Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Arthritis Rheumatol ; 69(6): 1246-1256, 2017 06.
Article in English | MEDLINE | ID: mdl-28235248

ABSTRACT

OBJECTIVE: The aggrecanase ADAMTS-5 and the collagenase matrix metalloproteinase 13 (MMP-13) are constitutively secreted by chondrocytes in normal cartilage, but rapidly endocytosed via the cell surface endocytic receptor low-density lipoprotein receptor-related protein 1 (LRP-1) and subsequently degraded. This endocytic system is impaired in osteoarthritic (OA) cartilage due to increased ectodomain shedding of LRP-1. The aim of this study was to identify the LRP-1 sheddase(s) in human cartilage and to test whether inhibition of LRP-1 shedding prevents cartilage degradation in OA. METHODS: Cell-associated LRP-1 and soluble LRP-1 (sLRP-1) released from human cartilage explants and chondrocytes were measured by Western blot analysis. LRP-1 sheddases were identified by proteinase inhibitor profiling and gene silencing with small interfering RNAs. Specific monoclonal antibodies were used to selectively inhibit the sheddases. Degradation of aggrecan and collagen in human OA cartilage was measured by Western blot analysis using an antibody against an aggrecan neoepitope and a hydroxyproline assay, respectively. RESULTS: Shedding of LRP-1 was increased in OA cartilage compared with normal tissue. Shed sLRP-1 bound to ADAMTS-5 and MMP-13 and prevented their endocytosis without interfering with their proteolytic activities. Two membrane-bound metalloproteinases, ADAM-17 and MMP-14, were identified as the LRP-1 sheddases in cartilage. Inhibition of their activities restored the endocytic capacity of chondrocytes and reduced degradation of aggrecan and collagen in OA cartilage. CONCLUSION: Shedding of LRP-1 is a key link to OA progression. Local inhibition of LRP-1 sheddase activities of ADAM-17 and MMP-14 is a unique way to reverse matrix degradation in OA cartilage and could be effective as a therapeutic approach.


Subject(s)
Antibodies, Monoclonal/pharmacology , Collagenases/drug effects , Low Density Lipoprotein Receptor-Related Protein-1/drug effects , Osteoarthritis/drug therapy , Proteolysis/drug effects , ADAM17 Protein/analysis , ADAM17 Protein/metabolism , ADAMTS5 Protein/metabolism , Adolescent , Adult , Aggrecans/drug effects , Cartilage, Articular/metabolism , Child , Chondrocytes/physiology , Collagen/drug effects , Endocytosis/drug effects , Female , Humans , Low Density Lipoprotein Receptor-Related Protein-1/physiology , Male , Matrix Metalloproteinase 13/metabolism , Matrix Metalloproteinase 14/analysis , Matrix Metalloproteinase 14/metabolism , Middle Aged , Osteoarthritis/physiopathology , Young Adult
2.
Oncotarget ; 7(13): 16773-92, 2016 Mar 29.
Article in English | MEDLINE | ID: mdl-26934448

ABSTRACT

The membrane-associated matrix metalloproteinase-14, MT1-MMP, has been implicated in pericellular proteolysis with an important role in cellular invasion of collagenous tissues. It is substantially upregulated in various cancers and rheumatoid arthritis, and has been considered as a potential therapeutic target. Here, we report the identification of antibody fragments to MT1-MMP that potently and specifically inhibit its cell surface functions. Lead antibody clones displayed inhibitory activity towards pro-MMP-2 activation, collagen-film degradation and gelatin-film degradation, and were shown to bind to the MT1-MMP catalytic domain outside the active site cleft, inhibiting binding to triple helical collagen. Affinity maturation using CDR3 randomization created a second generation of antibody fragments with dissociation constants down to 0.11 nM, corresponding to an improved affinity of 332-fold with the ability to interfere with cell-surface MT1-MMP functions, displaying IC50 values down to 5 nM. Importantly, the new inhibitors were able to inhibit collagen invasion by tumor-cells in vitro and in vivo primary tumor growth and metastasis of MDA-MB-231 cells in a mouse orthotopic xenograft model. Herein is the first demonstration that an inhibitory antibody targeting sites outside the catalytic cleft of MT1-MMP can effectively abrogate its in vivo activity during tumorigenesis and metastasis.


Subject(s)
Antineoplastic Agents/pharmacology , Matrix Metalloproteinase 14 , Single-Chain Antibodies/pharmacology , Animals , Antibody Affinity , Cell Line, Tumor , Humans , Mice , Mice, SCID , Neoplasm Invasiveness/pathology , Xenograft Model Antitumor Assays
3.
Thromb Haemost ; 114(1): 139-49, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25855589

ABSTRACT

Recombinant tissue-type plasminogen activator (tPA, trade name Alteplase), currently the only drug approved by the US Food and Drug Administration and the European Medicines Agency for the treatment of cerebral ischaemic stroke, has been implicated in a number of adverse effects reportedly mediated by interactions with the low-density lipoprotein (LDL) family receptors, including neuronal cell death and an increased risk of cerebral haemorrhage. The tissue-type plasminogen activator is the principal initiator of thrombolysis in human physiology, an effect that is mediated directly via localised activation of the plasmin zymogen plasminogen at the surface of fibrin clots in the vascular lumen. Here, we sought to identify a ligand to tPA capable of inhibiting the relevant LDL family receptors without interfering with the fibrinolytic activity of tPA. Systematic evolution of ligands by exponential enrichment (SELEX) was employed to isolate tPA-binding RNA aptamers, which were characterised in biochemical assays of tPA association to low density lipoprotein receptor-related protein-1 (LRP-1, an LDL receptor family member); tPA-mediated in vitro and ex vivo clot lysis; and tPA-mediated plasminogen activation in the absence and presence of a stimulating soluble fibrin fragment. Two aptamers, K18 and K32, had minimal effects on clot lysis, but were able to efficiently inhibit tPA-LRP-1 association and LDL receptor family-mediated endocytosis in human vascular endothelial cells and astrocytes. These observations suggest that coadministration alongside tPA may be a viable strategy to improve the safety of thrombolytic treatment of cerebral ischaemic stroke by restricting tPA activity to the vascular lumen.


Subject(s)
Aptamers, Nucleotide/metabolism , Endocytosis , Endothelial Cells/metabolism , Fibrinolytic Agents/metabolism , Receptors, LDL/metabolism , Thrombolytic Therapy/methods , Tissue Plasminogen Activator/metabolism , Aptamers, Nucleotide/chemistry , Aptamers, Nucleotide/genetics , Astrocytes/drug effects , Astrocytes/metabolism , Binding Sites , Binding, Competitive , Cells, Cultured , Computer-Aided Design , Endothelial Cells/drug effects , Fibrin/metabolism , Fibrinolysis/drug effects , Fibrinolytic Agents/toxicity , Humans , Ligands , Low Density Lipoprotein Receptor-Related Protein-1 , Nucleic Acid Conformation , Plasminogen/metabolism , Protein Binding , SELEX Aptamer Technique , Structure-Activity Relationship , Thrombolytic Therapy/adverse effects , Time Factors , Tissue Plasminogen Activator/genetics , Tissue Plasminogen Activator/toxicity , Tumor Suppressor Proteins/metabolism
4.
PLoS One ; 10(3): e0119207, 2015.
Article in English | MEDLINE | ID: mdl-25793507

ABSTRACT

Nucleic acid aptamer selection is a powerful strategy for the development of regulatory agents for molecular intervention. Accordingly, aptamers have proven their diligence in the intervention with serine protease activities, which play important roles in physiology and pathophysiology. Nonetheless, there are only a few studies on the molecular basis underlying aptamer-protease interactions and the associated mechanisms of inhibition. In the present study, we use site-directed mutagenesis to delineate the binding sites of two 2´-fluoropyrimidine RNA aptamers (upanap-12 and upanap-126) with therapeutic potential, both binding to the serine protease urokinase-type plasminogen activator (uPA). We determine the subsequent impact of aptamer binding on the well-established molecular interactions (plasmin, PAI-1, uPAR, and LRP-1A) controlling uPA activities. One of the aptamers (upanap-126) binds to the area around the C-terminal α-helix in pro-uPA, while the other aptamer (upanap-12) binds to both the ß-hairpin of the growth factor domain and the kringle domain of uPA. Based on the mapping studies, combined with data from small-angle X-ray scattering analysis, we construct a model for the upanap-12:pro-uPA complex. The results suggest and highlight that the size and shape of an aptamer as well as the domain organization of a multi-domain protein such as uPA, may provide the basis for extensive sterical interference with protein ligand interactions considered distant from the aptamer binding site.


Subject(s)
Aptamers, Nucleotide/chemistry , Aptamers, Nucleotide/metabolism , Urokinase-Type Plasminogen Activator/metabolism , Binding Sites , Catalytic Domain , Humans , Models, Molecular , Mutagenesis, Site-Directed , SELEX Aptamer Technique , Scattering, Small Angle , Urokinase-Type Plasminogen Activator/chemistry , X-Ray Diffraction
5.
Protein Eng Des Sel ; 27(6): 179-90, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24769623

ABSTRACT

We previously showed that a human anti-TACE antibody, D1(A12), is a potent inhibitor of TNF-α converting enzyme (TACE) ectodomain proteolysis and has pharmacokinetic properties suitable for studies of the inhibition of TACE-dependent growth factor shedding in relation to possible therapeutic applications. However, the lack of murine TACE immunoreactivity limits pre-clinical in vivo studies to human xenograft models which are poor analogies to in situ pathology and are not considered clinically predictive. Here, to overcome these limitations, we set out to develop a 'mouse and human cross-reactive' specific anti-TACE antibody. We first re-investigated the originally selected anti-TACE ectodomain phage-display clones, and isolated a lead 'mouse-human cross-reactive' anti-TACE scFv, clone A9. We reformatted scFv-A9 into an IgG2 framework for comprehensive biochemical and cellular characterization and further demonstrated that A9 is an exosite TACE inhibitor. However, surface plasmon resonance analysis and quenched-fluorescent (QF) peptide assay indicated that IgG reformatting of A9 caused low binding affinity and an 80-fold reduction in TACE ectodomain inhibition, severely limiting its efficacy. To address this, we constructed second generation phage-display randomization libraries focused on the complementarity-determining region 3, and carried out affinity selections shuffling between human and mouse TACE ectodomain as antigen in addition to an off-rate selection to increase the chance of affinity improvement. The bespoke 'three-step' selections enabled a 100-fold affinity enhancement of A9 IgG, and also improved its IC50 in a QF peptide assay to 0.2 nM. In human and mouse cancer cell assays, matured A9 IgG showed significant cell-surface TACE inhibition as a monotherapy or combination therapy with chemotherapeutic agent. Collectively, these data suggest that we successfully developed an exosite inhibitor of TACE with sub-nanomolar affinity, which possesses both murine and human immunoreactive properties that can be used for in vivo application in murine pre-clinical cancer models.


Subject(s)
ADAM Proteins/immunology , Antineoplastic Agents/pharmacology , Single-Chain Antibodies/pharmacology , ADAM Proteins/antagonists & inhibitors , ADAM Proteins/chemistry , ADAM Proteins/metabolism , ADAM17 Protein , Allosteric Regulation , Animals , Antibody Specificity , Antineoplastic Agents/chemistry , Cell Line, Tumor , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Drug Screening Assays, Antitumor , HEK293 Cells , Humans , Immunotherapy , Inhibitory Concentration 50 , Kinetics , Mice , Protein Binding , Single-Chain Antibodies/chemistry , Gemcitabine
6.
Biochem J ; 449(1): 161-6, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23016918

ABSTRACT

An important regulatory mechanism of serine proteases is the proteolytic conversion of the inactive pro-enzyme, or zymogen, into the active enzyme. This activation process is generally considered an irreversible process. In the present study, we demonstrate that an active enzyme can be converted back into its zymogen form. We determined the crystal structure of uPA (urokinase-type plasminogen activator) in complex with an inhibitory antibody, revealing that the antibody 'rezymogenizes' already activated uPA. The present study demonstrates a new regulatory mechanism of protease activity, which is also an extreme case of protein allostery. Mechanistically, the antibody binds a single surface-exposed loop, named the autolysis loop, thereby preventing the stabilization of uPA in its active conformation. We argue that this autolysis loop is a key structural element for rezymogenation of other proteases, and will be a new target site for pharmacological intervention with serine protease activity.


Subject(s)
Antibodies, Monoclonal/pharmacology , Enzyme Precursors/antagonists & inhibitors , Enzyme Precursors/metabolism , Urokinase-Type Plasminogen Activator/metabolism , Antibodies, Monoclonal/chemistry , Cell Line, Tumor , Crystallography, X-Ray , Enzyme Induction/physiology , Enzyme Precursors/chemistry , Enzyme Stability/physiology , Humans , Proteolysis , Urokinase-Type Plasminogen Activator/chemistry
7.
Mol Cancer Res ; 10(12): 1532-43, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23038812

ABSTRACT

Data accumulated over the latest two decades have established that the serine protease urokinase-type plasminogen activator (uPA) is a potential therapeutic target in cancer. When designing inhibitors of the proteolytic activity of serine proteases, obtaining sufficient specificity is problematic, because the topology of the proteases' active sites are highly similar. In an effort to generate highly specific uPA inhibitors with new inhibitory modalities, we isolated uPA-binding RNA aptamers by screening a library of 35 nucleotides long 2'-fluoro-pyrimidine RNA molecules using a version of human pro-uPA lacking the epidermal growth factor-like and kringle domains as bait. One pro-uPA-binding aptamer sequence, referred to as upanap-126, proved to be highly specific for human uPA. Upanap-126 delayed the proteolytic conversion of human pro-uPA to active uPA, but did not inhibit plasminogen activation catalyzed by two-chain uPA. The aptamer also inhibited the binding of pro-uPA to uPAR and the binding of vitronectin to the preformed pro-uPA/uPAR complex, both in cell-free systems and on cell surfaces. Furthermore, upanap-126 inhibited human tumor cell invasion in vitro in the Matrigel assay and in vivo in the chick embryo assay of cell escape from microtumors. Finally, upanap-126 significantly reduced the levels of tumor cell intravasation and dissemination in the chick embryo model of spontaneous metastasis. Together, our findings show that usage of upanap-126 represents a novel multifunctional mechanistic modality for inhibition of uPA-dependent processes involved in tumor cell spread.


Subject(s)
Aptamers, Nucleotide/genetics , Aptamers, Nucleotide/pharmacology , Prostatic Neoplasms/pathology , Prostatic Neoplasms/therapy , Urokinase-Type Plasminogen Activator/antagonists & inhibitors , Urokinase-Type Plasminogen Activator/genetics , Animals , Cell Line, Tumor , Cell-Free System , Chick Embryo , Epidermal Growth Factor/genetics , Epidermal Growth Factor/metabolism , HEK293 Cells , Humans , Male , Molecular Targeted Therapy , Neoplasm Invasiveness , Plasminogen/genetics , Plasminogen/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Serine Proteases/genetics , Serine Proteases/metabolism , Urokinase-Type Plasminogen Activator/metabolism
8.
Neoplasia ; 13(9): 806-21, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21969814

ABSTRACT

Urokinase-type plasminogen activator (uPA) and plasmin have long been implicated in cancer progression. However, the precise contributions of the uPA/plasmin system to specific steps involved in cancer cell dissemination have not been fully established. Herein, we have used a highly disseminating variant of the human PC-3 prostate carcinoma cell line, PC-hi/diss, as a prototype of aggressive carcinomas to investigate the mechanisms whereby pro-uPA activation and uPA-generated plasmin functionally contribute to specific stages of metastasis. The PC-hi/diss cells secrete and activate significant amounts of pro-uPA, leading to efficient generation of plasmin in solution and at the cell surface. In a mouse orthotopic xenograft model, treatment with the specific pro-uPA activation-blocking antibody mAb-112 significantly inhibited local invasion and distant metastasis of the PC-hi/diss cells. To mechanistically examine the uPA/plasmin-mediated aspects of tumor cell dissemination, the anti-pro-uPA mAb-112 and the potent serine protease inhibitor, aprotinin, were used in parallel in a number of in vivo assays modeling various rate-limiting steps in early metastatic spread. Our findings demonstrate that, by generating plasmin, activated tumor-derived uPA facilitates early stages of PC-hi/diss dissemination, specifically the escape from the primary tumor and tumor cell intravasation. Moreover, through a series of in vitro and in vivo analyses, we suggest that PC-hi/diss-invasive escape and dissemination may be enhanced by cleavage of stromal fibronectin by uPA-generated plasmin. Together, our findings point to inhibition of pro-uPA activation at the apex of the uPA/plasmin cascade as a therapy-valid approach to control onset of tumor escape and ensuing metastatic spread.


Subject(s)
Fibrinolysin/metabolism , Neoplasm Metastasis , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Urokinase-Type Plasminogen Activator/metabolism , Animals , Antibodies, Monoclonal/immunology , Aprotinin/pharmacology , Cell Line, Tumor , Cell Movement , Chick Embryo , Enzyme Activation , Fibronectins/metabolism , Humans , Male , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Invasiveness , Serine Proteinase Inhibitors/pharmacology , Transplantation, Heterologous , Tumor Escape , Urokinase-Type Plasminogen Activator/immunology
9.
Biochem J ; 438(1): 39-51, 2011 Aug 15.
Article in English | MEDLINE | ID: mdl-21635223

ABSTRACT

Tight regulation of serine proteases is essential for their physiological function, and unbalanced states of protease activity have been implicated in a variety of human diseases. One key example is the presence of uPA (urokinase-type plasminogen activator) in different human cancer types, with high levels correlating with a poor prognosis. This observation has stimulated efforts into finding new principles for intervening with uPA's activity. In the present study we characterize the so-called autolysis loop in the catalytic domain of uPA as a potential inhibitory target. This loop was found to harbour the epitopes for three conformation-specific monoclonal antibodies, two with a preference for the zymogen form pro-uPA, and one with a preference for active uPA. All three antibodies were shown to have overlapping epitopes, with three common residues being crucial for all three antibodies, demonstrating a direct link between conformational changes of the autolysis loop and the creation of a catalytically mature active site. All three antibodies are potent inhibitors of uPA activity, the two pro-uPA-specific ones by inhibiting conversion of pro-uPA to active uPA and the active uPA-specific antibody by shielding the access of plasminogen to the active site. Furthermore, using immunofluorescence, the conformation-specific antibodies mAb-112 and mAb-12E6B10 enabled us to selectively stain pro-uPA or active uPA on the surface of cultured cells. Moreover, in various independent model systems, the antibodies inhibited tumour cell invasion and dissemination, providing evidence for the feasibility of pharmaceutical intervention with serine protease activity by targeting surface loops that undergo conformational changes during zymogen activation.


Subject(s)
Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/pharmacology , Autolysis/drug therapy , Enzyme Precursors/antagonists & inhibitors , Urokinase-Type Plasminogen Activator/antagonists & inhibitors , Animals , Cell Movement , Chick Embryo , Enzyme Activation/drug effects , Enzyme Precursors/immunology , Enzyme Precursors/metabolism , Fibrinolytic Agents/pharmacology , Humans , Hydrolysis , Kinetics , Mice , Mice, Inbred BALB C , Plasminogen/metabolism , Receptors, Urokinase Plasminogen Activator/metabolism , Surface Plasmon Resonance , Urokinase-Type Plasminogen Activator/immunology , Urokinase-Type Plasminogen Activator/metabolism
10.
J Mol Biol ; 411(2): 417-29, 2011 Aug 12.
Article in English | MEDLINE | ID: mdl-21669207

ABSTRACT

A key regulatory step for serine proteases of the trypsin clan is activation of the initially secreted zymogens, leading to an increase in activity by orders of magnitude. Zymogen activation occurs by cleavage of a single peptide bond near the N-terminus of the catalytic domain. Besides the catalytic domain, most serine proteases have N-terminal A-chains with independently folded domains. Little is known about how zymogen activation affects the interplay between domains. This question is investigated with urokinase-type plasminogen activator (uPA), which has an epidermal growth factor domain and a kringle domain, connected to the catalytic domain by a 15-residue linker. uPA has been implicated under several pathological conditions, and one possibility for pharmacological control is targeting the conversion of the zymogen pro-uPA to active uPA. Therefore, a small-angle X-ray scattering study of the conformations of pro-uPA and uPA in solution was performed. Structural models for the proteins were derived using available atomic-resolution structures for the various domains. Active uPA was found to be flexible with a random conformation of the amino-terminal fragment domain with respect to the serine protease domain. In contrast, pro-uPA was observed to be rigid, with the amino-terminal fragment domain in a fixed position with respect to the serine protease domain. Analytical ultracentrifugation analysis supported the observed difference between pro-uPA and uPA in overall shape and size seen with small-angle X-ray scattering. Upon association of either of two monoclonal Fab (fragment antigen-binding) fragments that are directed against the catalytic domain of, respectively, pro-uPA and uPA, rigid structures were formed.


Subject(s)
Enzyme Precursors/chemistry , Serine Proteases/metabolism , Urokinase-Type Plasminogen Activator/chemistry , Enzyme Precursors/metabolism , Humans , Models, Molecular , Protein Conformation , Protein Structure, Tertiary , Scattering, Small Angle , Ultracentrifugation , Urokinase-Type Plasminogen Activator/metabolism
11.
Am J Pathol ; 175(4): 1638-52, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19729488

ABSTRACT

To analyze the process of tumor cell intravasation, we used the human tumor-chick embryo spontaneous metastasis model to select in vivo high (PC-hi/diss) and low (PC-lo/diss) disseminating variants from the human PC-3 prostate carcinoma cell line. These variants dramatically differed in their intravasation and dissemination capacities in both chick embryo and mouse spontaneous metastasis models. Concomitant with enhanced intravasation, PC-hi/diss exhibited increased angiogenic potential in avian and murine models. PC-hi/diss angiogenesis and intravasation were dependent on increased secretion of vascular endothelial growth factor (VEGF), since treating developing tumors with a function-blocking anti-VEGF antibody simultaneously inhibited both processes without affecting primary tumor growth. PC-hi/diss cells were also more migratory and invasive, suggestive of heightened ability to escape from primary tumors due to matrix-degrading activity. Consistent with this suggestion, PC-hi/diss cells produced more of the serine protease urokinase-type plasminogen activator (uPA) as compared with PC-lo/diss. The functional role of uPA in PC-hi/diss dissemination was confirmed by inhibition of invasion, angiogenesis, and intravasation with specific function-blocking antibodies that prevented uPA activation and blocked uPA activity. These processes were similarly sensitive to aprotinin, a potent inhibitor of serine proteases, including uPA-generated plasmin. Thus, our comparison of the PC-3 intravasation variants points to key roles for the uPA-plasmin system in PC-hi/diss intravasation, possibly via (1) promoting tumor cell matrix invasion and (2) facilitating development of VEGF-dependent angiogenic blood vessels.


Subject(s)
Neovascularization, Pathologic/pathology , Prostatic Neoplasms/blood supply , Prostatic Neoplasms/pathology , Urokinase-Type Plasminogen Activator/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , Cell Adhesion , Cell Line, Tumor , Cell Movement , Chick Embryo , Disease Models, Animal , Humans , Male , Mice , Neoplasm Invasiveness , Neoplasm Metastasis , Vascular Endothelial Growth Factor A/biosynthesis
12.
Biochemistry ; 48(40): 9606-17, 2009 Oct 13.
Article in English | MEDLINE | ID: mdl-19705874

ABSTRACT

Serine proteases are initially synthesized as single-chain proenzymes with activities that are many orders of magnitude lower than those of the mature enzyme. Proteolytic cleavage of an exposed loop liberates a new amino terminus that inserts into a hydrophobic pocket and forms a stabilizing salt bridge with a ubiquitously conserved aspartate residue, resulting in a conformational change organizing the mature oxyanion hole. In a decisive 1976 work, Huber and Bode [Bode, W., and Huber, R. (1976) FEBS Lett. 68, 231-236] demonstrated that peptides sequentially similar to the new amino terminus in combination with a catalytic site inhibitor could specifically induce a trypsin-like conformation in trypsinogen. We now demonstrate that an Ile-Ile or Ile-Val dipeptide can induce limited enzyme activity in the single-chain zymogen form of urokinase-type plasminogen activator (uPA) or its K158A variant, which cannot be activated proteolytically. Furthermore, the slow formation of a covalent serpin-protease complex between single-chain uPA and PAI-1 is significantly accelerated in the presence of specific dipeptide sequences. The technique of using a dipeptide mimic as a surrogate for the liberated amino terminus further provides a novel means by which to covalently label the immature active site of single-chain uPA with a fluorescent probe, permitting fluorescence approaches for direct observations of conformational changes within the protease domain during zymogen activation. These data demonstrate the structural plasticity of the protease domain, reinforce the notion of "molecular sexuality", and provide a novel way of studying conformational changes of zymogens during proteolytic activation.


Subject(s)
Dipeptides/physiology , Peptide Hydrolases , Urokinase-Type Plasminogen Activator/biosynthesis , Urokinase-Type Plasminogen Activator/chemistry , Animals , Catalysis , Cattle , Chromogenic Compounds/chemistry , Chromogenic Compounds/metabolism , Dipeptides/chemistry , Dipeptides/metabolism , Enzyme Induction , Enzyme Precursors/chemistry , Enzyme Precursors/metabolism , Humans , Peptide Hydrolases/chemistry , Peptide Hydrolases/physiology , Substrate Specificity
13.
J Biol Chem ; 284(7): 4647-57, 2009 Feb 13.
Article in English | MEDLINE | ID: mdl-19047064

ABSTRACT

Serine proteases are secreted from cells as single-chain zymogens, typically having activities orders of magnitude lower than those of the mature two-chain enzymes. Activation occurs by a conformational change initiated by cleavage of a specific peptide bond. We have derived a monoclonal antibody (mAb-112) which binds with subnanomolar affinity to pro-uPA, the zymogen form of urokinase-type plasminogen activator (uPA). We mapped the epitope of the antibody to the autolysis loop, one of the structural elements known to change conformation during zymogen activation. A mechanistic evaluation with biophysical methods elucidated a novel bifunctional inhibitory mechanism whereby mAb-112 not only delays the proteolytic conversion of single-chain pro-uPA into the two-chain form but also subsequently averts the conformational transition to a mature protease by sequestering the two-chain form in a zymogen-like, noncatalytic state. Functional studies employing two variants of human HT-1080 cells, exhibiting high and low levels of dissemination in a chorioallantoic membrane assay, demonstrate that mAb-112 is an effective inhibitor of tumor cell intravasation. These findings show that pharmacological interference with zymogen activation is a plausible and robust means to regulate uPA activity and the downstream effects of plasminogen activation in the spread of cancer and other processes of pathological tissue remodeling. A strategy that targets regions related to pro-enzyme activation likely provide a unique inhibitor-protease interaction surface and is, thus, expected to enhance the chances of engineering high inhibitor specificity. Our results provide new information about the structural flexibility underlying the equilibrium between active and inactive forms of serine proteases.


Subject(s)
Antibodies, Monoclonal/pharmacology , Enzyme Precursors/antagonists & inhibitors , Models, Chemical , Urokinase-Type Plasminogen Activator/antagonists & inhibitors , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Cell Line, Tumor , Chick Embryo , Enzyme Activation/drug effects , Enzyme Precursors/immunology , Enzyme Precursors/metabolism , Epitope Mapping/methods , Epitopes/immunology , Humans , Neoplasms/drug therapy , Neoplasms/enzymology , Neoplasms/immunology , Urokinase-Type Plasminogen Activator/immunology , Urokinase-Type Plasminogen Activator/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...