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.
Arterioscler Thromb Vasc Biol ; 21(11): 1777-82, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11701465

ABSTRACT

Adenoviral vectors are promising agents for vascular gene transfer. Their use, however, is limited by inflammatory host responses, neointima formation, and brevity of transgene expression. Inclusion of the immunomodulatory adenoviral E3 genes in a vector might prevent inflammation and neointima formation and prolong transgene expression. We compared 2 adenoviral vectors in a model of in vivo gene transfer to rabbit arteries. Both vectors expressed a luciferase reporter gene. One vector (AdE3Luc) contained the adenovirus early 3 (E3) region and the other (AdRSVLuc) lacked E3. Expression of E3 genes by AdE3Luc was confirmed in vitro and in vivo. Arteries transduced with AdE3Luc had substantially and significantly less inflammation (fewer T cells and lower levels of vascular cell adhesion molecule-1 and intercellular adhesion molecule 1 expression) and decreased neointima formation 14 days after gene transfer. Luciferase expression from the 2 vectors was equivalent, however, at both 3 and 14 days after gene transfer. Expression of E3 had no systemic immunosuppressive effects, as measured by peripheral blood counts and by assays for serum antibodies to adenovirus. We conclude that expression of E3 significantly decreases adenovirus-induced arterial wall inflammation and neointima formation. Because inflammation and neointima formation are major barriers to the clinical application of adenoviral vectors, use of E3-containing vectors improves the promise of adenovirus-mediated arterial gene transfer.


Subject(s)
Adenoviridae/genetics , Adenovirus E3 Proteins/genetics , Arterial Occlusive Diseases/therapy , Arteritis/therapy , Genetic Therapy/methods , Adenoviridae/immunology , Adenoviridae/metabolism , Adenovirus E3 Proteins/biosynthesis , Animals , Antibodies, Viral/biosynthesis , Arterial Occlusive Diseases/metabolism , Arterial Occlusive Diseases/pathology , Arteries/metabolism , Arteries/pathology , Arteritis/immunology , Arteritis/pathology , CHO Cells , Cell Adhesion Molecules/metabolism , Cell Line , Cricetinae , DNA, Viral/genetics , Genetic Vectors , Luciferases/biosynthesis , Luciferases/genetics , RNA, Viral/biosynthesis , Rabbits , Transduction, Genetic
2.
Arterioscler Thromb Vasc Biol ; 20(6): 1452-8, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10845857

ABSTRACT

The utility of adenoviral vectors for arterial gene transfer is limited by the brevity of their expression and by inflammatory host responses. As a step toward circumventing these difficulties, we used a rabbit model of in vivo arterial gene transfer to test 3 second-generation vectors: a vector containing a temperature-sensitive mutation in the E2A region, a vector deleted of E2A, and a vector that expresses the immunomodulatory 19-kDa glycoprotein (gp19k) from adenovirus 2. Compared with similar first-generation vectors, the second-generation vectors did not significantly prolong beta-galactosidase transgene expression or decrease inflammation in the artery wall. Although cyclophosphamide ablated the immune and inflammatory responses to adenovirus infusion, it only marginally prolonged transgene expression (94% drop in expression between 3 and 14 days). In experiments performed with "null" adenoviral vectors (no transgene), loss of vector DNA from the arterial wall was also rapid (>99% decrease between 1 hour and 14 days), unrelated to dose, and only marginally blunted by cyclophosphamide. Thus, the early loss of transgene expression after adenoviral arterial gene transfer is due primarily to loss of vector DNA, is not correlated with the presence of local vascular inflammation, and cannot be prevented by use of E2A-defective viruses, expression of gp19k, or cyclophosphamide-mediated immunosuppression. Adenovirus-induced vascular inflammation can be prevented by cyclophosphamide treatment or by lowering the dose of infused virus. However, stabilization of adenovirus-mediated transgene expression in the arterial wall is a more elusive goal and will require novel approaches that prevent the early loss of vector DNA.


Subject(s)
Adenoviridae/genetics , Arteries/metabolism , DNA-Binding Proteins , DNA/metabolism , Gene Expression , Gene Transfer Techniques , Genetic Vectors , Animals , Basic Helix-Loop-Helix Transcription Factors , Cyclophosphamide/pharmacology , Genetic Vectors/adverse effects , Immunosuppressive Agents/pharmacology , Male , Rabbits , Transcription Factors/genetics , Vasculitis/etiology , beta-Galactosidase/genetics
3.
Arterioscler Thromb Vasc Biol ; 20(2): 298-308, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10669624

ABSTRACT

Fas ligand (FasL) is expressed by cells of the arterial wall and is present in human atherosclerotic lesions. However, the role of FasL in modifying the initiation and progression of atherosclerosis is unclear. To investigate the role of arterial FasL expression in the development of atherosclerosis, we first established a model of primary lesion formation in rabbit carotid arteries. In this model, infusion of adenoviral vectors into surgically isolated, nondenuded arteries of hypercholesterolemic rabbits leads to the formation of human-like early atherosclerotic lesions. Expression of FasL in arterial endothelium in this model decreased T-cell infiltration and expression of vascular cell adhesion molecule-1 but did not affect expression of intercellular adhesion molecule-1. Intimal lesions grew more rapidly in FasL-transduced arteries than in arteries transduced with a control adenovirus that did not express a transgene. Total intimal macrophage accumulation was increased in FasL-transduced arteries; however, the proportion of lesion area occupied by macrophages was not elevated. The accelerated lesion growth was primarily due to the accumulation of intimal smooth muscle cells with a synthetic proliferative phenotype. There was no significant apoptosis in FasL-transduced or control arteries and no granulocytic infiltrates. Thus, the net result of elevated FasL expression is to accelerate atherosclerotic lesion growth by increasing lesion cellularity. Vascular expression of FasL may contribute to the progression of atherosclerosis.


Subject(s)
Arteries/metabolism , Arteriosclerosis/etiology , Hypercholesterolemia/complications , Hypercholesterolemia/metabolism , Membrane Glycoproteins/metabolism , Adenoviridae/physiology , Animals , Apoptosis/physiology , Arteriosclerosis/metabolism , Arteriosclerosis/pathology , Cell Division/drug effects , Endothelium, Vascular/metabolism , Fas Ligand Protein , Gene Expression , Macrophages/pathology , Membrane Glycoproteins/genetics , Membrane Glycoproteins/pharmacology , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/physiology , Rabbits , T-Lymphocytes/physiology , Transgenes/genetics , Vascular Cell Adhesion Molecule-1/metabolism
4.
Hum Gene Ther ; 10(9): 1469-78, 1999 Jun 10.
Article in English | MEDLINE | ID: mdl-10395372

ABSTRACT

The vessel wall fibrinolytic system plays an important role in maintaining the arterial phenotype and in regulating the arterial response to injury. Plasminogen activator inhibitor type 1 (PAI-1) regulates tissue fibrinolysis and is expressed in arterial tissue; however, its biological role remains uncertain. To help elucidate the role of PAI-1 in the artery wall, and to begin to clarify whether manipulation of vascular PAI-1 expression might be a target for gene therapy, we used adenoviral vectors to increase expression of rat PAI-1 in rat carotid arteries. Infusion of an adenoviral vector in which PAI-1 expression was driven by a promoter derived from the Rous sarcoma virus (RSV) did not increase PAI-1 expression above endogenous levels. To improve PAI-1 expression, we modified the vector by (1) truncating the 3' untranslated region of PAI-1 to increase the mRNA half-life, (2) substituting the SRalpha or the cytomegalovirus (CMV) promoter for the RSV promoter, (3) including an intron in the expression cassette, and (4) altering the direction of transcription of the transgene cassette. The optimal expression vector, revealed by in vitro studies, contained the CMV promoter, an intron, and a truncated PAI-1 mRNA. This vector increased PAI-1 expression by 30-fold over control levels in vitro and by 1.6 to 2-fold over endogenous levels in vivo. This vector will be useful for elucidating the role of PAI-1 in arterial pathobiology. Because genes that are important in maintaining the vascular phenotype are likely to be expressed in the vasculature, the technical issues of how to increase in vivo expression of endogenous genes are highly relevant to the development of genetic therapies for vascular disease.


Subject(s)
Adenoviruses, Human , Carotid Arteries , Gene Transfer Techniques , Genetic Vectors , Plasminogen Activator Inhibitor 1/genetics , Animals , Avian Sarcoma Viruses/genetics , Cells, Cultured , Culture Techniques , Cytomegalovirus/genetics , Gene Transfer Techniques/standards , Humans , Male , Muscle, Smooth, Vascular/cytology , Plasminogen Activator Inhibitor 1/metabolism , Promoter Regions, Genetic , Rats , Rats, Sprague-Dawley
5.
J Vasc Surg ; 29(3): 543-50, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10069919

ABSTRACT

PURPOSE: Adenovirus-mediated arterial gene transfer is a promising tool in the study of vascular biology and the development of vascular gene therapy. However, intraluminal delivery of adenoviral vectors causes vascular inflammation and neointimal formation. Whether these complications could be avoided and gene transfer efficiency maintained by means of delivering adenoviral vectors via the adventitia was studied. METHODS: Replication-defective adenoviral vectors encoding a beta-galactosidase (beta-gal) gene (AdRSVnLacZ) or without a recombinant gene (AdNull) were infused into the lumen or the adventitia of rabbit carotid arteries. Two days after infusion of either AdRSVnLacZ (n = 8 adventitial, n = 8 luminal) or AdNull (n = 4 luminal), recombinant gene expression was quantitated by histochemistry (performed on tissue sections) and with a beta-gal activity assay (performed on vessel extracts). Inflammation caused by adenovirus infusion was assessed 14 days after infusion of either AdNull (n = 6) or vehicle (n = 6) into the carotid adventitia. Inflammation was assessed by means of examination of histologic sections for the presence of neointimal formation and infiltrating T cells and for the expression of markers of vascular cell activation (ICAM-1 and VCAM-1). To measure the systemic immune response to adventitial infusion of adenovirus, plasma samples (n = 3) were drawn 14 days after infusion of AdNull and assayed for neutralizing antibodies. RESULTS: Two days after luminal infusion of AdRSVnLacZ, approximately 30% of luminal endothelial cells expressed beta-gal. Similarly, 2 days after infusion of AdRSVnLacZ to the adventitia, approximately 30% of adventitial cells expressed beta-gal. beta-gal expression was present in the carotid adventitia, the internal jugular vein adventitia, and the vagus nerve perineurium. Elevated beta-gal activity (50- to 80-fold more than background; P <.05) was detected in extracts made from all AdRSVnLacZ-transduced arteries. The amount of recombinant protein expression per vessel did not differ significantly between vessels transduced via the adventitia (17.1 mU/mg total protein [range, 8.1 to 71.5]) and those transduced via a luminal approach (10.0 mU/mg total protein [range, 3.9 to 42.6]). Notably, adventitial delivery of AdNull did not cause neointimal formation. In addition, vascular inflammation in arteries transduced via the adventitia (ie, T-cell infiltrates and ICAM-1 expression) was confined to the adventitia, sparing both the intima and media. Antiadenoviral neutralizing antibodies were present in all rabbits after adventitial delivery of AdNull. CONCLUSION: Infusion of adenoviral vectors into the carotid artery adventitia achieves recombinant gene expression at a level equivalent to that achieved by means of intraluminal vector infusion. Because adventitial gene transfer can be performed by means of direct application during open surgical procedures, this technically simple procedure may be more clinically applicable than intraluminal delivery. Moreover, despite the generation of a systemic immune response, adventitial infusion had no detectable pathologic effects on the vascular intima or media. For these reasons, adventitial gene delivery may be a particularly useful experimental and clinical tool.


Subject(s)
Adenoviridae/genetics , Arteritis/prevention & control , Carotid Arteries , DNA, Viral/genetics , Elastic Tissue , Gene Transfer Techniques , Genetic Vectors/administration & dosage , Animals , Antibodies, Viral/blood , Arteritis/pathology , Carotid Arteries/enzymology , Carotid Arteries/pathology , DNA, Recombinant/genetics , Elastic Tissue/enzymology , Elastic Tissue/pathology , Endothelium, Vascular/enzymology , Endothelium, Vascular/pathology , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Viral , Genetic Vectors/adverse effects , Histocytochemistry , Infusions, Intra-Arterial/adverse effects , Intercellular Adhesion Molecule-1/genetics , Jugular Veins/enzymology , Male , Pharmaceutical Vehicles , Rabbits , T-Lymphocytes/pathology , Tunica Intima/pathology , Tunica Media/pathology , Vagus Nerve/enzymology , Vascular Cell Adhesion Molecule-1/genetics , beta-Galactosidase/analysis , beta-Galactosidase/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...