Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Circulation ; 94(12): 3281-90, 1996 Dec 15.
Article in English | MEDLINE | ID: mdl-8989142

ABSTRACT

BACKGROUND: Striated muscle has been shown to be capable of taking up and expressing foreign genes transferred in the form of naked plasmid DNA, although typically with a low level of gene expression. In the case of genes that encode secreted proteins, however, low transfection efficiency may not preclude bio-activity of the secreted gene product. Accordingly, we investigated the hypothesis that intramuscular (IM) gene therapy with naked plasmid DNA encoding vascular endothelial growth factor (VEGF) could augment collateral development and tissue perfusion in an animal model of hindlimb ischemia. METHODS AND RESULTS: Ten days after ischemia was induced in one rabbit hindlimb, 500 micrograms of phVEGF165, or the reporter gene LacZ, was injected IM into the ischemic hindlimb muscles. Thirty days later, angiographically recognizable collateral vessels and histologically identifiable capillaries were increased in VEGF transfectants compared with controls. This augmented vascularity improved perfusion to the ischemic limb, documented by a superior calf blood pressure ratio for phVEGF165 (0.85 +/- 0.05) versus controls (0.64 +/- 0.05, P < .01), improved blood flow in the ischemic limb (measured with an intra-arterial Doppler wire) at rest (phVEGF165 = 21.3 +/- 3.9 mL/min, control = 14.6 +/- 1.6 mL/min, P < .01) and after a vasodilator (phVEGF165 = 54.2 +/- 12.0 mL/min, control = 37.3 +/- 8.9 mL/min, P < .01) and increased microspheres in the adductor (phVEGF165 = 4.3 +/- 1.6 mL.min-1.100 g of tissue-1, control = 2.9 +/- 1.2 mL.min-1.100 g of tissue-1, P < .05) and gastrocnemius (phVEGF165 = 3.9 +/- 1.0 mL.min-1.100 g of tissue-1, control = 2.8 +/- 1.4 mL.min-1.100 g of tissue-1, P < .05) muscles of the ischemic limb. CONCLUSIONS: Ischemic skeletal muscle represents a promising target for gene therapy with naked plasmid DNA. IM transfection of genes encoding angiogenic cytokines, particularly those that are naturally secreted by intact cells, may constitute an alternative treatment strategy for patients with extensive peripheral vascular disease in whom the use of intravascular catheter-based gene transfer is compromised and/or prohibited.


Subject(s)
Collateral Circulation , DNA, Complementary/administration & dosage , Endothelial Growth Factors/biosynthesis , Endothelial Growth Factors/genetics , Genetic Therapy , Ischemia/therapy , Lymphokines/biosynthesis , Lymphokines/genetics , Muscle, Skeletal/blood supply , Transfection , Animals , Base Sequence , Blood Pressure , DNA Primers , Gene Expression , Genes, Reporter , Hindlimb/blood supply , Humans , Injections, Intramuscular , Ischemia/physiopathology , Male , Plasmids/administration & dosage , Polymerase Chain Reaction , RNA, Messenger/analysis , RNA, Messenger/biosynthesis , Rabbits , Recombinant Proteins/biosynthesis , Regional Blood Flow , Transcription, Genetic , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors , beta-Galactosidase/biosynthesis
2.
Circulation ; 94(12): 3291-302, 1996 Dec 15.
Article in English | MEDLINE | ID: mdl-8989143

ABSTRACT

BACKGROUND: Delinquent reendothelialization (rET) has been shown to have a permissive, if not facilitatory, impact on smooth muscle cell proliferation. This inverse relation has been attributed to certain functions of the endothelium, including barrier regulation of permeability, thrombogenicity, and leukocyte adherence, as well as production of growth-inhibitory molecules. Accordingly, the present investigation was designed to test the hypothesis that an endothelial cell (EC) mitogen could serve as the basis for a novel gene therapy strategy designed to facilitate EC regeneration, reduce neointimal thickening, and promote recovery of EC dysfunction after balloon injury. METHODS AND RESULTS: New Zealand White rabbits underwent simultaneous balloon injury and gene transfer of one femoral artery with phVEGF165, encoding the 165-amino acid isoform of vascular endothelial growth factor (VEGF), or pGSVLacZ. In each animal transfected with phVEGF165 or pGSVLacZ, the contralateral femoral artery was also subjected to balloon injury but not to gene transfer. For pGSVLacZ, rET remained incomplete at 4 weeks after transfection; in contrast, phVEGF165 produced prompt rET, which was 95% complete by 1 week. Furthermore, rET in the contralateral, balloon-injured, nontransfected limb of the VEGF group was similarly accelerated. Consequently, intimal thickening was diminished, thrombotic occlusion was less frequent, and recovery of EC-dependent vasomotor reactivity was accelerated in VEGF transfectants compared with control animals. A similar benefit was observed for the contralateral, balloon-injured, nontransfected limb. CONCLUSIONS: Catheter-mediated, site-specific arterial gene transfer of phVEGF165 can accelerate rET at local and remote sites, leading to inhibition of neointimal thickening, reduction in thrombogenicity, and restoration of endothelium-dependent vasomotor reactivity. These findings support the notion that gene transfer encoding for an EC-specific mitogen may be useful for preventing the complications, including restenosis, of balloon angioplasty.


Subject(s)
Endothelial Growth Factors/biosynthesis , Endothelial Growth Factors/genetics , Endothelium, Vascular/pathology , Femoral Artery/pathology , Genetic Therapy , Lymphokines/biosynthesis , Lymphokines/genetics , Neovascularization, Physiologic , Regeneration , Transfection , Angioplasty, Balloon , Animals , Base Sequence , DNA Primers , DNA, Complementary/administration & dosage , Gene Expression , Genes, Reporter , Humans , Lac Operon , Male , Plasmids/administration & dosage , Polymerase Chain Reaction , Rabbits , Recombinant Proteins/biosynthesis , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
3.
Am J Pathol ; 147(6): 1649-60, 1995 Dec.
Article in English | MEDLINE | ID: mdl-7495290

ABSTRACT

Proliferation of vascular cells has been previously shown to contribute to spontaneous development of coronary collaterals. Recent studies from several laboratories have established that collateral artery growth in both the heart and limb can be enhanced by administration of angiogenic growth factors, or therapeutic angiogenesis. In this study, we sought (1) to define the extent and time course of endothelial cell (EC) and smooth muscle cell (SMC) proliferation accompanying spontaneous collateral development during limb ischemia and (2) to determine the extent to which proliferative activity of ECs and SMCs is augmented during therapeutic angiogenesis with vascular endothelial growth factor (VEGF), a heparin-binding EC-specific mitogen. Ten days after induction of limb ischemia by surgically excising the femoral artery of rabbits, either VEGF (500 to 1000 micrograms) or saline was administered as a bolus into the iliac artery of the ischemic limb. Cellular proliferation was evaluated by bromodeoxyuridine labeling for 24 hours at day 0 (immediately before VEGF administration) and at days 3, 5, and 7 after VEGF, EC proliferation in the midzone collaterals of VEGF-treated animals increased 2.8-fold at day 5 (P < 0.05 versus control), and returned to baseline levels by day 7. SMC proliferation in midzone collaterals also increased 2.7-fold in response to VEGF (P < 0.05). No significant increase in EC or SMC proliferation was observed in either the stem or re-entry collaterals of VEGF-treated animals compared with untreated ischemic control animals. Reduction of hemodynamic deficit in the ischemic limb measured by lower limb blood pressure was documented at day 7 after VEGF (P < 0.01 versus untreated, ischemic control). These data thus (1) establish the contribution of cellular proliferation to collateral vessel development in limb ischemia and (2) support the concept that augmented cellular proliferation contributes to the enhanced formation of collateral vessels after therapeutic angiogenesis with VEGF.


Subject(s)
Endothelial Growth Factors/pharmacology , Endothelium, Vascular/drug effects , Extremities/blood supply , Lymphokines/pharmacology , Neovascularization, Physiologic/physiology , Animals , Cell Division/drug effects , Cell Division/physiology , Collateral Circulation/drug effects , Collateral Circulation/physiology , Extremities/injuries , Male , Models, Cardiovascular , Muscle, Smooth/drug effects , Neovascularization, Physiologic/drug effects , Rabbits , Time Factors , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
4.
Circulation ; 91(11): 2793-801, 1995 Jun 01.
Article in English | MEDLINE | ID: mdl-7758186

ABSTRACT

BACKGROUND: Most strategies designed to reduce restenosis by the use of pharmacological or biological reagents involve direct inhibition of vascular smooth muscle cell (SMC) proliferation. Alternatively, SMC proliferation might be indirectly inhibited if reendothelialization could be specifically facilitated at sites of balloon-induced arterial injury. Accordingly, we investigated the hypothesis that application of an endothelial cell (EC)-specific mitogen to a freshly denuded intimal surface could accelerate reendothelialization and thereby attenuate intimal hyperplasia. METHODS AND RESULTS: The left carotid artery of 31 Sprague-Dawley rats was subjected to balloon injury, after which 16 rats were treated with a 30-minute incubation with 100 micrograms of vascular endothelial growth factor (VEGF), an EC-specific mitogen. Control animals (n = 15) received a 30-minute incubation with 0.9% saline. At 2 weeks after balloon injury, carotid artery reendothelialization was markedly superior in the VEGF-treated group compared with the control group (14.59 +/- 1.12 versus 7.96 +/- 0.51 mm2, P < 0.005). The extent of reendothelialization measured at 4 weeks after balloon injury remained superior for arteries treated with VEGF (18.04 +/- 0.90 mm2) versus saline (13.42 +/- 0.84 mm2, P < .005). Neointimal thickening was correspondingly attenuated to a statistically significant degree in arteries treated with VEGF versus the control group at both the 2-week and 4-week time points. Immunostaining for proliferating cell nuclear antigen (PCNA) disclosed a threefold increase in PCNA-positive cells in the neointima of control arteries versus VEGF-treated arteries at 2 weeks after injury. CONCLUSIONS: Application of VEGF, an EC-specific growth regulatory molecule, may be effectively used in vivo to promote reendothelialization and thereby indirectly attenuate neointimal thickening due to SMC proliferation.


Subject(s)
Carotid Artery Injuries , Catheterization , Endothelial Growth Factors/administration & dosage , Endothelium, Vascular/physiology , Lymphokines/administration & dosage , Tunica Intima/drug effects , Animals , Carotid Artery, Common/ultrastructure , Endothelial Growth Factors/therapeutic use , Hyperplasia , Lymphokines/therapeutic use , Male , Microscopy, Electron , Proliferating Cell Nuclear Antigen/analysis , Rats , Rats, Sprague-Dawley , Regeneration/physiology , Tunica Intima/pathology , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
6.
Lab Invest ; 71(3): 387-91, 1994 Sep.
Article in English | MEDLINE | ID: mdl-7523763

ABSTRACT

BACKGROUND: Little information exists regarding the time course of gene expression after arterial transfection. Accordingly, we sought to determine the time course of gene expression after liposome-mediated arterial gene transfer (lipofectin) using an arterial organ culture model. EXPERIMENTAL DESIGN: Explanted segments of rabbit descending thoracic aorta were maintained in organ culture. Arterial gene transfer, facilitated by cationic liposomes (Lipofectin), was performed with the plasmid pXGH5 encoding the human growth hormone (hGH) under the control of mouse metallothionein-1 promoter. RESULTS: The time course of hGH production after transfection with the plasmid pXGH5 was evaluated. Significant levels (181.0 +/- 33.9 ng/24 hours/gm) of hGH were detected within 24 hours post-transfection, reached a peak on day 7 (238.4 +/- 35.3 ng/24 hours/gm), and declined after day 10. At day 21, hGH could be observed in 50% of the arteries. Immunostaining with a monoclonal antibody for hGH revealed that only a small number of arterial cells (< 1%) were responsible for production of hGH. CONCLUSIONS: The organ culture model is a feasible and efficient means for investigating the kinetics of arterial gene transfer. Transfection of pXGH5 results in significant levels of hGH for up to 3 weeks, despite anatomic evidence of only limited gene expression. These data thus support the notion that the magnitude and/or duration of gene expression may be disproportionately high, relative to anatomic assessment of transfection efficiency in the case of a transgene encoding for a secreted protein.


Subject(s)
Arteries/metabolism , Gene Transfer Techniques , Recombinant Proteins/metabolism , Animals , Aorta, Thoracic , Drug Carriers , Growth Hormone/genetics , Growth Hormone/metabolism , Humans , Immunologic Techniques , Liposomes , Male , Organ Culture Techniques , Phosphatidylethanolamines/pharmacology , Plasmids , Rabbits , Staining and Labeling , Time Factors , Transfection
7.
Antiviral Res ; 23(1): 33-44, 1994 Jan.
Article in English | MEDLINE | ID: mdl-8141591

ABSTRACT

Recombinant bovine interferon-alpha I1 (rBoIFN-alpha) has known antiviral and immunomodulatory effects which have been exploited to reduce clinical disease in a number of clinical situations including bovine respiratory diseases. A slow release rBoIFN-alpha formulation may be of value to reduce bovine respiratory disease under field conditions by extending the period of protection, and hence improving the prophylactic benefits of rBoIFN-alpha. In this report, we describe a formulation of rBoIFN-alpha in sesame oil containing calcium stearate which can successfully sustain the release of rBoIFN-alpha over an 8-day period. Recombinant bovine IFN-alpha could be measured in serum for 8 days following treatment with an initial burst of release 6 h after injection. After a single subcutaneous depot injection of 50 mg and 100 mg of rBoIFN-alpha, initial serum levels reached 12-15 ng/ml and 25 ng/ml respectively. Correlating with this burst of release, there was a decrease in the number of circulating CD4-CD8- gamma delta+ T lymphocytes, and a slight neutropenia. No alterations in other cell phenotypes tested (CD4, CD8, CD2, CD6, B cells, monocytes or MHC class II) were observed, nor were there changes in lymphokine activated killer (LAK), natural killer (NK) cell activity, or oxygen radical formation (assessed by reduction of nitroblue tetrazolium). However, despite the rapid and short-lived burst of rBoIFN-alpha, levels of 2-5 oligoadenylate (2-5 A) synthetase remained elevated for 8 days. The sustained increase of 2-5 A synthetase was not due to the high initial dose released during the burst 6-12 h after injection, since injection of a bioavailable equivalent dose of interferon induced a significant rise in 2-5 A synthetase activity for 4 days only. As 2-5 A synthetase is known to be a correlate of antiviral activity, we propose that this formulation of rBoIFN-alpha may be one approach to increase the window of protection, leading to more effective prevention of bovine respiratory disease.


Subject(s)
2',5'-Oligoadenylate Synthetase/blood , Interferon Type I/administration & dosage , Animals , Cattle , Delayed-Action Preparations , Interferon Type I/adverse effects , Interferon Type I/blood , Leukocyte Count , Neutropenia/chemically induced , Recombinant Proteins , T-Lymphocytes/cytology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...