Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
1.
Biochem J ; 477(9): 1701-1719, 2020 05 15.
Article in English | MEDLINE | ID: mdl-32296833

ABSTRACT

To facilitate investigations of protein-protein interactions (PPIs), we developed a novel platform for quantitative mapping of protein binding specificity landscapes, which combines the multi-target screening of a mutagenesis library into high- and low-affinity populations with sophisticated next-generation sequencing analysis. Importantly, this method generates accurate models to predict affinity and specificity values for any mutation within a protein complex, and requires only a few experimental binding affinity measurements using purified proteins for calibration. We demonstrated the utility of the approach by mapping quantitative landscapes for interactions between the N-terminal domain of the tissue inhibitor of metalloproteinase 2 (N-TIMP2) and three matrix metalloproteinases (MMPs) having homologous structures but different affinities (MMP-1, MMP-3, and MMP-14). The binding landscapes for N-TIMP2/MMP-1 and N-TIMP2/MMP-3 showed the PPIs to be almost fully optimized, with most single mutations giving a loss of affinity. In contrast, the non-optimized PPI for N-TIMP2/MMP-14 was reflected in a wide range of binding affinities, where single mutations exhibited a far more attenuated effect on the PPI. Our new platform reliably and comprehensively identified not only hot- and cold-spot residues, but also specificity-switch mutations that shape target affinity and specificity. Thus, our approach provides a methodology giving an unprecedentedly rich quantitative analysis of the binding specificity landscape, which will broaden the understanding of the mechanisms and evolutionary origins of specific PPIs and facilitate the rational design of specific inhibitors for structurally similar target proteins.


Subject(s)
Matrix Metalloproteinase 14/metabolism , Matrix Metalloproteinase 1/metabolism , Protein Interaction Domains and Motifs , Protein Interaction Mapping/methods , Tissue Inhibitor of Metalloproteinase-2/genetics , Computational Biology/methods , Gene Library , High-Throughput Nucleotide Sequencing , Matrix Metalloproteinase 3/metabolism , Mutagenesis , Mutation , Protein Engineering/methods , Tissue Inhibitor of Metalloproteinase-2/chemistry , Tissue Inhibitor of Metalloproteinase-2/metabolism
2.
FEBS Lett ; 592(14): 2499-2511, 2018 07.
Article in English | MEDLINE | ID: mdl-29933498

ABSTRACT

Due in part to the needs of the biopharmaceutical industry, there has been an increased drive to generate high quality recombinant proteins in large amounts. However, achieving high yields can be a challenge as the novelty and increased complexity of new targets often makes them 'difficult-to-express'. This study aimed to define the molecular features that restrict the production of a model 'difficult-to-express' recombinant protein, Tissue Inhibitor Metalloproteinase-3 (TIMP-3). Building from experimental data, computational approaches were used to rationalize the redesign of this recombinant target to generate a chimera with enhanced secretion. The results highlight the importance of early identification of unfavourable sequence attributes, enabling the generation of engineered protein forms that bypass 'secretory' bottlenecks and result in efficient recombinant protein production.


Subject(s)
Cloning, Molecular/methods , Protein Engineering/methods , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Amino Acid Sequence , Animals , CHO Cells , Computational Biology , Cricetinae , Cricetulus , Gene Expression , Humans , Mice , Models, Biological , Protein Transport/genetics , Recombinant Proteins/chemistry , Secretory Pathway/genetics , Synthetic Biology/methods , Tissue Inhibitor of Metalloproteinase-2/chemistry , Tissue Inhibitor of Metalloproteinase-2/genetics , Tissue Inhibitor of Metalloproteinase-2/metabolism , Tissue Inhibitor of Metalloproteinase-3/chemistry , Tissue Inhibitor of Metalloproteinase-3/genetics , Tissue Inhibitor of Metalloproteinase-3/metabolism
3.
Biomacromolecules ; 19(4): 1285-1293, 2018 04 09.
Article in English | MEDLINE | ID: mdl-29505725

ABSTRACT

Tissue inhibitor of metalloproteinase 2 (TIMP-2) is an endogenous 22 kDa proteinase inhibitor, demonstrating antitumorigenic, antimetastatic and antiangiogenic activities in vitro and in vivo. Recombinant TIMP-2 is currently undergoing preclinical testing in multiple, murine tumor models. Here we report the development of an inert, injectable peptide hydrogel matrix enabling encapsulation and sustained release of TIMP-2. We studied the TIMP-2 release profile from four ß-hairpin peptide gels of varying net electrostatic charge. A negatively charged peptide gel (designated AcVES3) enabling encapsulation of 4 mg/mL of TIMP-2, without effects on rheological properties, facilitated the slow sustained release (0.9%/d) of TIMP-2 over 28 d. Released TIMP-2 is structurally intact and maintains the ability to inhibit MMP activity, as well as suppress lung cancer cell proliferation in vitro. These findings suggest that the AcVES3 hydrogel will be useful as an injectable vehicle for systemic delivery of TIMP-2 in vivo for ongoing preclinical development.


Subject(s)
Gene Transfer Techniques , Lung Neoplasms/genetics , Recombinant Proteins/genetics , Tissue Inhibitor of Metalloproteinase-2/genetics , Cell Proliferation/drug effects , Humans , Hydrogel, Polyethylene Glycol Dimethacrylate/chemistry , Hydrogel, Polyethylene Glycol Dimethacrylate/therapeutic use , Lung Neoplasms/therapy , Peptides/chemistry , Peptides/genetics , Peptides/therapeutic use , Recombinant Proteins/chemistry , Recombinant Proteins/therapeutic use , Rheology , Static Electricity , Tissue Inhibitor of Metalloproteinase-2/chemistry , Tissue Inhibitor of Metalloproteinase-2/therapeutic use
4.
Eur Rev Med Pharmacol Sci ; 22(5): 1315-1322, 2018 03.
Article in English | MEDLINE | ID: mdl-29565489

ABSTRACT

OBJECTIVE: To clarify the role of long non-coding RNA (lncRNA) GACAT3 in invasion and metastasis of non-small cell lung cancer (NSCLC) and its effect on radiotherapy. PATIENTS AND METHODS: The expression of GACAT3 and TIMP2 in cells and tissues of NSCLC were detected by quantitative Real-time PCR (qRT-PCR). The influence of GACAT3 on cell proliferation and the capacity of colony formation were estimated by MTT test and colony forming experiment respectively. Luciferase reporting assay was used to confirm the correlation between GACAT3 and TIMP2. In addition, we observed the influence of GACAT3 on radiosensitivity of NSCLC cells. RESULTS: Using lncRNA array analysis, we found that GACAT3 expression increased significantly. Further studies showed that overexpression of ectopic GACAT3 in A549 cells promoted cell proliferation and migration, and enhanced the sensitivity of lung cancer cells to radiotherapy. TIMP2, confirmed a direct target of GACAT3 by bioinformatics analysis and our experiments, may be involved in the GACAT3-induced upregulation of MMP10. CONCLUSIONS: LncRNA GACAT3 may be a potential biomarker for the evaluation of curative effect and prognosis of lung cancer.


Subject(s)
Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/pathology , RNA, Long Noncoding/metabolism , 3' Untranslated Regions , A549 Cells , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/radiotherapy , Cell Movement/radiation effects , Cell Proliferation/radiation effects , Female , Humans , Lung Neoplasms/genetics , Lung Neoplasms/radiotherapy , Male , Matrix Metalloproteinase 10/metabolism , Middle Aged , Prognosis , RNA, Long Noncoding/chemistry , RNA, Long Noncoding/genetics , Radiation, Ionizing , Tissue Inhibitor of Metalloproteinase-2/chemistry , Tissue Inhibitor of Metalloproteinase-2/genetics , Tissue Inhibitor of Metalloproteinase-2/metabolism , Up-Regulation
5.
FEBS Lett ; 592(7): 1122-1134, 2018 04.
Article in English | MEDLINE | ID: mdl-29473954

ABSTRACT

MMP-14 and MMP-9 are two well-established cancer targets for which no specific clinically relevant inhibitor is available. Using a powerful combination of computational design and yeast surface display technology, we engineered such an inhibitor starting from a nonspecific MMP inhibitor, N-TIMP2. The engineered purified N-TIMP2 variants showed enhanced specificity toward MMP-14 and MMP-9 relative to a panel of off-target MMPs. MMP-specific N-TIMP2 sequence signatures were obtained that could be understood from the structural perspective of MMP/N-TIMP2 interactions. Our MMP-9 inhibitor exhibited 1000-fold preference for MMP-9 vs. MMP-14, which is likely to translate into significant differences under physiological conditions. Our results provide new insights regarding evolution of promiscuous proteins and optimization strategies for design of inhibitors with single-target specificities.


Subject(s)
Matrix Metalloproteinase 14/chemistry , Matrix Metalloproteinase 9/chemistry , Matrix Metalloproteinase Inhibitors/chemistry , Tissue Inhibitor of Metalloproteinase-2/chemistry , Humans , Matrix Metalloproteinase 14/chemical synthesis , Protein Binding
6.
Biochemistry ; 56(49): 6423-6433, 2017 Dec 12.
Article in English | MEDLINE | ID: mdl-29140689

ABSTRACT

Tissue inhibitor of metalloprotease-2 (TIMP-2) is a secreted 21 kDa multifunctional protein first described as an endogenous inhibitor of matrix metalloproteinases (MMPs) that prevents breakdown of the extracellular matrix often observed in chronic diseases. TIMP-2 diminishes the level of growth factor-mediated cell proliferation in vitro, as well as neoangiogenesis and tumor growth in vivo independent of its MMP inhibitory activity. These physiological properties make TIMP-2 an excellent candidate for further preclinical development as a biologic therapy of cancer. Here we present a straightforward bioprocessing methodology that yields >35 mg/L recombinant human TIMP-2 6XHis-tagged protein (rhTIMP-2) from suspension cultures of HEK-293-F cells. Enhanced rhTIMP-2-6XHis yields were achieved by optimization of both TIMP-2 cDNA codon sequence and cell culture conditions. Using a two-step chromatographic process, we achieved >95% purity with minimal processing losses. Purified rhTIMP-2-6XHis was free of mouse antigen contamination. Circular dichroism spectroscopy indicated a well-folded rhTIMP-2-6XHis that is highly stable and refractory to pH changes. Two-dimensional heteronuclear single-quantum coherence nuclear magnetic resonance of full length rhTIMP-2-6XHis also indicated a monodisperse, well-folded protein preparation. Purified rhTIMP-2-6XHis inhibited MMP-2 enzymatic activity in a dose-dependent fashion with an IC50 of ∼1.4 nM. Pretreatment of A549 lung cancer and JygMC(A) triple-negative breast cancer cells with rhTIMP-2-6XHis in low-nanomolar amounts inhibited EGF-induced proliferation to basal (unstimulated) levels. This study therefore not only offers a robust bioprocess methodology for rhTIMP-2 production but also characterizes critical physicochemical and biological attributes that are useful for monitoring quality control of the production process.


Subject(s)
Protein Engineering/methods , Tissue Inhibitor of Metalloproteinase-2/chemistry , Tissue Inhibitor of Metalloproteinase-2/pharmacology , Animals , Cell Proliferation/drug effects , HEK293 Cells , Humans , Mice , Neoplasms/drug therapy , Neoplasms/physiopathology , Protein Folding , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Tissue Inhibitor of Metalloproteinase-2/genetics , Tissue Inhibitor of Metalloproteinase-2/metabolism
7.
Microb Cell Fact ; 16(1): 73, 2017 Apr 28.
Article in English | MEDLINE | ID: mdl-28454584

ABSTRACT

BACKGROUND: As regulators of multifunctional metalloproteinases including MMP, ADAM and ADAMTS families, tissue inhibitors of metalloproteinases (TIMPs) play a pivotal role in extracellular matrix remodeling, which is involved in a wide variety of physiological processes. Since abnormal metalloproteinase activities are related to numerous diseases such as arthritis, cancer, atherosclerosis, and neurological disorders, TIMPs and their engineered mutants hold therapeutic potential and thus have been extensively studied. Traditional productions of functional TIMPs and their N-terminal inhibitory domains (N-TIMPs) rely on costly and time-consuming insect and mammalian cell systems, or tedious and inefficient refolding from denatured inclusion bodies. The later process is also associated with heterogeneous products and batch-to-batch variation. RESULTS: In this study, we developed a simple approach to directly produce high yields of active TIMPs in the periplasmic space of Escherichia coli without refolding. Facilitated by disulfide isomerase (DsbC) co-expression in protease-deficient strain BL21 (DE3), N-TIMP-1/-2 and TIMP-2 which contain multiple disulfide bonds were produced without unwanted truncations. 0.2-1.4 mg purified monomeric TIMPs were typically yielded per liter of culture media. Periplasmically produced TIMPs exhibited expected inhibition potencies towards MMP-1/2/7/14, and were functional in competitive ELISA to elucidate the binding epitopes of MMP specific antibodies. In addition, prepared N-TIMPs were fully active in a cellular context, i.e. regulating cancer cell morphology and migration in 2D and 3D bioassays. CONCLUSION: Periplasmic expression in E. coli is an excellent strategy to recombinantly produce active TIMPs and N-TIMPs.


Subject(s)
Escherichia coli/metabolism , Periplasm/enzymology , Tissue Inhibitor of Metalloproteinase-1/genetics , Tissue Inhibitor of Metalloproteinase-1/metabolism , Tissue Inhibitor of Metalloproteinase-2/genetics , Tissue Inhibitor of Metalloproteinase-2/metabolism , Cloning, Molecular , Epitopes/immunology , Humans , Metalloproteases/antagonists & inhibitors , Periplasm/metabolism , Protein Disulfide-Isomerases/genetics , Protein Disulfide-Isomerases/metabolism , Protein Folding , Recombinant Proteins/metabolism , Solubility , Tissue Inhibitor of Metalloproteinase-1/chemistry , Tissue Inhibitor of Metalloproteinase-1/pharmacology , Tissue Inhibitor of Metalloproteinase-2/chemistry , Tissue Inhibitor of Metalloproteinase-2/pharmacology
8.
J Biol Chem ; 292(8): 3481-3495, 2017 02 24.
Article in English | MEDLINE | ID: mdl-28087697

ABSTRACT

Degradation of the extracellular matrices in the human body is controlled by matrix metalloproteinases (MMPs), a family of more than 20 homologous enzymes. Imbalance in MMP activity can result in many diseases, such as arthritis, cardiovascular diseases, neurological disorders, fibrosis, and cancers. Thus, MMPs present attractive targets for drug design and have been a focus for inhibitor design for as long as 3 decades. Yet, to date, all MMP inhibitors have failed in clinical trials because of their broad activity against numerous MMP family members and the serious side effects of the proposed treatment. In this study, we integrated a computational method and a yeast surface display technique to obtain highly specific inhibitors of MMP-14 by modifying the natural non-specific broad MMP inhibitor protein N-TIMP2 to interact optimally with MMP-14. We identified an N-TIMP2 mutant, with five mutations in its interface, that has an MMP-14 inhibition constant (Ki ) of 0.9 pm, the strongest MMP-14 inhibitor reported so far. Compared with wild-type N-TIMP2, this variant displays ∼900-fold improved affinity toward MMP-14 and up to 16,000-fold greater specificity toward MMP-14 relative to other MMPs. In an in vitro and cell-based model of MMP-dependent breast cancer cellular invasiveness, this N-TIMP2 mutant acted as a functional inhibitor. Thus, our study demonstrates the enormous potential of a combined computational/directed evolution approach to protein engineering. Furthermore, it offers fundamental clues into the molecular basis of MMP regulation by N-TIMP2 and identifies a promising MMP-14 inhibitor as a starting point for the development of protein-based anticancer therapeutics.


Subject(s)
Drug Design , Matrix Metalloproteinase 14/metabolism , Matrix Metalloproteinase Inhibitors/chemistry , Matrix Metalloproteinase Inhibitors/pharmacology , Tissue Inhibitor of Metalloproteinase-2/chemistry , Tissue Inhibitor of Metalloproteinase-2/pharmacology , Amino Acid Sequence , Animals , Cattle , Crystallography, X-Ray , Directed Molecular Evolution , Humans , Matrix Metalloproteinase 14/chemistry , Matrix Metalloproteinase Inhibitors/metabolism , Molecular Docking Simulation , Mutation , Tissue Inhibitor of Metalloproteinase-2/genetics
9.
J Biol Chem ; 291(21): 11348-58, 2016 May 20.
Article in English | MEDLINE | ID: mdl-27033700

ABSTRACT

The four tissue inhibitors of metalloproteinases (TIMPs) are potent inhibitors of the many matrixins (MMPs), except that TIMP1 weakly inhibits some MMPs, including MMP14. The broad-spectrum inhibition of MMPs by TIMPs and their N-domains (NTIMPs) is consistent with the previous isothermal titration calorimetric finding that their interactions are entropy-driven but differ in contributions from solvent and conformational entropy (ΔSsolv, ΔSconf), estimated using heat capacity changes (ΔCp). Selective engineered NTIMPs have potential applications for treating MMP-related diseases, including cancer and cardiomyopathy. Here we report isothermal titration calorimetric studies of the effects of selectivity-modifying mutations in NTIMP1 and NTIMP2 on the thermodynamics of their interactions with MMP1, MMP3, and MMP14. The weak inhibition of MMP14 by NTIMP1 reflects a large conformational entropy penalty for binding. The T98L mutation, peripheral to the NTIMP1 reactive site, enhances binding by increasing ΔSsolv but also reduces ΔSconf However, the same mutation increases NTIMP1 binding to MMP3 in an interaction that has an unusual positive ΔCp This indicates a decrease in solvent entropy compensated by increased conformational entropy, possibly reflecting interactions involving alternative conformers. The NTIMP2 mutant, S2D/S4A is a selective MMP1 inhibitor through electrostatic effects of a unique MMP-1 arginine. Asp-2 increases reactive site polarity, reducing ΔCp, but increases conformational entropy to maintain strong binding to MMP1. There is a strong negative correlation between ΔSsolv and ΔSconf for all characterized interactions, but the data for each MMP have characteristic ranges, reflecting intrinsic differences in the structures and dynamics of their free and inhibitor-bound forms.


Subject(s)
Matrix Metalloproteinase 14/chemistry , Matrix Metalloproteinase 14/metabolism , Matrix Metalloproteinase 1/chemistry , Matrix Metalloproteinase 1/metabolism , Matrix Metalloproteinase 3/chemistry , Matrix Metalloproteinase 3/metabolism , Tissue Inhibitor of Metalloproteinase-1/chemistry , Tissue Inhibitor of Metalloproteinase-1/metabolism , Tissue Inhibitor of Metalloproteinase-2/chemistry , Tissue Inhibitor of Metalloproteinase-2/metabolism , Humans , Matrix Metalloproteinase 1/genetics , Matrix Metalloproteinase 14/genetics , Matrix Metalloproteinase 3/genetics , Models, Molecular , Mutagenesis, Site-Directed , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Thermodynamics , Tissue Inhibitor of Metalloproteinase-1/genetics , Tissue Inhibitor of Metalloproteinase-2/genetics
10.
PLoS One ; 9(4): e93712, 2014.
Article in English | MEDLINE | ID: mdl-24710006

ABSTRACT

Multispecific proteins play a major role in controlling various functions such as signaling, regulation of transcription/translation, and immune response. Hence, a thorough understanding of the atomic-level principles governing multispecific interactions is important not only for the advancement of basic science but also for applied research such as drug design. Here, we study evolution of an exemplary multispecific protein, a Tissue Inhibitor of Matrix Metalloproteinases 2 (TIMP2) that binds with comparable affinities to more than twenty-six members of the Matrix Metalloproteinase (MMP) and the related ADAMs families. We postulate that due to its multispecific nature, TIMP2 is not optimized to bind to any individual MMP type, but rather embodies a compromise required for interactions with all MMPs. To explore this hypothesis, we perform computational saturation mutagenesis of the TIMP2 binding interface and predict changes in free energy of binding to eight MMP targets. Computational results reveal the non-optimality of the TIMP2 binding interface for all studied proteins, identifying many affinity-enhancing mutations at multiple positions. Several TIMP2 point mutants predicted to enhance binding affinity and/or binding specificity towards MMP14 were selected for experimental verification. Experimental results show high abundance of affinity-enhancing mutations in TIMP2, with some point mutations producing more than ten-fold improvement in affinity to MMP14. Our computational and experimental results collaboratively demonstrate that the TIMP2 sequence lies far from the fitness maximum when interacting with its target enzymes. This non-optimality of the binding interface and high potential for improvement might characterize all proteins evolved for binding to multiple targets.


Subject(s)
Matrix Metalloproteinase 14/chemistry , Matrix Metalloproteinase 14/genetics , Molecular Dynamics Simulation , Point Mutation , Tissue Inhibitor of Metalloproteinase-2/chemistry , Tissue Inhibitor of Metalloproteinase-2/genetics , Amino Acid Substitution , Humans , Matrix Metalloproteinase 14/metabolism , Mutagenesis , Protein Binding , Protein Structure, Quaternary , Tissue Inhibitor of Metalloproteinase-2/metabolism
11.
PLoS One ; 8(9): e75836, 2013.
Article in English | MEDLINE | ID: mdl-24073280

ABSTRACT

Matrix metalloproteinases (MMPs) play central roles in vertebrate tissue development, remodeling, and repair. The endogenous tissue inhibitors of metalloproteinases (TIMPs) regulate proteolytic activity by binding tightly to the MMP active site. While each of the four TIMPs can inhibit most MMPs, binding data reveal tremendous heterogeneity in affinities of different TIMP/MMP pairs, and the structural features that differentiate stronger from weaker complexes are poorly understood. Here we report the crystal structure of the comparatively weakly bound human MMP-10/TIMP-2 complex at 2.1 Å resolution. Comparison with previously reported structures of MMP-3/TIMP-1, MT1-MMP/TIMP-2, MMP-13/TIMP-2, and MMP-10/TIMP-1 complexes offers insights into the structural basis of binding selectivity. Our analyses identify a group of highly conserved contacts at the heart of MMP/TIMP complexes that define the conserved mechanism of inhibition, as well as a second category of diverse adventitious contacts at the periphery of the interfaces. The AB loop of the TIMP N-terminal domain and the contact loops of the TIMP C-terminal domain form highly variable peripheral contacts that can be considered as separate exosite interactions. In some complexes these exosite contacts are extensive, while in other complexes the AB loop or C-terminal domain contacts are greatly reduced and appear to contribute little to complex stability. Our data suggest that exosite interactions can enhance MMP/TIMP binding, although in the relatively weakly bound MMP-10/TIMP-2 complex they are not well optimized to do so. Formation of highly variable exosite interactions may provide a general mechanism by which TIMPs are fine-tuned for distinct regulatory roles in biology.


Subject(s)
Matrix Metalloproteinase 10/chemistry , Matrix Metalloproteinases/chemistry , Tissue Inhibitor of Metalloproteinase-2/chemistry , Tissue Inhibitor of Metalloproteinases/chemistry , Binding Sites , Catalytic Domain , Crystallography, X-Ray , Humans , Matrix Metalloproteinase 10/metabolism , Matrix Metalloproteinases/metabolism , Protein Binding , Protein Structure, Tertiary , Substrate Specificity , Tissue Inhibitor of Metalloproteinase-2/metabolism , Tissue Inhibitor of Metalloproteinases/metabolism
12.
Biochem Soc Trans ; 41(5): 1166-9, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24059503

ABSTRACT

Manipulations of PPIs (protein-protein interactions) are important for many biological applications such as synthetic biology and drug design. Combinatorial methods have been traditionally used for such manipulations, failing, however, to explain the effects achieved. We developed a computational method for prediction of changes in free energy of binding due to mutation that bring about deeper understanding of the molecular forces underlying binding interactions. Our method could be used for computational scanning of binding interfaces and subsequent analysis of the interfacial sequence optimality. The computational method was validated in two biological systems. Computational saturated mutagenesis of a high-affinity complex between an enzyme AChE (acetylcholinesterase) and a snake toxin Fas (fasciculin) revealed the optimal nature of this interface with only a few predicted affinity-enhancing mutations. Binding measurements confirmed high optimality of this interface and identified a few mutations that could further improve interaction fitness. Computational interface scanning of a medium-affinity complex between TIMP-2 (tissue inhibitor of metalloproteinases-2) and MMP (matrix metalloproteinase) 14 revealed a non-optimal nature of the binding interface with multiple mutations predicted to stabilize the complex. Experimental results corroborated our computational predictions, identifying a large number of mutations that improve the binding affinity for this interaction and some mutations that enhance binding specificity. Overall, our computational protocol greatly facilitates the discovery of affinity- and specificity-enhancing mutations and thus could be applied for design of potent and highly specific inhibitors of any PPI.


Subject(s)
Computational Biology , Protein Conformation , Protein Interaction Maps/genetics , Humans , Matrix Metalloproteinase 14/chemistry , Matrix Metalloproteinase 14/genetics , Matrix Metalloproteinase 9/chemistry , Matrix Metalloproteinase 9/genetics , Mutation , Sensitivity and Specificity , Tissue Inhibitor of Metalloproteinase-2/chemistry , Tissue Inhibitor of Metalloproteinase-2/genetics
13.
FEBS J ; 280(1): 139-59, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23121732

ABSTRACT

Artificial miniproteins that are able to target catalytic sites of matrix metalloproteinases (MMPs) were designed using a functional motif-grafting approach. The motif corresponded to the four N-terminal residues of TIMP-2, a broad-spectrum protein inhibitor of MMPs. Scaffolds that are able to reproduce the functional topology of this motif were obtained by exhaustive screening of the Protein Data Bank (PDB) using STAMPS software (search for three-dimensional atom motifs in protein structures). Ten artificial protein binders were produced. The designed proteins bind catalytic sites of MMPs with affinities ranging from 450 nm to 450 µm prior to optimization. The crystal structure of one artificial binder in complex with the catalytic domain of MMP-12 showed that the inter-molecular interactions established by the functional motif in the artificial binder corresponded to those found in the MMP-14-TIMP-2 complex, albeit with some differences in geometry. Molecular dynamics simulations of the ten binders in complex with MMP-14 suggested that these scaffolds may allow partial reproduction of native inter-molecular interactions, but differences in geometry and stability may contribute to the lower affinity of the artificial protein binders compared to the natural protein binder. Nevertheless, these results show that the in silico design method used provides sets of protein binders that target a specific binding site with a good rate of success. This approach may constitute the first step of an efficient hybrid computational/experimental approach to protein binder design.


Subject(s)
Matrix Metalloproteinase Inhibitors/chemistry , Molecular Dynamics Simulation , Recombinant Fusion Proteins/chemistry , Amino Acid Motifs , Amino Acid Sequence , Catalytic Domain , Crystallography, X-Ray , Databases, Protein , Humans , Hydrogen Bonding , Matrix Metalloproteinase 12/chemistry , Matrix Metalloproteinase 14/chemistry , Matrix Metalloproteinase Inhibitors/chemical synthesis , Molecular Sequence Data , Protein Binding , Protein Engineering , Protein Stability , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/chemical synthesis , Tissue Inhibitor of Metalloproteinase-2/chemistry
14.
PLoS One ; 7(5): e36707, 2012.
Article in English | MEDLINE | ID: mdl-22693555

ABSTRACT

BACKGROUND: Frog metamorphosis is totally dependent on thyroid hormone (T3) and mimics the postembryonic period around birth in mammals. It is an excellent model to study the molecular basis of postembryonic development in vertebrate. We and others have shown that many, if not all, matrix metalloproteinases (MMPs), which cleave proteins of the extracellular matrix as well as other substrates, are induced by T3 and important for metamorphosis. MMP activity can be inhibited by tissue inhibitors of metalloproteinase (TIMPs). There are 4 TIMPs in vertebrates and their roles in postembryonic development are poorly studied. METHODOLOGY/PRINCIPAL FINDINGS: We analyzed the TIMP2 genes in Xenopus laevis and the highly related species Xenopus tropicalis and discovered that TIMP2 is a single copy gene in Xenopus tropicalis as in mammals but is duplicated in Xenopus laevis. Furthermore, the TIMP2 locus in Xenopus tropicalis genome is different from that in human, suggesting an evolutionary reorganization of the locus. More importantly, we found that the duplicated TIMP2 genes were similarly regulated in the developing limb, remodeling intestine, resorbing tail during metamorphosis. Unexpectedly, like its MMP target genes, the TIMP2 genes were upregulated by T3 during both natural and T3-induced metamorphosis. CONCLUSIONS/SIGNIFICANCE: Our results indicate that TIMP2 is highly conserved among vertebrates and that the TIMP2 locus underwent a chromosomal reorganization during evolution. Furthermore, the unexpected upregulation of TIMP2 genes during metamorphosis suggests that proper balance of MMP activity is important for metamorphosis.


Subject(s)
Matrix Metalloproteinases/metabolism , Tissue Inhibitor of Metalloproteinase-2/genetics , Tissue Inhibitor of Metalloproteinase-2/metabolism , Xenopus Proteins/genetics , Xenopus Proteins/metabolism , Xenopus laevis/growth & development , Xenopus laevis/metabolism , Amino Acid Sequence , Animals , Gene Duplication , Gene Expression Regulation, Developmental/drug effects , Genetic Loci/genetics , Humans , Metamorphosis, Biological/drug effects , Molecular Sequence Data , Organ Specificity , Tissue Inhibitor of Metalloproteinase-2/chemistry , Triiodothyronine/pharmacology , Xenopus Proteins/chemistry , Xenopus laevis/genetics
15.
J Biol Chem ; 287(19): 15935-46, 2012 May 04.
Article in English | MEDLINE | ID: mdl-22427646

ABSTRACT

Matrix metalloproteinase 10 (MMP-10, stromelysin-2) is a secreted metalloproteinase with functions in skeletal development, wound healing, and vascular remodeling; its overexpression is also implicated in lung tumorigenesis and tumor progression. To understand the regulation of MMP-10 by tissue inhibitors of metalloproteinases (TIMPs), we have assessed equilibrium inhibition constants (K(i)) of putative physiological inhibitors TIMP-1 and TIMP-2 for the active catalytic domain of human MMP-10 (MMP-10cd) using multiple kinetic approaches. We find that TIMP-1 inhibits the MMP-10cd with a K(i) of 1.1 × 10(-9) M; this interaction is 10-fold weaker than the inhibition of the similar MMP-3 (stromelysin-1) catalytic domain (MMP-3cd) by TIMP-1. TIMP-2 inhibits the MMP-10cd with a K(i) of 5.8 × 10(-9) M, which is again 10-fold weaker than the inhibition of MMP-3cd by this inhibitor (K(i) = 5.5 × 10(-10) M). We solved the x-ray crystal structure of TIMP-1 bound to the MMP-10cd at 1.9 Å resolution; the structure was solved by molecular replacement and refined with an R-factor of 0.215 (R(free) = 0.266). Comparing our structure of MMP-10cd·TIMP-1 with the previously solved structure of MMP-3cd·TIMP-1 (Protein Data Bank entry 1UEA), we see substantial differences at the binding interface that provide insight into the differential binding of stromelysin family members to TIMP-1. This structural information may ultimately assist in the design of more selective TIMP-based inhibitors tailored for specificity toward individual members of the stromelysin family, with potential therapeutic applications.


Subject(s)
Matrix Metalloproteinase 10/chemistry , Protein Structure, Tertiary , Tissue Inhibitor of Metalloproteinase-1/chemistry , Tissue Inhibitor of Metalloproteinase-2/chemistry , Binding Sites/genetics , Binding, Competitive , Catalytic Domain , Crystallography, X-Ray , HEK293 Cells , Humans , Kinetics , Matrix Metalloproteinase 10/genetics , Matrix Metalloproteinase 10/metabolism , Matrix Metalloproteinase 3/chemistry , Matrix Metalloproteinase 3/genetics , Matrix Metalloproteinase 3/metabolism , Models, Molecular , Mutation , Tissue Inhibitor of Metalloproteinase-1/genetics , Tissue Inhibitor of Metalloproteinase-1/metabolism , Tissue Inhibitor of Metalloproteinase-2/genetics , Tissue Inhibitor of Metalloproteinase-2/metabolism
16.
BMC Musculoskelet Disord ; 12: 273, 2011 Nov 28.
Article in English | MEDLINE | ID: mdl-22122951

ABSTRACT

BACKGROUND: Several symptom-relieving interventions have been shown to be efficacious among osteoarthritis (OA) patients with knee effusion; however, not every symptomatic knee OA patient has clinical effusion. Results may be over-generalized since it is unclear if effused knees represent a unique pathological condition or subset compared to knees without effusion. The primary purpose of this study was to determine if biochemical differences existed between OA knees with and without effusion. METHODS: The present cross-sectional study consisted of 22 volunteers (11 with knee effusion, 11 without knee effusion) with confirmed late-stage radiographic knee OA (Kellgren-Lawrence score ≥ 3). Synovial fluid samples were collected and analyzed using a custom multiplex enzyme-linked immunosorbent assay to determine eight specific biomarker concentrations (e.g., catabolic, anabolic). RESULTS: Matrix metalloproteinase (MMP)-3, tissue inhibitor of MMPs (TIMP)-1, TIMP-2, and interleukin-10 were significantly higher in the knees with effusion than in the knees without effusion. CONCLUSIONS: The biochemical differences that existed between knees with and without effusion provide support that OA subsets may exist, characterized by distinct biochemical characteristics and clinical findings (e.g., effusion).


Subject(s)
Osteoarthritis, Knee/metabolism , Synovial Fluid/chemistry , Synovial Fluid/physiology , Aged , Biomarkers/chemistry , Biomarkers/metabolism , Exudates and Transudates/chemistry , Exudates and Transudates/physiology , Female , Humans , Interleukin-10/chemistry , Interleukin-10/physiology , Male , Matrix Metalloproteinase 3/chemistry , Matrix Metalloproteinase 3/physiology , Middle Aged , Osteoarthritis, Knee/diagnostic imaging , Osteoarthritis, Knee/pathology , Pilot Projects , Radiography , Synovial Fluid/enzymology , Tissue Inhibitor of Metalloproteinase-1/chemistry , Tissue Inhibitor of Metalloproteinase-1/physiology , Tissue Inhibitor of Metalloproteinase-2/chemistry , Tissue Inhibitor of Metalloproteinase-2/physiology
17.
Peptides ; 32(9): 1840-8, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21871510

ABSTRACT

Tissue inhibitor of metalloproteinases-2 (TIMP-2) inhibits angiogenesis by several mechanisms involving either MMP inhibition or direct endothelial cell binding. The primary aim of this study was to identify the TIMP-2 region involved in binding to the previously identified receptor integrin α3ß1, and to determine whether synthetic peptides derived from this region retained angio-inhibitory and tumor suppressor activity. We demonstrated that the N-terminal domain of TIMP-2 (N-TIMP-2) binds to α3ß1 and inhibits vascular endothelial growth factor-stimulated endothelial cell growth in vitro, suggesting that both the α3ß1-binding domain and the growth suppressor activity of TIMP-2 localize to the N-terminal domain. Using a peptide array approach we identify a 24 amino acid region of TIMP-2 primary sequence, consisting of residues Ile43-Ala66, which shows α3ß1-binding activity. Subsequently we demonstrate that synthetic peptides from this region compete for TIMP-2 binding to α3ß1 and suppress endothelial growth in vitro. We define a minimal peptide sequence (peptide 8-9) that possesses both angio-inhibitory and, using a murine xenograft model of Kaposi's sarcoma, anti-tumorigenic activity in vivo. Thus, both the α3ß1-binding and the angio-inhibitory activities co-localize to a solvent exposed, flexible region in the TIMP-2 primary sequence that is unique in amino acid sequence compared with other members of the TIMP family. Furthermore, comparison of the TIMP-2 and TIMP-1 protein 3-D structures in this region also identified unique structural differences. Our findings demonstrate that the integrin binding, tumor growth suppressor and in vivo angio-inhibitory activities of TIMP-2 are intimately associated within a unique sequence/structural loop (B-C loop).


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Integrin alpha3beta1/metabolism , Tissue Inhibitor of Metalloproteinase-2/metabolism , Amino Acid Sequence , Animals , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Humans , Mice , Mice, Nude , Microarray Analysis , Molecular Sequence Data , Neovascularization, Pathologic/metabolism , Peptides/administration & dosage , Peptides/chemical synthesis , Peptides/pharmacology , Protein Binding , Sarcoma, Kaposi/pathology , Tissue Inhibitor of Metalloproteinase-1/chemistry , Tissue Inhibitor of Metalloproteinase-1/metabolism , Tissue Inhibitor of Metalloproteinase-2/chemistry , Vascular Endothelial Growth Factor A/pharmacology
18.
J Biol Chem ; 286(19): 16891-9, 2011 May 13.
Article in English | MEDLINE | ID: mdl-21454617

ABSTRACT

The avid binding of tissue inhibitors of metalloproteinases (TIMPs) to matrix metalloproteinases (MMPs) is crucial for the regulation of pericellular and extracellular proteolysis. The interactions of the catalytic domain (cd) of MMP-1 with the inhibitory domains of TIMP-1 and TIMP-2 (N-TIMPs) and MMP-3cd with N-TIMP-2 have been characterized by isothermal titration calorimetry and compared with published data for the N-TIMP-1/MMP-3cd interaction. All interactions are largely driven by increases in entropy but there are significant differences in the profiles for the interactions of both N-TIMPs with MMP-1cd as compared with MMP-3cd; the enthalpy change ranges from small for MMP-1cd to highly unfavorable for MMP-3cd (-0.1 ± 0.7 versus 6.0 ± 0.5 kcal mol(-1)). The heat capacity change (ΔC(p)) of binding to MMP-1cd (temperature dependence of ΔH) is large and negative (-210 ± 20 cal K(-1) mol(-1)), indicating a large hydrophobic contribution, whereas the ΔC(p) values for the binding to MMP-3cd are much smaller (-53 ± 3 cal K(-1) mol(-1)), and some of the entropy increase may arise from increased conformational entropy. Apart from differences in ionization effects, it appears that the properties of the MMP may have a predominant influence in the thermodynamic profiles for these N-TIMP/MMP interactions.


Subject(s)
Matrix Metalloproteinase 1/metabolism , Matrix Metalloproteinase 3/metabolism , Calorimetry/methods , Catalytic Domain , Entropy , Humans , Models, Biological , Molecular Conformation , Protease Inhibitors/pharmacology , Protein Interaction Mapping , Protein Structure, Tertiary , Temperature , Thermodynamics , Tissue Inhibitor of Metalloproteinase-1/chemistry , Tissue Inhibitor of Metalloproteinase-2/chemistry
19.
Mycoses ; 54(4): 325-30, 2011 Jul.
Article in English | MEDLINE | ID: mdl-20406392

ABSTRACT

Matrix metalloproteinase (MMP)-9 activity is controlled by the balance between MMP-9 and its major tissue inhibitor of metalloproteinases (TIMPs). We hypothesised whether Candida proteinases may affect local tissue inflammatory processes by modifying these molecules. The effects of sonicated cells and concentrated growth media of six Candida species on MMP-9, TIMP-1 and TIMP-2 were tested. Incubated samples were analysed by Western blot and detected by enhanced chemoluminescence techniques. The residual activity of degraded TIMP-1 was evaluated by a casein degradation assay. The proteinase activity of the microbial strains was also assessed by a fluorimetric assay, and the action of inhibitors on MMP-14 and Candida parapsilosis Cp2 was demonstrated. Cell fractions of both strains of C. parapsilosis exerted a weak ability to convert 92-kDa proMMP-9 to 86-kDa active form. Cell fractions of both strains of Candida albicans, C. parapsilosis Cp2, Candida glabrata reference strain, and both strains of Candida krusei fragmented TIMP-1 (28 kDa) to a 24-kDa species, which associated with reduced inhibitory activity on MMP-9 caseinolysis. Our findings indicate that Candida can participate in tissue inflammation by modifying the host's MMP-9 and their inhibitors. A rapid fluorimetric assay can be adapted for Candida proteinases.


Subject(s)
Candida/enzymology , Enzyme Precursors/metabolism , Matrix Metalloproteinase 9/metabolism , Peptide Hydrolases/metabolism , Tissue Inhibitor of Metalloproteinase-1/metabolism , Tissue Inhibitor of Metalloproteinase-2/metabolism , Virulence Factors/metabolism , Blotting, Western , Enzyme Precursors/chemistry , Fluorometry , Humans , Luminescent Measurements , Matrix Metalloproteinase 9/chemistry , Molecular Weight , Tissue Inhibitor of Metalloproteinase-1/chemistry , Tissue Inhibitor of Metalloproteinase-2/chemistry
20.
Int Immunol ; 22(8): 637-49, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20501611

ABSTRACT

Polymorphonuclear neutrophils (PMNs) are the first line of defense against invading organisms in humans; in addition, PMNs contribute to the linking of innate and adaptive immunity. To fulfill their biological behavior, PMNs utilize an arsenal of proteolytic enzymes, including members of the matrix metalloproteinase family of zinc-dependent endopeptidases. PMNs express high levels of MT6-MMP (MMP-25), a glycosyl-phosphatidylinositol-anchored MMP, that belongs to the subfamily of membrane-anchored matrix metalloproteinases. Due to the paucity of information on MT6-MMP in primary cells, we set to investigate the localization and potential function of MT6-MMP in human PMNs. We found that MT6-MMP is present in the membrane, granules and nuclear/endoplasmic reticulum/Golgi fractions of PMNs where it is displayed as a disulfide-linked homodimer of 120 kDa. Stimulation of PMNs resulted in secretion of active MT6-MMP into the supernatants. Membrane-bound MT6-MMP, conversely, is located in the lipid rafts of resting PMNs and stimulation does not alter this location. In addition, TIMP-2, a natural inhibitor of MT6-MMP, does not co-localize with it in the lipid rafts. Interestingly, living PMNs do not display MT6-MMP on the cell surface. However, induction of apoptosis induces MT6-MMP relocation on PMNs' cell surface. Our studies suggest that metalloproteinases may play a role in respiratory burst and IL-8 secretion, but not chemotaxis or granulocyte macrophage colony-stimulating factor-induced survival. Collectively, these results provide new insights on the role of MT6-MMP in the physiology of human PMNs.


Subject(s)
Apoptosis , Matrix Metalloproteinases, Membrane-Associated/metabolism , Membrane Microdomains/metabolism , Neutrophils/metabolism , Cells, Cultured , GPI-Linked Proteins/chemistry , GPI-Linked Proteins/metabolism , Humans , Immunoblotting , Matrix Metalloproteinases, Membrane-Associated/chemistry , Membrane Microdomains/chemistry , Microscopy, Confocal , Neutrophils/chemistry , Protein Transport , Tissue Inhibitor of Metalloproteinase-2/chemistry , Tissue Inhibitor of Metalloproteinase-2/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...