129 results on '"Zolotukhin I"'
Search Results
2. High-titer recombinant adeno-associated virus production utilizing a recombinant herpes simplex virus type I vector expressing AAV-2 Rep and Cap
- Author
-
Conway, J E, Rhys, CMJ ap, Zolotukhin, I, Zolotukhin, S, Muzyczka, N, Hayward, G S, and Byrne, B J
- Published
- 1999
- Full Text
- View/download PDF
3. Recombinant adeno-associated virus purification using novel methods improves infectious titer and yield
- Author
-
Zolotukhin, S, Byrne, B J, Mason, E, Zolotukhin, I, Potter, M, Chesnut, K, Summerford, C, Samulski, R J, and Muzyczka, N
- Published
- 1999
- Full Text
- View/download PDF
4. Incomplete elimination of viral genomes is associated with chronic inflammation in nonhuman primate livers after AAV-mediated gene transfer.
- Author
-
Pichard V, Guilbaud M, Devaux M, Jaulin N, Journou M, Cospolite M, Garcia A, Ferry N, Michalak-Provost S, Gernoux G, and Adjali O
- Abstract
The liver is a unique organ where immunity can be biased toward ineffective response notably in the context of viral infections. Chronic viral hepatitis depends on the inability of the T-cell immune response to eradicate antigen. In the case of recombinant Adeno-Associated-Virus, used for therapeutic gene transfer, conflicting reports describe tolerance induction to different transgene products while other studies have shown conventional cytotoxic CD8
+ T cell responses with a rapid loss of transgene expression. We performed a 1 year follow up of 6 non-human primates after all animals received an rAAV8 vector carrying the GFP transgene at doses of 7×1012 vg/kg. We report that despite anti-GFP peripheral cellular response and loss of hepatic transgene expression, we were still able to detect persisting viral genomes in the liver until 1-year post-injection. These viral genomes were associated with liver inflammation, fibrosis and signs of CD8 T cell exhaustion, including high expression of PD-1. Our study shows that AAV8-mediated gene transfer can results to loss of transgene expression in liver and chronic inflammation several months after gene transfer., Competing Interests: Competing interests: The author declares no competing interests., (© 2025. The Author(s).)- Published
- 2025
- Full Text
- View/download PDF
5. Site-specific modifications to AAV8 capsid yields enhanced brain transduction in the neonatal MPS IIIB mouse.
- Author
-
Gilkes JA, Judkins BL, Herrera BN, Mandel RJ, Boye SL, Boye SE, Srivastava A, and Heldermon CD
- Subjects
- Animals, Brain, Dependovirus genetics, Genetic Vectors genetics, Mice, Tissue Distribution, Transduction, Genetic, Capsid, Mucopolysaccharidosis III genetics, Mucopolysaccharidosis III therapy
- Abstract
Mucopolysaccharidosis type IIIB (MPS IIIB) is an autosomal recessive lysosomal disease caused by defective production of the enzyme α-N-acetylglucosaminidase. It is characterized by severe and complex central nervous system degeneration. Effective therapies will likely target early onset disease and overcome the blood-brain barrier. Modifications of adeno-associated viral (AAV) vector capsids that enhance transduction efficiency have been described in the retina. Herein, we describe for the first time, a transduction assessment of two intracranially administered adeno-associated virus serotype 8 variants, in which specific surface-exposed tyrosine (Y) and threonine (T) residues were substituted with phenylalanine (F) and valine (V) residues, respectively. A double-mutant (Y444 + 733F) and a triple-mutant (Y444 + 733F + T494V) AAV8 were evaluated for their efficacy for the potential treatment of MPS IIIB in a neonatal setting. We evaluated biodistribution and transduction profiles of both variants compared to the unmodified parental AAV8, and assessed whether the method of vector administration would modulate their utility. Vectors were administered through four intracranial routes: six sites (IC6), thalamic (T), intracerebroventricular, and ventral tegmental area into neonatal mice. Overall, we conclude that the IC6 method resulted in the widest biodistribution within the brain. Noteworthy, we demonstrate that GFP intensity was significantly more robust with AAV8 (double Y-F + T-V) compared to AAV8 (double Y-F). This provides proof of concept for the enhanced utility of IC6 administration of the capsid modified AAV8 (double Y-F + T-V) as a valid therapeutic approach for the treatment of MPS IIIB, with further implications for other monogenic diseases., (© 2020. The Author(s), under exclusive licence to Springer Nature Limited.)
- Published
- 2021
- Full Text
- View/download PDF
6. Superior human hepatocyte transduction with adeno-associated virus vector serotype 7.
- Author
-
Shao W, Pei X, Cui C, Askew C, Dobbins A, Chen X, Abajas YL, Gerber DA, Samulski RJ, Nichols TC, and Li C
- Subjects
- Animals, Antibodies, Neutralizing metabolism, Case-Control Studies, Cell Line, Dependovirus immunology, Female, Green Fluorescent Proteins metabolism, HEK293 Cells, Hepatocytes cytology, Humans, Mice, Serogroup, Transduction, Genetic, Dependovirus physiology, Genetic Vectors administration & dosage, Green Fluorescent Proteins genetics, Hemophilia A immunology, Hepatocytes virology
- Abstract
Although therapeutic outcomes have been achieved in hemophilia patients after delivery of clotting factor genes to the liver using adeno-associated virus (AAV) vectors, it is well known that the preclinical results generated from hemophilia animal models have not been directly predictive of successful translation in humans. To address this discrepancy humanized mouse models have recently been used to predict AAV transduction efficiency for human hepatocytes. In this study we evaluated AAV vector transduction from several serotypes in human liver hepatocytes xenografted into chimeric mice. After systemic administration of AAV vectors encoding a GFP transgene in humanized mice, the liver was harvested for either immunohistochemistry staining or flow cytometry assay for AAV human hepatocyte transduction analysis. We observed that AAV7 consistently transduced human hepatocytes more efficiently than other serotypes in both immunohistochemistry assay and flow cytometry analysis. To better assess the future application of AAV7 for systemic administration in the treatment of hemophilia or other liver diseases, we analyzed the prevalence of neutralizing antibodies (NAbs) to AAV7 in sera from healthy subjects and patients with hemophilia. In the general population, the prevalence of NAbs to AAV7 was lower than that of AAV2 or AAV3B. However, a higher prevalence of AAV7 NAbs was found in patients with hemophilia. In summary, results from this study suggest that AAV7 vectors should be considered as an effective vehicle for human liver targeting in future clinical trials.
- Published
- 2019
- Full Text
- View/download PDF
7. Successes and challenges in clinical gene therapy.
- Author
-
Kohn DB, Chen YY, and Spencer MJ
- Subjects
- Humans, T-Lymphocytes, Hematopoietic Stem Cells, Genetic Vectors genetics, Dependovirus genetics, Gene Editing, Genetic Therapy, Neoplasms
- Abstract
Despite the ups and downs in the field over three decades, the science of gene therapy has continued to advance and provide enduring treatments for increasing number of diseases. There are active clinical trials approaching a variety of inherited and acquired disorders of different organ systems. Approaches include ex vivo modification of hematologic stem cells (HSC), T lymphocytes and other immune cells, as well as in vivo delivery of genes or gene editing reagents to the relevant target cells by either local or systemic administration. In this article, we highlight success and ongoing challenges in three areas of high activity in gene therapy: inherited blood cell diseases by targeting hematopoietic stem cells, malignant disorders using immune effector cells genetically modified with chimeric antigen receptors, and ophthalmologic, neurologic, and coagulation disorders using in vivo administration of adeno-associated virus (AAV) vectors. In recent years, there have been true cures for many of these diseases, with sustained clinical benefit that exceed those from other medical approaches. Each of these treatments faces ongoing challenges, namely their high one-time costs and the complexity of manufacturing the therapeutic agents, which are biological viruses and cell products, at pharmacologic standards of quality and consistency. New models of reimbursement are needed to make these innovative treatments widely available to patients in need., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
8. First use of gene therapy to treat growth hormone resistant dwarfism in a mouse model.
- Author
-
Sia KC, Gan SU, Mohd Rodhi SH, Fu ZY, Kopchick JJ, Waters MJ, and Lee KO
- Subjects
- Animals, Disease Models, Animal, Genetic Therapy, Humans, Insulin-Like Growth Factor I genetics, Insulin-Like Growth Factor I metabolism, Insulin-Like Growth Factor I therapeutic use, Mice, Receptors, Somatotropin genetics, Receptors, Somatotropin metabolism, Receptors, Somatotropin therapeutic use, Growth Hormone genetics, Growth Hormone therapeutic use, Laron Syndrome drug therapy, Laron Syndrome therapy
- Abstract
The only treatment tested for growth hormone receptor (GHR) defective Laron Syndrome (LS) is injections of recombinant insulin-like-growth factor 1 (rhIGF1). The response is suboptimal and associated with progressive obesity. In this study, we treated 4-5-week-old Laron dwarf mice (GHR-/-) with an adeno-associated virus expressing murine GHR (AAV-GHR) injection at a dose of 4 × 10
10 vector genome per mouse. Serum growth hormone (GH) levels decreased, and GH-responsive IGF1, IGF binding protein 3 (IGFBP3) and acid labile subunit (ALS) increased. There was a significant but limited increase in body weight and length, similar to the response to rhIGF1 treatment in LS patients. All the major organs increased in weight except the brain. Our study is the first to use gene therapy to treat GH-receptor deficiency. We propose that gene therapy with AAV-GHR may eventually be useful for the treatment of human LS., (© 2022. The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
9. XIAP gene therapy effects on retinal ganglion cell structure and function in a mouse model of glaucoma.
- Author
-
Visuvanathan S, Baker AN, Lagali PS, Coupland SG, Miller G, Hauswirth WW, and Tsilfidis C
- Subjects
- Animals, Axons, Disease Models, Animal, Genetic Therapy, Intraocular Pressure, Mice, Retinal Ganglion Cells metabolism, Glaucoma genetics, Glaucoma therapy, Neurodegenerative Diseases
- Abstract
Glaucoma is a prevalent neurodegenerative disease that is characterized by progressive visual field loss. It is the leading cause of irreversible blindness in the world. The main risk factor for glaucoma is elevated intraocular pressure that results in the damage and death of retinal ganglion cells (RGCs) and their axons. The death of RGCs has been shown to be apoptotic. We tested the hypothesis that blocking the activation of apoptosis may be an effective strategy to prevent RGC death and preserve functional vision in glaucoma. In the magnetic microbead mouse model of induced ocular hypertension, inhibition of RGC apoptosis was targeted through viral-mediated ocular delivery of the X-linked inhibitor of apoptosis (XIAP) gene, a potent caspase inhibitor. Pattern electroretinograms revealed that XIAP therapy resulted in significant protection of both somal and axonal RGC function in glaucomatous eyes. Histology confirmed that the treated optic nerves showed preservation of axon counts and reduced glial cell infiltration. These results show that XIAP is able to provide both functional and structural protection of RGCs in the microbead model of glaucoma and provide important proof-of-principle for XIAP's efficacy as a neuroprotective treatment for glaucoma., (© 2021. The Author(s), under exclusive licence to Springer Nature Limited.)
- Published
- 2022
- Full Text
- View/download PDF
10. Antiviral immunity and nucleic acid sensing in haematopoietic stem cell gene engineering.
- Author
-
Piras F and Kajaste-Rudnitski A
- Subjects
- Genetic Therapy, Genetic Vectors, Hematopoietic Stem Cells, Humans, Nucleic Acids genetics
- Abstract
The low gene manipulation efficiency of human hematopoietic stem and progenitor cells (HSPC) remains a major hurdle for sustainable and broad clinical application of innovative therapies for a wide range of disorders. Given that all current and emerging gene transfer and editing technologies are bound to expose HSPC to exogenous nucleic acids and most often also to viral vectors, we reason that host antiviral factors and nucleic acid sensors play a pivotal role in the efficacy of HSPC genetic manipulation. Here, we review recent progress in our understanding of vector-host interactions and innate immunity in HSPC upon gene engineering and discuss how dissecting this crosstalk can guide the development of more stealth and efficient gene therapy approaches in the future.
- Published
- 2021
- Full Text
- View/download PDF
11. Transduction optimization of AAV vectors for human gene therapy of glaucoma and their reversed cell entry characteristics.
- Author
-
Rodriguez-Estevez L, Asokan P, and Borrás T
- Subjects
- Aged, Cells, Cultured, Female, Humans, Male, Middle Aged, Trabecular Meshwork cytology, Trabecular Meshwork metabolism, Transduction, Genetic standards, Dependovirus genetics, Genetic Therapy methods, Genetic Vectors genetics, Glaucoma therapy, Transduction, Genetic methods
- Abstract
The trabecular meshwork (TM) of the eye is responsible for maintaining physiological intraocular pressure (IOP). Dysfunction of this tissue results in elevated IOP, subsequent optic nerve damage and glaucoma, the world's leading cause of irreversible blindness. IOP regulation by delivering candidate TM genes would offer an enormous clinical advantage to the current daily-drops/surgery treatment. Initially, we showed that a double-stranded AAV2 (scAAV2) transduced the human TM very efficiently, while its single-stranded form (ssAAV2) did not. Here, we quantified transduction and entry of single- and double-strand serotypes 1, 2.5, 5, 6, 8, and 9 in primary, single individual-derived human TM cells (HTM). scAAV2 exhibited highest transduction in all individuals, distantly followed by scAAV2.5, scAAV6, and scAAV5. Transduction of scAAV1, scAAV8, and scAAV9 was negligible. None of the ssAAV serotypes transduced, but their cell entries were significantly higher than those of their corresponding scAAV. Tyrosine scAAV2 capsid mutants increased transduction in HTM cultured cells and all TM-outflow layers of perfused postmortem human eyes. These studies provide the first serotype optimization for gene therapy of glaucoma in humans. They further reveal biological differences between the AAV forms in HTM cells, whose understanding could contribute to the development of gene therapy of glaucoma.
- Published
- 2020
- Full Text
- View/download PDF
12. Treatment of multifocal breast cancer by systemic delivery of dual-targeted adeno-associated viral vectors.
- Author
-
Trepel, M, Körbelin, J, Spies, E, Heckmann, M B, Hunger, A, Fehse, B, Katus, H A, Kleinschmidt, J A, Müller, O J, and Michelfelder, S
- Subjects
BREAST cancer patients ,BREAST cancer treatment ,DRUG delivery systems ,ADENOVIRUSES ,MICRORNA ,GENETIC transformation ,THERAPEUTICS - Abstract
Adeno-associated viral (AAV) vectors yield high potential for clinical gene therapy but, like for other vectors systems, they frequently do not sufficiently transduce the target tissue and their unspecific tropism prevents their application for multifocal diseases such as disseminated cancer. Targeted AAV vectors have been obtained from random AAV display peptide libraries but so far, all vector variants selected from AAV libraries upon systemic administration in vivo retained some collateral tropism, frequently the heart. Here we explored, if this impediment can be overcome by microRNA-regulated transgene cassettes as the combination of library-derived capsid targeting and micro-RNA control has not been evaluated so far. We used a tumor-targeted AAV capsid variant (ESGLSQS) selected from random AAV-display peptide libraries in vivo with remaining off-target tropism toward the heart and regulated targeted transgene expression in vivo by complementary target elements for heart-specific microRNA (miRT-1d). Although this vector still maintained its strong transduction capacity for tumor target tissue after intravenous injection, transgene expression in the heart was almost completely abrogated. This strong and completely tumor-specific transgene expression was used for therapeutic gene transfer in an aggressive multifocal, transgenic, polyoma middle T-induced, murine breast cancer model. A therapeutic suicide gene, delivered systemically by this dual-targeted AAV vector to multifocal breast cancer, significantly inhibited tumor growth after one single vector administration while avoiding side effects compared with untargeted vectors. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
13. The MRI contrast agent gadoteridol enhances distribution of rAAV1 in the rat hippocampus.
- Author
-
Hullinger, R, Ugalde, J, Purón-Sierra, L, Osting, S, and Burger, C
- Subjects
CONTRAST media ,MAGNETIC resonance imaging ,ADENO-associated virus ,HIPPOCAMPUS (Brain) ,GADOLINIUM ,GENETIC vectors ,GENETIC transduction - Abstract
Contrast agents are commonly used in combination with magnetic resonance imaging (MRI) to monitor the distribution of molecules in the brain. Recent experiments conducted in our laboratory have shown that co-infusion of recombinant Adeno-associated virus serotype 5 (rAAV5) and the MRI contrast agent gadoteridol (Gd) enhances vector transduction in the rat striatum. The goal of this study was to determine whether gadoteridol may also be used as a tool to enhance transduction efficiency of rAAV1 and rAAV5 within the rat hippocampus. We show that Gd/rAAV1-GFP but not Gd/rAAV5-GFP co-infusion results in significantly higher distribution of the transgene both in the injected hemisphere as well as in the contralateral side and adjacent areas of cortex along the injection track. We also show that Gd/rAAV1-GFP co-infusion has no deleterious effect on hippocampal function as assessed by two tests of spatial memory formation. This work indicates that Gd can be exploited as a method to increase transduction efficiency of AAV1 in the hippocampus for animal studies. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
14. Modulation of feeding by chronic rAAV expression of a relaxin-3 peptide agonist in rat hypothalamus.
- Author
-
Ganella, D E, Callander, G E, Ma, S, Bye, C R, Gundlach, A L, and Bathgate, R A D
- Subjects
ADENO-associated virus ,GENE expression in viruses ,RELAXIN ,PEPTIDES ,LABORATORY rats ,HYPOTHALAMUS ,G protein coupled receptors ,NEUROPEPTIDES ,BRAIN stem - Abstract
Relaxin-3 is a neuropeptide that is abundantly expressed by discrete brainstem neuron populations that broadly innervate forebrain areas rich in the relaxin-3 G-protein-coupled-receptor, RXFP3. Acute and subchronic central administration of synthetic relaxin-3 or an RXFP3-selective agonist peptide, R3/I5, increase feeding and body weight in rats. Intrahypothalamic injection of relaxin-3 also increases feeding. In this study, we developed a recombinant adeno-associated virus 1/2 (rAAV1/2) vector that drives expression and constitutive secretion of bioactive R3/I5 and assessed the effect of intrahypothalamic injections on daily food intake and body weight gain in adult male rats over 8 weeks. In vitro testing revealed that the vector rAAV1/2-fibronectin (FIB)-R3/I5 directs the constitutive secretion of bioactive R3/I5 peptide. Bilateral injection of rAAV1/2-FIB-R3/I5 vector into the paraventricular nucleus produced an increase in daily food intake and body weight gain (P<0.01, ∼23%, respectively), relative to control treatment. In a separate cohort of rats, quantitative polymerase chain reaction analysis of hypothalamic mRNA revealed strong expression of R3/I5 transgene at 3 months post-rAAV1/2-FIB-R3/I5 infusion. Levels of mRNA transcripts for the relaxin-3 receptor RXFP3, the hypothalamic 'feeding' peptides neuropeptide Y, AgRP and POMC, and the reproductive hormone, GnRH, were all similar to control, whereas vasopressin and oxytocin (OT) mRNA levels were reduced by ∼25% (P=0.051) and ∼50% (P<0.005), respectively, in rAAV1/2-FIB-R3/I5-treated rats (at 12 weeks, n=9/8 rats per group). These data demonstrate for the first time that R3/I5 is effective in modulating feeding in the rat by chronic hypothalamic RXFP3 activation and suggest a potential underlying mechanism involving altered OT signalling. Importantly, there was no desensitization of the feeding response over the treatment period and no apparent deleterious health effects, indicating that targeting the relaxin-3-RXFP3 system may be an effective long-term therapy for eating disorders. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
15. Hairpin-end conformation of adeno-associated virus genome determines interactions with DNA-repair pathways.
- Author
-
Cataldi, M P and McCarty, D M
- Subjects
VIRAL genomes ,CONFORMATIONAL analysis ,ADENO-associated virus ,DNA repair ,REPEATED sequence (Genetics) ,DNA replication ,VIRION - Abstract
The palindromic terminal repeats (TRs) of adeno-associated virus (AAV) form DNA hairpins (HPs) are essential for replication and for priming the conversion of single-stranded virion DNA to double strand. In recombinant AAV (rAAV) gene-delivery vectors, they are targets for the DNA-repair pathways leading to circularization, concatemerization and, infrequently, chromosomal integration. We investigated the effect of the TR HP on recombination by comparing specific DNA substrates transfected into wild-type and DNA-repair-deficient cells. DNA molecules with the TR sequences constrained in the T-shaped HP conformation at one or both ends were subject to a loss of gene expression, which was partially relieved in ataxia telangiectasia mutated (ATM
−/− ) cells. The ATM-dependent effect was mediated by transcriptional silencing of a subset of HP-containing molecules in cis rather than a loss of DNA, and was dependent on the specific T-shaped structure of the HP and not the primary sequence. DNA molecules with simple U-shaped HP ends were unaffected by ATM-dependent silencing. The silenced molecules remained in a linear conformation, in contrast to the expressed molecules, which were circularized. In the absence of ATM activity, this subset remained linear but was actively expressed. DNA molecules with the TR sequence in the open duplex conformation, or without TR sequences, were unaffected by ATM mutation and were predominantly converted to circular forms. A separate HP-specific effect in normal cells resulted in a loss of DNA substrate in the nucleus and was ATM independent. These results suggest that the presence of the HP structure on rAAV vector genomes subjects them to specific, and sometimes unproductive, DNA-repair/recombination pathways. [ABSTRACT FROM AUTHOR]- Published
- 2013
- Full Text
- View/download PDF
16. scAAV-mediated gene transfer of interleukin-1-receptor antagonist to synovium and articular cartilage in large mammalian joints.
- Author
-
Watson, R S, Broome, T A, Levings, P P, Rice, B L, Kay, J D, Smith, A D, Gouze, E, Gouze, J-N, Dacanay, E A, Hauswirth, W W, Nickerson, D M, Dark, M J, Colahan, P T, and Ghivizzani, S C
- Subjects
GENETIC transformation ,INTERLEUKIN-1 receptors ,CHEMICAL inhibitors ,GREEN fluorescent protein ,ADENO-associated virus ,ARTICULAR cartilage - Abstract
With the long-term goal of developing a gene-based treatment for osteoarthritis (OA), we performed studies to evaluate the equine joint as a model for adeno-associated virus (AAV)-mediated gene transfer to large, weight-bearing human joints. A self-complementary AAV2 vector containing the coding regions for human interleukin-1-receptor antagonist (hIL-1Ra) or green fluorescent protein was packaged in AAV capsid serotypes 1, 2, 5, 8 and 9. Following infection of human and equine synovial fibroblasts in culture, we found that both were only receptive to transduction with AAV1, 2 and 5. For these serotypes, however, transgene expression from the equine cells was consistently at least 10-fold higher. Analyses of AAV surface receptor molecules and intracellular trafficking of vector genomes implicate enhanced viral uptake by the equine cells. Following delivery of 1 × 10
11 vector genomes of serotypes 2, 5 and 8 into the forelimb joints of the horse, all three enabled hIL-1Ra expression at biologically relevant levels and effectively transduced the same cell types, primarily synovial fibroblasts and, to a lesser degree, chondrocytes in articular cartilage. These results provide optimism that AAV vectors can be effectively adapted for gene delivery to large human joints affected by OA. [ABSTRACT FROM AUTHOR]- Published
- 2013
- Full Text
- View/download PDF
17. Endocytic processing of adeno-associated virus type 8 vectors for transduction of target cells.
- Author
-
Liu, Y, Joo, K-I, and Wang, P
- Subjects
ENDOCYTOSIS ,ADENO-associated virus ,GENETIC transduction ,GENE targeting ,SEROTYPES ,CLATHRIN ,CYTOSKELETON ,VIRUSES - Abstract
We investigated the transduction of HEK293T cells permissive to adeno-associated virus serotype 8 (AAV8) to understand the mechanisms underlying its endocytic processing. Results showed that AAV8 enters cells through clathrin-mediated endocytosis followed by trafficking through various endosomal compartments. Interestingly, compared to the relatively well-characterized AAV2, a distinct involvement of late endosomes was observed for AAV8 trafficking within the target cell. AAV8 particles were also shown to exploit the cytoskeleton network to facilitate their transport within cells. Moreover, the cellular factors involved during endosomal escape were examined by an in vitro membrane permeabilization assay. Our data demonstrated that an acidic endosomal environment was required for AAV2 penetration through endosomal membranes and that the cellular endoprotease furin could promote AAV2 escape from the early endosomes. In contrast, these factors were not sufficient for AAV8 penetration through endosomal membranes. We further found that the ubiquitin-proteasome system is likely involved in the intracellular transport of AAV8 to nucleus. Taken together, our data have shed some light on the intracellular trafficking pathways of AAV8, which, in turn, could provide insight for potentializing AAV-mediated gene delivery. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
18. Retina transduction by rAAV2 after intravitreal injection: comparison between mouse and rat.
- Author
-
Dias MS, Araujo VG, Vasconcelos T, Li Q, Hauswirth WW, Linden R, and Petrs-Silva H
- Subjects
- Animals, Electroretinography, Genetic Therapy, Genistein administration & dosage, Imatinib Mesylate administration & dosage, Intravitreal Injections, Mice, Mutation, Rats, Transduction, Genetic, Adjuvants, Pharmaceutic administration & dosage, Dependovirus genetics, Genetic Vectors administration & dosage, Protein Kinase Inhibitors administration & dosage, Retina virology
- Abstract
Adeno-associated virus vectors (rAAV) are currently the most common vehicle used in clinical trials of retinal gene therapy, usually delivered through subretinal injections to target cells of the outer retina. However, targeting the inner retina requires intravitreal injections, a simple and safe procedure, which is effective for transducing the rodent retina, but still of low efficiency in the eyes of primates. We investigated whether adjuvant pharmacological agents may enhance rAAV transduction of the retinas of mouse and rat after intravitreal delivery. Tyrosine kinase inhibitors were highly efficient in mice, especially imatinib and genistein, and promoted transduction even of the outer retina. In rats, however, we report that they were not effective. Even with direct proteasomal inhibition in rats, the effects upon transduction were only minimal and restricted to the inner retina. Even tyrosine capsid mutant rAAVs in rats had a transduction profile similar to wtAAV. Thus, the differences between mouse and rat, in both eye size and the inner limiting membrane, compromise the efficiency of AAV vectors penetration from the vitreous into the retina, and impact the efficacy of strategies developed to enhance intravitreal retinal rAAV transduction. Further improvement of strategies, then are required.
- Published
- 2019
- Full Text
- View/download PDF
19. AAV-based neonatal gene therapy for hemophilia A: long-term correction and avoidance of immune responses in mice.
- Author
-
Hu, C and Lipshutz, G S
- Subjects
GENE therapy ,HEMOPHILIA in children ,IMMUNE response ,ANTIGENS ,IMMUNOGLOBULINS ,GENE expression ,LABORATORY mice ,THERAPEUTICS - Abstract
Hemophilia A gene therapy has been hampered by immune responses to vector-associated antigens and by neutralizing antibodies or inhibitors against the factor VIII (FVIII) protein; these 'inhibitors' more commonly affect hemophilia A patients than those with hemophilia B. A gene replacement strategy beginning in the neonatal period may avoid the development of these immune responses and lead to prolonged expression with correction of phenotype, thereby avoiding long-term consequences. A serotype rh10 adeno-associated virus (AAV) was developed splitting the FVIII coding sequence into heavy and light chains with the chicken β-actin promoter/CMV enhancer for dual recombinant adeno-associated viral vector delivery. Virions of each FVIII chain were co-injected intravenously into mice on the second day of life. Mice express sustained levels of FVIII antigen 5% up to 22 months of life without development of antibodies against FVIII. Phenotypic correction was manifest in all AAV-FVIII-treated mice as demonstrated by functional assay and reduction in bleeding time. This study demonstrates the use of AAV in a gene replacement strategy in neonatal mice that establishes both long-term phenotypic correction of hemophilia A and lack of antibody development against FVIII in this disease model where AAV is administered shortly after birth. These studies support the consideration of gene replacement therapy for diseases that are diagnosed in utero or in the early neonatal period. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
20. Intracellular transport of recombinant adeno-associated virus vectors.
- Author
-
Nonnenmacher, M and Weber, T
- Subjects
ADENO-associated virus ,GENE therapy ,CLINICAL trials ,PARKINSON'S disease ,HEART failure ,ENDOCYTOSIS ,RECOMBINANT viruses - Abstract
Recombinant adeno-associated viral vectors (rAAVs) have been widely used for gene delivery in animal models, and are currently evaluated for human gene therapy after successful clinical trials in the treatment of inherited, degenerative or acquired diseases, such as Leber congenital amaurosis, Parkinson disease or heart failure. However, limitations in vector tropism, such as limited tissue specificity and insufficient transduction efficiencies of particular tissues and cell types, still preclude therapeutic applications in certain tissues. Wild-type adeno-associated viruses (AAVs) are defective viruses that require the presence of a helper virus to complete their life cycle. On the one hand, this unique property makes AAV vectors one of the safest available viral vectors for gene delivery. On the other, it also represents a potential obstacle because rAAV vectors have to overcome several biological barriers in the absence of a helper virus to transduce successfully a cell. Consequently, a better understanding of the cellular roadblocks that limit rAAV gene delivery is crucial and, during the last 15 years, numerous studies resulted in an expanding body of knowledge of the intracellular trafficking pathways of rAAV vectors. This review describes our current understanding of the mechanisms involved in rAAV attachment to target cells, endocytosis, intracellular trafficking, capsid processing, nuclear import and genome release with an emphasis on the most recent discoveries in the field and the emerging strategies used to improve the efficiency of AAV-derived vectors. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
21. Development of optimized AAV3 serotype vectors: mechanism of high-efficiency transduction of human liver cancer cells.
- Author
-
Cheng, B, Ling, C, Dai, Y, Lu, Y, Glushakova, L G, Gee, S W Y, McGoogan, K E, Aslanidi, G V, Park, M, Stacpoole, P W, Siemann, D, Liu, C, and Srivastava, A
- Subjects
GENETIC vectors ,GENETIC transduction ,LIVER cancer ,CANCER cells ,MOLECULAR genetics ,ADENOVIRUSES ,SEROTYPES ,HEPATOCYTE growth factor ,TRANSGENE expression ,GENE therapy - Abstract
Our recent studies have revealed that among the 10 different commonly used adeno-associated virus (AAV) serotypes, AAV3 vectors transduce human liver cancer cells extremely efficiently because these cells express high levels of human hepatocyte growth factor receptor (hHGFR), and AAV3 utilizes hHGFR as a cellular co-receptor for viral entry. In this report, we provide further evidence that both extracellular as well as intracellular kinase domains of hHGFR are involved in AAV3 vector entry and AAV3-mediated transgene expression. We also document that AAV3 vectors are targeted for degradation by the host cell proteasome machinery, and that site-directed mutagenesis of surface-exposed tyrosine (Y) to phenylalanine (F) residues on AAV3 capsids significantly improves the transduction efficiency of Y701F, Y705F and Y731F mutant AAV3 vectors. The transduction efficiency of the Y705+731F double-mutant vector is significantly higher than each of the single mutants in liver cancer cells in vitro. In immunodeficient mouse xenograft models, direct intratumoral injection of AAV3 vectors also led to high-efficiency transduction of human liver tumor cells in vivo. We also document here that the optimized tyrosine-mutant AAV3 vectors lead to increased transduction efficiency following both intratumoral and tail-vein injections in vivo. The optimized tyrosine-mutant AAV3 serotype vectors containing proapoptotic genes should prove useful for the potential gene therapy of human liver cancers. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
22. Elastin-like polypeptide matrices for enhancing adeno-associated virus-mediated gene delivery to human neural stem cells.
- Author
-
Kim, J-S, Chu, H S, Park, K I, Won, J-I, and Jang, J-H
- Subjects
ELASTIN ,POLYPEPTIDES ,ADENO-associated virus ,GENE therapy ,NEURAL stem cells ,GENETIC vectors ,GENE targeting ,FIBROBLASTS ,TISSUE culture ,REGENERATIVE medicine - Abstract
The successful development of efficient and safe gene delivery vectors continues to be a major obstacle to gene delivery in stem cells. In this study, we have developed an elastin-like polypeptide (ELP)-mediated adeno-associated virus (AAV) delivery system for transducing fibroblasts and human neural stem cells (hNSCs). AAVs have significant promise as therapeutic vectors because of their safety and potential for use in gene targeting in stem cell research. ELP has been recently employed as a biologically inspired 'smart' biomaterial that exhibits an inverse temperature phase transition, thereby demonstrating promise as a novel drug carrier. The ELP that was investigated in this study was composed of a repetitive penta-peptide with [Val-Pro-Gly-Val-Gly]. A novel AAV variant, AAV r3.45, which was previously engineered by directed evolution to enhance transduction in rat NSCs, was nonspecifically immobilized onto ELPs that were adsorbed beforehand on a tissue culture polystyrene surface (TCPS). The presence of different ELP quantities on the TCPS led to variations in surface morphology, roughness and wettability, which were ultimately key factors in the modulation of cellular transduction. Importantly, with substantially reduced viral quantities compared with bolus delivery, ELP-mediated AAV delivery significantly enhanced delivery efficiency in fibroblasts and hNSCs, which have great potential for use in tissue engineering applications and neurodegenerative disorder treatments, respectively. The enhancement of cellular transduction in stem cells, as well as the feasibility of ELPs for utilization in three-dimensional scaffolds, will contribute to the advancement of gene therapy for stem cell research and tissue regenerative medicine. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
23. Peptide affinity reagents for AAV capsid recognition and purification.
- Author
-
Pulicherla, N and Asokan, A
- Subjects
PEPTIDES ,MIRIDAE ,SEROTYPES ,ION exchange chromatography ,BIOLOGICAL reagents ,VIRAL replication ,CHEMICAL affinity - Abstract
We report the discovery of AAV capsid-binding peptides identified through phage panning. The heptapeptide motif GYVSRHP selectively recognized AAV serotype 8 capsids and blocked transduction in vitro. Recombinant AAV8 vectors were purified directly from crude cell lysate and supernatant through sequential application of peptide affinity and anion exchange chromatography. Peptide affinity reagents may serve as useful alternatives to monoclonal antibodies in AAV capsid recognition, and offer readily scalable solutions for purification of clinical grade AAV vectors. [ABSTRACT FROM AUTHOR]
- Published
- 2011
- Full Text
- View/download PDF
24. Improvement of the mdx mouse dystrophic phenotype by systemic in utero AAV8 delivery of a minidystrophin gene.
- Author
-
Koppanati, B. M., Li, J., Reay, D. P., Wang, B., Daood, M., Zheng, H., Xiao, X., Watchko, J. F., and Clemens, P. R.
- Subjects
DUCHENNE muscular dystrophy ,MUSCLE diseases ,STRIATED muscle ,HUMAN chromosome abnormality diagnosis ,CYTOMEGALOVIRUSES ,INTRAPERITONEAL injections - Abstract
Duchenne muscular dystrophy (DMD) is a devastating primary muscle disease with pathological changes in skeletal muscle that are ongoing at the time of birth. Progressive deterioration in striated muscle function in affected individuals ultimately results in early death due to cardio-pulmonary failure. As affected individuals can be identified before birth by prenatal genetic testing for DMD, gene replacement treatment can be started in utero. This approach offers the possibility of preventing pathological changes in muscle that begin early in life. To test in utero gene transfer in the mdx mouse model of DMD, a minidystrophin gene driven by the human cytomegalovirus promoter was delivered systemically by an intraperitoneal injection to the fetus at embryonic day 16. Treated mdx mice studied at 9 weeks after birth showed widespread expression of recombinant dystrophin in skeletal muscle, restoration of the dystrophin-associated glycoprotein complex in dystrophin-expressing muscle fibers, improved muscle pathology, and functional benefit to the transduced diaphragm compared with untreated littermate controls. These results support the potential of the AAV8 vector to efficiently cross the blood vessel barrier to achieve systemic gene transfer to skeletal muscle in utero in a mouse model of muscular dystrophy, to significantly improve the dystrophic phenotype and to ameliorate the processes that lead to exhaustion of the skeletal muscle regenerative capacity. [ABSTRACT FROM AUTHOR]
- Published
- 2010
- Full Text
- View/download PDF
25. rAAV2/5 gene-targeting to rods:dose-dependent efficiency and complications associated with different promoters.
- Author
-
Beltran, W. A., Boye, S. L., Boye, S. E., Chiodo, V. A., Lewin, A. S., Hauswirth, W. W., and Aguirre, G. D.
- Subjects
GENE therapy ,CYTOMEGALOVIRUS retinitis ,RETINAL ganglion cells ,GENETIC transduction ,OPSINS - Abstract
A prerequisite for using corrective gene therapy to treat humans with inherited retinal degenerative diseases that primarily affect rods is to develop viral vectors that target specifically this population of photoreceptors. The delivery of a viral vector with photoreceptor tropism coupled with a rod-specific promoter is likely to be the safest and most efficient approach to target expression of the therapeutic gene to rods. Three promoters that included a fragment of the proximal mouse opsin promoter (mOP), the human G-protein-coupled receptor protein kinase 1 promoter (hGRK1), or the cytomegalovirus immediate early enhancer combined with the chicken β actin proximal promoter CBA were evaluated for their specificity and robustness in driving GFP reporter gene expression in rods, when packaged in a recombinant adeno-associated viral vector of serotype 2/5 (AAV2/5), and delivered via subretinal injection to the normal canine retina. Photoreceptor-specific promoters (mOP, hGRK1) targeted robust GFP expression to rods, whereas the ubiquitously expressed CBA promoter led to transgene expression in the retinal pigment epithelium, rods, cones and rare Müller, horizontal and ganglion cells. Late onset inflammation was frequently observed both clinically and histologically with all three constructs when the highest viral titers were injected. Cone loss in the injected regions of the retinas that received the highest titers occurred with both the hGRK1 and CBA promoters. Efficient and specific rod transduction, together with preservation of retinal structure was achieved with both mOP and hGRK1 promoters when viral titers in the order of 10
11 vg ml–1 were used. [ABSTRACT FROM AUTHOR]- Published
- 2010
- Full Text
- View/download PDF
26. Near-perfect infectivity of wild-type AAV as benchmark for infectivity of recombinant AAV vectors.
- Author
-
Zeltner, N., Kohlbrenner, E., Clément, N., Weber, T., and Linden, R. M.
- Subjects
IMMUNOLOGY ,GENE therapy ,GENETICS ,GENETIC transformation ,CLINICAL medicine - Abstract
Viral vectors derived from adeno-associated viruses (AAVs) are widely used for gene transfer both in vitro and in vivo. The increasing use of AAV as a gene transfer vector, as well as recently shown immunological complications in clinical trials, highlight the necessity to define the specific activity of vector preparations beyond current standards. In this report, we determined the infectious, physical and genome-containing particle titers of several wild-type AAV type 2 (wtAAV2) and recombinant AAV type 2 (rAAV2) preparations that were produced and purified by standard methods. We found that the infectivity of wtAAV2 approaches a physical-to-infectious particle ratio of one. This near-perfect physical-to-infectious particle ratio defines a ‘ceiling’ for the theoretically achievable quality of recombinant AAV vectors. In comparison, for rAAV2, only approximately 50 out of 100 viral particles contained a genome and, more strikingly, only approximately 1 of the 100 viral particles was infectious. Our findings suggest that current strategies for rAAV vector design, production and/or purification should be amenable to improvements. Ultimately, this could result in the generation of near-perfect vector particles, a prospect with significant implications for gene therapy. [ABSTRACT FROM AUTHOR]
- Published
- 2010
- Full Text
- View/download PDF
27. IL-10 delivery by AAV5 vector attenuates inflammation in mice with pseudomonas pneumonia.
- Author
-
Buff, S. M., Yu, H., McCall, J. N., Caldwell, S. M., Ferkol, T. W., Flotte, T. R., and Virella-Lowell, I. L.
- Subjects
PSEUDOMONAS aeruginosa ,LUNG diseases ,CYSTIC fibrosis ,PATHOGENIC microorganisms ,CYTOKINES - Abstract
Lung infections with Pseudomonas aeruginosa and other pathogens in cystic fibrosis (CF) cause progressive airway obstruction and tissue damage, the predominant cause of morbidity and mortality in CF. We investigated whether a recombinant adeno-associated virus type 5 (AAV5) vector expressing murine interleukin (IL)-10 (AAV5.Cβ-mIL-10), a regulatory/anti-inflammatory cytokine, could decrease airway inflammation in IL-10 knockout mice chronically infected with mucoid P. aeruginosa. Mice that received AAV5.Cβ-mIL10 through intratracheal inoculation produced IL-10 at an average of 25 000 pg/ml in the epithelial lining fluid (ELF) and 12 000 pg/g-lung tissue 6 weeks post-vector delivery, significantly higher levels than in placebo-treated mice. At 3 days post-infection, proinflammatory cytokines (IL-1β, tumor necrosis factor (TNF)-α, macrophage inhibitory protein (MIP)-1α and (KC) in the ELF and lung homogenate were decreased (1–9 folds) in the AAV5.Cβ-mIL10-treated mice accompanied by less pronounced and more localized neutrophil infiltration in lung sections, when compared with placebo-treated mice. These results suggest that AAV5.Cβ-mIL10 induces IL-10 levels in the lungs mediating a significant anti-inflammatory response and making AAV-IL-10 gene transfer a potentially useful therapy in the treatment of CF lung disease. [ABSTRACT FROM AUTHOR]
- Published
- 2010
- Full Text
- View/download PDF
28. Lack of humoral immune response to the tetracycline (Tet) activator in rats injected intracranially with Tet-off rAAV vectors.
- Author
-
Han, Y., Chang, Q. A., Virag, T., West, N. C., George, D., Castro, M. G., and Bohn, M. C.
- Subjects
TRANSGENE expression ,GENE therapy ,IMMUNE response ,PARKINSON'S disease ,INTRAMUSCULAR injections - Abstract
The ability to safely control transgene expression from viral vectors is a long-term goal in the gene therapy field. We have previously reported tight regulation of GFP expression in rat brain using a self-regulating tet-off rAAV vector. The immune responses against tet regulatory elements observed by other groups in nonhuman primates after intramuscular injection of tet-on encoding vectors raise concerns about the clinical value of tet-regulated vectors. However, previous studies have not examined immune responses following injection of AAV vectors into brain. Therefore, rat striatum was injected with tet-off rAAV harboring a therapeutic gene for Parkinson's disease, either hAADC or hGDNF. The expression of each gene was tightly controlled by the tet-off regulatory system. Using an ELISA developed with purified GST–tTA protein, no detectable immunogenicity against tTA was observed in sera of rats that received an intrastriatal injection of either vector. In contrast, sera from rats intradermally injected with an adenovirus containing either tTA or rtTA, as positive controls, had readily detectable antibodies. These observations suggest that tet-off rAAV vectors do not elicit an immune response when injected into rat brain and that these may offer safer vectors for Parkinson's disease than vectors with constitutive expression. [ABSTRACT FROM AUTHOR]
- Published
- 2010
- Full Text
- View/download PDF
29. Neonatal gene transfer using lentiviral vector for murine Pompe disease: long-term expression and glycogen reduction.
- Author
-
Kyosen, S. O., Iizuka, S., Kobayashi, H., Kimura, T, Fukuda, T., Shen, J., Shimada, Y., Ida, H., Eto, Y., and Ohashi, T.
- Subjects
GLYCOGEN storage disease type II ,GENETIC transformation ,LYSOSOMAL storage diseases ,GLYCOGEN ,TRANSGENE expression - Abstract
Pompe disease results from the deficiency of the lysosomal enzyme acid α-glucosidase (GAA), leading to accumulated glycogen in the heart and the skeletal muscles, which causes cardiomyopathy and muscle weakness. In this study, we tested the feasibility of gene therapy for Pompe disease using a lentivirus vector (LV). Newborn GAA knockout mice were treated with intravenous injection of LV encoding human GAA (hGAA) through the facial superficial temporal vein. The transgene expression in the tissues was analyzed up to 24 weeks after treatment. Our results showed that the recombinant LV was efficient not only in increasing the GAA activity in tissues but also in decreasing their glycogen content. The examination of histological sections showed clearence of the glycogen storage in skeletal and cardiac muscles 16 and 24 weeks after a single vector injection. Levels of expressed hGAA could be detected in serum of treated animals until 24 weeks. No significant immune reaction to transgene was detected in most treated animals. Therefore, we show that LV-mediated delivery system was effective in correcting the biochemical abnormalities and that this gene transfer system might be suitable for further studies on delivering GAA to Pompe disease mouse models. [ABSTRACT FROM AUTHOR]
- Published
- 2010
- Full Text
- View/download PDF
30. Mannitol-facilitated CNS entry of rAAV2 vector significantly delayed the neurological disease progression in MPS IIIB mice.
- Author
-
McCarty, D. M., DiRosario, J., Gulaid, K., Muenzer, J., and Fu, H.
- Subjects
MUCOPOLYSACCHARIDOSIS ,CARBOHYDRATE metabolism disorders ,MICROBIAL genetics ,LABORATORY mice ,GENE therapy - Abstract
The presence of the blood–brain barrier (BBB) presents the most critical challenge in therapeutic development for mucopolysaccharidosis (MPS) IIIB, a lysosomal storage disease with severe neurological manifestation, because of α-N-acetylglucosaminidase (NaGlu) deficiency. Earlier, we showed a global central nervous system (CNS) transduction in mice by mannitol-facilitated entry of intravenous (IV)-delivered recombinant adeno-associated viral serotype 2 (rAAV2) vector. In this study, we optimized the approach and showed that the maximal transduction in the CNS occurred when the rAAV2 vector was IV injected at 8 min after mannitol administration, and was approximately 10-fold more efficient than IV delivery of the vector at 5 or 10 min after mannitol infusion. Using this optimal (8 min) regimen, a single IV infusion of rAAV2-CMV-hNaGlu vector is therapeutically beneficial for treating the CNS disease of MPS IIIB in adult mice, with significantly extended survival, improved behavioral performance, and reduction of brain lysosomal storage pathology. The therapeutic benefit correlated with maximal delivery to the CNS, but not peripheral tissues. This milestone data shows the first effective gene delivery across the BBB to treat CNS disease. The critical timing of vector delivery and mannitol infusion highlights the important contribution of this pretreatment to successful intervention, and the long history of safe use of mannitol in patients bodes well for its application in CNS gene therapy. [ABSTRACT FROM AUTHOR]
- Published
- 2009
- Full Text
- View/download PDF
31. Fragile X mental retardation protein replacement restores hippocampal synaptic function in a mouse model of fragile X syndrome.
- Author
-
Zeier, Z., Kumar, A., Bodhinathan, K., Feller, J. A., Foster, T. C., and Bloom, D. C.
- Subjects
INTELLECTUAL disabilities ,X chromosome abnormalities ,PROTEINS ,COGNITION disorders ,CENTRAL nervous system diseases ,GENOTYPE-environment interaction - Abstract
Fragile X syndrome (FXS) is caused by a mutation that silences the fragile X mental retardation gene (FMR1), which encodes the fragile X mental retardation protein (FMRP). To determine whether FMRP replacement can rescue phenotypic deficits in a fmr1-knockout (KO) mouse model of FXS, we constructed an adeno-associated virus-based viral vector that expresses the major central nervous system (CNS) isoform of FMRP. Using this vector, we tested whether FMRP replacement could rescue the fmr1-KO phenotype of enhanced long-term depression (LTD), a form of synaptic plasticity that may be linked to cognitive impairments associated with FXS. Extracellular excitatory postsynaptic field potentials were recorded from CA3–CA1 synaptic contacts in hippocampal slices from wild-type (WT) and fmr1-KO mice in the presence of AP-5 and anisomycin. Paired-pulse low-frequency stimulation (PP-LFS)-induced LTD is enhanced in slices obtained from fmr1 KO compared with WT mice. Analyses of hippocampal synaptic function in fmr1-KO mice that received hippocampal injections of vector showed that the PP-LFS-induced LTD was restored to WT levels. These results indicate that expression of the major CNS isoform of FMRP alone is sufficient to rescue this phenotype and suggest that post-developmental protein replacement may have the potential to improve cognitive function in FXS. [ABSTRACT FROM AUTHOR]
- Published
- 2009
- Full Text
- View/download PDF
32. Indoleamine 2,3-dioxygenase attenuates inhibitor development in gene-therapy-treated hemophilia A mice.
- Author
-
Liu, L., Liu, H., Mah, C., and Fletcher, B. S.
- Subjects
HEMOPHILIA ,T cells ,APOPTOSIS ,KYNURENINE ,IMMUNOGLOBULINS - Abstract
A serious impediment to gene and protein replacement therapy in hemophilia A is the development of inhibitors. Mechanisms responsible for inhibitor development include T-cell-dependent adaptive immune responses and the CD28–B7 signaling pathway that eventually leads to the formation of antibodies directed against factor VIII (FVIII). Indoleamine 2,3-dioxygenase (IDO) is a potent immunosuppressive enzyme that can inhibit T-cell responses and induce T-cell apoptosis by regulation of tryptophan metabolism. Kynurenine, one of the metabolites of tryptophan, has been implicated as an immune modulator. Here we hypothesize that co-delivery of the genes for FVIII and IDO can attenuate inhibitor formation. Using transposon-based gene delivery, we observed long-term therapeutic FVIII expression and significantly reduced inhibitor titers when the genes were co-delivered. Co-expression of FVIII and IDO in the liver was associated with increased plasma kynurenine levels, an inhibition of T-cell infiltration and increased apoptosis of T cells within the liver. These experiments suggest that modulation of tryptophan catabolism through IDO expression provides a novel strategy to reduce inhibitor development in hemophilia gene/protein therapy.Gene Therapy (2009) 16, 724–733; doi:10.1038/gt.2009.13; published online 5 March 2009 [ABSTRACT FROM AUTHOR]
- Published
- 2009
- Full Text
- View/download PDF
33. Tailoring the AAV vector capsid for gene therapy.
- Author
-
Vandenberghe, L. H., Wilson, J. M., and Gao, G.
- Subjects
GENE therapy ,GENETIC transformation ,MIRIDAE ,MAMMALS ,GENETIC vectors - Abstract
A number of preclinical studies have shown the adeno-associated virus (AAV) to be an efficient vehicle for gene therapy. Clinical studies successfully demonstrated its potential for in vivo gene transfer. The complexity of host–vector interactions when progressing from small to large animal models, and eventually to humans, has impeded translation of AAV technology to the clinic. One approach to address this complexity has been to explore the biological characteristics of variations in AAV capsid structure. Initial strategies characterized the naturally occurring capsid variants from mammalian species. The structural and functional knowledge gathered on these natural AAV variants as vectors has led to the first series of second-generation vectors that aim at specifically improving certain properties by rational design of the capsid. A third exciting approach uses directed evolution to isolate vectors that are able to overcome selective pressures applied in the laboratory and thereby steer the capsid to evolve toward improved functionality.Gene Therapy (2009) 16, 311–319; doi:10.1038/gt.2008.170; published online 4 December 2008 [ABSTRACT FROM AUTHOR]
- Published
- 2009
- Full Text
- View/download PDF
34. An efficient rHSV-based complementation system for the production of multiple rAAV vector serotypes.
- Author
-
Kang, W., Wang, L., Harrell, H., Liu, J., Thomas, D. L., Mayfield, T. L., Scotti, M. M., Ye, G. J., Veres, G., and Knop, D. R.
- Subjects
RECOMBINANT viruses ,HERPES simplex virus ,HERPESVIRUSES ,VIRUS diseases ,GENE transfection ,VIRAL genetics - Abstract
Recombinant herpes simplex virus type 1 (rHSV)-assisted recombinant adeno-associated virus (rAAV) vector production provides a highly efficient and scalable method for manufacture of clinical grade rAAV vectors. Here, we present an rHSV co-infection system for rAAV production, which uses two ICP27-deficient rHSV constructs, one bearing the rep2 and cap (1, 2 or 9) genes of rAAV, and the second bearing an AAV2 ITR-gene of interest (GOI) cassette. The optimum rAAV production parameters were defined by producing rAAV2/GFP in HEK293 cells, yielding greater than 9000 infectious particles per cell with a 14:1 DNase resistance particle to infectious particle (DRP/ip) ratio. The optimized co-infection parameters were then used to generate large-scale stocks of rAAV1/AAT, which encode the human α-1-antitrypsin (hAAT) protein, and purified by column chromatography. The purified vector was extensively characterized by rAAV- and rHSV-specific assays and compared to transfection-made vector for in vivo efficacy in mice through intramuscular injection. The co-infection method was also used to produce rAAV9/AAT for comparison to rAAV1/AAT in vivo. Intramuscular administration of 1 × 10
11 DRP per animal of rHSV-produced rAAV1/AAT and rAAV9/AAT resulted in hAAT protein expression of 5.4 × 104 and 9.4 × 105 ng ml−1 serum respectively, the latter being clinically relevant.Gene Therapy (2009) 16, 229–239; doi:10.1038/gt.2008.158; published online 16 October 2008 [ABSTRACT FROM AUTHOR]- Published
- 2009
- Full Text
- View/download PDF
35. Novel anti-VEGF chimeric molecules delivered by AAV vectors for inhibition of retinal neovascularization.
- Author
-
Pechan, P., Rubin, H., Lukason, M., Ardinger, J., DuFresne, E., Hauswirth, W. W., Wadsworth, S. C., and Scaria, A.
- Subjects
VASCULAR endothelial growth factors ,NEOVASCULARIZATION inhibitors ,RETROLENTAL fibroplasia ,IMMUNOGLOBULIN genes ,GENE therapy ,SCIENTIFIC method - Abstract
Vascular endothelial growth factor (VEGF) is important in pathological neovascularization, which is a key component of diseases such as the wet form of age-related macular degeneration, proliferative diabetic retinopathy and cancer. One of the most potent naturally occurring VEGF binders is VEGF receptor Flt-1. We have generated two novel chimeric VEGF-binding molecules, sFLT01 and sFLT02, which consist of the second immunoglobulin (IgG)-like domain of Flt-1 fused either to a human IgG1 Fc or solely to the CH3 domain of IgG1 Fc through a polyglycine linker 9Gly. In vitro analysis showed that these novel molecules are high-affinity VEGF binders. We have demonstrated that adeno-associated virus serotype 2 (AAV2)-mediated intravitreal gene delivery of sFLT01 efficiently inhibits angiogenesis in the mouse oxygen-induced retinopathy model. There were no histological observations of toxicity upon persistent ocular expression of sFLT01 for up to 12 months following intravitreal AAV2-based delivery in the rodent eye. Our data suggest that AAV2-mediated intravitreal gene delivery of our novel molecules may be a safe and effective treatment for retinal neovascularization.Gene Therapy (2009) 16, 10–16; doi:10.1038/gt.2008.115; published online 17 July 2008 [ABSTRACT FROM AUTHOR]
- Published
- 2009
- Full Text
- View/download PDF
36. Recirculating cardiac delivery of AAV2/1SERCA2a improves myocardial function in an experimental model of heart failure in large animals.
- Author
-
Byrne, M. J., Power, J. M., Preovolos, A., Mariani, J. A., Hajjar, R. J., and Kaye, D. M.
- Subjects
HEART failure ,CARDIAC arrest ,EXCITATION (Physiology) ,SARCOPLASMIC reticulum ,HISTOPATHOLOGY - Abstract
Abnormal excitation–contraction coupling is a key pathophysiologic component of heart failure (HF), and at a molecular level reduced expression of the sarcoplasmic reticulum (SR) Ca
2+ ATPase (SERCA2a) is a major contributor. Previous studies in small animals have suggested that restoration of SERCA function is beneficial in HF. Despite this promise, the means by which this information might be translated into potential clinical application remains uncertain. Using a recently established cardiac-directed recirculating method of gene delivery, we administered adeno-associated virus 2 (AAV2)/1SERCA2a to sheep with pacing-induced HF. We explored the effects of differing doses of AAV2/1SERCA2a (low 1 × 1010 d.r.p.; medium 1 × 1012 d.r.p. and high 1 × 1013 d.r.p.) in conjunction with an intra-coronary delivery group (2.5 × 1013 d.r.p.). At the end of the study, haemodynamic, echocardiographic, histopathologic and molecular biologic assessments were performed. Cardiac recirculation delivery of AAV2/1SERCA2a elicited a dose-dependent improvement in cardiac performance determined by left ventricular pressure analysis, (+d P/d tmax ; low dose −220±70, P>0.05; medium dose 125±53, P<0.05; high dose 287±104, P<0.05) and echocardiographically (fractional shortening: low dose −3±2, P>0.05; medium dose 1±2, P>0.05; high dose 6.5±3.9, P<0.05). In addition to favourable haemodynamic effects, brain natriuretic peptide expression was reduced consistent with reversal of the HF molecular phenotype. In contrast, direct intra-coronary infusion did not elicit any effect on ventricular function. As such, AAV2/1SERCA2a elicits favourable functional and molecular actions when delivered in a mechanically targeted manner in an experimental model of HF. These observations lay a platform for potential clinical translation.Gene Therapy (2008) 15, 1550–1557; doi:10.1038/gt.2008.120; published online 24 July 2008 [ABSTRACT FROM AUTHOR]- Published
- 2008
- Full Text
- View/download PDF
37. Cancer gene therapy using mesenchymal stem cells expressing interferon-β in a mouse prostate cancer lung metastasis model.
- Author
-
Ren, C, Kumar, S, Chanda, D, Kallman, L, Chen, J, Mountz, J D, and Ponnazhagan, S
- Subjects
CANCER treatment ,GENE therapy ,STEM cells ,INTERFERONS ,PROSTATE cancer ,METASTASIS ,LUNGS ,ANIMAL models in research ,MICE - Abstract
Cell-based therapy for cancer is a promising new field. Among cell types that can be used for this purpose, mesenchymal stem cells (MSCs) appear to hold great advantage for reasons including easier propagation in culture, possible genetic modification to express therapeutic proteins and preferential homing to sites of cancer growth upon in vivo transfer. The present study evaluated the potential of genetically modified MSC, constitutively expressing interferon (IFN)-β, in an immunocompetent mouse model of prostate cancer lung metastasis. A recombinant adeno-associated virus (rAAV) encoding mouse IFN-β was constructed and initially tested in vitro for high-level expression and bioactivity of the transgenic protein. MSCs were transduced by the rAAV-IFN-β or green fluorescent protein ex vivo and used as cellular vehicles to target lung metastasis of TRAMP-C2 prostate cancer cells in a therapy model. Cohorts of mice were killed on days 30 and 75 to determine the effect of therapy by measurement of tumor volume, histology, immunohistochemistry, enzyme-linked immunosorbent assay and flow cytometry. Results indicated a significant reduction in tumor volume in lungs following IFN-β-expressing MSC therapy. Immunohistochemistry of the lung demonstrated increased tumor cell apoptosis and decreased tumor cell proliferation and blood vessel counts. A significant increase in the natural kill cell activity was observed following IFN-β therapy correlating the antitumor effect. Systemic level of IFN-β was not significantly elevated from this targeted cell therapy. These data demonstrate the potential of MSC-based IFN-β therapy for prostate cancer lung metastasis.Gene Therapy (2008) 15, 1446–1453; doi:10.1038/gt.2008.101; published online 3 July 2008 [ABSTRACT FROM AUTHOR]
- Published
- 2008
- Full Text
- View/download PDF
38. Targeting gene expression to cones with human cone opsin promoters in recombinant AAV.
- Author
-
Komáromy, A. M., Alexander, J. J., Cooper, A. E., Chiodo, V. A., Acland, G. M., Hauswirth, W. W., and Aguirre, G. D.
- Subjects
RETINAL (Visual pigment) ,VISUAL pigments ,PHOTORECEPTORS ,GENE expression ,DOG diseases ,ACHROMATISM ,GENE therapy - Abstract
Specific cone-directed therapy is of high priority in the treatment of human hereditary retinal diseases. However, not much information exists about the specific targeting of photoreceptor subclasses. Three versions of the human red cone opsin promoter (PR0.5, 3LCR-PR0.5 and PR2.1), and the human blue cone opsin promoter HB569, were evaluated for their specificity and robustness in targeting green fluorescent protein (GFP) gene expression to subclasses of cones in the canine retina when used in recombinant adeno-associated viral vectors of serotype 5. The vectors were administered by subretinal injection. The promoter PR2.1 led to most effective and specific expression of GFP in the long- and medium-wavelength-absorbing cones (L/M cones) of normal and diseased retinas. The PR0.5 promoter was not effective. Adding three copies of the 35-bp LCR in front of PR0.5 lead to weak GFP expression in L/M cones. The HB569 promoter was not specific, and GFP was expressed in a few L/M cones, some rods and the retinal pigment epithelium. These results suggest that L/M cones, the predominant class of cone photoreceptors in the retinas of dogs and most mammalian species can be successfully targeted using the human red cone opsin promoter.Gene Therapy (2008) 15, 1049–1055; doi:10.1038/gt.2008.32; published online 13 March 2008 [ABSTRACT FROM AUTHOR]
- Published
- 2008
- Full Text
- View/download PDF
39. Toward exascale production of recombinant adeno-associated virus for gene transfer applications.
- Author
-
Cecchini, S., Negrete, A., and Kotin, R. M.
- Subjects
GENETIC vectors ,MICROBIAL genetic engineering ,CLINICAL trials ,DUCHENNE muscular dystrophy ,TISSUE culture - Abstract
To gain acceptance as a medical treatment, adeno-associated virus (AAV) vectors require a scalable and economical production method. Recent developments indicate that recombinant AAV (rAAV) production in insect cells is compatible with current good manufacturing practice production on an industrial scale. This platform can fully support development of rAAV therapeutics from tissue culture to small animal models, to large animal models, to toxicology studies, to Phase I clinical trials and beyond. Efforts to characterize, optimize and develop insect cell-based rAAV production have culminated in successful bioreactor-scale production of rAAV, with total yields potentially capable of approaching the ‘exa-(10
18 ) scale.’ These advances in large-scale AAV production will allow us to address specific catastrophic, intractable human diseases such as Duchenne muscular dystrophy, for which large amounts of recombinant vector are essential for successful outcome.Gene Therapy (2008) 15, 823–830; doi:10.1038/gt.2008.61; published online 10 April 2008 [ABSTRACT FROM AUTHOR]- Published
- 2008
- Full Text
- View/download PDF
40. Potential of AAV vectors in the treatment of metabolic disease.
- Author
-
Alexander, I. E., Cunningham, S. C., Logan, G. J., and Christodoulou, J.
- Subjects
METABOLIC disorders ,GENE therapy ,PHENOTYPES ,CENTRAL nervous system ,LIPOPROTEIN lipase - Abstract
Inborn errors of metabolism are collectively common, frequently severe and in many instances difficult or impossible to treat. Accordingly, there is a compelling need to explore novel therapeutic modalities, including gene therapy, and examine multiple phenotypes where the risks of experimental therapy are outweighed by potential benefits to trial participants. Among available gene delivery systems recombinant AAV shows special promise for the treatment of metabolic disease given the unprecedented efficiencies achieved in transducing key target tissues, such as liver and muscle, in small animal models. To date over 30 metabolic disease phenotypes have been investigated in small animal studies with complete phenotype correction being achieved in a substantial proportion. Achieving adequately widespread transduction within the central nervous system, however, remains a major challenge, and will be critical to realization of the therapeutic potential of gene therapy for many of the most clinically troubling metabolic disease phenotypes. Despite the relatively low immunogenicity of AAV vectors, immune responses are also emerging as a factor requiring special attention as efforts accelerate toward human clinical translation. Four metabolic disease phenotypes have reached phase I or I/II trials with one, targeting lipoprotein lipase deficiency, showing exciting early evidence of efficacy.Gene Therapy (2008) 15, 831–839; doi:10.1038/gt.2008.64; published online 10 April 2008 [ABSTRACT FROM AUTHOR]
- Published
- 2008
- Full Text
- View/download PDF
41. Manufacturing and characterizing AAV-based vectors for use in clinical studies.
- Author
-
Wright, J. F.
- Subjects
ADENOMA ,GENE therapy ,CLINICAL medicine ,GENETIC engineering ,THERAPEUTICS - Abstract
Recombinant adeno-associated virus (AAV)-based vectors expressing therapeutic gene products have shown great promise for human gene therapy. A major challenge for translation of promising research to clinical development is the manufacture and certification of AAV vectors for clinical use. This review summarizes relevant aspects of current Good Manufacturing Practice, focusing on considerations and challenges specific for recombinant AAV.Gene Therapy (2008) 15, 840–848; doi:10.1038/gt.2008.65; published online 17 April 2008 [ABSTRACT FROM AUTHOR]
- Published
- 2008
- Full Text
- View/download PDF
42. Systemic AAV-9 transduction in mice is influenced by animal age but not by the route of administration.
- Author
-
Bostick, B., Ghosh, A., Yue, Y., Long, C., and Duan, D.
- Subjects
VIRUS diseases ,GENETIC transformation ,AGE ,ALKALINE phosphatase ,NEWBORN infants ,MUSCLES ,MICE ,AORTA ,LIVER ,KIDNEYS - Abstract
Adeno-associated virus (AAV) serotype-9 (AAV-9) has attracted great attention as an optimal vehicle for body-wide gene delivery. Here we examined the effect of animal age (newborn vs adult) and the route of administration (intravenous vs intra-arterial) on systemic AAV-9 transduction. We delivered an alkaline phosphatase (AP) reporter gene AAV vector (AV.RSV.AP) to either newborn (via either the facial vein or the left ventricular cavity) or adult (via tail vein) C57Bl/10 mice. At 12 weeks’ postinfection, we examined the AP expression. We observed efficient transduction in multiple skeletal muscles and the heart, irrespective of the age or delivery route. However, the soleus muscle, which consists mainly of slow-twitch myofibers, was poorly transduced. Besides striated muscle, we also found consistent high-level transduction in the lung. Abundant AP-positive cells were seen in alveolar cells and vasculature, but not in bronchioles. Interestingly, several organs demonstrated an age-dependent profile. In particular, the aorta, liver and kidney were preferentially transduced in adult mice while the inner layer of retina was strongly transduced only following the neonatal administration. Taken together, our results demonstrate the robustness of intravascular AAV-9 delivery for muscle and lung gene therapy applications. The unique expression patterns in the aorta, liver, kidney and retina call for special attention when designing AAV-9 gene therapy applications for these organs.Gene Therapy (2007) 14, 1605–1609; doi:10.1038/sj.gt.3303029; published online 27 September 2007 [ABSTRACT FROM AUTHOR]
- Published
- 2007
- Full Text
- View/download PDF
43. Differential internalization and nuclear uncoating of self-complementary adeno-associated virus pseudotype vectors as determinants of cardiac cell transduction.
- Author
-
Sipo, I., Fechner, H., Pinkert, S., Suckau, L., Wang, X., Weger, S., and Poller, W.
- Subjects
CELLULAR mechanics ,HEART cells ,MICROBIAL genetics ,GENETIC transduction ,BACTERIOPHAGES ,TRANSGENE expression ,GENE therapy - Abstract
Recently it was shown that several new pseudotyped adeno-associated virus (AAV) vectors support cardioselective expression of transgenes. The molecular mechanisms underlying this propensity for cardiac cell transduction are not well understood. We comparatively analyzed AAV vector attachment, internalization, intracellular trafficking, and nuclear uncoating of recombinant self-complementary (sc) AAV2.2 versus pseudotyped scAAV2.6 vectors expressing green fluorescence protein (GFP) in cells of cardiac origin. In cardiac-derived HL-1 cells and primary neonatal rat cardiomyocytes (PNCMs), expression of GFP increased rapidly after incubation with scAAV2.6-GFP, but remained low after scAAV2.2-GFP. Internalization of scAAV2.6-GFP was more efficient than that of scAAV2.2-GFP. Nuclear translocation was similarly efficient for both, but differential nuclear uncoating rates emerged as a key additional determinant of transduction: 30% of all scAAV2.6-GFP genomes translocated to the nucleus became uncoated within 48 h, but only 16% of scAAV2.2-GFP genomes. In contrast to this situation in cells of cardiac origin, scAAV2.2-GFP displayed more efficient internalization and similar (tumor cell line HeLa) or higher (human microvascular endothelial cell (HMEC)) uncoating rates than scAAV.2.6-GFP in non-cardiac cell types. In summary, both internalization and nuclear uncoating are key determinants of cardiac transduction by scAAV2.6 vectors. Any in vitro screening for the AAV pseudotype most suitable for cardiac gene therapy – which is desirable since it may allow significant reductions in vector load in upcoming clinical trials – needs to quantitate both key steps in transduction.Gene Therapy (2007) 14, 1319–1329; doi:10.1038/sj.gt.3302987; published online 5 July 2007 [ABSTRACT FROM AUTHOR]
- Published
- 2007
- Full Text
- View/download PDF
44. Significantly increased lifespan and improved behavioral performances by rAAV gene delivery in adult mucopolysaccharidosis IIIB mice.
- Author
-
Fu, H., Kang, L., Jennings, J. S., Moy, S. S., Perez, A, DiRosario, J., McCarty, D. M., and Muenzer, J.
- Subjects
MUCOPOLYSACCHARIDOSIS ,LYSOSOMAL storage diseases ,INBORN errors of metabolism ,CENTRAL nervous system ,GENE therapy ,GENETIC engineering - Abstract
Mucopolysaccharidosis (MPS) IIIB is an inherited lysosomal storage disease, caused by the deficiency of α-N-acetylglucosaminidase (NaGlu), resulting in severe global neurological involvement with high mortality. One major hurdle in therapeutic development for MPS IIIB is the presence of the blood–brain barrier, which impedes the global central nervous system (CNS) delivery of therapeutic materials. In this study, we used a minimal invasive strategy, combining an intravenous (i.v.) and an intracisternal (i.c.) injection, following an i.v. infusion of mannitol, to complement the CNS delivery of adeno-associated viral (AAV) vector for treating MPS IIIB in young adult mice. This treatment resulted in a significantly prolonged lifespan of MPS IIIB mice (11.1–19.5 months), compared with that without treatment (7.9–11.3), and correlated with significantly improved behavioral performances, the restoration of functional NaGlu, and variable correction of lysosomal storage pathology in the CNS, as well as in different somatic tissues. This study demonstrated the great potential of combining i.v. and i.c. administration for improving rAAV CNS gene delivery and developing rAAV gene therapy for treating MPS IIIB in patients.Gene Therapy (2007) 14, 1065–1077; doi:10.1038/sj.gt.3302961; published online 26 April 2007 [ABSTRACT FROM AUTHOR]
- Published
- 2007
- Full Text
- View/download PDF
45. Intraperitoneal gene therapy by rAAV provides long-term survival against epithelial ovarian cancer independently of survivin pathway.
- Author
-
Isayeva, T., Ren, C., and Ponnazhagan, S.
- Subjects
CANCER treatment ,GENE therapy ,OVARIAN cancer ,PACLITAXEL ,DISEASES in women ,GENETIC transformation ,DRUG therapy - Abstract
Epithelial ovarian carcinoma is the leading cause of death from gynecological malignancies. Owing to the lack of an effective screening method, insidious onset, and non-specific symptoms, a majority of women present with advanced stage disease. Despite improvements from cytoreductive surgery and chemotherapy, recurrent disease remains a formidable challenge. In the present study, we demonstrate for the first time that stable intra-abdominal genetic transfer of endostatin and angiostatin (E+A) by recombinant adeno-associated virus (rAAV) provides sustained antitumor effects on the growth and dissemination of epithelial ovarian cancer in a mouse model. Further, when combined with paclitaxel (taxol), the effect of this therapy was dramatically increased and resulted in long-term tumor-free survival overcoming prior limitations of chemotherapy and gene therapy. The combined effects of angiosuppressive therapy and chemotherapy were found to be independently of survivin pathway. Evidence for the superior effects of the combination therapy was indicated by significantly lower ascites volume with less hemorrhage and tumor conglomerates, lower ascites vascular endothelial growth factor, higher tumor cell apoptosis and decreased blood vasculature, and long-term disease-free survival. Histopathology of visceral organs and liver enzyme assays indicated no toxicity or pathology.Gene Therapy (2007) 14, 138–146. doi:10.1038/sj.gt.3302853; published online 31 August 2006 [ABSTRACT FROM AUTHOR]
- Published
- 2007
- Full Text
- View/download PDF
46. Long-term persistence of gene expression from adeno-associated virus serotype 5 in the mouse airways.
- Author
-
Sumner-Jones, S. G., Davies, L. A., Varathalingam, A., Gill, D. R., and Hyde, S. C.
- Subjects
GENE expression ,TRANSGENE expression ,AIRWAY (Anatomy) ,GENOMES ,EPITHELIAL cells ,MICE - Abstract
Recombinant adeno-associated virus vectors based on serotype 2 (rAAV2) have been used to deliver transgenes to the airways in a variety of pre-clinical and clinical studies. Gene transfer in these studies has been severely restricted by the basolateral localization of rAAV2 receptors. Here, we studied vectors constructed from the AAV5 genome and capsid, which utilize N-linked sialic acid-containing receptors found on the apical surface of airway epithelial cells. We investigated gene transfer efficacy and duration of transgene expression following delivery of rAAV5/5 vectors to the mouse respiratory tract. Robust, dose-dependent transgene expression was observed in the epithelium lining the nose for at least 32 weeks, and for at least 52 weeks in the lung. Importantly, in the lung, transgene expression mediated by rAAV5/5 was 40-fold greater than by rAAV2/2 vectors. A distinct cellular preference for rAAV5/5-mediated transduction was observed, with transgene expression being predominantly restricted to sustentacular cells of the olfactory epithelium in the nose and alveolar type II cells in the lung. Administration of rAAV5/5 vectors to both the nose and lungs led to the rapid development of rAAV5/5-neutralizing antibodies, suggesting that repeated administration may be severely hampered by host immune responses.Gene Therapy (2006) 13, 1703–1713. doi:10.1038/sj.gt.3302815; published online 20 July 2006 [ABSTRACT FROM AUTHOR]
- Published
- 2006
- Full Text
- View/download PDF
47. Using magnetic forces to enhance non-viral gene transfer to airway epithelium in vivo.
- Author
-
Xenariou, S., Griesenbach, U., Ferrari, S., P.Dean, Scheule, R. K., Cheng, S. H., Geddes, D. M., Plank, C., and Alton, E. W. F. W
- Subjects
GENETIC transformation ,VIRAL genetics ,EPITHELIUM ,PLASMIDS ,MAGNETIC fields ,GENE expression - Abstract
We have assessed whether magnetic forces (magnetofection) can enhance non-viral gene transfer to the airways. TransMAG
PEI , a superparamagnetic particle was coupled to Lipofectamine 2000 or cationic lipid 67 (GL67)/plasmid DNA (pDNA) liposome complexes. In vitro transfection with these formulations resulted in approximately 300- and 30-fold increase in reporter gene expression, respectively, after exposure to a magnetic field, but only at suboptimal pDNA concentrations. Because GL67 has been formulated for in vivo use, we next assessed TransMAGPEI in the murine nasal epithelium in vivo, and compared this to naked pDNA. At the concentrations required for in vivo experiments, precipitation of magnetic complexes was seen. After extensive optimization, addition of non-precipitated magnetic particles resulted in approximately seven- and 90-fold decrease in gene expression for naked pDNA and GL67/pDNA liposome complexes, respectively, compared to non-magnetic particles. Thus, whereas exposure to a magnetic field improved in vitro transfection efficiency, translation to the in vivo setting remains difficult.Gene Therapy (2006) 13, 1545–1552. doi:10.1038/sj.gt.3302803; published online 1 June 2006 [ABSTRACT FROM AUTHOR]- Published
- 2006
- Full Text
- View/download PDF
48. Long-term expression and repeated administration of AAV type 1, 2 and 5 vectors in skeletal muscle of immunocompetent adult mice.
- Author
-
Rivière, C, Danos, O, and Douar, A M
- Subjects
GENE therapy ,HELA cells ,DENDRITIC cells ,IMMUNE response ,GENE expression - Abstract
Adeno-associated viral (AAV) vectors promote long-term gene transfer into muscle in many animal species. Increased expression levels may be obtained by using alternative serotypes in combination with repeated administrations. Here we compared AAV vectors based on serotypes 1, 2 and 5 in immunocompetent mice and assessed the feasibility of multiple administrations of either identical (readministration) or different (cross-administration) serotype-based vectors. A 1-year-long dose–response study confirmed the superiority of recombinant (r)AAV1, achieving transduction levels 5 to 10-fold higher than rAAV2 and rAAV5 in mouse skeletal muscle, respectively. Repeated administration demonstrated that increased gene transfer level was achieved with a second injection of rAAV1 following the first administration of rAAV2 or rAAV5. A readministration study with a vector encoding a different gene allowed the evaluation of gene expression from the second vector only. All three rAAVs were inhibited when the animals were previously exposed to the same serotype. In contrast, no significant change in gene expression from the second vector was observed in cross-administration. A humoral immune response was elicited against the viral capsid for all three serotypes following the initial exposure. Neutralizing antibody (NAB) levels correlated with the vector dose injected. No significant cross-reactivity of NAB from a given serotype toward another was observed in vitro. These data provide the first direct comparative evaluation of re- and cross-administration of rAAV1, rAAV2 and rAAV5 in muscle, and further indicate that rAAV1 is capable of transducing muscle tissue when cross-administered.Gene Therapy (2006) 13, 1300–1308. doi:10.1038/sj.gt.3302766; published online 11 May 2006 [ABSTRACT FROM AUTHOR]
- Published
- 2006
- Full Text
- View/download PDF
49. Restoration of fatty aldehyde dehydrogenase deficiency in Sjögren–Larsson syndrome.
- Author
-
Haug, S and Braun-Falco, M
- Subjects
SJOGREN'S syndrome ,GENE therapy ,NEUROCUTANEOUS disorders ,GENETIC mutation ,ALDEHYDES ,FATTY acids - Abstract
Sjögren–Larsson syndrome (SLS) is an autosomal recessive neurocutaneous disorder caused by mutation in the ALDH3A2 gene that codes for human fatty aldehyde dehydrogenase (FALDH). Sjögren–Larsson syndrome patients lack FALDH, which catalyzes the oxidation of long-chain aliphatic aldehydes to fatty acids. The impaired FALDH activity leads to congenital ichthyosis, mental retardation and spasticity. The current lack of treatment is an impetus to develop gene therapy strategies by introducing functional FALDH into defective cells. We delivered human FALDH into keratinocytes of SLS patients using recombinant adeno-associated virus-2 vectors. Transduction of SLS keratinocytes resulted in an augmentation of FALDH activity comparable to phenotypically normal heterozygous carriers. Toxicity of long-chain aldehydes for FALDH-deficient cells decreased almost to the level of unaffected keratinocytes. Three-dimensional culture of corrected SLS keratinocytes revealed an ameliorated FALDH expression. These studies demonstrate the restoration of FALDH in human SLS cells supporting the concept of gene therapy as a potential future treatment option for SLS.Gene Therapy (2006) 13, 1021–1026. doi:10.1038/sj.gt.3302743; published online 9 March 2006 [ABSTRACT FROM AUTHOR]
- Published
- 2006
- Full Text
- View/download PDF
50. Intrathecal administration of AAV vectors for the treatment of lysosomal storage in the brains of MPS I mice.
- Author
-
Watson, G., Bastacky, J., Belichenko, P., Buddhikot, M., Jungles, S., Vellard, M., Mobley, W. C., and Kakkis, E.
- Subjects
MUCOPOLYSACCHARIDOSIS ,LYSOSOMAL storage diseases ,HISTOPATHOLOGY ,NEURONS ,NEUROGLIA ,CEREBELLUM ,GENE therapy - Abstract
Mucopolysaccharidosis type I (MPS I) is caused by an inherited deficiency of α-L-iduronidase (IDUA). The result is a progressive, lysosomal storage disease with central nervous system (CNS) as well as systemic involvement. To target gene therapy to the CNS, recombinant adeno-associated virus (AAV) vectors carrying IDUA sequence were administered to MPS I mice via injection into cerebrospinal fluid. In contrast to intravenous administration, this intrathecal administration was effective in generating widespread IDUA activity in the brain, with the cerebellum and olfactory bulbs having highest activities. In general, IDUA levels correlated with vector dose, although this correlation was obscured in cerebellum by particularly high variability. High doses of vector (4 × 10
10 particles) provided IDUA levels approaching or exceeding normal levels in the brain. Histopathology indicated that the number of cells with storage vacuoles was reduced extensively or was eliminated entirely. Elimination of storage material in Purkinje cells was particularly dramatic. A lower vector dose (2 × 109 particles) reduced both the number of storage cells and the extent of storage per cell, but the effect was not complete. Some perivascular cells with storage persisted, and this cell type appeared to be more resistant to treatment than neurons or glial cells. We conclude that intrathecal administration of AAV-IDUA delivers vector to brain cells, and that this route of administration is both minimally invasive and effective.Gene Therapy (2006) 13, 917–925. doi:10.1038/sj.gt.3302735; published online 16 February 2006 [ABSTRACT FROM AUTHOR]- Published
- 2006
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.