Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Cardiovasc Res ; 49(2): 288-97, 2001 Feb 01.
Article in English | MEDLINE | ID: mdl-11164839

ABSTRACT

OBJECTIVE: We examined the functional consequences of expressing adult rabbit fast skeletal sarcoplasmic reticulum (SR) Ca(2+)-ATPase (SERCA1a) in isolated adult rat ventricular myocytes. METHODS: Myocytes were infected with a recombinant adenovirus harboring SERCA1a. Then 2 days after myocyte infection, protein expression was estimated using Western blot and SDS-PAGE analysis. We also measured the ATP-dependent oxalate-facilitated Ca(2+) uptake of myocyte homogenates and monitored Ca(2+) transient in myocytes loaded with the Ca(2+) dye, indo-1. RESULTS: SERCA1a gene expression resulted in a 36% increase in the total SERCA protein level in infected myocytes compared to controls (P<0.01), while SERCA2 and phospholamban levels did not change. This increase was associated with a 42% rise in SR Ca(2+) uptake (P<0.01), while tau (the time constant of Ca(2+) transient decay), and the time to peak fell by 32% (P<0.01) and 38% (P<0.001), respectively. Increasing the frequency of stimulation from 0.2 to 2 Hz decreased tau in both cell types (P<0.01). However, the decrease was much smaller in infected (P<0.01) than in uninfected cells (P<0.001). Isoproterenol (1 microM) further decreased tau in infected myocytes by 23% (P<0.05). In these cells, the diastolic [Ca(2+)](i) decreased by 50% (P<0.05) while the systolic [Ca(2+)](i) increased by 19% (P<0.05). No difference was found in the speed of SR Ca(2+) reloading after caffeine washout between the two cell types. CONCLUSION: Adenovirus-mediated SERCA1a gene transfer to adult rat ventricular myocytes enhances SR Ca(2+) handling to a degree similar to that observed following physiological stimulation.


Subject(s)
Adenoviridae/genetics , Calcium-Transporting ATPases/genetics , Calcium/metabolism , Genetic Vectors/administration & dosage , Myocardium/metabolism , Sarcoplasmic Reticulum/enzymology , Analysis of Variance , Animals , Blotting, Western , Calcium-Transporting ATPases/analysis , Calcium-Transporting ATPases/metabolism , Cells, Cultured , Gene Transfer Techniques , Male , Microscopy, Confocal , Myocardium/enzymology , Rats , Rats, Wistar , Sarcoplasmic Reticulum Calcium-Transporting ATPases
2.
Gene Ther ; 6(7): 1210-21, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10455429

ABSTRACT

The present work studies the effects of a replication-deficient adenovirus (Ad), Ad-RSVbFGF, bearing the human basic fibroblast growth factor (bFGF) cDNA, as a potential vector for therapeutic angiogenesis of ischemic diseases. The different isoforms of the protein were expressed from the viral vector in various cell types and, although the cytoplasmic isoform does not possess a signal peptide, we observed its release from a muscle cell line. The proteins were fully functional when tested in a long-term survival assay of quiescent fibroblasts. After endothelial cell infection with Ad-RSVbFGF, we observed an 80&percnt increase in the mean length of the capillary-like tubes that differentiated in a three-dimensional model of angiogenesis. We evaluated angiogenesis directly in mice 14 days after subcutaneous injection of Matrigel plugs containing Ad-RSVbFGF. A marked neovascularization was observed in the Matrigel plugs and in the surrounding tissues. Finally, the recombinant virus was injected into the hindlimb muscles of mdx mice. A 2.5-fold increase in bFGF content of the muscle was observed 6 days after injection, without any significant variations detected in the animal sera. Immunohistological detection showed an increased number of large-caliber vessels in the treated muscles as compared with control muscles. These results demonstrate that Ad-mediated transfer of the human bFGF gene can induce angiogenesis in muscle, making this tissue a potential target for the treatment of ischemic diseases.


Subject(s)
Adenoviridae/genetics , Fibroblast Growth Factor 2/genetics , Gene Transfer Techniques , Genetic Vectors/administration & dosage , Muscle, Skeletal/blood supply , Neovascularization, Physiologic , Animals , Blotting, Western , Fibroblast Growth Factor 2/analysis , Genetic Therapy/methods , Humans , Ischemia/therapy , Mice , Protein Isoforms/analysis , Protein Isoforms/genetics
3.
Hum Gene Ther ; 9(13): 1919-28, 1998 Sep 01.
Article in English | MEDLINE | ID: mdl-9741430

ABSTRACT

Adenoviruses are attractive vectors for gene transfer into cardiac muscle. However, their promiscuous tissue tropism, which leads to an ectopic expression of the transgene, is a considerable limitation. To restrict expression to cardiomyocytes, we have constructed two recombinant adenoviruses (Ad-MLC2-250betagal and Ad-MLC2-2100betagal) containing the beta-galactosidase reporter gene under the control of the 250- or 2100-bp rat ventricle-specific cardiac myosin light chain-2v promoter (MLC-2v). Our in vitro and in vivo data have evidenced that the 2100-bp promoter allows stronger beta-galactosidase activity than the 250-bp promoter and that the deleted promoter allows a weak beta-galactosidase expression in skeletal muscle-derived cells in vitro. In contrast to the in vitro results, the highly deleted MLC-2v promoter of 250 pb conserved its heart specificity in in ovo and in vivo when introduced into the adenovirus genome, indicating that the specificity of this promoter is neither altered by the inverted terminal repeat nor by the enhancer of the Ela promoter, both of which located in the 5' flanking region of the promoter. Systemic injections of both recombinant adenoviruses into chicken embryos showed beta-galactosidase expression mainly in the right ventricle of the heart. We have confirmed the cardiac specificity of both promoters in mammalian species after injection of both recombinant adenoviruses into the heart of adult rats in vivo. The comparison of both promoters in vitro and in vivo has shown that the 250-bp MLC-2v promoter is 80% less active than the 2100-bp MLC-2v promoter and has enabled us to conclude that the MLC-2v promoter of 2100 bp is the most appropriate for efficient expression of a reporter gene or a therapeutic cardiac gene (e.g., SERCA2a or minidystrophin gene).


Subject(s)
Adenoviridae/genetics , Cardiac Myosins , Gene Transfer Techniques , Genetic Vectors/genetics , Heart Ventricles/metabolism , Myosin Light Chains/genetics , Adenovirus E1A Proteins/genetics , Animals , Cells, Cultured , Chick Embryo , DNA, Viral/analysis , Enhancer Elements, Genetic/genetics , Female , Gene Expression Regulation/genetics , Genes, Reporter/genetics , Muscle, Skeletal/metabolism , Myocardium/cytology , Organ Specificity , Promoter Regions, Genetic/genetics , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Terminal Repeat Sequences/genetics , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
4.
Gene Ther ; 5(8): 1105-13, 1998 Aug.
Article in English | MEDLINE | ID: mdl-10326034

ABSTRACT

AdmATF is a recombinant adenovirus encoding a secreted version of the amino-terminal fragment (ATF) of murine urokinase (uPA). This defective adenovirus was used in three murine models to assess the antitumoral effects associated with local or systemic delivery of ATF, a broad cell invasion inhibitor that antagonizes uPA binding to its cell surface receptor (uPAR). A single intratumoral injection of AdmATF into pre-established MDA-MB-231 human breast xenografts grown in athymic mice, or into pre-established C57/BL6 syngeneic Lewis lung carcinoma resulted in a specific arrest of tumor growth. Neovascularization within and at the vicinity of the injection site was also suppressed, suggesting that AdmATF inhibited primary tumor growth by targeting angiogenesis. AdmATF also interfered with tumor cell establishment at distant sites: (1) lung dissemination of Lewis lung carcinoma cells was significantly reduced following intratumoral injection at the primary site; and (2) systemic administration of AdmATF inhibited subsequent liver metastasis in a LS174T human colon carcinoma xenograft model. These data outline the potential of using a recombinant adenovirus directing the secretion of an antagonist of cell-associated uPA for cancer gene therapy.


Subject(s)
Adenoviridae/genetics , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Neoplasms/therapy , Neovascularization, Pathologic/prevention & control , Urokinase-Type Plasminogen Activator/genetics , Animals , Breast Neoplasms/pathology , Breast Neoplasms/therapy , Female , Genetic Engineering/methods , Humans , Injections, Intralesional , Injections, Intramuscular , Liver Neoplasms/secondary , Liver Neoplasms/therapy , Lung Neoplasms/pathology , Lung Neoplasms/therapy , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Plasminogen Inactivators/pharmacology , Rats , Rats, Inbred Lew , Receptors, Cell Surface/antagonists & inhibitors , Receptors, Urokinase Plasminogen Activator , Tumor Cells, Cultured , Urokinase-Type Plasminogen Activator/antagonists & inhibitors
5.
Biochim Biophys Acta ; 1330(1): 8-16, 1997 Nov 13.
Article in English | MEDLINE | ID: mdl-9375808

ABSTRACT

Substantial effort has been focused on the development of highly efficient gene transfer strategies. Although viral and non-viral methods have been elaborated, mechanisms of gene delivery are still poorly understood. We exploited our recent observation that replication-deficient type 5 adenovirus dramatically enhances lipofectAMINE-mediated gene transfer (lipoadenofection) in differentiated cells to elucidate the mechanism of adenovirus action in this process. Heat-induced denaturation of viral capsid abolishes adenovirus action whereas inactivation of viral genome by short treatment with UV has no effect. Electron microscopic observations reveal the formation of a complex containing adenovirus and lipofectAMINE which probably carries DNA into cells via endocytosis. Anti-adenovirus antiserum or monoclonal anti-alpha(v)beta3 integrin antibody inhibits lipoadenofection, at least partially. Neutralization of endosomal compartments with chloroquine, ammonium chloride or monensin does not prevent adenovirus improvement of gene transfer. Hence, adenovirus-lipofectAMINE-DNA complexes in which viral particles are each encompassed by three lipid layers, penetrate cells via an endocytic pathway involving probably the adenovirus receptor and alpha(v)beta3 integrin. The resulting efficient transfer and expression of plasmid DNA proceeds from a mechanism in which adenoviral endosomolytic activity appears to be required while viral genome is not essential.


Subject(s)
Adenoviridae/genetics , Gene Transfer Techniques , Liposomes , Capsid/genetics , Cell Line , Chloramphenicol O-Acetyltransferase/genetics , DNA, Viral , Endocytosis , Genome, Viral , Humans , Hydrogen-Ion Concentration , beta-Galactosidase/genetics
6.
C R Acad Sci III ; 320(2): 103-12, 1997 Feb.
Article in English | MEDLINE | ID: mdl-9181118

ABSTRACT

Adenoviruses are very attractive vectors for gene transfer into the cardiac muscle; however, their promiscuous tissue tropism, leading to an ectopic expression of the transgene, is a considerable practical limitation. To restrict expression of a reporter gene in cultured cardiomyocytes and in the heart of the rat, we have constructed a recombinant adenovirus (Ad-MLC2 beta gal) containing the beta-galactosidase gene under the control of the rat ventricle-specific cardiac myosin light chain 2 (MLC-2v) promoter. We show in this work that the MLC-2v promoter inside the adenoviral genome retains its cardiac specificity in vitro in cultured cardiomyocytes as well as in vivo in the animal heart. Northern blot studies after Ad-MLC2 beta gal infection show significant transcription only in cells derived from the cardiac muscle and not from the skeletal muscle. Quantitative analysis of the beta-galactosidase activity in a number of cell lines also confirms this result. The level of beta-galactosidase expression in rat neonatal cardiomyocytes infected with Ad-MLC2 beta gal is 8% of that found when primary cells are infected with Ad-RSV beta gal (containing a beta-galactosidase gene under the control of the Rous sarcoma virus promoter). The cardiomyocytes-specific expression is also found after injection of Ad-MLC2 beta gal directly into the rat myocardium, although the viral genome can be detected by polymerase chain reaction (PCR) in other tissues. Lack of expression after direct injection into liver and skeletal muscle confirms these results. The use of a tissue-specific promoter is a first step to restrict transgene expression to a particular cell type of the targeted tissue.


Subject(s)
Mastadenovirus/genetics , Myocardium/cytology , Promoter Regions, Genetic/genetics , Animals , Cell Line, Transformed , Cells, Cultured , Gene Expression , Gene Transfer Techniques , In Vitro Techniques , Liver/cytology , Muscle, Skeletal/cytology , Myosin Light Chains/genetics , Rats , Recombination, Genetic , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
8.
J Natl Cancer Inst ; 88(24): 1857-63, 1996 Dec 18.
Article in English | MEDLINE | ID: mdl-8961977

ABSTRACT

BACKGROUND: Despite vigorous efforts at curbing tobacco consumption and aggressive combined-modality treatment programs, both the incidence of and the mortality from lung cancer have remained virtually unchanged in the last 10 years. More effective innovative therapies are clearly needed. The direct transfer into tumor cells of tumor suppressor genes or toxic gene products that specifically promote tumor cell death and spare nonmalignant cells is a potentially novel anticancer treatment approach that should be investigated. PURPOSE: On the basis of compelling preclinical data, we initiated a phase I study involving six patients with inoperable lung cancer and an endobronchial lesion accessible by bronchoscopy. Our purpose was to evaluate the feasibility, tolerance, and clinical, biologic, and immunologic effects of the intratumoral administration of a recombinant, replication-deficient adenovirus (rAd.RSV beta-gal), using the Rous sarcoma virus promoter to drive transcription of the Escherichia coli lacZ marker gene that encodes for the bacterial enzyme beta-galactosidase (beta-gal). METHODS: From June 1994 through April 1995, six patients (five males and one female) were enrolled in the trial. A single dose of recombinant virus suspension containing 10(7) or 10(8) plaque-forming units (PFU) was injected intratumorally into two successive cohorts of three patients. Eligible patients received concomitant chemotherapy. Patients were kept under isolation conditions from 3 days before the injection was given until virus excretion was undetectable. Biopsy specimens of the tumor and surrounding mucosa were collected on the 8th day and at 1, 2, and 3 months after injection. They were analyzed by cell culture, polymerase chain reaction (PCR), and beta-gal expression for the presence of recombinant adenovirus. So that the risk of virus recombination or complementation could be minimized, wildtype adenovirus carriers among the hospital staff (identified by PCR) were excluded from contact with patients who were potentially excreting recombinant virus. RESULTS: beta-gal was expressed in tumor biopsy specimens of three patients (one who received the 10(7) PFU dose level and two who received 10(8)). Bronchoalveolar lavage specimens collected immediately after injection were positive for recombinant adenovirus when analyzed in culture and by PCR. All biologic fluids were negative for recombinant virus as judged by PCR after day 12, with the exception of bronchoalveolar lavage specimens (positive PCR up to 90 days in two of three patients treated with 10(8) PFU). The blood samples obtained from the three patients treated with 10(8) PFU showed positive PCR results immediately after virus injection. Patients were kept in isolation for a median of 17 days. The most common toxic effects were moderate bleeding (occurring in two patients) during bronchoscopy and fever (seen in four patients). Endoscopic and clinically objective antitumor responses were seen in four patients, including one patient who showed a complete response by pathologic evaluation. The median survival for the patients was 12.5 months (range, 3-16+ months). Throughout the study, hospital staff remained negative for recombinant adenovirus infection. CONCLUSIONS: This ongoing phase I study has demonstrated that a recombinant adenovirus-mediated marker gene, such as rAd.RSV beta-gal, can be safely introduced into humans and that the gene product is expressed by lung tumor cells of the host.


Subject(s)
Bronchial Neoplasms/therapy , Carcinoma/therapy , Genetic Therapy/methods , Lung Neoplasms/therapy , beta-Galactosidase/genetics , Adenoviridae , Bronchial Neoplasms/enzymology , Bronchoalveolar Lavage Fluid , Bronchoscopy , Carcinoma/enzymology , Feasibility Studies , Gene Transfer Techniques , Genetic Vectors , Humans , Lung Neoplasms/enzymology , Polymerase Chain Reaction
9.
Proc Natl Acad Sci U S A ; 93(8): 3570-4, 1996 Apr 16.
Article in English | MEDLINE | ID: mdl-8622977

ABSTRACT

Fast skeletal muscles of mdx (X chromosome-linked muscular dystrophy) mice were injected after birth with a recombinant adenovirus containing a minidys- trophin gene, a 6.3-kbp cDNA coding for the N- and C-terminal ends of dystrophin. Adult muscles were challenged by forced lengthening during tetanic contractions. Stretch-induced mechanical and histological damages were much reduced in injected muscles, in direct proportion of the Miniber of fibers expressing minidystrophin. Damaged fibers were preferentially found among minidystrophin-negative regions. Minidystrostrophin confers an important functional and structural protection of limb muscles against high mechanical stress, even after a partial somatic gene transfer.


Subject(s)
Dystrophin/genetics , Gene Transfer Techniques , Genetic Therapy , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Animal/therapy , Adenoviridae/genetics , Animals , Genetic Vectors , In Vitro Techniques , Mice , Mice, Inbred mdx , Muscle Contraction , Muscle Fibers, Fast-Twitch/physiology , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Muscular Dystrophy, Animal/physiopathology , Stress, Mechanical
10.
C R Seances Soc Biol Fil ; 190(1): 13-31, 1996.
Article in French | MEDLINE | ID: mdl-8881265

ABSTRACT

This review focuses on the general properties of adenovirus vectors and their application to muscle gene transfer. In the prospect of a treatment for Duchenne muscular dystrophy, in vivo expression of the dystrophin gene in animal models remains the main concern. After the presentation of the two partners: adenovirus and muscle, we summarize our results of gene transfer in mice. We have evaluated efficiency of systemic and intramuscular injections, the impact of age at injection, the duration of expression in adult normal and genetically-immunosuppressed SCID mice, using a recombinant adenovirus expressing a reporter gene. After adenovirus-mediated transfer of a mini-model, we have shown an efficient and stable expression of the transgene, a long-term correction of the degeneration process and a functional protection of the treated muscle. The discussion focuses on the problems and the perspectives for gene therapy: safety problems, improvement of safety and vector capacity, host immune response, delivery to muscle and muscle-targeted expression.


Subject(s)
Adenoviridae/genetics , Gene Transfer Techniques , Genetic Vectors , Muscle, Skeletal , Muscular Dystrophies/therapy , Adenoviridae/immunology , Animals , Antibody Formation , Clinical Trials as Topic , Disease Models, Animal , Graft vs Host Disease/immunology , Humans , Mice , Mice, SCID , Muscle, Skeletal/physiology
11.
Gene Ther ; 2(1): 16-21, 1995 Jan.
Article in English | MEDLINE | ID: mdl-7712329

ABSTRACT

Direct introduction of exogenous genes into pre-existent tumors could provide an effective therapeutic approach for treatment of localized tumors. In this report we show that direct intratumoral delivery in animals of a replication-deficient adenovirus vector harboring the murine interleukin (IL)-2 gene (AD-mIL2) causes complete disappearance of P815 murine mastocytoma tumors in up to 75% of cases. Histological analysis of treated tumors revealed the presence of several zones of necrosis and the infiltration of macrophages and T cells. Moreover, the successfully treated animals develop a long lasting state of immunity during which further challenges with the tumor cells are rejected. To our knowledge this is the first successful in vivo treatment of an established tumor using adenoviral gene therapy methods.


Subject(s)
Adenoviruses, Human/genetics , Gene Transfer Techniques , Genetic Therapy/methods , Genetic Vectors , Interleukin-2/genetics , Mast-Cell Sarcoma/therapy , Animals , Cell Division , Cell Size , DNA, Recombinant/genetics , Immunohistochemistry , Interleukin-2/biosynthesis , Mast-Cell Sarcoma/immunology , Mast-Cell Sarcoma/pathology , Mast-Cell Sarcoma/virology , Mice , Mice, Inbred DBA
12.
Circulation ; 90(4): 1648-56, 1994 Oct.
Article in English | MEDLINE | ID: mdl-7923648

ABSTRACT

BACKGROUND: Previous investigations in live animals convincingly established that arterial gene transfer, while feasible, was compromised by a low transfection efficiency. More recent studies have shown that transfection efficiency may be substantially augmented by the use of recombinant adenoviral vectors. Most in vivo transfections reported to date, however, have used direct (operative) administration of the adenoviral vector. Clinical applications of arterial gene transfer (such as prevention of restenosis), however, would require local percutaneous delivery of the transgene. The present study was designed to extend in vivo intraoperative findings to percutaneous delivery system and to assess whether gene transfer remains site specific. METHODS AND RESULTS: A recombinant, replication-defective adenovirus modified to include an expression cassette for nucleus-targeted beta-galactosidase was introduced into rabbit iliac arteries in vivo using either a double-balloon catheter (DBC, n = 27) or a hydrogel-coated balloon catheter (HBC, n = 27). Contralateral arteries-normal, endothelium-denuded, or sham-transfected with a control adenoviral vector-served as controls. beta-Galactosidase expression was assessed by X-Gal staining. Cell-transduction efficiency was measured by morphometric analysis. Polymerase chain reaction (PCR) and histochemistry were used to detect the presence and/or expression of viral DNA in remote organs. Transgene expression was detected in all cases (46 of 46) between 3 and 14 days after transfection but was in no case detectable 28 days after transfection. In the DBC group, transgene expression was limited to endothelial cells when the endothelium was left intact and to rare medial cells (< 2.2%) when it had been removed. In contrast, HBC delivery resulted in transduction of up to 9.6% of medial smooth muscle cells (P = .0001). Optimized PCR and histochemistry failed to detect evidence of extra-arterial transfection except in a small number of cells (between 1 in 3 x 10(2) and 1 in 3 x 10(5) cells) in the livers of 2 animals in the DBC group. CONCLUSIONS: (1) Efficient, adenovirus-mediated, arterial gene transfer to endothelial and/or smooth muscle cells is feasible by percutaneous, clinically applicable techniques. (2) Consistent transfection of medial smooth muscle cells may be achieved when the endothelial layer is abraded. (3) Medial transfection is more efficient when an HBC, rather than a DBC, is used. (4) Percutaneous delivery of the adenoviral vector via HBC results in site-specific arterial gene transfer. Very-low-level extra-arterial transfection may occur, however, when the DBC is used.


Subject(s)
Adenoviridae/genetics , Arteries/physiology , Endothelium, Vascular/physiology , Gene Transfer Techniques , Genetic Vectors , Muscle, Smooth, Vascular/physiology , Animals , Base Sequence , Catheterization/instrumentation , Cells, Cultured , Endothelium, Vascular/cytology , Gene Expression , Histocytochemistry , Hydrogel, Polyethylene Glycol Dimethacrylate , Lac Operon , Molecular Probes/genetics , Molecular Sequence Data , Muscle, Smooth, Vascular/cytology , Polyethylene Glycols , Rabbits , Transfection
13.
Gene Ther ; 1 Suppl 1: S53-4, 1994.
Article in English | MEDLINE | ID: mdl-8542399

ABSTRACT

Due to their quiescent nature and spatial complexity, many target tissues for gene therapy will require novel strategies. An alternative to ex vivo gene transfer, providing many technical advantages and possibly allowing sufficient transfer of the therapeutic gene, is direct in vivo delivery of the vehicle. For a favorable outcome, this procedure is dependent on a high-titer vector, fully competent before post-mitotic cells. In view of the restrictions with the use of retroviruses, we investigated the potentials of adenovirus. Adenoviruses have as primary targets of infection the differentiated epithelial cell. The large DNA genome of the virus hints to a large cloning capacity. Furthermore, the wild type adenovirus has been largely used in man as a vaccine against adenovirus-induced respiratory disease. Taken together, the biological characteristics of adenovirus and the precedent of administration to humans are suggestive of adenovirus-based gene therapy for diseases involving a variety of quiescent tissues. The use of a replication-defective adenovirus carrying a gene encoding a nuclearly-targeted beta-galactosidase Ad.RSV beta gal demonstrated that replication-defective adenovirus offers an efficient means to transfer a gene for extended periods of time in the liver, muscle, lung and brain (1-6).


Subject(s)
Adenoviridae/genetics , Dystrophin/genetics , Gene Transfer Techniques , Genetic Vectors , Muscle, Skeletal/metabolism , Animals , Cell Line , Humans , Mice , Mice, Inbred mdx , Muscle, Skeletal/enzymology , beta-Galactosidase/genetics
14.
Hum Gene Ther ; 4(6): 703-11, 1993 Dec.
Article in English | MEDLINE | ID: mdl-8186285

ABSTRACT

The murine mastocytoma cell line P815 was used as a model to evaluate the effect on its tumorigenic capacity following interleukin-2 (IL-2) gene transfer into the tumor cells using a replication-defective adenovirus vector. The data show that P815 cells infected in vitro with this recombinant adenovirus secreted significant amounts of functional IL-2 as tested on CTL-L2 cells. Furthermore, when injected into syngeneic DBA/2 mice, the tumorigenic phenotype is lost in up to 80% of the animals. The rejection of the infected cells was host dependent, because co-injection at the same site or concomitant injection at the opposite side of the animal with a tumorigenic dose of noninfected P815 cells did not lead to tumor development in 50-70% of the mice. Moreover, protected animals developed a long-lasting state of immunization against the P815 tumor cells and their splenocytes were able to transfer the immunity to syngeneic naive recipients.


Subject(s)
Adenoviridae/genetics , Gene Transfer Techniques , Genetic Therapy , Interleukin-2/genetics , Neoplasms, Experimental/therapy , Animals , Cell Transplantation , Feasibility Studies , Genetic Vectors , Humans , Interleukin-2/therapeutic use , Mice , Mice, Inbred DBA , Neoplasms, Experimental/immunology , Phenotype , Recombination, Genetic , Spleen/cytology , Tumor Cells, Cultured
15.
Nat Genet ; 5(2): 130-4, 1993 Oct.
Article in English | MEDLINE | ID: mdl-8252038

ABSTRACT

Duchene muscular dystrophy (DMD) is a fatal progressive X-linked muscle disorder, caused by mutations in the dystrophin gene. We have investigated adenovirus-mediated transfer of a dystrophin minigene in a mutant mouse lacking dystrophin, the mdx mouse. We report here that six months after a single intramuscular injection of a recombinant adenovirus containing a human dystrophin minigene, a large number of dystrophin-positive fibres are still detected in the injected muscles. Moreover, although the minigene encodes a truncated protein, its expression is able to protect the fibres efficiently against the degeneration process that affects the dystrophin-deficient mdx myofibres.


Subject(s)
Adenoviridae/genetics , Dystrophin/genetics , Genetic Therapy , Muscular Dystrophies/therapy , Transfection , Animals , Genes, Viral , Humans , Mice , Mice, Transgenic , Muscular Dystrophies/genetics , Muscular Dystrophies/pathology , Time Factors , beta-Galactosidase/genetics
16.
Biochem Biophys Res Commun ; 195(3): 1174-83, 1993 Sep 30.
Article in English | MEDLINE | ID: mdl-8216246

ABSTRACT

The efficiency of gene transfer into human blood monocyte-derived macrophages has been evaluated using a replication-defective adenovirus vector harboring a lac Z gene of E. coli as a reporter gene. Whereas, no beta-galactosidase activity was found in freshly infected purified monocytes, 40% to 80% of infected macrophages which derived from these monocytes showed a beta-galactosidase activity, 2 to 4 days after infection and lasted for at least 3 weeks. Moreover, beta-galactosidase activity was found in infected monocyte/macrophages 7 days after their injection into a human tumor preestablished in nude mice. These data indicate that it is possible to transfer and stably express a gene of potential therapeutical function into human monocyte-derived macrophages using an adenovirus vector.


Subject(s)
Adenoviridae/genetics , Genetic Vectors , Macrophages/microbiology , Monocytes/microbiology , Transfection/methods , Animals , Cell Line , Histocytochemistry , Humans , Immunity, Cellular , Mice , Mice, Nude , Neoplasms, Experimental , Recombinant Fusion Proteins/biosynthesis , beta-Galactosidase/biosynthesis , beta-Galactosidase/genetics , beta-Galactosidase/isolation & purification
17.
Nucleic Acids Res ; 21(14): 3249-55, 1993 Jul 11.
Article in English | MEDLINE | ID: mdl-8341599

ABSTRACT

A hammerhead ribozyme designed to cleave in trans the R region of HIV-1 RNA was inserted into a eukaryotic expression vector. This ribozyme was studied in vitro using the T3 RNA polymerase promoter located upstream of the eukaryotic promoter. The ribozyme showed no activity against its specific target sequence under any condition tested. To decrease the influence of potential cis inhibitory sequences in such a ribozyme transcript, a specific target sequence was inserted upstream of the ribozyme-coding sequence. This insertion allowed the release by cis cleavage of a short RNA bearing ribozyme activity and able to cleave in trans an external RNA target. The cis cleavage reaction generated two RNA molecules: the shorter RNA species, which included the catalytic domain, showed a trans cleavage reaction. This self-cleavable ribozyme was active in vitro at 37 degrees C against three distinct HIV-1 transcripts sharing the specific target sequence. Ribozyme activity was thus attained by self-cleavage of the ribozyme-containing sequence from the longer vector transcript.


Subject(s)
HIV-1/enzymology , RNA, Catalytic/metabolism , RNA, Viral/metabolism , Base Sequence , Binding, Competitive , Cloning, Molecular , DNA, Viral , Enzyme Activation , Molecular Sequence Data , Nucleic Acid Conformation , Temperature
18.
J Gen Virol ; 74 ( Pt 3): 501-7, 1993 Mar.
Article in English | MEDLINE | ID: mdl-8383183

ABSTRACT

A replication-defective recombinant adenovirus (Ad) expressing the full length Epstein-Barr virus (EBV) major envelope glycoprotein gp340/220 was tested for its ability to protect against EBV-induced lymphoma in the cottontop tamarin. Antibody responses against Ad capsid proteins and EBV gp340/220 were observed but these antibodies did not neutralize EBV in vitro. However, all immunized animals were protected against challenge following three intramuscular doses of the recombinant Ad. These data indicate that the recombinant Ad is potentially a useful vector for vaccination.


Subject(s)
Antigens, Viral/immunology , Herpesvirus 4, Human/immunology , Lymphoma/prevention & control , Tumor Virus Infections/prevention & control , Viral Matrix Proteins/immunology , Adenoviridae , Animals , Antibodies, Viral/biosynthesis , Antigens, Viral/biosynthesis , Genetic Vectors , Lymphoma/microbiology , Recombinant Proteins/biosynthesis , Recombinant Proteins/immunology , Saguinus , Vaccines, Synthetic/immunology , Viral Matrix Proteins/biosynthesis , Viral Vaccines/immunology , Virus Replication
19.
Nature ; 361(6413): 647-50, 1993 Feb 18.
Article in English | MEDLINE | ID: mdl-8437625

ABSTRACT

Duchenne progressive muscular dystrophy is a lethal and common X-linked genetic disease caused by the absence of dystrophin, a 427K protein encoded by a 14 kilobase transcript. Two approaches have been proposed to correct the dystrophin deficiency in muscle. The first, myoblast transfer therapy, uses cells from normal donors, whereas the second involves direct intramuscular injection of recombinant plasmids expressing dystrophin. Adenovirus is an efficient vector for in vivo expression of various foreign genes. It has recently been demonstrated that a recombinant adenovirus expressing the lac-Z reporter gene can infect stably many mouse tissues, particularly muscle and heart. We have tested the ability of a recombinant adenovirus, containing a 6.3 kilobase pair Becker-like dystrophin complementary DNA driven by the Rous sarcoma virus promoter to direct the expression of a 'minidystrophin' in infected 293 cells and C2 myoblasts, and in the mdx mouse, after intramuscular injection. We report here that in vivo, we have obtained a sarcolemmal immunostaining in up to 50% of fibres of the injected muscle.


Subject(s)
Adenoviridae/genetics , Dystrophin/genetics , Muscles/metabolism , Muscular Dystrophies/genetics , Transfection , Animals , Base Sequence , Blotting, Western , Cell Line , DNA/genetics , Dystrophin/analysis , Fluorescent Antibody Technique , Gene Expression , Genetic Vectors , Humans , Mice , Mice, Mutant Strains , Molecular Sequence Data , Muscular Dystrophy, Animal/genetics , Polymerase Chain Reaction , Sarcolemma/metabolism
20.
J Virol ; 63(7): 3109-16, 1989 Jul.
Article in English | MEDLINE | ID: mdl-2542618

ABSTRACT

A spliced cDNA spanning the Epstein-Barr virus BZLF1 gene expresses the BZLF1 protein and is active in inducing the virus productive cycle. A deletion mutant which lacks the N-terminal half of the protein is inactive. Cotransfection experiments in EBV-negative B-lymphocyte cell lines demonstrated that the BZLF1 gene activates the promoter for the BSLF2 + BMLF1 gene in the absence of any other EBV gene product. These results confirmed that the spliced BZLF1 gene is the transactivating gene structure in BamHI-Z.


Subject(s)
Gene Expression Regulation , Genes, Viral , Genes , Herpesvirus 4, Human/genetics , Oncogene Proteins, Viral/genetics , Promoter Regions, Genetic , Virus Replication , Amino Acid Sequence , Base Sequence , Cell Line , DNA, Recombinant , Herpesvirus 4, Human/physiology , Humans , Molecular Sequence Data , Plasmids , RNA, Messenger/genetics , Restriction Mapping , Transcription, Genetic , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...