1
|
Yan Z, Vorhies K, Feng Z, Park SY, Choi SH, Zhang Y, Winter M, Sun X, Engelhardt JF. Recombinant Adeno-Associated Virus-Mediated Editing of the G551D Cystic Fibrosis Transmembrane Conductance Regulator Mutation in Ferret Airway Basal Cells. Hum Gene Ther 2022; 33:1023-1036. [PMID: 35686451 PMCID: PMC9595624 DOI: 10.1089/hum.2022.036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 05/03/2022] [Indexed: 12/30/2022] Open
Abstract
Mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene cause cystic fibrosis (CF), a chronic disease that affects multiple organs, including the lung. We developed a CF ferret model of a scarless G551→D substitution in CFTR (CFTRG551D-KI), enabling approaches to correct this gating mutation in CF airways via gene editing. Homology-directed repair (HDR) was tested in Cas9-expressing CF airway basal cells (Cas9-GKI) from this model, as well as reporter basal cells (Y66S-Cas9-GKI) that express an integrated nonfluorescent Y66S-EGFP (enhanced green fluorescent protein) mutant gene to facilitate rapid assessment of HDR by the restoration of fluorescence. Recombinant adeno-associated virus (rAAV) vectors were used to deliver two DNA templates and sgRNAs for dual-gene editing at the EGFP and CFTR genes, followed by fluorescence-activated cell sorting of EGFPY66S-corrected cells. When gene-edited airway basal cells were polarized at an air-liquid interface, unsorted and EGFPY66S-corrected sorted populations gave rise to 26.0% and 70.4% CFTR-mediated Cl- transport of that observed in non-CF cultures, respectively. The consequences of gene editing at the CFTRG551D locus by HDR and nonhomologous end joining (NHEJ) were assessed by targeted gene next-generation sequencing (NGS) against a specific amplicon. NGS revealed HDR corrections of 3.1% of G551 sequences in the unsorted population of rAAV-infected cells, and 18.4% in the EGFPY66S-corrected cells. However, the largest proportion of sequences had indels surrounding the CRISPR (clustered regularly interspaced short palindromic repeats) cut site, demonstrating that NHEJ was the dominant repair pathway. This approach to simultaneously coedit at two genomic loci using rAAV may have utility as a model system for optimizing gene-editing efficiencies in proliferating airway basal cells through the modulation of DNA repair pathways in favor of HDR.
Collapse
Affiliation(s)
- Ziying Yan
- Department of Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Kai Vorhies
- Department of Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Zehua Feng
- Department of Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Soo Yeun Park
- Department of Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Soon H. Choi
- Department of Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Yulong Zhang
- Department of Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Michael Winter
- Department of Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Xingshen Sun
- Department of Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - John F. Engelhardt
- Department of Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| |
Collapse
|
2
|
Abstract
Reporter gene assays are widely used in high-throughput screening (HTS) to identify compounds that modulate gene expression. Traditionally a reporter gene assay is built by cloning an endogenous promoter sequence or synthetic response elements in the regulatory region of a reporter gene to monitor transcriptional activity of a specific biological process (exogenous reporter assay). In contrast, an endogenous locus reporter has a reporter gene inserted in the endogenous gene locus that allows the reporter gene to be expressed under the control of the same regulatory elements as the endogenous gene, thus more accurately reflecting the changes seen in the regulation of the actual gene. In this chapter, we introduce some of the considerations behind building a reporter gene assay for high-throughput compound screening and describe the methods we have utilized to establish 1536-well format endogenous locus reporter and exogenous reporter assays for the screening of compounds that modulate Myc pathway activity.
Collapse
Affiliation(s)
- Yaping Liu
- Screening & Protein Sciences, Merck Research Labs, Merck & Co., Inc., West Point, PA, USA
| | - Jeffrey Hermes
- Screening and Translational Enzymology, Roche, Basel, Roche, Basel, Canton of Basel-Stadt, Switzerland
| | - Jing Li
- Screening & Protein Sciences, Merck Research Labs, Merck & Co., Inc., West Point, PA, USA
| | - Matthew Tudor
- Screening & Protein Sciences, Merck Research Labs, Merck & Co., Inc., West Point, PA, USA.
| |
Collapse
|
3
|
Mitchell AM, Moser R, Samulski RJ, Hirsch ML. Stimulation of AAV Gene Editing via DSB Repair. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016. [DOI: 10.1007/978-1-4939-3509-3_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
4
|
Genome Engineering Using Adeno-associated Virus: Basic and Clinical Research Applications. Mol Ther 2015; 24:458-64. [PMID: 26373345 DOI: 10.1038/mt.2015.151] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2015] [Accepted: 07/21/2015] [Indexed: 12/18/2022] Open
Abstract
In addition to their broad potential for therapeutic gene delivery, adeno-associated virus (AAV) vectors possess the innate ability to stimulate homologous recombination in mammalian cells at high efficiencies. This process--referred to as AAV-mediated gene targeting--has enabled the introduction of a diverse array of genomic modifications both in vitro and in vivo. With the recent emergence of targeted nucleases, AAV-mediated genome engineering is poised for clinical translation. Here, we review key properties of AAV vectors that underscore its unique utility in genome editing. We highlight the broad range of genome engineering applications facilitated by this technology and discuss the strong potential for unifying AAV with targeted nucleases for next-generation gene therapy.
Collapse
|
5
|
Deyle DR, Hansen RS, Cornea AM, Li LB, Burt AA, Alexander IE, Sandstrom RS, Stamatoyannopoulos JA, Wei CL, Russell DW. A genome-wide map of adeno-associated virus-mediated human gene targeting. Nat Struct Mol Biol 2014; 21:969-75. [PMID: 25282150 PMCID: PMC4405182 DOI: 10.1038/nsmb.2895] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 08/27/2014] [Indexed: 02/03/2023]
Abstract
To determine which genomic features promote homologous recombination, we created a genome-wide map of gene targeting sites. We used an adeno-associated virus vector to target identical loci introduced as transcriptionally active retroviral vectors. A comparison of ~2,000 targeted and untargeted sites showed that targeting occurred throughout the human genome and was not influenced by the presence of nearby CpG islands, sequence repeats or DNase I-hypersensitive sites. Targeted sites were preferentially located within transcription units, especially when the target loci were transcribed in the opposite orientation to their surrounding chromosomal genes. We determined the impact of DNA replication by mapping replication forks, which revealed a preference for recombination at target loci transcribed toward an incoming fork. Our results constitute the first genome-wide screen of gene targeting in mammalian cells and demonstrate a strong recombinogenic effect of colliding polymerases.
Collapse
Affiliation(s)
- David R Deyle
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - R Scott Hansen
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Anda M Cornea
- Department of Molecular and Cellular Biology, University of Washington, Seattle, Washington, USA
| | - Li B Li
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Amber A Burt
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Ian E Alexander
- Gene Therapy Research Unit, Children's Medical Research Institute, Westmead, New South Wales, Australia
| | - Richard S Sandstrom
- Department of Genome Sciences, University of Washington, Seattle, Washington, USA
| | | | - Chia-Lin Wei
- Genomic Technologies Department, Joint Genome Institute, Walnut Creek, California, USA
| | - David W Russell
- 1] Department of Medicine, University of Washington, Seattle, Washington, USA. [2] Department of Biochemistry, University of Washington, Seattle, Washington, USA
| |
Collapse
|
6
|
Deyle DR, Li LB, Ren G, Russell DW. The effects of polymorphisms on human gene targeting. Nucleic Acids Res 2013; 42:3119-24. [PMID: 24371280 PMCID: PMC3950700 DOI: 10.1093/nar/gkt1303] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
DNA mismatches that occur between vector homology arms and chromosomal target sequences reduce gene targeting frequencies in several species; however, this has not been reported in human cells. Here we demonstrate that even a single mismatched base pair can significantly decrease human gene targeting frequencies. In addition, we show that homology arm polymorphisms can be used to direct allele-specific targeting or to improve unfavorable vector designs that introduce deletions.
Collapse
Affiliation(s)
- David R Deyle
- Department of Medicine, University of Washington, Seattle, WA, 98195, USA and Department of Biochemistry, University of Washington, Seattle, WA, 98195, USA
| | | | | | | |
Collapse
|
7
|
Jin LF, Li F, Wang HP, Wei F, Qin P, Du LF. Ultrasound targeted microbubble destruction stimulates cellular endocytosis in facilitation of adeno-associated virus delivery. Int J Mol Sci 2013; 14:9737-50. [PMID: 23652832 PMCID: PMC3676809 DOI: 10.3390/ijms14059737] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2013] [Revised: 04/24/2013] [Accepted: 04/27/2013] [Indexed: 12/19/2022] Open
Abstract
The generally accepted mechanism for ultrasound targeted microbubble destruction (UTMD) to enhance drug and gene delivery is through sonoporation. However, passive uptake of adeno-associated virus (AAV) into cells following sonoporation does not adequately explain observations of enhanced transduction by UTMD. This study investigated alternative mechanisms of UTMD enhancement in AAV delivery. UTMD significantly enhanced transduction efficiency of AAV in a dose-dependent manner. UTMD stimulated a persistent uptake of AAV into the cytoplasm and nucleus. This phenomenon occurred over several hours, suggesting that some viral particles are endocytosed by cells rather than exclusively passing through pores created by sonoporation. Additionally, UTMD enhanced clathrin expression and accumulation at the plasma membrane suggesting greater clathrin-mediated endocytosis following UTMD. Transmission electron microscopy (TEM) revealed that UTMD stimulated formation of clathrin-coated pits (CPs) and uncoated pits (nCPs). Furthermore, inhibition of clathrin-mediated endocytosis partially blocked the enhancement of AAV uptake following UTMD. The results of this study implicate endocytosis as a mechanism that contributes to UTMD-enhanced AAV delivery.
Collapse
Affiliation(s)
- Li-Fang Jin
- Department of Ultrasound, Shanghai Jiaotong University Affiliated First People’s Hospital, 100 Haining Road, Shanghai 200080, China; E-Mails: (L.-F.J.); (F.L.)
| | - Fan Li
- Department of Ultrasound, Shanghai Jiaotong University Affiliated First People’s Hospital, 100 Haining Road, Shanghai 200080, China; E-Mails: (L.-F.J.); (F.L.)
| | - Hui-Ping Wang
- Experimental Research Center, Shanghai Jiaotong University Affiliated First People’s Hospital, 100 Haining Road, Shanghai 200080, China; E-Mails: (H.-P.W.); (F.W.)
| | - Fang Wei
- Experimental Research Center, Shanghai Jiaotong University Affiliated First People’s Hospital, 100 Haining Road, Shanghai 200080, China; E-Mails: (H.-P.W.); (F.W.)
| | - Peng Qin
- Department of Instrumentation Science and Engineering, Shanghai Jiaotong University, 800 Dongchuan Road, Shanghai 200240, China; E-Mail:
| | - Lian-Fang Du
- Department of Ultrasound, Shanghai Jiaotong University Affiliated First People’s Hospital, 100 Haining Road, Shanghai 200080, China; E-Mails: (L.-F.J.); (F.L.)
- Author to whom correspondence should be addressed; E-Mail: ; Tel./Fax: +86-21-6324-0090 (ext. 4417)
| |
Collapse
|
8
|
Luo Y, Kofod-Olsen E, Christensen R, Sørensen CB, Bolund L. Targeted genome editing by recombinant adeno-associated virus (rAAV) vectors for generating genetically modified pigs. J Genet Genomics 2012; 39:269-74. [PMID: 22749014 DOI: 10.1016/j.jgg.2012.05.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2012] [Accepted: 05/10/2012] [Indexed: 12/22/2022]
Abstract
Recombinant adeno-associated virus (rAAV) vectors have been extensively used for experimental gene therapy of inherited human diseases. Several advantages, such as simple vector construction, high targeting frequency by homologous recombination, and applicability to many cell types, make rAAV an attractive approach for targeted genome editing. Combined with cloning by somatic cell nuclear transfer (SCNT), this technology has recently been successfully adapted to generate gene-targeted pigs as models for cystic fibrosis, hereditary tyrosinemia type 1, and breast cancer. This review summarizes the development of rAAV for targeted genome editing in mammalian cells and provides strategies for enhancing the rAAV-mediated targeting frequency by homologous recombination. We discuss current development and application of the rAAV vectors for targeted genome editing in porcine primary fibroblasts, which are subsequently used as donor cells for SCNT to generate cloned genetically designed pigs and provide positive perspectives for the generation of gene-targeted pigs with rAAV in the future.
Collapse
Affiliation(s)
- Yonglun Luo
- Department of Biomedicine, Aarhus University, Wilhelm Meyers Alle 4, DK-8000 Aarhus C, Denmark.
| | | | | | | | | |
Collapse
|
9
|
Abstract
Gene targeting with adeno-associated virus (AAV) vectors has been demonstrated in multiple human cell types, with targeting frequencies ranging from 10(-5) to 10(-2) per infected cell. These targeting frequencies are 1-4 logs higher than those obtained by conventional transfection or electroporation approaches. A wide variety of different types of mutations can be introduced into chromosomal loci with high fidelity and without genotoxicity. Here we provide a detailed protocol for gene targeting in human cells with AAV vectors. We describe methods for vector design, stock preparation and titration. Optimized transduction protocols are provided for human pluripotent stem cells, mesenchymal stem cells, fibroblasts and transformed cell lines, as well as a method for identifying targeted clones by Southern blots. This protocol (from vector design through a single round of targeting and screening) can be completed in ∼10 weeks; each subsequent round of targeting and screening should take an additional 7 weeks.
Collapse
Affiliation(s)
- Iram F Khan
- Department of Medicine, University of Washington, Seattle, WA, USA
| | | | | |
Collapse
|
10
|
Jensen NM, Dalsgaard T, Jakobsen M, Nielsen RR, Sørensen CB, Bolund L, Jensen TG. An update on targeted gene repair in mammalian cells: methods and mechanisms. J Biomed Sci 2011; 18:10. [PMID: 21284895 PMCID: PMC3042377 DOI: 10.1186/1423-0127-18-10] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2010] [Accepted: 02/02/2011] [Indexed: 11/10/2022] Open
Abstract
Transfer of full-length genes including regulatory elements has been the preferred gene therapy strategy for clinical applications. However, with significant drawbacks emerging, targeted gene alteration (TGA) has recently become a promising alternative to this method. By means of TGA, endogenous DNA repair pathways of the cell are activated leading to specific genetic correction of single-base mutations in the genome. This strategy can be implemented using single-stranded oligodeoxyribonucleotides (ssODNs), small DNA fragments (SDFs), triplex-forming oligonucleotides (TFOs), adeno-associated virus vectors (AAVs) and zinc-finger nucleases (ZFNs). Despite difficulties in the use of TGA, including lack of knowledge on the repair mechanisms stimulated by the individual methods, the field holds great promise for the future. The objective of this review is to summarize and evaluate the different methods that exist within this particular area of human gene therapy research.
Collapse
Affiliation(s)
- Nanna M Jensen
- Institute of Human Genetics, The Bartholin Building, University of Aarhus, 8000 Aarhus C, Denmark
| | | | | | | | | | | | | |
Collapse
|
11
|
Gellhaus K, Cornu TI, Heilbronn R, Cathomen T. Fate of Recombinant Adeno-Associated Viral Vector Genomes During DNA Double-Strand Break-Induced Gene Targeting in Human Cells. Hum Gene Ther 2010; 21:543-53. [DOI: 10.1089/hum.2009.167] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Katharina Gellhaus
- Institute of Virology, Charité Medical School, Campus Benjamin Franklin, D-12203 Berlin, Germany
- Present address: Epiontis GmbH, D-12489 Berlin, Germany
| | - Tatjana I. Cornu
- Institute of Virology, Charité Medical School, Campus Benjamin Franklin, D-12203 Berlin, Germany
- Present address: Epiontis GmbH, D-12489 Berlin, Germany
| | - Regine Heilbronn
- Institute of Virology, Charité Medical School, Campus Benjamin Franklin, D-12203 Berlin, Germany
| | - Toni Cathomen
- Institute of Virology, Charité Medical School, Campus Benjamin Franklin, D-12203 Berlin, Germany
- Department of Experimental Hematology, Hannover Medical School, D-30625 Hannover, Germany
| |
Collapse
|
12
|
Paiboonsukwong K, Ohbayashi F, Shiiba H, Aizawa E, Yamashita T, Mitani K. Correction of mutant Fanconi anemia gene by homologous recombination in human hematopoietic cells using adeno-associated virus vector. J Gene Med 2010; 11:1012-9. [PMID: 19653252 DOI: 10.1002/jgm.1382] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND Adeno-associated virus (AAV) vectors have been shown to correct a variety of mutations in human cells by homologous recombination (HR) at high rates, which can overcome insertional mutagenesis and transgene silencing, two of the major hurdles in conventional gene addition therapy of inherited diseases. We examined an ability of AAV vectors to repair a mutation in human hematopoietic cells by HR. METHODS We infected a human B-lymphoblastoid cell line (BCL) derived from a normal subject with an AAV, which disrupts the hypoxanthine phosphoribosyl transferase1 (HPRT1) locus, to measure the frequency of AAV-mediated HR in BCL cells. We subsequently constructed an AAV vector encoding the normal sequences from the Fanconi anemia group A (FANCA) locus to correct a mutation in the gene in BCL derived from a FANCA patient. RESULTS Under optimal conditions, approximately 50% of BCL cells were transduced with an AAV serotype 2 (AAV-2) vector. In FANCA BCL cells, up to 0.016% of infected cells were gene-corrected by HR. AAV-mediated restoration of normal genotypic and phenotypic characteristics in FANCA-mutant cells was confirmed at the DNA, protein and functional levels. CONCLUSIONS The results obtained in the present study indicate that AAV vectors may be applicable for gene correction therapy of inherited hematopoietic disorders.
Collapse
Affiliation(s)
- Kittiphong Paiboonsukwong
- Gene Therapy Division, Research Center for Genomic Medicine, Saitama Medical University, Saitama, Japan
| | | | | | | | | | | |
Collapse
|
13
|
A comparison of synthetic oligodeoxynucleotides, DNA fragments and AAV-1 for targeted episomal and chromosomal gene repair. BMC Biotechnol 2009; 9:35. [PMID: 19379497 PMCID: PMC2676283 DOI: 10.1186/1472-6750-9-35] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2008] [Accepted: 04/20/2009] [Indexed: 12/14/2022] Open
Abstract
Background Current strategies for gene therapy of inherited diseases consist in adding functional copies of the gene that is defective. An attractive alternative to these approaches would be to correct the endogenous mutated gene in the affected individual. This study presents a quantitative comparison of the repair efficiency using different forms of donor nucleic acids, including synthetic DNA oligonucleotides, double stranded DNA fragments with sizes ranging from 200 to 2200 bp and sequences carried by a recombinant adeno-associated virus (rAAV-1). Evaluation of each gene repair strategy was carried out using two different reporter systems, a mutated eGFP gene or a dual construct with a functional eGFP and an inactive luciferase gene, in several different cell systems. Gene targeting events were scored either following transient co-transfection of reporter plasmids and donor DNAs, or in a system where a reporter construct was stably integrated into the chromosome. Results In both episomal and chromosomal assays, DNA fragments were more efficient at gene repair than oligonucleotides or rAAV-1. Furthermore, the gene targeting frequency could be significantly increased by using DNA repair stimulating drugs such as doxorubicin and phleomycin. Conclusion Our results show that it is possible to obtain repair frequencies of 1% of the transfected cell population under optimized transfection protocols when cells were pretreated with phleomycin using rAAV-1 and dsDNA fragments.
Collapse
|
14
|
Liu X, Luo M, Guo C, Yan Z, Wang Y, Lei-Butters DCM, Engelhardt JF. Analysis of adeno-associated virus progenitor cell transduction in mouse lung. Mol Ther 2008; 17:285-93. [PMID: 19034263 DOI: 10.1038/mt.2008.248] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Although recombinant adeno-associated virus (rAAV) has been widely used in lung gene therapy approaches, it remains unclear to what extent commonly used AAV serotypes transduce adult progenitors in the lung. In this study, we evaluated the life span and proliferative capacity of rAAV1-, 2-, and 5-transduced airway cells in mouse lung, using a LacZ-CRE reporter transgenic model and Cre-expressing rAAV. In this model, the expression of CRE recombinase led to permanent genetic marking of transduced cells and their descendants with LacZ. To investigate whether the rAAV-transduced cells included airway progenitors, we injured the airways of rAAV-infected mice with Naphthalene, while simultaneously labeling with 5-bromodeoxyuridine (BrdU) to identify slow-cycling progenitor/stem cells that entered the cell cycle and retained label. Both rAAV5 and rAAV1 vectors were capable of transducing a subset of long-lived Clara cells and alveolar type II (ATII) cells that retained nucleotide label and proliferated following lung injury. Importantly, rAAV1 and 5 appeared to preferentially transduce conducting airway epithelial progenitors that had the capacity to clonally expand, both in culture and in vivo following lung injury. These studies suggest that rAAV may be a useful vector for gene targeting of airway stem/progenitor cells.
Collapse
Affiliation(s)
- Xiaoming Liu
- Department of Anatomy and Cell Biology, College of Medicine, The University of Iowa, Iowa City, Iowa, USA
| | | | | | | | | | | | | |
Collapse
|
15
|
Schultz BR, Chamberlain JS. Recombinant adeno-associated virus transduction and integration. Mol Ther 2008; 16:1189-99. [PMID: 18500252 DOI: 10.1038/mt.2008.103] [Citation(s) in RCA: 143] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Recombinant adeno-associated virus (rAAV) holds promise as a gene therapy vector for a multitude of genetic disorders such as hemophilia, cystic fibrosis, and the muscular dystrophies. Given the variety of applications and tissue types toward which these vectors may be targeted, an understanding of rAAV transduction is crucial for the effective application of therapy. rAAV transduction mechanisms have been the subject of much study, resulting in a body of knowledge relating to events from virus-cell attachment through to vector genome conformation in the target cell nucleus. Instead of utilizing one mechanism in each phase of vector transduction, rAAV appears to employ multiple possible pathways toward transgene expression, in part dependent on rAAV serotype, dose, and target cell type. Once inside the nucleus, the rAAV genome exists in a predominantly episomal form; therefore, nondividing cells tend to be most stably transduced. However, rAAV has a low frequency of integration into the host cell genome, often in or near genes, and can be associated with host genome mutations. This review describes the current understanding of the mechanisms and rate-limiting steps involved in rAAV transduction.
Collapse
Affiliation(s)
- Brian R Schultz
- Molecular and Cellular Biology, Senator Paul D Wellstone Muscular Dystrophy Cooperative Research Center, University of Washington, Seattle, Washington, USA
| | | |
Collapse
|
16
|
Ghosh A, Duan D. Expanding adeno-associated viral vector capacity: a tale of two vectors. Biotechnol Genet Eng Rev 2008; 24:165-77. [PMID: 18059632 DOI: 10.1080/02648725.2007.10648098] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Affiliation(s)
- Arkasubhra Ghosh
- Department of Molecular Microbiology and Immunology, University of Missouri, School of Medicine, One Hospital Dr., Room M610G, MSB, Columbia, MO 65212, USA
| | | |
Collapse
|
17
|
Fernandez SL, Russell DW, Hurlin PJ. Development of human gene reporter cell lines using rAAV mediated homologous recombination. Biol Proced Online 2007; 9:84-90. [PMID: 18464937 PMCID: PMC2374725 DOI: 10.1251/bpo136] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2007] [Revised: 10/04/2007] [Accepted: 11/19/2007] [Indexed: 11/23/2022] Open
Abstract
Understanding mechanisms of gene regulation has broad therapeutic implications for human disease. Here we describe a novel method for generating human cell lines that serve as reporters of transcriptional activity. This method exploits the ability of recombinant adeno-associated virus to mediate the insertion of exogenous DNA sequences into specific genomic loci through homologous recombination. To overcome the severe size limitation of the rAAV for carrying exogenous DNA, an enhanced green fluorescent protein (EGFP)-Luciferase fusion gene was used as both a selectable marker and gene expression reporter. EGFP was used for selection of correctly targeted alleles by taking advantage of known regulatory conditions that activate transcription of specific genes. Using this method, we describe the generation of primary human fibroblasts that express EGFP-Luciferase under the control of the c-Myc oncogene.
Collapse
Affiliation(s)
- Sandra L Fernandez
- Shriners Hospitals for Children. Department of Cell Developmental Biology, Oregon Health & Science, Portland, OR 97201, USA
| | | | | |
Collapse
|
18
|
Cornu TI, Cathomen T. Targeted genome modifications using integrase-deficient lentiviral vectors. Mol Ther 2007; 15:2107-13. [PMID: 17998901 DOI: 10.1038/sj.mt.6300345] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Gene correction aims at repairing a defective gene directly in the cellular genome, which warrants tissue-specific and sustained expression of the repaired gene through its endogenous promoter. We have developed a novel system based on integrase-deficient lentiviral vectors (IDLVs) that allows us to correct an endogenous mutation using a strategy based on homologous recombination (HR). In a proof-of-concept approach, an IDLV encoding a repair template was co-delivered with an I-SceI nuclease expression vector to rescue a defective enhanced green fluorescent protein (EGFP) gene. Expression of the nuclease created a double-strand break within the target locus, which was crucial for stimulating IDLV-based gene repair. Stable gene correction was realized in up to 12% of the cells, depending on the vector dose, the nuclease expression levels, and the cell type. Genotypic analyses confirmed that gene correction was the result of genuine HR between the target locus and the IDLV repair template. This study presents IDLVs as valuable tools for introducing precise and permanent genetic modifications in human cells.
Collapse
Affiliation(s)
- Tatjana I Cornu
- Charité Medical School, Institute of Virology (CBF), Berlin, Germany
| | | |
Collapse
|
19
|
Liu X, Luo M, Guo C, Yan Z, Wang Y, Engelhardt JF. Comparative biology of rAAV transduction in ferret, pig and human airway epithelia. Gene Ther 2007; 14:1543-8. [PMID: 17728794 PMCID: PMC2121584 DOI: 10.1038/sj.gt.3303014] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Differences between rodent and human airway cell biology have made it difficult to translate recombinant adeno-associated virus (rAAV)-mediated gene therapies to the lung for cystic fibrosis (CF). As new ferret and pig models for CF become available, knowledge about host cell/vector interactions in these species will become increasingly important for testing potential gene therapies. To this end, we have compared the transduction biology of three rAAV serotypes (AAV1, 2 and 5) in human, ferret, pig and mouse-polarized airway epithelia. Our results indicate that apical transduction of ferret and pig airway epithelia with these rAAV serotypes closely mirrors that observed in human epithelia (rAAV1>rAAV2 congruent withrAAV5), while transduction of mouse epithelia was significantly different (rAAV1>rAAV5>>rAAV2). Similarly, ferret, pig and human epithelia also shared serotype-specific differences in the polarity (apical vs basolateral) and proteasome dependence of rAAV transduction. Despite these parallels, N-linked sialic acid receptors were required for rAAV1 and rAAV5 transduction of human and mouse airway epithelia, but not ferret or pig airway epithelia. Hence, although the airway tropisms of rAAV serotypes 1, 2 and 5 are conserved better among ferret, pig and human as compared to mouse, viral receptors/co-receptors appear to maintain considerable species diversity.
Collapse
Affiliation(s)
- X Liu
- Department of Anatomy & Cell Biology, College of Medicine, The University of Iowa, Iowa City, IA, USA
- Center for Gene Therapy of Cystic Fibrosis and Other Genetic Diseases, College of Medicine, The University of Iowa, Iowa City, IA, USA
| | - M Luo
- Department of Anatomy & Cell Biology, College of Medicine, The University of Iowa, Iowa City, IA, USA
| | - C Guo
- Department of Anatomy & Cell Biology, College of Medicine, The University of Iowa, Iowa City, IA, USA
- College of Life Science, Ningxia University, Yingchuan, Ningxia, China
| | - Z Yan
- Department of Anatomy & Cell Biology, College of Medicine, The University of Iowa, Iowa City, IA, USA
- Center for Gene Therapy of Cystic Fibrosis and Other Genetic Diseases, College of Medicine, The University of Iowa, Iowa City, IA, USA
| | - Y Wang
- College of Life Science, Ningxia University, Yingchuan, Ningxia, China
| | - JF Engelhardt
- Department of Anatomy & Cell Biology, College of Medicine, The University of Iowa, Iowa City, IA, USA
- College of Life Science, Ningxia University, Yingchuan, Ningxia, China
- Department of Internal Medicine, College of Medicine, The University of Iowa, Iowa City, IA, USA
- Center for Gene Therapy of Cystic Fibrosis and Other Genetic Diseases, College of Medicine, The University of Iowa, Iowa City, IA, USA
| |
Collapse
|
20
|
Alwin S, Gere MB, Guhl E, Effertz K, Barbas CF, Segal DJ, Weitzman MD, Cathomen T. Custom zinc-finger nucleases for use in human cells. Mol Ther 2006; 12:610-7. [PMID: 16039907 DOI: 10.1016/j.ymthe.2005.06.094] [Citation(s) in RCA: 135] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2005] [Revised: 06/03/2005] [Accepted: 06/06/2005] [Indexed: 12/21/2022] Open
Abstract
Genome engineering through homologous recombination (HR) is a powerful instrument for studying biological pathways or creating treatment options for genetic disorders. In mammalian cells HR is rare but the creation of targeted DNA double-strand breaks stimulates HR significantly. Here, we present a method to generate, evaluate, and optimize rationally designed endonucleases that promote HR. The DNA-binding domains were synthesized by assembling predefined zinc-finger modules selected by phage display. Attachment of a transcriptional activation domain allowed assessment of DNA binding in reporter assays, while fusion with an endonuclease domain created custom nucleases that were tested for their ability to stimulate HR in episomal and chromosomal gene repair assays. We demonstrate that specificity, expression kinetics, and protein design are crucial parameters for efficient gene repair and that our two-step assay allows one to go quickly from design to testing to successful employment of the custom nucleases in human cells.
Collapse
Affiliation(s)
- Stephen Alwin
- Institute of Virology, Charité Medical School, Campus Benjamin Franklin, Berlin, Germany
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Miller DG, Wang PR, Petek LM, Hirata RK, Sands MS, Russell DW. Gene targeting in vivo by adeno-associated virus vectors. Nat Biotechnol 2006; 24:1022-6. [PMID: 16878127 DOI: 10.1038/nbt1231] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2006] [Accepted: 06/15/2006] [Indexed: 12/28/2022]
Abstract
Therapeutic gene delivery typically involves the addition of a transgene expression cassette to mutant cells. This approach is complicated by transgene silencing, aberrant transcriptional regulation and insertional mutagenesis. An alternative strategy is to correct mutations through homologous recombination, allowing for normal regulation of gene expression from the endogenous locus. Adeno-associated virus (AAV) vectors containing single-stranded DNA efficiently transduce cells in vivo and have been shown to target homologous chromosomal sequences in cultured cells. To determine whether AAV-mediated gene targeting can occur in vivo, we developed a mouse model that contains a mutant, nuclear-localized lacZ gene inserted at the ubiquitously expressed ROSA26 locus. Foci of beta-galactosidase-positive hepatocytes were observed in these mice after injection with an AAV vector containing a lacZ gene fragment, and precise correction of the 4-bp deletion was demonstrated by gene sequencing. We also used AAV gene-targeting vectors to correct the naturally occurring GusB gene mutation responsible for murine mucopolysaccharidosis type VII.
Collapse
Affiliation(s)
- Daniel G Miller
- Department of Pediatrics, Division of Genetics and Developmental Medicine, University of Washington, 1705 NE Pacific St., Seattle, WA 98195-7720, USA
| | | | | | | | | | | |
Collapse
|
22
|
Vasileva A, Linden RM, Jessberger R. Homologous recombination is required for AAV-mediated gene targeting. Nucleic Acids Res 2006; 34:3345-60. [PMID: 16822856 PMCID: PMC1488886 DOI: 10.1093/nar/gkl455] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
High frequencies of gene targeting can be achieved by infection of mammalian cells with recombinant adeno-associated virus (rAAV) vectors [D. W. Russell and R. K. Hirata (1998) Nature Genet., 18, 325–330; D. W. Russell and R. K. Hirata (2000) J. Virol., 74, 4612–4620; R. Hirata et al. (2002) Nat. Biotechnol., 20, 735–738], but the mechanism of targeting is unclear and random integration often occurs in parallel. We assessed the role of specific DNA repair and recombination pathways in rAAV gene targeting by measuring correction of a mutated enhanced green fluorescent protein (EGFP) gene in cells where homologous recombination (HR) or non-homologous end-joining (NHEJ) had been suppressed by RNAi. EGFP-negative cells were transduced with rAAV vectors carrying a different inactivating deletion in the EGFP, and in parallel with rAAV vectors carrying red fluorescent protein (RFP). Expression of RFP accounted for viral transduction efficiency and long-term random integration. Approximately 0.02% of the infected GFP-negative cells were stably converted to GFP positive cells. Silencing of the essential NHEJ component DNA-PK had no significant effect on the frequency of targeting at any time point examined. Silencing of the SNF2/SWI2 family members RAD54L or RAD54B, which are important for HR, reduced the rate of stable rAAV gene targeting ∼5-fold. Further, partial silencing of the Rad51 paralogue XRCC3 completely abolished stable long-term EGFP expression. These results show that rAAV gene targeting requires the Rad51/Rad54 pathway of HR.
Collapse
Affiliation(s)
- Ana Vasileva
- Department of Gene and Cell Medicine, Mount Sinai School of MedicineNew York, NY 10029, USA
| | - R. Michael Linden
- Department of Gene and Cell Medicine, Mount Sinai School of MedicineNew York, NY 10029, USA
| | - Rolf Jessberger
- Department of Gene and Cell Medicine, Mount Sinai School of MedicineNew York, NY 10029, USA
- Institute of Physiological Chemistry, Medical Faculty Carl Gustav Carus, Dresden University of Technology01307 Dresden, Germany
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
23
|
Abstract
Vectors based on the adeno-associated virus (AAV) have attracted much attention as potent gene-delivery vehicles, mainly because of the persistence of this non-pathogenic virus in the host cell and its sustainable therapeutic gene expression. However, virus infection can be accompanied by potentially mutagenic random vector integration into the genome. A novel approach to AAV-mediated gene therapy based on gene targeting through homologous recombination allows efficient, high-fidelity, non-mutagenic gene repair in a host cell.
Collapse
Affiliation(s)
- Ana Vasileva
- Department of Gene and Cell Medicine, Mount Sinai School of Medicine, New York, USA
| | | |
Collapse
|
24
|
Lewin AS, Glazer PM, Milstone LM. Gene therapy for autosomal dominant disorders of keratin. J Investig Dermatol Symp Proc 2005; 10:47-61. [PMID: 16250209 DOI: 10.1111/j.1087-0024.2005.10207.x] [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] [Indexed: 12/31/2022]
Abstract
Dominant mutations that interfere with the assembly of keratin filaments cause painful and disfiguring epidermal diseases like pachyonychia congenita and epidermolysis bullosa simplex. Genetic therapies for such diseases must either suppress the production of the toxic proteins or correct the genetic defect in the chromosome. Because epidermal skin cells may be genetically modified in tissue culture or in situ, gene correction is a legitimate goal for keratin diseases. In addition, recent innovations, such as RNA interference in animals, make an RNA knockdown approach plausible in the near future. Although agents of RNA reduction (small interfering RNA, ribozymes, triplex oligonucleotides, or antisense DNA) can be delivered as nucleotides, the impermeability of the skin to large charged molecules presents a serious impediment. Using viral vectors to deliver genes for selective inhibitors of gene expression presents an attractive alternative for long-term treatment of genetic disease in the skin.
Collapse
MESH Headings
- Animals
- Darier Disease/genetics
- Darier Disease/therapy
- Dependovirus/genetics
- Ectodermal Dysplasia/genetics
- Ectodermal Dysplasia/therapy
- Epidermolysis Bullosa Simplex/genetics
- Epidermolysis Bullosa Simplex/therapy
- Gene Silencing
- Gene Targeting
- Genes, Dominant
- Genetic Therapy
- Genetic Vectors
- Humans
- Keratins/genetics
- Keratoderma, Palmoplantar/genetics
- Keratoderma, Palmoplantar/therapy
- Mice
- Mutation
- Nails, Malformed/genetics
- Nails, Malformed/therapy
- Oligonucleotides, Antisense/genetics
- Oligonucleotides, Antisense/therapeutic use
- RNA Interference
- RNA, Catalytic/genetics
- RNA, Catalytic/therapeutic use
- RNA, Small Interfering/genetics
- RNA, Small Interfering/therapeutic use
Collapse
Affiliation(s)
- Alfred S Lewin
- Department of Molecular Genetics and Microbiology, University of Florida College of Medicine, Gainesville, Florida 32610-0266, USA.
| | | | | |
Collapse
|
25
|
Trobridge G, Hirata RK, Russell DW. Gene targeting by adeno-associated virus vectors is cell-cycle dependent. Hum Gene Ther 2005; 16:522-6. [PMID: 15871683 DOI: 10.1089/hum.2005.16.522] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Adeno-associated virus (AAV) vectors can be used to introduce site-specific mutations into homologous chromosomal sequences. There are many potential applications of this technique, but the process of AAV-mediated gene targeting and factors that influence targeting efficiency are not completely understood. We investigated the dependence of AAV-mediated gene targeting on the host cell-cycle status. The frequency of gene targeting by AAV vectors was compared in dividing and serum-arrested normal human fibroblast cultures. Gene targeting occurred in arrested fibroblast cultures at 0.15 to 1.1% the frequency of dividing cultures, and only took place in cells that had undergone DNA synthesis. Gene targeting was also reduced when DNA synthesis was inhibited by hydroxyurea.
Collapse
Affiliation(s)
- Grant Trobridge
- Division of Hematology, Department of Medicine, University of Washington, Seattle, WA 98195, USA
| | | | | |
Collapse
|
26
|
Hendrie PC, Russell DW. Gene Targeting with Viral Vectors. Mol Ther 2005; 12:9-17. [PMID: 15932801 DOI: 10.1016/j.ymthe.2005.04.006] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2005] [Revised: 04/06/2005] [Accepted: 04/06/2005] [Indexed: 10/25/2022] Open
Abstract
Genetic manipulation of cells for scientific and therapeutic goals can be achieved by both gene-addition and gene-targeting methods. Gene targeting precisely alters a gene in its natural chromosome location, providing distinct advantages over gene-addition approaches. Classic gene-targeting delivery systems (microinjection, electroporation, or calcium phosphate transfection) have led to major scientific advances, but are too inefficient in their current state to be used for some applications, including gene therapy. This review describes the development of gene-targeting vectors based on three types of viruses (retrovirus, adenovirus, and adeno-associated virus) and discusses the design, possible mechanisms of action, and applications of gene-targeting vectors based on adeno-associated virus.
Collapse
Affiliation(s)
- Paul C Hendrie
- Division of Hematology, Department of Medicine, University of Washington, Seattle, WA 98195, USA
| | | |
Collapse
|
27
|
Suzuki I, Im S, Tada A, Scott C, Akcali C, Davis MB, Barsh G, Hearing V, Abdel-Malek Z. Participation of the melanocortin-1 receptor in the UV control of pigmentation. J Investig Dermatol Symp Proc 1999; 4:29-34. [PMID: 10537004 DOI: 10.1038/sj.jidsp.5640177] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The cloning of the melanocortin-1 receptor (MC1R) gene from human melanocytes and the demonstration that these cells respond to the melanocortins alpha-melanocyte stimulating hormone (alpha-MSH) and adrenocorticotropic hormone (ACTH) with increased proliferation and melanogenesis have renewed the interest in investigation the physiological role of these hormones in regulating human pigmentation. Alpha-melanocyte stimulating hormone and ACTH are both synthesized in the human epidermis, and their synthesis is upregulated by exposure to ultraviolet radiation (UVR). Activation of the MC1R by ligand binding results in stimulation of cAMP formation, which is a principal mechanism for inducing melanogenesis. The increase in cAMP is required for the pigmentary response of human melanocytes to UVR, and for allowing them to overcome the UVR-induced G1 arrest. Treatment of human melanocytes with alpha-MSH increases eumelanin synthesis, an effect that is expected to enhance photoprotection of the skin. Population studies have revealed more than 20 allelic variants of the MC1R gene. Some of these variants are overexpressed in individuals with skin type I or II, red hair, and poor tanning ability. Future studies will aim at further exploration of the role of these variants in MC1R function, and in determining constitutive human pigmentation, the response to sun exposure, and possibly the susceptibility to skin cancer.
Collapse
Affiliation(s)
- I Suzuki
- Department of Dermatology, University of Cincinnati, Ohio 45267-0592, USA
| | | | | | | | | | | | | | | | | |
Collapse
|