1
|
Gao Y, Guo L, Wang F, Wang Y, Li P, Zhang D. Development of mitochondrial gene-editing strategies and their potential applications in mitochondrial hereditary diseases: a review. Cytotherapy 2024; 26:11-24. [PMID: 37930294 DOI: 10.1016/j.jcyt.2023.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 10/12/2023] [Accepted: 10/13/2023] [Indexed: 11/07/2023]
Abstract
Mitochondrial DNA (mtDNA) is a critical genome contained within the mitochondria of eukaryotic cells, with many copies present in each mitochondrion. Mutations in mtDNA often are inherited and can lead to severe health problems, including various inherited diseases and premature aging. The lack of efficient repair mechanisms and the susceptibility of mtDNA to damage exacerbate the threat to human health. Heteroplasmy, the presence of different mtDNA genotypes within a single cell, increases the complexity of these diseases and requires an effective editing method for correction. Recently, gene-editing techniques, including programmable nucleases such as restriction endonuclease, zinc finger nuclease, transcription activator-like effector nuclease, clustered regularly interspaced short palindromic repeats/clustered regularly interspaced short palindromic repeats-associated 9 and base editors, have provided new tools for editing mtDNA in mammalian cells. Base editors are particularly promising because of their high efficiency and precision in correcting mtDNA mutations. In this review, we discuss the application of these techniques in mitochondrial gene editing and their limitations. We also explore the potential of base editors for mtDNA modification and discuss the opportunities and challenges associated with their application in mitochondrial gene editing. In conclusion, this review highlights the advancements, limitations and opportunities in current mitochondrial gene-editing technologies and approaches. Our insights aim to stimulate the development of new editing strategies that can ultimately alleviate the adverse effects of mitochondrial hereditary diseases.
Collapse
Affiliation(s)
- Yanyan Gao
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Linlin Guo
- The Affiliated Cardiovascular Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Fei Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Yin Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Peifeng Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Dejiu Zhang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China.
| |
Collapse
|
2
|
Mitochondrial Genome Editing to Treat Human Osteoarthritis-A Narrative Review. Int J Mol Sci 2022; 23:ijms23031467. [PMID: 35163384 PMCID: PMC8835930 DOI: 10.3390/ijms23031467] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 01/24/2022] [Accepted: 01/26/2022] [Indexed: 12/27/2022] Open
Abstract
Osteoarthritis (OA) is a severe, common chronic orthopaedic disorder characterised by a degradation of the articular cartilage with an incidence that increases over years. Despite the availability of various clinical options, none can stop the irreversible progression of the disease to definitely cure OA. Various mutations have been evidenced in the mitochondrial DNA (mtDNA) of cartilage cells (chondrocytes) in OA, leading to a dysfunction of the mitochondrial oxidative phosphorylation processes that significantly contributes to OA cartilage degeneration. The mitochondrial genome, therefore, represents a central, attractive target for therapy in OA, especially using genome editing procedures. In this narrative review article, we present and discuss the current advances and breakthroughs in mitochondrial genome editing as a potential, novel treatment to overcome mtDNA-related disorders such as OA. While still in its infancy and despite a number of challenges that need to be addressed (barriers to effective and site-specific mtDNA editing and repair), such a strategy has strong value to treat human OA in the future, especially using the groundbreaking clustered regularly interspaced short palindromic repeats (CRIPSR)/CRISPR-associated 9 (CRISPR/Cas9) technology and mitochondrial transplantation approaches.
Collapse
|
3
|
Kues WA, Kumar D, Selokar NL, Talluri TR. Applications of genome editing tools in stem cells towards regenerative medicine: An update. Curr Stem Cell Res Ther 2021; 17:267-279. [PMID: 34819011 DOI: 10.2174/1574888x16666211124095527] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 09/14/2021] [Accepted: 09/25/2021] [Indexed: 11/22/2022]
Abstract
Precise and site specific genome editing through application of emerging and modern gene engineering techniques, namely zinc finger nucleases (ZFNs), transcription activator-like effector nucleases (TALENs) and clustered regularly interspaced short palindromic repeats (CRISPR/Cas9) have swiftly progressed the application and use of the stem cell technology in the sphere of in-vitro disease modelling and regenerative medicine. Genome editing tools facilitate the manipulating of any gene in various types of cells with target specific nucleases. These tools aid in elucidating the genetics and etiology behind different diseases and have immense promise as novel therapeutics for correcting the genetic mutations, make alterations and cure diseases permanently that are not responding and resistant to traditional therapies. These genome engineering tools have evolved in the field of biomedical research and have also shown to have a significant improvement in clinical trials. However, their widespread use in research revealed potential safety issues, which need to be addressed before implementing such techniques in clinical purposes. Significant and valiant attempts are being made in order to surpass those hurdles. The current review outlines the advancements of several genome engineering tools and describes suitable strategies for their application towards regenerative medicine.
Collapse
Affiliation(s)
- Wilfried A Kues
- Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Department of Biotechnology, Stem Cell Physiology, Höltystr 10, 31535 Neustadt. Germany
| | - Dharmendra Kumar
- Animal Physiology and Reproduction Division, ICAR-Central Institute for Research on Buffaloes, Hisar-125001, Haryana. India
| | - Naresh L Selokar
- Animal Physiology and Reproduction Division, ICAR-Central Institute for Research on Buffaloes, Hisar-125001, Haryana. India
| | - Thirumala Rao Talluri
- Equine Production Campus, ICAR- National Research Centre on Equines, Bikaner-334001, Rajasthan. India
| |
Collapse
|
4
|
Kang S, Jeon S, Kim S, Chang YK, Kim YC. Development of a pVEC peptide-based ribonucleoprotein (RNP) delivery system for genome editing using CRISPR/Cas9 in Chlamydomonas reinhardtii. Sci Rep 2020; 10:22158. [PMID: 33335164 PMCID: PMC7747696 DOI: 10.1038/s41598-020-78968-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 11/19/2020] [Indexed: 12/13/2022] Open
Abstract
Recent technical advances related to the CRISPR/Cas9-based genome editing system have enabled sophisticated genome editing in microalgae. Although the demand for research on genome editing in microalgae has increased over time, methodological research has not been established to date for the delivery of a ribonucleoprotein (Cas9/sgRNA complex) using a cell penetrating peptide into microalgal cell lines. Here, we present a ribonucleoprotein delivery system for Chlamydomonas reinhardtii mediated by the cell penetrating peptide pVEC (LLIILRRRIRKQAHAHSK) which is in a non-covalent form. Using this technically simple method, the ribonucleoprotein was successfully delivered into C. reinhardtii. Gene Maa7 and FKB12 were disrupted, and their distinguishing patterns of Indel mutations were analyzed with the observation of several insertions of sequences not originating from the genome DNA, such as chloroplast DNA, into the expected loci. In addition, the cytotoxicity of Cas9 and the ribonucleoprotein was investigated according to the concentration and time in the algal cells. It was observed that Cas9 alone without the sgRNA induces a more severe cytotoxicity compared to the ribonucleoprotein. Our study will not only contribute to algal cell biology and its genetic engineering for further applications involving various organisms but will also provide a deeper understating of the basic science of the CRISPR/Cas9 system.
Collapse
Affiliation(s)
- Seongsu Kang
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 305-701, Republic of Korea
| | - Seungjib Jeon
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 305-701, Republic of Korea
| | - Seungcheol Kim
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 305-701, Republic of Korea
| | - Yong Keun Chang
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 305-701, Republic of Korea
| | - Yeu-Chun Kim
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 305-701, Republic of Korea.
| |
Collapse
|
5
|
Bukhari H, Müller T. Endogenous Fluorescence Tagging by CRISPR. Trends Cell Biol 2019; 29:912-928. [PMID: 31522960 DOI: 10.1016/j.tcb.2019.08.004] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 08/11/2019] [Accepted: 08/12/2019] [Indexed: 01/01/2023]
Abstract
Fluorescent proteins have revolutionized biomedical research as they are easy to use for protein tagging, cope without fixation or permeabilization, and thus, enable live cell imaging in various models. Current methods allow easy and quick integration of fluorescent markers to endogenous genes of interest. In this review, we introduce the three central methods, zinc finger nucleases (ZFNs), transcription activator-like effectors (TALENs), and CRISPR, that have been widely used to manipulate cells or organisms. Focusing on CRISPR technology, we give an overview on homology-directed repair (HDR)-, microhomology-mediated end joining (MMEJ)-, and nonhomologous end joining (NHEJ)-based strategies for the knock-in of markers, figure out recent developments of the technique for highly efficient knock-in, and demonstrate pros and cons. We highlight the unique aspects of fluorescent protein knock-ins and pinpoint specific improvements and perspectives, like the combination of editing with stem cell derived organoid development.
Collapse
Affiliation(s)
- Hassan Bukhari
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Molecular Biochemistry, Cell Signalling, Ruhr-University Bochum, Bochum, Germany
| | - Thorsten Müller
- Department of Molecular Biochemistry, Cell Signalling, Ruhr-University Bochum, Bochum, Germany; Institute of Psychiatric Phenomics and Genomics (IPPG), University Hospital, LMU Munich, Munich 80336, Germany.
| |
Collapse
|
6
|
Use of gene-editing technology to introduce targeted modifications in pigs. J Anim Sci Biotechnol 2018; 9:5. [PMID: 29423214 PMCID: PMC5787920 DOI: 10.1186/s40104-017-0228-7] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 12/22/2017] [Indexed: 01/06/2023] Open
Abstract
Pigs are an important resource in agriculture and serve as a model for human diseases. Due to their physiological and anatomical similarities with humans, pigs can recapitulate symptoms of human diseases, making them a useful model in biomedicine. However, in the past pig models have not been widely used partially because of the difficulty in genetic modification. The lack of true embryonic stem cells in pigs forced researchers to utilize genetic modification in somatic cells and somatic cell nuclear transfer (SCNT) to generate genetically engineered (GE) pigs carrying site-specific modifications. Although possible, this approach is extremely inefficient and GE pigs born through this method often presented developmental defects associated with the cloning process. Advancement in the gene-editing systems such as Zinc-Finger Nucleases (ZFNs), Transcription activator-like effector nucleases (TALENs), and the Clustered regularly interspaced short palindromic repeat (CRISPR)/CRISPR-associated 9 (Cas9) system have dramatically increased the efficiency of producing GE pigs. These gene-editing systems, specifically engineered endonucleases, are based on inducing double-stranded breaks (DSBs) at a specific location, and then site-specific modifications can be introduced through one of the two DNA repair pathways: non-homologous end joining (NHEJ) or homology direct repair (HDR). Random insertions or deletions (indels) can be introduced through NHEJ and specific nucleotide sequences can be introduced through HDR, if donor DNA is provided. Use of these engineered endonucleases provides a higher success in genetic modifications, multiallelic modification of the genome, and an opportunity to introduce site-specific modifications during embryogenesis, thus bypassing the need of SCNT in GE pig production. This review will provide a historical prospective of GE pig production and examples of how the gene-editing system, led by engineered endonucleases, have improved GE pig production. We will also present some of our current progress related to the optimal use of CRISPR/Cas9 system during embryogenesis.
Collapse
|
7
|
Xing J, Zhang C, Xu K, Hu L, Wang L, Zhang T, Ren C, Zhang Z. An Improved Genome Engineering Method Using Surrogate Reporter-Coupled Suicidal ZFNs. Methods Mol Biol 2018; 1867:175-183. [PMID: 30155823 DOI: 10.1007/978-1-4939-8799-3_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Using engineered nucleases such as zinc-finger nucleases (ZFNs) and TALE nuclease (TALEN) to accomplish genome editing often causes high cellular toxicity because of the consistent expression of artificial nucleases and off-targeting effect. And lacking selection marker in modified cells makes it hard to enrich these positive cells. Here we introduce a method by incorporating a surrogate reporter enrichment into a suicidal ZFN system, which is designed by a pair of ZFN expression cassettes flanked with its target sites. Our data demonstrated that this modified system achieved almost the same ZFN activity as the original method but reduced ~40% toxicity. This new suicidal ZFN expression system coupled with a surrogate reporter not only enables decreased cellular toxicity but also makes the genetic modified cells to be enriched by EGFP analysis.
Collapse
Affiliation(s)
- Jiani Xing
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Cunfang Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Kun Xu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Linyong Hu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Ling Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Tingting Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Chonghua Ren
- Genome Center and Department of Biochemistry and Molecular Medicine, University of California, Davis, CA, USA
- Guangzhou Key Laboratory of Insect Development Regulation and Application Research, South China Normal University, Guangzhou, China
| | - Zhiying Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China.
| |
Collapse
|
8
|
Song XQ, Su LN, Wei HP, Liu YH, Yin HF, Li JH, Zhu DX, Zhang AL. The effect of Id1gene silencing on the neural differentiation of MSCs. BIOTECHNOL BIOTEC EQ 2017. [DOI: 10.1080/13102818.2017.1286234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Affiliation(s)
- Xiao-qing Song
- Department of Biology, Basic Medical College, Hebei North University, Zhangjiakou, China
| | - Li-ning Su
- Department of Biology, Basic Medical College, Hebei North University, Zhangjiakou, China
| | - Hui-ping Wei
- Department of Biology, Basic Medical College, Hebei North University, Zhangjiakou, China
| | - Ying-hui Liu
- Department of Agriculture Science, Agriculture and Forestry College of Hebei North University, Zhangjiakou, China
| | - Hai-feng Yin
- Department of Biology, Basic Medical College, Hebei North University, Zhangjiakou, China
| | - Ji-hong Li
- Department of Biology, Basic Medical College, Hebei North University, Zhangjiakou, China
| | - Deng-xiang Zhu
- Department of Biology, Basic Medical College, Hebei North University, Zhangjiakou, China
| | - Ai-lan Zhang
- Department of Biology, Basic Medical College, Hebei North University, Zhangjiakou, China
| |
Collapse
|
9
|
Genome Editing for the β-Hemoglobinopathies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1013:203-217. [DOI: 10.1007/978-1-4939-7299-9_8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
|
10
|
Abstract
Duchenne muscular dystrophy (DMD) is a recessive lethal inherited muscular dystrophy caused by mutations in the gene encoding dystrophin, a protein required for muscle fibre integrity. So far, many approaches have been tested from the traditional gene addition to newer advanced approaches based on manipulation of the cellular machinery either at the gene transcription, mRNA processing or translation levels. Unfortunately, despite all these efforts, no efficient treatments for DMD are currently available. In this review, we highlight the most advanced therapeutic strategies under investigation as potential DMD treatments.
Collapse
Affiliation(s)
- Hayder Abdul-Razak
- School of Biological Sciences, Royal Holloway, University of London, Egham, Surrey, TW20 0EX, UK
| | - Alberto Malerba
- School of Biological Sciences, Royal Holloway, University of London, Egham, Surrey, TW20 0EX, UK
| | - George Dickson
- School of Biological Sciences, Royal Holloway, University of London, Egham, Surrey, TW20 0EX, UK
| |
Collapse
|
11
|
Cheng JK, Lewis AM, Kim DS, Dyess T, Alper HS. Identifying and retargeting transcriptional hot spots in the human genome. Biotechnol J 2016; 11:1100-9. [PMID: 27311394 DOI: 10.1002/biot.201600015] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Revised: 05/18/2016] [Accepted: 05/30/2016] [Indexed: 01/17/2023]
Abstract
Mammalian cell line development requires streamlined methodologies that will reduce both the cost and time to identify candidate cell lines. Improvements in site-specific genomic editing techniques can result in flexible, predictable, and robust cell line engineering. However, an outstanding question in the field is the specific site of integration. Here, we seek to identify productive loci within the human genome that will result in stable, high expression of heterologous DNA. Using an unbiased, random integration approach and a green fluorescent reporter construct, we identify ten single-integrant, recombinant human cell lines that exhibit stable, high-level expression. From these cell lines, eight unique corresponding integration loci were identified. These loci are concentrated in non-protein coding regions or intronic regions of protein coding genes. Expression mapping of the surrounding genes reveals minimal disruption of endogenous gene expression. Finally, we demonstrate that targeted de novo integration at one of the identified loci, the 12(th) exon-intron region of the GRIK1 gene on chromosome 21, results in superior expression and stability compared to the standard, illegitimate integration approach at levels approaching 4-fold. The information identified here along with recent advances in site-specific genomic editing techniques can lead to expedited cell line development.
Collapse
Affiliation(s)
- Joseph K Cheng
- Department of Chemical Engineering, The University of Texas at Austin, Austin, Texas, USA
| | - Amanda M Lewis
- Department of Chemical Engineering, The University of Texas at Austin, Austin, Texas, USA.,Biologics Development, Bristol-Myers Squibb, Devens, MA, USA
| | - Do Soon Kim
- Department of Chemical Engineering, The University of Texas at Austin, Austin, Texas, USA
| | - Timothy Dyess
- Department of Chemical Engineering, The University of Texas at Austin, Austin, Texas, USA
| | - Hal S Alper
- Department of Chemical Engineering, The University of Texas at Austin, Austin, Texas, USA. .,Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, Texas, USA.
| |
Collapse
|
12
|
Bevacqua RJ, Fernandez-Martín R, Savy V, Canel NG, Gismondi MI, Kues WA, Carlson DF, Fahrenkrug SC, Niemann H, Taboga OA, Ferraris S, Salamone DF. Efficient edition of the bovine PRNP prion gene in somatic cells and IVF embryos using the CRISPR/Cas9 system. Theriogenology 2016; 86:1886-1896.e1. [PMID: 27566851 DOI: 10.1016/j.theriogenology.2016.06.010] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 05/17/2016] [Accepted: 06/05/2016] [Indexed: 12/19/2022]
Abstract
The recently developed engineered nucleases, such as zinc-finger nucleases, transcription activator-like effector nucleases, and clustered regularly interspaced short palindromic repeat (CRISPR)/CRISPR-associated nuclease (Cas) 9, provide new opportunities for gene editing in a straightforward manner. However, few reports are available regarding CRISPR application and efficiency in cattle. Here, the CRISPR/Cas9 system was used with the aim of inducing knockout and knock-in alleles of the bovine PRNP gene, responsible for mad cow disease, both in bovine fetal fibroblasts and in IVF embryos. Five single-guide RNAs were designed to target 875 bp of PRNP exon 3, and all five were codelivered with Cas9. The feasibility of inducing homologous recombination (HR) was evaluated with a reporter vector carrying EGFP flanked by 1 kbp PRNP regions (pHRegfp). For somatic cells, plasmids coding for Cas9 and for each of the five single-guide RNAs (pCMVCas9 and pSPgRNAs) were transfected under two different conditions (1X and 2X). For IVF zygotes, cytoplasmic injection was conducted with either plasmids or mRNA. For plasmid injection groups, 1 pg pCMVCas9 + 0.1 pg of each pSPgRNA (DNA2X) was used per zygote. In the case of RNA, two amounts (RNA1X and RNA2X) were compared. To assess the occurrence of HR, a group additionally cotransfected or coinjected with pHRegfp plasmid was included. Somatic cell lysates were analyzed by polymerase chain reaction and surveyor assay. In the case of embryos, the in vitro development and the genotype of blastocysts were evaluated by polymerase chain reaction and sequencing. In somatic cells, 2X transfection resulted in indels and large deletions of the targeted PRNP region. Regarding embryo injection, higher blastocyst rates were obtained for RNA injected groups (46/103 [44.6%] and 55/116 [47.4%] for RNA1X and RNA2X) than for the DNA2X group (26/140 [18.6%], P < 0.05). In 46% (26/56) of the total sequenced blastocysts, specific gene editing was detected. The total number of genetic modifications (29) was higher than the total number of gene-edited embryos, as three blastocysts from the group RNA2X reported more than one type of modification. The modifications included indels (10/56; 17.9%) and large deletions (19/56; 33.9%). Moreover, it was possible to detect HR in 1/8 (12.5%) embryos treated with RNA2X. These results report that the CRISPR/Cas9 system can be applied for site-specific edition of the bovine genome, which could have a great impact on the development of large animals resistant to important zoonotic diseases.
Collapse
Affiliation(s)
- R J Bevacqua
- Animal Biotechnology Laboratory, INPA UBA-CONICET, Buenos Aires, Argentina
| | - R Fernandez-Martín
- Animal Biotechnology Laboratory, INPA UBA-CONICET, Buenos Aires, Argentina
| | - V Savy
- Animal Biotechnology Laboratory, INPA UBA-CONICET, Buenos Aires, Argentina
| | - N G Canel
- Animal Biotechnology Laboratory, INPA UBA-CONICET, Buenos Aires, Argentina
| | - M I Gismondi
- Instituto de Biotecnología, CICVyA, INTA-CONICET, Hurlingham, Argentina
| | - W A Kues
- Institute of Farm Animal Genetics (FLI), Mariensee, Hannover, Germany
| | | | | | - H Niemann
- Institute of Farm Animal Genetics (FLI), Mariensee, Hannover, Germany
| | - O A Taboga
- Instituto de Biotecnología, CICVyA, INTA-CONICET, Hurlingham, Argentina
| | - S Ferraris
- Cloning and Transgenesis Laboratory, Maimonides University, Buenos Aires, Argentina
| | - D F Salamone
- Animal Biotechnology Laboratory, INPA UBA-CONICET, Buenos Aires, Argentina.
| |
Collapse
|
13
|
Ethical and regulatory aspects of genome editing. Blood 2016; 127:2553-60. [PMID: 27053531 DOI: 10.1182/blood-2016-01-678136] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Accepted: 02/18/2016] [Indexed: 12/26/2022] Open
Abstract
Gene editing is a rapidly developing area of biotechnology in which the nucleotide sequence of the genome of living cells is precisely changed. The use of genome-editing technologies to modify various types of blood cells, including hematopoietic stem cells, has emerged as an important field of therapeutic development for hematopoietic disease. Although these technologies offer the potential for generation of transformative therapies for patients suffering from myriad disorders of hematopoiesis, their application for therapeutic modification of primary human cells is still in its infancy. Consequently, development of ethical and regulatory frameworks that ensure their safe and effective use is an increasingly important consideration. Here, we review a number of issues that have the potential to impact the clinical implementation of genome-editing technologies, and suggest paths forward for resolving them such that new therapies can be safely and rapidly translated to the clinic.
Collapse
|
14
|
Corrigan-Curay J, O'Reilly M, Kohn DB, Cannon PM, Bao G, Bushman FD, Carroll D, Cathomen T, Joung JK, Roth D, Sadelain M, Scharenberg AM, von Kalle C, Zhang F, Jambou R, Rosenthal E, Hassani M, Singh A, Porteus MH. Genome editing technologies: defining a path to clinic. Mol Ther 2016; 23:796-806. [PMID: 25943494 DOI: 10.1038/mt.2015.54] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Affiliation(s)
| | | | - Donald B Kohn
- University of California, Los Angeles, Los Angeles, California, USA
| | - Paula M Cannon
- Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Gang Bao
- Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Frederic D Bushman
- University of Pennsylvania, School of Medicine, Philadelphia, Pennsylvania, USA
| | - Dana Carroll
- University of Utah, School of Medicine, Salt Lake City, Utah, USA
| | - Toni Cathomen
- University Medical Center Freiberg, Freiberg, Germany
| | - J Keith Joung
- Massachusetts General Hospital, Charlestown, Massachusetts; Harvard Medical School, Boston, Massachusetts, USA
| | - David Roth
- University of Pennsylvania, School of Medicine, Philadelphia, Pennsylvania, USA
| | - Michel Sadelain
- Memorial Sloan-Kettering Cancer Center, New York, New York, USA
| | - Andrew M Scharenberg
- Seattle Children's Research Institute and University of Washington, School of Medicine, Seattle, Washington, USA
| | - Christof von Kalle
- National Center for Tumor Diseases and German Cancer Research Center, Heidelberg, Germany
| | - Feng Zhang
- Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts, USA
| | - Robert Jambou
- National Institutes of Health, Bethesda, Maryland, USA
| | | | - Morad Hassani
- National Institutes of Health, Bethesda, Maryland, USA
| | - Aparna Singh
- National Institutes of Health, Bethesda, Maryland, USA
| | | |
Collapse
|
15
|
Multi-reporter selection for the design of active and more specific zinc-finger nucleases for genome editing. Nat Commun 2016; 7:10194. [PMID: 26738816 PMCID: PMC4729830 DOI: 10.1038/ncomms10194] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 11/16/2015] [Indexed: 01/17/2023] Open
Abstract
Engineered nucleases have transformed biological research and offer great therapeutic potential by enabling the straightforward modification of desired genomic sequences. While many nuclease platforms have proven functional, all can produce unanticipated off-target lesions and have difficulty discriminating between homologous sequences, limiting their therapeutic application. Here we describe a multi-reporter selection system that allows the screening of large protein libraries to uncover variants able to discriminate between sequences with substantial homology. We have used this system to identify zinc-finger nucleases that exhibit high cleavage activity (up to 60% indels) at their targets within the CCR5 and HBB genes and strong discrimination against homologous sequences within CCR2 and HBD. An unbiased screen for off-target lesions using a novel set of CCR5-targeting nucleases confirms negligible CCR2 activity and demonstrates minimal off-target activity genome wide. This system offers a straightforward approach to generate nucleases that discriminate between similar targets and provide exceptional genome-wide specificity. Zinc finger nucleases have an established role in genome editing. Here, the authors report a strategy for identifying zinc finger nucleases that discriminate between desired targets and provide genome-wide specificity.
Collapse
|
16
|
Moehrle BM, Nattamai K, Brown A, Florian MC, Ryan M, Vogel M, Bliederhaeuser C, Soller K, Prows DR, Abdollahi A, Schleimer D, Walter D, Milsom MD, Stambrook P, Porteus M, Geiger H. Stem Cell-Specific Mechanisms Ensure Genomic Fidelity within HSCs and upon Aging of HSCs. Cell Rep 2015; 13:2412-2424. [PMID: 26686632 DOI: 10.1016/j.celrep.2015.11.030] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 08/13/2015] [Accepted: 11/08/2015] [Indexed: 01/22/2023] Open
Abstract
Whether aged hematopoietic stem and progenitor cells (HSPCs) have impaired DNA damage repair is controversial. Using a combination of DNA mutation indicator assays, we observe a 2- to 3-fold increase in the number of DNA mutations in the hematopoietic system upon aging. Young and aged hematopoietic stem cells (HSCs) and hematopoietic progenitor cells (HPCs) do not show an increase in mutation upon irradiation-induced DNA damage repair, and young and aged HSPCs respond very similarly to DNA damage with respect to cell-cycle checkpoint activation and apoptosis. Both young and aged HSPCs show impaired activation of the DNA-damage-induced G1-S checkpoint. Induction of chronic DNA double-strand breaks by zinc-finger nucleases suggests that HSPCs undergo apoptosis rather than faulty repair. These data reveal a protective mechanism in both the young and aged hematopoietic system against accumulation of mutations in response to DNA damage.
Collapse
Affiliation(s)
- Bettina M Moehrle
- Institute of Molecular Medicine, University of Ulm, 89081 Ulm, Germany
| | - Kalpana Nattamai
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center and University of Cincinnati, Cincinnati, OH 45229, USA
| | - Andreas Brown
- Institute of Molecular Medicine, University of Ulm, 89081 Ulm, Germany
| | - Maria C Florian
- Institute of Molecular Medicine, University of Ulm, 89081 Ulm, Germany
| | - Marnie Ryan
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center and University of Cincinnati, Cincinnati, OH 45229, USA
| | - Mona Vogel
- Institute of Molecular Medicine, University of Ulm, 89081 Ulm, Germany
| | | | - Karin Soller
- Institute of Molecular Medicine, University of Ulm, 89081 Ulm, Germany
| | - Daniel R Prows
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center and University of Cincinnati, Cincinnati, OH 45229, USA
| | - Amir Abdollahi
- German Cancer Consortium (DKTK), 69120 Heidelberg, Germany; Molecular and Translational Radiation Oncology, Heidelberg Ion Therapy Center (HIT), 69120 Heidelberg, Germany
| | - David Schleimer
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center and University of Cincinnati, Cincinnati, OH 45229, USA
| | - Dagmar Walter
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine gGmbH (HI-STEM), 69120 Heidelberg, Germany
| | - Michael D Milsom
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine gGmbH (HI-STEM), 69120 Heidelberg, Germany; Deutsches Krebsforschungszentrum (DKFZ), Division of Stem Cells and Cancer, Experimental Hematology Group, 69120 Heidelberg, Germany
| | - Peter Stambrook
- Department of Molecular Genetics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Matthew Porteus
- Department of Pediatrics, Stanford University, Stanford, CA 94305, USA
| | - Hartmut Geiger
- Institute of Molecular Medicine, University of Ulm, 89081 Ulm, Germany; Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center and University of Cincinnati, Cincinnati, OH 45229, USA.
| |
Collapse
|
17
|
Abstract
The ability to manipulate the genome with precise spatial and nucleotide resolution (genome editing) has been a powerful research tool. In the past decade, the tools and expertise for using genome editing in human somatic cells and pluripotent cells have increased to such an extent that the approach is now being developed widely as a strategy to treat human disease. The fundamental process depends on creating a site-specific DNA double-strand break (DSB) in the genome and then allowing the cell's endogenous DSB repair machinery to fix the break such that precise nucleotide changes are made to the DNA sequence. With the development and discovery of several different nuclease platforms and increasing knowledge of the parameters affecting different genome editing outcomes, genome editing frequencies now reach therapeutic relevance for a wide variety of diseases. Moreover, there is a series of complementary approaches to assessing the safety and toxicity of any genome editing process, irrespective of the underlying nuclease used. Finally, the development of genome editing has raised the issue of whether it should be used to engineer the human germline. Although such an approach could clearly prevent the birth of people with devastating and destructive genetic diseases, questions remain about whether human society is morally responsible enough to use this tool.
Collapse
Affiliation(s)
- Matthew Porteus
- Department of Pediatrics, Division of Stem Cell Transplantation and Regenerative Medicine, Stanford University, Stanford, California 94305;
| |
Collapse
|
18
|
Both TALENs and CRISPR/Cas9 directly target the HBB IVS2-654 (C > T) mutation in β-thalassemia-derived iPSCs. Sci Rep 2015; 5:12065. [PMID: 26156589 PMCID: PMC4496796 DOI: 10.1038/srep12065] [Citation(s) in RCA: 122] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Accepted: 06/16/2015] [Indexed: 12/19/2022] Open
Abstract
β-Thalassemia is one of the most common genetic blood diseases and is caused by either point mutations or deletions in the β-globin (HBB) gene. The generation of patient-specific induced pluripotent stem cells (iPSCs) and subsequent correction of the disease-causing mutations may be a potential therapeutic strategy for this disease. Due to the low efficiency of typical homologous recombination, endonucleases, including TALENs and CRISPR/Cas9, have been widely used to enhance the gene correction efficiency in patient-derived iPSCs. Here, we designed TALENs and CRISPR/Cas9 to directly target the intron2 mutation site IVS2-654 in the globin gene. We observed different frequencies of double-strand breaks (DSBs) at IVS2-654 loci using TALENs and CRISPR/Cas9, and TALENs mediated a higher homologous gene targeting efficiency compared to CRISPR/Cas9 when combined with the piggyBac transposon donor. In addition, more obvious off-target events were observed for CRISPR/Cas9 compared to TALENs. Finally, TALENs-corrected iPSC clones were selected for erythroblast differentiation using the OP9 co-culture system and detected relatively higher transcription of HBB than the uncorrected cells. This comparison of using TALENs or CRISPR/Cas9 to correct specific HBB mutations in patient-derived iPSCs will guide future applications of TALENs- or CRISPR/Cas9-based gene therapies in monogenic diseases.
Collapse
|
19
|
Strategies to increase genome editing frequencies and to facilitate the identification of edited cells. Methods Mol Biol 2015; 1239:281-9. [PMID: 25408413 DOI: 10.1007/978-1-4939-1862-1_16] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The power of genome editing is increasingly recognized as it has become more accessible to a wide range of scientists and a wider range of uses has been reported. Nonetheless, an important practical aspect of the strategy is develop methods to increase the frequency of genome editing or methods that enrich for genome-edited cells such that they can be more easily identified. This chapter discusses several different approaches including the use of cold-shock, exonucleases, surrogate markers, specialized donor vectors, and oligonucleotides to enhance the frequency of genome editing or to facilitate the identification of genome-edited cells.
Collapse
|
20
|
Semaan M, Ivanusic D, Denner J. Cytotoxic Effects during Knock Out of Multiple Porcine Endogenous Retrovirus (PERV) Sequences in the Pig Genome by Zinc Finger Nucleases (ZFN). PLoS One 2015; 10:e0122059. [PMID: 25909512 PMCID: PMC4409370 DOI: 10.1371/journal.pone.0122059] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Accepted: 02/10/2015] [Indexed: 01/20/2023] Open
Abstract
Xenotransplantation has been proposed as a solution to the shortage of suitable human donors for transplantation and pigs are currently favoured as donor animals. However, xenotransplantation may be associated with the transmission of zoonotic microorganisms. Whereas most porcine microorganisms representing a risk for the human recipient may be eliminated by designated pathogen free breeding, multiple copies of porcine endogenous retroviruses (PERVs) are integrated in the genome of all pigs and cannot be eliminated this way. PERVs are released as infectious particles and infect human cells. The zinc finger nuclease (ZFN) technology allows knocking out specifically cellular genes, however it was not yet used to eliminate multiple integrated proviral sequences with a strong conservation in the target sequence. To reduce the risk of horizontal PERV transmission and to knock out as many as possible proviruses, for the first time the powerful tool of the ZFN technology was used. ZFN were designed to bind specifically to sequences conserved in all known replication-competent proviruses. Expression and transport of the ZFN into the nucleus was shown by Western blot analysis, co-localisation analysis, PLA and FRET. Survival of transfected cells was analysed using fluorescent ZFN and cell counting. After transfection a strong expression of the ZFN proteins and a co-localisation of the expressed ZFN proteins were shown. However, expression of the ZFN was found to be extremely toxic for the transfected cells. The induced cytotoxicity was likely due to the specific cutting of the high copy number of the PERV proviruses, which is also commonly observed when ZFN with low specificity cleave numerous off-target sites in a genome. This is the first attempt to knock out multiple, nearly identical, genes in a cellular genome using ZFN. The attempt failed, and other strategies should be used to prevent PERV transmission.
Collapse
Affiliation(s)
| | - Daniel Ivanusic
- Robert Koch Institute, Nordufer 20, Berlin, Germany
- Freie Universität Berlin, Kaiserswerther Str. 16–18, Berlin, Germany
| | - Joachim Denner
- Robert Koch Institute, Nordufer 20, Berlin, Germany
- * E-mail:
| |
Collapse
|
21
|
Hendel A, Fine EJ, Bao G, Porteus MH. Quantifying on- and off-target genome editing. Trends Biotechnol 2015; 33:132-40. [PMID: 25595557 PMCID: PMC4308725 DOI: 10.1016/j.tibtech.2014.12.001] [Citation(s) in RCA: 112] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Revised: 12/02/2014] [Accepted: 12/07/2014] [Indexed: 12/13/2022]
Abstract
Genome editing with engineered nucleases is a rapidly growing field thanks to transformative technologies that allow researchers to precisely alter genomes for numerous applications including basic research, biotechnology, and human gene therapy. While the ability to make precise and controlled changes at specified sites throughout the genome has grown tremendously in recent years, we still lack a comprehensive and standardized battery of assays for measuring the different genome editing outcomes created at endogenous genomic loci. Here we review the existing assays for quantifying on- and off-target genome editing and describe their utility in advancing the technology. We also highlight unmet assay needs for quantifying on- and off-target genome editing outcomes and discuss their importance for the genome editing field.
Collapse
Affiliation(s)
- Ayal Hendel
- Department of Pediatrics, Stanford University, Stanford, CA 94305, USA
| | - Eli J Fine
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| | - Gang Bao
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| | - Matthew H Porteus
- Department of Pediatrics, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
22
|
Lim KI. Recent advances in developing molecular tools for targeted genome engineering of mammalian cells. BMB Rep 2015; 48:6-12. [PMID: 25104401 PMCID: PMC4345644 DOI: 10.5483/bmbrep.2015.48.1.165] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Indexed: 12/23/2022] Open
Abstract
Various biological molecules naturally existing in diversified species including fungi, bacteria, and bacteriophage have functionalities for DNA binding and processing. The biological molecules have been recently actively engineered for use in customized genome editing of mammalian cells as the molecule-encoding DNA sequence information and the underlying mechanisms how the molecules work are unveiled. Excitingly, multiple novel methods based on the newly constructed artificial molecular tools have enabled modifications of specific endogenous genetic elements in the genome context at efficiencies that are much higher than that of the conventional homologous recombination based methods. This minireview introduces the most recently spotlighted molecular genome engineering tools with their key features and ongoing modifications for better performance. Such ongoing efforts have mainly focused on the removal of the inherent DNA sequence recognition rigidity from the original molecular platforms, the addition of newly tailored targeting functions into the engineered molecules, and the enhancement of their targeting specificity. Effective targeted genome engineering of mammalian cells will enable not only sophisticated genetic studies in the context of the genome, but also widely-applicable universal therapeutics based on the pinpointing and correction of the disease-causing genetic elements within the genome in the near future.
Collapse
Affiliation(s)
- Kwang-il Lim
- Department of Medical and Pharmaceutical Sciences, College of Science, Sookmyung Women’s University, Seoul 140-742, Korea
| |
Collapse
|
23
|
Aluru N, Karchner SI, Franks DG, Nacci D, Champlin D, Hahn ME. Targeted mutagenesis of aryl hydrocarbon receptor 2a and 2b genes in Atlantic killifish (Fundulus heteroclitus). AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2015; 158:192-201. [PMID: 25481785 PMCID: PMC4272816 DOI: 10.1016/j.aquatox.2014.11.016] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Revised: 11/17/2014] [Accepted: 11/18/2014] [Indexed: 06/04/2023]
Abstract
Understanding molecular mechanisms of toxicity is facilitated by experimental manipulations, such as disruption of function by gene targeting, that are especially challenging in non-standard model species with limited genomic resources. While loss-of-function approaches have included gene knock-down using morpholino-modified oligonucleotides and random mutagenesis using mutagens or retroviruses, more recent approaches include targeted mutagenesis using zinc finger nuclease (ZFN), transcription activator-like effector nuclease (TALENs) and clustered regularly interspaced short palindromic repeats (CRISPR)-Cas9 technology. These latter methods provide more accessible opportunities to explore gene function in non-traditional model species. To facilitate evaluation of toxic mechanisms for important categories of aryl hydrocarbon pollutants, whose actions are known to be receptor mediated, we used ZFN and CRISPR-Cas9 approaches to generate aryl hydrocarbon receptor 2a (AHR2a) and AHR2b gene mutations in Atlantic killifish (Fundulus heteroclitus) embryos. This killifish is a particularly valuable non-traditional model, with multiple paralogs of AHR whose functions are not well characterized. In addition, some populations of this species have evolved resistance to toxicants such as halogenated aromatic hydrocarbons. AHR-null killifish will be valuable for characterizing the role of the individual AHR paralogs in evolved resistance, as well as in normal development. We first used five-finger ZFNs targeting exons 1 and 3 of AHR2a. Subsequently, CRISPR-Cas9 guide RNAs were designed to target regions in exon 2 and 3 of AHR2a and AHR2b. We successfully induced frameshift mutations in AHR2a exon 3 with ZFN and CRISPR-Cas9 guide RNAs, with mutation frequencies of 10% and 16%, respectively. In AHR2b, mutations were induced using CRISPR-Cas9 guide RNAs targeting sites in both exon 2 (17%) and exon 3 (63%). We screened AHR2b exon 2 CRISPR-Cas9-injected embryos for off-target effects in AHR paralogs. No mutations were observed in closely related AHR genes (AHR1a, AHR1b, AHR2a, AHRR) in the CRISPR-Cas9-injected embryos. Overall, our results demonstrate that targeted genome-editing methods are efficient in inducing mutations at specific loci in embryos of a non-traditional model species, without detectable off-target effects in paralogous genes.
Collapse
Affiliation(s)
- Neelakanteswar Aluru
- Biology Department, Woods Hole Oceanographic Institution, Woods Hole, MA 02543, USA; Superfund Research Program, Boston University School of Public Health, Boston, MA, USA.
| | - Sibel I Karchner
- Biology Department, Woods Hole Oceanographic Institution, Woods Hole, MA 02543, USA; Superfund Research Program, Boston University School of Public Health, Boston, MA, USA
| | - Diana G Franks
- Biology Department, Woods Hole Oceanographic Institution, Woods Hole, MA 02543, USA; Superfund Research Program, Boston University School of Public Health, Boston, MA, USA
| | - Diane Nacci
- Office of Research and Development, National Health and Environmental Effects Research Laboratory, Atlantic Ecology Division, Environmental Protection Agency, Narragansett, RI 02882, USA
| | - Denise Champlin
- Office of Research and Development, National Health and Environmental Effects Research Laboratory, Atlantic Ecology Division, Environmental Protection Agency, Narragansett, RI 02882, USA
| | - Mark E Hahn
- Biology Department, Woods Hole Oceanographic Institution, Woods Hole, MA 02543, USA; Superfund Research Program, Boston University School of Public Health, Boston, MA, USA
| |
Collapse
|
24
|
Zhang C, Xu K, Hu L, Wang L, Zhang T, Ren C, Zhang Z. A suicidal zinc finger nuclease expression coupled with a surrogate reporter for efficient genome engineering. Biotechnol Lett 2014; 37:299-305. [PMID: 25280729 DOI: 10.1007/s10529-014-1690-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Accepted: 09/25/2014] [Indexed: 11/28/2022]
Abstract
Genome editing with engineered nucleases, such as zinc-finger nucleases (ZFNs) and TALE nucleases, remains confronted with a high risk of cellular toxicity induced by off-targeting. Here we describe the construction of a suicidal nuclease expression vector in which a pair of ZFNs genes were flanked of its target sites. To further enrich the targeted cells, the suicidal ZFN expression cassette was also inserted within an eGFP reporter, to disrupt the ORF of eGFP gene. ZFN-associated toxicity was reduced by ~40 % with this new system, and the activities of ZFNs were ~4.5 % lower. We conclude that using this new suicidal ZFN expression and surrogate reporter system represents an improvement for genomic editing by reducing toxicity and allowing easy detection of edited cells by eGFP analysis.
Collapse
Affiliation(s)
- Cunfang Zhang
- College of Animal Science and Technology, Northwest A&F University, No. 22 Xinong Road, Yangling, 712100, Shaanxi, China,
| | | | | | | | | | | | | |
Collapse
|
25
|
Barker JC, Barker AD, Bills J, Huang J, Wight-Carter M, Delgado I, Noble DL, Huang LJ, Porteus MH, Davis KE. Genome Editing of Mouse Fibroblasts by Homologous Recombination for Sustained Secretion of PDGF-B and Augmentation of Wound Healing. Plast Reconstr Surg 2014; 134:389e-401e. [DOI: 10.1097/prs.0000000000000427] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
|
26
|
Enhanced gene disruption by programmable nucleases delivered by a minicircle vector. Gene Ther 2014; 21:921-30. [PMID: 25142139 DOI: 10.1038/gt.2014.76] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2013] [Revised: 06/18/2014] [Accepted: 07/09/2014] [Indexed: 12/21/2022]
Abstract
Targeted genetic modification using programmable nucleases such as zinc finger nucleases (ZFNs) and transcription activator-like effector nucleases (TALENs) is of great value in biomedical research, medicine and biotechnology. Minicircle vectors, which lack extraneous bacterial sequences, have several advantages over conventional plasmids for transgene delivery. Here, for the first time, we delivered programmable nucleases into human cells using transient transfection of a minicircle vector and compared the results with those obtained using a conventional plasmid. Surrogate reporter assays and T7 endonuclease analyses revealed that cells in the minicircle vector group displayed significantly higher mutation frequencies at the target sites than those in the conventional plasmid group. Quantitative PCR and reverse transcription-PCR showed higher vector copy number and programmable nuclease transcript levels, respectively, in 293T cells after minicircle versus conventional plasmid vector transfection. In addition, tryphan blue staining and flow cytometry after annexin V and propidium iodide staining showed that cell viability was also significantly higher in the minicircle group than in the conventional plasmid group. Taken together, our results show that gene disruption using minicircle vector-mediated delivery of ZFNs and TALENs is a more efficient, safer and less toxic method than using a conventional plasmid, and indicate that the minicircle vector could serve as an advanced delivery method for programmable nucleases.
Collapse
|
27
|
Wijshake T, Baker DJ, van de Sluis B. Endonucleases: new tools to edit the mouse genome. Biochim Biophys Acta Mol Basis Dis 2014; 1842:1942-1950. [PMID: 24794718 DOI: 10.1016/j.bbadis.2014.04.020] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Revised: 04/16/2014] [Accepted: 04/18/2014] [Indexed: 12/26/2022]
Abstract
Mouse transgenesis has been instrumental in determining the function of genes in the pathophysiology of human diseases and modification of genes by homologous recombination in mouse embryonic stem cells remains a widely used technology. However, this approach harbors a number of disadvantages, as it is time-consuming and quite laborious. Over the last decade a number of new genome editing technologies have been developed, including zinc finger nucleases (ZFNs), transcription activator-like effector nucleases (TALENs) and clustered regularly interspaced short palindromic repeats/CRISPR-associated (CRISPR/Cas). These systems are characterized by a designed DNA binding protein or RNA sequence fused or co-expressed with a non-specific endonuclease, respectively. The engineered DNA binding protein or RNA sequence guides the nuclease to a specific target sequence in the genome to induce a double strand break. The subsequent activation of the DNA repair machinery then enables the introduction of gene modifications at the target site, such as gene disruption, correction or insertion. Nuclease-mediated genome editing has numerous advantages over conventional gene targeting, including increased efficiency in gene editing, reduced generation time of mutant mice, and the ability to mutagenize multiple genes simultaneously. Although nuclease-driven modifications in the genome are a powerful tool to generate mutant mice, there are concerns about off-target cleavage, especially when using the CRISPR/Cas system. Here, we describe the basic principles of these new strategies in mouse genome manipulation, their inherent advantages, and their potential disadvantages compared to current technologies used to study gene function in mouse models. This article is part of a Special Issue entitled: From Genome to Function.
Collapse
Affiliation(s)
- Tobias Wijshake
- Molecular Genetics, University of Groningen, University Medical Center Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Darren J Baker
- Department of Pediatric and Adolescent Medicine, Mayo Clinic College of Medicine, 200 First St SW, Rochester, MN 55905, USA
| | - Bart van de Sluis
- Molecular Genetics, University of Groningen, University Medical Center Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands.
| |
Collapse
|
28
|
Bach C, Sherman W, Pallis J, Patra P, Bajwa H. Evaluation of novel design strategies for developing zinc finger nucleases tools for treating human diseases. BIOTECHNOLOGY RESEARCH INTERNATIONAL 2014; 2014:970595. [PMID: 24808958 PMCID: PMC3997970 DOI: 10.1155/2014/970595] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Revised: 01/02/2014] [Accepted: 01/02/2014] [Indexed: 11/24/2022]
Abstract
Zinc finger nucleases (ZFNs) are associated with cell death and apoptosis by binding at countless undesired locations. This cytotoxicity is associated with the binding ability of engineered zinc finger domains to bind dissimilar DNA sequences with high affinity. In general, binding preferences of transcription factors are associated with significant degenerated diversity and complexity which convolutes the design and engineering of precise DNA binding domains. Evolutionary success of natural zinc finger proteins, however, evinces that nature created specific evolutionary traits and strategies, such as modularity and rank-specific recognition to cope with binding complexity that are critical for creating clinical viable tools to precisely modify the human genome. Our findings indicate preservation of general modularity and significant alteration of the rank-specific binding preferences of the three-finger binding domain of transcription factor SP1 when exchanging amino acids in the 2nd finger.
Collapse
Affiliation(s)
- Christian Bach
- University of Bridgeport, Biomedical Engineering, 221 University Avenue, Bridgeport, CT 06604, USA
| | - William Sherman
- Physics Faculty, BHSEC Queens, 30-20 Thomson Avenue, Long Island City, NY 11101, USA
| | - Jani Pallis
- University of Bridgeport, Mechanical Engineering, 221 University Avenue, Bridgeport, CT 06604, USA
| | - Prabir Patra
- University of Bridgeport, Biomedical Engineering, 221 University Avenue, Bridgeport, CT 06604, USA
| | - Hassan Bajwa
- University of Bridgeport, Electrical Engineering, 221 University Avenue, Bridgeport, CT 06604, USA
| |
Collapse
|
29
|
Damian M, Porteus MH. A crisper look at genome editing: RNA-guided genome modification. Mol Ther 2013; 21:720-2. [PMID: 23542565 DOI: 10.1038/mt.2013.46] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Affiliation(s)
- Mara Damian
- Department of Pediatrics, Stanford University, Stanford, California 94305, USA
| | | |
Collapse
|
30
|
Voit RA, Hendel A, Pruett-Miller SM, Porteus MH. Nuclease-mediated gene editing by homologous recombination of the human globin locus. Nucleic Acids Res 2013; 42:1365-78. [PMID: 24157834 PMCID: PMC3902937 DOI: 10.1093/nar/gkt947] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Tal-effector nucleases (TALENs) are engineered proteins that can stimulate precise genome editing through specific DNA double-strand breaks. Sickle cell disease and β-thalassemia are common genetic disorders caused by mutations in β-globin, and we engineered a pair of highly active TALENs that induce modification of 54% of human β-globin alleles near the site of the sickle mutation. These TALENS stimulate targeted integration of therapeutic, full-length beta-globin cDNA to the endogenous β-globin locus in 19% of cells prior to selection as quantified by single molecule real-time sequencing. We also developed highly active TALENs to human γ-globin, a pharmacologic target in sickle cell disease therapy. Using the β-globin and γ-globin TALENs, we generated cell lines that express GFP under the control of the endogenous β-globin promoter and tdTomato under the control of the endogenous γ-globin promoter. With these fluorescent reporter cell lines, we screened a library of small molecule compounds for their differential effect on the transcriptional activity of the endogenous β- and γ-globin genes and identified several that preferentially upregulate γ-globin expression.
Collapse
Affiliation(s)
- Richard A Voit
- Department of Pediatrics, Stanford University, 1291 Welch Rd. Stanford, CA 94305, USA and Department of Pediatrics, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd. Dallas, TX 75390, USA
| | | | | | | |
Collapse
|
31
|
Abstract
The limited sequence similarity of protein sequences with known structures has led to an indispensable need for computational technology to predict their structures. Structural bioinformatics (SB) has become integral in elucidating the sequence-structure-function relationship of a protein. This report focuses on the applications of SB within the context of protein engineering including its limitation and future challenges.
Collapse
Affiliation(s)
- Yee Siew Choong
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, 11800, Minden, Pulau Pinang, Malaysia,
| | | | | |
Collapse
|
32
|
Owens JB, Mauro D, Stoytchev I, Bhakta MS, Kim MS, Segal DJ, Moisyadi S. Transcription activator like effector (TALE)-directed piggyBac transposition in human cells. Nucleic Acids Res 2013; 41:9197-207. [PMID: 23921635 PMCID: PMC3799441 DOI: 10.1093/nar/gkt677] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Revised: 07/09/2013] [Accepted: 07/10/2013] [Indexed: 12/13/2022] Open
Abstract
Insertional therapies have shown great potential for combating genetic disease and safer methods would undoubtedly broaden the variety of possible illness that can be treated. A major challenge that remains is reducing the risk of insertional mutagenesis due to random insertion by both viral and non-viral vectors. Targetable nucleases are capable of inducing double-stranded breaks to enhance homologous recombination for the introduction of transgenes at specific sequences. However, off-target DNA cleavages at unknown sites can lead to mutations that are difficult to detect. Alternatively, the piggyBac transposase is able perform all of the steps required for integration; therefore, cells confirmed to contain a single copy of a targeted transposon, for which its location is known, are likely to be devoid of aberrant genomic modifications. We aimed to retarget transposon insertions by comparing a series of novel hyperactive piggyBac constructs tethered to a custom transcription activator like effector DNA-binding domain designed to bind the first intron of the human CCR5 gene. Multiple targeting strategies were evaluated using combinations of both plasmid-DNA and transposase-protein relocalization to the target sequence. We demonstrated user-defined directed transposition to the CCR5 genomic safe harbor and isolated single-copy clones harboring targeted integrations.
Collapse
Affiliation(s)
- Jesse B. Owens
- Institute for Biogenesis Research, Department of Anatomy, Biochemistry and Physiology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI 96822, USA, Genome Center, Department of Biochemistry and Molecular Medicine, University of California, Davis, CA 95616, USA and Manoa BioSciences, Honolulu, HI 96819, USA
| | - Damiano Mauro
- Institute for Biogenesis Research, Department of Anatomy, Biochemistry and Physiology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI 96822, USA, Genome Center, Department of Biochemistry and Molecular Medicine, University of California, Davis, CA 95616, USA and Manoa BioSciences, Honolulu, HI 96819, USA
| | - Ilko Stoytchev
- Institute for Biogenesis Research, Department of Anatomy, Biochemistry and Physiology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI 96822, USA, Genome Center, Department of Biochemistry and Molecular Medicine, University of California, Davis, CA 95616, USA and Manoa BioSciences, Honolulu, HI 96819, USA
| | - Mital S. Bhakta
- Institute for Biogenesis Research, Department of Anatomy, Biochemistry and Physiology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI 96822, USA, Genome Center, Department of Biochemistry and Molecular Medicine, University of California, Davis, CA 95616, USA and Manoa BioSciences, Honolulu, HI 96819, USA
| | - Moon-Soo Kim
- Institute for Biogenesis Research, Department of Anatomy, Biochemistry and Physiology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI 96822, USA, Genome Center, Department of Biochemistry and Molecular Medicine, University of California, Davis, CA 95616, USA and Manoa BioSciences, Honolulu, HI 96819, USA
| | - David J. Segal
- Institute for Biogenesis Research, Department of Anatomy, Biochemistry and Physiology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI 96822, USA, Genome Center, Department of Biochemistry and Molecular Medicine, University of California, Davis, CA 95616, USA and Manoa BioSciences, Honolulu, HI 96819, USA
| | - Stefan Moisyadi
- Institute for Biogenesis Research, Department of Anatomy, Biochemistry and Physiology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI 96822, USA, Genome Center, Department of Biochemistry and Molecular Medicine, University of California, Davis, CA 95616, USA and Manoa BioSciences, Honolulu, HI 96819, USA
| |
Collapse
|
33
|
Chen Z, Jaafar L, Agyekum DG, Xiao H, Wade MF, Kumaran RI, Spector DL, Bao G, Porteus MH, Dynan WS, Meiler SE. Receptor-mediated delivery of engineered nucleases for genome modification. Nucleic Acids Res 2013; 41:e182. [PMID: 23956220 PMCID: PMC3799454 DOI: 10.1093/nar/gkt710] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Engineered nucleases, which incise the genome at predetermined sites, have a number of laboratory and clinical applications. There is, however, a need for better methods for controlled intracellular delivery of nucleases. Here, we demonstrate a method for ligand-mediated delivery of zinc finger nucleases (ZFN) proteins using transferrin receptor-mediated endocytosis. Uptake is rapid and efficient in established mammalian cell lines and in primary cells, including mouse and human hematopoietic stem-progenitor cell populations. In contrast to cDNA expression, ZFN protein levels decline rapidly following internalization, affording better temporal control of nuclease activity. We show that transferrin-mediated ZFN uptake leads to site-specific in situ cleavage of the target locus. Additionally, despite the much shorter duration of ZFN activity, the efficiency of gene correction approaches that seen with cDNA-mediated expression. The approach is flexible and general, with the potential for extension to other targeting ligands and nuclease architectures.
Collapse
Affiliation(s)
- Zhong Chen
- Department of Anesthesiology and Perioperative Medicine, Georgia Regents University, 1120 15th Street, Augusta, GA 30912, USA, Department of Radiation Oncology, Emory University School of Medicine, 4121 Rollins Research Center, 1510 Clifton Rd. NE, Atlanta, GA 30322, USA, Cold Spring Harbor Laboratory, 1 Bungtown Road, Cold Spring Harbor, New York 11724, USA, Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30322, USA, Department of Pediatrics-Divisions of Hematology/Oncology and Human Gene Therapy, Stanford University School of Medicine, Stanford, CA 94305, USA and Department of Biochemistry, Emory University School of Medicine, 4121 Rollins Research Center, 1510 Clifton Rd. NE, Atlanta, GA 30322, USA
| | - Lahcen Jaafar
- Department of Anesthesiology and Perioperative Medicine, Georgia Regents University, 1120 15th Street, Augusta, GA 30912, USA, Department of Radiation Oncology, Emory University School of Medicine, 4121 Rollins Research Center, 1510 Clifton Rd. NE, Atlanta, GA 30322, USA, Cold Spring Harbor Laboratory, 1 Bungtown Road, Cold Spring Harbor, New York 11724, USA, Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30322, USA, Department of Pediatrics-Divisions of Hematology/Oncology and Human Gene Therapy, Stanford University School of Medicine, Stanford, CA 94305, USA and Department of Biochemistry, Emory University School of Medicine, 4121 Rollins Research Center, 1510 Clifton Rd. NE, Atlanta, GA 30322, USA
| | - Davies G. Agyekum
- Department of Anesthesiology and Perioperative Medicine, Georgia Regents University, 1120 15th Street, Augusta, GA 30912, USA, Department of Radiation Oncology, Emory University School of Medicine, 4121 Rollins Research Center, 1510 Clifton Rd. NE, Atlanta, GA 30322, USA, Cold Spring Harbor Laboratory, 1 Bungtown Road, Cold Spring Harbor, New York 11724, USA, Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30322, USA, Department of Pediatrics-Divisions of Hematology/Oncology and Human Gene Therapy, Stanford University School of Medicine, Stanford, CA 94305, USA and Department of Biochemistry, Emory University School of Medicine, 4121 Rollins Research Center, 1510 Clifton Rd. NE, Atlanta, GA 30322, USA
| | - Haiyan Xiao
- Department of Anesthesiology and Perioperative Medicine, Georgia Regents University, 1120 15th Street, Augusta, GA 30912, USA, Department of Radiation Oncology, Emory University School of Medicine, 4121 Rollins Research Center, 1510 Clifton Rd. NE, Atlanta, GA 30322, USA, Cold Spring Harbor Laboratory, 1 Bungtown Road, Cold Spring Harbor, New York 11724, USA, Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30322, USA, Department of Pediatrics-Divisions of Hematology/Oncology and Human Gene Therapy, Stanford University School of Medicine, Stanford, CA 94305, USA and Department of Biochemistry, Emory University School of Medicine, 4121 Rollins Research Center, 1510 Clifton Rd. NE, Atlanta, GA 30322, USA
| | - Marlene F. Wade
- Department of Anesthesiology and Perioperative Medicine, Georgia Regents University, 1120 15th Street, Augusta, GA 30912, USA, Department of Radiation Oncology, Emory University School of Medicine, 4121 Rollins Research Center, 1510 Clifton Rd. NE, Atlanta, GA 30322, USA, Cold Spring Harbor Laboratory, 1 Bungtown Road, Cold Spring Harbor, New York 11724, USA, Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30322, USA, Department of Pediatrics-Divisions of Hematology/Oncology and Human Gene Therapy, Stanford University School of Medicine, Stanford, CA 94305, USA and Department of Biochemistry, Emory University School of Medicine, 4121 Rollins Research Center, 1510 Clifton Rd. NE, Atlanta, GA 30322, USA
| | - R. Ileng Kumaran
- Department of Anesthesiology and Perioperative Medicine, Georgia Regents University, 1120 15th Street, Augusta, GA 30912, USA, Department of Radiation Oncology, Emory University School of Medicine, 4121 Rollins Research Center, 1510 Clifton Rd. NE, Atlanta, GA 30322, USA, Cold Spring Harbor Laboratory, 1 Bungtown Road, Cold Spring Harbor, New York 11724, USA, Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30322, USA, Department of Pediatrics-Divisions of Hematology/Oncology and Human Gene Therapy, Stanford University School of Medicine, Stanford, CA 94305, USA and Department of Biochemistry, Emory University School of Medicine, 4121 Rollins Research Center, 1510 Clifton Rd. NE, Atlanta, GA 30322, USA
| | - David L. Spector
- Department of Anesthesiology and Perioperative Medicine, Georgia Regents University, 1120 15th Street, Augusta, GA 30912, USA, Department of Radiation Oncology, Emory University School of Medicine, 4121 Rollins Research Center, 1510 Clifton Rd. NE, Atlanta, GA 30322, USA, Cold Spring Harbor Laboratory, 1 Bungtown Road, Cold Spring Harbor, New York 11724, USA, Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30322, USA, Department of Pediatrics-Divisions of Hematology/Oncology and Human Gene Therapy, Stanford University School of Medicine, Stanford, CA 94305, USA and Department of Biochemistry, Emory University School of Medicine, 4121 Rollins Research Center, 1510 Clifton Rd. NE, Atlanta, GA 30322, USA
| | - Gang Bao
- Department of Anesthesiology and Perioperative Medicine, Georgia Regents University, 1120 15th Street, Augusta, GA 30912, USA, Department of Radiation Oncology, Emory University School of Medicine, 4121 Rollins Research Center, 1510 Clifton Rd. NE, Atlanta, GA 30322, USA, Cold Spring Harbor Laboratory, 1 Bungtown Road, Cold Spring Harbor, New York 11724, USA, Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30322, USA, Department of Pediatrics-Divisions of Hematology/Oncology and Human Gene Therapy, Stanford University School of Medicine, Stanford, CA 94305, USA and Department of Biochemistry, Emory University School of Medicine, 4121 Rollins Research Center, 1510 Clifton Rd. NE, Atlanta, GA 30322, USA
| | - Matthew H. Porteus
- Department of Anesthesiology and Perioperative Medicine, Georgia Regents University, 1120 15th Street, Augusta, GA 30912, USA, Department of Radiation Oncology, Emory University School of Medicine, 4121 Rollins Research Center, 1510 Clifton Rd. NE, Atlanta, GA 30322, USA, Cold Spring Harbor Laboratory, 1 Bungtown Road, Cold Spring Harbor, New York 11724, USA, Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30322, USA, Department of Pediatrics-Divisions of Hematology/Oncology and Human Gene Therapy, Stanford University School of Medicine, Stanford, CA 94305, USA and Department of Biochemistry, Emory University School of Medicine, 4121 Rollins Research Center, 1510 Clifton Rd. NE, Atlanta, GA 30322, USA
| | - William S. Dynan
- Department of Anesthesiology and Perioperative Medicine, Georgia Regents University, 1120 15th Street, Augusta, GA 30912, USA, Department of Radiation Oncology, Emory University School of Medicine, 4121 Rollins Research Center, 1510 Clifton Rd. NE, Atlanta, GA 30322, USA, Cold Spring Harbor Laboratory, 1 Bungtown Road, Cold Spring Harbor, New York 11724, USA, Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30322, USA, Department of Pediatrics-Divisions of Hematology/Oncology and Human Gene Therapy, Stanford University School of Medicine, Stanford, CA 94305, USA and Department of Biochemistry, Emory University School of Medicine, 4121 Rollins Research Center, 1510 Clifton Rd. NE, Atlanta, GA 30322, USA
- *To whom correspondence should be addressed. Tel: +1 706 721 3287; Fax: +1 706 434 7131;
| | - Steffen E. Meiler
- Department of Anesthesiology and Perioperative Medicine, Georgia Regents University, 1120 15th Street, Augusta, GA 30912, USA, Department of Radiation Oncology, Emory University School of Medicine, 4121 Rollins Research Center, 1510 Clifton Rd. NE, Atlanta, GA 30322, USA, Cold Spring Harbor Laboratory, 1 Bungtown Road, Cold Spring Harbor, New York 11724, USA, Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30322, USA, Department of Pediatrics-Divisions of Hematology/Oncology and Human Gene Therapy, Stanford University School of Medicine, Stanford, CA 94305, USA and Department of Biochemistry, Emory University School of Medicine, 4121 Rollins Research Center, 1510 Clifton Rd. NE, Atlanta, GA 30322, USA
- *To whom correspondence should be addressed. Tel: +1 706 721 3287; Fax: +1 706 434 7131;
| |
Collapse
|
34
|
Hauschild-Quintern J, Petersen B, Cost GJ, Niemann H. Gene knockout and knockin by zinc-finger nucleases: current status and perspectives. Cell Mol Life Sci 2013; 70:2969-83. [PMID: 23161061 PMCID: PMC11113862 DOI: 10.1007/s00018-012-1204-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2012] [Revised: 10/19/2012] [Accepted: 10/22/2012] [Indexed: 12/01/2022]
Abstract
Zinc-finger nucleases (ZFNs) are engineered site-specific DNA cleavage enzymes that may be designed to recognize long target sites and thus cut DNA with high specificity. ZFNs mediate permanent and targeted genetic alteration via induction of a double-strand break at a specific genomic site. Compared to conventional homology-based gene targeting, ZFNs can increase the targeting rate by up to 100,000-fold; gene disruption via mutagenic DNA repair is similarly efficient. The utility of ZFNs has been shown in many organisms, including insects, amphibians, plants, nematodes, and several mammals, including humans. This broad range of tractable species renders ZFNs a useful tool for improving the understanding of complex physiological systems, to produce transgenic animals, cell lines, and plants, and to treat human disease.
Collapse
Affiliation(s)
- J. Hauschild-Quintern
- Institute of Farm Animal Genetics, Friedrich-Loeffler-Institut, Mariensee, Hoeltystrasse 10, 31535 Neustadt a. Rbge., Germany
| | - B. Petersen
- Institute of Farm Animal Genetics, Friedrich-Loeffler-Institut, Mariensee, Hoeltystrasse 10, 31535 Neustadt a. Rbge., Germany
| | - G. J. Cost
- Sangamo BioSciences, 501 Canal Blvd., Richmond, CA 94804 USA
| | - H. Niemann
- Institute of Farm Animal Genetics, Friedrich-Loeffler-Institut, Mariensee, Hoeltystrasse 10, 31535 Neustadt a. Rbge., Germany
- Rebirth, Cluster of Excellence, Hannover Medical School, Hannover, Germany
| |
Collapse
|
35
|
Pillay LM, Selland LG, Fleisch VC, Leighton PLA, Cheng CS, Famulski JK, Ritzel RG, March LD, Wang H, Allison WT, Waskiewicz AJ. Evaluating the mutagenic activity of targeted endonucleases containing a Sharkey FokI cleavage domain variant in zebrafish. Zebrafish 2013; 10:353-64. [PMID: 23781947 DOI: 10.1089/zeb.2012.0832] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Synthetic targeted endonucleases such as zinc finger nucleases (ZFNs) and transcription activator-like effector nucleases (TALENs) have recently emerged as powerful tools for targeted mutagenesis, especially in organisms that are not amenable to embryonic stem cell manipulation. Both ZFNs and TALENs consist of DNA-binding arrays that are fused to the nonspecific FokI nuclease domain. In an effort to improve targeted endonuclease mutagenesis efficiency, we enhanced their catalytic activity using the Sharkey FokI nuclease domain variant. All constructs tested display increased DNA cleavage activity in vitro. We demonstrate that one out of four ZFN arrays containing the Sharkey FokI variant exhibits a dramatic increase in mutagenesis frequency in vivo in zebrafish. The other three ZFNs exhibit no significant alteration of activity in vivo. Conversely, we demonstrate that TALENs containing the Sharkey FokI variant exhibit absent or severely reduced in vivo mutagenic activity in zebrafish. Notably, Sharkey ZFNs and TALENs do not generate increased toxicity-related defects or mortality. Our results present Sharkey ZFNs as an effective alternative to conventional ZFNs, but advise against the use of Sharkey TALENs.
Collapse
Affiliation(s)
- Laura M Pillay
- Department of Biological Sciences, University of Alberta , Edmonton, Canada
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Richter S, Morrison S, Connor T, Su J, Print CG, Ronimus RS, McGee SL, Wilson WR. Zinc finger nuclease mediated knockout of ADP-dependent glucokinase in cancer cell lines: effects on cell survival and mitochondrial oxidative metabolism. PLoS One 2013; 8:e65267. [PMID: 23799003 PMCID: PMC3683018 DOI: 10.1371/journal.pone.0065267] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Accepted: 04/23/2013] [Indexed: 01/22/2023] Open
Abstract
Zinc finger nucleases (ZFN) are powerful tools for editing genes in cells. Here we use ZFNs to interrogate the biological function of ADPGK, which encodes an ADP-dependent glucokinase (ADPGK), in human tumour cell lines. The hypothesis we tested is that ADPGK utilises ADP to phosphorylate glucose under conditions where ATP becomes limiting, such as hypoxia. We characterised two ZFN knockout clones in each of two lines (H460 and HCT116). All four clones had frameshift mutations in all alleles at the target site in exon 1 of ADPGK, and were ADPGK-null by immunoblotting. ADPGK knockout had little or no effect on cell proliferation, but compromised the ability of H460 cells to survive siRNA silencing of hexokinase-2 under oxic conditions, with clonogenic survival falling from 21±3% for the parental line to 6.4±0.8% (p = 0.002) and 4.3±0.8% (p = 0.001) for the two knockouts. A similar increased sensitivity to clonogenic cell killing was observed under anoxia. No such changes were found when ADPGK was knocked out in HCT116 cells, for which the parental line was less sensitive than H460 to anoxia and to hexokinase-2 silencing. While knockout of ADPGK in HCT116 cells caused few changes in global gene expression, knockout of ADPGK in H460 cells caused notable up-regulation of mRNAs encoding cell adhesion proteins. Surprisingly, we could discern no consistent effect on glycolysis as measured by glucose consumption or lactate formation under anoxia, or extracellular acidification rate (Seahorse XF analyser) under oxic conditions in a variety of media. However, oxygen consumption rates were generally lower in the ADPGK knockouts, in some cases markedly so. Collectively, the results demonstrate that ADPGK can contribute to tumour cell survival under conditions of high glycolytic dependence, but the phenotype resulting from knockout of ADPGK is cell line dependent and appears to be unrelated to priming of glycolysis in these lines.
Collapse
Affiliation(s)
- Susan Richter
- Auckland Cancer Society Research Centre, The University of Auckland, Auckland, New Zealand
| | - Shona Morrison
- Metabolic Research Unit, School of Medicine, Deakin University, Geelong, Victoria, Australia
| | - Tim Connor
- Metabolic Research Unit, School of Medicine, Deakin University, Geelong, Victoria, Australia
| | - Jiechuang Su
- Auckland Cancer Society Research Centre, The University of Auckland, Auckland, New Zealand
| | - Cristin G. Print
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
- The New Zealand Bioinformatics Institute, The University of Auckland, Auckland, New Zealand
| | | | - Sean L. McGee
- Metabolic Research Unit, School of Medicine, Deakin University, Geelong, Victoria, Australia
| | - William R. Wilson
- Auckland Cancer Society Research Centre, The University of Auckland, Auckland, New Zealand
- * E-mail:
| |
Collapse
|
37
|
Osborn MJ, Starker CG, McElroy AN, Webber BR, Riddle MJ, Xia L, DeFeo AP, Gabriel R, Schmidt M, Von Kalle C, Carlson DF, Maeder ML, Joung JK, Wagner JE, Voytas DF, Blazar BR, Tolar J. TALEN-based gene correction for epidermolysis bullosa. Mol Ther 2013; 21:1151-9. [PMID: 23546300 PMCID: PMC3677309 DOI: 10.1038/mt.2013.56] [Citation(s) in RCA: 205] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Recessive dystrophic epidermolysis bullosa (RDEB) is characterized by a functional deficit of type VII collagen protein due to gene defects in the type VII collagen gene (COL7A1). Gene augmentation therapies are promising, but run the risk of insertional mutagenesis. To abrogate this risk, we explored the possibility of using engineered transcription activator-like effector nucleases (TALEN) for precise genome editing. We report the ability of TALEN to induce site-specific double-stranded DNA breaks (DSBs) leading to homology-directed repair (HDR) from an exogenous donor template. This process resulted in COL7A1 gene mutation correction in primary fibroblasts that were subsequently reprogrammed into inducible pluripotent stem cells and showed normal protein expression and deposition in a teratoma-based skin model in vivo. Deep sequencing-based genome-wide screening established a safety profile showing on-target activity and three off-target (OT) loci that, importantly, were at least 10 kb from a coding sequence. This study provides proof-of-concept for TALEN-mediated in situ correction of an endogenous patient-specific gene mutation and used an unbiased screen for comprehensive TALEN target mapping that will cooperatively facilitate translational application.
Collapse
Affiliation(s)
- Mark J Osborn
- Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, Minnesota, USA
| | - Colby G Starker
- Center for Genome Engineering, University of Minnesota, Minneapolis, Minnesota, USA
- Department of Genetics, Cell Biology & Development, University of Minnesota, Minneapolis, Minnesota, USA
| | - Amber N McElroy
- Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Beau R Webber
- Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Megan J Riddle
- Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Lily Xia
- Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Anthony P DeFeo
- Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Richard Gabriel
- Department of Translational Oncology, National Center for Tumor Diseases, Heidelberg, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Manfred Schmidt
- Department of Translational Oncology, National Center for Tumor Diseases, Heidelberg, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Christof Von Kalle
- Department of Translational Oncology, National Center for Tumor Diseases, Heidelberg, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Daniel F Carlson
- Center for Genome Engineering, University of Minnesota, Minneapolis, Minnesota, USA
| | - Morgan L Maeder
- Molecular Pathology Unit, Center for Computational & Integrative Biology, and Center for Cancer Research, Massachusetts General Hospital, Charlestown, Massachusetts, USA
- Program in Biological and Biomedical Sciences, Harvard Medical School, Boston, Massachusetts, USA
| | - J Keith Joung
- Molecular Pathology Unit, Center for Computational & Integrative Biology, and Center for Cancer Research, Massachusetts General Hospital, Charlestown, Massachusetts, USA
- Program in Biological and Biomedical Sciences, Harvard Medical School, Boston, Massachusetts, USA
- Department of Pathology, Harvard Medical School, Boston, MA, USA
| | - John E Wagner
- Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Daniel F Voytas
- Center for Genome Engineering, University of Minnesota, Minneapolis, Minnesota, USA
- Department of Genetics, Cell Biology & Development, University of Minnesota, Minneapolis, Minnesota, USA
| | - Bruce R Blazar
- Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Jakub Tolar
- Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
- Blood and Marrow Transplantation, University of Minnesota Medical School, MMC 366, 420 Delaware Street SE, Minneapolis, Minnesota 55455, USA. E-mail:
| |
Collapse
|
38
|
Wilson KA, McEwen AE, Pruett-Miller SM, Zhang J, Kildebeck EJ, Porteus MH. Expanding the Repertoire of Target Sites for Zinc Finger Nuclease-mediated Genome Modification. MOLECULAR THERAPY. NUCLEIC ACIDS 2013; 2:e88. [PMID: 23632390 PMCID: PMC3650245 DOI: 10.1038/mtna.2013.13] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/17/2012] [Accepted: 02/05/2013] [Indexed: 11/08/2022]
Abstract
Recent studies have shown that zinc finger nucleases (ZFNs) are powerful reagents for making site-specific genomic modifications. The generic structure of these enzymes includes a ZF DNA-binding domain and nuclease domain (Fn) are separated by an amino acid "linker" and cut genomic DNA at sites that have a generic structure (site1)-(spacer)-(site2) where the "spacer" separates the two binding sites. In this work, we compare the activity of ZFNs with different linkers on target sites with different spacer lengths. We found those nucleases with linkers' lengths of 2 or 4 amino acid (aa) efficiently cut at target sites with 5 or 6 base pair (bp) spacers, and that those ZFNs with a 5-aa linker length efficiently cut target sites with 6 or 7 bp spacers. In addition, we demonstrate that the Oligomerized Pool ENgineering (OPEN) platform used for making three-fingered ZF proteins (ZFPs) can be modified to incorporate modular assembly fingers (including those recognizing ANNs, CNNs, and TNNs) and we were able to generate nucleases that efficiently cut cognate target sites. The ability to use module fingers in the OPEN platform at target sites of 5-7 bp spacer lengths increases the probability of finding a ZFN target site to 1 in 4 bp. These findings significantly expand the range of sites that can be potentially targeted by these custom-engineered proteins.Molecular Therapy - Nucleic Acids (2013) 2, e88; doi:10.1038/mtna.2013.13; published online 30 April 2013.
Collapse
Affiliation(s)
- Kimberly A Wilson
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Abbye E McEwen
- Department of Medicine, Division of Pulmonary and Critical Care, Northwestern University, Chicago, Illinois, USA
| | - Shondra M Pruett-Miller
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Jiuli Zhang
- Department of Biological Sciences, Florida Atlantic University, Boca Raton, Florida, USA
| | - Eric J Kildebeck
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Matthew H Porteus
- Department of Pediatrics, Stanford University, Stanford, California, USA
| |
Collapse
|
39
|
Wilson KA, Chateau ML, Porteus MH. Design and Development of Artificial Zinc Finger Transcription Factors and Zinc Finger Nucleases to the hTERT Locus. MOLECULAR THERAPY. NUCLEIC ACIDS 2013; 2:e87. [PMID: 23612114 PMCID: PMC3650244 DOI: 10.1038/mtna.2013.12] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2012] [Accepted: 02/05/2013] [Indexed: 01/07/2023]
Abstract
The ability to direct human telomerase reverse transcriptase (hTERT) expression through either genetic control or tunable regulatory factors would advance not only our understanding of the transcriptional regulation of this gene, but also potentially produce new strategies for addressing telomerase-associated disease. In this work, we describe the engineering of artificial zinc finger transcription factors (ZFTFs) and ZF nucleases (ZFNs) to target sequences within the hTERT promoter and exon-1. We were able to identify several active ZFTFs that demonstrate a broadly tunable response when screened by a cell-based transcriptional reporter assay. Using the same DNA-binding domains, we generated ZFNs that were screened in combinatorial pairs in cell-based extrachromosomal single-strand annealing (SSA) assays and in gene-targeting assays using stably integrated constructs. Selected ZFN pairs were tested for the ability to induce sequence changes in a Cel1 assay and we observed frequencies of genomic modification up to 18.7% at the endogenous hTERT locus. These screening strategies have pinpointed several ZFN pairs that may be useful in gene editing of the hTERT locus. Our work provides a foundation for using engineered ZF proteins (ZFPs) for modulation of the hTERT locus.Molecular Therapy - Nucleic Acids (2013) 2, e87; doi:10.1038/mtna.2013.12; published online 23 April 2013.
Collapse
Affiliation(s)
- Kimberly A Wilson
- Department of Biochemistry, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA
- Department of Pediatrics, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA
| | - Morgan L Chateau
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Matthew H Porteus
- Department of Biochemistry, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA
- Department of Pediatrics, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA
- Department of Pediatrics, Stanford University, Stanford, California, USA
| |
Collapse
|
40
|
Nuclease Mediated Targeted Genome Modification in Mammalian Cells. SITE-DIRECTED INSERTION OF TRANSGENES 2013. [DOI: 10.1007/978-94-007-4531-5_13] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
41
|
Engineered Zinc Finger Nucleases for Targeted Genome Editing. SITE-DIRECTED INSERTION OF TRANSGENES 2013. [DOI: 10.1007/978-94-007-4531-5_5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
42
|
Ellis BL, Hirsch ML, Porter SN, Samulski RJ, Porteus MH. Zinc-finger nuclease-mediated gene correction using single AAV vector transduction and enhancement by Food and Drug Administration-approved drugs. Gene Ther 2013; 20:35-42. [PMID: 22257934 PMCID: PMC4957644 DOI: 10.1038/gt.2011.211] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2011] [Revised: 11/30/2011] [Accepted: 12/02/2011] [Indexed: 11/08/2022]
Abstract
An emerging strategy for the treatment of monogenic diseases uses genetic engineering to precisely correct the mutation(s) at the genome level. Recent advancements in this technology have demonstrated therapeutic levels of gene correction using a zinc-finger nuclease (ZFN)-induced DNA double-strand break in conjunction with an exogenous DNA donor substrate. This strategy requires efficient nucleic acid delivery and among viral vectors, recombinant adeno-associated virus (rAAV) has demonstrated clinical success without pathology. However, a major limitation of rAAV is the small DNA packaging capacity and to date, the use of rAAV for ZFN gene delivery has yet to be reported. Theoretically, an ideal situation is to deliver both ZFNs and the repair substrate in a single vector to avoid inefficient gene targeting and unwanted mutagenesis, both complications of a rAAV co-transduction strategy. Therefore, a rAAV format was generated in which a single polypeptide encodes the ZFN monomers connected by a ribosome skipping 2A peptide and furin cleavage sequence. On the basis of this arrangement, a DNA repair substrate of 750 nucleotides was also included in this vector. Efficient polypeptide processing to discrete ZFNs is demonstrated, as well as the ability of this single vector format to stimulate efficient gene targeting in a human cell line and mouse model derived fibroblasts. Additionally, we increased rAAV-mediated gene correction up to sixfold using a combination of Food and Drug Administration-approved drugs, which act at the level of AAV vector transduction. Collectively, these experiments demonstrate the ability to deliver ZFNs and a repair substrate by a single AAV vector and offer insights for the optimization of rAAV-mediated gene correction using drug therapy.
Collapse
Affiliation(s)
- BL Ellis
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - ML Hirsch
- UNC Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - SN Porter
- Department of Pediatrics, Stanford Medical School, Stanford, CA, USA
| | - RJ Samulski
- UNC Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - MH Porteus
- Department of Pediatrics, Stanford Medical School, Stanford, CA, USA
| |
Collapse
|
43
|
Nishijima KI, Iijima S. Transgenic chickens. Dev Growth Differ 2012; 55:207-16. [DOI: 10.1111/dgd.12032] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2012] [Revised: 11/05/2012] [Accepted: 11/20/2012] [Indexed: 12/01/2022]
Affiliation(s)
- Ken-ichi Nishijima
- Department of Biotechnology; Graduate School of Engineering; Nagoya University; Furo-cho, Chikusa-ku; Nagoya; 464-8603; Japan
| | - Shinji Iijima
- Department of Biotechnology; Graduate School of Engineering; Nagoya University; Furo-cho, Chikusa-ku; Nagoya; 464-8603; Japan
| |
Collapse
|
44
|
Obligate ligation-gated recombination (ObLiGaRe): custom-designed nuclease-mediated targeted integration through nonhomologous end joining. Genome Res 2012; 23:539-46. [PMID: 23152450 PMCID: PMC3589542 DOI: 10.1101/gr.145441.112] [Citation(s) in RCA: 233] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Custom-designed nucleases (CDNs) greatly facilitate genetic engineering by generating a targeted DNA double-strand break (DSB) in the genome. Once a DSB is created, specific modifications can be introduced around the breakage site during its repair by two major DNA damage repair (DDR) mechanisms: the dominant but error-prone nonhomologous end joining (NHEJ) pathway, and the less-frequent but precise homologous recombination (HR) pathway. Here we describe ObLiGaRe, a new method for site-specific gene insertions that uses the efficient NHEJ pathway and acts independently of HR. This method is applicable with both zinc finger nucleases (ZFNs) and Tale nucleases (TALENs), and has enabled us to insert a 15-kb inducible gene expression cassette at a defined locus in human cell lines. In addition, our experiments have revealed the previously underestimated error-free nature of NHEJ and provided new tools to further characterize this pathway under physiological and pathological conditions.
Collapse
|
45
|
Zhang W, Guo Y, Zhang C, Ji H, Meng W, Wang D, Li X, Mao Q, Xia H. Rescue the failed half-ZFN by a sensitive mammalian cell-based luciferase reporter system. PLoS One 2012; 7:e45169. [PMID: 23028823 PMCID: PMC3445457 DOI: 10.1371/journal.pone.0045169] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2012] [Accepted: 08/14/2012] [Indexed: 11/30/2022] Open
Abstract
ZFN technology is a powerful research tool and has been used for genome editing in cells lines, animals and plants. The generation of functional ZFNs for particular targets in mammalian genome is still challenging for an average research group. The modular-assembly method is relatively fast, easy-to-practice but has a high failure rate. Some recent studies suggested that a ZFP with low binding activity might be able to form a working ZFN pair with another binding active half-ZFP. In order to unveil the potential ZFP candidates among those with low binding activities, this paper established a highly sensitive mammalian cell-based transcriptional reporter system to assess the DNA binding activities of ZFPs by inserting multiple copies of ZFN target sequence fragment (TSF) of an interested gene (e. g., hPGRN or hVEGF). Our results showed that this system increased the screening sensitivity up to 50-fold and markedly amplified the differences in the binding activities between different ZFPs. We also found that the targeted chromosomal gene repair efficiency of each hPGRN or hVEGF ZFN pair was in proportion with the combination of the binding activities of the ZFL (Left zinc finger) and ZFR (Right zinc finger). A hPGRN ZFR with low binding ability was able to form a biological active ZFN if combined with a hPGRN ZFL with relatively high binding ability. Lastly, site-specific genome editing by hPGRN ZFNs generated by this system was confirmed by sequencing, and the PGRN knock-out cell line showed significantly decreased cell growth compared with the control. Our system will provide a valuable tool for further optimizing the nucleases with regard to specificity and cytotoxicity.
Collapse
Affiliation(s)
- Weifeng Zhang
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, Xi’an, Shaanxi, People’s Republic of China
| | - Yuanxu Guo
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, Xi’an, Shaanxi, People’s Republic of China
| | - Chen Zhang
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, Xi’an, Shaanxi, People’s Republic of China
| | - Haiyan Ji
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, Xi’an, Shaanxi, People’s Republic of China
| | - Wenpeng Meng
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, Xi’an, Shaanxi, People’s Republic of China
| | - Dongyang Wang
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, Xi’an, Shaanxi, People’s Republic of China
| | - Xing Li
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, Xi’an, Shaanxi, People’s Republic of China
| | - Qinwen Mao
- Departmet of Pathology, Northwestern University Feinberg School of Medicine Chicago, Chicago, Illinois, United States of America
- * E-mail: (QM); (HX)
| | - Haibin Xia
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, Xi’an, Shaanxi, People’s Republic of China
- * E-mail: (QM); (HX)
| |
Collapse
|
46
|
Wirt SE, Porteus MH. Development of nuclease-mediated site-specific genome modification. Curr Opin Immunol 2012; 24:609-16. [PMID: 22981684 DOI: 10.1016/j.coi.2012.08.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2012] [Accepted: 08/10/2012] [Indexed: 11/30/2022]
Abstract
Genome engineering is an emerging strategy to treat monogenic diseases that relies on the use of engineered nucleases to correct mutations at the nucleotide level. Zinc finger nucleases can be designed to stimulate homologous recombination-mediated gene targeting at a variety of loci, including genes known to cause the primary immunodeficiencies (PIDs). Recently, these nucleases have been used to correct disease-causing mutations in human cells, as well as to create new animal models for human disease. Although a number of hurdles remain before they can be used clinically, engineered nucleases hold increasing promise as a therapeutic tool, particularly for the PIDs.
Collapse
Affiliation(s)
- Stacey E Wirt
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | |
Collapse
|
47
|
Ansai S, Ochiai H, Kanie Y, Kamei Y, Gou Y, Kitano T, Yamamoto T, Kinoshita M. Targeted disruption of exogenous EGFP gene in medaka using zinc-finger nucleases. Dev Growth Differ 2012; 54:546-56. [PMID: 22642582 DOI: 10.1111/j.1440-169x.2012.01357.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2012] [Revised: 04/09/2012] [Accepted: 04/18/2012] [Indexed: 01/15/2023]
Abstract
Zinc-finger nucleases (ZFNs) are artificial enzymes that create site-specific double-strand breaks and thereby induce targeted genome editing. Here, we demonstrated successful gene disruption in somatic and germ cells of medaka (Oryzias latipes) using ZFN to target exogenous EGFP genes. Embryos that were injected with an RNA sequence pair coding for ZFNs showed mosaic loss of green fluorescent protein fluorescence in skeletal muscle. A number of mutations that included both deletions and insertions were identified within the ZFN target site in each embryo, whereas no mutations were found at the non-targeted sites. In addition, ZFN-induced mutations were introduced in germ cells and efficiently transmitted to the next generation. The mutation frequency varied (6-100%) in the germ cells from each founder, and a founder carried more than two types of mutation in germ cells. Our results have introduced the possibility of targeted gene disruption and reverse genetics in medaka.
Collapse
Affiliation(s)
- Satoshi Ansai
- Division of Applied Biosciences, Graduate School of Agriculture, Kyoto University, Kitashirakawa-Oiwake-cho, Sakyo-ku, Kyoto, 606-8502, Japan
| | | | | | | | | | | | | | | |
Collapse
|
48
|
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
|
49
|
Tzfira T, Weinthal D, Marton I, Zeevi V, Zuker A, Vainstein A. Genome modifications in plant cells by custom-made restriction enzymes. PLANT BIOTECHNOLOGY JOURNAL 2012; 10:373-89. [PMID: 22469004 DOI: 10.1111/j.1467-7652.2011.00672.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Genome editing, i.e. the ability to mutagenize, insert, delete and replace sequences, in living cells is a powerful and highly desirable method that could potentially revolutionize plant basic research and applied biotechnology. Indeed, various research groups from academia and industry are in a race to devise methods and develop tools that will enable not only site-specific mutagenesis but also controlled foreign DNA integration and replacement of native and transgene sequences by foreign DNA, in living plant cells. In recent years, much of the progress seen in gene targeting in plant cells has been attributed to the development of zinc finger nucleases and other novel restriction enzymes for use as molecular DNA scissors. The induction of double-strand breaks at specific genomic locations by zinc finger nucleases and other novel restriction enzymes results in a wide variety of genetic changes, which range from gene addition to the replacement, deletion and site-specific mutagenesis of endogenous and heterologous genes in living plant cells. In this review, we discuss the principles and tools for restriction enzyme-mediated gene targeting in plant cells, as well as their current and prospective use for gene targeting in model and crop plants.
Collapse
Affiliation(s)
- Tzvi Tzfira
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel.
| | | | | | | | | | | |
Collapse
|
50
|
Porteus M. Homologous recombination-based gene therapy for the primary immunodeficiencies. Ann N Y Acad Sci 2012; 1246:131-40. [PMID: 22236437 DOI: 10.1111/j.1749-6632.2011.06314.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The devastating nature of primary immunodeficiencies, the ability to cure primary immunodeficiencies by bone marrow transplantation, the ability of a small number of gene-corrected cells to reconstitute the immune system, and the overall suboptimal results of bone marrow transplantation for most patients with primary immunodeficiencies make the development of gene therapy for this class of diseases important. While there has been clear clinical benefit for a number of patients from viral-based gene therapy strategies, there have also been a significant number of serious adverse events, including the development of leukemia, from the approach. In this review, I discuss the development of nuclease-stimulated, homologous recombination-based approaches as a novel gene therapy strategy for the primary immunodeficiencies.
Collapse
Affiliation(s)
- Matthew Porteus
- Department of Pediatrics, Divisions of Cancer Biology, Hematology/Oncology, Human Gene Therapy, Stanford University, Stanford, California, USA.
| |
Collapse
|