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










Database
Publication year range
2.
Hepatology ; 33(1): 10-5, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11124814

ABSTRACT

Stable gene transfer into hepatocytes has been proposed to compensate for genetic deficiencies that affect liver function, or to deliver diffusible factors into the circulation. This strategy can be achieved using retroviral vectors; however, cell division must occur. We describe a simple and reproductive method that enables the induction of hepatocyte replication in a controlled fashion, thus allowing an efficient in vivo retroviral liver transduction that is applicable to mouse models of human genetic disorders. The approach is based on liver susceptibility to apoptosis via the Fas/CD95 pathway. We show that, 4 days following a single Fas agonist antibody (JO2) injection, hepatocyte replication occurs, the intensity of which is correlated with the level of the induced hepatic cytolysis. This treatment enables in vivo liver transduction, and its efficiency also correlates with the level of hepatic cytolysis. When recombinant retroviral vectors were infused intravenously during the period of hepatocyte replication, 15.4% +/- 1.7% of the hepatocytes were transduced, reaching up to 32.5%.


Subject(s)
Gene Transfer Techniques , Genetic Vectors , Liver Regeneration/physiology , Retroviridae/genetics , fas Receptor/physiology , Alanine Transaminase/blood , Animals , Antibodies, Monoclonal/pharmacology , Cell Division/drug effects , Dose-Response Relationship, Drug , Female , Hepatocytes/cytology , Injections , Mice , Mice, Inbred C57BL , Transduction, Genetic , Ultrafiltration , fas Receptor/immunology
3.
Hum Mol Genet ; 9(11): 1597-602, 2000 Jul 01.
Article in English | MEDLINE | ID: mdl-10861286

ABSTRACT

Liver repopulation constitutes an attractive approach for the treatment of liver disorders or of diseases requiring abundant secretion of an active protein. We have described previously a model of selective repopulation of a normal liver by Fas/CD95-resistant hepatocytes, in which we achieved up to 16% hepatocyte repopulation. In the present study, we investigated the therapeutic efficacy of this strategy. With this aim, apolipoprotein E (ApoE) knockout mice were transplanted with Fas/CD95-resistant hepatocytes which constitutively express ApoE. Transplanted mice were submitted to weekly injections of non-lethal doses of the Fas agonist antibody Jo2. After 8 weeks of treatment, we obtained up to 30% of the normal level of plasma ApoE. ApoE secretion was accompanied by a drastic and significant decrease in total plasma cholesterol, which even fell to normal levels. Moreover, this secretion was sufficient to markedly reduce the progression of atherosclerosis. These results demonstrate the efficacy of this repopulation approach for correcting a deficiency in a protein secreted by the liver.


Subject(s)
Cell Transplantation , Hypercholesterolemia/therapy , Liver/cytology , Animals , Apolipoproteins E/blood , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Arteriosclerosis/pathology , Cholesterol/blood , Disease Models, Animal , Humans , Lipoproteins/blood , Liver/metabolism , Liver/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Mutation , Proto-Oncogene Proteins c-bcl-2/metabolism
4.
Hum Mol Genet ; 8(5): 831-8, 1999 May.
Article in English | MEDLINE | ID: mdl-10196372

ABSTRACT

The severe neurodegenerative disorder, Tays-Sachs disease, is caused by a beta-hexosaminidase alpha-subunit deficiency which prevents the formation of lysosomal heterodimeric alpha-beta enzyme, hexosaminidase A (HexA). No treatment is available for this fatal disease; however, gene therapy could represent a therapeutic approach. We previously have constructed and characterized, in vitro, adenoviral and retroviral vectors coding for alpha- and beta-subunits of the human beta-hexosaminidases. Here, we have determined the in vivo strategy which leads to the highest HexA activity in the maximum number of tissues in hexA -deficient knock-out mice. We demonstrated that intravenous co-administration of adenoviral vectors coding for both alpha- and beta-subunits, resulting in preferential liver transduction, was essential to obtain the most successful results. Only the supply of both subunits allowed for HexA overexpression leading to massive secretion of the enzyme in serum, and full or partial enzymatic activity restoration in all peripheral tissues tested. The enzymatic correction was likely to be due to direct cellular transduction by adenoviral vectors and/or uptake of secreted HexA by different organs. These results confirmed that the liver was the preferential target organ to deliver a large amount of secreted proteins. In addition, the need to overexpress both subunits of heterodimeric proteins in order to obtain a high level of secretion in animals defective in only one subunit is emphasized. The endogenous non-defective subunit is otherwise limiting.


Subject(s)
Adenoviridae/genetics , Genetic Therapy/methods , Genetic Vectors/pharmacology , Tay-Sachs Disease/therapy , beta-N-Acetylhexosaminidases/genetics , Animals , Genetic Vectors/administration & dosage , Hexosaminidase A , Humans , Injections, Intramuscular , Isoenzymes , Liver/enzymology , Mice , Mice, Inbred Strains , Mice, Knockout , Muscle, Skeletal/enzymology , Tay-Sachs Disease/enzymology , Tay-Sachs Disease/genetics , Tissue Distribution , Transduction, Genetic , beta-N-Acetylhexosaminidases/deficiency , beta-N-Acetylhexosaminidases/metabolism
5.
Nat Med ; 4(10): 1185-8, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9771754

ABSTRACT

Hepatocyte transplantation might represent a potential therapeutic alternative to liver transplantation in the future; however, transplanted cells have a limited capacity to repopulate the liver, as they do not proliferate under normal conditions. Recently, studies in urokinase (uPA) transgenic mice and in fumarylacetoacetate hydrolase (FAH)-deficient mice have shown that the liver can be repopulated by genetically engineered hepatocytes harboring a selective advantage over resident hepatocytes. We have reported that transgenic mice expressing human Bcl-2 in their hepatocytes are protected from Fas/CD95-mediated liver apoptosis. We now show that Bcl-2 transplanted hepatocytes selectively repopulate the liver of mice treated with nonlethal doses of the anti-Fas antibody Jo2. FK 506 immunosuppressed mice were transplanted by splenic injection with Bcl-2 hepatocytes. The livers of female recipients were repopulated by male Bcl-2 transgenic hepatocytes, as much as 16%, after 8 to 12 administrations of Jo2. This only occurred after anti-Fas treatment, confirming that resistance to Fas-induced apoptosis constituted the selective advantage of these transplanted hepatocytes. Thus, we have demonstrated a method for increasing genetic reconstitution of the liver through selective repopulation with modified transgenic hepatocytes, which will allow optimization of cell and gene therapy in the liver.


Subject(s)
Apoptosis , Cell Transplantation/methods , Liver/cytology , Selection, Genetic , fas Receptor/metabolism , Animals , Chimera , Female , Genes, bcl-2 , Humans , Male , Mice , Mice, Inbred CBA , Mice, Transgenic
6.
Gene Ther ; 3(9): 769-74, 1996 Sep.
Article in English | MEDLINE | ID: mdl-8875224

ABSTRACT

Tay-Sachs disease (TSD) is a lysosomal storage disease due to hexosaminidase A deficiency caused by mutations in the gene for alpha-chain (Hex alpha). A human Hex alpha cDNA was subcloned into the adenoviral plasmid pAdRSV. Hex alpha. Replication-deficient adenovirus was generated by homologous recombination in 293 cells. Human fibroblasts from a patient suffering from TSD were infected with the recombinant adenovirus. TSD fibroblasts expressing the recombinant alpha-chain had an enzyme activity on the natural substrate ranging from 40 to 84% of the normal. The corrected cells secreted up to 25 times more Hex alpha than control fibroblasts. The Hex alpha encoded by the adenovirus was shown to be correctly transported into the lysosomes and to normalize the impaired degradation of GM2 ganglioside in TSD fibroblasts.


Subject(s)
Adenoviridae/genetics , Fibroblasts/enzymology , Gene Transfer Techniques , Genetic Vectors/genetics , Tay-Sachs Disease/genetics , beta-N-Acetylhexosaminidases/genetics , DNA, Complementary/genetics , G(M2) Ganglioside/metabolism , Gene Expression , Hexosaminidase A , Humans , Lysosomes/enzymology , RNA, Messenger/analysis , Tay-Sachs Disease/enzymology , beta-N-Acetylhexosaminidases/metabolism
7.
C R Seances Soc Biol Fil ; 190(1): 9-11, 1996.
Article in French | MEDLINE | ID: mdl-8881264

ABSTRACT

In hereditary neurological diseases, gene transfer into neurons is made difficult by: the nature of the cells (postmitotic cells, that cannot be cultured, genetically modified ex vivo, then retransplanted), sometimes, their widespread localization, the blood-brain barrier. However, three viral vectors derived from adenovirus, Herpes simplex virus and adeno-associated virus have been shown to be very efficient in transferring DNA into brain cells. All of these vectors can infect resting cells, especially neurons, and are efficient in vivo. Retroviral vectors which can infect dividing cells only are mainly used for ex vivo genetic modification of cells (neural progenitor cells, myoblasts, fibroblasts) followed by intracerebral transplantation. Alternatively, genetically modified cells can be transplanted in a peripheral site if the transgene product is able to cross the blood-brain barrier or to be transported retrogradely from the nerve terminals. We have especially investigated the potential interest of adenoviral vectors to transfer foreign genes into brain cells and to treat animal models of neurological diseases. These vectors allowed us to transfer the lacZ gene into any neural cell type, including neurons, glia, photoreceptors and olfactory receptors, ex vivo, in cell culture, and in vivo, by stereotactic administration. In addition, axonal transport of adenoviral vectors has been demonstrated, e.g. in the substantia nigra after injection into the striatum, in the olfactory bulb after intranasal instillation and in spinal motor neurons after intramuscular injection. After intracerebroventricular injection, ependymal cells are massively infected and express the transgene for several months, as this is also observed in neurons. Through the spinal canal and cerebrospinal fluid, the vector can diffuse to a considerable distance from the injection point, e.g. to the lumbar spinal cord after injection in the suboccipital region. To test the biological function of transgenes transferred through adenoviral vectors, we have constructed vectors with cDNAs or genes for various neutrophic factors: CNTF, NT3, BDNF and GDNF. These vectors were biologically active on target cells, ex vivo and in vivo. In the pmn mouse model of progressive motor neuronal degeneration, some of these vectors, alone or combined, allowed for prolongation of life of homozygous animals by more than two fold, and for decrease in the demyelination of phrenic nerve axons. Finally, we have also constructed an adenoviral vector carrying the alpha-hexosaminidase cDNA, encoding the enzyme subunit deficient in Tay Sachs patients. This vector permitted to normalize ganglioside metabolism in Tay Sachs fibroblasts and is currently tested in knock out mice deficient in hexosaminidase A. In spite of all these encouraging results, we are nevertheless aware that progress in vector design and delivery strategies will be needed before gene therapy can become a realistic therapeutical strategy in humans.


Subject(s)
Genetic Therapy , Hereditary Sensory and Autonomic Neuropathies/therapy , Adenoviridae/genetics , Animals , Genetic Vectors
SELECTION OF CITATIONS
SEARCH DETAIL
...