heritable connective tissue disorder, characterized by fragile hyperextensible skin and joint hypermobility. Approximately half of the classic EDS cases remain unexplained. Type V collagen controls collagen fibrillogenesis through its conserved α1(V)-N-propeptide domain. To gain an insight into the role of this domain, a yeast two-hybrid screen among proteins expressed in human dermal fibroblasts was performed utilizing the N-propeptide as a bait. We identified 12 interacting proteins, including extracellular matrix proteins and proteins involved in collagen biosynthesis. Eleven interactions were confirmed by surface plasmon resonance and/or co-immunoprecipitation: α1(I)- and α2(I)-collagen chains, α1(VI)-, α2(VI)- and α3(VI)-collagen chains, tenascin-C, fibronectin, PCPE-1 (procollagen C-proteinase enhancer-1), TIMP-1 (tissue inhibitor of metalloproteinases-1), MMP-2 (matrix metalloproteinase 2) and TGF-β1 (transforming growth factor β1). Solid-phase binding assays confirmed the involvement of the α1(V)-N-propeptide in the interaction between native type V collagen and type VI collagen, suggesting a bridging function of this protein complex in the cell-matrix environment. Enzymatic studies showed that processing of the α1(V)-N-propeptide by BMP-1 (bone morphogenetic protein 1)/procollagen C-proteinase is enhanced by PCPE-1. These interactions are likely to be involved in extracellular matrix homoeostasis and their disruption could explain the pathogenetic mechanism in unresolved classic EDS cases.
Collapse
|
62
|
Dubail J, Kesteloot F, Deroanne C, Motte P, Lambert V, Rakic JM, Lapière C, Nusgens B, Colige A. ADAMTS-2 functions as anti-angiogenic and anti-tumoral molecule independently of its catalytic activity. Cell Mol Life Sci 2010; 67:4213-32. [PMID: 20574651 PMCID: PMC11115784 DOI: 10.1007/s00018-010-0431-6] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2010] [Revised: 05/26/2010] [Accepted: 06/02/2010] [Indexed: 01/30/2023]
Abstract
ADAMTS-2 is a metalloproteinase that plays a key role in the processing of fibrillar procollagen precursors into mature collagen molecules by excising the amino-propeptide. We demonstrate that recombinant ADAMTS-2 is also able to reduce proliferation of endothelial cells, and to induce their retraction and detachment from the substrate resulting in apoptosis. Dephosphorylation of Erk1/2 and MLC largely precedes the ADAMTS-2 induced morphological alterations. In 3-D culture models, ADAMTS-2 strongly reduced branching of capillary-like structures formed by endothelial cells and their long-term maintenance and inhibited vessels formation in embryoid bodies (EB). Growth and vascularization of tumors formed in nude mice by HEK 293-EBNA cells expressing ADAMTS-2 were drastically reduced. A similar anti-tumoral activity was observed when using cells expressing recombinant deleted forms of ADAMTS-2, including catalytically inactive enzyme. Nucleolin, a nuclear protein also found to be associated with the cell membrane, was identified as a potential receptor mediating the antiangiogenic properties of ADAMTS-2.
Collapse
Affiliation(s)
- J. Dubail
- Laboratory of Connective Tissues Biology, GIGA-R, Tour de Pathologie, B23/3, 4000 Sart Tilman, Belgium
| | - F. Kesteloot
- Laboratory of Connective Tissues Biology, GIGA-R, Tour de Pathologie, B23/3, 4000 Sart Tilman, Belgium
| | - C. Deroanne
- Laboratory of Connective Tissues Biology, GIGA-R, Tour de Pathologie, B23/3, 4000 Sart Tilman, Belgium
| | - P. Motte
- Laboratory of Plant Cellular Biology, Sart Tilman, Belgium
| | - V. Lambert
- Laboratory of Development and Tumor Biology, University of Liège, Sart Tilman, Belgium
| | - J.-M. Rakic
- Department of Ophthalmology, University Hospital, Sart Tilman, Belgium
| | - C. Lapière
- Laboratory of Connective Tissues Biology, GIGA-R, Tour de Pathologie, B23/3, 4000 Sart Tilman, Belgium
| | - B. Nusgens
- Laboratory of Connective Tissues Biology, GIGA-R, Tour de Pathologie, B23/3, 4000 Sart Tilman, Belgium
| | - A. Colige
- Laboratory of Connective Tissues Biology, GIGA-R, Tour de Pathologie, B23/3, 4000 Sart Tilman, Belgium
| |
Collapse
|
63
|
Bekhouche M, Kronenberg D, Vadon-Le Goff S, Bijakowski C, Lim NH, Font B, Kessler E, Colige A, Nagase H, Murphy G, Hulmes DJS, Moali C. Role of the netrin-like domain of procollagen C-proteinase enhancer-1 in the control of metalloproteinase activity. J Biol Chem 2010; 285:15950-9. [PMID: 20207734 PMCID: PMC2871463 DOI: 10.1074/jbc.m109.086447] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
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.
Collapse
Affiliation(s)
- Mourad Bekhouche
- From the Institut de Biologie et Chimie des Protéines, CNRS/Université de Lyon UMR 5086, IFR128, 69367 Lyon, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
64
|
Paulissen G, Rocks N, Gueders MM, Crahay C, Quesada-Calvo F, Bekaert S, Hacha J, El Hour M, Foidart JM, Noel A, Cataldo DD. Role of ADAM and ADAMTS metalloproteinases in airway diseases. Respir Res 2009; 10:127. [PMID: 20034386 PMCID: PMC2805617 DOI: 10.1186/1465-9921-10-127] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2009] [Accepted: 12/24/2009] [Indexed: 12/16/2022] Open
Abstract
Lungs are exposed to the outside environment and therefore to toxic and infectious agents or allergens. This may lead to permanent activation of innate immune response elements. A Disintegrin And Metalloproteinases (ADAMs) and ADAMs with Thrombospondin motifs (ADAMTS) are proteinases closely related to Matrix Metalloproteinases (MMPs). These multifaceted molecules bear metalloproteinase and disintegrin domains endowing them with features of both proteinases and adhesion molecules. Proteinases of the ADAM family are associated to various physiological and pathological processes and display a wide spectrum of biological effects encompassing cell fusion, cell adhesion, "shedding process", cleavage of various substrates from the extracellular matrix, growth factors or cytokines... This review will focus on the putative roles of ADAM/ADAMTS proteinases in airway diseases such as asthma and COPD.
Collapse
Affiliation(s)
- Genevieve Paulissen
- Laboratory of Tumor and Development Biology, Groupe Interdisciplinaire de Génoprotéomique Appliquée- GIGA, University of Liège and CHU of Liège, Sart-Tilman, Belgium.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
65
|
Tian J, Pecaut MJ, Slater JM, Gridley DS. Spaceflight modulates expression of extracellular matrix, adhesion, and profibrotic molecules in mouse lung. J Appl Physiol (1985) 2009; 108:162-71. [PMID: 19850731 DOI: 10.1152/japplphysiol.00730.2009] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
NASA has reported pulmonary abnormalities in astronauts on space missions, but the molecular changes in lung tissue remain unknown. The goal of the present study was to explore the effects of spaceflight on expression of extracellular matrix (ECM), cell adhesion, and pro-fibrotic molecules in lungs of mice flown on Space Shuttle Endeavour (STS-118). C57BL/6Ntac mice housed in animal enclosure modules during a 13-day mission in space (FLT) were killed within hours after return; ground controls were treated similarly for comparison (GRD). Analysis of genes associated with ECM and adhesion molecules was performed according to quantitative RT-PCR. The data revealed that FLT lung samples had statistically significant transcriptional changes, i.e., at least 1.5-fold, in 25 out of 84 examined genes (P < 0.05); 15 genes were upregulated and 10 were downregulated. The genes that were upregulated by more than twofold were Ctgf, Mmp2, Ncam1, Sparc, Spock1, and Timp3, whereas the most downregulated genes were Lama1, Mmp3, Mmp7, vcam-1, and Sele. Histology showed profibrosis-like changes occurred in FLT mice, more abundant collagen accumulation around blood vessels, and thicker walls compared with lung samples from GRD mice. Immunohistochemistry was used to compare expression of six selected proteins associated with fibrosis. Immunoreactivity of four proteins (MMP-2, CTGF, TGF-beta1, and NCAM) was enhanced by spaceflight, whereas, no difference was detected in expression of MMP-7 and MMP-9 proteins between the FLT and GRD groups. Taken together, the data demonstrate that significant changes can be readily detected shortly after return from spaceflight in the expression of factors that can adversely influence lung function.
Collapse
Affiliation(s)
- Jian Tian
- Department of Radiation Medicine, Radiation Research Laboratories, Loma Linda Univ., Loma Linda, CA 92354, USA
| | | | | | | |
Collapse
|
66
|
Troeberg L, Fushimi K, Scilabra SD, Nakamura H, Dive V, Thøgersen IB, Enghild JJ, Nagase H. The C-terminal domains of ADAMTS-4 and ADAMTS-5 promote association with N-TIMP-3. Matrix Biol 2009; 28:463-9. [PMID: 19643179 PMCID: PMC2835468 DOI: 10.1016/j.matbio.2009.07.005] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2009] [Revised: 07/20/2009] [Accepted: 07/20/2009] [Indexed: 10/20/2022]
Abstract
We investigated whether the affinity of tissue inhibitor of metalloproteinases (TIMP)-3 for adamalysins with thrombospondin motifs (ADAMTS)-4 and ADAMTS-5 is affected by the non-catalytic ancillary domains of the enzymes. For this purpose, we first established a novel method of purifying recombinant FLAG-tagged TIMP-3 and its inhibitory N-terminal domain (N-TIMP-3) by treating transfected HEK293 cells with sodium chlorate to prevent heparan sulfate proteoglycan-mediated TIMP-3 internalization. TIMP-3 and N-TIMP-3 affinity for selected matrix metalloproteinases and forms of ADAMTS-4 and -5 lacking sequential C-terminal domains was determined. TIMP-3 and N-TIMP-3 displayed similar affinity for various matrix metalloproteinases as has been previously reported for E. coli-expressed N-TIMP-3. ADAMTS-4 and -5 were inhibited more strongly by N-TIMP-3 than by full-length TIMP-3. The C-terminal domains of the enzymes enhanced interaction with N-TIMP-3 and to a lesser extent with the full-length inhibitor. For example, N-TIMP-3 had 7.5-fold better K(i) value for full-length ADAMTS-5 than for the catalytic and disintegrin domain alone. We propose that the C-terminal domains of the enzymes affect the structure around the active site, favouring interaction with TIMP-3.
Collapse
Affiliation(s)
- Linda Troeberg
- Kennedy Institute of Rheumatology, Imperial College London, 65 Aspenlea Road, London, W6 8LH, UK
| | - Kazunari Fushimi
- Kennedy Institute of Rheumatology, Imperial College London, 65 Aspenlea Road, London, W6 8LH, UK
| | - Simone D. Scilabra
- Kennedy Institute of Rheumatology, Imperial College London, 65 Aspenlea Road, London, W6 8LH, UK
| | - Hiroyuki Nakamura
- Kennedy Institute of Rheumatology, Imperial College London, 65 Aspenlea Road, London, W6 8LH, UK
| | - Vincent Dive
- CEA, iBiTecS, Service d'Ingéníerie Moleculaires des Protéines, Gif Sur Yvette, F-91191, France
| | - Ida B. Thøgersen
- Department of Molecular Biology and Interdisciplinary Nanoscience Centre (iNANO), University of Aarhus, Gustav Wieds Vej 10C, DK-8000 Aarhus C, Denmark
| | - Jan J. Enghild
- Department of Molecular Biology and Interdisciplinary Nanoscience Centre (iNANO), University of Aarhus, Gustav Wieds Vej 10C, DK-8000 Aarhus C, Denmark
| | - Hideaki Nagase
- Kennedy Institute of Rheumatology, Imperial College London, 65 Aspenlea Road, London, W6 8LH, UK
| |
Collapse
|
67
|
Apte SS. A disintegrin-like and metalloprotease (reprolysin-type) with thrombospondin type 1 motif (ADAMTS) superfamily: functions and mechanisms. J Biol Chem 2009; 284:31493-7. [PMID: 19734141 DOI: 10.1074/jbc.r109.052340] [Citation(s) in RCA: 349] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Together with seven ADAMTS-like proteins, the 19 mammalian ADAMTS proteases constitute a superfamily. ADAMTS proteases are secreted zinc metalloproteases whose hallmark is an ancillary domain containing one or more thrombospondin type 1 repeats. ADAMTS-like proteins resemble ADAMTS ancillary domains and lack proteolytic activity. Vertebrate expansion of the superfamily reflects emergence of new substrates, duplication of proteolytic activities in new contexts, and cooperative functions of the duplicated genes. ADAMTS proteases are involved in maturation of procollagen and von Willebrand factor, as well as in extracellular matrix proteolysis relating to morphogenesis, angiogenesis, ovulation, cancer, and arthritis. New insights into ADAMTS mechanisms indicate significant regulatory roles for ADAMTS ancillary domains, propeptide processing, and glycosylation. ADAMTS-like proteins appear to have regulatory roles in the extracellular matrix.
Collapse
Affiliation(s)
- Suneel S Apte
- Department of Biomedical Engineering, Cleveland Clinic, Cleveland, Ohio 44195, USA.
| |
Collapse
|
68
|
Malfait F, Paepe AD. Bleeding in the heritable connective tissue disorders: Mechanisms, diagnosis and treatment. Blood Rev 2009. [DOI: 10.1016/j.blre.2009.06.001 doi:dx.doi.org] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
69
|
Malfait F, Paepe AD. Bleeding in the heritable connective tissue disorders: Mechanisms, diagnosis and treatment. Blood Rev 2009; 23:191-7. [DOI: 10.1016/j.blre.2009.06.001] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
70
|
Mineur P, Colige AC, Deroanne CF, Dubail J, Kesteloot F, Habraken Y, Noël A, Vöö S, Waltenberger J, Lapière CM, Nusgens BV, Lambert CA. Newly identified biologically active and proteolysis-resistant VEGF-A isoform VEGF111 is induced by genotoxic agents. ACTA ACUST UNITED AC 2008; 179:1261-73. [PMID: 18086921 PMCID: PMC2140032 DOI: 10.1083/jcb.200703052] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Ultraviolet B and genotoxic drugs induce the expression of a vascular endothelial growth factor A (VEGF-A) splice variant (VEGF111) encoded by exons 1-4 and 8 in many cultured cells. Although not detected in a series of normal human and mouse tissue, VEGF111 expression is induced in MCF-7 xenografts in nude mice upon treatment by camptothecin. The skipping of exons that contain proteolytic cleavage sites and extracellular matrix-binding domains makes VEGF111 diffusible and resistant to proteolysis. Recombinant VEGF111 activates VEGF receptor 2 (VEGF-R2) and extracellularly regulated kinase 1/2 in human umbilical vascular endothelial cells and porcine aortic endothelial cells expressing VEGF-R2. The mitogenic and chemotactic activity and VEGF111's ability to promote vascular network formation during embyonic stem cell differentiation are similar to those of VEGF121 and 165. Tumors in nude mice formed by HEK293 cells expressing VEGF111 develop a more widespread network of numerous small vessels in the peritumoral tissue than those expressing other isoforms. Its potent angiogenic activity and remarkable resistance to proteolysis makes VEGF111 a potential adverse factor during chemotherapy but a beneficial therapeutic tool for ischemic diseases.
Collapse
Affiliation(s)
- Pierre Mineur
- Laboratory of Connective Tissues Biology, University of Liège, B-4000 Liège, Belgium
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
71
|
Tissue-specific induction of ADAMTS2 in monocytes and macrophages by glucocorticoids. J Mol Med (Berl) 2007; 86:323-32. [DOI: 10.1007/s00109-007-0284-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2007] [Revised: 10/23/2007] [Accepted: 10/25/2007] [Indexed: 01/27/2023]
|
72
|
Kesteloot F, Desmoulière A, Leclercq I, Thiry M, Arrese JE, Prockop DJ, Lapière CM, Nusgens BV, Colige A. ADAM metallopeptidase with thrombospondin type 1 motif 2 inactivation reduces the extent and stability of carbon tetrachloride-induced hepatic fibrosis in mice. Hepatology 2007; 46:1620-31. [PMID: 17929299 DOI: 10.1002/hep.21868] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
UNLABELLED ADAMTS2 belongs to the "ADAM metallopeptidase with thrombospondin type 1 motif" (ADAMTS) family. Its primary function is to process collagen type I, II, III, and V precursors into mature molecules by excising the aminopropeptide. This process allows the correct assembly of collagen molecules into fibrils and fibers, which confers to connective tissues their architectural structure and mechanical resistance. To evaluate the impact of ADAMTS2 on the pathological accumulation of extracellular matrix proteins, mainly type I and III collagens, we evaluated carbon tetrachloride-induced liver fibrosis in ADAMTS2-deficient (TS2(-/-)) and wild-type (WT) mice. A single carbon tetrachloride injection caused a similar acute liver injury in deficient and WT mice. A chronic treatment induced collagen deposition in fibrous septa that were made of thinner and irregular fibers in TS2(-/-) mice. The rate of collagen deposition was slower in TS2(-/-) mice, and at an equivalent degree of fibrosis, the resorption of fibrous septa was slightly faster. Most of the genes involved in the development and reversion of the fibrosis were similarly regulated in TS2(-/-) and WT mice. CONCLUSION These data indicate that the extent of fibrosis is reduced in TS2(-/-) mice in comparison with their WT littermates. Inhibiting the maturation of fibrillar collagens may be a beneficial therapeutic approach to interfering with the development of fibrotic lesions.
Collapse
Affiliation(s)
- Frédéric Kesteloot
- Laboratory of Connective Tissues Biology, Interdisciplinary Cluster for Applied Genoproteomics/Center for Experimental Cancer Research, University of Liège, Liège, Belgium
| | | | | | | | | | | | | | | | | |
Collapse
|
73
|
Llamazares M, Obaya AJ, Moncada-Pazos A, Heljasvaara R, Espada J, López-Otín C, Cal S. The ADAMTS12 metalloproteinase exhibits anti-tumorigenic properties through modulation of the Ras-dependent ERK signalling pathway. J Cell Sci 2007; 120:3544-52. [PMID: 17895370 DOI: 10.1242/jcs.005751] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Members of the ADAMTS (a disintegrin and metalloproteinase with thrombospondin motifs) family of proteolytic enzymes are implicated in a variety of physiological processes, such as collagen maturation, organogenesis, angiogenesis, reproduction and inflammation. Moreover, deficiency or overexpression of certain ADAMTS proteins is directly involved in serious human diseases, including cancer. However, the functional roles of other family members, such as ADAMTS12, remain unknown. Here, by using different in vitro and in vivo approaches, we have evaluated the possible role of ADAMTS12 in the development and progression of cancer. First, we show that expression of ADAMTS12 in Madin-Darby canine kidney (MDCK) cells prevents the tumorigenic effects of hepatocyte growth factor (HGF) by blocking the activation of the Ras-MAPK signalling pathway and that this regulation involves the thrombospondin domains of the metalloproteinase. We also show that addition of recombinant human ADAMTS12 to bovine aortic endothelial cells (BAE-1 cells) abolishes their ability to form tubules upon stimulation with vascular endothelial growth factor (VEGF). Additionally, tumours induced in immunodeficient SCID mice injected with A549 cells overexpressing ADAMTS12 show a remarkable growth deficiency in comparison with tumours formed in animals injected with parental A549 cells. Overall, our data suggest that ADAMTS12 confers tumour-protective functions upon cells that produce this proteolytic enzyme.
Collapse
Affiliation(s)
- María Llamazares
- Departamento de Bioquímica y Biología Molecular, Instituto Universitario de Oncologia, Universidad de Oviedo, 33006-Oviedo, Asturias, Spain
| | | | | | | | | | | | | |
Collapse
|
74
|
Bonod-Bidaud C, Beraud M, Vaganay E, Delacoux F, Font B, Hulmes D, Ruggiero F. Enzymatic cleavage specificity of the proalpha1(V) chain processing analysed by site-directed mutagenesis. Biochem J 2007; 405:299-306. [PMID: 17407447 PMCID: PMC1904530 DOI: 10.1042/bj20070051] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The proteolytic processing of procollagen V is complex and depends on the activity of several enzymes among which the BMP-1 (bone morphogenetic protein-1)/tolloid metalloproteinase and the furin-like proprotein convertases. Few of these processing interactions could have been predicted by analysing the presence of conserved consensus sequences in the proalpha1(V) chain. In the present study we opted for a cell approach that allows a straightforward identification of processing interactions. A construct encompassing the complete N-terminal end of the proalpha1(V) chain, referred to as Nalpha1, was recombinantly expressed to be used for enzymatic assays and for antibody production. Structural analysis showed that Nalpha1 is a monomer composed of a compact globule and an extended tail, which correspond respectively to the non-collagenous Nalpha1 subdomains, TSPN-1 (thrombospondin-1 N-terminal domain-like) and the variable region. Nalpha1 was efficiently cleaved by BMP-1 indicating that the triple helix is not required for enzyme activity. By mutating residues flanking the cleavage site, we showed that the aspartate residue at position P2' is essential for BMP-1 activity. BMP-1 activity at the C-terminal end of the procollagen V was assessed by generating a furin double mutant (R1584A/R1585A). We showed that, in absence of furin activity, BMP-1 is capable of processing the C-propeptide even though less efficiently than furin. Altogether, our results provide new relevant information on this complex and poorly understood mechanism of enzymatic processing in procollagen V function.
Collapse
Affiliation(s)
- Christelle Bonod-Bidaud
- *Université de Lyon, Université Lyon 1, Lyon France
- †Institut de Biologie et Chimie des Protéines, UMR 5086 CNRS – Université Lyon 1, 7 passage du Vercors, 69367 Lyon Cedex 07, France
- ‡IFR 128 BioSciences Lyon-Gerland, 7 Passage du Vercors, 69367 Lyon Cedex 07, France
| | - Mickaël Beraud
- *Université de Lyon, Université Lyon 1, Lyon France
- †Institut de Biologie et Chimie des Protéines, UMR 5086 CNRS – Université Lyon 1, 7 passage du Vercors, 69367 Lyon Cedex 07, France
- ‡IFR 128 BioSciences Lyon-Gerland, 7 Passage du Vercors, 69367 Lyon Cedex 07, France
| | - Elisabeth Vaganay
- *Université de Lyon, Université Lyon 1, Lyon France
- †Institut de Biologie et Chimie des Protéines, UMR 5086 CNRS – Université Lyon 1, 7 passage du Vercors, 69367 Lyon Cedex 07, France
- ‡IFR 128 BioSciences Lyon-Gerland, 7 Passage du Vercors, 69367 Lyon Cedex 07, France
| | - Frédéric Delacoux
- *Université de Lyon, Université Lyon 1, Lyon France
- †Institut de Biologie et Chimie des Protéines, UMR 5086 CNRS – Université Lyon 1, 7 passage du Vercors, 69367 Lyon Cedex 07, France
- ‡IFR 128 BioSciences Lyon-Gerland, 7 Passage du Vercors, 69367 Lyon Cedex 07, France
| | - Bernard Font
- *Université de Lyon, Université Lyon 1, Lyon France
- †Institut de Biologie et Chimie des Protéines, UMR 5086 CNRS – Université Lyon 1, 7 passage du Vercors, 69367 Lyon Cedex 07, France
- ‡IFR 128 BioSciences Lyon-Gerland, 7 Passage du Vercors, 69367 Lyon Cedex 07, France
| | - David J. S. Hulmes
- *Université de Lyon, Université Lyon 1, Lyon France
- †Institut de Biologie et Chimie des Protéines, UMR 5086 CNRS – Université Lyon 1, 7 passage du Vercors, 69367 Lyon Cedex 07, France
- ‡IFR 128 BioSciences Lyon-Gerland, 7 Passage du Vercors, 69367 Lyon Cedex 07, France
| | - Florence Ruggiero
- *Université de Lyon, Université Lyon 1, Lyon France
- †Institut de Biologie et Chimie des Protéines, UMR 5086 CNRS – Université Lyon 1, 7 passage du Vercors, 69367 Lyon Cedex 07, France
- ‡IFR 128 BioSciences Lyon-Gerland, 7 Passage du Vercors, 69367 Lyon Cedex 07, France
- To whom correspondence should be addressed (email )
| |
Collapse
|
75
|
Affiliation(s)
- Karl E Kadler
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences, University of Manchester, Michael Smith Building, Oxford Road, Manchester, M13 9PT, UK.
| | | | | | | |
Collapse
|
76
|
Gendron C, Kashiwagi M, Lim NH, Enghild JJ, Thøgersen IB, Hughes C, Caterson B, Nagase H. Proteolytic activities of human ADAMTS-5: comparative studies with ADAMTS-4. J Biol Chem 2007; 282:18294-18306. [PMID: 17430884 DOI: 10.1074/jbc.m701523200] [Citation(s) in RCA: 193] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Aggrecanases have been characterized as proteinases that cleave the Glu373-Ala374 bond of the aggrecan core protein, and they are multidomain metalloproteinases belonging to the ADAMTS (adamalysin with thrombospondin type 1 motifs) family. The first aggrecanases discovered were ADAMTS-4 (aggrecanase 1) and ADAMTS-5 (aggrecanase 2). They contain a zinc catalytic domain followed by non-catalytic ancillary domains, including a disintegrin domain, a thrombospondin domain, a cysteine-rich domain, and a spacer domain. In the case of ADAMTS-5, a second thrombospondin domain follows the spacer domain. We previously reported that the non-catalytic domains of ADAMTS-4 influence both its extracellular matrix interaction and proteolytic abilities. Here we report the effects of these domains of ADAMTS-5 on the extracellular matrix interaction and proteolytic activities and compare them with those of ADAMTS-4. Although the spacer domain was critical for ADAMTS-4 localization in the matrix, the cysteine-rich domain influenced ADAMTS-5 localization. Similar to previous reports of other ADAMTS family members, very little proteolytic activity was detected with the ADAMTS-5 catalytic domain alone. The sequential inclusion of each carboxyl-terminal domain enhanced its activity against aggrecan, carboxymethylated transferrin, fibromodulin, decorin, biglycan, and fibronectin. Both ADAMTS-4 and -5 had a broad optimal activity at pH 7.0-9.5. Aggrecanolytic activities were sensitive to the NaCl concentration, but activities on non-aggrecan substrates, e.g. carboxymethylated transferrin, were not affected. Although ADAMTS-4 and ADAMTS-5 had similar general proteolytic activities, the aggrecanase activity of ADAMTS-5 was at least 1,000-fold greater than that of ADAMTS-4 under physiological conditions. Our studies suggest that ADAMTS-5 is a major aggrecanase in cartilage metabolism and pathology.
Collapse
Affiliation(s)
- Christi Gendron
- Kennedy Institute of Rheumatology Division, Imperial College London, London W6 8LH, United Kingdom
| | - Masahide Kashiwagi
- Kennedy Institute of Rheumatology Division, Imperial College London, London W6 8LH, United Kingdom
| | - Ngee Han Lim
- Kennedy Institute of Rheumatology Division, Imperial College London, London W6 8LH, United Kingdom
| | - Jan J Enghild
- Department of Molecular Biology, University of Aarhus, Science Park, DK-8000 Aarhus C, Denmark
| | - Ida B Thøgersen
- Department of Molecular Biology, University of Aarhus, Science Park, DK-8000 Aarhus C, Denmark
| | - Clare Hughes
- Connective Tissue Laboratories, Cardiff School of Biosciences, University of Cardiff, Cardiff CF1 3US, United Kingdom
| | - Bruce Caterson
- Connective Tissue Laboratories, Cardiff School of Biosciences, University of Cardiff, Cardiff CF1 3US, United Kingdom
| | - Hideaki Nagase
- Kennedy Institute of Rheumatology Division, Imperial College London, London W6 8LH, United Kingdom.
| |
Collapse
|
77
|
Zhang Y, Ge G, Greenspan DS. Inhibition of Bone Morphogenetic Protein 1 by Native and Altered Forms of α2-Macroglobulin. J Biol Chem 2006; 281:39096-104. [PMID: 17071617 DOI: 10.1074/jbc.m601362200] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The four mammalian bone morphogenetic protein 1 (BMP1)-like proteinases act to proteolytically convert procollagens to the major fibrous components of the extracellular matrix. They also activate lysyl oxidase, an enzyme necessary to the covalent cross-linking that gives collagen fibrils much of their tensile strength. Thus, these four proteinases are attractive targets for interventions designed to limit the excess formation of fibrous collagenous matrix that characterizes fibrosis. Although it has previously been reported that the serum protein alpha(2)-macroglobulin is unable to inhibit the astacin-like proteinases meprin alpha and meprin beta, we herein demonstrate alpha(2)-macroglobulin to be a potent inhibitor of the similar BMP1-like proteinases. BMP1 is shown to cleave the alpha(2)-macroglobulin "bait" region, at a single specific site, which resembles the sites at which BMP1-like proteinases cleave the C-propeptides of procollagens I-III. alpha(2)-Macroglobulin is an irreversible inhibitor that is shown to bind bone morphogenetic protein 1 in a covalent complex. It is also demonstrated that genetically modified alpha(2)-macroglobulin, in which the native bait region is replaced by sequences flanking the probiglycan BMP1 cleavage site, is enhanced approximately 24-fold in its ability to inhibit BMP1, and is capable of inhibiting the biosynthetic processing of procollagen I by cells. These findings suggest possible therapeutic interventions involving ectopic expression of modified versions of alpha(2)-macroglobulin in the treatment of fibrotic conditions.
Collapse
Affiliation(s)
- Yue Zhang
- Program in Cellular and Molecular Biology, the Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, Wisconsin 53706, USA
| | | | | |
Collapse
|
78
|
Liu YJ, Xu Y, Yu Q. Full-length ADAMTS-1 and the ADAMTS-1 fragments display pro- and antimetastatic activity, respectively. Oncogene 2006; 25:2452-67. [PMID: 16314835 PMCID: PMC2759703 DOI: 10.1038/sj.onc.1209287] [Citation(s) in RCA: 119] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
The exact role of a disintegrin and metalloproteinase with thrombospondin motifs-1 (ADAMTS-1) and the underlying mechanism of its involvement in tumor metastasis have not been established. We have now demonstrated that overexpression of ADAMTS-1 promotes pulmonary metastasis of TA3 mammary carcinoma and Lewis lung carcinoma cells and that a proteinase-dead mutant of ADAMTS-1 (ADAMTS-1E/Q) inhibits their metastasis, indicating that the prometastatic activity of ADAMTS-1 requires its metalloproteinase activity. Overexpression of ADAMTS-1 in these cells promoted tumor angiogenesis and invasion, shedding of the transmembrane precursors of heparin-binding epidermal growth factor (EGF) and amphiregulin (AR), and activation of the EGF receptor and ErbB-2, while overexpression of ADAMTS-1E/Q inhibited these events. Furthermore, we found that ADAMTS-1 undergoes auto-proteolytic cleavage to generate the NH(2)- and COOH-terminal cleavage fragments containing at least one thrombospondin-type-I-like motif and that overexpression of the NH(2)-terminal ADAMTS-1 fragment and the COOH-terminal ADAMTS-1 fragment can inhibit pulmonary tumor metastasis. These fragments also inhibited Erk1/2 kinase activation induced by soluble heparin-binding EGF and AR. Taken together, our results suggest that the proteolytic status of ADAMTS-1 determines its effect on tumor metastasis, and that the ADAMTS-1E/Q and the ADAMTS-1 fragments likely inhibit tumor metastasis by negatively regulating the availability and activity of soluble heparin-binding EGF and AR.
Collapse
Affiliation(s)
- Y-J Liu
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | | |
Collapse
|