Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Discov Med ; 36(180): 71-81, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38273747

ABSTRACT

BACKGROUND: Bladder cancer is a malignant tumor of the urinary and reproductive tract that seriously threatens human health. It is urgent to develop new drugs for bladder cancer. This study aims to explore whether curcumin could inhibit bladder cancer and the potential mechanism. METHODS: Firstly, network pharmacology was applied to explore the potential target of curcumin in bladder cancer. Among the potential target of curcumin on bladder cancer, the role of matrix metalloproteinase-14 (MMP14) was further explored by bioinformatic analysis and the expression of MMP14 was confirmed by immunohistochemistry staining. The effect of curcumin on bladder cancer was then studied using the cell counting kit-8 (CCK-8) assay, clone formation assay, apoptosis assay, and Transwell assay. Finally, AKT, MMP14, E-cadherin and N-cadherin were analyzed by Western blot assay to confirm whether curcumin could inhibit bladder cancer by inhibiting invasion via AKT/MMP14 pathway. RESULTS: In the present study, we found that the target of curcumin for bladder cancer includes signal transducer and activator of transcription 3 (STAT3), AKT, cyclin A2 (CCNA2), epidermal growth factor receptor (EGFR), E1A binding protein p300 (EP300) and MMP14. MMP14 was highly expressed in bladder cancer than in normal tissues and was associated with a worse prognosis (p < 0.05). Curcumin could inhibit the proliferation and migration of bladder cancer cells (p < 0.05), while promoting cell apoptosis by inhibiting the AKT/MMP14 pathway (p < 0.05). CONCLUSION: Curcumin could inhibit bladder cancer by inhibiting invasion through the AKT/MMP14 pathway.


Subject(s)
Curcumin , Urinary Bladder Neoplasms , Humans , Curcumin/pharmacology , Curcumin/therapeutic use , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-akt/pharmacology , Signal Transduction , Cell Proliferation , Matrix Metalloproteinase 14/metabolism , Matrix Metalloproteinase 14/pharmacology , Cell Line, Tumor , Cell Movement , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/pathology , Apoptosis
2.
F S Sci ; 5(1): 80-91, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38043603

ABSTRACT

OBJECTIVES: To assess the effect of simvastatin on uterine leiomyoma growth and extracellular matrix (ECM) deposition. DESIGN: Laboratory analysis of human leiomyoma cell culture, xenograft in a mouse model, and patient tissue from a clinical trial. SETTING: Academic research center. PATIENT(S): Tissue culture from human leiomyoma tissue and surgical leiomyoma tissue sections from a placebo-controlled randomized clinical trial. INTERVENTION(S): Simvastatin treatment. MAIN OUTCOME MEASURE(S): Serum concentrations, xenograft volumes, and protein expression. RESULTS: Mice xenografted with 3-dimensional human leiomyoma cultures were divided as follows: 7 untreated controls; 12 treated with activated simvastatin at 10 mg/kg body weight; and 15 at 20 mg/kg body weight. Simvastatin was detected in the serum of mice injected at the highest dose. Xenograft volumes were significantly smaller (mean 53% smaller at the highest concentration). There was dissolution of compact ECM, decreased ECM formation, and lower collagen protein expression in xenografts. Membrane type 1 matrix metalloproteinase was increased in vitro and in vivo. Matrix metalloproteinase 2 and low-density lipoprotein receptor-related protein 1 were increased in vitro. CONCLUSIONS: Simvastatin exhibited antitumoral activity with ECM degradation and decreased leiomyoma tumor volume in vivo. Activation of the matrix metalloproteinase 2, membrane type 1 matrix metalloproteinase, and low-density lipoprotein receptor-related protein 1 pathway may explain these findings.


Subject(s)
Leiomyoma , Uterine Neoplasms , Female , Humans , Mice , Animals , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 2/pharmacology , Simvastatin/pharmacology , Simvastatin/metabolism , Simvastatin/therapeutic use , Matrix Metalloproteinase 14/metabolism , Matrix Metalloproteinase 14/pharmacology , Uterine Neoplasms/drug therapy , Uterine Neoplasms/metabolism , Uterine Neoplasms/pathology , Leiomyoma/drug therapy , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Body Weight , Lipoproteins, LDL/metabolism , Lipoproteins, LDL/pharmacology , Lipoproteins, LDL/therapeutic use
3.
Wei Sheng Yan Jiu ; 52(6): 871-876, 2023 Nov.
Article in Chinese | MEDLINE | ID: mdl-38115649

ABSTRACT

OBJECTIVE: To observe the effect of the ubiquitination process on the expression of CD44 antigen(CD44) and matrix metalloproteinase-14(MMP14) in human bronchial epithelial(16HBE) malignantly transformed cells induced by glycidyl methacrylate(GMA). METHODS: Successfully resuscitated 16HBE cells were cultured using a final concentration of 8 µg/mL GMA as the treatment group and 1 µg/mL dimethyl sulfoxide as the solvent control group, each time stained for 72 h, and then stained again after an interval of 24 h. After repeating the staining three times, the cells were cultured in passages respectively. The 40th generation(P40) GMA-treated group and the same-generation solvent control group were subjected to soft agar colony formation assay and concanavalin A(ConA) agglutination test to confirm that the 40th generation of GMA-induced malignant transformed 16HBE cells possessed malignant transformed cell characteristics.5, 10, 20, 40, 60 µmol/L anacardic acid were used to inhibit the ubiquitination process of GMA-induced malignant transformed 16HBE cells. The protein expression of CD44 and MMP14 were detected by western blotting, while the transcript levels of CD44, MMP14, and TFAP2A were assessed by real-time fluorescence quantitative PCR(qPCR). RESULTS: (1) In the soft agar colony formation assay, the number of clones formed by the cells in the solvent control group was 22, and the number of clones created by the malignantly transformed cells in the GMA-treated group was 208. In the ConA agglutination test, the cells in the solvent control group were uniformly dispersed in ConA solution, and no obvious agglutination occurred for 30 min, whereas the cells in the GMA-treated group were agglutinated in the 5th min, and the agglutinated cells were larger and more rapidly agglutinated. The agglomerates were more significant and faster, and the sensitivity of agglutination was increased. (2) After differential inhibition of GMA-induced ubiquitination in malignantly transformed 16HBE cells, the expression levels of CD44 and MMP14 were reduced in GMA-induced malignantly transformed 16HBE cells compared with the control group(P<0.05). The transcript levels of MMP14 and CD44 decreased with increasing inhibitor concentration(P<0.05), and the transcript levels of the upstream transcription factor TFAP2A were also simultaneously reduced(P<0.05). CONCLUSION: Inhibition of the cellular ubiquitination process mediates the down-regulation of protein expression and transcriptional expression of CD44 and MMP14 in GMA-induced malignantly transformed 16HBE cells.


Subject(s)
Epithelial Cells , Matrix Metalloproteinase 14 , Humans , Matrix Metalloproteinase 14/metabolism , Matrix Metalloproteinase 14/pharmacology , Agar/adverse effects , Agar/metabolism , Hyaluronan Receptors/metabolism , Ubiquitination , Solvents/adverse effects , Solvents/metabolism , Cell Transformation, Neoplastic/chemically induced
4.
Autoimmunity ; 56(1): 2281223, 2023 12.
Article in English | MEDLINE | ID: mdl-37964516

ABSTRACT

Airway remodeling is an important pathologic factor in the progression of asthma. Abnormal proliferation and migration of airway smooth muscle cells (ASMCs) are important pathologic mechanisms in severe asthma. In the current study, claudin-1 (CLDN1) was identified as an asthma-related gene and was upregulated in ASMCs stimulated with platelet-derived growth factor BB (PDGF-BB). Cell counting kit-8 and EdU assays were used to evaluate cell proliferation, and transwell assay was carried out to analyze cell migration and invasion. The levels of inflammatory factors were detected using enzyme-linked immunosorbent assay. The results showed that CLDN1 knockdown inhibited the proliferation, migration, invasion, and inflammation of ASMCs treated with PDGF-BB, whereas overexpression of CLDN1 exhibited the opposite effects. Protein-protein interaction assay and co-immunoprecipitation revealed that CLDN1 directly interacted with matrix metalloproteinase 14 (MMP14). CLDN1 positively regulated MMP14 expression in asthma, and MMP14 overexpression reversed cell proliferation, migration, invasion, and inflammation induced by silenced CLDN1. Taken together, CLDN1 promotes PDGF-BB-induced cell proliferation, migration, invasion, and inflammatory responses of ASMCs by upregulating MMP14 expression, suggesting a potential role for CLDN1 in airway remodeling in asthma.


Subject(s)
Asthma , Matrix Metalloproteinase 14 , Humans , Becaplermin/pharmacology , Becaplermin/metabolism , Claudin-1/genetics , Claudin-1/metabolism , Matrix Metalloproteinase 14/genetics , Matrix Metalloproteinase 14/metabolism , Matrix Metalloproteinase 14/pharmacology , Airway Remodeling/genetics , Cell Proliferation/genetics , Asthma/genetics , Asthma/metabolism , Myocytes, Smooth Muscle/metabolism , Inflammation/metabolism , Cell Movement/genetics , Cells, Cultured
5.
Environ Pollut ; 313: 120140, 2022 Nov 15.
Article in English | MEDLINE | ID: mdl-36100121

ABSTRACT

TCDD (2,3,7,8-tetrachlorodibenzo-p-dioxin) and BDE-209 (decabromodiphenyl ether) are persistent organic pollutants (POPs) produced by industrial activities and associated with several diseases. TCDD is a known human carcinogen, but few studies investigated about the effects of exposure to both compounds, i.e., whether BDE-209 and TCDD can render tumor cells more aggressive and metastatic. In the current study we investigated if the exposure of B16-F1 and B16-F10 melanoma murine cells to environmental relevant concentrations of TCDD and BDE-209 at 24 h and 15-day exposure modulates the expression of genes related to metastasis, making the cells more aggressive. Both pollutants did not affect cell viability but lead to increase of cell proliferation, including the upregulation of vimentin, MMP2, MMP9, MMP14 and PGK1 gene expression and downregulation of E-cadherin, TIMP2, TIMP3 and RECK, strongly suggesting changes in cell phenotypes defined as epithelial to mesenchymal transition (EMT) in BDE-209 and TCDD-exposed cells. Foremost, increased expression of metalloproteinases and decreased expression of their inhibitors made B16-F1 cells similar the more aggressive B16-F10 cell line. Also, the higher secretion of extracellular vesicles by cells after acute exposure to BDE-209 could be related with the phenotype changes. These results are a strong indication of the potential of BDE-209 and TCDD to modulate cell phenotype, leading to a more aggressive profile.


Subject(s)
Environmental Pollutants , Melanoma , Polychlorinated Dibenzodioxins , Animals , Cadherins , Carcinogens , Environmental Pollutants/pharmacology , Epithelial-Mesenchymal Transition , GPI-Linked Proteins , Halogenated Diphenyl Ethers , Humans , Matrix Metalloproteinase 14/pharmacology , Matrix Metalloproteinase 2/pharmacology , Matrix Metalloproteinase 9 , Mice , Persistent Organic Pollutants , Polychlorinated Dibenzodioxins/toxicity , Vimentin/pharmacology
6.
Phytomedicine ; 100: 154062, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35366491

ABSTRACT

BACKGROUND: The current standard therapy for metastatic pancreatic cancer is ineffective, necessitating a new treatment approach for prognosis improvement. The urokinase-plasmin activator (uPA) is a critical factor in epithelial-mesenchymal transition (EMT) and cancer metastasis, but its underlying mechanisms in pancreatic cancer remains elusive. METHODS: We investigated uPA expression in our pancreatic cancer cohort. A bioinformatics approach was used to further determine the role of uPA in pancreatic cancer. We employed MiaPaCa-2 and PANC-1 cell lines to investigate how uPA regulates EMT and metastasis in pancreatic cancer and present a novel approach aimed at inhibiting uPA in pancreatic cancer. RESULTS: We observed that higher uPA mRNA expression was significantly associated with overall-poor survival and progression-free survival in pancreatic cancer. uPA was highly expressed in tumor tissue. Gene set enrichment analysis revealed a positive association between uPA mRNA expression and EMT and transforming growth factor ß (TGF-ß) signaling pathways. Moreover, shRNA-mediated uPA gene knockdown reduced plasmin, MMP14, and TGF-ß activation, leading to the inhibition of PANC-1 cells' EMT marker expression, migration, invasion, and cell viability. Notably, 4-acetyl-antroquinonol B (4-AAQB) treatment suppressed MiaPaCa-2 and PANC-1 cell migratory and invasive abilities by inhibiting the uPA/MMP14/TGF-ß axis through upregulation of miR-181d-5p. In the xenograft mouse model of orthotropic pancreatic cancer, 4-AAQB treatment has reduced tumor growth and metastasis rate by deactivating uPA and improving the survival of the mice model. CONCLUSION: Accordingly, to extent of our knowledge and previous studies, we demonstrated that 4-AAQB is an anti Pan-Cancer drug, and may inhibit pancreatic cancer EMT and metastasis and serve as a new therapeutic approach for patients with late-stage pancreatic cancer.


Subject(s)
Pancreatic Neoplasms , Urokinase-Type Plasminogen Activator , Animals , Cell Line, Tumor , Epithelial-Mesenchymal Transition , Fibrinolysin/pharmacology , Humans , Matrix Metalloproteinase 14/pharmacology , Mice , Pancreatic Neoplasms/pathology , RNA, Messenger , Transforming Growth Factor beta/metabolism , Ubiquinone/analogs & derivatives , Urokinase-Type Plasminogen Activator/genetics , Pancreatic Neoplasms
7.
Invest Ophthalmol Vis Sci ; 60(6): 2321-2329, 2019 05 01.
Article in English | MEDLINE | ID: mdl-31117124

ABSTRACT

Purpose: Investigate the impact matrix metalloproteinase 14 (MMP14) delivered via exosomes produced by corneal fibroblasts on vascular endothelial growth factor receptor 1 (VEGFR1) cleavage on endothelial cells, and other key processes of angiogenesis. Methods: Proteolysis of VEGFR1 and R2 by the catalytic domain of MMP14 was investigated via immunocytochemistry with anti-VEGFR1, anti-VEGFR2, and anti-MMP14 antibodies. Exosomes were isolated via precipitation and serial ultracentrifugation from wild-type (WT) and MMP14 exon4-deficient corneal fibroblasts. Transmission electron microscopy and nanotracking analysis were used to characterize the isolated exosomes. The presence of MMP14 in exosomes from WT fibroblasts was confirmed by Western blotting. VEGFR1 cleavage upon treatment with WT-derived exosomes, Δexon4-derived exosomes, or the pan-MMP inhibitor GM60001 was examined via in vitro proteolysis analysis using recombinant mouse (rm) VEGFR1/R2. Endothelial cell migration and proliferation were investigated using a Boyden chamber assay and BrdU incorporation, respectively. Results: WT-derived exosomes specifically cleaved rmVEGFR1 in vitro, whereas Δexon4-derived exosomes did not. Treatment with the pan-MMP inhibitor GM6001 effectively inhibited VEGFR1 cleavage by WT-derived exosomes, confirming the role of MMP14 in this cleavage. WT-derived exosomes induced greater endothelial cell migration (P < 0.01) and proliferation (P < 0.5) compared to Δexon4-derived exosomes. Conclusions: MMP14-containing exosomes may be involved in the regulation of corneal neovascularization through degradation of VEGFR1 and VEGFA-induced endothelial cell proliferation and migration.


Subject(s)
Cell Movement/drug effects , Cell Proliferation/drug effects , Endothelial Cells/metabolism , Exosomes/physiology , Matrix Metalloproteinase 14/pharmacology , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-1/metabolism , Cornea/cytology , Humans , Immunohistochemistry
8.
FASEB J ; 33(2): 2910-2927, 2019 02.
Article in English | MEDLINE | ID: mdl-30332299

ABSTRACT

We previously demonstrated that membrane type 1 (MT1) matrix metalloproteinase (MMP) was up-regulated in the hippocampus of the model of transgenic mice bearing 5 familial mutations on human amyloid precursor protein (APP) and presenilin 1 of Alzheimer disease (AD), and that the proteinase increased the levels of amyloid ß peptide (Aß) and its APP C-terminal fragment of 99 aa in a heterologous cell system. Here we provide further evidence that MT1-MMP interacts with APP and promotes amyloidogenesis in a proteolytic-dependent manner in Swedish APP-expressing human embryonic kidney 293 (HEKswe) cells. MT1-MMP-mediated processing of APP releases a soluble APP fragment, sAPP95. This process partly requires the activation of endogenous MMP-2 but is independent of ß-site APP cleaving enzyme 1 (BACE-1) or α-secretase activities. In contrast, MT1-MMP-mediated increase of Aß levels involved BACE-1 activity and was inhibited by tissue inhibitor of MMP-2, a natural inhibitor of both MT1-MMP and MMP-2. Interestingly, near abolishment of basal Aß production upon BACE-1 inhibition was rescued by MT1-MMP, indicating that the latter could mimic ß-secretase-like activity. Moreover, MT1-MMP promoted APP/Aß localization in endosomes, where Aß production mainly occurs. These data unveil new mechanistic insights to support the proamyloidogenic role of MT1-MMP based on APP processing and trafficking, and reinforce the idea that this proteinase may become a new potential therapeutic target in AD.-Paumier, J.-M., Py, N. A., González, L. G., Bernard, A., Stephan, D., Louis, L., Checler, F., Khrestchatisky, M., Baranger, K., Rivera, S. Proamyloidogenic effects of membrane type 1 matrix metalloproteinase involve MMP-2 and BACE-1 activities, and the modulation of APP trafficking.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/metabolism , Amyloid/chemistry , Aspartic Acid Endopeptidases/metabolism , Gene Expression Regulation/drug effects , Matrix Metalloproteinase 14/pharmacology , Matrix Metalloproteinase 2/metabolism , Alzheimer Disease , Amyloid Precursor Protein Secretases/genetics , Amyloid beta-Protein Precursor/genetics , Animals , Aspartic Acid Endopeptidases/genetics , HEK293 Cells , Humans , Matrix Metalloproteinase 2/genetics , Mice , Mice, Transgenic , Protein Transport
9.
Biophys Chem ; 234: 34-41, 2018 03.
Article in English | MEDLINE | ID: mdl-29407769

ABSTRACT

Membrane type-1 matrix metalloproteinase (MT1-MMP) is a transmembrane MMP which triggers intracellular signaling and regulates extracellular matrix proteolysis, two functions that are critical for tumor-associated angiogenesis and inflammation. While green tea catechins, particularly epigallocatechin gallate (EGCG), are considered very effective in preventing MT1-MMP-mediated functions, lack of structure-function studies and evidence regarding their direct interaction with MT1-MMP-mediated biological activities remain. Here, we assessed the impact in both cellular and biophysical assays of four ungallated catechins along with their gallated counterparts on MT1-MMP-mediated functions and molecular binding partners. Concanavalin-A (ConA) was used to trigger MT1-MMP-mediated proMMP-2 activation, expression of MT1-MMP and of endoplasmic reticulum stress biomarker GRP78 in U87 glioblastoma cells. We found that ConA-mediated MT1-MMP induction was inhibited by EGCG and catechin gallate (CG), that GRP78 induction was inhibited by EGCG, CG, and gallocatechin gallate (GCG), whereas proMMP-2 activation was inhibited by EGCG and GCG. Surface plasmon resonance was used to assess direct interaction between catechins and MT1-MMP interactors. We found that gallated catechins interacted better than their ungallated analogs with MT1-MMP as well as with MT1-MMP binding partners MMP-2, TIMP-2, MTCBP-1 and LRP1-clusterIV. Overall, current structure-function evidence supports a role for the galloyl moiety in both direct and indirect interactions of green tea catechins with MT1-MMP-mediated oncogenic processes.


Subject(s)
Catechin/analogs & derivatives , Matrix Metalloproteinase 14/metabolism , Tea/chemistry , Carcinogenesis/drug effects , Catechin/metabolism , Catechin/pharmacology , Cell Line, Tumor , Concanavalin A/pharmacology , Endoplasmic Reticulum Chaperone BiP , Enzyme Precursors/antagonists & inhibitors , Gelatinases/antagonists & inhibitors , Glioblastoma/pathology , Heat-Shock Proteins/antagonists & inhibitors , Humans , Matrix Metalloproteinase 14/pharmacology , Matrix Metalloproteinase Inhibitors/pharmacology , Protein Binding , Structure-Activity Relationship
10.
Biochem J ; 474(22): 3719-3732, 2017 11 01.
Article in English | MEDLINE | ID: mdl-28972070

ABSTRACT

Syndecans (SDCs) are transmembrane proteoglycans that are involved in cell adhesion and cell communication. Specifically, SDC2 plays a key role in tumorigenesis, metastasis, and angiogenesis. Previously, we found that rat SDC2 is shed by matrix metalloproteinase-7 (MMP-7) in colon cancer cells. Here, we analyzed the susceptibility of rat SDC2 to various MMPs. We found that the rat SDC2 ectodomain (ECD) fused to the C-terminal Fc region, which was expressed in mammalian cells, was cleaved more efficiently by MMP-14 than MMP-7. Likewise, when anchored on the surface of HeLa cells, rat SDC2 was cleaved more efficiently by the treatment of MMP-14 than MMP-7 and was shed more readily by membrane-anchored MMP-14 than soluble MMP-14. Furthermore, MMP-14 cleaved recombinant SDC2-ECD expressed in Escherichia coli into multiple fragments. Using N-terminal amino acid sequencing and the top-down proteomics approach, we determined that the major cleavage sites were S88↓L89, T98↓M99, T100↓L101, D132↓P133, and N148↓L149 for rat SDC2-ECD and S55↓G56, S65↓P66, P75↓K76, N92↓I93 D122↓P123, and S138↓L139 for human SDC2-ECD. Finally, the rat and human SDC2-ECD lost the ability to suppress vascular endothelial growth factor-induced formation of capillary-like tubes by human umbilical vein endothelial cells following cleavage by MMP-14, but its major cleavage-site mutant of rat SDC2-ECD did not. These results suggest that MMP-14 is a novel enzyme responsible for degrading SDC2 and impairing its physiological roles including angiogenesis.


Subject(s)
Human Umbilical Vein Endothelial Cells/physiology , Matrix Metalloproteinase 14/metabolism , Matrix Metalloproteinase 14/pharmacology , Syndecan-2/biosynthesis , Vascular Endothelial Growth Factor A/pharmacology , Animals , COS Cells , Chlorocebus aethiops , Dose-Response Relationship, Drug , HeLa Cells , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Insecta , Rats
11.
Cornea ; 30 Suppl 1: S45-9, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21912230

ABSTRACT

PURPOSE: Decorin is a small chondroitin sulfate proteoglycan that inhibits vascular endothelial cell migration and tube formation. Membrane type 1-matrix metalloproteinase (MT1-MMP) has been shown to be an important angiogenic enzyme in the cornea. We evaluated the specific role of MT1-MMP in decorin cleavage in the cornea. METHODS: Western blotting was used to evaluate decorin degradation by MT1-MMP. Aortic ring tube formation assays were used to assay the inhibitory effect of decorin and the stimulatory effect of MT1-MMP on vascular endothelial cells in vitro. Corneal micropocket assays using basic fibroblast growth factor (bFGF) were used to assess changes in the levels of decorin and MT1-MMP. RESULTS: MT1-MMP cleaves decorin in a time- and concentration-dependent manner in vitro. MT1-MMP levels were upregulated after in vivo bFGF pellet implantation in the cornea, and decorin cleavage products were detected in bFGF-implanted corneas but not in normal corneas. MT1-MMP reduced the inhibitory effects of decorin on aortic ring tube formation in vitro. CONCLUSION: MT1-MMP may play an essential role in angiogenesis through proteolytic processing of decorin in the cornea.


Subject(s)
Cornea/metabolism , Corneal Neovascularization/metabolism , Decorin/metabolism , Matrix Metalloproteinase 14/pharmacology , Animals , Blotting, Western , Cells, Cultured , Endothelial Cells/drug effects , Fibroblast Growth Factor 2/pharmacology , Matrix Metalloproteinase 14/physiology , Mice , Mice, Inbred C57BL
12.
Cancer Res ; 70(14): 6093-103, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-20587521

ABSTRACT

Epidermal growth factor (EGF) receptors (ErbB) and EGF family members represent promising targets for cancer therapy. Heparin-binding EGF (HB-EGF) is a member of the EGF family and is an important target for therapy in some types of human cancers. Processing of HB-EGF by proprotein convertases, and successively, by ADAM family proteases, generates a soluble growth factor that requires heparin as a cofactor. Although heparin potentiates HB-EGF activity in vitro, it is not clear how the heparin-binding activity of HB-EGF is regulated. Here, we show that membrane type 1-matrix metalloproteinase (MT1-MMP; MMP14), a potent invasion-promoting protease, markedly enhances HB-EGF-dependent tumor formation in mice. MT1-MMP additionally cleaves HB-EGF and removes the NH(2)-terminal 20 amino acids that are important for binding heparin. Consequently, the processing of HB-EGF by MT1-MMP converts HB-EGF into a heparin-independent growth factor with enhanced mitogenic activity, and thereby, expression of both proteins costimulates tumor cell growth in vitro and in vivo. The ErbB family of receptors expressed in human gastric carcinoma cells play a role in mediating enhanced HB-EGF activity by MT1-MMP during invasive cell growth in collagen. Thus, we shed light on a new mechanism whereby HB-EGF activity is regulated that should be considered when designing HB-EGF-targeted cancer therapy.


Subject(s)
Intercellular Signaling Peptides and Proteins/metabolism , Matrix Metalloproteinase 14/metabolism , Amino Acid Sequence , Animals , Cell Growth Processes/drug effects , Cell Growth Processes/physiology , Cell Line , Cell Line, Tumor , Dogs , Fibroblasts/cytology , Fibroblasts/drug effects , Gene Knockdown Techniques , Heparin-binding EGF-like Growth Factor , Humans , Intercellular Signaling Peptides and Proteins/biosynthesis , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/pharmacology , Matrix Metalloproteinase 14/biosynthesis , Matrix Metalloproteinase 14/genetics , Matrix Metalloproteinase 14/pharmacology , Mice , Molecular Sequence Data , Neoplasm Invasiveness , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Rats , Rats, Inbred BUF , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Transfection
13.
J Periodontal Res ; 45(3): 353-60, 2010 Jun.
Article in English | MEDLINE | ID: mdl-19909398

ABSTRACT

BACKGROUND AND OBJECTIVE: Emdogain (EMD), consisting mostly of amelogenin, is used in periodontal therapy to regenerate lost connective tissue. Emdogain is applied onto periodontally affected root surfaces, where it becomes exposed to proteolytic enzymes. In this study, we aimed to find out whether gingival crevicular fluid or matrix metalloproteinases (MMPs) could degrade EMD, and whether this degradation has consequences for in vitro cell proliferation. MATERIAL AND METHODS: We studied the effects of 156 gingival crevicular fluid samples collected from subjects with different stages of periodontal disease and from healthy control subjects and the effects of MMP-1, -2, -8, -9, -13 and -14 on the degradation of EMD using EMD-embedded zymography. The effects of gingival crevicular fluid with or without EMD and the effects of amelogenin on the proliferation of cultured periodontal ligament fibroblasts were studied by cell proliferation enzyme-linked immunosorbent assay kit. RESULTS: Degradation of Emdogain induced by gingival crevicular fluid was greater in samples from all stages of periodontal diseases compared with healthy control samples. Of the MMPs studied, only MMP-2 and MMP-8 showed limited EMD-degrading activities. One hundred micrograms per millilitre of EMD increased proliferation of periodontal ligament fibroblasts on average by 24% (confidence interval 0.60-0.64) and at 200 microg/mL by 30% (confidence interval 0.62-0.68) compared with control fibroblasts (confidence interval 0.48-0.52). However, gingival crevicular fluid (10 microg/mL) together with 100 microg/mL EMD induced the proliferation only by 6% (confidence interval 0.51-0.55) and with 200 microg/mL EMD by 12% (confidence interval 0.54-0.58). Amelogenin at 200 microg/mL decreased the proliferation of periodontal ligament fibroblasts by 54% (confidence interval 0.22-0.25). CONCLUSION: We suggest that diseased gingival crevicular fluid containing various proteases leads to degradation of EMD and decreased proliferation of periodontal ligament fibroblasts.


Subject(s)
Dental Enamel Proteins/metabolism , Fibroblasts/drug effects , Gingival Crevicular Fluid/metabolism , Periodontal Ligament/drug effects , Adolescent , Adult , Aggressive Periodontitis/metabolism , Alveolar Bone Loss/metabolism , Amelogenin/metabolism , Amelogenin/pharmacology , Cell Culture Techniques , Cell Proliferation/drug effects , Chronic Periodontitis/metabolism , Dental Enamel Proteins/pharmacology , Female , Fibroblasts/cytology , Gingival Crevicular Fluid/enzymology , Gingival Hemorrhage/metabolism , Gingivitis/metabolism , Humans , Male , Matrix Metalloproteinase 1/pharmacology , Matrix Metalloproteinase 13/pharmacology , Matrix Metalloproteinase 14/pharmacology , Matrix Metalloproteinase 2/pharmacology , Matrix Metalloproteinase 8/pharmacology , Matrix Metalloproteinase 9/pharmacology , Middle Aged , Periodontal Attachment Loss/metabolism , Periodontal Diseases/metabolism , Periodontal Ligament/cytology , Periodontal Pocket/metabolism , Young Adult
14.
Acta Biomater ; 5(6): 1898-904, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19307161

ABSTRACT

A novel matrix metalloproteinase sequestering biomaterial (MI Theramer beads) restored the epithelial barrier in a double chamber in vitro test system after disruption by Cytochalasin D and the secretion of the metalloproteinase MMP-2. MI beads are chemically modified (hydroxamated) poly(methacrylic acid-co-methyl methacrylate). We are exploring the utility of this material in inflammatory bowel disease (IBD), in which one manifestation is a compromised intestinal epithelial barrier. In a first step towards this goal we incubated MI beads (or polymethyl methacrylate control beads) with Caco-2 epithelial cells and mesenchymal 3T3 fibroblasts on two sides of a Matrigel((R))-coated filter and used Cytochalasin D (Cyto D) to activate MMP-2 (secreted by the 3T3 cells), disrupt actin filaments of Caco-2 cells and render the epithelial barrier leaky, as measured by dextran fluorescein equilibration. Addition of MI beads to Cyto D-treated cells inhibited active MMP-2 and prevented equilibration of dextran fluorescein. This study is the first step in showing a potential benefit to local (as opposed to systemic) inhibition of metalloproteinases in IBD or other intestinal inflammatory diseases.


Subject(s)
Biocompatible Materials/administration & dosage , Cell Membrane Permeability/drug effects , Epithelial Cells/physiology , Matrix Metalloproteinase 14/chemistry , Matrix Metalloproteinase 14/pharmacology , Caco-2 Cells , Epithelial Cells/cytology , Epithelial Cells/drug effects , Humans
15.
Curr Eye Res ; 33(11): 954-62, 2008 Nov.
Article in English | MEDLINE | ID: mdl-19085378

ABSTRACT

PURPOSE: To determine the effect of keratocyte-derived MT1-MMP on calf pulmonary artery endothelial cell (CPAE) proliferation and migration. METHODS: Keratocyte lines were generated from MT1-MMP knockout (KO) and wild type (WT) mice. WT keratocytes were transfected with WT or mutant MT1-MMP DNAs (DeltaTC or E240A). The effect of keratocyte-conditioned media on CPAE proliferation and migration was assayed. RESULTS: KO keratocyte conditioned media resulted in the greatest increase of CPAE cell proliferation (190.5+/-6.0%; p<0.01). WT keratocyte conditioned media showed higher CPAE proliferation (155.4+/-3.6%) than WT/MT1-MMP-transfected keratocytes (119.7+/-2.2%; p<0.001). Migration assays confirmed these findings. CONCLUSIONS: Keratocyte-derived MT1-MMP has anti-angiogenic effects in CPAE cells.


Subject(s)
Angiogenesis Inhibitors/deficiency , Angiogenesis Inhibitors/metabolism , Cornea/enzymology , Endothelial Cells/physiology , Matrix Metalloproteinase 14/deficiency , Matrix Metalloproteinase 14/metabolism , Pulmonary Artery/cytology , Angiogenesis Inhibitors/genetics , Angiogenesis Inhibitors/pharmacology , Animals , Cattle , Cell Line, Transformed , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Cornea/cytology , Culture Media, Conditioned/pharmacology , Endothelial Cells/cytology , Matrix Metalloproteinase 14/genetics , Matrix Metalloproteinase 14/pharmacology , Mice , Mice, Knockout , Mutation , Transfection , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL