Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
J Cancer Res Clin Oncol ; 149(10): 7235-7246, 2023 Aug.
Article in English | MEDLINE | ID: mdl-36905423

ABSTRACT

PURPOSE: This study aims to evaluate the value of tissue inhibitors of MMPs-2 (TIMP-2) to indicate 5-Fluorouracil (5-Fu) resistance status in colorectal cancer. METHODS: The 5-Fu resistance of colorectal cancer cell lines was detected using Cell-Counting Kit-8 (CCK-8) and calculated using IC50. Enzyme-linked immunosorbent assay (ELISA) and real time-quantitative polymerase chain reaction (RT-qPCR) were used to detect TIMP-2 expression level in the culture supernatant and serum. Twenty-two colorectal cancer patients' TIMP-2 levels and clinical characteristics were analyzed before and after chemotherapy. Additionally, the patient-derived xenograft (PDX) model of 5-Fu resistance was used to evaluate the feasibility of TIMP-2 as a predictive biomarker of 5-Fu resistance. RESULTS: Our experimental results display that TIMP-2 expression is elevated in colorectal cancer drug-resistant cell lines, and its expression level is closely related to 5-Fu resistance. Moreover, TIMP-2 in colorectal cancer patient serum undergoing 5-Fu-based chemotherapy could indicate their drug resistance status, and its efficacy is higher than CEA and CA19-9. Finally, PDX model animal experiments reveal that TIMP-2 can detect 5-Fu resistance in colorectal cancer earlier than tumor volume. CONCLUSION: TIMP-2 is a good indicator of 5-Fu resistance in colorectal cancer. Monitoring the serum TIMP-2 level can help the clinician identify 5-Fu resistance in colorectal cancer patients earlier during chemotherapy.


Subject(s)
Antimetabolites, Antineoplastic , Colorectal Neoplasms , Tissue Inhibitor of Metalloproteinase-2 , Animals , Humans , Antimetabolites, Antineoplastic/pharmacology , Antimetabolites, Antineoplastic/therapeutic use , Biomarkers , Cell Line, Tumor , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Drug Resistance, Neoplasm , Fluorouracil/pharmacology , Fluorouracil/therapeutic use , Tissue Inhibitor of Metalloproteinase-2/therapeutic use
2.
Cartilage ; 13(3): 19476035221115541, 2022.
Article in English | MEDLINE | ID: mdl-35932105

ABSTRACT

OBJECTIVE: The potential chondroprotective effect of celecoxib, a nonsteroidal anti-inflammatory drug and selective cyclooxygenase-2 inhibitor used to reduce pain and inflammation in knee osteoarthritis patients, is disputed. This study aimed at investigating the chondroprotective effects of celecoxib on (1) human articular cartilage explants and (2) in an in vivo osteoarthritis rat model. DESIGN: Articular cartilage explants from 16 osteoarthritis patients were cultured for 24 hours with celecoxib or vehicle. Secreted prostaglandins (prostaglandin E2, prostaglandin F2α, prostaglandin D2) and thromboxane B2 (TXB2) concentrations were determined in medium by ELISA, and protein regulation was measured with label-free proteomics. Cartilage samples from 7 of these patients were analyzed for gene expression using real-time quantitative polymerase chain reaction. To investigate the chondroprotective effect of celecoxib in vivo, 14 rats received an intra-articular injection of celecoxib or 0.9% NaCl after osteoarthritis induction by anterior cruciate ligament transection and partial medial meniscectomy (ACLT/pMMx model). Histopathological scoring was used to evaluate osteoarthritis severity 12 weeks after injection. RESULTS: Secretion of prostaglandins, target of Nesh-SH3 (ABI3BP), and osteonectin proteins decreased, whereas tissue inhibitor of metalloproteinase 2 (TIMP-2) increased significantly after celecoxib treatment in the human (ex vivo) explant culture. Gene expression of a disintegrin and metalloproteinase with thrombospondin motifs 4 and 5 (ADAMTS4/5) and metalloproteinase 13 (MMP13) was significantly reduced after celecoxib treatment in human cartilage explants. Cartilage degeneration was reduced significantly in an in vivo osteoarthritis knee rat model. CONCLUSIONS: Our data demonstrated that celecoxib acts chondroprotective on cartilage ex vivo and a single intra-articular bolus injection has a chondroprotective effect in vivo.


Subject(s)
Cartilage, Articular , Osteoarthritis, Knee , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cartilage, Articular/pathology , Celecoxib/metabolism , Celecoxib/pharmacology , Celecoxib/therapeutic use , Cyclooxygenase 2 Inhibitors/pharmacology , Cyclooxygenase 2 Inhibitors/therapeutic use , Humans , Metalloproteases/metabolism , Metalloproteases/pharmacology , Metalloproteases/therapeutic use , Osteoarthritis, Knee/pathology , Prostaglandins/metabolism , Prostaglandins/pharmacology , Prostaglandins/therapeutic use , Rats , Tissue Inhibitor of Metalloproteinase-2/metabolism , Tissue Inhibitor of Metalloproteinase-2/pharmacology , Tissue Inhibitor of Metalloproteinase-2/therapeutic use
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.
Nutr. hosp ; 34(3): 702-709, mayo-jun. 2017. tab, graf, ilus
Article in English | IBECS | ID: ibc-164130

ABSTRACT

Background: The intestinal dysbiosis is common in chronic liver disease and can induce to inflammatory responses and mediate the collagen deposition in the liver. Aim: To evaluate the probiotic Lactobacillus rhamnosus GG (LGG) for the treatment of liver fibrosis in a model of chronic cholestatic liver disease in rats. Methods: Male adult Wistar rats (n = 29) were submitted to common bile duct ligation (BDL groups) or manipulation of common bile duct without ligation (Ctrl groups).Two weeks after surgery, each group was randomly divided to receive 1 ml of PBS (Ctrl and BDL) or PBS containing 2.5 x 107 CFU of LGG (Ctrl-P and BDL-P) through gavages for 14 days. Euthanasia occurred 33 days after surgery when samples of blood and liver tissue were collected. Results: The hepatic gene expression of Tlr4, Tnfα, IL-6, Tgfβ, and metalloproteinase-2 and -9 were higher in the BDL groups in comparison to Ctrl. The ductular reaction evaluated by immunocontent of cytokeratin-7 (CK7) and the content of collagen were increased in BDL groups. Also, there was an imbalance in the antioxidant defenses (superoxide dismutase and catalase) and an increase in the oxidative stress marker sulfhydryl in BDL groups. The treatment with LGG significantly reduced gene expression of IL-6, collagen deposition, and ductular reaction in hepatic tissue of animals from BDL-P groups. Conclusion: The treatment with the probiotic LGG was able to reduce liver fibrosis, ductular reaction, and hepatic gene expression of IL-6 in a model of cholestatic liver disease in rats (AU)


Introducción: la disbiosis intestinal es común en la enfermedad hepática crónica y puede inducir respuestas inflamatorias y mediar la deposición hepática de colágeno. Objetivo: evaluar el efecto del probiótico Lactobacillus rhamnosus GG (LGG) en el tratamiento de la fibrosis hepática en un modelo de enfermedad hepática colestásica en ratas. Métodos: se sometió a ratas Wistar macho adultas (n = 29) a ligadura del conducto biliar común (grupo BDL) o a manipulación del conducto biliar sin ligadura (grupo Ctrl). Dos semanas después, cada grupo se dividió aleatoriamente para recibir 1 ml de PBS (Ctrl y BDL) o PBS con 2,5 x 107 UFC de LGG (Ctrl-P y BDL-P) durante 14 días. Se aplicó la eutanasia 33 días después de la cirugía y se recogieron muestras de sangre y de tejido hepático. Resultados: las expresiones hepáticas de Tlr4, Tnfα, IL-6, Tgfβ, metaloproteinasa-2 y -9 fueron mayores en los grupos BDL. La reacción ductular evaluada por el inmunocontenido de citoqueratina 7 (CK7) y el contenido de colágeno se aumentó en los grupos BDL. Además, hubo un desequilibrio en las defensas antioxidantes (superóxido dismutasa y catalasa) y un aumento en el estrés oxidativo (sulfhidrilo) en los grupos BDL. El tratamiento con LGG redujo la expresión génica de IL-6, la deposición de colágeno y la reacción ductular en el hígado de los animales del grupo BDL-P. Conclusión: el tratamiento con LGG redujo la expresión génica de IL-6 en el hígado, la fibrosis hepática y la reacción ductular en un modelo de enfermedad hepática colestásica en ratas (AU)


Subject(s)
Animals , Rats , Liver Cirrhosis/diet therapy , Liver Cirrhosis/veterinary , Liver Diseases/diet therapy , Liver Diseases/veterinary , Lacticaseibacillus rhamnosus/isolation & purification , Probiotics/administration & dosage , Lacticaseibacillus rhamnosus , Probiotics/therapeutic use , Tissue Inhibitor of Metalloproteinase-2/therapeutic use , Matrix Metalloproteinase 9/therapeutic use , Oxidative Stress , Rats, Wistar/physiology
5.
Invest Ophthalmol Vis Sci ; 58(4): 1971-1981, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28384717

ABSTRACT

Purpose: The purpose of this study was to determine the endogenous regulation pattern of tissue inhibitor of metalloproteinase-2 (TIMP-2) in the tree shrew sclera during myopia development and investigate the capacity of exogenous TIMP-2 to inhibit matrix metalloproteinase-2 (MMP-2) in vitro and both scleral collagen degradation and myopia development in vivo. Methods: TIMP-2 expression in the sclera during myopia development was assessed using polymerase chain reaction. In vitro TIMP-2 inhibition of MMP-2 was investigated using a gelatinase activity plate assay and zymography. Tree shrews were injected with a collagen precursor before undergoing monocular form deprivation and concurrent daily subconjunctival injections of either TIMP-2 or vehicle to the form-deprived eye. In vivo ocular biometry changes were monitored, and scleral tissue was collected after 12 days and assayed for collagen degradation. Results: The development of myopia was associated with a mean reduction in TIMP-2 mRNA expression after 5 days of form deprivation (P < 0.01). Both activation and activity of MMP-2 were inhibited by TIMP-2 with an IC50 of 10 to 20 and 2 nM, respectively. In vivo exogenous addition of TIMP-2 significantly reduced myopia development (P < 0.01), due to reduced vitreous chamber elongation (P < 0.01). In vivo TIMP-2 treatment also significantly inhibited posterior scleral collagen degradation relative to vehicle-treated eyes (P < 0.01), with levels similar to those in control eyes. Conclusions: Myopia development in mammals is associated with reduced expression of TIMP-2, which contributes to increased degradative activity in the sclera. It follows that replenishment of this TIMP-2 significantly reduced the rate of both scleral collagen degradation and myopia development.


Subject(s)
Gene Expression Regulation, Developmental , Myopia/genetics , RNA/genetics , Sclera/enzymology , Tissue Inhibitor of Metalloproteinase-2/genetics , Tissue Inhibitor of Metalloproteinase-2/therapeutic use , Animals , Animals, Newborn , Biomarkers/metabolism , Biometry , Cells, Cultured , Collagen/metabolism , Disease Models, Animal , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Matrix Metalloproteinase Inhibitors/therapeutic use , Myopia/drug therapy , Myopia/metabolism , RNA, Messenger , Real-Time Polymerase Chain Reaction , Tissue Inhibitor of Metalloproteinase-2/biosynthesis , Tupaia
6.
Am J Surg ; 211(1): 197-205, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26601648

ABSTRACT

BACKGROUND: Burns induce microvascular hyperpermeability. We hypothesize that this occurs partly through an imbalance between matrix metalloproteinases (MMPs) and endogenous MMP inhibitors such as tissue inhibitors of metalloproteinases (TIMPs), and that such derangements can be attenuated with the use of TIMP-2. METHOD: Rats underwent either sham or burn: serum and tissue were collected. Western blot was used to examine MMP-9 and TIMP-2 levels and MMP activity was assayed from lung tissue. Rat lung microvascular endothelial cells were used to assess monolayer permeability and evaluate the adherens junction proteins ß-catenin, vascular endothelial cadherin and filamentous actin after exposure to burn serum ± TIMP-2. RESULTS: Lung tissue from burn animals showed increased MMP activity, decreased levels of TIMP-2, and no difference in levels of active MMP-9 in burn vs control groups. Burn serum increased monolayer permeability, damaged adherens junction proteins, and incited actin stress fiber formation; TIMP-2 attenuated these derangements. CONCLUSIONS: Burns may lower TIMP-2 levels and increase MMP activity and that TIMP-2 application in vitro may attenuate burn-induced hyperpermeability and decreases damage to endothelial structural proteins. These links warrant further investigation.


Subject(s)
Burns/enzymology , Capillary Permeability/drug effects , Endothelial Cells/drug effects , Matrix Metalloproteinase 9/metabolism , Microvessels/drug effects , Protective Agents/pharmacology , Tissue Inhibitor of Metalloproteinase-2/pharmacology , Animals , Biomarkers/metabolism , Blotting, Western , Burns/drug therapy , Burns/physiopathology , Capillary Permeability/physiology , Cells, Cultured , Endothelial Cells/enzymology , Endothelial Cells/physiology , Lung/drug effects , Lung/enzymology , Lung/physiopathology , Male , Microvessels/enzymology , Microvessels/physiopathology , Protective Agents/metabolism , Protective Agents/therapeutic use , Rats , Rats, Sprague-Dawley , Tissue Inhibitor of Metalloproteinase-2/metabolism , Tissue Inhibitor of Metalloproteinase-2/therapeutic use
7.
PLoS One ; 7(4): e35710, 2012.
Article in English | MEDLINE | ID: mdl-22545131

ABSTRACT

TIMP-2 protein has been intensively studied as a promising anticancer candidate agent, but the in vivo mechanism underlying its anticancer effect has not been clearly elucidated by previous works. In this study, we investigated the mechanism underlying the anti-tumor effects of a TIMP-2 fusion protein conjugated with human serum albumin (HSA/TIMP-2). Systemic administration of HSA/TIMP-2 effectively inhibited tumor growth at a minimum effective dose of 60 mg/kg. The suppressive effect of HSA/TIMP-2 was accompanied by a marked reduction of in vivo vascularization. The anti-angiogenic activity of HSA/TIMP-2 was directly confirmed by CAM assays. In HSA/TIMP-2-treated tumor tissues, MMP-2 expression was profoundly decreased without a change in MT1-MMP expression of PECAM-1-positive cells. MMP-2 mRNA was also decreased by HSA/TIMP-2 treatment of human umbilical vein endothelial cells. Zymographic analysis showed that HSA/TIMP-2 substantially decreased extracellular pro-MMP-2 activity (94-99% reduction) and moderately decreased active MMP-2 activity (10-24% reduction), suggesting MT1-MMP-independent MMP-2 modulation. Furthermore, HSA/TIMP-2 had no effect on in vitro active MMP-2 activity and in vivo MMP-2 activity. These studies show that HSA/TIMP-2 potentiates anti-angiogenic activity by modulating MMP-2 expression, but not MMP-2 activity, to subsequently suppress tumor growth, suggesting an important role for MMP-2 expression rather than MMP-2 activity in anti-angiogenesis.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Gene Expression Regulation, Neoplastic/drug effects , Matrix Metalloproteinase 2/genetics , Neovascularization, Pathologic/drug therapy , Prostatic Neoplasms/drug therapy , Serum Albumin/therapeutic use , Tissue Inhibitor of Metalloproteinase-2/therapeutic use , Angiogenesis Inhibitors/genetics , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Human Umbilical Vein Endothelial Cells , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Prostate/blood supply , Prostate/drug effects , Prostate/metabolism , Prostate/pathology , Prostatic Neoplasms/blood supply , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Rats , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/therapeutic use , Serum Albumin/genetics , Tissue Inhibitor of Metalloproteinase-2/genetics
8.
Klin Monbl Augenheilkd ; 228(9): 754-61, 2011 Sep.
Article in German | MEDLINE | ID: mdl-21796575

ABSTRACT

This review summarises some recent aspects of myopia research. The following conclusions have been drawn. As long as myopia progression is visually controlled, at least three different interventions are possible: (i) spectacles/contact lenses which correct only the centre of the visual field and leave the periphery somewhat myopic, (ii) outdoor activity or equivalent temporary increase in illuminance, (iii) pharmacological intervention of retinal growth signals that are transmitted to the underlying sclera. Options (i) and (ii) can be used without risks although there is still room for improvement of the variables. Option (iii) has re-entered a new phase of orientation with new searches for candidate targets after previous testing with muskarinic antagonists (pirenzepine) in children did not enter phase 3 level. If myopia is outside the range over which it is visually controlled by emmetropisation (in the case of high and pathological myopias), in principle the possibility exists to improve the mechanical stability of the sclera pharmacologically. However, there is still a need for more research. Up to now, the mechanical weakness of the sclera in highly mopyic eyes is surgically stabilised by "scleral buckling". However, these procedures have found limited acceptance since the effects were not very reliable. In 40 - 50 % of the cases of high myopia, degenerative processes are found in the retina which can be seen as consequence of the mechanical tension in the fundus, but may also be indepedent of this factor (no significant correlation with axial length!). In part they can be slowed down by intravitreal anti-VEGF therapy. A long-term study from Denmark has shown that most patients with myopia of between 6-9 dpt during puberty reach retirement age without disabling visual loss.


Subject(s)
Myopia/therapy , Aged , Atropine/adverse effects , Atropine/therapeutic use , Axial Length, Eye/drug effects , Axial Length, Eye/physiology , Contact Lenses , Disease Progression , Eyeglasses , Follow-Up Studies , Humans , Intravitreal Injections , Muscarinic Antagonists/adverse effects , Muscarinic Antagonists/therapeutic use , Myopia/complications , Myopia/physiopathology , Prognosis , Retina/drug effects , Retina/physiopathology , Retinal Degeneration/etiology , Retinal Degeneration/physiopathology , Retinal Degeneration/prevention & control , Sclera/physiopathology , Scleral Buckling , Tissue Inhibitor of Metalloproteinase-2/adverse effects , Tissue Inhibitor of Metalloproteinase-2/therapeutic use , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/physiology , Visual Acuity/physiology , Visual Fields/physiology
9.
Clin Transl Sci ; 4(1): 24-31, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21348952

ABSTRACT

HYPOTHESIS: Adenoviral-mediated (AdV-T2) overexpression of TIMP-2 would blunt ventricular remodeling and improve survival in a murine model of chronic ischemic injury. METHODS: Male mice (n = 124) aged 10-14 weeks underwent either (1) left coronary artery ligation to induce myocardial infarction (MI group, n = 36), (2) myocardial injection of 6 × 10¹° viral particles of AdV-T2 immediately post-MI (MI + T2 group, n = 30), (3) myocardial injection of 6 × 10¹° viral particles of a control adenovirus (MI + Ct, n = 38), or 4) received no intervention (controls, n = 20). On post-MI day 7, surviving mice (n = 79) underwent echocardiographic, immunohistochemical, and biochemical analysis. RESULTS: In infarcted animals, the MI + T2 group demonstrated improved survival (p < 0.02), better preservation of developed pressure and ventricular diameter (p < 0.04), and the lowest expression and activity of MMP-2 and MMP-9 (p < 0.04) compared with MI and MI + Ct groups. All infarcted hearts displayed significantly increased inflammatory cell infiltration (p < 0.04 vs. control, MI, or MI + T2), with infiltration highest in the MI + Ct group and lowest in the MI + T2 group (p < 0.04). CONCLUSIONS: Adenoviral mediated myocardial delivery of the TIMP-2 gene improves post-MI survival and limits adverse remodeling in a murine model of MI.


Subject(s)
Gene Transfer Techniques , Genetic Therapy , Myocardial Infarction/physiopathology , Myocardial Infarction/therapy , Tissue Inhibitor of Metalloproteinase-2/genetics , Tissue Inhibitor of Metalloproteinase-2/therapeutic use , Ventricular Remodeling/physiology , Animals , Blotting, Western , Cardiomegaly/etiology , Cardiomegaly/pathology , Cardiomegaly/physiopathology , Collagen/metabolism , Dilatation , Immunohistochemistry , Male , Matrix Metalloproteinases/metabolism , Mice , Mice, Inbred C57BL , Myocardial Infarction/complications , Myocardium/enzymology , Myocardium/pathology , Survival Analysis
10.
J Oral Pathol Med ; 39(1): 94-102, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19895658

ABSTRACT

BACKGROUND: Ameloblastomas are odontogenic neoplasms characterized by local invasiveness. This study was conducted to develop a new animal model of ameloblastoma and to address the role of tissue inhibitor of metalloproteinase-2 (TIMP-2) and matrix metalloproteinase-2 (MMP-2) in the growth and invasiveness of ameloblastomas. METHOD: Donated fresh human ameloblastoma tissue was finely minced, screened, and subcutaneously implanted in three locations on each of 10 BALB/c-nu/nu nude mice. Newly established tumors on each mouse were injected with: (i) transfection reagent; (ii) liposome and transfection reagent; or (iii) liposome, transfection reagent, and the expression plasmid pcDNA3.1(+)/green fluorescent protein (GFP)-TIMP-2. Tumors were monitored for 5 weeks and excised for histopathology, RNA, and protein analyses. RESULTS: The ameloblastoma xenografts were established with high frequency and contained a variety of typical features, validating this new model system. Xenografts injected with the TIMP-2 expression plasmid showed reduced growth, increased TIMP-2 mRNA and protein, and decreased MMP-2 protein compared with the control groups. CONCLUSIONS: We successfully established a new experimental model of ameloblastoma consisting of subcutaneous human xenografts in nude mice. In addition, we demonstrated the successful introduction of the TIMP-2 gene in tumor xenograft cells in vivo, resulting in xenograft growth inhibition. This growth inhibition may have resulted from TIMP-2 overexpression specifically inhibiting MMP-2 protein expression and activity.


Subject(s)
Ameloblastoma/drug therapy , Neoplasm Transplantation , Soft Tissue Neoplasms/drug therapy , Tissue Inhibitor of Metalloproteinase-2/therapeutic use , Transplantation, Heterologous , Adult , Animals , Blotting, Western , Disease Models, Animal , Female , Genetic Vectors/therapeutic use , Humans , Matrix Metalloproteinase 2 , Matrix Metalloproteinase Inhibitors , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness , Plasmids/therapeutic use , Reverse Transcriptase Polymerase Chain Reaction , Subcutaneous Tissue/surgery , Tissue Inhibitor of Metalloproteinase-2/genetics , Transfection , Tumor Cells, Cultured
11.
J Oral Pathol Med ; 38(9): 731-6, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19473446

ABSTRACT

BACKGROUND: Ameloblastoma is an odontogenic benign tumor characterized by local invasiveness and most of its local recurrences clinically result from local invasion. This study used matrix metalloproteinase-2 (MMP-2) inhibitor I (MMP-2I) to investigate the role played by MMP-2 activity in the local invasiveness of ameloblastoma. METHODS: The cells and xenografts of ameloblastoma were treated with MMP-2I and treatment group were compared with the control group. In vitro, the invasive activity of tumor cells was assayed in transwell cell culture chamber. Gelatinolytic activity of gelatinases and MMP-2/tissue inhibitor of matrix metalloproteinase (TIMP-2) protein expression was detected using gelatin zymography and flow cytometry. The cell viability and adhesion were evaluated using methyl thiazol tetrazolium. In vivo, bilateral subrenal capsule xenograft transplantation of ameloblastoma was performed in 10 nude mice and the invasion of ameloblastoma into the renal parenchyma was observed. RESULTS: Active-MMP-2 of conditioned media was significantly lower in treatment group than in the control group. Accordingly, potential of in vitro cell invasion, adhesion and in vivo tumor invasion were also significantly lower in the treatment group than in the control group. CONCLUSIONS: Inhibitor of MMP-2 activity suppressed the local invasive capability of ameloblastoma by decreasing MMP-2 activity. MMP-2 activity is in relation with invasive capacity of ameloblastoma.


Subject(s)
Ameloblastoma/enzymology , Jaw Neoplasms/enzymology , Matrix Metalloproteinase Inhibitors , Tissue Inhibitor of Metalloproteinase-2/pharmacology , Ameloblastoma/drug therapy , Animals , Humans , Jaw Neoplasms/drug therapy , Matrix Metalloproteinase 2/physiology , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness , Neoplasm Transplantation , Tissue Inhibitor of Metalloproteinase-2/therapeutic use , Tumor Cells, Cultured
12.
J Nutr ; 137(6): 1423-9, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17513401

ABSTRACT

Conjugated linoleic acid (CLA) is a group of linoleic acid derivatives that has been implicated in animal studies to reduce a number of components of mammary tumorigenesis. Previously, we showed that CLA could alter the latency and metastasis of the highly metastatic transplantable line 4526 mouse mammary tumor. Several possible mechanisms have been proposed for the actions of CLA, but here we assessed how CLA may act to alter the expression and activity of matrix-modifying proteins within tumors from line 4526. In vitro, highly metastatic mouse mammary tumor cells had significantly decreased invasiveness after treatment with CLA, an indication that matrix-modifying proteins may have been altered. Using these same highly metastatic cells, primary tumors were grown in mice of separate groups fed 0, 0.1, 0.5, and 1% CLA (wt:wt) and evaluated for their levels and activities of matrix-modifying enzymes, enzyme inhibitors, and enzyme activators. The addition of CLA to the diet increased steady-state levels of messenger RNA (mRNA) of the matrix metalloproteinases (MMP) -2 and -9 in primary tumors removed from mice. However, western analysis revealed that although relative levels of the proform of MMP-9 were consistent with the mRNA observations, MMP-2 proform levels were actually decreased by dietary CLA. The activity of MMP-2 was barely detectable, but gelatin zymography and an in vitro activity assay showed that MMP-9 activity was significantly decreased by CLA. The steady-state mRNA and protein levels of tissue inhibitors of metalloproteinase-1 (TIMP-1) and TIMP-2, natural inhibitors of MMP, were increased at higher dietary CLA levels relative to low or no CLA. Suppression of MMP activity, therefore, may be 1 pathway through which CLA reduces tumor invasion and spread.


Subject(s)
Antineoplastic Agents/pharmacology , Linoleic Acids, Conjugated/pharmacology , Mammary Neoplasms, Animal/enzymology , Metalloproteases/antagonists & inhibitors , Tissue Inhibitor of Metalloproteinase-1/pharmacology , Tissue Inhibitor of Metalloproteinase-2/pharmacology , Animals , Antineoplastic Agents/therapeutic use , Diet , Female , Linoleic Acids, Conjugated/therapeutic use , Mammary Neoplasms, Animal/pathology , Mammary Neoplasms, Animal/prevention & control , Metalloproteases/metabolism , Mice , Mice, Inbred BALB C , Neoplasm Invasiveness , Tissue Inhibitor of Metalloproteinase-1/therapeutic use , Tissue Inhibitor of Metalloproteinase-2/therapeutic use
13.
Circulation ; 113(20): 2435-44, 2006 May 23.
Article in English | MEDLINE | ID: mdl-16702468

ABSTRACT

BACKGROUND: Matrix metalloproteinase (MMP)-associated extracellular matrix degradation is thought to contribute to the progression and rupture of atherosclerotic plaques. However, direct evidence of this concept remains elusive. We hypothesized that overexpression of tissue inhibitor of metalloproteinase (TIMP)-1 or TIMP-2 would attenuate atherosclerotic plaque development and instability in high fat-fed apolipoprotein E-knockout (apoE(-/-)) mice. METHODS AND RESULTS: Seventy male apoE(-/-) mice (n=10/group) fed a high-fat diet for 7 weeks were injected intravenously with first-generation adenoviruses expressing the gene for human TIMP-1 (RAdTIMP-1) or TIMP-2 (RAdTIMP-2) or a control adenovirus (RAd66) and were fed a high-fat diet for a further 4 weeks. Analysis of brachiocephalic artery plaques revealed that RAdTIMP-2 but not RAdTIMP-1 infection resulted in a marked reduction (48+/-13%, P<0.05) in lesion area compared with that in control animals. Markers associated with plaque instability, assessed by smooth muscle cell and macrophage content and the presence of buried fibrous caps, were significantly reduced by RAdTIMP-2. Effects on lesion size were not sustained with first-generation adenoviruses, but murine TIMP-2 overexpression mediated by helper-dependent adenoviral vectors exerted significant effects on plaques assessed 11 weeks after infection. In an attempt to determine the mechanism of action, we treated macrophages and macrophage-derived foam cells with exogenous TIMP-2 in vitro. TIMP-2 significantly inhibited migration and apoptosis of macrophages and foam cells, whereas TIMP-1 failed to exert similar effects. CONCLUSIONS: Overexpression of TIMP-2 but not TIMP-1 inhibits atherosclerotic plaque development and destabilisation, possibly through modulation of macrophage and foam cell behavior. Helper-dependent adenovirus technology is required for these effects to be maintained long term.


Subject(s)
Atherosclerosis/prevention & control , Enzyme Inhibitors/therapeutic use , Tissue Inhibitor of Metalloproteinase-2/genetics , Adenoviridae , Animals , Apolipoproteins E/deficiency , Atherosclerosis/physiopathology , Disease Models, Animal , Disease Progression , Genetic Vectors , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Tissue Inhibitor of Metalloproteinase-1/blood , Tissue Inhibitor of Metalloproteinase-2/therapeutic use
15.
J Gene Med ; 7(2): 145-57, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15546163

ABSTRACT

BACKGROUND: Previous studies have shown that TIMP-2 overexpression is a useful therapeutic tool for inhibiting tumor growth and invasion in animals. However, it has not been reported whether genetic manipulation for TIMP-2 overexpression can induce an inhibitory effect on spontaneous metastasis from the primary tumor site to other organs such as lungs or lymph nodes in an animal model. METHODS: The present studies describe the effects of retrovirus-mediated TIMP-2 gene transfer into human breast cancer cell lines on the in vitro invasion of the tumor cells or the in vivo growth in nude mouse. Here we also used retroviral-mediated TIMP-2 overexpression by intratumoral injection for suppression of metastasis in human breast carcinoma established in the mammary fat pad of nude mice. RESULTS: As expected, overexpression of TIMP-2 inhibited matrix metalloprotenase (MMP) activity and invasion of the tumor cells. Also, the growth rate of tumors grafted with the breast cancer cells transduced with the retrovirus vector encoding TIMP-2 cDNA was significantly slower than the growth rate of tumors grafted with the breast cancer cells transduced with a control retrovirus vector. Furthermore, single intratumoral injection of the TIMP-2 retrovirus-producing cells into human breast tumor tissue established in mammary fat pads of nude mice showed a dramatic decrease in size and number of lung metastatic tumors. CONCLUSIONS: Retrovirus-mediated TIMP-2 gene transfer into human breast cancer cells is able to down-regulate invasion and show that tumor-derived angiogenesis is reduced. In this model, retroviral-mediated transduction of TIMP-2 cDNA into a limited population of human tumor cells inhibits tumor growth and prevents distant pulmonary metastasis. These results indicate that it may not be necessary to deliver and express these genes in every single tumor cell as long as the level of expression in a limited number of transduced cells is sufficient to prevent the excessive breakdown of the extracellular matrix.


Subject(s)
Antineoplastic Agents/therapeutic use , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/secondary , Gene Expression Regulation, Neoplastic , Genetic Therapy/methods , Lung Neoplasms/secondary , Tissue Inhibitor of Metalloproteinase-2/therapeutic use , Animals , Antineoplastic Agents/metabolism , Blotting, Western , Carcinoma, Ductal, Breast/blood supply , Carcinoma, Ductal, Breast/therapy , Cell Line, Tumor , Cell Proliferation , Extracellular Matrix/metabolism , Female , Gene Transfer Techniques , Genetic Vectors/genetics , Humans , Hylobates , Immunohistochemistry , Lung Neoplasms/blood supply , Lung Neoplasms/therapy , Matrix Metalloproteinases, Membrane-Associated , Metalloendopeptidases/metabolism , Mice , Mice, Nude , Neoplasm Invasiveness/prevention & control , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/therapy , Retroviridae , Tissue Inhibitor of Metalloproteinase-2/genetics , Tissue Inhibitor of Metalloproteinase-2/metabolism
16.
Int J Cancer ; 105(3): 419-29, 2003 Jun 20.
Article in English | MEDLINE | ID: mdl-12704680

ABSTRACT

Gene transfer is an attractive approach to fight cancer by targeting cancer cells or their vasculature. Our study reports the inhibition of tumor growth and angiogenesis by a nonviral method using dendrimers associated with 36-mer anionic oligomers (ON36) for delivering angiostatin (Kringle 1-3) and tissue inhibitor of metalloproteinase (TIMP)-2 genes. The optimal concentrations of dendrimers and ON36 for an efficient green fluorescent protein (GFP) plasmid delivery in endothelial cells (HMEC-1) and cancer cells (MDA-MB-435) were first chosen. Then the efficacy of transfection was determined by testing angiostatin and TIMP-2 secretion by Western blot and the biologic effects were evaluated. Angiostatin gene transfer markedly reduced in vitro (i) HMEC-1 but not MDA-MB-435 proliferation; (ii) HMEC-1 and MDA-MB-435 wound healing reparation; and (iii) capillary tube formation. TIMP-2 gene transfer did not affect cell proliferation but strongly inhibited (i) wound healing of HMEC-1 and MDA-MB-435 cells; and (ii) capillary tube formation. Supernatants of transfected-MDA-MB-435 cells also inhibited the formation of angiogenic networks on Matrigel, indicating a paracrine effect. In vivo, intratumoral angiostatin or TIMP-2 gene delivery using dendrimers associated with ON36 effectively inhibited tumor growth by 71% and 84%, respectively. Combined gene transfer resulted in 96% inhibition of tumor growth. Tumor-associated vascularization was also greatly reduced. These findings provide a basis for the further development of nonviral delivery of genes to fight cancer.


Subject(s)
Genetic Therapy/methods , Neovascularization, Pathologic , Peptide Fragments/genetics , Plasminogen/genetics , Tissue Inhibitor of Metalloproteinase-2/genetics , Angiostatins , Animals , Blotting, Western , Cell Division , Collagen/pharmacology , Cytokines/metabolism , Drug Combinations , Endothelium/cytology , Flow Cytometry , Gene Transfer Techniques , Green Fluorescent Proteins , Humans , Laminin/pharmacology , Luminescent Proteins/metabolism , Mice , Mice, Nude , Neoplasms, Experimental/therapy , Peptide Fragments/therapeutic use , Plasmids/metabolism , Plasminogen/therapeutic use , Proteoglycans/pharmacology , Time Factors , Tissue Inhibitor of Metalloproteinase-2/therapeutic use , Transfection , Tumor Cells, Cultured , Wound Healing
17.
Pharmacol Res ; 46(2): 155-63, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12220955

ABSTRACT

Glioma, the most common form of brain tumor, has been shown mostly by in vitro studies to utilize matrix metalloproteinase (MMP) for invasive growth through degradation of the extracellular matrix. In order to examine the in vivo role of MMP, we established a rodent model of glioma progression using C6 rat glioma cells and analyzed the effect of tissue inhibitors of metalloproteinases (TIMPs). TIMP-2 rather than TIMP-1 caused significant reduction of the tumor size accompanied by the presence of degenerated blood vessels and ischemic necrosis. Because TIMP-2 inhibits MMP-2 preferentially, we then examined glioma growth in MMP-2-deficient mice and observed essentially identical consequences. While MMP-2 activity was present in the tumor and adjacent tissues of the wild-type mice, no MMP-2 activity was detected even in the tumor of the null mice, although C6 cells are known to express MMP-2. These observations suggest that glioma induces MMP-2 and utilizes its activity in the host tissue to support angiogenesis and to maintain angioarchitecture.


Subject(s)
Brain Neoplasms/enzymology , Corpus Striatum/enzymology , Dura Mater/enzymology , Glioma/enzymology , Matrix Metalloproteinase 2/metabolism , Neovascularization, Pathologic/drug therapy , Protease Inhibitors/therapeutic use , Animals , Brain Neoplasms/blood supply , Brain Neoplasms/genetics , Corpus Striatum/blood supply , Dura Mater/blood supply , Glioma/blood supply , Glioma/genetics , Humans , Male , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase Inhibitors , Mice , Mice, Knockout , Neoplasm Transplantation , Neovascularization, Pathologic/metabolism , Protease Inhibitors/pharmacology , Rats , Rats, Sprague-Dawley , Tissue Inhibitor of Metalloproteinase-1/pharmacology , Tissue Inhibitor of Metalloproteinase-1/therapeutic use , Tissue Inhibitor of Metalloproteinase-2/pharmacology , Tissue Inhibitor of Metalloproteinase-2/therapeutic use , Tumor Cells, Cultured
19.
Drugs Aging ; 11(3): 229-44, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9303281

ABSTRACT

Matrix metalloproteinases (MMPs), or matrixins, are a family of zinc endopeptidases that play a key role in both physiological and pathological tissue degradation. Normally, there is a careful balance between cell division, matrix synthesis and matrix degradation, which is under the control of cytokines, growth factors and cell matrix interactions. The MMPs are involved in remodelling during tissue morphogenesis and wound healing. Under pathological conditions, this balance is altered: in arthritis, there is uncontrolled destruction of cartilage; in cancer, increased matrix turnover is thought to promote tumour cell invasion. The demonstration of a functional role of MMPs in arthritis and tumour metastasis raises the possibility of therapeutic intervention using synthetic MMP inhibitors with appropriate selectivity and pharmacokinetics. As the process of drug discovery focuses on structure-based design, efforts to resolve the 3-dimensional structures of the MMP family have intensified. Several novel MMP inhibitors have been identified and are currently being investigated in clinical trials. The structural information that is rapidly accumulating will be useful in refining the available inhibitors to selectively target specific MMP family members. In this review, we focus on the role of MMPs and their inhibitors in tumour invasion, metastasis and angiogenesis, and examine how MMPs may be targeted to prevent cancer progression.


Subject(s)
Antineoplastic Agents/therapeutic use , Extracellular Matrix/enzymology , Gene Expression Regulation, Enzymologic/drug effects , Metalloendopeptidases/antagonists & inhibitors , Neoplasms/drug therapy , Neoplasms/enzymology , Protease Inhibitors/therapeutic use , Animals , Antineoplastic Agents/pharmacology , Humans , Metalloendopeptidases/biosynthesis , Metalloendopeptidases/classification , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/enzymology , Protease Inhibitors/pharmacology , Tissue Inhibitor of Metalloproteinase-2/therapeutic use , Tissue Inhibitor of Metalloproteinase-3/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...