Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters











Database
Publication year range
1.
Gene Ther ; 15(1): 1-11, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17637794

ABSTRACT

A single plasmid regulated expression vector based upon a mifepristone-inducible two plasmid system, termed pBRES, has been constructed and tested in mice using murine interferon-b (mIFNb) as the transgene. The expression of mIFNb in the circulation was followed by measuring the systemic induction of IP-10, a validated biomarker for mIFNb in mice. Long-term, inducible expression of mIFNb was demonstrated following a single intramuscular (i.m.) injection of the pBRES mIFNb plasmid vector into the hind limb of mice. Induction of mIFNb expression was achieved by administration of the small molecule inducer, mifepristone (MFP). Plasmid DNA and mIFNb mRNA levels in the injected muscles correlated with mIFNb expression as monitored by IP-10 over a 3-month time period. Renewable transgene expression was achieved following repeat administration of the plasmid at 3 months following the first plasmid injection. A dose-dependent increase in expression was demonstrated by varying the amount of injected plasmid or the amount of the inducer administered to the mice. Finally, the pBRES plasmid expressing mIFNb under control of the inducer, MFP, was shown to be efficacious in a murine model of experimental allergic encephalomyelitis, supporting the feasibility of gene-based therapeutic approaches for treating diseases such as multiple sclerosis.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/therapy , Gene Expression Regulation , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Interferon-beta/genetics , Plasmids/administration & dosage , Animals , Biomarkers/blood , Chemokine CXCL10/analysis , Disease Progression , Female , Injections, Intramuscular , Interferon-beta/blood , Mice , Mice, Inbred Strains , Mifepristone/administration & dosage , Multiple Sclerosis/therapy , Plasmids/analysis , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Transgenes
2.
Gene Ther ; 15(3): 224-32, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18033310

ABSTRACT

Vessel wall inflammation and matrix destruction are critical to abdominal aortic aneurysm (AAA) formation and rupture. We have previously shown that urokinase plasminogen activator (uPA) is highly expressed in experimental AAA and is essential for AAA formation and expansion. In this study, we examined the effects of overexpression of a natural inhibitor of uPA, plasminogen activator inhibitor-1 (PAI-1), on the development of angiotensin (Ang) II-induced AAA in ApoE-deficient (ApoE(-/-)) mice. Mice were treated with recombinant adenovirus containing either the human PAI-1 gene (Ad5.CMV.PAI-1) or the luciferase gene (Ad5.CMV.Luc) delivered either locally by intra-adventitial injection or systemically by tail vein injection. Our results show that local delivery of the PAI-1 gene completely prevented AAA formation (0 vs 55.6% in Ad5.CMV.Luc controls, P<0.05). In contrast, systemic delivery of the PAI-1 gene did not affect AAA incidence (78 vs 90% in Ad5.CMV.Luc controls, P=0.125). Local delivery of the PAI-1 gene 2 weeks after Ang II infusion prevented further expansion of small aneurysms, but had no significant effect on the progression of larger aneurysms. These data suggest that local PAI-1 gene transfer could be used to stabilize small AAA and reduce the rate of expansion and risk of rupture.


Subject(s)
Aortic Aneurysm, Abdominal/prevention & control , Genetic Therapy/methods , Plasminogen Activator Inhibitor 1/genetics , Transduction, Genetic/methods , Angiotensin II , Animals , Aorta, Abdominal/enzymology , Aorta, Abdominal/pathology , Aortic Aneurysm, Abdominal/metabolism , Aortic Aneurysm, Abdominal/pathology , Apolipoproteins E/genetics , Atherosclerosis/enzymology , Atherosclerosis/pathology , Cytomegalovirus/genetics , Fibrosis , Gene Expression , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Luciferases/genetics , Male , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Knockout , Plasminogen Activator Inhibitor 1/metabolism , Urokinase-Type Plasminogen Activator/metabolism
3.
Gene Ther ; 15(1): 54-60, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17960164

ABSTRACT

Neutralizing antibodies (nAB) at the time of administration hamper the effectiveness of adeno-associated virus (AAV) as a clinical DNA delivery system. The present study was designed to investigate if AAV re-administration in muscle tissue is dependent on the nAB titer. Recombinant (r)AAV serotype 1, as a promising candidate for targeting skeletal muscle, was used for gene delivery. C57Bl/6 mice were infected intramuscularly with doses between 1 x 10(9) and 5 x 10(10) virus particles (vp) of AAV1-expressing luciferase (AAV1-luc) or human interferon-beta (AAV1-hIFNbeta). Increasing transgene expression was observed over the first 2 months and anti-AAV1 nAB titers peaked between weeks 4 and 8. Six months after the first administration, 5 x 10(10) vp of AAV1-IFNbeta were re-administered. Following re-administration, nAB titers increased but did not significantly affect transgene expression from the AAV vector that had been administered first. In contrast, hIFNbeta expression originating from the second vector administration was significantly diminished and reflected the nAB titer present at the day of re-administration. The present study extends earlier observations that preexisting nAB affects AAV1 re-administration. The level of nAB is proportional to the virus dose used for the first injection and transgene expression following re-administration is dependent on preexisting nAB titer.


Subject(s)
Dependovirus/genetics , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Transduction, Genetic/methods , Animals , Antibodies/analysis , Antibody Formation , Dependovirus/immunology , Gene Expression , Genetic Engineering , Genetic Vectors/immunology , Humans , Injections, Intramuscular , Interferon-beta/genetics , Interferon-beta/immunology , Luciferases/genetics , Male , Mice , Mice, Inbred C57BL , Time Factors , Transgenes , Viral Load
4.
Handb Exp Pharmacol ; (180): 37-44, 2007.
Article in English | MEDLINE | ID: mdl-17554503

ABSTRACT

Mobilization and recruitment of bone marrow-derived progenitor cells (BMDPCs) play an important role in postischemic tissue repair. Patients with coronary artery disease (CAD) or peripheral vascular disease (PVD) exhibit endothelial dysfunction, and as a result are likely to have a reduced number of progenitor cells mobilized in their peripheral circulation following ischemic injury. Identification of eNOS independent pathways for BMDPC mobilization may have important therapeutic value in this patient population. To identify such mechanisms we investigated the effect of granulocyte-colony stimulating factor (GCSF) and stem cell factor (SCF) in eNOS-KO mice with and without surgical hind-limb ischemia. Our results suggest that BMDPC mobilization can be achieved via activation of NO-independent pathways.


Subject(s)
Bone Marrow Cells/cytology , Endothelial Cells/physiology , Hematopoietic Stem Cell Mobilization/methods , Nitric Oxide Synthase Type III/physiology , Nitric Oxide/physiology , Animals , Humans
5.
Gene Ther ; 13(18): 1342-50, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16642030

ABSTRACT

Gene delivery of angiogenic growth factors is a promising approach for the treatment of ischemic cardiovascular diseases. However, success of this new therapeutic principle is hindered by the lack of critical understanding as to how disease pathology affects the efficiency of gene delivery and/or the downstream signaling pathways of angiogenesis. Critical limb ischemia occurs in patients with advanced atherosclerosis often exhibiting deficiency in endothelial nitric oxide production. Similar to these patients, segmental femoral artery resection progresses into severe ischemic necrosis in mice deficient in endothelial nitric oxide synthase (ecNOS-KO) as well as in balb/c mice. We used these models to evaluate the influence of severe ischemia on transfection efficiency and duration of transgene expression in the skeletal muscle following plasmid injection in combination with electroporation. Subsequently, we also explored the potential therapeutic effect of the phosphomimetic mutant of ecNOS gene (NOS1177D) using optimized delivery parameters, and found significant benefit both in ecNOS-KO and balb/c mice. Our results indicate that NOS1177D gene delivery to the ischemic skeletal muscle can be efficient to reverse critical limb ischemia in pathological settings, which are refractory to treatments with a single growth factor, such as vascular endothelial growth factor.


Subject(s)
Genetic Therapy/methods , Ischemia/therapy , Muscle, Skeletal/metabolism , Nitric Oxide Synthase Type III/genetics , Transfection/methods , Vascular Endothelial Growth Factor A/metabolism , Animals , Electroporation , Endothelium, Vascular/metabolism , Gene Expression , Genetic Vectors , Hindlimb , Humans , Ischemia/metabolism , Ischemia/pathology , Laser-Doppler Flowmetry , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/pathology , Neovascularization, Physiologic , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/analysis , Nitric Oxide Synthase Type III/metabolism , Regional Blood Flow , Transgenes , Vasodilation
6.
Circ Res ; 88(9): 911-7, 2001 May 11.
Article in English | MEDLINE | ID: mdl-11349000

ABSTRACT

First-generation, E1-deleted adenoviral vectors (E1-AV) can transduce the vascular endothelium with high efficiency, but their use is limited by the resulting acute endothelial injury and the long-term development of intimal hyperplasia. To reduce the impact of viral proteins on the gene-modified cells, a second-generation adenoviral vector with an additional pair of deletions in the E4 region was developed. To determine whether this E1/E4-AV vector would be useful for vascular gene transfer, we directly compared the efficiency of gene transfer to uninjured rabbit carotid arteries using either an E1/E4-AV or an E1-AV vector encoding beta-galactosidase. Both vectors efficiently transduced vascular endothelium; however, the E1/E4-AV vector gene-modified vessels showed higher beta-galactosidase expression 10 days after gene transfer. Importantly, the E1/E4-AV vector produced substantially less endothelial cell activation, less inflammation, and reduced neointimal hyperplasia compared with the E1-AV vector-treated vessels. The E1-AV vector-transduced vessels also demonstrated significantly impaired endothelium-dependent relaxation whereas the E1/E4-AV vector did not impact vasomotor function, even at doses of virus in 5-fold excess of the amount required for >90% transduction of the endothelium. We conclude that the E1/E4-AV vector is superior to the E1-AV vector for vascular gene therapy because of the prolonged transgene expression, reduced vascular inflammation, reduced intimal hyperplasia, and maintenance of normal vasomotor function.


Subject(s)
Adenoviridae/genetics , Genetic Therapy , Vascular Diseases/therapy , Adenovirus E1 Proteins/genetics , Adenovirus E1A Proteins/genetics , Adenovirus E4 Proteins/genetics , Animals , Carotid Arteries/metabolism , Carotid Arteries/pathology , Carotid Arteries/physiopathology , Endothelium, Vascular/metabolism , Endothelium, Vascular/physiopathology , Gene Expression Regulation , Genetic Vectors/genetics , Immunohistochemistry , Inflammation/genetics , Inflammation/therapy , Intercellular Adhesion Molecule-1/metabolism , Male , Rabbits , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Transgenes/genetics , Tunica Intima/metabolism , Tunica Intima/pathology , Vascular Cell Adhesion Molecule-1/metabolism , Vascular Diseases/genetics , Vasodilation/genetics , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
7.
J Clin Invest ; 105(6): 741-8, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10727442

ABSTRACT

Reduced production of nitric oxide (NO) in the cirrhotic liver results from a defect in hepatic endothelial cell nitric oxide synthase (ecNOS) and appears to contribute to the high intrahepatic resistance and portal hypertension typical of cirrhosis. Therefore, we postulated that targeting a heterologous NOS isoform to sinusoidal endothelial cells or other perisinusoidal cells, such as hepatic stellate cells, would counter the defect in NO production and reduce resistance to blood flow. Recombinant adenovirus (Ad) carrying the neuronal NOS gene (nNOS) targeted liver sinusoidal endothelial cells, stellate cells, and hepatocytes more efficiently than the corresponding cells in cirrhotic livers, but transduction rates were substantial even in cirrhotic animals. Expression of nNOS in each liver cell type, whether from normal or injured liver, caused increased NO production and inhibited endothelin-1-induced contractility of perisinusoidal stellate cells. Finally, in 2 different in vivo models of cirrhosis and portal hypertension, transduction of livers with recombinant Ad.nNOS significantly reduced intrahepatic resistance and portal pressure. The data highlight the feasibility of gene transfer to diseased liver and hepatic cells and demonstrate the potential of a novel therapy for portal hypertension caused by cirrhosis.


Subject(s)
Genetic Therapy , Hypertension, Portal/therapy , Isoenzymes/genetics , Liver Cirrhosis, Experimental/complications , Nerve Tissue Proteins/genetics , Nitric Oxide Synthase/genetics , Adenoviridae/genetics , Animals , Cell Size , Cells, Cultured , DNA, Complementary/genetics , Endothelin-1/antagonists & inhibitors , Feasibility Studies , Genetic Vectors/genetics , Hypertension, Portal/etiology , Isoenzymes/physiology , Male , Nitric Oxide/physiology , Nitric Oxide Synthase/physiology , Nitric Oxide Synthase Type I , Nitrites/metabolism , Rats , Rats, Sprague-Dawley , Recombinant Fusion Proteins/physiology
8.
Circ Res ; 82(12): 1253-62, 1998 Jun 29.
Article in English | MEDLINE | ID: mdl-9648721

ABSTRACT

Acute injury after adenoviral vascular gene transfer remains incompletely characterized. Here, we describe the early response (< or =days) in 52 New Zealand White rabbits undergoing gene transfer (beta-galactosidase or empty vector) or sham procedures to both carotid arteries. After gene transfer, arteries were either left in vivo for 1 hour to 3 days (in vivo arteries) or were excised immediately after gene transfer and cultured (ex vivo arteries). Within 1 hour, in vivo arteries receiving infectious titers of > or = 4X10(9) plaque-forming units (pfu)/mL showed endothelial activation, with an acute inflammatory infiltrate developing by 6 hours. Ex vivo arteries showed endothelial activation but no inflammatory infiltrate. There were also significant differences in transgene expression between in vivo and ex vivo arteries. Ex vivo arteries showed titer-dependent increases in beta-galactosidase expression through 2X10(10) pfu/mL, whereas in in vivo arteries, titers above 4X10(9) pfu/mL merely increased acute inflammatory response, without increasing transgene expression. In vivo arteries showed significant time- and titer-dependent impairment in endothelium-dependent relaxation, with no effect on contraction or nitroprusside-induced relaxation. Interestingly, however, if rabbits were made neutropenic with vinblastine, their arteries maintained full endothelium-dependent relaxation, even after very high titer vascular infection (up to 1X10(11) pfu/mL). These findings show that recombinant adenovirus triggers an early inflammatory response, and it is the inflammatory response that in turn causes functional endothelial injury. This occurs at much lower titers than previously appreciated (though the precise threshold will undoubtedly vary between laboratories). However, titers below the inflammatory threshold produce excellent transgene expression without inflammation or vascular injury.


Subject(s)
Endothelium, Vascular/immunology , Gene Expression/immunology , Gene Transfer Techniques/adverse effects , Transgenes , Adenoviridae , Animals , Arteries/immunology , Arteries/pathology , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Genetic Vectors , Intercellular Adhesion Molecule-1/metabolism , Male , Rabbits , Transgenes/immunology , Vascular Cell Adhesion Molecule-1/metabolism , Vasculitis/etiology , Vasculitis/immunology
9.
Zhonghua Yi Xue Za Zhi ; 73(6): 352-4, 381-2, 1993 Jun.
Article in Chinese | MEDLINE | ID: mdl-8258106

ABSTRACT

Twenty-six patients with venous insufficiency and 15 normal controls were studied by local skin pathological and ultrastructure changes. Transcutaneous oxygen tension (Tcpo2) and tPA, PAI of venous blood were measured. Biopsies from the ulcerating and liposclerosis area showed pericapillary fibrins. The patients with severe venous disease resulting in liposclerosis or ulceration of the goiter area had Tcpo2 levels lower than the controls and patients with venous insufficiency but no skin changes. The more serious the skin changes in patients with venous insufficiency, the lower the fibrinolysis activity. We conclude that extravascular deposition of fibrins in patients with venous disease which block the diffusion and exchanges of oxygen between the blood and tissue is an important factor in the development of venous ulcerations. The decrease of fibrinolysis activity led to the decrease of removing pericapillary fibrins deposition, combined with extravascular fibrin caused venous ulceration.


Subject(s)
Skin/pathology , Varicose Ulcer/pathology , Venous Insufficiency/pathology , Aged , Blood Gas Monitoring, Transcutaneous , Fibrin/metabolism , Humans , Middle Aged , Plasminogen Inactivators/blood , Skin/ultrastructure , Tissue Plasminogen Activator/blood , Varicose Ulcer/blood , Varicose Ulcer/etiology
SELECTION OF CITATIONS
SEARCH DETAIL