Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Kosuke Yusa is active.

Publication


Featured researches published by Kosuke Yusa.


Nature Biotechnology | 2014

Genome-wide recessive genetic screening in mammalian cells with a lentiviral CRISPR-guide RNA library

Hiroko Koike-Yusa; Yilong Li; E-Pien Tan; Martin Del Castillo Velasco-Herrera; Kosuke Yusa

Identification of genes influencing a phenotype of interest is frequently achieved through genetic screening by RNA interference (RNAi) or knockouts. However, RNAi may only achieve partial depletion of gene activity, and knockout-based screens are difficult in diploid mammalian cells. Here we took advantage of the efficiency and high throughput of genome editing based on type II, clustered, regularly interspaced, short palindromic repeats (CRISPR)–CRISPR-associated (Cas) systems to introduce genome-wide targeted mutations in mouse embryonic stem cells (ESCs). We designed 87,897 guide RNAs (gRNAs) targeting 19,150 mouse protein-coding genes and used a lentiviral vector to express these gRNAs in ESCs that constitutively express Cas9. Screening the resulting ESC mutant libraries for resistance to either Clostridium septicum alpha-toxin or 6-thioguanine identified 27 known and 4 previously unknown genes implicated in these phenotypes. Our results demonstrate the potential for efficient loss-of-function screening using the CRISPR-Cas9 system.


Nature Methods | 2009

Generation of transgene-free induced pluripotent mouse stem cells by the piggyBac transposon

Kosuke Yusa; Roland Rad; Junji Takeda; Allan Bradley

Induced pluripotent stem cells (iPSCs) have been generated from somatic cells by transgenic expression of Oct4 (Pou5f1), Sox2, Klf4 and Myc. A major difficulty in the application of this technology for regenerative medicine, however, is the delivery of reprogramming factors. Whereas retroviral transduction increases the risk of tumorigenicity, transient expression methods have considerably lower reprogramming efficiencies. Here we describe an efficient piggyBac transposon–based approach to generate integration-free iPSCs. Transposons carrying 2A peptide–linked reprogramming factors induced reprogramming of mouse embryonic fibroblasts with equivalent efficiencies to retroviral transduction. We removed transposons from these primary iPSCs by re-expressing transposase. Transgene-free iPSCs could be identified by negative selection. piggyBac excised without a footprint, leaving the iPSC genome without any genetic alteration. iPSCs fulfilled all criteria of pluripotency, such as pluripotency gene expression, teratoma formation and contribution to chimeras. piggyBac transposon–based reprogramming may be used to generate therapeutically applicable iPSCs.


Nature | 2011

Targeted gene correction of α1-antitrypsin deficiency in induced pluripotent stem cells.

Kosuke Yusa; S. Tamir Rashid; Helene Strick-Marchand; Ignacio Varela; Pei Qi Liu; David Paschon; Elena Miranda; Adriana Ordóñez; Nicholas Hannan; Foad Rouhani; Sylvie Darche; Graeme J. M. Alexander; Stefan J. Marciniak; Noemi Fusaki; Mamoru Hasegawa; Michael C. Holmes; James P. Di Santo; David A. Lomas; Allan Bradley; Ludovic Vallier

Human induced pluripotent stem cells (iPSCs) represent a unique opportunity for regenerative medicine because they offer the prospect of generating unlimited quantities of cells for autologous transplantation, with potential application in treatments for a broad range of disorders. However, the use of human iPSCs in the context of genetically inherited human disease will require the correction of disease-causing mutations in a manner that is fully compatible with clinical applications. The methods currently available, such as homologous recombination, lack the necessary efficiency and also leave residual sequences in the targeted genome. Therefore, the development of new approaches to edit the mammalian genome is a prerequisite to delivering the clinical promise of human iPSCs. Here we show that a combination of zinc finger nucleases (ZFNs) and piggyBac technology in human iPSCs can achieve biallelic correction of a point mutation (Glu342Lys) in the α1-antitrypsin (A1AT, also known as SERPINA1) gene that is responsible for α1-antitrypsin deficiency. Genetic correction of human iPSCs restored the structure and function of A1AT in subsequently derived liver cells in vitro and in vivo. This approach is significantly more efficient than any other gene-targeting technology that is currently available and crucially prevents contamination of the host genome with residual non-human sequences. Our results provide the first proof of principle, to our knowledge, for the potential of combining human iPSCs with genetic correction to generate clinically relevant cells for autologous cell-based therapies.


Stem Cells | 2010

Butyrate Greatly Enhances Derivation of Human Induced Pluripotent Stem Cells by Promoting Epigenetic Remodeling and the Expression of Pluripotency-Associated Genes

Prashant Mali; Bin Kuan Chou; Jonathan Yen; Zhaohui Ye; Jizhong Zou; Sarah N. Dowey; Robert A. Brodsky; Joyce E. Ohm; Wayne Yu; Stephen B. Baylin; Kosuke Yusa; Allan Bradley; David J. Meyers; Chandrani Mukherjee; Philip A. Cole; Linzhao Cheng

We report here that butyrate, a naturally occurring fatty acid commonly used as a nutritional supplement and differentiation agent, greatly enhances the efficiency of induced pluripotent stem (iPS) cell derivation from human adult or fetal fibroblasts. After transient butyrate treatment, the iPS cell derivation efficiency is enhanced by 15‐ to 51‐fold using either retroviral or piggyBac transposon vectors expressing 4 to 5 reprogramming genes. Butyrate stimulation is more remarkable (>100‐ to 200‐fold) on reprogramming in the absence of either KLF4 or MYC transgene. Butyrate treatment did not negatively affect properties of iPS cell lines established by either 3 or 4 retroviral vectors or a single piggyBac DNA transposon vector. These characterized iPS cell lines, including those derived from an adult patient with sickle cell disease by either the piggyBac or retroviral vectors, show normal karyotypes and pluripotency. To gain insights into the underlying mechanisms of butyrate stimulation, we conducted genome‐wide gene expression and promoter DNA methylation microarrays and other epigenetic analyses on established iPS cells and cells from intermediate stages of the reprogramming process. By days 6 to 12 during reprogramming, butyrate treatment enhanced histone H3 acetylation, promoter DNA demethylation, and the expression of endogenous pluripotency‐associated genes, including DPPA2, whose overexpression partially substitutes for butyrate stimulation. Thus, butyrate as a cell permeable small molecule provides a simple tool to further investigate molecular mechanisms of cellular reprogramming. Moreover, butyrate stimulation provides an efficient method for reprogramming various human adult somatic cells, including cells from patients that are more refractory to reprogramming. STEM CELLS 2010;28:713–72028:713–720


Proceedings of the National Academy of Sciences of the United States of America | 2011

A hyperactive piggyBac transposase for mammalian applications

Kosuke Yusa; Liqin Zhou; Meng Amy Li; Allan Bradley; Nancy L. Craig

DNA transposons have been widely used for transgenesis and insertional mutagenesis in various organisms. Among the transposons active in mammalian cells, the moth-derived transposon piggyBac is most promising with its highly efficient transposition, large cargo capacity, and precise repair of the donor site. Here we report the generation of a hyperactive piggyBac transposase. The active transposition of piggyBac in multiple organisms allowed us to screen a transposase mutant library in yeast for hyperactive mutants and then to test candidates in mouse ES cells. We isolated 18 hyperactive mutants in yeast, among which five were also hyperactive in mammalian cells. By combining all mutations, a total of 7 aa substitutions, into a single reading frame, we generated a unique hyperactive piggyBac transposase with 17-fold and ninefold increases in excision and integration, respectively. We showed its applicability by demonstrating an increased efficiency of generation of transgene-free mouse induced pluripotent stem cells. We also analyzed whether this hyperactive piggyBac transposase affects the genomic integrity of the host cells. The frequency of footprints left by the hyperactive piggyBac transposase was as low as WT transposase (~1%) and we found no evidence that the expression of the transposase affects genomic integrity. This hyperactive piggyBac transposase expands the utility of the piggyBac transposon for applications in mammalian genetics and gene therapy.


Science | 2010

PiggyBac Transposon Mutagenesis: A Tool for Cancer Gene Discovery in Mice

Roland Rad; Lena Rad; Wei Wang; Juan Cadiñanos; George S. Vassiliou; Stephen A. Rice; Lia S. Campos; Kosuke Yusa; Ruby Banerjee; Meng Amy Li; Jorge de la Rosa; Alexander Strong; Dong Lu; Peter Ellis; Nathalie Conte; Fang Tang Yang; Pentao Liu; Allan Bradley

Piggybacking on Cancer Genes Transposons are mobile segments of DNA that can insert in or near important genes to cause mutations that disrupt gene function. Rad et al. (p. 1104, published online 14 October) adapted a mutagenic transposon called Piggybac, originally derived from a moth, into a tool for discovery of cancer-causing genes in mice. Mobilization of Piggybac in mice was associated with the development of leukemias and solid tumors. In many instances the causative mutations, which were identified by mapping the Piggybac integration sites, were within genes not previously implicated in cancer. Mutations induced by a transposable element in mice can be used to identify cancer-causing genes. Transposons are mobile DNA segments that can disrupt gene function by inserting in or near genes. Here, we show that insertional mutagenesis by the PiggyBac transposon can be used for cancer gene discovery in mice. PiggyBac transposition in genetically engineered transposon-transposase mice induced cancers whose type (hematopoietic versus solid) and latency were dependent on the regulatory elements introduced into transposons. Analysis of 63 hematopoietic tumors revealed that PiggyBac is capable of genome-wide mutagenesis. The PiggyBac screen uncovered many cancer genes not identified in previous retroviral or Sleeping Beauty transposon screens, including Spic, which encodes a PU.1-related transcription factor, and Hdac7, a histone deacetylase gene. PiggyBac and Sleeping Beauty have different integration preferences. To maximize the utility of the tool, we engineered 21 mouse lines to be compatible with both transposon systems in constitutive, tissue- or temporal-specific mutagenesis. Mice with different transposon types, copy numbers, and chromosomal locations support wide applicability.


Molecular and Cellular Biology | 2003

Characterization of Sleeping Beauty Transposition and Its Application to Genetic Screening in Mice

Kyoji Horie; Kosuke Yusa; Kojiro Yae; Junko Odajima; Sylvia E. J. Fischer; Vincent W. Keng; Tomoko Hayakawa; Sumi Mizuno; Gen Kondoh; Takashi W. Ijiri; Yoichi Matsuda; Ronald H.A. Plasterk; Junji Takeda

ABSTRACT The use of mutant mice plays a pivotal role in determining the function of genes, and the recently reported germ line transposition of the Sleeping Beauty (SB) transposon would provide a novel system to facilitate this approach. In this study, we characterized SB transposition in the mouse germ line and assessed its potential for generating mutant mice. Transposition sites not only were clustered within 3 Mb near the donor site but also were widely distributed outside this cluster, indicating that the SB transposon can be utilized for both region-specific and genome-wide mutagenesis. The complexity of transposition sites in the germ line was high enough for large-scale generation of mutant mice. Based on these initial results, we conducted germ line mutagenesis by using a gene trap scheme, and the use of a green fluorescent protein reporter made it possible to select for mutant mice rapidly and noninvasively. Interestingly, mice with mutations in the same gene, each with a different insertion site, were obtained by local transposition events, demonstrating the feasibility of the SB transposon system for region-specific mutagenesis. Our results indicate that the SB transposon system has unique features that complement other mutagenesis approaches.


Nucleic Acids Research | 2011

Mobilization of giant piggyBac transposons in the mouse genome

Meng Amy Li; Daniel J. Turner; Zemin Ning; Kosuke Yusa; Qi Liang; Sabine Eckert; Lena Rad; Tomas W. Fitzgerald; Nancy L. Craig; Allan Bradley

The development of technologies that allow the stable delivery of large genomic DNA fragments in mammalian systems is important for genetic studies as well as for applications in gene therapy. DNA transposons have emerged as flexible and efficient molecular vehicles to mediate stable cargo transfer. However, the ability to carry DNA fragments >10 kb is limited in most DNA transposons. Here, we show that the DNA transposon piggyBac can mobilize 100-kb DNA fragments in mouse embryonic stem (ES) cells, making it the only known transposon with such a large cargo capacity. The integrity of the cargo is maintained during transposition, the copy number can be controlled and the inserted giant transposons express the genomic cargo. Furthermore, these 100-kb transposons can also be excised from the genome without leaving a footprint. The development of piggyBac as a large cargo vector will facilitate a wider range of genetic and genomic applications.


Cancer Cell | 2013

A Genetic Progression Model of BrafV600E-Induced Intestinal Tumorigenesis Reveals Targets for Therapeutic Intervention

Roland Rad; Juan Cadiñanos; Lena Rad; Ignacio Varela; Alexander Strong; Lydia Kriegl; Fernando Constantino-Casas; Stefan Eser; Maren Hieber; Barbara Seidler; Stacey Price; Mario F. Fraga; Vincenzo Calvanese; Gary J. Hoffman; Hannes Ponstingl; Günter Schneider; Kosuke Yusa; C Grove; Roland M. Schmid; Wei Wang; George S. Vassiliou; Thomas Kirchner; Ultan McDermott; Pentao Liu; Dieter Saur; Allan Bradley

Summary We show that BRAFV600E initiates an alternative pathway to colorectal cancer (CRC), which progresses through a hyperplasia/adenoma/carcinoma sequence. This pathway underlies significant subsets of CRCs with distinctive pathomorphologic/genetic/epidemiologic/clinical characteristics. Genetic and functional analyses in mice revealed a series of stage-specific molecular alterations driving different phases of tumor evolution and uncovered mechanisms underlying this stage specificity. We further demonstrate dose-dependent effects of oncogenic signaling, with physiologic BrafV600E expression being sufficient for hyperplasia induction, but later stage intensified Mapk-signaling driving both tumor progression and activation of intrinsic tumor suppression. Such phenomena explain, for example, the inability of p53 to restrain tumor initiation as well as its importance in invasiveness control, and the late stage specificity of its somatic mutation. Finally, systematic drug screening revealed sensitivity of this CRC subtype to targeted therapeutics, including Mek or combinatorial PI3K/Braf inhibition.


Cell Reports | 2016

A CRISPR Dropout Screen Identifies Genetic Vulnerabilities and Therapeutic Targets in Acute Myeloid Leukemia

Konstantinos Tzelepis; Hiroko Koike-Yusa; Etienne De Braekeleer; Yilong Li; Emmanouil Metzakopian; Oliver M. Dovey; Annalisa Mupo; Vera Grinkevich; Meng Li; Milena Mazan; Malgorzata Gozdecka; Shuhei Ohnishi; Jonathan L. Cooper; Miten Patel; Thomas McKerrell; Bin Chen; Ana Filipa Domingues; Paolo Gallipoli; Sarah A. Teichmann; Hannes Ponstingl; Ultan McDermott; Julio Saez-Rodriguez; Brian J. P. Huntly; Francesco Iorio; Cristina Pina; George S. Vassiliou; Kosuke Yusa

Summary Acute myeloid leukemia (AML) is an aggressive cancer with a poor prognosis, for which mainstream treatments have not changed for decades. To identify additional therapeutic targets in AML, we optimize a genome-wide clustered regularly interspaced short palindromic repeats (CRISPR) screening platform and use it to identify genetic vulnerabilities in AML cells. We identify 492 AML-specific cell-essential genes, including several established therapeutic targets such as DOT1L, BCL2, and MEN1, and many other genes including clinically actionable candidates. We validate selected genes using genetic and pharmacological inhibition, and chose KAT2A as a candidate for downstream study. KAT2A inhibition demonstrated anti-AML activity by inducing myeloid differentiation and apoptosis, and suppressed the growth of primary human AMLs of diverse genotypes while sparing normal hemopoietic stem-progenitor cells. Our results propose that KAT2A inhibition should be investigated as a therapeutic strategy in AML and provide a large number of genetic vulnerabilities of this leukemia that can be pursued in downstream studies.

Collaboration


Dive into the Kosuke Yusa's collaboration.

Top Co-Authors

Avatar

Junji Takeda

Tokyo Medical and Dental University

View shared research outputs
Top Co-Authors

Avatar

Allan Bradley

Wellcome Trust Sanger Institute

View shared research outputs
Top Co-Authors

Avatar

Kyoji Horie

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Francesco Iorio

European Bioinformatics Institute

View shared research outputs
Top Co-Authors

Avatar

George S. Vassiliou

Wellcome Trust Sanger Institute

View shared research outputs
Top Co-Authors

Avatar

Hiroko Koike-Yusa

Wellcome Trust Sanger Institute

View shared research outputs
Top Co-Authors

Avatar

Yilong Li

Wellcome Trust Sanger Institute

View shared research outputs
Top Co-Authors

Avatar

Mathew J. Garnett

Wellcome Trust Sanger Institute

View shared research outputs
Top Co-Authors

Avatar

Ultan McDermott

Wellcome Trust Sanger Institute

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge