Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
Gene Ther ; 18(2): 182-8, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20962870

ABSTRACT

We have previously shown that recombinant Sendai virus (SeV) vector, derived from murine parainfluenza virus, is one of the most efficient vectors for airway gene transfer. We have also shown that SeV-mediated transfection on second administration, although reduced by 60% when compared with levels achieved after a single dose, is still high because of the efficient transfection achieved by SeV vector in murine airways. Here, we show that these levels further decrease on subsequent doses. In addition, we validated SeV vector repeat administration in a non-natural host model, the sheep. As part of these studies we first assessed viral stability in a Pari LC Plus nebuliser, a polyethylene catheter (PEC) and the Trudell AeroProbe. We also compared the distribution of gene expression after PEC and Trudell AeroProbe administration and quantified virus shedding after sheep transduction. In addition, we show that bronchial brushings and biopsies, collected in anaesthetized sheep, can be used to assess SeV-mediated gene expression over time. Similar to mice, gene expression in sheep was transient and had returned to baseline values by day 14. In conclusion, the SeV vector should be strongly considered for lung-related applications requiring a single administration of the vector even though it might not be suitable for diseases requiring repeat administration.


Subject(s)
Gene Expression , Genetic Vectors , Sendai virus/genetics , Sheep/genetics , Transduction, Genetic , Animals , Catheters , Female , Gene Transfer Techniques/instrumentation , Lung , Mice , Mice, Inbred BALB C , Models, Animal , Retreatment
2.
Gene Ther ; 14(9): 768-74, 2007 May.
Article in English | MEDLINE | ID: mdl-17301842

ABSTRACT

We have assessed if high-frequency ultrasound (US) can enhance nonviral gene transfer to the mouse lung. Cationic lipid GL67/pDNA, polyethylenimine (PEI)/pDNA and naked plasmid DNA (pDNA) were delivered via intranasal instillation, mixed with Optison microbubbles, and the animals were then exposed to 1 MHz US. Addition of Optison alone significantly reduced the transfection efficiency of all three gene transfer agents. US exposure did not increase GL67/pDNA or PEI/pDNA gene transfer compared to Optison-treated animals. However, it increased naked pDNA transfection efficiency by approximately 15-fold compared to Optison-treated animals, suggesting that despite ultrasound being attenuated by air in the lung, sufficient energy penetrates the tissue to increase gene transfer. US-induced lung haemorrhage, assessed histologically, increased with prolonged US exposure. The left lung was more affected than the right and this was mirrored by a lesser increase in naked pDNA gene transfer, in the left lung. The positive effect of US was dependent on Optison, as in its absence US did not increase naked pDNA transfection efficiency. We have thus established proof of principle that US can increase nonviral gene transfer, in the air-filled murine lung.


Subject(s)
Albumins , DNA/administration & dosage , Fluorocarbons , Genetic Therapy/methods , Lung/metabolism , Transfection/methods , Ultrasonics , Animals , DNA/genetics , Gene Expression , Luciferases/genetics , Lung Diseases/therapy , Male , Mice , Mice, Inbred BALB C , Polyethyleneimine
3.
Gene Ther ; 13(5): 449-56, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16319950

ABSTRACT

Sendai virus (SeV) is able to transfect airway epithelial cells efficiently in vivo. However, as with other viral vectors, repeated administration leads to reduced gene expression. We have investigated the impact of inducing immunological tolerance to immunodominant T-cell epitopes on gene expression following repeated administration. Immunodominant CD4 and CD8 T-cell peptide epitopes of SeV were administered to C57BL/6 mice intranasally 10 days before the first virus administration with transmission-incompetent F-protein-deleted DeltaF/SeV-GFP. At 21 days after the first virus administration, mice were again transfected with DeltaF/SeV. To avoid interference of anti-GFP antibodies, the second transfection was carried out with DeltaF/SeV-lacZ. At 2 days after the final transfection lung beta-galactosidase expression, T-cell proliferation and antibody responses were measured. A state of 'split tolerance' was achieved with reduced T-cell proliferation, but no impact on antiviral antibody production. There was no enhancement of expression on repeat administration; instead, T-cell tolerance was, paradoxically, associated with a more profound extinction of viral expression. Multiple immune mechanisms operate to eradicate viruses from the lung, and these findings indicate that impeding the adaptive T-cell response to the immunodominant viral epitope is not sufficient to prevent the process.


Subject(s)
Genetic Therapy/methods , Lung/immunology , Sendai virus/genetics , T-Lymphocytes/immunology , Viral Vaccines/administration & dosage , Animals , CD4 Antigens/immunology , CD8 Antigens/immunology , Cell Proliferation , Female , Gene Expression , Genetic Engineering , Immune Tolerance/genetics , Immunodominant Epitopes/immunology , Mice , Mice, Inbred C57BL , Sendai virus/immunology , Viral Vaccines/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...