1
|
Hutchinson-Gilford Progeria Syndrome-Current Status and Prospects for Gene Therapy Treatment. Cells 2019; 8:cells8020088. [PMID: 30691039 PMCID: PMC6406247 DOI: 10.3390/cells8020088] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Revised: 01/18/2019] [Accepted: 01/19/2019] [Indexed: 12/13/2022] Open
Abstract
Hutchinson-Gilford progeria syndrome (HGPS) is one of the most severe disorders among laminopathies—a heterogeneous group of genetic diseases with a molecular background based on mutations in the LMNA gene and genes coding for interacting proteins. HGPS is characterized by the presence of aging-associated symptoms, including lack of subcutaneous fat, alopecia, swollen veins, growth retardation, age spots, joint contractures, osteoporosis, cardiovascular pathology, and death due to heart attacks and strokes in childhood. LMNA codes for two major, alternatively spliced transcripts, give rise to lamin A and lamin C proteins. Mutations in the LMNA gene alone, depending on the nature and location, may result in the expression of abnormal protein or loss of protein expression and cause at least 11 disease phenotypes, differing in severity and affected tissue. LMNA gene-related HGPS is caused by a single mutation in the LMNA gene in exon 11. The mutation c.1824C > T results in activation of the cryptic donor splice site, which leads to the synthesis of progerin protein lacking 50 amino acids. The accumulation of progerin is the reason for appearance of the phenotype. In this review, we discuss current knowledge on the molecular mechanisms underlying the development of HGPS and provide a critical analysis of current research trends in this field. We also discuss the mouse models available so far, the current status of treatment of the disease, and future prospects for the development of efficient therapies, including gene therapy for HGPS.
Collapse
|
2
|
Morró M, Teichenne J, Jimenez V, Kratzer R, Marletta S, Maggioni L, Mallol C, Ruberte J, Kochanek S, Bosch F, Ayuso E. Pancreatic transduction by helper-dependent adenoviral vectors via intraductal delivery. Hum Gene Ther 2015; 25:824-36. [PMID: 25046147 DOI: 10.1089/hum.2013.182] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Pancreatic gene transfer could be useful to treat several diseases, such as diabetes mellitus, cystic fibrosis, chronic pancreatitis, or pancreatic cancer. Helper-dependent adenoviral vectors (HDAds) are promising tools for gene therapy because of their large cloning capacity, high levels of transgene expression, and long-term persistence in immunocompetent animals. Nevertheless, the ability of HDAds to transduce the pancreas in vivo has not been investigated yet. Here, we have generated HDAds carrying pancreas-specific expression cassettes, that is, driven either by the elastase or insulin promoter, using a novel and convenient plasmid family and homologous recombination in bacteria. These HDAds were delivered to the pancreas of immunocompetent mice via intrapancreatic duct injection. HDAds, encoding a CMV-GFP reporter cassette, were able to transduce acinar and islet cells, but transgene expression was lost 15 days postinjection in correlation with severe lymphocytic infiltration. When HDAds encoding GFP under the control of the specific elastase promoter were used, expression was detected in acinar cells, but similarly, the expression almost disappeared 30 days postinjection and lymphocytic infiltration was also observed. In contrast, long-term transgene expression (>8 months) was achieved with HDAds carrying the insulin promoter and the secretable alkaline phosphatase as the reporter gene. Notably, transduction of the liver, the preferred target for adenovirus, was minimal by this route of delivery. These data indicate that HDAds could be used for pancreatic gene therapy but that selection of the expression cassette is of critical importance to achieve long-term expression of the transgene in this tissue.
Collapse
Affiliation(s)
- Meritxell Morró
- 1 Center of Animal Biotechnology and Gene Therapy, Universitat Autònoma de Barcelona , Bellaterra 08193, Spain
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
3
|
Puttini S, van Zwieten RW, Saugy D, Lekka M, Hogger F, Ley D, Kulik AJ, Mermod N. MAR-mediated integration of plasmid vectors for in vivo gene transfer and regulation. BMC Mol Biol 2013; 14:26. [PMID: 24295286 PMCID: PMC4219123 DOI: 10.1186/1471-2199-14-26] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Accepted: 11/20/2013] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The in vivo transfer of naked plasmid DNA into organs such as muscles is commonly used to assess the expression of prophylactic or therapeutic genes in animal disease models. RESULTS In this study, we devised vectors allowing a tight regulation of transgene expression in mice from such non-viral vectors using a doxycycline-controlled network of activator and repressor proteins. Using these vectors, we demonstrate proper physiological response as consequence of the induced expression of two therapeutically relevant proteins, namely erythropoietin and utrophin. Kinetic studies showed that the induction of transgene expression was only transient, unless epigenetic regulatory elements termed Matrix Attachment Regions, or MAR, were inserted upstream of the regulated promoters. Using episomal plasmid rescue and quantitative PCR assays, we observed that similar amounts of plasmids remained in muscles after electrotransfer with or without MAR elements, but that a significant portion had integrated into the muscle fiber chromosomes. Interestingly, the MAR elements were found to promote plasmid genomic integration but to oppose silencing effects in vivo, thereby mediating long-term expression. CONCLUSIONS This study thus elucidates some of the determinants of transient or sustained expression from the use of non-viral regulated vectors in vivo.
Collapse
Affiliation(s)
- Stefania Puttini
- Institute of Biotechnology, University of Lausanne, Lausanne, Switzerland.
| | | | | | | | | | | | | | | |
Collapse
|
4
|
Robert MA, Zeng Y, Raymond B, Desfossé L, Mairey E, Tremblay JP, Massie B, Gilbert R. Efficacy and site-specificity of adenoviral vector integration mediated by the phage φC31 integrase. Hum Gene Ther Methods 2013. [PMID: 23194172 DOI: 10.1089/hgtb.2012.122] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Adenoviral vectors deleted of all their viral genes (helper-dependent [HD]) are efficient gene-transfer vehicles. Because transgene expression is rapidly lost in actively dividing cells, we investigated the feasibility of using phage φC31 integrase (φC31-Int) to integrate an HD carrying an attB site and the puromycin resistance gene into human cells (HeLa) and murine myoblasts (C2C12) by co-infection with a second HD-expressing φC31-Int. Because the HD genome is linear, we also investigated whether its circularization, through expression of Cre using a third HD, affects integration. Efficacy and specificity were determined by scoring the number of puromycin-resistant colonies and by sequencing integration sites. Unexpectedly, circularization of HD was unnecessary and it even reduced the integration efficacy. The maximum integration efficacy achieved was 0.5% in HeLa cells and 0.1% in C2C12 myoblasts. Up to 76% of the integration events occurred at pseudo attP sites and previously characterized hotspots were found. A small (two- to three-fold) increase in the number of γ-H2AX positive foci, accompanied by no noticeable change in γ-H2AX expression, indicated the low genotoxicity of φC31-Int. In conclusion, integration of HD mediated by φC31-Int is an attractive alternative to engineer cells, because it permits site-specific integration of large DNA fragments with low genotoxicity.
Collapse
Affiliation(s)
- Marc-André Robert
- Biotechnology Research Institute, National Research Council Canada, Montréal, Canada, H4P 2R2
| | | | | | | | | | | | | | | |
Collapse
|
5
|
Robert MA, Lin Y, Bendjelloul M, Zeng Y, Dessolin S, Broussau S, Larochelle N, Nalbantoglu J, Massie B, Gilbert R. Strength and muscle specificity of a compact promoter derived from the slow troponin I gene in the context of episomal (gutless adenovirus) and integrating (lentiviral) vectors. J Gene Med 2013; 14:746-60. [PMID: 23071006 DOI: 10.1002/jgm.2675] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2012] [Revised: 08/16/2012] [Accepted: 10/12/2012] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Gutless adenovirus (helper-dependent adenoviral vector; HDAd) and lentiviral vectors (LV) are attractive vectors for the gene therapy of muscle diseases. Because the organization of their DNA (episomal versus integrated) differs, we investigated whether the strength and specificity of ΔUSEx3, a novel muscle-specific promoter previously tested with plasmid, were maintained in the context of these vectors. METHODS Two HDAds expressing β-galactosidase regulated by ΔUSEx3 or CAG [cytomegalovirus (CMV) enhancer/β-actin promoter], and three LV expressing green fluorescent protein regulated by ΔUSEx3, CMV or a modified skeletal α-actin promoter (SPcΔ5-12), were constructed. Gene expression was compared in cell culture and after intravenous (HDAd only) and intramuscular injection of mice. RESULTS Irrespective of the vector used, ΔUSEx3 remained poorly active in nonmuscle cells and tissues. In myotubes, ΔUSEx3 was as strong as CMV and SPcΔ5-12, although it was ten-fold weaker than CAG, a proven powerful promoter in muscle. In cell culture, ΔUSEx3 activity in the context of LV was more stable than CMV, indicating it is less prone to silencing. In the context of HDAd, the behavior of ΔUSEx3 in skeletal muscle mirrored that of cell culture (10% of the CAG activity and half the number of transduced fibers). Surprisingly, in muscles treated with LV, ΔUSEx3 activity was five-fold lower than SPcΔ5-12. CONCLUSIONS The data obtained in the present study confirm that ΔUSEx3 is a strong and robust muscle-specific promoter in the context of HDAd (cell culture and in vivo) and LV (cell culture). However, it was less efficient in vivo in the context of LV.
Collapse
Affiliation(s)
- Marc-André Robert
- Biotechnology Research Institute, National Research Council Canada, Montreal, Québec, Canada. renald
| | | | | | | | | | | | | | | | | | | |
Collapse
|
6
|
Wang N, Lu Y, Pinard M, Pilotte A, Gilbert R, Massie B, Brodt P. Sustained production of a soluble IGF-I receptor by gutless adenovirus-transduced host cells protects from tumor growth in the liver. Cancer Gene Ther 2013; 20:229-36. [PMID: 23470563 DOI: 10.1038/cgt.2013.10] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The IGF-I receptor (IGF-IR) has an important role in malignant disease and is the target of several drugs presently in clinical trials. Gene therapy has been explored as cancer treatment, mainly for delivery of genes that induce cell death or enhance the immunological response to cancer. Previously, we have shown that the implantation of autologous bone-marrow stromal cells producing a soluble form of IGF-IR (sIGFIR) inhibited experimental liver metastasis of several tumor types in mice. Here, we evaluated the utility of adenovirus-based gene delivery for generating therapeutically effective plasma levels of this decoy. We constructed a third generation gutless adenovirus expressing sIGFIR and found that HEK-293 cells transduced by this, but not control adenoviruses, secreted soluble receptor protein that blocked IGF-I-induced tumor cell migration, proliferation and survival in vitro. Following virus injection in vivo, viral DNA was detectable by PCR in several host organs, particularly the liver, and this resulted in the production of measurable sIGFIR plasma levels for up to 21 days post injection. In mice producing virus-encoded sIGFIR, experimental liver metastasis was inhibited, indicating that sIGFIR levels were therapeutically effective. The results show that adenovirus-based delivery of inhibitory soluble proteins can provide an effective anticancer strategy.
Collapse
Affiliation(s)
- N Wang
- Department of Surgery, McGill University Health Center, Royal Victoria Hospital, Montreal, Quebec, Canada
| | | | | | | | | | | | | |
Collapse
|
7
|
Blain M, Zeng Y, Bendjelloul M, Hallauer PL, Kumar A, Hastings KE, Karpati G, Massie B, Gilbert R. Strong Muscle-Specific Regulatory Cassettes Based on Multiple Copies of the Human Slow Troponin I Gene Upstream Enhancer. Hum Gene Ther 2010; 21:127-34. [DOI: 10.1089/hum.2008.208] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Marilyne Blain
- Biotechnology Research Institute, National Research Council Canada, Montreal, Quebec H4P 2R2, Canada
- Department of Molecular Biology, University of Montreal, Montreal, Quebec H3C 3J7, Canada
| | - Yue Zeng
- Biotechnology Research Institute, National Research Council Canada, Montreal, Quebec H4P 2R2, Canada
- Neuromuscular Research Group, Montreal Neurological Institute, McGill University, Montréal, Quebec H3A 2B4, Canada
| | - Mehdi Bendjelloul
- Biotechnology Research Institute, National Research Council Canada, Montreal, Quebec H4P 2R2, Canada
- Neuromuscular Research Group, Montreal Neurological Institute, McGill University, Montréal, Quebec H3A 2B4, Canada
| | - Patricia L. Hallauer
- Neuromuscular Research Group, Montreal Neurological Institute, McGill University, Montréal, Quebec H3A 2B4, Canada
| | - Angela Kumar
- Neuromuscular Research Group, Montreal Neurological Institute, McGill University, Montréal, Quebec H3A 2B4, Canada
| | - Kenneth E.M. Hastings
- Neuromuscular Research Group, Montreal Neurological Institute, McGill University, Montréal, Quebec H3A 2B4, Canada
| | - George Karpati
- Neuromuscular Research Group, Montreal Neurological Institute, McGill University, Montréal, Quebec H3A 2B4, Canada
| | - Bernard Massie
- Biotechnology Research Institute, National Research Council Canada, Montreal, Quebec H4P 2R2, Canada
- Department of Microbiology and Immunology, University of Montreal, Montreal, Quebec H3C 3J7, Canada
| | - Rénald Gilbert
- Biotechnology Research Institute, National Research Council Canada, Montreal, Quebec H4P 2R2, Canada
- Neuromuscular Research Group, Montreal Neurological Institute, McGill University, Montréal, Quebec H3A 2B4, Canada
| |
Collapse
|
8
|
Lu Y, Tian C, Danialou G, Gilbert R, Petrof BJ, Karpati G, Nalbantoglu J. Targeting artificial transcription factors to the utrophin A promoter: effects on dystrophic pathology and muscle function. J Biol Chem 2008; 283:34720-7. [PMID: 18945675 PMCID: PMC3259868 DOI: 10.1074/jbc.m804518200] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2008] [Revised: 10/15/2008] [Indexed: 12/21/2022] Open
Abstract
Duchenne muscular dystrophy is caused by a genetic defect in the dystrophin gene. The absence of dystrophin results in muscle fiber necrosis and regeneration, leading to progressive muscle fiber loss. Utrophin is a close analogue of dystrophin. A substantial, ectopic expression of utrophin in the extrasynaptic sarcolemma of dystrophin-deficient muscle fibers can prevent deleterious effects of dystrophin deficiency. An alternative approach for the extrasynaptic up-regulation of utrophin involves the augmentation of utrophin transcription via the endogenous utrophin A promoter using custom-designed transcriptional activator proteins with zinc finger (ZFP) motifs. We tested a panel of custom-designed ZFP for their ability to activate the utrophin A promoter. Expression of one such ZFP efficiently increased, in a time-dependent manner, utrophin transcript and protein levels both in vitro and in vivo. In dystrophic mouse (mdx) muscles, administration of adenoviral vectors expressing this ZFP led to significant enhancement of muscle function with decreased necrosis, restoration of the dystrophin-associated proteins, and improved resistance to eccentric contractions. These studies provide evidence that specifically designed ZFPs can act as strong transcriptional activators of the utrophin A promoter. These may thus serve as attractive therapeutic agents for dystrophin deficiency states such as Duchenne muscular dystrophy.
Collapse
Affiliation(s)
- Yifan Lu
- Montreal Neurological Institute and
Department of Neurology & Neurosurgery, McGill University, Montreal,
Quebec H3A 2B4, the Respiratory Division, McGill
University Health Center and Meakins-Christie Laboratories, McGill University,
Montreal, Quebec H3A 1A1, and the Biotechnology
Research Institute, National Research Council Canada, Montreal, Quebec H4P
2R2, Canada
| | - Chai Tian
- Montreal Neurological Institute and
Department of Neurology & Neurosurgery, McGill University, Montreal,
Quebec H3A 2B4, the Respiratory Division, McGill
University Health Center and Meakins-Christie Laboratories, McGill University,
Montreal, Quebec H3A 1A1, and the Biotechnology
Research Institute, National Research Council Canada, Montreal, Quebec H4P
2R2, Canada
| | - Gawiyou Danialou
- Montreal Neurological Institute and
Department of Neurology & Neurosurgery, McGill University, Montreal,
Quebec H3A 2B4, the Respiratory Division, McGill
University Health Center and Meakins-Christie Laboratories, McGill University,
Montreal, Quebec H3A 1A1, and the Biotechnology
Research Institute, National Research Council Canada, Montreal, Quebec H4P
2R2, Canada
| | - Rénald Gilbert
- Montreal Neurological Institute and
Department of Neurology & Neurosurgery, McGill University, Montreal,
Quebec H3A 2B4, the Respiratory Division, McGill
University Health Center and Meakins-Christie Laboratories, McGill University,
Montreal, Quebec H3A 1A1, and the Biotechnology
Research Institute, National Research Council Canada, Montreal, Quebec H4P
2R2, Canada
| | - Basil J. Petrof
- Montreal Neurological Institute and
Department of Neurology & Neurosurgery, McGill University, Montreal,
Quebec H3A 2B4, the Respiratory Division, McGill
University Health Center and Meakins-Christie Laboratories, McGill University,
Montreal, Quebec H3A 1A1, and the Biotechnology
Research Institute, National Research Council Canada, Montreal, Quebec H4P
2R2, Canada
| | - George Karpati
- Montreal Neurological Institute and
Department of Neurology & Neurosurgery, McGill University, Montreal,
Quebec H3A 2B4, the Respiratory Division, McGill
University Health Center and Meakins-Christie Laboratories, McGill University,
Montreal, Quebec H3A 1A1, and the Biotechnology
Research Institute, National Research Council Canada, Montreal, Quebec H4P
2R2, Canada
| | - Josephine Nalbantoglu
- Montreal Neurological Institute and
Department of Neurology & Neurosurgery, McGill University, Montreal,
Quebec H3A 2B4, the Respiratory Division, McGill
University Health Center and Meakins-Christie Laboratories, McGill University,
Montreal, Quebec H3A 1A1, and the Biotechnology
Research Institute, National Research Council Canada, Montreal, Quebec H4P
2R2, Canada
| |
Collapse
|
9
|
Meneses-Acosta A, Dormond E, Jacob D, Tom R, Bernier A, Perret S, St-Laurent G, Durocher Y, Gilbert R, Kamen A. Development of a suspension serum-free helper-dependent adenovirus production system and assessment of co-infection conditions. J Virol Methods 2008; 148:106-14. [PMID: 18079009 DOI: 10.1016/j.jviromet.2007.10.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2007] [Revised: 10/20/2007] [Accepted: 10/26/2007] [Indexed: 12/14/2022]
Abstract
Helper-dependent adenovirus (HDAd), deleted in all viral protein-coding sequences has been designed to reduce immune response and favor long-term expression of therapeutic genes in clinical programs. Its production requires co-infection of E1-complementing cells with helper adenovirus (HAd). Significant progresses have been made in the molecular design of HDAd, but large scale production remains a challenge. In this work, a scalable system for HDAd production is designed and evaluated focusing on the co-infection step. A human embryo kidney 293 (293) derived cell line, the 293SF/FLPe was generated to produce efficiently HDAd while restricting the packaging of HAd. This cell line was adapted to grow in suspension and in serum-free medium. Multiplicity of infection (MOI) of HDAd ranging from 0.1 to 50 was evaluated in presence of HAd at a MOI of 5. Optimal MOIs for HDAd amplification were found in the range of 5-10. HAd contamination was only 1%. These results were validated in a 3 L bioreactor under controlled operating conditions where a higher HDAd yield of 2.6 x 10(9) viral particles (VP)/mL or 3.5 x 10(8) infectious units (IU)/mL of HDAd was obtained.
Collapse
Affiliation(s)
- Angélica Meneses-Acosta
- Animal Cell Technology Group, Biotechnology Research Institute, National Research Council, 6100 Royalmount Avenue, Montreal, Quebec, Canada H4P 2R2
| | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Kawano R, Ishizaki M, Maeda Y, Uchida Y, Kimura E, Uchino M. Transduction of full-length dystrophin to multiple skeletal muscles improves motor performance and life span in utrophin/dystrophin double knockout mice. Mol Ther 2008; 16:825-31. [PMID: 18334987 DOI: 10.1038/mt.2008.23] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is a fatal, progressive, muscle-wasting disease caused by defects in the dystrophin. No viral vector except the helper-dependent adenovirus vector (HDAdv) can package 14-kilobase (kb) full-length dystrophin complementary DNA (cDNA), and HDAdv is considerably safer than old-generation adenovirus vectors because of the large-size deletion in its genome. We have generated HDAdv that carries myc-tagged murine full-length dystrophin cDNA (HDAdv-myc-mFLdys). We injected it into multiple proximal muscles of 7-day-old utrophin/dystrophin double knockout mice (dko mice) (which typically show symptoms quite similar to human DMD) because the proximal muscles are affected in DMD patients. Eight weeks after the injections, the transduced dystrophin was widely expressed, and we found a significant reduction in centrally nucleated myofibers and the restoration of the dystrophin-associated proteins, beta-dystroglycan (beta-DG) and alpha-sarcoglycan (alpha-SG), as well as neuronal nitric oxide synthase (nNOS). The injected dko mice also showed an increase in body weight, an improvement in motor performance, and a prolongation of life span. Using HDAdv, we could treat DMD model mice even by transferring the therapeutic gene into multiple skeletal muscles. Our results suggest that multiple intramuscular administrations of HDAdv carrying full-length dystrophin cDNA may reduce symptoms and compensate for lost functions in DMD patients.
Collapse
Affiliation(s)
- Ryoko Kawano
- Department of Neurology, Kumamoto University Graduate School of Medical Science, Kumamoto, Japan
| | | | | | | | | | | |
Collapse
|
11
|
Deol JR, Danialou G, Larochelle N, Bourget M, Moon JS, Liu AB, Gilbert R, Petrof BJ, Nalbantoglu J, Karpati G. Successful compensation for dystrophin deficiency by a helper-dependent adenovirus expressing full-length utrophin. Mol Ther 2007; 15:1767-74. [PMID: 17667948 DOI: 10.1038/sj.mt.6300260] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Helper-dependent adenovirus vector (AdV)-mediated full-length dystrophin expression leads to significant mitigation of the dystrophic phenotype of the mdx mouse. However, dystrophin, as a neoantigen, elicits antibody formation. As an alternative approach, we evaluated gene transfer of full-length murine utrophin, a functional homologue of dystrophin that is normally present only at the neuromuscular junction. A single injection in the tibialis anterior (TA) muscle of the helper-dependent adenovirus vector encoding utrophin provided very good transduction, with 58% of fibers demonstrating sarcolemmal utrophin expression in the neonates, and 35% utrophin-positive (Utr(+)) fibers in adults. The presence of utrophin prevented extensive necrosis in the neonates, halted further necrosis in the adults, and led to restoration of sarcolemmal expression of dystrophin-associated proteins up to 1 year after injection. Marked physiological improvement was observed in both neonates and adults. Neither increased humoral responses nor cellular immune responses were evident. However, there was a time-related decline of the initial high utrophin expression. Although viral DNA persisted in animals that were injected in the neonatal stage, viral DNA levels decreased in muscles of adult mice. These results demonstrate that although utrophin gene transfer leads to amelioration of the dystrophic phenotype, the effects are not sustained upon loss of utrophin expression.
Collapse
Affiliation(s)
- Jatinderpal R Deol
- Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Uchida Y, Maeda Y, Kimura E, Yamashita S, Nishida Y, Arima T, Hirano T, Uyama E, Mita S, Uchino M. Effective repetitive dystrophin gene transfer into skeletal muscle of adult mdx mice using a helper-dependent adenovirus vector expressing the coxsackievirus and adenovirus receptor (CAR) and dystrophin. J Gene Med 2007; 7:1010-22. [PMID: 15756716 DOI: 10.1002/jgm.745] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND The helper-dependent adenovirus (HDAd) vector is less immunogenic and has a larger cloning capacity of up to 37 kb enough to carry the full-length dystrophin cDNA. However, high and long-term expression of dystrophin transduced to mature muscle still remains difficult. One of the main reasons for this is that the expression of the coxsackievirus and adenovirus receptor (CAR) is very low in mature muscle. METHODS We have constructed two different HDAd vectors. One contains the LacZ and the murine full-length dystrophin expression cassette (HDAdLacZ-dys), and the other is a new, improved vector containing the CAR and the dystrophin expression cassette (HDAdCAR-dys). RESULTS We initially demonstrated high dystrophin expression and prevention of the dystrophic pathology in mdx muscle injected during the neonatal phase with HDAdLacZ-dys. Furthermore, we demonstrated that repeated injections of HDAdCAR-dys into mature muscle led to approximately nine times greater dystrophin-positive fibers in number than a single injection, thereby recovering the expression of dystrophin-associated proteins. This data has also shown that HDAdCAR-dys enabled administration of adenovirus (Ad) vector to the host with pre-existing immunity to the same serotype of Ad. CONCLUSIONS Repetitive injections of the HDAd vector containing the CAR and the dystrophin expression cassette could improve the efficiency of subsequent dystrophin gene transfer to mature mdx muscle. This result suggests that our new HDAd vector will provide a novel gene therapy strategy for Duchenne muscular dystrophy, raising the prospects for gene therapy of other hereditary myopathies.
Collapse
MESH Headings
- Adenoviridae/genetics
- Animals
- COS Cells
- Chlorocebus aethiops
- Coxsackie and Adenovirus Receptor-Like Membrane Protein
- Dystrophin/deficiency
- Dystrophin/genetics
- Dystrophin/metabolism
- Gene Transfer Techniques
- Genes, Reporter
- Genetic Vectors/genetics
- Helper Viruses/genetics
- Helper Viruses/physiology
- Humans
- Immunity, Cellular/immunology
- Injections, Intramuscular
- Mice
- Mice, Inbred C57BL
- Mice, Inbred mdx
- Muscle, Skeletal/cytology
- Muscle, Skeletal/immunology
- Muscle, Skeletal/metabolism
- Muscular Dystrophy, Duchenne/pathology
- Muscular Dystrophy, Duchenne/prevention & control
- Receptors, Virus/genetics
- Receptors, Virus/metabolism
- Recombinant Fusion Proteins
Collapse
Affiliation(s)
- Yuji Uchida
- Department of Neurology, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto 860-0811, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Adriaansen J, Vervoordeldonk MJBM, Tak PP. Gene therapy as a therapeutic approach for the treatment of rheumatoid arthritis: innovative vectors and therapeutic genes. Rheumatology (Oxford) 2006; 45:656-68. [PMID: 16510530 DOI: 10.1093/rheumatology/kel047] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
In recent years, significant progress has been made in the treatment of rheumatoid arthritis (RA). In addition to conventional therapy, novel biologicals targeting tumour necrosis factor-alpha have successfully entered the clinic. However, the majority of the patients still has some actively inflamed joints and some patients suffer from side-effects associated with the high systemic dosages needed to achieve therapeutic levels in the joints. In addition, due to of the short half-life of these proteins there is a need for continuous, multiple injections of the recombinant protein. An alternative approach might be the use of gene transfer to deliver therapeutic genes locally at the site of inflammation. Several viral and non-viral vectors are being used in animal models of RA. The first gene therapy trials for RA have already entered the clinic. New vectors inducing long-term and regulated gene expression in specific tissue are under development, resulting in more efficient gene transfer, for example by using distinct serotypes of viral vectors such as adeno-associated virus. This review gives an overview of some promising vectors used in RA research. Furthermore, several therapeutic genes are discussed that could be used for gene therapy in RA patients.
Collapse
Affiliation(s)
- J Adriaansen
- Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, Amsterdam, The Netherlands
| | | | | |
Collapse
|
14
|
Gonçalves MAFV, Holkers M, Cudré-Mauroux C, van Nierop GP, Knaän-Shanzer S, van der Velde I, Valerio D, de Vries AAF. Transduction of myogenic cells by retargeted dual high-capacity hybrid viral vectors: robust dystrophin synthesis in duchenne muscular dystrophy muscle cells. Mol Ther 2006; 13:976-86. [PMID: 16443396 DOI: 10.1016/j.ymthe.2005.11.018] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2005] [Revised: 10/25/2005] [Accepted: 11/13/2005] [Indexed: 11/24/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is caused by mutations in the dystrophin gene (DMD), making it amenable to gene- or cell-based therapies. Another possible treatment entails the combination of both principles by transplantation of autologous myogenic cells after their genetic complementation. This approach requires efficient and stable transduction of these cells with recombinant DMD. Recently, we generated a dual high-capacity (hc) adenovirus (Ad)-adeno-associated virus (AAV) hybrid vector (HV) that can deliver two full-length dystrophin-encoding modules into target cells. We showed that HV transduction of human cells containing AAV Rep proteins leads to the insertion of foreign DNA into the AAVS1 locus. Here, we improved HV entry into muscle cells from DMD patients. After having verified that these cells barely express the coxsackie B virus and Ad receptor (CAR), which constitutes the attachment molecule for Ad serotype 5 (Ad5) fibers, we equipped dual hcAd/AAV HV particles with Ad serotype 50 fiber domains to achieve CAR-independent uptake. These retargeted vectors complemented much more efficiently the genetic defect of dystrophin-defective myoblasts and myotubes than their isogenic counterparts with conventional Ad5 fibers. Importantly, the accumulation of beta-dystroglycan along the membranes of vector-treated DMD myotubes indicated proper assembly of dystrophin-associated glycoprotein complexes.
Collapse
Affiliation(s)
- Manuel A F V Gonçalves
- Gene Therapy Section, Department of Molecular Cell Biology, Leiden University Medical Center, Wassenaarseweg 72, 2333 AL Leiden, The Netherlands.
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Mian A, Guenther M, Finegold M, Ng P, Rodgers J, Lee B. Toxicity and adaptive immune response to intracellular transgenes delivered by helper-dependent vs. first generation adenoviral vectors. Mol Genet Metab 2005; 84:278-88. [PMID: 15694178 DOI: 10.1016/j.ymgme.2004.11.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2004] [Revised: 11/08/2004] [Accepted: 11/09/2004] [Indexed: 11/26/2022]
Abstract
The host immune response to intracellular transgenes delivered by helper-dependent (HDV) vs. first generation (FGV) adenoviral vectors has been relatively unstudied. Previous studies showed short-term correction of bovine and murine argininosuccinate synthetase (ASS) deficiency after first generation adenoviral-mediated liver gene therapy. To determine whether the host adaptive immune response against the intracellular transgene human ASS (hASS) contributed to loss of gene expression in this setting, the same vector (FGV-CAG-hASS) was injected into Rag-/- (immunodeficient) mice. As in wild-type C57BL/6 (B6) mice, Rag-/- mice also showed significant loss of hASS expression and vector by week 4 post-injection, with concomitant elevation of liver enzymes and disruption of liver architecture. Therefore, direct toxicity due to vector rather than adaptive immune response against hASS primarily accounted for loss of expression with FGVs. In contrast to hASS, beta-galactosidase is strongly immunogenic and activates the host adaptive immune response. Loss of transgene expression was observed in B6 mice with either a FGV or a HDV expressing beta-galactosidase. However, the drop in gene expression observed with the HDV was primarily due to the adaptive immune response, since both beta-galactosidase expression and vector genome were sustained in immunodeficient mice treated with HDV. As expected, with weakly immunogenic hASS, vector genome and hASS expression were sustained with a HDV in spite of ubiquitous expression of the transgene. Therefore, viral gene expression is a primary determinant of intermediate and chronic toxicities at day 3 and week 4 post-injection. However, even in the absence of viral gene expression, strongly immunogenic intracellular transgenes can stimulate clearance of transduced hepatocytes.
Collapse
Affiliation(s)
- Asad Mian
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | | | | | | | | | | |
Collapse
|
16
|
Gonçalves MAFV, van Nierop GP, Tijssen MR, Lefesvre P, Knaän-Shanzer S, van der Velde I, van Bekkum DW, Valerio D, de Vries AAF. Transfer of the full-length dystrophin-coding sequence into muscle cells by a dual high-capacity hybrid viral vector with site-specific integration ability. J Virol 2005; 79:3146-62. [PMID: 15709034 PMCID: PMC548431 DOI: 10.1128/jvi.79.5.3146-3162.2005] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2004] [Accepted: 10/08/2004] [Indexed: 11/20/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is caused by mutations in the DMD gene, making it a potential target for gene therapy. There is, however, a scarcity of vectors that can accommodate the 14-kb DMD cDNA and permanently genetically correct muscle tissue in vivo or proliferating myogenic progenitors in vitro for use in autologous transplantation. Here, a dual high-capacity adenovirus-adeno-associated virus (hcAd/AAV) vector with two full-length human dystrophin-coding sequences flanked by AAV integration-enhancing elements is presented. These vectors are generated from input linear monomeric DNA molecules consisting of the Ad origin of replication and packaging signal followed by the recently identified AAV DNA integration efficiency element (p5IEE), the transgene(s) of interest, and the AAV inverted terminal repeat (ITR). After infection of producer cells with a helper Ad vector, the Ad DNA replication machinery, in concert with the AAV ITR-dependent dimerization, leads to the assembly of vector genomes with a tail-to-tail configuration that are efficiently amplified and packaged into Ad capsids. These dual hcAd/AAV hybrid vectors were used to express the dystrophin-coding sequence in rat cardiomyocytes in vitro and to restore dystrophin synthesis in the muscle tissues of mdx mice in vivo. Introduction into human cells of chimeric genomes, which contain a structure reminiscent of AAV proviral DNA, resulted in AAV Rep-dependent targeted DNA integration into the AAVS1 locus on chromosome 19. Dual hcAd/AAV hybrid vectors may thus be particularly useful to develop safe treatment modalities for diseases such as DMD that rely on efficient transfer and stable expression of large genes.
Collapse
Affiliation(s)
- Manuel A F V Gonçalves
- Gene Therapy Section, Department of Molecular Cell Biology, Leiden University Medical Center, Wassenaarseweg 72, 2333 AL Leiden, The Netherlands.
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Harui A, Roth MD, Kiertscher SM, Mitani K, Basak SK. Vaccination with helper-dependent adenovirus enhances the generation of transgene-specific CTL. Gene Ther 2005; 11:1617-26. [PMID: 15295617 DOI: 10.1038/sj.gt.3302332] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Recombinant adenoviral vectors (AdV) have been used experimentally as vaccines to present antigenic transgenes in vivo. However, administration of first-generation vectors (FG-AdV) is often limited by their induction of antiviral immunity. To address this limitation, helper-dependent vectors (HD-AdV) were developed that lack viral coding regions. While the administration of HD-AdV results in long-term gene expression in vivo, their utility as immunogens has never been examined. Direct vaccination with 10(8) blue-forming units (BFU) of HD-AdV injected into C57BL/6 mice lead to superior transgene-specific CTL and antibody responses when compared to the same amount of a FG-AdV. The antibody responses to viral antigens were high in response to both the vectors. As a mechanism to reduce viral exposure, dendritic cells (DC) were transduced with HD-AdV in vitro and then used as a cell-based vaccine. DC transduced with HD-AdV expressed higher levels of transgene-specific mRNA and up to 1200-fold higher levels of transgene protein than did DC transduced with a FG-AdV. In addition, HD-AdV-transduced DC stimulated superior transgene-specific CTL responses when administered in vivo, an effect that was further enhanced by maturing the DC with LPS prior to administration. In contrast to direct immunization with HD-AdV, vaccination with HD-AdV-transduced DC was associated with limited antibody responses against the AdV. We conclude that HD-AdV stimulates superior transgene-specific immune responses when compared to a FG-AdV, and that immunization with a DC-based vaccine maintains this efficacy while limiting antiviral reactivity.
Collapse
Affiliation(s)
- A Harui
- Pulmonary & Critical Care Medicine, UCLA School of Medicine, Los Angeles, CA 90095-1690, USA
| | | | | | | | | |
Collapse
|
18
|
Abstract
Helper-dependent adenoviral vectors possess a number of characteristics that make them attractive gene therapy vectors. These vectors are completely devoid of viral coding sequences and are able to mediate high-efficiency transduction in vivo to direct sustain high-level transgene expression with negligible chronic toxicity. This review focuses on advances in helper-dependent adenoviral vector technology, selected examples of in vivo studies of particular interest, and the issue of vector-mediated acute toxicity.
Collapse
Affiliation(s)
- Donna J Palmer
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | | |
Collapse
|
19
|
Louboutin JP, Wang L, Wilson JM. Gene transfer into skeletal muscle using novel AAV serotypes. J Gene Med 2005; 7:442-51. [PMID: 15517544 DOI: 10.1002/jgm.686] [Citation(s) in RCA: 95] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Skeletal muscle is an interesting target for gene delivery because of its mass and because the vectors can be delivered in a noninvasive way. Adeno-associated virus (AAV) vectors are capable of transducing skeletal muscle fibers and achieving stable and safe transgene expression. To date, most animal experiments using AAV have been based on AAV serotype 2, but some recent studies have demonstrated that AAV1 is more efficient than AAV2/2 in transducing muscle fibers. Recently, novel AAVs (AAV7 and AAV8) were isolated from rhesus macaques. METHODS We injected three different muscles (gastrocnemius, soleus, biceps femoris) of immunocompetent C57BL/6 mice with different pseudotyped AAV serotypes (AAV2/1, AAV2/2, AAV2/5, AAV2/7 and AAV2/8) and quantitatively compared the different gene transfer efficiencies. RESULTS The efficiencies of transduction in skeletal muscle with AAV2/7 and AAV2/8 were similar to AAV2/1, and higher than that seen with AAV2/2 and AAV2/5. All serotypes were able to transduce both slow and fast muscle fibers similarly at the vector titer used (1x10(11) genome copies per mouse). Despite a limited inflammatory response (slightly higher when using AAV2/2, AAV2/7 and AAV2/8 vectors than AAV2/1 and AAV2/5), transgene expression was observed throughout the length of the experiment. DISCUSSION These results show that AAV2/7 and AAV2/8 are able to transduce muscle fibers of immunocompetent mice very efficiently, offering new perspectives in gene transfer of skeletal muscle.
Collapse
Affiliation(s)
- Jean-Pierre Louboutin
- Gene Therapy Program, Department of Medicine, Division of Medical Genetics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | | |
Collapse
|
20
|
Jiang Z, Schiedner G, Gilchrist SC, Kochanek S, Clemens PR. CTLA4Ig delivered by high-capacity adenoviral vector induces stable expression of dystrophin in mdx mouse muscle. Gene Ther 2004; 11:1453-61. [PMID: 15269713 DOI: 10.1038/sj.gt.3302315] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Adenoviral (Ad) vector-mediated gene delivery of normal, full-length dystrophin to skeletal muscle provides a promising strategy for the treatment of Duchenne muscular dystrophy (DMD), an X-linked recessive, dystrophin-deficient muscle disease. Studies in animal models suggest that successful DMD gene therapy by Ad vector-mediated gene transfer would be precluded by cellular and humoral immune responses induced by vector capsid and transgene proteins. To address the immunity induced by Ad vector-mediated dystrophin gene delivery to dystrophic muscle, we developed high-capacity adenoviral (HC-Ad) vectors expressing mouse dystrophin driven by the muscle creatine kinase promoter (AdmDys) and mCTLA4Ig (AdmCTLA4Ig) individually, or together from one vector (AdmCTLA4Ig/mDys). We found stable expression of dystrophin protein in the tibialis anterior muscles of mdx mice, coinjected with AdmCTLA4Ig and AdmDys, or injected alone with AdmCTLA4Ig/mDys, whereas the expression of dystrophin protein in the control group coinjected with AdmDys and an empty vector decreased by at least 50% between 2 and 8 weeks after administration. Additionally, we observed reductions in Ad vector-induced Th1 and Th2 cytokines, Ad vector-specific cytotoxic T lymphocyte activation and neutralizing anti-Ad antibodies in both experimental groups that received a mCTLA4Ig-expressing vector as compared to the control group. This study demonstrates that the coexpression of mCTLA4Ig and dystrophin in skeletal muscle provided by HC-Ad vector-mediated gene transfer can provide stable expression of dystrophin in immunocompetent, adult mdx mouse muscle and applies a potentially powerful strategy to overcome adaptive immunity induced by Ad vector-mediated dystrophin gene delivery toward the ultimate goal of treatment for DMD.
Collapse
Affiliation(s)
- Z Jiang
- Department of Neurology, School of Medicine, University of Pittsburgh, PA 15213, USA
| | | | | | | | | |
Collapse
|
21
|
Basak SK, Kiertscher SM, Harui A, Roth MD. Modifying Adenoviral Vectors for Use as Gene-Based Cancer Vaccines. Viral Immunol 2004; 17:182-96. [PMID: 15279698 DOI: 10.1089/0882824041310603] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The past decade has produced significant advances in our understanding of antigen-presenting cells, tumor antigens, and other components of the immune response to cancer. Gene-based vaccination is emerging as one of the more promising approaches for loading dendritic cells (DC) with tumor-associated antigens. In this respect, it is proposed that adenoviral (AdV) vectors can deliver high antigen concentrations, promote effective processing and MHC expression, and stimulate potent cell-mediated immunity. While AdV vectors have performed well in pre-clinical vaccine models, their application to patient care has limitations. The in vivo administration of AdV vectors is associated with both innate and adaptive host responses that result in tissue inflammation and injury, viral neutralization, and premature clearance of AdV-transduced cells. A variety of strategies have been developed to address these limitations. The ideal vaccine would avoid vector-related immune responses, have relative specificity for transducing DC, and induce high levels of transgene expression. This review describes the range of host responses to AdV vaccines, identifies strategies to reduce viral recognition and enhance transgene antigen expression, and suggests future approaches to vector development and administration. There is every reason to believe that safer and more effective forms of AdV-based vaccines can be developed and applied to patient therapy.
Collapse
Affiliation(s)
- Saroj K Basak
- Pulmonary and Critical Care Medicine and the Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA, Los Angeles, California 90095-1690, USA
| | | | | | | |
Collapse
|
22
|
Dudley RWR, Lu Y, Gilbert R, Matecki S, Nalbantoglu J, Petrof BJ, Karpati G. Sustained Improvement of Muscle Function One Year After Full-Length Dystrophin Gene Transfer intomdxMice by a Gutted Helper-Dependent Adenoviral Vector. Hum Gene Ther 2004; 15:145-56. [PMID: 14975187 DOI: 10.1089/104303404772679959] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Dystrophin gene transfer using helper-dependent adenoviral vectors (HDAd) deleted of all viral genes is a promising option to treat muscles in Duchenne muscular dystrophy (DMD). Previously, we reported high-level dystrophin expression and functional correction of dystrophin-deficient (mdx) mouse muscle 60 days after gene transfer with an HDAd encoding two full-length murine dystrophin cDNAs (referred to as HDCBDysM). In the present study, we tested the long-term efficacy of HDCBDysM by examining muscle contractility parameters and the stability of dystrophin expression 1 year after injection into neonatal mdx muscles. At this point, HDCBDysM-treated muscles averaged 52% dystrophin-positive fibers. Treated muscles also displayed significantly greater isometric force production as well as greater resistance to the force deficits and damage caused by eccentric contractions. The level of protection against eccentric contraction-induced force deficits correlated with the percentage of dystrophin-positive fibers. Furthermore, HDCBDysM treatment restored the dystrophin-glycoprotein complex (DGC) to the sarcolemma and improved other aspects of mdx muscle histopathology examined (central nucleation, muscle hypertrophy, and mononuclear [phagocytic] cell infiltration). These improvements occurred despite the induction of a humoral response against murine dystrophin. Our results indicate that major therapeutic benefits of HDCBDysM are maintained for a long period of the animals' lifespan and suggest that HDCBDys holds promise for treating DMD by gene therapy.
Collapse
Affiliation(s)
- Roy W R Dudley
- Respiratory Division, McGill University Health Center and Meakins-Christie Laboratories, McGill University, Montréal, Québec, Canada, H3A 1A1
| | | | | | | | | | | | | |
Collapse
|
23
|
Kobinger GP, Louboutin JP, Barton ER, Sweeney HL, Wilson JM. Correction of the Dystrophic Phenotype byIn VivoTargeting of Muscle Progenitor Cells. Hum Gene Ther 2003; 14:1441-9. [PMID: 14577924 DOI: 10.1089/104303403769211655] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Successful gene therapy for most inherited diseases will require stable expression of the therapeutic gene. This can be addressed with integrating or self-replicating viruses by targeting postmitotic cells that have a long lifetime or stem cells that can replenish defective tissue with corrected cells. In this study, we explore the possibility of targeting a muscle stem cell population in situ through in vivo administration of vector. To develop this concept, we selected a mouse model of muscular dystrophy (mdx mice) that undergoes rapid turnover of muscle fibers. In vivo targeting of muscle progenitor cells, notably satellite cells, with a pseudotyped lentiviral vector encoding the minidystrophin restores dystrophin expression and provides functional correction in skeletal muscle of mdx mice. This study shows that progenitor cells can be genetically engineered in vivo and subsequently proliferate into terminally differentiated tissue carrying the genetic graft in a way that stably corrects function.
Collapse
Affiliation(s)
- Gary P Kobinger
- Gene Therapy Program, Division of Medical Genetics, University of Pennsylvania Health System, Philadelphia, Pennsylvania 19104, USA
| | | | | | | | | |
Collapse
|