Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Acta Academiae Medicinae Sinicae ; (6): 633-640, 2015.
Article in Chinese | WPRIM | ID: wpr-289933

ABSTRACT

<p><b>OBJECTIVE</b>To investgate the effects of rapamycin(RPM)and RPM-loaded poly(lactic-co-glycolic)acid(PLGA)nanoparticles(NPs)on the apoptosis of human umbilical arterial vascular smooth muscle cells(HUASMCs)in vitro and expression of bcl-2 and p27(kip1) protein.</p><p><b>METHODS</b>HUASMCs were cultured in vitro and divided to RPM and RPM-PLGA-NPs groups treated at 3 different concentration by 12 and 24 hours,with M231-smooth muscle growth supplements medium and null-PLGA-NPs treated groups as controlled. The apoptosis of HUASMCs was determined by terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick end labeling staining and flow cytometry. The expressions of bcl-2 and p27(kip1) were detected by streptacidin/peroxidase immunohistochemical method. The effect on cellular proliferation was assessed by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromidecolorimetry.</p><p><b>RESULTS</b>The proliferation of HUASMCs was inhibited by RPM and RPM-PLGA-NPs in a dose-dependent manner. DNA electrophoresis showed DNA ladder in RPM and RPM-PLGA-NPs groups and classical scalar strips in control groups. The apoptotic indexes of RPM 100 ng/ml group and RPM-PLGA-NPs 500 ng/ml group detected by flow cytometry were(45.45<2.36)% and(35.04<5.64)%,respectively,which were significantly higher than that of M231-smooth muscle growth supplements control group [(2.60<0.95)%,all P<0.01]. The apoptotic indexes of groups incubated with RPM and RPM-PLGA-NPs for 24 hours were significantly higher than those of groups which incubated for 12 hours(P<0.05,P<0.01). The positive expression indexes(PEI)of p27(kip1) and bcl-2 protein were higher in RPM and RPM-PLGA-NPs groups than that of control groups. The Spearman's rank correlation coefficient test showed that there was no significant correlation between the PEI of p27(kip1) and the apoptotic indexes in the RPM group and RPM-PLGA-NPs group(P>0.05).</p><p><b>CONCLUSIONS</b>Rapamycin-loaded PLGA nanoparticles and rapamycin have similar effects in inhibiting proliferation and inducing apoptosis;meanwhile,they upregulate the expression of p27(kip1) protein without downregulating the expression of bcl-2 protein in HUASMCs in vitro. RPM-PLGA-NPs has more potent pro-apoptotic effect than equivalent dose of RPM but is not linearly correlated with the p27(kip1) expression level.</p>


Subject(s)
Humans , Apoptosis , Cell Proliferation , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p27 , In Situ Nick-End Labeling , Lactic Acid , Muscle, Smooth, Vascular , Myocytes, Smooth Muscle , Nanoparticles , Polyglycolic Acid , Sirolimus , Umbilical Arteries
2.
Acta Academiae Medicinae Sinicae ; (6): 32-38, 2010.
Article in Chinese | WPRIM | ID: wpr-301598

ABSTRACT

<p><b>OBJECTIVE</b>To evaluate the effects of rapamycin (RPM)-loaded poly (lactic-co- glycolic) acid (PLGA) nanoparticles (NPs) on the proliferation, distribution of cell cycle, and expression of p27 protein in human umbilical arterial vascular smooth muscle cell (HUASMC) in vitro.</p><p><b>METHODS</b>The primarily culture model of HUASMC was successfully established by explant-attached method in vitro. The cells were administrated with different doses of RPM, and RPM-PLGA NPs were observed as treat groups compared with PLGA NPs and M231-SMGs medium cultured group. The effect of RPM-PLGA NPs on proliferation of HUASMC was assessed using 3-(4,5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide (MTT) colorimetry method. The influences of RPM-PLGA NPs on the cell cycle and cellular growth kinetics of HUASMCs were tested by flow cytometry. The effect of RPM-PLGA NPs on the expression of p27 protein of HUASMCs was assessed through an immunohistochemical method.</p><p><b>RESULTS</b>Compared with the control group, the proliferation of HUASMCs was inhibited by 50 microg/L and higher concentration of RPM-PLGA NPs in a dose-dependent manner (P < 0.05). The numbers of cells entering cell cycle of S/G2/M phases were significantly lower in RPM-PLGA NPs and RPM treated groups. Histologically, the expression of p27 were up-regulated in 500 microg/L RPM-PLGA NPs and 100 microg/L RPM treated group (all P < 0.01 ) when compared with the control group.</p><p><b>CONCLUSIONS</b>RPM-PLGA NPs has a similar effects as RPM in inhibiting the growth of in vitro cultured HUASMC. It can remarkably suppress the expression of in vitro cultured HUASMC p27 protein, arrest its cell cycle at G1/S phase, and inhibit its proliferation.</p>


Subject(s)
Humans , Cell Cycle , Cell Proliferation , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p27 , Metabolism , Drug Carriers , Lactic Acid , Muscle, Smooth, Vascular , Cell Biology , Myocytes, Smooth Muscle , Cell Biology , Metabolism , Nanoparticles , Polyglycolic Acid , Sirolimus , Pharmacology , Umbilical Arteries , Cell Biology
3.
Acta Academiae Medicinae Sinicae ; (6): 578-582, 2008.
Article in Chinese | WPRIM | ID: wpr-270645

ABSTRACT

<p><b>OBJECTIVE</b>To prepare a biodegradable drug-eluting stent in myocardium channel and evaluate its effect on myocardium channel after transmyocardial revascularization (TMR).</p><p><b>METHODS</b>A biodegradable drug-eluting stent was prepared using poly (epsilon-caprolactone) (PCL), bovine serum albumin (BSA), and poly (D, L-lactide-co-glycolide) (PLGA) as material of stent, model protein drug, and drug carrier respectively. The amount of BSA in stent and in vitro released BSA of stent were determined by the Coomassie brilliant blue assay. The mechanical strength of stent was tested by universal material testing machines. The material and structure of stent was characterized by nuclear magnetic resonance spectroscopy. The effect of stent on myocardium channel after TMR was evaluated in vivo by a standard animal model of chronic myocardial ischemia in miniswine.</p><p><b>RESULTS</b>The stent could carry 13.1 microg BSA per mg of stent and the stent could release about 95% of BSA after 30 days. The stent diminished 80% of initial scale under the stress of 1.7 Mpa. It also kept the myocardium channel patency after TMR.</p><p><b>CONCLUSIONS</b>A biodegradable drug-eluting stent in myocardium channel was successfully prepared. It can sustain the pressure from the heart and achieve the controlled release of drug. The stent can ensure the myocardium channel patency after TMR.</p>


Subject(s)
Animals , Humans , Biocompatible Materials , Chemistry , Blood Vessel Prosthesis , Caproates , Chemistry , Cardiac Surgical Procedures , Disease Models, Animal , Drug Delivery Systems , Heart , Lactones , Chemistry , Myocardial Ischemia , Drug Therapy , General Surgery , Myocardial Revascularization , Random Allocation , Swine , Swine, Miniature
4.
Acta Academiae Medicinae Sinicae ; (6): 491-497, 2008.
Article in Chinese | WPRIM | ID: wpr-270663

ABSTRACT

<p><b>OBJECTIVE</b>To sought to engineer and characterize a biodegradable nanoparticles (NPs) containing rapamycin which use poly (lactic-co-glycolic) acid (PLGA) as the carrier matrix and to assess its in vivo release characteristics by local drug delivery system intravascularly.</p><p><b>METHODS</b>Rapamycin-loaded PLGA NPs were prepared by an emulsification/solvent evaporation technique, and NPs size distribution was assessed by submicro laser defractometer. The particle morphology was observed by scanning electron microscopy. In vitro release from the NPs was performed in TE buffer at 37 degrees C under rotation utilizing double-chamber diffusion cells on a shake stander. In vivo NPs intravascular local delivery were performed by DISPATCH catheter in New Zealand rabbit abdominal aorta and Chinese experimental mini-pigs coronary artery models.</p><p><b>RESULTS</b>Biodegradable rapamycin loaded PLGA NPs were constructed successfully by emulsification solvent-evaporation technique. The diameter of rapamycin-PLGA NPs was around 246.8 nm with very narrow size distribution, and rapamycin-NPs showed good spherical shape with smooth uniform surface. Rapamycin loaded in NPs were around was 19.42%. Encapsulation efficiency of drug was over 77.53%. The in vitro release of rapamycin from NPs showed that 75% of the drug was sustained released over 2 weeks and controlled release in a linear pattern. After a single 10 minutes infusion of rapamycin-PLGA NPs suspension (5 mg/ml) under 20.27 kPa through DISPATCH catherter in vivo, the mean rapamycin levels at 7 day and 14 day were (2.438 +/- 0.439) and (0.529 +/- 0.144) microg/mg of the dry-weight of the artery segments (2 cm) which local delivery were administrated.</p><p><b>CONCLUSIONS</b>PLGA NPs controlled drug delivery system for intraarterial local anti-proliferative drug delivery can potentially improve local drug concentration and prolong drug residence time in animal model in vivo. It should be appropriate for further study of its therapy efficiency in human.</p>


Subject(s)
Animals , Rabbits , Aorta, Abdominal , Coronary Vessels , Drug Carriers , Chemistry , Drug Delivery Systems , Methods , Infusions, Intra-Arterial , Lactic Acid , Chemistry , Nanoparticles , Chemistry , Particle Size , Polyglycolic Acid , Chemistry , Sirolimus , Pharmacokinetics , Swine , Swine, Miniature
5.
Acta Pharmaceutica Sinica ; (12): 81-86, 2007.
Article in Chinese | WPRIM | ID: wpr-281921

ABSTRACT

The novel paclitaxel-loaded nanopaticle through surface modification with didodecylmethylammonium bromide (DMAB) was prepared and its prevenative against neointimal formation in a rabbit carotid artery injury model was tested. Paclitaxel-loaded nanoparticles were prepared from oil-water emulsions using biodegradable poly (lactic acid-co-glycolic acid) (PLGA). Specific additive for surface conjugation was added after particle formation. To enhance arterial retention using a cationic surfactant, DMAB, was used. The size and distribution, surface morphology and surface charge of the paclitaxel-loaded nanoparticles were then investigated by laser light scattering, scanning electron microscope and zeta potential analyzer. The drug encapsulation efficiency (EE) and in vitro release profile were measured by high-performance liquid chromatography (HPLC). Balloon injured rabbit carotid arteries were treated with single infusion of the paclitaxel-loaded NP suspension and observed for 28 days. The inhibitory effects of vascular smooth muscle cell migration and proliferation were evaluated as end-point. The NPs showed spherical shape with diameter ranging from 200 to 500 nm. The negatively charged PLGA NPs shifted to positive after the DMAB modification. The in vitro drug release profile showed a triphasic release pattern. 28 days later, morphologic analysis revealed that the inhibitory effect of intima proliferation is dose-dependent, and the 30 mg x mL(-1) nanoparticle concentration suspension could completely inhibit proliferation of intima. Paclitaxel-loaded nanoparticles through surface modification with DMAB provide an effective means of inhibiting proliferation response to vascular injury in the rabbit.


Subject(s)
Animals , Female , Male , Rabbits , Antineoplastic Agents, Phytogenic , Chemistry , Therapeutic Uses , Carotid Artery Injuries , Pathology , Catheterization , Delayed-Action Preparations , Drug Compounding , Drug Delivery Systems , Hyperplasia , Lactic Acid , Chemistry , Nanoparticles , Nanotechnology , Paclitaxel , Chemistry , Therapeutic Uses , Particle Size , Polyglycolic Acid , Chemistry , Tunica Intima , Wounds and Injuries , Pathology
6.
Acta Academiae Medicinae Sinicae ; (6): 475-480, 2006.
Article in Chinese | WPRIM | ID: wpr-313749

ABSTRACT

<p><b>OBJECTIVE</b>To evaluate the effect of antisense monocyte chemotactic protein-1 (A-MCP-1) nanoparticles (NPs) as gene carrier on gene transfer in two kinds of animal models.</p><p><b>METHODS</b>Poly (lactic acid-co-glycolic acid) (PLGA) was used to make the NPs loaded with A-MCP-1 through a double-emulsion/solvent evaporation technique. NPs size was assessed by dynamic laser defractometer. The particle morphology was observed by scanning electron microscopy. DNA content in the NPs was measured by dissolving known amounts of NPs in chloroform and extracting DNA with water. In vitro release was performed in tris-EDTA buffer at 37 degrees C using double-chamber diffusion cells. The receiver buffer was replaced daily. The A-MCP-1 NPs was transfected into the cultured smooth muscle cells. PCR was used to evaluate the transfection of A-MCP-1. Cationic lipid (Lipofectamine) was used to transfect A-MCP-1 as control. After 48 hours incubation, cells were digested and examined by polymerase chain reaction. Twenty New Zealand white rabbits under jugular vein to artery bypass grafting procedure were divided into four groups: the first group received grafts treated with A-MCP-1 NPs, the second group received grafts treated with cationic liposome (dioleoyl trimethyl ammonium propane)-A-MCP-1, the third group received grafts treated with plasmid DNA, and the fourth group received grafts without transfection as control. Fourteen days after surgery the grafts were harvested. The expression of A-MCP-1 and its effect on MCP-1 in vein grafts were detected by dot blotting. The morphology of the grafts was investigated. To establish abdominal aortic aneurysms rats model, rats were randomly divided into three groups: A-MCP-1 NPs injection group, shame NPs injection group and control groups (without injection). Two weeks after surgery, diameter of abdominal aorta was measured and aortic tissue was obtained for PCR analysis to evaluate the A-MCP-1 expression. Western blot were applied to detect the inhibitory effect to the expression of MCP-1 mRNA and CD68 protein by A-MCP-1 NPs.</p><p><b>RESULTS</b>NPs size ranged 198nm to 205nm with average around 201.4 nm. DNA content in the NPs was 4.14%. NPs showed steady release rate in vitro in Tris-EDTA solution. It released faster in the first week then maintained a slowly sustained release up to 16 days. In cell culture A-MCP-1 gene successfully transfected into smooth muscle cells by NPs vector. In vein grafting animal model, A-MCP-1 expression was detected in the vascular walls of NPs and cationic lipid treated groups. The degree of vascular hyperplasia in the gene NPs treated group was significantly lower than that in control group. There was no significant difference in the inhibition of intimal hyperplasia between NPs and cationic lipid treated groups. Two weeks after transfection in abdominal aortic aneurysm rats models, the abdominal aortic diameter of A-MCP-1 NPs injection group was (1.79 +/- 0.12) mm, significantly smaller than that of control groups [shame NPs group was (2.58 +/- 0.21) mm, and saline group was (2.63 +/- 0.29) mm] (P < 0.01). The expressions of MCP-1 mRNA and CD68 protein in A-MCP-1 NPs injection group were 12.5 +/- 1.5 and 17.6 +/- 2.1, which were much lower than those in control group [in shame NPs group, which were 35.7 +/- 4.5, 42.3 +/- 5.7 (P < 0.01), and saline group which is 32.4 +/- 3.9, 39.8 +/- 4.8 (P < 0.01)]. Specific band of A-MCP-1 was detected only in the A-MCP-1 NPs injection group by PCR.</p><p><b>CONCLUSION</b>A-MCP-1 gene NPs can be successfully used in rabbit vein grafting model and abdominal aortic aneurysm rats models, and may be potentially applied in clinical practice.</p>


Subject(s)
Animals , Rabbits , Rats , Aortic Aneurysm , Genetics , Chemokine CCL2 , Genetics , Metabolism , Gene Transfer Techniques , Genetic Vectors , Lactic Acid , Chemistry , Models, Animal , Nanoparticles , Oligonucleotides, Antisense , Genetics , Polyglycolic Acid , Chemistry , Polymers , Chemistry , Transfection
7.
Acta Academiae Medicinae Sinicae ; (6): 665-669, 2006.
Article in Chinese | WPRIM | ID: wpr-313710

ABSTRACT

<p><b>OBJECTIVE</b>To explore the feasibility of using an endovascular metal stent as a highly efficient and site-specific gene delivery system.</p><p><b>METHODS</b>Stents were formulated with a collagen coating. Anti-DNA monoclonal antibodies were covalently bound to the collagen surface by a cross linking reagent of N-succinimidyl 3-(2-pyridyldithio) propionate (SPDP). Binding capacity and stability of antibody and plasmid DNA on stents were quantified by radioactive labeling. The gene transduction efficiency was evaluated in cell culture and in rabbits.</p><p><b>RESULTS</b>The amount of antibodies binding on collagen matrix through SPDP reaction was 15 times higher than that of through physical absorption (P < 0.005). The binding stability of plasmid was significantly better than the control groups (P < 0.01). There was no harmful effect on cell growth with the anti-DNA antibody modified stents. The stents retrieved from cell culture after 72 hours of incubation in A10 cells showed numerous transducted cells only infiltrating the surface coating indicating a highly localized and efficient gene delivery pattern. Results of in vivo gene transfer by this modified stent revealed (2.8 +/- 0.7)% of total cells transduction and the higher transduction location was neointimal layer (about 7%). No distal spread of vector was detectable in the anti-DNA antibody modified stent implantation animals.</p><p><b>CONCLUSIONS</b>Anti-DNA antibody modified stents represent a novel highly efficient and site-specific gene delivery system which can deliver various kinds of plasmid vectors. The release of plasmid DNA tethered on the stents could be controlled in some conditions. This novel system provided a novel platform for cardiovascular site-specific gene therapy.</p>


Subject(s)
Animals , Male , Mice , Rabbits , Antibodies, Antinuclear , Allergy and Immunology , Antibodies, Monoclonal , Allergy and Immunology , Cells, Cultured , Coated Materials, Biocompatible , Collagen , DNA , Genetics , Gene Transfer Techniques , Genetic Vectors , Plasmids , Stainless Steel , Stents
8.
Acta Academiae Medicinae Sinicae ; (6): 682-685, 2006.
Article in Chinese | WPRIM | ID: wpr-313707

ABSTRACT

<p><b>OBJECTIVE</b>To explore the feasibility of utilizing two implantable devices made from modified polyurethane films with antibody tethered replication-defective adenoviruses encoding for green fluorescent protein (AdGFP) as gene delivery platforms.</p><p><b>METHODS</b>Intra-aortic button implants of collagen-coated polyurethane films with antibody tethered AdGFP were sutured into the infrarenal aorta of adult pigs and pulmonary valve leaflet in juvenile sheep was replaced by polyurethane pulmonary valve cusp replacement with antibody-tethered AdGFP. After seven days, the buttons, prosthetic leaflets, and their surrounding tissues were explanted and evaluated for biocompatibility and AdGFP-mediated gene transfer by fluorescent microscopy and PCR analysis.</p><p><b>RESULTS</b>In vivo analysis of gene transfer from collagen-coated polyurethane films in pig infrarenal aorta implants, one week explants of the collagen-coated polyurethane films demonstrated (14.2 +/- 2.5)% of neointimal cells on the surface of the implant. In sheep pulmonary valve leaflet replacement studies, polyurethane films with antibody tethered AdGFP vector demonstrated (25.1 +/- 5.7)% of cells attached to polyurethane valve leaflets were transduced in one week. PCR analyses showed that GFP DNA was not detectable in blood or distal tissues.</p><p><b>CONCLUSION</b>Site-specific intravascular delivery of adenoviral vectors for gene therapy can be achieved with these two kinds of polyurethane implants utilizing the antivector antibody tethering mechanism.</p>


Subject(s)
Animals , Male , Adenoviridae , Genetics , Blood Vessel Prosthesis , Gene Transfer Techniques , Genetic Therapy , Methods , Genetic Vectors , Green Fluorescent Proteins , Genetics , Heart Valve Prosthesis , Polyurethanes , Chemistry , Prosthesis Implantation , Sheep , Swine
9.
Acta Academiae Medicinae Sinicae ; (6): 845-848, 2006.
Article in Chinese | WPRIM | ID: wpr-313671

ABSTRACT

Electroactive and/or electrically conductive polymers have shown potential applications in the culture of excitable cells and as the electroactive scaffolds for neuronal or cardiac tissue engineering. The biocompatibility of the conductive polymer can be improved by covalently grafting or blending with oligo- or polypeptides. The new progresses in this area on two types of conductive polymers, polypyrrole and polyaniline (PANi) are reviewed in this paper. The studies of oligopeptide-modified PANi and electrospun PANi/gelatin nanofibers are highlighted.


Subject(s)
Animals , Mice , Rats , Aniline Compounds , Chemistry , Biocompatible Materials , Chemistry , Cells, Cultured , Materials Testing , Polymers , Chemistry , Pyrroles , Chemistry , Tissue Engineering
10.
Acta Academiae Medicinae Sinicae ; (6): 350-354, 2006.
Article in Chinese | WPRIM | ID: wpr-281200

ABSTRACT

<p><b>OBJECTIVE</b>To study the feasibility of delivering viral gene vector from a collagen-coated polyurethane (PU) film through a mechanism involving monoclonal antiviral antibody tethering.</p><p><b>METHODS</b>Anti-adenoviral monoclonal antibodies were covalently bound to the collagen-coated PU surface. These antibodies enabled tethering of replication defective adenoviruses through highly specific antigen-antibody affinity. The PU film-based gene delivery using antibody-tethered adenovirus encoding green fluorescent protein (GEP) was tested in rat arterial smooth muscle cell (A10 cell) culture in vitro. The virus binding stability was studied by incubating the collagen-coated PU film in PBS solution at 37 degrees C for 20 days, followed with A10 cell cultures with the incubated films and the corresponding buffer solution.</p><p><b>RESULTS</b>PU films with antibody-tethered adenovirus encoding GFP demonstrated efficient and highly localized gene delivery to A10 cells. Virus binding was stable for at least 10 days at physiological conditions, more than 77% of the originally bound virus remained in the film after 15 day's incubation.</p><p><b>CONCLUSION</b>Gene delivery using PU film-based anti-viral antibody tethering of vectors exhibited potentials of applications in a wide array of single or multiple therapeutic gene strategies, and in further stent-based gene delivery therapeutic strategies.</p>


Subject(s)
Humans , Adenoviridae , Genetics , Antibodies, Monoclonal , Allergy and Immunology , Antibody Specificity , Allergy and Immunology , Gene Transfer Techniques , Genetic Therapy , Genetic Vectors , Polyurethanes , Chemistry , Protein Binding
11.
Acta Pharmaceutica Sinica ; (12): 557-562, 2005.
Article in Chinese | WPRIM | ID: wpr-353473

ABSTRACT

<p><b>AIM</b>To evaluate the effects of an array of additives on drug release from double-layered poly(lactic-co-glycolic acid) (PLGA) matrices.</p><p><b>METHODS</b>Additives differing in molecular size, hydrophilicity and steric configuration were selected for this study. An anti-proliferative 2-aminochromone, U-86983 (U-86, Pharmacia and Upjohn), was used as a model agent because of our interest in investigating local drug delivery systems for the inhibition of restenosis.</p><p><b>RESULTS</b>In vitro release of U-86 PLGA matrices without additive showed a typical biphasic release kinetics, i.e. a slow diffusion release (Phase I) followed by a fast erosion-mediated release (Phase II). The water-soluble additives in PLGA matrices changed the biphasic release pattern to a near monophasic profile by increasing the release of the Phase I. Increasing the ratio of additives to PLGA in matrices caused a significant increase in U-86 release rates. A high molecular weight water-soluble additive, Pluronic F127, resulted in a matrix showing perfect zero-order release kinetics. The morphologic evaluation of matrices using scanning electron microscopy indicated that the water-soluble additives were leachable and thus generated a highly porous structure in the matrices. Conclusion Water-solubility, molecular size and steric configuration of additives are the important determinants in generating various types of pore structures in polymer matrix which in turn affect the release mechanism and release kinetics.</p>


Subject(s)
Biocompatible Materials , Chemistry , Chromones , Chemistry , Delayed-Action Preparations , Drug Carriers , Drug Delivery Systems , Excipients , Chemistry , Lactic Acid , Chemistry , Molecular Weight , Morpholines , Chemistry , Pharmacokinetics , Poloxamer , Chemistry , Polyglycolic Acid , Chemistry , Polymers , Chemistry , Tartrates , Chemistry
12.
Acta Academiae Medicinae Sinicae ; (6): 718-722, 2005.
Article in Chinese | WPRIM | ID: wpr-318829

ABSTRACT

<p><b>OBJECTIVE</b>To evaluate the feasibility and stability of chemically conjugating IgM on collagen films.</p><p><b>METHODS</b>IgM was labeled with 125I using the chloramine-T method. Six collagen films were randomly divided into two groups. In chemical coupling group 125I-labeled IgM was chemically coupled with the films through N-succinmiclyl-3- (2-pyridyl-dithio) propionate reaction. In control group 125I-labeled IgM was absorbed onto collagen films. The amount of IgM on the collagen films and the amount of IgM remained on the films after extensive rinsing with phosphate buffered saline were monitored by counting the radioactivity of 125I.</p><p><b>RESULTS</b>The amount of antibodies loaded onto collagen films in the chemical coupling group was 15 times higher than that on the control films, showing significant statistical difference (P < 0.01). And the stability of conjugation antibodies on collagen films was significantly better than the control films.</p><p><b>CONCLUSION</b>Chemical coupling is an effective approach to immobilize antibodies on collagen for further plasmid DNA tethering.</p>


Subject(s)
Animals , Cattle , Mice , Angioplasty, Balloon, Coronary , Antibodies, Antinuclear , Metabolism , Coated Materials, Biocompatible , Chemistry , Metabolism , Collagen , Chemistry , Metabolism , Genetic Vectors , Immunoglobulin M , Metabolism , In Vitro Techniques , Protein Binding , Stents , Surface Properties
13.
Acta Academiae Medicinae Sinicae ; (6): 190-193, 2005.
Article in Chinese | WPRIM | ID: wpr-343741

ABSTRACT

<p><b>OBJECTIVE</b>To explore the possibility of using an endovascular microcoil as a gene delivery system.</p><p><b>METHODS</b>Anti-adenoviral monoclonal antibodies were covalently attached to the collagen-coated surface of platinum microcoil. These antibodies were used to tether adenovirus encoding green fluorescent protein (Ad-GFP). Cell culture studies with rat arterial smooth muscle cells (A10) assessed transduction on or near the coil. Platinum coils coated with Ad-GFP were implanted into the ligated common carotid artery (CCA) of adult rats in a model of arterial stasis and pressurization. After 7 days, CCA segments were harvested, and coils were removed for histopathology and GFP expression studies, while organs were evaluated by polymerase chain reaction to assess viral biodistribution.</p><p><b>RESULTS</b>In cell culture studies, GFP-positive smooth muscle cells were detected only on the platinum coil surface. After 7 days, GFP was detected on the harvested platinum coil and in the organizing thrombus within the CCA according to fluorescence microscopy and immunohistochemistry. Morphometric analyses revealed that (13.3 +/- 2.0)% of cells within the organized thrombus were transduced with Ad-GFP via the gene delivery system. Ad-GFP was not detectable by polymerase chain reaction in lung, liver, or kidney.</p><p><b>CONCLUSIONS</b>Gene delivery endovascular microcoils represents an interventional device-based gene therapy system that can serve as a suitable platform for either single or multiple gene therapy vectors.</p>


Subject(s)
Animals , Rats , Adenoviridae , Genetics , Allergy and Immunology , Aneurysm , General Surgery , Antibodies, Viral , Chemistry , Metabolism , Biological Availability , Carotid Artery, Common , Metabolism , General Surgery , Cells, Cultured , Drug Delivery Systems , Embolization, Therapeutic , Methods , Endothelial Growth Factors , Therapeutic Uses , Genetic Therapy , Methods , Genetic Vectors , Chemistry , Green Fluorescent Proteins , Muscle, Smooth, Vascular , Cell Biology , Platinum , Chemistry , Rats, Sprague-Dawley , Recombinant Proteins , Therapeutic Uses , Transduction, Genetic , Methods
14.
Chinese Journal of Surgery ; (12): 58-61, 2004.
Article in Chinese | WPRIM | ID: wpr-311151

ABSTRACT

<p><b>OBJECTIVE</b>To experimentally investigate direct intramuscular gene transfer of nanoparticles encoding vascular endothelial growth factor for the treatment of peripheral artery disease.</p><p><b>METHODS</b>The human VEGF(165) cDNA was cloned into the eukaryotic expression vector PIRES2 under the control of cytomegalovirus promoter/enhancer. The recombinant gene was transferred into a rabbit model of chronic hindlimb ischemia by naked plasmid and nanoparticle respectively. Ischemia was induced in the hindlimb of New Zealand White rabbits by ligation of the distal external iliac artery and complete excision of the femoral artery and all its branches. At day 7 postoperation animals received VEGF(165) plasmid (10 intramuscular) or nanoparticle-VEGF(165) (8 intramuscular). With RT-PCR, immunohistochemistry analysis, and angiography, the expression and biological effects of VEGF(165) gene in experimental animals were investigated.</p><p><b>RESULTS</b>Two weeks after initiation of therapy, angiography showed that the transfer of VEGF(165) gene stimulated the formation of focal neovessels and established collateral circulation. The adductor muscle of ischemic limbs was histologically examined at day 14. Capillary density was increased among VEGF(165)-transfected rabbits, especially Nano-VEGF(165)-treated animals (Naked VEGF(165) plasmid = 50.18 per mm(2), Nano-VEGF(165) = 81.22 per mm(2), Control = 29.54 per mm(2), P < 0.05). RT-PCR showed that the transcription and expression of VEGF(165) gene in experimental group were significantly higher than those of control groups.</p><p><b>CONCLUSIONS</b>Intramuscular administration of VEGF(165) induces collateral artery augmentation in the rabbit model of chronic limb ischemia. Nanoparticle can act as a vector to transfect specific gene and it will benefit gene transfer.</p>


Subject(s)
Animals , Male , Rabbits , Angiography , Capsules , Chronic Disease , Disease Models, Animal , Genetic Therapy , Methods , Hindlimb , Immunohistochemistry , Ischemia , Genetics , Therapeutics , Nanotechnology , Particle Size , Reverse Transcriptase Polymerase Chain Reaction , Treatment Outcome , Vascular Endothelial Growth Factor A , Genetics , Therapeutic Uses
SELECTION OF CITATIONS
SEARCH DETAIL