Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Gene Ther ; 11 Suppl 1: S109-21, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15454965

ABSTRACT

Duchenne muscular dystrophy (DMD) is a lethal genetic muscle disorder caused by recessive mutations in the dystrophin gene. The size of the gene (2.4 Mb) and mRNA (14 kb) in addition to immunogenicity problems and inefficient transduction of mature myofibres by currently available vector systems are formidable obstacles to the development of efficient gene therapy approaches. Adeno-associated viral (AAV) vectors overcome many of the problems associated with other vector systems (nonpathogenicity and minimal immunogenicity, extensive cell and tissue tropism) but accommodate limited transgene capacity (<5 kb). As a result of these observations, a number of laboratories worldwide have engineered a series of microdystrophin cDNAs based on genotype-phenotype relationship in Duchenne (DMD) and Becker (BMD) dystrophic patients, and transgenic studies in mdx mice. Recent progress in characterization of AAV serotypes from various species has demonstrated that alternative AAV serotypes are far more efficient in transducing muscle than the traditionally used AAV2. This article summarizes the current progress in the field of recombinant adeno-associated viral (rAAV) delivery for DMD, including optimization of recombinant AAV-microdystrophin vector systems/cassettes targeting the skeletal and cardiac musculature.


Subject(s)
Dependovirus/genetics , Dystrophin/genetics , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Muscular Dystrophy, Duchenne/therapy , Animals , Dystrophin/metabolism , Genetic Engineering , Genetic Vectors/genetics , Humans , Mice , Mice, Inbred mdx , Models, Animal , Muscle, Skeletal/metabolism , Myocardium/metabolism
2.
Gene Ther ; 9(1): 21-9, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11850719

ABSTRACT

Apolipoprotein E (apoE) is a multifunctional plasma glycoprotein involved in lipoprotein metabolism and a range of cell signalling phenomena. ApoE-deficient (apoE(-/-)) mice exhibit severe hypercholesterolaemia and are an excellent model of human atherosclerosis. ApoE somatic gene transfer and bone marrow transplantation in apoE(-/-) mice results in reversal of hypercholesterolaemia, inhibition of atherogenesis and regression of atherosclerotic plaque density. Replication defective adeno-associated virus vectors (rAAVs) are an attractive system currently in clinical trial for muscle-based heterologous gene therapy to express secreted recombinant plasma proteins. Here we have applied rAAV transduction of skeletal muscle to express wild-type (epsilon3) and a defective receptor-binding mutant (epsilon2) human apoE transgene in apoE(-/-) mice. In treated animals, apoE mRNA was present in transduced muscles and, although plasma levels of recombinant apoE fell below the detection levels of our ELISA (ie <10 ng/ml), circulating antibodies to human apoE and rAAV were induced. Up to 3 months after a single administration of rAAV/apoE3, a significant reduction in atherosclerotic plaque density in aortas of treated animals was observed (approximately 30%), indicating that low-level rAAV-mediated apoE3 expression from skeletal muscle can retard atherosclerotic progression in this well-defined genetic model.


Subject(s)
Apolipoproteins E/genetics , Arteriosclerosis/therapy , Dependovirus/genetics , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Animals , Antibodies/blood , Aorta/pathology , Apolipoproteins E/analysis , Apolipoproteins E/immunology , Arteriosclerosis/pathology , Blotting, Western/methods , Enzyme-Linked Immunosorbent Assay/methods , Humans , Mice , Mice, Knockout , Transduction, Genetic/methods , Transgenes
3.
Gene Ther ; 8(20): 1580-6, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11704819

ABSTRACT

We have examined the ability of proliferating myoblasts and post-mitotic, differentiated myotubes to produce retroviral vector using hybrid adeno-retroviral vectors as templates. We show that production of retroviral vector from myoblasts peaks 48 h after adenoviral infection at 4.8 x 10(4) cfu/ml and is scarcely detectable by 96 h. Both fully and partially differentiated myotubes were able to generate a sustained increase in the levels of retroviral vector compared with myoblasts peaking 48 h at 1.4 x 10(5) cfu/ml and 1.8 x 10(5) cfu/ml, respectively. Addition of the cell cycle inhibitor aphidicolin (5 microg/ml) had no effect on the production of retroviral vector from fully differentiated myotubes, but resulted in an 80% increase in vector production from partially differentiated myotubes. Thus indicating that retroviral vector production is more efficient in post-mitotic myotubes and is independent of muscle cell cycle progression.


Subject(s)
Adenoviridae/genetics , Genetic Therapy/methods , Genetic Vectors/genetics , Muscle Fibers, Skeletal/virology , Muscular Dystrophy, Duchenne/therapy , Retroviridae/genetics , 3T3 Cells , Animals , Antiviral Agents/pharmacology , Aphidicolin/pharmacology , Cell Differentiation , Cell Line , Gene Expression , Genetic Engineering/methods , Genetic Vectors/pharmacology , Green Fluorescent Proteins , Humans , Luminescent Proteins/genetics , Mice , Microscopy, Fluorescence , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/ultrastructure
4.
J Biol Chem ; 276(49): 46011-6, 2001 Dec 07.
Article in English | MEDLINE | ID: mdl-11590165

ABSTRACT

Sub-endothelial infiltration of monocytes occurs early in atherogenesis and is facilitated by cell adhesion molecules that are up-regulated on activated endothelium. Apolipoprotein E (apoE) helps protect against atherosclerosis, in part, because apoE particles secreted by macrophages have local beneficial effects at lesion sites. Here, we hypothesize that such protection includes anti-inflammatory actions and investigate whether cell-derived apoE can inhibit tumor necrosis factor-alpha-mediated up-regulation of vascular cell adhesion molecule-1 (VCAM-1) in human umbilical vein endothelial cells (HUVECs). Two models were used to mimic endothelial exposure to macrophage-derived apoE. In the first, HUVECs were transiently transfected to secrete apoE; VCAM-1 induction inversely correlated with secretion of apoE into the media (r = -0.76, p < 0.001). In the second, incubation of HUVECs with media from recombinant Chinese hamster ovary (CHO) cells expressing apoE (CHO(apoE)) also reduced VCAM-1 in a dose-dependent manner (r = -0.70, p < 0.001). Characterization of CHO(apoE) cell-derived apoE revealed several similarities to apoE particles secreted by human blood monocyte-derived macrophages. The suppression of endothelial activation by apoE most likely occurs via stimulation of endothelial nitric oxide synthase; apoE increased levels of intracellular nitric oxide and its surrogate marker, cyclic guanosine monophosphate, while the nitric oxide synthase inhibitor, ethyl-isothiourea, blocked its effect. We propose that apoE secreted locally at lesion sites by macrophages may be anti-inflammatory by stimulating endothelium to release NO and suppress VCAM-1 expression.


Subject(s)
Apolipoproteins E/physiology , Down-Regulation/physiology , Endothelium, Vascular/metabolism , Vascular Cell Adhesion Molecule-1/genetics , Animals , CHO Cells , Cricetinae , Endothelium, Vascular/cytology , Humans , Nitric Oxide/metabolism , Transfection
5.
Hum Mol Genet ; 9(17): 2545-51, 2000 Oct 12.
Article in English | MEDLINE | ID: mdl-11030760

ABSTRACT

Apolipoprotein-E (apoE) protects against coronary artery disease via hepatic removal of atherogenic remnant lipoproteins, sequestration of cholesterol from vessel walls and local anti-oxidant, anti-platelet and anti-inflammatory actions. ApoE gene transfer may thus ameliorate a hyperlipidaemic profile and have beneficial effects at lesion sites to prevent or regress atherosclerosis, a concept endorsed by adenoviral-mediated hepatic expression studies. Here, using plasmid vectors expressing allelic human apoE2 or apoE3 isoforms, skeletal muscle was evaluated as an effective secretory platform for apoE gene augmentation. Transfected myoblasts and myotubes were found to efficiently secrete recombinant apoE in vitro as spherical 10-16 nm lipoprotein particles with pre-beta mobility. Intramuscular plasmid injection in apoE(-/-) mice, which develop spontaneous atherosclerotic plaque and xanthoma resulted in expression and secretion of apoE. Human apoE mRNA was detected by RT-PCR in injected muscles and, although concentrations of apoE3, which is rapidly cleared from plasma, were near ELISA detection limits, levels of plasma apoE2 were measurable (17.5 +/- 4.3 ng/ml). To assess whether muscle-based expression of apoE2 could inhibit atherogenesis, long-term follow-up studies were conducted. Although hyperlipidaemia was not reduced in treated animals, end-point pathology showed clear retardation of atherosclerotic and xanthomatous lesions. Up to 9 months following a single apoE2 plasmid administration, atherosclerotic lesion coverage in proximal aorta was significantly reduced by 20-30% (P < 0.01), whereas development of gross dorsal xanthoma (>5 mm diameter) was effectively reduced to zero. We conclude that expression of apoE from ectopic muscle sites has therapeutic potential to limit progression of atherosclerosis.


Subject(s)
Apolipoproteins E/genetics , Arteriosclerosis/therapy , Genetic Therapy , Muscle, Skeletal/metabolism , Plasmids , Xanthomatosis/therapy , Animals , Apolipoprotein E2 , Apolipoprotein E3 , Apolipoproteins E/blood , Apolipoproteins E/deficiency , Apolipoproteins E/metabolism , Cell Line , Disease Progression , Gene Expression , Gene Transfer Techniques , Genetic Vectors , Humans , Hyperlipidemias/metabolism , Hyperlipidemias/therapy , Mice , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transfection
6.
Int J Mol Med ; 6(4): 363-75, 2000 Oct.
Article in English | MEDLINE | ID: mdl-10998427

ABSTRACT

Adeno-associated virus (AAV), a defective parvovirus, was discovered more than 30 years ago. Interest in this virus for human gene therapy applications focuses on its non-pathogenicity, broad tropism and infectivity, site-specific integration and long-term persistence. The field of rAAV research has considerably advanced: titers of 1014 p/ml have been achieved, plasmid systems devised to produce helper-free viruses, chimaeric vectors combining properties of rAAV ITRs and large sequence capacity from Ad/HS vectors in parallel with the revolutionary intron strategy based on heterodimerisation of the forming concatamers have expanded the vector capacity. Muscle cells and neurons (post-mitotic cells) are amongst the most efficient targets of rAAV delivery and AAV receptors and co-receptors have been identified. This review will describe advances in the field of rAAV technology that overcome certain limitations of the vector as a gene delivery system and overview applications involving these recombinant vectors for the treatment of acquired and inherited diseases.


Subject(s)
Adenoviridae/genetics , Genetic Therapy , Genetic Vectors/genetics , Animals , Disease , Gene Transfer Techniques , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/therapy , Humans , Therapeutics
SELECTION OF CITATIONS
SEARCH DETAIL
...