201
|
Abstract
Many devastating human diseases are caused by mutations in a single gene that prevent a somatic cell from carrying out its essential functions, or by genetic changes acquired as a result of infectious disease or in the course of cell transformation. Targeted gene therapies have emerged as potential strategies for treatment of such diseases. These therapies depend upon rare-cutting endonucleases to cleave at specific sites in or near disease genes. Targeted gene correction provides a template for homology-directed repair, enabling the cell's own repair pathways to erase the mutation and replace it with the correct sequence. Targeted gene disruption ablates the disease gene, disabling its function. Gene targeting can also promote other kinds of genome engineering, including mutation, insertion, or gene deletion. Targeted gene therapies present significant advantages compared to approaches to gene therapy that depend upon delivery of stably expressing transgenes. Recent progress has been fueled by advances in nuclease discovery and design, and by new strategies that maximize efficiency of targeting and minimize off-target damage. Future progress will build on deeper mechanistic understanding of critical factors and pathways.
Collapse
Affiliation(s)
- Olivier Humbert
- Departments of Immunology and Biochemistry, University of Washington School of Medicine, Seattle, WA 98195, USA
| | | | | |
Collapse
|
202
|
Gruenert DC, Sargent RG. Virus-mediated Genetic Surgery: Homologous Recombination With a Little "Helper" From My Friends. MOLECULAR THERAPY. NUCLEIC ACIDS 2012; 1:e2. [PMID: 23344619 PMCID: PMC3381596 DOI: 10.1038/mtna.2011.7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Affiliation(s)
- Dieter C Gruenert
- 1] Department of Otolaryngology-Head and Neck Surgery, University of California, San Francisco, San Francisco, California, USA [2] Department of Laboratory Medicine, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Helen Diller Family Comprehensive Cancer Center, Institute for Human Genetics, Cardiovascular Research Institute, University of California, San Francisco, San Francisco, California, USA
| | | |
Collapse
|
203
|
Deem AK, Li X, Tyler JK. Epigenetic regulation of genomic integrity. Chromosoma 2012; 121:131-51. [PMID: 22249206 DOI: 10.1007/s00412-011-0358-1] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2011] [Revised: 12/16/2011] [Accepted: 12/19/2011] [Indexed: 10/14/2022]
Abstract
Inefficient and inaccurate repair of DNA damage is the principal cause of DNA mutations, chromosomal aberrations, and carcinogenesis. Numerous multiple-step DNA repair pathways exist whose deployment depends on the nature of the DNA lesion. Common to all eukaryotic DNA repair pathways is the need to unravel the compacted chromatin structure to facilitate access of the repair machinery to the DNA and restoration of the original chromatin state afterward. Accordingly, our cells utilize a plethora of coordinated mechanisms to locally open up the chromatin structure to reveal the underlying DNA sequence and to orchestrate the efficient and accurate repair of DNA lesions. Here we review changes to the chromatin structure that are intrinsic to the DNA damage response and the available mechanistic insight into how these chromatin changes facilitate distinct stages of the DNA damage repair pathways to maintain genomic stability.
Collapse
Affiliation(s)
- Angela K Deem
- Department of Biochemistry and Molecular Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | | |
Collapse
|
204
|
Abstract
Zinc-finger nucleases (ZFNs) are targetable DNA cleavage reagents that have been adopted as gene-targeting tools. ZFN-induced double-strand breaks are subject to cellular DNA repair processes that lead to both targeted mutagenesis and targeted gene replacement at remarkably high frequencies. This article briefly reviews the history of ZFN development and summarizes applications that have been made to genome editing in many different organisms and situations. Considerable progress has been made in methods for deriving zinc-finger sets for new genomic targets, but approaches to design and selection are still being perfected. An issue that needs more attention is the extent to which available mechanisms of double-strand break repair limit the scope and utility of ZFN-initiated events. The bright prospects for future applications of ZFNs, including human gene therapy, are discussed.
Collapse
|
205
|
Muñoz NM, Beard BC, Ryu BY, Luche RM, Trobridge GD, Rawlings DJ, Scharenberg AM, Kiem HP. Novel reporter systems for facile evaluation of I-SceI-mediated genome editing. Nucleic Acids Res 2011; 40:e14. [PMID: 22110042 PMCID: PMC3258163 DOI: 10.1093/nar/gkr897] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Two major limitations to achieve efficient homing endonuclease-stimulated gene correction using retroviral vectors are low frequency of gene targeting and random integration of the targeting vectors. To overcome these issues, we developed a reporter system for quick and facile testing of novel strategies to promote the selection of cells that undergo targeted gene repair and to minimize the persistence of random integrations and non-homologous end-joining events. In this system, the gene target has an I-SceI site upstream of an EGFP reporter; and the repair template includes a non-functional EGFP gene, the positive selection transgene MGMTP140K tagged with mCherry, and the inducible Caspase-9 suicide gene. Using this dual fluorescent reporter system it is possible to detect properly targeted integration. Furthermore, this reporter system provides an efficient approach to enrich for gene correction events and to deplete events produced by random integration. We have also developed a second reporter system containing MGMTP140K in the integrated target locus, which allows for selection of primary cells with the integrated gene target after transplantation. This system is particularly useful for testing repair strategies in primary hematopoietic stem cells. Thus, our reporter systems should allow for more efficient gene correction with less unwanted off target effects.
Collapse
Affiliation(s)
- Nina M Muñoz
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | | | | | | | | | | | | | | |
Collapse
|
206
|
Chan F, Hauswirth WW, Wensel TG, Wilson JH. Efficient mutagenesis of the rhodopsin gene in rod photoreceptor neurons in mice. Nucleic Acids Res 2011; 39:5955-66. [PMID: 21478169 PMCID: PMC3152346 DOI: 10.1093/nar/gkr196] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2011] [Revised: 02/24/2011] [Accepted: 03/17/2011] [Indexed: 11/13/2022] Open
Abstract
Dominant mutations in the rhodopsin gene, which is expressed in rod photoreceptor cells, are a major cause of the hereditary-blinding disease, autosomal dominant retinitis pigmentosa. Therapeutic strategies designed to edit such mutations will likely depend on the introduction of double-strand breaks and their subsequent repair by homologous recombination or non-homologous end joining. At present, the break repair capabilities of mature neurons, in general, and rod cells, in particular, are undefined. To detect break repair, we generated mice that carry a modified human rhodopsin-GFP fusion gene at the normal mouse rhodopsin locus. The rhodopsin-GFP gene carries tandem copies of exon 2, with an ISceI recognition site situated between them. An ISceI-induced break can be repaired either by non-homologous end joining or by recombination between the duplicated segments, generating a functional rhodopsin-GFP gene. We introduced breaks using recombinant adeno-associated virus to transduce the gene encoding ISceI nuclease. We found that virtually 100% of transduced rod cells were mutated at the ISceI site, with ∼85% of the genomes altered by end joining and ∼15% by the single-strand annealing pathway of homologous recombination. These studies establish that the genomes of terminally differentiated rod cells can be efficiently edited in living organisms.
Collapse
Affiliation(s)
- Fung Chan
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX 77030 and Department of Ophthalmology, University of Florida, Gainesville, FL 32610, USA
| | - William W. Hauswirth
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX 77030 and Department of Ophthalmology, University of Florida, Gainesville, FL 32610, USA
| | - Theodore G. Wensel
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX 77030 and Department of Ophthalmology, University of Florida, Gainesville, FL 32610, USA
| | - John H. Wilson
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX 77030 and Department of Ophthalmology, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
207
|
Rahman SH, Maeder ML, Joung JK, Cathomen T. Zinc-finger nucleases for somatic gene therapy: the next frontier. Hum Gene Ther 2011; 22:925-33. [PMID: 21631241 PMCID: PMC3159524 DOI: 10.1089/hum.2011.087] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2011] [Accepted: 06/01/2011] [Indexed: 12/12/2022] Open
Abstract
Zinc-finger nucleases (ZFNs) are a powerful tool that can be used to edit the human genome ad libitum. The technology has experienced remarkable development in the last few years with regard to both the target site specificity and the engineering platforms used to generate zinc-finger proteins. As a result, two phase I clinical trials aimed at knocking out the CCR5 receptor in T cells isolated from HIV patients to protect these lymphocytes from infection with the virus have been initiated. Moreover, ZFNs have been successfully employed to knockout or correct disease-related genes in human stem cells, including hematopoietic precursor cells and induced pluripotent stem cells. Targeted genome engineering approaches in multipotent and pluripotent stem cells hold great promise for future strategies geared toward correcting inborn mutations for personalized cell replacement therapies. This review describes how ZFNs have been applied to models of gene therapy, discusses the opportunities and the risks associated with this novel technology, and suggests future directions for their safe application in therapeutic genome engineering.
Collapse
Affiliation(s)
- Shamim H. Rahman
- Department of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany
| | - Morgan L. Maeder
- Molecular Pathology Unit, Center for Cancer Research, and Center for Computational and Integrative Biology, Massachusetts General Hospital, Charlestown, MA 02129
- Biological and Biomedical Sciences Program, Harvard Medical School, Boston, MA 02115
| | - J. Keith Joung
- Molecular Pathology Unit, Center for Cancer Research, and Center for Computational and Integrative Biology, Massachusetts General Hospital, Charlestown, MA 02129
- Biological and Biomedical Sciences Program, Harvard Medical School, Boston, MA 02115
- Department of Pathology, Harvard Medical School, Boston, MA 02115
| | - Toni Cathomen
- Department of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany
| |
Collapse
|
208
|
Chen F, Pruett-Miller SM, Huang Y, Gjoka M, Duda K, Taunton J, Collingwood TN, Frodin M, Davis GD. High-frequency genome editing using ssDNA oligonucleotides with zinc-finger nucleases. Nat Methods 2011; 8:753-5. [PMID: 21765410 PMCID: PMC3617923 DOI: 10.1038/nmeth.1653] [Citation(s) in RCA: 365] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2011] [Accepted: 06/27/2011] [Indexed: 11/09/2022]
Abstract
Zinc-finger nucleases (ZFNs) have enabled highly efficient gene targeting in multiple cell types and organisms. Here we describe methods for using simple ssDNA oligonucleotides in tandem with ZFNs to efficiently produce human cell lines with three distinct genetic outcomes: (i) targeted point mutation, (ii) targeted genomic deletion of up to 100 kb and (iii) targeted insertion of small genetic elements concomitant with large genomic deletions.
Collapse
Affiliation(s)
- Fuqiang Chen
- Sigma-Aldrich Biotechnology, St. Louis, Missouri, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
209
|
Sargent RG, Kim S, Gruenert DC. Oligo/polynucleotide-based gene modification: strategies and therapeutic potential. Oligonucleotides 2011; 21:55-75. [PMID: 21417933 DOI: 10.1089/oli.2010.0273] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Oligonucleotide- and polynucleotide-based gene modification strategies were developed as an alternative to transgene-based and classical gene targeting-based gene therapy approaches for treatment of genetic disorders. Unlike the transgene-based strategies, oligo/polynucleotide gene targeting approaches maintain gene integrity and the relationship between the protein coding and gene-specific regulatory sequences. Oligo/polynucleotide-based gene modification also has several advantages over classical vector-based homologous recombination approaches. These include essentially complete homology to the target sequence and the potential to rapidly engineer patient-specific oligo/polynucleotide gene modification reagents. Several oligo/polynucleotide-based approaches have been shown to successfully mediate sequence-specific modification of genomic DNA in mammalian cells. The strategies involve the use of polynucleotide small DNA fragments, triplex-forming oligonucleotides, and single-stranded oligodeoxynucleotides to mediate homologous exchange. The primary focus of this review will be on the mechanistic aspects of the small fragment homologous replacement, triplex-forming oligonucleotide-mediated, and single-stranded oligodeoxynucleotide-mediated gene modification strategies as it relates to their therapeutic potential.
Collapse
Affiliation(s)
- R Geoffrey Sargent
- Department of Otolaryngology-Head and Neck Surgery, University of California , San Francisco, California 94115, USA
| | | | | |
Collapse
|
210
|
Silva G, Poirot L, Galetto R, Smith J, Montoya G, Duchateau P, Pâques F. Meganucleases and other tools for targeted genome engineering: perspectives and challenges for gene therapy. Curr Gene Ther 2011; 11:11-27. [PMID: 21182466 PMCID: PMC3267165 DOI: 10.2174/156652311794520111] [Citation(s) in RCA: 252] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2010] [Revised: 12/10/2010] [Accepted: 12/10/2010] [Indexed: 12/17/2022]
Abstract
The importance of safer approaches for gene therapy has been underscored by a series of severe adverse events (SAEs) observed in patients involved in clinical trials for Severe Combined Immune Deficiency Disease (SCID) and Chromic Granulomatous Disease (CGD). While a new generation of viral vectors is in the process of replacing the classical gamma-retrovirus-based approach, a number of strategies have emerged based on non-viral vectorization and/or targeted insertion aimed at achieving safer gene transfer. Currently, these methods display lower efficacies than viral transduction although many of them can yield more than 1% of engineered cells in vitro. Nuclease-based approaches, wherein an endonuclease is used to trigger site-specific genome editing, can significantly increase the percentage of targeted cells. These methods therefore provide a real alternative to classical gene transfer as well as gene editing. However, the first endonuclease to be in clinic today is not used for gene transfer, but to inactivate a gene (CCR5) required for HIV infection. Here, we review these alternative approaches, with a special emphasis on meganucleases, a family of naturally occurring rare-cutting endonucleases, and speculate on their current and future potential.
Collapse
Affiliation(s)
- George Silva
- Cellectis, 102 Avenue Gaston Roussel, 93 235 Romainville, Cedex, France
| | - Laurent Poirot
- Cellectis Genome Surgery, 102 Avenue Gaston Roussel, 93 235 Romainville, Cedex, France
| | - Roman Galetto
- Cellectis Genome Surgery, 102 Avenue Gaston Roussel, 93 235 Romainville, Cedex, France
| | - Julianne Smith
- Cellectis Genome Surgery, 102 Avenue Gaston Roussel, 93 235 Romainville, Cedex, France
| | - Guillermo Montoya
- Macromolecular Crystallography Group, Structural Biology and Biocomputing Programme, Spanish National Cancer Centre (CNIO), Melchor Fdez. Almagro 3, 28029 Madrid, Spain
| | | | - Frédéric Pâques
- Cellectis Genome Surgery, 102 Avenue Gaston Roussel, 93 235 Romainville, Cedex, France
- Cellectis, 102 Avenue Gaston Roussel, 93 235 Romainville, Cedex, France
| |
Collapse
|
211
|
Stoddard BL. Homing endonucleases: from microbial genetic invaders to reagents for targeted DNA modification. Structure 2011; 19:7-15. [PMID: 21220111 PMCID: PMC3038549 DOI: 10.1016/j.str.2010.12.003] [Citation(s) in RCA: 227] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2010] [Revised: 12/14/2010] [Accepted: 12/15/2010] [Indexed: 12/23/2022]
Abstract
Homing endonucleases are microbial DNA-cleaving enzymes that mobilize their own reading frames by generating double strand breaks at specific genomic invasion sites. These proteins display an economy of size, and yet recognize long DNA sequences (typically 20 to 30 base pairs). They exhibit a wide range of fidelity at individual nucleotide positions in a manner that is strongly influenced by host constraints on the coding sequence of the targeted gene. The activity of these proteins leads to site-specific recombination events that can result in the insertion, deletion, mutation, or correction of DNA sequences. Over the past fifteen years, the crystal structures of representatives from several homing endonuclease families have been solved, and methods have been described to create variants of these enzymes that cleave novel DNA targets. Engineered homing endonucleases proteins are now being used to generate targeted genomic modifications for a variety of biotech and medical applications.
Collapse
Affiliation(s)
- Barry L Stoddard
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue N., A3-025, Seattle, WA 98109, USA.
| |
Collapse
|
212
|
Abstract
Herpes simplex virus type 1 (HSV1) is a major health problem. As for most viral diseases, current antiviral treatments are based on the inhibition of viral replication once it has already started. As a consequence, they impair neither the viral cycle at its early stages nor the latent form of the virus, and thus cannot be considered as real preventive treatments. Latent HSV1 virus could be addressed by rare cutting endonucleases, such as meganucleases. With the aim of a proof of concept study, we generated several meganucleases recognizing HSV1 sequences, and assessed their antiviral activity in cultured cells. We demonstrate that expression of these proteins in African green monkey kidney fibroblast (COS-7) and BSR cells inhibits infection by HSV1, at low and moderate multiplicities of infection (MOIs), inducing a significant reduction of the viral load. Furthermore, the remaining viral genomes display a high rate of mutation (up to 16%) at the meganuclease cleavage site, consistent with a mechanism of action based on the cleavage of the viral genome. This specific mechanism of action qualifies meganucleases as an alternative class of antiviral agent, with the potential to address replicative as well as latent DNA viral forms.
Collapse
|
213
|
Abstract
The laboratory rat is rapidly gaining momentum as a mammalian genetic model organism. Although traditional forward genetic approaches are well established, recent technological developments have enabled efficient gene targeting and mutant generation. Here we outline the current status, possibilities and application of these techniques in the rat.
Collapse
Affiliation(s)
- Ruben van Boxtel
- Hubrecht Institute for Developmental Biology and Stem Cell Research, Cancer Genomics Center, Royal Netherlands Academy of Sciences and University Medical Center Utrecht, Uppsalalaan 8, 3584 CT Utrecht, The Netherlands
| | | |
Collapse
|
214
|
Urnov FD, Rebar EJ, Holmes MC, Zhang HS, Gregory PD. Genome editing with engineered zinc finger nucleases. Nat Rev Genet 2010; 11:636-46. [PMID: 20717154 DOI: 10.1038/nrg2842] [Citation(s) in RCA: 1542] [Impact Index Per Article: 102.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Reverse genetics in model organisms such as Drosophila melanogaster, Arabidopsis thaliana, zebrafish and rats, efficient genome engineering in human embryonic stem and induced pluripotent stem cells, targeted integration in crop plants, and HIV resistance in immune cells - this broad range of outcomes has resulted from the application of the same core technology: targeted genome cleavage by engineered, sequence-specific zinc finger nucleases followed by gene modification during subsequent repair. Such 'genome editing' is now established in human cells and a number of model organisms, thus opening the door to a range of new experimental and therapeutic possibilities.
Collapse
Affiliation(s)
- Fyodor D Urnov
- Sangamo BioSciences Inc., Richmond, California 94804, USA
| | | | | | | | | |
Collapse
|
215
|
Ding Z, Zhang J, Shao J. [ERCC1 expression as a predictor of survival after operation in stage I non-small cell lung cancer patients]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2010; 13:522-5. [PMID: 20677653 PMCID: PMC6000703 DOI: 10.3779/j.issn.1009-3419.2010.05.26] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND AND OBJECTIVE Proteins of the nucleotide excision repair pathway can repair DNA damage. The excision repair cross-complementing (ERCC) gene family reduce damagement of DNA by nucleotide excision and repair. The aim of this study is to investigate the expressions of ERCC1 (members of DNA repair gene family) in patients with non-small cell lung cancer (NSCLC) as well as their clinical prognostic significance. METHODS Expression levels ofERCC1 were detected by IHC in 118 stage I NSCLC patients. Kaplan-Meier survival curve, and Cox multivariate regression analysis were used for statistical analysis. RESULTS The patients with high expression of ERCC1 had significantly longer survival time than those with low expression of ERCC1, and Cox multivariate regression analysis showed that expression of RRM1 was an independent prognostic factor for NSCLC patients. CONCLUSION NSCLC patients with high ERCC1 expression have a better survival when compared to patients with low ERCC1 expression. Therefore, an intact DNA repair mechanism may reduce the accumulation of genetic aberrations that are thought to contribute to a tumor malignant potential and therefore the risk of relapse after definitive treatment.
Collapse
Affiliation(s)
- Zhengping Ding
- Department of Thoracic Surgery, Shanghai Chest Hospital/Shanghai Lung Tumor Clinical Medical Center, Shanghai 200030, China
| | | | | |
Collapse
|
216
|
DeKelver RC, Choi VM, Moehle EA, Paschon DE, Hockemeyer D, Meijsing SH, Sancak Y, Cui X, Steine EJ, Miller JC, Tam P, Bartsevich VV, Meng X, Rupniewski I, Gopalan SM, Sun HC, Pitz KJ, Rock JM, Zhang L, Davis GD, Rebar EJ, Cheeseman IM, Yamamoto KR, Sabatini DM, Jaenisch R, Gregory PD, Urnov FD. Functional genomics, proteomics, and regulatory DNA analysis in isogenic settings using zinc finger nuclease-driven transgenesis into a safe harbor locus in the human genome. Genome Res 2010; 20:1133-42. [PMID: 20508142 PMCID: PMC2909576 DOI: 10.1101/gr.106773.110] [Citation(s) in RCA: 244] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2010] [Accepted: 05/11/2010] [Indexed: 11/25/2022]
Abstract
Isogenic settings are routine in model organisms, yet remain elusive for genetic experiments on human cells. We describe the use of designed zinc finger nucleases (ZFNs) for efficient transgenesis without drug selection into the PPP1R12C gene, a "safe harbor" locus known as AAVS1. ZFNs enable targeted transgenesis at a frequency of up to 15% following transient transfection of both transformed and primary human cells, including fibroblasts and hES cells. When added to this locus, transgenes such as expression cassettes for shRNAs, small-molecule-responsive cDNA expression cassettes, and reporter constructs, exhibit consistent expression and sustained function over 50 cell generations. By avoiding random integration and drug selection, this method allows bona fide isogenic settings for high-throughput functional genomics, proteomics, and regulatory DNA analysis in essentially any transformed human cell type and in primary cells.
Collapse
Affiliation(s)
- Russell C. DeKelver
- Sangamo BioSciences, Inc., Point Richmond Tech Center, Richmond, California 94804, USA
| | - Vivian M. Choi
- Sangamo BioSciences, Inc., Point Richmond Tech Center, Richmond, California 94804, USA
| | - Erica A. Moehle
- Sangamo BioSciences, Inc., Point Richmond Tech Center, Richmond, California 94804, USA
| | - David E. Paschon
- Sangamo BioSciences, Inc., Point Richmond Tech Center, Richmond, California 94804, USA
| | - Dirk Hockemeyer
- The Whitehead Institute for Biomedical Research, Cambridge, Massachusetts 02142, USA
| | - Sebastiaan H. Meijsing
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, California 94158, USA
| | - Yasemin Sancak
- The Whitehead Institute for Biomedical Research, Cambridge, Massachusetts 02142, USA
| | - Xiaoxia Cui
- Sigma-Aldrich Research Biotechnology, St. Louis, Missouri 63103, USA
| | - Eveline J. Steine
- The Whitehead Institute for Biomedical Research, Cambridge, Massachusetts 02142, USA
| | - Jeffrey C. Miller
- Sangamo BioSciences, Inc., Point Richmond Tech Center, Richmond, California 94804, USA
| | - Phillip Tam
- Sangamo BioSciences, Inc., Point Richmond Tech Center, Richmond, California 94804, USA
| | - Victor V. Bartsevich
- Sangamo BioSciences, Inc., Point Richmond Tech Center, Richmond, California 94804, USA
| | - Xiangdong Meng
- Sangamo BioSciences, Inc., Point Richmond Tech Center, Richmond, California 94804, USA
| | - Igor Rupniewski
- Sangamo BioSciences, Inc., Point Richmond Tech Center, Richmond, California 94804, USA
| | - Sunita M. Gopalan
- Sangamo BioSciences, Inc., Point Richmond Tech Center, Richmond, California 94804, USA
| | - Helena C. Sun
- Sangamo BioSciences, Inc., Point Richmond Tech Center, Richmond, California 94804, USA
| | - Kathleen J. Pitz
- Sangamo BioSciences, Inc., Point Richmond Tech Center, Richmond, California 94804, USA
| | - Jeremy M. Rock
- Sangamo BioSciences, Inc., Point Richmond Tech Center, Richmond, California 94804, USA
| | - Lei Zhang
- Sangamo BioSciences, Inc., Point Richmond Tech Center, Richmond, California 94804, USA
| | - Gregory D. Davis
- Sigma-Aldrich Research Biotechnology, St. Louis, Missouri 63103, USA
| | - Edward J. Rebar
- Sangamo BioSciences, Inc., Point Richmond Tech Center, Richmond, California 94804, USA
| | - Iain M. Cheeseman
- The Whitehead Institute for Biomedical Research, Cambridge, Massachusetts 02142, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | - Keith R. Yamamoto
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, California 94158, USA
| | - David M. Sabatini
- The Whitehead Institute for Biomedical Research, Cambridge, Massachusetts 02142, USA
| | - Rudolf Jaenisch
- The Whitehead Institute for Biomedical Research, Cambridge, Massachusetts 02142, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | - Philip D. Gregory
- Sangamo BioSciences, Inc., Point Richmond Tech Center, Richmond, California 94804, USA
| | - Fyodor D. Urnov
- Sangamo BioSciences, Inc., Point Richmond Tech Center, Richmond, California 94804, USA
| |
Collapse
|
217
|
Greenwald DL, Cashman SM, Kumar-Singh R. Engineered zinc finger nuclease-mediated homologous recombination of the human rhodopsin gene. Invest Ophthalmol Vis Sci 2010; 51:6374-80. [PMID: 20671268 DOI: 10.1167/iovs.10-5781] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
PURPOSE Novel zinc finger nucleases (ZFNs) were designed to target the human rhodopsin gene and induce homologous recombination of a donor DNA fragment. METHODS Three-finger zinc finger nucleases were designed based on previously published guidelines. To assay for ZFN specificity, the authors generated human embryonic retinoblast cell lines stably expressing a Pro23His rhodopsin, the most common mutation associated with autosomal dominant retinitis pigmentosa in North America. They report quantification of these rhodopsin-specific ZFNs to induce a targeted double-strand break in the human genome, demonstrate their ability to induce homologous recombination of a donor DNA fragment, and report the quantification of the frequency of ZFN-mediated homologous recombination. RESULTS Compared with endogenous homologous recombination, the authors observed a 12-fold increase in homologous recombination and an absolute frequency of ZFN-directed homologous recombination as high as 17% in the human rhodopsin gene. CONCLUSIONS ZFNs are chimeric proteins with significant potential for the treatment of inherited diseases. In this study, the authors report the design of novel ZFNs targeting the human rhodopsin gene. These ZFNs may be useful for the treatment of retinal diseases such as retinitis pigmentosa, one of the most common causes of inherited blindness in the developed world. Herein, they also report on several aspects of donor fragment design and in vitro conditions that facilitate ZFN-mediated homologous recombination.
Collapse
Affiliation(s)
- David L Greenwald
- Department of Ophthalmology, Tufts University School of Medicine, Boston, MA 02111, USA
| | | | | |
Collapse
|
218
|
Olsen PA, Gelazauskaite M, Randøl M, Krauss S. Analysis of illegitimate genomic integration mediated by zinc-finger nucleases: implications for specificity of targeted gene correction. BMC Mol Biol 2010; 11:35. [PMID: 20459736 PMCID: PMC2875229 DOI: 10.1186/1471-2199-11-35] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2009] [Accepted: 05/10/2010] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Formation of site specific genomic double strand breaks (DSBs), induced by the expression of a pair of engineered zinc-finger nucleases (ZFNs), dramatically increases the rates of homologous recombination (HR) between a specific genomic target and a donor plasmid. However, for the safe use of ZFN induced HR in practical applications, possible adverse effects of the technology such as cytotoxicity and genotoxicity need to be well understood. In this work, off-target activity of a pair of ZFNs has been examined by measuring the ratio between HR and illegitimate genomic integration in cells that are growing exponentially, and in cells that have been arrested in the G2/M phase. RESULTS A reporter cell line that contained consensus ZFN binding sites in an enhanced green fluorescent protein (EGFP) reporter gene was used to measure ratios between HR and non-homologous integration of a plasmid template. Both in human cells (HEK 293) containing the consensus ZFN binding sites and in cells lacking the ZFN binding sites, a 3.5 fold increase in the level of illegitimate integration was observed upon ZFN expression. Since the reporter gene containing the consensus ZFN target sites was found to be intact in cells where illegitimate integration had occurred, increased rates of illegitimate integration most likely resulted from the formation of off-target genomic DSBs. Additionally, in a fraction of the ZFN treated cells the co-occurrence of both specific HR and illegitimate integration was observed. As a mean to minimize unspecific effects, cell cycle manipulation of the target cells by induction of a transient G2/M cell cycle arrest was shown to stimulate the activity of HR while having little effect on the levels of illegitimate integration, thus resulting in a nearly eight fold increase in the ratio between the two processes. CONCLUSIONS The demonstration that ZFN expression, in addition to stimulating specific gene targeting by HR, leads to increased rates of illegitimate integration emphasizes the importance of careful characterization of ZFN treated cells. In order to reduce off-target events, reversible cell cycle arrest of the target cells in the G2/M phase is an efficient way for increasing the ratio between specific HR and illegitimate integration.
Collapse
Affiliation(s)
- Petter A Olsen
- Section for Cellular and Genetic Therapy, Institute of Microbiology, Oslo University Hospital, Rikshospitalet, Gausdadalleen 21, 0349 Oslo, Norway
- University of Oslo, 0027 Oslo, Norway
| | - Monika Gelazauskaite
- Section for Cellular and Genetic Therapy, Institute of Microbiology, Oslo University Hospital, Rikshospitalet, Gausdadalleen 21, 0349 Oslo, Norway
- University of Oslo, 0027 Oslo, Norway
| | - Markus Randøl
- Section for Cellular and Genetic Therapy, Institute of Microbiology, Oslo University Hospital, Rikshospitalet, Gausdadalleen 21, 0349 Oslo, Norway
- University of Oslo, 0027 Oslo, Norway
| | - Stefan Krauss
- Section for Cellular and Genetic Therapy, Institute of Microbiology, Oslo University Hospital, Rikshospitalet, Gausdadalleen 21, 0349 Oslo, Norway
- University of Oslo, 0027 Oslo, Norway
| |
Collapse
|
219
|
Petek LM, Russell DW, Miller DG. Frequent endonuclease cleavage at off-target locations in vivo. Mol Ther 2010; 18:983-6. [PMID: 20216527 PMCID: PMC2890094 DOI: 10.1038/mt.2010.35] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2009] [Accepted: 02/10/2010] [Indexed: 11/09/2022] Open
Abstract
Target-site DNA breaks increase recombination frequencies, however, the specificity of the enzymes used to create them remains poorly defined. The location and frequency of off-target cleavage events are especially important when rare-cutting endonucleases are used in clinical settings. Here, we identify noncanonical cleavage sites of I-SceI that are frequently cut in the human genome by localizing adeno-associated virus (AAV) vector-chromosome junctions, demonstrating the importance of in vivo characterization of enzyme cleavage specificity.
Collapse
Affiliation(s)
- Lisa M Petek
- Department of Pediatrics, Division of Genetic Medicine, University of Washington, Seattle, Washington, USA
| | | | | |
Collapse
|
220
|
Gene correction by homologous recombination with zinc finger nucleases in primary cells from a mouse model of a generic recessive genetic disease. Mol Ther 2010; 18:1103-10. [PMID: 20389291 DOI: 10.1038/mt.2010.57] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Zinc Finger nucleases (ZFNs) have been used to create precise genome modifications at frequencies that might be therapeutically useful in gene therapy. We created a mouse model of a generic recessive genetic disease to establish a preclinical system to develop the use of ZFN-mediated gene correction for gene therapy. We knocked a mutated GFP gene into the ROSA26 locus in murine embryonic stem (ES) cells and used these cells to create a transgenic mouse. We used ZFNs to determine the frequency of gene correction by gene targeting in different primary cells from this model. We achieved targeting frequencies from 0.17 to 6% in different cell types, including primary fibroblasts and astrocytes. We demonstrate that ex vivo gene-corrected fibroblasts can be transplanted back into a mouse where they retained the corrected phenotype. In addition, we achieved targeting frequencies of over 1% in ES cells, and the targeted ES cells retained the ability to differentiate into cell types from all three germline lineages. In summary, potentially therapeutically relevant frequencies of ZFN-mediated gene targeting can be achieved in a variety of primary cells and these cells can then be transplanted back into a recipient.
Collapse
|
221
|
Grizot S, Epinat JC, Thomas S, Duclert A, Rolland S, Pâques F, Duchateau P. Generation of redesigned homing endonucleases comprising DNA-binding domains derived from two different scaffolds. Nucleic Acids Res 2010; 38:2006-18. [PMID: 20026587 PMCID: PMC2847234 DOI: 10.1093/nar/gkp1171] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2009] [Revised: 11/27/2009] [Accepted: 11/27/2009] [Indexed: 11/14/2022] Open
Abstract
Homing endonucleases have become valuable tools for genome engineering. Their sequence recognition repertoires can be expanded by modifying their specificities or by creating chimeric proteins through domain swapping between two subdomains of different homing endonucleases. Here, we show that these two approaches can be combined to create engineered meganucleases with new specificities. We demonstrate the modularity of the chimeric DmoCre meganuclease previously described, by successfully assembling mutants with locally altered specificities affecting both I-DmoI and I-CreI subdomains in order to create active meganucleases with altered specificities. Moreover these new engineered DmoCre variants appear highly specific and present a low toxicity level, similar to I-SceI, and can induce efficient homologous recombination events in mammalian cells. The DmoCre based meganucleases can therefore offer new possibilities for various genome engineering applications.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Philippe Duchateau
- Cellectis SA, 102 Avenue Gaston Roussel, 93235 Romainville Cedex, France
| |
Collapse
|
222
|
Efficient repair of DNA double-strand breaks in malignant cells with structural instability. Mutat Res 2010; 683:115-22. [PMID: 19909760 DOI: 10.1016/j.mrfmmm.2009.10.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2009] [Revised: 10/22/2009] [Accepted: 10/30/2009] [Indexed: 01/08/2023]
Abstract
Aberrant repair of DNA double-strand breaks (DSBs) is thought to be important in the generation of gross chromosomal rearrangements (GCRs). To examine how DNA DSBs might lead to GCRs, we investigated the repair of a single DNA DSB in a structurally unstable cell line. An I-SceI recognition site was introduced into OVCAR-8 cells between a constitutive promoter (EF1alpha) and the Herpes simplex virus thymidine kinase (TK) gene, which confers sensitivity to gancyclovir (GCV). Expression of I-SceI in these cells caused a single DSB. Clones with aberrant repair could acquire resistance to GCV by separation of the EF1alpha promoter from the TK gene, or deletion of either the EF1alpha promoter or the TK gene. All mutations that we identified were interstitial deletions. Treatment of cells with etoposide or bleomycin, agents known to produce DNA DSBs following expression of I-SceI also did not generate GCRs. Because we identified solely interstitial deletions using the aforementioned negative selection system, we developed a positive selection system to produce GCR. A construct containing an I-SceI restriction site immediately followed by a hygromycin phosphotransferase cDNA, with no promoter, was stably integrated into OVCAR-8 cells. DNA DSBs were produced by an I-SceI expression vector. None of the hygromycin resistant clones recovered had linked the hygromycin phosphotransferase cDNA to an endogenous promoter, but had instead captured a portion of the I-SceI expression vector. These results indicate that even in a structurally unstable malignant cell line, the majority of DNA DSBs are repaired by religation of the two broken chromosome ends, without the introduction of a GCR.
Collapse
|
223
|
Moynahan ME, Jasin M. Mitotic homologous recombination maintains genomic stability and suppresses tumorigenesis. Nat Rev Mol Cell Biol 2010; 11:196-207. [PMID: 20177395 PMCID: PMC3261768 DOI: 10.1038/nrm2851] [Citation(s) in RCA: 703] [Impact Index Per Article: 46.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Mitotic homologous recombination promotes genome stability through the precise repair of DNA double-strand breaks and other lesions that are encountered during normal cellular metabolism and from exogenous insults. As a result, homologous recombination repair is essential during proliferative stages in development and during somatic cell renewal in adults to protect against cell death and mutagenic outcomes from DNA damage. Mutations in mammalian genes encoding homologous recombination proteins, including BRCA1, BRCA2 and PALB2, are associated with developmental abnormalities and tumorigenesis. Recent advances have provided a clearer understanding of the connections between these proteins and of the key steps of homologous recombination and DNA strand exchange.
Collapse
|
224
|
A dual-activation, adenoviral-based system for the controlled induction of DNA double-strand breaks by the restriction endonuclease SacI. Biotechniques 2010; 47:847-54. [PMID: 19852768 DOI: 10.2144/000113237] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Spontaneous damage to DNA is frequent and may lead to cell death, cell senescence, or mutations. DNA double-strand breaks (DSBs) are of special interest because they are highly toxic and have been implicated in neurodegeneration, cancer, and aging. Until now, there has not been a reliable system allowing tunable induction of random DSBs without affecting other macromolecules or cell functions. Here, we describe an adenoviral-based, doxycycline-mediated, and tamoxifen-dependent system for quantitative introduction of DSBs in mammalian cells. We generated a single adenoviral vector containing a tet-inducible, composite SacI restriction endonuclease/estrogen receptor (ERT2) gene, and a constitutively expressed reverse transactivator (rtTA) gene. Transduced mouse embryonic fibroblasts-as well as mouse liver cells in vivo-demonstrated a high level of DSBs in response to treatment with doxycycline and tamoxifen. We show that the amount of induced DSBs can be titrated by doxycycline dose and duration of treatment. This system should be useful for studying the processing of randomly induced DSBs and their effects on cell fate, without the side effects normally associated with radiation or chemical treatment.
Collapse
|
225
|
Wu CC, Liu MT, Chang YT, Fang CY, Chou SP, Liao HW, Kuo KL, Hsu SL, Chen YR, Wang PW, Chen YL, Chuang HY, Lee CH, Chen M, Wayne Chang WS, Chen JY. Epstein-Barr virus DNase (BGLF5) induces genomic instability in human epithelial cells. Nucleic Acids Res 2009; 38:1932-49. [PMID: 20034954 PMCID: PMC2847232 DOI: 10.1093/nar/gkp1169] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Epstein–Barr Virus (EBV) DNase (BGLF5) is an alkaline nuclease and has been suggested to be important in the viral life cycle. However, its effect on host cells remains unknown. Serological and histopathological studies implied that EBV DNase seems to be correlated with carcinogenesis. Therefore, we investigate the effect of EBV DNase on epithelial cells. Here, we report that expression of EBV DNase induces increased formation of micronucleus, an indicator of genomic instability, in human epithelial cells. We also demonstrate, using γH2AX formation and comet assay, that EBV DNase induces DNA damage. Furthermore, using host cell reactivation assay, we find that EBV DNase expression repressed damaged DNA repair in various epithelial cells. Western blot and quantitative PCR analyses reveal that expression of repair-related genes is reduced significantly in cells expressing EBV DNase. Host shut-off mutants eliminate shut-off expression of repair genes and repress damaged DNA repair, suggesting that shut-off function of BGLF5 contributes to repression of DNA repair. In addition, EBV DNase caused chromosomal aberrations and increased the microsatellite instability (MSI) and frequency of genetic mutation in human epithelial cells. Together, we propose that EBV DNase induces genomic instability in epithelial cells, which may be through induction of DNA damage and also repression of DNA repair, subsequently increases MSI and genetic mutations, and may contribute consequently to the carcinogenesis of human epithelial cells.
Collapse
Affiliation(s)
- Chung-Chun Wu
- National Institute of Cancer Research, National Health Research Institutes, Zhunan, Taiwan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
226
|
Manthey GM, Naik N, Bailis AM. Msh2 blocks an alternative mechanism for non-homologous tail removal during single-strand annealing in Saccharomyces cerevisiae. PLoS One 2009; 4:e7488. [PMID: 19834615 PMCID: PMC2759526 DOI: 10.1371/journal.pone.0007488] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2009] [Accepted: 09/25/2009] [Indexed: 11/19/2022] Open
Abstract
Chromosomal translocations are frequently observed in cells exposed to agents that cause DNA double-strand breaks (DSBs), such as ionizing radiation and chemotherapeutic drugs, and are often associated with tumors in mammals. Recently, translocation formation in the budding yeast, Saccharomyces cerevisiae, has been found to occur at high frequencies following the creation of multiple DSBs adjacent to repetitive sequences on non-homologous chromosomes. The genetic control of translocation formation and the chromosome complements of the clones that contain translocations suggest that translocation formation occurs by single-strand annealing (SSA). Among the factors important for translocation formation by SSA is the central mismatch repair (MMR) and homologous recombination (HR) factor, Msh2. Here we describe the effects of several msh2 missense mutations on translocation formation that suggest that Msh2 has separable functions in stabilizing annealed single strands, and removing non-homologous sequences from their ends. Additionally, interactions between the msh2 alleles and a null allele of RAD1, which encodes a subunit of a nuclease critical for the removal of non-homologous tails suggest that Msh2 blocks an alternative mechanism for removing these sequences. These results suggest that Msh2 plays multiple roles in the formation of chromosomal translocations following acute levels of DNA damage.
Collapse
Affiliation(s)
- Glenn M. Manthey
- Division of Molecular Biology, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, United States of America
| | - Nilan Naik
- Scripps College Post-Baccalaureate Premedical Program, Claremont, California, United States of America
| | - Adam M. Bailis
- Division of Molecular Biology, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, United States of America
- * E-mail:
| |
Collapse
|
227
|
Abstract
DNA chromosomal DSBs (double-strand breaks) are potentially hazardous DNA lesions, and their accurate repair is essential for the successful maintenance and propagation of genetic information. Two major pathways have evolved to repair DSBs: HR (homologous recombination) and NHEJ (non-homologous end-joining). Depending on the context in which the break is encountered, HR and NHEJ may either compete or co-operate to fix DSBs in eukaryotic cells. Defects in either pathway are strongly associated with human disease, including immunodeficiency and cancer predisposition. Here we review the current knowledge of how NHEJ and HR are controlled in somatic mammalian cells, and discuss the role of the chromatin context in regulating each pathway. We also review evidence for both co-operation and competition between the two pathways.
Collapse
|
228
|
Kikuchi K, Abdel-Aziz HI, Taniguchi Y, Yamazoe M, Takeda S, Hirota K. Bloom DNA helicase facilitates homologous recombination between diverged homologous sequences. J Biol Chem 2009; 284:26360-7. [PMID: 19661064 DOI: 10.1074/jbc.m109.029348] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Bloom syndrome caused by inactivation of the Bloom DNA helicase (Blm) is characterized by increases in the level of sister chromatid exchange, homologous recombination (HR) associated with cross-over. It is therefore believed that Blm works as an anti-recombinase. Meanwhile, in Drosophila, DmBlm is required specifically to promote the synthesis-dependent strand anneal (SDSA), a type of HR not associating with cross-over. However, conservation of Blm function in SDSA through higher eukaryotes has been a matter of debate. Here, we demonstrate the function of Blm in SDSA type HR in chicken DT40 B lymphocyte line, where Ig gene conversion diversifies the immunoglobulin V gene through intragenic HR between diverged homologous segments. This reaction is initiated by the activation-induced cytidine deaminase enzyme-mediated uracil formation at the V gene, which in turn converts into abasic site, presumably leading to a single strand gap. Ig gene conversion frequency was drastically reduced in BLM(-/-) cells. In addition, BLM(-/-) cells used limited donor segments harboring higher identity compared with other segments in Ig gene conversion event, suggesting that Blm can promote HR between diverged sequences. To further understand the role of Blm in HR between diverged homologous sequences, we measured the frequency of gene targeting induced by an I-SceI-endonuclease-mediated double-strand break. BLM(-/-) cells showed a severer defect in the gene targeting frequency as the number of heterologous sequences increased at the double-strand break site. Conversely, the overexpression of Blm, even an ATPase-defective mutant, strongly stimulated gene targeting. In summary, Blm promotes HR between diverged sequences through a novel ATPase-independent mechanism.
Collapse
Affiliation(s)
- Koji Kikuchi
- Department of Radiation Genetics, Graduate School of Medicine, Kyoto University, Yoshidakonoe, Kyoto 606-8501, Japan
| | | | | | | | | | | |
Collapse
|
229
|
Grizot S, Smith J, Daboussi F, Prieto J, Redondo P, Merino N, Villate M, Thomas S, Lemaire L, Montoya G, Blanco FJ, Pâques F, Duchateau P. Efficient targeting of a SCID gene by an engineered single-chain homing endonuclease. Nucleic Acids Res 2009; 37:5405-19. [PMID: 19584299 PMCID: PMC2760784 DOI: 10.1093/nar/gkp548] [Citation(s) in RCA: 123] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Sequence-specific endonucleases recognizing long target sequences are emerging as powerful tools for genome engineering. These endonucleases could be used to correct deleterious mutations or to inactivate viruses, in a new approach to molecular medicine. However, such applications are highly demanding in terms of safety. Mutations in the human RAG1 gene cause severe combined immunodeficiency (SCID). Using the I-CreI dimeric LAGLIDADG meganuclease as a scaffold, we describe here the engineering of a series of endonucleases cleaving the human RAG1 gene, including obligate heterodimers and single-chain molecules. We show that a novel single-chain design, in which two different monomers are linked to form a single molecule, can induce high levels of recombination while safeguarding more effectively against potential genotoxicity. We provide here the first demonstration that an engineered meganuclease can induce targeted recombination at an endogenous locus in up to 6% of transfected human cells. These properties rank this new generation of endonucleases among the best molecular scissors available for genome surgery strategies, potentially avoiding the deleterious effects of previous gene therapy approaches.
Collapse
Affiliation(s)
- Sylvestre Grizot
- Cellectis SA, Cellectis Genome Surgery, 93235 Romainville, France
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
230
|
Chromosomal translocations induced at specified loci in human stem cells. Proc Natl Acad Sci U S A 2009; 106:10620-5. [PMID: 19549848 DOI: 10.1073/pnas.0902076106] [Citation(s) in RCA: 164] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The precise genetic manipulation of stem and precursor cells offers extraordinary potential for the analysis, prevention, and treatment of human malignancies. Chromosomal translocations are hallmarks of several tumor types where they are thought to have arisen in stem or precursor cells. Although approaches exist to study factors involved in translocation formation in mouse cells, approaches in human cells have been lacking, especially in relevant cell types. The technology of zinc finger nucleases (ZFNs) allows DNA double-strand breaks (DSBs) to be introduced into specified chromosomal loci. We harnessed this technology to induce chromosomal translocations in human cells by generating concurrent DSBs at 2 endogenous loci, the PPP1R12C/p84 gene on chromosome 19 and the IL2Rgamma gene on the X chromosome. Translocation breakpoint junctions for t(19;X) were detected with nested quantitative PCR in a high throughput 96-well format using denaturation curves and DNA sequencing in a variety of human cell types, including embryonic stem (hES) cells and hES cell-derived mesenchymal precursor cells. Although readily detected, translocations were less frequent than repair of a single DSB by gene targeting or nonhomologous end-joining, neither of which leads to gross chromosomal rearrangements. While previous studies have relied on laborious genetic modification of cells and extensive growth in culture, the approach described in this report is readily applicable to primary human cells, including multipotent and pluripotent cells, to uncover both the underlying mechanisms and phenotypic consequences of targeted translocations and other genomic rearrangements.
Collapse
|
231
|
Glover L, Horn D. Site-specific DNA double-strand breaks greatly increase stable transformation efficiency in Trypanosoma brucei. Mol Biochem Parasitol 2009; 166:194-7. [PMID: 19459229 PMCID: PMC2691778 DOI: 10.1016/j.molbiopara.2009.03.010] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Genetic manipulation in African trypanosomes typically relies upon electroporation with chromosomal integration of DNA constructs by homologous recombination. Relatively little is known about chromosomal recombination and repair in these organisms however and low transformation efficiency and position effects can limit forward genetic approaches. In yeast and mammalian cells, site-specific DNA double-strand breaks (DSBs) stimulate targeted integration through homologous recombination-based repair where the exogenous DNA serves as the template. We have explored the effect of DSBs on targeted integration in bloodstream-form Trypanosoma brucei, focusing on the ribosomal RNA-spacer target commonly used to integrate recombinant constructs. DSB-repair within the ribosomal RNA tandem gene-repeats is likely dominated by single-strand annealing allowing approximately 80% of cells to survive the break. In the presence of exogenous DNA, transformation efficiency is increased approximately 250-fold by DSB-induction. In the example presented, more than 1% of cells that survive the procedure were transformed generating 80,000 transformants from a typical experiment.
Collapse
Affiliation(s)
- Lucy Glover
- London School of Hygiene & Tropical Medicine, Keppel Street, London, WC1E 7HT, UK
| | | |
Collapse
|
232
|
McConnell Smith A, Takeuchi R, Pellenz S, Davis L, Maizels N, Monnat RJ, Stoddard BL. Generation of a nicking enzyme that stimulates site-specific gene conversion from the I-AniI LAGLIDADG homing endonuclease. Proc Natl Acad Sci U S A 2009; 106:5099-104. [PMID: 19276110 PMCID: PMC2664052 DOI: 10.1073/pnas.0810588106] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2008] [Indexed: 11/18/2022] Open
Abstract
Homing endonucleases stimulate gene conversion by generating site-specific DNA double-strand breaks that are repaired by homologous recombination. These enzymes are potentially valuable tools for targeted gene correction and genome engineering. We have engineered a variant of the I-AniI homing endonuclease that nicks its cognate target site. This variant contains a mutation of a basic residue essential for proton transfer and solvent activation in one active site. The cleavage mechanism, DNA-binding affinity, and substrate specificity profile of the nickase are similar to the wild-type enzyme. I-AniI nickase stimulates targeted gene correction in human cells, in cis and in trans, at approximately 1/4 the efficiency of the wild-type enzyme. The development of sequence-specific nicking enzymes like the I-AniI nickase will facilitate comparative analyses of DNA repair and mutagenesis induced by single- or double-strand breaks.
Collapse
Affiliation(s)
- Audrey McConnell Smith
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue, North Seattle, WA 98109
- Graduate Program in Molecular and Cellular Biology, University of Washington, Seattle, WA 98195
- Northwest Genome Engineering Consortium, Seattle, WA 98101; and
| | - Ryo Takeuchi
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue, North Seattle, WA 98109
- Northwest Genome Engineering Consortium, Seattle, WA 98101; and
| | - Stefan Pellenz
- Northwest Genome Engineering Consortium, Seattle, WA 98101; and
- Departments of Pathology and Genome Sciences, and
| | - Luther Davis
- Northwest Genome Engineering Consortium, Seattle, WA 98101; and
- Immunology and Biochemisty, University of Washington Medical School, Seattle, WA 98195
| | - Nancy Maizels
- Graduate Program in Molecular and Cellular Biology, University of Washington, Seattle, WA 98195
- Northwest Genome Engineering Consortium, Seattle, WA 98101; and
- Immunology and Biochemisty, University of Washington Medical School, Seattle, WA 98195
| | - Raymond J. Monnat
- Graduate Program in Molecular and Cellular Biology, University of Washington, Seattle, WA 98195
- Northwest Genome Engineering Consortium, Seattle, WA 98101; and
- Departments of Pathology and Genome Sciences, and
| | - Barry L. Stoddard
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue, North Seattle, WA 98109
- Graduate Program in Molecular and Cellular Biology, University of Washington, Seattle, WA 98195
- Northwest Genome Engineering Consortium, Seattle, WA 98101; and
| |
Collapse
|
233
|
Chen Z, Wen F, Sun N, Zhao H. Directed evolution of homing endonuclease I-SceI with altered sequence specificity. Protein Eng Des Sel 2009; 22:249-56. [PMID: 19176595 DOI: 10.1093/protein/gzp001] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Homing endonucleases recognize specific long DNA sequences and catalyze double-stranded breaks that significantly stimulate homologous recombination, representing an attractive tool for genome targeting and editing. We previously described a two-plasmid selection system that couples enzymatic DNA cleavage with the survival of host cells, and enables directed evolution of homing endonucleases with altered cleavage sequence specificity. Using this selection system, we successfully evolved mutant I-SceI homing endonucleases with greatly increased cleavage activity towards a new target DNA sequence that differs from the wild-type cleavage sequence by 4 bp. The most highly evolved mutant showed a survival rate approximately 100-fold higher than that of wild-type I-SceI enzyme. The degree of selectivity displayed by a mutant isolated from one round of saturation mutagenesis for the new target sequence is comparable to that of wild-type I-SceI for the natural sequence. These results highlight the ability and efficiency of our selection system for engineering homing endonucleases with novel DNA cleavage specificities. The mutant identified from this study can potentially be used in vivo for targeting the new cleavage sequence within genomic DNA.
Collapse
Affiliation(s)
- Zhilei Chen
- Center for Biophysics and Computational Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | | | | | | |
Collapse
|
234
|
Mao Z, Bozzella M, Seluanov A, Gorbunova V. DNA repair by nonhomologous end joining and homologous recombination during cell cycle in human cells. Cell Cycle 2008; 7:2902-6. [PMID: 18769152 PMCID: PMC2754209 DOI: 10.4161/cc.7.18.6679] [Citation(s) in RCA: 481] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
DNA double-strand breaks (DSBs) are dangerous lesions that can lead to potentially oncogenic genomic rearrangements or cell death. The two major pathways for repair of DSBs are nonhomologous end joining (NHEJ) and homologous recombination (HR). NHEJ is an intrinsically error-prone pathway while HR results in accurate repair. To understand the origin of genomic instability in human cells it is important to know the contribution of each DSB repair pathway. Studies of rodent cells and human cancer cell lines have shown that the choice between NHEJ or HR pathways depends on cell cycle stage. Surprisingly, cell cycle regulation of DSB repair has not been examined in normal human cells with intact cell cycle checkpoints. Here we measured the efficiency of NHEJ and HR at different cell cycle stages in hTERT-immortalized diploid human fibroblasts. We utilized cells with chromosomally-integrated fluorescent reporter cassettes, in which a unique DSB is introduced by a rare-cutting endonuclease. We show that NHEJ is active throughout the cell cycle, and its activity increases as cells progress from G1 to G2/M (G1 < S < G2/M). HR is nearly absent in G1, most active in the S phase, and declines in G2/M. Thus, in G2/M NHEJ is elevated, while HR is on decline. This is in contrast to a general belief that NHEJ is most active in G1, while HR is active in S, G2 and M. The overall efficiency of NHEJ was higher than HR at all cell cycle stages. We conclude that human somatic cells utilize error-prone NHEJ as the major DSB repair pathway at all cell cycle stages, while HR is used, primarily, in the S phase.
Collapse
Affiliation(s)
| | | | - Andrei Seluanov
- Department of Biology, University of Rochester, Rochester, New York 14627, USA
| | - Vera Gorbunova
- Department of Biology, University of Rochester, Rochester, New York 14627, USA
| |
Collapse
|
235
|
Affiliation(s)
- John H Wilson
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA.
| |
Collapse
|
236
|
Nergadze SG, Santagostino MA, Salzano A, Mondello C, Giulotto E. Contribution of telomerase RNA retrotranscription to DNA double-strand break repair during mammalian genome evolution. Genome Biol 2008; 8:R260. [PMID: 18067655 PMCID: PMC2246262 DOI: 10.1186/gb-2007-8-12-r260] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2007] [Revised: 11/28/2007] [Accepted: 12/07/2007] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND In vertebrates, tandem arrays of TTAGGG hexamers are present at both telomeres and intrachromosomal sites (interstitial telomeric sequences (ITSs)). We previously showed that, in primates, ITSs were inserted during the repair of DNA double-strand breaks and proposed that they could arise from either the capture of telomeric fragments or the action of telomerase. RESULTS An extensive comparative analysis of two primate (Homo sapiens and Pan troglodytes) and two rodent (Mus musculus and Rattus norvegicus) genomes allowed us to describe organization and insertion mechanisms of all the informative ITSs present in the four species. Two novel observations support the hypothesis of telomerase involvement in ITS insertion: in a highly significant fraction of informative loci, the ITSs were introduced at break sites where a few nucleotides homologous to the telomeric hexamer were exposed; in the rodent genomes, complex ITS loci are present in which a retrotranscribed fragment of the telomerase RNA, far away from the canonical template, was inserted together with the telomeric repeats. Moreover, mutational analysis of the TTAGGG arrays in the different species suggests that they were inserted as exact telomeric hexamers, further supporting the participation of telomerase in ITS formation. CONCLUSION These results strongly suggest that telomerase was utilized, in some instances, for the repair of DNA double-strand breaks occurring in the genomes of rodents and primates during evolution. The presence, in the rodent genomes, of sequences retrotranscribed from the telomerase RNA strengthens the hypothesis of the origin of telomerase from an ancient retrotransposon.
Collapse
Affiliation(s)
- Solomon G Nergadze
- Dipartimento di Genetica e Microbiologia 'Adriano Buzzati-Traverso', Università degli Studi di Pavia, Via Ferrata, 27100 Pavia, Italy
| | | | | | | | | |
Collapse
|
237
|
Majumdar A, Muniandy PA, Liu J, Liu JL, Liu ST, Cuenoud B, Seidman MM. Targeted gene knock in and sequence modulation mediated by a psoralen-linked triplex-forming oligonucleotide. J Biol Chem 2008; 283:11244-52. [PMID: 18303025 DOI: 10.1074/jbc.m800607200] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Information from exogenous donor DNA can be introduced into the genome via homology-directed repair (HDR) pathways. These pathways are stimulated by double strand breaks and by DNA damage such as interstrand cross-links. We have employed triple helix-forming oligonucleotides linked to psoralen (pso-TFO) to introduce a DNA interstrand cross-link at a specific site in the genome of living mammalian cells. Co-introduction of duplex DNA with target region homology resulted in precise knock in of the donor at frequencies 2-3 orders of magnitude greater than with donor alone. Knock-in was eliminated in cells deficient in ERCC1-XPF, which is involved in recombinational pathways as well as cross-link repair. Separately, single strand oligonucleotide donors (SSO) were co-introduced with the pso-TFO. These were 10-fold more active than the duplex knock-in donor. SSO efficacy was further elevated in cells deficient in ERCC1-XPF, in contrast to the duplex donor. Resected single strand ends have been implicated as critical intermediates in sequence modulation by SSO, as well as duplex donor knock in. We asked whether there would be a competition between the donor species for these ends if both were present with the pso-TFO. The frequency of duplex donor knock in was unaffected by a 100-fold molar excess of the SSO. The same result was obtained when the homing endonuclease I-SceI was used to initiate HDR at the target site. We conclude that the entry of double strand breaks into distinct HDR pathways is controlled by factors other than the nucleic acid partners in those pathways.
Collapse
Affiliation(s)
- Alokes Majumdar
- Laboratory of Molecular Gerontology, NIA, National Institutes of Health, Baltimore, Maryland 21224, USA
| | | | | | | | | | | | | |
Collapse
|
238
|
Fajardo-Sanchez E, Stricher F, Pâques F, Isalan M, Serrano L. Computer design of obligate heterodimer meganucleases allows efficient cutting of custom DNA sequences. Nucleic Acids Res 2008; 36:2163-73. [PMID: 18276641 PMCID: PMC2367722 DOI: 10.1093/nar/gkn059] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Meganucleases cut long (>12 bp) unique sequences in genomes and can be used to induce targeted genome engineering by homologous recombination in the vicinity of their cleavage site. However, the use of natural meganucleases is limited by the repertoire of their target sequences, and considerable efforts have been made to engineer redesigned meganucleases cleaving chosen targets. Homodimeric meganucleases such as I-CreI have provided a scaffold, but can only be modified to recognize new quasi-palindromic DNA sequences, limiting their general applicability. Other groups have used dimer-interface redesign and peptide linkage to control heterodimerization between related meganucleases such as I-DmoI and I-CreI, but until now there has been no application of this aimed specifically at the scaffolds from existing combinatorial libraries of I-CreI. Here, we show that engineering meganucleases to form obligate heterodimers results in functional endonucleases that cut non-palindromic sequences. The protein design algorithm (FoldX v2.7) was used to design specific heterodimer interfaces between two meganuclease monomers, which were themselves engineered to recognize different DNA sequences. The new monomers favour functional heterodimer formation and prevent homodimer site recognition. This design massively increases the potential repertoire of DNA sequences that can be specifically targeted by designed I-CreI meganucleases and opens the way to safer targeted genome engineering.
Collapse
Affiliation(s)
- Emmanuel Fajardo-Sanchez
- Structural Biology and Biocomputing Program, European Molecular Biology Laboratory (EMBL), Meyerhofstrasse 1, D-69117 Heidelberg, Germany
| | | | | | | | | |
Collapse
|
239
|
Eklund JL, Ulge UY, Eastberg J, Monnat RJ. Altered target site specificity variants of the I-PpoI His-Cys box homing endonuclease. Nucleic Acids Res 2007; 35:5839-50. [PMID: 17720708 PMCID: PMC2034468 DOI: 10.1093/nar/gkm624] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
We used a yeast one-hybrid assay to isolate and characterize variants of the eukaryotic homing endonuclease I-PpoI that were able to bind a mutant, cleavage-resistant I-PpoI target or ‘homing’ site DNA in vivo. Native I-PpoI recognizes and cleaves a semi-palindromic 15-bp target site with high specificity in vivo and in vitro. This target site is present in the 28S or equivalent large subunit rDNA genes of all eukaryotes. I-PpoI variants able to bind mutant target site DNA had from 1 to 8 amino acid substitutions in the DNA–protein interface. Biochemical characterization of these proteins revealed a wide range of site–binding affinities and site discrimination. One-third of variants were able to cleave target site DNA, but there was no systematic relationship between site-binding affinity and site cleavage. Computational modeling of several variants provided mechanistic insight into how amino acid substitutions that contact, or are adjacent to, specific target site DNA base pairs determine I-PpoI site-binding affinity and site discrimination, and may affect cleavage efficiency.
Collapse
Affiliation(s)
- Jennifer L. Eklund
- Department of Genome Sciences, Department of Pathology, the Molecular and Cellular Biology Program, University of Washington, Seattle, WA and Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Umut Y. Ulge
- Department of Genome Sciences, Department of Pathology, the Molecular and Cellular Biology Program, University of Washington, Seattle, WA and Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Jennifer Eastberg
- Department of Genome Sciences, Department of Pathology, the Molecular and Cellular Biology Program, University of Washington, Seattle, WA and Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Raymond J. Monnat
- Department of Genome Sciences, Department of Pathology, the Molecular and Cellular Biology Program, University of Washington, Seattle, WA and Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- *To whom correspondence should be addressed. 206 616 7392206 543 3967
| |
Collapse
|
240
|
Stephanou NC, Gao F, Bongiorno P, Ehrt S, Schnappinger D, Shuman S, Glickman MS. Mycobacterial nonhomologous end joining mediates mutagenic repair of chromosomal double-strand DNA breaks. J Bacteriol 2007; 189:5237-46. [PMID: 17496093 PMCID: PMC1951864 DOI: 10.1128/jb.00332-07] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2007] [Accepted: 04/30/2007] [Indexed: 12/18/2022] Open
Abstract
Bacterial nonhomologous end joining (NHEJ) is a recently described DNA repair pathway best characterized in mycobacteria. Bacterial NHEJ proteins LigD and Ku have been analyzed biochemically, and their roles in linear plasmid repair in vivo have been verified genetically; yet the contributions of NHEJ to repair of chromosomal DNA damage are unknown. Here we use an extensive set of NHEJ- and homologous recombination (HR)-deficient Mycobacterium smegmatis strains to probe the importance of HR and NHEJ in repairing diverse types of chromosomal DNA damage. An M. smegmatis Delta recA Delta ku double mutant has no apparent growth defect in vitro. Loss of the NHEJ components Ku and LigD had no effect on sensitivity to UV radiation, methyl methanesulfonate, or quinolone antibiotics. NHEJ deficiency had no effect on sensitivity to ionizing radiation in logarithmic- or early-stationary-phase cells but was required for ionizing radiation resistance in late stationary phase in 7H9 but not LB medium. In addition, NHEJ components were required for repair of I-SceI mediated chromosomal double-strand breaks (DSBs), and in the absence of HR, the NHEJ pathway rapidly mutates the chromosomal break site. The molecular outcomes of NHEJ-mediated chromosomal DSB repair involve predominantly single-nucleotide insertions at the break site, similar to previous findings using plasmid substrates. These findings demonstrate that prokaryotic NHEJ is specifically required for DSB repair in late stationary phase and can mediate mutagenic repair of homing endonuclease-generated chromosomal DSBs.
Collapse
Affiliation(s)
- Nicolas C Stephanou
- Immunology Program, Memorial Sloan-Kettering Cancer Center, New York, New York 10021, USA
| | | | | | | | | | | | | |
Collapse
|
241
|
Francis R, Richardson C. Multipotent hematopoietic cells susceptible to alternative double-strand break repair pathways that promote genome rearrangements. Genes Dev 2007; 21:1064-74. [PMID: 17473170 PMCID: PMC1855232 DOI: 10.1101/gad.1522807] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Chromosomal translocations are a hallmark of hematopoietic malignancies. The initial molecular events or pathways that lead to translocations in hematopoietic cells are largely unknown, particularly in the stem cell-enriched population postulated to be the initial target for these events. We used in vitro differentiation of mouse embryonic stem (ES) cells combined with an I-SceI endonuclease double-strand break (DSB) repair assay to determine the relative susceptibility of isogenic hematopoietic subpopulations to DSB-induced translocations and the mechanisms that generate them. DSB-induced reciprocal translocations were frequently observed in multipotent progenitors but significantly suppressed in actively proliferating myeloid cells. Most reciprocal translocations in multipotent progenitors resulted from repair consistent with single-strand annealing followed by gap repair. Overexpression of Rad51, a protein central to DNA strand exchange and recombination, did not further increase the frequency of recovered translocations but did increase the frequency of long-tract gene conversion events associated with loss of heterozygosity and tandem duplications. These data directly demonstrate that hematopoietic multipotent progenitor cells are particularly susceptible to the formation of chromosomal rearrangements analogous to those observed in human hematopoietic malignancies. This particular subpopulation apparently represents a window of opportunity for the initiation of potentially oncogenic events following DNA damage.
Collapse
Affiliation(s)
- Richard Francis
- Institute for Cancer Genetics, Department of Pathology, Columbia University, New York, New York 10032, USA
| | - Christine Richardson
- Institute for Cancer Genetics, Department of Pathology, Columbia University, New York, New York 10032, USA
- Corresponding author.E-MAIL ; FAX (704) 687-3128
| |
Collapse
|
242
|
Soutoglou E, Dorn JF, Sengupta K, Jasin M, Nussenzweig A, Ried T, Danuser G, Misteli T. Positional stability of single double-strand breaks in mammalian cells. Nat Cell Biol 2007; 9:675-82. [PMID: 17486118 PMCID: PMC2442898 DOI: 10.1038/ncb1591] [Citation(s) in RCA: 404] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2007] [Accepted: 03/30/2007] [Indexed: 11/09/2022]
Abstract
Formation of cancerous translocations requires the illegitimate joining of chromosomes containing double-strand breaks (DSBs). It is unknown how broken chromosome ends find their translocation partners within the cell nucleus. Here, we have visualized and quantitatively analysed the dynamics of single DSBs in living mammalian cells. We demonstrate that broken ends are positionally stable and unable to roam the cell nucleus. Immobilization of broken chromosome ends requires the DNA-end binding protein Ku80, but is independent of DNA repair factors, H2AX, the MRN complex and the cohesion complex. DSBs preferentially undergo translocations with neighbouring chromosomes and loss of local positional constraint correlates with elevated genomic instability. These results support a contact-first model in which chromosome translocations predominantly form among spatially proximal DSBs.
Collapse
Affiliation(s)
- Evi Soutoglou
- National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Jonas F. Dorn
- The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Kundan Sengupta
- Genetics Branch, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Maria Jasin
- Memorial Sloan Kettering Cancer Center, New York, NY 10021, USA
| | - Andre Nussenzweig
- Experimental Immunology Branch, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Thomas Ried
- Genetics Branch, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | | | - Tom Misteli
- National Cancer Institute, NIH, Bethesda, MD 20892, USA
| |
Collapse
|
243
|
Prieto J, Redondo P, Padró D, Arnould S, Epinat JC, Pâques F, Blanco FJ, Montoya G. The C-terminal loop of the homing endonuclease I-CreI is essential for site recognition, DNA binding and cleavage. Nucleic Acids Res 2007; 35:3262-71. [PMID: 17452357 PMCID: PMC1904291 DOI: 10.1093/nar/gkm183] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Meganucleases are sequence-specific endonucleases with large cleavage sites that can be used to induce efficient homologous gene targeting in cultured cells and plants. These enzymes open novel perspectives for genome engineering in a wide range of fields, including gene therapy. A new crystal structure of the I-CreI dimer without DNA has allowed the comparison with the DNA-bound protein. The C-terminal loop displays a different conformation, which suggests its implication in DNA binding. A site-directed mutagenesis study in this region demonstrates that whereas the C-terminal helix is negligible for DNA binding, the final C-terminal loop is essential in DNA binding and cleavage. We have identified two regions that comprise the Ser138-Lys139 and Lys142-Thr143 pairs whose double mutation affect DNA binding in vitro and abolish cleavage in vivo. However, the mutation of only one residue in these sites allows DNA binding in vitro and cleavage in vivo. These findings demonstrate that the C-terminal loop of I-CreI endonuclease plays a fundamental role in its catalytic mechanism and suggest this novel site as a region to take into account for engineering new endonucleases with tailored specificity.
Collapse
Affiliation(s)
- Jesús Prieto
- Spanish National Cancer Center (CNIO), Structural Biology and Biocomputing Programme, NMR Group and Macromolecular Crystallography Group, c/Melchor Fdez. Almagro 3, 28029-Madrid, Spain and CELLECTIS S.A., 102 route de Noisy 93235 Romainville, France
| | - Pilar Redondo
- Spanish National Cancer Center (CNIO), Structural Biology and Biocomputing Programme, NMR Group and Macromolecular Crystallography Group, c/Melchor Fdez. Almagro 3, 28029-Madrid, Spain and CELLECTIS S.A., 102 route de Noisy 93235 Romainville, France
| | - Daniel Padró
- Spanish National Cancer Center (CNIO), Structural Biology and Biocomputing Programme, NMR Group and Macromolecular Crystallography Group, c/Melchor Fdez. Almagro 3, 28029-Madrid, Spain and CELLECTIS S.A., 102 route de Noisy 93235 Romainville, France
| | - Sylvain Arnould
- Spanish National Cancer Center (CNIO), Structural Biology and Biocomputing Programme, NMR Group and Macromolecular Crystallography Group, c/Melchor Fdez. Almagro 3, 28029-Madrid, Spain and CELLECTIS S.A., 102 route de Noisy 93235 Romainville, France
| | - Jean-Charles Epinat
- Spanish National Cancer Center (CNIO), Structural Biology and Biocomputing Programme, NMR Group and Macromolecular Crystallography Group, c/Melchor Fdez. Almagro 3, 28029-Madrid, Spain and CELLECTIS S.A., 102 route de Noisy 93235 Romainville, France
| | - Frédéric Pâques
- Spanish National Cancer Center (CNIO), Structural Biology and Biocomputing Programme, NMR Group and Macromolecular Crystallography Group, c/Melchor Fdez. Almagro 3, 28029-Madrid, Spain and CELLECTIS S.A., 102 route de Noisy 93235 Romainville, France
| | - Francisco J. Blanco
- Spanish National Cancer Center (CNIO), Structural Biology and Biocomputing Programme, NMR Group and Macromolecular Crystallography Group, c/Melchor Fdez. Almagro 3, 28029-Madrid, Spain and CELLECTIS S.A., 102 route de Noisy 93235 Romainville, France
| | - Guillermo Montoya
- Spanish National Cancer Center (CNIO), Structural Biology and Biocomputing Programme, NMR Group and Macromolecular Crystallography Group, c/Melchor Fdez. Almagro 3, 28029-Madrid, Spain and CELLECTIS S.A., 102 route de Noisy 93235 Romainville, France
- *To whom correspondence should be addressed. Tel:00 34 912246900; Fax: 00 34 912246976;
| |
Collapse
|
244
|
Saberi A, Hochegger H, Szuts D, Lan L, Yasui A, Sale JE, Taniguchi Y, Murakawa Y, Zeng W, Yokomori K, Helleday T, Teraoka H, Arakawa H, Buerstedde JM, Takeda S. RAD18 and poly(ADP-ribose) polymerase independently suppress the access of nonhomologous end joining to double-strand breaks and facilitate homologous recombination-mediated repair. Mol Cell Biol 2007; 27:2562-71. [PMID: 17242200 PMCID: PMC1899888 DOI: 10.1128/mcb.01243-06] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2006] [Revised: 08/07/2006] [Accepted: 11/17/2006] [Indexed: 01/05/2023] Open
Abstract
The Saccharomyces cerevisiae RAD18 gene is essential for postreplication repair but is not required for homologous recombination (HR), which is the major double-strand break (DSB) repair pathway in yeast. Accordingly, yeast rad18 mutants are tolerant of camptothecin (CPT), a topoisomerase I inhibitor, which induces DSBs by blocking replication. Surprisingly, mammalian cells and chicken DT40 cells deficient in Rad18 display reduced HR-dependent repair and are hypersensitive to CPT. Deletion of nonhomologous end joining (NHEJ), a major DSB repair pathway in vertebrates, in rad18-deficient DT40 cells completely restored HR-mediated DSB repair, suggesting that vertebrate Rad18 regulates the balance between NHEJ and HR. We previously reported that loss of NHEJ normalized the CPT sensitivity of cells deficient in poly(ADP-ribose) polymerase 1 (PARP1). Concomitant deletion of Rad18 and PARP1 synergistically increased CPT sensitivity, and additional inactivation of NHEJ normalized this hypersensitivity, indicating their parallel actions. In conclusion, higher-eukaryotic cells separately employ PARP1 and Rad18 to suppress the toxic effects of NHEJ during the HR reaction at stalled replication forks.
Collapse
Affiliation(s)
- Alihossein Saberi
- CREST Research Project, Radiation Genetics, Faculty of Medicine, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
245
|
Moehle EA, Rock JM, Lee YL, Jouvenot Y, DeKelver RC, Gregory PD, Urnov FD, Holmes MC. Targeted gene addition into a specified location in the human genome using designed zinc finger nucleases. Proc Natl Acad Sci U S A 2007; 104:3055-60. [PMID: 17360608 PMCID: PMC1802009 DOI: 10.1073/pnas.0611478104] [Citation(s) in RCA: 280] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Efficient incorporation of novel DNA sequences into a specific site in the genome of living human cells remains a challenge despite its potential utility to genetic medicine, biotechnology, and basic research. We find that a precisely placed double-strand break induced by engineered zinc finger nucleases (ZFNs) can stimulate integration of long DNA stretches into a predetermined genomic location, resulting in high-efficiency site-specific gene addition. Using an extrachromosomal DNA donor carrying a 12-bp tag, a 900-bp ORF, or a 1.5-kb promoter-transcription unit flanked by locus-specific homology arms, we find targeted integration frequencies of 15%, 6%, and 5%, respectively, within 72 h of treatment, and with no selection for the desired event. Importantly, we find that the integration event occurs in a homology-directed manner and leads to the accurate reconstruction of the donor-specified genotype at the endogenous chromosomal locus, and hence presumably results from synthesis-dependent strand annealing repair of the break using the donor DNA as a template. This site-specific gene addition occurs with no measurable increase in the rate of random integration. Remarkably, we also find that ZFNs can drive the addition of an 8-kb sequence carrying three distinct promoter-transcription units into an endogenous locus at a frequency of 6%, also in the absence of any selection. These data reveal the surprising versatility of the specialized polymerase machinery involved in double-strand break repair, illuminate a powerful approach to mammalian cell engineering, and open the possibility of ZFN-driven gene addition therapy for human genetic disease.
Collapse
Affiliation(s)
- Erica A. Moehle
- Sangamo BioSciences, Inc., Point Richmond Technology Center, 501 Canal Boulevard, Suite A100, Richmond, CA 94804
| | - Jeremy M. Rock
- Sangamo BioSciences, Inc., Point Richmond Technology Center, 501 Canal Boulevard, Suite A100, Richmond, CA 94804
| | - Ya-Li Lee
- Sangamo BioSciences, Inc., Point Richmond Technology Center, 501 Canal Boulevard, Suite A100, Richmond, CA 94804
| | - Yann Jouvenot
- Sangamo BioSciences, Inc., Point Richmond Technology Center, 501 Canal Boulevard, Suite A100, Richmond, CA 94804
| | - Russell C. DeKelver
- Sangamo BioSciences, Inc., Point Richmond Technology Center, 501 Canal Boulevard, Suite A100, Richmond, CA 94804
| | - Philip D. Gregory
- Sangamo BioSciences, Inc., Point Richmond Technology Center, 501 Canal Boulevard, Suite A100, Richmond, CA 94804
| | - Fyodor D. Urnov
- Sangamo BioSciences, Inc., Point Richmond Technology Center, 501 Canal Boulevard, Suite A100, Richmond, CA 94804
- *To whom correspondence should be addressed. E-mail:
| | - Michael C. Holmes
- Sangamo BioSciences, Inc., Point Richmond Technology Center, 501 Canal Boulevard, Suite A100, Richmond, CA 94804
| |
Collapse
|
246
|
Smith J, Grizot S, Arnould S, Duclert A, Epinat JC, Chames P, Prieto J, Redondo P, Blanco FJ, Bravo J, Montoya G, Pâques F, Duchateau P. A combinatorial approach to create artificial homing endonucleases cleaving chosen sequences. Nucleic Acids Res 2006; 34:e149. [PMID: 17130168 PMCID: PMC1702487 DOI: 10.1093/nar/gkl720] [Citation(s) in RCA: 213] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Meganucleases, or homing endonucleases (HEs) are sequence-specific endonucleases with large (>14 bp) cleavage sites that can be used to induce efficient homologous gene targeting in cultured cells and plants. These findings have opened novel perspectives for genome engineering in a wide range of fields, including gene therapy. However, the number of identified HEs does not match the diversity of genomic sequences, and the probability of finding a homing site in a chosen gene is extremely low. Therefore, the design of artificial endonucleases with chosen specificities is under intense investigation. In this report, we describe the first artificial HEs whose specificity has been entirely redesigned to cleave a naturally occurring sequence. First, hundreds of novel endonucleases with locally altered substrate specificity were derived from I-CreI, a Chlamydomonas reinhardti protein belonging to the LAGLIDADG family of HEs. Second, distinct DNA-binding subdomains were identified within the protein. Third, we used these findings to assemble four sets of mutations into heterodimeric endonucleases cleaving a model target or a sequence from the human RAG1 gene. These results demonstrate that the plasticity of LAGLIDADG endonucleases allows extensive engineering, and provide a general method to create novel endonucleases with tailored specificities.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Jesús Prieto
- Structural Biology and Biocomputing Programme, Centro Nacional de Investigaciones Oncológicas (CNIO)C/ Melchor Fdez Almagro, 28029 Madrid, Spain
| | - Pilar Redondo
- Structural Biology and Biocomputing Programme, Centro Nacional de Investigaciones Oncológicas (CNIO)C/ Melchor Fdez Almagro, 28029 Madrid, Spain
| | - Francisco J. Blanco
- Structural Biology and Biocomputing Programme, Centro Nacional de Investigaciones Oncológicas (CNIO)C/ Melchor Fdez Almagro, 28029 Madrid, Spain
| | - Jerónimo Bravo
- Structural Biology and Biocomputing Programme, Centro Nacional de Investigaciones Oncológicas (CNIO)C/ Melchor Fdez Almagro, 28029 Madrid, Spain
| | - Guillermo Montoya
- Structural Biology and Biocomputing Programme, Centro Nacional de Investigaciones Oncológicas (CNIO)C/ Melchor Fdez Almagro, 28029 Madrid, Spain
| | - Frédéric Pâques
- To whom correspondence should be addressed. Tel: +33 1 41 83 99 00; Fax: +33 1 41 83 99 03;
| | | |
Collapse
|
247
|
Nagaraju G, Odate S, Xie A, Scully R. Differential regulation of short- and long-tract gene conversion between sister chromatids by Rad51C. Mol Cell Biol 2006; 26:8075-86. [PMID: 16954385 PMCID: PMC1636746 DOI: 10.1128/mcb.01235-06] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2006] [Revised: 08/02/2006] [Accepted: 08/23/2006] [Indexed: 01/05/2023] Open
Abstract
The Rad51 paralog Rad51C has been implicated in the control of homologous recombination. To study the role of Rad51C in vivo in mammalian cells, we analyzed short-tract and long-tract gene conversion between sister chromatids in hamster Rad51C(-/-) CL-V4B cells in response to a site-specific chromosomal double-strand break. Gene conversion was inefficient in these cells and was specifically restored by expression of wild-type Rad51C. Surprisingly, gene conversions in CL-V4B cells were biased in favor of long-tract gene conversion, in comparison to controls expressing wild-type Rad51C. These long-tract events were not associated with crossing over between sister chromatids. Analysis of gene conversion tract lengths in CL-V4B cells lacking Rad51C revealed a bimodal frequency distribution, with almost all gene conversions being either less than 1 kb or greater than 3.2 kb in length. These results indicate that Rad51C plays a pivotal role in determining the "choice" between short- and long-tract gene conversion and in suppressing gene amplifications associated with sister chromatid recombination.
Collapse
Affiliation(s)
- Ganesh Nagaraju
- Department of Medicine, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | | | | | | |
Collapse
|
248
|
Abstract
Although notable progress has been made in the treatment of non-small-cell lung cancer (NSCLC) in recent years, this disease is still associated with a poor prognosis for most patients. Modern techniques have facilitated the identification of specific genetic factors that may play a role in disease progression and patient response to therapy, prompting research efforts to identify the clinical predictors of outcome for NSCLC. Recent evidence suggests that the application of a pharmacogenomic approach has the potential to greatly improve survival in certain subpopulations of patients with NSCLC, which could profoundly influence the decision-making process used in evolving treatment strategies for this malignancy.
Collapse
Affiliation(s)
- Gerold Bepler
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA.
| |
Collapse
|
249
|
Beumer K, Bhattacharyya G, Bibikova M, Trautman JK, Carroll D. Efficient gene targeting in Drosophila with zinc-finger nucleases. Genetics 2006; 172:2391-403. [PMID: 16452139 PMCID: PMC1456366 DOI: 10.1534/genetics.105.052829] [Citation(s) in RCA: 179] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
This report describes high-frequency germline gene targeting at two genomic loci in Drosophila melanogaster, y and ry. In the best case, nearly all induced parents produced mutant progeny; 25% of their offspring were new mutants and most of these were targeted gene replacements resulting from homologous recombination (HR) with a marked donor DNA. The procedure that generates these high frequencies relies on cleavage of the target by designed zinc-finger nucleases (ZFNs) and production of a linear donor in situ. Increased induction of ZFN expression led to higher frequencies of gene targeting, demonstrating the beneficial effect of activating the target. In the absence of a homologous donor DNA, ZFN cleavage led to the recovery of new mutants at three loci-y, ry and bw-through nonhomologous end joining (NHEJ) after cleavage. Because zinc fingers can be directed to a broad range of DNA sequences and targeting is very efficient, this approach promises to allow genetic manipulation of many different genes, even in cases where the mutant phenotype cannot be predicted.
Collapse
Affiliation(s)
- Kelly Beumer
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, Utah 84112, USA
| | | | | | | | | |
Collapse
|
250
|
Chames P, Epinat JC, Guillier S, Patin A, Lacroix E, Pâques F. In vivo selection of engineered homing endonucleases using double-strand break induced homologous recombination. Nucleic Acids Res 2005; 33:e178. [PMID: 16306233 PMCID: PMC1289081 DOI: 10.1093/nar/gni175] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
Homing endonucleases, endonucleases capable of recognizing long DNA sequences, have been shown to be a tool of choice for precise and efficient genome engineering. Consequently, the possibility to engineer novel endonucleases with tailored specificities is under strong investigation. In this report, we present a simple and efficient method to select meganucleases from libraries of variants, based on their cleavage properties. The method has the advantage of directly selecting for the ability to induce double-strand break induced homologous recombination in a eukaryotic environment. Model selections demonstrated high levels of enrichments. Moreover, this method compared favorably with phage display for enrichment of active mutants from a mutant library. This approach makes possible the exploration of large sequence spaces and thereby represents a valuable tool for genome engineering.
Collapse
Affiliation(s)
| | | | | | | | | | - Frédéric Pâques
- To whom correspondence should be addressed. Tel: +33 1 41 83 99 00; Fax: +33 1 41 83 99 03;
| |
Collapse
|