1
|
AAV2-mediated follistatin overexpression induces ovine primary myoblasts proliferation. BMC Biotechnol 2014; 14:87. [PMID: 25330993 PMCID: PMC4210504 DOI: 10.1186/s12896-014-0087-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Accepted: 10/06/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Follistatin (FST) has been shown to bind to some TGF-β family members and can function as a potent myostatin (MSTN) antagonist. Recent studies have revealed that over-expression of FST by adeno-associated viruses increases muscle growth in mice, humans and nonhuman primates. In the present study, to determine the effect of FST on ovine primary myoblast (OPM) proliferation, FST was over-expressed using an adeno-associated virus serotype 2 (AAV 2) vector. RESULTS Western blot results showed that AAV induced the expression of FST protein in transduced OPM cells. Real-time quantitative PCR results indicated that over-expression of FST resulted in a dramatic increase in Akt I and CDK2 expression and a decrease in p21 expression. Moreover, cell cycle analysis confirmed that FST down-regulated p21, a CDK inhibitor, and increased the level of CDK2 expression in OPM cells. Hence, follistatin positively regulated the G1 to S progression. Our results showed that FST induced proliferation through a down-regulation of p21, as only the p21 expression level was down-regulated as a result of FST over-expression in myoblasts, whereas no change was observed in the level of p57 expression. CONCLUSIONS These results expanded our understanding of the regulation mechanism of FST in ovine primary myoblasts. Our results provide the first evidence that the AAV viral system can be used for gene transfer in ovine myoblast cells. Moreover, the results showed that an AAV vector can successfully induce the expression of FST in OPM cells in vitro. These findings demonstrated that FST over-expression induces proliferation through a down-regulation of the p21 gene under proliferating conditions.
Collapse
|
2
|
Gao K, Li M, Zhong L, Su Q, Li J, Li S, He R, Zhang Y, Hendricks G, Wang J, Gao G. Empty Virions In AAV8 Vector Preparations Reduce Transduction Efficiency And May Cause Total Viral Particle Dose-Limiting Side-Effects. Mol Ther Methods Clin Dev 2014; 1:20139. [PMID: 25485285 PMCID: PMC4255953 DOI: 10.1038/mtm.2013.9] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Accepted: 11/12/2013] [Indexed: 02/05/2023]
Abstract
Empty virions are inadvertent by-products of recombinant adeno-associated virus (rAAV) packaging process, resulting in vector lots with mixtures of full and empty virions at variable ratios. Impact of empty virions on the efficiency and side-effects of rAAV transduction has not been well characterized. Here, we generated partially and completely empty AAV8 virions, fully packaged rAAV8 lots as well as mixtures of empty and fully packaged virions with variable ratios of empty virions (REVs). The aforementioned dosing formulations of rAAV8 expressing either cellular (EGFP or nuclear-targeted (n) LacZ) or secreted (human α1-antitrypsin, hA1AT) reporter genes were intravenously injected into two different mouse strains, followed by analyses of transgene expressions and serum alanine aminotransferase (ALT) levels at different time points. We found that addition of empty particles to the fixed doses of rAAV8 preparations repressed liver transduction up to 64% (serum hA1AT) and 44% (nLacZ) in C57BL/6 mice, respectively. The similar trend in inhibiting EGFP expression together with concurrent elevations of serum ATL levels were observed in the BALB/c mice, indicating that empty particles may also exacerbate side-effects of rAAV8EGFP transduction. Our results suggest that removal of empty particles from rAAV preparations may improve efficacy and safety of AAV in clinical applications.
Collapse
Affiliation(s)
- Kai Gao
- Gene Therapy Center, University of Massachusetts Medical School, Worcester, Massachusetts, USA
- National Institutes for Food and Drug Control, Beijing, China
| | - Mengxin Li
- Gene Therapy Center, University of Massachusetts Medical School, Worcester, Massachusetts, USA
- Viral Vector Core, University of Massachusetts Medical School, Worcester, Massachusetts, USA
- Department of Microbiology and Physiology Systems, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Li Zhong
- Gene Therapy Center, University of Massachusetts Medical School, Worcester, Massachusetts, USA
- Viral Vector Core, University of Massachusetts Medical School, Worcester, Massachusetts, USA
- Division of Hematology/Oncology, Department of Pediatrics, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Qin Su
- Gene Therapy Center, University of Massachusetts Medical School, Worcester, Massachusetts, USA
- Viral Vector Core, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Jia Li
- Gene Therapy Center, University of Massachusetts Medical School, Worcester, Massachusetts, USA
- Department of Microbiology and Physiology Systems, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Shaoyong Li
- Gene Therapy Center, University of Massachusetts Medical School, Worcester, Massachusetts, USA
- Department of Microbiology and Physiology Systems, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Ran He
- Gene Therapy Center, University of Massachusetts Medical School, Worcester, Massachusetts, USA
- Viral Vector Core, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Yu Zhang
- Gene Therapy Center, University of Massachusetts Medical School, Worcester, Massachusetts, USA
- Department of Oncology, Guizhou People’s Hospital, Guiyang, Guizhou, China
| | - Gregory Hendricks
- Department of Cell and Developmental Biology, Electron Microscopy Core Facility, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Junzhi Wang
- National Institutes for Food and Drug Control, Beijing, China
| | - Guangping Gao
- Gene Therapy Center, University of Massachusetts Medical School, Worcester, Massachusetts, USA
- Viral Vector Core, University of Massachusetts Medical School, Worcester, Massachusetts, USA
- Department of Microbiology and Physiology Systems, University of Massachusetts Medical School, Worcester, Massachusetts, USA
- Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
3
|
Song L, Kauss MA, Kopin E, Chandra M, Ul-Hasan T, Miller E, Jayandharan GR, Rivers AE, Aslanidi GV, Ling C, Li B, Ma W, Li X, Andino LM, Zhong L, Tarantal AF, Yoder MC, Wong KK, Tan M, Chatterjee S, Srivastava A. Optimizing the transduction efficiency of capsid-modified AAV6 serotype vectors in primary human hematopoietic stem cells in vitro and in a xenograft mouse model in vivo. Cytotherapy 2013; 15:986-98. [PMID: 23830234 PMCID: PMC3711144 DOI: 10.1016/j.jcyt.2013.04.003] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2012] [Revised: 04/01/2013] [Accepted: 04/02/2013] [Indexed: 12/18/2022]
Abstract
BACKGROUND AIMS Although recombinant adeno-associated virus serotype 2 (AAV2) vectors have gained attention because of their safety and efficacy in numerous phase I/II clinical trials, their transduction efficiency in hematopoietic stem cells (HSCs) has been reported to be low. Only a few additional AAV serotype vectors have been evaluated, and comparative analyses of their transduction efficiency in HSCs from different species have not been performed. METHODS We evaluated the transduction efficiency of all available AAV serotype vectors (AAV1 through AAV10) in primary mouse, cynomolgus monkey and human HSCs. The transduction efficiency of the optimized AAV vectors was also evaluated in human HSCs in a murine xenograft model in vivo. RESULTS We observed that although there are only six amino acid differences between AAV1 and AAV6, AAV1, but not AAV6, transduced mouse HSCs well, whereas AAV6, but not AAV1, transduced human HSCs well. None of the 10 serotypes transduced cynomolgus monkey HSCs in vitro. We also evaluated the transduction efficiency of AAV6 vectors containing mutations in surface-exposed tyrosine residues. We observed that tyrosine (Y) to phenylalanine (F) point mutations in residues 445, 705 and 731 led to a significant increase in transgene expression in human HSCs in vitro and in a mouse xenograft model in vivo. CONCLUSIONS These studies suggest that the tyrosine-mutant AAV6 serotype vectors are the most promising vectors for transducing human HSCs and that it is possible to increase further the transduction efficiency of these vectors for their potential use in HSC-based gene therapy in humans.
Collapse
Affiliation(s)
- Liujiang Song
- Experimental Hematology Laboratory, Department of Physiology, School of Basic Medical Sciences, Central South University, Changsha 410078, China
- Division of Cellular and Molecular Therapy, Department of Pediatrics, City of Hope Medical Center, Duarte, CA, USA
- Powell Gene Therapy Center, University of Florida College of Medicine, Gainesville, FL 32611, USA
- Genetics Institute, University of Florida College of Medicine, Gainesville, FL 32611, USA
- Shenzhen Institute of Xiangya Biomedicine, Shenzhen 518057, China
| | - M. Ariel Kauss
- Department of Virology, Beckman Research Institute of the City of Hope, Duarte, CA 91010, USA
| | - Etana Kopin
- Department of Virology, Beckman Research Institute of the City of Hope, Duarte, CA 91010, USA
| | - Manasa Chandra
- Department of Virology, Beckman Research Institute of the City of Hope, Duarte, CA 91010, USA
| | - Taihra Ul-Hasan
- Department of Virology, Beckman Research Institute of the City of Hope, Duarte, CA 91010, USA
| | - Erin Miller
- Department of Virology, Beckman Research Institute of the City of Hope, Duarte, CA 91010, USA
| | - Giridhara R. Jayandharan
- Division of Cellular and Molecular Therapy, Department of Pediatrics, City of Hope Medical Center, Duarte, CA, USA
- Powell Gene Therapy Center, University of Florida College of Medicine, Gainesville, FL 32611, USA
- Genetics Institute, University of Florida College of Medicine, Gainesville, FL 32611, USA
- Department of Haematology and Centre for Stem Cell Research, Christian Medical College, Vellore, Tamil Nadu, India
| | - Angela E. Rivers
- Division of Cellular and Molecular Therapy, Department of Pediatrics, City of Hope Medical Center, Duarte, CA, USA
- Powell Gene Therapy Center, University of Florida College of Medicine, Gainesville, FL 32611, USA
- Genetics Institute, University of Florida College of Medicine, Gainesville, FL 32611, USA
- Division of Hematology/Oncology, Department of Pediatrics, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - George V. Aslanidi
- Division of Cellular and Molecular Therapy, Department of Pediatrics, City of Hope Medical Center, Duarte, CA, USA
- Powell Gene Therapy Center, University of Florida College of Medicine, Gainesville, FL 32611, USA
- Genetics Institute, University of Florida College of Medicine, Gainesville, FL 32611, USA
| | - Chen Ling
- Division of Cellular and Molecular Therapy, Department of Pediatrics, City of Hope Medical Center, Duarte, CA, USA
- Powell Gene Therapy Center, University of Florida College of Medicine, Gainesville, FL 32611, USA
- Genetics Institute, University of Florida College of Medicine, Gainesville, FL 32611, USA
| | - Baozheng Li
- Division of Cellular and Molecular Therapy, Department of Pediatrics, City of Hope Medical Center, Duarte, CA, USA
- Powell Gene Therapy Center, University of Florida College of Medicine, Gainesville, FL 32611, USA
- Genetics Institute, University of Florida College of Medicine, Gainesville, FL 32611, USA
| | - Wenqin Ma
- Division of Cellular and Molecular Therapy, Department of Pediatrics, City of Hope Medical Center, Duarte, CA, USA
- Powell Gene Therapy Center, University of Florida College of Medicine, Gainesville, FL 32611, USA
- Genetics Institute, University of Florida College of Medicine, Gainesville, FL 32611, USA
| | - Xiaomiao Li
- Division of Cellular and Molecular Therapy, Department of Pediatrics, City of Hope Medical Center, Duarte, CA, USA
- Powell Gene Therapy Center, University of Florida College of Medicine, Gainesville, FL 32611, USA
- Genetics Institute, University of Florida College of Medicine, Gainesville, FL 32611, USA
| | - Lourdes M. Andino
- Division of Cellular and Molecular Therapy, Department of Pediatrics, City of Hope Medical Center, Duarte, CA, USA
- Powell Gene Therapy Center, University of Florida College of Medicine, Gainesville, FL 32611, USA
- Genetics Institute, University of Florida College of Medicine, Gainesville, FL 32611, USA
- Department of Pharmacology and Therapeutics, University of Florida College of Medicine, Gainesville, FL 32611, USA
| | - Li Zhong
- Division of Cellular and Molecular Therapy, Department of Pediatrics, City of Hope Medical Center, Duarte, CA, USA
- Powell Gene Therapy Center, University of Florida College of Medicine, Gainesville, FL 32611, USA
- Genetics Institute, University of Florida College of Medicine, Gainesville, FL 32611, USA
- Gene Therapy Center and Department of Pediatrics, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Alice F. Tarantal
- Center for Fetal Monkey Gene Transfer for Heart, Lung, and Blood Diseases, California National Primate Research Center, and Departments of Pediatrics and Cell Biology and Human Anatomy, University of California, Davis, CA 95616, USA
| | - Mervin C. Yoder
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Kamehameha K. Wong
- Department of Virology, Beckman Research Institute of the City of Hope, Duarte, CA 91010, USA
- Division of Hematology/Stem Cell Transplantation, City of Hope Medical Center, Duarte, CA, USA
| | - Mengqun Tan
- Experimental Hematology Laboratory, Department of Physiology, School of Basic Medical Sciences, Central South University, Changsha 410078, China
- Shenzhen Institute of Xiangya Biomedicine, Shenzhen 518057, China
| | - Saswati Chatterjee
- Department of Virology, Beckman Research Institute of the City of Hope, Duarte, CA 91010, USA
| | - Arun Srivastava
- Division of Cellular and Molecular Therapy, Department of Pediatrics, City of Hope Medical Center, Duarte, CA, USA
- Powell Gene Therapy Center, University of Florida College of Medicine, Gainesville, FL 32611, USA
- Genetics Institute, University of Florida College of Medicine, Gainesville, FL 32611, USA
- Department of Molecular Genetics and Microbiology, University of Florida College of Medicine, Gainesville, FL 32611, USA
- Shands Cancer Center, University of Florida College of Medicine, Gainesville, FL 32611, USA
| |
Collapse
|
4
|
Zhong L, Jayandharan GR, Aslanidi GV, Zolotukhin S, Herzog RW, Srivastava A. Development of Novel Recombinant AAV Vectors and Strategies for the Potential Gene Therapy of Hemophilia. ACTA ACUST UNITED AC 2012; S1. [PMID: 23264889 DOI: 10.4172/2157-7412.s1-008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Recombinant vectors based on a non-pathogenic human parvovirus, the adeno-associated virus (AAV), have gained attention as a potentially safe and useful alternative to the more commonly used retroviral and adenoviral vectors. AAV vectors are currently in use in Phase I/II clinical trials for gene therapy of a number of diseases such as cystic fibrosis, α-1 antitrypsin deficiency, muscular dystrophy, Batten's disease, and Parkinson's disease, and have shown efficacy in patients with Leber's congenital amaurosis, and hemophilia B. For patients with hemophilia B, however, relatively large vector doses are needed to achieve therapeutic benefits. Large vector doses also trigger an immune response as significant fraction of the vectors fails to traffic efficiently to the nucleus, and is targeted for degradation by the host cell proteasome machinery. With a better understanding of the various steps in the life cycle of AAV vectors, strategies leading to the development of novel AAV vectors that are capable of high-efficiency transduction at lower doses are needed. In this review, we summarize our strategies to develop novel AAV vectors for the potential gene therapy of both hemophilia B and hemophilia A, based on our recent studies on the basic molecular biology of AAV. These strategies, including the development of novel AAV vectors by site-directed mutagenesis of critical surface-exposed tyrosine residues on AAV2 capsids to circumvent the ubiquitination step and the use of different AAV serotypes and self-complementary (sc) AAV2 vectors, and their use as helper vectors to circumvent the obstacles of second-strand DNA synthesis of single-stranded (ss) AAV, should dramatically accelerate the progress towards the potential gene therapy of both hemophilia A and hemophilia B.
Collapse
Affiliation(s)
- Li Zhong
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL, USA ; Division of Hematology/Oncology, Department of Medicine, and Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, USA
| | | | | | | | | | | |
Collapse
|
5
|
Abstract
The cytopathic effects induced during parvovirus infection have been widely documented. Parvovirus infection-induced cell death is often directly associated with disease outcomes (e.g., anemia resulting from loss of erythroid progenitors during parvovirus B19 infection). Apoptosis is the major form of cell death induced by parvovirus infection. However, nonapoptotic cell death, namely necrosis, has also been reported during infection of the minute virus of mice, parvovirus H-1 and bovine parvovirus. Recent studies have revealed multiple mechanisms underlying the cell death during parvovirus infection. These mechanisms vary in different parvoviruses, although the large nonstructural protein (NS)1 and the small NS proteins (e.g., the 11 kDa of parvovirus B19), as well as replication of the viral genome, are responsible for causing infection-induced cell death. Cell cycle arrest is also common, and contributes to the cytopathic effects induced during parvovirus infection. While viral NS proteins have been indicated to induce cell cycle arrest, increasing evidence suggests that a cellular DNA damage response triggered by an invading single-stranded parvoviral genome is the major inducer of cell cycle arrest in parvovirus-infected cells. Apparently, in response to infection, cell death and cell cycle arrest of parvovirus-infected cells are beneficial to the viral cell lifecycle (e.g., viral DNA replication and virus egress). In this article, we will discuss recent advances in the understanding of the mechanisms underlying parvovirus infection-induced cell death and cell cycle arrest.
Collapse
Affiliation(s)
- Aaron Yun Chen
- Department of Microbiology, Molecular Genetics & Immunology, University of Kansas Medical Center, Mail Stop 3029, 3901 Rainbow Blvd, Kansas City, KS 66160, USA
| | | |
Collapse
|
6
|
Honaramooz A, Megee S, Zeng W, Destrempes MM, Overton SA, Luo J, Galantino-Homer H, Modelski M, Chen F, Blash S, Melican DT, Gavin WG, Ayres S, Yang F, Wang PJ, Echelard Y, Dobrinski I. Adeno‐associated virus (AAV)‐mediated transduction of male germ line stem cells results in transgene transmission after germ cell transplantation. FASEB J 2007; 22:374-82. [PMID: 17873102 DOI: 10.1096/fj.07-8935com] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
We explored whether exposure of mammalian germ line stem cells to adeno-associated virus (AAV), a gene therapy vector, would lead to stable transduction and transgene transmission. Mouse germ cells harvested from experimentally induced cryptorchid donor testes were exposed in vitro to AAV vectors carrying a GFP transgene and transplanted to germ cell-depleted syngeneic recipient testes, resulting in colonization of the recipient testes by transgenic donor cells. Mating of recipient males to wild-type females yielded 10% transgenic offspring. To broaden the approach to nonrodent species, AAV-transduced germ cells from goats were transplanted to recipient males in which endogenous germ cells had been depleted by fractionated testicular irradiation. Transgenic germ cells colonized recipient testes and produced transgenic sperm. When semen was used for in vitro fertilization (IVF), 10% of embryos were transgenic. Here, we report for the first time that AAV-mediated transduction of mammalian germ cells leads to transmission of the transgene through the male germ line. Equally important, this is also the first report of transgenesis via germ cell transplantation in a nonrodent species, a promising approach to generate transgenic large animal models for biomedical research.
Collapse
Affiliation(s)
- Ali Honaramooz
- Center for Animal Transgenesis and Germ Cell Research, University of Pennsylvania, 382 W. State Road, Kennett Square, PA 19348, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Yan Z, Lei-Butters DCM, Liu X, Zhang Y, Zhang L, Luo M, Zak R, Engelhardt JF. Unique biologic properties of recombinant AAV1 transduction in polarized human airway epithelia. J Biol Chem 2006; 281:29684-92. [PMID: 16899463 PMCID: PMC1712671 DOI: 10.1074/jbc.m604099200] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
The choice of adeno-associated virus serotypes for clinical applications is influenced by the animal model and model system used to evaluate various serotypes. In the present study, we sought to compare the biologic properties of rAAV2/1, rAAV2/2, and rAAV2/5 transduction in polarized human airway epithelia using viruses purified by a newly developed common column chromatography method. Results demonstrated that apical transduction of human airway epithelia with rAAV2/1 was 100-fold more efficient than rAAV2/2 and rAAV2/5. This transduction profile in human airway epithelia (rAAV2/1 >> rAAV2/2 = rAAV2/5) was significantly different from that seen following nasal administration of these vectors to mouse lung (rAAV2/5 > rAAV2/1 >> rAAV2/2), emphasizing differences in transduction of these serotypes between these two species. In stark contrast to rAAV2/2 and rAAV2/5, rAAV2/1 transduced both the apical and basolateral membrane of human airway epithelia with similar efficiency. However, the overall level of transduction across serotypes did not correlate with vector internalization. We hypothesized that differences in post-entry processing of these serotypes might influence the efficiency of apical transduction. To this end, we tested the effectiveness of proteasome inhibitors to augment nuclear translocation and gene expression from the three serotypes. Augmentation of rAAV2/1 apical transduction of human polarized airway epithelia was 10-fold lower than that for rAAV2/2 and rAAV2/5. Cellular fractionation studies demonstrated that proteasome inhibitors more significantly enhanced rAAV2/2 and rAAV2/5 translocation to the nucleus than rAAV2/1. These results demonstrate that AAV1 transduction biology in human airway epithelia differs from that of AAV2 and AAV5 by virtue of altered ubiquitin/proteasome sensitivities that influence nuclear translocation.
Collapse
Affiliation(s)
- Ziying Yan
- Department of Anatomy and Cell Biology, University of Iowa College of Medicine, Iowa City, Iowa 52242, USA
| | | | | | | | | | | | | | | |
Collapse
|
8
|
Fleurence E, Riviere C, Lacaze-Masmonteil T, Franco-Motoya ML, Waszak P, Bourbon J, Danos O, Douar AM, Delacourt C. Comparative Efficacy of Intratracheal Adeno-Associated Virus Administration to Newborn Rats. Hum Gene Ther 2005; 16:1298-306. [PMID: 16259563 DOI: 10.1089/hum.2005.16.1298] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Transient local overexpression of genes that promote lung defense or repair may help to protect or promote alveolar development in premature neonates. We showed that the use of adenoviral vectors in neonates was limited by the induction of lung growth disorders. In the present work we compare the efficiency of gene transfer to the neonatal lung by three adeno-associated viral vectors: rAAV1, rAAV2, and rAAV5. Transduction efficiency was first measured in vitro, by infecting A549 immortalized human lung epithelial cells, and primary epithelial and mesenchymal cells isolated from human fetal lung. AAV vectors yielded similar low levels of luciferase gene expression in the different cell types. In vivo transduction efficiency was evaluated in newborn rats, with AAV-LacZ vectors being intratracheally instilled at 3 days of age. Both rAAV5 and rAAV1, but not rAAV2, induced significant lung beta-galactosidase expression, which persisted on day 35. Highest beta- galactosidase levels were measured with rAAV5, but remained far lower than those obtained with adenoviral vectors. A transient increase in alveolar macrophages was observed on day 6, but not on day 8, after rAAV5-LacZ instillation. Morphometric evaluation of lung structures was performed on day 21, and showed no altered lung growth. We conclude that rAAV1 or rAAV5 was more efficient at mediating gene transfer in the neonatal lung than was rAAV2, without adversely affecting lung development. However, in vivo transgene expression was relatively low, and needs to be improved for future therapeutic use of these adeno-associated vectors.
Collapse
|
9
|
Fleurence E, Riviere C, Lacaze-Masmonteil T, Franco-Motoya ML, Waszak P, Bourbon J, Danos O, Douar AM, Delacourt C. Comparative Efficacy of Intratracheal Adeno-Associated Virus Administration to Newborn Rats. Hum Gene Ther 2005. [DOI: 10.1089/hum.2005.16.ft-136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
10
|
Flotte TR, Berns KI. Adeno-associated virus: a ubiquitous commensal of mammals. Hum Gene Ther 2005; 16:401-7. [PMID: 15871671 DOI: 10.1089/hum.2005.16.401] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Affiliation(s)
- Terence R Flotte
- Department of Pediatrics, and Genetics Institute, University of Florida, Gainesville, FL 32610, USA.
| | | |
Collapse
|
11
|
Kahl CA, Pollok K, Haneline LS, Cornetta K. Lentiviral vectors pseudotyped with glycoproteins from Ross River and vesicular stomatitis viruses: variable transduction related to cell type and culture conditions. Mol Ther 2005; 11:470-82. [PMID: 15727944 DOI: 10.1016/j.ymthe.2004.08.032] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2004] [Revised: 08/13/2004] [Accepted: 08/13/2004] [Indexed: 10/25/2022] Open
Abstract
HIV-1-derived lentiviral vectors have been pseudotyped with various envelope glycoproteins to alter their host range. Previously, we found that envelope glycoproteins derived from the alphavirus Ross River virus (RRV) can pseudotype lentiviral vectors and mediate efficient transduction of a variety of epithelial and fibroblast-derived cell lines. In this study, we have investigated transduction of hematopoietic cells using RRV-pseudotyped vectors encoding the enhanced green fluorescent protein (EGFP). RRV-mediated transduction of human CD34+ cord blood cells and progenitors was very inefficient, even at multiplicities of infection of 100 (0.4% EGFP-positive progenitor colonies). Inefficient transduction was also observed in a variety of hematopoietic cell lines. However, two erythroleukemia-derived cell lines and monocytic cells that were driven to macrophage-like differentiation were moderately transduced. Transduction of hematopoietic cells with a control VSV-G-pseudotyped lentiviral vector was generally efficient, but unexpectedly decreased up to threefold upon stimulation of lymphocytic cell lines or primary murine bone marrow cells. Also, the tested hematopoietic cell lines were essentially nonpermissive for adeno-associated type 2 (AAV) vectors, and this was not affected by lineage, activity, or differentiation. Treatment of permissive 293 cells with proteases revealed that transduction with both the RRV- and the VSV-G-pseudotyped vectors in part depends on the presence of cell surface proteins. These results show a severely restricted ability of RRV glycoproteins to mediate transduction in hematopoietic cells that is likely due to specific receptor requirements that differ from those of VSV-G and AAV. Conversely, transduction with the VSV glycoprotein is affected by cellular activation more than widely believed. Our findings suggest that the envelope glycoproteins and culture conditions employed need to be carefully evaluated for each application. Furthermore, the uniquely restricted host range of RRV-pseudotyped vectors may aid in the design of novel cell-selective transduction strategies.
Collapse
Affiliation(s)
- Christoph A Kahl
- Department of Medical and Molecular Genetics, Indianapolis, IN 46202, USA
| | | | | | | |
Collapse
|
12
|
Yotnda P, Zompeta C, Heslop HE, Andreeff M, Brenner MK, Marini F. Comparison of the efficiency of transduction of leukemic cells by fiber-modified adenoviruses. Hum Gene Ther 2005; 15:1229-42. [PMID: 15684699 DOI: 10.1089/hum.2004.15.1229] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Efficient gene transfer with adenoviral type 5 (Ad5) vectors depends on the initial attachment of their fiber, which binds the coxsackie-adenovirus receptor (CAR), and their subsequent internalization, mediated by the interaction of viral penton base with target cell alphav integrins. We previously demonstrated that human leukemic cells lack these receptors and are therefore resistant to Ad5 transduction, limiting efforts to genetically modify these cells. Human leukemic blasts are, however, susceptible to transduction with an adenovector made CAR independent by substitution of a chimeric Ad5/35 fiber [Yotnda et al. (2001). Gene Ther. 8, 930-937]. Other receptors can also be targeted with recombinant ligand moieties incorporated into adenovirus fiber. We have determined which of these fiber-modified adenovectors is most effective at modifying human primary leukemia cells, and lines. We used a replication-incompetent Ad5-beta-gal vector, in which the Ad5 fiber was replaced with fiber from various adenovirus serotypes (Ad35 and Ad11), or modified either with variable length polylysine (K4, K7, K21) or RGD-4C peptide. All the modified fiber vectors transduced primary leukemia cells and cell lines more efficiently than Ad5. Polylysine-substituted Ad5F/K21 and peptide-modified Ad5F/RGD vectors were most effective overall (up to 100% efficiency), whereas Ad5F/RGD was the most effective at transducing B cell acute lymphoblastic leukemia cells (90% efficiency). Ad5F/K21 and Ad5F/RGD should be of value for the genetic modification of human primary leukemia cells.
Collapse
Affiliation(s)
- P Yotnda
- Center for Cell and Gene Therapy, Baylor College of Medicine, Methodist Hospital, and Texas Children's Hospital, Houston, TX 77030, USA.
| | | | | | | | | | | |
Collapse
|
13
|
Yotnda P, Zompeta C, Heslop H, Andreeff M, Brenner M, Marini F. Comparison of the Efficiency of Transduction of Leukemic Cells by Fiber-Modified Adenoviruses. Hum Gene Ther 2004. [DOI: 10.1089/hum.2004.15.ft-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
14
|
Song S, Witek RP, Lu Y, Choi YK, Zheng D, Jorgensen M, Li C, Flotte TR, Petersen BE. Ex vivo transduced liver progenitor cells as a platform for gene therapy in mice. Hepatology 2004; 40:918-24. [PMID: 15382177 DOI: 10.1002/hep.20404] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/07/2022]
Abstract
Allogeneic stem cell-based transplants may be limited by allograft rejection, as is seen with conventional organ transplantation. One way to avert such a response is to use autologous stem cells, but that may carry the risk of recurrence of the original disease, particularly in the context of a genetic defect. We investigated the potential for gene modification of autologous stem cells to avoid both problems, using recombinant adenoassociated virus vector expressing human alpha1-antitrypsin in murine liver progenitor cells. We showed that recombinant adenoassociated virus 1 was the most efficient vector for liver progenitor cell transduction among five different serotypes of recombinant adenoassociated virus vectors. Ex vivo infected green fluorescent protein-positive liver progenitor cells from C57BL/6 mice with recombinant adenoassociated virus 1-vector-expressing human alpha1 antitrypsin were transplanted into the liver of monocrotaline-treated and partial-hepatectomized C57BL/6 recipients. Using green fluorescent protein as a donor marker, we were able to determine that at 18 weeks after transplantation, approximately 40% to 50% of the regenerated liver was green fluorescent protein positive. In addition, transgene expression (serum human alpha1-antitrypsin) was sustained for the length of the study (18 weeks after transplantation). Immunostaining revealed approximately 5% to 10% of repopulating liver cells expressing human alpha1-antitrypsin. In conclusion, this study demonstrated the feasibility of long-term engraftment and stability of transgene expression from genetically modified liver progenitor cells with a recombinant adenoassociated virus vector and implies a novel approach to gene therapy for treatment of liver diseases, such as alpha1-antitrypsin deficiency.
Collapse
Affiliation(s)
- Sihong Song
- Department of Pharmaceutics, University of Florida, Gainesville, FL 32610, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Abstract
Over the past two decades, the ability to transfer genes into hematopoietic stem cells (HSCs) has provided new insights into the behavior of individual stem cells and offered a novel approach for the treatment of various inherited or acquired disorders. At present, gene transfer into HSCs has been achieved mainly using modified retroviruses. While retrovirus-based vectors could efficiently transduce murine HSCs, extrapolation of these methods to large mammals and human clinical trials resulted in very low numbers of gene-marked engrafted cells. In addition, in vitro progenitor assays used to optimize gene transfer procedures were found to poorly predict the outcome of stem cell gene transfer. The focus rapidly turned to the development of superior and more relevant preclinical assays in human stem cell gene transfer research. Xenogeneic transplant models and large animal transplantation system have been invaluable. The development of better assays for evaluating human gene therapy protocols and a better understanding of stem cell and vector biology has culminated over the past decade in multiple strategies to improve gene transfer efficiency into HSCs. Improved gene transfer vectors, optimization of cytokine combination, and incorporation of a recombinant fragment of fibronectin during transduction are examples of novel successful additions to the early gene transfer protocols that have contributed to the first unequivocal clinical benefits resulting from genetic manipulation of HSC.
Collapse
Affiliation(s)
- André Larochelle
- Hematology Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | |
Collapse
|
16
|
Rucker M, Fraites TJ, Porvasnik SL, Lewis MA, Zolotukhin I, Cloutier DA, Byrne BJ. Rescue of enzyme deficiency in embryonic diaphragm in a mouse model of metabolic myopathy: Pompe disease. Development 2004; 131:3007-19. [PMID: 15169761 DOI: 10.1242/dev.01169] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Several human genetic diseases that affect striated muscle have been modeled by creating knockout mouse strains. However, many of these are perinatal lethal mutations that result in death from respiratory distress within hours after birth. As the diaphragm muscle does not contract until birth, the sudden increase in diaphragm activity creates permanent injury to the muscle causing it to fail to meet respiratory demands. Therefore, the impact of these mutations remains hidden throughout embryonic development and early death prevents investigators from performing detailed studies of other striated muscle groups past the neonatal stage. Glycogen storage disease type II (GSDII), caused by a deficiency in acid alpha-glucosidase (GAA), leads to lysosomal accumulation of glycogen in all cell types and abnormal myofibrillogenesis in striated muscle. Contractile function of the diaphragm muscle is severely affected in both infantile-onset and late-onset individuals, with death often resulting from respiratory failure. The knockout mouse model of GSDII survives well into adulthood despite the gradual weakening of all striated muscle groups. Using this model, we investigated the delivery of recombinant adeno-associated virus (rAAV) vectors encoding the human GAA cDNA to the developing embryo. Results indicate specific high-level transduction of diaphragm tissue, leading to activity levels up to 10-fold higher than normal and restoration of normal contractile function. Up to an estimated 50 vector copies per diploid genome were quantified in treated diaphragms. Histological glycogen staining of treated diaphragms revealed prevention of lysosomal glycogen accumulation in almost all fibers when compared with untreated controls. This method could be employed with disease models where specific rescue of the diaphragm would allow for increased survival and thus further investigation into the impact of the gene deletion on other striated muscle groups.
Collapse
Affiliation(s)
- Mary Rucker
- Powell Gene Therapy Center, and Departments of Molecular Genetics and Microbiology and Pediatrics, University of Florida College of Medicine, Gainesville, Florida 32610, USA
| | | | | | | | | | | | | |
Collapse
|
17
|
Zhang W, Singam R, Hellermann G, Kong X, Juan HS, Lockey RF, Wu SJ, Porter K, Mohapatra SS. Attenuation of dengue virus infection by adeno-associated virus-mediated siRNA delivery. GENETIC VACCINES AND THERAPY 2004; 2:8. [PMID: 15301687 PMCID: PMC514572 DOI: 10.1186/1479-0556-2-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2004] [Accepted: 08/09/2004] [Indexed: 11/25/2022]
Abstract
Background The need for safe and effective treatment of dengue virus (DEN), a class A agent that causes dengue hemorrhagic fever/dengue shock syndrome, has been a critical global priority. An effective vaccine for DEN is not yet available. In this study the possibility of attenuating DEN infection using adeno-associated virus (AAV)-encoded short interfering RNAs (siRNA) was examined in Vero cells and human dendritic cells (DCs). Methods A cassette encoding siRNA targeted to a 3' untranslated sequence common to all DEN serotypes was designed and tested for its ability to attenuate DEN infection by use of AAV delivery. Results Vero cells or DCs infected with AAV-siRNA showed a significant, dose-dependent reduction in DEN infection. Treatment of DCs with AAV-siRNA also decreased the DEN-induced apoptosis of DCs and did not induce significant inflammation. Conclusion These results demonstrate that AAV-mediated siRNA delivery is capable of reducing DEN infection in cells and may be useful in decreasing DEN replication in humans.
Collapse
Affiliation(s)
- Weidong Zhang
- Division of Allergy and Immunology-JMC Airway Disease Research Center, Department of Internal Medicine, University of South Florida; VA Hospital Tampa, FL, USA
| | - Rajeswari Singam
- Division of Allergy and Immunology-JMC Airway Disease Research Center, Department of Internal Medicine, University of South Florida; VA Hospital Tampa, FL, USA
| | - Gary Hellermann
- Division of Allergy and Immunology-JMC Airway Disease Research Center, Department of Internal Medicine, University of South Florida; VA Hospital Tampa, FL, USA
| | - Xiaoyuan Kong
- Division of Allergy and Immunology-JMC Airway Disease Research Center, Department of Internal Medicine, University of South Florida; VA Hospital Tampa, FL, USA
| | - Homero San Juan
- Division of Allergy and Immunology-JMC Airway Disease Research Center, Department of Internal Medicine, University of South Florida; VA Hospital Tampa, FL, USA
| | - Richard F Lockey
- Division of Allergy and Immunology-JMC Airway Disease Research Center, Department of Internal Medicine, University of South Florida; VA Hospital Tampa, FL, USA
| | - Shuen-Ju Wu
- Viral Diseases Department, Naval Medical Research Center, Silver Spring, Maryland, USA
| | - Kevin Porter
- Viral Diseases Department, Naval Medical Research Center, Silver Spring, Maryland, USA
| | - Shyam S Mohapatra
- Division of Allergy and Immunology-JMC Airway Disease Research Center, Department of Internal Medicine, University of South Florida; VA Hospital Tampa, FL, USA
| |
Collapse
|
18
|
Flotte TR. Gene therapy progress and prospects: recombinant adeno-associated virus (rAAV) vectors. Gene Ther 2004; 11:805-10. [PMID: 15042119 DOI: 10.1038/sj.gt.3302233] [Citation(s) in RCA: 107] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- T R Flotte
- Department of Pediatrics and the Powell Gene Therapy Center, University of Florida, Gainesville, FL, USA
| |
Collapse
|
19
|
Zhang X, Nakaoka T, Nishishita T, Watanabe N, Igura K, Shinomiya KI, Takahashi TA, Yamashita N. Efficient adeno-associated virus-mediated gene expression in human placenta-derived mesenchymal cells. Microbiol Immunol 2003; 47:109-16. [PMID: 12636261 DOI: 10.1111/j.1348-0421.2003.tb02793.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Mesenchymal cells from various sources are pluripotent and are attractive sources for cell transplantation. In this study, we analyzed recombinant adeno-associated virus (rAAV)-mediated gene expression in human placenta-derived mesenchymal cells (hPDMCs), which reside in placental villi. After transduction of AV-CAG-EGFP, a rAAV expressing enhanced green fluorescence protein (EGFP), hPDMCs showed much higher level of EGFP expression than human umbilical vein endothelial cells or rat aortic smooth muscle cells. The number of EGFP-positive hPDMCs infected by AV-CAG-EGFP alone did not increase significantly by coinfection of adenovirus, which enhanced expression level of the rAAV vector. Moreover, flow cytometric analysis showed discrete positive fraction of EGFP-expressing hPDMCs, which is about 15-20% of the cells infected with AV-CAG-EGFP. Therefore, some cell population in hPDMCs might be highly susceptible to rAAV-mediated gene transduction. In addition, stable EGFP expressions were observed in about 1% of hPDMCs infected with AV-CAG-EGFP at 4 weeks post-infection. Collectively, hPDMCs have characters favorable for rAAV-mediated gene expression.
Collapse
Affiliation(s)
- Xiaohong Zhang
- Department of Advanced Medical Science, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo 108-8639, Japan
| | | | | | | | | | | | | | | |
Collapse
|