Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 39
Filtrer
1.
Catheter Cardiovasc Interv ; 54(4): 533-8, 2001 Dec.
Article de Anglais | MEDLINE | ID: mdl-11747197

RÉSUMÉ

Early studies have indicated no correlation between the amount of mechanical injury and the level of myocardial gene expression following direct plasmid vector injection. Recently, however, evidence suggests that combined laser myocardial injury and plasmid-based gene delivery exert synergistic effects on gene expression and activity. The purpose of the study was to determine whether laser-induced myocardial injury followed by transendocardial gene transfer increases gene expression compared to gene transfer alone. We assessed the ability of a plasmid vector to express its transgene after injection into porcine ischemic myocardium with and without preceding laser myocardial injury. Thirteen animals had transendocardial injections of the luciferase reporter gene in a plamid vector using a catheter-based injection system. Injections (0.5 mg per animal, 50 microg per injection site) were divided into 10 sites in the ischemic territory. Eight animals underwent transendocardial laser injury of the ischemic region (2 Joule per pulse x 10 sites) prior to gene delivery. In five animals, gene injection sites were dispersed between laser channels, and in three animals laser and gene delivery were applied in close proximity (< 5 mm) or at the same location. Luciferase activity was measured at 3 and 7 days. Luciferase expression in ischemic zones was markedly elevated at day 3 and 7, and similar whether animals were pretreated using laser injury followed by gene transfer compared to gene transfer alone. Neither same-spot injection nor dispersed gene delivery were associated with augmented gene expression compared to gene transfer alone. Using the above-described catheter-based approach to combine localized laser injury and injection of naked DNA into ischemic myocardium, laser injury did not augment gene expression above levels present with gene transfer alone.


Sujet(s)
Thérapie génétique/méthodes , Lésions traumatiques du coeur/étiologie , Thérapie laser , Ischémie myocardique/génétique , Ischémie myocardique/chirurgie , Myocarde/métabolisme , Plasmides/génétique , Plasmides/usage thérapeutique , Animaux , Techniques électrophysiologiques cardiaques , Expression des gènes/génétique , Techniques de transfert de gènes , Vecteurs génétiques/génétique , Vecteurs génétiques/usage thérapeutique , Ventricules cardiaques/traumatismes , Luciferases/biosynthèse , Luciferases/génétique , Modèles cardiovasculaires , Suidae , Résultat thérapeutique
2.
Coron Artery Dis ; 11(8): 615-9, 2000 Dec.
Article de Anglais | MEDLINE | ID: mdl-11107509

RÉSUMÉ

BACKGROUND: Direct transfer of genes holds promise for the sustained delivery of therapeutic proteins to treat cardiovascular diseases. This can be accomplished by several approaches, including use of adenoviral vectors and naked plasmid DNA vectors. We previously demonstrated achieval of effective delivery of genes into the myocardium with a left ventricular-guided catheter-based approach using an adenoviral vector. OBJECTIVE: To evaluate the levels and duration of expression of genes induced after injection of a specific plasmid vector, using the same delivery platform as that in our previous work. METHODS: The pCOR plasmids are narrow-host-range plasmid vectors designed for nonviral gene therapy. We tested the ability of the pCOR plasmid vector to express its transgene after injection into the myocardium of pigs with chronic experimental ischemia using a catheter-based transendocardial delivery system. Four animals were subjected to transendocardial injections of the luciferase reporter pCOR gene into ischemic and nonischemic zones using the Biosense intramyocardial injection catheter. Injections (1 mg per animal, 50 micrograms per injection site) were performed at 20 sites in ischemic and nonischemic zones. Measurements of luciferase activity were performed 3 and 7 days thereafter. RESULTS: We observed high levels of expression of luciferase gene in ischemic and nonischemic regions (on days 3 and 7, respectively, in ischemic zone 58,237 and 33,709 pg; in nonischemic zone 39,928 and 46,036 pg). Control noninjected samples from the left and right ventricles contained no detectable luciferase activity. CONCLUSIONS: With a catheter-based approach, the pCOR plasmid was successfully used to deliver genes into designated myocardial regions, and provides sustained expression of protein for at least 7 days, of roughly similar magnitudes in ischemic and nonischemic myocardium.


Sujet(s)
Techniques de transfert de gènes , Thérapie génétique/méthodes , Vecteurs génétiques , Luciferases/génétique , Ischémie myocardique/thérapie , Plasmides , Animaux , Suidae , Facteurs temps
3.
Gene Ther ; 7(16): 1353-61, 2000 Aug.
Article de Anglais | MEDLINE | ID: mdl-10981661

RÉSUMÉ

Though the efficacy of intravascular gene transfer has been demonstrated in native vessels following acute injury, this methodology has not been validated in complex models of vascular injury that more closely mimic clinical angioplasty procedures. Previous studies have shown that Gax gene overexpression modulates the injury-induced remodeling of the vessel in rat carotid and normal rabbit iliac arteries. Here, we evaluated the effect of the Gax gene delivery in atheromatous stented vessels. Rabbits were fed 120 g daily of 1% cholesterol diet for 3 weeks. At 1 week they underwent initial injury on the external iliac artery, then balloon angioplasty was performed at 3 weeks at the same site with a 2.5 mm diameter channel balloon catheter (three times 1 min at 6 atm). Either saline (n = 4) or the control viral construct Ad-CMVluc (5 x 109 p.f.u.) (n = 5) or Ad-CMVGax (5 x 10(9) p.f.u.) (n = 4) was delivered with a poloxamer mixture via a channel balloon (6 atm, 30 min), and a 15 mm long Palmaz-Schatz stent (PS154) was then deployed at the site (1 min, 8 atm). Arteries were analyzed 1 month later. At 1 month, the Ad-CMVGax treated arteries exhibited a lower maximal intimal area (1. 15+/-0.1 mm2) than saline (1.87+/-0.15 mm2, P = 0.007) or Ad-CMVluc-treated vessels (1.98+/-0.31 mm2, P = 0.04). Likewise Ad-CMVGax-treated vessels displayed a lower maximal percentage cross-sectional area narrowing (35.1+/-3.5%) than saline (65.3+/-9.4%, P = 0.01) or Ad-CMVluc-treated vessels (62.7+/-6.7%, P = 0.02). Angiographic analysis revealed larger minimal lumen diameter in Ad-CMVGax treated arteries (2.0+/-0.1 mm) than saline (1.14+/-0.36 mm, P = 0.06) or Ad-CMVluc-treated vessels (1.23+/-0.25 mm, P = 0.02). Overexpression of the Gax gene inhibits neointimal hyperplasia and lumen loss in atheromatous stented rabbit iliac arteries.


Sujet(s)
Adenoviridae/génétique , Artériosclérose/thérapie , Techniques de transfert de gènes , Thérapie génétique/méthodes , Vecteurs génétiques/administration et posologie , Protéines à homéodomaine/génétique , Protéines du muscle/génétique , Animaux , Artériosclérose/imagerie diagnostique , Artériosclérose/anatomopathologie , Expression des gènes , Hyperplasie , Artère iliaque/imagerie diagnostique , Artère iliaque/traumatismes , Artère iliaque/anatomopathologie , Mâle , Lapins , Radiographie , Récidive , Statistique non paramétrique , Endoprothèses , beta-Galactosidase/génétique
4.
Circ Res ; 87(6): 448-52, 2000 Sep 15.
Article de Anglais | MEDLINE | ID: mdl-10988235

RÉSUMÉ

Ischemia induces both hypoxia and inflammation that trigger angiogenesis. The inflammatory reaction is modulated by production of anti-inflammatory cytokines. This study examined the potential role of a major anti-inflammatory cytokine, interleukin (IL)-10, on angiogenesis in a model of surgically induced hindlimb ischemia. Ischemia was produced by artery femoral occlusion in both C57BL/6J IL-10(+/+) and IL-10(-/-) mice. After 28 days, angiogenesis was quantified by microangiography, capillary, and arteriole density measurement and laser Doppler perfusion imaging. The protein levels of IL-10 and vascular endothelial growth factor (VEGF) were determined by Western blot analysis in hindlimbs. IL-10 was markedly expressed in the ischemic hindlimb of IL-10(+/+) mice. Angiogenesis in the ischemic hindlimb was significantly increased in IL-10(-/-) compared with IL-10(+/+) mice. Indeed, angiographic data showed that vessel density in the ischemic leg was 10.2+/-0.1% and 5.7+/-0.4% in IL-10(-/-) and IL-10(+/+) mice, respectively (P:<0.01). This corresponded to improved ischemic/nonischemic leg perfusion ratio by 1.4-fold in IL-10(-/-) mice compared with IL-10(+/+) mice (0.87+/-0. 05 versus 0.63+/-0.01, respectively; P:<0.01). Revascularization was associated with a 1.8-fold increase in tissue VEGF protein level in IL-10(-/-) mice compared with IL-10(+/+) mice (P:<0.01). In vivo electrotransfer of murine IL-10 cDNA in IL-10(-/-) mice significantly inhibited both the angiogenic process and the rise in VEGF protein level observed in IL-10(-/-) mice. No changes in vessel density or VEGF content were observed in the nonischemic hindlimb. These findings underscore the antiangiogenic effect of IL-10 associated with the downregulation of VEGF expression and suggest a role for the inflammatory balance in the modulation of ischemia-induced angiogenesis.


Sujet(s)
Membre pelvien/vascularisation , Interleukine-10/métabolisme , Ischémie/physiopathologie , Néovascularisation physiologique , Animaux , Artérioles/physiologie , Vaisseaux capillaires/physiologie , ADN complémentaire/génétique , Facteurs de croissance endothéliale/métabolisme , Techniques de transfert de gènes , Interleukine-10/génétique , Ischémie/génétique , Fluxmétrie laser Doppler , Lymphokines/métabolisme , Mâle , Souris , Souris de lignée C57BL , Muscles squelettiques/métabolisme , Facteur de croissance endothéliale vasculaire de type A , Facteurs de croissance endothéliale vasculaire
5.
Curr Cardiol Rep ; 2(1): 39-47, 2000 Jan.
Article de Anglais | MEDLINE | ID: mdl-10980871

RÉSUMÉ

Several phase I/II clinical trials are currently ongoing in gene therapy of cardiovascular disease. Whereas the indications vary, including peripheral artery disease, ischemic heart disease, post-angioplasty restenosis, and vein graft failure, these trials are mostly based on the use of adenoviral vectors and nonviral vectors. Novel vectors aimed at improving the efficacy and safety of gene delivery in target organs, such as heart, skeletal muscle, vasculature, and liver, have been recently generated. Some of them have already been successfully validated in preclinical models of cardiovascular disease. This review focuses on the most recent advances in vector development that could substantially increase the spectrum of cardiovascular pathologies amenable to gene transfer-based treatments.


Sujet(s)
Maladies cardiovasculaires/thérapie , Thérapie génétique , Vecteurs génétiques , Adenoviridae/génétique , Dependovirus/génétique , Régulation de l'expression des gènes , Techniques de transfert de gènes , Humains , Lentivirus/génétique , Virus de la leucémie murine/génétique , Plasmides , Transduction génétique
6.
J Biol Chem ; 275(38): 29643-7, 2000 Sep 22.
Article de Anglais | MEDLINE | ID: mdl-10882714

RÉSUMÉ

Previous studies have indicated that advanced age is associated with impaired angiogenesis in part because of reduced levels of vascular endothelial growth factor (VEGF) expression. To investigate potential mechanisms responsible for this age-dependent defect in VEGF expression, aortic smooth muscle cells isolated from young rabbits (ages 6-8 months) or old rabbits (ages 4-5 years) were exposed to normoxic (21% oxygen) or hypoxic (0.1% oxygen) conditions. Hypoxia-induced VEGF expression was significantly lower in old versus young cells. VEGF mRNA stability in hypoxic conditions was similar in both young and old cells. However, transient transfection with a luciferase reporter gene that was transcriptionally regulated by the VEGF promoter revealed a significant defect in VEGF up-regulation following hypoxia in old versus young cells (a 43 versus 117% increase in luciferase activity, p < 0.05); this difference was not seen when a deletion construct lacking the hypoxia-inducible 1 (HIF-1) binding site was used. Moreover, although HIF-1 alpha-mRNA expression was shown to be similar in young and old smooth muscle cells, HIF-1 alpha protein and DNA binding activity were significantly reduced in old versus young smooth muscle cells that were exposed to hypoxia. We propose that age-dependent reduction in hypoxia-induced VEGF expression results from reduced HIF-1 activity and may explain the previously described age-dependent impairment of angiogenesis in response to ischemia.


Sujet(s)
Vieillissement/physiologie , Protéines de liaison à l'ADN/génétique , Facteurs de croissance endothéliale/génétique , Régulation de l'expression des gènes/physiologie , Lymphokines/génétique , Protéines nucléaires/génétique , Animaux , Hypoxie cellulaire/génétique , Cellules cultivées , Protéines de liaison à l'ADN/métabolisme , Facteurs de croissance endothéliale/biosynthèse , Facteur-1 induit par l'hypoxie , Sous-unité alpha du facteur-1 induit par l'hypoxie , Lymphokines/biosynthèse , Protéines nucléaires/métabolisme , Lapins , Facteurs de transcription/génétique , Facteurs de transcription/métabolisme , Facteur de croissance endothéliale vasculaire de type A , Facteurs de croissance endothéliale vasculaire
7.
Arterioscler Thromb Vasc Biol ; 20(2): 435-42, 2000 Feb.
Article de Anglais | MEDLINE | ID: mdl-10669641

RÉSUMÉ

The apolipoprotein E (apoE)-deficient mouse is a relevant animal model of human atherosclerosis. Although the prevention of atherosclerosis development has been documented after somatic gene transfer into animal models, regression of lesions remains to be demonstrated. Thus, we used this genetically defined mouse model nn the nude background to show atherosclerosis regression. ApoE-deficient nude mice were infected with 5 x 10(8) or 10(9) plaque-forming units of a first-generation adenovirus encoding human apoE cDNA. The secretion of human apoE resulted in a rapid decrease of total cholesterol, which normalized the hypercholesterolemic phenotype within 14 days (from 600+/-100 to <100 microg/mL). Transgene expression was observed during a period of >4 months, with a normalization of cholesterol and triglyceride levels during 5 months. At that time, we successfully reinjected the recombinant adenovirus and observed the appearance of the human protein as well as the correction of lipoprotein phenotype. In mice killed 6 months-after the first infection, we observed a dose-dependent regression of fatty streak lesions in the aorta. We showed sustained expression of a transgene with a first-generation adenoviral vector and a correction of dyslipoproteinemia phenotype leading to lesion regression. These data demonstrate that somatic gene transfer can induce plaque regression.


Sujet(s)
Apolipoprotéines E/déficit , Apolipoprotéines E/génétique , Artériosclérose/métabolisme , Artériosclérose/anatomopathologie , Techniques de transfert de gènes , Animaux , Apolipoprotéines E/métabolisme , Artériosclérose/génétique , Humains , Lipoprotéines/sang , Souris , Souris de lignée C57BL , Souris nude , Souris transgéniques/génétique , Distribution tissulaire
8.
J Clin Invest ; 104(10): 1469-80, 1999 Nov.
Article de Anglais | MEDLINE | ID: mdl-10562309

RÉSUMÉ

Homeobox transcription factors specify body plan by regulating differentiation, proliferation, and migration at a cellular level. The homeobox transcription factor Gax is expressed in quiescent vascular smooth muscle cells (VSMCs), and its expression is downregulated by vascular injury or other conditions that lead to VSMC proliferation. Previous investigations demonstrate that Gax may regulate VSMC proliferation by upregulating the cyclin-dependent kinase (cdk) inhibitor p21. Here we examined whether Gax influences VSMC migration, a key feature in the development of stenotic lesions after balloon injury. Transduction of a Gax cDNA inhibited the migratory response of VSMCs toward PDGF-BB, basic fibroblast growth factor, or hepatocyte growth factor/scatter factor. Gax expression also inhibited migration of NIH.3T3 fibroblasts and embryonic fibroblasts lacking p53. Gax was unable to inhibit the migration of fibroblasts lacking p21, but this effect could be restored in these cells by providing exogenous p21 or by overexpressing another cdk inhibitor, p16. Flow cytometric analysis implicated a Gax-mediated downregulation of alpha(v)beta(3) and alpha(v)beta(5) integrin expression in VSMCs as a potential cause for reduced cell motility. Gax specifically downregulated beta(3) and beta(5) in VSMCs in culture and after acute vascular injury in vivo. Repression of integrin expression was also found in NIH 3T3 cells and p53 knockout fibroblasts, but not in p21-knockout fibroblasts, unless these cells express exogenous p21 or p16. These data suggest that cycle progression, integrin expression, and cell migration can be regulated in VSMCs by the homeobox gene product Gax.


Sujet(s)
Régulation de l'expression des gènes , Protéines à homéodomaine/métabolisme , Intégrines/génétique , Protéines du muscle/métabolisme , Muscles lisses vasculaires/physiologie , Récepteur vitronectine/génétique , Facteurs de transcription/métabolisme , Cellules 3T3 , Animaux , Aorte thoracique/cytologie , Aorte thoracique/physiologie , Bécaplermine , Mouvement cellulaire/effets des médicaments et des substances chimiques , Survie cellulaire , Cellules cultivées , Facteur de croissance fibroblastique de type 2/pharmacologie , Gènes homéotiques , Facteur de croissance des hépatocytes/pharmacologie , Protéines à homéodomaine/génétique , Cinétique , Mâle , Souris , Protéines du muscle/génétique , Muscles lisses vasculaires/cytologie , Muscles lisses vasculaires/effets des médicaments et des substances chimiques , Facteur de croissance dérivé des plaquettes/métabolisme , Facteur de croissance dérivé des plaquettes/pharmacologie , Protéines proto-oncogènes c-sis , Rats , Rat Sprague-Dawley , Récepteurs aux facteurs de croissance dérivés des plaquettes/métabolisme , Protéines recombinantes/métabolisme , Transfection
9.
Gene Ther ; 6(5): 758-63, 1999 May.
Article de Anglais | MEDLINE | ID: mdl-10505098

RÉSUMÉ

Adenovirus-mediated gene delivery in animal models of vascular injury has provided insights into the mechanisms underlying vessel wall pathologies. We have previously demonstrated that overexpression of the Gax transcription factor inhibits neointimal formation in rat and rabbit models of arterial injury. Here, we evaluate potential mechanisms for the reduction in stenotic lesion size due to Gax overexpression. At 3, 7 and 14 days after injury the Ad-Gax-infected arteries displayed a marked decrease in medial vascular smooth muscle cell number (3 days, 54% reduction P < 0.01; 7 days, 41% reduction P < 0.003; 14 days, 49% reduction P < 0.02). At 3 days after injury, PCNA expression was attenuated in the Ad-Gax-treated vessels compared with control vessels (65% reduction P < 0.02), indicating a reduction in cellular proliferation. At 7 days and 14 days after injury Ad-Gax-infected arteries exhibited elevated number of TUNEL-positive medial VSMCs compared with control-treated arteries (7 days, 9.2-fold increase P < 0.03; 14 days, 17.2-fold increase P < 0.03), indicating an induction of apoptotic cell death. These data suggest that deregulated Gax expression induces first cell cycle arrest and then apoptosis in the vascular smooth muscle cells that contribute to the neointimal layer. Therefore, the efficacy of this therapeutic strategy appears to result from the ability of the Gax transcriptional regulator to modulate multiple cellular responses.


Sujet(s)
Adenoviridae/génétique , Sténose carotidienne/thérapie , Thérapie génétique/méthodes , Vecteurs génétiques/administration et posologie , Muscles lisses vasculaires/anatomopathologie , Facteurs de transcription/génétique , Animaux , Apoptose/génétique , Lésions traumatiques de l'artère carotide/anatomopathologie , Sténose carotidienne/anatomopathologie , Division cellulaire/génétique , Expression des gènes , Immunohistochimie , Mâle , Modèles biologiques , Antigène nucléaire de prolifération cellulaire/analyse , Rats , Rat Sprague-Dawley
10.
Circulation ; 99(18): 2445-51, 1999 May 11.
Article de Anglais | MEDLINE | ID: mdl-10318668

RÉSUMÉ

BACKGROUND: In humans, fibrates are frequently used normolipidemic drugs. Fibrates act by regulating genes involved in lipoprotein metabolism via activation of the peroxisome proliferator-activated receptor-alpha (PPARalpha) in liver. In rodents, however, fibrates induce a peroxisome proliferation, leading to hepatomegaly and possibly hepatocarcinogenesis. Although this peroxisome proliferative response appears not to occur in humans, it remains controversial whether the beneficial effects of fibrates on lipoprotein metabolism can occur dissociated from such undesirable peroxisomal response. Here, we assessed the influence of fenofibrate on lipoprotein metabolism and peroxisome proliferation in the rabbit, an animal that, contrary to rodents and similar to humans, is less sensitive to peroxisome proliferators. METHODS AND RESULTS: First, we demonstrate that in normal rabbits, fenofibrate given at a high dose for 2 weeks does not influence serum concentrations or intestinal mRNA levels of the HDL apolipoprotein apoA-I. Therefore, the study was continued with human apoA-I transgenic rabbits that overexpress the human apoA-I gene under control of its homologous promoter, including its PPAR-response elements. In these animals, fenofibrate increases serum human apoA-I concentrations via an increased expression of the human apoA-I gene in liver. Interestingly, liver weight or mRNA levels and activity of fatty acyl-CoA oxidase, a rate-limiting and marker enzyme of peroxisomal beta-oxidation, remain unchanged after fenofibrate. CONCLUSIONS: Expression of the human apoA-I transgene in rabbit liver suffices to confer fibrate-mediated induction of serum apoA-I. Furthermore, these data provide in vivo evidence that the beneficial effects of fibrates on lipoprotein metabolism occur mechanistically dissociated from any deleterious activity on peroxisome proliferation and possibly hepatocarcinogenesis.


Sujet(s)
Anticholestérolémiants/usage thérapeutique , Apolipoprotéine A-I/métabolisme , Fénofibrate/usage thérapeutique , Microcorps/effets des médicaments et des substances chimiques , Proliférateurs des péroxysomes/pharmacologie , Lapins/métabolisme , Acyl-CoA oxidase , Animaux , Animal génétiquement modifié , Anticholestérolémiants/pharmacologie , Apolipoprotéine A-I/génétique , Cholestérol HDL/sang , Résistance aux substances , Fénofibrate/pharmacologie , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Humains , Muqueuse intestinale/métabolisme , Intestins/effets des médicaments et des substances chimiques , Lipoprotein lipase/métabolisme , Lipoprotéines HDL/métabolisme , Foie/effets des médicaments et des substances chimiques , Foie/enzymologie , Foie/anatomopathologie , Tumeurs expérimentales du foie/induit chimiquement , Taille d'organe/effets des médicaments et des substances chimiques , Spécificité d'organe , Oxidoreductases/analyse , Proliférateurs des péroxysomes/toxicité , Protéines de fusion recombinantes/métabolisme , Rodentia/métabolisme , Spécificité d'espèce
11.
Proc Natl Acad Sci U S A ; 96(8): 4262-7, 1999 Apr 13.
Article de Anglais | MEDLINE | ID: mdl-10200250

RÉSUMÉ

Gene delivery to skeletal muscle is a promising strategy for the treatment of muscle disorders and for the systemic secretion of therapeutic proteins. However, present DNA delivery technologies have to be improved with regard to both the level of expression and interindividual variability. We report very efficient plasmid DNA transfer in muscle fibers by using square-wave electric pulses of low field strength (less than 300 V/cm) and of long duration (more than 1 ms). Contrary to the electropermeabilization-induced uptake of small molecules into muscle fibers, plasmid DNA has to be present in the tissue during the electric pulses, suggesting a direct effect of the electric field on DNA during electrotransfer. This i.m. electrotransfer method increases reporter and therapeutic gene expression by several orders of magnitude in various muscles in mouse, rat, rabbit, and monkey. Moreover, i.m. electrotransfer strongly decreases variability. Stability of expression was observed for at least 9 months. With a pCMV-FGF1 plasmid coding for fibroblast growth factor 1, this protein was immunodetected in the majority of muscle fibers subjected to the electric pulses. DNA electrotransfer in muscle may have broad applications in gene therapy and in physiological, pharmacological, and developmental studies.


Sujet(s)
Stimulation électrique/méthodes , Techniques de transfert de gènes , Muscles squelettiques/physiologie , Animaux , Électroporation/méthodes , Gènes rapporteurs , Haplorhini , Humains , Luciferases/génétique , Luciferases/métabolisme , Souris , Souris de lignée C57BL , Fibres musculaires squelettiques/physiologie , Lapins , Rats , Protéines de fusion recombinantes/biosynthèse , Protéines recombinantes/métabolisme , beta-Galactosidase/biosynthèse , beta-Galactosidase/génétique
12.
Circulation ; 99(1): 105-10, 1999.
Article de Anglais | MEDLINE | ID: mdl-9884386

RÉSUMÉ

BACKGROUND: Apolipoprotein (apo) A-I is the major component of HDL, and it displays antiatherogenic properties. METHODS AND RESULTS: The human apoA-I gene has been transferred into different mouse models by use of a recombinant adenovirus under the control of an RSV-LTR promoter (AV RSV apoA-I). Administration of AV RSV apoA-I to C57BL/6 mice resulted in moderate expression of human apoA-I for 3 weeks, leading to a transient elevation (40% at day 11 after injection) of HDL cholesterol concentration. In contrast, administration of AV RSV apoA-I to human apoA-I-transgenic mice induced a large increase of human apoA-I and HDL cholesterol concentrations (300% and 360%, respectively, at day 14 after injection) for 10 weeks, indicating that an immune response to the transgene was one major hurdle for long-term duration of expression. Recombinant adenovirus expressing human apolipoprotein A-I (AV RSV apoA-I) was also injected into human apoA-I-transgenic/apoE-deficient mice, which are prone to develop atherosclerosis. Over a 6-week period, overexpression of human apoA-I inhibited fatty streak lesion formation by 56% in comparison with control. CONCLUSIONS: Somatic gene transfer of human apoA-I prevents the development of atherosclerosis in the mouse model.


Sujet(s)
Apolipoprotéine A-I/génétique , Artériosclérose/thérapie , Techniques de transfert de gènes , Adenoviridae/génétique , Animaux , Apolipoprotéines E/déficit , Modèles animaux de maladie humaine , Évolution de la maladie , Vecteurs génétiques , Humains , Souris , Souris de lignée C57BL , Valeurs de référence
13.
Clin Invest Med ; 21(4-5): 172-85, 1998.
Article de Anglais | MEDLINE | ID: mdl-9800066

RÉSUMÉ

The authors previously demonstrated that the gene for human lipoprotein lipase (hLPL), an enzyme crucial to the breakdown of triglyceride (TG)-rich dietary fats, corrects the hypertriglyceridemia in lipoprotein lipase (LPL)-deficient knockout mice after adenoviral (Ad)-mediated LPL gene transfer. They have now extended their observations to primary cultured mouse hepatocytes and intact animals of normal LPL genotype, and confirm effective overexpression of hLPL from the liver and a sustained TG-lowering effect in plasma over 60 days. A typical first-generation Ad-vector containing the hLPL cDNA (Ad-LPL) resulted in efficient gene transfer into isolated mouse hepatocytes and significant de novo synthesis of active hLPL protein. In this experiment, 5 x 10(9) viral particles (5 x 10(7) pfu) of either Ad-LPL or an Ad-LacZ control vector were injected into CD1 mice of normal LPL genotype. Hepatic expression of hLPL was confirmed at Day 7 postinjection by in situ hybridization and direct measurement of LPL in the liver. This correlated with a total LPL activity (human + mouse) in postheparin plasma (PHP) of 1020.5 standard deviation [SD] 93.6 mU/mL, versus 479.5 SD 129.7 mU/mL (p < 0.001) in Ad-LacZ controls at Day 7. Respective hLPL activity comprised 49% of the total. Significantly raised levels of hLPL protein mass persisted until Day 60. Corresponding plasma TGs decreased to 39% of Ad-LacZ controls at Day 7, and, despite absent hLPL activity from Day 28 on, serum TGs remained significantly lower in Ad-LPL mice up to Day 42. Fast phase liquid chromatography analysis showed a dramatic depletion in TG-rich lipoproteins, mainly very low density lipoproteins (VLDL) and chylomicron fractions. Therefore, Ad-mediated overexpression of hepatic LPL was found to significantly decrease plasma TG levels unrelated to primary LPL deficiency.


Sujet(s)
Adenoviridae/génétique , Techniques de transfert de gènes , Lipolyse/génétique , Lipoprotein lipase/génétique , Foie/enzymologie , Animaux , Cellules cultivées , Chylomicron/sang , Régulation de l'expression des gènes codant pour des enzymes/génétique , Humains , Hybridation in situ , Lipoprotein lipase/métabolisme , Lipoprotéines/sang , Foie/virologie , Mâle , Souris , Lignées consanguines de souris , Triglycéride/sang
14.
EMBO J ; 17(13): 3576-86, 1998 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-9649428

RÉSUMÉ

Tissues with the highest rates of proliferation typically exhibit the highest frequencies of apoptosis, but the mechanisms that coordinate these processes are largely unknown. The homeodomain protein Gax is down-regulated when quiescent cells are stimulated to proliferate, and constitutive Gax expression inhibits cell proliferation in a p21(WAF/CIP)-dependent manner. To understand how mitogen-induced proliferation influences the apoptotic process, we investigated the effects of deregulated Gax expression on cell viability. Forced Gax expression induced apoptosis in mitogen-activated cultures, but quiescent cultures were resistant to cell death. Though mitogen activation was required for apoptosis, neither the cdk inhibitor p21(WAF/CIP) nor the tumor suppressor p53 was required for Gax-induced cell death. Arrest in G1 or S phases of the cell cycle with chemical inhibitors also did not affect apoptosis, further suggesting that Gax-mediated cell death is independent of cell cycle activity. Forced Gax expression led to Bcl-2 down-regulation and Bax up-regulation in mitogen-activated, but not quiescent cultures. Mouse embryonic fibroblasts homozygous null for the Bax gene were refractive to Gax-induced apoptosis, demonstrating the functional significance of this regulation. These data suggest that the homeostatic balance between cell growth and death can be controlled by mitogen-dependent pathways that circumvent the cell cycle to alter Bcl-2 family protein expression.


Sujet(s)
Apoptose , Cycle cellulaire , Protéines à homéodomaine/métabolisme , Protéines du muscle/métabolisme , Protéines proto-oncogènes c-bcl-2/métabolisme , Protéines proto-oncogènes/métabolisme , Animaux , Cellules cultivées , Inhibiteur p21 de kinase cycline-dépendante , Cyclines/génétique , Cyclines/métabolisme , Phase G1 , Protéines à homéodomaine/génétique , Hydroxy-urée/pharmacologie , Mâle , Souris , Mitogènes/pharmacologie , Protéines du muscle/génétique , Muscles lisses vasculaires/cytologie , Phénotype , Polyènes/pharmacologie , Protéines proto-oncogènes/génétique , Rats , Rat Sprague-Dawley , Phase S , Sérumalbumine bovine/pharmacologie , Sirolimus , Protéine p53 suppresseur de tumeur/génétique , Protéine p53 suppresseur de tumeur/métabolisme , Protéine Bax
15.
Hum Gene Ther ; 9(7): 1013-24, 1998 May 01.
Article de Anglais | MEDLINE | ID: mdl-9607413

RÉSUMÉ

UNLABELLED: Reduction in transfection time and the ability to perform gene transfer in conjunction with endovascular stent implantation constitute two important challenges for percutaneous adenovirus-mediated gene transfer to vessel walls. Studies have suggested that the use of biocompatible polyol poloxamer 407 could be useful. We first evaluated the use of poloxamer 407 for percutaneous gene transfer in nonstented rabbit iliac arteries. A 200-microl mixture of Ad-RSVbetagal or Ad-CMVLuc in either phosphate-buffered saline (PBS) or 20% poloxamer was delivered. After 3 days, gene transfection was evaluated by X-Gal staining or measurement of luciferase activity. Poloxamer use resulted in a 3- to 15-fold increase in the percentage of transfected cells (X-Gal, p = 0.001) and a 16-fold increase in protein product (luciferase activity, p = 0.03), and allowed a decrease in transfection time from 30 to 5 min with minimal reduction in transfection efficiency. We then evaluated the feasibility of percutaneous gene transfer, using Ad-RSVbetagal diluted in pure PBS or 20% poloxamer, in conjunction with stent implantation. Gene delivery was performed either immediately before (pre-) or after (post-) stent implantation. When adenoviruses were diluted in PBS, gene transfer had a low efficiency (prestent, 0.3%; poststent, 0.2%; NS). With poloxamer, the efficacy was much higher (p = 0.0001) and similar "pre" (2.2%) or "post" (1.7%) stent delivery (NS). CONCLUSIONS: (1) The use of poloxamer, rather than PBS, as a vehicle increases the efficacy of percutaneous adenovirus-mediated gene transfer and reduces transfection time; (2) gene transfer performed during stent implantation with poloxamer is feasible and achieves a significant level of gene expression. Thus percutaneous gene delivery is applicable to conventional stents and could present an attractive method by which to achieve local biological effects in a stent environment.


Sujet(s)
Adénovirus humains , Matériaux biocompatibles , Techniques de transfert de gènes , Vecteurs génétiques , Artère iliaque/métabolisme , Poloxalène , Endoprothèses , Transfection , Angioplastie coronaire par ballonnet , Animaux , Femelle , Gènes rapporteurs , Humains , Opéron lac , Mesures de luminescence , Lapins , Facteurs temps
16.
Biochim Biophys Acta ; 1391(3): 329-36, 1998 Apr 22.
Article de Anglais | MEDLINE | ID: mdl-9555083

RÉSUMÉ

Rat hepatocytes cocultured with rat liver epithelial cells (RLEC) were used to investigate the influence of all-trans retinoic acid (RA) on the regulation of apolipoproteins (Apo) A-I and A-II gene expression, the major protein constituent of high-density lipoproteins. In contrast to rat hepatocytes in conventional primary culture, Apo A-I and Apo A-II gene expression remained high and stable for several days in parenchymal cells in coculture. Treatment of cocultured rat hepatocytes with RA resulted in a specific decrease in Apo A-I mRNA levels whereas no marked difference in Apo A-II mRNA levels was observed. Such a negative effect of RA was already detected as early as 2 days of treatment and was effective for the entire experimental period (6 days). As controls, RARbeta mRNA levels increased whereas those of GAPDH mRNA were not affected by the RA treatment. The decrease in Apo A-I mRNA levels was associated with lower amounts of Apo A-I secreted in the culture medium within day 1 of treatment. This effect required active transcription and protein synthesis. These results show that, contrary to primary pure hepatocyte cultures and hepatoma cell lines, cocultures of rat hepatocytes reproduce the in vivo results suggesting that only well differentiated hepatocytes may correctly respond to RA. Furthermore, they demonstrate that RA can directly act on hepatocytes and differently affect Apo A-I and Apo A-II gene expression.


Sujet(s)
Apolipoprotéine A-I/génétique , Régulation négative/génétique , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Foie/métabolisme , Trétinoïne/pharmacologie , Albumines/effets des médicaments et des substances chimiques , Albumines/métabolisme , Animaux , Apolipoprotéine A-I/biosynthèse , Apolipoprotéine A-I/effets des médicaments et des substances chimiques , Cellules cultivées , Techniques de coculture , Régulation négative/effets des médicaments et des substances chimiques , Cellules épithéliales/métabolisme , Foie/cytologie , Foie/effets des médicaments et des substances chimiques , Mâle , Rats , Rat Sprague-Dawley
17.
Gene Ther ; 5(8): 1023-30, 1998 Aug.
Article de Anglais | MEDLINE | ID: mdl-10326024

RÉSUMÉ

The endothelium and internal elastic lamina (IEL) appear to be the main barriers to adenovirus-mediated gene transfer to medial smooth muscle cells (SMC). The present randomized study tested whether controlled incubation with elastase enhanced the efficiency of catheter-based gene transfer to medial SMC by adenoviral vectors. After an initial safety dose ranging study, rabbits underwent balloon abrasion of the iliac endothelium followed by local incubation of either elastase (2 x 10(-7) IU over 5 min) or saline using a double balloon catheter (DBC). Then, adenoviral vectors (5 x 10(9) p.f.u.) carrying Cmv-Luc or RSV-beta gal reporter genes were instilled for 30 min. Three days later, the number of medial SMC expressing lacZ was increased in the elastase-treated arteries compared with saline-treated arteries (7.2 +/- 2.5 versus 2.3 +/- 0.9 cells per section, P = 0.003). Likewise, the amount of luciferase protein product was increased (70 +/- 32 versus 36 +/- 15 pg luciferase/mg tissue, P = 0.03). No vessel enlargement, light or electron microscopic evidence of injury or inflammation was seen in elastase-treated arteries up to 7 weeks. Preincubation with elastase increased transduction efficiency of catheter-based gene delivery of replication-defective adenoviral vectors to rabbit iliac arteries without detectable arterial damage.


Sujet(s)
Adenoviridae , Techniques de transfert de gènes , Thérapie génétique/méthodes , Vecteurs génétiques/administration et posologie , Muscles lisses vasculaires/enzymologie , Pancreatic elastase/administration et posologie , Animaux , Cathétérisme , Élasticité , Gènes rapporteurs , Artère iliaque/anatomie et histologie , Artère iliaque/traumatismes , Luciferases/métabolisme , Microscopie électronique , Muscles lisses vasculaires/anatomie et histologie , Pancreatic elastase/métabolisme , Projets pilotes , Lapins , Répartition aléatoire , beta-Galactosidase/métabolisme
18.
Arterioscler Thromb Vasc Biol ; 17(11): 2532-9, 1997 Nov.
Article de Anglais | MEDLINE | ID: mdl-9409224

RÉSUMÉ

Humans homozygous or heterozygous for mutations in the lipoprotein lipase (LPL) gene demonstrate significant disturbances in plasma lipoproteins, including raised triglyceride (TG) and reduced HDL cholesterol levels. In this study we explored the feasibility of adenovirus-mediated gene replacement therapy for LPL deficiency. A total of 5 x 10(9) plaque-forming units (pfu) of an E1/E3-deleted adenovirus expressing either human LPL (Ad-LPL) or the bacterial beta-galactosidase gene (Ad-LacZ) as a control were administered to mice heterozygous for targeted disruption in the LPL gene (n = 57). Peak expression of total postheparin plasma LPL activity was observed at day 7 in Ad-LPL mice versus Ad-LacZ controls (834 +/- 133 vs 313 +/- 89 mU/mL, P < .01), and correlated with human-specific LPL activity (522 +/- 219 mU/mL) and mass (9214 +/- 782 ng/mL), a change that was significant to 14 and 42 days, respectively. At day 7, plasma TGs were significantly reduced relative to Ad-LacZ mice (0.17 +/- 0.07 vs 1.90 +/- 0.89 mmol/L, P < .01) but returned to endogenous levels by day 42. Ectopic liver expression of human LPL was confirmed by in situ hybridization analysis and from raised LPL activity and mass in liver homogenates. Analysis of plasma lipoprotein composition revealed a marked decrease in VLDL-derived TGs. Severely impaired oral and intravenous fat-load tolerance in LPL-deficient mice was subsequently corrected after Ad-LPL administration and closely paralleled that observed in wild-type mice. These findings suggest that liver-targeted adenovirus-mediated LPL gene transfer offers an effective means for transient correction of altered lipoprotein metabolism and impaired fat tolerance due to LPL deficiency.


Sujet(s)
Thérapie génétique , Hyperlipoprotéinémie de type I/thérapie , Hypertriglycéridémie/thérapie , Lipoprotein lipase/génétique , Adenoviridae/génétique , Animaux , Matières grasses alimentaires , Vecteurs génétiques/génétique , Hétérozygote , Humains , Hypertriglycéridémie/étiologie , Lipoprotein lipase/sang , Lipoprotein lipase/physiologie , Foie/enzymologie , Foie/métabolisme , Mâle , Souris , Souris de lignée C57BL , Souris knockout , Protéines de fusion recombinantes/physiologie
19.
Development ; 124(21): 4405-13, 1997 Nov.
Article de Anglais | MEDLINE | ID: mdl-9334288

RÉSUMÉ

The development of the tubular heart into a complex four-chambered organ requires precise temporal and region-specific regulation of cell proliferation, migration, death and differentiation. While the regulatory mechanisms in heart morphogenesis are not well understood, increasing attention has focused on the homeodomain proteins, which are generally linked to morphogenetic processes. The homeodomain containing gene Gax has been shown to be expressed in heart and smooth muscle tissues. In this study, the Gax protein was detected in the nuclei of myocardial cells relatively late in chicken heart development, at a time when myocyte proliferation is declining. To test the hypothesis that the Gax protein functions as a negative regulator of cardiomyocyte proliferation, a replication-defective adenovirus was used to force its precocious nuclear expression during chicken heart morphogenesis. In experiments in which Gax- and beta-galactosidase-expressing adenoviruses were co-injected, clonal expansion of myocytes was reduced, consistent with inhibition of myocyte proliferation. This effect on proliferation was corroborated by the finding that the percentage of exogenous Gax-expressing myocytes that were positive for the cell cycle marker PCNA decreased over time and was lower than in control myocytes. The precocious nuclear expression of Gax in tubular hearts resulted in abnormal heart morphology, including small ventricles with rounded apices, a thinned compact zone and coarse trabeculae. These results suggest a role for the Gax protein in heart morphogenesis causing proliferating cardiomyocytes to withdraw from the cell cycle, thus influencing the size and shape that the heart ultimately attains.


Sujet(s)
Coeur/croissance et développement , Protéines à homéodomaine/génétique , Protéines du muscle/génétique , Myocarde/cytologie , Myocarde/métabolisme , Adenoviridae/génétique , Animaux , Division cellulaire , Embryon de poulet , Régulation de l'expression des gènes au cours du développement , Vecteurs génétiques/génétique , Cardiopathies congénitales/génétique , Protéines à homéodomaine/métabolisme , Protéines du muscle/métabolisme
20.
Genes Dev ; 11(13): 1674-89, 1997 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-9224717

RÉSUMÉ

gax, a diverged homeobox gene expressed in vascular smooth muscle cells (VSMCs), is down-regulated in vitro by mitogen stimulation and in vivo in response to vascular injury that leads to cellular proliferation. Recombinant Gax protein microinjected into VSMCs and fibroblasts inhibited the mitogen-induced entry into S-phase when introduced either during quiescence or early stages of G1. Overexpression of gax with a replication-defective adenovirus vector resulted in G0/G1 cell cycle arrest of VSMCs and fibroblasts. The gax-induced growth inhibition correlated with a p53-independent up-regulation of the cyclin-dependent kinase inhibitor p21. Gax overexpression also led to an association of p21 with cdk2 complexes and a decrease in cdk2 activity. Fibroblasts deficient in p21 were not susceptible to a reduction in cdk2 activity or growth inhibition by gax overexpression. Localized delivery of the virus to denuded rat carotid arteries significantly reduced neointima formation and luminal narrowing. These data indicate that gax overexpression can inhibit cell proliferation in a p21-dependent manner and can modulate injury-induced changes in vessel wall morphology that result from excessive cellular proliferation.


Sujet(s)
Kinases CDC2-CDC28 , Cyclines/physiologie , Protéines à homéodomaine/physiologie , Protéines du muscle/physiologie , Adenoviridae/génétique , Adenoviridae/physiologie , Angioplastie par ballonnet/effets indésirables , Animaux , Lésions traumatiques de l'artère carotide , Division cellulaire , Lignée cellulaire , Kinase-2 cycline-dépendante , Inhibiteur p21 de kinase cycline-dépendante , Kinases cyclines-dépendantes/antagonistes et inhibiteurs , Kinases cyclines-dépendantes/génétique , Virus défectifs/génétique , Virus défectifs/physiologie , Régulation de l'expression des gènes , Vecteurs génétiques , Protéines à homéodomaine/génétique , Humains , Protéines du muscle/génétique , Muscles lisses vasculaires/cytologie , Muscles lisses vasculaires/métabolisme , Protein-Serine-Threonine Kinases/antagonistes et inhibiteurs , Protein-Serine-Threonine Kinases/génétique , Rats , Protéines de fusion recombinantes/génétique , Protéine p53 suppresseur de tumeur/génétique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE