Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Ann Oncol ; 31(12): 1679-1692, 2020 12.
Article in English | MEDLINE | ID: mdl-32918998

ABSTRACT

Glioblastoma represents the most common primary malignancy of the central nervous system in adults and remains a largely incurable disease. The elucidation of disease subtypes based on mutational profiling, gene expression and DNA methylation has so far failed to translate into improved clinical outcomes. However, new knowledge emerging from the subtyping effort in the IDH-wild-type setting may provide directions for future precision therapies. Here, we review recent learnings in the field, and further consider how tumour microenvironment differences across subtypes may reveal novel contexts of vulnerability. We discuss recent treatment approaches and ongoing trials in the IDH-wild-type glioblastoma setting, and propose an integrated discovery stratagem incorporating multi-omics, single-cell technologies and computational approaches.


Subject(s)
Brain Neoplasms , Glioblastoma , Adult , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , DNA Methylation , Glioblastoma/drug therapy , Glioblastoma/genetics , Humans , Isocitrate Dehydrogenase/genetics , Mutation , Precision Medicine , Tumor Microenvironment
2.
Stem Cells Int ; 2016: 5623235, 2016.
Article in English | MEDLINE | ID: mdl-27274737

ABSTRACT

Serum-free culture methods for patient-derived primary glioma cultures, selecting for glioma stem-like cells (GSCs), are becoming the gold standard in neurooncology research. These GSCs can be implemented in drug screens to detect patient-specific responses, potentially bridging the translational gap to personalized medicine. Since numerous compounds are available, a rapid and reliable readout for drug efficacies is required. This can be done using approaches that measure viability, confluency, cytotoxicity, or apoptosis. To determine which assay is best suitable for drug screening, 10 different assays were systematically tested on established glioma cell lines and validated on a panel of GSCs. General applicability was assessed using distinct treatment modalities, being temozolomide, radiation, rapamycin, and the oncolytic adenovirus Delta24-RGD. The apoptosis and cytotoxicity assays did not unequivocally detect responses and were excluded from further testing. The NADH- and ATP-based viability assays revealed comparable readout for all treatments; however, the latter had smaller standard deviations and direct readout. Importantly, drugs that interfere with cell metabolism require alternative techniques such as confluency monitoring to accurately measure treatment effects. Taken together, our data suggest that the combination of ATP luminescence assays with confluency monitoring provides the most specific and reproducible readout for drug screening on primary GSCs.

3.
Gene Ther ; 22(12): 947-59, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26196249

ABSTRACT

Oncolytic viruses (OV) have broad potential as an adjuvant for the treatment of solid tumors. The present study addresses the feasibility of clinically applicable drugs to enhance the oncolytic potential of the OV Delta24-RGD in glioblastoma. In total, 446 drugs were screened for their viral sensitizing properties in glioblastoma stem-like cells (GSCs) in vitro. Validation was done for 10 drugs to determine synergy based on the Chou Talalay assay. Mechanistic studies were undertaken to assess viability, replication efficacy, viral infection enhancement and cell death pathway induction in a selected panel of drugs. Four viral sensitizers (fluphenazine, indirubin, lofepramine and ranolazine) were demonstrated to reproducibly synergize with Delta24-RGD in multiple assays. After validation, we underscored general applicability by testing candidate drugs in a broader context of a panel of different GSCs, various solid tumor models and multiple OVs. Overall, this study identified four viral sensitizers, which synergize with Delta24-RGD and two other strains of OVs. The viral sensitizers interact with infection, replication and cell death pathways to enhance efficacy of the OV.


Subject(s)
Glioblastoma/therapy , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/virology , Oncolytic Virotherapy/methods , Oncolytic Viruses/drug effects , Brain Neoplasms/drug therapy , Brain Neoplasms/therapy , Brain Neoplasms/virology , Cell Line, Tumor , Drug Evaluation, Preclinical , Fluphenazine/pharmacology , Glioblastoma/drug therapy , Glioblastoma/virology , HCT116 Cells , Humans , Indoles/pharmacology , Oncolytic Viruses/physiology , Virus Replication/drug effects
4.
J Neuroimmunol ; 278: 271-6, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25468776

ABSTRACT

Inflammation plays a key role in the pathophysiology of Glioblastoma Multiforme (GBM). Here we focus on the contribution of the so far largely ignored complement system. ELISA and immunohistochemistry were combined to assess levels and localization of critical components of the initiation- and effector pathways of the complement cascade in sera and tumor tissue from GBM patients and matched controls. Serum levels of factor-B were decreased in GBM patients whereas C1q levels were increased. C1q and factor-B deposited in the tumor tissue. Deposition of C3 and C5b-9 suggests local complement activation.MBL deficiency, based on serum levels, was significantly less frequent among GBM patients compared to controls (14% vs. 33%). Therefore low levels of MBL may protect against the initiation/progression of GBM.


Subject(s)
Brain Neoplasms , Complement Activation , Glioblastoma , Adult , Brain Neoplasms/blood , Brain Neoplasms/immunology , Brain Neoplasms/physiopathology , Complement C1q/metabolism , Complement Factor B/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Glioblastoma/blood , Glioblastoma/immunology , Glioblastoma/physiopathology , Humans , Male , Mannose-Binding Lectin/metabolism , Middle Aged , Survival Analysis
5.
Cancer Gene Ther ; 20(9): 507-13, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23907517

ABSTRACT

Glioblastoma (GB) is a devastating disease for which new treatment modalities are needed. Efficacious therapy requires the removal of stem-cell like cells, these cells drive tumor progression because of their ability to self-renew and differentiate. In glioblastoma, the GB stem-like cells (GSC) form a small population of tumor cells and possess high resistance to chemo and radiation therapies. To assess the sensitivity of GSC to reovirus-mediated cytolysis, a panel of GSC cultures was exposed to wild-type reovirus Type 3 Dearing (T3D) and its junction adhesion molecule-A (JAM-A)-independent mutant, jin-1. Several parameters were evaluated, including the fraction of cells expressing the JAM-A reovirus receptor, the fraction of cells synthesizing reovirus proteins, the number of infectious reovirus particles required to reduce cell viability, the amount of infectious progeny reovirus produced and the capacity of the reoviruses to infect the GSC in 3-dimensional (3D) tumor cell spheroids. Our data demonstrate a marked heterogeneity in the susceptibility of the cultures to reovirus-induced cytolysis. While in monolayer cultures the jin-1 reovirus was generally more cytolytic than the wild-type reovirus T3D, in the 3D GSC spheroids, these viruses were equally effective. Despite the variation in reovirus sensitivity between the different GSC cultures, our data support the use of reovirus as an oncolytic agent. It remains to be established whether the variation in the reovirus sensitivity correlates with a patient's response to reovirus therapy. Moreover, our data show that the expression of the JAM-A receptor is not a major determinant of reovirus sensitivity in 3D GSC cultures.


Subject(s)
Brain Neoplasms/metabolism , Glioblastoma/metabolism , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/virology , Reoviridae/physiology , Viral Tropism , Adult , Aged , Brain Neoplasms/virology , Cell Adhesion Molecules/metabolism , Cell Line, Tumor , Cytopathogenic Effect, Viral , Female , Glioblastoma/virology , Humans , Male , Middle Aged , Oncolytic Viruses/physiology , Receptors, Cell Surface/metabolism , Spheroids, Cellular , Tumor Cells, Cultured
6.
Cancer Biol Ther ; 7(5): 786-93, 2008 May.
Article in English | MEDLINE | ID: mdl-18756624

ABSTRACT

Malignant glioma continues to be a major target for gene therapy and virotherapy due to its aggressive growth and the current lack of effective treatment. However, these approaches have been hampered by inefficient infection of glioma cells by viral vectors,particularly vectors derived from serotype 5 adenoviruses (Ad5). This results from limited cell surface expression of the primary adenovirus receptor, coxsackie-adenovirus-receptor (CAR), on tumor cells. To circumvent this problem, Ad fiber pseudotyping,the genetic replacement of either the entire fiber or fiber knob domain with its structural counterpart from another human Ad serotype that recognizes a cellular receptor other than CAR, has been shown to enhance Ad infectivity in a variety of tumor types,including human glioma. Here, we have extended the paradigm of genetic pseudotyping to include fiber domains from non-human or"xenotype" Ads for infectivity enhancement of human glioma cell populations. In this study, we evaluated the gene transfer efficiency of a panel of Ad vectors which express one of five different "xenotype"fiber knob domains, including those derived from murine,ovine, porcine and canine species, in both human glioma cell lines as well as primary glioma tumor cells from patients. Adenovirus vectors displaying either canine Ad or porcine Ad fiber elements had the highest gene transfer to both glioma cell lines and primary tumor cells. The correlation between the viral infectivity of modified adenovirus vectors and expression of human CAR and CD46(an adenovirus type B receptor) on the surfaces of tumor cells was also analyzed. Taken together, human adenovirus vectors modified with "xenotype" fiber elements could be excellent candidates to target human glioma.


Subject(s)
Adenoviridae/metabolism , Brain Neoplasms/metabolism , Glioma/metabolism , Animals , Cell Line, Tumor , Constitutive Androstane Receptor , Cytomegalovirus/metabolism , Gene Transfer Techniques , Genetic Therapy/methods , Genetic Vectors , Humans , Membrane Cofactor Protein/metabolism , Mice , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factors/metabolism , Viruses/metabolism
7.
Bone ; 40(4): 797-812, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17189720

ABSTRACT

This review begins with an introduction to the malignant bone tumor, osteosarcoma [OS] and then moves to a discussion of the commonly used vectors for gene transfer. We first briefly highlight non-viral vectors including polymeric and liposomal delivery systems but concentrate predominantly on the 5 leading viral vectors used in cancer gene therapy, specifically retroviruses, adeno-associated viruses, herpes viruses and lentiviruses with the most detailed analysis reserved for adenoviruses. The 3 main strategies for gene therapy in osteosarcoma are next summarized. As part of this review, the several prodrug-converting enzymes utilized in OS suicide gene therapy are examined. The text then turns to a discussion of adenovirus-mediated gene transfer and the need for tumor targeting via transductional or transcriptional approaches. Because of practical problems with use of replication-incompetent viruses in achieving complete tumor kill in vivo, virotherapy utilizing replication competent viruses has come to the fore. This topic is, thus, next reviewed which allows for a natural transition to a discussion of armed therapeutic viruses many of which are conditionally replicating adenoviruses carrying transgenes with established anti-tumor efficacy. We recognize that several other issues have arisen which hamper progress in the field of cancer gene therapy. We, therefore, review viral-induced toxicity in the host and vector delivery issues which have been found to potentially influence safety. We end with a brief perspective including commenting on animal models used in examining delivery strategies for osteosarcoma gene therapy. The challenges remaining are touched upon most especially the need to deal with pulmonary metastatic disease from OS.


Subject(s)
Bone Neoplasms/therapy , Genetic Therapy/methods , Osteosarcoma/therapy , Genes, Transgenic, Suicide , Genetic Therapy/adverse effects , Genetic Vectors , Humans , Mutation , Oncolytic Virotherapy , Safety , Viruses/genetics
8.
Oncol Res ; 16(10): 471-7, 2007.
Article in English | MEDLINE | ID: mdl-18196871

ABSTRACT

Multicellular tumor spheroids are used as a model to assess the efficacy of replicating oncolytic adenoviruses. As most assays used to assess cellular viability are unsuitable for oncolytic viruses because of ongoing viral replication, we have used positron emission tomography (PET) to sequentially determine the incorporation of 18F-labeled deoxyglucose (18F-DG) as a measure of viability and compared the results to more commonly used assays for measuring the effect of oncolytic therapy. Glioma monolayer cultures and spheroids were infected with wild-type replicating adenovirus and viability was measured by 18F-DG incorporation, WST-1 assay, crystal violet assay, and spheroid volume 2 to 10 days following infection. Results show that volume measurements in adenovirus-infected spheroids are confounded by the cytopathic effect occurring in infected cells. 18F-DG PET provides a useful method to assess small differences in cell number and viability following oncolytic viral therapy in glioma monolayer cultures and spheroids without the need for disintegration of these cultures. Moreover, using 18F-DG PET, repeated sequential measurements of spheroid viability can be made, decreasing the required number of spheroids per experiment. This is a valuable feature when using spheroids derived from limited amounts of patient material.


Subject(s)
Adenoviridae/physiology , Fluorodeoxyglucose F18 , Glioma/diagnostic imaging , Glioma/therapy , Oncolytic Virotherapy/methods , Glioma/genetics , Glioma/virology , Humans , Positron-Emission Tomography/methods , Spheroids, Cellular , Tumor Cells, Cultured
9.
Gene Ther ; 10(23): 1982-91, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14528322

ABSTRACT

Conditionally replicative adenoviruses (CRAds) are potentially useful agents for anticancer virotherapy approaches. However, lack of coxsackievirus and adenovirus receptor (CAR) expression on many primary tumor cells limits the oncolytic potency of CRAds. This makes the concept of targeting, that is, redirecting infection via CAR-independent entry pathways, relevant for CRAd development. Bispecific adapter molecules constitute highly versatile means for adenovirus targeting. Here, we constructed a CRAd with the Delta24 E1A mutation that produces a bispecific single-chain antibody directed towards the adenovirus fiber knob and the epidermal growth factor receptor (EGFR). This EGFR-targeted CRAd exhibited increased infection efficiency and oncolytic replication on CAR-deficient cancer cells and augmented lateral spread in CAR-deficient 3-D tumor spheroids in vitro. When compared to its parent control with native tropism, the new CRAd exhibited similar cytotoxicity on CAR-positive cancer cells, but up to 1000-fold enhanced oncolytic potency on CAR-deficient, EGFR-positive cancer cells. In addition, EGFR-targeted CRAd killed primary human CAR-deficient brain tumor specimens that were refractory to the parent control virus. We conclude, therefore, that CRAds expressing bispecific targeting adapter molecules are promising agents for cancer treatment. Their use is likely to result in enhanced oncolytic replication in cancerous tissues and thus in more effective tumor regression.


Subject(s)
Adenovirus E1A Proteins/genetics , ErbB Receptors/metabolism , Genetic Therapy/methods , Genetic Vectors/genetics , Neoplasms/therapy , Receptors, Virus/genetics , Animals , Cell Line, Tumor , Coxsackie and Adenovirus Receptor-Like Membrane Protein , ErbB Receptors/immunology , Gene Targeting , Humans , Immunoglobulin Fc Fragments/immunology , Neoplasms/immunology , Neoplasms/metabolism , Receptors, Virus/deficiency , Virus Replication
10.
Circ Res ; 91(10): 945-52, 2002 Nov 15.
Article in English | MEDLINE | ID: mdl-12433840

ABSTRACT

Proteases of the plasminogen activator (PA) and matrix metalloproteinase (MMP) system play an important role in smooth muscle cell (SMC) migration and neointima formation after vascular injury. Inhibition of either PAs or MMPs has previously been shown to result in decreased neointima formation in vivo. To inhibit both protease systems simultaneously, a novel hybrid protein, TIMP-1.ATF, was constructed consisting of the tissue inhibitor of metalloproteinase-1 (TIMP-1) domain, as MMP inhibitor, linked to the receptor-binding amino terminal fragment (ATF) of urokinase. By binding to the u-PA receptor this protein will not only anchor the TIMP-1 moiety directly to the cell surface, it will also prevent the local activation of plasminogen by blocking the binding of urokinase-type plasminogen activator (u-PA) to its receptor. Adenoviral expression of TIMP-1.ATF was used to inhibit SMC migration and neointima formation in human saphenous vein segments in vitro. SMC migration was inhibited by 65% in Ad.TIMP-1.ATF-infected cells. Infection with adenoviral vectors encoding the individual domains, Ad.TIMP-1 and Ad.ATF, reduced migration by 32% and 52%, respectively. Neointima formation in saphenous vein organ cultures infected with Ad.TIMP-1.ATF was inhibited by 72% compared with 42% reduction after Ad.TIMP-1 infection and 34% after Ad.ATF infection. These data show that binding of TIMP-1.ATF hybrid protein to the u-PA receptor at the cell surface strongly enhances the inhibitory effect of TIMP-1 on neointima formation in human saphenous vein cultures.


Subject(s)
Muscle, Smooth, Vascular/metabolism , Recombinant Fusion Proteins/biosynthesis , Tissue Inhibitor of Metalloproteinase-1/genetics , Tunica Intima/metabolism , Urokinase-Type Plasminogen Activator/genetics , Adenoviridae/genetics , Animals , CHO Cells/cytology , CHO Cells/drug effects , CHO Cells/metabolism , Cell Division/drug effects , Cell Division/physiology , Cell Membrane/metabolism , Cell Movement/drug effects , Cell Movement/physiology , Cells, Cultured , Cricetinae , Culture Media, Conditioned/pharmacology , Enzyme Activation/drug effects , Flow Cytometry , Gene Transfer Techniques , Humans , In Vitro Techniques , Matrix Metalloproteinase 13 , Matrix Metalloproteinase Inhibitors , Muscle, Smooth, Vascular/cytology , Protein Structure, Tertiary/physiology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, Cell Surface/metabolism , Receptors, Urokinase Plasminogen Activator , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/pharmacology , Saphenous Vein/cytology , Saphenous Vein/drug effects , Saphenous Vein/metabolism , Tissue Inhibitor of Metalloproteinase-1/pharmacology , Tunica Intima/drug effects
11.
Expert Opin Biol Ther ; 2(8): 943-52, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12517272

ABSTRACT

Standard therapies are not capable of curing patients with malignant glioma; more than 90% of patients die within 2 years after diagnosis. Gene therapy appeared as a promising new approach for this disease. However, results of clinical trials with replication deficient viral vectors were disappointing. The main reasons being poor transduction efficiency of adenovirus towards glioma cells and limited spread and distribution of the vector in the tumour. With the increasing knowledge of viral genetics and its functions, an attractive alternative tool to kill malignant glioma cells has been developed: Replicating adenovirus as an oncolytic agent. This type of therapy, also referred to as virotherapy, has the potential to overcome some of the limitations connected with replication deficient adenoviral vectors. In this review the authors describe the latest developments in strategies that are being used to create a tumour- or glioma-selective replicating adenovirus. Special attention is given to the methods of viral delivery to an infiltrating tumour in the brain, regarding optimal dose and toxicity. Furthermore, the role of conventional antitumour treatments, such as irradiation and chemotherapy, in enhancing the effect of virotherapy is being emphasised.


Subject(s)
Adenoviridae , Brain Neoplasms/therapy , Viruses , Animals , Cell Line, Tumor , Glioma/therapy , Humans
12.
Gene Ther ; 8(7): 534-41, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11319620

ABSTRACT

Injury-induced neointimal development results from migration and proliferation of vascular smooth muscle cells (SMC). Cell migration requires controlled proteolytic degradation of extracellular matrix surrounding the cell. Plasmin is a major contributor to this process by degrading various matrix proteins directly, or indirectly by activating matrix metalloproteinases. This makes it an attractive target for inhibition by gene transfer. An adenoviral vector, Ad.ATF.BPTI, was constructed encoding a hybrid protein, which consists of the aminoterminal fragment (ATF) of urokinase-type plasminogen activator (u-PA) linked to bovine pancreas trypsin inhibitor (BPTI), a potent inhibitor of plasmin. This hybrid protein binds to the u-PA receptor, thereby inhibiting plasmin activity at the cell surface, and was found to be a potent inhibitor of cell migration in vitro. Local infection with Ad.ATF.BPTI of balloon-injured rat carotid artery resulted in detectable expression of ATF.BPTI mRNA and protein in the vessel wall. Morphometric analysis of arterial cross-sections revealed that delivery of Ad.ATF.BPTI to the carotid artery wall at the time of balloon injury inhibited neointima formation by 53% (P < 0.01) at 14 days and 19% (P = NS) at 28 days after injury when compared with control vector-infected arteries. Intima/media ratios were decreased by 60% (P < 0.01) and 35% (P < 0.05) at 14 and 28 days, respectively, when compared with control vector-infected arteries. Furthermore, a small but significant increase in medial area was found in the Ad.ATF.BPTI-treated arteries at 28 days (P < 0.05). These results show that local infection of the vessel wall with Ad.ATF.BPTI reduces neointima formation, presumably by inhibiting SMC migration, thereby offering a novel therapeutic approach to inhibiting neointima development.


Subject(s)
Angioplasty, Balloon/adverse effects , Aprotinin/genetics , Carotid Stenosis/prevention & control , Fibrinolysin/antagonists & inhibitors , Genetic Therapy/methods , Adenoviridae/genetics , Animals , Aprotinin/metabolism , Carotid Arteries/metabolism , Carotid Stenosis/pathology , Carotid Stenosis/therapy , Cell Culture Techniques , Gene Expression , Gene Transfer Techniques , Genetic Vectors , Humans , Male , Peptide Fragments/genetics , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Recurrence , Tunica Intima/pathology , Tunica Media/pathology , Urokinase-Type Plasminogen Activator/genetics
13.
Circulation ; 103(4): 562-9, 2001 Jan 30.
Article in English | MEDLINE | ID: mdl-11157723

ABSTRACT

BACKGROUND: Smooth muscle cell migration, in addition to proliferation, contributes to a large extent to the neointima formed in humans after balloon angioplasty or bypass surgery. Plasminogen activator/plasmin-mediated proteolysis is an important mediator of this smooth muscle cell migration. Here, we report the construction of a novel hybrid protein designed to inhibit the activity of cell surface-bound plasmin, which cannot be inhibited by its natural inhibitors, such as alpha(2)-antiplasmin. This hybrid protein, consisting of the receptor-binding amino-terminal fragment of uPA (ATF), linked to the potent protease inhibitor bovine pancreas trypsin inhibitor (BPTI), can inhibit plasmin activity at the cell surface. METHODS AND RESULTS: The effect of adenovirus-mediated ATF.BPTI expression on neointima formation was tested in human saphenous vein organ cultures. Infection of human saphenous vein segments with Ad.CMV.ATF.BPTI (5x10(9) pfu/mL) resulted in 87.5+/-3.8% (mean+/-SEM, n=10) inhibition of neointima formation after 5 weeks, whereas Ad.CMV.ATF or Ad.CMV.BPTI virus had only minimal or no effect on neointima formation. The efficacy of ATF.BPTI in vivo was demonstrated in a murine model for neointima formation. Neointima formation in the femoral artery of mice, induced by placement of a polyethylene cuff, was strongly inhibited (93.9+/-2%) after infection with Ad.CMV.mATF.BPTI, a variant of ATF.BPTI able to bind specifically to murine uPA receptor; Ad.CMV.mATF and Ad.CMV.BPTI had no significant effect. CONCLUSIONS: These data provide evidence that adenoviral transfer of a hybrid protein that binds selectively to the uPA receptor and inhibits plasmin activity directly on the cell surface is a powerful approach to inhibiting neointima formation and restenosis.


Subject(s)
Aprotinin/physiology , Blood Vessels/physiology , Tunica Intima/growth & development , Urokinase-Type Plasminogen Activator/physiology , Adenoviridae/genetics , Animals , Aprotinin/genetics , CHO Cells , Cattle , Cricetinae , Femoral Artery/growth & development , Femoral Artery/injuries , Femoral Vein/cytology , Femoral Vein/metabolism , Fibrinolysin/metabolism , Gene Expression , Genetic Vectors/genetics , Humans , Mice , Mice, Inbred C57BL , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Organ Culture Techniques , Peptide Fragments/genetics , Peptide Fragments/physiology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/physiology , Saphenous Vein/cytology , Transfection , Urokinase-Type Plasminogen Activator/chemistry , Urokinase-Type Plasminogen Activator/genetics
14.
Thromb Haemost ; 84(3): 460-7, 2000 Sep.
Article in English | MEDLINE | ID: mdl-11019972

ABSTRACT

Smooth muscle cell migration plays a role in the development of intimal hyperplasia. Given the established role of the plasminogen activation system in cell migration, an approach to therapy is to overexpress an inhibitor of plasmin. Therefore, an adenoviral vector was constructed encoding the hybrid protein ATF.BPTI, which contains the active domain of bovine pancreas trypsin inhibitor (BPTI), fused to ATF, the amino terminal fragment or receptor-binding domain of u-PA. Adenoviral vectors expressing ATF and BPTI individually were also constructed, and a fourth vector was constructed encoding ATF.BPTI linked by an internal ribosomal entry site to Green Fluorescent Protein (ABIG). Both the expression and functionality of the recombinant proteins were established in human vascular smooth muscle cells. Adenoviral gene transfer of ATF.BPTI inhibited SMC migration more efficiently than the expression of ATF or BPTI individually. Expression of ABIG resulted in the co-expression of ATF.BPTI and Green Fluorescent Protein, thereby providing a tool to monitor transfection efficiency and the behavior of the transfected cells.


Subject(s)
Antifibrinolytic Agents/metabolism , Gene Transfer Techniques , Luminescent Proteins/genetics , Trypsin Inhibitors/genetics , Urokinase-Type Plasminogen Activator/genetics , Adenoviridae/genetics , Animals , Cattle , Cell Division/drug effects , Cell Movement/drug effects , Encephalomyocarditis virus/genetics , Fibrinolysin/antagonists & inhibitors , Fibrinolysin/metabolism , Fibrinolytic Agents/metabolism , Genetic Vectors , Green Fluorescent Proteins , Humans , Indicators and Reagents , Luminescent Proteins/metabolism , Membrane Proteins/metabolism , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Pancreas , Plasminogen Activators/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Saphenous Vein/cytology , Saphenous Vein/metabolism , Trypsin Inhibitors/metabolism , Trypsin Inhibitors/pharmacology , Urokinase-Type Plasminogen Activator/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...