scispace - formally typeset
Search or ask a question
Author

Jiyeon Kweon

Other affiliations: Seoul National University
Bio: Jiyeon Kweon is an academic researcher from University of Ulsan. The author has contributed to research in topics: CRISPR & Cas9. The author has an hindex of 13, co-authored 21 publications receiving 2439 citations. Previous affiliations of Jiyeon Kweon include Seoul National University.

Papers
More filters
Journal ArticleDOI
TL;DR: Off-target effects of RGENs can be reduced below the detection limits of deep sequencing by choosing unique target sequences in the genome and modifying both guide RNA and Cas9, and paired nickases induced chromosomal deletions in a targeted manner without causing unwanted translocations.
Abstract: RNA-guided endonucleases (RGENs), derived from the prokaryotic adaptive immune system known as CRISPR/Cas, enable targeted genome engineering in cells and organisms. RGENs are ribonucleoproteins that consist of guide RNA and Cas9, a protein component originated from Streptococcus pyogenes. These enzymes cleave chromosomal DNA, whose sequence is complementary, to guide RNA in a targeted manner, producing site-specific DNA double-strand breaks (DSBs), the repair of which gives rise to targeted genome modifications. Despite broad interest in RGEN-mediated genome editing, these nucleases are limited by off-target mutations and unwanted chromosomal translocations associated with off-target DNA cleavages. Here, we show that off-target effects of RGENs can be reduced below the detection limits of deep sequencing by choosing unique target sequences in the genome and modifying both guide RNA and Cas9. We found that both the composition and structure of guide RNA can affect RGEN activities in cells to reduce off-target effects. RGENs efficiently discriminated on-target sites from off-target sites that differ by two bases. Furthermore, exome sequencing analysis showed that no off-target mutations were induced by two RGENs in four clonal populations of mutant cells. In addition, paired Cas9 nickases, composed of D10A Cas9 and guide RNA, which generate two single-strand breaks (SSBs) or nicks on different DNA strands, were highly specific in human cells, avoiding off-target mutations without sacrificing genome-editing efficiency. Interestingly, paired nickases induced chromosomal deletions in a targeted manner without causing unwanted translocations. Our results highlight the importance of choosing unique target sequences and optimizing guide RNA and Cas9 to avoid or reduce RGEN-induced off-target mutations.

1,332 citations

Journal ArticleDOI
TL;DR: A genome-scale collection of TALENs for efficient and scalable gene targeting in human cells is presented and single- and double-gene-knockout cells in which NF-κB signaling pathways were disrupted showed unambiguous suppression of signal transduction.
Abstract: A collection of TALENs targeted to 18,740 human protein-coding genes will facilitate genetic engineering of human cells.

382 citations

Journal ArticleDOI
TL;DR: A simple formula and a computer program are developed to predict the deletion patterns at a given nuclease target site that are associated with microhomology of at least two bases, which can be predicted to achieve efficient gene disruption in cell lines and whole organisms.
Abstract: To the Editor: Programmable nucleases such as Cas9 RNA-guided engineered nucleases (RGENs)1 enable gene knockout in cultured cells and organisms by producing site-specific DNA double-strand breaks, whose repair via error-prone nonhomologous end joining gives rise to small insertions and deletions (indels) at target sites, often causing frameshift mutations in a protein-coding sequence2. The efficiency of this method can be reduced by in-frame mutations via microhomology-mediated end joining3,4 (Fig. 1a). Here we present a computer program that assists in the choice of Cas9 nuclease, zinc-finger nuclease and transcription activator–like effector nuclease (TALEN) target sites, using microhomology prediction to achieve efficient gene disruption in cell lines and whole organisms. First we examined the mutations induced by ten TALENs and ten RGENs in human cells via deep sequencing (Supplementary Table 1 and Supplementary Methods). We focused our analysis on deletions because they are much more prevalent than insertions (98.7% vs. 1.3%, respectively, for TALENs; 75.1% vs. 24.9% for RGENs) and because microhomology is irrelevant for insertions. In aggregate, microhomologies of 2–8 bases were found in 44.3% and 52.7% of all deletions induced by TALENs and RGENs, respectively (Supplementary Fig. 1 and Supplementary Table 2). Thus, 43.7% (0.987 × 0.443) and 39.6% (0.751 × 0.527) of all the mutations induced by TALENs and RGENs, respectively, were associated with microhomology. At a given nuclease target site, the effect of these microhomologyassociated deletions can be predicted. In the extreme cases, (i) all deletions cause frameshifts in a protein-coding gene or (ii) no deletions cause frameshifts. In contrast, one-third of microhomologyindependent deletions result in in-frame mutations. Assuming that ~60% of indels are microhomology independent on average, the fraction of in-frame mutations at a given site can range from 20% (60%/3 + 0%) to 60% (60%/3 + 40%), a threefold difference between the two extreme cases. Because most eukaryotic cells are diploid rather than haploid, the fraction of null cells carrying two outof-frame mutations can range from 16% (0.40 × 0.40) to 64% (0.80 × 0.80), depending on the choice of target site. A careful analysis of indel sequences also revealed that the frequency of microhomology-associated deletions depends on both the size of the microhomology and the length of the deletion (Supplementary Fig. 2). On the basis of these observations, we developed a simple formula and a computer program (Supplementary Fig. 3) to predict the deletion patterns at a given nuclease target site that are associated with microhomology of at least two bases (Fig. 1b and Supplementary Note). We assigned a pattern score to each deletion pattern and a microhomology score (equaling the sum of pattern scores) to each target site. We then obtained an out-of-frame score at a given site by dividing the sum of pattern scores assigned to frameshifting deletions by the microhomology score. To evaluate the utility of our scoring system, we arbitrarily chose two target sites in exons, one with a high score (top 20%) and the other with a low score (bottom 20%) in each of nine human genes. We targeted a total of 6 and 12 sites in human cells with RGENs and TALENs, respectively (Supplementary Table 3), and then analyzed the mutant patterns by deep sequencing (Supplementary Table 4). High-score sites produced out-of-frame indels much more frequently than did low-score sites in all nine pairs (Fig. 1c), even at two adjacent target sites separated by 29 bases in the MCM6 gene (Supplementary Fig. 4). On average, the high-score sites and low-score sites produced frameshifting indels at frequencies of 79.3% and 42.5%, respectively (Student’s t-test, P < 0.01). We then tested in HeLa cells 68 new RGENs that target different genes (Supplementary Table 5). Again, out-of-frame scores correlated well with the frequencies of frameshifting indels or deletions (Pearson coefficient = 0.717 and 0.732, respectively) (Fig. 1d and Supplementary Fig. 5). The frequencies of out-of-frame indels ranged from 38.7% to 94.0%. In a diploid human cell, the probability of obtaining null clones would thus range from 15.0% (0.387 × 0.387) to 88.4%. Most cancer cell lines including HeLa are multi-ploid (>3n), making it even more important to choose high-score sites. We expect that the scoring system would work even better for TALENs because TALENs induce microhomology-independent insertions much less frequently than do RGENs (Supplementary Fig. 1b). We also analyzed the genotypes of 81 mice carrying mutations produced via TALENs5 or RGENs6, from our previous studies. The frequencies of out-of-frame deletions correlated well with predicted scores (Pearson coefficient = 0.996; Supplementary Fig. 6). In summary, we developed a scoring system to estimate the frequency of microhomology-associated deletions at nuclease

330 citations

Journal ArticleDOI
TL;DR: It is found that zinc finger nucleases designed to target two different sites in a human chromosome could introduce two concurrent double-strand breaks, whose repair via non-homologous end-joining (NHEJ) gives rise to targeted duplications and inversions of the genomic segments of up to a mega base pair in length between the two sites.
Abstract: Despite the recent discoveries of and interest in numerous structural variations (SVs)—which include duplications and inversions—in the human and other higher eukaryotic genomes, little is known about the etiology and biology of these SVs, partly due to the lack of molecular tools with which to create individual SVs in cultured cells and model organisms. Here, we present a novel method of inducing duplications and inversions in a targeted manner without pre-manipulation of the genome. We found that zinc finger nucleases (ZFNs) designed to target two different sites in a human chromosome could introduce two concurrent double-strand breaks, whose repair via non-homologous end-joining (NHEJ) gives rise to targeted duplications and inversions of the genomic segments of up to a mega base pair (bp) in length between the two sites. Furthermore, we demonstrated that a ZFN pair could induce the inversion of a 140-kbp chromosomal segment that contains a portion of the blood coagulation factor VIII gene to mimic the inversion genotype that is associated with some cases of severe hemophilia A. This same ZFN pair could be used, in theory, to revert the inverted region to restore genomic integrity in these hemophilia A patients. We propose that ZFNs can be employed as molecular tools to study mechanisms of chromosomal rearrangements and to create SVs in a predetermined manner so as to study their biological roles. In addition, our method raises the possibility of correcting genetic defects caused by chromosomal rearrangements and holds new promise in gene and cell therapy.

179 citations

Journal ArticleDOI
TL;DR: It is shown that TALENs can be used both for creating disease models associated with chromosomal rearrangements in iPSCs and for correcting genetic defects caused by chromosomal inversions, as an iPSC-based novel therapeutic option for the treatment of hemophilia A.
Abstract: Hemophilia A, one of the most common genetic bleeding disorders, is caused by various mutations in the blood coagulation factor VIII (F8) gene. Among the genotypes that result in hemophilia A, two different types of chromosomal inversions that involve a portion of the F8 gene are most frequent, accounting for almost half of all severe hemophilia A cases. In this study, we used a transcription activator-like effector nuclease (TALEN) pair to invert a 140-kbp chromosomal segment that spans the portion of the F8 gene in human induced pluripotent stem cells (iPSCs) to create a hemophilia A model cell line. In addition, we reverted the inverted segment back to its normal orientation in the hemophilia model iPSCs using the same TALEN pair. Importantly, we detected the F8 mRNA in cells derived from the reverted iPSCs lines, but not in those derived from the clones with the inverted segment. Thus, we showed that TALENs can be used both for creating disease models associated with chromosomal rearrangements in iPSCs and for correcting genetic defects caused by chromosomal inversions. This strategy provides an iPSC-based novel therapeutic option for the treatment of hemophilia A and other genetic diseases caused by chromosomal inversions.

139 citations


Cited by
More filters
Journal ArticleDOI
28 Nov 2014-Science
TL;DR: The power of the CRISPR-Cas9 technology to systematically analyze gene functions in mammalian cells, study genomic rearrangements and the progression of cancers or other diseases, and potentially correct genetic mutations responsible for inherited disorders is illustrated.
Abstract: The advent of facile genome engineering using the bacterial RNA-guided CRISPR-Cas9 system in animals and plants is transforming biology. We review the history of CRISPR (clustered regularly interspaced palindromic repeat) biology from its initial discovery through the elucidation of the CRISPR-Cas9 enzyme mechanism, which has set the stage for remarkable developments using this technology to modify, regulate, or mark genomic loci in a wide variety of cells and organisms from all three domains of life. These results highlight a new era in which genomic manipulation is no longer a bottleneck to experiments, paving the way toward fundamental discoveries in biology, with applications in all branches of biotechnology, as well as strategies for human therapeutics.

4,774 citations

Journal ArticleDOI
TL;DR: A review of achievements made possible by site-specific nuclease technologies and applications of these reagents for genetic analysis and manipulation, including the therapeutic potential of ZFNs and TALENs, and future prospects for the field are discussed.

3,235 citations

01 Jun 2005

3,154 citations

Journal ArticleDOI
TL;DR: A modified version of the CRISPR-Cas9 system has been developed to recruit heterologous domains that can regulate endogenous gene expression or label specific genomic loci in living cells, which will undoubtedly transform biological research and spur the development of novel molecular therapeutics for human disease.
Abstract: Targeted genome editing using engineered nucleases has rapidly gone from being a niche technology to a mainstream method used by many biological researchers. This widespread adoption has been largely fueled by the emergence of the clustered, regularly interspaced, short palindromic repeat (CRISPR) technology, an important new approach for generating RNA-guided nucleases, such as Cas9, with customizable specificities. Genome editing mediated by these nucleases has been used to rapidly, easily and efficiently modify endogenous genes in a wide variety of biomedically important cell types and in organisms that have traditionally been challenging to manipulate genetically. Furthermore, a modified version of the CRISPR-Cas9 system has been developed to recruit heterologous domains that can regulate endogenous gene expression or label specific genomic loci in living cells. Although the genome-wide specificities of CRISPR-Cas9 systems remain to be fully defined, the power of these systems to perform targeted, highly efficient alterations of genome sequence and gene expression will undoubtedly transform biological research and spur the development of novel molecular therapeutics for human disease.

2,930 citations

Journal ArticleDOI
TL;DR: Recently devised sgRNA design rules are used to create human and mouse genome-wide libraries, perform positive and negative selection screens and observe that the use of these rules produced improved results, and a metric to predict off-target sites is developed.
Abstract: CRISPR-Cas9-based genetic screens are a powerful new tool in biology. By simply altering the sequence of the single-guide RNA (sgRNA), one can reprogram Cas9 to target different sites in the genome with relative ease, but the on-target activity and off-target effects of individual sgRNAs can vary widely. Here, we use recently devised sgRNA design rules to create human and mouse genome-wide libraries, perform positive and negative selection screens and observe that the use of these rules produced improved results. Additionally, we profile the off-target activity of thousands of sgRNAs and develop a metric to predict off-target sites. We incorporate these findings from large-scale, empirical data to improve our computational design rules and create optimized sgRNA libraries that maximize on-target activity and minimize off-target effects to enable more effective and efficient genetic screens and genome engineering.

2,866 citations