1
|
Xu J, Shi P, Yang L, Cui H. Basic mechanism of mobilizing cell movement during invasion of glioblastoma and target selection of targeted therapy. J Adv Res 2025:S2090-1232(25)00286-3. [PMID: 40345646 DOI: 10.1016/j.jare.2025.04.042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 04/09/2025] [Accepted: 04/27/2025] [Indexed: 05/11/2025] Open
Abstract
BACKGROUND Glioblastoma (GBM), also known as glioblastoma multiforme, is a rapidly growing and highly invasive malignant tumor. Due to the inability to clearly distinguish between glioblastoma and normal tissue, surgery cannot achieve safe resection, often leading to poor patient prognosis and inevitable tumor recurrence. According to previous studies, GBM invasion is related to intercellular adhesion, matrix degradation, extracellular matrix and its related adhesion molecules, as well as the molecular matrix of protein hydrolases in the microenvironment of GBM cells and stromal cells. AIM OF REVIEW The aim is to enhance our understanding of the molecular mechanisms underlying GBM invasion and to advance research on targeted therapies for inhibiting GBM invasion. KEY SCIENTIFIC CONCEPTS OF REVIEW This article describes the protein hydrolases that may affect GBM cell invasion, changes in the cytoskeleton during motility, and the regulatory mechanisms of intracellular signaling pathways in GBM invasion. In addition, we also explored the possibility of targeted therapy against invasion related molecules in GBM.
Collapse
Affiliation(s)
- Jie Xu
- Cancer Center, Medical Research Institute, Southwest University, Chongqing 400716, China; Jinfeng Laboratory, Chongqing 401329, China
| | - Pengfei Shi
- Cancer Center, Medical Research Institute, Southwest University, Chongqing 400716, China; Jinfeng Laboratory, Chongqing 401329, China.
| | - Liqun Yang
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400716, China.
| | - Hongjuan Cui
- Cancer Center, Medical Research Institute, Southwest University, Chongqing 400716, China; Jinfeng Laboratory, Chongqing 401329, China; State Key Laboratory of Resource Insects, Southwest University, Chongqing 400716, China.
| |
Collapse
|
2
|
Doddawad V, Shivananda S, Kalabharathi HL, Shetty A, Sowmya S, Sowmya HK. Matrix metalloproteinases in oral cancer: A catabolic activity on extracellular matrix components. BIOMEDICAL AND BIOTECHNOLOGY RESEARCH JOURNAL (BBRJ) 2023. [DOI: 10.4103/bbrj.bbrj_10_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/16/2023]
|
3
|
Remacle AG, Cieplak P, Nam DH, Shiryaev SA, Ge X, Strongin AY. Selective function-blocking monoclonal human antibody highlights the important role of membrane type-1 matrix metalloproteinase (MT1-MMP) in metastasis. Oncotarget 2018; 8:2781-2799. [PMID: 27835863 PMCID: PMC5356841 DOI: 10.18632/oncotarget.13157] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 10/13/2016] [Indexed: 02/05/2023] Open
Abstract
The invasion-promoting MT1-MMP is a cell surface-associated collagenase with a plethora of critical cellular functions. There is a consensus that MT1-MMP is a key protease in aberrant pericellular proteolysis in migrating cancer cells and, accordingly, a promising drug target. Because of high homology in the MMP family and a limited success in the design of selective small-molecule inhibitors, it became evident that the inhibitor specificity is required for selective and successful MT1-MMP therapies. Using the human Fab antibody library (over 1.25×109 individual variants) that exhibited the extended, 23-27 residue long, VH CDR-H3 segments, we isolated a panel of the inhibitory antibody fragments, from which the 3A2 Fab outperformed others as a specific and potent, low nanomolar range, inhibitor of MT1-MMP. Here, we report the in-depth characterization of the 3A2 antibody. Our multiple in vitro and cell-based tests and assays, and extensive structural modeling of the antibody/protease interactions suggest that the antibody epitope involves the residues proximal to the protease catalytic site and that, in contrast with tissue inhibitor-2 of MMPs (TIMP-2), the 3A2 Fab inactivates the protease functionality by binding to the catalytic domain outside the active site cavity. In agreement with the studies in metastasis by others, our animal studies in acute pulmonary melanoma metastasis support a key role of MT1-MMP in metastatic process. Conversely, the selective anti-MT1-MMP monotherapy significantly alleviated melanoma metastatic burden. It is likely that further affinity maturation of the 3A2 Fab will result in the lead inhibitor and a proof-of-concept for MT1-MMP targeting in metastatic cancers.
Collapse
Affiliation(s)
- Albert G Remacle
- Infectious and Inflammatory Disease Center/Cancer Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Piotr Cieplak
- Infectious and Inflammatory Disease Center/Cancer Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Dong Hyun Nam
- Department of Chemical and Environmental Engineering, University of California, Riverside, Riverside, CA 92512, USA
| | - Sergey A Shiryaev
- Infectious and Inflammatory Disease Center/Cancer Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Xin Ge
- Department of Chemical and Environmental Engineering, University of California, Riverside, Riverside, CA 92512, USA
| | - Alex Y Strongin
- Infectious and Inflammatory Disease Center/Cancer Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| |
Collapse
|
4
|
Park HS, Quan KT, Han JH, Jung SH, Lee DH, Jo E, Lim TW, Heo KS, Na M, Myung CS. Rubiarbonone C inhibits platelet-derived growth factor-induced proliferation and migration of vascular smooth muscle cells through the focal adhesion kinase, MAPK and STAT3 Tyr 705 signalling pathways. Br J Pharmacol 2017; 174:4140-4154. [PMID: 28832962 DOI: 10.1111/bph.13986] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 08/07/2017] [Accepted: 08/07/2017] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND AND PURPOSE The proliferation and migration of vascular smooth muscle cells (VSMCs) induced by platelet-derived growth factor (PDGF) are important steps in cardiovascular diseases, including neointimal lesion formation, myocardial infarction and atherosclerosis. Here, we evaluated the rubiarbonone C-mediated signalling pathways that regulate PDGF-induced VSMC proliferation and migration. EXPERIMENTAL APPROACH Cell proliferation and migration were measured in cells treated with rubiarbonone C followed by PDGF BB using the MTT assay, [3 H]-thymidine incorporation, flow cytometry and wound-healing migration assay, MMP gelatin zymography, a fluorescence assay for F-actin. Western blotting of molecules including MAPK, focal adhesion kinase (FAK) and STAT3 and an immunofluorescence assay using anti-PCNA and -STAT3 antibodies were performed to evaluate rubiarbonone C signalling pathway(s). The medial thickness of the carotid artery was evaluated using a mouse carotid ligation model. KEY RESULTS Rubiarbonone C inhibited PDGF-induced VSMC proliferation and migration and diminished the ligation-induced increase in medial thickness of the carotid artery. In PDGF-stimulated VSMCs rubiarbonone C decreased the following: (i) levels of cyclin-dependent kinases, cyclins, PCNA and hyperphosphorylated retinoblastoma protein; (ii) levels and activity of MMP2 and MMP9; (iii) activation of MAPK; (iv) F-actin reorganization, by reducing FAK activation; (v) activation of STAT3. CONCLUSIONS AND IMPLICATIONS These findings suggest that rubiarbonone C inhibits the proliferation and migration of VSMCs by inhibiting the FAK, MAPK and STAT3 signalling pathways. Therefore, rubiarbonone C could be a good candidate for the treatment of cardiovascular disease.
Collapse
Affiliation(s)
- Hyun-Soo Park
- Department of Pharmacology, Chungnam National University College of Pharmacy, Daejeon, Korea
| | - Khong Trong Quan
- Department of Pharmacognosy, Chungnam National University College of Pharmacy, Daejeon, Korea.,Department of Pharmaceutical Analysis and Standardization, National Institute of Medicinal Materials, Hanoi, Vietnam
| | - Joo-Hui Han
- Department of Pharmacology, Chungnam National University College of Pharmacy, Daejeon, Korea
| | - Sang-Hyuk Jung
- Department of Pharmacology, Chungnam National University College of Pharmacy, Daejeon, Korea
| | - Do-Hyung Lee
- Department of Pharmacology, Chungnam National University College of Pharmacy, Daejeon, Korea
| | - Eunji Jo
- Department of Pharmacology, Chungnam National University College of Pharmacy, Daejeon, Korea
| | - Tae-Wan Lim
- Department of Pharmacology, Chungnam National University College of Pharmacy, Daejeon, Korea
| | - Kyung-Sun Heo
- Department of Pharmacology, Chungnam National University College of Pharmacy, Daejeon, Korea
| | - MinKyun Na
- Department of Pharmacognosy, Chungnam National University College of Pharmacy, Daejeon, Korea.,Institute of Drug Research and Development, Chungnam National University, Daejeon, Korea
| | - Chang-Seon Myung
- Department of Pharmacology, Chungnam National University College of Pharmacy, Daejeon, Korea.,Institute of Drug Research and Development, Chungnam National University, Daejeon, Korea
| |
Collapse
|
5
|
Ohnishi YI, Iwatsuki K, Ishihara M, Ohkawa T, Kinoshita M, Shinzawa K, Fujimoto Y, Yoshimine T. Promotion of astrocytoma cell invasion by micro RNA–22 targeting of tissue inhibitor of matrix metalloproteinase–2. J Neurosurg Spine 2017; 26:396-403. [DOI: 10.3171/2016.8.spine16248] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECTIVE
Diffuse astrocytomas (DAs) have a high recurrence rate due to diffuse infiltration into the brain and spinal cord. Micro RNAs (miRNAs) are small noncoding RNAs that regulate gene expression by binding to complementary sequences of target messenger RNA (mRNA). It has been reported that miRNA-22 (miR-22) is involved in the invasion of some cancer cell lines. The aim of this study was to identify the biological effects of miR-22 in regard to the invasion of human DAs.
METHODS
The authors evaluated whether the level of miR-22 is elevated in human spinal DAs by using miRNA chips. Next, the role of miR-22 in 1321N1 human astrocytoma cells was investigated. Finally, to elucidate whether miR-22 promotes invasion by astrocytoma cells in vivo, the authors transplanted miR-22 overexpressed astrocytoma cells into mouse thoracic spinal cord.
RESULTS
The miR-22 significantly upregulated the invasion capacity of 1321N1 cells. Computational in silico analysis predicted that tissue inhibitor of matrix metalloproteinase–2 (TIMP2) is a target gene of miR-22. This was confirmed by quantitative reverse transcription polymerase chain reaction and Western blotting, which showed that miR-22 inhibited TIMP2 mRNA and protein expression, respectively. Luciferase reporter assays demonstrated that miR-22 directly bound the 3′-untranslated regions of TIMP2. The authors further showed that miR-22 promoted invasiveness in 1321N1 astrocytoma cells when transplanted into mouse spinal cord.
CONCLUSIONS
These data suggest that miR-22 acts to regulate invasion of 1321N1 astrocytoma cells by targeting TIMP2 expression. Additional studies with more cases and cell lines are required to elucidate the findings of this study for a novel treatment target for spinal DAs.
Collapse
Affiliation(s)
| | | | | | - Toshika Ohkawa
- 2Department of Neurosurgery, Yao Municipal Hospital; and
| | - Manabu Kinoshita
- 3Department of Neurosurgery, Osaka Medical Center for Cancer and Cardiovascular Diseases, Osaka, Japan
| | - Koei Shinzawa
- 4Molecular Genetics, Osaka University Medical School
| | | | | |
Collapse
|
6
|
Chang JH, Huang YH, Cunningham CM, Han KY, Chang M, Seiki M, Zhou Z, Azar DT. Matrix metalloproteinase 14 modulates signal transduction and angiogenesis in the cornea. Surv Ophthalmol 2015; 61:478-97. [PMID: 26647161 DOI: 10.1016/j.survophthal.2015.11.006] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2015] [Revised: 11/12/2015] [Accepted: 11/23/2015] [Indexed: 11/16/2022]
Abstract
The cornea is transparent and avascular, and retention of these characteristics is critical to maintaining vision clarity. Under normal conditions, wound healing in response to corneal injury occurs without the formation of new blood vessels; however, neovascularization may be induced during corneal wound healing when the balance between proangiogenic and antiangiogenic mediators is disrupted to favor angiogenesis. Matrix metalloproteinases (MMPs), which are key factors in extracellular matrix remodeling and angiogenesis, contribute to the maintenance of this balance, and in pathologic instances, can contribute to its disruption. Here, we elaborate on the facilitative role of MMPs, specifically MMP-14, in corneal neovascularization. MMP-14 is a transmembrane MMP that is critically involved in extracellular matrix proteolysis, exosome transport, and cellular migration and invasion, processes that are critical for angiogenesis. To aid in developing efficacious therapies that promote healing without neovascularization, it is important to understand and further investigate the complex pathways related to MMP-14 signaling, which can also involve vascular endothelial growth factor, basic fibroblast growth factor, Wnt/β-catenin, transforming growth factor, platelet-derived growth factor, hepatocyte growth factor or chemokines, epidermal growth factor, prostaglandin E2, thrombin, integrins, Notch, Toll-like receptors, PI3k/Akt, Src, RhoA/RhoA kinase, and extracellular signal-related kinase. The involvement and potential contribution of these signaling molecules or proteins in neovascularization are the focus of the present review.
Collapse
Affiliation(s)
- Jin-Hong Chang
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Yu-Hui Huang
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Christy M Cunningham
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Kyu-Yeon Han
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Michael Chang
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Motoharu Seiki
- Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Zhongjun Zhou
- Department of Biochemistry, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Dimitri T Azar
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, University of Illinois at Chicago, Chicago, Illinois, USA.
| |
Collapse
|
7
|
XIA DONG, FENG LIBO, WU XIAOLONG, XIA GUODONG, XU LIANG. Microencapsulation of recombinant adenovirus within poly-DL-lactide-poly(ethylene glycol) microspheres for enhanced gene transfection efficiency and inhibitory effects on hepatocellular carcinoma cells in vitro. Mol Med Rep 2015; 12:2336-42. [DOI: 10.3892/mmr.2015.3578] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2013] [Accepted: 06/17/2014] [Indexed: 11/06/2022] Open
|
8
|
Aoki M, Miyake K, Ogawa R, Dohi T, Akaishi S, Hyakusoku H, Shimada T. siRNA Knockdown of Tissue Inhibitor of Metalloproteinase-1 in Keloid Fibroblasts Leads to Degradation of Collagen Type I. J Invest Dermatol 2014; 134:818-826. [DOI: 10.1038/jid.2013.396] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2013] [Revised: 08/19/2013] [Accepted: 09/03/2013] [Indexed: 12/19/2022]
|
9
|
Remacle AG, Shiryaev SA, Golubkov VS, Freskos JN, Brown MA, Karwa AS, Naik AD, Howard CP, Sympson CJ, Strongin AY. Non-destructive and selective imaging of the functionally active, pro-invasive membrane type-1 matrix metalloproteinase (MT1-MMP) enzyme in cancer cells. J Biol Chem 2013; 288:20568-80. [PMID: 23733191 DOI: 10.1074/jbc.m113.471508] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Proteolytic activity of cell surface-associated MT1-matrix metalloproteinase (MMP) (MMP-14) is directly related to cell migration, invasion, and metastasis. MT1-MMP is regulated as a proteinase by activation and conversion of the latent proenzyme into the active enzyme, and also via inhibition by tissue inhibitors of MMPs (TIMPs) and self-proteolysis. MT1-MMP is also regulated as a membrane protein through its internalization and recycling. Routine immunohistochemistry, flow cytometry, reverse transcription-PCR, and immunoblotting methodologies do not allow quantitative imaging and assessment of the cell-surface levels of the active, TIMP-free MT1-MMP enzyme. Here, we developed a fluorescent reporter prototype that targets the cellular active MT1-MMP enzyme alone. The reporter (MP-3653) represents a liposome tagged with a fluorochrome and functionalized with a PEG chain spacer linked to an inhibitory hydroxamate warhead. Our studies using the MP-3653 reporter and its inactive derivative demonstrated that MP-3653 can be efficiently used not only to visualize the trafficking of MT1-MMP through the cell compartment, but also to quantify the femtomolar range amounts of the cell surface-associated active MT1-MMP enzyme in multiple cancer cell types, including breast carcinoma, fibrosarcoma, and melanoma. Thus, the levels of the naturally expressed, fully functional, active cellular MT1-MMP enzyme are roughly equal to 1 × 10(5) molecules/cell, whereas these levels are in a 1 × 10(6) range in the cells with the enforced MT1-MMP expression. We suggest that the reporter we developed will contribute to the laboratory studies of MT1-MMP and then, ultimately, to the design of novel, more efficient prognostic approaches and personalized cancer therapies.
Collapse
Affiliation(s)
- Albert G Remacle
- Sanford-Burnham Medical Research Institute, La Jolla, California 92037, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Abstract
The peritoneal metastatic route of cancer dissemination is shared by cancers of the ovary and gastrointestinal tract. Once initiated, peritoneal metastasis typically proceeds rapidly in a feed-forward manner. Several factors contribute to this efficient progression. In peritoneal metastasis, cancer cells exfoliate into the peritoneal fluid and spread locally, transported by peritoneal fluid. Inflammatory cytokines released by tumor and immune cells compromise the protective, anti-adhesive mesothelial cell layer that lines the peritoneal cavity, exposing the underlying extracellular matrix to which cancer cells readily attach. The peritoneum is further rendered receptive to metastatic implantation and growth by myofibroblastic cell behaviors also stimulated by inflammatory cytokines. Individual cancer cells suspended in peritoneal fluid can aggregate to form multicellular spheroids. This cellular arrangement imparts resistance to anoikis, apoptosis, and chemotherapeutics. Emerging evidence indicates that compact spheroid formation is preferentially accomplished by cancer cells with high invasive capacity and contractile behaviors. This review focuses on the pathological alterations to the peritoneum and the properties of cancer cells that in combination drive peritoneal metastasis.
Collapse
|
11
|
Jung YS, Liu XW, Chirco R, Warner RB, Fridman R, Kim HRC. TIMP-1 induces an EMT-like phenotypic conversion in MDCK cells independent of its MMP-inhibitory domain. PLoS One 2012; 7:e38773. [PMID: 22701711 PMCID: PMC3372473 DOI: 10.1371/journal.pone.0038773] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2011] [Accepted: 05/12/2012] [Indexed: 12/16/2022] Open
Abstract
Matrix metalloproteinases (MMPs) and their endogenous inhibitors (TIMPs) regulate epithelial-mesenchymal transition (EMT) critical for the development of epithelial organs as well as cancer cell invasion. TIMP-1 is frequently overexpressed in several types of human cancers and serves as a prognostic marker. The present study investigates the roles of TIMP-1 on the EMT process and formation of the lumen-like structure in a 3D Matrigel culture of MDCK cells. We show that TIMP-1 overexpression effectively prevents cell polarization and acinar-like structure formation. TIMP-1 induces expression of the developmental EMT transcription factors such as SLUG, TWIST, ZEB1 and ZEB2, leading to downregulation of epithelial marker and upregulation of mesenchymal markers. Importantly, TIMP-1's ability to induce the EMT-like process is independent of its MMP-inhibitory domain. To our surprise, TIMP-1 induces migratory and invasive properties in MDCK cells. Here, we present a novel finding that TIMP-1 signaling upregulates MT1-MMP and MMP-2 expression, and potentiates MT1-MMP activation of pro-MMP-2, contributing to tumor cell invasion. In spite of the fact that TIMP-1, as opposed to TIMP-2, does not interact with and inhibit MT1-MMP, TIMP-1 may act as a key regulator of MT1-MMP/MMP-2 axis. Collectively, our findings suggest a model in which TIMP-1 functions as a signaling molecule and also as an endogenous inhibitor of MMPs. This concept represents a paradigm shift in the current view of TIMP-1/MT1-MMP interactions and functions during cancer development/progression.
Collapse
Affiliation(s)
- Young Suk Jung
- Department of Pathology, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Xu-Wen Liu
- Department of Pathology, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Rosemarie Chirco
- Department of Pathology, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Richard B. Warner
- Department of Pathology, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Rafael Fridman
- Department of Pathology, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Hyeong-Reh Choi Kim
- Department of Pathology, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan, United States of America
- * E-mail:
| |
Collapse
|
12
|
Matrix metalloproteinases and their tissue inhibitors in hypertension-related pregnancy complications. J Hum Hypertens 2012; 27:72-8. [DOI: 10.1038/jhh.2012.8] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
13
|
Díaz N, Suárez D. Alternative interdomain configurations of the full-length MMP-2 enzyme explored by molecular dynamics simulations. J Phys Chem B 2012; 116:2677-86. [PMID: 22324833 DOI: 10.1021/jp211088d] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Conformational freedom between the different domains of the matrix metalloproteinase family of enzymes has been repeatedly invoked to explain the mechanism of hydrolysis of some of their most complex macromolecular substrates. This proposed interdomain motion has been experimentally confirmed to occur in solution for matrix metalloproteinases MMP-1, MMP-9, and MMP-12. In this work, we computationally assess the likely conformational freedom in aqueous solution of the full-length form of the MMP-2 enzyme in the absence of its pro-peptide domain. To this end, we perform molecular dynamics (MD) simulations and approximate free energy analyses in four different arrangements of the protein domains that correspond to (a) the compact conformation observed in the X-ray structure; (b) an initially elongated structure in which the hemopexin (HPX) domain is separated from the catalytic (CAT) and fibronectin domains; and (c-d) two alternative conformations suggested by protein-protein docking calculations. Overall, our results indicate that the interdomain flexibility is very likely a general property of the MMP-2 enzyme in solution.
Collapse
Affiliation(s)
- Natalia Díaz
- Departamento de Química Física y Analítica, Julián Clavería 8, Universidad de Oviedo, Oviedo (Asturias), 33006 Spain.
| | | |
Collapse
|